NZ626580B2 - Variant activin receptor polypeptides, alone or in combination with chemotherapy, and uses thereof - Google Patents

Variant activin receptor polypeptides, alone or in combination with chemotherapy, and uses thereof Download PDF

Info

Publication number
NZ626580B2
NZ626580B2 NZ626580A NZ62658012A NZ626580B2 NZ 626580 B2 NZ626580 B2 NZ 626580B2 NZ 626580 A NZ626580 A NZ 626580A NZ 62658012 A NZ62658012 A NZ 62658012A NZ 626580 B2 NZ626580 B2 NZ 626580B2
Authority
NZ
New Zealand
Prior art keywords
vactriib
polypeptide
seq
set forth
amino acid
Prior art date
Application number
NZ626580A
Other versions
NZ626580A (en
Inventor
Huiquan Han
Keith Soo Nyung Kwak
John Lu
Jeonghoon Sun
Lei Ting Tony Tam
Xiaolan Zhou
Original Assignee
Amgen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US13/329,897 external-priority patent/US8501678B2/en
Application filed by Amgen Inc filed Critical Amgen Inc
Priority claimed from PCT/US2012/070571 external-priority patent/WO2013106175A1/en
Publication of NZ626580A publication Critical patent/NZ626580A/en
Publication of NZ626580B2 publication Critical patent/NZ626580B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/655Azo (—N=N—), diazo (=N2), azoxy (>N—O—N< or N(=O)—N<), azido (—N3) or diazoamino (—N=N—N<) compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/179Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators

Abstract

Disclosed is a pharmaceutical composition comprising (i) an isolated protein comprising a variant activin IIB receptor polypeptide (vActRIIB), wherein vActRIIB comprises a polypeptide sequence having at least 95% identity to the amino acid sequence set forth at amino acids 25 through 134 of the sequence set forth in SEQ ID NO:18, wherein the polypeptide comprises an amino acid substitution at position 28, and wherein the polypeptide is capable of binding myostatin, activin A, or GDF-11, and (ii) a chemotherapeutic agent. Also disclosed is the use of such compositions in the manufacture of a medicament for treating cancer. ence set forth in SEQ ID NO:18, wherein the polypeptide comprises an amino acid substitution at position 28, and wherein the polypeptide is capable of binding myostatin, activin A, or GDF-11, and (ii) a chemotherapeutic agent. Also disclosed is the use of such compositions in the manufacture of a medicament for treating cancer.

Description

VARIANT ACTIVIN RECEPTOR POLYPEPTIDES, ALONE OR IN COMBINATION WITH CHEMOTHERAPY, AND USES THEREOF CROSS-REFERENCE TO RELATED APPLICATIONS [000 1] This application claims the right of ty based on US. Serial No. 13/329,897, filed December 19, 2011, the entire disclosure of which is herein incorporated by reference, in its entirety, for all purposes.
This application is d to US. Serial No. 13/080,515, filed April 5, 2011, now pending, which is a divisional of US. Serial No. 12/074,877, filed March 5, 2008, now d as U. S. Patent No. 7,947,646 which claims the benefit of United States provisional application serial number 61/065,474, filed February 11, 2008, and United States provisional application serial number 60/905,459, filed March 6, 2007, the disclosures of which are relied upon and incorporated by reference herein.
REFERENCE TO THE SEQUENCE LISTING The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled AUS- CIP_SeqList.txt which is 265,085 bytes in size, and was created December 19, 2011. The information in the electronic format of the Sequence Listing is orated herein by reference in its entirety FIELD OF THE INVENTION The technical field of this invention relates to transforming growth factor-B (TGF-B) family s and soluble TGF-B receptors, as well as methods of modulating the activities of TGF—B family members for the treatment of various disorders.
OUND OF THE INVENTION The orming growth factor B (TGF-B) family of proteins includes the orming growth factors-B (TGF-B), ns, bone morphogenic ns (BMP), nerve growth factors (NGFs), brain-derived neurotrophic factor (BDNF), and growth/differentiation factors (GDFs). These family members are involved in the tion of a wide range of biological processes including cell proliferation, differentiation, and other functions.
Growth/differentiation factor 8 (GDF-8), also referred to as myostatin, is a TGF-B family member expressed for the most part in the cells of developing and adult skeletal muscle tissue. Myostatin appears to play an essential role in negatively controlling skeletal muscle growth (McPherron et al., Nature (London) 387, 83-90 ). Antagonizing myostatin has been shown to increase lean muscle mass in animals (McFerron et al., supra, s et al., Science 296:1486 (2002)).
Another member of the TGF-B family of proteins is a related growth/ differentiation factor, GDF-11. GDF-11 has imately 90% identity of the amino acid sequence of myostatin. GDF-11 has a role in the axial patterning in developing animals (Oh et al, Genes Dev 11:1812-26 (1997)), and also appears to play a role in skeletal muscle development and .
Activins A, B and AB are the homodimers and heterdimer respectively of two polypeptide chains, BA and BB (Vale et al. Nature 321, 776-779 (1986), Ling et al., Nature 321, 779-782 ). Activins were originally discovered as gonadal peptides ed in the tion of follicle stimulating hormone synthesis, and are now believed to be involved in the regulation of a number of biological ties. Activin A is a predominant form of activin.
Activin, myostatin, GDF-11 and other members of the TGF- B superfamily bind and signal through a combination of activin type II and activin type IIB receptors, both of which are transmembrane serine/threonine s son et al., J. Biol. Chem. 279, 28036- 28044 (2004)). linking studies have determined that myostatin is capable of binding the activin type 11 receptors ActRIIA and ActRIIB in vitro (Lee et al., PNAS USA 98:9306-11 (2001)). There is also evidence that GDF-11 binds to both ActRIIA and ActRIIB (Oh et al., Genes Dev 16:2749-54 ). [00 1 0] TGF-B protein expression is known to be associated with a variety of diseases and disorders. ore, therapeutic molecules capable of antagonizing several TGF-B proteins simultaneously may be particularly effective for these diseases and disorders. [00 1 1] In addition, the production of protein therapeutics can be complicated by problems ing during the expression and purification of the protein. One problem is the aggregation of proteins during sion or purif1cation. The accumulation of high levels of protein during cell culture conditions may lead to aggregation. Purif1cation processes may expose the protein to onal factors promoting further aggregation (Cromwell, M.E.M. et al., The AAPS Journal 8:E572-E579, 2006). Attempts can be made to te the factors that cause aggregation, however, a need exists for proteins designed to have a decreased tendency to form aggregates. The present invention fillfillS the need for therapeutic molecules that bind to multiple ligands, and have reduced aggregation and thus improved manufacturability, in order to efficiently produce proteins useful for treating TGF-B related disease states.
SUMMARY OF THE INVENTION [00 1 2] The present ion provides an isolated protein sing a variant human activin receptor IIB (designated vActRIIB or sActRIIB) polypeptide. As used herein the term vActRIIB polypeptide refers to both human vActRIIB polypeptides and human vActRIIBS polypeptides. In one embodiment, the protein comprises a polypeptide having an amino acid sequence of SEQ ID NOS: 2 or 18 in which amino acids at either position E28 or R40, or both position E28 and R40 are substituted with another non-native amino acid, and wherein the polypeptide is capable of binding myostatin, activin A, or GDF-l I. In one embodiment the protein comprises a polypeptide having an amino acid sequence of SEQ ID NOS: 2 or 18 in which the amino acids at positions E28 or R40, or both E28 and R40 are substituted with a non- native amino acid, and wherein the signal peptide is removed, and wherein the polypeptide is capable of binding myostatin, activin A, or GDF-l I. In one ment the protein comprises a polypeptide having an amino acid ce of SEQ ID NOS: 2 or 18 in which amino acids at positions E28 or R40, or both E28 and R40 are substituted with another amino acid, wherein the signal sequence is removed, and N-terminal of the mature polypeptide is truncated, and wherein the polypeptide is capable of binding myostatin, activin A, or GDF-l I. In one embodiment the inal mature truncated vActRIIB polypeptide lacks the N-terminal four amino acids or the N-terminal six amino acids of the mature sequence, wherein the polypeptide is capable of binding myostatin, activin A, or GDF-l I. In one embodiment, the substitution at position E28 is selected from the group consisting of W, Y and A. In a further embodiment, the substitution at position E28 is selected from the group of amino acids consisting of A, F, Q, V, I, L, M, K, H, W and Y. In a r embodiment, the substitution at position R40 is selected from the group of amino acids ting of G, Q, M, H, K and N. In a further embodiment the substitution at position E28 is selected from the group of amino acids consisting of A, F, Q, V, I, L, M, K, H, W and Y and the substitution at position R40 is selected from the group of amino acids consisting of A, G, Q, M, H, K and N. In a further embodiment the ptide further comprises a heterologous protein. In one embodiment, the logous n is an Fc domain. In a further ment, the Fc domain is a human IgG Fc domain. [00 1 3] In one embodiment, the protein ses polypeptides having an amino acid sequence set forth in SEQ ID NOS: 4, 6, 8, 10, l2, l4, 16, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 52, 54, 56, 60, 62, 64, 66, 70, 72, 87, 88, 91, 93, 95, and 97. [00 1 4] In r ment, the protein comprises a polypeptide encoded by the polynucleotide having the sequence set forth in SEQ ID NOS: 3, 5, 7, 9, ll, 13, 15, 19, 21, 23, , 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 51, 53, 55, 59, 61, 63, 65, 67, 69, 71, 92, 94, 96 or its complement. [00 1 5] In r aspect the present invention es an isolated nucleic acid molecule comprising a polynucleotide encoding a vActRIIB polypeptide. In one ment, the nucleic acid molecule comprises a polynucleotide having the c acid sequence set forth in SEQ ID NOS: 3, 5, 7, 9, 11, 13, 15, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 51, 53, 55, 59, 61, 63, 65, 69, 71, 92, 94, 96 or its complement. [00 1 6] In r embodiment, the nucleic acid le comprises a polynucleotide encoding a polypeptide consisting of the amino acid sequence set forth in the group consisting of SEQ ID NOS: 4, 6, 8, 10, 12, 14, 16, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 52, 54, 56, 60, 62, 64, 66, 68, 70, 72, 87, 88, 91, 93, 95, and 97. In a further embodiment, the nucleic acid molecule filrther comprises a transcriptional or translational regulatory sequence.
In r aspect a recombinant vector sing the vActRIIB nucleic acid molecule is ed. In r aspect, host cells comprising the recombinant vectors are provided, and methods of producing the vActRIIB polypeptides are provided. [00 1 7] The present invention fiarther provides a composition containing at least one vActRIIB polypeptide or protein of the present invention. In one embodiment, the composition is a pharmaceutical composition containing the vActRIIB polypeptide or protein in admixture with a pharmaceutically acceptable carrier. [00 1 8] In another aspect, the present invention provides a method of treating or preventing a muscle wasting disease in a t suffering from such a disorder by administering a therapeutic composition containing a vActRIIB polypeptide or n to the subject. The muscle wasting disease includes or results from, but is not limited to, the following conditions: cancer cachexia, muscular dystrophy, amyotrophic lateral sclerosis, congestive obstructive pulmonary disease, chronic heart failure, chemical ia, cachexia from HIV/AIDS, renal failure, uremia, rheumatoid arthritis, age-related sarcopenia, age-related frailty, organ atrophy, carpal tunnel syndrome, androgen deprivation, and muscle-wasting due to inactivity from prolonged bed rest, spinal chord injury, stroke, bone fracture, and aging. The muscle wasting may also result from weightlessness due to space flight, insulin resistance, muscle wasting due to burns, androgen deprivation, and other disorders. In r aspect, the present invention es a method of treating a e correlated to sion of activin A.
In one embodiment, the disease is . In another aspect, the present invention provides a method of treating a metabolic disorder comprising administering a therapeutic composition to a subject in need of such treatment, wherein the metabolic disorder is selected from bone loss, 2012/070571 diabetes, obesity, impaired e tolerance, hyperglycemia, androgen deprivation, and metabolic syndrome. In another aspect, the present invention provides a method of gene therapy comprising administering a vector encoding a vActRIIB polypeptide or protein of the present invention to a subject in need thereof, wherein the vector is capable of expressing the vActRIIB polypeptide or protein in the subject. [00 1 9] In r ment, a pharmaceutical composition is provided comprising i) a variant activin IIB receptor polypeptide (vActRIIB) wherein said polypeptide comprises the polypeptide sequence of SEQ ID NO: 18 except for a single amino acid substitution at position 28, wherein the ptide is e of binding tin, activin A, or GDF-l l, and ii) a chemotherapeutic agent. The variant activin IIB receptor of the pharmaceutical composition can have a substitution at position 28 of the vActRIIB polypeptide that is selected from the group consisting of A, F, Q, V, I, L, M, K, H, W and Y for E. In another embodiment the substitution at on 28 of the vActRIIB polypeptide is selected from the group of amino acids ting of A, W and Y for E or the substitution at position 28 of the vActRIIB polypeptide is W. In yet other embodiments, the vActRIIB polypeptide lacks the inal signal sequence. In yet other embodiments, the pharmaceutical also comprises a pharmaceutically acceptable carrier, a chemotherapeutic and the activin IIB receptor polypeptide t.
In another aspect, the chemotherapeutic agent of the pharmaceutical composition is a side analogue. In other embodiments, the chemotherapeutic agent can be 5-fluorouracil or dacarbazine. [002 1] In another embodiment, a method of inhibiting myostatin activity in a subject in need of such treatment is provided comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition comprising i) a variant activin IIB receptor ptide (vActRIIB) n said ptide comprises the polypeptide sequence of SEQ ID NO: 18 except for a single amino acid substitution at position 28, wherein the polypeptide is capable of binding myostatin, activin A, or GDF-l l, and ii) a chemotherapeutic agent. The variant activin IIB receptor of the pharmaceutical composition can have a substitution at position 28 of the vActRIIB polypeptide that is selected from the group consisting of A, E, Q, V, I, L, M, K, H, W and Y for E. In other embodiment the substitution at position 28 of the vActRIIB polypeptide is selected from the group of amino acids consisting of A, W and Y for E or the substitution at position 28 of the vActRIIB polypeptide is W. In yet other embodiments, the vActRIIB ptide lacks the N—terminal signal sequence.
In another aspect, a method of treating a disease in which activin is over- expressed in a subject in need of such treatment is provided comprising administering to a subject a eutically effective amount of the composition comprising i) a variant activin IIB receptor polypeptide IIB) wherein said ptide ses the polypeptide ce of SEQ ID NO: 18 except for a single amino acid substitution at position 28, n the polypeptide is capable of binding myostatin, activin A, or GDF-l l, and ii) a chemotherapeutic agent. The t activin IIB receptor of the pharmaceutical composition can have a substitution at on 28 of the vActRIIB polypeptide that is selected from the group consisting of A, F, Q, V, I, L, M, K, H, W and Y for E. In other embodiment the substitution at position 28 of the vActRIIB polypeptide is selected from the group of amino acids consisting of A, W and Y for E or the substitution at position 28 of the vActRIIB polypeptide is W. In yet other ments, the vActRIIB polypeptide lacks the N—terminal signal sequence.
In an aspect of the invention, the above method can be used to treat cancers.
The cancers can be testicular cancer, ovarian cancer or melanoma.
In another embodiment, a method of reducing the size of a tumor mass in a subject in need of such treatment is provided comprising administering an ive amount of a pharmaceutical ition comprising i) a variant activin IIB receptor polypeptide (vActRIIB) wherein said polypeptide comprises the polypeptide sequence of SEQ ID NO: 18 except for a single amino acid substitution at position 28, wherein the polypeptide is capable of binding myostatin, activin A, or GDF-l l, and ii) a chemotherapeutic agent. The variant activin IIB or of the ceutical composition can have a substitution at position 28 of the IB polypeptide that is ed from the group consisting of A, E, Q, V, I, L, M, K, H, W and Y for E. In other embodiment the substitution at position 28 of the vActRIIB polypeptide is selected from the group of amino acids consisting of A, W and Y for E or the substitution at position 28 of the vActRIIB polypeptide is W. In yet other embodiments, the vActRIIB polypeptide lacks the N—terminal signal sequence. The tumor mass can result from testicular or ovarian cancer or a melanoma.
In yet another embodiment, a method of treating melanoma, in a subject in need of such treatment is provided comprising administering to said subject a therapeutically effective amount of a pharmaceutical composition comprising a variant activin IIB receptor polypeptide IIB) wherein said polypeptide comprises the polypeptide sequence of SEQ ID NO: 18 except for a single amino acid substitution at position 28, wherein the polypeptide is capable of binding myostatin, activin A, or GDF-l l.
In another embodiment, a method of treating a condition having overexpression of angiogenesis factors, in a subject in need of such ent is provided comprising administering to said subject a therapeutically effective amount of a pharmaceutical composition comprising a t activin IIB receptor polypeptide (vActRIIB) wherein said polypeptide comprises the polypeptide sequence of SEQ ID NO: 18 except for a single amino acid substitution at position 28, wherein the polypeptide is capable of binding myostatin, n A, or GDF-l l. The condition can be cancer and the cancer can be ovarian cancer.
VEGF-A or Ang-l can be overexpressed.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1. Figure 1 shows the amino acid ce of wild-type e ActRIIB-human IgGch (SEQ ID NO: 98). The signal peptide sequence is in bold, followed by the mature ActRIIB extracellular , and the human IgG1 Fc in italics, ing a partial hinge region. Amino acids E28 and R40 are ined. The linker sequence GGGGS (SEQ ID NO: 75) is in italics and underlined.
Figure 2. Figure 2 shows the amino acid sequence of e ActRIIBS-human IgGch (SEQ ID NO: 99). The signal peptide sequence is in bold, followed by the mature ActRIIBS soluble domain, and the human IgG1 Fc, including a partial hinge region, is in italics. E28 and R40 are underlined. The linker sequence ) (SEQ ID NO: 75) is in italics and underlined.
Figure 3. Figure 3 shows the effect of soluble vActRIIB-Fc E28W treatment on testicular (Figure 3A) and ovarian (Figure 3B) mass in inhibin-(x knockout mice.
Figures 4A-4B. Figure 4 shows the effect of soluble vActRIIB-Fc E28W treatment on survival rates in male (Figure 4A) and female (Figure 4B) inhibin-(x knockout mice. [003 1] Figure 5. Figure 5 shows the effect of soluble vActRIIB-Fc E28W treatment on body weight in colon 26 tumor-bearing mice.
Figure 6. Figure 6 shows the effect of soluble vActRIIB-Fc E28W treatment on the survival of colon 26 tumor bearing mice.
Figures 7A-7D show activin levels in mice (Figure 7A) and the effect of IB (E28W) on ovarian cancer (OC) (Figures 7A-7D) in KO mice. Figure 7A shows levels of activin in normal and ovarian cancer. ***P<0.001, Student t-test; n=20. Figure 7B shows the effect of sActRIIB on activin levels. Values are mean i SEM. ***: P<0.001 vs. WT control. n=6-12. Figure 7C shows s in ovarian mass with sActRIIB. Values are mean i SEM. ***: P<0.001 vs. WT control. n=6-10. Figure 7D shows ovarian tumors in treated and non-treated mice. Scale bar = 10 mm Figures 8A-8B show ActiVin A blockade abolishes overexpression ofVEGF levels in ovarian tumors (Figure 8A) and activation of e-3 in testicular tumors following treatment with sActRIIB (E28W) e 8B). Figure 8A - Values are mean SEM. *: P<0.05; Student t-test. n=10. Figure 8B- ** P<0.01, Student t-test.
Figures 9A-9B show the effect of activin nist IB (E28W) and cytotoxic chemotherapeutic agents on tumor growth. Figure 9A shows the effect of sActRIIB and S-Fu on TOV-21G tumor growth inhibition in nude mice. s in tumor volumes were recorded longitudinally. ***: P<0.001 vs. PBS; #: P<0.05 vs. 5-Fu; 1: P<0.01 vs. sActRIIB; Repeated measures ANOVA; n=12. Figure 9B shows the effect of sActRIIB and dacarbazine on the growth of G361 human melanoma xenografts in nude mice. Results of tumor volume (mm3) are expressed as the mean :: SEM. *: P<0.05 vs. PBS group. Statistics based on repeated measures ANOVA. n=8.
DETAILED DESCRIPTION [003 6] ns comprising variant human n IIB receptor (vActRIIB; also referred to as sActRIIB) polypeptides are disclosed. These proteins and polypeptides are terized by their ability to bind to at least one of three TGF-B proteins, myostatin (GDF-8), activin A, and GDF-11, and to inhibit the activities of these proteins. These proteins and polypeptides also exhibit a reduced tendency to aggregate compared to polypeptides not containing the modifications disclosed herein. The modifications consist of amino acid substitutions at positions 28, 40, or both 28 and 40 with reference to wild type ActRIIB of accession number NP_OOlO97 (SEQ ID NO: 47), and the extracellular domain of ActRIIB (SEQ ID NO: 18) or ActRIIBS (SEQ ID NO: 2). [003 7] As used herein the term “TGF-B family members” or “TGF-B proteins” refers to the structurally related growth factors of the transforming growth factor family including activins, and growth and differential factor (GDF) proteins (Kingsley et al. Genes DeV. 8: 133- 146 (1994), McPherron et al. Growth s and cytokines in health and disease, Vol. 1B, D.
LeRoith and C.Bondy. ed., JAI Press Inc., Greenwich, Conn, USA: pp 357-393). [003 8] GDF-8, also ed to as myostatin, is a ve regulator of skeletal muscle tissue (McPherron et al. PNAS USA 94: 12457-12461 (1997)). Myostatin is synthesized as an inactive protein complex approximately 375 amino acids in length, having GenBank Accession No: AAB86694 (SEQ ID NO: 49) for human. The precursor protein is activated by proteolytic cleavage at a tetrabasic sing site to produce an N—terminal inactive prodomain and an approximately 109 amino acid C-terminal protein which dimerizes to form a homodimer of about 25 kDa. This homodimer is the mature, biologically active protein (Zimmers et al., Science 296, 1486 (2002)). [003 9] As used herein, the term “prodomain” or “propeptide” refers to the inactive N- terminal protein which is cleaved off to release the active C-terminal protein. As used herein the term atin” or “mature myostatin” refers to the mature, biologically active C-terminal polypeptide, in monomer, dimer or other form, as well as biologically active fragments or related polypeptides including allelic variants, splice variants, and fusion peptides and polypeptides. The mature myostatin has been reported to have 100% ce identity among many s including human, mouse, chicken, porcine, , and rat (Lee et al., PNAS 98, 9306 (2001)).
As used herein GDF-ll refers to the BMP (bone morphogenic protein) having Swissprot accession number 095390 (SEQ ID NO: 50), as well as variants and species homologs of that protein. GDF-ll has approximately 90% ty to myostatin at the amino acid level. GDF-ll is involved in the tion of anterior/posterior patterning of the axial skeleton (McPherron et al, Nature Genet. 22 (93): 260-264 (1999); Gamer et al, Dev. Biol. 208 (1), 222-232 (1999)) but postnatal functions are unknown. [004 1] Activin A is the homodimer of the ptide chains BA. As used herein the term “activin A” refers to the activin protein having GenBank Accession No: NM_002192 (SEQ ID NO: 48), as well as variants and species gs of that protein.
ACTIVIN RECEPTORS As used herein, the term activin type II B receptors (ActRIIB) refers to human n receptors having accession number NP_001097 (SEQ ID NO: 47). The term soluble ActRIIB encompasses the extracellular domain of ActRIIB (SEQ ID NO: 18), B5 (SEQ ID NO: 2) and these sequences wherein the arginine at position 64 is substituted with alanine, as well as.
VARIANT SOLUBLE ActRIIB PTIDES [0 043] The t invention provides isolated ns comprising human variant soluble ActIIB receptor polypeptides (referred to herein as vActRIIB polypeptides, or variant polypeptides or IB). As used herein the term “VActRIIB protein” refers to a protein comprising a vActRIIB polypeptide. As used herein the term “isolated” refers to a protein or polypeptide molecule purified to some degree from endogenous material. These polypeptides and proteins are characterized as having the y to bind and inhibit the activity of any one of activin A, myostatin, or GDF-l 1. In some embodiments, the binding affinity of the variant polypeptides for activin A, myostatin, or GDF-ll is improved ed to wild-type polypeptides.
In one embodiment, the VActRIIB polypeptide has the amino acid sequence of SEQ ID NOS: 2 or 18 in which amino acids at either position E28 or R40, or both position E28 and R40 are substituted with another non-native amino acid, and wherein the ptide is capable of binding myostatin, activin A, or GDF-l 1. In another embodiment, the vActRIIB polypeptides are the mature versions, or the truncated mature versions of these sequences. As used herein the term “mature VActRIIB polypeptide” refers to the polypeptide having the amino acid signal sequence removed. In one embodiment, the mature sequences are, for example, amino acids 19 through 160 of SEQ ID NO: 2, and amino acids 19 through 134 of SEQ ID NO: 18, n one or both amino acids at ons 28 and 40 are substituted with another non-native amino acid and the polypeptides retain the ability to bind to activin A, myostatin, or GDF-l 1. As used herein the term truncated mature VActRIIB polypeptide refers to the polypeptide having the signal sequence and in addition amino acids from the N-terminal of the mature polypeptide removed. In one embodiment, the mature N-terminal 4 amino acids or the N-terminal 6 amino acids of the mature polypeptide are d. In this embodiment, the truncated mature sequences are, for example, amino acids 23 through 160 of SEQ ID NO: 2, or amino acids 25 through 160 of SEQ ID NO: 2; and amino acids 23 through 134 of SEQ ID NO: 18, or amino acids 25 h 134 of SEQ ID NO: 18 wherein one or both amino acids at positions 28 and 40 are tuted with non-wild type amino acids which retain the ability to bind to activin A, tin, or GDF-l 1. As used herein, the term “position 28” and “position 40” (that is, E28 and R40) refers to the amino acid position with reference to the ces SEQ ID NOS: 2 and 18 that include an 18 amino acid signal sequence. For consistency, if mature VActRIIB polypeptides have substitutions at position 10 and/or position 22, or ted mature polypeptides have substitutions at position 6 and/or position 18, or substitutions at positions 4 and/or position 16 with respect to the mature or truncated mature sequences, these variants will still be referred to with t to the fill length SEQ ID NOS: 2 and 18, or as shown in Figure l or 2, i.e., the amino acid substitution at position E28 and/or R40. Such mature embodiments or N-terminal truncated embodiments are exemplified below. 2012/070571 In one embodiment, the substitution at position E28 is selected from the group of amino acids consisting of W, Y and A. In one embodiment, the substitution at position 28 is W. In a filrther embodiment the tution at position 28 is selected from the group of amino acids consisting of A, F, Q, V, I, L, M, K, H, W and Y. In a filrther embodiment, the substitution at position 40 is selected from the group of amino acids consisting of G, Q, M, H, K and N. In a filrther embodiment the substitution at position 28 is selected from the group of amino acids consisting of A, F, Q, V, I, L, M, K, H, W and Y and the substitution at position 40 is ed from the group of amino acids consisting of A, G, Q, M, H, K, and N. In one embodiment, the protein comprises polypeptides having an amino acid sequence selected from the group consisting of SEQ ID NOS: 4, 6, 8, 10, l2, l4, 16, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 52, 54, 56, 60, 62, 64, 66, 68, 70, 72, 87, 88, 91, 93, 95, and 97. In another embodiment, the protein comprises a ptide encoded by the polynucleotide having the sequence set forth in the group consisting of SEQ ID NOS: 3, 5, 7, 9, ll, 13, 15, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 51, 53, 55, 59, 61, 63, 65, 67, 69, 71, 92, 94, 96 or its complement.
In one embodiment, the signal sequences are removed from the vActRIIB ptide, leaving the mature variant polypeptides. Various signal peptides can be used in the preparation of the polypeptides of the instant application. The signal peptides can have the sequence shown in Figures 1 and 2 (SEQ ID NO: 73), or alternative signal sequences such as SEQ ID NO: 74, the signal sequence for SEQ ID NOS: 2 and 18. Any other signal peptides useful for sing vActRIIB or vActRIIB5 polypeptides may be used.
In another ment, the vActRIIB polypeptides have sequences that are substantially similar to SEQ ID NOS: 2 and 18. As used herein the term “substantially similar” refers to polypeptides having at least about 80% identity, at least about 85% identity, at least about 90% identity, at least about 95% identity, at least about 98% identity, or at least about 99% identity to the amino acid sequence set forth in SEQ ID NOS: 2 and 18, and wherein one or both amino acids at positions 28 and/or 40 are substituted with non-wild type amino acids, wherein the polypeptide retains the activity of the polypeptide of SEQ ID NOS: 2 and 18, that is the ability to bind and inhibit myostatin, activin A or GDF-l 1. In addition, the term vActRIIB ptide encompasses fragments of SEQ ID NOS: 2 or 18 such as N and C terminal tions containing the tutions at position 28 and /or 40 described herein, wherein the ptide is capable of binding and inhibiting myostatin, activin A or GDF-l l.
As used herein the term “derivative” of the IB and vActRIIB5 polypeptides refers to the ment of at least one additional chemical moiety, or at least one WO 06175 additional polypeptide to form covalent or aggregate conjugates such as glycosyl groups, lipids, acetyl groups, or C-terminal or N-terminal fusion polypeptides, conjugation to PEG molecules, and other modifications which are bed more fully below. Variant ActRIIB receptor ptides (vActRIIB) can also include additional modifications and derivatives, including modifications to the C and N termini which arise from processing due to expression in various cell types such as ian cells, E. coli, yeasts and other recombinant host cells. Further included are vActRIIB polypeptide nts and polypeptides comprising inactivated N- glycosylation site(s), inactivated protease processing site(s), or conservative amino acid substitution(s), of the polypeptide sequences set forth in SEQ ID NOS: 4, 6, 8, 10, 12, l4, 16, , 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 52, 54, 56, 60, 62, 64, 66, 68, 70, 72, 87, 88, 91, 93, 95, and 97.
As used herein, the term a “vActRIIB or vActRIIBS polypeptide activity” or “a biological activity of a soluble ActRIIB or ActRIIBS polypeptide” refers to one or more in vitro or in vivo activities of the vActRIIB and vActRIIBS polypeptides including but not d to those demonstrated in the Example below. Activities of the vActRIIB polypeptides include, but are not limited to, the ability to bind to myostatin or activin A or GDF-l l, and the ability to reduce or neutralize an activity of myostatin or n A or GDF-l 1. As used herein, the term “capable of binding” to myostatin, activin A, or GDF-ll refers to binding measured by methods known in the art, such as the Biacore method described in Example 2 below. Also, in Example 2, the pMARE C2Cl2 cell-based assay measures n A neutralizing activity, myostatin neutralizing activity, and GDF-ll neutralizing activity. In vivo activities e but are not limited to increasing body weight, increasing lean muscle mass, increasing skeletal muscle mass, decreasing fat mass as demonstrated in animal models below and as known in the art. Biological activities further include reducing or preventing cachexia caused by certain types of tumors, preventing the growth of certain types of tumors, and increasing survival of certain animal models. Further discussion of the IB polypeptide activities is provided below.
The polypeptides of the present invention further comprise heterologous polypeptides attached to the vActRIIB polypeptide either directly or through a linker sequence to form a fusion protein. As used herein the term “fusion protein” refers to a n having a heterologous polypeptide attached via recombinant DNA techniques. logous polypeptides include but are not limited to EC polypeptides, His tags, and leucine zipper domains to promote erization and stabilization of the t ActRIIB ptides as bed in, for example, WO 00/29581, which is herein orated by reference. In one WO 06175 2012/070571 embodiment, the heterologous polypeptide is an EC ptide or domain. In one embodiment, the Fc domain is selected from a human IgGl, IgG2, and IgG4 Fc domain. These are provided in SEQ ID NOS: 80, 82 and 84. The vActRIIB can further comprise all or a n of the hinge ce of the IgGl, IgG2, or IgG4 adjacent to its respective IgG Fc region. The full hinge sequence for IgGl, IgG2, and IgG4 are provided in SEQ ID NOS: 76, 77, and 78 respectively. [005 1] The vActRIIB polypeptide can optionally fiarther comprise a “linker” sequence.
Linkers serve primarily as a spacer between a polypeptide and a second heterologous polypeptide or other type of fusion or between two or more variant ActRIIB polypeptides. In one embodiment, the linker is made up of amino acids linked together by peptide bonds, ably from 1 to 20 amino acids linked by peptide bonds, wherein the amino acids are selected from the 20 naturally occurring amino acids. One or more of these amino acids may be glycosylated, as is understood by those in the art. In one embodiment, the l to 20 amino acids are selected from glycine, alanine, proline, asparagine, ine, and lysine. Preferably, a linker is made up of a ty of amino acids that are sterically unhindered, such as glycine and alanine. Exemplary linkers are polyglycines (particularly (Gly)5, (Gly)8, poly(Gly-Ala), and polyalanines. One exemplary suitable linker as shown in the Examples below is (Gly)4Ser (SEQ ID NO: 75). In a r embodiment, vActRIIB can comprise a hinge linker, that is a linker sequence are provided adjacent to the hinge region, as exemplified in SEQ ID NO: 79.
The linkers are also non-peptide linkers. For example, alkyl linkers such as - NH-(CH2)s-C(O)-, wherein s = 2-20 can be used. These alkyl linkers may further be substituted by any non-sterically hindering group such as lower alkyl (e.g., Cl-C6) lower acyl, halogen (e.g., Cl, Br), CN, NH2, phenyl, etc.
In one embodiment the vActRIIB polypeptides can be attached to an EC polypeptide, directly or via a linker, or via a hinge linker. In one embodiment, the Fe is a human IgG Fc. vActRIIB attached to Fe include for example, vActRIIB-IgGch, E28A (SEQ ID NO: 60); vActRIIB-IgGch, E28W (SEQ ID NO: 62), vActRIIB-IgGch, E28Y (SEQ ID NO: 64), vActRIIB-IgG Fc, R40G (SEQ ID NO: 66), vActRIIBS-IgGch, E28A (SEQ ID NO: 70), and IBS-IgGch E28W (SEQ ID NO: 72), as shown in Tables 1 and 2, and described in the Examples herein. Further embodiments include vActRIIB-IgG2 Fc, EQ ID NO: 91), vActRIIB-IgG2 Fc, E28Y (SEQ ID NO: 93), and vActRIIB-IgG2 Fc (SEQ ID NO: 95). The variants have been demonstrated to produce less aggregation compared to the wild type ActRIIB-IgG2 IgG2, as trated in the Examples below. 2012/070571 The vActRIIB polypeptides disclosed herein can also be attached to a non- polypeptide molecule for the purpose of conferring desired properties such as reducing degradation and/or increasing half-life, reducing toxicity, reducing immunogenicity, and/or increasing the biological activity of the ActRIIB polypeptides. Exemplary molecules e but are not limited to linear polymers such as polyethylene glycol (PEG), polylysine, a dextran; a lipid; a cholesterol group (such as a steroid); a carbohydrate, or an accharide molecule. [0 0 5 5] In another aspect, the present invention es ed nucleic acid molecules comprising cleotides encoding the vActRIIB polypeptides of the present invention. As used herein the term ted” refers to nucleic acid molecules purified to some degree from endogenous material. In one embodiment, the nucleic acid molecule of the present invention comprises a polynucleotide encoding the polypeptides of SEQ ID NOS: 4, 6, 8, 10, 12, 14, 16, ,22,24,26,28,30,32,34,36,38,40,42,44,46,52,54,56,60,62,64,66,68,70,72,87, 88, 91, 93, 95, and 97. Due to the known degeneracy of the genetic code, wherein more than one codon can encode the same amino acid, a DNA sequence can vary from that shown in SEQ I[)}U)S:3,5,7,9,11,13,15,19,21,23,25,27,29,31,33,35,37,39,41,43,45,51,53,55, 59, 61, 63, 65, 67, 69, 71, 92, 94, and 96 or the mentary strand of SEQ ID NOS: 3, 5, 7, 9,11,13,15,19,21,23,25,27,29,31,33,35,37,39,41,43,45,51,53,55,59,61,63,65,67, 69, 71, 92, 94, and 96, and still encode a polypeptide having the amino acid sequence of SEQ I[)}M)S:4,6,8,10,12,14,16,20,22,24,26,28,30,32,34,36,38,40,42,44,46,52,54,56, 60,62,64,66,68,70,72,87,88,91,93,95,andl¥1 SudivafiantDDLAsequencescaniesuh from silent mutations occurring during production, or can be the product of deliberate nnuagenesnsofthesesequences In another embodiment the nucleic acid molecule of the present invention comprises a polynucleotide having the cleotide sequence set forth in SEQ ID NOS: 3, 5, 7,9,11,13,15,19,21,23,25,27,29,31,33,35,37,39,41,43,45,51,53,55,59,61,63,65, 67, 69, 71, 92, 94, and 96 or the complementary strand of SEQ ID NOS: 3, 5, 7, 9, 11, 13, 15, 19,21,23,25,27,29,31,33,35,37,39,41,43,45,51,53,55,59,61,63,65,67,69,71,92, 94, and 96. In another embodiment, the present invention provides nucleic acid molecules which hybridize under stringent or moderate ions with the polypeptide-encoding regions ofSEKQI[)}M)S:3,5,7,9,11,13,15,19,21,23,25,27,29,31,33,35,37,39,41,43,45,51, 53,55,59,61,63,65,67,69,71,92,94,and96\Nhemfintheencodedpoqupfideconqwmesan amino acid sequence as set forth in SEQ ID NOS: 4, 6, 8, 10, 12, 14, 16, 20, 22, 24, 26, 28, 30, 36,38,40,42,44,46,52,54,56,60,62,64,66,68,70,72,87,88,91,93,95,and97 and n the encoded polypeptide maintains an activity of a vActRIIB polypeptide. c acid molecules of the invention include DNA in both -stranded and double-stranded form, as well as the RNA complement f. DNA includes, for example, cDNA, genomic DNA, synthetic DNA, DNA amplified by PCR, and combinations f. Genomic DNA may be isolated by conventional techniques, such as by using the DNA of SEQ ID NOS:l or 17, or a suitable nt thereof, as a probe. Genomic DNA encoding ActRIIB polypeptides is obtained from genomic libraries which are available for a number of species. Synthetic DNA is available from chemical synthesis of overlapping oligonucleotide fragments ed by assembly of the nts to reconstitute part or all of the coding regions and flanking sequences. RNA may be obtained from procaryotic expression vectors which direct high-level synthesis ofmRNA, such as vectors using T7 promoters and RNA polymerase. cDNA is obtained from libraries prepared from mRNA isolated from various tissues that express ActRIIB. The DNA molecules of the ion include filll length genes as well as polynucleotides and fragments thereof The full length gene may also include sequences encoding the N-terminal signal ce.
In another aspect of the present invention, expression vectors containing the nucleic acid sequences are also ed, and host cells transformed with such vectors and s of producing the vActRIIB polypeptides are also provided. The term “expression vector” refers to a plasmid, phage, virus or vector for expressing a polypeptide from a polynucleotide ce. Vectors for the sion of the vActRIIB ptides contain at a minimum ces required for vector propagation and for expression of the cloned insert.
An sion vector comprises a transcriptional unit comprising an assembly of (l) a genetic element or elements having a regulatory role in gene expression, for example, promoters or enhancers, (2) a sequence that encodes vActRIIB polypeptides to be transcribed into mRNA and translated into protein, and (3) appropriate transcription initiation and termination sequences. These sequences may fiarther include a selection marker. s suitable for expression in host cells are readily available and the nucleic acid molecules are inserted into the vectors using standard recombinant DNA ques. Such vectors can include promoters which filnction in specific tissues, and viral vectors for the expression of vActRIIB in targeted human or animal cells. An exemplary expression vector suitable for expression of vActRIIB is the pDSRa, (described in W0 90/14363, herein incorporated by reference) and its derivatives, containing vActRHB polynucleotides, as well as any additional suitable vectors known in the art or described below.
The application further provides methods of making vActRHB polypeptides. A variety of other expression/host systems may be utilized. These systems include but are not limited to microorganisms such as bacteria ormed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast ormed with yeast expression vectors; insect cell systems infected with virus sion vectors (e.g., baculovirus); plant cell systems transfected with virus expression s (e.g., ower mosaic virus, CaMV; o mosaic virus, TMV) or transformed with bacterial sion vectors (e.g., Ti or pBR322 plasmid); or animal cell systems. Mammalian cells useful in recombinant protein production include but are not limited to VERO cells, HeLa cells, Chinese hamster ovary (CHO) cell lines, or their derivatives such as Veggie CH0 and related cell lines which grow in serum-free media (see Rasmussen et al., 1998, Cytotechnology 28:31) or CHO strain DX-B11, which is deficient in DHFR (see Urlaub et al., 1980, Proc. Natl. Acad. Sci. USA 77:4216-20) COS cells such as the COS-7 line ofmonkey kidney cells (ATCC CRL 1651) (see Gluzman et al., 1981, Cell 23:175), W138, BHK, HepG2, 3T3 (ATCC CCL 163), RIN, MDCK, A549, PC12, K562, L cells, C127 cells, BHK (ATCC CRL 10) cell lines, the CV1/EBNA cell line derived from the African green monkey kidney cell line CV1 (ATCC CCL 70) (see McMahan et al., 1991, EMBO J. 10:2821), human embryonic kidney cells such as 293, 293 EBNA or MSR 293, human epidermal A431 cells, human Colo205 cells, other transformed primate cell lines, normal diploid cells, cell strains derived from in vitro culture of primary tissue, primary explants, HL-60, U937, HaK or Jurkat cells. Mammalian expression allows for the production of secreted or soluble polypeptides which may be recovered from the growth medium.
Using an appropriate host-vector system, vActRIIB polypeptides are produced recombinantly by culturing a host cell transformed with an expression vector ning the nucleic acid molecules of the present invention under conditions allowing for production.
Transformed cells can be used for long-term, high-yield polypeptide production. Once such cells are transformed with vectors that contain selectable markers as well as the desired expression cassette, the cells can be allowed to grow for 1-2 days in an enriched media before they are switched to selective media. The selectable marker is designed to allow growth and recovery of cells that successfully express the uced sequences. Resistant clumps of stably transformed cells can be proliferated using tissue culture techniques appropriate to the cell line employed. An overview of expression of recombinant proteins is found in Methods of Enzymology, v. 185, Goeddell, D.V., ed., Academic Press (1990). [006 1] In some cases, such as in expression using procaryotic systems, the expressed polypeptides of this ion may need to be “refolded” and ed into a proper tertiary structure and 1de linkages generated in order to be biologically active. Refolding can be accomplished using a number of procedures well known in the art. Such methods include, for example, exposing the solubilized polypeptide to a pH usually above 7 in the presence of a chaotropic agent. The ion of chaotrope is similar to the choices used for inclusion body solubilization, however a chaotrope is typically used at a lower concentration. Exemplary opic agents are guanidine and urea. In most cases, the refolding/oxidation solution will also contain a reducing agent plus its oxidized form in a specific ratio to generate a particular redox potential which allows for disulfide shuffling to occur for the formation of cysteine bridges. Some ly used redox couples include cysteine/cystamine, glutathione/dithiobisGSH, cupric chloride, dithiothreitol DTT/dithiane DTT, and 2- mercaptoethanol (bME)/dithio-bME. In many instances, a vent may be used to increase the efficiency of the ing. Commonly used cosolvents e glycerol, polyethylene glycol of various molecular weights, and arginine.
In addition, the polypeptides can be synthesized in solution or on a solid support in accordance with conventional techniques. Various automatic synthesizers are commercially available and can be used in accordance with known ols. See, for example, Stewart and Young, Solid Phase Peptide Synthesis, 2d.Ed., Pierce Chemical Co. (1984); Tam et al., J Am Chem Soc, 42, (1983); eld, Science 232:341-347 (1986); Barany and eld, The Peptides, Gross and Meienhofer, eds, ic Press, New York, 1-284; Barany et al., Int J Pep Protein Res, 30:705-739 (1987).
The polypeptides and proteins of the present invention can be d according to n ation techniques are well known to those of skill in the art. These ques involve, at one level, the crude fractionation of the proteinaceous and non-proteinaceous fractions. Having separated the peptide polypeptides from other proteins, the peptide or polypeptide of interest can be further purified using chromatographic and electrophoretic techniques to achieve partial or complete purification (or ation to homogeneity). The term “isolated polypeptide” or “purified polypeptide” as used herein, is intended to refer to a composition, isolatable from other components, wherein the ptide is purified to any degree relative to its naturally-obtainable state. A purified polypeptide therefore also refers to a polypeptide that is free from the environment in which it may naturally occur. Generally, “purified” will refer to a polypeptide composition that has been subjected to fractionation to remove various other components, and which composition substantially retains its expressed biological activity. Where the term “substantially purified” is used, this designation will refer to a peptide or polypeptide composition in which the polypeptide or peptide forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 85%, or about 90% or more of the proteins in the composition. 2012/070571 Various techniques suitable for use in purification will be well known to those of skill in the art. These include, for example, itation with ammonium sulphate, PEG, antibodies oprecipitation) and the like or by heat denaturation, followed by centrifugation; tography such as affinity chromatography in-A columns), ion exchange, gel filtration, reverse phase, hydroxylapatite, hydrophobic interaction chromatography; isoelectric focusing; gel electrophoresis; and combinations of these techniques. As is generally known in the art, it is believed that the order of conducting the various purification steps may be changed, or that certain steps may be omitted, and still result in a le method for the preparation of a substantially purified polypeptide. Exemplary purification steps are provided in the Examples below.
Various methods for quantifying the degree of purification of polypeptide will be known to those of skill in the art in light of the present disclosure. These include, for example, determining the specific binding activity of an active fraction, or assessing the amount of peptide or polypeptide within a fraction by SDS/PAGE analysis. A preferred method for assessing the purity of a polypeptide fiaction is to calculate the binding ty of the n, to compare it to the g activity of the initial extract, and to thus calculate the degree of purification, herein assessed by a “-fold purification number.” The actual units used to represent the amount of binding activity will, of course, be dependent upon the particular assay que chosen to follow the ation and whether or not the polypeptide or peptide exhibits a detectable binding ty. t activin type IIB polypeptides bind to ligands that activate - degradation cascades. vActRIIB polypeptides capable of binding and inhibiting the activity of the ligands activin A, myostatin, and/or GDF-l l, have therapeutic potential against the diseases that involve muscle atrophy, as well as the ent of certain cancers, e.g. ovarian tumors prostate tumors and melanoma, and other diseases as shown in the Examples below. [0 0 6 7] However, aggregation can occur when expressing or purifying wild-type ActRIIB or ActRHBS polypeptides. This aggregation includes structured oligomer formation during expression and non-structured aggregate generation both during expression and after polypeptide purification.
The combined approaches of structure analysis, molecular modeling, and mass spectrometry have indicated that multimerization may arise in ActRIIB polypeptides via olecular disulfide bond formation aided by electrostatic and hydrogen bonding interactions between nonglycosylated ActRIIB polypeptides. cant hydrogen bonds exist at the interface of two ActRIIB molecules; between E28 side chain in one ActRIIB and R40 side chain in the other ActRIIB, for example. In addition, critical electrostatic interactions exist between E28 in one B and R40 in the other B.
These electrostatic interactions may significantly contribute to increase the tion of temporal B dimers, resulting in promotion of noncovalent and/or covalent bond formation between ActRIIB units. The interaction between residues 28 and 40 is the most critical among these interactions as these two residues are involved in double hydrogen bonds and a strong electrostatic interaction. The residues 28 and 40 are involved in ActRIIB:ActRIIB interactions and not in ActRIIB:ligand ctions. Thus, residues 28 and 40 can be substituted with tive amino acids according to invention, to improve the lity, and reduce the aggregation of the receptor polypeptides. Therefore, E28 and R40 were substituted respectively with other possible natural amino acids, sed, and tested by Biacore as shown below. Biacore determined binding are shown in Tables 1A and 1B in Example 2 below.
Furthermore, percent aggregation of the vActRIIB polypeptides are determined below.
The results in the Examples below show reduced ation for vActRIIB polypeptides and proteins having the amino acid substitutions described , while retaining the ability to bind and neutralize myostatin, activin A, or GDF-l 1.
ANTIBODIES [007 1] The present invention filrther includes antibodies which bind to variant ActRIIB polypeptides, including those that specifically bind to the vActRIIB polypeptides of the present invention. As used herein the term “specifically binds” refers to antibodies having a binding affinity (Ka) for vActRIIB polypeptides of 106 M-l or greater. As used herein, the term “antibody” refers to intact antibodies including polyclonal antibodies (see, for example Antibodies: A Laboratory Manual, Harlow and Lane (eds), Cold Spring Harbor Press, (1988)), and onal antibodies (see, for example, US. Patent Nos. RE 32,011, 4,902,614, 4,543,439, and 4,411,993, and Monoclonal Antibodies: A New Dimension in Biological Analysis, Plenum Press, Kennett, McKeam and Bechtol (eds.) (1980)). As used herein, the term “antibody” also refers to a fragment of an antibody such as F(ab), F(ab’), F(ab’)2, Fv, Fe, and single chain antibodies which are produced by recombinant DNA techniques or by enzymatic or al cleavage of intact dies. The term “antibody” also refers to bispecific or bifunctional antibodies, which are an artificial hybrid antibody having two ent heavy/light chain pairs and two different binding sites. Bispecific antibodies can be produced by a variety of s including fusion of hybridomas or linking of Fab’ fragments.
(See Songsivilai et al, Clin. Exp. Immunol. 79:315-321 (1990), ny et al., J.
Immunol. 148: 1547-1553 (1992)).
As used herein the term “antibody” also refers to chimeric antibodies, that is, antibodies having a human constant dy globin domain coupled to one or more man le antibody immunoglobin domain, or fragments f (see, for example, US. Patent No. 5,595,898 and US. Patent No. 5,693,493). Antibodies also refers to “humanized” antibodies (see, for example, US. Pat. No. 567 and W0 94/10332), minibodies (WO 94/09817), maxibodies, and antibodies produced by transgenic animals, in which a transgenic animal containing a proportion of the human antibody producing genes but deficient in the production of endogenous antibodies are capable of producing human antibodies (see, for example, Mendez et al., Nature Genetics 15:146-156 (1997), and US.
Patent No. 6,300,129). The term odies” also includes multimeric dies, or a higher order complex of proteins such as heterdimeric antibodies, and anti-idiotypic antibodies.
“Antibodies” also includes anti-idiotypic antibodies. The antibodies against V ActRIIB can be used, for example, to identify and quantitate vActRIIB in vitro and in vivo.
Also ed are polyclonal antibodies from any mammal, for example mouse and rat antibodies, and rabbit antibodies, that bind specifically to the vActRIIB polypeptides described , including SEQ ID NOS: 4, 6, 8, 10, 12, l4, 16, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 52, 54, 56, 60, 62, 64, 66, 68, 70, 72, 87, 88, 91, 93, 95, and 97.
Such antibodies find use as research tools and in quantitative assays for detecting and assaying the polypeptides disclosed herein. Such antibodies are made using methods described above and as known in the art.
PHARMACEUTICAL COMPOSITIONS Pharmaceutical compositions containing the vActRIIB proteins and polypeptides of the present invention are also provided. Such compositions comprise a therapeutically or prophylactically effective amount of the polypeptide or protein in admixture with ceutically acceptable materials, and physiologically acceptable formulation materials. The pharmaceutical composition may contain formulation materials for modifying, maintaining or ving, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, tion or penetration of the composition. Suitable formulation materials include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, ne or lysine); crobials; antioxidants (such as ascorbic acid, sodium sulfite or sodium hydrogen-sulfite); buffers (such as borate, bicarbonate, Tris-HCl, citrates, phosphates, other organic acids); bulking agents (such as ol or glycine), chelating agents (such as ethylenediamine tetraacetic acid (EDTA)); complexing agents (such as caffeine, polyvinylpyrrolidone, beta-cyclodextrin or ypropyl-beta- cyclodextrin); fillers; monosaccharides; disaccharides and other carbohydrates (such as glucose, mannose, or dextrins); proteins (such as serum albumin, gelatin or immunoglobulins); coloring; flavoring and diluting agents; emulsifying ; hydrophilic polymers (such as polyvinylpyrrolidone); low molecular weight polypeptides; salt-forming counterions (such as sodium); preservatives (such as benzalkonium chloride, benzoic acid, salicylic acid, thimerosal, phenethyl l, methylparaben, propylparaben, chlorhexidine, sorbic acid or en peroxide); solvents (such as glycerin, propylene glycol or polyethylene glycol); sugar alcohols (such as mannitol or sorbitol); suspending agents; surfactants or wetting agents (such as pluronics, PEG, sorbitan esters, polysorbates such as polysorbate 20, polysorbate 80, triton, tromethamine, lecithin, cholesterol, tyloxapal); stability ing agents (sucrose or ol); tonicity enhancing agents (such as alkali metal halides (preferably sodium or potassium chloride, mannitol sorbitol); ry vehicles; diluents; excipients and/or pharmaceutical adjuvants. (Remington’s Pharmaceutical Sciences, 18th Edition, A.R. Gennaro, ed., Mack Publishing Company, 1990).
The optimal pharmaceutical composition will be determined by one d in the art depending upon, for example, the intended route of administration, ry format, and desired dosage. See for example, Remington’s Pharmaceutical Sciences, supra. Such compositions may influence the physical state, stability, rate of in vivo e, and rate of in vivo clearance of the polypeptide. For e, suitable compositions may be water for injection, physiological saline solution for parenteral administration.
The primary vehicle or carrier in a ceutical composition may be either aqueous or non-aqueous in nature. For example, a le vehicle or carrier may be water for injection, physiological saline solution or artificial cerebrospinal fluid, possibly mented with other materials common in compositions for parenteral administration. l buffered saline or saline mixed with serum albumin are further exemplary vehicles. Other exemplary pharmaceutical compositions comprise Tris buffer of about pH 7.0-8.5, or acetate buffer of about pH 4.0-5.5, which may fiarther include sorbitol or a suitable substitute thereof In one embodiment of the t invention, compositions may be prepared for storage by mixing the selected composition having the desired degree of purity with optional formulation agents (Remington’s Pharmaceutical Sciences, supra) in the form of a lyophilized cake or an aqueous 2012/070571 solution. Further, the therapeutic composition may be formulated as a lyophilizate using appropriate excipients such as sucrose.
The formulations can be delivered in a variety of methods, for example, by inhalation therapy, orally, or by ion. When parenteral administration is contemplated, the therapeutic compositions for use in this invention may be in the form of a pyrogen-free, parenterally acceptable aqueous on comprising the desired polypeptide in a pharmaceutically acceptable vehicle. A particularly le vehicle for eral injection is sterile distilled water in which a polypeptide is formulated as a sterile, isotonic solution, properly preserved. Yet another preparation can involve the ation of the d le with an agent, such as injectable microspheres, bio-erodible particles, polymeric compounds (polylactic acid, polyglycolic acid), beads, or liposomes, that provides for the lled or sustained e of the product which may then be delivered via a depot injection. Hyaluronic acid may also be used, and this may have the effect of promoting sustained duration in the circulation. Other suitable means for the introduction of the desired molecule include implantable drug delivery devices.
In another aspect, pharmaceutical formulations suitable for injectable administration may be formulated in aqueous solutions, preferably in logically compatible buffers such as Hanks’ solution, Ringer’s solution, or physiologically buffered saline. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection sions. Suitable lipophilic solvents or vehicles include fatty oils, such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate, triglycerides, or liposomes. Non-lipid tionic amino polymers may also be used for delivery. Optionally, the suspension may also contain suitable stabilizers or agents to increase the lity of the compounds and allow for the preparation of highly concentrated solutions. In another embodiment, a pharmaceutical composition may be formulated for inhalation. Inhalation solutions may also be formulated with a propellant for aerosol delivery. In yet another embodiment, solutions may be nebulized.
Pulmonary administration is further described in PCT Application No. PCT/U894/001875, which describes pulmonary delivery of chemically modified proteins.
It is also contemplated that certain formulations may be stered orally. In one embodiment of the present invention, molecules that are administered in this fashion can be formulated with or without those carriers customarily used in the compounding of solid dosage forms such as s and capsules. For example, a e may be ed to release the active portion of the formulation at the point in the gastrointestinal tract when bioavailability is maximized and pre-systemic degradation is minimized. Additional agents can be included to facilitate tion of the therapeutic molecule. Diluents, ngs, low melting point waxes, vegetable oils, lubricants, suspending agents, tablet egrating agents, and binders may also be employed. Pharmaceutical compositions for oral administration can also be formulated using pharmaceutically acceptable carriers well known in the art in dosages suitable for oral administration. Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, sions, and the like, for ingestion by the patient. [008 1] Pharmaceutical preparations for oral use can be ed through combining active compounds with solid excipient and sing the resultant mixture of granules (optionally, after grinding) to obtain tablets or dragee cores. Suitable auxiliaries can be added, if desired. Suitable ents include carbohydrate or protein fillers, such as sugars, including lactose, sucrose, mannitol, and sorbitol; starch from corn, wheat, rice, potato, or other plants; cellulose, such as methyl cellulose, hydroxypropylmethyl-cellulose, or sodium carboxymethylcellulose; gums, including arabic and tragacanth; and proteins, such as gelatin and collagen. If desired, disintegrating or solubilizing agents may be added, such as the cross- linked polyvinyl pyrrolidone, agar, and alginic acid or a salt thereof, such as sodium alginate.
Dragee cores may be used in conjunction with suitable coatings, such as concentrated sugar ons, which may also contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or um dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or ts may be added to the tablets or dragee coatings for product fication or to characterize the quantity of active compound, i.e., .
Pharmaceutical preparations that can be used orally also include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a coating, such as ol or sorbitol. Push-fit capsules can contain active ients mixed with fillers or binders, such as lactose or starches, lubricants, such as talc or ium stearate, and, optionally, stabilizers. In soft capsules, the active compounds may be ved or suspended in suitable liquids, such as fatty oils, liquid, or liquid polyethylene glycol with or without stabilizers.
Additional pharmaceutical compositions will be evident to those skilled in the art, including formulations ing polypeptides in sustained- or controlled-delivery formulations. Techniques for formulating a variety of other sustained- or controlled-delivery 2012/070571 means, such as liposome carriers, bio-erodible microparticles or porous beads and depot injections, are also known to those skilled in the art. See for example, PCT/US93/00829 that describes controlled release of porous polymeric microparticles for the delivery of pharmaceutical compositions. Additional examples of sustained-release preparations include semipermeable polymer matrices in the form of shaped es, e. g. films, or microcapsules.
Sustained release es may include polyesters, hydrogels, polylactides (US. 3,773,919, EP 58,481), copolymers of L-glutamic acid and gamma ethyl-L-glutamate (Sidman et al., Biopolymers, 22:547-556 (1983), poly (2-hydroxyethyl-methacrylate) (Langer et al., J.
Biomed. Mater. Res., 15:167-277, (1981); Langer et al., Chem. Tech.,12:98-105(1982)), ne vinyl acetate (Langer et al., supra) or poly-D(-)hydroxybutyric acid (EP 133,988).
Sustained-release compositions also include liposomes, which can be prepared by any of l methods known in the art. See e.g., Eppstein et al., PNAS (USA), 823688 ; EP 36,676; EP 88,046; EP 143,949. [0 0 8 5] The pharmaceutical composition to be used for in vivo stration typically must be sterile. This may be accomplished by filtration through sterile filtration nes.
Where the composition is lyophilized, sterilization using this method may be conducted either prior to or following lyophilization and reconstitution. The ition for parenteral administration may be stored in lyophilized form or in solution. In addition, parenteral compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper able by a hypodermic injection needle.
Once the pharmaceutical composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or a dehydrated or lyophilized powder. Such formulations may be stored either in a ready-to-use form or in a form (e.g., lyophilized) requiring reconstitution prior to administration.
In a specific embodiment, the present invention is directed to kits for producing a single-dose administration unit. The kits may each contain both a first container having a dried n and a second container having an aqueous formulation. Also included within the scope of this invention are kits containing single and multi-chambered pre-filled syringes (e.g., liquid syringes and lyosyringes).
An effective amount of a pharmaceutical composition to be ed therapeutically will depend, for example, upon the eutic context and objectives. One skilled in the art will appreciate that the appropriate dosage levels for treatment will thus vary ing, in part, upon the molecule delivered, the indication for which the polypeptide is being used, the route of administration, and the size (body weight, body surface or organ size) and condition (the age and general health) of the patient. ingly, the clinician may titer the dosage and modify the route of administration to obtain the optimal therapeutic . A typical dosage may range from about 0.1mg/kg to up to about 100 mg/kg or more, depending on the factors mentioned above. Polypeptide compositions may be preferably injected or administered intravenously. Long-acting pharmaceutical compositions may be administered every three to four days, every week, or biweekly depending on the half-life and clearance rate of the particular formulation. The frequency of dosing will depend upon the pharmacokinetic parameters of the polypeptide in the formulation used. Typically, a composition is administered until a dosage is reached that achieves the desired effect. The composition may therefore be administered as a single dose, or as multiple doses (at the same or different concentrations/dosages) over time, or as a continuous infusion. Further refinement of the appropriate dosage is routinely made. Appropriate dosages may be ascertained through use of appropriate dose-response data.
The route of administration of the ceutical composition is in accord with known s, e.g. orally, through injection by intravenous, intraperitoneal, intracerebral (intra-parenchymal), intracerebroventricular, intramuscular, intra-ocular, intraarterial, intraportal, intralesional routes, intramedullary, intrathecal, entricular, transdermal, subcutaneous, or intraperitoneal; as well as intranasal, l, topical, sublingual, urethral, vaginal, or rectal means, by sustained release systems or by implantation devices. Where desired, the itions may be administered by bolus injection or continuously by infusion, or by implantation device. Alternatively or additionally, the ition may be administered locally via implantation of a membrane, sponge, or another appropriate al on to which the desired molecule has been absorbed or encapsulated. Where an implantation device is used, the device may be implanted into any le tissue or organ, and ry of the desired le may be via diffiJsion, timed-release bolus, or continuous administration.
In some cases, the vActRIIB polypeptides of the present invention can be delivered by implanting certain cells that have been genetically engineered, using s such as those described herein, to express and secrete the polypeptide. Such cells may be animal or human cells, and may be autologous, heterologous, or xenogeneic. Optionally, the cells may be immortalized. In order to decrease the chance of an immunological response, the cells may be encapsulated to avoid infiltration of surrounding tissues. The encapsulation als are typically biocompatible, semi-permeable polymeric ures or membranes that allow the release of the polypeptide product(s) but prevent the destruction of the cells by the patient’s immune system or by other detrimental s from the nding tissues. [009 1] vActRIIB gene therapy in vivo is also envisioned wherein a nucleic acid molecule ng vActRIIB, or a derivative of vActRIIB is introduced directly into the subject. For example, a nucleic acid sequence encoding a vActRIIB is introduced into target cells via local injection of a c acid construct with or without an appropriate delivery vector, such as an adeno-associated virus vector. Alternative viral vectors include, but are not limited to, retroviruses, adenovirus, herpes simplex, virus and papilloma virus vectors.
Physical er of the virus vector may be achieved in vivo by local injection of the desired c acid construct or other appropriate delivery vector containing the desired nucleic acid sequence, liposome-mediated transfer, direct injection (naked DNA), or microparticle bombardment (gene-gun).
USES OF vActRIIB COMPOSITIONS The present invention provides methods and pharmaceutical compositions for reducing or neutralizing the amount or activity of myostatin, activin A, or GDF-ll in vivo and in vitro by contacting the polypeptides with IB polypeptide. vActRIIB polypeptides have a high affinity for myostatin, activin A, and GDF-l l, and are capable of reducing and inhibiting the biological activities of at least one of myostatin, n A and GDF-l 1. In some embodiments, the vActRIIB polypeptides exhibit improved activity compared with the wild type ActRIIB polypeptides. This is demonstrated in the Examples below.
In one aspect, the present ion provides methods and reagents for ng myostatin-related and/or activin A related disorders in a subject in need of such a treatment by stering an effective dosage of a vActRIIB ition to the subject. As used herein the term “subject” refers to any animal, such as mammals including humans.
The compositions of the present invention are used to increase lean muscle mass as a percentage of body weight and decrease fat mass as percentage of body weight.
The disorders that can be d by a vActRIIB ition include but are not limited to various forms of muscle wasting, as well as metabolic disorders such as diabetes and related ers, and bone degenerative diseases such as osteoporosis. The vActRIIB itions have been demonstrated to be effective in treating muscle wasting disorders in various disease models set forth in Example 3 below. This demonstrated in the treatment of muscle wasting in inhibin-(x knockout mice, treatment of muscle wasting in 26 cancer ia models, prevention of muscular atrophy in hind limb suspension model, treatment of OXV female showing increase in lean muscle mass, decrease in fat mass and increase in bone mineral content.
Muscle wasting disorders also include phies such as Duchenne’s ar dystrophy, progressive muscular dystrophy, Becker’s type muscular dystrophy, Dej erine- Landouzy muscular dystrophy, Erb’s muscular dystrophy, and infantile neuroaxonal muscular dystrophy. Additional muscle wasting disorders arise from chronic diseases or disorders such as amyotrophic lateral sclerosis, congestive obstructive pulmonary disease, cancer, AIDS, renal failure, organ atrophy, androgen deprivation, and rheumatoid tis.
Over-expression of myostatin and/or activin may bute to cachexia, a severe muscle and fat wasting syndrome. The effectiveness of the vActRIIB polypeptides in treating cachexias in animal models is shown in Example 3 below. Cachexia also arises due to rheumatoid arthritis, diabetic nephropathy, renal failure, chemotherapy, injury due to burns, as well as other causes. In another e, serum and intramuscular trations of myostatin-immunoreactive protein was found to be increased in men exhibiting AIDS-related muscle wasting and was inversely d to fat-free mass (Gonzalez-Cadavid et al., PNAS USA 95: 1493 8-14943 (1998)). Myostatin levels have also been shown to increase in response to burns es, resulting in a catabolic muscle effect (Lang et al, FASEB J 15, 1807-1809 (2001)). Additional conditions resulting in muscle wasting may arise from inactivity due to disability such as confinement in a wheelchair, prolonged bed rest due to stroke, illness, spinal chord injury, bone fracture or trauma, and muscular atrophy in a microgravity nment (space flight). For example, plasma myostatin immunoreactive protein was found to increase after ged bed rest (Zachwiej a et al. J Gravit Physiol. 6(2): 1 l(l999). It was also found that the muscles of rats exposed to a microgravity environment during a space e flight expressed an increased amount of tin compared with the muscles of rats which were not exposed (Lalani et al., J.Endocrin 167 (3):417-28 (2000)).
In addition, age-related increases in fat to muscle ratios, and age-related muscular y appear to be related to myostatin. For example, the average serum myostatin- immunoreactive protein increased with age in groups of young (19-35 yr old), middle-aged (3 6- 75 yr old), and elderly (76-92 yr old) men and women, while the average muscle mass and fat- free mass declined with age in these groups (Yarasheski et al. J Nutr Aging 6(5):343-8 (2002)).
In addition, myostatin has now been found to be sed at low levels in heart muscle and expression is lated in cardiomyocytes after infarct (Sharma et al., J Cell Physiol. 180 (1): 1-9 (1999)). Therefore, reducing myostatin levels in the heart muscle may improve recovery of heart muscle after infarct.
Myostatin also appears to influence lic disorders including type 2 diabetes, noninsulin-dependent diabetes mellitus, hyperglycemia, and y. For example, lack of myostatin has been shown to improve the obese and diabetic phenotypes of two mouse models (Yen et al. FASEB J. 8:479 . It has been demonstrated in US. application serial no: 1 1/590,962, US. application publication No: 2007/01 17130, AAV-ActRIIBS vectors increases the muscle to fat ratio in an animal, in particular for obese animal models. The vActRIIB polypeptides of the present disclosure, such as SEQ ID NOS: 4, 6, 8, 10, 12, 14, 16, , 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 52, 54, 56, 60, 62, 64, 66, 68, 70, 72, 87, 88, 91, 93, 95 are suitable for such uses. Therefore, decreasing fat composition by administering the compositions of the present invention will e diabetes, y, and hyperglycemic conditions in animals. In addition, compositions containing the vActRIIB polypeptides may decrease food intake in obese individuals, as demonstrated in US. application serial no: 1 1/590,962, US. ation publication No: 2007/01 17130 for the ActRIIBS polypeptide. [0 0 1 0 0] Administering the ActRIIB polypeptides of the present invention may improve bone strength and reduce osteoporosis and other degenerative bone diseases. This has been demonstrated in the OVX mouse model described below. It has been also been found, for example, that myostatin-deficient mice showed increased mineral content and y of the mouse humerus and increased mineral content of both trabecular and cortical bone at the regions where the muscles , as well as increased muscle mass (Hamrick et al. Calcif Tissue Int 71(1):63-8 (2002)). In addition, the vActRIIB compositions of the present invention can be used to treat the effects of androgen ation such as androgen deprivation therapy used for the treatment of prostate cancer. [00 1 0 1] The present invention also provides methods and compositions for increasing muscle mass in food animals by administering an effective dosage of the vActRIIB proteins to the animal. Since the mature C-terminal tin polypeptide is identical in all species tested, vActRIIB polypeptides would be ed to be effective for increasing muscle mass and reducing fat in any agriculturally important species including cattle, chicken, turkeys, and pigs. [00 1 02] The vActRIIB polypeptides and itions of the present invention also antagonize the activity of activin A. n A is known to be expressed in certain types of cancers, particularly gonadal tumors such as ovarian carcinomas, and to cause severe cachexia.
(Ciprano et al. Endocrinol 141 (7):2319-27 (2000), Shou et al., Endocrinol 138 (11):5000-5 (1997); Coerver et al, Mol inol 10(5):534-43 (1996); Ito et al. British J Cancer 82(8): 1415-20 (2000), Lambert-Messerlian, et al, logic Oncology 74:93-7 (1999). In Example 3 below, the vActRIIB ptides of the present invention have been demonstrated to be effective in treating severe ia, reducing tumor size, and prolonging survival in WO 06175 inhibin-u knockout mice models and colon-26 cancer cachexia mouse models. Therefore, the compositions of the present disclosure can be used to treat conditions related to activin A overexpression, as well as myostatin expression, such as cachexia from certain cancers and the treatment of certain gonadal type tumors and melanoma. [00 1 03] The compositions of the present disclosure may be used alone or in combination with other therapeutic agents to enhance their therapeutic effects or decrease ial side effects. These properties include increased activity, increased solubility, reduced degradation, increased half-life, reduced toxicity, and reduced immunogenicity. Thus the compositions of the t disclosure are useful for extended treatment s. In addition, the properties of hydrophilicity and hydrophobicity of the compounds of the invention are well balanced, thereby ing their utility for both in vitro and especially in vivo uses. Specifically, compounds of the disclosure have an appropriate degree of solubility in aqueous media that permits absorption and ilability in the body, while also having a degree of lity in lipids that permits the compounds to traverse the cell membrane to a putative site of action, such as a particular muscle mass. [00 1 04] The vActRIIB polypeptides and itions of the present invention can be used in ation with herapeutic agents for treating . Chemotherapeutics can include anti-neoplastic drugs. herapeutics can also include alkylating agents, anti- metabolites, plant alkaloids and terpenoids. [00 1 05] Alkylating agents can comprise cisplatin and caroplatin. Anti-metabolites can include purines (e. g. azathiprine or mercaptopurine) or pyrimidines. Additionally, chemo- therapeutic agents can be nucleoside analogues, e.g. 5-fluorocil. Vinca alkaloids, such as vincristine or vinblastine can also be used. [00 1 0 6] Additional anti-neoplastic agents can include, for example, alkylating agents including: nitrogen mustards, such as mechlorethamine, cyclophosphamide, ifosfamide, melphalan and chlorambucil; oureas, such as carmustine (BCNU), lomustine (CCNU), and semustine (methyl-CCNU); ethylenimines/methylmelamine such as thriethylenemelamine (TEM), triethylene, thiophosphoramide (thiotepa), hexamethylmelamine (HMM, altretamine); alkyl sulfonates such as an; triazines such as dacarbazine ; antimetabolites including folic acid analogs such as methotrexate and trimetrexate, pyrimidine analogs such as S-fluorouracil, fluorodeoxyuridine, gemcitabine, cytosine arabinoside (AraC, cytarabine), 5- azacytidine, 2,2’-difluorodeoxycytidine, purine analogs such as 6-mercaptopurine, 6- anine, azathioprine, 2’-deoxycoformycin (pentostatin), erythrohydroxynonyladenine (EHNA), fludarabine phosphate, and 2-chlorodeoxyadenosine (cladribine, 2-CdA); natural _ 29 _ products including totic drugs such as paclitaxel, vinca alkaloids including vinblastine (VLB), vincristine, and vinorelbine, taxotere, estramustine, and estramustine phosphate; ppipodophylotoxins such as etoposide and teniposide; antibiotics such as actimomycin D, daunomycin (rubidomycin), doxorubicin, ntrone, idarubicin, bleomycins, ycin amycin), cinC, and actinomycin; enzymes such as L-asparaginase; biological response rs such as interferon-alpha, IL-2, G-CSF and GM-CSF; miscellaneous agents including platinium coordination complexes such as cisplatin and carboplatin, anthracenediones such as mitoxantrone, tuted urea such as hydroxyurea, methylhydrazine derivatives including N—methylhydrazine (MIH) and procarbazine, adrenocortical suppressants such as mitotane (o,p’-DDD) and aminoglutethimide; hormones and antagonists including adrenocorticosteroid antagonists such as prednisone and equivalents, dexamethasone and aminoglutethimide; progestin such as hydroxyprogesterone caproate, medroxyprogesterone acetate and megestrol acetate; estrogen such as diethylstilbestrol and ethinyl estradiol equivalents; antiestrogen such as tamoxifen; androgens including testosterone propionate and fluoxymesterone/equivalents; antiandrogens such as flutamide, gonadotropin-releasing hormone analogs and leuprolide; and non-steroidal antiandrogens such as flutamide. [00 1 0 7] In addition, the vActRHB polypeptides ofthe t invention are useful for detecting and quantitating myostatin, activin A, or GDF-ll in any number of assays. In general, the ActRIIB polypeptides of the present invention are useful as capture agents to bind and immobilize tin, activin A, or GDF-ll in a variety of assays, r to those described, for example, in Asai, ed., Methods in Cell Biology, 37, Antibodies in Cell Biology, Academic Press, Inc., New York (1993). The polypeptides may be labeled in some manner or may react with a third molecule such as an antibody which is labeled to enable myostatin to be detected and quantitated. For example, a polypeptide or a third molecule can be modified with a detectable moiety, such as biotin, which can then be bound by a fourth molecule, such as enzyme-labeled streptavidin, or other proteins. (Akerstrom, J Immunol 135:25 89 (1985); Chaubert, Mod Pathol 5 (1997)). [00 1 0 8] The invention having been described, the following examples are d by way of ration, and not limitation.
EXAMPLES E 1: EXPRESSION AND PURIFICATION OF VACTRIIB POLYPEPTIDES [00 1 0 9] The ing methods were used for expressing and purifying the variant ActRIIB polypeptides. [00 1 1 0] The cDNA of the human actiVin type IIB receptor was isolated from a cDNA library of human testis origin (Clontech, Inc.) and cloned as described in US. application serial no: 11/590,962, US. application publication No: 2007/0117130.
DETERMINATION OF AMINO ACID SUBSTITUTIONS [00 1 1 1] The combined approaches of structure analysis, molecular modeling, and mass spectrometry ted that aggregation (oligomerization) may arise in ActRIIB through olecular disulfide bond formation triggered by electrostatic and H-bonding interactions between nonglycosylated ActRIIB molecules. The residues 28 and 40 were determined to be involved in ActRIIB:ActRIIB interactions and not in ActRIIB interactions with its ligands. [00 1 1 2] Initially, E28 and R40 on ActRIIB-Fc were substituted with A at each position.
Light scattering and mass spectrometry analyses confirmed that the n of fully ylated VActRIIB-IgGch, E28A, and VActRIIB-IgGch R40A was significantly increased compared to wild-type protein. E28A and R40A VActRIIB-IgGch were ted at 37 °C for 6 days, resulting little or no aggregation compared to wild type. Amino acid substitutions at positions 28 and 40 (with respect to SEQ ID NOS: 2 and 18 with the signal sequence) were made to alleviate or prevent aggregation that can occur during expression or purification of the wild-type ActRIIB (SEQ ID NOS: 2 and 18). This ation has been identified as structured oligomer formation during sion and non-structured aggregate generation both during expression and after protein purification. [00 1 1 3] Aggregation at different stages of the production and ation processes was determined using size exclusion tography according to the ure below. [00 1 1 4] The following exemplary method was used to produce the variant ActRIIB polypeptides (VActRIIB and VActRIIBS). Polynucleotides encoding the VActRIIB, E28W (SEQ ID NO: 23) were fused to polynucleotides encoding human IgG1 Fc domain (SEQ ID NO: 82) or polynucleotides encoding human IgG2 Fc (SEQ ID NO: 84), Via a hinge linker sequence (nucleotides encoding SEQ ID NO: 79) using PCR overlap extension using primers containing the mutation resulting in E28W. The fill polynucleotide sequence is SEQ ID NO: 61. Double stranded DNA fragments were subcloned into pTTS (Biotechnology Research Institute, National ch Council Canada (NRCC), 6100 Avenue Royalmount, Montreal (Quebec) Canada H4P 2R2), pDSROL (described in WO/9014363) and/or derivatives of pDSR0L.
In other embodiments, polynucleotides encoding IB polypeptides were attached to polynucleotides encoding linker GGGGS (SEQ ID NO: 75) or multimers thereof, and or hinge linkers (such as SEQ ID NO: 79). [00 1 1 5] Transient expression of engineered vActRIIB-Fc and vActRIIB5-Fc was carried out as follows. [00 1 1 6] The engineered variants of the above two molecules were expressed transiently in serum-free suspension adapted 293-6E cells (National Research Council of Canada, Ottawa, Canada) maintained in FreeStyleTM medium (Invitrogen ation, Carlsbad, CA) supplemented with 250 ug/ml geneticin (Invitrogen) and 0.1% Pluronic F68 (Invitrogen).
Transfections were performed as 1L cultures. Briefly, the cell inoculum was grown to 1.1 X 106 cells/ml in a 4L fernbach shake flask (Corning, Inc.). The shake flask culture was maintained on an Innova 2150 shaker platform (News Brunswick Scientific, Edison, NJ) at 65 RPM which was placed in a fied incubator maintained at 37°C and 5% CO2. At the time of transfection, the 293-6E cells were diluted to 1.0 X 106 cells/ml. [00 1 1 7] The transfection complexes were formed in 100 ml FreeStyle medium. 1 mg plasmid DNA was first added to the medium ed by 3 ml of FuGene HD ection reagent (Roche Applied Science, Indianapolis, IN). The transfection complex was ted at room temperature for approximately 15 minutes and then added to the cells in the shake flask.
Twenty-hour hours post ection, 20% (w/v) of peptone TNl (OrganoTechnie S.A., TeknieScience, QC, Canada) was added to reach a final concentration of 0.5% (w/v). The transfection/expression was performed for 4-7 days, after which the conditioned medium was harvested by centrifilgation at 4,000 RPM for 60 minutes at 4°C. [00 1 1 8] Stable ection and expression was carried out as follows. The vActRIIB- human (hu) IgG2-Fc cell lines were created by transfecting stable CHO host cells with the sion ds pDC323-vActRIIB (E28W)—hngG2 EC and pDC324-vActRIIB (E28W)- hngG2 Fc (according to Bianchi et al., Biotech and Bioengineering, 84(4):439-444 (2003)) using a standard electroporation procedure. After transfection of the host cell line with the sion plasmids the cells were grown in serum-free selection medium without GHT for 2-3 weeks to allow for selection of the plasmid and recovery of the cells. Cell are selected until they achieved greater than 85% viability. This pool of transfected cells was then cultured in medium containing 150 nM methotrexate.
CELL LINE CLONING [00 1 1 9] A cell bank was made of selected clones according to the ing procedure.
An amplified pool of stable transfected cells was seeded in 96-well plates, and ate clones were evaluated for growth and productivity mance in small-scale studies. Pre-master cell banks (PMCB) of approximately 60 vials were prepared from the chosen clone. All PMCBs were tested for sterility, mycoplasma and viruses. [00 1 2 0] A vActRIIB-Fc expressing cell line was scaled up using a typical fed-batch process. Cells were inoculated into a Wave bioreactor (Wave h LLC). Culture was fed three times with bolus feeds. 10L were harvested on day 10, the remainder was harvested on day 11; both harvests underwent depth filtration followed by sterile filtration. The conditioned media was filtered through a 10 inch 0.45/02 micron pre filter, ed by a filtration through a 6 inch 0.2 micron filter.
PROTEIN PURIFICATION [00 1 2 1] Approximately 5 L of the conditioned medium ning ActRIIB-Fc (both IgG1 and IgG2), ActRIIBS-Fc (both IgG1 and IgG2), and variants of these were concentrated using a 5 ft2 10K ne tangential flow filter (Pall). The concentrated material was applied to a 5 mL Protein A High Performance ColumnTM (GE care) which had been equilibrated with PBS cco’s with no magnesium chloride or calcium chloride). After washing the column with the equilibration buffer until the absorbance at 280 nm (OD280) was less than 0.1, the bound protein was eluted with 0.1 M glycine-HCl, pH 2.7, and immediately neutralized with 1 M Tris-HCl, pH 8.5. The neutralized eluted pool was concentrated to a volume of 1 ml and applied to a 320ml Sephacryl -200 column (GE Healthcare) that was equilibrated in PBS cco’s with no magnesium chloride or calcium chloride. A 4-20% SDS PAGE gels (Invitrogen) were run to determine which fractions to pool. These polypeptides were tested for activity, and degree of aggregation, as shown below. [00 1 22] Optionally, the polypeptides can be further purified, using for example, using a Shp-Sepharose column. Concentration was determined using OD280.
EXAMPLE 2: IN VITRO ACTIVITY ASSAYS [00 1 23] Samples of vActRIIB polypeptides d as described above were diluted with phosphate-buffered saline (PBS: 2.67 mM potassium chloride, 138 mM sodium chloride, 1.47 mM potassium phosphate monobasic, 8.1 mM sodium phosphate dibasic, pH 7.4) to 0.2 mg/ml, ted at 37 °C for 6 days, then applied to MALDI-MS (matrix-assisted laser _ 33 _ desorption/ionization mass ometry), SEC and/or SEC-LS analyses. The aggregation of the wt and variant polypeptides after the protein A purification step were determined using SEC or SEC-LS, and the lar weight of the molecules confirmed using the MALDI-MS procedure described below. [00 1 24] Size exclusion chromatography (SEC). Experiments were performed on an Agilent 1100 HPLC system with two s (TOSOHAAS G3000swxl, 7.8 X 300 mm) in tandem. 2x PBS was used as the mobile phase at 0.5 ute. [00 1 2 5] Size exclusion chromatography-light scattering (SEC-LS). Experiments were performed on an Agilent 1100 HPLC system with a Superdex-200 gel ion column (Amersham Pharmacia, Waukesha, WI). The s were then passed through a Wyatt miniDawn LS laser light scattering detector and Wyatt Optilab DSP Refractometer (Wyatt Technology Co., Santa Barbara, CA) to determine the molecular mass. PBS was used as the mobile phase at 0.4 ml/minute. [00 1 2 6] Matrix-Assisted laser Desorption /Ionization Mass Spectrometry. s were mixed (1 : l) with sinapinic acid and applied to MALDI-MS (Applied tems Voyager System 2009). This procedure was used to check molecular weight of the molecules. [00 1 2 7] Determination of binding affinity, and IC50 values for activin and myostatin were obtained as described below. [00 1 2 8] Qualitative BIAcore® Assay. E28 and R40 were substituted respectively with other natural amino acids in fusions with IgGl PC as bed above. These were generated with or without linkers, as shown in the Tables below. Each vActRIIB-IgGch sample from conditioned media was captured on goat uman IgGl Fc antibody (Jackson Immuno Research, cat# 109098, lot 63550) coated CM5 surface. 20 nM of Activin A was ed over captured sample surfaces using BIACore2000 (BIACore Life Sciences, Piscataway, New ). The resulting sensorgrams were normalized to the captured RL (500 RU) of vActRIIB-IgGch variants. The normalized binding response (RU) for some variants are shown in Table 2, and is further bed below. Relative binding y for activin was also determined by Biacore measurements using conditioned media obtained from mammalian cell expression. Activin A (20 nM) was used to capture soluble receptor polypeptide in the conditioned media and measured SPR signals were normalized. Normalized SPR of +++++: > 60,++++140——60,+++:20——40,++:10——20,+:5——10,-:<5. [00 1 2 9] Table 1A and Table 1B summarize the results of the relative binding data. The table below shows that certain embodiments of the vActRIIB-IgGch in particular bound to activin A with higher affinity than wild type, or retained comparable affinity with the wild type.
WO 06175 Table 1A: Wild-type and Engineered ActRHB-IgGl Fc Binding (Stable Transfectants) Relative . Linker (SEQ actiVin -cno nnnnns nonconzo -cno s noon -CH0 ActRIIBS E28A GGGGS -CHO ActRIIBS E28W R4OA GGGGS -CHO ActRIIB none (E28) GGGGS -CHO ActRIIB E28A GGGGS -CHO ActRIIB E28A 2(GGGGS) cos nnnnn <n2s> cos nnnnn noon cos ActRIIB none (E28) R4OA Table 1B: Wild-type and Engineered ActRHB-IgGl Fc Binding (Transient Transfectants) Relative . Linker (SEQ actiVin bindin_ cos ActRHB E28W GGGGS cos ActRHB E28Y GGGGS cos ActRIIB none (E28) R4OG GGGGS cos ActRHB E28F GGGGS cos ActRIIB none (E28) GGGGS cos —————-—+ cos nnnnnn noon ocoos — . Relative L1nker (SEQ CHO Expression Molecule Res28 Res40 activin ID NO. 75) bindin' cos —————- C2C12 CELL BASED ACTIVITY ASSAY [00 1 3 0] vActRIIB5-IgG1Fc and vActRIIB-IgGch variants were generated as described above. The y of these variants to inhibit the binding of activin A or myostatin to the activin IIB receptor was tested using a cell bascd activity assay as described below. [0 0 1 3 1] A myostatin/activin/GDF-l 1-rcsponsivc reporter cell line was ted by transfection of C2C12 myoblast cells (ATCC No: CRL-1772) with a pMARE-luc uct.
The pMARE-luc uct is made by cloning twclvc s of thc CAGA sequence, representing the myostatin/activin response clcmcnts (Dcnnlcr ct al. EMBO 17: 3091-3100 (1998)) into a pLuc-MCS reporter vector (Stratagcnc cat # 219087) upstream of the TATA box.
The C2C12 cells naturally express activin or IIB on their cell surface. When myostatin/activinA/GDF-ll binds the cell ors, the Smad pathway is activated, and phosphorylatcd Smad binds to the response element (Macias-Silva ct al. Cell 87: 1215 (1996)), resulting in the expression of thc lucifcrasc gcnc. Lucifcrasc activity was then measured using a commercial rasc reporter assay kit (cat # E4550, Promega, Madison, WI) according to manufacturer’s protocol. A stable line of C2C12 cells that has been transfcctcd with pMARE- luc (C2C12/pMARE) was used to measure activity according to the following procedure.
Reporter cells were plated into 96 well cultures. Screening using dilutions of the wild type and variant ActRIIB-IgGl Fc fusions constructed as described above was performed with the concentration fixed at 4 nM activin. Activin A was cubated with the receptors at several concentrations. Activin activity was measured by determining the rase activity in the treated cultures. The IC50 values were determined for each polypeptide. These are shown in Table 2. The same procedure was followed for ActRIIB-hngG2 Fc fusions produced as described above for the determination of myostatin. Protein A purified wt and variants were used in the determination of IC50 values for myostatin using the same methodology. For this determination, the polypeptides were pre-incubated with 4 nM myostatin. In addition, the degree of aggregation was ined using the procedures described above. These values are given in Table 3 below. [00 1 3 2] Out of the set of ActRIIB5-IgG1 Fc variants that are listed in Table 1A, several ActRIIB-IgGl Fc variants and three ActRIIB5-IgG1 Fc variants together with the wild type polypeptides were fiarther purified and analyzed by SPR (surface plasmon resonance) at 20 nM activin A. Table 2 shows the SPR binding affinity of selected vActRIIB-IgGl Fc polypeptides for activin. n A (20 nM) was used to capture vActRIIB polypeptides in the samples and ed SPR signals were normalized. IC50 values were obtained from cell-based activin inhibition assays bed above. Standard errors are less than 10% for all results.
Table 2 . SPR normalized RU Var1ant IC50 (nM) Act1v1n. .
(RU 2 response unit) ActRIIB-IgGch (SEQ 35 8.20 ID NO:58) vActRIIB-IgGch, 20 25.30 E28A (SEQ ID NO:60) vActRIIB-IgGch, 128 2.07 E28W (SEQ ID NO:62) vActRIIB-IgGch, 115 2.10 E28Y (SEQ ID NO:64) vActRIIB-IgGch, 18 R40G (SEQ ID NO:66) ActRIIB5-IgG1Fc (SEQ 37 ID NO:68) IB5-IgG1Fc, 8 E28A (SEQ ID NO:70) vActRIIB5-IgG1Fc, 45 16.86 E28W (SEQ ID NO:72) 2012/070571 [00 133] As shown above in Table 2 above, the ICSO value of vActRIIB-IgGch (E28W) for blocking activin was 2.07 nM and the ICSO value of vActRIIB-IgGch (E28Y) was 2.1 nM ed to wild type. Furthermore, the E28W and E28Y variants ofvActRHB-IgGch were stable and not aggregated once purified. [00 1 3 4] The ICSO value in a myostatin blocking cell based assay was determined for additional variant polypeptides as well. These variants were the mature truncated vActRHB polypeptides lacking the signal sequence and the first six amino acids of the N—terminal. These sequences are shown in Table 3. Table 3 shows the percent aggregation of the protein after protein A purification, and the ICSO value with respect to myostatin. It can be seen that the percent aggregation is much less for the variant polypeptides ed with the wild type.
Similar results were obtained for mature truncated vActRHB polypeptides without the signal sequence and the N—terminal four amino acids, and with identical tutions as shown below.
Table 3 IC50 (HM) Ac\tfl:rIiI:fiFc 0 ActRIIB a re/Oation myostatin gg g cell based hActRIIB- ETREZSCIYYNANWEL GGGGS PSVFLFPP hIngFc ERTNQSGLERCEGEQ VECPPC KPKDTLMISRTPEV (SEQ ID DKRLHCYASWRNSS P (SEQ TCVVVDVSHEDPE NO: 89) GTIELVKKGCWLDDF ID VQFNW Y VDGVEV NCYDRQECVATEENP NO:79) HNAKTKPREEQFN CEGNFCNER STFRVVSVLTVVH FTHLPEAGGPEVTYE QDWLNGKEYKCK PPPTAPT (SEQ ID NO: PAPEIKTIS 86) KTKGQPREPQVYT LPPSREEMTKNQVS LTCLVKGFYPSDIA VEWESNGQPENNY KTTPPMLDSDGSFF LYSKLTVDKSRWQ QGNVFSCSVMHEA LHNHYTQKSLSLSP GK (SEQ ID NO: 80) IC50 (HM) AcéRHBEFC 0A) ActRIIB myostatin arian aggrega 1011t' cell based hActRIIB- ETRW28CIYYNANWE GGGSV APPVAGPSVFLFPP hIngFc LERTNQSGLRCEGEQ ECPPCP KPKDTLMISRTPEV (E28W) DKRLHCYASWRNSS (SEQ ID TCVVVDVSHEDPE (SEQ ID GTIELVKKGCWLDDF NO:79) VQFNWYVDGVEV N09 1) NCYDRQECVATEENP HNAKTKPREEQFN QVYFCCCEGNFCNER SVLTVVH FTHLPEAGGPEVTYE QDWLNGKEYKCK PPPTAPT (SEQ ID NO: VSNKGLPAPIEKTIS 87) KTKGQPREPQVYT LPPSREEMTKNQVS LTCLVKGFYPSDIA VEWESNGQPENNY KTTPPMLDSDGSFF LYSKLTVDKSRWQ QGNVFSCSVMHEA LHNHYTQKSLSLSP GK SEQ ID NO: 80 hActRIIB- ETRY28CIYYNANWEL GGGGS APPVAGPSVFLFPP hIngFc ERTNQSGLERCEGEQ VECPPC KPKDTLMISRTPEV (E28Y) DKRLHCYASWRNSS P (SEQ TCVVVDVSHEDPE (SEQ ID GTIELVKKGCWLDDF ID VQFNWYVDGVEV NO: 93) CYDRQECVATEENPQ NO:79) HNAKTKPREEQFN EGNFCNERF STFRVVSVLTVVH THLPEAGGPEVTYEP QDWLNGKEYKCK PPTAPT (SEQ ID VSNKGLPAPIEKTIS NO:88) KTKGQPREPQVYT LPPSREEMTKNQVS LTCLVKGFYPSDIA VEWESNGQPENNY KTTPPMLDSDGSFF LYSKLTVDKSRWQ QGNVFSCSVMHEA QKSLSLSP [00 1 3 5] Table 4 fies the sequences corresponding to SEQ ID NOS: 1-99 in the sequence listing.
Table 4 SEQ Description ID NO 1 BS extracellular domain, .01 tide 2 ActRIIBS ellular domain, .01 ooetide 3 VActRIIBS E28A ol nucleotide 4 VActRIIBS E28A ool ooetide VActRIIBS E28A and R40A ool nucleotide 6 VActRIIBS E28A and R40A ool ooetide 7 VActRIIBS E28W ool nucleotide 8 VActRIIBS E28W ool ooetide 9 VActRIIBS E28Y ol nucleotide SEQ Description ID NO VActRIIBS E28Y tide 11 VActRIIBS E28X wherein X is A, F, Q, V, I, L, M, K, H, W or Y .01 nucleotide 12 VActRIIBS E28X wherein X is A, F, Q, V, I, L, M, K, H, W or Y .01 ..etide 13 VActRIIBS E28X and R40X, wherein X(28) is A, F, Q, V, I, L, M, K, H, W or Y wherein X(40) is A, G, Q, M, H, K or N .01 nucleotide 14 VActRIIBS E28X and R40X, wherein X(28) is A, F, Q, V, I, L, M, K, H, W or Y wherein X(40) is A, G, Q, M, H, K or N .01 ..etide VActRIIBS R40X wherein X is G, Q, M, H, K or N 0.1 nucleotide 16 VActRIIBS R40X wherein X is G, Q, M, H, K or N .01 .e.tide 17 ActRIIB extracellular domain, .01 nucleotide 18 ActRIIB extracellular domain, .01 .e.tide 19 VActRIIB E28A .01 nucleotide VActRIIB E28A .01 .e.tide 21 VActRIIB E28A and R40A .01 nucleotide 22 VActRIIB E28A and R40A .01 .e.tide 23 IB E28W .01 nucleotide 24 VActRHB E28W .01 .e.tide VActRIIB E28Y .01 nucleotide 26 VActRIIB E28Y .01 .e.tide 27 VActRIIB E28X wherein X is A, F, Q, V, I, L, M, K, H, W or Y .01 nucleotide 28 VActRIIB E28X wherein X is A, F, Q, V, I, L, M, K, H, W or Y .01 .e.tide 29 VActRIIB E28X and R40X, wherein X(28) is A, F, Q, V, I, L, M, K, H, Y or W wherein X(40) is A, G, Q, M, H, K or N .01 nucleotide VActRIIB E28X and R40X, wherein X(28) is A, F, Q, V, I, L, M, K, H, Y or W wherein X(40) is A, G, Q, M, H, K or N .01 e 31 VActRIIB R40X wherein X is G, Q, M, H, K or N .01 tide 32 VActRIIB R40X wherein X is G, Q, M, H, K or N .01 .e.tide 33 VActRIIB R64A, E28A .01 nucleotide 34 VActRIIB R64A, E28A .01 .e.tide VActRIIB R64A, E28A and R40A .01 tide 36 IB R64A, E28A and R40A .01 .e.tide 37 VActRIIB R64A, E28W .01 nucleotide 38 VActRIIB R64A, E28W .01 e 39 VActRIIB R64A, E28Y .01 tide 40 VActRIIB R64A, E28Y .01 .e.tide WO 06175 SEQ Description ID NO 41 VActRIIB R64A, E28X wherein X is A, F, Q, V, I, L, M, K, H, Y or W .01 nucleotide 42 VActRIIB R64A, E28X wherein X is A, F, Q, V, I, L, M, K, H, Y or W .01 ..etide 43 IB R64A, E28X and R40X, wherein X(28) is A, F, Q, V, I, L, M, K, H, W or Y wherein X(40) is A, G, Q, M, H, K or N .01 nucleotide 44 VActRIIB R64A, E28X and R40X, wherein X(28) is A, F, Q, V, I, L, M, K, H, W or Y wherein X(40) is A, G, Q, M, H, K or N .01 ..etide 45 VActRIIB R64A, R40X wherein X is G, Q, M, H, K or N 0.1 tide 46 IB R64A, R40X wherein X is G, Q, M, H, K or N .01 .e.tide 47 Secuence Accession NP 001097 Wild t .e ActRIIB .01 .e.tide 48 Secuence Accession NM 002192 ActiVin A .01 .e.tide 49 Secuence Accession AAB86694 M ostatin .01 .e.tide 50 ce Accession 095390 GDF-11 .01 .e t.ide 51 IB5 E28X and R40X, wherein X = an amino acid .01 nucleotide 52 IB5 E28X and R40X, wherein X = an amino acid .01 .e.tide 53 VActRIIB E28X and R40X, wherein X = an amino acid .01 nucleotide 54 VActRIIB E28X and R40X, wherein X = an amino acid .01 .e t.ide 55 VActRIIB R64A, E28X and R40X, wherein X = any amino acid .01 nucleotide 56 VActRIIB R64A, E28X and R40X, wherein X = an amino acid .01 .e.tide 57 ActRIIB-IG1Fc rnature .01 nucleotide 58 ActRIIB-I_G1Fc rnature .01 .e.tide 59 VActRIIB-I_G1Fc E28A E10A rnature .01 nucleotide 60 VActRIIB-I_G1Fc E28A E10A rnature .01 .e t.ide 61 VActRIIB-IGch E28W E10W rnature .01 nucleotide 62 VActRIIB-I_G1Fc E28W E10W e .01 .e.tide 63 VActRIIB-I_G1Fc E28Y E10Y rnature .01 tide 64 VActRIIB-I_G1Fc E28Y E10Y rnature .01 .e t.ide 65 VActRIIB-IGch R40G R22G rnature .01 nucleotide 66 VActRIIB-I_G1Fc rnature R40G R22G rnature .01 .e.tide 67 VActRIIB5-IGch rnature .01 nucleotide 68 VActRIIB5-I_G1Fc rnature .01 .e.tide 69 VActRIIB5-I_G1Fc E28A E10A rnature .01 nucleotide 70 IB5-I_G1Fc E28A E10A rnature .01 .e.tide 71 VActRIIB5-IG1Fc E28W E10W rnature .01 nucleotide E10W 72 VActRIIB5-I_G1Fc E28W E10W rnature .01 .e.tide E10W 73 Sinal secuence shown in Fiures 1 and 2 74 Alternative sinal se . uence 75 Linker 76 C0rn.lete hin_e re ions for IG1 77 C0rn.lete hine reion for I _G2 78 C0rn.lete hine reion for I _G4 SEQ Description ID NO 79 Hine linker 80 LG2 Fc 0nl nnetide 81 LG2 Fc nucleotide deenerate 82 LG1 Fc 0nl nnetide 83 IG1 Fc 0nl nucleotide 84 LG4 Fc 0nl nnetide 85 LG4 Fc 0nl nucleotide—deenerate 86 ActRIIB mature truncated wt 0nl nentide 87 VActRIIB E4W E28W mature truncated 0nl nentide 88 IB E4Y E28Y mature truncated 0nl nentide 89 ActRIIB-LG2Fc mature ted 0nl nentide 90 ActRIIB-IG2 Fc mature ted 0ol nucleotide deenerate 91 VActRIIB-LG2Fc E4W E28W mature truncated 0nl nentide 92 VActRIIB-LG2Fc E4W E28W mature truncated 0nl nucleotide 93 IB-LG2Fc E4Y E28Y mature truncated 0nl nentide 94 VActRIIB-LG2Fc E4Y E28Y mature truncated 0nl nucleotide 95 VActRIIB-IG2 Fc E4A E28A mature truncated 0nl nentide 96 VActRIIB-IgG2 Fc (E4A) E28A mature truncated polynucleotide deenerate 97 VActRIIB—IgG2 Fc (E4X) E28X—wherein X is A, F, Q, V, I, L, M, K, H, W or Y mature truncated 0nl ne tnide 98 Fi_ure 1—ActRIIB-IG1 Fe 99 Fi_ure 2—ActRIIB5-IG1 Fc EXAMPLE 3: IN VIVO ENTS USING VActRIIB [00 1 3 6] All of the following animal studies were carried out using the mature truncated VActRIIB-IgG2 Fc (E28W) polypeptide, (SEQ ID NO: 91), according to the procedures described below.
ENT OF MUSCLE WASTING IN INHIBIN-OL DEFICIENT MICE [00 1 3 7] Inhibin-(x is a naturally occurring tor of actiVin A. Mice lacking inhibin-(x show significantly elevated n A levels in circulation and suffer from a lethal wasting syndrome which is associated with the spontaneous ion of tumors such as ovarian, testicular cancers and adrenal cancers (Matzuk et al., PNAS 91(19):8817-21 (1994), Cipriano et al. Endocrinology 121(7): 2319-27(2000), Matzuk et al., Nature 02):3 13-9 (1992)). For the ing experiments, inhibin-u knockout mice (C57BL/6J) were obtained from Charles River Laboratories. The effects of the VActRIIB-IgG2 Fc E28W (SEQ ID NO: 91) (hereinafter E28W, or E28W polypeptide, or soluble receptor E28W) on body weight and muscle mass was examined in inhibin-OL knockout mice. A 14-day single-inj ection study in 8-week-old male inhibin-01 knockout mice was performed. At 8 weeks of age, the male inhibin-01 knockout mice had lost more than 25% of body weight compared to age-matched wild-type littermate control mice. 5 of the knockout mice were given a single subcutaneous injection of E28W (30 mg/kg), while 5 knockout mice were aneously administered an equal volume of PBS (vehicle) on day 0. As a baseline control, 5 age-matched wild-type mice were administered a single subcutaneous injection of vehicle on day 0. The mice were weighed at day 0, day 7 and day 14.
At the end of day 14, all the mice were sacrificed, and their lean carcass weights and calf muscle mass were analyzed via necropsy. Over the l4-day study period, the average body weight of the vehicle-treated knockout mice d by approximately 1.1 g from 22.5 g on day 0 to 21 .4 g on day 14. In st, the average body weight of the E28W-treated knockout mice showed a dramatic gain by 11 g from 22.1 g on day 0 to 33.1 g on day 14. Terminal necropsy is revealed that the E28W polypeptide virtually doubled the lean carcass weight and calf muscle mass in the inhibin-01 knockout mice, as shown below. The average lean carcass weight of the E28W-treated knockout mice was approximately 14.9 g compared with approximately 8.0 g for the e-treated knockout mice, and approximately 12.1 g for vehicle-treated wild-type control mice. The average calf muscle weight (from both legs) of the reated knockout mice was imately 426 mg compared with approximately 209 mg for the vehicle-treated knockout mice, and approximately 324 mg for vehicle-treated wild- type control mice. These results demonstrate the effectiveness of the E28W polypeptide for the treatment of disease states of weight loss and muscle wasting and are summarized in the Table below. _WTplusVehicle KO plus Vehicle KO plus E28W Body Weight *# 28.64 ——/— 1.11 21.36 +/— 099* 33.10 ——/— 1.56 Lean Carcass (g) 12.07 ——/— 0.36 8.00 +/— 029* *# 14.90 ——/— 0.77 CalfMuscle (g) 0.324--/— 0.014 0.209 +/— 0012* 0.426 --/— 0.024*# *2 P<0.05 vs. WT + Veh; #: P<0.05 vs. KO + Veh. [00 1 3 8] The effects of the administration of the E28W polypeptide on the rates of ion of ular and ovarian tumors were examined in male and female inhibin-01 KO mice, respectively. In this study, 11 of the inhibin-01 knockout mice, including -old males (n=5) and 9-week-old s (n=6), were treated with a single subcutaneous ion of E28W (30 mg/kg), while another 11 of the inhibin-01 knockout mice, including age-matched males (n=5 ) and females (n=6) received a single injection of an equal volume of PBS WO 06175 (vehicle). In addition, 11 of the wild-type littermate control mice, including age-matched males (n=5) and females (n=6) were administered a single injection of vehicle. Two weeks after the treatment, the mice were sacrificed and subjected to necropsy to examine the rates of ion of visually identifiable testicular and ovarian tumors. It was observed that 10 of the 11 e- treated knockout mice developed identifiable tumors. Specifically, testicular and ovarian tumor formations were found in 5 out of the 5 males and 5 out of the 6 females examined, respectively. The sizes of these tumors were found to be 2-3 fold larger than the corresponding normal testis or ovary in wild-type control mice. This is shown in Figure 3. Only 10% (1 out of ll) of the E28W-treated inhibin-(x knockout mice showed visible tumor formation.
Specifically, in females, 1 out of 6 of the E28W-treated ut mice developed an identifiable ovarian tumor, whereas 5 out of the 6 of the untreated female uts had little or no change in the size or gross logy of the ovary compared with age-matched wild- type ls. 5 out of 5 of the E28W-treated male knockout mice showed no visible tumors with little or no change in the size or gross logy of the testis in comparison to the age- matched wild-type controls. These s demonstrate that E28W administration was effective in reducing the formation of testicular and ovarian tumors in the inhibin-(x KO mice, and also in reducing the formation of melanomas, suggesting a clinical utility for the soluble receptor therapy in cancer treatment. [00 1 3 9] The effectiveness of the E28W polypeptide in treating anorexia was examined in male inhibin-(x ut mice. In this study, food consumption in the n-(x knockout mice (n=5) was significantly reduced compared to that of the age-matched wild-type mice (n=lO). It was observed that the food intake of the E28W treated inhibin-(x knockout mice was significantly increased during a 3-week period examined. The average weekly food intake in the E28W-treated knockout mice increased to a level slightly higher than that in the age- matched wild-type control mice, and was about 50% greater than the average weekly food intake of the knockout mice treated with the vehicle. Thus, the data show that the E28W treatment was highly effective in ameliorating ia in the inhibin-(x KO mice. [00 1 4 0] The effect of the E28W treatment on al was examined in male and female n-u KO mice, respectively. For males, 25 inhibin-u KO mice around 50 days of age were administered the E28W polypeptide (10 mg/kg/wk, SC), while 26 age-matched inhibin-(x KO mice received vehicle (PBS). 19 aged-matched wild-type male mice received vehicle and were used as baseline control. The vehicle-treated knockout mice started dying on day 15 of the study d 65 days of age). By day 34 of the experiment (around 84 days of age), 50 % of the vehicle-treated knockout mice had died, and by day 78 (around 128 days of age), 100% of them had died. In contrast, none of the 25 knockout mice treated with the E28W polypeptide, or of the 19 wild-type control mice treated with e, died before day 78 of the study (around 128 days of age). In the E28W-treated knockout mice, 1 out of 25 died on day 78 of the study (around 128 days of age) and 24 out of 25 survived beyond day 100 (around 150 days of age). No vehicle-treated wild type mice died during the 100-day study period. Similar survival results were obtained in female inhibin-(x KO mice. 22 of female inhibin-(x KO mice of approximately 50 days of age were treated with E28W (10 mg/kg/wk, SC), while 23 female inhibin-(x KO mice of the same age were treated with PBS (vehicle). In the me, 17 of wild type female control mice were d with vehicle. The vehicle-treated female knockout mice began dying on day 40 of the study (around 90 days of age). By day 58 of the experiment (around 108 days of age), 50% of the vehicle-treated female knockout mice had died, and by day 86 of the study (about 136 days of age) 100% of them had died. In contrast, only about 5% (1 out of 22) of the E28W-treated female knockout mice had died while about 90% (20 out of 22) survived beyond day 120 of the study (about 170 days of age). No vehicle-treated wild type mice died during the l20-day study period. ore, the data demonstrate that the E28W polypeptide therapy is effective in dramatically prolonging the survival of both male and female inhibin-(x knockout mice. A schematic plot of the survival curves for both the male and female ut mice is provided in Figure 4.
TREATMENT OF MUSCLE WASTING IN COLON-26 TUMOR G MICE [00 1 4 1] Colon-26 tumor bearing mice is a widely used preclinical animal model for ng cancer cachexia (Fujita et al., Int J Cancer 68(5):637-43 (1996), Kwak et al., Cancer Research 64(22):8193-8 (2004)). The effect of E28W polypeptide on body weight change, muscle mass and survival rate were studied in the tumor-bearing mice. Colon-26 (C-26) tumor cells were subcutaneously implanted into 40 10-week-old, male CDF1 mice at 0.5 x 106 cells per mouse. The tumor implantation was performed on day 0. Beginning on day 5 after tumor implantation, twenty C-26 mice were treated weekly with a subcutaneous injection of 10 mg/kg vActRIIB IgG2 Fc E28W (SEQ ID NO: 91), while twenty C-26 mice were treated with a vehicle (PBS). At the same time, 10 age and weight matched normal mice were treated with a vehicle (PBS) only. Body weight and food intake were determined 3 times per week. The tumor-bearing mice were inspected twice daily for survival. Tumor sizes were measured using rs (Ultra-Cal IV IP65 electronic caliper, Fred V Fowler Co. Boston MA) connected to a PC er and values were automatically recorded to a worksheet in a Microsoft Excel data file. As shown in Figure 5, two weeks after tumor tation, the mice bearing C-26 tumors developed severe cacheXia and lost their body weight ically. E28W treatment effectively mitigated the body weight loss in the tumor-bearing mice. The average body weight of the bearing mice treated with E28W was significantly higher than that of the tumor- bearing mice treated with vehicle (p<0.001, from day 7 to day 33 after tumor-inoculation.
Unpaired T test, Graph pad Software Inc. San Diego CA). [00 1 42] There was no difference in tumor size between the E28W polypeptide treated and vehicle treated groups indicating that the treatment had no effect on C-26 tumor growth.
Terminal necropsy analysis showed that the average lean carcass mass and calf muscle weight of the E28W -treated C-26 tumor-bearing mice were significantly higher than those of the tumor-bearing mice treated with e (p<0.001 for both lean carcass and calf ). The effect of the E28W on survival of the C-26 tumor-bearing mice is shown in Figure 6. The vehicle treated mice began dying at about day 14 post tumor implantation. At day 35 post tumor implantation, all 20 vehicle treated C-26 tumor-bearing mice died; however 17 out of 20 C-26 tumor-bearing mice treated with E28W were still surviving. Thus, the E28W treatment led to a significant prolongation of survival of the C-26 tumor-bearing mice 001, chi- square test). Therefore, the E28W polypeptide was not only effective in maintaining body weight and muscle mass but also in prolonging the survival of the C-26 tumor-bearing mice.
ENT OF HINDLIMB SUSPENSION MICE [00 1 43] The mb suspension mouse model was used to examine the effect of the vActRIIB-IgG2 Fc E28W (SEQ ID NO: 91) on muscle mass in disuse state. Hindlimb suspension procedure is essentially the same as previously reported by Carlson CJ el al (Carlson CJ, Booth FW and Gordon SE: Am J Physiol Regul Integr Comp l. 277: R601- RR606, l999).Nine-week-old female C57BL/6 mice were used for the study. A total of 60 mice were divided into three groups as follows: 1. Non-suspended ne control group (20 mice) treated with vehicle (PB S), 2. Hindlimb suspension group (20 mice) treated with vehicle, and 3. Hindlimb suspension mice group (20 mice) treated with vActRIIB-IgG2 Fc, E28W. Specifically, a single SC injection of either 30 mg/kg of vActRIIB-IgG 2 Fc E28W or vehicle was given to the above described groups respectively, at the time of the initiation of hindlimb suspension. Body weight s were measured udinally 2-3 times per week. mice from each group were sacrificed at the following 4 different time points: day 1, day 3, day 7 and day 14. Calf muscle weights were determined via necropsy. [00 1 44] As shown in the Table below, hindlimb suspension led to a significant loss in body weight up to 10%. Treatment of the hindlimb suspended mice with vActRIIB-IgG2 Fe E28W led to a significant body weight gain to a level higher than either the vehicle treated mb suspension group or the non-suspended baseline control group as analyzed by ANOVA measurement. During the ek study period, the e body weight gain of the vActRIIB-IgG2 Fc E28W (SEQ ID NO: 91) treatment group was 12.6% in comparison to the 0.2% drop in the vehicle-treated suspension group and 4.8% weight gain in the non- suspened baseline control group, respectively. Time-course necropsy results showed that the mb muscle mass changed in parallel to the body weights. Treatment of the suspended mice with vActRIIB-IgG2 Fc (E28W) tely mitigated the muscle loss. Therefore, the results of this experiment show that E28W is effective in the treatment of muscle atrophy associated with disuse.
Group/days (body day 3 (%) day 7 (%) day 14 (%) weight change %) Non-HS + PBS HS -- vehicle 400% HS -- E28W, 30 -9.7% 2.1% 12.6% mg/ml TREATMENT OF OVX MICE [00 145] Ovariectomized female C57Bl6 mice (OVX) are considered to be a model for female hypogonadism and osteoporesis. 24 female C57Bl6 mice were ovariectomized at age 3 months and allowed to recover for 3 months. At age 6 months, 24 OVX mice as well as 24 age-matched sham operated l C57Bl6 mice were measured for longitudinal changes in body weight, muscle, and fat mass by NMR and bone mass (PIXImus—GE LUNAR Corporation) over a 3 month treatment period. At the end of the period, the animals were ced, and the bone tissues harvested during terminal necropsy and subjected to Faxitron X-ray and microCT (Faxitron X-ray Corporation and GE Medical system) analysis. It was demonstrated to the E28W variant receptor (SEQ ID NO: 91) was effective at increasing body , specifically lean skeletal muscle mass, and bone mass, while decreasing fat content of the mice to the level seen in ariectomized mice. Specifically, over a 12 week period, lean muscle mass was increased from 20 g to 27.0 g for OVX mice d with E28W, compared with 20 g to 27.5 g for sham operated mice treated with E28W, compared with almost no increase in lean muscle mass for OVX plus vehicle or sham plus vehicle (about 19 grams for OVX plus vehicle and about 20 g for wild type plus vehicle). In the same study, OVX mice treated with E28W showed d fat mass from 8 g average per animal to about 4 g average per , comparable with the sham operated animals, by the end of the 12 week study. The OVX mice treated with the vehicle, in contrast, did not lose fat mass at any time during the study. Finally, bone mass was increased in the OVX mice treated with E28W compared with vehicle treated OVX mice. Analysis of femur/tibia BMC (Bone mineral t) of the dissected bone harvested during terminal necropsy was determined by pQCT analysis (Peripheral Quantitative Computed Tomography). The OVX mice treated with E28W sed BMC of about 0.045 g/cm to about 0.055 g/cm at the end of the 12 week study, which is comparable to the final BMC of sham operated vehicle treated animals. The OVX mice treated with vehicle showed about the same BMC of 0.045 g/cm at the end of the 12 week study. The E28W d wild type mice showed an increase ofBMC from about 0.054 g/cm to about 0.065 g/cm at the end of the 12 week study. These studies demonstrate the effectiveness of the E28W polypeptide as potential treatments of frailty, osteoporesis, and obesity in aging.
EXAMPLE 4: TREATMENT OF CANCER WITH SACTRIIB (E28W — SEQ ID NO. 91) [00 1 4 6] Activin levels in mice with n cancer (OC) was investigated (Figure 7A).
Circulating activin A levels were significantly elevated in mice with ovarian cancer (OC) compared to normal control subjects (Normal). Serum activin A was measured by ELISA.
The effect of n cancer treatment with sActRIIB (E28W) (SEQ ID. NO: 91) is shown in Figures 7B-7D. 12-week-old female KO mice with established ovarian tumors were d for 14 days with a single injection of IB or PBS. l, age-matched female WT mates received PBS. It can be seen in Figure 7B that the elevated circulating levels of activin A in the KO mice was rapidly reduced to the WT control level by sActRIIB treatment.
Serum n A was measured by ELISA at day 0, 1 and 14 after treatment. [00 1 4 7] In Figure 7C it can be seen that sActRIIB treatment rapidly reduced the n tumor mass in the KO mice to the WT control level. The weights of ovaries (left and right) of individual animals were analyzed by cohorts via necropsy at day 0, 1 and 14 after treatment.
Representative gross morphology images of ovaries depicting the ic regression of the advanced ovarian tumors in KO mice in response to 14-day sActRIIB treatment. (Figure 7D) [00 1 4 8] Several additional observation were made in KO mice treated with sActRIIB (data not shown). Overexpression of Activin A mRNA was seen in ovarian tumors ofKO mice. The overexpression was prevented by sActRIIB treatment. Additionally, sActRIIB treatment completely blocked the increase in phospho-Smad2 in ovarian tumors in KO mice.
Also, the severe loss of E-cadherin in ovarian tumors of KO mice was reversed by sActRIIB 2012/070571 treatment. y, representative immunohistochemical images showed the complete disappearance of E-cadherin immunoreactivity from ovarian sections ofKO mice and the reappearance of strong E-cadherin staining after l4-day treatment with sActRIIB. [00 1 4 9] Figure 8A shows that Activin A blockade can abolish the overexpression of VEGF in ovarian tumors in n-(x KO mice. l2-week-old female KO mice with established ovarian tumors were d for 14 days with a single injection of sActRIIB or PBS. As controls, age-matched female WT littermates received PBS. ELISA data reveals that the serum VEGF levels are markedly elevated in the KO mice but are reduced to the WT control levels after sActRIIB treatment. [0 0 1 5 0] Additionally, in data not shown, it was observed that immunohistochemical staining images demonstrated dramatically increased VEGF and Ang-l immunoreactivities in the ovarian tumor sections in KO mice, which were completely abolished by sActRIIB treatment. Western blot analysis also revealed that the expressions of tumor angiogenesis- related proteins in, Osteropontin, l and IGFBP-2 were highly induced in ovarian tumors in KO mice but were decreased to the WT control levels after sActRIIB treatment. [00 1 5 1] Immunohistochemical analysis shows that sActRIIB treatment leads to caspase- 3 tion in testicular tumors in the KO mice. In Figure 8B arrows point to the active caspase-3 immunoreactivity in the sActRIIB-treated ovarian tumor section where residual tumor cells are clustered. There was an absence of active caspase-3 immunostaining in the PBS-treated ovarian tumor section. The histogram on the right shows the tative analysis of active caspase-3 based on multiple ovarian sections from 3 animals in each group. [00 1 52] In another experiment, it was shown that n A antagonism suppressed angiopoeitin-l overexpression and prevented neovascularization in the tumor microenvironment in TOV-21G human ovarian cancer xenografts. Implantation of TOV-21G ovarian cancer xenografts resulted in elevated activin A in circulation in CD1 nude mice.
Activin A was measured by ELISA . Administration of activin A nist, sActRIIB suppressedTOV-ZlG tumor growth in CD1 nude mice. The G xenografts-bearing mice were treated with sActRIIB, or with PBS at day 12 post tumor implantation. [00 1 53] Blocking activin A had no direct effect on the proliferation in vitro of TOV-21G cells in cell es. Growth of TOV 21G cells was monitored “by real time in vitro micro- imaging” using the IncuCyte system. The n A inhibitors were added at the time of cell g. Staurosporin, as a positive control for inhibition of cell growth, was used to demonstrate the ility of the monitoring system. [0 0 1 5 4] Blocking activin A suppressed Ang-l overexpression and inhibited tumor neovascularization in TOV-21G n tumor xenografts. TOV-21G afts-bearing nude mice were treated with sActRIIB or PBS at day 12 post tumor implantation. The TOV-ZlG tumors were isolated at different time points after the treatment and then subjected to immunohistochemical staining for Ang-l and CD31 as a marker of neovasculature . Cell nuclei were counterstained. [00 1 5 5] Caspase-3 tion and cancer cell apoptosis in G xenograft tumors also followed blocking of activin A. TOV-ZlG tumor sections were histochemically examined for caspase-3 activation using active caspasespecif1c antibody and for cell apoptosis using TUNEL staining. sActRIIB induced active caspase-3 and cell apoptosis. [00 1 5 6] The experiments demonstrate that activin A is capable of stimulating angiogenesis factor overexpression (e. g., VEGF-A and Ang-l) by multiple cell types (i.e., cancer, endothelial, fibroblast and tic cells), whereas blocking the elevated activin A in tumors prevented overexpression of angiogenic factors and as a consequence activated caspase- 3 and cell apoptosis, leading to tumor ssion.
EXAMPLE 5: OVARIAN AND TESTICULAR MORPHOLOGY [00 1 5 7] Histological examination of ovarian and testicular tumors in lO-week-old male and l4-week-old female inhibin-(x KO mice that had been treated for two weeks with PBS or sActRIIB along with age-matched WT controls was performed. 8-week-old male and 12- week-old female inhibin-(x KO mice received a single injection of sActRIIB or PBS. 14 days after the treatment, ovarian and testicular organs were collected by necropsy. Tissue sections were subjected to Periodic Acid-Schiff Staining for ovaries and Masson’s Trichrome Staining for testes. [00 1 5 8] In the untreated female KO mice, the y enlarged ovaries were predominantly filled with solid cancer mass and large hemorrhages with hardly any recognizable les left. However, after the sActRIIB treatment, the ovaries were normal in size, and their morphology appeared relatively normal with many recognizable follicles, minimal cancer invasion and few hemorrhages. This microscopic analysis thus orates our findings on tumor weight and gross morphology and confirmed that the sActRIIB treatment caused a dramatic cancer regression in the testes and ovaries in the KO animals. In the untreated male KO mice, the normal testicular structures were no longer recognizable by light microscopy, as the seminiferous tubules were mostly ed by a e, undifferentiated cancer mass. However, after the sActRIIB treatment, the testicular ures appeared vely normal with most of the ferous tubules largely intact, although a few small areas still contained residual cancer cells, and the number of spermatogonia cells in some tubules was reduced. [0 0 1 5 9] It was also noted that sActRIIB ent rapidly reduced the testicular tumor mass in the KO mice to the WT control level. The s of testes (left and right) of individual animals were analyzed by cohorts via sy immediately before and 14 days after ent. For the experiment P<0.001 vs. WT l. n=6-12. [0 0 1 6 0] Additionally, it was noted that sActRIIB abolished VEGF and Ang-l overexpression and induced caspase-3 activation in testicular tumors in n-u KO-mice.
Angiopoietin-2 transcripts were sed toward control levels after sActRIIB treatment in both ovarian and testicular tumors.
EXAMPLE 6: EFFECT OF THE COMBINATION OF sActRHB AND A HERAPEUTIC [00 1 6 1] The effect of activin antagonist sActRIIB and cytotoxic chemotherapeutic agent -Fluorouracil on TOV-21G tumor growth inhibition in nude mice. e 9A) TOV-21G xenografts-bearing nude mice were treated at day 12 post tumor implantation with IB, 5- Fu, or combination of sActRIIB and 5-Fu, respectively. Changes in tumor volumes were recorded longitudinally. It can be observed in Figure 9A that sActRIIB by itself reduces tumor volume 34% and 5-fluorouracil (5-FU) by itself reduces tumor volume by 49%. Combining the two together, however, decrease tumor volume by 74%. [00 1 62] Figure 9B shows the effects of sActRIIB and dacarbazine on growth inhibition of G361 human melanoma aft in nude mice. BALB/c nude mice were treated with sActRIIB, dacarbazine, or sActRIIB plus dacarbazine. For dacarbazine combination study, BALB/c nude mice implanted with G361 xenograft (5x106 cells/mouse) were treated with sActRIIB only (10 mg/kg, SC, lX/wk), dacarbazine only (4 consecutive days of daily IP injection at 5 mg/kg), or sActRIIB and 5-FU combination beginning at day 18 post tumor tation. Tumor volumes were measured longitudinally by electronic caliper up to day 26 after implantation. [00 1 63] Given that the chemotherapeutic and IB presumably act by different mechanisms, it would not necessarily be expected that the demonstrated results, i.e. both agents in combination would affect overall tumor growth r than either agent individually. [00 1 64] Ovarian cancer is the deadliest of all gynecologic cancers. Activin A, a gonadal cytokine that serves important functions in ting the menstrual cycle, is expressed strongly in many malignancies including ovarian cancer. Neutralizing activin A eradicates ovarian tumors established in inhibin-def1cient mice and markedly impedes the growth of multiple activin A-secreting ovarian xenograft tumors in nude mice. Blocking activin A appears to have no direct effect on the eration in vitro of ovarian cancer cells in cell cultures, but, in viva, it completely abolishes the pression of angiogenesis factors (i.e., VEGF and angiopoietins) and inhibits neovascularization in the tumor microenvironment. It, y, induces caspase-3 activation and cancer cell apoptosis in ovarian tumors. This profound effect in viva can be explained, at least in part, by the ability of tumor-derived activin A to trigger the overproduction of angiogenesis s in the host’s elial cells, which reside in the tumor microenvironment. In addition to the ovarian tumors, activin blockade was also found to inhibit the in viva growth of at least two other cancer types (testicular tumors and melanoma) that secrete n A. Importantly, the tumor-inhibitory effect of activin antagonism was found to be, at least, additive to that of S-fiuorouracil or dacarbazine chemotherapy. These findings demonstrate a function of activin A as a critical or of tumor enesis and tumorigenesis. Blocking the elevated n A with sActRIIB, therefore, appears to be a promising new approach to combat ovarian cancer, testicular cancer, melanoma and possibly other malignancies. [00 1 6 5] The present invention is not to be limited in scope by the specific embodiments described herein, which are ed as single illustrations of individual aspects of the invention, and filnctionally equivalent methods and ents are within the scope of the invention. Indeed, various modifications of the invention, in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the appended claims.

Claims (29)

THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:
1. A pharmaceutical composition comprising (i) an isolated protein comprising a variant activin IIB receptor polypeptide (vActRIIB), wherein vActRIIB comprises a ptide sequence having at least 95% identity to the amino acid sequence set forth at amino acids 25 through 134 of the sequence set forth in SEQ ID NO:18, wherein the polypeptide comprises an amino acid substitution at position 28, and wherein the polypeptide is capable of binding myostatin, activin A, or GDF-11, and (ii) a chemotherapeutic agent.
2. The pharmaceutical ition according to claim 1, wherein the substitution at position 28 of the IB polypeptide is selected from the group consisting of A, F, Q, V, I, L, M, K, H, W and Y for E.
3. The pharmaceutical composition according to claim 1 or 2, wherein the substitution at on 28 of the vActRIIB polypeptide is selected from the group of amino acids consisting of A, W and Y for E.
4. The pharmaceutical composition according to any one of claims 1 to 3, wherein the tution at position 28 of the vActRIIB polypeptide is W.
5. The ceutical composition according to any one of claims 1 to 4, wherein the chemotherapeutic agent is a side analogue, 5-fluorouracil, and/or dacarbazine.
6. The pharmaceutical composition according to any one of claims 1 to 5, wherein the polypeptide sequence has at least 99% identity to the amino acid sequence set forth at amino acids 25 through 134 of the sequence set forth in SEQ ID NO: 18, wherein vActRIIB comprises a W amino acid substitution at position 28, wherein the isolated n further comprises an Fc domain linked to vActRIIB via a linker, wherein the sequence of the Fc domain is shown in SEQ ID NO:80, and wherein the sequence of the linker is shown in SEQ ID NO:79.
7. The pharmaceutical composition according to any one of claims 1 to 5, wherein vActRIIB comprises a polypeptide sequence having at least 97% identity to the amino acid sequence set forth at amino acids 25 through 134 of the sequence set forth in SEQ ID NO: 18.
8. The ceutical composition according to any one of claims 1 to 5, wherein vActRIIB comprises a polypeptide sequence having at least 98% identity to the amino acid sequence set forth at amino acids 25 through 134 of the ce set forth in SEQ ID NO: 18.
9. Use of an isolated protein comprising a variant activin IIB receptor ptide (vActRIIB) in the preparation of a medicament for treating cancer in a subject in combination with a chemotherapeutic agent, wherein the vActRIIB comprises a polypeptide sequence having at least 95% identity to the amino acid sequence set forth at amino acids 25 through 134 of the sequence set forth in SEQ ID NO:18, wherein the polypeptide comprises an amino acid substitution at position 28, and wherein the polypeptide is capable of binding myostatin, activin A, or GDF-11.
10. The use according to claim 9, wherein the subject is receiving treatment with the chemotherapeutic agent.
11. Use of a chemotherapeutic agent in the preparation of a medicament for treating cancer in a subject in combination with an isolated protein comprising a t activin IIB receptor polypeptide (vActRIIB), n the vActRIIB ses a polypeptide sequence having at least 95% ty to the amino acid sequence set forth at amino acids 25 through 134 of the sequence set forth in SEQ ID NO:18, wherein the polypeptide comprises an amino acid substitution at position 28, and wherein the polypeptide is capable of binding myostatin, n A, or GDF-11.
12. The use according to claim 9, wherein the subject is receiving treatment with the isolated protein.
13. The use according to any one of claims 9 to 12, wherein treatment of the cancer comprises administering to the subject the isolated protein and the herapeutic agent
14. The use according to claim 13, n the isolated protein and the chemotherapeutic agent are administered to the subject concurrently, consecutively or sequentially.
15. Use of (i) an isolated protein comprising a variant activin IIB receptor polypeptide (vActRIIB) and (ii) a chemotherapeutic agent in the preparation of a medicament for treating cancer in a subject suffering therefrom, wherein the vActRIIB comprises a polypeptide sequence having at least 95% identity to the amino acid sequence set forth at amino acids 25 through 134 of the sequence set forth in SEQ ID NO:18, wherein the polypeptide comprises an amino acid substitution at position 28, and wherein the polypeptide is capable of g myostatin, activin A, or GDF-11.
16. The use according to any one of claims 9 to 15, wherein the substitution at position 28 of the IB polypeptide is selected from the group consisting of A, F, Q, V, I, L, M, K, H, W and Y for E.
17. The use ing to any one of claims 9 to 16, wherein the substitution at position 28 of the vActRIIB polypeptide is selected from the group of amino acids consisting of A, W and Y for E.
18. The use according to any one of claims 9 to 17, wherein the tution at position 28 of the vActRIIB ptide is W.
19. The use according to any one of claims 9 to 18, wherein vActRIIB comprises a polypeptide sequence having at least 97% identity to the amino acid sequence set forth at amino acids 25 through 134 of the ce set forth in SEQ ID NO: 18.
20. The use according to any one of claims 9 to 19, wherein vActRIIB comprises a polypeptide sequence having at least 98% identity to the amino acid sequence set forth at amino acids 25 through 134 of the sequence set forth in SEQ ID NO: 18.
21. The use according to any one of claims 9 to 20, wherein vActRIIB comprises a polypeptide sequence having at least 99% identity to the amino acid sequence set forth at amino acids 25 through 134 of the sequence set forth in SEQ ID NO: 18, wherein vActRIIB comprises a W amino acid tution at position 28, wherein the isolated protein further comprises an Fc domain linked to vActRIIB via a linker, wherein the sequence of the Fc domain is shown in SEQ ID NO:80, and wherein the sequence of the linker is shown in SEQ ID NO:79.
22. The use according to any one of claims 9 to 21, wherein the herapeutic agent is a side analogue, 5-fluorouracil, and/or dacarbazine.
23. The use according to any one of claim 9 to 22, wherein the cancer is solid tumor .
24. The use according to any one of claim 9 to 22, wherein the cancer is ovarian cancer.
25. The use according to any one of claim 9 to 22, wherein the cancer is ular cancer.
26. The use according to any one of claims 9 to 25, wherein treating the cancer comprises reducing the size of a tumor mass in the subject.
27. The use according to claim 26, wherein the tumor mass is an ovarian tumor mass or a solid tumor mass
28. The pharmaceutical composition according to any one of claims 1 to 8 as described in any example .
29. The use according to any one of claims 9 to 27 as described in any example hereof.
NZ626580A 2011-12-19 2012-12-19 Variant activin receptor polypeptides, alone or in combination with chemotherapy, and uses thereof NZ626580B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US13/329,897 2011-12-19
US13/329,897 US8501678B2 (en) 2007-03-06 2011-12-19 Variant activin receptor polypeptides and uses thereof
PCT/US2012/070571 WO2013106175A1 (en) 2011-12-19 2012-12-19 Variant activin receptor polypeptides, alone or in combination with chemotherapy, and uses thereof

Publications (2)

Publication Number Publication Date
NZ626580A NZ626580A (en) 2016-07-29
NZ626580B2 true NZ626580B2 (en) 2016-11-01

Family

ID=

Similar Documents

Publication Publication Date Title
AU2017248486B2 (en) Variant activin receptor polypeptides, alone or in combination with chemotherapy, and uses thereof
US10407487B2 (en) Variant activin receptor
US8999917B2 (en) Variant activin receptor polypeptides and uses thereof
EP3101029B1 (en) Variants of activin iib receptor polypeptides and uses thereof
NZ626580B2 (en) Variant activin receptor polypeptides, alone or in combination with chemotherapy, and uses thereof
AU2016210719A1 (en) Variants of activin IIB receptor polypeptides and uses thereof