AU644389C - A mammalian cytokine, IL-11 - Google Patents

A mammalian cytokine, IL-11

Info

Publication number
AU644389C
AU644389C AU67578/90A AU6757890A AU644389C AU 644389 C AU644389 C AU 644389C AU 67578/90 A AU67578/90 A AU 67578/90A AU 6757890 A AU6757890 A AU 6757890A AU 644389 C AU644389 C AU 644389C
Authority
AU
Australia
Prior art keywords
cells
cell
sequence
expression
dna sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired
Application number
AU67578/90A
Other versions
AU6757890A (en
AU644389B2 (en
Inventor
Frances K Bennett
Stephan R. Paul
Yu-Chung Yang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Childrens Medical Center Corp
Genetics Institute LLC
Original Assignee
Childrens Medical Center Corp
Genetics Institute LLC
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 US07/526,474 external-priority patent/US5215895A/en
Application filed by Childrens Medical Center Corp, Genetics Institute LLC filed Critical Childrens Medical Center Corp
Publication of AU6757890A publication Critical patent/AU6757890A/en
Assigned to GENETICS INSTITUTE INC., CHILDREN'S MEDICAL CENTER CORPORATION reassignment GENETICS INSTITUTE INC. Amend patent request/document other than specification (104) Assignors: GENETICS INSTITUTE INC.
Application granted granted Critical
Publication of AU644389B2 publication Critical patent/AU644389B2/en
Publication of AU644389C publication Critical patent/AU644389C/en
Assigned to GENETICS INSTITUTE, LLC, CHILDREN'S MEDICAL CENTER CORPORATION reassignment GENETICS INSTITUTE, LLC Request to Amend Deed and Register Assignors: CHILDREN'S MEDICAL CENTER CORPORATION, GENETICS INSTITUTE INC.
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Description

A MAMMALIAN CYTOKINE, IL-11 The present invention relates to a novel cytokine that stimulates the function of cells of the immune and hematopoietic systems, and to processes for obtaining the factor and producing it by recombinant genetic engineering techniques. Background of the Invention
A growing family of regulatory proteins that deliver signals between cells of the immune system has been identified. These regulatory molecules are known as cytokines. Many of the cytokines have been found to control the growth and development, as well as the biological activities of cells of the hematopoietic and immune systems. These regulatory molecules include all of the colony-stimulating factors (e.g., GM-CSF, G-CSF, M-CSF, and ulti CSF or interleukin-3) , the interleukins (IL-1 through IL-9) , the interferons (alpha, beta and gamma) , the tumor necrosis factors (alpha and beta) , erythropoietin, macrophage inhibitory proteins, the tumor growth factors and leukemia inhibitory factor (LIF) . These cytokines exhibit a wide range of biological activities with target cells from bone marrow, peripheral blood, fetal liver, and other lymphoid or hematopoietic organs. See, e.g., F. R. Balkwill and F. Burke,
Immunology Today, 10.(9) :299 (1989) ; G. Wong and S. Clark, Immunology Today, 9_(5):137 (1988); and S. C. Clark and R. Ka en, Science, 236:1229-1237 (1987).
The biochemical and biological identification and characterization of certain cytokines was hampered by the small quantities of the naturally occurring factors available from natural sources, e.g., blood and urine. Many of the cytokines have recently been molecularly cloned, heterologously expressed and purified to homogeneity. Several of these purified factors have been found to demonstrate regulatory effects on the hematopoietic and immune systems in vivo, including GM- CSF, M-CSF, G-CSF, IL-1, IL-2, IL-3, IL-6, IL-7, TNF, the interferons and erythropoietin. There remains a need in the art for additional proteins purified from their natural sources or otherwise produced in homogeneous form, which are capable of stimulating or enhancing immune responsiveness and hematopoietic cell development, which are suitable for pharmaceutical use. Summary of the Invention
In one aspect the present invention provides a novel mammalian cytokine, called IL-11, which is substantially free from other mammalian proteins. This protein may be produced by recombinant genetic engineering techniques. It may also be purified from cell sources producing the factor naturally or upon induction with other factors. IL-11 may also be synthesized by chemical techniques, or a combination of the above-listed techniques.
Active, mature mammalian IL-11 is an approximately 178 a ino acid protein, characterized by an apparent molecular weight of approximately 20 kd, as determined by analyzing 35S methionine labelled supernatant fluid derived from IL-11 cDNA transfected
COS-1 cells on sodium dodecylsulfate polyacrylamide gel electrophoresis. The calculated molecular weight for the mature protein is also approximately 20 kd.
The IL-11 protein of this invention has displayed biological activities in various assays, which indicate its role as a general stimulator of a variety of hematopoietic and immune functions. The IL-11 protein of this invention demonstrates proliferative activity in an IL-6 dependent mouse plasmacytoma cell line, T1165. I - 11 has also demonstrated in preliminary assays, the ability to stimulate, either directly or indirectly, the maturation of B cells. Specifically, IL-11 is believed to stimulate the T cell dependent development of B cells. It has further demonstrated synergy with IL-3 in an assay stimulating megakaryocyte proliferation, but may act on other lineages as well. Another aspect of the present invention is a
DNA sequence that encodes the expression of a mammalian IL-11 protein. This DNA sequence may include an isolated DNA sequence that encodes the expression of a mammalian IL-11 protein as described above. The DNA sequence coding for active IL-11 is characterized as comprising the sar.e or substantially the same nucleotide sequence in Table I or fragments thereof. This DNA sequence may include 5* and 3 ' mammalian non-coding sequences flanking the IL-11 coding sequence. The DNA sequence may also encode an amino terminal signal peptide. Table I illustrates these non-coding 51 and 31 flanking sequences and a signal sequence of mammalian IL-11 isolated from the primate cell line PU34 and expressed in COS-1 cells. It is understood that the DNA sequence of this invention may, however, exclude some or all of these flanking or signal sequences. Moreover the DNA sequence of the present invention which encodes a biologically active mammal La IL-11 protein may also comprise DNA capable of hybridizing under appropriate conditions, or which would be capable of hybridizing under said conditions, but for the degeneracy of the genetic code, to an isolated DNA sequence of Table I. Thus, the DNA sequence of this invention may include or contain modifications in the non-coding sequences, signal sequences or coding sequences based on allelic variation, species variation or deliberate modification. Also provided by the present invention is a recombinant DNA molecule comprising vector DNA and a DNA sequence encoding mammalian IL-11. The DNA molecule provides the IL-11 DNA in operative association with a regulatory sequence capable of directing the replication and expression of IL-11 in a selected host cell. Host cells transformed with such DNA molecules for use in expressing recombinant IL-11 protein are also provided by the present invention.
The DNA molecules and transformed cells of the invention are employed in another aspect, a novel process for producing recombinant mammalian IL-11 protein, or peptide fragments thereof. In this process a cell line transformed with a DNA sequence encoding expression of IL-11 protein or a fragment thereof (or a recombinant DNA molecule as described above) in operative association with a suitable regulatory or expression control sequence capable of controlling expression of the protein is cultured under appropriate conditions permitting expression of the recombinant DNA. The expressed IL-11 protein is then harvested from the host cell or culture medium by suitable conventional means. This claimed process may employ a number of known cells as host cells for expression of the protein. Presently preferred cell lines for producing IL-11 are mammalian cell lines and bacterial cells.
Another aspect of this invention provides pharmaceutical compositions containing a therapeutically effective amount of mammalian IL-11 or of one or more biologically active peptide fragments thereof. These proteins or peptide fragments may be presented in a pharmaceutically acceptable vehicle. These pharmaceutical compositions may be employed, alone or in combination with other suitable pharmaceutical agents, in methods for treating disease states characterized by a deficiency in the number or level of activity of hematopoietic cells. Pharmaceutical compositions containing IL-11 may be also be employed for the treatment of disorders of the immune system, such as immunodeficiencies.
IL-11 containing compositions may be used to stimulate megakaryocyte growth and differentiation in synergy with IL-3. Additional areas of use are in platelet formation, acquired chemotherapeutic or bone marrow related thrombocytopenia. IL-11 is also likely to operate as an effector molecule to improve the function of other cytokines. IL-11 compositions may also be useful in directly or indirectly stimulating the production or function of B cells. Thus IL-11 compositions may be employed in therapies for cancer, the treatment of infections, acceleration of wound healing and in stimulating the immune system in general. IL-11 may also be used in potentiating the immune response to certain antigens, particularly vaccines. A further aspect of the invention, therefore, is a method for treating these and/or other pathological states by administering to a patient a therapeutically effective amount of IL-11 or a peptide fragment thereof in a suitable pharmaceutical carrier. These therapeutic methods may include administering simultaneously or sequentially with IL-11 or a peptide fragment thereof an effective amount of at least one other cytokine, hematopoietin, interleukin, growth factor, or antibody. Still another aspect of the present invention are antibodies directed against mammalian IL-11 or a peptide thereof. As part of this aspect, therefore, the invention claims cell lines capable of secreting such antibodies and methods for their production.
Other aspects and advantages of the present invention are described further in the following detailed description of preferred embodiments of the present invention. Brief Description of the Drawings
Fig. 1 graphically depicts the enhancement of the development of murine NP-reactive B cells by pClR6- transfected cos-1 cell conditioned medium in the murine plaque-forming assay. Fig. 2 graphically depicts the enhancement of the development of IL-3-dependent murine megakaryocyte colonies by pClR6-transfected cos-1 cell conditioned medium in the murine fibrin clot assay.
Detailed Description of the Invention
The present invention provides a biologically active mammalian cytokine, IL-11, in a form substantially free from association with other mammalian proteins and proteinaceous materials. This protein can be produced via recombinant techniques to enable large quantity production of pure, active IL-11 useful for therapeutic applications. Alternatively, this protein may be obtained as a homogeneous protein purified from a mammalian cell line secreting or expressing it. Further IL-11 or active fragments thereof may be chemically synthesized.
Mammalian IL-11 was initially isolated from a primate cell line developed by placing bone marrow cells from a healthy macaque monkey in long term culture and infecting them with the retrovirus U19-5 [Dr. Roger Cone, Tufts Medical School] . After incubation with the appropriate antibiotic, a live cell line designated PU34 was selected for its growth characteristics and induced with IL-1 alpha expressed in E. coli. Conditioned medium showed activity in a proliferation assay with IL-6 dependent mouse plasmacytoma cells in the presence of neutralizing antibody to IL-6. A cDNA library was prepared from IL-1-stimulated (2u/ml IL-1 for 24 hours) PU34 cell mRNA according to the expression cloning method previously described in, e.g., G. G. Wong et al, Science, 218.1810-815 (1985); Y. C. Yang et al, Cell. 47:3-10 (1986); and A. E. Namen et al, Nature, 333:571-573 (1988) .
The library was constructed in an expression vector which permits the expression of cDNA inserts in mammalian cells, e.g. COS-1 cells. Screening of the library was performed by transfecting COS-1 cells with 5μg of DNA prepared from pools of 200-500 cDNA clones. By assaying the supernatant fluid for activity in the T1165 assay, cDNA clones expressing IL-11 activity were identified.
An isolated clone having T1165 activity was called pPU34-TRA (also called pClR6) and was sequenced. Table I illustrates the cDNA sequence and the amino acid sequence (single letter code) of both the primate and human clones of the IL-11 polypeptide. The nucleotide sequence from position 1-721 for the primate sequence was obtained from pClR6. The remainder, from nucleotides 721-1102 was sequenced from a second primate cDNA isolated by hybridization with pClR6. A human cDNA encoding the plasmacytoma stimulatory activity of IL-11 was isolated from a cDNA library prepared from the human lung cell line, MRC5 [described by Jacobs et al, Nature, 227: 43 (1970) by direct hybridization with the insert from pPU34-TRA (pClR6) . The differences found in the human IL-11 nucleotide sequence are indicated in Table I above the primate sequence and the resulting changes in amino acid sequences are indicated below the appropriate amino acid in the primate sequence.
The primate nucleotide sequence comprises 1100 base pairs. The primate sequence contains a 5' non- coding sequence of 72 base pairs. The sequence of Table I also shows a 3- non-coding sequence of 431 bases. The human nucleotide sequence similarly contained a single long reading frame of 597 nucleotides.
Both the primate and the human sequences are characterized by a single long open reading frame predicting an unprocessed 199 amino acid polypeptide which begins at primate nucleotide position 73 in Table I. The first 21 amino acids from positions (1) Met to position (21) Ala in the predicted amino acid sequence of IL-11 from both the primate and human clones contain a stretch of hydrophobic amino acids that resembles a conventional mammalian secretory leader sequence [D. Perlman et al, J. Mol. Biol.. 167:391-409 (1983)]. The N-terminal of the mature IL-11 protein (underlined in Table I) and consists of the amino acid sequence PRO-GLY- PRO-PRO-PRO-GLY. The protein is first synthesized as a precursor of 199 amino acids which gets proteolytically cleaved between nucleotides #134-135, to yield a mature 178 amino acid polypeptide beginning with the sequence Pro-Gly at amino acid positions 22-23 and terminating after amino acid position 199 at the TGA termination triplet at nucleotide positions 671-672. The calculated molecular mass of the mature protein correspond well with the apparent molecular weight of a novel protein band revealed by SDS-PAGE (reducing conditions) of supernatant fluid derived from IL-11 cDNA transfected COS-1 cells, that is, approximately 20 kd in both cases.
TABLE I
Primate and Human IL-11 sequence, 5« - 3*
Differences between human and primate sequence are indicated by bases above primate nucleotide sequence and amino acids below primate amino acid sequence.
GGGAAGGTGG AAGGGTTAAA GGCCCCCGGC TCCCTGCCCC 40
CTGCCCTGGG GAACCCCTGG CCCTGCGGGG AC ATG AAC TGT GTT 84
Met Asn Cys Val 1 TGC CGC CTG GTC CTG GTC GTG CTG AGC CTG TGG CCA GAT 123 Cys Arg Leu Val Leu Val Val Leu Ser Leu Trp Pro Asp 5 10 15
signal sequence cleavage site C 4- C ACA GCT GTT GCC CCT GGG CCA CCA CCT GGC TCC CCT CGA 162 Thr Ala Val Ala Pro Gly Pro Pro Pro Glv Ser Pro Arg 20 25 Pro 30
N terminal mature IL-11 T
GCT TCC CCA GAC CCT CGG GCC GAG CTG GAC AGC ACC GTG 201 Ala Ser Pro Asp Pro Arg Arg Glu Leu Asp Ser Thr Val Val 35 40
C CTC CTG ACC CGC TCT CTC CTG GAG GAC ACG CGG CAG CTG 240 Leu Leu Thr Arg Ser Leu Leu Glu Asp Thr Arg Gin Leu 45 50 Ala 55
G GC G
ACT ATA CAG CTG AAG GAC AAA TTC CCA GCT GAC GGG GAC 279 Thr lie Gin Leu Lys Asp Lys Phe Pro Ala Asp Gly Asp Ala Ala 60 Arg 65
T CAC AAC CTG GAT TCC CTG CCC ACC CTG GCC ATG AGC GCG 318 His Asn Leu Asp Ser Leu Pro Thr Leu Ala Met Ser Ala 70 75 80
A G GGG GCA CTG GGA GCT CTA CAG CTC CCG AGT GTG CTG ACA 357 Gly Ala Leu Gly Ala Leu Gin Leu Pro Ser Val Leu Thr 85 90 Gly 95 AGG CTG CGA GCG GAC CTA CTG TCC TAC CTG CGG CAT GTG 396 Arg Leu Arg Ala Asp Leu Leu Ser Tyr Leu Arg His Val
100 105
C GGT
CAG TGG CTG CGT CGG GCA ATG GGC TCT TCC CTG AAG ACC 435 Gin Trp Leu Arg Arg Ala Met Gly Ser Ser Leu Lys Thr 110 Gly 120
115
C G A
CTG GAG CCT GAG CTG GGC ACC CTG CAG ACC CGG CTG GAC 474
Leu Glu Pro Glu Leu Gly Thr Leu Gin Thr Arg Leu Asp 125 130 Ala
CGG CTG CTG CGC CGG CTG CAG CTC CTG ATG TCC CGC CTG 513 Arg Leu Leu Arg Arg Leu Gin Leu Leu Met Ser Arg Leu 135 140 145
CA G GCC CTG CCC CAG CTG CCC CCA GAC CCG CCG GCG CCC CCG 552
Ala Leu Pro Gin Leu Pro Pro Asp Pro Pro Ala Pro Pro
150 Pro 155 160
G CTG GCG CCC CCC TCC TCA ACC TGG GGG GGC ATC AGG GCC 591 Leu Ala Pro Pro Ser Ser Thr Trp Gly Gly lie Arg Ala
165 Ala 170
GCC CAC GCC ATC CTG GGG GGG CTG CAC CTG ACA CTT GAC 630 Ala His Ala lie Leu Gly Gly Leu His Leu Thr Leu Asp
175 180 185
A G
TGG GCC GTG AGG GGG CTA CTG CTG CTG AAG ACT CGG CTG 669
Trp Ala Val Arg Gly Leu Leu Leu Leu Lys Thr Arg Leu 190 195 199
TGA CCCGAGGCCC AGAGCCACCA CCGTCCTTCC 702 End
AAAGCCACAT CTTATTTATT TATTTATTTC GGTACTGGGG 742 GCGAAACAGC CAGGTGATCC CCCTGCCTTT AGCTCCCCCT 782
AGTTAGAGAC AGTCCTTCCG TGAGGCTGGG GGGCATCTGT 822
GCCTTATTTA TACTTATTTA TTTCAGGAGC GGGGGTGGGC 862 TCCTGGGTCC CCGAGGAGGA GGGAGCTGGG GTCCCGGATT 902
CTTGTGTCCA CAGACTTCTG CCCTGGCTCC TCCCCCTCGA 942
GGCCTGGGCA GGAATACATA CTATTTATTT AAGCAATTAC 982
TTTTCATGTT GGGGTGGGGA GGGAGGGGAA AGGGAAGCCT 1022
GGGTTTTTGT ACAAAAATGT GAGAAACCTT TGTGAGACGG 1062
AGAACAAGGA ATTAAATGTG TCATACATAA AAAAAAAA 1100
The nucleotide sequence of IL-11 cDNA has been compared with the nucleotide sequences recorded in Genbank. No significant similarities in nucleotide sequence were found with the published DNA sequences of other proteins. Only mild homology was found between the leader sequence of IL-11 and those of gamma interferon and IL-6. No significant homology was found between the coding sequence of IL-11 and any other published polypeptide sequence. Additionally, as described in more detail in
Example 11, IL-11 is a synergistic factor for IL-3- dependent proliferation of primitive progenitors. A result of the synergism is the shortening of the G0 period of the stem cells. In at least one culture system, IL-11, like IL-6, acts synergistically with IL-3 in support of megakaryocyte colony formation [S.R. Paul et al, Proc. Natl. Acad. Sci. U.S.A.. 87:7512-7516 (1990)]. Thus, it appears that IL-11, as well as G-CSF and IL-6, interacts with early and late hempoietic lineages. However, in constrast to IL-6, which is also such a synergistic factor, IL-11 preferentially stimulates only macrophage proliferation in secondary cultures of pooled blast cells. Thus, IL-11 appears to be distinct from other known lymphokines, factors and proteins. IL-11 is also implicated in playing a role within the lymphoid lineages, resulting in stimulation of multiple arms of the defense system. Thus, IL-11 is expected to be useful in the manipulation of stem cells for both experimental and clinical purposes.
The biological activity of the mammalian IL-11 encoded by this sequence was detected in the functional polypeptides produced by mammalian cells transfected with the cloned sequence under the control of appropriate expression control sequences. The cloned primate sequence in plasmid pPU34-TRA (pClR6) as reported in Table I was deposited with the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Maryland on November 14, 1989 under ATCC No. 68172. The cloned human sequence, illustrated in Table I by the modifications from the primate sequence on both the nucleotide and amino acid levels, was deposited with the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Maryland on March 30, 1990 under ATCC No. 68284.
The IL-11 polypeptide is active in the T1165 assay, described below. In initial studies, IL-11 was found to significantly enhance the formation of immunoglobulin-secreting B cells in a standard murine spleen cell plaque formation assay, even at final dilutions as high as 1:500. This system measures the development of B cells in culture that respond to a specific immunogen, 4-hydroxy-3-rιitrophenyi-acetyi- modified horse red blood cells (NP-HRBC) in the context of the normal cellular constituents of the spleen. Thy 1 complement-mediated depletion of T cells from the spleen cell cultures resulted in complete abrogation of the response, demonstrating that the increase in NP- responding B cells, even in the presence of the primate IL-11, depends at least in part on the presence of T cells. The activity of IL-11 is therefore not attributable to a direct B cell mitogenic effect because B cell itogens, such as lipopolysaccharide, stimulate the formation of NP-specific plaque forming cells in the absence of T cells. Thus IL-11 may regulate the proliferation, differentiation and activation of T and B lymphocytes.
Analysis of the effects of the IL-11 in a variety of hematopoietic culture systems revealed striking effects on egakaryocyte development. With murine bone marrow cells as targets, IL-11 had little effect alone, but stimulated by threefold megakaryocyte colony formation supported by IL-3. CFU-Meg formation with IL-3 and IL-11 exceeded that of aplastic canine serum which is used as a positive control. The IL-11 polypeptides provided herein also include factors encoded by sequences similar to that of recombinant IL-11 in Table I, but into which modifications are naturally provided or deliberately engineered. Thus the present invention also encompasses these novel DNA sequences, free of association with DNA sequences encoding other primate proteins, and coding on expression for IL-11 polypeptides. These DNA sequences include sequences the same or substantially the same as the above-identified DNA sequence and fragments thereof, and those sequences which hybridize under stringent hybridization conditions [see, T. Maniatis et al, Molecular Cloning (A Laboratory Manual) , Cold Spring
Harbor Laboratory (1982) , pages 387 to 389] to the DNA sequence of Table I. An example of one such stringent hybridization condition is hybridization at 4XSSC at 65°C, followed by a washing in 0.1XSSC at 65*C for an hour. Alternatively an exemplary stringent hybridization condition is 50% formamide, 4XSSC at 42°C.
DNA sequences which hybridize to the sequences for IL-11 or active fragments thereof under relaxed hybridization conditions and which code on expression for IL-11 peptides having IL-11 biological properties also encode novel IL-11 polypeptides. Examples of such non- stringent hybridization conditions are 4XSSC at 50°C or hybridization with 30-40% formamide at 42°C. For example, a DNA sequence which shares regions of significant homology with the sequences of IL-11 and encodes a protein having one or more IL-11 biological properties clearly encodes a IL-11 polypeptide even if such a DNA sequence would not stringently hybridize to the IL-11 sequence of Table I or to fragments thereof encoding peptides with IL-11 activity.
Similarly, DNA sequences which code for IL-11 polypeptides but which differ in codon sequence due to the degeneracies of the genetic code are also encompassed by this invention. Allelic variations (naturally- occurring base changes in the species population which may or may not result in an amino acid change) in DNA sequences encoding the IL-11 protein sequences and peptide fragments thereof evidencing IL-11 biological activity are also included in the present invention as well as analogs or derivatives thereof. Other variations in the DNA sequence of IL-11 which are caused by point mutations or by induced modifications to enhance certain characteristics of the IL-11 protein, such as the biological activity, half-life or production of the polypeptides encoded thereby are also encompassed in the invention. In addition to the use of the cDNA sequence above in recombinant techniques, IL-11 polypeptides of this invention may also be produced by known conventional chemical synthesis. Methods for constructing the polypeptides of the present invention by synthetic means are known to those of skill in the art. The synthetically-constructed IL-11 polypeptide sequences or fragments thereof which duplicate or partially duplicate continuous sequences of the amino acid residues of Table I are also part of this invention. The synthetically- constructed IL-11 polypeptide sequences, by virtue of sharing primary, secondary, or tertiary structural and confor ational characteristics with natural IL-11 polypeptides may possess IL-11 biological properties in common therewith. Thus, they may be employed as biologically active or immunological substitutes for natural, purified IL-11 polypeptides in therapeutic and immunological processes.
Modifications in the protein, peptide or DNA sequences of IL-11 or active fragments thereof can be made by one skilled in the art using known techniques. Modifications of interest in the IL-11 sequences may include the replacement, insertion or deletion of one or more selected amino acid residues in the coding sequences. Mutagenic techniques for such replacement, insertion or deletion are well known to one skilled in the art. [See, e.g.. United States patent 4,518,584.] Other specific mutations of the sequences of the IL-11 polypeptide described herein may involve, e.g., the insertion of one or more glycosylation sites. An asparagine-1inked glycosylation recognition site can be inserted into the sequence by the deletion, substitution or addition of amino acids into the peptide sequence or nucleotides into the DNA sequence. Such changes may be made at any site of the molecule that is modified by addition of O-linked carbohydrate or at other sites in the molecule. Expression of such altered nucleotide or peptide sequences produces variants which may be glycosylated at those sites and which may have altered or improved pharmacological or biologic properties. Additional analogs and derivatives of the sequence of IL-11 which would be expected to retain IL-11 activity in whole or in part may also be easily made by one of skill in the art given the disclosures herein. One such modification may be the attachment of polyethylene glycol (PEG) onto existing lysine residues in the IL-11 sequence or the insertion of one or more lysine residues or other amino acid residues that can react with PEG or PEG derivatives into the sequence by conventional techniques to enable the attachment of PEG moieties. Such modifications are believed to be encompassed by this invention.
The present invention also provides a method for producing IL-11 polypeptides or active fragments thereof. One method of the present invention involves introducing the cDNA encoding an IL-11 polypeptide into an expression vector to make an expression system for IL- 11. A selected host cell is transformed with the vector and cultured. The method of this present invention therefore comprises culturing a suitable cell or cell line, which has been transformed with a DNA sequence coding on expression for an IL-11 polypeptide or a fragment thereof under the control of known regulatory sequences. The expressed factor is then recovered, isolated and purified from the culture medium ( or from the cell, if expressed intracellularly) by appropriate means known to one of skill in the art. Suitable cells or cell lines for this method may be mammalian cells, such as Chinese hamster ovary cells (CHO) or 3T3 cells. The selection of suitable mammalian host cells and methods for transformation, culture, amplification, screening and product production and purification are known in the art. See, e.g., Gething and Sambrook, Nature, 293:620-625 (1981), or alternatively, Kaufman et al, Mol. Cell. Biol. , 5_(7) :1750-1759 (1985) or Howley et al, U. S. Patent 4,419,446. Other suitable mammalian cell lines are the monkey COS-l cell line and the CV-1 cell line. Further exemplary mammalian host cells include particularly primate cell lines and rodent cell lines, including transformed cell lines. Normal diploid cells, cell strains derived from in vitro culture of primary tissue, as well as primary explants, are also suitable. Candidate cells may be genotypically deficient in the selection gene, or may contain a dominantly acting selection gene. Other suitable mammalian cell lines include but are not limited to, HeLa, mouse L-929 cells, 3T3 lines derived from Swiss, Balb-c or NIH mice, BHK or HaK hamster cell lines.
Similarly useful as host cells suitable for the present invention are bacterial cells. For example, the various strains of IL. coli (e.g., HB101, and MC1061) are well-known as host cells in the field of biotechnology. Various strains of B^ subtilis. Pseudomonas. other bacilli and the like may also be employed in this method.
Many strains of yeast cells known to those skilled in the art are also available as host cells for expression of the polypeptides of the present invention. Additionally, where desired, insect cells may be utilized as host cells in the method of the present invention. See, e.g. Miller et al, Genetic Engineering, 8.:277-298 (Plenum Press 1986) and references cited therein.
The present invention also provides recombinant DNA molecules, or vectors, for use in the method of expression of novel IL-11 polypeptides. These vectors contain the novel isolated DNA sequences which code for IL-11 polypeptides of the invention. Alternatively, vectors incorporating modified sequences as described above are also embodiments of the present invention and useful in the production of IL-11 polypeptides. The vector employed in the method also contains selected regulatory sequences in operative association with the DNA coding sequences of the invention and capable of directing the replication and expression thereof in selected host cells.
The vector used in the examples below is pXM [Y. C. Yang et al, Cell, 47:3-10 (1986)]. The mammalian cell expression vectors described herein may be synthesized by techniques well known to those skilled in this art. The components of the vectors, e.g. replicons, selection genes, enhancers, promoters, and the like, may be obtained from natural sources or synthesized by known procedures. See, Kaufman et al, J. Mol. Biol.. 159:511- 521 (1982) ; and Kaufman, Proc. Natl. Acad. Sci.. USA, 2.:689-693 (1985). Alternatively, the vector DNA may include all or part of the bovine papilloma virus genome [Lusky et al. Cell, 3_6:391-401 (1984)] and be carried in cell lines such as C127 mouse cells as a stable episomal element. The transformation of these vectors into appropriate host cells can result in expression of the IL-11 polypeptides.
Other appropriate expression vectors of whien numerous types are known in the art for mammalian, insect, yeast, fungal and bacterial expression can also be used for this purpose. IL-11, purified to homogeneity from cell sources or produced recombinantly or synthetically, may be used in a pharmaceutical preparation or formulation to treat immune deficiencies or disorders. IL-11 may also be employed to treat deficiencies in hematopoietic progenitor or stem cells, or disorders relating thereto. IL-11 compositions may be employed in methods for treating cancer and other pathological states resulting from disease, exposure to radiation or drugs, and including for example, leukopenia, bacterial and viral infections, anemia, B cell or T cell deficiencies, including immune cell or hematopoietic cell deficiency following a bone marrow transplantation. IL-11 may also be used to potentiate the immune response to a variety of vaccines creating longer lasting and more effective immunity. As mentioned previously, IL-11 compositions may be employed to stimulate development of B cells, and megakaryocytes. Therapeutic treatment of such disease states with these IL-11 polypeptide compositions may avoid undesirable side effects caused by treatment with presently available drugs.
The polypeptides of the present invention may also be employed, alone or in combination with other cytokines, hematopoietins, interleukins, growth factors or antibodies in the treatment of the above-identified conditions.
The present invention also provides methods and therapeutic compositions for treating the conditions referred to above. Such compositions comprise a therapeutically effective amount of an IL-11 polypeptide of the present invention in admixture with a pharmaceutically acceptable carrier. This composition can be systematically administered parenterally.
Alternatively, the composition may be administered intravenously. If desirable, the composition may be administered subcutaneously or topically, e.g., at the site of a wound. When systematically administered, the therapeutic composition for use in this invention is in the form of a pyrogen-free, parenterally acceptable aqueous solution. The preparation of such a pharmaceutically acceptable protein solution, having due regard to pH, isotonicity, stability and the like, is within the skill of the art.
The dosage regimen involved in a method for treating the above-described conditions will be determined by the attending physician considering various factors which modify the action of drugs, e.g. the condition, body weight, sex and diet of the patient, the severity of any infection, time of administration and other clinical factors. Generally, the daily regimen should be in the range of 1-1000 micrograms of polypeptide or 50 to 5000 units (i.e., a unit being the concentration of polypeptide which leads to half maximal stimulation in the T1165 assay) of polypeptide per kilogram of body weight.
The therapeutic method and compositions of the present invention may also include co-administration with other human factors. Exemplary cytokines or hematopoietins for such use include the known factors IL- 1 through IL-9, GM-CSF, G-CSF, M-CSF, MIF, Meg-CSF, the interferons, TNF and erythropoietin. Particularly desirable candidates for participation in IL-11 therapy may include IL-3 and IL-6. Growth factors like B cell growth factor, B cell differentiation factor, or eosinophil differentiation factors may also prove useful in co-administration with IL-11. The dosage recited above would be adjusted to compensate for such additional components in the therapeutic composition. Progress of the treated patient can be monitored by conventional methods.
Other uses for these novel polypeptides are in the development of antibodies generated by standard methods for jln vivo or in vitro diagnostic or therapeutic use. Such antibodies may include both monoclonal and polyclonal antibodies, as well as chimeric antibodies or "recombinant1* antibodies generated by known techniques. Also provided by this invention are the cell lines generated by presenting IL-11 or a fragment thereof as an antigen to a selected mammal, followed by fusing cells of the animal with certain cancer cells to create immortalized cell lines by known techniques. The methods employed to generate such cell lines and antibodies directed against all or portions of a mammalian IL-11 polypeptide of the present invention are also encompassed by this invention.
The antibodies of the present invention may be utilized for .in vivo and in vitro diagnostic purposes, such as by associating the antibodies with detectable labels or label systems. Alternatively these antibodies may be employed for .in vivo and in vitro therapeutic purposes, such as by association with certain toxic or therapeutic compounds or moieties known to those of skill in this art.
The following examples illustratively describe the cloning, expression and production of mammalian IL-11 and other methods and products of the present invention. These examples are for illustration and do not limit the scope of the present invention.
Example 1 - Isolation of mRNA and Construction of cDNA Library
A primate cell line, pU34, was developed and was found to elaborate significant activity in the T1165 assay of Example 7 in the presence of neutralizing antibody to IL-6. The PU-34 stromal cell line was derived from a long term primate marrow culture by immortalization with a defective amphotropic transforming retroviral vector. The U19 retrovirus plasmid was constructed as previously reported [P. S. Jat et al, J\_ of Virol. , .59:746-750 (1986)] and contains SV40 large T antigen sequence and the neo-phosphotransferase sequence encoding G418-resistance expressed off the Moloney murine leukemia virus long terminal repeat. An amphotropic producer clone was generated by infection of the packaging cell line ψAM [R. Cone et al, Proc. Natl. Acad. Sci. , USA. £1:6349-6353 (1984)] with ecotropic viral harvest from ψ2U19-5 [P. S. Jat, cited above] followed by selection in 0.75 g/ l G418.
One clone ψAMU19-BL produces recombinant SV40 virus at a titer of 5X103 G418-resistant CFU/ml when assayed on NIH/3T3 cells. Long term marrow cultures (LTMC) were established using standard methods and maintained in Iscove's Modified Dulbecco's Medium (IMDM) supplemented with 10% fetal calf serum, 10% horse serum, 100 units/ml penicillin and 100 μg/ l streptomycin (Sigma Chemical Co., St. Louis, MO) complete long term culture medium.
The LTMC adherent layer was infected 7 and 10 days after establishment with 2 ml of ψAMU19-BL viral stock in the presence of 8 μg/ml of polybrene (Aldrich Chemical Co., Inc., Milwaukee, Wl) for 2.5 hours at 33°C. Beginning three days after infection, the cultures were selected in 0.5 mg/ l G418. Fourteen days after infection G418-resistant colonies were picked and expanded in multiwell plates (Corning Glassware, Corning, NY) .
The conditioned medium from one cell line, designated PU-34, was extensively analyzed based on its ability to support progenitor cells in long term cultures. This cell line demonstrated the capacity to maintain multipotent human and primate progenitor cells for up to three weeks in culture. In addition to known growth factor activities including IL-6, IL-7, GM-CSF, M- CSF, G-CSF and LIF/HILDA, the ILl-α-stimulated PU-34 conditioned medium proved capable of stimulating the proliferation of the T1165 murine plasmacytoma cell line, which is normally responsive to IL-6 [R. P. Nordan et al, cited above] , even in the presence of a neutralizing antiserum against human IL-6. This bioassay was used during expression cloning of a cDNA library generated from PU-34. The bioassay is described in detail in Example 7 below. The cDNA library from PU-34 cells was prepared as follows: PU-34 cells were stimulated for 24 hours with ILl-α at a concentration of 2 units/ml. Poly adenylated RNA (poly A+ RNA) was prepared from these cells by standard methods. Total RNA was extracted according to the method of Chirgwin et al, Biochemistry, 18_:5294-5299 (1979) from the stimulated pU34 cells. mRNA was prepared by oligo(dT)-cellulose chromatography [H. Aviv et al, Proc. Natl. Acad. Sci. USA. 69:1408-1412 (1972) ] . Five micrograms of mRNA was used to synthesize double-stranded cDNA as described by Wong et al, cited above, with DNA polymerase I and RNAse H in the second strand reaction [T. Maniatis et al, cited above] . The Cos-1 cell expression vector pXM [Y. C. Yang et al, Cell 47: 3-10 (1986) ] was linearized at the unique Xho I site and ligated to equimolar amounts of semi-Xho I adapted cDNA. The ligation reaction was used to transform competent E^. coli (strain HB101) [Y. C. Yang et al, cited above] to generate a library of approximately 500,000 ampicillin-resistant colonies.
Example 2 - DNA Preparation and COS-1 Cell Transfection The expression cloning system previously described by G. G. Wong et al, cited above, was employed to isolate a cDNA encoding the IL-11 activity as follows. Bacterial colonies were replicated onto nitrocellulose filters. Colonies from each filter were scraped into L-broth and plas id DNA was isolated by previously described methods [J. A. Meyers et al, J. Bacteriol.. 127:1529-1536 (1976)]. Each primary DNA sample was prepared from a pool of 200-500 colonies. Five micrograms of each plasmid DNA was used to transfect Cos-1 cells by the diethylaminoethyl-dextran (DEAE) protocol with the addition of 0.1 mM chloroquine [L. M. Sompayrac et al, Proc. Natl. Acad. Sci. USA. 28:7575-7578 (1981) and H. Luthman et al, Nucl. Acids Res.. 11:1295-1308 (1983)]; Y. C. Yang et al, cited above] . Culture supernatant from transfected Cos-l cells was harvested 72 hours after transfection and assayed for T1165 stimulatory activity (see Example 7) .
Of the 317 pools screened, plasmid DNA from the two positive pools which contained detectable levels of IL-6 (as determined by neutralization with anti-IL-6 antibody) and residual activity in the T1165 assay in the presence of anti-IL-6 antibody, were re-transfected into COS-1 cells and transfected supernatants were re-screened for activity in the T1165 assay. One pool with such activity was selected and subdivided to contain fewer number of clones. A pool from this group was selected which demonstrated higher activity in the assay than the total collection of pools. Individul colonies were picked from this pool. Their DNAs were prepared, transfected, and the transfected supernatants were examined for activity in the T1165 assay. Two positive clones were identified, one expressing IL-6 activity and the other expressing activity unneutralized by anti-IL-6 antibodies. This latter pool was subdivided and the transfection process repeated until a single positive plasmid, called alternatively pClR6 or pPU34-TRA, was obtained which encoded the novel T1165 proliferation activity. This clone was re-examined in the assay of Example 7.
The activity from the conditioned medium from pClR6-transfected Cos-1 cells was also compared with other cytokines, e.g., murine and human IL-6 and murine GM-CSF. The conditioned medium stimulated measurable incorporation of 3H-thymidine by T1165 cells, even at final dilutions up to 1:1000. At optimal concentrations the novel cytokine supported incorporation that was more than 100 fold above background levels.
The insert of this cDNA was sequenced by the dideoxy chain termination method on super-coiled templates with synthetic oligonucleotide primers [F. Sanger et al, Proc. Natl. Acad. Sci. , USA, 74:5463-5467
(1977)]. The nucleotide sequence of the pClR6 cDNA shown in Table I contains a single long open reading frame of 597 nucleotides encoding a predicted 199 amino acid polypeptide. Located immediately adjacent to the putative initiation codon is a stretch of 17-20 hydrophobic amino acids that resembles a conventional protein secretory leader sequence.
Although the initial cDNA clone, pClR6, proved to be incomplete, analysis of additional cDNAs revealed that this transcript contains approximately 420 base pairs of 3 • noncoding sequence with multiple copies of the RNA instability sequence, ATTTA, believed to be an important regulatory element for cytokine gene expression [G. Shaw et al, Cell. 46:659-667 (1986)].
Example 3 - Protein Analysis
The polypeptide encoded by the cDNA of pPU34- TRA was identified using pulse-labeling experiments. Forty-eight hours after induction with chloroquine, culture supernatant from COS-1 cells transfected with recombinant DNA of IL-11 clones was removed and cells were pulse-labeled with 0.5 mCi [35S] methionine in 1.0 ml of DMEM for four hours at 37°C. Ten microliter samples of the radiolabelled supernatant were collected and subjected to a 15% SDS-PAGE with the Laemmli buffer system on a 12% gel [U. K. Laemmli, Natur , 227:680-685 (1970)]. After electrophoresis, the gel was immersed in a fluorography enhancing solution (Enhance; New England Nuclear, Boston, MA) , dried, and exposed to X-ray film. SDS-polyacrylamide gel electrophoresis (SDS- PAGE) analysis of conditioned medium from 35S-methionine labelled pClR6-transfected cos-1 cells revealed the presence of a prominent 20 kD species that was not present in mock-transfected controls consistent with the molecular mass expected for an approximately 180 amino acid secreted protein.
This size estimate as well as the lack of heterogeneity of the expressed protein are in accordance with the absence of the consensus sequence (Asn-X- Thr/Ser) [R. J. Winzler in "Hormonal Proteins and Peptides", ed. Li, C. H. (Academic Press, New York), pp. 1 (1973) ] for the addition of asparagine-linked carbohydrate. The predicted amino acid sequence of the mature protein includes no cysteine residues, a feature not found with any other cytokine gene.
Example 4 - Human Cell Lines Expressing IL-11
Two human cell lines have been identified as sources of at least one species of IL-11. Specifically, the human lung fibroblast cell line, MRC-5, available from the American Type Culture Collection under Accession number ATCC CCL 171, when induced with one unit/ml of recombinant human IL-1-alpha (Genetics Institute, Inc.) and 10"7M phorbol 12-13 dibutyrate (Sigma) , has been tested on the T1165 assay. The induced conditioned medium was observed to exhibit greater cpm on the assay than does IL-6 at saturation levels, i.e., similar activity to that exhibited by the induced conditioned medium of PU34. It has been noted that the presence of IL-11 will enhance a low IL-6 signal. In addition, as detailed below, Northern blot of this cell line reveals the presence of message for IL-11.
Additionally the human trophoblastic cell line, TPA30-1, available from the ATCC under Accession Number CRL 1583 also reveals uninduced the presence of IL-11 message in Northern blots.
Other human sources for IL-11 may also be available and easily identified given the teachings of the present invention.
Example 5 - RNA Analysis A. PU34
Five micrograms of total cellular RNA from bacterial IL-1-alpha induced PU34 cells was electrophoresed through 1.2% agarose gel containing 2.2M formaldehyde [H. Lehrach et al. Biochemistry, 16:4743 (1977)]. The formaldehyde-denatured RNA was transferred to nylon filter (Zetabind; Cuno, Meriden, CT) as described [E. M. Southern, J. Mol. Biol. r 98:503-517 (1975)] and probed with 32P-labelled cDNA probes.
A cDNA probe was made by cleaving cDNA inserts from the vector with Xho I restriction enzyme and labelled the inserts with 32P-dCTP using random oligonucleotides as primers in the presence of the large fragment of DNA polymerase I [A. P. Feinberg et al, Analy. Biochemistry. 132:6-13 (1983)]. The nylon filter was prehybridized for 4 hours at 65°C, hybridized with 32P-dCTP labelled cDNA probe in hybridization solution consisted of 4XSSC, 0.5% SDS 5X Denhardt's solution and 100 ug/ l denatured salmon sperm DNA for 16 hours at 65°C. Other probes used included human (rh) IL-lα, rhlL- 2, rhIL-3, rhIL-4 , rhIL-5, rhIL-6, rhIL-7, rhIL-9, rhGM- CSF, rhM-CSF, LIF/HILDA and primate IL-11.
After hybridization, the filter was washed two times with 2XSSC/0.1% SDS for 30 minutes at 65°C, then with 0.2XSSC/0.1% SDS for 30 minutes at 65°C. The filter was then dried and applied to X-ray film in the presence of a calcium tungstate intensifying screen at -70°C.
This Northern blot analysis revealed that PU34 mRNA contained two species of IL-11 transcripts, with message sizes of approximately 2.5 kb and approximately 1.5 kb which hybridize with the pCIR6 probe. The size of the cDNA sequence of Table I above correlates well with the smaller message. This difference results from alternative splicing to yield additional 3! noncoding sequences in the larger transcript as demonstrated by isolation and analysis of additional cDNA clones. The presence of the two transcripts by PU34 cells appears to be IL-lα regulated since neither transcript was evident in the absence of IL-lα induction. Neither transcript was identified by RNA blot analysis in preparations of mRNA from the human T cell lines C10-MJ2 [Leary et al, Blood, 69.:953 (1987)], C5-MJ2 [Arya et al, Science, 223:1086 (1984)], and Mo [Golde et al, Proc. Natl. Acad. Sci.. USA. 77:593 (1980)] from lectin-stimulated human peripheral blood lymphocytes or from human placental. Thus, it appears the only identified source of IL-11 is mesenchymal-derived adherent cells. B. MRC-5
The human fetal lung fibroblast cell line (MRC- 5) as described by Jacobs et al, Nature, 227:43 (1970) was found to express both transcripts following stimulation with 50 ng/ml phorbal myristate acetate (PMA) and 1 unit/ml IL-lα.
As described above for PU34 RNA, two species of transcripts were identified with identical message sizes of approximately 2.5 kb and approximately 1.5 kb in this cell line. Analysis of the human cDNA sequence isolated from the MRC-5 cell line revealed that the primate and human coding regions share approximately 95% identity at the nucleotide level. C. TPA30-1
When the same procedures were performed on the human SV40-transformed trophoblastic cell line, TPA30-1, using the same probe, only the larger approximately 2.3 kb IL-11 message, was identified.
Example 6 - DNA Seguence Analysis
The nucleotide sequence of the cDNA clone of pPU34-TRA was determined as described [G. G. Wong et al and Y. C. Yang et al, cited above] by generating ordered sets of overlapping fragments via Bal 31 nuclease digestion and subcloning into M13 vector [M. Poncz et al, Proc. Natl. Acad. Sci. USA. 29:4298-4302 (1982); and J. Messing et al, Gene, l :269-276 (1982)]. Single-stranded DNA was prepared, and the nucleotide sequence was determined by the dideoxynucleotide chain-termination procedure [F. Sanger et al, Proc. Natl. Acad. Sci. USA, 24:5463-5467 (1977)]. This nucleotide sequence appears in Table I above.
Example 7. Biological Activity in Assays. A. T1165 Proliferation Assay
T1165 IL-6 dependent murine plasmacytoma cells R. P. Nordan et al, Science, 233:566 '(1986) ; and obtained from Dr. Nordan, National Institutes of Health] are routinely grown in RPMI supplemented with 10% heat- inactivated fetal calf serum, 2 mM glutamine, 100 u/ml penicillin, 100 μg/ml streptomycin (all Gibco, Grand Island, NY) , 5X10"5 M beta ercaptoethanol (Sigma Chemical Co., St. Louis, MO), and supplemented with 10-20 U/ml recombinant human IL-6 produced in CHO cells (Genetics Institute, Inc.). Two to four days following passage, the cells are removed from culture, washed to remove residual IL-6 and resuspended at a concentration of 7.5 X 104 to 1 X 105 cells/ml.
Serial dilutions of the sample to be assayed (either PU34 conditioned medium or pClR6-transfected Cos cell conditioned medium) are made in duplicate in 100 μl of culture medium without IL-6 on 96-well microtiter plates. lOOμl of the above cell suspension is then added to each well and the plates are incubated at 37°C for 2-3 days; 0.5μCi of 3H-thymidine [DuPont, Wilmington, DE] is added per well for the final six hours of the assay. Cells are harvested onto GFC type C filter paper (LKB) , washed with water and ethanol and dried. The filters are then immersed in scintillation fluid and counted on an LKB flatbed scintillation counter. Proliferation is measured by 3H-thymidine uptake.
Induced conditioned medium from the PU34 cells caused greater proliferation of the T1165 cells than saturating levels of IL-6, suggesting the presence of another factor. When assayed in the presence of antibody to human IL-6, a low but significant activity remained in the conditioned medium. Fractionated samples of conditioned medium from IL-1-induced PU34 containing very low levels of IL-6 were also assayed with and without antibody to human IL-6 and the results suggested the presence of a factor that was proliferative to a low degree by itself and capable of synergizing with low levels of IL-6.
COS cell supernatants from transfection of the pU34 library were also assayed for activity, alone and in the presence of a cocktail of antibody to human IL-6 plus suboptimal levels of murine IL-6. The antibody is capable of neutralizing primate IL-6 produced by the PU34 cells, but not able to neutralize murine IL-6. Therefore, a synergizing factor could be screened for without interference from the PU34 IL-6 present in the library.
The mature IL-11 protein of Table I is characterized by a half-maximal activity of 100 dilution units per ml in this assay.
B. B Cell Plague Forming Assay A B Cell plaque forming assay was performed on
COS cell expressed 11-10 according to the procedures described in R. M. O'Hara et al, J. Immunol. , 141:2935- 2942 (1988) . The murine plaque forming assay was performed by incubating 7.5 X106 spleen cells from naive C57B1/6 mice with 3X106 4-hydroxy-3-nitrophenyl-acetyl- modified horse red blood cells (NP-HRBC) in 0.75 ml Mishell-Dutton media [R. I. Mishell et al, J. Exp. Med. , 126:423-442 (1967)] supplemented with 5% fetal calf serum with or without test samples (COS cell conditioned media containing IL-11) for 5 days. NP-coupled horse red blood cells (H-RBC) or sheep red blood cells (S-RBC) were prepared by reaction of 10 mg NP-O-Succinimide (Cambridge Biochemical, Inc., Cambridge, England) in dimethyl formamide (Sigma Chemical Co., St. Louis, Missouri) with 1 ml packed H-RBC or S-RBC (Colorado Serum Co., Denver, CO) as has been described previously [P. B. Hausman et al, J. of Immunol.. 134:1388-1396 (1985)].
These cultures were fed daily by addition of 0.1 ml supplemental medium containing 5% fetal calf serum without test samples (the conditioned media) . NP- responsive B-cells were identified at the end of the culture period using the NP-coupled-sheep RBC plaque assay as described by Dresser et al in "Handbook in Experimental Immunology" (D. M. Weir, Blackwell, Oxford) , p. 271 (1973) with the percent response calculated by comparing the numbers of plaques obtained from cultures supported with the conditioned medium containing IL-11 with those cultures supplemented with medium alone. In a typical experiment, background responses in the absence of exogenous factors yielded 6000 NP-specific plaque forming cells per 7.5 X 106 cells plated. The results of such as assay can be seen in
Fig. 1. The percent of control response is the increase in the development of NP-responsive B cells in 5 day cultures of naive spleen cells stimulated with NP-HRBC supported by the indicated dilution of pClR6-transfected cos-1 cell conditioned medium compared to control cultures supplemented with medium alone. COS-produced mammalian IL-11 produces a 2 and one-half to 3-fold increase in plaque forming units/culture in this assay, indicating the IL-11 plays either a direct role in B cell stimulation and differentiation, or an indirect role in stimulating T cells to secrete other cytokines which influence the B cell response.
C. Murine Fibrin Clot Assay COS cell produced mammalian IL-11 was also examined for activity in the megakaryocyte colony formation assay performed substantially as described in S. Kuriya et al, Ex . Hematol.. JL5_:896-901 (1987) and modified by the addition of 2% calf serum. Briefly described, the murine colony forming unit megakaryocyte (CFU-Meg) assay was performed by plating 2.5 X 105 murine bone marrow cells in 0.4 ml of IMDM supplemented with 20% fetal calf serum in 6 well dishes. Clot formation was initiated by addition of 0.25 mg fibrinogen and 0.25 units thrombin (Sigma Chemical Co., St. Louis, MO) at 37°C. Test samples at various dilutions were added to the fibrin clot and cultures subsequently incubated for 6 days at 37°C. The clots were fixed with 2.5% glutaraldehyde and stained with 0.5 mg/ml acetylthiocholine iodide as described in S. Kuriya et al, cited above and A. Nakeff et al, Proc. Soc. Exp. Biol. Med. , 151:587-590 (1976). Positive colonies (containing only megakaryocytes) were enumerated under direct microscopy. Colony numbers were evaluated in duplicate. Fig. 2 illustrates the results. The colony number represents the total number of megakaryocyte colonies (acetylcholinesterase positive cells) in 6 day cultures of mouse bone marrow cells supported by: (1) a 1:10 dilution of canine aplastic anemia serum; (2) 150 units/ml murine IL-3; (3) no stimulus; and dilutions of (4) 1:10 or (5) 1:50 pClR6-transfected-cos-l cell conditioned medium alone or dilutions of (6) 1:10 or (7) 1:50 of pClR6-transfected-cos-l cell conditioned medium supplemented with 150 units/ml murine IL-3. When IL-11 was tested in this assay alone, little response was detected. However, when IL-11 was tested in this assay in the presence of recombinant murine IL-3, the assay results demonstrated that the combination of IL-11 and IL-3 stimulated the production and maturation of megakaryocyte cells in this assay to a significant degree. This assay demonstrated that mammalian IL-11 has a synergistic effect with IL-3 in the stimulation of megakaryocyte development.
Example 8 - Obtaining Human IL-11 To obtain the cloned sequence for human IL-11, the PU34 IL-11 cDNA which hybridized to the human IL-11 mRNA in Example 5 above, was employed to screen a cDNA library prepared from the human lung fibroblast cell line, MRC-5, described above. Recombinants from this library were plated and duplicate nitrocellulose replicase made of the plates. These replicase were hybridized overnight at 65°C in standard hybridization solution (4XSSC) with the mammalian IL-11 cDNA labelled with 32P-dCTP using the random priming labelling technique [A. P. Feinberg, cited above] . The filters were then washed in 0.2XSSC at the same temperature until the background radioactivity was lowered to an acceptable level to permit detection of specifically hybridizing sequences. Colonies found to hybridize to the mammalian IL-11 probe on the duplicate filters were picked and used to prepare plasmid DNA.
The full sequence for human IL-11 was determined according to methods analogous to those described above for the isolation of mammalian IL-ll from the PU34 cell line. The human sequence is shown also in Table I. Where the human sequence nucleotides differed from the primate sequence, the human nucleotide is provided above the primate nucleotide sequence in Table I.
Alternatively, oligonucleotides may be constructed from the sequence of Table I with appropriate restriction sites for subcloning purposes, and the Polymerase Chain Reaction employed to generate the human DNA sequence for IL-11. For example, the following oligonucleotides are synthesized:
5' oligonucleotide: 51 ATGGATCCACATGAACTGTGTTTGCCG 3' 3' oligonucleotide: 5' TCAAGCTTTCACAGCCGAGTCTTCAGC 3'. These oligonucleotides are then employed in the
Polymerase Chain Reaction in the cDNA library of MRC-5 or TPA30-1, to obtain the DNA sequence for human IL-ll therefrom. The PCR technique is performed according to procedures now standard in the art. The PCR product obtained is then subcloned into an appropriately-digested pXM, or other, expression vector. For the above oligonucleotides, the pXM vector would be digested with BamHI and Hindlll for the subcloning.
Still a third method to obtain the sequence of human IL-11 involves screening a human genomic library using the sequence of Table I as a probe. Example 9 - Expression of Recombinant IL-11
To produce recombinant mammalian IL-11 including the human factor, the cDNA encoding it is transferred into an appropriate expression vector of which numerous types are known in the art for mammalian, insect, yeast, fungal and bacterial expression by standard molecular biology techniques. See, e.g., Y. C. Yang εt al., Cell, 42:3-10 (1986). As described previously for mammalian IL-11, the cDNA for human IL-11 is synthesized using standard techniques and cloned into the expression vector, pXM (Yang et al., cited above). This vector permits the expression of cDNA inserts in mammalian cells, e.g., COS- 1 cells. pXM contains the SV40 enhancer, major adenovirus late promoter, tripartite leader sequence, and small hybrid intervening sequence, the DHFR coding sequence, SV40 late message poly A addition site and adenovirus Val gene. This vector may be linearized with the endonuclease enzyme Xhol and ligated to equimolar amounts of IL-11 cDNA which has been previously modified by the addition of synthetic oligonucleotides that generate complementary Xhol cohesive ends. Such oligonucleotides are commercially available [Colla orative Research, Lexington, MA].
Another vector which has been shown to express cytokines well in CHO cells is pEMC2Bl. This vector may be derived from pMT2pc which has been deposited with the American Type Culture Collection (ATCC) , Rockville, MD (USA) under Accession Number ATCC 40348. The DNA is linearized by digestion of the plasmid with Pstl. The DNA is then blunted using T4 DNA polymerase. An oligonucleotide 5' TGCAGGCGAGCCTGAATTCCTCGA 3 ' is then ligated into the DNA, recreating the Pstl site at the 5- end and adding an EcoRI site and Xhol site before the ATG of the DHFR cDNA. This plasmid is called pMT21. pMT21 is cut with EcoRI and Xhol which cleaves the plasmid at two adjacent cloning sites. An EMCV fragment of 508 base pairs was cut from pMT2ECAT1 [S.K. Jong et al., J. Virol. , «.:1651-1660 (1989)] with the restriction enzymes EcoRI and Taqαl. A pair of oligonucleotides 68 nucleotides in length were synthesized to duplicate the EMCV sequence up to the ATG. The ATG was changed to an ATT, and a C is added, creating a Xhol site at the 31 end. A Taqαl site is situated at the 5' end. The sequences of the oligonucleotides were:
5' CGAGGTTAAAAAACGTCTAGGCCCCCCGAACCACGGGGACGTGGTTTTCCTTT GAAAAACACGATTGC 3' and its complementary strand. Ligation of the pMT21 EcoRI-to-XhoI fragment to the EMCV EcoRI-to-Taqαl fragment and to the Taqαl/Xhol oligonucleotides produced the vector pEMC2Bl. This vector contains the SV40 origin of replication and enhancer, the adenovirus major late promoter, a cDNA copy of the majority of the adenovirus tripartite leader sequence, a small hybrid intervening sequence, an SV40 polyadenylation signal and the adenovirus VA I gene, DHFR and ?-lactamase markers and an EMC sequence, in appropriate relationships to direct the high level expression of the desired cDNA in mammalian cells. The EMC2B1 vector is linearized with the endonuclease enzyme EcoRI and subsequently ligated in equimolar amount separately to the cDNA encoding IL-11 that was previously modified by addition of synthetic oligonucleotides that generate EcoRI complementary ends to generate constructs for expression.
The desired vector containing IL-11 is then introduced into appropriate host cells by conventional genetic engineering techniques. The transformed cells are cultured and the expressed IL-11 is recovered and purified from the culture medium using standard techniques.
A. Mammalian Cell Expression
To obtain expression of the IL-11 polypeptide in mammalian host cells, the pXM vector containing the IL-11 DNA sequence is transfected onto COS cells as described in Example 2. The conditioned medium from the transfected COS cells contains IL-11 biological activity as measured in the T1165 assay. Similarly the pEMC-2Bl construct containing the cDNA for IL-11 is transfected into CHO cells. One skilled in the art can also construct other mammalian expression vectors comparable to the pXM/IL-11 vector by, e.g., inserting the DNA sequence of IL-11 from the respective plasmids with Xhol and employing well- known recombinant genetic engineering techniques and other known vectors, such as pJL3 and pJL4 [Gough et al., EMBO J. , 4.:645-653 (1985)] and pMT2 (starting with pMT2- VWF, ATCC #67122; see PCT application PCT/US87/00033) .
The transformation of these vectors into appropriate host cells can result in expression of the IL-11 polypeptides. Mammalian host cells other than COS cells may also be employed in IL-11 expression. For example, preferably for stable integration of the vector DNA, and for subsequent amplification of the integrated vector DNA, both by conventional methods, CHO cells may be employed as a mammalian host cell of choice.
Once the vectors and host cells are selected and transformed, stable transformants are screened for expression of the IL-11 by standard immunological or enzymatic assays. The presence of the DNA or mRNA encoding the IL-11 polypeptides may be detected by standard procedures such as Southern or Northern blotting. Transient expression of the DNA encoding the polypeptides during the several days after introduction of the expression vector DNA into suitable host cells is measured without selection by activity or immunologic assay, e.g., the T1165 assay, of the proteins in the culture medium.
B. Bacterial Expression Systems Similarly, one skilled in the art could manipulate the sequence of IL-11 by eliminating any mammalian regulatory sequences flanking the coding sequences and inserting bacterial sequences to create bacterial vectors for intracellular or extracellular expression of the IL-11 polypeptides of the invention by bacterial cells.
The DNA encoding the factor may be further modified to contain different codons for bacterial expression as is known in the art. Preferably the mature IL-11 sequence (the nucleotide encoding amino acids 21 to 199 in Table I) is operatively linked inframe to a nucleotide sequence encoding a secretory leader polypeptide permitting bacterial expression, secretion and processing of the mature variant protein, also as is known in the art. The compounds expressed in bacterial host cells may then be recovered, purified, and/or characterized with respect to physiochemical, biochemical and/or clinical parameters, all by known methods. Alternatively, the IL-11 may be expressed as a cytoplasmic protein in E. coli. In this case, the molecule would most likely have to be refolded after complete denaturation with guanidine hydrochloride, a process also known in the art. The presently preferred method for expression of IL-11 in E. coli, involves removing the first 31 codons of the human IL-11 sequence.
The following sequence is then attached at codon 32 of the mature human IL-11 sequence:
ATG CCA GGT CCA CCA CCA GGT CCA CCT CGA GTT 1 5 10
C. Insect or Yeast Cell Expression
Similar manipulations can be performed for the construction of an insect vector [See, e.g., procedures described in published European patent application 155,476] for expression in insect cells. A yeast vector could also be constructed employing yeast regulatory sequences for intracellular or extracellular expression of the proteins of the present invention by yeast cells. [See, e.g., procedures described in published PCT application WO 86/00639 and European patent application EP 123,289. ] Example 10 - Construction of CHO Cell Lines Expressing High Levels of IL-11
One method for producing high levels of the IL- 11 polypeptides of the invention from mammalian cells involves the construction of cells containing multiple copies of the heterologous IL-11 gene. The heterologous gene can be linked to an amplifiable marker, e.g., the dihydrofolate reductase (DHFR) gene for which cells containing increased gene copies can be selected for propagation in increasing concentrations of methotrexate (MTX) according to the procedures of Kaufman & Sharp, J. Mol. Biol.. (1982) supr . This approach can be employed with a number of different cell types. Alternatively, the IL-11 cDNA and drug resistance selection gene (e.g., DHFR) may be introduced into the same vector. A preferred vector for this approach is pEMC2Bl.
For example, the pXM vector containing a IL-ll gene in operative association with other plasmid sequences enabling expression thereof and the DHFR expression plasmid pAdA26SV(A)3 (Kaufman & Sharp, Mol. Cell Biol. , 2(9) :1598-1608 (1983) can be co-introduced into DHFR-deficient CHO cells, DUKX-BII, by calcium phosphate coprecipitation and transfection.
Alternatively, the pEMC-2Bl vector containing the IL-11 gene in operative association with other plasmid sequences enabling expression thereof is introduced into DHFR-deficient CHO cells, DUKX-BII, by protoplast fusion and transfection. The IL-11 gene and DHFR marker gene are both efficiently expressed when IL-9 is introduced into pEMC2Bl. The IL-11 gene may be introduced into pMT2 as previously mentioned and the resultant vector used in place of pXM/IL-11 and pAdA26SV(A)3.
DHFR expressing transformants are selected for growth in alpha media with dialyzed fetal calf serum. Transformants are checked for expression of IL-9 by bioassay, immunoassay or RNA blotting and positive pools are subsequently selected for amplification by growth in increasing concentrations of MTX (sequential steps in 0.02, 0.2, 1.0 and 5 uM MTX) as described in Kaufman et al, Mol. Cell Biol.. 5_:1750 (1983). The amplified lines are cloned, and biologically active IL-11 polypeptide expression is monitored by the T1165 assay. IL-11 polypeptide expression is expected to increase with increasing levels of MTX resistance. In any of the expression systems described above, the resulting cell lines can be further amplified by appropriate drug selection, resulting cell lines recloned and the level of expression assessed using the T1165 assay described herein.
The IL-11 expressing CHO cell lines can be adapted to growth in serum-free medium. Homogeneous IL- 11 can be isolated from conditioned medium from the cell line using methods familiar in the art, including techniques such as lectin-affinity chromatography, reverse phase HPLC, FPLC and the like.
Example 11 - Effect of IL-11 on Proliferation in Culture of Early Murine Progenitors
Methylcellulose cell cultures were established in 35 mm Lux suspension culture dishes (#5221R, Nunc,
Inc. Naperville, IL) .
5-Fluorouracil (5-FU) (Adria Laboratories,
Columbia, OH) was administered intravenously through the tail veins of 10 to 15 week old female BDF1 mice [ARS Sprague Dawley, Indianapolis, IN] at 150 mg/kg body weight [T. Suda et al, J. Cell. Physiol.. 117:308-318
(1983) and G. S. Hodgson et al. Nature, 281:381-382 (1979)]. Single cell suspensions were prepared from pooled femurs or spleens of three mice. Light density (<1,077) mononuclear cells were collected from the interface of Ficoll-Paque after centrifugation at 400 g. After overnight adherence of these cells to plastic dishes, nonadherent mononuclear (bone marrow and spleen) cells were harvested 2 and 5 days after 5-FU injection, respectively.
One ml of culture contained 2 x 104 marrow cells from normal mice, 5 x 104 marrow cells or 1 x 106 spleen cells from 5-FU-treated mice, α-medium (Flow Laboratories, Inc., McLean, VA) , 1.2% 1,500 cps methylcellulose (Fisher Scientific Co., Norcross, GA) , 30% fetal calf serum (FCS) (Hyclone Laboratories, Inc. , Logan, UT) , 1% deionized fraction V bovine serum albumin (BSA) (Sigma Chemical Co., St. Louis, MO), 1 x 10"4 M 2- mercaptoethanol (Eastman Organic Chemicals, Rochester, NY) and hemopoietic factors. Dishes were incubated at 37°C in a humidified atmosphere flushed with 5% C02. Except for megakaryocyte colonies, colonies consisting of 50 or more cells were scored on an inverted microscope on the specified day of incubation. Megakaryocyte colonies were scored when they contained four or more εgakaryocytes. Abbreviations for colony types are as follows: GM, granulocyte/macrophage; Mast, mast cell colonies; E, erythroid bursts; M, megakaryocyte colonies; GMM, granulocyte/macrophage/megakaryocyte colonies [T. Nakahata et al, J. Cell. Phvsiol. , 111:239-246 (1982)]; GEMM, granulocyte/erythrocyte/macrophage/megakaryocyte colonies [T. Nakahata et al, cited above; and A. A.
Fauser et al, Blood, 5_2:1243-1248 (1978)]; and Bl, blast cell colonies [T. Nakahata et al, Proc. Natl. Acad. Sci. USA, 29:3843-3847 (1982); and T. Suda et al, cited above] . The hemopoietic potential of the blast cell colonies was determined by blast cell colony replating. Between days 5 and 15 of incubation, individual blast cell colonies containing 50 to 150 cells were picked with an Eppendorf pipet and replated in secondary methylcellulose cultures containing 2 U/ml human urinary erythropoietin (Ep) [activity of 370 U per g, available from Dr. Makoto Kawakita, Kumamoto University Medical School, Kumamoto, Japan], 1% (v/v) concentrated (X20) supernatant of cultures of WEHI-3 cells. Blast cells were also used as pure target populations of hemopoietic cells to determine whether the observed effects of IL-11 were direct or due to the release of other factors. One million day-4 post-5-FU spleen cells were cultured in the presence of 100 U/ml of recombinant murine IL-3. IL-3 was conditioned by Chinese Hamster Ovary (CHO) cells that had been genetically engineered to produce murine IL-3 to high titer (approximately 30,000 U/ml). On day 8 of culture, individual blast cell colonies (between 50 to 150 cells) were picked from cultures, pooled, washed twice with medium and replated in secondary cultures each containing different combinations of factors.
Recombinant human IL-6 with specific activity of 4 x 106 U/mg protein was expressed in E^. coli. IL-11 was medium conditioned (CM) by COS-1 cells transfected with cDNA encoding the murine plasmacytoma-stimulatory activity [S. R. Paul et al, Proc. Natl. Acad. Sci. USA, in press (1990) ] . A. Colony Formation from Marrow
Cells of Normal Mice
Colony formation from normal marrow cells was supported by IL-11. In the presence or absence of 2 U/ml Ep, IL-11 gave rise to colonies in a dose-dependent manner. A 1:100 dilution of IL-11 supported maximal colony formation. However, the total number of colonies detected on day 8 or day 16 of incubation with IL-11 was significantly fewer than in cultures with IL-3. The colonies found in IL-11-containing cultures were predominantly of the GM type, although some multilineage (GMM and GEMM) colonies were also observed. IL-11 in a 1:100 dilution supported formation of three blast cell colonies on day 16 of incubation.
B. Colony Formation from Marrow Cells of 5-FU-Treated Mice
Colony formation from marrow cells harvested two days after injection of 150 mg/kg 5-FU [T. Suda et al., cited above; and G. S. Hodgson et al., cited above] in cultures established in the presence of IL-11, IL-6, IL-3 singly and in various combinations was studied to determine whether IL-11 acts synergistically with IL-3 in supporting the proliferation of primitive progenitors.
Addition of IL-11 at final dilutions of 1:100 and 1:1,000 to an optimal concentration of IL-3 significantly enhanced colony formation. In particular, in the presence of a 1:100 dilution of IL-11 and IL-3 the kinetics of colony formation was accelerated as compared to that supported by the individual factors. The time course of colony formation as well as the total number of colonies supported were similar to those observed with the combination of IL-6 and IL-3. IL-11 alone in a 1:100 dilution supported scant colony formation after a long period of incubation. These results indicated that IL-11 enhances IL-3-dependent proliferation of primitive progenitors. The effects of a combination of IL-11 and IL-6 on the kinetics of colony formation from day-2 post-5-FU marrow cells relative to the effects of individual synergistic factors were tested separately. IL-6 and IL- 11 significantly accelerated IL-3-dependent colony formation. The effects of the combination of IL-6 and IL-11, however, did not differ from those of individual factors.
C. Serial Observations of Blast Cell Colony Development from Daγ-4 Post-5-
FU Spleen Cells
The growth rates of individual blast cell colonies were serially plotted through culture mapping studies. The results indicated that the synergistic effect of IL-11 results from a decrease in the time stem cells spend in the dormant state, an effect very similar to that observed with IL-6 or G-CSF because the growth rates were not statistically different in these culture systems. D. Comparison of the Replating
Potentials of Blast Cell Colonies The proliferative potentials of blast cell colonies that respond to IL-11 and IL-6 were tested by replating experiments. Significant variations in the secondary replating efficiencies were seen among individual blast cell colonies as reported previously [K. Ikebuchi et al., Blood. 12.:2007-2014 (1988)]. There was, however, no significant differences in the replating efficiencies of blast cell colonies grown in the three different primary culture conditions.
Similar to previous observations [K. Ikebuchi et al., cited above], the percentages of secondary GEMM colonies in secondary colonies and the incidences of secondary GEMM colonies per blast cell colonies were significantly higher from the primary blast cell colonies identified in cultures containing IL-11 or IL-6 than those seen in cultures containing IL-3 alone. There were no significant differences in these parameters between cultures containing IL-11 plus IL-3 and cultures containing IL-6 plus IL-3.
These results indicated that the synergistic activities of IL-11 and IL-6 are similar and that the increases in the incidences of secondary GEMM colonies may be due to shortening of the G. period of stem cells during blast cell colony formation [K. Ikebuchi et al., cited above] . E. Replating Studies of Pooled
Blast Cells Target cells obtained by pooling blast cells from early stages of cultures supported by IL-3 were used to compare the direct effects of IL-11 and IL-6 on GM colony formation. Pooled blast cells are devoid of stromal cells and express very high replating efficiencies.
Blast cell colonies containing 50 to 150 cells identified in cultures containing IL-3 were picked and pooled, and were replated in secondary cultures containing IL-11, IL-6 or IL-3 in the presence of 2 U/ml Ep. These date indicate that at least 70% of the blast cells are hemopoietic progenitors. While the combination of IL-3 and Ep supported formation of a variety of single lineage and multilineage colonies, IL-11 and Ep supported the formation of only macrophage colonies. The combination of IL-6 and Ep supported formation of a similar number of pure macrophage colonies but also neutrophil/macrophage colonies. The macrophage colonies supported by IL-11 were smaller than the macrophage colonies supported by _-.L—6. These results indicated that IL-11 and IL-6 interact with overlapping but different progenitor subsets and that IL-11 preferentially supports the macrophage progenitor population. F. The Effects of Neutralizing Anti-
IL-6 Antibody on the Synergistic Effects of IL-11
In order to confirm that the direct colony-supporting ability between IL-11 and IL-6 was not a result of the crude nature of the Cos cell CM, neutralizing anti-IL-6 antibody, which is known to inhibit Cos-derived IL-6, was used to study the synergistic effects of IL-11 and IL-6 on IL3-dependent proliferation from dormant progenitors. In the presence of IL-6 or IL-11 and in the absence of antibodies, colony development from day-4 post-5-FU spleen cells was significantly hastened as indicated by the number of colonies on day 8, When anti- IL-6 antibody was present, the synergistic effects of IL- 6 were completely abrogated while the effects of IL-11 were not. The effects of the antibody persisted until day 16. These results excluded the possibility that the apparent synergistic effects in Cos cell CM were mediated The conditioned medium (CM) of COS cells transfected with IL-11 cDNA has been found to augment IL- 3-dependent proliferation of multipotential progenitors in culture, an activity originally associated with IL-6. The mechanism of the augmentation appears to be shortening of the G„ period of dormant stem cells.
The foregoing descriptions detail presently preferred embodiments of the invention. Numerous modifications and variations in practice of this invention are expected to occur to those skilled in the art. Such modifications and variations are encompassed within the following claims.

Claims (24)

1. Mammalian IL-11 substantially free from association with other proteinaceous materials.
2. The factor according to claim 1 comprising the same or substantially the same sequence of amino acids from amino acid #21 to amino acid #199 of Table I, or a biologically active fragment thereof.
3. The factor according to claim 2 comprising the human sequence.
4. The factor according to claim 1 encoded by all or a portion of the same or substantially the same DNA sequence as in Table I, a fragment thereof, or a DNA sequence capable of hybridizing thereto.
5. The factor according to claim 1 having one or more of the following characteristics:
(1) an apparent molecular weight under reducing conditions on SDS PAGE of approximately 20 kd;
(2) a calculated molecular weight of approximately 20 kd;
(3) biological activity in a T1165 assay; (4) biological activity in a megakaryocyte colony forming assay in the presence of IL-3 ;
(5) biological activity on a B cell plaque forming assay.
6. The factor according to claim 1 produced by culturing a cell transformed with a DNA sequence encoding expression of an IL-11 polypeptide in operative association with an expression control sequence capable of directing the replication and expression of IL-11, and recovering from the conditioned medium thereof an IL-11 protein.
7. The factor according to claim 5 wherein said DNA sequence comprises the same or substantially the same DNA sequence of Table I, a fragment thereof, or a DNA sequence capable of hybridizing thereto.
8. The factor according to claim 1 which is human IL-11.
9. A process for producing IL-11 or a fragment thereof comprising culturing a cell line transformed with a cDNA sequence encoding expression of IL-11 or a fragment thereof in operative association with an expression control sequence capable of directing the replication and expression of IL-11.
10. A DNA sequence coding for IL-11 and comprising all or a portion of the same or substantially the same sequence of nucleotide bases as in Table I, a fragment thereof, or a DNA sequence capable of hybridizing thereto.
11. A cell transformed with the DNA sequence of claim 10 in operative association with an expression control sequence.
12. The cell according to claim 11 comprising a mammalian or bacterial cell.
13. A plasmid vector comprising a DNA sequence of claim 10.
14. A pharmaceutical composition comprising a therapeutically effective amount of IL-11 or a biologically active fragment thereof in a pharmaceutically effective vehicle.
15. The composition according to claim 14 further comprising therapeutically effective amounts of an additional cytokine, hematopoietin, growth factor or antibody.
16. The composition according to claim 15 where said cytokine is selected from the group consisting of
IL-1 through IL-9, GM-CSF, G-CSF, M-CSF, the interferons, Meg-CSF, MIF, LIF, TNF, and erythropoietin.
17. The composition according to claim 16 wherein said cytokine is IL-3 or IL-6.
18. A method for stimulating the immune system or treating disorders relating thereto comprising administering to a patient an effective amount of IL-11 or a fragment thereof.
19. The method according to claim 18 further comprising administering simultaneously or sequentially with said IL-11 an effective amount of at least one hematopoietin, cytokine, growth factor or antibody.
20. The method according to claim 19 wherein said hematopoietin is IL-3 or IL-6.
21. A method for stimulating the hematopoietic system or treating disorders related thereto comprising administering to a patient an effective amount of IL-11 or a fragment thereof.
22. The method according to claim 21 further comprising administering simultaneously or sequentially with said IL-ll an effective amount of at least one hematopoietin, cytokine, growth factor or antibody.
23. The method according to claim 22 wherein said hematopoietin is IL-3 or IL-6.
24. Homogeneous mammalian IL-11 having biological activity in the T1165 assay in the absence of IL-6.
AU67578/90A 1989-11-22 1990-11-20 A mammalian cytokine, IL-11 Expired AU644389C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US44110089A 1989-11-22 1989-11-22
US526474 1990-05-21
US07/526,474 US5215895A (en) 1989-11-22 1990-05-21 Dna encoding a mammalian cytokine, interleukin-11
US441100 1990-05-21
PCT/US1990/006803 WO1991007495A1 (en) 1989-11-22 1990-11-20 A mammalian cytokine, il-11

Publications (3)

Publication Number Publication Date
AU6757890A AU6757890A (en) 1991-06-13
AU644389B2 AU644389B2 (en) 1993-12-09
AU644389C true AU644389C (en) 1995-03-23

Family

ID=

Similar Documents

Publication Publication Date Title
EP0504177B1 (en) A mammalian cytokine, il-11
AU637620B2 (en) A human cytokine, interleukin-9
US4959455A (en) Primate hematopoietic growth factors IL-3 and pharmaceutical compositions
US5457038A (en) Natural killer stimulatory factor
US4877729A (en) Recombinant DNA encoding novel family of primate hematopoietic growth factors
US20070104680A1 (en) Antibodies to natural killer stimulatory factor
IL83137A (en) Non-glycosylated il-6 polypeptide pharmaceutical composition containing it dna sequence coding for its bacterial expression vector containing said sequence and bacterial cells transformed by them
US5371193A (en) Mammalian cytokine, IL-11
WO1990011301A1 (en) Human macrophage migration inhibitory factor
US5414071A (en) Human cytokine IL-9
AU644389C (en) A mammalian cytokine, IL-11
US6348191B1 (en) Production and use of IL-6
US6787645B1 (en) DNA encoding human JE cytokine
US6284237B1 (en) Methods of treatment using IL-6
IE871727L (en) Novel family of primate il3-like hematopoietic growth¹factors.