Title of Invention

IL-7 VARIANTS WITH REDUCED IMMUNOGENICITY

Abstract A polypeptide comprising a modified human IL-7 molecule or an active portion thereof having a T-cell epitope modified to reduce an anti-IL-7 T-cell response, said polypeptide further comprising an Fc portion of an immunoglobulin molecule fused via its C-terminal to the N-terminal of said modified IL-7 molecule, wherein said fused Fc-IL7 molecule is selected from the group consisting of: (i) huFcy2(h)(FN>AQ) - L - IL-7(F39P, F57N, L128S), wherein Fcy2(h) is a human Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2 comprising the mutations F296A and N297Q, L is a linker with the sequence GGGGSGGGG, and IL-7(F39P, F57N, L128S) is IL-7 containing the mutations F39P, F57N, and L128S; said molecule having the sequence as depicted in Fig. 31; (ii) huFcy2(h)(FN>AQ) - L - IL-7(F39P, F57N, L77D, L128S), wherein Fcy2(h) is a human Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2 comprising the mutations F296A and N297Q, L is a linker with the sequence GGGGSGGGG, and IL-7(F39P, F57N, L77D L128S) is IL-7 containing the mutations F39P, F57N, L77D and L128S; said molecule having the sequence as depicted in Fig. 32; (iii) huFcy2(h) - L - IL-7 (F39P, F57N, L77D, L128S), wherein Fcy2(h) is a human Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2, L is a linker with the sequence GGGGSGGGG, and IL-7(F39P, F57N, L77D, L128S) is IL-7 containing the mutations F39P, F57N, L77D and L128S; said molecule having the sequence as depicted in Fig. 33; and (iv) huFcy2(h) - L - IL-7 (F39P, F57N, L128S), wherein Fcy2(h) is a human Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2 L is a linker with the sequence GGGGSGGGG, and IL-7(F39P, F57N, L128S) is IL-7 containing the mutations F39P, F57N, and L128S; said molecule having the sequence as depicted in Fig. 34.
Full Text WO 2006/061219 PCT/EP2005/013145
1
IL-7 Variants with Reduced Immunogenicity
Cross Reference to Related Applications
[0001] This application claims priority to and the benefit of U.S. Provisional
Patent Application No. 60/634,470, filed on December 9, 2004, the entire
disclosure of which is incorporated by reference herein.
Field of the Invention
[0002] The invention relates generally to IL-7 molecules modified to reduce
their immunogenicity. These molecules include also fusion proteins comprising
said modified IL-7 molecules and immunoglobulin molecules or portions thereof,
especially corresponding Fc fusion proteins.
Background
[0003] Cytokines are stimulators of the immune system and are thus useful
as drugs. For example, interferon-alpha (IFN-a), interferon-beta (IFN-fi),
inierleuk/n-2 (IL-2), and granuiocyte/macrophage-colony stimulating factor (GM-
CSF) are all approved drugs used to treat viral infections, cancer, immune system
misregulation such as autoimmune disease, and to promote recovery of the
immune system after cancer chemotherapy. Unfortunately, these proteins can
stimulate an immune response against themselves, causing patients to develop
antibodies against the therapeutic protein. These antibodies can also inhibit
function of the same protein endogenously produced within the patient, resulting
in potential long-term consequences for patient health.
[0004] lnterleukin-7 is a cytokine that promotes survival and proliferation of T-
cells, B-cells, and other immune cells. It is also potentially a therapeutic protein
to treat patients whose immune systems have been damaged by cancer
chemotherapy, HIV infection, or other diseases, disorders, or chemical
exposures. However, based on its immunostimulatory properties, therapeutically
administered IL-7 is expected to induce an antibody response against itself.
Therefore, there is a need in the art for improved versions of IL-7 that are less
immunogenic, but that retain the property of stimulating the immune system.
CONFIRMATION COPY

WO 2006/061219 PCT/EP2005/013145
2
Summary of the Invention
[0005] The present invention is directed to interleukin-7 (IL-7) which has been
modified to reduce its immunogenicity in comparison to wild-type IL-7. More
specifically, the IL-7 proteins of the invention are modified to remove potential T-
cell epitopes. As a result, IL-7 proteins, including immunoglobulin -IL7 fusion
proteins, preferably Fc-IL7 fusion proteins, of the invention have improved
biological properties compared to wild-type IL-7.
[0006] Accordingly, in one aspect, the invention features a polypeptide at
least 80% identical to a human IL-7 moijety or an active portion thereof,
comprising an amino acid substitution at one or more residues corresponding to
Gln22, Leu24, He30, Phe39, Met54, Phe57, Arg58, Ala60, Leu63, Lys68, Met69,
Leu77, Ile88, Val96, Leu 104, Leu 128, Met147, Thr149, or Lys150. These amino
acid modifications can be used singly or in combination to reduce an anti-IL-7 T-
cell response. Thus, the invention encompasses IL-7 moieties with for example,
one, at least two, at least four, or at least eight amino acid modifications at
positions selected from Gin22, Leu24, He30, Phe39, Met54, Phe57, Arg58, Ala60,
Leu63, Lys68, Met 69, Leu77, Ile88, Val96, Leu104, Leu128, Met147, Thr149,
and Lys150. In one embodiment, the IL-7 moiety incorporates one, two, three,
four, five or more of the following substitutions: Gln22Asp, Leu24Asp, IIe30Thr,
Phe39Pro, Met54Ala, Phe57Lys, Phe57Asn, Arg58Asp, Ala60Ser, Arg61Glu,
Leu77Asp, Leu104Ser, Leu104Vai, Leu128Ala, Leu128Val, Leu128Pro,
Leu128Ser, Met147Lys, Thr149Ser, or Lys150Stop.
[0007] In one embodiment, the polypeptide contains a substitution or
substitutions at one or more at Phe39, Phe57, Leu77, and Leu 128. In a further
embodiment, the polypeptide has one or more of substitutions Phe39Pro,
Phe57Asn, Leu77Asp, and Leu128Ser. In another embodiment, the polypeptide
includes the substitutions Phe39Pro, Phe57Asn] Leu77Asp, and Leu128Ser,
while in a further embodiment, the polypeptide includes the substitutions
Phe39Pro, Phe57Asn, and Leu128Ser.
Preferred substitutions according to this invention are at postions:
Phe39, or Phe57, or Leu77, or Leu128;
Phe39 and Phe57, or Phe39 and Leu77, or Phe57 and Leu77,or Plne39 and
Leu128, or Phe57 and Leu128, or Leu77 and Leu128;
Phe39 and Phe 57 and Leu77, or Phe39 and Phe57 and Leu128, or

WO 2006/061219 PCT/EP2005/013145
3
Phe57 and Leu77 and Leu128, or Phe39 and Leu77 and Leu128;
Phe39 and Phe 57 and Leu77 and Leu128.
Preferred specific substitutions according to this invention are:
Phe39Pro, or Phe57Lys, or Leu77Asp, or Leu128Ser;
Phe39Pro and Phe57Lys, or Phe39Pro and Leu77Asp, or Phe57Lys and
Leu77Asp, or Phe39Pro and Leu128Ser, or Phe57Lys and Leu128Ser, or
Leu77Asp and Leu128Ser;
Phe39Pr and Phe 57Lys and Leu77Asp, or Phe39Pro and Phe57Lys and
Leu128Ser, or
Phe57Lys and Leu77Asp and Leu128Ser, or Phe39Pro and Leu77Asp and
Leu128Ser;
Phe39Pro and Phe 57Lys and Leu77Asp and Leu128Ser.
[0008] In certain embodiments of the invention, the polypeptide with at least
80% identity with a human IL-7 moiety further comprises an immunoglobulin (Ig)
moiety, such as a human Ig moiety, In one embodiment, the Ig moiety is lgG2.
In some embodiments, the Ig moiety is an Fc portion. The invention also relates
to a cell comprising a nucleic acid sequence encoding a polypeptide modified
according to the invention. In one embodiment, the cell is a prokaryotic cell.
[0009] In a further embodiment, the polypeptide has at least 90% identity to a
human IL-7 moiety or an active portion thereof, while in another embodiment, the
polypeptide has at least 95% identity to a human IL-7 moiety or an active portion
thereof.
[0010] The invention also features a method of treating a patient comprising
administering a therapeutically effective amount of a polypeptide of the invention
to, for example, a patient diagnosed with cancer or HIV. In one embodiment, the
invention provides for administration of between about 0.01 and about 10
mg/kg/day or between 0.01 and 10.00 mg/kg/day of a polypeptide of the
invention.
Brief Description of the Drawings
[0011] Figure 1 depicts the amino acid sequence for human IL-7. The signal
sequence is shown in bold. Also depicted in bold and italics is a stretch of
eighteen amino acids which can be deleted from the IL-7 sequence.

WO 2006/061219 PCT/EP2005/013145
4
[0012] Figure 2 depicts the amino acid sequence for cow IL-7. The signal
sequence is shown in bold.
[0013] Figure 3 depicts the amino acid sequence for sheep IL-7. The signal
sequence is shown in bold.
[0014] Figure 4 depicts the amino acid sequence of an exemplary
deimmunized human IL-7 wherein T cell epitope sequences have been modified.
[0015] Figure 5 depicts the amino acid sequence of a bacterially produced
deimmunized human IL-7.
[0016] Figure 6 depicts the nucleic acid sequence encoding mature IL-7
incorporating codons for the mutations F39P, F57N, and L128S.
[0017] Figure 7 depicts the amino acid sequence of mature IL-7 with the
mutations F39P, F57N and L128S.
[0018] Figure 8 depicts the nucleic acid sequence encoding mature IL-7
incorporating codons for the mutations F39P, F57N, L77D, and L128S.
[0019] Figure 9 depicts the amino acid sequence of mature IL-7 with the
mutations F39P, F57N, L77D and L128S.
[0020] Figure 10 depicts the nucleic acid sequence encoding bacterially
produced deimmunized IL-7 (bDel-IL-7), codon-optimized for £. co//with codons
for the amino acid mutations K68D, M69D, I88T, V96G.
[0021] Figure 11 depicts the nucleic acid sequence encoding a mature IL-7
variant, deimmunized IL-7 (De!-lL-7), with codons for the amino acid mutations
K68D, M69D, I88T, V96G.
[0022] Figure 12 depicts the amino acid sequence of FCK1-IL-7, where the Fc
portion consists of a K1 hinge, y\ CH2, and y\ CH3 region.
[0023] Figure 13 depicts the amino acid sequence of human
Fcy2(h)(FN>AQ)-IL-7, which is an Fc portion with a K1 hinge, a y2 CH2 domain,
and a y2 CH3 domain. The Fc portion incorporates the amino acid mutations
F296A and N297Q.
[0024] Figure 14 depicts the amino acid sequence of human FC)/1-(linker1)-
IL-7, which is an Fc portion consisting of a y1 hinge, CH1, and CH2 region
connected to an IL-7 moiety via a poiypeptide linker of amino acid sequence
GGGGSGGGGSGGGGS.
[0025] Figure 15 depicts the amino acid sequence of human Fcy1 (YN>AQ)-
(iinker 2)-lL-7, which Is a yi Fc portion with a y\ hinge, CH1, and CH2 region,

WO 2006/061219 PCT/EP2005/013145
5
incorporating the mutations Y296A and N297Q, connected to an IL-7 moiety via a
polypeptide linker of amino acid sequence GGGGSGGGG.
[0026] Figure 16 depicts the amino acid sequence of human Fcj/1 (YN>AQ,d)-
(linker 2)-IL-7, which is a K1 FC portion with a y\ hinge, CH1, and CH2 domains,
incorporating the mutations Y296A and N297Q, as well deletion of the C-terminal
lysine and preceding glycine of the Fc moiety. The Fc portion is connected to an
IL-7 moiety via a polypeptide linker of amino acid sequence GGGGSGGGG.
[0027] Figure 17 depicts the nucleic acid sequence of Fcy1, an Fc portion
with a hinge, CH1 domain and CH2 domain, all of lgG1.
[0028] Figure 18 depicts the nucleic acid sequence of Fcp1(YN>AQ), which is
an Fc portion with a hinge, CH1 domain and CH2 domain, all of lgG1. The Fc
portion incorporates the mutations Tyr296Ala and Asn297Gln.
[0029] Figure 19 depicts the nucleic acid sequence of Fcy2(h), which is an Fc
portion with an lgG1 hinge and lgG2 CH2 and CH3 domains.
[0030] Figure 20 depicts the nucleic acid sequence of FcK2(h){FN>AQ),
which is an Fc portion with an lgG1 hinge and lgG2 CH2 and CH3 domains. The
Fc portion incorporates the mutations F296A and N297Q.
[0031] Figure 21 depicts the amino acid sequence of mature human
deimmunized IL-7.1, wherein the IL-7 incorporates the substitutions L24D, M54A,
F57K, A60S, R61E, M147K, T149S, and deletes residues K150, E151, and H152.
[0032] Figure 22 depicts the nucleic acid sequence encoding the amino acid
sequence of Figure 21.
[0033] Figure 23 depicts the amino acid sequence of mature human
deimmunized IL-7.2 which incorporates the substitutions D76N, L77D, T87Q,
i88T, V96G, L119S, L128V, M147K, T149S, and deletes K150, E151, and H152.
[0034] Figure 24 depicts the nucleic acid sequence encoding the amino acid
sequence of Figure 23.
[0035] Figure 25 depicts the amino acid sequence of mature human
deimmunized IL-7.3 which incorporates the substitutions L24D, I30T, F39P,
M54A, F57K, A60S, R61E, M68D, N69D, L77D, T87Q, I88T, V96G, L119S,
L128A, M147K, T149S, and deletes K150, E151, and H152.
[0036] Figure 26 depicts the nucleic acid sequence encoding the amino acid
sequence of Figure 25.

WO 2006/061219 PCT/EP2005/013145
6
[0037] Figure 27 depicts the nucleic acid sequence encoding the linker
sequence GGGGSGGGG followed by mature human IL-7 containing the amino
acid substitution F39P, F57N, and L128S (PNS), and which contains flanking
restriction sites Xma I and Xho I at the 51 and 3! ends respectively.
[0038] Figure 28 depicts the nucleic acid sequence of mature
huFcy2(h)(FN>AQ)(linker2)-IL-7(F39P, F57N, L77D, L128S) which is a human Fc
portion with an lgG1 hinge, and lgG2 CH2 and CH3 domains incorporating the
mutations F296A and N297Q connected to the N-terminus of a human IL-7
moiety incorporating the mutations F39P, F57N, L77D, and L128S. The Fc
portion and the IL-7 moiety are connected by a linker sequence GGGGSGGGG.
[0039] Figure 29 depicts the nucleic acid sequence of mature huFcK2(h)-
(Iinker2)-IL-7(F39P, F57N, L77D, L128S) which is a human Fc portion with an
lgG1 hinge, and lgG2 CH2 and CH3 domains connected to the N-terminus of a
human IL-7 moiety incorporating the mutations F39P, F57N, L77D and L128S.
The Fc portion and the IL-7 moiety are connected by a linker sequence
GGGGSGGGG.
[0040] Figure 30 depicts the nucleic acid sequence of mature huFcy2(h)-
(Iinker2)-IL-7(F39P, F57N, L128S) which is a human Fc portion with an lgG1
hinge, and lgG2 CH2 and CH3 domains connected to the N-terminus of a human
IL-7 moiety incorporating the mutations F39P, F57N, and L128S. The Fc portion
and the IL-7 moiety are connected by a linker sequence GGGGSGGGG.
[0041] Figure 31 depicts the amino acid sequence of mature
huFcy2(h)(FN>AQ)-(linker2)-IL-7(F39P, F57N, L128S), which is a human Fc
portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2. The Fc portion
contains the mutations F296A and N297Q. The Fc portion is linked to the IL-7
moiety via a linker of sequence GGGGSGGGG. The IL-7 moiety contains the
mutations F39P, F57N, and L128S.
[0042] Figure 32 depicts the amino acid sequence of mature
huFcy2(h)(FN>AQMIinker2)-IL~7(F39P, F57N, L77D, L128S), which is a human
Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2. The Fc
portion contains the mutations F296A and N297Q. The Fc portion is linked to the
IL-7 moiety via a linker of sequence GGGGSGGGG. The IL-7 moiety contains
the mutations F39P, F57N, L77D and L128S.

WO 2006/061219 PCT/EP2005/013145
7
[0043] Figure 33 depicts the amino acid sequence of mature huFcy2(h)-
(Iinker2)-IL-7(F39P, F57N, L77D, L128S), which is a human Fc portion with a
hinge of lgG1 and CH2 and CH3 domains of lgG2. The Fc portion is linked to the
IL-7 moiety via a linker of sequence GGGGSGGGG. The IL-7 moiety contains
the mutations F39P, F57N, L77D and L128S.
[0044] Figure 34 depicts the amino acid sequence of mature huFcy2(h)
(Iinker2)-IL-7(F39P, F57N, L128S), which is a human Fc portion with a hinge of
igG1 and CH2 and CH3 domains of lgG2. The Fc portion is linked to the IL-7
moiety via a linker of sequence GGGGSGGGG. The IL-7 moiety contains the
mutations F39P, F57N, L77D and L128S.
[0045] Figure 35 is an amino acid sequence alignment of IL-7 proteins from
human, chimpanzee, baboon, macaque, bovine, pig, sheep, rat, and murine
sources.
[0046] Figure 36 depicts Fc-IL-7 plasma concentrations in μg/ml for both test
mice and control mice administered Fc-IL-7 subcutaneously.
[0047] Figure 37 shows values for the average fold change in plasma Fc-IL-7
concentrations between day 0 and day 2, and between day 2 and 4 in test mice
administered Fc-IL-7 subcutaneously (SC).
[0048] Figure 38 depicts the average organ weights of organs taken from test
mice sacrificed on day 7 compared to the average organ weights of mice in the
control group.
[0049] Figure 39 depicts a comparison of the frequency of granulocyte Gr-1 +
cells in cells/μL in the peripheral blood of mice from the control group, 0.5 mg/kg
dosage group, 5.0 mg/kg dosage group, and 25 mg/kg dosage group on day 7.
[0050] Figure 40 depicts a comparison of the frequency of CD19+ cells in
cellslμL in the peripheral blood of mice from the control group, 0.5 mg/kg dosage
group, 5.0 mg/kg dosage group, and 25 mg/kg dosage group on day 7.
[0051] Figure 41 depicts a comparison of the frequency of CD4+ cells in
cells/μL in the peripheral blood of test from the control group, 0.5 mg/kg dosage
group, 5.0 mg/kg dosage group, and 25 mg/kg dosage group on day 7.
[0052] Figure 42 depicts a comparison of the frequency of CD8+ cells in
cells/μL in the peripheral blood of test from the control group, 0.5 mg/kg dosage
group, 5.0 mg/kg dosage group, and 25 mg/kg dosage group on day 7.

WO 2006/061219 PCT/EP2005/013145
8
[0053] Figure 43 depicts the activity of Fc-lL-7 as compared to wild type IL-7
based on incorporation of tritiated thymidine in counts per minute versus IL-7/Fc-
IL-7 concentration in a standard cell proliferation assay.
Detailed Description of the Invention
[0054] The invention is directed to IL-7 proteins that have reduced
immunogenicity as compared to wild-type IL-7, as well as methods for making
and using such proteins. More specifically, the invention provides mutations
within IL-7 moieties that have the effect of reducing the immunogenicity of IL-7
itself, primarily by removing T-cell epitopes within IL-7 that may stimulate to an
immune response. The invention also encompasses fusion proteins
incorporating IL-7 moieties modified according to the teachings of the invention.
[0055] T-cell epitopes can be identified by a variety of computer and non-
computer methods, including predictions based on structure-based computer
modeling or by synthesis of peptides and testing for binding to specific MHC
Class II molecules or in an immunogenicity assay. According to the invention, a
potential T-cell epitope is a sequence that, when considered as an isolated
peptide, is predicted to bind to an MHC Class II molecule or an equivalent in a
non-human species. A potential T-cell epitope is defined without consideration of
other aspects of antigen processing, such as the efficiency of protein uptake into
antigen-presenting cells, the efficiency of cleavage at sites in an intact protein to
yield a peptide that can bind to MHC Class II, and so on. Thus, the set of T-cell
epitopes that are actually presented on MHC Class II after administration of a
protein to an animal is a subset of the potential T-cell epitopes. According to the
invention, a T-cell epitope is an epitope on a protein that interacts with an MHC
class II molecule. Without wishing to be bound by theory, it is understood that a
T-cell epitope is an amino acid sequence in a protein that failed to undergo the
negative T-cell selection process during T-cell development and therefore will be
expected to be presented by an MHC Class II molecule and recognized by a T-
cell receptor.
[0056] B-cell epitopes are also identified by a variety of computer and non-
computer methods, including predictions based on structure-based computer
modeling or by synthesis of peptides and testing for binding to specific B-cell
antigen receptor molecules or in an immunogenicity assay. According to the

WO 2006/061219 PCT/EP2005/013145
9
invention, a potential B-cell epitope is a sequence that, when considered as an
isolated peptide, is predicted to bind to a B-celi antigen receptor or an equivalent
in a non-human species. A B-cell epitope is an epitope that does bind or is
recognized by a B-cell antigen receptor and is a subset of potential B-ce!l
epitopes.
[0057] The invention provides methods related to reducing the
immunogenicity of IL-7. According to one embodiment of the invention, potential
non-self T-cell epitopes are identified in sequences of IL-7. For example,
potential non-self T-cell epitopes are identified by computational methods based
on modeling peptide binding to MHC Class II molecules. Substitutions are then
made such that the ability of peptides containing potential T-cell epitopes to bind
to MHC Class II is reduced or eliminated. This process of identifying and
modifying peptides which bind to MHC Class II is termed "de-immunization" and
the resultant modified protein molecules are termed "de-immunized."
[0058] According to the invention, MHC Class II binding can be removed in
situations where a protein is to be produced in bacteria or in an organism that
does not generate a mammalian glycosylation pattern, such as yeast or insect
cells.
[0059] The invention provides non-computer methods for reducing or
eliminating the number of T-cell epitopes in IL-7 without requiring elaborate
computer simulations or protein three-dimensional structures. In one
embodiment, a method of the invention takes advantage of the fact that a core
segment of nine amino acids interacts with both the MHC class II molecule as
well as the T-cell receptor during antigen presentation. The most N-termina!
amino acid, the "anchor" position, binds to a deep pocket within the MHC class II
molecule. One of the following amino acids is typically present at the anchor
position, which is important for binding to an MHC class II molecule: leucine,
valine, isoleucine, methionine, phenylalanine, tyrosine and tryptophan. According
to the invention, an additional 2 to 3 amino acids adjacent to the core 9 amino
acids also affect the interaction with MHC molecules.
[0060] A general method of the invention includes mutating any leucines,
valines, isoleucines, methionines, phenylalanines, tyrosines or tryptophans that
occur in IL-7. In one embodiment, one or more of these amino acids in a
candidate T-cell epitope is mutated to a threonine, an alanine or a proline,

WO 2006/061219 PCT/EP2005/013145
10
thereby retaining some of the hydrophobic nature of the amino acid that is
replaced. In further embodiments of the invention, one or more of the above-
mentioned amino acids is deleted from a candidate T-cell epitope or potential T-
cell epitope, or replaced with an appropriate amino acid analog. According to the
invention, if an amino acid is deleted to destroy a potential T-cell epitope, care
should be taken not to generate a new T-cell epitope that includes amino acids
near the deletion.
[0061] Thus, the invention provides nucleic acid sequences and proteins that
are useful in construction of less immunogenic IL-7 proteins. Specifically, the
invention provides proteins with mutations of leucines, valines, isoleucines,
methionines, phenylalanines, tyrosines, or tryptophans. Any aliphatic or aromatic
residue (leucine, valine, isoleucine, methionine, phenylaianine, tryptophan or
tyrosine) presents a high risk of creating an MHC binding peptide with the amino
acid in the first position (anchor position) that binds the pocket of the MHC
molecule. Therefore, substitution of any of the above-mentioned annino acids,
with an amino acid that is not one of the above-mentioned amino acids, or with
alanine, praline, orthreonine, will remove a candidate T-cell epitope.
[0062] The proteins can be human proteins with sequences that generally
correspond to sequences found in the human body. The invention also provides
nucleic acid sequences encoding such proteins. The nucleic acid sequences for
this aspect of the invention may exist as plasmids, PCR-generated fragments, or
nucleic acids produced by chemicai synthesis.
[0063] As used herein, the term "interleukin-7" or "IL-7" means IL-7
polypeptides and derivatives and analogs thereof having substantial amino acid
sequence identity to wild-type mature mammalian IL-7. For example, IL-7 refers
to an amino acid sequence of a recombinant or non-recombinant polypeptide
having an amino acid sequence of: i) a native or naturally-occurring allelic variant
of an IL-7 polypeptide, ii) a biologically active fragment of an IL-7 polypeptide, iii)
a biologically active polypeptide analog of an IL-7 polypeptide, or iv) a biologically
active variant of an IL-7 polypeptide.
[0064] IL-7 polypeptides modified according to the invention can be derived
from any species, e.g., human, cow or sheep. IL-7 nucleic acid and amino acid
sequences are well known in the art. For example, the human IL-7 amino acid
sequence has a Genbank accession number of NM 000880 (SEQ ID NO:1) and

WO 2006/061219 PCT/EP2005/013145
11
is shown in Figure 1; the mouse IL-7 amino acid sequence has a Genbank
accession number of NM 008371; the rat IL-7 amino acid sequence has a
Genbank accession number of AF 367210; the cow IL-7 amino acid sequence
has a Genbank accession number of NM 173924 (SEQ ID NO:2) and is shown in
Figure 2; and the sheep IL-7 amino acid sequence has a Genbank accession
number of U10089 (SEQ ID NO:3) and is shown in Figure 3. The signal
sequence for each of the polypeptide species is shown in bold in each of the
figures and is typically not included where the IL-7 portion is fused C-terminal to
the carrier protein.
[0065] In addition, in Figure 35, an alignment of various mammalian IL-7
sequences is shown. IL-7 from non-human primates is generally more than 90%
identical to human IL-7. Although the murine IL-7 sequence is the most divergent
from the human IL-7 sequence, with less than 70% identity, it is nevertheless
capable or activating the human IL-7 receptor. Therefore, IL-7 moieties from a
range of species are particularly useful in accordance with the teachings of the
invention.
[0066] A "variant" of an IL-7 protein is defined as an IL-7 amino acid
sequence that is altered by one or more amino acids as compared to wild-type IL-
7. The variant can have "conservative" changes, wherein a substituted amino
acid has similar structural or chemical properties, e.g., replacement of leucine
with isoleucine. More rarely, a variant can have "nonconservative" changes, e.g.,
replacement of a glycine with a tryptophan. Similar minor variations can also
include amino acid deletions or insertions, or both.
[0067] Variant IL-7 proteins also include polypeptides that have at least about
70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%,
92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity with
wild-type IL-7. To determine the percent identity of two amino acid sequences or
of two nucleic acids, the sequences are aligned for optimal comparison purposes
(e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid
sequence for optimal alignment with a second amino acid or nucleic acid
sequence). The percent identity between the two sequences is a function of the
number of identical positions shared by the sequences (i.e.., % homology = (# of
identical positions/total # of positions)times 100). The determination of percent
homology between two sequences can be accomplished using a mathematical

WO 2006/061219 PCT/EP2005/013145
12
algorithm. A non-limiting example of a mathematical algorithm utilized for the
comparison of two sequences is the algorithm of Karlin and Altschul, (1990) Proc.
Natl. Acad. Sci. USA, 87:2264-68, modified as in Karlin and Altschul, (1993) Proc.
Natl. Acad. Sci. USA, 90:5873-77. Such an algorithm is incorporated into the
NBLAST and XBLAST programs of Altschul et a/., (1990) J. Mol. Biol., 215:403-
10. BLAST nucleotide searches can be performed with the NBLAST program,
score=100, wordlength=12. BLAST protein searches can be performed with the
XBLAST program, score=50, wordlength=3. To obtain gapped alignments for
comparison purposes, Gapped BLAST can be utilized as described in Altschul et
al., (1997) Nucleic Acids Research, 25(17):3389-3402. When utilizing BLAST
and Gapped BLAST programs, the default parameters of the respective programs
(e.g., XBLAST and NBLAST) can be used.
[0068] Furthermore, the invention also includes IL-7 fusion proteins wherein
the IL-7 moiety contains a deletion and which retain comparable activity
compared to the corresponding unmodified IL-7 fusion proteins. For example, the
invention provides a form of lg-IL-7 or IL-7 in which the IL-7 moiety contains an
eighteen amino acid internal deletion corresponding to the sequence
VKGRKPAALGEAQPTKSL (SEQ ID NO:25), of wild-type human IL-7. (See SEQ
ID NO:1). In addition, the invention provides an active form of IL-7 wherein
Lys150 is deleted. Glu151and His152 may also be deleted in conjunction with
Lys150 while still leaving an active form of IL-7.
[0069] Throughout this application, the positions of amino acid residues in the
IL-7 sequence are given with reference to the mature human IL-7 protein. For
example, the cysteine in the N-terminal sequence MDCDIEGK...(SEQ ID NO:22)
of bacterially produced human IL-7 protein, which includes a start methionine, is
still referred to as Cys2.
Modifying IL-7 Proteins
[0070] One aspect of the invention derives from the insight that IL-7 produced
by bacterial expression will not contain post-translational modifications that are
characteristic of eukaryotes, such as mammals. For example, IL-7 contains three
predicted N-linked glycosylation sites at positions 70, 91, and 116. In an Fc-IL-7
fusion protein expressed in mammalian cells, the asparagines at positions 70 and
91 are glycosylated, while the asparagine at position 116 is not. It is likely that IL-
7 endogenously produced in the human body is also N-glycosylated, at least at

WO 2006/061219 PCT/EP2005/013145
13
positions 70 and 91, and possibly at position 116. These N-linked glycosylations
are not present in bacterially produced lL-7, and represent sequences that might
be recognized by the human immune system as "non-self," i.e., not normally
present in the human body. As such, the invention encompasses deimmunizing
these potential epitope regions on IL-7 to reduce the immunogenicity of IL-7 and
related proteins.
[0071] According to the invention, T-cell epitopes are present in IL-7 that
include positions 70 and 91, as described in Table 1. The epitopes shown in
Table 1 are defined in terms of a minimal 9-mer peptide, with the strong MHC
Class II anchor residue in the first position.
Table 1

[0072] According to the invention, one method for reducing the
immunogenicity of bacterially produced IL-7 is to introduce one or more of the
following mutations: Leu63Ala, Leu63Val, Leu63Pro, Leu63Thr, Lys68Asp,
Met69Asp, Lys68Glu, Met69Glu, He88Thr, lle88Ala, He88Val, and Val96Gly.
Other mutations may be introduced at positions 63, 68, 69, 88 and/or 94. Some
mutations are particularly useful in combination, such as the pairs Lys68Asp
coupled with Met69Asp and/or He88Thr coupled with Val96Gly.
[0073] When these mutations are introduced into IL-7 or a fusion protein
comprising IL-7, the resulting mutant protein generally has enough IL-7 biological
activity to be useful as a therapeutic protein. In fact, the biological activity of the
IL-7 moiety is at least 10%, 20%, 50%, 70%, 80%, 90%, 95%, 99% or 100% in
comparison to the biological activity of wild type IL-7. Activity of the IL-7 of the
invention can be tested in an in vitro or in vivo assay. Example 9 shows an assay
for testing biological activity of the IL-7 variants of the invention.
[0074] In addition, the mutations generally allow proper folding of the IL-7
moiety so that a pure protein, largely free of high-molecular weight aggregates

WO 2006/061219 PCT/EP2005/013145
14
and incorrectly disulfide-bonded forms, may be isolated. However, the folding
and biological activity that results from any particular combination should be
tested, for example as illustrated in the Examples, to verify that the desired
activity is obtained.
[0075] According to the invention, an alternative strategy for reducing the
immunogenicity of bacterially produced IL-7 is to alter Asn70 and Asn91 to
aspartic acid. Without wishing to be bound by theory, the mutation of Asn70 and
Asn91 to aspartic acid may be useful for the following reasons.
[0076] The immunogenicity of an exogenously administered therapeutic
protein is mediated, in part, through the presentation of T-cell epitopes derived
from the therapeutic protein. Such presentation is thought to occur through the
following mechanism. A therapeutic protein is taken up by an antigen-presenting
cell (APC), such as a dendritic cell, macrophage, or B-cell by endocytosis. The
protein is transported into a series of vesicles termed endosomes, including the
early, middle and late endosomes. In these vesicles, the environment becomes
progressively more harsh and less favorable for extracellular, disulfide-bonded
proteins that may be stably folded at neutral pH. Proteases termed cathepsins
degrade internalized proteins into small peptides. A proportion of these protein
fragments then become bound by MHC Class I! proteins which transport the
fragments to the cell surface as MHC Class ll/peptide complexes. Such
complexes are recognized by T-cell receptors on CD4+ T-cells.
[0077] In the case of peptides deriving from foreign proteins, presentation of
an MHC Class ll/peptide complex may stimulate an immune response. However,
in the case of peptides deriving from self proteins, there are multiple mechanisms
by which T-cells recognizing MHC Class ll/peptide complexes are deleted or
prevented from activating an immune response.
[0078] With the preceding two paragraphs taken as background, it is
important to consider how an N-glycosylated protein would be processed in the
endosome. Such a protein could be degraded into N-linked oligosaccharide-
containing peptides that could bind to MHC Class II molecules. According to an
insight of the invention, the endosome also contains an endoglycosidase that
sometimes removes the oligosaccharide from asparagine, and in doing so,
converts the asparagine into aspartic acid. Thus, self protein sequences that
contain asparagine-iinked oiigosaccharides may be presented by MHC Class II

WO 2006/061219 PCT/EP2005/013145
15
as peptides containing the asparagine linked to an oligosaccharide, or as
corresponding peptides containing aspartic acid instead of asparagine.
[0079] As part of the invention, it is also recognized that this strategy for
reducing the immunogenicity of mammalian proteins that are expressed in
bacteria may be applied in a general manner. Specifically, the substitution of
aspartic acid for asparagine at a site of N-linked glycosylation generally has the
effect of reducing the immunogenicity of a mammalian protein that is expressed
in a prokaryote.
[0080] The invention contains additional mutations that reduce the
immunogenicity of IL-7 and IL-7-containing fusion proteins when expressed in
either bacterial or mammalian cells. These mutations include those listed in
Table 2 below. An IL-7 or IL-7 containing fusion protein may comprise one or
more of these mutations. For example, in one embodiment, IL-7 is modified to
incorporate one or more of L24D, M54A, F57K, A60S, R61E, M147K, and T149S,
with K150, E151 and H152 being deleted. In another embodiment, IL-7 is
modified to incorporate one or more of D76N, L77D, T87Q, I88T, V96G, L119S,
M147K, and T149S, with K150, E151 and H152 being deleted. In a further
embodiment, IL-7 can be modified to incorporate one or more of L24D, I30T,
F39P, M54A, F57K, A60S, R61E, M68D, N69D, L77D, T87Q, I88T, V96G,
L119S, L128A, M147K, and T149S, with K150, E151 and H152 being deleted.
[0081] In another embodiment, an IL-7 molecule or an IL-7 containing fusion
protein may include mutations to one or more of residues 39, 57, 77 and/or 128 of
IL-7. For example, IL-7 in one embodiment, includes a mutation at residue 39. in
another embodiment, IL-7 includes a mutation at residue 57. In a further
embodiment, IL-7 includes mutations at both residues 39 and 57. In yet another
embodiment, IL-7 includes mutations at residues 39, 57 and 128, while in another
embodiment, IL-7 includes mutations at residues 39, 57 and 77. In yet another
embodiment, IL-7 includes mutations at residues 39, 57,77 and 128. In a further
embodiment, the phenylalanine residue at position 39 is replaced by a proline
residue (F39P). In another embodiment, the phenylalanine residue at 57 is
replaced by an asparagines residue (F57N). In another embodiment, the leucine
residue at position 77 is replaced by aspartic acid (L77D). In yet another
embodiment, the leucine residue at position 128 is replaced by serine (L128S).

WO 2006/061219 PCT/EP2005/013145
16

Verification of the Reduced Immunogenicity of the Proteins of the Invention
[0082] To check that a mutation of the invention has indeed resulted in
reduced immunogenicity, standard experimental tests, which are well known in
the art, may be employed. For example, a T-cell stimulation assay may be used
(e.g. Jones et ai, (2004), J. Interferon Cvtokine Res., 24:560). In such an assay,
human peripheral blood mononuclear cells (PBMCs) are obtained and cultured
according to standard conditions. After an optional pre-stimulation, a peptide
corresponding to a potential MHC Class II epitope is added to the culture of
PBMCs; the PBMCs are further incubated, and at a later time tritiated thymidine
is added. The peptide may be a minima! 9-mer, or may have about 10 to 15 or
more amino acids. After further incubation of the cells, incorporation of tritiated
thymidine into DNA is then measured by standard techniques.
[0083] The T-ce!l stimulation assay is thought to work by the following
mechanisms. First, if a peptide is used as a stimulator, the peptide must first bind
to an MHC Class II molecule present on a cell among the PBMCs. Second, the

WO 2006/061219 PCT/EP2005/013145
17
MHC Class ll/peptide complex must interact productively with a T-celi receptor on
a CD4+ T-cell. If the test peptide is unable to bind sufficiently tightly to an MHC
Class II molecule, no signal will result. If the peptide is able to bind an MHC
Class II molecule and there are T-cells expressing an appropriately rearranged T-
cell receptor capable of recognizing a particular MHC Class ll/peptide complex, a
signal should result. However, if such T-cells have been deleted as a result of a
negative selection process, no signal will result. These mechanisms are
considered relevant to the immunogenicity of a protein sequence, as inferred
from the stimulation or lack of stimulation by a given peptide.
[0084] If recognizing T-cells are present in very low numbers in the PBMC
population for stochastic reasons relating to failure of an appropriate T-cell
receptor to take place or proliferation of other, unrelated T-cells followed by
homeostasis of the T-cell population, there may also be no signal even though a
signal is expected. Thus, false negative results may occur. Based on these
considerations, it is important to use a large number of different sources of
PBMCs and to test these samples independently. It is also generally useful to
test PBMCs from an ethnically diverse set of humans, and to determine the MHC
Class II alleles present in each PBMC population.
[0085] The standard T-cell assay has the disadvantage that the tritium
incorporation signal is often only two-fold greater than the background
incorporation. The proteins and peptides of the invention may also be tested in a
modified T-cell assay in which, for example, purified CD4+ T-cells and purified
dendritic cells are co-cultured in the presence of the test peptide, followed by
exposure to tritiated thymidine and then assayed for tritiated thymidine
incorporation. This second assay has the advantage that tritiated thymidine
incorporation into irrelevant cells, such as CD8+ T-cells, is essentially eliminated
and background is thus reduced.
[0086] A third assay involves the testing of a candidate protein with reduced
immunogenicity in an animal such as a primate. Such an assay would generally
involve the testing of an entire IL-7 protein or IL-7-containing fusion protein in
which the IL-7 moiety had been designed by testing individual component
peptides for potential immunogenicity in a cell-based assay such as one
described above. Once such a candidate IL-7-containing protein is designed and
expressed, the protein is tested for immunogenicity by injection into an animal.

WO 2006/061219 PCT/EP2005/013145
18
[0087] Injection of the modified IL-7-containing protein is generally performed
in the same manner as the anticipated route of delivery during therapeutic use in
humans. For example, intradermal, subcutaneous, intramuscular, intraperitoneal
injection or intravenous infusion may be used. If more than one administration is
used, the administrations may be by different routes.
[0088] For immunogenicity testing purposes, it may be useful to coadminister
an adjuvant to increase the signal and minimize the number of animals that need
to be used. If an adjuvant is used, it is possible to use an adjuvant lacking a
protein component, such as non-coding DNA with unmethylated CpG
dinucleotides, bacterial lipid A, N-formyl methionine, or other bacterial non-protein
components. Without wishing to be bound by theory, the rationale for avoiding
protein-containing adjuvants is that other proteins may provide T-cell epitopes
that will ultimately contribute to an antibody response against the candidate
protein.
[0089] After one or more administrations of the candidate IL-7-containing
protein, the presence of anti-IL-7 antibodies is tested according to standard
techniques, such as the ELISA method. It is found that the altered IL-7-
containing molecules of the invention induce antibody formation less frequently,
and to a lesser extent, than corresponding molecules containing normal human
IL-7.
[0090] Many of the proteins of the invention alter surface residues of IL-7. It
is contemplated that the proteins of the invention, while being less immunogenic
than corresponding proteins containing human IL-7, may still occasionally induce
formation of antibodies. Because the B-cell epitopes of the proteins of the
invention are generally different from those of unmodified IL-7, antibodies to the
proteins of the invention will generally not cross-react with endogenous IL-7, and
formation of antibodies to the proteins of the invention will have no long-term
consequences for the health of the patient.
Fc-IL-7 Fusion Proteins
[0091] A key aspect of the invention is that IL-7 modified according to the
invention may be fused to a carrier protein to create a fusion protein. In one
embodiment, the carrier protein is disposed towards the N-terminus of the fusion
protein and the IL-7 is disposed towards the C-terminus. In another embodiment,

WO 2006/061219 PCT/EP2005/013145
19
the IL-7 is disposed towards the N-terminus of the fusion protein and the carrier
protein is disposed towards the C-terminus.
[0092] The carrier protein can be any polypeptide covalently fused to the IL-7
protein. In one embodiment, the carrier protein is albumin, for example, human
serum albumin. The albumin moiety may be fused to the C-terminal or N-terminal
end of the IL-7 moiety. In another embodiment, the carrier protein is an
immunoglobulin (Ig) moiety, such as an ig heavy chain. The Ig chain may be
derived from IgA, IgD, IgE, IgG, or IgM. According to the invention, the Ig moiety
may be an intact antibody and may direct the IL-7 fusion protein to specific target
sites in the body. Fusion proteins making use of antibody targeting are known to
those in the art.
[0093] In one embodiment, the Ig moiety comprises an Fc region. As used
herein, "Fc portion" encompasses domains derived from the constant region of an
immunoglobulin, such as a human immunoglobulin, including a fragment, analog,
variant, mutant or derivative of the constant region. Suitable immunoglobulins
include lgG1, lgG2, lgG3, lgG4, and other classes. The constant region of an
immunoglobulin is defined as a naturally-occurring or synthetically-produced
polypeptide homologous to the immunoglobulin C-terminal region, and can
include a hinge, a CH2 domain, a CH3 domain, or a CH4 domain, separately or in
any combination. In the present invention, the Fc portion typically includes at
least a CH2 domain. For example, the Fc portion can include hinge-CH2-CH3.
Alternatively, the Fc portion can include all or a portion of the hinge region, the
CH2 domain and/or the CH3 domain. Methods for making Fc-IL-7 fusion proteins
are disclosed in U.S. Provisional Patent Application No. 60/533,406.
[0094] The constant region of an immunoglobulin is responsible for many
important antibody functions including Fc receptor (FcR) binding and complement
fixation. There are five major classes of heavy chain constant region, classified
as IgA, IgG, IgD, IgE, and IgM. For example, IgG is separated into four y
subclasses: y 1, y2, y3, and yA, also known as IgGl, lgG2, lgG3, and lgG4,
respectively.
[0095] IgG molecules interact with multiple classes of cellular receptors,
including three classes of FCK receptors (Fc y R) specific for the IgG class of
antibody, namely Fcj/RI, FcyRII, and Fc^RIII. The important sequences for the
binding of IgG to the FcyR receptors have been reported to be located in the CH2

WO 2006/061219 PCT/EP2005/013145
20
and CH3 domains. The serum half-life of an antibody is influenced by the ability
of that antibody to bind to an Fc receptor (FcR). Similarly, the serum half-life of
immunoglobulin fusion proteins is also influenced by the ability to bind to such
receptors (Gillies et a/., (1999) Cancer Res. 59:2159-66). Compared to those of
lgG1, CH2 and CH3 domains of lgG2 and lgG4 have biochemically undetectable
or reduced binding affinity to Fc receptors. It has been reported that
immunoglobulin fusion proteins containing CH2 and CH3 domains of lgG2 or
lgG4 had longer serum half-lives compared to the corresponding fusion proteins
containing CH2 and CH3 domains of lgG1 (U.S. Patent No. 5,541,087; Lo et ai,
(1998) Protein Engineering. 11:495-500). Accordingly, in certain embodiments of
the invention, CH2 and CH3 domains are derived from an antibody isotype with
reduced receptor binding affinity and effector functions, such as, for example,
lgG2 or lgG4.
[0096] The hinge region is normally located C-terminal to the CH1 domain of
the heavy chain constant region. In the IgG isotypes, disulfide bonds typically
occur within this hinge region, permitting the final tetrameric molecule to form.
This region is dominated by pralines, serines and threonines. When included in
the present invention, the hinge region is typically at least homologous to the
naturally-occurring immunoglobulin region that includes the cysteine residues to
form disulfide bonds linking the two Fc moieties. Representative sequences of
hinge regions for human and mouse immunoglobulins are known in the art and
can be found in Borrebaeck, C. A. K., ed., (1992) Antibody Engineering, A
Practical Guide, W. H. Freeman and Co. Suitable hinge regions for the present
invention can be derived from lgG1, lgG2, lgG3, lgG4, and other immunoglobulin
classes.
[0097] The lgG1 hinge region has three cysteines, two of which are involved
in disulfide bonds between the two heavy chains of the immunoglobulin. These
same cysteines permit efficient and consistent disulfide bonding formation of an
Fc portion. Therefore, a hinge region of the present invention in one embodiment
is derived from lgG1, such as human lgG1. When the lgG1 hinge is used, the
first cysteine can be mutated to another amino acid, such as serine.
[0098] The lgG2 isotype hinge region has four disulfide bonds that tend to
promote oligomerization and possibly incorrect disulfide bonding during secretion
in recombinant systems. A suitable hinge region can be derived from an lgG2

WO 2006/061219 PCT/EP2005/013145
21
hinge. In one embodiment, the first two cysteines of the lgG2 hinge are mutated
to another amino acid.
[0099] The hinge region of lgG4 is known to form interchain disulfide bonds
inefficiently. However, a suitable hinge region for the present invention can be
derived from the lgG4 hinge region, and can contain a mutation that enhances
correct formation of disulfide bonds between heavy chain-derived moieties (Angal
et a/., (1993) Mol. Immunol., 30:105-8).
[00100] In accordance with the present invention, the Fc portion can contain
CH2 and/or CH3 and/or CH4 domains and a hinge region that are derived from
different antibody isotypes, i.e., a hybrid Fc portion. For example, in one
embodiment, the Fc portion contains CH2 and/or CH3 domains derived from
lgG2 or lgG4 and a mutant hinge region derived from lgG1. Alternatively, a
mutant hinge region from another igG subclass is used in a hybrid Fc portion.
For example, a mutant form of the lgG4 hinge that allows efficient disulfide
bonding between the two heavy chains can be used. A mutant hinge can also be
derived from an lgG2 hinge in which the first two cysteines are each mutated to
another amino acid. Such hybrid Fc portions facilitate high-level expression and
improve the correct assembly of the Fc-IL-7 fusion proteins. Assembly of such
hybrid Fc portions is known in the art and has been described in U.S. Published
Patent Application No. 2003-0044423.
[00101] In some embodiments, the Fc portion contains amino acid
modifications that generally extend the serum half-life of an Fc fusion protein.
Such amino acid modifications include mutations substantially decreasing or
eliminating Fc receptor binding or complement fixing activity. For example, the
glycosylation site within the Fc portion of an immunoglobulin heavy chain can be
removed. In lgG1, the glycosylation site is Asn297 within the amino acid
sequence GIn-Tyr-Asn-Ser (SEQ ID NO:26). In other immunoglobulin isotypes,
the glycosylation site corresponds to Asn297 of lgG1. For example, in lgG2 and
lgG4, the glycosylation site is the asparagine within the amino acid sequence
Gln-Phe-Asn-Ser (SEQ ID NO:28). Accordingly, a mutation of Asn297 of lgG1
removes the glycosylation site in an Fc portion derived from lgG1. In one
embodiment, Asn297 is replaced with Gin. In other embodiments, the tyrosine
within the amino acid sequence GIn-Tyr-Asn-Ser (SEQ ID NO:26) is further
mutated to eliminate a potential non-self T-cell epitope resulting from asparagine

WO 2006/061219 PCT/EP2005/013145
22
mutation. For example, the amino acid sequence Gln-Tyr-Asn-Ser (SEQ ID
NO:26) within an lgG1 heavy chain can be replaced with a Gln-Ala-Gln-Ser (SEQ
ID NO:27) amino acid sequence.
[00102] Similarly, in lgG2 or lgG4, a mutation of asparagine within the amino
acid sequence Gln-Phe-Asn-Ser (SEQ ID NO:28) removes the glycosylation site
in an Fc portion derived from lgG2 or lgG4 heavy chain. In one embodiment, the
asparagine is replaced with a glutamine. In other embodiments, the
phenylalanine within the amino acid sequence Gln-Phe-Asn-Ser (SEQ ID NO:28)
is further mutated to eliminate a potential non-self T-cell epitope resulting from
asparagine mutation. For example, the amino acid sequence Gln-Phe-Asn-Ser
(SEQ ID NO:28) within an lgG2 or lgG4 heavy chain can be replaced with a Gln-
Ala-Gln-Ser (SEQ ID NO:27) amino acid sequence. Other mutations that are
useful in reducing Fc receptor binding are disclosed in U.S. Patent Application
No. 09/256,156.
[00103] It has also been observed that alteration of amino acids near the
junction of the Fc portion and the non-Fc portion can dramatically increase the
serum half-life of the Fc fusion protein. (U.S. Published Patent Application No.
2002-0147311). Accordingly, the junction region of an Fc-IL-7 or IL-7-Fc fusion
protein of the present invention can contain alterations that, relative to the
naturally-occurring sequences of an immunoglobulin heavy chain and IL-7, lie
within about 10 amino acids of the junction point. These amino acid changes can
cause an increase in hydrophobicity by, for example, changing the C-terminal
lysine of the Fc portion to a hydrophobic amino acid such as alanine or leucine.
(See e.g. SEQ ID NO:34). In yet another embodiment of the invention, the C-
terminal lysine and preceding glycine of the Fc portion is deleted. (See e.g. SEQ
IDNO:35).
[00104] In other embodiments, the Fc portion contains amino acid alterations
of the Leu-Ser-Leu-Ser segment near the C-terminus of the Fc portion of an
immunoglobulin heavy chain. The amino acid substitutions of the Leu-Ser-Leu-
Ser (SEQ ID NO:29) segment eliminate potential junctional T-cell epitopes. In
one embodiment, the Leu-Ser-Leu-Ser (SEQ ID NO:29) amino acid sequence
near the C-terminus of the Fc portion is replaced with an Ala-Thr-Ala-Thr (SEQ ID
NO:30) amino acid sequence. In other embodiments, the amino acids within the
Leu-Ser-Leu-Ser (SEQ ID NO:29) segment are replaced with other amino acids

WO 2006/061219 PCT/EP2005/013145
23
such as glycine or praline. Detailed methods of generating amino acid
substitutions of the Leu-Ser-Leu-Ser (SEQ ID NO:29) segment near the C-
terminus of an lgG1, lgG2, lgG3, lgG4, or other immunoglobulin class molecules,
as well as other exemplary modifications for altering junctional T-cell epitopes,
have been described in U.S. Published Patent Application No. 2003-0166877.
[00105] In one embodiment, a spacer or linker peptide is inserted between the
carrier protein and the IL-7 protein. The spacer or linker peptide can be non-
charged or non-polar or hydrophobic. The length of a spacer or linker peptide is
between 1 and about 100 amino acids, or between 1 and about 50 amino acids,
or between 1 and about 25 amino acids, or between 1 and about 15 amino acids.
In one embodiment, the spacer contains a sequence (G4S)n, where n is less than
10. In another embodiment, the linker sequence is GGGGSGGGG (SEQ ID
NO:67). In yet another embodiment, the spacer contains a motif that is
recognized as an N-linked glycosylation site. In another embodiment of the
invention, the carrier protein and the IL-7 fusion protein are joined via a spacer or
linker peptide. In an alternative embodiment of the invention, the carrier protein
and IL-7 fusion protein are separated by a synthetic spacer, for example a PNA
spacer. The spacer can be non-charged, or non-polar or hydrophobic.
Production of IL-7 fusion proteins
[00106] Fusion proteins containing IL-7 modified according to the teachings of
the invention can be synthesized by the non-limiting methods described herein.
Assays useful for testing pharmacokinetic activities of fusion proteins containing
IL-7 modified according to the invention in in vivo animal models are also
described herein.
[00107] The IL-7 fusion proteins of the invention can be produced using
recombinant expression vectors known in the art. The term "expression vector"
refers to a replicable DNA construct used to express DNA which encodes the
desired IL-7 fusion protein and which includes a transcriptional unit comprising an
assembly of (1) genetic element(s) having a regulatory role in gene expression,
for example, promoters, operators, or enhancers, operatively linked to (2) a DNA
sequence encoding the desired IL-7 fusion protein which is transcribed into
mRNA and translated into protein, and (3) appropriate transcription and
translation initiation and termination sequences. The choice of promoter and
other regulatory elements generally varies according to the intended host cell.

WO 2006/061219 PCT/EP2005/013145
24
[00108] The nucleic acid encoding the IL-7 fusion protein is transfected into a
host cell using recombinant DNA techniques. In the context of the present
invention, the foreign DNA includes a sequence encoding the inventive proteins.
Suitable host cells include prokaryotic, yeast or higher eukaryotic cells. In one
embodiment, the host is a prokaryotic organism.
[00109] The recombinant IL-7 fusion proteins can be expressed in yeast hosts,
such as from Saccharomyces species, such as S. cerevisiae. Yeast of other
genera such as Pichia or Kluyveromyces may also be employed. Yeast vectors
will generally contain an origin of replication from a yeast plasmid or an
autonomously replicating sequence (ARS), a promoter, DNA encoding the IL-7
fusion protein, sequences for polyadenylation and transcription termination and a
selection gene. Suitable promoter sequences in yeast vectors include the
promoters for metallothionein, 3-phosphoglycerate kinase or other glycolytic
enzymes, such as enolase, glyceraldehyde-3-phosphate dehydrogenase,
hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-4-phosphate
isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate
isomerase, phosphoglucose isomerase and glucokinase.
[00110] Various mammalian or insect cell culture systems can be employed to
express recombinant protein. Baculovirus systems for production of proteins in
insect cells are well known in the art. Examples of suitable mammalian host cell
lines include NS/0 cells, L cells, C127, 3T3, Chinese hamster ovary (CHO), HeLa,
and BHK cell lines. Additional suitable mammalian host cells include CV-1 cells
(ATCC CCL70) and COS-7 cells both derived from monkey kidney. Another
suitable monkey kidney cell line, CV-1/EBNA, was derived by transfection of the
CV-1 cell line with a gene encoding Epstein-Barr virus nuclear antigen-1 (EBNA-
1) and with a vector containing CMV regulatory sequences (McMahan etal.,
(1991), EMBQ J., 10:2821). The EBNA-1 gene allows for episomal replication of
expression vectors, such as HAV-EO or pDC406, that contain the EBV origin of
replication.
[00111] Mammalian expression vectors may comprise non-transcribed
elements such as an origin of replication, a suitable promoter and enhancer
linked to the gene to be expressed, and other 5' or 3' flanking nontranscribed
sequences, and 5! or 3' nontranslated sequences, such as necessary ribosome
binding sites, a poly-adenylation site, splice donor and acceptor sites, and

WO 2006/061219 PCT/EP2005/013145
25
transcriptional termination sequences. Commonly used promoters and
enhancers are derived from Polyoma, Adenovirus 2, Simian Virus 40 (SV40), and
human cytomegalovirus. DNA sequences derived from the SV40 viral genome,
for example, SV40 origin, early and late promoter, enhancer, splice, and
polyadenylation sites may be used to provide the other genetic elements required
for expression of a heterologous DNA sequence.
[00112] When secretion of the IL-7 fusion protein from the host cell is desired,
the expression vector may comprise DNA encoding a signal or leader peptide. In
the present invention the native signal sequence of IL-7 can be used, or
alternatively, a heterologous signal sequence may be added, such as the signal
sequence from interleukin-4.
[00113] The present invention also provides a process for preparing the
recombinant proteins of the present invention including culturing a host cell
transformed with an expression vector comprising a DNA sequence that encodes
the IL-7 fusion protein under conditions that promote expression. The desired
protein is then purified from culture media or cell extracts. For example,
supernatants from expression systems that secrete recombinant protein into the
culture medium can be first concentrated using a commercially available protein
concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
Following the concentration step, the concentrate can be applied to a suitable
purification matrix, as known in the art.
[00114] An "isolated" or "purified" IL-7 fusion protein or biologically active
portion thereof is substantially free of cellular material or other contaminating
proteins from the cell or tissue source from which the IL-7 fusion protein is
derived, or substantially free from chemical precursors or other chemicals when
chemically synthesized. The language "substantially free of cellular material"
includes preparations of IL-7 fusion protein in which the protein is separated from
cellular components of the cells from which it is isolated or recombinantly
produced. In one embodiment, the language "substantially free of cellular
material" includes preparations of IL-7 fusion protein having less than about 30%
(by dry weight) of non-lL-7 fusion protein (also referred to herein as a
"contaminating protein"), less than about 20% of non-IL-7 fusion protein, less than
about 10% of non-IL-7 fusion protein, or less than about 5% non-IL-7 fusion
protein. When the IL-7 fusion protein or biologically active portion thereof is

WO 2006/061219 PCT/EP2005/013145
26
purified from a recombinant source, it is, in one embodiment, substantially free of
culture medium, i.e.., culture medium represents less than about 20%, less than
about 10%, or less than about 5% of the volume of the protein preparation.
[00115] The term "substantially pure lg-IL-7 fusion protein" or "substantially
pure IL-7 fusion protein" refers to a preparation in which the IL-7 comprising
fusion protein constitutes at least 60%, 70%, 80%, 90%, 95% or 99% of the
proteins in the preparation.
Methods of Treatment Using IL-7 Proteins
[00116] The IL-7 proteins, including fusion proteins, of the invention are useful
in treating immune deficiencies and in accelerating the natural reconstitution of
the immune system that occurs, for example, after diseases or treatments that
are immunosuppressive in nature. For example, IL-7 proteins can be used to
treat viral infections, immune disorders, and to enhance the growth (including
proliferation) of specific cell types. Moreover, the IL-7 proteins can be in the
treatment of cancers such as bladder cancer, lung cancer, brain cancer, breast
cancer, skin cancer, and prostate cancer. In one example, it is useful to treat
patients who have undergone one or more cycles of chemotherapy with IL-7
proteins as described above to help their immune cells replenish. Alternatively, it
is also useful to administer the IL-7 proteins described above to patients with HIV,
the elderly, patients receiving a transplant or other patients with suppressed
immune system function.
Administration
[00117] Both the IL-7 and IL-7 fusion proteins of the invention can be
incorporated into a pharmaceutical composition suitable for administration. Such
compositions typically comprise IL-7 or an IL-7 fusion protein and a
pharmaceutically-acceptable carrier. As used herein the language
"pharmaceutically-acceptable carrier" is intended to include any and all solvents,
dispersion media, coatings, antibacterial and antifungal agents, isotonic and
absorption delaying agents, and the like, compatible with pharmaceutical
administration. The use of such media and agents for pharmaceutically active
substances is well known in the art.
[0118] A pharmaceutical composition of the invention is formulated to be compatible
with its intended route of administration. Examples of routes of administration include
parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation),

WO 2006/061219 PCT/EP2005/013145
27
transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions
used for parenteral, intradermal, or subcutaneous application can include the following
components: a sterile diluent such as water for injection, saline solution, fixed oils,
polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial
agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or
sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as
acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium
chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or
sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable
syringes or multiple dose vials made of glass or plastic.
[0119] Medicaments that contain the IL-7 proteins of the invention can have a
concentration of 0.01 to 100% (w/w), though the amount varies according to the
dosage form of the medicaments.
[0120] Administration dose depends on the body weight of the patients, the
seriousness of the disease, and the doctor's opinion. However, it is generally
advisable to administer between about 0.01 to about 10 mg/kg body weight a
day, about 0.02 to about 2 mg/kg/day in case of injection, or about 0.5 mg/kg/day.
The dose can be administered once or several times daily according to the
seriousness of the disease and the doctor's opinion.
[0121] Compositions of the invention are useful when co-administered with
one or more other therapeutic agents, for example, a molecule also known to be
useful to replenish blood cells. For example, the molecule may be erythropoietin
which is known to be used to replenish red blood cells, G-CSF which is used to
replenish neutrophils or GM-CSF which is used to replenish granulocytes and
macrophages.
[0122] Aspects of invention are further illustrated by the following examples.
Examples
Example 1: Identification of T-cell epitopes by computational methods
[0123] According to the invention, epitopes of IL-7 can be modified using
methods for introducing mutations into proteins to modulate their interaction with
the immune system. These methods are similar to those disclosed in U.S.
Published Patent Application No. 2003-0166877. According to the invention,
known methods in the art that can be adapted according to the invention include

WO 2006/061219 PCT/EP2005/013145
28
those described in the prior art (WO 92/10755 and WO 96/40792 (Novo Nordisk),
EP 0519 596 (Merck & Co.), EP 0699 755(Centro de Immunologia Moelcular),
WO 98/52976 and WO 98/59244 (Biovation Ltd.) or related methods.
[0124] Advantageous mutant proteins, however, can be obtained if the
identification of said epitopes is realized by the following method which is
described herewith in detail and applied to IL-7. There are a number of factors
that play important roles in determining the total structure of a protein,
polypeptide or immunoglobulin. First, the peptide bond, i.e.., that bond which
joins the amino acids in the chain together, is a covalent bond. This bond is
planar in structure, essentially a substituted amide. An "amide" is any of a group
of organic compounds containing the grouping -CONH-.

[0125] The planar peptide bond linking Ca of adjacent amino acids may be
represented as depicted below:
[0126] Because the O=C and the C-N atoms lie in a relatively rigid plane, free
rotation does not occur about these axes. Hence, a plane schematically depicted
by the interrupted line is sometimes referred to as an "amide" or "peptide plane"
plane wherein lie the oxygen (O), carbon (C), nitrogen (N), and hydrogen (H)
atoms of the peptide backbone. At opposite corners of this amide plane are
located the Ca atoms. Since there is substantially no rotation about the O=C and
C-N atoms in the peptide or amide plane, a polypeptide chain thus comprises a
series of planar peptide linkages joining the Ca atoms.
[0127] A second factor that pla n important role in defining the total
structure or conformation of a polypeptide or protein is the angle of rotation of
each amide plane about the common Ca linkage. The terms "angle of rotation"
and "torsion angle" are hereinafter regarded as equivalent terms. Assuming that
the O, C, N, and H atoms remain in the amide plane (which is usually a valid
assumption, although there may be some slight deviations from planarity of these
atoms for some conformations), these angles of rotation define the N and R

WO 2006/061219 PCT/EP2005/013145
29
polypeptide's backbone conformation, i.e.., the structure as it exists between
adjacent residues. These two angles are known as and \\i. A set of the angles
i, Ψi where the subscript i represents a particular residue of a polypeptide chain,
thus effectively defines the polypeptide secondary structure. The conventions
used in defining the , Ψ angles, i.e.., the reference points at which the amide
planes form a zero degree angle, and the definition of which angle is , and which
angle is Ψ, for a given polypeptide, are defined in the literature. (See, e.g.,
Ramachandran et a/., (1968), Adv. Prot. Chem. 23:283-437, at pages 285-94).
[0128] The method can be applied to any protein, and is based in part upon
the discovery that in humans the primary Pocket 1 anchor position of MHC Class
II molecule binding grooves has a well designed specificity for particular amino
acid side chains. The specificity of this pocket is determined by the identity of the
amino acid at position 86 of the beta chain of the MHC Class II molecule. This
site is located at the bottom of Pocket 1 and determines the size of the side chain
that can be accommodated by this pocket. Marshall, J. Immunol., (1994),
152:4946-4956. If this residue is a glycine, then all hydrophobic aliphatic and
aromatic amino acids (hydrophobic aliphatics being: valine, leucine, isoleucine,
methionine and aromatics being: phenylalanine, tyrosine and tryptophan) can be
accommodated in the pocket, with a preference being for the aromatic side
chains. If this pocket residue is a valine, then the side chain of this amino acid
protrudes into the pocket and restricts the size of peptide side chains that can be
accommodated such that only hydrophobic aliphatic side chains can be
accommodated. Therefore, in an amino acid residue sequence, wherever an
amino acid with a hydrophobic aliphatic or aromatic side chain is found, there is
the potential for a MHC Class II restricted T-cell epitope. If the side-chain is
hydrophobic aliphatic, however, it is approximately twice as likely to be
associated with a T-cell epitope than an aromatic side chain (assuming an
approximately even distribution of Pocket 1 types throughout the global
population).
[0129] An exemplary computational method profiles the likelihood of peptide
regions of IL-7 to contain T-cell epitopes as follows: (1) The primary sequence of
a peptide segment of predetermined length is scanned, and all hydrophobic
aliphatic and aromatic side chains present are identified. (2) The hydrophobic

WO 2006/061219 PCT/EP2005/013145
30
aliphatic side chains are assigned a value greater than that for the aromatic side
chains; preferably about twice the value assigned to the aromatic side chains,
e.g., a value of,2 for a hydrophobic aliphatic side chain and a value of 1 for an
aromatic side chain. (3) The values determined to be present are summed for
each overlapping amino acid residue segment (window) of predetermined uniform
length within the peptide, and the total value for a particular segment (window) is
assigned to a single amino acid residue at an intermediate position of the
segment (window), preferably to a residue at about the midpoint of the sampled
segment (window). This procedure is repeated for each sampled overlapping
amino acid residue segment (window). Thus, each amino acid residue of the
peptide is assigned a value that relates to the likelihood of a T-cell epitope being
present in that particular segment (window). (4) The values calculated and
assigned as described in Step 3, above, can be plotted against the amino acid
coordinates of the entire amino acid residue sequence being assessed. (5) All
portions of the sequence which have a score of a predetermined value, e.g., a
value of 1, are deemed likely to contain a T-cell epitope and can be modified, if
desired.
[0130] This particular aspect of the present invention provides a general
method by which T-cell epitopes of lL-7 can be described. Modifications to the
peptide in these regions have the potential to modify the MHC Class II binding
characteristics.
[0131] According to another aspect of the present invention, T-cell epitopes
can be predicted with greater accuracy by the use of a more sophisticated
computational method which takes into account the interactions of peptides with
models of MHC Class II alleles.
[0132] The computational prediction of T-cell epitopes present within a
peptide according to this particular aspect contemplates the construction of
models of at least 42 MHC Class II alleles based upon the structures of all known
MHC Class II molecules and a method for the use of these models in the
computational identification of T-cell epitopes, the construction of libraries of
peptide backbones for each model in order to allow for the known variability in
relative peptide backbone alpha carbon (Ca) positions, the construction of
libraries of amino-acid side chain conformations for each backbone dock with
each model for each of the 20 amino-acid alternatives at positions critical for the

WO 2006/061219 PCT/EP2005/013145
31
interaction between peptide and MHC Class II molecule, and the use of these
libraries of backbones and side-chain conformations in conjunction with a scoring
function to select the optimum backbone and side-chain conformation for a
particular peptide docked with a particular MHC Class II molecule and the
derivation of a binding score from this interaction.
[0133] Models of MHC Class II molecules can be derived via homology
modeling from a number of similar structures found in the Brookhaven Protein
Data Bank ("PDB"). These may be made by the use of semi-automatic homology
modeling software (Modeller et al., (1993), J. Mol. Biol., 234:779-815) which
incorporates a simulated annealing function, in conjunction with the CHARMm
force-field for energy minimization (available from Molecular Simulations Inc., San
Diego, Ca.). Alternative modeling methods can be utilized as well.
[0134] Other computational methods which use libraries of experimentally
derived binding data of each amino-acid alternative at each position in the binding
groove for a small set of MHC Class II molecules (Marshall et al., (1995), Biomed.
Pept. Proteins Nucleic Acids, 1(3):157-162) are known, as are yet other
computational methods which use similar experimental binding data in order to
define the binding characteristics of particular types of binding pockets within the
groove, again using a relatively small subset of MHC Class II molecules, and then
'mixing and matching1 pocket types from this pocket library to artificially create
further 'virtual' MHC Class II molecules (Stumiolo et al., (1999), Nat. Biotech,
17(6): 555-561. Both methods suffer the major disadvantage that, due to the
complexity of the assays and the need to synthesize large numbers of peptide
variants, only a small number of MHC Class II molecules can be experimentally
scanned. Therefore the first method can only make predictions for a small
number of MHC Class II molecules. The second method also makes the
assumption that a pocket lined with similar amino-acids in one molecule will have
the same binding characteristics when in the context of a different Class II allele
and suffers further disadvantages in that only those MHC Class II molecules can
be 'virtually' created which contain pockets contained within the pocket library.
Using the modeling approach described herein, the structure of any number and
type of MHC Class II molecules can be deduced, therefore alleles can be
specifically selected to be representative of the global population. In addition, the
number of MHC Class II molecules scanned can be increased by making further

WO 2006/061219 PCT/EP2005/013145
32
models further than having to generate additional data via complex
experimentation.
[0135] The use of a backbone library allows for variation in the positions of
the Ca atoms of the various peptides being scanned when docked with particular
MHC Class II molecules. This is again in contrast to the alternative
computational methods described above which rely on the use of simplified
peptide backbones for scanning amino-acid binding in particular pockets. These
simplified backbones are not likely to be representative of backbone
conformations found in 'real' peptides leading to inaccuracies in prediction of
peptide binding. The present backbone library is created by superposing the
backbones of all peptides bound to MHC Class II molecules found within the
Protein Data Bank and noting the root mean square (RMS) deviation between the
Ca atoms of each of the eleven amino-acids located within the binding groove.
While this library can be derived from a small number of suitable available mouse
and human structures (currently 13), in order to allow for the possibility of even
greater variability, the RMS figure for each C"- a position is increased by 50%.
The average Ca position of each amino-acid is then determined and a sphere
drawn around this point whose radius equals the RMS deviation at that position
plus 50%. This sphere represents all allowed Ca positions.
[0136] Working from the Ca with the least RMS deviation (that of the amino-
acid in Pocket 1 as mentioned above, equivalent to Position 2 of the 11 residues
in the binding groove), the sphere is three-dimensionally gridded, and each vertex
within the grid is then used as a possible location for a Ca of that amino-acid.
The subsequent amide plane, corresponding to the peptide bond to the
subsequent amino-acid is grafted onto each of these Cas and the and Ψ angles
are rotated step-wise at set intervals in order to position the subsequent Ca. If
the subsequent Ca falls within the 'sphere of allowed positions' for this Ca than
the orientation of the dipeptide is accepted, whereas if it falls outside the sphere
then the dipeptide is rejected. This process is then repeated for each of the
subsequent Ca positions, such that the peptide grows from the Pocket 1 Ca
'seed1, until all nine subsequent Cas have been positioned from all possible
permutations of the preceding Cas. The process is then repeated once more for

WO 2006/061219 PCT/EP2005/013145
33
the single Ca preceding pocket 1 to create a library of backbone Ca positions
located within the binding groove.
[0137] The number of backbones generated is dependent upon several
factors: The size of the 'spheres of allowed positions'; the fineness of the
gridding of the 'primary sphere' at the Pocket 1 position; the fineness of the step-
wise rotation of the and Ψ angles used to position subsequent Cas. Using this
process, a large library of backbones can be created. The larger the backbone
library, the more likely it will be that the optimum fit will be found for a particular
peptide within the binding groove of an MHC Class il molecule. Inasmuch as all
backbones will not be suitable for docking with all the models of MHC Class II
molecules due to clashes with amino-acids of the binding domains, for each allele
a subset of the library is created comprising backbones which can be
accommodated by that allele. The use of the backbone library, in conjunction
with the models of MHC Class II molecules creates an exhaustive database
consisting of allowed side chain conformations for each amino-acid in each
position of the binding groove for each MHC Class II molecule docked with each
allowed backbone. This data set is generated using a simple steric overlap
function where a MHC Class II molecule is docked with a backbone and an
amino-acid side chain is grafted onto the backbone at the desired position. Each
of the rotatable bonds of the side chain is rotated step-wise at set intervals and
the resultant positions of the atoms dependent upon that bond noted. The
interaction of the atom with atoms of side-chains of the binding groove is noted
and positions are either accepted or rejected according to the following criteria:
the sum total of the overlap of all atoms so far positioned must not exceed a pre-
determined value. Thus the stringency of the conformational search is a function
of the interval used in the step-wise rotation of the bond and the pre-cletermined
limit for the total overlap. This latter value can be small if it is known that a
particular pocket.is rigid; however, the stringency can be relaxed if the positions
of pocket side-chains are known to be relatively flexible. Thus allowances can be
made to imitate variations in flexibility within pockets of the binding groove. This
conformationa! search is then repeated for every amino-acid at every position of
each backbone when docked with each of the MHC Class II molecules to create
the exhaustive database of side-chain conformations.

WO 2006/061219 PCT/EP2005/013145
34
[0138] A suitable mathematical expression is used to estimate the energy of
binding between models of MHC Class II molecules in conjunction with peptide
ligand conformations which have to be empirically derived by scanning the large
database of backbone/side-chain conformations described above. Thus a protein
is scanned for potential T-cell epitopes by subjecting each possible peptide of
length varying between 9 and 20 amino-acids (although the length is kept
constant for each scan) to the following computations: An MHC Class II molecule
is selected together with a peptide backbone allowed for that molecule and the
side-chains corresponding to the desired peptide sequence are grafted on. Atom
identity and interatomic distance data relating to a particular side-chain at a
particular position on the backbone are collected for each allowed conformation
of that amino-acid (obtained from the database described above). This is
repeated for each side-chain along the backbone and peptide scores derived
using a scoring function. The best score for that backbone is retained and the
process repeated for each allowed backbone for the selected model. The scores
from all allowed backbones are compared and the highest score is deemed to be
the peptide score for the desired peptide in that MHC Class II model. This
process is then repeated for each model with every possible peptide derived from
the protein being scanned, and the scores for peptides versus models are
displayed.
[0139] In the context of the present invention, each ligand presented for the
binding affinity calculation is an amino-acid segment selected from a peptide or
protein as discussed above. Thus, the ligand is a selected stretch of amino acids
about 9 to 20 amino acids in length derived from a peptide, polypeptide or protein
of known sequence. The terms "amino acids" and "residues" are hereinafter
regarded as equivalent terms. The ligand, in the form of the consecutive amino
acids of the peptide to be examined grafted onto a backbone from the backbone
library, is positioned in the binding cleft of an MHC Class II molecule from the
MHC Class II molecule mode! library via the coordinates of the C"-a atoms of the
peptide backbone and an allowed conformation for each side-chain is selected
from the database of allowed conformations. The relevant atom identities and
interatomic distances are also retrieved from this database and used to calculate
the peptide binding score. Ligands with a high binding affinity for the MHC Class
II binding pocket are flagged as candidates for site-directed mutagenesis. Amino-

WO 2006/061219 PCT/EP2005/013145
35
acid substitutions are made in the flagged ligand (and hence in the protein of
interest) which is then retested using the scoring function in order to determine
changes which reduce the binding affinity below a predetermined threshold value.
These changes can then be incorporated into the protein of interest to remove T-
cell epitopes.
[0140] Binding between the peptide ligand and the binding groove of MHC
Class II molecules involves non-covalent interactions including, but not limited to:
hydrogen bonds, electrostatic interactions, hydrophobic (lipophilic) interactions
and van der Waal's interactions. These are included in the peptide scoring
function as described in detail below. It should be understood that a hydrogen
bond is a non-covalent bond which can be formed between polar or charged
groups and consists of a hydrogen atom shared by two other atoms. The
hydrogen of the hydrogen donor has a positive charge where the hydrogen
acceptor has a partial negative charge. For the purposes of peptide/protein
interactions, hydrogen bond donors may be either nitrogens with hydrogen
attached or hydrogens attached to oxygen or nitrogen. Hydrogen bond acceptor
atoms may be oxygens not attached to hydrogen, nitrogens with no hydrogens
attached and one or two connections, or sulphurs with only one connection.
Certain atoms, such as oxygens attached to hydrogens or imine nitrogens {e.g.
C=NH) may be both hydrogen acceptors or donors. Hydrogen bond energies
range from 3 to 7 Kcal/mol and are much stronger than van der Waal's bonds, but
weaker than covalent bonds. Hydrogen bonds are also highly directional and are
at their strongest when the donor atom, hydrogen atom and acceptor atom are
co-linear. Electrostatic bonds are formed between oppositely charged ion pairs
and the strength of the interaction is inversely proportional to the square of the
distance between the atoms according to Coulomb's law. The optimal distance
between ion pairs is about 2.8A. In protein/peptide interactions,, electrostatic
bonds may be formed between arginine, histidine or lysine and aspartate or
glutamate. The strength of the bond will depend upon the pKa of the ionizing
group and the dielectric constant of the medium although they are approximately
similar in strength to hydrogen bonds.
[0141] Lipophilic interactions are favorable hydrophobic-hydrophobic contacts
that occur between the protein and the peptide ligand. Usually, these will occur
between hydrophobic amino acid side chains of the peptide buried within the

WO 2006/061219 PCT/EP2005/013145
36
pockets of the binding groove such that they are not exposed to solvent.
Exposure of the hydrophobic residues to solvent is highly unfavorable since the
surrounding solvent molecules are forced to hydrogen bond with each other
forming cage-like clathrate structures. The resultant decrease in entropy is highly
unfavorable. Lipophilic atoms may be sulphurs which are neither polar nor
hydrogen acceptors and carbon atoms which are not polar.
[0142] van der Waal's bonds are non-specific forces found between atoms
which are 3- 4A apart. They are weaker and less specific than hydrogen and
electrostatic bonds. The distribution of electronic charge around an atom
changes with time and, at any instant, the charge distribution is not symmetric.
This transient asymmetry in electronic charge induces a similar asymmetry in
neighboring atoms. The resultant attractive forces between atoms reaches a
maximum at the van der Waal's contact distance but diminishes very rapidly at
about 1Ǻ to about 2Ǻ. Conversely, as atoms become separated by less than the
contact distance, increasingly strong repulsive forces become dominant as the
outer electron clouds of the atoms overlap. Although the attractive forces are
relatively weak compared to electrostatic and hydrogen bonds (about 0.6
Kcal/mol), the repulsive forces in particular may be very important in determining
whether a peptide ligand may bind successfully to a protein.
[0143] In one embodiment, the Bohm scoring function (SCORE1 approach)
is used to estimate the binding constant. (Bohm, H.J.. (1994), J. Comput. Aided
Moi. Des., 8(3):243-256) which is hereby incorporated in its entirety). In another
embodiment, the scoring function (SCORE2 approach) is used to estimate the
binding affinities as an indicator of a ligand containing a T-cell epitope (Bohm,
H.J., (1998), J. Comput. Aided Mol. Des., 12(4):309-323) which is hereby
incorporated in its entirety). However, the Bohm scoring functions as described
in the above references are used to estimate the binding affinity of a ligand to a
protein where it is already known that the ligand successfully binds to the protein
and the protein/ligand complex has had its structure solved, the solved structure
being present in the Protein Data Bank ("PDB"). Therefore, the scoring function
has been developed with the benefit of known positive binding data. In order to
allow for discrimination between positive and negative binders, a repulsion term
must be added to the equation. In addition, a more satisfactory estimate of
binding energy is achieved by computing the lipophilic interactions in a pairwise

WO 2006/061219 PCT/EP2005/013145
37
manner rather than using the area based energy term of the above Bohm
functions. Therefore, in one embodiment, the binding energy is estimated using a
modified Bohm scoring function. In the modified Bohm scoring function, the
binding energy between protein and ligand (∆Gbind) is estimated considering the
following parameters: The reduction of binding energy due to the overall loss of
translational and rotational entropy of the ligand (∆G0); contributions from ideal
hydrogen bonds (∆Ghb) where at least one partner is neutral; contributions from
unperturbed ionic interactions (AGiOnic); lipophilic interactions between lipophilic
ligand atoms and lipophilic acceptor atoms (AG|jP0); the loss of binding energy
due to the freezing of internal degrees of freedom in the ligand, i.e.., the freedom
of rotation about each C-C bond is reduced (∆Grot); the energy of the interaction
between the protein and ligand (Evdw). Consideration of these terms gives
equation 1:
(∆Gbind)=(∆Go)+(∆GhbXNhb)+(∆GionicXNioniC)+(∆GlipoXNlipo)+(∆Grot+NrQt)+(Evdw)-
where N is the number of qualifying interactions for a specific term and, in one
embodiment, ∆Go, ∆Ghb, ∆GiOnic. ∆Glipo and ∆Grat are constants which are given
the values: 5.4, -4.7, -4.7, -0.17, and 1.4, respectively.
[0144] The term Nhb is calculated according to equation 2:
Nhb = ∑h-bondsf( ∆R, ∆α) X f(Nneighb) X fpcs
[0145] f(AR, Aa) is a penalty function which accounts for large deviations of
hydrogen bonds from ideality and is calculated according to equation 3:
f(∆R,∆-α) = f1( ∆R) x f2(∆ α)
where: fl(∆R) =1 if ∆R or = 1 - (∆R - TOL)/0.4 if AR or = 0 if ∆R >0.4 + TOL
and: f2(∆α) = 1 if ∆α or = 1 -(∆ α - 30)/50 if ∆α or = 0 if ∆α >80°
[0146] TOL is the tolerated deviation in hydrogen bond length = 0.25Ǻ;
∆R is the deviation of the H-O/N hydrogen bond length from the ideal value = 1.9Ǻ;
∆α is the deviation of the hydrogen bond angle [0147] f(Nneighb) distinguishes between concave and convex parts of a protein
surface and therefore assigns greater weight to polar interactions found in

WO 2006/061219 PCT/EP2005/013145
38
pockets rather than those found at the protein surface. This function is calculated
according to equation 4 below:
f(Nneighb) = (Nneighb/Nneighb,o)a where α = 0.5.
[0148] Nneighb is the number of non-hydrogen protein atoms that are closer
than 5Ǻ to any given protein atom.
[0149] Neeighb,o is a constant = 25
[0150] fpcs is a function which allows for the polar contact surface area per
hydrogen bond and therefore distinguishes between strong and weak hydrogen
bonds and its value is determined according to the following criteria:
fpcs= β When APolar/NHB or fPcs= I when Apolar/NHB > 10 Ǻ2
Apolar is the size of the polar protein-ligand contact surface
NHB is the number of hydrogen bonds
β is a constant whose value = 1.2
[0151] For the implementation of the modified Bohm scoring function, the
contributions from ionic interactions, ∆GiOnic, are computed in a similar fashion to
those from hydrogen bonds described above since the same geometry
dependency is assumed.
[0152] The term Nlipo is calculated according to equation 5 below:
Nlipo = ∑ILf(riL)
[0153] f(riL) is calculated for all lipophilic ligand atoms, I, and all lipophilic
protein atoms, L, according to the following criteria:
f(rIL) =1 when riL R1
f(rIL> =0 when rIL >= R2
where: R1=r,vdw + rLvdw +0.5
and R2 = R1 + 3.0
and rivdw is the van der Waal's radius of atom I
and rLVdw is the van der Waal's radius of atom L
[0154] The term Nrot is the number of ratable bonds of the amino acid side
chain and is taken to be the number of acyclic sp3 - sp3 and sp3 - sp2 bonds.
Rotations of terminal -CH3 or -NH3 are not taken into account.
[0155] The final term, Evdw, is calculated according to equation 6 below:

WO 2006/061219 PCT/EP2005/013145
39
Evdw = ε1ε2((r1vdw +r2vdw)12/r12 - (nvdw +r2vdw)6/r6), where:
ε1 and ε2 are constants dependent upon atom identity;
r1vdw +r2vdw are the van der Waal's atomic radii; and
r is the distance between a pair of atoms.
[0156] With regard to equation 6, in one embodiment, the constants ε1 and ε2
are given the atom values: C: 0.245, N: 0.283, O: 0.316, S: 0.316, respectively
(i.e.. for atoms of Carbon, Nitrogen, Oxygen and Sulfur, respectively). With
regards to equations 5 and 6, the van der Waal's radii are given the atom values
C: 1.85, N: 1.75, O: 1.60, S: 2.00 Ǻ.
[0157] It should be understood that all predetermined values and constants
given in the equations above are determined within the constraints of current
understandings of protein ligand interactions with particular regard to the type of
computation being undertaken herein.
[0158] As described above, the scoring function is applied to data extracted
from the database of side-chain conformations, atom identities, and interatomic
distances. For the purposes of the present description, the number of MHC
Class II molecules included in this database is 42 models plus four solved
structures. It should be apparent from the above descriptions that the modular
nature of the construction of the computational method of the present invention
means that new models can simply be added and scanned with the peptide
backbone library and side-chain conformational search function to create
additional data sets which can be processed by the peptide scoring function as
described above. This allows for the repertoire of scanned MHC Class II
molecules to easily be increased, or structures and associated data to be
replaced if data are available to create more accurate models of the existing
alleles.
[0159] The present prediction method can be calibrated against a data set
comprising a large number of peptides whose affinity for various MHC Class II
molecules has previously been experimentally determined. By comparison of
calculated versus experimental data, a cut of value can be determined above
which it is known that ail experimentally determined T-cell epitopes are correctly
predicted.

WO 2006/061219 PCT/EP2005/013145
40
[0160] It should be understood that, although the above scoring function is
relatively simple compared to some sophisticated methodologies that are
available, the calculations are performed extremely rapidly. It should also be
understood that the objective is not to calculate the true binding energy perse for
each peptide docked in the binding groove of a selected MHC Class II protein.
The underlying objective is to obtain comparative binding energy data as an aid
to predicting the location of T-cell epitopes based on the primary structure (i.e..
amino acid sequence) of a selected protein. A relatively high binding energy or a
binding energy above a selected threshold value would suggest the presence of a
T-ce!l epitope in the iigand. The ligand may then be subjected to at least one
round of amino-acid substitution and the binding energy recalculated. Due to the
rapid nature of the calculations, these manipulations of the peptide sequence can
be performed interactively within the program's user interface on cost-effectively
available computer hardware. Major investment in computer hardware is thus not
required.
[0161] It would be apparent to one skilled in the art that other available
software could be used for the same purposes. In particular, more sophisticated
software which is capable of docking ligands into protein binding-sites may be
used in conjunction with energy minimization. Examples of docking software are:
DOCK (Kuntz et al., (1982), J. Mol. BioL 161:269-288), LUDI (Bohm, H.J.,
(1994), J. Comput Aided Mol. Des.. 8:623-632) and FLEXX (Rarey et al., (1995),
ISMB, 3:300-308). Examples of molecular modeling and manipulation software
include: AMBER (Tripos) and CHARMm (Molecular Simulations Inc.). The use of
these computational methods would severely limit the throughput of the method
of this invention due to the lengths of processing time required to make the
necessary calculations. However, it is feasible that such methods could be used
as a 'secondary screen' to obtain more accurate calculations of binding energy
for peptides which are found to be 'positive binders' via the method of the present
invention. The limitation of processing time for sophisticated molecular mechanic
or molecular dynamic calculations is one which is defined both by the design of
the software which makes these calculations and the current technology
limitations of computer hardware. It may be anticipated that, in the future, with
the writing of more efficient code and the continuing increases in speed of
computer processors, it may become feasible to make such calculations within a

WO 2006/061219 PCT/EP2005/013145
41
more manageable time-frame. Further information on energy functions applied to
macromolecules and consideration of the various interactions that take place
within a folded protein structure can be found in: Brooks etal., (1983), J. Comput.
Chem., 4:187-217 and further information concerning general protein-ligand
interactions can be found in: Dauber-Osguthorpe etal., (1988), Proteins, 4(1):31-
47. Useful background information can also be found, for example, in Fasman,
G.D., ed., Prediction of Protein Structure and the Principles of Protein
Conformation, Plenum Press, New York, ISBN: 0-306 4313-9.
Example 2: In vitro analysis of IL-7 derived peptides as potential CD4+ T helper
cell epitopes by unfractionated PBMC cultures
[0162] Based on in silico predictions that sequences surrounding N-linked
glycosylation sites of the IL-7 protein are immunogenic, peptides encompassing
these regions, spanning for example Leu63 to Ser71 (LRQFLKMNS SEQ ID
NO:4) or Ile88 to Val96 (ILLNCTGQV SEQ ID NO:7) in mature human IL-7
protein, are analyzed for their immunogenicity, which is measured by their ability
to induce T-cell proliferation in vitro. In essence, PBMCs isolated from human
blood are incubated with individual overlapping 15-mer peptides, and proliferative
responses are measured by 3H-thymidine incorporation. In principle, T-cells
within the mixture of PBMCs will only proliferate if they recognize individual
peptide-MHC complexes on autologous APCs (antigen presenting cells), and
thus proliferation is an indication of peptide immunogenicity.
[0163] For example, 15-mer peptides that are staggered by three amino acids
and span the region from, for example, Met54 to Leu80 in human IL-7 are
synthesized (Pepscan Systems, Netherlands), resuspended in DMSO (Sigma
Chemical, St. Louis, MO, U.S.A.), and used at a final concentration of 5 //M in
0.5% DMSO in culture media.
[0164] PBMCs are isolated from peripheral blood from healthy donors by
Ficoll-Hypaque gradient centrifugation, and are stored frozen in liquid nitrogen. In
addition, each PBMC sample is HLA typed, using a SSP PCR typing kit (Bio-
Synthesis, Lewisville, TX) on DNA isolated with a QiaAmp Tissue Kit (Qiagen,
Valencia, CA).
[0165] In a typical proliferation assay, each of the overlapping 15-mer
peptides is assayed in sextuplicate PBMC cultures derived from 40 naTve donors.

WO 2006/061219 PCT/EP2005/013145
42
Briefly, 2 x105 PBMCs, thawed rapidly before use, are mixed with 5 pM of each
peptide and incubated at 37°C in 5% CO2 for 7 days. As a positive control,
samples are incubated with the tetanus toxin derived peptide MQYIKANSKFIGI
(SEQ ID NO:15), whereas negative control samples are incubated with 0.5%
DMSO. During the last 12 hours of incubation the cultures are pulsed with of
[methyl-3H]thymidine (0.5 //Ci/well) (NEN Life Science Products, Boston, MA), the
cultures are harvested onto filter mats and thymidine incorporation is measured
as counts per minute (CPM) using a Wallac microplate beta top plate scintillation
counter (Perkin Elmer, Boston, MA). The stimulatory index for each peptide is
calculated by dividing the CPM value of a given peptide divided by the CPM value
obtained from negative controls.
[0166] It is found that the stimulatory index of the positive control peptide is
significantly greater than 1 (the stimulatory index of the negative control), and
peptides containing the core sequence LRQFLKMNS (SEQ ID NO:4),
FLKMNSTGD (SEQ ID NO:5) or LKMNSTGDF (SEQ ID NO:6), such as, for
example peptides ARKLRQFLKMNSTGD (SEQ ID NO:10),
LRQFLKMNSTGDFDL (SEQ ID NO:11), or FLKMNSTGDFDLHLL (SEQ ID
NO: 12), have an increase in stimulatory index. Therefore peptide sequences
such as LRQFLKMNS (SEQ ID NO:4) or LKMNSTGDF (SEQ ID NO:6) indeed
represent potential T-cell epitopes.
[0167] A similar analysis is performed with a series of 15-mer peptides
encompassing a region defined by the core peptides ILLNCTGQV (SEQ ID NO:7)
and LLNCTGQVK (SEQ ID NO:8), and it is found that these peptide sequences
as well represent potential T-cell epitopes.
Example 3: Mapping of CD4+ T helper cell epitopes using differentiated human
dendritic cells (DCs) in vitro
[0168] Mature dendritic cells (DCs) are potent antigen-presenting cells
(APCs) that present antigenic peptides or whole proteins to T-cells efficiently.
Isolated DCs, pulsed with antigenic peptides in vitro, are used to induce primary
T-cell responses that can be measured in in vitro proliferation assays.
Differentiated DCs are generated, for example by the following procedure: first,
human monocytes are generated by allowing PBMCs to adhere to plastic tissue
culture flasks or by purifying CD14+ PBMCs with magnetically labeled antibodies

WO 2006/061219 PCT/EP2005/013145
43
(Miltenyi Biotec, Auburn, CA). The purified monocytes (0.5 to 1.5 x 10° cells/ml)
are then cultured in AIM V media (GIBCO BRL, Grand Island, NY, U.S.A.)
containing 1000 U7ml GM-CSF (Endogen; Woburn, MA) and 500 U/ml IL-4
(Endogen; Woburn, MA) for 3 days. Subsequently, these immature DCs are
pulsed with 5 ^/g/ml of experimental or control peptides and further incubated with
a combination of 1000 U/ml TNF-a (Endogen; Woburn, MA), 1000 U/ml GM-CSF
and 500 U/ml IL-4 for another 48 hours. Mature DCs are monitored by high
surface expression levels of CD80+, CD86+ and HLA-DR.
[0169] These mature antigen-pulsed DCs are irradiated with 4200 Rads and
are used in a proliferation assay with purified autologous CD4+ T-cells. (CD4+ T-
cells are purified with magnetically labeled antibodies (Miltenyi Biotec, Auburn,
CA), using frozen PBMC aliquots from the same donor that provides monocytes
for the in vitro DC differentiation.) In a typical assay, antigen-pulsed DCs (2 *
105/mL) are incubated together with autologous CD4+ T-cells (2 x 106 cells/mL) in
round-bottomed 96-well plates at 37°C in 5% CO2 for 7 days, [methyl-
3H]thymidine (NEN Life Science Products, Boston, MA) is added during the last
12 hours of incubation at 0.5 pCi/well, the samples are harvested, lysed onto
glass filters, and 3H-thymidine incorporation is measured in a scintillation counter.
[0170] 15-mer peptides, as described for Example 1, are tested in this assay
and compared to reference peptides and other controls. It is found that this
assay is more sensitive than the assay described in Example 1, allowing better
differentiation between the ability of individual peptides to induce T-cell
proliferation. It is found, that IL-7 peptides containing the core sequences
LRQFLKMNS (SEQ ID NO:4), FLKMNSTGD (SEQ ID NO:5), LKMNSTGDF
(SEQ ID NO:6), ILLNCTGQV(SEQ ID NO:7) or LLNCTGQVK (SEQ ID NO:8) do
indeed induce significant T-cell proliferation and therefore these sequences do
represent potential T-cell epitopes.
Example 4: In vitro analysis of de-immunizing amino acid substitutions in IL-7
[0171] Amino acid substitutions in the peptide regions described above which
are considered to render the IL-7 protein less immunogenic are tested in in vitro
assays, as described in Example 1 and Example 2. For example, the variant IL-7
peptide encompassing the sequence LRQFLDDNS (SEQ ID NO: 13) is expected
to generate a significantly decreased T-cell proliferative response compared to

WO 2006/061219 PCT/EP2005/013145
44
the wild-type parental peptide encompassing LRQFLKMNS (SEQ ID NO:4).
Similarly, the variant IL-7 peptide encompassing the sequence TLLMCTGQG
(SEQ ID NO: 14) is expected to generate a significantly decreased T-cell
proliferative response compared to the wild-type parental peptide encompassing
ILLNCTGQV (SEQ ID NO:7)
[0172] A series of IL-7 derived 15-mer peptides is synthesized that
encompass the variant IL-7 sequences LRQFLDDNS (SEQ ID NO:13) or
TLLNCTGQG (SEQ ID NO:14), as described in Example 1. In addition, the
variant IL-7 proteins, or fusion proteins containing variant IL-7, which include
substitutions of the invention are produced either in a prokaryotic or eukaryotic
expression system (Del-IL-7's). For example, a variant IL-7 is produced that
includes the amino acid substitutions K68D, M69D, I88T and V94G. (In addition,
the prokaryotically produced IL-7 proteins include a start methionine.) These
peptides and purified proteins, and their parental counterparts, are tested in their
ability to induce T-cell proliferation, in assays with whole PBMC cultures as
described in Example 1 or by pulsing human DCs as described in Example 2. 25
μg/ml of protein is used to stimulate PBMCs or to pulse DCs.
[0173] It is found that in general, peptides derived from the variant IL-7
sequences have a significantly reduced ability to induce T-cell proliferation
compared to the corresponding peptides derived from the wild-type human IL-7
protein. Therefore, these variant peptide sequences are much poorer potential T-
cell epitopes. Likewise, the bacterially produced variant IL-7 protein also has a
reduced ability to induce T-cell proliferation than wild-type IL-7, indicating that
these mutated regions may be significant contributors to the immunogenicity of
prokaryotically produced IL-7.
EXAMPLE 5: Analysis of IL-7 derived peptides as potential B-cell epitopes
[0174] For bacterially-produced, unglycosylated human IL-7 protein,
sequences surrounding N-Iinked glycosylation sites of IL-7 may be recognized as
"non-self by the human immune system, and elicit an antibody response.
Essentially, to assess if these sequences represent linear B-cell epitopes,
peptides spanning these sequences are used to immunize rabbits, and the
reactivity of resulting antibodies toward bacterially-produced native human IL-7

WO 2006/061219 PCT/EP2005/013145
45
and denatured human IL-7 is tested. As a further control, a native eukaryotically-
produced glycosylated huFc-IL-7 fusion protein is used.
[0175] Methods and materials to raise polyclonal antibodies against a specific
peptide antigen in, for example, rabbits, and their subsequent purification are
generally known to those skilled in the art, and references thereto may be found,
for example, in: Antibodies: A Laboratory Manual, E. Hariow and D. Lane, Cold
Spring Harbor Press.
[0176] Briefly, in one example, a peptide containing the core sequence
FLKMNSTGD (SEQ ID NO:5), such as LRQFLKMNSTGDFDL[C] (SEQ ID NO:
18) or [CjLRQFLKMNSTGDFDL (SEQ ID NO:19) is coupled via an added
terminal cysteine to three different carrier proteins, keyhole limpet hemocyanin
(KLH, EMD Biosciences, San Diego, CA), BSA (EMD Biosciences, San Diego,
CA), and ovalbumin (Pierce, Rockford, IL) using a coupling agent such as SMCC
(Pierce, Rockford, IL), and multiple rabbits are immunized by successive
injections with each of the peptide conjugate in the presence of adjuvant. The
immune response is boosted with further injections of one of the peptide-carrier
conjugates at monthly intervals, and the resulting antiserum from each rabbit is
affinity-purified over a Sulfo-Link column (Pierce, Rockford, IL) to which the
peptide is coupled, and the antibody is further concentrated over a hydroxyapatite
column (Bio-Rad Laboratories, Hercules, CA).
[0177] The purified antibodies are tested against bacterially-produced native
human IL-7, denatured human IL-7 and a eukaryotically-produced glycosylated
huFc-IL-7 fusion protein in an ELISA assay, following standard procedures.
Briefly, ELISA plates, coated with the purified protein preparations, are incubated
with the test antibody samples, the plates are washed, incubated with a
secondary antibody such as horseradish peroxidase-coupled anti-rabbit IgG,
washed again and incubated with a chromogenic substrate solution to indicate
the concentration of bound antibody.
[0178] Similarly, antibodies are raised to other peptides encompassing the
...MNSTG...(SEQ ID NO:20) glycosylation site (at Asn70) in human IL-7, or to
peptides encompassing the ...LNCTG...(SEQ ID NO:21) glycosylation site (at
Asn91). Using this approach, it is found that generally the denatured bacterially-
produced human IL-7 is well recognized by the antibodies and that the
glycosylated huFc-IL-7 fusion protein is not. Peptides that give rise to antibodies

WO 2006/061219 PCT/EP2005/013145
46
reacting with the bacterially-produced native human IL-7 protein indicate that a
linear B-cell epitope at the glycosylation site is recognized. It is further found that,
in a cell-based proliferation assay as described in Example 9, the antibodies
raised against peptides of this Example have the effect of inhibiting IL-7-
stimulated cell proliferation. This result indicates that these antibodies have
neutralizing activity.
EXAMPLE 6: Construction of human IL-7 variants that lack potential T-cell
epitopes
[0179] Nucleic acids are constructed that encode versions of human IL-7
variants either suitable for bacterial expression or suitable for eukaryotic
expression, for instance as a fusion protein. For example, nucleic acids encoding
a mature human IL-7 variant containing the substitutions K68D, M69D, I88T, and
V96G (Del-IL-7 SEQ ID NO:16) are constructed, using standard methods familiar
to those skilled in the art. Figure 11 shows an example of such a DNA sequence
encoding a mature IL-7 variant, Del-IL-7, with codon substitutions of amino acid
residues K68D, M69D, I88T, and V96G (SEQ ID NO:24).
[0180] For bacterial expression, the protein sequence of Del-IL-7 including a
start methionine (bDel-IL-7 SEQ ID NO:17) is reverse-translated using a codon
bias appropriate for optimal E. coli expression. The resulting nucleic acid
sequence is further adapted to include desired (or to exclude undesired) features
such as a stop codon or restriction sites, and sequences are added that facilitate
cloning into a bacterial expression vector, for example an appropriate vector from
the pET series (EMD Biosciences, San Diego, CA). The nucleic acid sequence is
synthesized by total gene synthesis (Blue Heron Biotechnology, Bothell, WA) and
inserted into the expression vector. An example of a DNA sequence encoding
bDel-IL-7, codon-optimized for E. coli with codon substitutions of amino acid
residues K68D, M69D, I88T, V96G, is shown in Figure 10 (SEQ ID NO:23).
[0181] For eukaryotic expression as a huFc-Dei-IL-7 fusion protein, the
nucleic acid sequence of the mature human IL-7 is modified to incorporate
codons for desired amino acid mutations of the invention as described above.
(See e.g. SEQ ID NO:24). The sequence is further adapted to incorporate
flanking sequences with unique restriction sites for insertion as a Xma I / Xho I
fragment in-frame into a pdCs-huFc expression vector encoding the hinge, CH2

WO 2006/061219 PCT/EP2005/013145
47
and CH3 region of lgG1 (see Lo et a/., (1998), Protein Engineering 11:495), and
is synthesized by total gene synthesis (Blue Heron Biotechnology, Bothell, WA).
The synthetic Xma I / Xho I Del-IL-7 fragment is then cloned into the pdCs-huFc
vector, yielding an expression plasmid encoding huFc-Del-lL-7. Other IL-7 and
Fc-IL-7 variants of the invention can be produced by similar methods.
[0182] Specifically, nucleic acids encoding the human deimmunized Fc-IL-7
fusion proteins huFcy2(h)(FN>AQ)-(linker2)-IL-7(PNS) and huFcy2(h)(FN>AQ)-
(Iinker2)-IL-7(PNDS) were generated as follows. huFcy2(h)(FN>AQ)-(linker2)-IL-
7(PNS) is a human Fc-IL-7 fusion protein comprising the N-terminus of human IL-
7 genetically fused to the C-terminus of a human lgG2 Fc domain with an lgG1
hinge via a linker sequence GGGGSGGGG. The Fc portion contains the
mutations Phe296Ala and Asn297Gln. The IL-7 portion contains the mutations
F39P, F57N and L128S. huFcy2(h)(FN>AQ)-(linker2)-IL-7(PNDS) is the same as
huFcy2(hXFN>AQ)-(linker2)-IL-7(PNS), but for containing an additional mutation
in the IL-7 moiety, L77D. The sequence also contains codons for the mutation of
the LSLS sequence near the C-terminus of the Fc portion to be replaced by
ATAT. In addition, the nucleic acid sequence includes a codon to replace the C-
terminal lysine of the Fc portion with an alanine residue.
[0183] A nucleic acid of the sequence presented in Figure 27 was
synthesized de novo (Blue Heron Biotechnology, Bothell, WA), which encodes
the linker sequence GGGGSGGGG followed by mature human IL-7 containing
the amino acid substitutions F39P, F57N, and L128S (IL-7(PNS)) and which
contains flanking restriction sites Xma I and Xho I at the 5'- and 3' ends,
respectively. This purified Xma I / Xho I fragment was ligated to a likewise
digested and purified vector fragment of the pdCs-huFc series, pdC10-
huFcy2(h)(FN>AQ), generating a plasmid encoding huFcy2(h)(FN>AQ)-(linker2)-
IL-7(PNS). Lo et a/., (1998), Protein Engineering 11:495. The coding sequence
was ascertained by sequencing.
[0184] The further introduction of the substitution L77D into IL-7(PNS) was
performed by standard PCR mutagenesis methods, using mutagenic primers
M(s) (5'-TGACTTTGATGACCACCTGTTAAAAGTTTC-3' (SEQ ID NO: 50);
mutated codon underlined) and M(a)
(5'-AACAGGTGGICATCAAAGTCACCAGTGC-31) (SEQ ID NO:51). Briefly,

WO 2006/061219 PCT/EP2005/013145
48
separate PCR reactions were performed on a plasmid template containing
(Iinker2)-IL-7(PNS), one with M(s) and the downstream primer
5' - CTCGAGTCAGTGTTCTTTAGTGCCCATC - 3' (SEQ ID NO:52), the other
with M(a) and the upstream primer
5' - CCCGGGTGCTGGAGGTGGAGGATCAGGTG- 3' (SEQ ID NO:53), the PCR
fragments were purified and combined as the template for a secound round of
PCR using again the upstream primer
5' - CCCGGGTGCTGGAGGTGGAGGATCAGGTG- 3'(SEQ ID NO:54) and the
downstream primer 5' - CTCGAGTCAGTGTTCTTTAGTGCCCATC - 3'(SEQ ID
NO:55). The resultant purified fragment was inserted into a TA cloning vector
pCR2.1 ((Invitrogen, Carlsbad, CA), and its sequence was confirmed. An Xma I /
Xho I fragment encoding (Iinker2)-IL-7(PNDS) was excised, and ligated to a
likewise digested and purified vector fragment of the pdCs-huFc series, pdC10-
huFcy2(h)(FN>AQ), generating a plasmid encoding huFcy2(h)(FN>AQ)-(linker2)-
IL-7(PNDS).
[0185] Similarly, plasmids encoding variants of these fusion proteins differing
in the Fc moiety are obtained; for example a plasmid encoding
huFcy2(h)(iinker2)-IL-7(PNDS) is obtained by ligating an Xma I /Xho I fragment
encoding (Iinker2)-IL-7(PNDS) to a likewise digested and purified vector fragment
of the pdCs-huFc series, pdC10-huFcy2(h).
EXAMPLE 7: Expression and purification of lL-7 variants
[0186] For eukaryotic expression of the huFc-Del-IL-7 fusion protein,
electroporation is used to introduce the DNA encoding the fusion protein into a
mouse myeloma NS/0 cell line. To perform electroporation NS/0 cells are grown
in Dulbecco's modified Eagle's medium supplemented with 10% heat-inactivated
fetal bovine serum, 2 mM glutamine and penicillin/streptomycin. About 5x106
cells are washed once with PBS and resuspended in 0.5 ml PBS. 10 ug of
linearized plasmid DNA for huFc-Del-IL-7 is then incubated with the cells in a
Gene Pulser Cuvette (0.4 cm electrode gap, BioRad) on ice for 10 min.
Electroporation is performed using a Gene Pulser (BioRad, Hercules, CA) with
settings at 0.25 V and 500 μF. Cells are allowed to recover for 10 min on ice,
after which they are resuspended in growth medium and plated onto two 96 well
plates.

WO 2006/061219 PCT/EP2005/013145
49
[0187] Stably transfected clones are selected by their growth in the presence
of 100 nM methotrexate (MTX), which is added to the growth medium two days
post-transfectiom The cells are fed every 3 days two to three more times, and
MTX-resistant clones appear in 2 to 3 weeks. Supematants from clones are
assayed by anti-Fc ELISA to identify clones that produced high amounts of the IL-
7 fusion protein. High producing clones are isolated and propagated in growth
medium containing 100 nM MTX. Typically, a serum-free growth medium, such
as H-SFM or CD medium (Life Technologies), is used.
[0188] A standard purification of Fc-containing fusion proteins is performed
based on the affinity of the Fc protein moiety for Protein A. Briefly, NS/0 cells
expressing the fusion protein, such as huFc-Del-IL-7, are grown in tissue culture
medium and the supernatant containing the expressed protein is collected and
loaded onto a pre-equilibrated Fast Flow Protein A Sepharose column. The
column is then washed extensively with buffer (such as 100 mM sodium
phosphate, 150 mM NaCI at neutral pH). Bound protein is eluted at a low pH (pH
2.5 - 3) in same buffer as above and fractions are immediately neutralized.
[0189] Bacterial expression and purification of bDel-IL-7 is performed
essentially as described by Cosenza et al. for bacterially-produced IL--7 (see
Cosenza et al., (1997) JBC, 272:32995). In essence, bDel-IL-7 is isolated from
inclusion bodies, denatured and refolded. Briefly, bacterial expression cultures
transformed with the expression vector encoding, for example, bDel-IL-7 are
grown to mid-log phase and recombinant protein expression is induced.
Following induction, the bacteria are harvested and lysed by sonication, and
inclusion bodies are isolated in buffer A (50 mM Tris HCI (7.5), 5 mM EDTA, 20%
sucrose). After extensive washes, the inclusion bodies are resuspended in a
guanidine denaturation buffer (50 mM Tris-HCI (pH8.0), 5 M guanidine HCI, 5 mM
EDTA), briefly sonicated and reduced in 6 mM DTT. Denatured bDel-IL-7 protein
is then further purified by denaturing size exclusion HPLC. The protein is then
refolded in refolding buffer (50 mM glycine, 30 mM NaOH, 0.4 M L-arginine, 1
mM DTT, pH 10), dialyzed into a phosphate buffer, and further purified by size
exclusion HPLC.

WO 2006/061219 PCT/EP2005/013145
50
EXAMPLE 8: Biochemical analysis of IL-7 variants
[0190] The effect of the introduced mutations on the integrity of IL-7 proteins
is assessed by routine reducing and non-reducing SDS-PAGE analysis and size
exclusion chromatography.
[0191] For example, the fusion protein huFc-Del-IL-7, expressed from NS/0
cells, is captured on Protein A Sepharose beads (Repligen, Needham, MA) from
the tissue culture medium into which it is secreted, and eluted by boiling in protein
sample buffer, with or without a reducing agent such as /?-mercaptoethanol. The
sample is fractionated by SDS-PAGE and the protein bands are visualized by
Coomassie staining. It is expected that a fusion protein containing IL-7 mutations
that sufficiently interfere with proper folding is more likely to show degradation
products by SDS-PAGE.
[0192] Purified huFc-Del-IL-7 is also analyzed by size exclusion
chromatography (SEC) to assess the extent to which the fusion protein is
aggregated. Briefly, the cell culture supernatant is loaded onto a pre-equilibrated
Fast-Flow Protein A Sepharose column, the column is washed extensively in a
physiological buffer (such as 100 mM Sodium Phosphate, 150 mM NaCI at
neutral pH), and the bound protein is eluted at about pH 2.5 to 3 in same salt
buffer as above. Fractions are immediately neutralized, peak fractions are
pooled, and an aliquot is fractionated over an analytical SEC column.
EXAMPLE 9: In vitro activity of IL-7 variants
[0193] To determine whether the IL-7 variants containing mutations of the
invention retain their cytokine activity in vitro cellular proliferation bioassays are
performed. Human PBMC (Peripheral Blood Mononuclear Cells) are activated by
PHA-P to produce cells which are responsive to IL-7. Proliferation is measured in
a standard thymidine incorporation assay.
[0194] For example, the cytokine activity of huFc-Del-IL-7 and bDel-IL-7 is
determined. Briefly, PBMC's are first incubated for five days with 10
microgram/ml PHA-P, cells are washed and then incubated in medium with huFc-
Del-IL-7 or bDe!-IL-7, prepared as a dilution series, for a total of 48 hours. During
the final 12 hours, the samples are pulsed with 0.3 JJC\ of [methyl-3H]thymidine
(Dupont-NEN-027). Cells are then washed extensively, harvested and lysed onto
glass filters. 3H-thymidine incorporated into DNA is measured in a scintillation

WO 2006/061219 PCT/EP2005/013145
51
counter. As a standard, wild type hulL-7 protein, obtained from R&D Systems
(Minneapolis, MN), or obtained from the National Institute for Biological
Standards and Control (NIBSC), is assayed.
[0195] An ED50 value of cell proliferation for huFc-Del-IL-7 or bDel-lL-7 is
obtained from plotting a dose response curve according to standard techniques,
and determining the protein concentration that results in half-maximal response.
Example 10: Induction of anti-human IL-7 antibodies in monkeys by wild-type IL-
7 and IL-7 variants
[0196] It is known that bacterial-derived wild-type human IL-7 administered to
monkeys often results in neutralizing anti-human IL-7 antibody titers (Storek et
a/., (2003), Blood, 101:4209; Fry etai, (2003), Blood, 101:2294). Thus, the
propensity of prokaryotically produced variant IL-7 and wild-type IL-7 proteins, as
well as eukaryotically produced fusion proteins containing wild-type or variant IL-7
polypeptides, to induce neutralizing antibodies in nonhuman primates is
assessed. In a typical experiment, rhesus macaques are injected with 40//g/kg
of the protein samples subcutaneously once a day for four weeks. For example,
the protein samples are commercially available prokaryotically-produced IL-7
(PeproTech, Rocky Hill, NJ), the prokaryotically produced variant IL-7(K68D,
M69D, I88T, V94G), and the equivalent Fc-IL-7 fusion proteins produced in a
mammalian expression system. At regular intervals, serum is obtained from the
animals, and serum concentrations of antibodies against human IL-7 are
measured by ELISA using human IL-7 coated 96 well plates (Nunc, Naperville,
IL). Typically, serial dilutions of each serum sample are added to each well in
triplicate for two hours, washed with 0.05% Tween (Tween 20) in PBS and
blocked with 1% BSA/1% goat serum in PBS. To each sample a horseradish
peroxidase-conjugated anti-macaque IgG is added (1:60,000 in sample buffer),
incubated at 37°C for 2 hr, and the plate is washed 8 times with 0.05% Tween in
PBS. Samples are then assayed using the colorimetric substrate solution OPD
(o-phenylenediamine dihydrochloride) by measuring the OD at 490 nm,
subtracting the background OD reading at 650 nm.
[0197] It is found that prokaryotically produced wild-type IL-7 protein indeed
gives rise to high anti-IL-7 antibody titers. In contrast, the antibody titers of the
prokaryotically produced variant IL-7 gives rise to significantly lower titers of anti

WO 2006/061219 PCT/EP2005/013145
52
IL-7 antibodies. It is also found that the differences in the levels of anti IL-7
antibody titers produced by animals administered mammalian produced wild-type
and variant Fc-IL-7 fusion proteins, (with mutations around the N-linked
glycosylation sites) are not as pronounced. This result may indicate that the lack
of glycosylation at these sites in the prokaryotically produced proteins contributes
to the immunogenicity of these proteins.
Example 11: Acute tolerability of Fc-IL-7 in immunocompetent mice
[0198] The Fc-IL-7 fusion protein huFcK2(h)(N>Q)(linker2)-hulL-7 was
prepared according to the method described in Example No. 6, purified, then
formulated in 50mM phosphate, 150mM sodium chloride pH 7.00, 0,05% (v/v)
Tween 80. The protein concentrations of diluted solutions were determined using
the absorbance at 280nm and the theoretical extinction coefficient of 0.98
mg/OD280, based on the known protein sequence. For dosing mice, an aliquot of
each sample was removed from stock vials and diluted with 0.9% saline within
one hour of dosing.
[0199] C57B1/6 mice (Charles River Laboratories, Wilmington, MA), 17
weeks of aged were divided into groups of 2 mice each and were administered
the Fc-IL-7 fusion proteins subcutaneously for 5 consecutive days. Groups
received dosages of either 0.5, 5.0, 25 mg/kg or the vehicle control each day. All
mice survived the treatment through day 7, at which point the mice were
sacrificed.
[0200] Fc-IL-7 plasma levels were determined by obtaining blood samples
from the retro-orbital sinus 6 hours after dosing on days 0, 2, 4, and 7. Blood
samples were collected in tubes containing heparin to prevent clotting. Cells
were removed by centrifugation and the concentration of intact Fc-IL-7 fusion
protein in the plasma was measured using standard ELISA procedures. Plasma
levels of Fc-IL-7 in //g///l for test mice are shown in Figure 36. The plasma of
mice showed a dose dependent increase in Fc-IL-7 concentration at all time
points tested and further increased following each dose. However, as shown in
Figure 37, the magnitude of the increase lessened after each dosing.
[0201] The functional activity of Fc-IL-7 was confirmed by measuring
increases in B cells and T cells on day 7 following the initiation of dosing. Since
IL-7 boosts the production of immune effector cells such as B cells and T cells,

WO 2006/061219 PCT/EP2005/013145
53
the cellularity and weight of the spleen is expected to increase. Mice were
sacrificed on day 7 and organs were removed and weighed. Figure 38 shows the
average organ weights on day 7. As expected, spleen weight increased 3 to 5
fold 1 week after the initial dose. Lung weights increased 2 fold following the 2
higher doses of Fc-IL-7 due to lymphotcytic infiltration. No weight changes were
observed in the kidney or liver.
[0202] The response of B cells (CD19+, CD4+, CD8+ and granulocytes (Gr-
I +)) in all groups was observed on day 7. Fig. 39 shows the frequency of Gr-1 +
cells in the peripheral blood of two mice in each group. As the data shows,
granulocytes were generally unresponsive to Fc-IL-7. Fig. 40 shows the
frequency of CD19+ cells in the peripheral blood of two mice in each group. Fig.
41 shows the frequency of CD4+ cells in the peripheral blood of two mice in each
group, while Fig. 42 shows the frequency of CD8+ cells in the peripheral blood of
two mice in each group. The increases in B cell (Fig. 40 and T cell (Figs. 41 and
42 numbers were maximal for the 5 mg/kg dosage group with each mouse tested
showing significantly increased T cell and B cell numbers over the control group
and the 0.5 mg/kg dosage group. However, T cell numbers either declined or
increased for mice in the 25 mg/kg dosage group. All measurements of cells are
shown in cells per/;L of blood.
Example 12. Assessment of Human Fc-IL-7 Activity
[0203] The biological activity of the Fc-IL-7 fusion protein tested in Example
11 was measured by tritiated thymidine uptake in a standard cell proliferation
assay using peripheral blood mononuclear cell (PBMC) PHA blasts according to
the method described in Yokota et al, (1986), Proc. Natl. Acad. Sci. USA,
83:5894; and Stern et al., (1990), Proc. Natl. Acad. Sci. USA, 87:6808-6812, with
human IL-7 used as a standard. As shown in Figure 43, cellular proliferation as
measured by the uptake of tritiated thymidine for the Fc-IL-7 molecule is similar to
that of the standard NIBSC human IL-7 (World Health Organization), indicating
that the activity of the Fc-iL-7 molecule is similar to wild-type human IL-7.
Equivalents
[0204] The invention may be embodied in other specific forms without
departing from the spirit or essential characteristics thereof. The foregoing

WO 2006/061219 PCT/EP2005/013145
54
embodiments are therefore to be considered in all respects illustrative rather than
limiting on the invention described herein. Scope of the invention is thus
indicated by the appended claims rather than by the foregoing description, and all
changes which come within the meaning and range of equivalency of the claims
are intended to be embraced therein.

WO 2006/061219 PCT/EP2005/013145
55
WE CLAIM:
1. A polypeptide comprising a modified human IL-7 molecule or an active portion thereof having a T-cell epitope modified to reduce an anti-IL-7 T-cell response, said polypeptide further comprising an Fc portion of an immunoglobulin molecule fused via its C-terminal to the N-terminal of said modified IL-7 molecule, wherein said fused Fc-IL7 molecule is selected from the group consisting of:
(i) huFcy2(h)(FN>AQ) - L - IL-7(F39P, F57N, L128S),
wherein Fcy2(h) is a human Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2 comprising the mutations F296A and N297Q, L is a linker with the sequence GGGGSGGGG, and IL-7(F39P, F57N, L128S) is IL-7 containing the mutations F39P, F57N, and L128S; said molecule having the sequence as depicted in Fig. 31; (ii) huFcy2(h)(FN>AQ) - L - IL-7(F39P, F57N, L77D, L128S),
wherein Fcy2(h) is a human Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2 comprising the mutations F296A and N297Q, L is a linker with the sequence GGGGSGGGG, and IL-7(F39P, F57N, L77D L128S) is IL-7 containing the mutations F39P, F57N, L77D and L128S; said molecule having the sequence as depicted in Fig. 32; (iii) huFcy2(h) - L - IL-7 (F39P, F57N, L77D, L128S),
wherein Fcy2(h) is a human Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2, L is a linker with the sequence GGGGSGGGG, and IL-7(F39P, F57N, L77D, L128S) is IL-7

containing the mutations F39P, F57N, L77D and L128S; said molecule having the sequence as depicted in Fig. 33; and
(iv) huFcy2(h) - L - IL-7 (F39P, F57N, L128S),
wherein Fcy2(h) is a human Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2 L is a linker with the sequence GGGGSGGGG, and IL-7(F39P, F57N, L128S) is IL-7 containing the mutations F39P, F57N, and L128S; said molecule having the sequence as depicted in Fig. 34.
2. A polypeptide as claimed in claim 1, wherein said immunoglobulin molecule is a human
immunoglobulin.
3. A polypeptide as claimed in claim 1 or 2, wherein said immunoglobulin molecule is IgG2.
4. A nucleic acid molecule encoding a polypeptide as claimed in any one as claimed in claims 1-3.

5. An expression vector comprising a nucleic acid molecule as claimed in claim 4.
6. A prokaryotic cell comprising an expression vector as claimed in claim 5.

A polypeptide comprising a modified human IL-7 molecule or an active portion thereof having a T-cell epitope modified to reduce an anti-IL-7 T-cell response, said polypeptide further comprising an Fc portion of an immunoglobulin molecule fused via its C-terminal to the N-terminal of said modified IL-7 molecule, wherein said fused Fc-IL7 molecule is selected from the group consisting of:
(i) huFcy2(h)(FN>AQ) - L - IL-7(F39P, F57N, L128S),
wherein Fcy2(h) is a human Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2 comprising the mutations F296A and N297Q, L is a linker with the sequence GGGGSGGGG, and IL-7(F39P, F57N, L128S) is IL-7 containing the mutations F39P, F57N, and L128S; said molecule having the sequence as depicted in Fig. 31;
(ii) huFcy2(h)(FN>AQ) - L - IL-7(F39P, F57N, L77D, L128S),
wherein Fcy2(h) is a human Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2 comprising the mutations F296A and N297Q, L is a linker with the sequence GGGGSGGGG, and IL-7(F39P, F57N, L77D L128S) is IL-7 containing the mutations F39P, F57N, L77D and L128S; said molecule having the sequence as depicted in Fig. 32;
(iii) huFcy2(h) - L - IL-7 (F39P, F57N, L77D, L128S),
wherein Fcy2(h) is a human Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2, L is a linker with the sequence GGGGSGGGG, and IL-7(F39P, F57N, L77D, L128S) is IL-7 containing the mutations F39P, F57N, L77D and L128S; said molecule having the sequence as depicted in Fig. 33; and
(iv) huFcy2(h) - L - IL-7 (F39P, F57N, L128S),
wherein Fcy2(h) is a human Fc portion with a hinge of lgG1 and CH2 and CH3 domains of lgG2 L is a linker with the sequence GGGGSGGGG, and IL-7(F39P, F57N, L128S) is IL-7 containing the mutations F39P, F57N, and L128S; said molecule having the sequence as depicted in Fig. 34.

Documents:

02516-kolnp-2007-abstract.pdf

02516-kolnp-2007-claims.pdf

02516-kolnp-2007-correspondence others.pdf

02516-kolnp-2007-description complete.pdf

02516-kolnp-2007-drawings.pdf

02516-kolnp-2007-form 1.pdf

02516-kolnp-2007-form 2.pdf

02516-kolnp-2007-form 3.pdf

02516-kolnp-2007-form 5.pdf

02516-kolnp-2007-gpa.pdf

02516-kolnp-2007-international publication.pdf

02516-kolnp-2007-international search report.pdf

02516-kolnp-2007-pct request.pdf

02516-kolnp-2007-sequence listing.pdf

2516-KOLNP-2007-(20-06-2012)-CORRESPONDENCE.pdf

2516-KOLNP-2007-(20-06-2012)-FORM-1.pdf

2516-KOLNP-2007-(24-07-2012)-CORRESPONDENCE.pdf

2516-KOLNP-2007-(24-07-2012)-FORM-1.pdf

2516-KOLNP-2007-(30-03-2012)-ABSTRACT.pdf

2516-KOLNP-2007-(30-03-2012)-AMANDED CLAIMS.pdf

2516-KOLNP-2007-(30-03-2012)-CORRESPONDENCE.pdf

2516-KOLNP-2007-(30-03-2012)-DESCRIPTION (COMPLETE).pdf

2516-KOLNP-2007-(30-03-2012)-EXAMINATION REPORT REPLY RECEIVED.pdf

2516-KOLNP-2007-(30-03-2012)-FORM-1-1.pdf

2516-KOLNP-2007-(30-03-2012)-FORM-1.pdf

2516-KOLNP-2007-(30-03-2012)-FORM-2.pdf

2516-KOLNP-2007-(30-03-2012)-FORM-3.pdf

2516-KOLNP-2007-(30-03-2012)-OTHERS PCT FORM.pdf

2516-KOLNP-2007-(30-03-2012)-OTHERS.pdf

2516-KOLNP-2007-(30-03-2012)-PA-CERTIFIED COPIES.pdf

2516-KOLNP-2007-(30-03-2012)-PETITION UNDER RULE 137-1.pdf

2516-KOLNP-2007-(30-03-2012)-PETITION UNDER RULE 137.pdf

2516-kolnp-2007-form-18.pdf


Patent Number 254410
Indian Patent Application Number 2516/KOLNP/2007
PG Journal Number 44/2012
Publication Date 02-Nov-2012
Grant Date 31-Oct-2012
Date of Filing 06-Jul-2007
Name of Patentee MERCK PATENT GMBH
Applicant Address FRANKFURTER STRASSE 250 64293 DARMSTADT
Inventors:
# Inventor's Name Inventor's Address
1 GILLIES, STEPHEN, D 47 SWANSON LANE, CARLISLE, MA 01741
2 WAY, JEFFREY, C 108, FAYERWEATHER STREET, UNIT 2, CAMBRIDGE, MA 02138
PCT International Classification Number C07K 14/54
PCT International Application Number PCT/EP2005/013145
PCT International Filing date 2005-12-08
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/634,470 2004-12-09 U.S.A.