Title of Invention

ANTI-IL 17 ANTIBODIES

Abstract Anti-IL-17 antibodies are identified that are characterized as having a high affinity and slow off rate for human IL-17. The antibodies of the invention may be chimeric, humanized or fully human antibodies, immunoconjugates of the antibodies or antigen- binding fragments thereof. The antibodies of the invention are useful in particular for treating autoimmune, inflammatory, cell proliferative and developmental disorders.
Full Text ANTI-IL-ANTIBODIES
FIELD OF THE INVENTION
The present invention is in the field of medicine, particularly in the field of
monoclonal antibodies against human IL-17. The invention relates to neutralizing anti-
IL-17 monoclonal antibodies that bind with high affinity to an IL-17 non-linear or
conformational antigenic epitope comprising amino acids DGNVDYH. The antibodies of
the invention may be chimeric, humanized or human antibodies, immunoconjugates of
the antibodies or antigen-binding fragments thereof and are useful as a medicament for
the treatment of autoimmune, inflammatory, cell proliferative and developmental
disorders.
BACKGROUND OF THE INVENTION
The IL-17 family of cytokines presently includes EL-17A, IL-17B, IL-17C, IL-
17D, EL-17E and IL-17F. All IL-17 family members have four highly conserved cysteine
residues that are involved in the formation of intrachain disulfide linkages and have two
or more cysteine residues that may be involved in interchain disulfide linkages. Members
of the IL-17 family have no sequence similarity to any other known cytokines. However,
a viral homologue of IL-17A was found in open reading frame 13 of herpesvirus saimiri
(Yao, Z. et al., Immunity, 3:811,1995) and has 72% amino acid residue identity to human
IL-17 A. Multiple functions have been reported for the IL-17 family members that mainly
involve regulation of the immune response.
Interleukin 17 (IL-17, also referred to as IL-17 A) is a 20-30 kD homodimeric
glycoprotein produced predominantly by activated CD4+ T cells and functions as a
proinflammatory cytokine. When a particular IL-17 family member is referred to simply
as "IL-17," it is understood that the family member referred to is DL-17A. IL-17 is
secreted by activated T cells at sites of inflammation not in the systemic circulation. IL-
17 binds to a type I transmembrane receptor termed IL-17R which is a large ubiquitously
expressed protein that demonstrates no significant sequence similarity to other known
cytokine receptors. IL-17 has multiple biologic properties including upregulating
adhesion molecules and inducing the production of multiple inflammatory cytokines and
chemokines from various cell types including synoviocytes, chondroctes, fibroblasts,

endothelial cells, epithelial cells, keratinocytes, and macrophages. Also, IL-17 induces
recruitment of neutrophils to an inflammatory site through induction of chemokine
release, stimulates production of prostaglandins and metalloproteinases, and inhibits
proteoglycan synthesis. Furthermore, IL-17 plays an important role in the maturation of
hematopoietic progenitor cells. It has been demonstrated that IL-17 has signaling roles in
different organs and tissues including lung, articular cartilage, bone, brain, hematopoietic
cells, kidney, skin and intestine. For a review of IL-17 bioactivity see, e.g., Kolls and
Linden, Immunity 21:467-476,2004, or Fossiez, et al. Int. Rev. Immunol. 16:541,1998.
Increased levels of IL-17 (i.e., EL-17A) have been associated with several
conditions, diseases or disorders including airway inflammation, rheumatoid arthritis
("RA"), osteoarthritis, bone erosion, intraperitoneal abscesses and adhesions,
inflammatory bowel disorder ("IBD"), allograft rejection, psoriasis, certain types of
cancer, angiogenesis, atherosclerosis and multiple sclerosis ("MS") (for a review see
Witkowski, et al, Cell. Mot Life Sci. 61:567-579,2004). Both EL-17 and IL-17R are up-
regulated in the synovial tissue of RA patients. Blocking an IL-17 bioactivity by binding
an IL-17 specific antibody or soluble receptor to IL-17 reduces inflammation and bone
erosion in various animal arthritis models. (See, e.g., Lubberts et al, Arthritis &
Rheumatism, 50:650-659,2004). Furthermore, IL-17 has IL-ip independent effects on
collagen matrix breakdown and inflammation and joint damage, while IL-17 has synergy
with TNF-a to amplify inflammation.
Thus, given its localized distribution at the site of inflammation, IL-17 appears to
be a novel target for the treatment of RA and other inflammatory or autoimmune diseases
with a potentially greater safety profile than drugs that target the systemic circulation of
pro-inflammatory cytokines such as TNF-a. Current FDA approved byproducts
(ENBREL*, REMICADE* and HUMIRA* antibodies) that bind to and neutralize TNF-a
have demonstrated efficacy in reducing signs and symptoms of RA and in slowing
progression of the disease in a subset of RA patients. However, not all RA patients
respond equally to inhibition of a TNF-a bioactivity with these byproducts.
Additionally, IL-17 mRNA is increased in multiple sclerosis lesions and in mononuclear
cells in the blood and cerebrospinal fluid of MS patients, particularly during clinical
exacerbation. Accordingly, there is a need for compositions that antagonize or neutralize
the activity of IL-17 in order to treat disorders, diseases or conditions wherein the

presence of IL-17 bioactivity causes or contributes to an undesirable pathological effect
or wherein a decrease in IL-17 bioactivity contributes to a desirable therapeutic effect,
including inflammatory disorders, cell proliferative and developmental disorders and
autoimmune disorders such as RA and MS and IBD.
There is a need for a neutralizing anti-IL-17 antibody that specifically binds IL-17
of human origin as well as IL-17 of a non-human mammal thereby allowing the antibody
to be used in preclinical and clinical in vivo studies. Furthermore, there is a need for an
IL-17-specific antibody which binds EL-17 with a high affinity and/or has a slow off rate
thereby allowing the effective therapeutic dose to be minimized resulting in less frequent
dosing with such an antibody than with an antibody that binds IL-17 with a lesser affinity
(i.e., a higher KD) and/or has a faster off rate. A high affinity IL-17-specific antibody is
also desirable in that it may allow the antibody to be administered to a patient
subcutaneously rather than intravenously. There is also a need for an IL-17-specific
antibody with a low ICJO value in an IL-17 bioactivity assay in order to generate a
therapeutic anti-IL-17 antibody with a minimum effective therapeutic dose. It is also
desirable to provide an antibody specific to IL-17 where an immune response to the
antibody evoked by a patient receiving the antibody is reduced to a minimum. The
present invention satisfies these needs and provides related advantages.
SUMMARY OF THE INVENTION
Antibodies of the invention are chimeric, humanized, or fully human anti-IL-17
monoclonal antibodies, and antigen-binding portions thereof, that bind a non-linear
epitope comprising IL-17 amino acids DGNVDYH (SEQ ID NO: 276) and antagonize or
neutralize at least one in vitro or in vivo biological activity associated with IL-17 or a
portion thereof
In one embodiment, antibodies of the invention have an ICJO of less than or equal
to about 1 nM, 900 pM, 800 pM, 700 pM, 600pM, 560 pM or 500 pM in an in vitro IL-8
reporter assay as described, for example, in Example 6A herein or less than or equal to
560 pM in an in vitro GROa Reporter Assay as described, for example, in Example 6B
herein.
In another embodiment, antibodies of the invention are characterized by a strong
binding affinity (KD) for human EL-17, i.e., less than about 7 pM, 6.5 pM, 6.0 pM, 5.5

pM, 5.0 pM, 4.5 pM or 4.0 pM. Alternatively, the antibodies of the invention are
characterized by a KD for human IL-17 of no greater than about 7 pM, 6.5 pM, 6.0 pM,
5.5 pM, 5.0 pM, 4.5 pM or preferably no greater than about 4.0 pM. Preferably the
antibodies of the invention are further characterized with a karate from human IL-17 of
less than2 x 10s s"1.
In another embodiment, an anti-IL-17 antibody of the invention is characterized
by specifically binding human IL-17 as well as cynomoigus monkey IL-17 while not
binding mouse or rat IL-17 at levels greater than background. Additionally, an anti-IL-
17 antibody of the invention binds human IL-17 (i.e., IL-17A) but does not bind human
DL-17B, C, D.Eor F.
In one embodiment, an anti-IL-17 monoclonal antibody of the invention
comprises a light chain variable region ("LCVR") polypeptide comprising 3 CDR
sequences which are present together in a Fab listed in Table 3 hereinbelow and which
are present in the antibody of the invention in the same CDR position as in the Fab listed
in Table 3. Preferably an anti-IL-17 monoclonal antibody of the invention comprises a
LCVR polypeptide with an amino acid sequence selected from the group consisting of
SEQ ID NOs: 178-243.
In another embodiment, an anti-IL-17 monoclonal antibody of the invention
comprises a heavy chain variable region ("HCVR") polypeptide comprising 3 CDRs
which are present together in a Fab listed in Table 2 hereinbelow and which are present in
the antibody of the invention in the same CDR position as in the Fab listed in Table 2.
Preferably an anti-IL-17 monoclonal antibody of the invention comprises a HCVR
polypeptide with an amino acid sequence selected from the group consisting of SEQ ID
NOs: 56-121.
In another embodiment, an anti-IL-17 monoclonal antibody of the invention
comprises a LCVR polypeptide comprising 3 CDRs which are present together in a Fab
listed in Table 3 and which are present in the antibody of the invention in the same CDR
position as in the Fab listed in Table 3 and further comprises a HCVR polypeptide
comprising 3 CDRs which are present together in a Fab listed in Table 2 and which are
present in the antibody of the invention in the same CDR position as in the Fab listed in
Table 2. Preferably the 6 CDRs of an antibody of the invention, or functional fragment

thereof, exist together in a Fab listed in Table 1 hereinbelow and are present in the
antibody of the invention in the same CDR position as in the Fab listed in Table 1.
In a preferred embodiment, an anti-EL-17 monoclonal antibody of the invention
comprises (i) a LCVR polypeptide with an amino acid sequence selected from the group
consisting of SEQ ID NOs: 178-243 and (ii) a HCVR polypeptide with an amino acid
sequence selected from the group consisting of SEQ ID NOs: 56-121. In a more
preferred embodiment, an antibody of the invention comprising an LCVR polypeptide
with an amino acid sequence selected from the group consisting of SEQ ID NOs: 178-243
further comprises the HCVR polypeptide selected from the group consisting of SEQ ID
NOs: S6-121 that is present in a Fab listed in Table 1 that comprises the particular LCVR
present in the antibody.
In another embodiment, a monoclonal antibody of the invention is one which can
compete for binding to human EL-17, or a portion of human EL-17, with a competing
antibody wherein the competing antibody comprises two polypeptides with the amino
acid sequences of SEQ ID NOs: 241 and 118.
In another embodiment, a LCVR of an anti-IL-17 monoclonal antibody of the
invention comprises 1,2 or 3 peptides, preferably 3 peptides, selected from the group
consisting of peptides with a sequence as shown in (a) SEQ ID NOs: 122-149; (b) SEQ
ID NOs: 150-167, and (c) SEQ ID NOs:168-177 (i.e., one peptide from (a), one peptide
from (b) and one peptide from (c) for an antibody comprising 3 said peptides). A peptide
with the sequence shown in SEQ ID NOs: 122-149, when present in an antibody of the
invention, is at CDRL1. A peptide with the sequence shown in SEQ ID NOs: 150-167,
when present in an antibody of the invention, is at CDRL2. A peptide with the sequence
shown in SEQ ID NOs: 150-167, when present in an antibody of the invention, is at
CDRL3.
In another embodiment, a HCVR of an anti-IL-17 monoclonal antibody of the
invention comprises 1,2 or 3 peptides, preferably 3 peptides, selected from the group
consisting of peptides with a sequence as shown in (a) SEQ ID NOs: 11-28; (b) SEQ ID
NOs: 29-32, and (c) SEQ ID NOs: 33-55 and 261 (i.e., one peptide from (a), one peptide
from (b) and one peptide from (c) for an antibody comprising 3 said peptides). A peptide
with the sequence shown in SEQ ID NOs: 11-28, when present in said antibody, is at
CDRH1. A peptide with the sequence shown in SEQ ID NOs: 29-32, when present in

said antibody, is at CDRH2. A peptide with the sequence shown in SEQ ID NOs: 33-55
and 261, when present in said antibody, is at CDRH3.
The present invention further provides an anti-DL-17 monoclonal antibody
comprising six peptides selected from the group consisting of peptides with a sequence as
shown in (a) SEQ ID NOs: 122-149; (b) SEQ ID NOs: 150-167, (c) SEQ ID NOs:168-
177, (d) SEQ ID NOs: 11-28; (e) SEQ ID NOs: 29-32, and (f) SEQ ID NOs: 33-55 and
261 (i.e., one peptide from each of (a-f)); preferably the six peptides coexist in a Fab
listed in Table 1 herein. A peptide with the sequence shown in SEQ ID NOs: 122-149,
when present in an antibody of the invention, is at CDRL1. A peptide with the sequence
shown in SEQ ID NOs: 150-167, when present in an antibody of the invention, is at
CDRL2. A peptide with the sequence shown in SEQ ID NOs: 150-167, when present in
an antibody of the invention, is at CDRL3. A peptide with the sequence shown in SEQ
ID NOs: 11-28, when present in said antibody, is at CDRH1. A peptide with the
sequence shown in SEQ ID NOs: 29-32, when present in said antibody, is at CDRH2. A
peptide with the sequence shown in SEQ ID NOs: 33-55 and 261, when present in said
antibody, is at CDRH3.
The present invention further provides an anti-IL-17 monoclonal antibody
comprising the six peptides with the sequences as shown in SEQ ID NOs: 247,248,249,
244,245 and 246. The peptide with the sequence shown in SEQ ID NO: 247 is at
CDRL1. The peptide with the sequence shown in SEQ ID NO: 248 is at CDRL2. The
peptide with the sequence shown in SEQ ID NO: 249 is at CDRL3. The peptide with the
sequence shown in SEQ ID NO: 244 is at CDRH1. The peptide with the sequence shown
in SEQ ID NO: 245 is at CDRH2. The peptide with the sequence shown in SEQ ID NO:
246 is atCDRH3.
An anti-EL-17 monoclonal antibody of the invention may comprise or consist of
an intact antibody {i.e., full length), a substantially intact antibody or an antigen-binding
portion thereof, e.g., a Fab fragment, a F(ab*)2 fragment or a single chain Fv fragment
Furthermore, an antibody of the invention may be labeled with a detectable label,
immobilized on a solid phase and/or conjugated with a heterologous compound, e.g., an
enzyme, toxin or polyethylene glycol molecule.
In another embodiment, the invention provides a method of preparing an anti-IL-
17 monoclonal antibody of the invention comprising maintaining a host cell of the

invention (i.e., host cell that has been transformed, transduced or infected with a vector
(or vectors) of the invention expressing an antibody of the invention) under conditions
appropriate for expression of a monoclonal antibody of the invention, whereby such
antibody is expressed. The method may further comprise the step of isolating the
monoclonal antibody of the invention from the cell or preferably from the culture media
in which the cell is grown.
Diagnostic uses for monoclonal antibodies of the invention are contemplated In
one diagnostic application, the invention provides a method for determining the level of
IL-17 protein in a sample comprising exposing a sample to be tested to an anti-IL-17
antibody of the invention under binding conditions and determining specific binding of
the antibody to the sample. An anti-IL-17 antibody of the invention may be used to
determine the levels of IL-17 in test samples by comparing test sample values to a
standard curve generated by binding said antibody to samples with known amounts of IL-
17. The invention further provides a kit comprising an antibody of the invention and,
preferably, instructions for using the antibody to detect IL-17 protein in a sample.
The invention provides a composition, preferably a pharmaceutical composition,
comprising an anti-IL-17 monoclonal antibody of the invention. The pharmaceutical
composition of the invention may further comprise a pharmaceutically acceptable carrier,
excipient and/or diluent. In said pharmaceutical composition, the anti-IL-17 monoclonal
antibody of the invention is the sole active ingredient. Preferably the pharmaceutical
composition comprises a homogeneous or substantially homogeneous population of an
anti-IL-17 monoclonal antibody of the invention. The composition for therapeutic use is
physiologically compatible, sterile and may be lyophilized and optionally supplied with
an appropriate diluent
The invention provides a method of inhibiting at least one IL-17 bioactivity in an
animal, preferably a mammal, more preferably a human, in need thereof comprising
administering a therapeutically effective amount, or IL-17 neutralizing amount, of an
anti-IL-17 monoclonal antibody of the invention to said animal. The invention further
provides a method of treating a disease or disorder ameliorated by neutralizing or
antagonizing an IL-17 bioactivity, e.g., inhibition of signal transduction resulting from
the binding of IL-17 to its receptor, that comprises administering to a patient (e.g., a

human) in need of such treatment or prevention a therapeutically effective amount of IL-
17 neutralizing amount, of a monoclonal antibody of the invention.
The invention embodies an anti-DL-17 monoclonal antibody of the invention for
use in the manufacture of a medicament for administration to a mammal, preferably a
human, for the treatment of, e.g., an autoimmune disorder or inflammation disorder or
cell-proliferation disorder.
The invention further embodies an article of manufacture comprising a packaging
material and an antibody of the invention contained within said packaging material,
wherein the packaging material comprises a package insert which indicates that the
antibody specifically neutralizes an IL-17 activity or decreases the level of functional EL-
17 present in the system.
The invention further provides isolated nucleic acid molecules encoding an
antibody of the invention or light chain or heavy chain thereof; a vector (or vectors)
comprising said nucleic acid, optionally operably linked to control sequences recognized
by a host cell transformed with the vector; a host cell comprising that vector; a process
for producing an antibody of the invention comprising culturing the host cell so that the
nucleic acid is expressed and, optionally, recovering the antibody from the host cell
culture medium.
The invention further provides isolated nucleic acid molecules encoding
cynomolgus monkey IL-17 (SEQ ID NO: 253) or rabbit IL-17 (SEQ ID NO: 251); the IL-
17 protein encoded by the monkey or rabbit nucleic acid (SEQ ID NOs: 10 or 9
respectively); vectors comprising said nucleic acid molecule; host cell comprising said
vector; and a process for producing cynomolgus monkey IL-17 or rabbit IL-17.
BRIEF DESRIF1 ION OF THE DRAWINGS
FIG. 1 shows the amino acid sequence alignment of members of the human IL-17
family of proteins (IL-17, IL-17B, IL-17C, IL-17D, IL-17E and EL-17F).
FIG. 2 shows the amino acid sequence alignment of EL-17 from human, rabbit, rat,
cynomolgus monkey and murine species.

DETAILED DESCRIPTION OF THE INVENTION
The invention presents chimeric, humanized or fully human anti-IL-17
monoclonal antibodies, or antigen-binding portions thereof, able to neutralize or
antagonize at least one IL-17 activity in vitro and/or in vivo. Preferably, such antibodies
of the invention are further characterized as having an IC50 less than about 600 or S60 pM
in e.g., an in vitro IL-8 reporter assay or GROa reporter assay (see, e.g., Example 6)
and/or preferably having a strong binding affinity with IL-17 of less than 4 pM. The
antibodies of the invention are further characterized in that they specifically bind human
and cynomolgus monkey IL-17 (SEQ ID NOs: 1 and 10 respectively) but do not bind
murine or rat IL-17 (SEQ ID NOs: 7 and 8 respectively). The antigenic epitope to which
monoclonal antibodies of the invention bind is a non-linear epitope of human (and
monkey) IL-17 and comprises residues DGNVDYH (SEQ ID NO: 276) of IL-17. An
antibody of the invention makes contact with the peptide DGNVDYH when it is in the
context of the full-length IL-17.
Definitions
"Interleukin 17" also referred to as "IL-17" or "IL-17A" is a 20-30 kD
glycosylated homodimeric protein. The human IL-17 gene codes for a 155 amino acid
protein that has a 19 amino acid signal sequence and a 136 amino acid mature segment.
Human IL-17 shows amino acid sequence identity of 62.5% and 58% to the mouse and
rat amino acid IL-17 sequences, respectively as shown in Figure 2. Human IL-17 shows
amino acid sequence identity of 97.4% to the cynomolgus monkey IL-17.
A full-length antibody as it exists naturally is an immunoglobulin molecule
comprised of four peptide chains, two heavy (H) chains (about 50-70 kDa when full
length) and two light (L) chains (about 25 kDa when full length) interconnected by
disulfide bonds. The amino terminal portion of each chain includes a variable region of
about 100-110 or more amino acids primarily responsible for antigen recognition. The
carboxy-terminal portion of each chain defines a constant region primarily responsible for
effector function.
Light chains are classified as kappa or lambda and characterized by a particular
constant region. Each light chain is comprised of an N-terminal light chain variable
region (herein "LCVR") and a light chain constant region comprised of one domain, CL.

Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, and define the
antibody's isotype as IgG, IgM, IgA, IgD, and IgE, respectively and several of these may
be further divided into subclasses (isotypes) e.g., IgGi, IgG2, IgG3, IgG4, IgAi and IgA2.
Each heavy chain type is characterized by a particular constant region. Each heavy chain
is comprised of an N-terminal heavy chain variable region (herein "HCVR") and a heavy
chain constant region. The heavy chain constant region is comprised of three domains
(CHI, CH2, and CH3) for IgG, IgD, and IgA; and 4 domains (CHI, CH2, CH3, and CH4)
for IgM and IgE.
The HCVR and LCVR regions can be further subdivided into regions of
hypervariability, termed complementarity determining regions ("CDRs"), interspersed
with regions that are more conserved, termed framework regions ("FR"). Each HCVR
and LCVR is composed of three CDRs and four FRs, arranged from amino-terminus to
carboxy-terminus in the following order FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
For full-length antibodies of the invention the light chains preferably comprise,
downstream of FR4, a polypeptide with the sequence shown in SEQ ID NO: 277. For
full-length antibodies of the invention the heavy chains preferably comprise, downstream
of FR4, a polypeptide with the sequence shown in SEQ ID NO: 278. Herein the 3 CDRs
of the heavy chain are referred to as "CDRH1, CDRH2, and CDRH3" and the 3 CDRs of
the light chain are referred to as "CDRL1, CDRL2 and CDRL3." The CDRs contain
most of the residues which form specific interactions with the antigen. The numbering
and positioning of CDR amino acid residues within the HCVR and LCVR regions is in
accordance with the well-known Kabat numbering convention.
The term "antibody," in reference to an anti-IL-17 monoclonal antibody of the
invention (or simply "antibody of the invention"), as used herein, refers to a monoclonal
antibody. A "monoclonal antibody" as used herein refers to a rodent, preferably murine
antibody, a chimeric antibody, a humanized antibody or a fully human antibody, unless
otherwise indicated herein. Monoclonal antibodies of the invention can be produced
using e.g., hybridoma techniques well known in the art, as well as recombinant
technologies, phage display technologies, synthetic or recombinant technologies or
combinations of such technologies readily known in the art The term "monoclonal
antibody" as used herein is not limited to antibodies produced through hybridoma
technology. "Monoclonal antibody" refers to an antibody that is derived from a single

copy or clone, including e.g., eukaiyotic, prokaryotic, orphage clone, and not the method
by which it is produced. A "monoclonal antibody" can be an intact antibody (comprising
a complete or full-length Fc region), a substantially intact antibody, or a portion or
fragment of an antibody comprising an antigen-binding portion, e;g., a Fab fragment,
Fab' fragment or F(ab'>2 fragment of a murine antibody or of a chimeric, humanized or
human antibody. The "Fab" fragment contains a variable and constant domain of the light
chain and a variable domain and the first constant domain (CHI) of the heavy chain.
"F(abV antibody fragments comprise a pair of Fab fragments which are generally
covalently linked near their carboxy termini by hinge cysteines between them. Other
chemical couplings of antibody fragments are also known in the art.
The variable region of each light-heavy chain pair forms an antigen-binding site
of the antibody. Thus, an intact IgG antibody has two binding sites. Except in
bifunctional or bispecific antibodies, the two antigen-binging sites of the antibody are the
same. As used herein, the "antigen-binding portion" or "antigen-binding region" or
"antigen-binding domain" refers interchangeably to that portion of an antibody molecule
which contains the amino acid residues that interact with an antigen and confer on the
antibody its specificity and affinity for the antigen. This antibody portion includes the
"framework" amino acid residues necessary to maintain the proper conformation of the
antigen-binding residues. Preferably, the CDRs of the antigen-binding region of the
antibodies of the invention are entirely or substantially of murine origin, optionally with
certain amino acid residues altered, e.g., substituted with a different amino acid residue,
(see e.g., Tables 2 and 3) to optimize a particular property of the antibody, e.g., KD, KB,
ICso. Preferably the framework regions of antibodies of the invention are of human origin
or substantially of human origin (at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99%
of human origin. Preferred framework regions of antibodies of the invention have the
following sequences: SEQ ID NOs: 262 (HCVR FR1X 263 (HCVR FR2), 264 (HCVR
FR3), 265 (HCVR FR4), 266 (LCVR FR1), 267 (LCVR FR2), 268 (LCVR FR3), 269
(LCVR FR4) and follow Kabat numbering. In other embodiments, the antigen-binding
region of an IL-17 antibody of the invention can be derived from other non-human
species including, but not limited to, rabbit, rat or hamster. Alternatively, the antigen-
binding region can be derived from human sequence.

Furthermore, a "monoclonal antibody" as used herein can be a single chain Fv
fragment that may be produced by joining the DNA encoding a LCVR and the DNA
encoding a HCVR with a linker sequence. (See, Pluckthun, The Pharmacology of
Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New
York, pp 269-315,1994). It is understood that regardless of whether fragments are
specified, the term "antibody" as used herein includes such fragments as well as single
chain forms. As long as the protein retains the ability to specifically or preferentially
bind its intended target {i.e., epitope or antigen), it is included within the term "antibody."
Antibodies may or may not be glycosylated and still fall within the bounds of the
invention.
A population of "monoclonal antibodies," refers to a homogeneous or
substantially homogeneous antibody population (i.e., at least about 85%, 90%, 91%, 92%,
93%, 94%, 95%, 96%, more preferably at least about 97% or 98% or most preferably at
least 99% of the antibodies in the population would compete in an ELISA assay for the
same antigen or epitope or more preferably the antibodies are identical in amino acid
sequence. Antibodies may or may not be glycosylated and still fall within the bounds of
the invention. Monoclonal antibodies may be homogeneous if they have identical amino
acid sequence although they may differ in a post-translational modification, e.g.,
glycosylation pattern.
A 'Variant" antibody, refers herein to a molecule which differs in amino acid
sequence from a "parent" antibody amino acid sequence by virtue of addition, deletion
and/or substitution of one or more amino acid residue(s) of the parent antibody sequence.
In a preferred embodiment, the variant antibody comprises at least one amino acid (e.g.,
from one to about ten, and preferably 2,3,4,5,6,7 or 8) addition, deletion and/or
substitution in the CDR regions of the parent antibody. Identity or homology with
respect to the variant antibody sequence is defined herein as the percentage of amino acid
residues in the variant antibody sequence that are identical with the parent antibody
residues after aligning the sequences and introducing gaps, if necessary, to achieve the
maximum percent sequence identity. The variant antibody retains the ability to bind the
antigen, or preferably, the epitope, to which the parent antibody binds and preferably has
at least one property or bioactivity that is superior to that of the parent antibody. For
example, the variant antibody preferably has stronger binding affinity, slower off-rate,

lower IC50 or enhanced ability to inhibit an antigen bioactivity than does the parent
antibody. A variant antibody of particular interest herein is one which displays at least
about 2-fold, preferably at least about S-fold, 10-fold or 20-fold enhancement in a
property or bioactivity when compared to the parent antibody.
The "parent" antibody herein is one which is encoded by an amino acid sequence
used for the preparation of a variant antibody. The parent antibody may have framework
sequence of murine origin, but preferably the framework sequence is entirely or
substantially of human origin. The parent antibody may be a murine, chimeric,
humanized or human antibody.
The term "specifically binds" as used herein refers to the situation in which one
member of a specific binding pair does not significantly bind to molecules other than its
specific binding partner(s). The term is also applicable where e.g., an antigen-binding
domain of an antibody of the invention is specific for a particular epitope that is carried
by a number of antigens, in which case the specific antibody carrying the antigen-binding
domain will be able to bind to the various antigens carrying the epitope. Accordingly a
monoclonal antibody of the invention specifically binds human IL-17 (i.e., IL-17A) while
it does not specifically bind human nL-17B,IL-17C,IL-17D,IL-17E,IL-17F. Further, a
monoclonal antibody of the invention specifically binds human IL-17 and cynomolgus
monkey IL-17 but does not specifically bind rat IL-17 or murine IL-17. Further a
monoclonal antibody of the invention specifically binds a non-linear or conformational
human IL-17 epitope comprising amino acids DGNDYH but does not bind a human IL-
17 epitope which does not comprise amino acids DGNDYH.
The term "preferentially binds" as used herein, refers to the situation in which an
antibody binds a specific antigen at least about 20% greater, preferably at least about
50%, 2-fold, 20-fold, 50-fold or 100-fold greater than it binds a different antigen as
measured by a technique available in the art, e.g., competition ELISA or Ko measurement
with a BIACORE or KINEXA assay. An antibody may preferentially bind one epitope
within an antigen over a different epitope within the same antigen. Accordingly an
antibody of the invention preferentially binds human IL-17 over rabbit IL-17.
The term "epitope" refers to that portion of a molecule capable of being
recognized by and bound by an antibody at one or more of the antibody's antigen-binding
regions. Epitopes often consist of a chemically active surface grouping of molecules such

as amino acids or sugar side chains and have specific three-dimensional structural
characteristics as well as specific charge characteristics. By "inhibiting epitope" and/or
"neutralizing epitope" is intended an epitope, which when in the context of the intact
antigenic molecule and when bound by an antibody specific to the epitope, results in loss
or diminution of a biological activity of the molecule in vivo or in vitro or in an organism
containing the molecule.
The term "epitope," as used herein, further refers to a portion of a polypeptide
having antigenic and/or immunogenic activity in an animal, preferably a mammal, e.g., a
mouse or a human. The term "antigenic epitope," as used herein, is defined as a portion
of a polypeptide to which an antibody can specifically bind as determined by any method
well known in the art, for example, by conventional immunoassays. Antigenic epitopes
need not necessarily be immunogenic, buy may be immunogenic. An "immunogenic
epitope," as used herein, is defined as a portion of a polypeptide that elicits an antibody
response in an animal, as determined by any method known in the art A "nonlinear
epitope" or "conformational epitope" comprises noncontiguous polypeptides (or amino
acids) within the antigenic protein to which an antibody specific to the epitope binds.
The phrases "biological property" or "biological characteristic," or the terms
"activity" or "bioactivity," in reference to an antibody of the present invention, are used
interchangeably herein and include, but are not limited to, epitope/antigen affinity and
specificity, ability to neutralize or antagonize an activity of EL-17 in vivo or in vitro, ICso,
in vivo stability of the antibody and the immunogenic properties of the antibody. Other
identifiable biological properties or characteristics of an antibody recognized in the art
include, for example, cross-reactivity, (ie., with non-human homologs of the targeted
peptidc, or with other proteins or tissues, generally), and ability to preserve high
expression levels of protein in mammalian cells. The aforementioned properties or
characteristics can be observed, measured or assessed using art-recognized techniques
t
including, but not limited to, ELISA, competitive ELISA, BIACORE or KINEXA surface
plasmon resonance analysis, in vitro or in vivo neutralization assays without limit,
receptor binding, cytokine or growth factor production and/or secretion, signal
transduction and immunohistochemistry with tissue sections from different sources
including human, primate, or any other source.

The term "inhibit" or "neutralize" as used herein with respect to an activity of an
antibody of the invention means the ability to substantially antagonize, prohibit, prevent,
restrain, slow, disrupt, eliminate, stop, or reverse e.g., progression or severity of that
which is being inhibited including, but not limited to, a biological activity (e.g., an IL-17
activity) or property, a disease or a condition. The inhibition or neutralization of an IL-
17 activity resulting from binding of an antibody of the invention with IL-17 is preferably
at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or higher.
The term "isolated" when used in relation to a nucleic acid or protein (e.g., an
antibody) refers to a nucleic acid molecule or protein that is identified and separated from
at least one contaminant with which it is ordinarily associated in its natural source.
Preferably, an "isolated antibody" is an antibody that is substantially free of other
antibodies having different antigenic specificities (e.g., pharmaceutical compositions of
the invention comprise an isolated antibody that specifically binds IL-17 and is
substantially free of antibodies that specifically bind antigens other than IL-17).
The terms "Kabat numbering" and "Kabat labeling" are used interchangeably
herein. These terms, which are recognized in the art, refer to a system of numbering
amino acid residues which are more variable (i.e., hypervariable) than other amino acid
residues in the heavy and light chain variable regions of an antibody (Kabat, et al., Ann.
NYAcad. Sci. 190:382-93 (1971); Kabat, et al, Sequences of Proteins o/Immunologicat
Interest, Fifth Edition, U.S. Department of Health and Human Services, NEK Publication
No. 91-3242 (1991)).
A polynucleotide is "operably linked" to another polynucleotide when it is placed
into a functional relationship with the other polynucleotide. For example, a promoter or
enhancer is operably linked to a coding sequence if it affects the transcription of the
sequence. A peptide is "operably linked" to another peptide when the polynucleotides
encoding them are operably linked, preferably they are in the same open reading frame.
The terms "individual," "subject," and "patient," used interchangeably herein,
refer to a mammal, including, but not limited to, murines, simians, humans, mammalian
farm animals, mammalian sport animals, and mammalian pets; preferably the term refers
to humans. In a certain embodiment, the subject, preferably a mammal, preferably a
human, is further characterized with a disease or disorder or condition that would benefit
from a decreased bioactivity of DL-17.

The term "vector" includes a nucleic acid molecule capable of transporting
another nucleic acid to which it has been linked including, but not limited to, plasmids
and viral vectors. Certain vectors are capable of autonomous replication in a host cell into
which they are introduced while other vectors can be integrated into the genome of a host
cell upon introduction into the host cell, and thereby, are replicated along with the host
genome. Moreover, certain vectors are capable of directing the expression of genes to
which they are operably linked. Such vectors are referred to herein as "recombinant
expression vectors" (or simply "expression vectors") and exemplary vectors are well
known in the art.
As used herein, the expressions "cell," "host cell," "cell line," and "cell culture"
are used interchangeably and include an individual cell or cell culture that is a recipient of
any isolated polynucleotide of the invention or any recombinant vectors) comprising a
sequence encoding a HCVR, LCVR or monoclonal antibody of the invention. Host cells
include progeny of a single host cell, and the progeny may not necessarily be completely
identical (in morphology or in total DNA complement) to the original parent cell due to
natural, accidental, or deliberate mutation and/or change. A host cell includes cells
transformed, transduced or infected with a recombinant vector or a polynucleotide
expressing a monoclonal antibody of the invention or a light chain or heavy chain thereof.
A host cell which comprises a recombinant vector of the invention, either stably
incorporated into the host chromosome or not, may also be referred to as a "recombinant
host cell". Preferred host cells for use in the invention are CHO cells (e.g., ATCC CRL-
9096), NSO cells, SP2/0 cells, COS cells (ATCC e.g., CRL-1650, CRL-1651) and HeLa
(ATCC CCL-2). Additional host cells for use in the invention include plant cells, yeast
cells, other mammalian cells and prokaryotic cells.
Antibody Characterization
The present invention relates to isolated, monoclonal antibodies that specifically
bind human EL-17 (i.e., IL-17A) with high affinity. The antibodies of the invention are
preferably chimeric, humanized or human antibodies or antigen-binding portions thereof.
Furthermore, antibodies of the invention neutralize or antagonize at least one EL-17
biological activity in vivo and/or in vitro. Specific binding of an anti-IL-17 monoclonal
antibody of the invention, (including antigen-binding portions thereof) to DL-17 allows

said antibody to be used as a therapeutic for IL-17-associated diseases and disorders, i.e.,
conditions, diseases or disorders which benefit from inhibition of an JL-17 biological
activity.
The antigenic IL-17 epitope to which the antibodies of the invention bind is a non-
linear epitope that comprises amino acids ADGNVDYHMN (SEQ ID NO: 266), more
preferably amino acids DGNVDYH (SEQ ID NO: 267) of human IL-17. Antibodies
which bind said epitope, specifically and preferentially bind human IL-17 and
cynomolgus monkey IL-17 as compared to their binding murine IL-17 or rat EL-17. The
monoclonal antibodies of the invention bind human IL-17 at least 5, 10, 20, 30,40, 50,
60, 70, 80, 90, or 100-fold greater (e.g., greater affinity or greater specificity) than with
which it binds murine IL-17 or rat DL-17; more preferably at least 150, 200, 250, 300,
350, 400,450, 500, 550 or 600-fold greater than with which it binds murine IL-17 or rat
IL-17 even more preferably it does not bind murine IL-17 or rat EL-17 at levels greater
than background levels as determined e.g., by ELISA assay, competition ELISA assay or
KD values in a BIACORE or KINEXA assay.
In a preferred embodiment, the invention provides an anti-IL-17 monoclonal
antibody that possesses a strong binding affinity for human EL-17, i.e., binds human IL-
17, or a portion thereof comprising DGNVDYH (SEQ ID NO: 267) [i.e., antibody
contacts the DGNVDYH polypeptide], with a binding affinity (Ko) for human IL-17 of
less than about 7 pM, 6.5 pM or 6 pM, preferably less than about 5.5 pM, 5 pM or 4.5 pM
and most preferably less than about 4 pM. Alternatively, the antibodies of the invention
are characterized by a KD for human IL-17 of no greater than about 7 pM, 6.5 pM or 6
pM, preferably no greater than about 5.5 pM, 5 pM or 4.5 pM and most preferably no
greater than about 4 pM. Antibody affinities may be determined as described in the
examples hereinbelow or other methods available in the art Preferably the anti-IL-17
antibodies of the invention which possess a strong binding affinity as described above
also bind a non-linear human IL-17 epitope (hat comprises amino acids
ADGNVDYHMN (SEQ ID NO: 266), more preferably amino acids DGNVDYH (SEQ
ID NO: 267), wherein the antibody makes contact with the polypeptide DGNVDYH.
In one embodiment, the antibodies of the invention have an off rate (koff) for
human IL-17 of less than 5 x 10s, 4 x 105, 3 x 105 or 2 x 10'5 s-1. In a preferred
embodiment, the antibodies of the invention characterized by possessing a strong binding

affinity for human IL-17 as described above (KD less than about 7 pM or 6 pM,
preferably less than about 5 pM or 4.5 pM and most preferably less than about 4 pM) also
have an off rate (lea) for human IL-17 of less than 5 x 10"5,4 x 1O'$, 3 x 10'5 or 2 x 10'5 s'
1 and even more preferably also bind a non-linear human IL-17 epitope that comprises
amino acids ADGNVDYHMN (SEQ ID NO: 266), more preferably amino acids
DGNVDYH (SEQ ID NO: 267) of human IL-17.
In another embodiment, the antibodies of the invention have an ICso of less than
lnM, 900 pM, 800 pM, 700 pM, 650 pM, 600 pM, 560 pM, 550 pM or 500 pM in e.g., an
in vitro IL-8 reporter assay or less than about 560 pM in a GROa reporter assay (see
Example 6). In a preferred embodiment, the antibodies of the invention are characterized
by possessing a strong binding affinity for human IL-17 as described above (KD less than
about 7 pM or 6 pM, preferably less than about 5 pM or 4.5 pM and most preferably less
than about 4 pM) and also have an ICS0 of less than lnM, 900 pM, 800 pM, 700 pM, 650
pM, 600 pM, 560 pM, 550 pM or 500 pM in e.g., an in vitro IL-8 reporter assay or less
than about 560 pM in a GROa reporter assay and even more preferably also have an off
rate (lea) for human IL-17 of less than 5 x 10"5,4 x 10'5,3 x 10s or 2 x 10"s s'1 and even
more preferably also bind a non-linear human IL-17 epitope that comprises amino acids
DGNVDYH (SEQ ID NO: 267) of human IL-17 wherein the antibody contacts the
DGNVDYH polypeptide.
The most preferred embodiment of the invention is an anti-IL-17 antibody
comprising a light chain amino acid sequence consisting of SEQ ID NO: 279 and a heavy
chain amino acid sequence consisting of SEQ ID NO: 280. Preferably this antibody
comprises two identical light chains and two identical heavy chains. Preferably the light
chain with amino acid sequence as shown in SEQ ID NO: 279 is encoded by a nucleic
acid comprising the sequence shown in SEQ ID NO: 281 (including signal sequence) or
SEQ ID NO: 283 (without the signal sequence). Preferably the heavy chain with amino
acid sequence as shown in SEQ ID NO: 280 is encoded by a nucleic acid comprising the
sequence shown in SEQ ID NO: 282 (including the signal sequence) or SEQ ID NO: 284
(without the signal sequence).
Monoclonal antibodies ("mAbs") may be made using the hybridoma method
widely known in the art (see e.g., Kohler et al., Nature, 256:495, 1975) or may be made
by recombinant DNA methods (e.g., as in U.S. Patent No. 4,816,567). Generally, a

hybridoma can be produced by fusing a suitable immortal cell line (e.g., a myeloma cell
line such as SP2/0) with antibody producing cells of the immunized animal. The
antibody producing cell, preferably those of the spleen or lymph nodes, are obtained from
animals immunized with the antigen of interest. The fused cells (hybridomas) can be
isolated using selective culture conditions, and cloned by limiting dilution. Culture
medium in which hybridoma cells are growing is assayed for production of monoclonal
antibodies directed against the antigen. Preferably, the binding specificity of mAbs
produced by hybridoma cells is determined by immunoprecipitation or by an in vitro
binding assay, such as radioimmunoassay or ELISA. Cells which produce antibodies
with the desired binding properties can be selected by a suitable assay. Methods for such
isolation and screening are well known in the art.
Other suitable methods of producing or isolating antibodies of the invention,
including human or artificial antibodies, can be used, including, for example, methods
which select a recombinant antibody (e.g., single chain Fv or Fab) from a library, or
which rely upon immunization of transgenic animals (e.g., mice) capable of producing a
repertoire of human antibodies (see e.g., Jakobovits etal, Proc. Natl. Acad. Sci. USA,
90:2551-2555,1993; Jakobovits etal, Nature, 362:255-258, 1993; U.S. Patent Numbers
5,545,806 and 5,545,807).
Single chain antibodies, and chimeric, humanized or primatized (CDR-grafted)
antibodies, as well as chimeric or CDR-grafted single chain antibodies, and the like,
comprising portions derived from different species, are also encompassed by the present
invention and the term "antibody". The various portions of these antibodies can be joined
together chemically by conventional techniques, synthetically, or can be prepared as a
contiguous protein using genetic engineering techniques. For example, nucleic acids
encoding a chimeric or humanized chain can be expressed to produce a contiguous
protein. See e.g., U.S. Patent No. 4,816,567; European Patent No. 125,023 Bl; U.S.
Patent No. 4,816,397; European Patent No. 120,694 Bl; WO 86/01533; European Patent
No. 194,276 Bl; U.S. Patent No. 5,225,539; European Patent No. 239,400 Bl and U.S.
Patent Nos. 5,585,089 and 5,698,762.
In addition, functional fragments of antibodies (i.e., antigen-binding fragments),
including fragments of chimeric, humanized, primatized or single chain antibodies, can
also be produced and fall within the scope of the invention. Preferred functional

fragments retain an antigen-binding function of a corresponding full-length antibody.
Particularly preferred functional fragments retain the ability to inhibit one or more
functions or bioactivities characteristic of a mammalian mature EL-17, preferably human
IL-17, such as a binding activity, a signaling activity, and/or stimulation or inhibition of a
cellular response. For example, in one embodiment, a functional fragment can inhibit the
interaction of mature IL-17 with its receptor and/or can inhibit one or more receptor-
mediated functions.
Antibody portions capable of binding to human IL-17 include, but are not limited
to, Fv, Fab, Fab' and F(ab')2 fragments and are encompassed by the invention. Such
fragments can be produced by enzymatic cleavage or by recombinant techniques. For
instance, papain or pepsin cleavage of an intact antibody can generate Fab or F(ab')2
fragments, respectively. Papain digestion of antibodies produces two identical antigen-
binding fragments, called "Fab" fragments, each with a single antigen-binding site. The
Fab fragment also contains the constant domain of the light chain and the first constant
domain (CHI) of the heavy chain. Pepsin treatment yields an F(ab)2 fragment that has
two antigen-combining sites and is still capable of cross-linking antigen.
"Fv" is the minimum antibody fragment which contains a complete antigen-
recognition and -binding site. This region consists of a dimer of one heavy- and one
light-chain variable domain in tight, non-covalent association. It is in this configuration
that the three CDRs of each variable domain interact to define an antigen-binding site on
the surface of the VH-VL dimer. Collectively, the six CDRs confer antigen-binding
specificity to the antibody. To overcome the tendency of non-covalently linked HCVR
and LCVR domains in the Fv to dissociate when co-expressed in a host cell, a single
chain Fv fragment (scFv) can be constructed in which a flexible and adequately long
polypeptide links either the C-terminus of the HCVR to the N-terminus of the LCVR or
the C-terminus of the LCVR to the N-terminus of the HCVR, A commonly used linker is
a 15-residue (Gly4Ser)3 peptide. For a review of sFv, see Pluckthun in The
Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-
Verlag, New York, pp. 269-315 (1994). Antibodies can also be produced in a variety of
truncated forms using antibody genes in which one or more stop codons has been
introduced upstream of the natural stop site. For example, a chimeric gene encoding a

F(ab*)2 heavy chain portion can be designed to include DNA sequences encoding the CH1
domain and hinge region of the heavy chain.
Selection of antibody fragments from libraries using enrichment technologies such
as phage-display (Matthews DJ and Wells JA. Science. 260:1113-7,1993), ribosome
display (Hanes, et al., Proa Natl. Acad. Sci., (USA) 95:14130-5,1998), bacterial display
(Samuelson P., et al., Journal of Biotechnology. 96:129-54,2002) or yeast display (Kieke
M.C., et al., Protein Engineering, 10:1303-10,1997) have proven to be successful
alternatives to classical hybridoma technology (Review: Little M. et a]., Immunology
Today, 21:364-70,2000).
Variant Antibodies
A murine monoclonal antibody or a human antibody (produced e.g., in a
transgenic mouse) raised against IL-17 may be a parent antibody. A parent antibody may
be further altered to create a chimeric or humanized form of the antibody or other variant
form of the antibody using methods available in the art, e.g., PCR mutagenesis. Such
chimeric, humanized, or otherwise variant antibodies, may serve as parent antibodies for
further variation or mutagenesis. Parent antibodies of the invention may be mutagenized,
e.g., within the CDR domain(s) (see, e.g., Tables 2 and 3) to create variant antibodies that
may be screened for presence of a property of interest, e.g., binding affinity (lower KD),
IC50, specificity, preferential binding, etc. Preferably the property of interest in the
variant antibody is an improvement over that property in the parent antibody. An amino
acid substitution variant antibody is preferred and has at least 1,2,3,4,5,6,7,8,9 or 10
amino acid residues) of the parent antibody molecule removed and a different residue
inserted in its place. The site of greatest interest for substitutional mutagenesis is one or
more CDR regions, but FR alterations are also contemplated. Conservative amino acid
substitutions are preferred; although, for more substantial changes, non-conservative
amino acid changes may be introduced and the resulting antibodies screened for the
property of interest.
A convenient way for generating substitution variants of a parent antibody is
affinity maturation using phage display. Briefly, a polynucleotide molecule encoding a
parent antibody is mutated within one or more CDR regions to generate all possible
amino acid substitutions at each amino acid residue at which a substitution is desired.

The antibody variants thus generated are displayed in a monovalent fashion from
filamentous phage particles as fusions to the gene III product of M13 packaged within
each particle. The phage-displayed variant antibodies are then screened for their
biological activity (e.g., binding affinity, specificity, IC50)- In order to identify candidate
CDR region sites for modification, alanine scanning mutagens can be performed to
identify CDR region residues contributing significantly to antigen binding.
Alternatively, or in addition, it may be beneficial to analyze a crystal structure of
the antigen-antibody complex to identify contact points between the antibody and IL-17.
Such contact residues and neighboring residues are candidates for substitution according
to the techniques elaborated herein or known in the art Alternatively, or in addition,
random mutagenesis or point mutagenesis may be performed, on one or more
polynucleotide molecules encoding at least one CDR. The mutagenesis may be
performed, at one or more positions, either while the CDR is operably linked to the
framework region within the variable region or while the CDR is independent of other
variable region sequence and then the altered CDR returned to the variable region using
recombinant DNA technology. Once such variant antibodies are generated the panel of
variants is subjected to screening for a property or activity of interest and antibodies with
superior properties in one or more relevant assays may be selected for further
development.
Any cysteine residue not involved in maintaining the proper conformation of an
anti-IL-17 antibody of the invention may be substituted, generally with serine, to improve
the oxidative stability of the molecule and prevent aberrant crosslinking. Conversely,
cysteine bond(s) may be added to the antibody to improve its stability (particularly where
the antibody is an antibody fragment such as an Fv fragment).
Another type of amino acid variant of the antibody alters the original
glycosylation pattern of the antibody. By altering is meant deleting one or more
carbohydrate moieties found in the antibody, and/or adding one or more glycosylation
sites that are not present in the parent antibody. Glycosylation of antibodies is typically
either N-linked or O-linked. N-linked refers to the attachment of the carbohydrate moiety
to the side chain of an asparagine residue. The tripeptide sequences asparagine-X-serine
and asparagine-X-threonine, where X is any amino acid except proline, are the
recognition sequences for enzymatic attachment of the carbohydrate moiety to the

asparagines side chain. Thus the presence of either of these tripeptide sequences in a
polypeptide creates a potential glycosylation site. O-linked glycosylation refers to the
attachment of one of the sugars N-aceylgalactosamine, galactose, or xylose to a
hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-
hydroxylysine may also be used.
Addition of glycosylation sites to the antibody is conveniently accomplished by
altering the amino acid sequence such that it contains one or more of the above-described
tripeptide sequences (for N-linked glycosylation sites). The alteration may also be made
by the addition of, or substitution by, one or more serine or threonine residues to the
sequence of the original antibody (for O-linked glycosylation sites).
Sequence
A preferred monoclonal antibody of the invention comprises a LCVR comprising
a peptide with a sequence selected from the group consisting of SEQ ID NOs: 178-243
and/or a HCVR comprising a peptide with a sequence selected from the group consisting
of SEQ ID NOs: 56-121. In a preferred embodiment, an antibody of the invention
comprises a LCVR comprising a peptide with a sequence selected from the group
consisting of SEQ ID NOs: 178-243 and further comprises a HCVR comprising a peptide
with a sequence selected from the group consisting of SEQ ID NOs: 56-121, wherein the
HCVR and LCVR present in an antibody of the invention exist together in a Fab listed in
Table 1. For example, an antibody of the invention comprising an LCVR polypeptide
with an amino acid sequence of SEQ ID NO: 178, preferably further comprises a HCVR
polypeptide comprising an amino acid sequence selected from the group consisting of
SEQ ID NOs: 56,60, 68-93 and 95. Furthermore, an antibody of the invention
comprising an LCVR polypeptide with an amino acid sequence of SEQ ID NO: 241,
preferably further comprises a HCVR polypeptide comprising an amino acid sequence
selected from the group consisting of SEQ ID NOs: 118 and 106. The skilled artisan will
appreciate that the antibodies of the invention are not limited to the specific sequences of
HCVR and LCVR as listed in Table 1 herein, but also include variants of these sequences
that, when present in an anti-IL-17 antibody of the invention, retain or improve antigen
binding ability and at least one other functional property of the parent antibody, e.g.,

epitope specificity, ability to compete with the parent antibody for binding to IL-17, IC50
and/or KD or koff values for binding human IL-17.
Furthermore, a monoclonal antibody of the invention is one that is competitively
inhibited from binding human IL-17 (or a portion thereof comprising DGNVDYH) by a
competing monoclonal antibody wherein the competing monoclonal antibody comprises
two polypeptides with the amino acid sequences shown in SEQ ID NOs: 241 (LCVR) and
118 (HCVR). Such competitive inhibition between antibodies may be measured by
assays in the art, e.g., a competition ELISA assay.
Preferably, an antibody of the invention which competes with the competing
antibody defined above is further characterized by specifically binding human IL-17 but
not binding human IL-17B, IL-17C, IL-17D, IL-17E or IL-17F. Additionally, the
antibody is further characterized by specifically binding human IL-17 and cynomolgus
monkey IL-17 but not binding rat IL-17 or mouse IL-17 at levels greater than
background.
More preferably, an antibody of the invention which competes for binding to human IL-
17 with a competing antibody comprising amino acid sequences shown in SEQ ID NOs:
241 and 118 is further characterized by binding a human IL-17 nonlinear epitope
comprising amino acids DGNVDYH (SEQ ID NO: 276). Even more preferably, an
antibody of the invention which competes for binding to human IL-17 with a competing
antibody comprising amino acid sequences shown in SEQ ID NOs: 241 and 118 is further
characterized by having a KD for human IL-17 of less than about 7 pM, 6.5 pM or 6 pM,
preferably less than about S.S pM, S pM or 4.S pM and most preferably less than about 4
pM and/or is characterized by an IC50, preferably in an in vitro IL-8 reporter assay, that is
less than 700 pM, 650 pM, 600 pM, 560 pM, 550 pM or 500 pM, or by an IC50 in an in
vitro GROα reporter assay of less than about 560 pM, and/or has an off rate (koff) for
human IL-17 of less than 5 x 10-5, 4 x 10-5, 3 x 10-5 or 2 x 10-5 8-1.
In one embodiment, an anti-IL-17 antibody of the invention has a heavy chain
variable region and a light chain variable region, wherein the heavy chain variable region
comprises CDR regions with the following ammo acid sequences: CDRH1 (SEQ ID NO:
244), CDRH2 (SEQ ID NO: 245), and CDRH3 (SEQ ID NO: 246); and/or wherein the
light chain variable region comprises CDR regions with the following amino acid
sequences: CDRL1 (SEQ ID NO: 247), CDRL2 (SEQ ID NO: 248) and CDRL3 (SEQ

ID NO: 249). Preferably, the six CDRs of an antibody of the invention exist together as
in a Fab listed in Table 1 herein. Even more preferably, the heavy chain CDRS are in the
context of the following framework sequences: FR1 with SEQ ID NO: 262, FR2 with
SEQ ID NO: 263, FR3 with SEQ ID NO: 264 and FR4 with SEQ ID NO: 265 and the
light chain CDRs are in the context of the following framework sequences: FR1 with
SEQ ID NO: 266, FR2 with SEQ ID NO: 267, FR3 with SEQ ID NO: 268 and FR4 with
SEQ ID NO: 269, wherein the order from the amino terminus is FR1-CDR1-FR2-CDR2-
FR3-CDR3-FR4.
It is further contemplated that an anti-IL-17 antibody of the invention comprises a
HCVR comprising a CDRH1 comprising a sequence selected from the group consisting
of SEQ ID NOs: 11 -28, and/or a CDRH2 comprising a sequence selected from the group
consisting of SEQ ID NOs: 29-32, and/or a CDRH3 comprising a sequence selected from
the group consisting of SEQ ID NOs: 33-55 and 261. In another embodiment, an anti-IL-
17 antibody of the invention comprises a LCVR comprising a CDRL1 comprising a
sequence selected from the group consisting of SEQ ID NOs: 122-149, and/or a CDRL2
comprising a sequence selected from the group consisting of SEQ ID NOs: 150-167,
and/or a CDRL3 comprising a sequence selected from the group consisting of SEQ ID
NOs: 168-177. In a preferred embodiment, an anti-IL-17 antibody of the invention
comprises a HCVR comprising a CDRH1 comprising a sequence selected from the group
consisting of SEQ ID NOs: 11-28, and/or a CDRH2 comprising a sequence selected from
the group consisting of SEQ ID NOs: 29-32, and/or a CDRH3 comprising a sequence
selected from the group consisting of SEQ ID NOs: 33-55 and 261, and further comprises
a LCVR comprising a CDRL1 comprising a sequence selected from the group consisting
of SEQ ID NOs: 122-149, and/or a CDRL2 comprising a sequence selected from the
group consisting of SEQ ID NOs: 150-167, and/or a CDRL3 comprising a sequence
selected fiom the group consisting of SEQ ID NOs: 168-177.
The composition comprising a CDR of the invention will generally be an antibody
heavy or light chain sequence or a substantial portion thereof, in which the CDR is
located at a location consistent with Kabat numbering. The three CDR regions for each
chain, heavy an light, are provided in a framework region as a contiguous sequence
represented by the following formula: FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4. The
heavy chain or light chain FR1, FR2, FR3 and FR4 combine to form the complete

framework region of an antibody when arranged as a contiguous sequence with the CDRs
in the order stated. Preferably the framework regions of an antibody of the invention are
of human origin or substantially of human origin (i.e., greater than about 80,82,85, 87,
90, 92,95,97%).
In a humanized antibody for therapeutic use in humans, the framework sequence
is preferably entirely or substantially of human origin. Preferably the light chain
framework region of a humanized, human or chimeric antibody of the invention
comprises FR1 with SEQ ID NO: 266, FR2 with SEQ ID NO: 267, FR3 with SEQ ID
NO: 268 and FR4 with SEQ ID NO: 269. Preferably the heavy chain framework region
of a humanized, human or chimeric antibody of the invention comprises FR1 with SEQ
ID NO: 262, FR2 with SEQ ID NO: 263, FR3 with SEQ ID NO: 264, and FR4 with SEQ
ID NO: 265. For example, a preferred embodiment of LCVR of antibody 126 of the
invention, as described in Tables 1,2 and 3 herein comprises (polypeptides in order from
N-terminus) FR1 with SEQ ID NO: 266, CDR1 with SEQ ID NO: 131, FR2 with SEQ ID
NO: 267, CDR2 with SEQ ID NO: 167, FR3 with SEQ ID NO: 268, CDR3 with SEQ ID
NO: 168 and FR4 with SEQ ID NO: 269. The entire LCVR sequence, operably linked to
a human kappa constant region is as shown in SEQ ID NO: 274. Further, a preferred
embodiment of HCVR of antibody 126 of the invention comprises (in order from N-
terminus) FR1 with SEQ ID NO: 262, CDRl with SEQ ID NO: 26, FR2 with SEQ ID
NO: 262, CDR2 with SEQ ID NO: 30, FR3 with SEQ ID NO: 264, CDR3 with SEQ ID
NO: 52 and FR4 with SEQ ID NO: 265. The entire HCVR sequence, operably linked to a
human IgG4 Fc region is as shown in SEQ ID NO: 273.
In one embodiment, an anti-IL-17 antibody of the invention, wherein all or a
portion of the variable region is limited by a particular sequence as shown by a SEQ ID
NO herein (see, e.g., Tables 1-3) is further characterized by being a chimeric, humanized,
or fully human antibody or antigen-binding portion thereof that antagonizes or neutralizes
at least one human IL-17 activity in vivo or in vitro. An IL-17 antibody of the invention,
wherein all or a portion of the variable region is limited by a particular sequence as
shown by a SEQ ID NO herein is further characterized by specifically binding human IL-
17 but not binding human IL-17B, IL-17C, IL-17D, IL-17E or IL-17F. Additionally, the
antibody is further characterized by specifically binding human IL-17 and cynomolgus

monkey IL-17 but not binding rat IL-17 or mouse IL-17 at levels greater than
background.
More preferably, such antibody is further characterized by binding a human IL-17
nonlinear epitope comprising amino acids DGNVDYH (SEQ ID NO: 276) wherein the
antibody makes contact with the polypeptide with SEQ ID NO: 276. Even more
preferably, such antibody is further characterized by having a KD for human IL-17 of less
than about 7 pM, 6.5 pM or 6 pM, preferably less than about 5.5 pM, 5 pM or 4.5 pM and
most preferably less than about 4 pM and/or is characterized by an ICso, preferably in an
in vitro IL-8 reporter assay, that is less than 700 pM, 650 pM, 600 pM, 560 pM, 550 pM
or 500 pM, or an IC50 in an in vitro GROa reporter assay of less than about 560 pM,
and/or has an off rate (koff) for human IL-17 of less than 5 x 10-5, 4 x 10-5,3 x 10-5 or 2 x
10-5,s-1
Antibody Expression
The present invention is also directed to cell lines that express an anti-IL-17
monoclonal antibody of the invention or portion thereof. Creation and isolation of cell
lines producing a monoclonal antibody of the invention can be accomplished using
standard techniques known in the art Preferred cell lines include COS, CHO, SP2/0,
NSO and yeast (available from public repositories such as ATCC, American Type Culture
Collection, Manassas, VA).
A wide variety of host expression systems can be used to express an antibody of
the present invention including prokaryotic and eukaryotic expression systems (such as
yeast, baculovirus, plant, mammalian and other animal cells, transgenic animals, and
hybridoma cells), as well as phage display expression systems. An example of a suitable
bacterial expression vector is pUC119 and a suitable eukaryotic expression vector is a
modified pcDNA3.1 vector with a weakened dhfr selection system. Other antibody
expression systems are also known in the art and are contemplated herein.
An antibody of the invention can be prepared by recombinant expression of
immunoglobulin light and heavy chain genes in a host cell. To express an antibody
recombinantly, a host cell is transformed, transduced, infected or the like with one or
more recombinant expression vectors carrying DNA fragments encoding the
immunoglobulin light and/or heavy chains of the antibody such that the light and/or

heavy chains are expressed in the host cell. The heavy chain and the light chain may be
expressed independently from different promoters to which they are operably linked in
one vector or, alternatively, the heavy chain and the light chain may be expressed
independently from different promoters to which they are operably linked in two vectors
- one expressing the heavy chain and one expressing the light chain. Optionally the
heavy chain and light chain may be expressed in different host cells. Preferably, the
recombinant antibodies are secreted into the medium in which the host cells are cultured,
from which the antibodies can be recovered or purified. Standard recombinant DNA
methodologies are used to obtain antibody heavy and light chain genes, incorporate these
genes into recombinant expression vectors, and introduce the vectors into host cells.
Such standard recombinant DNA technologies are described, for example, in Sambrook,
Fritsch, and Maniatis (Eds.), Molecular Cloning; A Laboratory Manual, Second Edition,
Cold Spring Harbor, N.Y., 1989; Ausubel, et al (Eds.) Current Protocols in Molecular
Biology, Greene Publishing Associates, 1989.
An isolated DNA encoding a HCVR region can be converted to a full-length
heavy chain gene by operably linking the HCVR-encoding DNA to another DNA
molecule encoding heavy chain constant regions (CH1, CH2, and CH3). The sequences
of human heavy chain constant region genes are known in the art. See, e.g., Kabat, et al.,
Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of
Health and Human Services, NIH Publication No. 91-3242 (1991). DNA fragments
encompassing these regions can be obtained e.g., by standard PCR amplification. The
heavy chain constant region can be of any type, (e.g., IgG, IgA, IgE, IgM or IgD), class
(e.g., IgG1, IgG2, IgG3 and IgG-0 or subclass constant region and any allotypic variant
thereof as described in Kabat (supra). Alternatively, the antigen binding portion can be a
Fab fragment, Fab' fragment, F(ab')2 fragment, Fd, or a single chain Fv fragment (scFv).
For a Fab fragment heavy chain gene, the HCVR-encoding DNA may be operably linked
to another DNA molecule encoding only a heavy chain CH1 constant region.
An isolated DNA encoding a LCVR region may be converted to a full-length light
chain gene (as well as to a Fab light chain gene) by operably linking the LCVR-encoding
DNA to another DNA molecule encoding a light chain constant region, CL. The
sequences of human light chain constant region genes are known in the art See, e.g.,
Kabat, supra. DNA fragments encompassing these regions can be obtained by standard

PCR amplification. The light chain constant region can be a kappa or lambda constant
region.
To create an scFv gene, the HCVR- and LCVR-encoding DNA fragments are
operably linked to another fragment encoding a flexible linker, e.g., encoding the amino
acid sequence (Gly4-Ser)3, such that the HCVR and LCVR sequences can be expressed as
a contiguous single-chain protein, with the LCVR and HCVR regions joined by the
flexible linker. See, e.g.. Bird, et ah, Science 242:423-6,1988; Huston, et al, Proc Nail.
Acad. Sci. USA 85:5879-83,1988; McCafferty, etal., Nature 348:552-4,1990.
In one embodiment, the invention provides a vector, preferably (but not limited
to) a plasmid, a recombinant expression vector, a yeast expression vector, or a retroviral
expression vector comprising a polynucleotide encoding an anti-IL-17 monoclonal
antibody of the invention. Alternatively, a vector of the invention comprises a
polynucleotide encoding an LCVR and/or a polynucleotide encoding an HCVR of the
invention. When both an LCVR and an HCVR encoding sequence are present in the
same vector, they may be transcribed independently, each from a separate promoter to
which it is operably linked. If the sequences encoding the LCVR and HCVR are present
in the same vector and transcribed from one promoter to which they are both operably
linked, the LCVR may be 5' to the HCVR or the LCVR may be 3' to the HCVR,
furthermore, the LCVR and HCVR coding region in the vector may be separated by a
linker sequence of any size or content, preferably such linker, when present, is a
polynucleotide encoding an internal ribosome entry site.
To express an antibody of the invention, a DNA encoding a partial or full-length
light and/or heavy chain, obtained as described above, are inserted into an expression
vector such that the gene is operably linked to transcriptional and translational control
sequences. The expression vector and expression control sequences are chosen to be
compatible with the expression host cell used. The antibody light chain gene and the
antibody heavy chain gene can be inserted into separate vectors or, more typically, both
genes are inserted into the same expression vector. The antibody genes are inserted into
the expression vector by standard methods. Additionally, the recombinant expression
vector can encode a signal peptide that facilitates secretion of the anti-IL-17 monoclonal
antibody light and/or heavy chain from a host cell. The anti-IL-17 monoclonal antibody
light and/or heavy chain gene can be cloned into the vector such that the signal peptide is

operably linked in-frame to the amino terminus of the antibody chain gene. The signal
peptide can be an immunoglobulin signal peptide or aheterologous signal peptide.
In addition to the antibody heavy and/or light chain gene(s), a recombinant
expression vector of the invention carries regulatory sequences that control the expression
of the antibody chain gene(s) in a host cell. The term "regulatory sequence" is intended
to include promoters, enhancers and other expression control elements (eg.,
polyadenylation signals), as needed, that control the transcription or translation of the
antibody chain gene(s). The design of the expression vector, including the selection of
regulatory sequences may depend on such factors as the choice of the host cell to be
transformed, the level of expression of protein desired. Preferred regulatory sequences
for mammalian host cell expression include viral elements that direct high levels of
protein expression in mammalian cells, such as promoters and/or enhancers derived from
cytomegalovirus (CMV), Simian Virus 40 (SV40), adenovirus, (e.g., the adenovirus
major late promoter (AdMLP)) and polyoma virus.
In addition to the antibody heavy and/or light chain genes and regulatory
sequences, the recombinant expression vectors of the invention may carry additional
sequences, such as sequences that regulate replication of the vector in host cells (e.g.,
origins of replication) and one or more selectable marker genes. The selectable marker
gene facilitates selection of host cells into which the vector has been introduced. For
example, typically the selectable marker gene confers resistance to drugs, such as G418,
hygromycin, or methotrexate, on a host cell into which the vector has been introduced.
Preferred selectable marker genes include the dihydrofolate reductase (dhfr) gene (for use
in dhfr-minus host cells with methotrexate selection/amplification), the neo gene (for
G418 selection), and glutamine synthetase (GS) in a GS-negative cell line (such as NSO)
for selection/amplification.
For expression of the light and/or heavy chains, the expression vectors) encoding
the heavy and/or light chains is introduced into a host cell by standard techniques e.g.,
electroporation, calcium phosphate precipitation, DEAE-dextran transfection,
transduction, infection and the like. Although it is theoretically possible to express the
antibodies of the invention in either prokaryotic or eukaryotic host cells, eukaryotic cells
are preferred, and most preferably mammalian host cells, because such cells are more
likely to assemble and secrete a properly folded and immunologically active antibody.

Preferred mammalian host cells for expressing the recombinant antibodies of the
invention include Chinese Hamster Ovary (CHO cells) [including dhfr minus CHO cells,
described in Urlaub and Chasin, Proc Natl Acad. Sci. USA 77:4216-20,1980, used with
a DHFR selectable marker, e.g., as described in Kaufman and Sharp, J. Mol. Biol.
159:601-21,1982] NSO myeloma cells, COS cells, and SP2/O cells. When recombinant
expression vectors encoding antibody genes are introduced into mammalian host cells,
the antibodies are produced by culturing the host cells for a period of time sufficient to
allow for expression of the antibody in the host cells or, more preferably, secretion of the
antibody into the culture medium in which the host cells are grown under appropriate
conditions known in the art. Antibodies can be recovered from the host cell and/or the
culture medium using standard purification methods.
Host cells can also be used to produce portions, or fragments, of intact antibodies,
eg., Fab fragments or scFv molecules by techniques that are conventional. It will be
understood by a skilled artisan that variations on the above procedure are within the scope
of the present invention. For example, it may be desirable to transfect a host cell with
DNA encoding either the light chain or the heavy chain of an antibody of this invention.
Recombinant DNA technology may also be used to remove some or all the DNA
encoding either or both of the light and heavy chains that is not necessary for binding to
IL-17. The molecules expressed from such truncated DNA molecules are also
encompassed by the antibodies of the invention.
The invention provides a host cell comprising a nucleic acid molecule of the
present invention. Preferably a host cell of the invention comprises one or more vectors
or constructs comprising a nucleic acid molecule of the present invention. The host cell
of the invention is a cell into which a vector of the invention has been introduced, said
vector comprising a polynucleotide encoding a LCVR of an antibody of the invention
and/or a polynucleotide encoding a HCVR of the invention. The invention also provides
a host cell into which two vectors of the invention have been introduced; one comprising
a polynucleotide encoding a LCVR of an antibody of the invention and one comprising a
polynucleotide encoding a HCVR present in an antibody of the invention and each
operably linked to a promoter sequence. The host cell types include mammalian,
bacterial, plant and yeast cells. Preferably the host cell is a CHO cell, a COS cell, a
SP2/0 cell, a NSO cell, a yeast cell or a derivative or progeny of any preferred cell type.

In a preferred system for recombinant expression of an antibody of the invention,
a recombinant expression vector encoding both the antibody heavy chain and the
antibody light chain is introduced into dhfr-minus CHO cells by e.g., calcium phosphate-
mediated transfection. Within the recombinant expression vector, the antibody heavy and
light chain genes are each operably linked to enhancer/promoter regulatory elements
(e.g., derived from SV40, CMV, adenovirus and the like, such as a CMV
enhancer/AdMLP promoter regulatory element or an SV40 enhancer/AdMLP promoter
regulatory element) to drive high levels of transcription of the genes. The recombinant
expression vector also carries a dhfr gene, which allows for selection of CHO cells that
have been transfected with the vector using methotrexate selection/amplification. The
selected transformant host cells are cultured to allow for expression of the antibody heavy
and light chains and intact antibody is recovered from the culture medium. Standard
molecular biology techniques are used to prepare the recombinant expression vector,
transfect the host cells, select for transformants, culture the host cells and recover the
antibody from the culture medium. Antibodies, or antigen-binding portions thereof, of
the invention can be expressed in an animal (e.g., a mouse) that is transgenic for human
immunoglobulin genes (see, e.g., Taylor, et al., Nucleic Acids Res. 20:6287-95, 1992).
Once expressed, the intact antibodies, their dimers, individual light and heavy
chains, or other immunoglobulin forms of the present invention can be purified according
to standard procedures of the art, including ammonium sulfate precipitation, ion
exchange, affinity, reverse phase, hydrophobic interaction column chromatography, gel
electrophoresis and the like. Substantially pure immunoglobulins of at least about 90%,
92%, 94% or 96% homogeneity are preferred, and 98 to 99% or more homogeneity most
preferred, for pharmaceutical uses. Once purified, partially or to homogeneity as desired,
the peptides may then be used therapeutically or prophylactically, as directed herein.
Chimeric antibody
As used herein, the term "chimeric antibody" includes monovalent, divalent or
polyvalent immunoglobulins. A monovalent chimeric antibody is a dimer formed by a
chimeric heavy chain associated through disulfide bridges with a chimeric light chain. A
divalent chimeric antibody is a tetramer formed by two heavy chain-light chain dimers
associated through at least one disulfide bridge.

A chimeric heavy chain of an antibody comprises an antigen-binding region
derived from the heavy chain of a non-human antibody specific for IL-17, which is
operably linked to at least a portion of a human, or substantially human (or species
different from that from which the antigen-binding region was derived), heavy chain
constant region such as CH1 or CH2, or preferably to a full-length heavy chain constant
region. A chimeric light chain of an antibody for use in humans comprises an antigen-
binding region derived entirely or substantially from the light chain of a non-human
antibody specific for IL-17, operably linked to at least a portion of a human, or
substantially human (or species different from that from which the antigen-binding region
was derived), light chain constant region (CL), or preferably to a full-length light chain
constant region. Antibodies, fragments or derivatives having chimeric heavy chains and
light chains of the same or different variable region binding specificity, can also be
prepared by appropriate association of the individual polypeptide chains, according to
known method steps.
With this approach, hosts expressing chimeric heavy chains are separately
cultured from hosts expressing chimeric light chains, and the immunoglobulin chains are
separately recovered and then associated. Alternatively, the hosts can be co-cultured and
the chains allowed to associate spontaneously in the culture medium, followed by
recovery of the assembled immunoglobulin or fragment. Methods for producing chimeric
antibodies are known in the art (see, e.g., U.S. Patent Nos.: 6,284,471; 5,807,715;
4,816,567; and 4,816,397).
Humanized antibodies
Preferably an antibody of the invention to be used for therapeutic purposes, would
have the sequence of the framework and constant region (to the extent it exists in the
antibody) derived from the mammal in which it would be used as a therapeutic so as to
decrease the possibility mat the mammal would illicit an immune response against the
therapeutic antibody. Humanized antibodies are of particular interest since they are
considered to be valuable for therapeutic application and avoid the human anti-mouse
antibody response frequently observed with rodent antibodies. Additionally, in
humanized antibodies the effector portion of the antibody is of human origin so it may
interact better with the other parts of the human immune system (e.g., destroy the target

cells more efficiently by complement-dependent cytotoxicity or antibody-dependent
cellular cytotoxicity). Also, injected humanized antibodies may have a half-life more
like that of naturally occurring human antibodies than do e.g., murine antibodies, thereby
allowing smaller and less frequent doses to be given. The term "humanized antibody" as
used herein refers to an antibody comprising portions of antibodies of different origin,
wherein at least one portion is of human origin. For example, the humanized antibody
can comprise portions derived from an antibody of nonhuman origin with the requisite
specificity, such as a mouse, and from an antibody of human origin, joined together
chemically by conventional techniques (e.g., synthetic) or prepared as a contiguous
polypeptide using genetic engineering techniques.
Preferably, a "humanized antibody" has CDRs that originate (or substantially
originate) from a non-human antibody (preferably a mouse monoclonal antibody) while
framework and constant region, to the extent it is present, (or a significant or substantial
portion thereof, i.e., at least about 90%, 92%, 94%, 95%, 96%, 97%, 98% or 99%) are
encoded by nucleic acid sequence information that occurs in the human germline
immunoglobulin region (see, e.g., the International ImMunoGeneTics Database) or in
recombined or mutated forms thereof whether or not said antibodies are produced in
human cell.
The CDRs of a humanized antibody may be altered or optimized from the CDRs
of a non-human parent antibody from which they originated to generate desired
properties, e.g., specificity, affinity and/or preferential binding. Altered or optimized
CDRs may have amino acid substitutions, additions and/or deletions when compared to a
parent CDRs, preferably about 1,2,3,4, 5, 6,7, 8,9 or 10 total within the six CDR
domains. For example, the amino acid positions of CDRs that are underlined and in bold
print in Tables 2 and 3 are positions which have been altered from the CDRs as shown in
Fab 1 of Tables 2 and 3. Alternatively murine antibody 2321 may be a parent antibody
for comparison of CDRs of an antibody of the invention.
Humanized forms of non-human (e.g., murine) antibodies include an intact
antibody, a substantially intact antibody, a portion of an antibody comprising an antigen-
binding site, or a portion of an antibody comprising a Fab fragment, Fab' fragment,
F(ab')2, or a single chain Fv fragment Humanized antibodies preferably contain minimal
sequence derived from non-human immunoglobulin. Humanized antibodies may also

comprise residues which are found neither in the recipient antibody nor in the imported
CDR or framework sequences. In general, the humanized antibody will comprise
substantially all of at least one, and typically two, variable domains, in which all or
substantially all of the amino acids in the CDR regions correspond to those of a non-
human immunoglobulin and all or substantially all of the amino acids in the FR regions
are those of a human immunoglobulin consensus sequence. The humanized antibody
optimally also comprises at least a portion of an immunoglobulin constant region (Fc),
typically that of a human immunoglobulin. [Jones et al., Nature, 321:522-525,1986;
Riechmann et al., Nature, 332:323-329,1988; and Presta, Curr. Op. Struct. Biol, 2:593-
596, 1992.]
Humanized antibodies may be subjected to in vitro mutagenesis using methods of
routine use in the art (or, when an animal transgenic for human Ig sequences is used, in
vivo somatic mutagenesis) and, thus, the framework region amino acid sequences of the
HCVR and LCVR regions of the humanized recombinant antibodies are sequences that,
while derived from those related to human germline HCVR and LCVR sequences, may
not naturally exist within the human antibody germline repertoire in vivo. It is
contemplated that such amino acid sequences of the HCVR and LCVR framework
regions of the humanized recombinant antibodies are at least 90%, 92%, 94%, 95%, 96%,
98% or most preferably at least 99% identical to a human germline sequence. Preferably,
those framework residues of the parent antibody (e.g., murine antibody or generally the
antibody from which the humanized antibody is derived) which maintain or affect
combining-site structures will be retained. These residues may be identified e.g., by X-
ray crystallography of the parent antibody or Fab fragment, thereby identifying the three-
dimensional structure of the antigen-binding site. One strategy to humanize antibodies is
to choose a human germline sequence with the greatest homology to the framework of the
parent antibody as the framework to receive the donor CDRs. This germline approach is
based on the same rationale as the best-fit strategy, but only germline sequences are
searched in the databases.
The humanized antibody of the present invention may comprise or be derived
from a human germline light chain framework. In particular embodiments, the light chain
germline sequence is selected from human VK sequences including, but not limited to,
Al, A10, All, A14, A17, A18, A19, A2, A20, A23, A26, A27, A3, A30, A5, A7, B2, B3,

L1, L10, L11, L12, L14, L15, L16, L18, L19, L2, L20, L22, L23, L24, L25, L4/18a, L5,
L6, L8, L9, 01, O11, 012,014,018,02,04, and 08. In certain embodiments, this light
chain human germline framework is selected from V1-11, V1-13, V1-16, V1-17, V1-18,
V1-19, V1-2, V1-20, V1-22, VI-3, V1-4, V1-5, V1-7, V1-9, V2-1, V2-11, V2-13, V2-14,
V2-15, V2-17, V2-19, V2-6, V2-7, V2-8, V3-2, V3-3, V3-4, V4-1, V4-2, V4-3, V4-4,
V4-6, V5-1, V5-2, V5-4, and V5-6.
In other embodiments, the humanized antibody of the present invention may
comprise or be derived from a human germline heavy chain framework. In particular
embodiments, this heavy chain human germline framework is selected from VH1-18,
VH1-2, VH1-24, VH1-3, VH1-45, VH1-46, VH1-58, VH1-69, VH1-8, VH2-26, VH2-5,
VH2-70, VH3-11, VH3-13, VH3-15, VH3-16, VH3-20, VH3-21, VH3-23, VH3-30,
VH3-33, VH3-35, VH3-38, VH3-43, VH3-48, VH3-49, VH3-53, VH3-64, VH3-66,
VH3-7, VH3-72, VH3-73, VH3-74, VH3-9, VH4-28, VH4-31, VH4-34, VH4-39, VH4-4,
VH4-59, VH4-61, VH5-51, VH6-1, and VH7-81. See PCT WO 2005/005604 for a
description of the different germline sequences.
In particular embodiments, the light chain variable region and/or heavy chain
variable region comprises a framework region or at least a portion of a framework region
(e.g., containing 2 or 3 subregions, such as FR2 and FR3). In certain embodiments, at
least FRL1, FRL2, FRL3, or FRL4 is fully human. In other embodiments, at least FRH1,
FRH2, FRH3, or FRH4 is fully human. In some embodiments, at least FRL1, FRL2,
FRL3, or FRL4 is a germline sequence (e.g., human germline) or comprises human
consensus sequences for the particular framework. In other embodiments, at least FRH1,
FRH2, FRH3, or FRH4 is a germline sequence (e.g., human germline) or comprises
human consensus sequences for the particular framework. In preferred embodiments, all
of the framework region is human framework region.
In general, humanized antibodies may be produced by obtaining nucleic acid
sequences encoding the HCVR and LCVR of an antibody, e.g., a murine antibody or
antibody made by a hybridoma, which binds a IL-17 epitope of the invention, identifying
the CDRs in said HCVR and LCVR (nonhuman), and grafting such CDR-encoding
nucleic acid sequences onto selected human framework-encoding nucleic acid sequences.
Optionally, a CDR region may be optimized by mutagenizing randomnly or at particular
locations in order to substitute one or more amino acids in the CDR with a different

amino acid prior to grafting the CDR region into the framework region. Alternatively, a
CDR region may be optimized subsequent to insertion into the human framework region
using methods available to one of skill in the art Preferably, the human framework amino
acid sequences are selected such that the resulting antibody is likely to be suitable for in
vivo administration in humans. This can be determined, e.g.. based on previous usage of
antibodies containing such human framework sequence. Preferably, the human
framework sequence will not itself be significantly immunogenic.
Alternatively, the amino acid sequences of the frameworks for the antibody to be
humanized may be compared to those of known human framework sequences the human
framework sequences to be used for CDR-grafting and selected based on their comprising
sequences highly similar to those of the parent antibody, e.g., a tnurine antibody which
binds IL-17 (e.g., an antibody comprising a HCVR with SEQ ID NO: 270 and further
comprising a LCVR with SEQ ID NO: 271). Numerous human framework sequences
have been isolated and their sequences reported in the art. This enhances the likelihood
that the resultant CDR-grafted humanized antibody, which contains CDRs of the parent
(e.g., murine) or optimized CDRs of the parent antibody grafted onto selected human
frameworks (and possibly also the human constant region) will substantially retain the
antigen binding structure and thus retain the binding affinity of the parent antibody. To
retain a significant degree of antigen binding affinity, the selected human framework
regions will preferably be those that are expected to be suitable for in vivo administration,
i.e., not immunogenic.
In either method, the DNA sequence encoding the HCVR and LCVR regions of
the preferably murine anti-IL-17 antibody are obtained. Methods for cloning nucleic acid
sequences encoding immunoglobulins are known in the art. Such methods may, for
example, involve the amplification of the immunoglobulin-encoding sequences to be
cloned using appropriate primers by polymerase chain reaction (PCR). Primers suitable
for amplifying immunoglobulin nucleic acid sequences, and specifically murine HCVR
and LCVR sequences have been reported in the literature. After such immunoglobulin-
encoding sequences have been cloned, they will be sequences by methods well known in -
the art
After the CDR-encoding sequences are grafted onto the selected human
framework encoding sequences, the resultant DNA sequences encoding the "humanized"

variable heavy and variable light sequences are then expressed to produce a humanized
Fv or humanized antibody that binds IL-17. The humanized HCVR and LCVR may be
expressed as part of a whole anti-IL-17 antibody molecule, ie., as a fusion protein with
human constant domain sequences whose encoding DNA sequences have been obtained
from a commercially available library or which have been obtained using, e.g., one of the
above described methods for obtaining DNA sequences, or are in the art However, the
HCVR and LCVR sequences can also be expressed in the absence of constant sequences
to produce a humanized anti-IL-17 Fv. Nevertheless, fusion of human constant
sequences onto the variable region is potentially desirable because the resultant
humanized anti-IL- 17antibody may possess human effector functions.
Methods for synthesizing DNA encoding a protein of known sequence are well
known in the art. Using such methods, DNA sequences which encode the subject
humanized HCVR and LCVR sequences (with or without constant regions) are
synthesized, and then expressed in a vector system suitable for expression of recombinant
antibodies. This may be effected in any vector system which provides for the subject
humanized HCVR and LCVR sequences to be expressed as a fusion protein with human
constant domain sequences and to associate to produce functional (antigen binding)
antibodies or antibody fragments.
Human constant domain sequences are known in the art, and have been reported
in the literature. Preferred human constant light chain sequences include the kappa and
lambda constant light chain sequences. Preferred human constant heavy chain sequences
include human IgG1, human IgG2, human IgG3, human IgC4 (see, e.g., Seq ID NOs: 257-
260 respectively) and mutated versions thereof which provide for altered effector
function, e.g., enhanced in vivo half-life, reduced Fc receptor binding, altered
deamidation profile and the like.
If present, human framework regions are preferably derived from a human
antibody variable region having sequence similarity to the analogous or equivalent region
of the antigen binding region donor (i.e., the parent antibody). Other sources of
framework regions for portions of human origin of a humanized antibody include human
variable consensus sequences (see e.g., Kettleborough, C.A. et el. Protein Engineering
4:773-783 (1991); Carter et al.. WO 94/04679. For example, the sequence of the
antibody or variable region used to obtain the nonhuman portion can be compared to

human sequences as described in Kabat et al. Sequences of Proteins of Immunological
Interest, Fifth Edition, NIH, U.S. Government Printing Office (1991). In a particularly
preferred embodiment, the framework regions of a humanized antibody chain are derived
from a human variable region having at least about 60% overall sequence identity,
preferably at least about 70%, 80%, or 90% overall sequence identity and more
preferably at least about 85% overall sequence identity, with the variable region of the
nonhuman donor. A human portion can also be derived from a human antibody having at
least about 65% sequence identity, and preferably at least about 70% sequence identity,
within the particular portion (e.g., FR) being used, when compared to the equivalent
portion (e.g., FR) of the nonhuman donor.
References further describing methods involved in humanizing a mouse antibody
that may be used are e.g., Queen et al., Proc. Natl Acad. Sci. USA 88:2869, 1991; U.S.
Pat. No. 5,693,761; U.S. Pat No. 4,816,397; U.S. Pat No. 5,225,539; computer programs
ABMOD and ENCAD as described in Levitt, M., J. Mol. Biol. 168:595-620,1983;
humanization can be essentially performed following the method of Winter and co-
workers (Jones et al., Nature, 321:522-525, 1986; Riechmann et al., Nature, 332:323-327,
1988; Verhoeyen et al., Science, 239:1534-1536,1988).
Human Antibodies
As an alternative to humanization, human antibodies can be generated. Human
antibodies can be produced using various techniques known in the art, including phage
display libraries (Hoogenboom and Winter, J. Mol. Biol, 227:381, 1991; Marks et al., J.
Mol Biol., 222:581,1991). The techniques of Cole et al. and Boemer et al. are also
available for the preparation of human monoclonal antibodies (Cole et al., Monoclonal
Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985) and Boemer et al., /.
Immunol, 147:86-95,1991). Similarly, human antibodies can be made by introducing
human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous
immunoglobulin genes have been partially or complete inactivated. Upon immunization,
e.g., with an antigen comprising an irnmunogenic ephope of the invention, a full
repertoire of human antibody production is obtained, which closely resembles that seen in
humans in all respects, including gene rearrangement, assembly and antibody repertoire.
This approach is described, for example, in U.S. Pat. Nos. 5,545,807; 5,545,806;

5,569,825; 5,589,369; 5,591,669; 5,625,126; 5,633,425; 5,661,016, and in the following
scientific publications: Marks et al., BioTechnology 10:779-783,1992; Lonberg et al,
Nature 368: 856-859,1994; Morrison, Nature 368: 812-13,1994; Fishwild et al., Nature
Biotechnology 14:845-51,1996; Neater get, Nature Biotechnology 14: 826(1996);
Lonberg and Huszar, Intern. Rev. Immunol. 13: 65-93 (1995) and Jobkobovits et al.,
Proc. Natl. Acad. Sci., USA, 90:2551,1993.
Human immunoglobulin genes introduced into the mouse, thus creating transgenic
mice capable of responding to antigens with antibodies having human sequences are also
described in Bruggemann et al. (Proc. Natl Acad. Sci. USA 86:6709-6713, 1989). There
are several strategies that exist for the generation of mammals that produce human
antibodies. In particular, there is the "minilocus" approach in which an exogenous Ig
locus is mimicked through the inclusion of pieces (e.g., individual genes) from the Ig
locus (see, e.g., U.S. Pat. NOs. 5,545,807,5,545,806,5,625,825,5,625,126,5,633,425,
5,661,016,5,770,429,5,789,650, and 5,814,318,5,612,205, 5,721,367, 5,789,215), YAC
introduction of large and substantially germline fragments of the Ig loci (See Mendez et
al. Nature Genetics 15:146-156,1997; Green and Jakobovits J. Exp. Med. 188:483-495,
1998), and introduction of entire or substantially entire loci through the use of microcell
fusion (see European Patent Application No. EP 0 843 961 A1).
Any transgenic mouse capable of responding to immunization with antibodies
having human sequences may be used to produce an anti-IL-17 antibody of the invention
when using methods available to one skilled in the art, e.g., when such mouse is
immunized with a polypeptide comprising an immunogenic epitope of the invention.
Uses
Antibodies of the present invention are useful in therapeutic, prophylactic,
diagnostic and research applications as described herein. An antibody of the invention
may be used to diagnose a disorder or disease associated with the expression of human
IL-17. In a similar manner, the antibody of the invention can be used in an assay to
monitor IL-17 levels in a subject being tested for an IL-17-associated condition.
Research applications include methods that utilize the antibody of the invention and a
label to detect IL-17 in a sample, e.g., in a human body fluid or in a cell or tissue extract.
Antibodies of the invention may by used with or without modification, and are labeled by

covalent or non-covalent attachment of a detectable moiety. The detectable moiety can
be any one that is capable of producing, either directly or indirectly, a detectable signal.
For example, the detectable moiety may be a radioisotope such as, e.g., 3H 14C, 32P, 35S,
or l23I, a fluorescent or chemiluminescent compound, such as fluorescein isothiocyanate,
rhodamine, or luciferin; or an enzyme, such as alkaline phosphatase, beta-galactosidase,
or horseradish peroxidase. Any method known in the art for separately conjugating the
antibody to the detectable moiety may be employed, including those methods described
by Hunter, et al., Nature 144:945,1962; David, et al., Biochemistry 13:1014,1974; Pain,
et al., J. Immunol Meth. 40:219,1981; and Nygren, J. Histochem. and Cytochem. 30:
407, 1982.
A variety of conventional protocols for measuring IL-17, including e.g., ELISAs,
RIAs, and FACS, are known in the art and provide a basis for diagnosing altered or
abnormal levels of IL-17 expression. Normal or standard expression values are
established using any art known technique, eg., by combining a sample comprising a IL-
17 polypeptide with, e.g., antibodies under conditions suitable to form a antigen:antibody
complex. The antibody is directly or indirectly labeled with a detectable substance to
facilitate detection of the bound or unbound antibody. Suitable detectable substances
include various enzymes, prosthetic groups, fluorescent materials, luminescent materials
and radioactive materials. Examples of suitable enzymes include horseradish peroxidase,
alkaline phosphatase, β-galactosidase, or acetylcholinesterase; examples of suitable
prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of
suitable fluorescent materials include umbelliferone, fluorescein, fluorescein
isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or
phycoerythrin; an example of a luminescent material includes luminol; and examples of a
radioactive material include l2SL l31I,35S, or 3H. (See, eg., Zola, Monoclonal Antibodies:
A Manual of Techniques, CRC Press, Inc. (1987)).The amount of a standard complex
formed is quantitated by various methods, such as, e.g., photometric means. Amounts of
IL-17 polypeptide expressed in samples are then compared with the standard values.
As a matter of convenience, the antibody of the present invention can be provided
in a kit, a packaged combination of reagents in predetermined amounts with instructions
for performing the diagnostic assay. Where the antibody is labeled with an enzyme, the
kit will include substrates and cofactors required by the enzyme (e.g., a substrate

precursor which provides the detectable chromophore or fluorophore). In addition, other
additives may be included such as stabilizers, buffers (e.g., a blocking buffer or lysis
buffer) and the like. The relative amounts of the various reagents may be varied widely
to provide for concentrations in solution of the reagents which substantially optimize the
sensitivity of the assay. Particularly, the reagents may be provided as dry powders,
usually lyophilized, including excipients which on dissolution will provide a reagent
solution having the appropriate concentration.
Therapeutic Uses for the Antibody
IL-17 is a pro-inflammatory cytokine secreted by activated T cells at sites of
inflammation, not in the systemic circulation; it is not readily detectable in the serum or
tissues of a healthy person. IL-17 upregulates adhesion molecules, induces production of
multiple inflammatory cytokines and chemokines from various cell types including
synoviocytes, chondroctes, fibroblasts, endothelial cells, epithelial cells, thereby inducing
recruitment of neutrophils to an inflammatory site, stimulates the production of
prostaglandins and metalloproteinases, and inhibits proteogrycan synthesis. Furthermore,
IL-17 plays an important role in the maturation of hematopoietic progenitor cells. IL-17
has signaling roles in different organs and tissues including lung, articular cartilage, bone,
brain, hematopoietic cells, kidney, skin and intestine. IL-17 shares 15-27% amino acid
homology with IL-17 B, C and E and 44-50% amino acid homology with IL-17 D and F.
IL-17 binds to the IL-17 receptor with low affinity (about 1 nM), while other IL-17
family members do not bind to the IL-17 receptor.
Increased levels of IL-17 (i.e., IL-17 A) have been associated with several
conditions including airway inflammation, RA, osteoarthritis, bone erosion,
intraperitoneal abscesses and adhesions, IBD, allograft rejection, psoriasis, certain types
of cancer, angiogenesis, atherosclerosis and MS. Both IL-17 and IL-17R are up-
regulated in the synovial tissue of RA patients. IL-17 exerts its role in pathogenesis of
RA through IL-l-p and TNF-a dependent and independent pathways. IL-17 stimulates
secretion of other cytokines and chemokines, e.g., TNF-α, IL-1β, IL-6, IL-8 and Gro-α.
IL-17 directly contributes to disease progression in RA. Injection of IL-17 into the
mouse knee promotes joint destruction independently of IL-1β activity (Ann. Rheum.
Dis. 59:529-32,2000). Anti-IL-1β antibody has no effect on IL-17 induced inflammation

and joint damage (J. Immunol. 167:1004-1013,2001). In an SCW-induced murine
arthritis model, IL-17 induced inflammatory cell infiltration and proteoglycan depletion
in wild-type and IL-1β knock out and TNF-α knock out mice. 11-17 knock out mice are
phenotypically normal in the absence of antigenic challenge, but have markedly reduced
arthritis following type II collagen immunization (J. Immunol. 171:6173-6177,2003).
Multiple sclerosis ("MS") is an autoimmune disease characterized by central
nervous system ("CNS") inflammation with damage to the myelin sheath surrounding
axons. A hallmark of MS is that T cells infiltrate into the CNS. MS affects more than
350,000 persons in the U.S. and 2.5 million worldwide. There are many forms and the
most common is relapsing/remitting disease ("RRMS) followed by a secondary
progressive stage. Current therapeutics consist of Interferon-P (AVONEX,
BETASERON and REBIF) that reduces the relapse/exacerbation rate by 31%-34%, but
may produce flu-like symptoms and/or synthesis of neutralizing antibodies (e.g., about
15% of patients receiving AVONEX produce neutralizing antibodies in 18 months.
TYSABRI, approved by the FDA for RRMS was subsequently removed from the market
due to CNS immunosuppression concerns. There is still an unmet medical need in the
treatment of MS. IL-17 mRNA is increased in MS lesions and in mononuclear cells
(MNC) in blood and cerebrospinal fluid from MS patients. Higher numbers of IL-17
mRNA-expressing blood MNC are detected during MS clinical exacerbation compared to
remission (Multiple Sclerosis, 5:101-104,1999). Furthermore, experimental autoimmune
encephalomyelitis ("EAE"), a preclinical animal model for MS is significantly suppressed
in IL-17 knockout mice.
Therefore, a pharmaceutical composition comprising an anti-IL-17 monoclonal
antibody of the invention may be useful for the treatment or prevention of conditions
wherein the presence of IL-17 causes or contributes to undesirable pathological effects or
decrease of IL-17 activity has a therapeutic benefit in mammals, preferably humans,
including, but not limited to, airway inflammation, asthma, RA, osteoarthritis, bone
erosion, intraperitoneal abscesses and adhesions, IBD, allograft rejection, psoriasis,
certain types of cancer, angiogenesis, atherosclerosis and MS, as well as other
inflammatory disorders, conditions, diseases or states including without limit:
erythematosus, response to allergen exposure, Helicobacter pylori associated gastritis,
bronchial asthma, and allograft rejection (e.g., renal), systemic lupus erythematosis and

lupus nephritis. The use of an anti-IL-17 monoclonal antibody of the present invention
for treating or preventing of at least one of the aforementioned disorders in which IL-17
activity is detrimental or which benefits for decreased levels of bioactive IL-17 is
contemplated herein. Additionally, the use of an anti-IL-17 monoclonal antibody of the
present invention for use in the manufacture of a medicament for the treatment of at least
one of the aforementioned disorders is contemplated.
As used herein, the terms "treatment", "treating", and the like, refer to obtaining a
desired pharmacologic and/or physiologic effect The effect may be prophylactic in terms
of completely or partially preventing a disease or symptom thereof and/or may be
therapeutic in terms of a partial or complete cure for a disease and/or adverse affect
attributable to the disease. Treatment", as used herein, includes administration of a
compound of the present invention for treatment of a disease or condition in a mammal,
particularly in a human, and includes: (a) preventing the disease from occurring in a
subject which may be predisposed to the disease but has not yet been diagnosed as having
it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease,
i.e., causing regression of the disease or disorder or alleviating symptoms or
complications thereof. Dosage regimens may be adjusted to provide the optimum desired
response (e.g., a therapeutic or prophylactic response). For example, a single bolus may
be administered, several divided doses may be administered over time or the dose may be
proportionally reduced or increased as indicated by the exigencies of the therapeutic
situation.
Pharmaceutical Composition
An antibody of the invention can be incorporated into pharmaceutical
compositions suitable for administration to a subject (see, e.g., Example 14). The
compounds of the invention may be administered alone or in combination with a
pharmaceutically acceptable carrier, diluent, and/or excipient, in single or multiple doses.
The pharmaceutical compositions for administration are designed to be appropriate for
the selected mode of administration, and pharmaceutically acceptable diluent, carrier,
and/or excipients such as dispersing agents, buffers, surfactants, preservatives,
solubilizing agents, isotonicity agents, stabilizing agents and the like are used as
appropriate (See, e.g., Example 14 herein). Said compositions are designed in

accordance with conventional techniques as in e.g.. Remington. The Science and Practice
of Pharmacy, 19th Edition, Gennaro, Ed., Mack Publishing Co., Easton, PA 1995 which
provides a compendium of formulation techniques as are generally known to
practitioners.
A pharmaceutical composition comprising an anti-IL-17 monoclonal antibody of
the present invention can be administered to a subject at risk for or exhibiting pathologies
as described herein using standard administration techniques including oral, intravenous,
intraperitoneal, subcutaneous, pulmonary, transdermal, intramuscular, intranasal, buccal,
sublingual, or suppository administration.
A pharmaceutical composition of the invention preferably is a "therapeutically
effective amount" or a "prophylactically effective amount" of an antibody of the
invention. A "therapeutically effective amount" refers to an amount effective, at dosages
and for periods of time necessary, to achieve the desired therapeutic result. A
therapeutically effective amount of the antibody may vary according to factors such as the
disease state, age, sex, and weight of the individual, and the ability of the antibody or
antibody portion to elicit a desired response in the individual. A therapeutically effective
amount is also one in which any toxic or detrimental effect of the antibody, are
outweighed by the therapeutically beneficial effects. A "prophylactically effective
amount" refers to an amount effective, at dosages and for periods of time necessary, to
achieve the desired prophylactic result. Typically, since a prophylactic dose is used in
subjects prior to or at an earlier stage of disease, the prophylactically effective amount
will be less than the therapeutically effective amount
A therapeutically-effective or prophylactically-effective amount is at least the
minimal dose, but less than atoxic dose, of an active agent which is necessary to impart
therapeutic benefit to a subject. Stated another way, a therapeutically-effective amount of
an antibody of the invention is an amount which in mammals, preferably humans,
decreases an IL-17 bioactivity, e.g., binding to IL17R, wherein the presence of IL-17
causes or contributes to undesirable pathological effects or decrease in IL-17 levels
results in a beneficial therapeutic effect in a mammal, preferably a human.
The route of administration of an antibody of the present invention may be oral,
parenteral, by inhalation, or topical. Preferably, the antibodies of the invention can be
incorporated into a pharmaceutical composition suitable for parenteral administration.

The term parenteral as used herein includes intravenous, intramuscular, subcutaneous,
rectal, vaginal, or intraperitoneal administration. Peripheral systemic delivery by
intravenous or intraperitoneal or subcutaneous injection is preferred. Suitable vehicles
for such injections are straightforward in the art.
The pharmaceutical composition typically must be sterile and stable under the
conditions of manufacture and storage in the container provided, including e.g., a sealed
vial or syringe. Therefore, pharmaceutical compositions may be sterile filtered after
making the formulation, or otherwise made microbiologically acceptable. A typical
composition for intravenous infusion could have a volume as much as 250-1000 ml of
fluid, such as sterile Ringer's solution, physiological saline, dextrose solution and Hank's
solution and a therapeutically effective dose, (e.g., 1 to 100 mg/ml, or more) of antibody
concentration. Dose may vary depending on the type and severity of the disease. As is
well known in the medical arts, dosages for any one subject depends upon many factors,
including the patient's size, body surface area, age, the particular compound to be
administered, sex, time and route of administration, general health, and other drugs being
administered concurrently. A typical dose can be, for example, in the range of 0.001 to
1000 μg; however, doses below or above this exemplary range are envisioned, especially
considering the aforementioned factors. The dairy parenteral dosage regimen can be
about 0.1 μg/kg to about 100 mg/kg of total body weight, preferably from about 0.3 μg/kg
to about 10 mg/kg and more preferably from about 1 μg/kg to 1 mg/kg, even more
preferably from about 0.5 to 10 mg/kg body weight per day. Progress may be monitored
by periodic assessment For repeated administrations over several days or longer,
depending on the condition, the treatment is repeated until a desired suppression of
disease symptoms occurs. However, other dosage regimens may be useful and are not
excluded herefrom. The desired dosage can be delivered by a single bolus administration,
by multiple bolus administrations, or by continuous infusion administration of antibody,
depending on the pattern of pharmacokinetic decay that the practitioner wishes to
achieve.
These suggested amounts of antibody are subject to a great deal of therapeutic
discretion. The key factor in selecting an appropriate dose and scheduling is the result
obtained. Factors for consideration in this context include the particular disorder being
treated, the particular mammal being treated, the clinical condition of the individual

patient, the cause of the disorder, the site of delivery of the antibody, the particular type
of antibody, the method of administration, the scheduling of administration, and other
factors known to medical practitioners.
Therapeutic agents of the invention may be frozen or lyophilized for storage and
reconstituted in a suitable sterile carrier prior to use. Lyophilization and reconstitutton
can lead to varying degrees of antibody activity loss. Dosages may have to be adjusted to
compensate. Generally, pH between 6 and 8 is preferred.
Article of Manufacture.
In another embodiment of the invention, an article of manufacture containing
materials useful for the treatment or prevention of the disorders or conditions described
above is provided. The article of manufacture comprises a container and a label. Suitable
containers include, for example, bottles, vials, syringes, and test tubes. The containers
may be formed from a variety of materials such as glass or plastic. The container holds a
composition of an antibody of the invention which is effective for preventing or treating
the disorder or condition and may have a sterile access port (for example the container
may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic
injection needle). The active agent in the composition is an anti-IL-17 antibody of the
invention. The label on, or associated with, the container indicates that the composition
is used for treating the condition of choice. The article of manufacture may further
comprise a second container comprising a pharmaceutically-acceptable buffer, such as
phosphate-buffered saline, Ringer's solution and dextrose solution. It may further
include other materials desirable from a commercial and user standpoint, including other
buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
The following examples are offered for illustrative purposes only, and are not
intended to limit the scope of the present invention in any way.


















Example 1 ELISA I: Antibody Binding to IL-17 of Various Species
An exemplary ELISA assay for measuring binding of antibodies to IL-17 uses
sealed Costar 3366 microtiter plates that are coated overnight at 4°C with 50 μl of 1.0
μg/ml human IL-17 per well (R&D Systems, #317-IL/CF) in carbonate coating buffer (50
mM sodium carbonate, pH 9.0). Alternatively, mouse, rat, rabbit or cynomolgus monkey
IL-17 are used. Human IL-22 (R&D Systems) is used as a control antigen. Rabbit and
cynomolgus monkey IL-17 are not commercially available and therefore require cloning
and expression, or artificial synthesis, according to methods known in the art making use
of the amino acid sequences for IL-17 of the various species provided in Figure 2 (Seq ED
NOs: 9 and 10). Exemplary nucleotide sequences encoding IL-17 of the various species
are shown in SEQ ID NOs: 250-254.
The plate is subsequently blocked by adding 100 ul blocking buffer (Pierce
#37515). The plate is incubated for 1 hour at 37°C then washed three times in wash
buffer (PBS pH 7.4 and 0.05% Tween). Then, 50 μl of either sample antibody or control
antibody (diluted to various concentrations in PBS pH 7.4, e.g., 2,0.4,0.08,0.016,
0.0032 and 0 μg/ml) is added to each well and the plate is further incubated for 1 hour at
37°C. The plate is then washed three times with wash buffer before adding 50 μl per well
of anti-human kappa-alkaline phosphatase conjugated diluted to 1:1000 in PBS pH 7.4.
The test samples are incubated for 1 hour at 37°C. Then p-nitrophenyl phosphate
disodium salt (PNPP, Pierce #37620) is freshly made by dissolving in diethanolamine
substrate buffer according to manufacturer's instruction and 50 μl is added to each well.
Color development is allowed to proceed for about 10 minutes at room temperature then
color signal is measured at an absorbance of 405 nm using any appropriate ELISA plate
reader. The degree of binding is proportional to color signal production.
Antibodies of the invention bind human IL-17 in an ELISA assay as described
herein, but do not bind rat or mouse IL-17. It is anticipated, given the Biacore data of
Example 4 demonstrating that antibodies of the invention bind human and monkey IL-17,
that the antibodies of the invention would also demonstrate binding to monkey IL-17 in
an ELISA assay as described herein.
Example 2 ELISA II: Antibody Binding to Proteins of IL-17 Family

An ELISA is used to measure whether antibodies of the invention selectively
and/or preferentially bind particular human IL-17 members (e.g., IL-17A, IL-17B, IL-
17C, IL-17D, IL-17E or DL-17F) or human IL-22 (negative control).
In an exemplary assay, ELISA plate wells (Nunc Immuno Maxisorp) are coated
with 100 μl (0-5 μg/ml in IX coating buffer (BioFx)) of IL-17 family member proteins
(R&D Systems) sealed and incubated overnight at 4°C. The solution in the well is
removed by flicking and blocking buffer (200 |xl 1.5% BSA in PBS) is added. The plates
are incubated on a rotating shaker for 30 minutes at room temperature. Then 100 μl of an
antibody to be tested is added per well at varying concentrations (e.g., 2,0.4,0.08,0.016,
0.0032 and 0 μg/ml). The plates are again incubated overnight (4°C) followed by
warming on a rotating shaker (60 min room temp). Each plate-well is then washed five
times with buffer (IX Ish buffer, BioFX). After washing, an appropriate commercially
available HRP-conjugated secondary antibody (1:2000 in PBS with 1.5% BSA) is added
(100 μl/well). Plates are re-incubated on a rotating shaker (60 min. room temp.) followed
by buffer washing (5X) as described above. The colorimetric signal is developed by
adding TMB (100 μl/well) until saturation (approx 3-5 min.) then further development is
ended by adding stop solution (100 μl/well, BioFX). The color signal is measured at 450
nm absorbance using any appropriate ELISA plate reader. The degree of binding is
proportional to color signal production. Antibodies of the invention (e.g., Fabs 103, 104,
118, 121, 126 and 131 as described in Table 1) specifically bind human IL-17 (i.e., IL-
17 A), but, under similar conditions, do not bind at greater than background levels to
human IL-17B, human DL-17C, human IL-17D, human IL-17E, human IL-17F, murine
IL-17 or human IL-22.
Example 3 Isolation and activation of cells for cloning IL-17
A. Rat splenocytes
Using sterile forceps and scissors, remove spleen of a rat sacrificed by COj inhalation
and put the spleen into a tube containing 5 ml RPMI1640 media + 10% fetal bovine
serum and penicillin/streptomycin (media solution). Pour contents of the tube into a 10
cm Petri dish and remove fat from spleen. Homogenize the spleen gentry using a pair of
fully frosted, pre-autoclaved microscopy slides. Wash cells off slides using media
solution, pipette a few times and filter cells through cell strainer (Fisher Scientific).

Wash cells once with media solution, count cells and resuspend them to a final
concentration of 2 x 107 cells/ml in 80 ml. Add cell solution to a T150 flask, add
Concanavalin A to a final concentration of 3 ng/ml and incubate at 37°C for about 15
hours. Harvest the cells, wash with PBS, freeze the cell pellet on dry ice and proceed
immediately to standard RNA isolation procedures.
B. Cynomolgus monkey and rabbit peripheral blood mononuclear cells (PBMC)
Load about 7 ml whole blood from cynomolgus monkey or 10 ml whole blood from
New Zealand white rabbit into a BD Vacutainer™ CPT™ System for separation of
mononuclear cells from whole blood. Centrifuge the CPT cell preparation tube for 20
min at 1500 x gravity in a horizontal swinging bucket rotor. Collect lymphocytes and
monocytes at the interface, wash twice with media solution, count and resuspend in media
solution at a final concentration of 106 cells/mL Add Concanavalin A to a final
concentration of 3 ug/ml and incubate at 37°C for about 15 hours. Harvest the cells,
wash with PBS, freeze the cell pellet on dry ice and proceed immediately to standard
RNA isolation procedures.
Example 4 Measuring Binding Kinetic Constants
A BIACORE® 2000 instrument is used to measure antigen-antibody binding
kinetics and affinity. The instrument utilizes the optical properties of surface plasmon
resonance to detect alteration in protein concentration of interacting molecules within a
dextran biosensor matrix. Except as noted, all reagents and materials are purchased from
BIACORE® AB. All measurements are performed at 25°C. Samples are resuspended in
HBS-EP buffer to a final concentration of 2 μg/ml (150 mM sodium chloride, 3 mM
EDTA, 0.005% (w/v) surfactant P-20, and 10 mM HEPES, pH 7.4). Protein A is
immobilized on flow cells 1 to 4 of a CM4 sensor chip at a level of 500 response units
using an amine coupling kit
Binding is evaluated using multiple analytical cycles. Each cycle is performed at
a flow rate of 50 μl/minute and consists of the following steps: injection of about 20 μl of
an antibody composition at 2 ug/ml aiming at a capture of 100-200 response units,
injection of 250 μl of human IL-17, Cynomalgus monkey IL-17, New Zealand white
rabbit IL-17, rat IL-17 or mouse IL-17 (starting at 10 nM and using two-fold serial
dilutions for each cycle) followed by 20 minutes for dissociation, and regeneration using

30 μl of 10 mM glycine hydrochloride, pH 1.5. Association and dissociation rates for
each cycle are evaluated using a "1:1 with mass transfer" binding model in the
BIAevaluation software.
Full-length mAbs 103,104, 118, 121, 126 and 131 (see Table 1) having an IgG4
Fc region exhibit high affinity binding to human IL-17 and to monkey IL-17 with a KD
less than 5 pM, a Koff slower than 2 x 10-5s-1 and a K™ of at least 5 x 106 M-1s-1. The KD
and koirare improved (i.e., lower KD, slower koff) in these variant mAbs over Fab 2321
mAb (parent Fab of e.g., Fab 103 and 104) comprising a murine variable region [Seq ID
Nos: 261 (VH of 2321), 262 (VL of 2321)], a human IgG4 heavy chain constant region
(SEQ ID NO: 260) and a kappa light chain constant regions (SEQ ID NO: 272).
Antibodies of the invention exhibit binding no greater than background levels to mouse
IL-17 or rat IL-17; no binding is detected up to 200 nM mouse IL-17 and no binding is
detected up to 1 μM rat IL-17. When the full-length mAbs 103, 104, 121 and 126 are
tested, under the same conditions described above, for binding to cynomolgus monkey
IL-17 and rabbit IL-17; binding to rabbit IL-17 is weak and biphasic while binding to
monkey IL-17 is similar to binding to humaa Specific values for certain mabs (values
are reported as mean ± standard error of mean) of the invention when tested in this assay
are listed in Table 4 below. It is contemplated that Fc regions other than that of IgG4
would not significantly affect KD and koff



Example 5 IL-17 Receptor/Anti-IL-17 Antibody Binding Competition Studies
This example demonstrates that the antibodies of the invention compete for
binding to IL-17 with the IL-17 receptor.
BIACORE binding studies are performed using the IL-17 receptor Fc-fusion
protein (R&D #177-IR). To demonstrate that the IL-17 receptor Fc-fusion protein binds
human IL-17, a BIACORE assay is performed in BIACORE binding buffer (HBS-EP) +
1 mg/ml BSA at 25°C on a BIACORE 2000 instrument. A CM4 chip is used with
approximately 600 response units of Protein A immobilized on flow cells 1,2 and 3 of
the chip. Approximately 100 response units of IL-17 receptor Fc-fusion protein is
captured on flow cell 2 of the chip. Human IL-17 is then exposed to flow cells 1 and 2 in
concentrations ranging from 600 nM to 9.4 nM. After each 250 ul injection of human IL-
17, the complex is allowed to dissociate for about 12 minutes by running buffer across the
chip. At the end of the dissociation, a 20 μl injection of 100 mM glycine pH 1.5 is used
to regenerate the chip before the next cycle of binding begins. Flow cell 1 is used as a
reference flow cell. The data is fit using the "Bivalent analyte" model in the
BIAevaluation Version 3.2 software. The results indicate that this interaction has an on-
rate of 1.06 x 105 M-1s-1 a fast off-rate of 20.3 s-1 and a slow off-rate of 1.63 x 10-4 s-1.
Therefore, mis interaction has a KD or binding affinity of 1.5 nM and 0.19 mM which is
much weaker than the binding affinities of the antibodies of the invention to human IL-
17.
Binding for the competition experiment is also measured in HBS-EP +1 mg/ml
BSA at 25°C on a BIACORE 2000 instrument with a CM4 chip. Approximately 1000
response units of an antibody of the invention is immobilized on flow cells 2,3 and 4 of
the chip; flow cell 1 is left blank. Using a flow rate of 50 μl/ml, 25 μl of 500 nM human
IL-17 is injected over all four flow cells, forming the antibody:antigen complex on the
surface of the chip. After the injection is complete and the complex formed, 250 μl of

500 nM human IL-17 receptor Fc fusion protein is injected over all four flow cells. At
the end of this injection a 25 μl injection 100 mM glycine pH 1.5 is used to regenerate the
chip. The same binding experiment is then repeated using a 250 ΜL injection of buffer
rather than IL-17 receptor Fc fusion protein.
The binding profiles for both the receptor injection over the antibody:antigen
complex and for the buffer control injection over the antibody :antigen complex are
identical. This indicates that there are no binding sites available for the dimeric IL-17 to
bind to its receptor once it is bound to an antibody of the invention. This result also
Indicates that the receptor is not able to "pull" IL-17 away from any of the antibodies
once the complex is formed. These antibodies can inhibit human IL-17 from binding to
its receptor, therefore neutralizing biological activity of human IL-17.
Example 6A In vitro IL-8 Reporter Assay
To test the ability of an antibody of the invention to neutralize or antagonize an
IL-17 bioactivity, one can utilize the IL-8 reporter assay described herein. This approach
can also be used to determine the potency of Fabs or mAbs of the invention in a cell-
based assay. The human HS27 cell line (ATCC #CRL-1634) secretes IL-8 in response to
IL-17. The IL-17-induced IL-8 secretion is inhibited by neutralizing anti-IL-17
antibodies (See, e.g., J. Imm. 155:5483-5486,1995 or Cytokine 9:794-800,1997).
Accordingly, IL-17-induced IL-8 secretion should proceed unconstrained if sufficient IL-
17 is added to HS27 cells in the absence of neutralizing anti-IL-17 antibody.
HS27 cells are maintained in assay medium: DMEM high glucose medium
lacking phenol red (Invitrogen #31053-028) with 10% fetal bovine serum, 4 mM L-
glutamine, 1 mM sodium pyruvate, penicillin G (100 U/500 ml) and streptomycin (100
μg/500 ml). Cells are grown in T150 flasks until they are about 80-90% confluent the
day of the assay. Human IL-17 (R&D Systems, #317-IL-050) is reconstituted in sterile
PBS without Ca2+ and Mg2+ stored frozen, freshly thawed for use and diluted to 200
ng/ml in assay medium. A 50 ul aliquot of the diluted IL-17 is added to each well of a
96-well flat-bottom tissue culture plate (Falcon #35-3072) with the outer wells left empty.
Duplicate wells are used for a media-only control (100 μl/well) and IL-17-only control
(100 μl/well). Testing is carried out in duplicate or triplicate. Sterile full-length mAb
proteins are diluted to a maximum concentration of 24 μg/ml in assay media. Serial

dilutions (typically 1:5) are made in a separate assay plate and 50 μl of the Fab samples at
the various dilutions are added to the wells containing IL-17 then incubated at 37°C for 1
hour. Assay medium alone is used as a negative control.
HS27 cells (typically about 20,000 cells in 100 μl assay medium) are added to
each well of the plate containing Fab + IL-17 (or controls) and incubated for about 48
hours at 37°C. The media supernatants are then collected after centrifugation of the 96
well plates for 5 minutes at 500 times gravity and diluted 1:15 or 1:10 in assay media.
The level of IL-17 neutralization is measured by determination of IL-8 amounts in
supernatant using a commercial ELISA kit according to manufacturer's instruction except
assay medium is substituted for standard diluent and substrate volume is 100 μl/well
(R&D Systems, ELISA D-8000C or R&D DuoSet ELISA #DY208hIL-8). ELISA
measurements (450 nm) are taken on a microplate reader. Calibration curves are obtained
using a 4-parameter logistic fit with IL-8 values (pg/ml) determined from the calibration
curves using standard statistical techniques. IC50 values are obtained using standard
statistical techniques.
Full-length mabs 103,104,121 and 126 of the invention (with IgG4 Fc region),
when tested in the assay described (2-4 replications), have an average IC50 (based on an
estimated molecular weight of 150 kD for each mAb) of between 450 and 500 pM with
the range of all measured values between 365 and 618 pM.
Example 6B In vitro GROa Reporter Assay
To test the ability of an antibody of the invention to neutralize or antagonize an
IL-17 bioactivity, one can utilize the following cell-based assay. IL-17 can stimulate
epithelial cells and other cells to secrete GROa. The ability of an antibody of the
invention to neutralize IL-17 - induced GROa secretion from the human colorectal
adenocarcinoma epithelial cell line HT-29 is tested in mis assay.
To test whether human IL-17 dose-dependently induced GROa secretion from
HT-29 cells, recombinant IL-17 (R&D Systems #317-IL-050/CF; reconstituted in sterile
Dulbecco's PBS without Ca2+ and Mg2+ (D-PBS)) is diluted (to 4.5 μg/ml; 3X the
highest test concentration) in assay/culture medium (McCoy's 5A (Invitrogen); 10% FBS
(Invitrogen); penicillin G (100 U/500 ml); and streptomycin (100 ug/500 mL IL-17 is
further diluted serially (1:5) in assay medium. Various concentrations of IL-17 (0.096

ng/ml - 1,500 ng/ml; 3.0 pM - 46,875 pM) are dispensed (50 μl each) into inner wells of
a tissue-culture treated 96-well plate. Assay medium (50 μl) is dispensed into 3 wells for
a "medium alone" treatment. Testing is carried out in triplicate (3 wells per treatment).
The plate containing IL-17 in assay medium is incubated for approx. 60-90 minutes at
37°C, 5% CO2, before the addition of HT-29 cells.
For evaluation of an antibody of the invention, e.g., mAb 126 with an IgG4 Fc
region, a concentration of IL-17 that gave approximately 70% of maximal GROa
secretion from HT-29 cells is used (60 ng/ml). Recombinant human IL-17 (R&D
Systems) is diluted (to 240 ng/ml; 4X working concentration) in assay/culture medium.
Diluted DL-17 is dispensed (50 μl) into 60 separate inner wells of tissue-culture treated
96-well plates (Becton Dickinson Falcon #35-3072). Assay medium (50 μl) is dispensed
into 3 wells for a "medium alone" treatment.
A dose range of an antibody of the invention to be tested is typically from 2.56 -
40,000 pM. In a separate dilution plate, the antibody of the invention and control
antibody (sterile, in 1X PBS, pH 7.4) are diluted to 160,000 pM in assay medium. The
antibody of the invention and control antibody are further diluted serially (1:5) in assay
medium. Each test concentration of the antibody of the invention to be tested is then
added (50 μl) to wells containing IL-17. Testing is typically carried out in triplicate.
Assay medium alone (50 μl) is used for "medium alone" and "IL-17 alone" controls.
Plates containing IL-17 and antibody of the invention mixtures are incubated for 60-90
minutes at 37°C, 5% CO2, before the addition of HT-29 cells.
HT-29 cells (human colorectal adenocarcinoma epithelial cells, ATCC #HTB-38),
are maintained in culture/assay medium in tissue culture-treated flasks using standard
techniques. HT-29 cells are grown in tissue culture flasks until they were 50-80%
confluent on the day of the assay. On the day of the assay, the cells are rinsed with HBSS
(Cambrex #CC-5024) and detached from the culture flasks with trypsin + EDTA. The
trypsin is inactivated with complete assay medium. HT-29 cells are then centrifuged at
SOOXg for 5 min. at RT. The cell pellet is then re-suspended in assay medium and 20,000
HT-29 cells (in 100 μl) are added to each treatment well of the 96-well plates. An equal
volume of D-PBS is added to each of the unused edge wells (without cells) to reduce

edge effects resulting from evaporation. The 96-well plates were placed in a tissue culture
incubator (37°C, 5% CO2) for approximately 48 hours.
At the end of the assay, the plates are centrifuged (SOOXg for 5 min. at RT), and
the cell culture media is transferred to polypropylene 96-well plates. GROa levels are
measured with a GROa sandwich ELISA (R+D Systems DuoSet #DY275), as per the
manufacturer's instructions, except for: using assay medium as the standard diluent, using
IX ELISA wash buffer from BioFX Labs, using a sample and standard volume of SO μl
per well, using a substrate from BioFX Labs (HRP substrate, #TMBW-1000-01), and
using a stop solution from BioFX Labs (#LSTP-1000-01) (100 μl per well). At the end of
the ELISA reactions, plates are read at 450 nm on a microplate reader. Calibration curves
for GROa are obtained by performing a 4-parameter logistic fit GROa values
(concentration in pg/ml) for the samples are obtained from the calibration curves. The
human colorectal adenocarcinoma epithelial cell line HT-29 secreted GROa when
stimulated with IL-17, in a dose-dependent manner (Table 5). Control human IgG4 did
not cause a decrease in IL-17 - induced GROa secretion. These results (Table 6)
demonstrate that mAb 126 is able to completely neutralize human IL-17 - induced GROa
secretion from HT-29 cells in vitro using the conditions described. The IC50 value for
mAb 126 in this assay is approximately 560 pM.



Human IL-17 is able to bind and stimulate the mouse IL-17 receptor, leading to an
elevation and subsequent secretion of mouse KC (CXCL1) chemokine. Time and dose
ranging experiments are undertaken to identify the optimal dose of human IL-17 and the
optimal time for induction of mouse KC. These experiments indicate that a 150 μg/kg
dose of human IL-17 and a time of 2 hours post IL-17 administration gives maximal
levels of KC in mouse serum. Full-length antibodies of the present invention (e.g., Fab
126 or Fab 121 with HCVR operably linked to human IgG4 Fc, SEQ ID NO:260 [or SEQ
ID NO: 278] and the LCVR operably linked to a human kappa constant region, SEQ ID
NO: 263 [or SEQ ID NO: 277]) are administered intravenously to mice at 1,10,100 and
1000 μg/kg) one hour prior to a subcutaneous injection of human IL-17. At two hours
after human IL-17 administration, the mice are sacrificed and KC levels are determined
by ELISA using a commercially available kit according to manufacturer's instruction
(KC Quantikine, R&D). Isotype matched antibodies are used as negative controls. The
antibodies block the ability of human IL-17 to stimulate the mouse IL-17 receptor,
leading to inhibition of an elevation of mouse KC, in a dose dependent manner. Mabl26
(a full length antibody comprising Fab 126), at a dose of 20 ug/mouse under the
conditions described, decreases the mean KC level by approximately four-fold compared
to a control antibody which had no effect. Mab 121, at a dose of 20 ug/mouse under the
conditions described, decreases the mean KC level by approximately three-fold compared
to a control antibody.
Example 8 Epitope Mapping
Two of the anti-IL-17 antibodies (Fab 126 and Fab 104) are used to determine that
the humanization and optimization of the parent murine Fab (2321, SEQ IDNOs: 261

and 262) do not alter the epitope-binding ability of the Fabs resulting from humanization
and optimization of the parent The humanized, optimized Fabs bind to the same epitope
as does the parent murine Fab as determined by a standard competition ELISA or by H-D
exchange and mass spec analysis for epitope mapping (See, e.g., Hoofnagle, A., et al.,
Methods Mol. Biol. 250:283-298,2004; Hoomagle, A., et al., Ann. Rev. Btophys. Biomol.
Struct., 32:1-25,2003; Baerga-Ortiz, A., et aL, Protein Sci. 11:1300-8,2002) therefore,
Fabs 1-132 of the invention derived from the same parent Fab, would be expected to bind
the same epitope.
Using the H-D exchange and mass spec assay (H/DXMS) to map the epitope, it is
determined that amino acids between 80 and 89 [ADGNVDYHMN (SEQ ID NO: 266)]
of human IL-17 (SEQ ID NO: 1) are comprised within the discontinuous epitope to which
the antibodies of the invention bind. DGNVDYH (SEQ ID NO: 267) is an essential
sequence comprised within the discontinuous epitope to which antibodies of the invention
bind based on comparison of sequence variation of IL-17 among different species and
binding capability. Changing the amino acid sequence of SEQ ID NO: 267 within the
context of entire IL-17 sequence, results in no detectable binding to the altered IL-17 by
an antibody of the invention. Antibodies of the invention do not bind to rat or mouse IL-
17 at levels greater than control antibody.
H/DXMS assay is used to identify regions of IL-17 to which antibodies of the
invention bind. The rate of amide hydrogen exchange rate is dependent on the structure
and solvent accessibility of the amid hydrogen. Free IL-17 or IL-17: antibody complex in
water is mixed with deuterated water (D2O) to allow exchange of amide protons by
deuterium. Those backbone amide groups that participate in protein binding are
protected from exchange and remain protonated. These regions are then identified by
peptic proteolysis, coupled with LC and electrospray ionization mass spectrometry.
Human IL-17 containing a C-terminal His and Flag tag (IL-17-Flis) is expressed and
purified from GS-CHO cells using an IMAC column. Two 10 fig aliquots (7.7 μl) of IL-
17-Flis solution is transferred into 2 Microcon, and 100 ug of either mAb 104 or mAb
126 (molar ratio of IL-17/Mab -1/2) is added into Microcoa Twenty μg of IL-17-Flis
solution is transferred into another Microcon and no antibody added. Then lx PBS buffer
is added into each Microcon to the final volume of -180 μl and centrifuged at 14,000g for
14 min. Then 150 ml of lx PBS buffer is added into each Microcon and centrifuged at

14,000g for 14 rain. These steps are necessary to ensure the free antigen and the
antigenrantibody complex are in identical buffer conditions.
The protein portion is collected and the final volume is adjusted to SO ul (complex)
or 80 ul (IL-17-Flis only) with lxPBS. Six microliters of IL-17-Flis or complex of IL-
17-Flis and mAb complex are transferred into a micro plastic vial, and 14 ul of 100%
D2O is added into it, resulting in 70% D2O in the sample. The solution is incubated at
ambient temperature for 10 min. The exchange is immediately quenched, digested by
adding 20 ul of 1% formic acid solution and 2 ul of 2 mg/ml pepsin solution, and
incubated at ambient temperature for 30 sec or at 0°C for 10 min. The digest is
immediately injected onto a column manually. Waters 2795 HPLC and Micromass LTC
Premier are used for all assays. HPLC stream from HPLC pump is connected to a metal
tube (about 1 ml), to manual injector, to a Zorbax C18 column (2.1x50mm) running
under these settings (Column Temperature:0°C; Mobile Phase C: 0.15% formic acid in
H2O, D: 0.12% Formic acid in ACN; Run Time:23 min). The column is equilibrated
with 98% A (0.15% formic acid aqueous solution) and 2% B (0.12% formic acid in
acetonitrile) at a flow rate of 0.2 ml/min. A gradient elution is performed from 2% to
10% B over 0.5 min, then to 40% B over 14.5 min, then to 90% B over 1 min with 2 min
hold, and then returned to 2%B in 1 min.). The sample from HPLC is analyzed by mass
spectrometer operated with these settings (Ion Mode: Positive; Mass Scan Range: 300-
2000; Sample Cone Voltage: 80; Desolvation Gas Flow (L/Hr):700; Desolvation Temp:
300°C). The metal tube, injector loop and column are submerged in ice water throughout
the assay. Mass spectrum of each peptic peptide of IL-17 is obtained after H/D exchange
with or without an anti-IL-17 mAb tested. For small peptides, the average mass of each
peptide is calculated based on its isotopic ions and intensities. For larger peptides, the
average masses are obtained from deconvoluted mass spectra after internal calibration.
When antibody forms a complex with IL-17, the binding region (epitope) of IL-17
is protected from solvent This leads to slower amide hydrogen exchange rates when
compared to those of IL-17 alone. By comparing the mass of peptides from the free and
from the complex after deuterium exchange, the peptides protected by complex formation
should be different from the corresponding peptide in free IL-17. Table 7 below lists
mass differences that are obtained by H/DXMS for peptic peptides of IL-17. These peptic
peptides cover the whole sequence of IL-17-Flis. As the data in the table demonstrate,

the mass difference of IL-17-Flis peptide between the complex and itself is similar for
both antibodies tested, i.e. they bind the same epitope. A major mass difference is found
for the peptic peptide 24-87+117-133 (i.e., amino acids 24 to 87 and 117 to 133 of IL-17)
(these two peptides are connected through disulfide bond) and 66-87+117-134,
suggesting residues within these regions are involved in binding. Since those peptic
peptides are quite large, other enzymatic digests are necessary to narrow down specific
amino acid residues involved in binding. In addition to this data, the antibodies of the
invention do not bind to other member of IL-17 family (IL-17 B,C,D,E, and F) and they
also do not bind to mouse or rat IL-17. These data together with sequence comparison
and examination of IL-17 homology structural model suggest that residues 80-89 are
comprised within a non-linear epitope of IL-17 to which the antibodies of the invention
bind.

Example 9 IL-17 Expression in Cancer Tissues
Various human non-cancerous and cancerous cell lysates are tested for the
presence of IL-17 protein. Tissues (approximately 50-100 mg piece) are snap-frozen on
dry ice, thawed on ice and lysed in 350 μl TPER buffer (Pierce #78510) including
protease inhibitors (Pierce #78410) and phosphatase inhibitors in tubes containing
ceramic lysing beads (Qbiogene #6913-050; 1.4 mm ceramic beads in 2.0 ml tubes). The
tubes are placed on ice for 5-10 min then centrifuged at 13,000 x gravity for 10 min at

4°C and the material transferred to new tubes to remove debris. Recentrifuge as
described and transfer to new tube. Protein concentration is determined using a standard
BSA method. The samples are analyzed for IL-17 using a commercial IL-17 ELISA kit
according to manufacturer's instructions (R&D #DY317 using wash buffer, substrate
solution and stop solution from BioFX Labs). IL-17 levels are normalized to total protein
concentration. IL-17 levels are increased between two- and three-fold in cancerous colon
tissue (60 samples tested) as compared to normal colon tissue (63 samples tested). IL-17
levels are increased on average three- to four-fold in cancerous kidney tissue (21 samples
tested) as compared to normal kidney tissue (21 samples tested). IL-17 levels in
cancerous prostate tissue (44 samples tested) are increased as compared to the normal
prostate tissue (7 samples tested). The IL-17 levels were not elevated in other types of
tumor tissue tested including breast, neck, lung, larynx, thyroid, tongue, ovary and brain.
Example 10 IL-17 Activation of Microglial Cells
IL-17 induces a tnurine brain microglia cell line (BV-2) to secrete IFNand IL-
12p70. The BV-2 murine microglial cell line [obtained from Scios, with permission from
Elisabeta Blasi (Microbiology University of Perugia, Italy) who originally isolated them
(E. Blasi et al., J. Immunology 1990,27:229-237)] are cultured on poly-D-lysine coated
tissue culture flasks, to no greater than 60% confluence in high-glucose DMEM
(Invitrogen #31053-028) with 2 mM L-glutamine (Invitrogen/GEBCO #25030-081), 10%
FBS (heat inactivated; Invitrogen/GIBCO #10082-147), 1 mM sodium pyruvate
(Invitrogen/GIBCO #11360-070), 100 μg/ml Nonnocin (InvivoGen) at 37°C, 5% CO*
On day 0 of the assay, BV-2 cells are rinsed (Dulbecco's PBS without Ca2+ and
Mg2+; Invitrogen), detached (0.25% trypsin + EDTA) followed by trypsin inactivation
then centrifuged (500Xg 5 min. at RT). The resulting cell pellet is re-suspended to a cell
density of~7,000 cells/100 μl culture medium. 100 ul of cell suspension is dispensed into
60 separate inner wells of poly-D-lysine coated tissue-culture treated 96-well plates.
Plates are incubated as described, for approx. 48 hrs before treatment with IL-17.
On day 2 of the assay, recombinant mouse IL-17 (mIL-17) (carrier-free; R&D
Systems); reconstituted in sterile Dulbecco's PBS without Ca2+ and Mg2+ is diluted in a
polypropylene plate to 1.5 μg/ml (the highest test concentration) in culture medium.
Mouse IL-17 is further serially diluted in the polypropylene plate. A positive control is

LPS diluted in culture medium to 1 μg/ml (the highest test concentration). Assay medium
is used as a negative control. Medium is gently aspirated from the cells, before adding
treatments (150 jilAvell). Testing is carried out in triplicate (3 wells per treatment).
Separate replicate plates are incubated for either 24 hr. or 48 hr. at 37°C, 5% CO2.
On days 3 and day 4 of the assay, plates are centrifuged (500Xg for 5 min. RT),
then cell culture media is transferred to polypropylene 96-well plates, which are sealed
and frozen (-80°C). Media samples are thawed and assayed for cytokine and chemokine
levels with a murine 22-plex multiplex kit (Linco), as per the manufacturer's instructions
(except: a black-walled polycarbonate filter plate (Millipore) replaces the filter plate
included in the kit). Fluorescence is read on a Luminex® instrument (50 beads per bead
set, low RP1 gain setting). Data is shown in Table 8 below.
Standard curves are obtained using a four- or five-parameter logistic fit. IFNγ and
IL-12p70 values (pg/ml) are determined from the standard curves using standard
statistical techniques.


IBD is a chroNIc inflammatory disease which includes Crohn's Disease and
Ulcerative Colitis. IL-17 protein levels are significantly elevated in the sera and in colon
tissues from both ulcerative colitis and Crohn's disease patients. However, IL-17 is not
detectable in the sera from normal individuals, or patients with infectious colitis or
ischaemic colitis The DSS (Dextran Sodium Sulfate) model is one of the oldest and most
representative pre-clinical model for irritable bowel disease (IBD). In the DSS model
(see, e.g., FASEB Journal 2004; 18:1550-1552) both acute and chronic inflammatory
lesions are induced. Mice have a high degree of uniformity of the legions with losing
body weight and colon length. It is reproducible in respect of time course and severity
among individual mice. For disease induction, mice receive 5% DSS 30-40 Kd) in
drinking water for 7 days. Disease Activity Index (DAI) including hemoccult positive or
rectal bleeding, loose stool and loss of body weight (5-8%) is observed at about day 8.
Body weights of mice are monitored every day for 2 weeks. Mice are sacrificed from
about day 12 to about day 15. DL-17 protein is significantly increased in DSS-treated
colon versus narve colon. Treatment with IL-17 antibody may reduce the disease activity
index.
Example 12 EAE Model for Multiple Sclerosis
EAE is a CD4+ T cell-mediated demyelinating disease of the central nervous
system (CNS) that serves as a model for MS in humans. The pathogenic mechanisms of
EAE development include antigen-specific T cell activation and Th1 differentiation
followed by T cell and macrophage infiltration into the CNS. IL-17 contributes to the
pathology of multiple sclerosis (MS). Microarray analysis of MS lesions of human
patients have demonstrated an increase of IL-17 (Lock, et al. Nat Med. 8:500-508,2002).
IL-17 mRNA-expressing mononuclear cells (MNC) in blood and cerebrospinal fluid are
significantly elevated in number in MS patients and higher numbers of IL-17 mRNA-
expressing blood MNC were detected during MS clinical exacerbation compared to
remission (Matusevicius, et al. Multiple Sclerosis. 5:1-1-104,1999). EAE is
significantly suppressed in IL-17 knockout mice (Nakae et al., J. Immun. 171:6173-
6177).
The example described here demonstrates that IL-17 protein is increased in the
spinal cord of EAE mice and treatment with an anti-murine IL-17 antibody reduces the

EAE score in the active EAE model. For disease induction, 8-9 week old female
C57BL/6 mice are subcutaneously immunized on day 0 with either (i) 200 μl of 5 mg/ml
pertussis toxin (PT) and Complete Freund's Adjuvant (CFA) or (ii) PT, CFA and 300
μg/200 μl of MOG35-55 (myelin oligodendrocyte glycoprotein emulsified in CFA
containing 5 mg/ml of heat inactivated Mycobacterium tuberculosis). On day 2, mice are
treated again with PT. Mice are scored throughout the study for levels of paralysis.
Disease is expected in the group receiving MOG. A rat anti-murine IL-17 monoclonal
IgG1 antibody or isotype control antibody is administered to mice on days 1,7 and 15
(BD Biosciences for rat anti-murine IL-17 antibody). Mice receiving MOG are sacrificed
when clinical score reaches between 1-3 (on a scale of 0-4); this is between days 14-31
for study 1 in Table 9 below and between days 14-16 for study 2 in Table 10 below.
Clinical signs of disease develop about day 10. Individual animals are subjectively
scored by at least 2 scorers independently and blinded to the identity of treatment groups
according to clinical CNS disease severity. Grade 0 is normal; Grade 1 is completely
limp tail; Grade 2 is unilateral partial hind limb weakness; Grade 3 is complete hind limb
paralysis; and Grade 4 is moribund, (see J. Exp. Med. 194: 873-881,2001). A control
mouse is sacrificed on the same day as a MOG-treated mouse. Spinal cords are isolated
at the time of sacrifice and flash frozen to be used for IL-17 protein analysis by ELISA.
IL-17 antibody treatment group has significantly lower disease scores as compared to
isotype control group.
Ly sates of each whole spinal cord are made in 1 ml (study 1 in Table 9 below) or
0.4 ml (study 2 in Table 7 below) TPER protein extraction reagent (Pierce #78510) with
complete protease inhibitors (Roche Applied Science #11697498). in 2 ml tubes
containing ceramic beads (lysing matrix D, QBiogene #6913050), and FastPrep
instrument (BiolOl) for 30 seconds at a scale of 5.5. After lysis, samples are centrifuged
(5 min. at 14,000 rpm in a microfuge) to remove debris. Supematants are transferred to
new microfuge tubes. Total protein concentration in each lysate is determined with a
BCA protein assay kit (Pierce #23225), vising the microplate protocol of the
manufacturer. Lysates are frozen and stored at -80°C.
After thawing lysates on ice, and clarifying by centrifugation, mouse IL-17 levels
are measured in undiluted samples by ELISA (R&D Quantikine #M1700) as per
manufacturer's instructions. Standard curves are obtained using a four-parameter logistic

fit. IL-17 values are detennined from the standard curves using standard statistical
techniques. IL-17 levels are normalized to protein concentration in each sample and
expressed as pg IL-17/ml total protein in each lysate in Tables 9 and 10 below. As
demonstrated by the data in the tables, increased levels of IL-17 were detected in EAE
mice.

Example 13 Collagen-induced arthritis model
Collagen-induced arthritis (CIA) is a widely used rodent model for rheumatoid
arthritis ("RA") and has histopathological features in common with human RA.
Experimental arthritis, induced in DBA/1 mice by immunization and boosting with
emulsions of type n collagen, is a polyarthritic disease characterized by inflammation of
the small joints and progressive erosion of cartilage and bone (Trentham, D, et al, J. Exp.
Med. 146:857-858,1977). Recently, Lubberts, et al, (Arthritis & Rheumatism, 50:650-
659,2004; incorporated herein) demonstrated that poryclonal rabbit anti-murine IL-17
antibody, administered either at the onset or at a later stage of murine CIA, ameliorated
clinical signs of arthritis.
In the CIA model, mice given a single injection of rat anti-murine IL-17 IgG2a
mAb intraperitoneally (8mg/kg R&D, MAB421 clone 50104.11) show significantly lower
clinical scores than mice injected with 16 mg/kg control IgG2a. The acute phase

reactant, C-reactive protein (CRP), is an accepted index of disease activity in RA patients.
Similar to CRP, murine serum amyloid protein (SAP) serves as an indicator of disease in
the murine CIA model (Bliven, M., et al, Arthritis & Rheumatism, 29:1131-1138,1986).
In animals treated with 8 mg/kg of anti-murine IL-17, SAP levels were significantly
lower than in those treated with control antibody. Moreover, the decrease in clinical
scores and SAP values are comparable to an anti-mouse IL-1β group (8 mg/kg) used as a
positive control. Finally, significant reduction in synovial inflammation at 8mg/kg
antibody and bone resorption at 16 mg/kg antibody is present compared to that of mice
treated with control antibody. A dose response study may be conducted in the CIA model
with anti-murine IL-17 antibody (e.g., at 0.1, 1 and 8 mg/kg). The clinical scores for rat
anti-murine IL-17 display a trend for dose responsiveness. A similar assay may be
performed in cynomolgus monkeys as a model for RA using an antibody of the invention.
Example 14 Anti-IL-17 mAb Purification
A vector expressing a mAb of the invention is stably incorporated into an
appropriate host cell, (e.g., CHO DG44 (dhfr-) cells (Chasin) or NSO cells) using
standard procedures and purified using Protein A affinity column. Briefly, clarified
conditioned media is applied to a 5 ml HiTrap rProtein A Sepharose FF column
(Amersham Biosciences) that has been equilibrated with PBS (pH 7.4). The column is
washed with 5 column volumes of equilibration buffer at a flow rate of 110 cm/hr to wash
out nonspecific binding components. The bound antibody is eluted using a linear pH
gradient (0.1 M sodium phosphate buffer pH 6.8 to 0.1 M sodium citrate buffer pH 2.5).
The main protein peak in the elution is collected and its pH adjusted to neutrality with 1
M Tris buffer (pH 8.5). The protein pool is concentrated to 1-2 mg/ml using 10K
Vivaspin membrane (Vivasciences) and sterile filtered (0.45 μm) before storage at 4°C.
For large preparations of a mAb of the invention, the cell free concentrate is
purified over three sequential chromatography columns (Protein A, Anion Exchange, and
Hydrophobic Interaction chromatography). The purity of the mAb after these
chromatography steps is greater than 99% as assessed by analytical size exclusion
chromatography. The mAb is exchanged into a buffer as listed below depending upon the
concentration of the antibody. Chemical stability results indicate a preferred pH between
6.0 and 7.0 (inclusive); although for 20 mg/ml preparations, the pH may be between 5.5

and 7.0 (inclusive, e.g., 5.5., 5.6,5.7., 5.8,5.9,6.0,6.1,6.2, 6.3,6.4,6.5, 6.6,6.7, 6.8,6.,
or 7.0). For lyophilized product, a sodium chloride level of 90-30 mM (90,85, 80,75,
70, 65, 60,55, 50,45,40,35 or 30 mM or any value between 30 and 90 mM) is
preferred, while for a liquid formulation (e.g., to be administered subcutaneously) a
sodium chloride level of 100 - 150 mM (100,110,120,130,140, or 150 mM or any value
between 100 and 150 mM) is preferred. The product is then concentrated to a final
concentration of about 10,20 or 25 nag/ml (alternatively higher, 30,40, 50,60, 70, 80,90,
100, 110, 120,130, 140,150 mg/ml or higher) and sterile filtered. The filtered product
may be immediately frozen at -70°C or may be lyophilized. A minimal weight ratio of
1:2 for antibody to lyoprotectant, (e.g., sucrose or trehalose) is needed for stable
lyophilized formulation but is not required for a liquid formulation. Additionally, 0.02%
surfactant (w/v), i.e., polysorbate-80, is added for both solution formulations and
solutions to be lyophilized. The lyophilized material is resuspended in sterile Water for
Injection or sterile 0.9% sodium chloride prior to administration.

Serum pharmacokinetics of antibodies of the invention (e.g., mAb 126 and 121
[IgG4 Fc region with Fab 126 or 121 respectively]) are determined after intravenous or
subcutaneous administration in male cynomolgus monkeys. Concentrations of the
antibodies in the serum are determined using a standard antigen-capture ELJSA assay in
which plates are coated with human IL-17 and bound serum antibody is detected using an
anti-IgG4 secondary antibody. Following intravenous administration of 1 mg/kg, mAb
126 is eliminated with a mean half-life of 6.5 days and mAb 121 is eliminated with a
mean half-life of about 11 days. Following subcutaneous administration of 1 mg/kg,

mAb 126 has a mean elimination half-life of 10.3 days and mAb 121 has a mean
elimination half-life of 13 days.
Example 16 Tumor Xenograft Model
To establish tumor xenograft models with which to test the antitumor activity of
anti-IL-17 antibodies of the invention, 5 million HCT116 colorectal carcinoma cells are
mixed with Matrigel and subcutaneously injected into the left flank of 56-week-old
female athymic (nu/nu) mice ( Charles River laboratories, Wilmington. MA). Mice are
treated by subcutaneous injection every 7 days with control antibodies (e.g., human IgG4
and mouse IgG1), 4 mg/kg anti-human-IL-17, 8 mg/kg anti-mouse IL-17, or combination
of 4 mg/kg anti-human-IL-17 and 8 mg/kg anti-mouse-IL-17 for 4 weeks. The first
antibody administration starts one day prior to implanting the cells. Tumors are measured
twice each week with a caliper and body weight is monitored twice a week. Plasma is
collected from each mouse at day 34 and KC levels are measured using a KC ELISA kit
according to manufacturer's instructions (R&D System). In comparison with control
IgG-injected mice, mice treated with the combination of anti-human IL-17 antibody and
anti-mouse-IL-17 antibody have significantly reduced tumor volume. Furthermore, mice
treated with both anti-human IL17 antibody and anti-mouse IL17 antibody have
dramatically decreased plasma KC. The mice treated with either 4 mg/kg anti-human
IL17 antibody or 8 mg/kg anti-mouse IL 1 7 antibody revealed no significant reduction in
tumor volumes and plasma KC levels. Data are shown in Tables 12 and 13 herein.
To measure IL17 level in the tumors, the tumors from mouse xenograft models are
prepared largely as described in Example 9. For protein measurement, tumor lysates are
diluted 1:10 in TPER + IX Halt in a polypropylene 96-well dilution plate. Protein
concentration is determined by using the microplate protocol of the Coomassie Plus
Protein Assay (Pierce #23236). BSA standard is diluted in TPER + Halt IL-17 protein
levels are determined using human and mouse IL-17 ELISA kits from R&D System as
per manufacturer's instructions (human IL-17 DuoSet ELISA, R+D Systems, Cat
#DY317; mouse IL-17 ELISA, R+D System, Cat # 421). Both human sad mouse IL-17
were increased in tumors from HCT116 and HT29 colon tumor xenograft models as
compared to H460 lung tumor xenograft model.



We Claim:
1. A humanized anti-IL-17 monoclonal antibody, wherein the antibody binds human
IL-17 with a KD less than 7.0 pM.
2. The antibody of claim 1, wherein said antibody further has a dissociation rate
constant or koff rate of less than 5 x 10-5 s-1.
3. The antibody according to any one of claims 1 -2, wherein said antibody
specifically binds a non-linear epitope of human IL-17, wherein the epitope
comprises a polypeptide with SEQ ID NO: 276.
4. The antibody according to any one of claims 1 to 3, wherein said antibody
neutralizes IL-17 bioactivity in an in vitro IL-8 reporter assay at an IC50 of less
than 1 nM.
5. An anti-IL-17 monoclonal antibody, wherein said antibody competes for binding
to human IL-17 with a humanized anti-IL-17 antibody comprising two
polypeptides with the amino acid sequences shown in SEQ ID NO: 241 and SEQ
ID NO: 118.
6. A humanized anti-IL-17 monoclonal antibody comprising a heavy chain variable
region (HCVR) comprising a polypeptide with an amino acid sequence selected
from the group consisting of SEQ ID NOs: 56-121.
7. A humanized anti-IL-17 monoclonal antibody comprising a light chain variable
region (LCVR) comprising a polypeptide with an amino acid sequence selected
from the group consisting of SEQ ID NOs: 178-243.
8. A humanized anti-IL-17 antibody comprising a CDRL1 polypeptide with SEQ ID
NO: 247, a CDRL2 polypeptide with SEQ ID NO: 248, a CDRL3 polypeptide
with SEQ ID NO: 249, a CDRH1 polypeptide with SEQ ID NO: 244, a CDRH2
polypeptide SEQ ID NO: 245, and a CDRH3 polypeptide with SEQ ID NO: 246.

9. The humanized monoclonal antibody according to any one of claims 1 to 8,
wherein said antibody is a humanized or fully human antibody or antigen-binding
portion thereof.
10. The antibody according to any one of claims 1 to 9, wherein the antibody is a
humanized full-length antibody, a humanized substantially intact antibody, a Fab
fragment, a F(ab')2 fragment or a single chain Fv fragment.
11. The antibody according to any one of claims 1 to 10, wherein the antibody further
comprises a heavy chain constant region selected from the group consisting of
IgG,, IgG2, IgG3, IgG4, IgA, IgE, IgM and IgD.
12. A composition comprising the antibody according to any one of claims 1 to 11
and wherein said composition further comprises a pharmaceutically acceptable
carrier.
13. An antibody according to any one of claims 1 to 11, for use as a medicament.
14. Use of an effective amount of the antibody according to any one of claims 1 to 11
in the preparation of a medicament for the treatment of one or more conditions
selected from the group consisting of rheumatoid arthritis, inflammatory bowel
disorder, psoriasis, and multiple sclerosis.

Anti-IL-17 antibodies are identified that are characterized as having a high affinity and
slow off rate for human IL-17. The antibodies of the invention may be chimeric,
humanized or fully human antibodies, immunoconjugates of the antibodies or antigen-
binding fragments thereof. The antibodies of the invention are useful in particular for
treating autoimmune, inflammatory, cell proliferative and developmental disorders.

Documents:

01838-kolnp-2008-abstract.pdf

01838-kolnp-2008-claims.pdf

01838-kolnp-2008-correspondence others.pdf

01838-kolnp-2008-description complete.pdf

01838-kolnp-2008-drawings.pdf

01838-kolnp-2008-form 1.pdf

01838-kolnp-2008-form 2.pdf

01838-kolnp-2008-form 3.pdf

01838-kolnp-2008-form 5.pdf

01838-kolnp-2008-gfa.pdf

01838-kolnp-2008-international publication.pdf

01838-kolnp-2008-international search report.pdf

01838-kolnp-2008-pct priority document notification.pdf

01838-kolnp-2008-pct request form.pdf

1838-KOLNP-2008-(07-10-2011)-CORRESPONDENCE.pdf

1838-KOLNP-2008-(07-10-2011)-FORM 3.pdf

1838-KOLNP-2008-(08-04-2014)-CORRESPONDENCE.pdf

1838-KOLNP-2008-(08-09-2011)-ABSTRACT.pdf

1838-KOLNP-2008-(08-09-2011)-AMANDED CLAIMS.pdf

1838-KOLNP-2008-(08-09-2011)-CORRESPONDENCE.pdf

1838-KOLNP-2008-(08-09-2011)-DESCRIPTION (COMPLETE).pdf

1838-KOLNP-2008-(08-09-2011)-DRAWINGS.pdf

1838-KOLNP-2008-(08-09-2011)-FORM 1.pdf

1838-KOLNP-2008-(08-09-2011)-FORM 2.pdf

1838-KOLNP-2008-(08-09-2011)-FORM 3.pdf

1838-KOLNP-2008-(08-09-2011)-OTHERS.pdf

1838-KOLNP-2008-(08-09-2011)-PA.pdf

1838-KOLNP-2008-(08-09-2011)-PETITION UNDER RULE 137-1.1.pdf

1838-KOLNP-2008-(08-09-2011)-PETITION UNDER RULE 137.pdf

1838-KOLNP-2008-(08-09-2011)-PETITION UNDER RULE 138.pdf

1838-KOLNP-2008-(12-12-2014)-CORRESPONDENCE.pdf

1838-KOLNP-2008-(25-02-2013)-CORRESPONDENCE.pdf

1838-KOLNP-2008-(25-02-2013)-FORM 1.pdf

1838-KOLNP-2008-(26-11-2014)-CLAIMS.pdf

1838-KOLNP-2008-(26-11-2014)-CORRESPONDENCE.pdf

1838-KOLNP-2008-(26-11-2014)-OTHERS.pdf

1838-KOLNP-2008-ASSIGNMENT.pdf

1838-KOLNP-2008-CORRESPONDENCE 1.2.pdf

1838-KOLNP-2008-CORRESPONDENCE 1.3.pdf

1838-KOLNP-2008-CORRESPONDENCE OTHERS 1.1.pdf

1838-KOLNP-2008-CORRESPONDENCE.pdf

1838-KOLNP-2008-FORM 13-1.1..pdf

1838-KOLNP-2008-FORM 13.pdf

1838-kolnp-2008-form 18.pdf

1838-KOLNP-2008-OTHERS.pdf

1838-KOLNP-2008-PA.pdf

1838-KOLNP-2008-SEQUENCE LISTING.pdf


Patent Number 265028
Indian Patent Application Number 1838/KOLNP/2008
PG Journal Number 06/2015
Publication Date 06-Feb-2015
Grant Date 02-Feb-2015
Date of Filing 09-May-2008
Name of Patentee ELI LILLY AND COMPANY
Applicant Address LILLY CORPORATE CENTER, CITY OF INDIANAPOLIS, STATE OF INDIANA
Inventors:
# Inventor's Name Inventor's Address
1 HUANG, LIHUA 3536 CORSHAM CIRCLE, CARMEL, INDIANA 46032
2 LU, JIRONG 13793 STONE DRIVE, CARMEL, INDIANA 46032
3 NG, KINGMAN 10745 PUTNAM PLACE, CARMEL, INDIANA 46032
4 ALLAN, BARRETT 542 SAN DIEGUITO DRIVE, ENCINITAS, CALIFORNIA 92024
5 LIU, LING 3393 KILKENNY CIRCLE, CARMEL, INDIANA 46032
6 TETREAULT, JONATHAN, WENDELL 3232 DEVEREAUX DRIVE, INDIANAPOLIS, INDIANA 46228
7 WERNER, ANDREW, GORDON 1550 NORTHBROOK DRIVE, INDIANAPOLIS, INDIANA 46260
8 CHOW, CHI-KIN 7348 WINDSOR LAKES DRIVE, INDIANAPOLIS, INDIANA 46237
PCT International Classification Number C07K 16/24
PCT International Application Number PCT/US2006/061586
PCT International Filing date 2006-12-05
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/749,953 2005-12-13 U.S.A.
2 60/801,948 2006-05-19 U.S.A.