Language selection

Search

Patent 2647282 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2647282
(54) English Title: CTLA4 ANTIBODY COMBINATION THERAPY
(54) French Title: POLYTHERAPIE A BASE D'UN ANTICORPS ANTI-CTLA4
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 39/395 (2006.01)
  • A61P 31/18 (2006.01)
  • A61P 35/00 (2006.01)
  • C07K 16/28 (2006.01)
(72) Inventors :
  • GOMEZ-NAVARRO, JESUS (United States of America)
(73) Owners :
  • PFIZER PRODUCTS INC. (United States of America)
(71) Applicants :
  • PFIZER PRODUCTS INC. (United States of America)
(74) Agent: SMART & BIGGAR
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2007-03-26
(87) Open to Public Inspection: 2007-10-11
Examination requested: 2008-09-24
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/IB2007/000860
(87) International Publication Number: WO2007/113648
(85) National Entry: 2008-09-24

(30) Application Priority Data:
Application No. Country/Territory Date
60/789,662 United States of America 2006-04-05

Abstracts

English Abstract

The invention relates to administration of an anti-CTLA4 antibody, particularly human antibodies to human CTLA4, such as those having amino acid sequences of antibodies 3.1.1, 4.1.1, 4.8.1, 4.10.2, 4.13.1, 4.14.3, 6.1.1, 11.6.1, 11.7.1, 12.3.1.1, 12.9.1.1, CP-675,206 (11.2.1), and ipilimumab, in combination with a therapeutic agent for treatment of cancer. An exemplary method of the invention comprises administering an anti-CTLA4 antibody, preferably, CP-675,206, and a chemotherapeutic agent, preferably, gemcitabine, for treatment of pancreatic cancer, among other treatment modalities.


French Abstract

L'invention concerne l'administration d'un anticorps anti-CTLA4, en particulier d'anticorps humains dirigés contre le CTLA4 humain, tels que ceux comportant les séquences d'acides aminés des anticorps 3.1.1, 4.1.1, 4.8.1, 4.10.2, 4.13.1, 4.14.3, 6.1.1, 11.6.1, 11.7.1, 12.3.1.1, 12.9.1.1, CP-675,206 (11.2.1) et ipilimumab, en association avec un agent thérapeutique pour traiter le cancer. Un procédé selon l'invention donné à titre d'exemple consiste à administrer un anticorps anti-CTLA4, de préférence le CP-675,206, et un agent chimiothérapeutique, de préférence la gemcitabine, pour traiter le cancer du pancréas, parmi d'autres modalités de traitement.

Claims

Note: Claims are shown in the official language in which they were submitted.




-112-

CLAIMS

1. A method for the treatment of cancer in a patient in need of such
treatment,
said method comprising administering to said patient a therapeutically
effective amount of
CP-675,206 anti-CTLA4 antibody in combination with a therapeutically effective
amount of at
least one therapeutic agent, wherein said cancer and said agent are selected
from the group
consisting of:
(a) the cancer is non-Hodgkin's lymphoma (NHL) and wherein the agent is
rituximab;
(b) the cancer is NHL and the agent is cyclophosphamide, doxorubicin,
vincristine,
and prednisone (CHOP);
(c) the cancer is NHL and the agent is cyclophosphamide, doxorubicin,
vincristine,
prednisone and rituximab (CHOP-R);
(d) the cancer is lung cancer and the agent is bevacizumab;
(e) the cancer is non-small cell lung cancer (NSCLC) and the agent is
gefitinib;
(f) the cancer is NSCLC and the agent is bevacizumab;
(g) the cancer is NSCLC and the agent is a taxane and gemcitabine, and further

wherein the taxane is selected from the group consisting of docetaxel and
paclitaxel;
(h) the cancer is NSCLC and the agent is a taxane and a platinum compound;
(i) the cancer is NSCLC and the agent is docetaxel;
(j) the cancer is NSCLC and the agent is erlotinib;
(k) the cancer is NSCLC and the agent is pemetrexed;
(l) the cancer is NSCLC and the agent is a platinum compound;
(m) the cancer is gastric cancer and the agent is irinotecan;
(n) the cancer is gastric cancer and the agent is fluorouracil and leucovorin;
(o) the cancer is liver cancer and the agent is doxorubicin, ifosfamide and
vincristine;
(p) the cancer is liver cancer and the agent is doxorubicin and vincristine;
(q) the cancer is colorectal carcinoma (CRC) and the agent is fluorouracil;
(r) the cancer is CRC and the agent is capecitabine;
(s) the cancer is CRC and the agent is fluorouracil, leucovorin, and
oxaliplatin
(FOLFOX);
(t) the cancer is CRC and the agent is fluorouracil, leucovorin, and
irinotecan
(FOLFIRI);
(u) the cancer is CRC and the agent is cetuximab;
(v) the cancer is chronic myeloid leukemia (CML) and the agent is imatinib
mesylate;
(w) the cancer is chronic lymphocytic leukemia (CLL) and the agent is imatinib

mesylate;
(x) the cancer is pancreatic cancer and the agent is gemcitabine;


-113-
(y) the cancer is breast cancer and the agent is a taxane;
(z) the cancer is breast cancer and the agent is cyclophosphamide, doxorubicin
and
a taxane;
(aa) the cancer is breast cancer and the agent is selected from the group
consisting
of tamoxifen, anastrazole, letrozole, and fulvestrant;
(bb) the cancer is breast cancer and the agent is trastuzumab;
(cc) the cancer is breast cancer and the agent is bevacizumab;
(dd) the cancer is breast cancer and the agent is cetuximab;
(ee) the cancer is breast cancer and the agent is axitinib;
(ff) the cancer is bladder cancer and the agent is Bacillus Calmette-Guerin
(BCG);
(gg) the cancer is bladder cancer and the agent is gemcitabine and cisplatin;
(hh) the cancer is melanoma and the agent is interferon alpha;
(ii) the cancer is multiple myeloma and the agent is bortezomib;
(jj) the cancer is multiple myeloma and the agent is dexamethasone and
thalidomide;
and
(kk) the cancer is ovarian cancer and the agent is carboplatin and paclitaxel.
2. The method of claim 1, wherein said treatment is selected from the group
consisting of neoadjuvant therapy, adjuvant therapy, first-line therapy,
second-line therapy,
and third-line therapy.
3. The method of claim 1, wherein said agent is administered sequentially or
contemporaneously with said antibody.
4. The method of claim 1 according to subparagraph (i), wherein said taxane is

paclitaxel and wherein said platinum compound is carboplatin.
5. The method of claim 4, wherein said method further comprises administering
at least one agent selected from the group consisting of bevacizumab,
PF03512676, and
sunitinib.
6. The method of claim 1(i), further comprising administering at least one
agent
selected from the group consisting of erlotinib and pemetrexed, wherein said
treatment
comprises second line therapy.
7. The method of claim 1, wherein said therapeutically effective amount of
said
antibody, or portion thereof, ranges from about 1 mg/kg to 40 mg/kg.
8. The method of claim 7, wherein said amount ranges from about 3 mg/kg to
15 mg/kg.
9. The method of claim 1 according to subparagraph (s), wherein said
treatment is selected from the group consisting of first line therapy and
adjuvant therapy
following surgical resection of a primary colon tumor.


-114-
10. The method of claim 1 according to subparagraph (x), wherein said
pancreatic cancer is selected from the group consisting of non-resectable
Stage II, locally
advanced Stage III, and metastatic Stage IV and wherein said treatment
comprises first line
therapy.
11. The method of claim 1 according to subparagraph (x), wherein said antibody

is administered after administration of the last dose of gemcitabine.
12. The method of claim 17, wherein said antibody is administered at least
about
three weeks after and within at least six weeks of administration of the last
dose of
gemcitabine.
13. The method of claim 1 according to subparagraph (y), wherein said
treatment
comprises first line therapy.
14. The method of claim 1 according to subparagraph (kk), wherein said
treatment comprises first line therapy.
15. The method of claim 1 according to subparagraph (I), wherein said NSCLC
responded or remained stable after about six cycles of administration of said
platinum
compound and further wherein said antibody is administered after said platinum-
based
therapy.
16. A method for preventing or treating infection by HIV or for preventing,
treating
or delaying the onset of AIDS in a patient in need thereof, said method
comprising
administering to said patient a therapeutically effective amount of anti-CTLA4
antibody CP-
675,206 and further comprising administering a therapeutically effective
amount of at least
one antiviral agent selected from the group consisting of an HIV protease
inhibitor, a non-
nucleoside reverse transcriptase inhibitor, a nucleoside/nucleotide reverse
transcriptase
inhibitor, a CCR5 antagonist, an inhibitor of gp120 interaction with CD4, an
HIV fusion
inhibitor, a HIV integrase inhibitor, an RNaseH inhibitor, a prenylation
inhibitor, and a
maturation inhibitor.
17. The method of claim 16, wherein said CCR5 antagonist is maraviroc.
18. The method of claim 17, said method further comprising assessing the co-
receptor tropism of said HIV.
19. A method for preventing or treating infection by HIV or for preventing,
treating
or delaying the onset of AIDS in a patient in need thereof, said method
comprising
administering to said patient a therapeutically effective amount of an anti-
CTLA4 antibody and
maraviroc.
20. The method of claim 19, wherein said anti-CTLA4 antibody is selected from
the group consisting of CP-675,206 and ipilimumab.

Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
CTLA4 ANTIBODY COMBINATION THERAPY
Background of the Invention
The present invention relates to uses and compositions comprising an anti-
CTLA4
antibody and at least one therapeutic agent for treatment of cancer. The
invention further
relates to administering, in addition to the antibody-therapeutic agent
inhibitor combination, at
least one additional therapy such as, among others, additional therapeutic
agents, radiation
and stem cell transplant.
Current antitumor agents act by a variety of mechanisms that inhibit cancer
cell
growth and division, ultimately destroying the malignant cell. However,
because these
cytotoxic agents are generally not selective for neoplastic cells, they
destroy normal cells,
disrupt physiologic functions, and are often associated with adverse effects.
An alternative
approach to cancer therapy is to target the immune system ("immunotherapy")
rather than the
tumor itself so that the patient's own immune system attacks tumors while
sparing non-tumor
cells.
One cancer immunotherapy approach targets cytotoxic T lymphocyte-associated
antigen 4 (CTLA4; CD152), which is a cell surface receptor expressed on
activated T cells.
Binding of CTLA4 to its natural ligands, B7.1 (CD80) and B7.2 (CD86), delivers
a negative
regulatory signal to T cells, and blocking this negative signal results in
enhanced T cell
immune function and antitumor activity in animal models (Thompson and Allison
Immunity
7:445-450 (1997); McCoy and LeGros Immunol.& Cell BioL 77:1-10 (1999)).
Several studies
have demonstrated that CTLA4 blockade using antibodies markedly enhances T
cell-
mediated killing of tumors and can induce antitumor immunity (see, e.g., Leach
et al., Science
271:1734-1736 (1996); Kwon et al. Proc. Natl. Acad. Sci. USA 94:8099-8103
(1997); Kwon et
al., Natl. Acad. Sci. USA 96:15074-15079 (1999); Yang et al. Cancer Res
57:4036-41 (1997);
Hurwitz et al. Proc. Natl. Acad. Sci. USA 95:10067-71 (1998)). CTLA4
antibodies and their
uses are described in, e.g., the following applications and patents: U.S.
Patent Application
No. 09/472,087, now issued as U.S. Patent No. 6,682,736; Int. Appi. No.
PCT/US99/30895
(published June 29, 2000, as WO 00/37504); U.S. Pat. Appi. No. 10/612,497
(published
November 18, 2004, as US 2004/0228858); U.S. Pat. Appi. No. 10/776,649
(published
November 18, 2004, as US 2004/0228861); Int. Appl. No. lnt. Appl. No.
PCT/US00/23356
(published March 1, 2001, as WO 01/14424) (e.g., antibody 10D1, also known as
MDX-010,
and ipilimumab, Medarex, Princeton, NJ); Int. Appi. No. PCT/US99/28739
(published June 8,
2000, as WO 00/32231); U.S. Pat. Nos. 5,811,097, 5,855,887, 6,051,227, and
6,207,156; U.S.
Pat. No. 5,844,095, to Linsley et al.; Int. Appi. No. PCT/US92/05202
(published Jan. 7, 1993, as
WO 93/00431); U.S. Patent Appl. No. 10/153,382 (published May 8, 2003, as US
2003/0086930); U.S. Pat. Appl. No. 10/673,738 (published February 24, 2005 as
US


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 2-

2005/0042223); U.S. Pat. Appl. No. 11/085,368 (published October 13, 2005, as
US
2005/0226875); U.S. Pat. Appl. No. 60/624,856 (filed Nov. 4, 2004), now
published as
W02006/048749, May 11, 2006; U.S. Pat. Appl. No. 60/664,364 (filed March 23,
2005) and
U.S. Pat. Appl. No. 60/711,707 (filed Aug. 26, 2005) now published as WO
2006/101691 Sept.
28, 2006; U.S. Pat. Appl. No. 60/664,653 (filed March 23, 2005) now published
as WO
2006/101692, Sept. 28, 2006; U.S. Pat. Appl. No. 60/697,082 (filed July 7,
2005) now WO
2007/008463 published Jan. 18, 2007.
Despite the successes of currently available anti-cancer treatments, complete
responses to these treatments or prolonged survival are infrequently observed,
and the
patient population refractory to these treatments is still large. Thus, there
is an unmet need
for the development of new therapeutic regimens, particularly those capable of
augmenting or
potentiating the anti-tumor activity of other anti-neoplastic agents while
reducing the cytotoxic
side effects of current chemotherapeutics, and the present invention meets
this need.
Summary of the Invention
The present invention includes a method for the treatment of cancer in a
patient in
need of such treatment. The method comprises administering to the patient a
therapeutically
effective amount of an anti-CTLA4 antibody, e.g., ipilimumab (also referred to
as MDX-010)
and CP-675,206 (also referred to as 11.2.1. and ticilimumab), in combination
with a
therapeutically effective amount of at least one therapeutic agent, wherein
the cancer and the
agent are selected from the group consisting of:
(a) the cancer is non-Hodgkin's lymphoma (NHL) and wherein the agent is
rituximab;
(b) the cancer is NHL and the agent is cyclophosphamide, doxorubicin,
vincristine,
and prednisone (CHOP);
(c) the cancer is NHL and the agent is cyclophosphamide, doxorubicin,
vincristine,
prednisone and rituximab (CHOP-R);
(d) the cancer is lung cancer and the agent is bevacizumab;
(e) the cancer is non-small cell lung cancer (NSCLC) and the agent is
gefitinib;
(f) the cancer is NSCLC and the agent is bevacizumab;
(g) the cancer is NSCLC and the agent is a taxane and gemcitabine, and further
wherein the taxane is selected from the group consisting of docetaxel and
paclitaxel;
(h) the cancer is NSCLC and the agent is a taxane and a platinum compound;
(i) the cancer is NSCLC and the agent is docetaxel;
(j) the cancer is NSCLC and the agent is eriotinib;
(k) the cancer is NSCLC and the agent is pemetrexed;
(I) the cancer is NSCLC and the agent is a platinum compound;
(m) the cancer is gastric cancer and the agent is irinotecan;


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 3-

(n) the cancer is gastric cancer and the agent is fluorouracil and leucovorin;
(o) the cancer is liver cancer and the agent is doxorubicin, ifosfamide and
vincristine;
(p) the cancer is liver cancer and the agent is doxorubicin and vincristine;
(q) the cancer is colorectal carcinoma (CRC) and the agent is fluorouracil;
(r) the cancer is CRC and the agent is capecitabine;
(s) the cancer is CRC and the agent is fluorouracil, leucovorin, and
oxaliplatin
(FOLFOX);
(t) the cancer is CRC and the agent is fluorouracil, leucovorin, and
irinotecan
(FOLFIRI);
(u) the cancer is CRC and the agent is cetuximab;
(v) the cancer is chronic myeloid leukemia (CML) and the agent is imatinib
mesylate;
(w) the cancer is chronic lymphocytic leukemia (CLL) and the agent is imatinib
mesylate;
(x) the cancer is pancreatic cancer and the agent is gemcitabine;
(y) the cancer is breast cancer and the agent is a taxane;
(z) the cancer is breast cancer and the agent is cyclophosphamide, doxorubicin
and
a taxane;
(aa) the cancer is breast cancer and the agent is selected from the group
consisting
of tamoxifen, anastrazole, letrozole, and fulvestrant;
(bb) the cancer is breast cancer and the agent is trastuzumab;
(cc) the cancer is breast cancer and the agent is bevacizumab;
(dd) the cancer is breast cancer and the agent is cetuximab;
(ee) the cancer is breast cancer and the agent is axitinib;
(ff) the cancer is bladder cancer and the agent is Bacillus Calmette-Guerin
(BCG);
(gg) the cancer is bladder cancer and the agent is gemcitabine and cisplatin;
(hh) the cancer is melanoma and the agent is interferon alpha;
(ii) the cancer is multiple myeloma and the agent is bortezomib;
(jj) the cancer is multiple myeloma and the agent is dexamethasone and
thalidomide;
and
(kk) the cancer is ovarian cancer and the agent is carboplatin and paclitaxel.

In one aspect, the treatment is selected from the group consisting of
neoadjuvant
therapy, adjuvant therapy, first-line therapy, second-line therapy, and third-
line therapy.
In another aspect, the agent is administered sequentially or contemporaneously
with
the antibody.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-4-
In yet another aspect, the taxane is paclitaxel and the platinum compound is
carboplatin.
In one aspect, the method further comprises administering at least one agent
selected from the group consisting of bevacizumab, PF03512676, and sunitinib.
In another aspect, the method further comprises administering at least one
agent
selected from the group consisting of erlotinib and pemetrexed, wherein the
treatment
comprises second line therapy.
In one aspect, the therapeutically effective amount of the antibody ranges
from about
1 mg/kg to 40 mg/kg.
In another aspect, the therapeutically effective amount of the antibody ranges
from
about 3 mg/kg to 15 mg/kg.
In one aspect, the NHL is indolent NHL and the treatment comprises first line
therapy.
In another aspect, the NHL is aggressive NHL and the treatment comprises
second
line therapy.
In a further aspect, the treatment comprises first line therapy.
In one aspect, the cancer is CRC and the agent is capecitabine and the
treatment
comprises first line therapy.
In another aspect, the cancer is CRC and the agent is FOLFOX and the treatment
is
selected from the group consisting of first line therapy and adjuvant therapy
following surgical
resection of a primary colon tumor.
In one aspect, the cancer is CML and the agent is imatinib mesylate and the
treatment comprises first line therapy.
In another aspect, the cancer is CLL and the agent is imatinib mesylate and
the
treatment comprises first line therapy.
In a further aspect, the cancer is pancreatic cancer and the agent is
gemcitabine and
the pancreatic cancer is selected from the group consisting of non-resectable
Stage II, locally
advanced Stage III, and metastatic Stage IV and wherein the treatment
comprises first line
therapy.
In another aspect, the cancer is breast cancer and the agent is a taxane and
the
treatment comprises first line therapy.
In one aspect, the cancer is ovarian cancer and the agent is carboplatin and
paclitaxel and the treatment comprises first line therapy.
The invention includes a kit for the treatment of NSCLC comprising a
therapeutically
effective amount of CP-675,206; a therapeutically effective amount of a
carboplatin; a
therapeutically effective amount of paclitaxel; an applicator; and an
instructional material for
the use of the kit.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 5-

In one aspect, the kit further comprises a therapeutically effective amount of
at least
one agent selected from the group consisting of bevacizumab, sunitinib, and
PF03512676.
The invention includes a kit for the treatment of NSCLC comprising a
therapeutically
effective amount of CP-675,206; a therapeutically effective amount of an agent
selected from
the group consisting of docetaxel, erlotinib and pemetrexed; an applicator;
and an
instructional material for the use of the kit.
The invention includes a kit for the treatment of CRC comprising a
therapeutically
effective amount of CP-675,206; a therapeutically effective amount of
carboplatin; a
therapeutically effective amount of paclitaxel; an applicator; and an
instructional material for
the use of the kit.
The invention includes a kit for the treatment of CRC comprising a
therapeutically
effective amount of CP-675,206; a therapeutically effective amount of a
fluorouracil; a
therapeutically effective amount of leucovorin; a therapeutically effective
amount of
oxaliplatin; an applicator; and an instructional material for the use of the
kit.
The invention includes a kit for the treatment of pancreatic cancer comprising
a
therapeutically effective amount of CP-675,206; a therapeutically effective
amount of
gemcitabine; an applicator; and an instructional material for the use of the
kit.
The invention includes a kit for the treatment of ovarian cancer comprising a
therapeutically effective amount of CP-675,206; a therapeutically effective
amount of
carboplatin; a therapeutically effective amount of paclitaxel; an applicator;
and an instructional
material for the use of the kit.
The invention includes a method for preventing or treating infection by HIV or
for
preventing, treating or delaying the onset of AIDS in a patient in need
thereof. The method
comprises administering to the patient a therapeutically effective amount of
anti-CTLA4
antibody CP-675,206 and further comprises administering a therapeutically
effective amount
of at least one antiviral agent selected from the group consisting of an HIV
protease inhibitor,
a non-nucleoside reverse transcriptase inhibitor, a nucleoside/nucleotide
reverse
transcriptase inhibitor, a CCR5 antagonist, an inhibitor of gp120 interaction
with CD4, an HIV
fusion inhibitor, a HIV integrase inhibitor, an RNaseH inhibitor, a
prenylation inhibitor, and a
maturation inhibitor.
In one aspect, the CCR5 antagonist is maraviroc.
In another aspect, the method further comprises assessing the co-receptor
tropism of
the HIV.
The invention includes a method for preventing or treating infection by HIV or
for
preventing, treating or delaying the onset of AIDS in a patient in need
thereof, where the


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-6-
method comprises administering to the patient a therapeutically effective
amount of an anti-
CTLA4 antibody and maraviroc.
In one aspect, the anti-CTLA4 antibody is selected from the group consisting
of CP-
675,206 and ipilimumab.
Alternate embodiments of the invention are described below, such as those
employing alternate anti-CTLA4 antibodies and involving different methods.

Brief Description of the Drawings
The foregoing summary, as well as the following detailed description of the
invention,
will be better understood when read in conjunction with the appended drawings.
For the
purpose of illustrating the invention there are shown in the drawings
embodiment(s) which are
presently preferred. It should be understood, however, that the invention is
not limited to the
precise arrangements and instrumentalities shown.
In the drawings:
Figure 1, comprising Figures 1A-1 D, shows the nucleotide and amino acid
sequences
of anti-CTLA4 antibody 4.1.1. Figure 1A shows the full length nucleotide
sequence for the
4.1.1 heavy chain (SEQ ID NO:1). Figure 1B shows the full length amino acid
sequence for
the 4.1.1 heavy chain (SEQ ID NO:2), and the amino acid sequence for the 4.1.1
heavy chain
variable region (SEQ ID NO:3) designated between brackets "[ ]". The amino
acid sequence
of each 4.1.1 heavy chain CDR is underlined. The CDR sequences are as follows:
CDRI:
GFTFSSHGMH (SEQ ID NO:4); CDR2: VIWYDGRNKYYADSV (SEQ ID NO:5); and CDR3:.
GGHFGPFDY (SEQ ID NO:6). Figure 1 C shows the nucleotide sequence for the
4.1.1 light
chain (SEQ ID NO:7). Figure 1 D shows the amino acid sequence of the full
length 4.1.1 light
chain (SEQ ID NO:8), and the variable region as indicated between brackets "[
]" (SEQ ID
NO:9). The amino acid sequence of each CDR is indicated as follows: CDRI:
RASQSISSSFLA (SEQ ID NO:10); CDR2: GASSRAT (SEQ ID NO:11); and CDR3:
QQYGTSPWT (SEQ ID NO:12).
Figure 2, comprising Figures 2A-2D, shows the nucleotide and amino acid
sequences
of anti-CTLA4 antibody 4.13.1. Figure 2A shows the full length nucleotide
sequence for the
4.13.1 heavy chain (SEQ ID NO:13). Figure 2B shows the full length amino acid
sequence
for the 4.13.1 heavy chain (SEQ ID NO:14), and the amino acid sequence for the
4.13.1
heavy chain variable region (SEQ ID NO:15) designated between brackets "[ ]".
The amino
acid sequence of each 4.13.1 heavy chain CDR is underlined. The CDR sequences
are as
follows: CDRI: GFTFSSHGIH (SEQ ID NO:16); CDR2: VIWYDGRNKDYADSV (SEQ ID
NO:12); and CDR3: VAPLGPLDY (SEQ ID NO:18). Figure 2C shows the nucleotide


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 7-

sequence for the 4.13.1 light chain (SEQ ID NO:19). Figure 2D shows the amino
acid
sequence of the full length 4.13.1 light chain (SEQ ID NO:20), and the
variable region as
indicated between brackets "[ ]" (SEQ ID NO:21). The amino acid sequence of
each CDR is
indicated as follows: CDR1: RASQSVSSYLA (SEQ ID NO:22); CDR2: GASSRAT (SEQ ID
NO:23); and CDR3: QQYGRSPFT (SEQ ID NO:24).
Figure 3, comprising Figures 3A-3D, shows the nucleotide and amino acid
sequences
of anti-CTLA4 antibody CP-675,206. Figure 3A shows the full length nucleotide
sequence for
the CP-675,206 heavy chain (SEQ ID NO:25). Figure 3B shows the full length
amino acid
sequence for the CP-675,206 heavy chain (SEQ ID NO:26), and the amino acid
sequence for
the CP-675,206 heavy chain variable region (SEQ ID NO:27) designated between
brackets "[
]". The amino acid sequence of each CP-675,206 heavy chain CDR is underlined.
The CDR
sequences are as follows: CDR1: GFTFSSYGMH (SEQ ID NO:28); CDR2:
VIWYDGSNKYYADSV (SEQ ID NO:29); and CDR3: DPRGATLYYYYYGMDV (SEQ ID
NO:30). Figure 3C shows the nucleotide sequence for the CP-675,206 light chain
(SEQ ID
NO:31). Figure 3D shows the amino acid sequence of the full length CP-675,206
light chain
(SEQ ID NO:32), and the variable region as indicated between brackets "[ ]"
(SEQ ID NO:33).
The amino acid sequence of each CDR is indicated as follows: CDRI: RASQSINSYLD
{SEQ
ID NO:34); CDR2: AASSLQS (SEQ ID NO:35); and CDR3: QQYYSTPFT (SEQ ID NO:36).
Figure 4 is a graph depicting the plasma serum levels in Rhesus monkeys
following
exposure to an anti-CTLA4 antibody. All Rhesus monkeys were administered
influenza
vaccine FLUZONE intramuscularly (IM) on week 0 and again on week 4. In
addition to
FLUZONE, animals in Group 1(animal number 1(closed circle), 2 (closed square),
and 3
(closed triangle)) received a single dose of anti-CTL4 antibody 4.1.1 (also
referred to as CP-
642,570) at 5 mg/kg intravenously (IV) on week 0. Animals in Group 3 (animal
number 4
(open circle), 5 (open square), 6 (open triangle), and 7 (marked by "x"))
received a single
dose of an irrelevant human antibody (anti-KLH antibody) at 5 mg/kg IV on week
0.
Figure 5 is a graph depicting the serum level of a neopterin, a marker which
is
suggestive of immune activity, in Rhesus monkeys immunized with FLUZONE IM on
week 0
and week 4 and administered anti-CTLA4 antibody 4.1.1 at 5 mg/kg IV on week 0.
Animal
number AG40 (closed diamond), number AK80 (closed square) and number AM90
(closed
triangle) were in group I and received anti-CTLA4 antibody and FLUZONE on day
0 followed
by FLUZONE on week 0. Animal number AK76 (x), number AM84 (*), and number BC13
(open circle) were in control group 2 and were administered FLUZONE on week 0
and week 4
and also administered an irrelevant human antibody (anti-KLH antibody) at 5
mg/ml IV on
week 0 of the protocol.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 8-

Figure 6 is a graph depicting the level of a 2-5 adenylate synthetase in white
blood
cell pellets isolated from the blood of Rhesus monkeys immunized with FLUZONE
IM on
weeks 0 and 4 and further administered anti-CTLA4 antibody 4.1.1 (5mg/kg IV)
on week 0.
Peripheral blood cells were obtained from each animal in each group 1(FLUZONE
IM weeks
0 and 4 and anti-CTLA4 antibody week 0) and group 2 (FLUZONE IM on weeks 0 and
4 and
an irrelevant antibody (anti-KLH 5 mg/kg IV) administered on week 0) at week 2
(light gray
shading), week 4 (darker gray shading) and week 6 (unshaded bars). Where the
values
exceeded 4000 pmol/dI, the values are shown in each bar. The cells were
pelleted and the
level of 2-5 adenylate synthetase in the pellets obtained from each animal was
assessed.
Figure 7 is a graph depicting the IgG anti-FLUZONE titer in Rhesus monkeys
administered anti-CTLA4 antibody. Serum anti-FLUZONE IgG titers were assessed
for
animals in Group 1(open triangle; administered FLUZONE on weeks 0 and 4 and
administered anti-CTLA4 antibody on week 0), Group 2 (open square;
administered
FLUZONE on weeks 0 and 4 and administered an irrelevant human anti-KLH
antibody on
week 0), Group 3 (closed circle; administered FLUZONE on weeks 0 and 4 and
administered
anti-CTLA4 antibody on week 4), Group 4 (open circle; administered FLUZONE on
weeks 0
and 4 and administered an irrelevant human anti-KLH antibody on week 4) and
Group 5
(closed triangle; administered FLUZONE on weeks 0 and 4 but no antibody was
administered). FLUZONE was administered IM and antibodies were administered IV
at 5
mg/kg. Anti-FLUZONE IgG titers were assessed one week before immunization with
FLUZONE (week -1, also referred to as "prebleed") and then at week 0, week 2,
week 4,
week 6 and week 8. Animals in Group 3 (immunized with FLUZONE on weeks 0 and 4
and
administered anti-CTLA4 antibody on week 4) demonstrated increased anti-
FLUZONE IgG
serum titers compared with animals in the other groups.
Figure 8 is a graph depicting the anti-FLUZONE IgG serum titer in Rhesus
monkeys
immunized with FLUZONE and on weeks 0 and 4 and administered anti-CLTA4
antibody on
week 0 (Group 1), immunized with FLUZONE on weeks 0 and 4 and administered an
irrelevant antibody on week 0 (Group 2), immunized with FLUZONE on weeks 0 and
4 and
administered anti-CTLA4 antibody on week 4 (Group 3), immunized with FLUZONE
on weeks
0 and 4 and administered an irrelevant antibody on week 0 (Group 4), and
immunized with
FLUZONE on weeks 0 and 4 (Group 5). The data points are separated by animal
group and
the titers for each individual animal within each group for the following data
points: prebleed
(week -1; +), week 0 (open triangle), week 2 (closed triangle), week 4 (open
squares), week 6
(closed circle) and week 8 (open circle).
Figure 9 is a bar graph depicting the anti-FLUZONE IgG antibody titers at week
6 in
Rhesus monkeys immunized with FLUZONE on weeks 0 and 4 (Groups 1-5). Group 1


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 9-

animals received anti-CTLA4 (5 mg/kg IV) on week 0. Group 2 animals received
irrelevant
antibody anti-KLH (5 mg/kg IV) on week 0. Group 3 animals received anti-CTLA4
(5 mg/kg IV)
on week 4. Group 4 animals received irrelevant antibody anti-KLH (5 mg/kg IV)
on week 4.
Group 5 animals received only FLUZONE immunizations on weeks 0 and 4.
Detailed Description Of The Invention
Unless otherwise defined herein, scientific and technical terms used in
connection
with the present invention shall have the meanings that are commonly
understood by those of
ordinary skill in the art. Further, unless otherwise required by context,
singular terms shall
include pluralities and plural terms shall include the singular. Generally,
nomenclatures used
in connection with, and techniques of, cell and tissue culture, molecular
biology, immunology,
microbiology, genetics and protein and nucleic acid chemistry and
hybridization described
herein are those well known and commonly used in the art.
The methods and techniques of the present invention are generally performed
according to methods well known in the art and as described in various general
and more
specific references that are cited and discussed throughout the present
specification unless
otherwise indicated. Such references include, e.g., Sambrook and Russell,
Molecular
Cloning, A Laboratory Approach, Cold Spring Harbor Press, Cold Spring Harbor,
NY (2001),
Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, NY
(2002), and
Harlow and Lane Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory
Press,
Cold Spring Harbor, NY (1990), which are incorporated herein by reference.
Enzymatic
reactions and purification techniques are performed according to
manufacturer's
specifications, as commonly accomplished in the art or as described herein.
The
nomenclatures used in connection with, and the laboratory procedures and
techniques of,
analytical chemistry, synthetic organic chemistry, and medicinal and
pharmaceutical
chemistry described herein are those well known and commonly used in the art.
Standard
techniques are used for chemical syntheses, chemical analyses, pharmaceutical
preparation,
formulation, and delivery, and treatment of patients.
As used herein, each of the following terms has the meaning associated with it
in this
section.
The articles "a" and "an" are used herein to refer to one or to more than one
(Le., to at
least one) of the grammatical object of the article. By way of example, "an
element" means
one element or more than one element.
As used herein, the twenty conventional amino acids and their abbreviations
follow
conventional usage. See Immunology--A Synthesis (2nd Edition, E. S. Golub and
D. R. Gren,


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 10-

Eds., Sinauer Associates, Sunderland, Mass. (1991)), which is incorporated
herein by
reference.
A "conservative amino acid substitution" is one in which an amino acid residue
is
substituted by another amino acid residue having a side chain R group with
similar chemical
properties (e.g., charge or hydrophobicity). In general, a conservative amino
acid substitution
will not substantially change the functional properties of a protein. In cases
where two or more
amino acid sequences differ from each other by conservative substitutions, the
percent
sequence identity or degree of similarity may be adjusted upwards to correct
for the
conservative nature of the substitution. Means for making this adjustment are
well-known to
those of skill in the art. See, e.g., Pearson, Methods Mol. Biol. 243:307-31
(1994).
Examples of groups of amino acids that have side chains with similar chemical
properties include 1) aliphatic side chains: glycine, alanine, valine,
leucine, and isoleucine; 2)
aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side
chains:
asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine,
and tryptophan;
5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains:
aspartic acid and
glutamic acid; and 7) sulfur-containing side chains: cysteine and methionine.
Preferred
conservative amino acids substitution groups are: valine-leucine-isoleucine,
phenylalanine-
tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-
glutamine.
Alternatively, a conservative replacement is any change having a positive
value in the
PAM250 log-likelihood matrix disclosed in Gonnet et al., Science 256:1443-45
(1992), herein
incorporated by reference. A "moderately conservative" replacement is any
change having a
nonnegative value in the PAM250 log-likelihood matrix.
Preferred amino acid substitutions are those which: (1) reduce susceptibility
to
proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding
affinity for forming protein
complexes, and (4) confer or modify other physicochemical or functional
properties of such
analogs. Analogs comprising substitutions, deletions, and/or insertions can
include various
muteins of a sequence other than the specified peptide sequence. For example,
single or
multiple amino acid substitutions (preferably conservative amino acid
substitutions) may be
made in the specified sequence (preferably in the portion of the polypeptide
outside the
domain(s) forming intermolecular contacts, e.g., outside of the CDRs). A
conservative amino
acid substitution should not substantially change the structural
characteristics of the parent
sequence (e.g., a replacement amino acid should not tend to break a helix that
occurs in the
parent sequence, or disrupt other types of secondary structure that
characterizes the parent
sequence). Examples of art-recognized polypeptide secondary and tertiary
structures are
described in Proteins, Structures and Molecular Principles (Creighton, Ed., W.
H. Freeman
and Company, New York (1984)); Introduction to Protein Structure (C. Branden
and J. Tooze,


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 11-

eds., Garland Publishing, New York, N.Y. (1991)); and Thornton et al., Nature
354:105
(1991), which are each incorporated herein by reference.
Sequence similarity for polypeptides is typically measured using sequence
analysis
software. Protein analysis software matches similar sequences using measures
of similarity
assigned to various substitutions, deletions and other modifications,
including conservative
amino acid substitutions. For instance, Genetics Computer Group (GCG available
from
Genetics Computer Group, Inc.), also referred to as the Wisconsin Package, is
an integrated
software package of over 130 programs for accessing, analyzing and
manipulating nucleotide
and protein sequences. GCG contains programs such as "Gap" and "Bestfit" which
can be
used with default parameters to determine sequence similarity, homology and/or
sequence
identity between closely related polypeptides, such as homologous polypeptides
from
different species of organisms or between a wild type protein and a mutein
thereof. See, e.g.,
GCG version 6.1, version 7.0, version 9.1, and version 10Ø
Polypeptide sequences also can be compared using FASTA, a program in GCG,
using default or recommended parameters. FASTA (e.g., FASTA2 and FASTA3)
provides
alignments and percent sequence identity of the regions of the best overlap
between the
query and search sequences (Pearson, Methods EnzymoL 183:63-98 (1990);
Pearson,
Methods Mol. Biol. 132:185-219 (2000)). Another preferred algorithm when
comparing a
sequence of the invention to a database containing a large number of sequences
from
different organisms is the computer program BLAST, especially blastp or
tblastn, using
default parameters. See, e.g., Altschul et al., J. MoL Biol. 215:403-410
(1990); Altschul et al.,
Nucleic Acids Res. 25:3389-402 (1997); herein incorporated by reference.
An intact "antibody" comprises at least two heavy (H) chains and two light (L)
chains
inter-connected by disulfide bonds. See generally, Fundamental Immunology, Ch.
7 (Paul,
W., ed., 2nd ed. Raven Press, N.Y. (1989)) (incorporated by reference in its
entirety for all
purposes). Each heavy chain is comprised of a heavy chain variable region
(HCVR or VH) and
a heavy chain constant region (CH). The heavy chain constant region is
comprised of three
domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain
variable region
(LCVR or VL) and a light chain constant region. The light chain constant
region is comprised
of one domain, CL. The VH and VL regions can be further subdivided into
regions of
hypervariability, termed complementarity determining regions (CDR),
interspersed with
regions that are more conserved, termed framework regions (FR). Each VH and VL
is
composed of three CDRs and four FRs, arranged from amino-terminus to carboxyl-
terminus
in the following order: FRI, CDR1, FR2, CDR2, FR3, CDR3, FR4. The assignment
of amino
acids to each domain is in accordance with the definitions of Kabat, Sequences
of Proteins of


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 12-

Immunological Interest (National Institutes of Health, Bethesda, MD (1987 and
1991)), or
Chothia & Lesk, J. MoL Biol. 196:901-917 (1987); Chothia et al., Nature
342:878-883 (1989).
The term "antigen-binding portion" of an antibody (or simply "antibody
portion"), as
used herein, refers to one or more fragments of an antibody that retain the
ability to
specifically bind to an antigen (e.g., CTLA4). It has been shown that the
antigen-binding
function of an antibody can be performed by fragments of a full-length
antibody. Examples of
binding fragments encompassed within the term "antigen-binding portion" of an
antibody
include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL
and CH1
domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab
fragments linked by a
disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH
and CH1 domains;
(iv) a Fv fragment consisting of the VL and VH domains of a single arm of an
antibody, (v) a
dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH
domain; and
(vi) an isolated complementarity determining region (CDR). Furthermore,
although the two
domains of the Fv fragment, VL and VH, are coded for by separate genes, they
can be joined,
using recombinant methods, by a synthetic linker that enables them to be made
as a single
protein chain in which the VL and VH regions pair to form monovalent molecules
(known as
single chain Fv (scFv)); see e.g., Bird et al. Science 242:423-426 (1988) and
Huston et al.
Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988)). Such single chain antibodies
are also
intended to be encompassed within the term "antigen-binding portion" of an
antibody. Other
forms of single chain antibodies, such as diabodies are also encompassed.
Diabodies are
bivalent, bispecific antibodies in which VH and VL domains are expressed on a
single
polypeptide chain, but using a linker that is too short to allow for pairing
between the two
domains on the same chain, thereby forcing the domains to pair with
complementary domains
of another chain and creating two antigen binding sites (see e.g., Holliger et
al. Proc. Natl.
Acad. Scf. USA 90:6444-6448 (1993); Poljak et al. Structure 2:1121-1123
(1994)).
Still further, an antibody or antigen-binding portion thereof may be part of
larger
immunoadhesion molecules, formed by covalent or noncovalent association of the
antibody or
antibody portion with one or more other proteins or peptides. Examples of such
immunoadhesion molecules include use of the streptavidin core region to make a
tetrameric
scFv molecule (Kipriyanov et al. Human Antibodies and Hybridomas 6:93-101
(1995)) and
use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag
to make
bivalent and biotinylated scFv molecules (Kipriyanov et al. Mol. Immunol.
31:1047-1058
(1994)). Other examples include where one or more CDRs from an antibody are
incorporated
into a molecule either covalently or noncovalently to make it an immunoadhesin
that
specifically binds to an antigen of interest, such as CTLA4. In such
embodiments, the CDR(s)
may be incorporated as part of a larger polypeptide chain, may be covalently
linked to another


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-13-
polypeptide chain, or may be incorporated noncovalently. Antibody portions,
such as Fab and
F(ab')2 fragments, can be prepared from whole antibodies using conventional
techniques,
such as papain or pepsin digestion, respectively, of whole antibodies.
Moreover, antibodies,
antibody portions and immunoadhesion molecules can be obtained using standard
recombinant DNA techniques, as described herein.
Where an "antibody" is referred to herein with respect to the present
invention, it
should be understood that an antigen-binding portion thereof may also be used.
An antigen-
binding portion competes with the intact antibody for specific binding. See
generally,
Fundamental Immunology, Ch. 7 (Paul, W., ed., 2nd ed., Raven Press, N.Y.
(1989))
(incorporated by reference in its entirety for all purposes). Antigen-binding
portions may be
produced by recombinant DNA techniques or by enzymatic or chemical cleavage of
intact
antibodies. In some embodiments, antigen-binding portions include Fab, Fab',
F(ab')2, Fd, Fv,
dAb, and complementarity determining region (CDR) fragments, single-chain
antibodies
(scFv), chimeric antibodies, diabodies and polypeptides that contain at least
a portion of an
antibody that is sufficient to confer specific antigen binding to the
polypeptide. In
embodiments having one or more binding sites, the binding sites may be
identical to one
another or may be different.
The terms "human antibody" or "human sequence antibody", as used
interchangeably
herein, include antibodies having variable and constant regions (if present)
derived from
human germline immunoglobulin sequences. The human sequence antibodies of the
invention may include amino acid residues not encoded by human germline
immunoglobulin
sequences (e.g., mutations introduced by random or site-specific mutagenesis
in vitro or by
somatic mutation in vivo). However, the term "human antibody", as used herein,
is not
intended to include "chimeric" antibodies in which CDR sequences derived from
the germline
of another mammalian species, such as a mouse, have been grafted onto human
framework
sequences (i.e., "humanized" or PRIMATIZEDT"' antibodies).
The term "chimeric antibody" as used herein means an antibody that comprises
regions from two or more different antibodies. In one embodiment, one or more
of the CDRs
are derived from a human anti-CTLA4 antibody. In another embodiment, all of
the CDRs are
derived from a human anti-CTLA4 antibody. In another embodiment, the CDRs from
more
than one human anti-CTLA4 antibodies are combined in a chimeric human
antibody. For
instance, a chimeric antibody may comprise a CDRI from the light chain of a
first human anti-
CD40 antibody, a CDR2 from the light chain of a second human anti-CTLA4
antibody and a
CDR3 and CDR3 from the light chain of a third human anti-CTLA4 antibody, and
the CDRs
from the heavy chain may be derived from one or more other anti-CD40
antibodies. Further,


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-14-
the framework regions may be derived from one of the same anti-CTLA4
antibodies or from
one or more different human(s).
Moreover, as discussed previously herein, chimeric antibody includes an
antibody
comprising a portion derived from the germline sequences of more than one
species.
"Glycoform" refers to a complex oligosaccharide structure comprising linkages
of
various carbohydrate units. Such structures are described in, e.g., Essentials
of Glycobiology
Varki et al., eds., Cold Spring Harbor Laboratory Press, Cold Spring Harbor,
NY (1999),
which also provides a review of standard glycobiology nomenclature. Such
glycoforms
include, but are not limited to, G2, GI, GO, G-1, and G-2 (see, e.g.,
International Patent
Publication No. WO 99/22764).
"Glycosylation pattern" is defined as the pattern of carbohydrate units that
are
covalently attached to a protein (e.g., the glycoform) as well as to the
site(s) to which the
glycoform(s) are covalently attached to the peptide backbone of a protein,
more specifically to
an immunoglobulin protein.
It is likely that antibodies expressed by different cell lines or in
transgenic animals will
have different glycoforms and/or glycosylation patterns compared with each
other. However,
all antibodies encoded by the nucleic acid molecules provided herein, or
comprising the
amino acid sequences provided herein are part of the instant invention,
regardless of the
glycosylation of the antibodies.
By the term "effective amount", or "therapeutically effective amount," as used
herein,
is meant an amount that when administered to a mammal, preferably a human,
mediates a
detectable therapeutic response compared to the response detected in the
absence of the
compound. A therapeutic response, such as, but not limited to, inhibition of
and/or decreased
tumor growth, tumor size, metastasis, and the like, can be readily assessed by
a plethora of
art-recognized methods, including, e.g., such methods as disclosed herein.
The skilled artisan would understand that the effective amount of the compound
or
composition administered herein varies and can be readily determined based on
a number of
factors such as the disease or condition being treated, the stage of the
disease, the age and
health and physical condition of the mammal being treated, the severity of the
disease, the
particular compound being administered, and the like.
By the term "compete", as used herein with regard to an antibody, is meant
that a first
antibody, or an antigen-binding portion thereof, competes for binding with a
second antibody,
or an antigen-binding portion thereof, where binding of the first antibody
with its cognate
epitope is detectably decreased in the presence of the second antibody
compared to the
binding of the first antibody in the absence of the second antibody. The
alternative, where the
binding of the second antibody to its epitope is also detectably decreased in
the presence of


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 15-

the first antibody, can, but need not be the case. That is, a first antibody
can inhibit the
binding of a second antibody to its epitope without that second antibody
inhibiting the binding
of the first antibody to its respective epitope. However, where each antibody
detectably
inhibits the binding of the other antibody with its cognate epitope or ligand,
whether to the
same, greater, or lesser extent, the antibodies are said to "cross-compete"
with each other for
binding of their respective epitope(s). For instance, cross-competing
antibodies can bind to
the epitope, or potion of the epitope, to which the antibodies used in the
invention bind. Use
of both competing and cross-competing antibodies is encompassed by the present
invention.
Regardless of the mechanism by which such competition or cross-competition
occurs (e.g.,
steric hindrance, conformational change, or binding to a common epitope, or
portion thereof,
and the like), the skilled artisan would appreciate, based upon the teachings
provided herein,
that such competing and/or cross-competing antibodies are encompassed and can
be useful
for the methods disclosed herein.
The term "epitope" includes any protein determinant capable of specific
binding to an
immunoglobulin or T-cell receptor. Epitopic determinants usually consist of
chemically active
surface groupings of molecules such as amino acids or sugar side chains and
usually have
specific three dimensional structural characteristics, as well as specific
charge characteristics.
Conformational and nonconformational epitopes are distinguished in that the
binding to the
former but not the latter is lost in the presence of denaturing solvents.
"Instructional material," as that term is used herein, includes a publication,
a
recording, a diagram, or any other medium of expression which can be used to
communicate
the usefulness of the compound, combination, and/or composition 6f the
invention in the kit
for affecting, alleviating or treating the various diseases or disorders
recited herein.
Optionally, or alternately, the instructional material can describe one or
more methods of
alleviating the diseases or disorders in a cell, a tissue, or a mammal,
including as disclosed
elsewhere herein.
The instructional material of the kit may, for example, be affixed to a
container that
contains the compound and/or composition of the invention or be shipped
together with a
container which contains the compound and/or composition. Alternatively, the
instructional
material may be shipped separately from the container with the intention that
the recipient
uses the instructional material and the compound cooperatively.
Except when noted, the terms "patient" or "subject" are used interchangeably
and
refer to mammals such as human patients and non-human primates, as well as
veterinary
subjects such as rabbits, rats, and mice, and other animals. Preferably,
patient refers to a
human.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-16-
Conventional notation is used herein to portray polypeptide sequences: the
left-hand
end of a polypeptide sequence is the amino-terminus; the right-hand end of a
polypeptide
sequence is the carboxyl-terminus.
By the phrase "specifically binds," as used herein, is meant a compound, e.g.,
a
protein, a nucleic acid, an antibody, and the like, which recognizes and binds
a specific
molecule, but does not substantially recognize or bind other molecules in a
sample. For
instance, an antibody or a peptide inhibitor which recognizes and binds a
cognate ligand (e.g.,
an anti-CTLA4 antibody that binds with its cognate antigen, CTLA4) in a
sample, but does not
substantially recognize or bind other molecules in the sample. Thus, under
designated assay
conditions, the specified binding moiety (e.g., an antibody or an antigen-
binding portion
thereof) binds preferentially to a particular target molecule and does not
bind in a significant
amount to other components present in a test sample. A variety of assay
formats may be
used to select an antibody that specifically binds a molecule of interest. For
example, solid-
phase ELISA immunoassay, immunoprecipitation, BlAcore, FACS, and Western blot
analysis
are among many assays that may be used to identify an antibody that
specifically reacts with
CTLA4. Typically, a specific or selective reaction will be at least twice
background signal or
noise and more typically more than 10 times background, even more
specifically, an antibody
is said to "specifically bind" an antigen when the equilibrium dissociation
constant (KD) is <- 1
pM, preferably s 100 nM and most preferably <_ 10 nM.
The term "KD" refers to the equilibrium dissociation constant of a particular
antibody-
antigen interaction.
As used herein, "substantially pure" means an object species is the
predominant
species present (Le., on a molar basis it is more abundant than any other
individual species in
the composition), and preferably a substantially purified fraction is a
composition wherein the
object species (e.g., an anti-CTLA4 antibody) comprises at least about 50
percent (on a molar
basis) of all macromolecular species present. Generally, a substantially pure
composition will
comprise more than about 80 percent of all macromolecular species present in
the
composition, more preferably more than about 85%, 90%, 95%, and 99%. Most
preferably,
the object species is purified to essential homogeneity (contaminant species
cannot be
detected in the composition by conventional detection methods) wherein the
composition
consists essentially of a single macromolecular species.
As used herein, to "treat" means reducing the frequency with which symptoms of
a
disease (Le., tumor growth and/or metastasis, or other effect mediated by the
numbers and/or
activity of immune cells, and the like) are experienced by a patient. The term
includes the
administration of the compounds or agents of the present invention to prevent
or delay the
onset of the symptoms, complications, or biochemical indicia of a disease,
alleviating the


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 17-

symptoms or arresting or inhibiting further development of the disease,
condition, or disorder.
Treatment may be prophylactic (to prevent or delay the onset of the disease,
or to prevent the
manifestation of clinical or subclinical symptoms thereof) or therapeutic
suppression or
alleviation of symptoms after the manifestation of the disease.
"Combination therapy" embraces the administration of an immunostimulatory anti-

CTLA4 antibody, preferably, CP-675,206, and another therapeutic agent as part
of a specific
treatment regimen optionally including a maintenance phase, intended to
provide a beneficial
effect from the co-action of these therapeutic agents. The beneficial effect
of the combination
includes, but is not limited to, pharmacokinetic or pharmacodynamic co-action
resulting from
the combination of therapeutic agents. Administration of these therapeutic
agents in
combination typically is carried out over a defined time period (usually
minutes, hours, days or
weeks depending upon the combination selected). "Combination therapy"
generally is not
intended to encompass the administration of two or more of these therapeutic
agents as part
of separate monotherapy regimens that incidentally and arbitrarily result in
the combinations
of the present invention.
"Combination therapy" embraces administration of these therapeutic agents in a
sequential manner, that is, wherein each therapeutic agent is administered at
a different time,
as well as administration of these therapeutic agents, or at least two of the
therapeutic
agents, in a substantially simultaneous manner. Sequential or substantially
simultaneous
administration of each therapeutic agent can be effected by any appropriate
route including,
but not limited to, oral routes, intravenous routes, intramuscular,
subcutaneous routes, and
direct absorption through mucous membrane tissues. The therapeutic agents can
be
administered by the same route or by different routes. For example, a first
therapeutic agent
(e.g., a chemotherapeutic agent) can be administered orally, and a second
agent (e.g., anti-
CTLA4 antibody) can be administered intravenously. Further, a first
therapeutic agent of the
combination selected may be administered by intravenous injection while the
other
therapeutic agents of the combination may be administered orally.
Alternatively, for example,
both the therapeutic agents may be administered by intravenous or subcutaneous
injection.
In the present specification the term "sequential" means, unless otherwise
specified,
characterized by a regular sequence or order, e.g., if a dosage regimen
includes the
administration of an anti-CTLA4 antibody and a chemotherapeutic agent, a
sequential dosage
regimen could include administration of the anti-CTLA4 antibody before,
simultaneously,
substantially simultaneously, or after administration of the chemotherapeutic
agent, but both
agents will be administered in a regular sequence or order. The term
"separate" means,
unless otherwise specified, to keep apart one from the other. The term
"simultaneously"
means, unless otherwise specified, happening or done at the same time, i.e.,
the compounds


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 18-

of the invention are administered at the same time. The term "substantially
simultaneously"
means that the compounds are administered within minutes of each other (e.g.,
within 10
minutes of each other) and intends to embrace joint administration as well as
consecutive
administration, but if the administration is consecutive it is separated in
time for only a short
period (e.g., the time it would take a medical practitioner to administer two
compounds
separately). As used herein, concurrent administration and substantially
simultaneous
administration are used interchangeably. Sequential administration refers to
temporally
separated administration of the anti-CTLA4 antibody and the chemotherapeutic
agent.
"Combination therapy" also can embrace the administration of the therapeutic
agents
as described above in further combination with other biologically active
ingredients (such as,
but not limited to, a second and different antineoplastic agent, a dendritic
cell vaccine or other
tumor vaccine) and non-drug therapies (such as, but not limited to, surgery or
radiation
treatment). Where the combination therapy further comprises radiation
treatment,, the
radiation treatment may be conducted at any suitable time so long as a
beneficial effect from
the co-action of the combination of the therapeutic agents and radiation
treatment is
achieved. For example, in appropriate cases, the beneficial effect is still
achieved when the
radiation treatment is temporally removed from the administration of the
therapeutic agents,
perhaps by days or even weeks.
As used herein, the term "adjuvant therapy" refers to treatment given after
the primary
treatment, including, without limitation, radiation, chemotherapy, hormone
therapy, etc. The
goal of adjuvant therapy is to increase the patients' chances of remission or
cure, to increase
the patients' overall survival benefit, and to help decrease the risk of
recurrence. Therefore, it
will be understood that if the anti-CTLA4 antibody and therapeutic agent
combination is
administered as adjuvant therapy, the combination may be administered to the
patient after
the primary treatment, e.g., the patient is given a regimen of surgery,
radiation and/or
chemotherapy, followed by a course of a combination of an anti-CTLA4 antibody
and a
therapeutic agent. In this regard, the dose of anti-CTLA4 antibody and
therapeutic agent may
be considered a therapeutic dose or a maintenance dose, depending on the goals
of the
adjuvant therapy. The term "neoadjuvant therapy" refers to treatment given
before the
primary treatment, including, without limitation, surgery, radiation,
chemotherapy, etc. In the
neoadjuvant setting, the dose of anti-CTLA4 antibody and therapeutic agent is
considered a
therapeutic dose.
The term "first-line therapy" refers to the first type of therapy given for a
condition or
disease, or the first therapy of choice for the treatment of a particular type
of cancer. It
necessarily follows that the term "second-line therapy" therefore refers to
the treatment given
when the initial or first-line therapy is unsuccessful, and "third-line
therapy" refers to a


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 19-

treatment or treatment regimen that is given when both the initial treatment
and the
subsequent treatment are unsuccessful.

1. Combination Therapy
A. Cancer
CTLA4 antibodies described herein can be used to treat a wide variety of
cancers.
Without wishing to be bound by any particular theory, administration of a
therapeutic agent,
when administered with an immunoenhancing anti-CTLA4 antibody, may provide a
synergistic
effect. Without wishing to be bound by any theory of the invention, the agent
that mediates
an anti-tumor effect may mediate a decrease in tumor load, may mediate an
increase of
tumor antigens in the host antigen-presentation route, may decrease
inflammation such that
the antibody and/or other therapeutic agents may better penetrate the tumor,
and/or may
mediate a decrease in immune-suppressive tumor factors. Thus, the combination
of an anti-
CTLA4 antibody and at least one therapeutic agent may mediate a synergistic
therapeutic
effect thereby providing a benefit to a patient in need thereof which is
greater than either
compound alone.
Cancers that may be treated include, but are not limited to human sarcomas and
carcinomas, e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma,
osteogenic
sarcoma, Kaposi's sarcoma, chordoma, angiosarcoma, endotheliosarcoma,
lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma,
erythroblastoma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, a primary or
secondary brain tumor, colon carcinoma, pancreatic cancer, breast cancer,
ovarian cancer,
uterine cancer, cervical cancer, prostate cancer, skin cancer, bone cancer,
cancer of the
small intestine, cancer of the anal region, cancer of the head or neck,
gastrointestinal (gastric,
colorectal, and duodenal) cancer, esophageal cancer, squamous cell carcinoma,
basal cell
carcinoma, adenocarcinoma, sweat gland carcinoma, cutaneous or intraocular
melanoma,
carcinoma of the endometrium, carcinoma of the vagina, carcinoma of the
fallopian tubes,
carcinoma of the vulva, carcinoma of the cervix, neoplasms of the central
nervous system
(CNS), including primary or secondary CNS tumors, spinal axis tumors, primary
CNS
lymphoma, brain stem glioma, pituitary adenoma, sebaceous gland carcinoma,
papillary
carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma,
bronchogenic carcinoma, stomach cancer, colon cancer, cancer of the rectum,
renal cell
carcinoma (RCC), hepatoma, bile duct carcinoma, gall bladder cancer, liver
cancer, kidney
cancer, gastro-esophageal cancer, choriocarcinoma, seminoma, embryonal
carcinoma,
Wilms' tumor, testicular tumor, cancer of the penis, lung carcinoma, small
cell lung carcinoma
(SCLC), non-small cell lung cancer (NSCLC), thyroid cancer, cancer of the
parathyroid gland,


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 20-

carcinoid tumors, cancer of the urethra, cancer of the ureter, cancer of the
renal pelvis,
cancer of the endocrine system, cancer of the adrenal gland, pancreatic
endocrine tumors
(such as pheochromocytoma, insulinoma, vasoactive intestinal peptide tumor,
islet cell tumor
and glucagonoma), bladder carcinoma, epithelial carcinoma, glioma,
astrocytoma, pituitary
adenoma, adrenocortical cancer, medulloblastoma, craniopharyngioma,
ependymoma,
pinealoma, tumors of the blood vessels (including benign and malignant tumors
such as
hemangioma, hemangiosarcoma, hemangioblastoma and lobular capillary
hemangioma),
acoustic neuroma, oligodendroglioma, meningioma, melanoma, cholangiocarcinoma,
neuroblastoma, retinoblastoma, leukemias, acute lymphocytic leukemia, acute
myelocytic
leukemia, myeloblastic leukemia, promyelocytic leukemia, myelomonocytic
leukemia,
monocytic leukemia, erythroleukemia, chronic leukemia, lymphocytic leukemia,
chronic
myeloid leukemia, chronic myelogenous leukemia, chronic myelocytic leukemia,
chronic
granulocytic leukemia, chronic lymphocytic leukemia, lymphocytic lymphomas,
polycythemia
vera, lymphoma, Hodgkin's disease lymphoma, non-Hodgkin's disease lymphoma,
cutaneous
T-cell lymphoma (CTCL), cutaneous B-cell lymphoma, multiple myeloma,
Waldenstrom's
macroglobulinemia, heavy chain disease, soft tissue sarcomas, gastrointestinal
stromal
tumors (GIST), glioblastomas, or a combination of one or more of the foregoing
cancers.
In preferred embodiments, the cancers include non-Hodgkin's lymphoma, non-
small
cell lung cancer, colorectal carcinoma, chronic myeloid leukemia, chronic
lymphocytic
leukemia, pancreatic cancer, breast cancer and ovarian cancer.
Various preferred combination therapies are described herein for a variety of
cancers;
however, in certain embodiments described herein, the invention is not limited
to these, or
any other, combination therapies.
1. Non-Hodgkin's lymphoma (NHL)
In one embodiment, the invention includes administering CP-675,206 to treat
NHL, in
combination with at least one therapeutic agent, preferably, rituximab
(RITUXAN; Genentech,
San Francisco, CA).
The combination can be administered to treat a patient afflicted with indolent
NHL or
aggressive NHL. In one embodiment, the NHL is indolent NHL, and the
combination of CP-
675,206 and rituximab comprises first-line therapy. In a further embodiment,
the combination
therapy is administered as second-line therapy following cyclophosphamide,
doxorubicin,
vincristine, and prednisone (CHOP) and/or CHOP-R (cyclophosphamide,
doxorubicin,
vincristine, and prednisone with rituximab). In yet a further embodiment, the
combination is
followed by administration of rituximab as a single agent (SA) therapy. The
combination of
rituximab and CP-675,206 may increase and/or prolong an anti-tumor cell
response thereby
providing a therapeutic benefit to a patient suffering from NHL.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-21-
In one embodiment, a combination of CP-675,206 and rituximab is administered
as a
second-line therapy to treat aggressive NHL refractory to CHOP-R. In another
embodiment,
where the treatment is second-line therapy for aggressive NHL refractory to
CHOP-R, the
method further comprises administering rituximab as a single agent therapy
following
administration of the CP-675,206/rituximab combination.
In another aspect, the combination of CP-675,206 and rituximab is administered
to a
patient as a second-line therapy following another treatment including, but
not limited to, a
chemotherapeutic therapy or a bone marrow transplant. In one aspect, the CP-
675,206
rituximab combination is administered following high-dose bone marrow ablative
chemotherapy and bone marrow transplant (BMT). In another aspect, the
combination is
administered as a first-line therapy prior to bone marrow ablation
chemotherapy and BMT.
Methods well-known in the art for assessing therapeutic strategies for
treatment of NHL,
which consider various known factors such as age and condition of the patient,
stage of the
disease, and the like, are available to determine when the combination therapy
is indicated to
effectively treat a patient afflicted with aggressive NHL.
2. Non-small cell lung cancer (NSCLC)
In one embodiment, the invention includes a method for treating NSCLC
comprising
administering a combination comprising CP-675,206 and a therapeutic agent to a
patient in
need of treatment. In one aspect, the agent is a non-immunesuppressive
chemotherapeutic
agent, such as, but not limited to, gefitinib (IRESSA). The combination of CP-
675,206 and
therapeutic agent may increase the immune response to the tumor due to, inter
alia,
increased release of tumor antigen associated with the cytotoxic effect of the
chemotherapeutic agent. Thus, the skilled artisan, based upon the disclosure
provided
herein, would appreciate that while any therapeutic agent that mediates cell
death and/or
release of tumor antigen can be used with an anti-CTLA4 antibody to provide an
increased
immune response to the tumor and, thereby, providing a therapeutic benefit to
the patient.
Moreover, the skilled artisan would further understand once armed with the
teachings
provided herein, that the therapeutic effect can be further enhanced wherein
the therapeutic
agent does not decrease the immune enhancing effect of the antibody. While
gefitinib is a
chemotherapeutic agent known not to decrease the immune response in a patient,
the
invention is not limited to this, or any other, particular therapeutic agent.
Rather, in an
embodiment, the invention encompasses administration of CP-675,206 and a
therapeutic
agent that may mediate a therapeutic response to a NSCLC tumor.
In one embodiment of the invention, treatment comprises administration
combination
therapy comprising CP-675,206 and platinum-based chemotherapy. In one aspect,
CP-
675,206 is administered to a NSCLC patient following a course of platinum-
based therapy as


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 22-

an adjuvant therapy. That is, a patient that has been administered a course of
platinum-
based therapy and where the disease has responded or remained stable is
administered CP-
675,206. In one aspect, the patient NSCLC is Stage Illb (with effusion) or
Stage IV disease
that has responded or remained stable after about six cycles of a platinum-
containing
regimen. In a further aspect, the patient is administered CP-675,206 at least
about three
weeks after the last dose of platinum-based chemotherapy. In yet another
aspect, CP-
675,206 is administered within about six weeks after the last dose of platinum-
based
chemotherapy. In another aspect, CP-675,206 is administered at least about
three weeks
after but within six weeks of the last dose of platinum-based therapy. In one
aspect of the
invention, platinum-based chemotherapy comprises a platinum-based compound
selected
from such compounds as cisplatin, carboplatin (PARAPLATIN), eptaplatin,
lobaplatin,
nedaplatin, oxaliplatin (ELOXATIN, Sanofi), streptozocin, satraplatin (JM-1
26).
In another embodiment of the present invention, treatment for lung cancer
comprises
administration of bevacizumab (AVASTIN) and CP-675,206. In one aspect, the
lung cancer
comprises non-small cell and small cell lung cancer. In another aspect, the
lung cancer
comprises small cell lung cancer.
In another embodiment, the cancer is NSCLC and the treatment encompasses
administration of CP-675,206 in combination with a-taxane, where a taxane can
include, but is
not limited to, docetaxel (TAXOTERE), and paclitaxel (TAXOL), in further
combination with
gemcitabine (GEMZAR).
In another embodiment of the invention, the invention encompasses a method for
treating NSCLC comprising administration of a CP-675,206, a taxane (e.g.,
docetaxel and
paclitaxel), and a platinum compound. In one aspect, the method for treating
NSCLC is a first
line therapy comprising administering a combination of CP-675,206, a taxane
and a platinum
compound where the taxane is paclitaxel and the platin is carboplatin
(carbo/paclitaxel). In
another aspect, the combination of CP-675,206, paclitaxel and carboplatin is
administered
contemporaneously, or in sequence. In yet a further aspect, the
paclitaxel/carboplatin
combination is administered prior to or following administration of CP-
675,206. That is, the
antibody is contemporaneously administered in combination with the
carbo/paclitaxel therapy
or it is administered following carbo/paclitaxel therapy. The interval between
administration of
carbo/paclitaxel therapy and administration of the antibody may be readily
determined by one
skilled in the art based upon well-known methods. In one aspect, the cancer is
locally
advanced Stage Ilib NSCLC. In another aspect, the NSCLC is metastatic Stage IV
NSCLC.
In another aspect, the CP-675,206-carboplatin-paclitaxel combination is
administered with
bevacizumab. In another aspect, the CP-675,206-carboplatin-paclitaxel
combination is


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 23-

administered with sunitinib (SU11248). In a further aspect, the CP-675,206-
carboplatin-
paclitaxel combination is administered with PF03512676 (CpG-7909).
In another embodiment of the invention, the therapy is administration of
docetaxel
(TAXOTERE) and CP-675,206 as second line therapy for NSCLC for patients with
locally
advanced Stage IIIb or metastatic Stage IV disease after failure of prior
platinum-based
chemotherapy. In one aspect, CP-675,206 is administered contemporaneously with
docetaxel therapy. In another aspect, CP-675,206 is administered following
docetaxel
therapy. In yet another aspect, the NSCLC comprises locally advanced Stage
Illb or
metastatic Stage IV non-small cell lung cancer after failure of prior platinum-
based
chemotherapy.
In yet another embodiment, the invention includes treatment for NSCLC where
the
therapy comprises administration of CP-675,206 and eriotinib (TARCEVA) as
second line
therapy. In one aspect, CP-675,206 is contemporaneously administered with
eriotinib. In
another aspect, CP-675,206 is administered following erlotinib therapy.
In a further embodiment, the invention includes a second line therapy for
NSCLC
comprising administration of CP-675,206 and pemetrexed (ALIMTA). In one
aspect, CP-
675,206 is administered contemporaneously with pemetrexed. In another aspect,
CP-
675,206 is administered following pemetrexed therapy. In yet another aspect,
the NSCLC is
locally advanced Stage IIIb. In yet a further aspect, the NSCLC is metastatic
Stage IV non-
small cell lung cancer after failure of prior platinum-based chemotherapy.
In another embodiment, the invention includes treatment of locally advanced
Stage
IIIb or metastatic Stage IV NSCLC after failure of prior platinum-based
chemotherapy and
epidermal growth factor receptor inhibition-based therapy where the method is
a third line
treatment. In one aspect, CP-675,206 is administered as a single agent. In
another aspect,
CP-675,206 is administered in combination with a therapeutic agent.
3. Colorectal carcinoma (CRC)
In one embodiment, the invention includes a method for treating cancer of the
colon
and/or rectum. In one embodiment, the method comprises administering a
combination
comprising CP-675,206 and fluorouracil (5FU) as first line therapy for CRC
patients as first
line therapy of CRC patients intolerant of oxaliplatin (ELOXATIN) or
irinotecan (CAMPTO). In
another embodiment, the treatment comprises administering a combination of CP-
675,206
and capecitabine (XELODA) as first line therapy for patients intolerant of
oxaliplatin or
irinotecan.
In another embodiment, the therapy comprises first line treatment of patients
with
metastatic carcinoma of the colon or rectum comprising administration of CP-
675,206 and
FOLFOX (fluorouracil, leucovorin, and oxaliplatin). In one aspect, CP-675,206
is


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 24-

administered contemporaneously with FOLFOX therapy. In another aspect, CP-
675,206 is
administered following FOLFOX therapy.
In another aspect, the treatment comprises adjuvant therapy of patients with
Stage III
colon cancer who have undergone complete resection of the primary tumor. In a
further
aspect , the therapy comprises administration of CP-675,206 and FOLFOX
(fluorouracil,
leucovorin, and oxaliplatin). In one aspect, CP-675,206 is administered
contemporaneously
with FOLFOX therapy. In another aspect, CP-675,206 is administered following
FOLFOX
therapy.
In another embodiment, the therapy comprises first line treatment of patients
with
metastatic CRC comprising administration of CP-675,206 and FOLFIRI
(fluorouracil,
leucovorin, and irinotecan). In one aspect, CP-675,206 is administered
contemporaneously
with FOLFIRI therapy. In another aspect, CP-675,206 is administered following
FOLFIRI
therapy.
In another embodiment of the invention, the therapy comprises treatment of CRC
comprising administration of CP-675,206 and cetuximab (ERBITUX, ImClone). In
one aspect,
CP-675,206 is administered contemporaneously with cetuximab. In another
aspect, CP-
675,206 is administered following cetuximab.
In another embodiment, the invention encompasses a method of treating CRC
comprising administering a combination of CP-675,206 and fluorouracil and
capecitabine as
adjuvant therapy. Coadministration of a chemotherapeutic 'agent, which can
mediate
increased release of a tumor antigen, and CP-675,206 may mediate an increased
immune
response to the tumor thereby providing a therapeutic benefit to a patient
afflicted therewith.
4. Leukemia
In one embodiment, the invention includes a method of treating chronic myeloid
leukemia (CML). In one aspect, the method comprises administering a
combination of CP-
675,206 and imatinib mesylate (GLEEVEC, Novartis) as first line therapy of
CML.
In another embodiment, the invention includes a method for treating chronic
Iymphocytic leukemia (CLL). In one aspect, the treatment comprises
administration of CP-
675,206 and imatinib mesylate as first line therapy.
5. Pancreatic cancer
In one embodiment, the invention includes a method for treating pancreatic
cancer
comprising administering a combination of CP-675,206 and gemcitabine (GEMZAR).
In one
aspect, the therapy is a first line treatment for patients with locally
advanced (non-resectable
Stage II or Stage III) adenocarcinoma of the pancreas. In another aspect, the
cancer is
metastatic (Stage IV) adenocarcinoma of the pancreas. In yet another aspect,
gemcitabine is


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 25-

administered contemporaneously with CP-675,206. In yet a further aspect, CP-
675,206 is
administered following administration of gemcitabine.
In one embodiment of the invention, CP-675,206 is administered after at least
one
course of gemcitabine. In one aspect, gemcitabine is administered
intravenously at a dose of
about 1000 mg/m2 once per week for up to seven (7) weeks or until toxicity
necessitates
reducing or withholding a dose. The administration phase is followed by a one
week resting
period during which gemcitabine is not administered. Subsequent cycles of
gemcitabine
administration consist of i.v. infusions once weekly for three consecutive
weeks followed by
one week resting period. In a further aspect, CP-675,206 is administered after
a full course
(i.e., seven weeks administration and one week resting period) of gemcitabine.
In another
embodiment, CP-675,206 is administered every three weeks thereafter. In yet
another
aspect, the combination of gemcitabine (e.g., three weeks administration
followed by one
week rest) and CP-675,206 (e.g., every three weeks) is continued until disease
progression
or intolerable toxicity.
6. Breast cancer
In one embodiment, the invention includes a method to treat locally advanced
or
metastatic triple receptor negative breast cancer. In one aspect, the method
is a first line
therapy. In another aspect, the therapy comprises administering CP-675,206 and
a taxane
(e.g., docetaxel and paclitaxel). In one aspect, CP-675,206 taxane are
administered
contemporaneously. In another aspect, CP-675,206 is administered following
administration
of the taxane.
In another embodiment, the therapy comprises adjuvant treatment for breast
cancer
comprising administration of CP-675,206 and cyclophosphamide, doxorubicin and
a taxane.
In one aspect, the therapy comprises adjuvant therapy.
In another embodiment, therapy comprises administration of CP-675,206 and
tamoxifen (NOVALDEX), anastrazole (ARIMIDEX), letrozole (FEMARA), exemestane
(AROMASIN), or fulvestrant (FASLODEX), or a combination thereof. In one
aspect, the
therapy is adjuvant therapy for metastatic breast cancer.
In another embodiment of the present invention, the therapy for breast cancer
comprises administration of CP-675,206 and trastuzumab (HERCEPTIN). In one
aspect, the
antibodies are administered contemporaneously. In another aspect, trastuzumab
is
administered followed by administration of CP-675,206. In a further aspect,
the CP-675,206-
trastuzumab combination therapy comprises adjuvant therapy and first line
therapy for
metastatic breast cancer.
In yet another embodiment, the therapy comprises administering CP-675,206 and
bevacizumab. In one aspect, CP-675,206 and bevacizumab are administered


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 26-

contemporaneously. In another aspect, CP-675,206 is administered following
administration
of bevacizumab.
In one embodiment of the invention, the therapy for treating breast cancer
comprises
administering CP-675,206 and cetuximab. In one aspect, CP-675,206 and
cetuximab are
administered contemporaneously. In another aspect, CP-675,206 is administered
following
administration of cetuximab.
In another embodiment of the invention, the therapy for treating breast cancer
comprises administering CP-675,206 and axitinib. In one aspect, CP-675,206 and
axitinib are
administered contemporaneously. In another aspect, CP-675,206 is administered
following
administration of axitinib.
7. Ovarian cancer
In one embodiment, the invention includes a method for treatment carcinoma of
the
ovary. In one embodiment, therapy comprises administration of CP-675,206 and
carboplatin
and paclitaxel. In one aspect, CP-675,206 is administered contemporaneously
with
carboplatin and paclitaxel. In another aspect, CP-675,206 is administered
following
administration of carboplatin and paclitaxel. In yet another aspect, the
treatment is a first
line treatment for advanced carcinoma of the ovary.
In another embodiment of the invention, therapy comprises second line
treatment for
patients who have progressed (e.g., as indicated by tumor assessment, CA-125
doubling
value, CT scan, and the like) following paclitaxel-based therapy. In one
aspect, therapy
comprises administration of CP-675,206 as a single agent. In another aspect,
therapy
comprises administration of CP-675,206 in combination with an agent selected
from the group
consisting of altretamine, anastrozole, bevacizumab, carboplatin, cisplatin,
cyclophosphamide, liposomal doxorubicin, docetaxel, gemcitabine, ifosfamide,
irinotecan,
letrozole, melphalan, oral etoposide, oxaliplatin, tamoxifen, topotecan, and
vinorelbine, and
any combination thereof.
8. Gastric cancer
In one embodiment of the present invention, the treatment comprises gastric
cancer
and therapy comprises administering CP-675,206 and irinotecan. In one aspect,
CP-675,206
and irinotecan are administered contemporaneously. In another aspect, CP-
675,206 is
administered following administration of irinotecan.
In another embodiment, treatment comprises administration of CP-675,206,
fluorouracil and leucovorin. In one aspect, CP-675,206, fluorouracil and
leucovorin are
administered contemporaneously. In another aspect, CP-675,206 is administered
following
administration of fluorouracil and leucovorin.
9. Liver cancer


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 27-

In one embodiment of the invention, the cancer is liver cancer and therapy
comprises
administering CP-675,206 and doxorubicin, ifosfamide and vincristine. In one
aspect, CP-
675,206, and doxorubicin, ifosfamide and vincristine are administered
contemporaneously. In
another aspect, CP-675,206 is administered following administration of
doxorubicin,
ifosfamide and vincristine.
In another embodiment, the cancer is liver cancer and therapy comprises
administering CP-675,206, doxorubicin, and vincristine. In one aspect, CP-
675,206, and
doxorubicin, ifosfamide and vincristine are administered contemporaneously. In
another
aspect, CP-675,206 is administered following administration of doxorubicin and
vincristine.
10. Metastatic lesions
In one embodiment of the present invention, CP-675,206 is administered to
treat
metastases where the metastases is to the brain and/or bone metastasis. In one
aspect, the
brain and bone metastases are treated using CP-675,206 regardless of the site
of the primary
tumor from which the cancer metastasized.
In one aspect, brain metastases are treated using a combination of surgical
resection
and/or radiotherapeutic ablation in further combination with an anti-CTLA4
antibody,
preferably, CP-675,206. That is, where multiple metastases and/or lesions
greater than about
3 centimeters in diameter are present, whole brain radiation may be
administered followed by
administration of anti-CTLA4 antibody. In a further aspect, the antibody is
administered
preceding radiation. In yet a further aspect, the antibody is administered at
least about 3
mg/kg, more preferably, at least about 6 mg/kg, even more preferably, at least
about 10
mg/kg, and more preferably, at least about 15 mg/kg. In another aspect, the
antibody is
administered every three weeks, more preferably, every four weeks, and even
more
preferably, every three months.
In one embodiment, where there are fewer than about 5 brain lesions and all
lesions
are less than about three centimeres in maximal diameter, stereotactic
radiosurgery (SRS) is
administered followed by administration of anti-CTLA4 antibody. In one aspect,
the antibody
is administered before SRS. In another aspect, the antibody is administered
before and after
SRS.
In another embodiment, surgery, whole brain radiation and SRS may be combined
in
any order to treat brain metastases, which treatment is further combined with
antibody
therapy. In one aspect, the antibody therapy is administered after
radiation/surgery/SRS.
Alternatively, antibody surgery is administered before radiation/surgery/SRS
and may be
administered before and after such therapy.
This is because, as would be understood by the skilled artisan, brain
metastases are
common in melanoma. While SRS may achieve local control of the brain
metastases, most


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 28-

patients die of systemic metastases and/or reseeding of the CNS occurs due to
poor
management of the systemic disease. Accordingly, in one embodiment, the
invention
provides a combination therapy to control the local brain metastases and to
prevent reseeding
of the CNS and control the systemic disease thereby providing a therapeutic
benefit to a
patient in need thereof.
11. Bladder cancer
In one embodiment, CP-675,206 is administered in combination with BCG to treat
bladder cancer. In one aspect, CP-675,206 and BCG are administered
contemporaneously.
In another aspect, CP-675,206 is administered following administration of BCG.
In another embodiment, treatment of bladder cancer comprises administering CP-
675,206 and gemcitabine and cisplatin. In one aspect, CP-675,206, gemcitabine
and
cisplatin are administered contemporaneously. In another aspect, CP-675,206 is
administered following administration of gemcitabine and cisplatin.
12. Melanoma
The present invention encompasses combination of CP-675,206 and interferon
alpha
for treating melanoma. In one aspect, CP-675,206 and interferon alpha are
administered
contemporaneously. In another aspect, CP-675,206 is administered following
administration
of interferon alpha. In a further aspect, interferon alpha is administered as
a high dose.
13. Multiple myeloma
In one embodiment of the present invention, treatment for multiple myeloma
comprises administration of CP-675,206 and bortezomib. In one aspect, CP-
675,206 and
bortezomib are administered contemporaneously. In another aspect, CP-675,206
is
administered following administration of bortezomib.
In another embodiment, therapy comprises administration of CP-675,206 and
dexamethasone and thalidomide. In one aspect, CP-675,206, dexamethasone and
thalidomide are administered contemporaneously. In another aspect, CP-675,206
is
administered following administration of dexamethasone and thalidomide.
In alternate embodiments of the invention discussed above, anti-CTLA4
antibodies
other than CP-675,206 may be used in the methods of the invention. Alternate
antibodies are
described herein, including the antibodies described in U.S. Patent
Application No.
09/472,087, now issued as U.S. Patent No. 6,682,736; Int. Appi. No.
PCT/US99/30895
(published June 29, 2000, as WO 00/37504); U.S. Pat. Appl. No. 10/612,497
(published
November 18, 2004, as US 2004/0228858); U.S. Pat. Appl. No. 10/776,649
(published
November 18, 2004, as US 2004/0228861); lnt. Appi. No. Int. Appl. No.
PCT/USOO/23356
(published March 1, 2001, as WO 01/14424) (e.g., antibody 10D1, also known as
MDX-010,
and referred to herein as ipilimumab, Medarex, Princeton, NJ); Int. Appl. No.
PCT/US99/28739


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 29-

(published June 8, 2000, as WO 00/32231); U.S. Pat. Nos. 5,811,097, 5,855,887,
6,051,227,
and 6,207,156; U.S. Pat. No. 5,844,095, to Linsley et al.; Int. Appl. No.
PCT/US92/05202
(published Jan. 7, 1993, as WO 93/00431); U.S. Patent Appl. No. 10/153,382
(published May 8,
2003, as US 2003/0086930); U.S. Pat. Appl. No. 10/673,738 (published February
24, 2005 as
US 2005/0042223); U.S. Pat. Appi. No. 11/085,368 (published October 13, 2005,
as US
2005/0226875); U.S. Pat. Appl. No. 60/624,856 (filed Nov. 4, 2004); U.S. Pat.
Appl. No.
60/664,364 (filed March 23, 2005); U.S. Pat. Appl. No. 60/664,653 (filed March
23, 2005); U.S.
Pat. Appl. No. 60/697,082 (filed July 7, 2005); U.S. Pat. Appl. No. 60/711,707
(filed Aug. 26,
2005). More preferably, the antibodies are described in U.S. Patent No.
6,682,736, and WO
01/14424. Even more preferably, the antibodies include an antibody having the
heavy and
light chain amino acid sequences of an antibody selected from the group
consisting of 4.1.1,
4.8.1, 4.10.2, 4.13.1, 4.14.3, 6.1.1, 11.2.1 (CP-675,206 or ticilimumab),
11.6.1, 11.7.1.,
12.3.1.1,12.9.1.1, and ipilimumab.
B. Pathogens
Because of the immune-enhancing effects of CTLA4-blockade, it would be
understood by the skilled artisan armed with the disclosure provided herein
that the present
invention encompasses administration of anti-CTLA4 antibody in combination
with standard of
care therapy to treat a variety of pathogen-associated diseases, disorders or
conditions,
where an immune response to the pathogen would provide a therapeutic benefit.
These
include, but are not limited to, bacterial, viral, fungal, parasitic, and
other pathogenic disease.
Treatment of infectious disease comprising anti-CTLA4 antibody blockade is
discussed in,
e.g., WO 03/086459, published on Oct. 23, 2003.
Similar to use of CTLA4 blockade in combination to treat a tumor discussed
previously, anti-CTLA4 antibody blockade can be used in combination with
vaccines and
other therapeutic agents, including standard of care therapy, to treat a
disease, disorder or
condition associated with a pathogens, or a toxin derived therefrom. That is,
previous studies
have demonstrated that anti-CTLA4 antibody blockade can be useful in treatment
of
infectious parasites (McCoy et al., J. Exp. Med. 186:183-187 (1997); Murphy et
al., J.
Immunol. 161:4153-4160 (1998)). Examples of pathogens for which this
therapeutic approach
may be particularly useful, include, but are not limited to HIV, Hepatitis (A,
B, & C), Influenza,
Herpes, Giardia, Malaria, Leishmania, Staphylococcus aureus, Pseudomonas
aureginosa,
and Helicobacter pylori.
Some examples of viruses treatable by methods of the invention include
hepatitis (A,
B, or C), herpes virus (e.g., VZV, HSV-1, HAV-6, HSV-II, and CMV, Epstein Barr
virus),
adenovirus, influenza virus, flaviviruses, echovirus, ebola virus, rhinovirus,
coxsackie virus,
cornovirus, respiratory syncytial virus, mumps virus, rotavirus, measles
virus, rubella virus,


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 30-

parvovirus, vaccinia virus, poxvirus, HTLV virus, dengue virus,
papillomavirus, molluscum
virus, poliovirus, rabies virus, JC virus and arboviral encephalitis virus.
In one embodiment, bacterial infection treatable by methods of the invention
include
chiamydia, rickettsial bacteria, mycobacteria, staphylococci, streptococci,
pneumonococci,
meningococci and conococci, klebsiella, proteus, serratia, pseudomonas,
legionella,
diphtheria, salmonella, bacilli, helicobacter, cholera, tetanus, botulism,
anthrax, plague,
leptospirosis, and Lyme Disease bacteria (e.g., Borrelia burgdorferi).
In another embodiment, fungal infection treatable according to the methods of
the
invention include, but are not limited to,
Candida (albicans, krusei, glabrata, tropicalis, etc.), Cryptococcus
neoformans, Aspergillus
(fumigatus, niger, etc.), Genus Mucorales (mucor, absidia, rhizophus),
Sporothrix schenkii,
Blastomyces dermatitidis, Paracoccidioides brasiliensis, Coccidioides immitis
and
Histoplasma capsulatum.
In yet another embodiment, parasitic infection treatable by methods of the
invention
include, among others, Entamoeba histolytica, Balantidium coli,
Naegleriafowleri,
Acanthamoeba sp., Giardia lambia, Cryptosporidium sp., Pneumocystis carinii,
Plasmodium
vivax, Babesia microti, Trypanosoma brucei, Trypanosoma cruzi, Leishmania
donovani,
Toxoplasma gondi, Nippostrongylus brasiliensis.
1. HIV/AIDS
Studies suggest that CTLA4 blockade may be useful for treatment of viral
disease,
more specifically, studies in rhesus macaques demonstrated that anti-CTLA4
(i.e.,
ipilimumab), combined with antiretroviral therapy ("ART"), was associated with
increased
antiviral response. See Hryniewicz et al., Blood 108:3834-3842 (2006). More
particularly,
CTLA4 blockade not only decreased viral load and increased virus-specific
effector T cells, it
also decreased the level of immunosuppressive molecules (e.g., transforming
growth factor
beta [TGF-(3] and indoleamine 2,3-dioxygenase [IDO]), in SIVma1251-infected
macaques also
receiving ART (didanosine, stavudine and tenofovir [PMPA]). These data suggest
that anti-
CTLA4 antibody, in combination with antiviral therapy, may provide a
therapeutic benefit in
treatment of HIV infection in humans.
In one embodiment of the invention, anti-CTLA4 antibodies, in particular CP-
675,206,
and their pharmaceutically acceptable salts, solvates and derivatives, may be
administered
alone or as part of a combination therapy. Thus included within the scope of
the present
invention are embodiments comprising co-administration of, and compositions
which contain,
in addition to a compound of the invention, one or more additional therapeutic
agents.
Such multiple drug regimens, often referred to as combination therapy, may be
used
in the treatment and prevention of infection and multiplication of the human
immunodeficiency


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-31-
virus, HIV, and related pathogenic retroviruses within a patient in need of
treatment or one at
risk of becoming such a patient. The ability of such retroviral pathogens to
evolve within a
relatively short period of time into strains resistant to any monotherapy
which has been
administered to said patient is well known in the literature. A recommended
treatment for HIV
is a combination drug treatment called Highly Active Anti-Retroviral Therapy
("HAART").
HAART combines three or more HIV drugs. Thus, the methods of treatment and
pharmaceutical compositions of the present invention may employ a compound of
the
invention in the form of monotherapy, but said methods and compositions may
also be used
in the form of combination therapy in which anti-CTLA4 antibody is co-
administered in
combination with one or more additional therapeutic agents such as those
described in detail
further herein.
The therapeutic agents that may be used in combination with the anti-CTLA4
antibody include, but are not limited to, those useful as HIV protease
inhibitors (PIs), non-
nucleoside reverse transcriptase inhibitors (NNRTIs), nucleoside/nucleotide
reverse
transcriptase inhibitors (NRTIs), CCR5 antagonists, agents which inhibit the
interaction of
gp120 with CD4, other agents which inhibit the entry of HIV into a target cell
(such as fusion
inhibitors), inhibitors of HIV integrase, RNaseH inhibitors, prenylation
inhibitors, maturation
inhibitors which act by interfering with production of the HIV capsid protein,
compounds useful
as anti-infectives, and others as described below.
It will be appreciated by a person skilled in the art, that a combination drug
treatment,
as described herein above, may comprise two or more compounds having the same,
or
different, mechanism of action. Thus, by way of illustration only, a
combination may comprise
a compound of the invention and: one or more NRTIs; one or more NRTis and a
PI; one or
more NRTIs and another CCR5 antagonist; a PI; a PI and an NNRTI; an NNRTI; and
so on.
Examples of Pls include, but are not limited to, amprenavir (141 W94), CGP-
73547,
CGP-61755, DMP-450 (mozenavir), nelfinavir, ritonavir, saquinavir (invirase),
lopinavir, TMC-
126, atazanavir, palinavir, GS-3333, KN 1-413, KNI-272, LG-71350, CGP-61755,
PD 173606,
PD 177298, PD 178390, PD 178392, U-140690, ABT-378, DMP-450, AG-1776, MK-944,
becanavir (formerly known as VX-478, GW640385), indinavir, tipranavir, TMC-1
14, DPC-681,
DPC-684, fosamprenavir calcium (Lexiva), benzenesulfonamide derivatives
disclosed in WO
03/053435, R-944, Ro-03-34649, VX-385, GS-224338, OPT-TL3, PL-100, PPL-100, SM-

309515, AG-148, DG-35-VIII, DMP-850, GW-5950X, KNI-1039, L-756423, LB-71262,
LP-130,
RS-344, SE-063, UIC-94-003, Vb-19038, A-77003, BMS-182193, BMS-186318, SM-
309515,
JE-2147, GS-9005.
Examples of NRTis include, but are not limited to, abacavir, GS-840,
lamivudine,
adefovir dipivoxil, beta-fluoro-ddA, zalcitabine, didanosine, stavudine,
zidovudine, tenofovir


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 32-
(9-[9(R)-2-(phosphonomethoxy)propyl]adenine; PMPA), tenofovir disoproxil
fumarate (Viread;
Gliead Sciences), amdoxovir (DAPD), SPD-754, SPD-756, racivir, reverset (DPC-
817), MIV-
210 (FLG), beta-L-Fd4C (ACH-126443), MIV-310 (alovudine, FLT), dOTC, DAPD,
entecavir,
GS-7340, emtricitabine (FTC).
Examples of NNRTIs include, but are not limited to, efavirenz, HBY-097,
nevirapine,
TMC-120 (dapivirine), TMC-125, etravirine, delavirdine, DPC-083, DPC-961,
capravirine,
rilpivirine, 5-{[3,5-Diethyl-1-(2-hydroxyethyl)-1 H-pyrazol-4-
yl]oxy}isophthalonitrile or
pharmaceutically acceptable salts, solvates or derivatives thereof; GW-678248,
GW-695634,
MIV-150, calanolide, and tricyclic pyrimidinone derivatives as disclosed in WO
03/062238.
Examples of CCR5 antagonists include, but are not limited to, TAK-779, SC-
351125,
ancriviroc (formerly known as SCH-C), vicriviroc (formerly known as SCH-D),
maraviroc,
NCB-9471, CCR5mAb004, PRO-140, aplaviroc (also known as GW-873140, Ono-4128,
AK-
602), AMD-887 CMPD-167, methyl 1-endo-{8-[(3S)-3-(acetylamino)-3-(3-
fluorophenyl)propyl]-
8-azabicyclo[3.2.1 ]oct-3-yl}-2-methyl-4,5,6,7-tetrahyd ro-1 H-im idazo[4, 5-
c]pyrid ine-5-
carboxylate or pharmaceutically acceptable salts, solvates or derivatives
thereof, methyl 3-
endo-{8-[(3S)-3-(acetamido)-3-(3-fluorophenyl)propyl]-8-azabicyclo[3.2.1 ]oct-
3-yl}-2-methyl-
4,5,6,7-tetrahydro-3H-imidazo[4,5-c]pyridine-5-carboxylate or pharmaceutically
acceptable
salts, solvates or derivatives thereof, ethyl 1 -endo-{8-[(3S)-3-(acetylamino)-
3-(3-
fluorophenyl)propyl]-8-azabicyclo[3.2. 1 ]oct-3-yl}-2-methyl-4,5,6,7-
tetrahydro-1 H-im idazo[4,5-
c]pyridine-5-carboxylate or pharmaceutically acceptable salts, solvates or
derivatives thereof,
and /V {(1S)-3-[3-endo-(5-Isobutyryl-2-methyl-4,5,6,7-tetrahydro-1H-
imidazo[4,5-c]pyridin-1-
yl)-8-azabicyclo[3.2.1 ]oct-8-yl]-1-(3-fluorophenyl)propyl}acetamide) or
pharmaceutically
acceptable salts, solvates or derivatives thereof.
In one embodiment, the anti-CTLA4 antibody, preferably CP-675,206, is
administered
in combination with maraviroc. In another embodiment, anti-CTLA4
antibody/maraviroc
combination therapy comprises assessing whether is expressed on the surface of
the
patient's cells. In yet another embodiment, the therapy comprises assessing
the co-receptor
(i.e., CXCR4 or CCR5) tropism of the HIV virus to determine whether the
patient is infected
with an HIV that uses the CCR5 co-receptor, the CXCR4 co-receptor, or both
(dual/mixed
tropic) to determined whether the patient is a candidate for maraviroc
therapy. In another
embodiment, the co-receptor tropism assay is TROFILE (Monogram Biosciences,
Inc., San
Francisco, CA). This is because treatment with maraviroc inhibits CCR5-
mediated entry into
cells. Thus, assessing CCR5 expression by a patient's cell and/or assessing
the co-receptor
tropism of the virus optimizes the treatment options for the patient.
Examples of entry and fusion inhibitors include, but are not limited to, BMS-
806,
BMS-488043, 5-{(1S)-2-[(2R)-4-Benzoyl-2-methyl-piperazin-1-yl]-1-methyl-2-oxo-
ethoxy}-4-


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 33-

methoxy-pyridine-2-carboxylic acid methylamide and 4-{(1S)-2-[(2R)-4-Benzoyl-2-
methyl-
piperazin-1-yl]-1-methyl-2-oxo-ethoxy}-3-methoxy-N-methyl-benzamide,
enfuvirtide (T-20),
sifuvirtide SP-01A, T1249, PRO 542, AMD-3100, soluble CD4, compounds disclosed
in JP
2003171381, and compounds disclosed in JP 2003119137.
Examples of inhibitors of HIV integrase include, but are not limited to, L-
000870810
GW-810781, 1,5-naphthyridine-3-carboxamide derivatives disclosed in WO
03/062204,
compounds disclosed in WO 03/047564, compounds disclosed in WO 03/049690, and
5-
hydroxypyrimidine-4-carboxamide derivatives disclosed in WO 03/035076, MK-0518
(5-(1,1-
dioxo-1,2-thiazinan-2-yl)-N- (4-fluorobenzyl)-8-hydroxy-1,6-naphthyridine-7-
carboxamide-
disclosed in WO 03016315), GS-9137 (JTK-303).
Examples of prenylation inhibitors include, but are not limited to, HMG CoA
reductase
inhibitors, such as statins (e.g., atorvastatin).
Examples of maturation inhibitors include 3-0-(3'3'-dimethylsuccinyl) betulic
acid
(otherwise known as PA-457) and alphaHGA.
II. Dosage Regimens
Dosage regimens may be adjusted to provide the optimum desired 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. It is especially advantageous to formulate
parenteral compositions in
dosage unit form for ease of administration and uniformity of dosage. Dosage
unit form as used
herein refers to physically discrete units suited as unitary dosages for the
mammalian subjects
to be treated; each unit containing a predetermined quantity of active
compound calculated to
produce the desired therapeutic effect in association with the required
pharmaceutical carrier.
The specification for the dosage unit forms of the invention are dictated by
and directly
dependent on (a) the unique characteristics of the antibody and the particular
therapeutic or
prophylactic effect to be achieved, and (b) the limitations inherent in the
art of compounding
such an active compound for the treatment of sensitivity in individuals.
An exemplary, non limiting range for a therapeutically effective amount of
ticilimumab
administered according to the invention is at least about 1 mg/kg, at least
about 5 mg/kg, at
least about 10 mg/kg, more than about 10 mg/kg, or at least about 15 mg/kg,
for example about
1-30 mg/kg, or for example about 1-25 mg/kg, or for example about 1-20 mg/kg,
or for example
about 5-20 mg/kg, or for example about 10-20 mg/kg, or for example about 15-20
mg/kg, or for
example, about 15 mg/kg. It is to be noted that dosage values may vary with
the type and
severity of the condition to be alleviated, and may include single or multiple
doses. It is to be
further understood that for any particular subject, specific dosage regimens
should be adjusted


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 34-

over time according to the individual need and the professional judgment of
the person
administering or supervising the administration of the compositions, and that
dosage ranges set
forth herein are exemplary only and are not intended to limit the scope or
practice of the claimed
composition. Determining appropriate dosages and regiments for administration
of the antibody
are well-known in the relevant art and would be understood to be encompassed
by the skilled
artisan once provided the teachings disclosed herein.
In one embodiment, CP-675,206 is administered in an intravenous formulation as
a
sterile aqueous solution containing about 5 to 20 mg/mI of antibody, in an
appropriate buffer
system.
In one embodiment, for administration of low doses, part of the dose is
administered by
an intravenous bolus and the rest by infusion of the antibody formulation. For
example, a 0.01
mg/kg intravenous injection of the antibody may be given as a bolus, and the
rest of a
predetermined antibody dose may be administered by intravenous injection. In
another
embodiment, the entire low dose is administered as a single bolus injection.
For higher doses,
e.g., 3 mg/kg, the antibody is not administered as a bolus, but the entire
amount is administered
by infusion. A predetermined dose of the antibody may be administered, for
example, over a
period of about an hour and a half to about five hours.
The present invention relates to administering a combination of an anti-CTLA4
antibody
and at least one therapeutic agent. In one embodiment, the antibody is CP-
675,206. In
alternative embodiments of the invention, the antibody is selected from
various anti-CTLA4
antibodies, including, but not limited to, ipilimumab. In one aspect, the
antibody is ipilimumab
and the dose is about 3 mg/kg. In another aspect, the dose of ipilimumab is at
least about 10
mg/kg.
In one embodiment of the present invention, the antibody (e.g., CP-675,206,
ipilimumab, and the like) is administered approximately every three weeks,
more preferably, for
about four cycles followed by every three months thereafter. In one aspect of
this embodiment,
the antibody is administered at about 10 mg/kg.
The skilled artisan would appreciate that the combination of CP-675,206 and a
therapeutic agent can be administered simultaneously or the antibody and the
therapeutic agent
can be administered at different times. For instance, in one embodiment, the
antibody is
administered as a single injection and/or infusion and the therapeutic agent
(e.g., gemcitabine)
is administered once per day commencing before, during, or after
administration of the
antibody. However, the present invention is not limited to any particular
dosage or
administration regimen for a therapeutic agent. Rather, the optimal dose,
route and regimen for
administration of the antibody and the therapeutic agent can be readily
determined by one of
ordinary skill in the relevant art using well-known methods.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 35-

For instance, a single dose or multiples doses of the antibody may be
administered.
Alternatively, at least one dose, or at least three, six or 12 doses may be
administered.The
doses may be administered, for example, every two weeks, every three weeks,
monthly,
every twenty days, every 25 days, every 28 days, every 30 days, every 40 days,
every 6
weeks, every 50 days, every two months, every 70 days, every 80 days, every
three months,
every six months or yearly. In one aspect, the antibody is administered once
every three
weeks, preferably for four cycles, and then every three months thereafter. In
addition, the
second therapeutic agent can be administered daily, several times or once per
day, weekly,
every other week, every third week, every fourth week, monthly, every three
months, every
six months, once per year, or any other period that provides a therapeutic
benefit to the
patient as determined by the skilled practitioner.
In one aspect, CP-675,206 is administered once per month. In another aspect,
CP-
675,206 is administered every three months. In yet another aspect, ipilimumab
is administered
once per month.
The antibody can be administered until disease progression, or intolerable
toxicity, or
up to 12 consecutive cycles, whichever time is shorter. The antibody can also
be
administered using a regimen comprising administration of a loading dose
followed by a lower
dose. Repeat courses of at least one, and more preferably, several cycles of
antibody and a
therapeutic agent can be administered to a patient that experiences a tumor
recurrence who
had previously derived benefit from administration of the combination of an
anti-CTLA4
antibody and another therapeutic agent, or the antibody or therapeutic agent
not administered
in combination previously.
In one embodiment, a single injection comprising CP-675,206 is administered to
a
patient intravenously at a dose of about 3 mg/kg every twenty-eight days. In
another
embodiment, an anti-CTLA4 antibody is administered to a patient intravenously
at a dose of
about 3 mg/kg, preferably, about 6 mg/kg, more preferably, about 10 mg/kg,
preferably, about
15 mg/kg, every three weeks. After four administrations, the antibody is then
administered
every three months.
The invention encompasses administration of an anti-CTLA4 antibody, preferably
CP-
675,206, in combination with a wide plethora of agents. The skilled artisan
would appreciate,
once armed with the teachings provided herein, that any art-recognized dosing
regimen for the
agent may be used. Dosing regimens for chemotherapeutic and other agents
described herein
may be found in treatises well known in the art, including, but not limited
to, Cancer: Principles
and Practice of Oncology, 7th edition, DeVita, Hellman, and Rosenberg,
editors, Lippincott,
Williams & Wilkins, 2004, and Cancer Chemotherapy and Biotherapy Principles
and Practice,
4 'h edition. Chabner, Longo, editors, Lippincott, Williams & Wilkins, 2005.
Additionally,


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 36-

administration of many agents disclosed herein may be performed based upon the
FDA-
approved drug label instructions. Such label instructions are readily and
publicly available from
the website of the Department of Health and Sciences, U.S. Food and Drug
Administration,
Center for Drug Evaluation and Research website, which may be searched
according to drug
name or active ingredient. Thus, the skilled artisan would understand the
dosing regimen to be
used in combination therapy for various diseases comprising administration of
an anti-CTLA4
antibody and a therapeutic agent. The dose may be adjusted as known in the art
based-on,
among other factors, toxicity, if any, and therapeutic effectiveness. Thus,
dosing regimens for
many chemotherapeutic, and other agents related to cancer treatment, are well
known in the art
and are described in numerous publications, including those described herein.
See also
International Publication No. PCT/US03/12802 (published Nov. 6, 2003, as WO
03/090686),
which is incorporated by reference herein for all purposes, for a list of
exemplary dosing
regimens for various chemotherapeutic agents (e.g., at pages 72 through 74).
In some embodiments, the antibody-therapeutic agent combination can be
administered as an adjuvant therapy, or it can be administered to the patient
as a neoadjuvant
therapy prior to surgery, radiation therapy, or any,other treatment, in order
to sensitize the tumor
cells or to otherwise confer a therapeutic benefit to the patient.
Further, in some embodiments, the combination can be administered as a first
line
therapy, or as a second line or third line therapy, such as, but not limited
to, once previous
therapy(ies) has failed. Alternatively, the combination can be administered
concurrently with a
first line therapy, and or at any point during therapy, following initial
treatment. Thus, a
combination of an anti-CTLA4 antibody and a therapeutic agent can provide a
therapeutic
benefit once a previous therapy has failed, once systemic adjuvant therapy
using another
therapeutic agent (e.g., temozolomide, leuprolide, paclitaxel, imatinib
mesylate, gefitinib, and
the like) has failed. Therefore, the invention encompasses administration of
an anti-CTLA4
antibody and a therapeutic agent in combination with or following additional
therapy, including,
but not limited to, a different therapeutic agent (e.g., a chemotherapeutic,
an antibody, an
immunomodulator, a cytokine, and the like) as would be appreciated by one
skilled in the art
based upon the disclosure provided herein.
III. Anti-CTLA4 Antibodies
The invention encompasses alternate embodiments employing CTLA4 antibodies in
addition to CP-675,206. In one embodiment, the CTLA4 antibody comprises a
heavy chain
wherein the amino acid sequence of the VH comprises the amino acid sequences
set forth in
SEQ ID NOs:3, 15 and 27. In yet another embodiment, the VL of the CTLA4
antibody
comprises the amino acid sequences set forth in SEQ ID NOs:9, 21 and 33. More
preferably,


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-37-
the VH and VL regions of the antibody comprise the amino acid sequences set
forth in SEQ ID
NO:3 (VH 4.1.1) and SEQ ID N0:9 (VL 4.1.1), respectively; the amino acid
sequences set
forth in SEQ ID N0:15 (VH 4.13.1) and SEQ ID NO:21 (VL 4.13.1), respectively;
and the
amino acid sequences set forth in SEQ ID NO:27 (VH 11.2.1) and SEQ ID NO:33
(VL 11.2.1),
respectively. Most preferably, the antibody is CP-675,206, which has the heavy
and light
chain amino acid sequences of antibody 11.2.1 (ticilimumab).
In yet another embodiment, the amino acid sequence of the heavy chain
comprises
the amino acid sequence encoded by a nucleic acid comprising the nucleic acid
sequences
set forth in SEQ ID NOs:1, 13, and 25. In yet another embodiment, the light
chain comprises
the amino acid sequence encoded by a nucleic acid comprising the nucleic acid
sequences
set forth in SEQ ID NOs:7, 19 and 31. More preferably, the heavy and light
chains comprise
the amino acid sequences encoded by nucleic acids comprising the nucleic acid
sequences
set forth in SEQ ID N0:1 (heavy chain 4.1.1) and SEQ ID N0:7 (light chain
4.1.1),
respectively; the nucleic acid sequences set forth in SEQ ID N0:13 (heavy
chain 4.13.1) and
SEQ ID N0:19 (light chain 4.13.1), respectively; and the nucleic acid
sequences set forth in
SEQ ID N0:25 (heavy chain 11.2.1) and SEQ ID N0:31 (light chain 11.2.1),
respectively.
Furthermore, the antibody can comprise a heavy chain amino acid sequence
comprising human CDR amino acid sequences derived from the VH 3-30 or 3-33
gene, or
conservative substitutions or somatic mutations therein. It is understood that
the VH 3-33 gene
encodes from FRI through FR3 of the heavy chain variable region of an antibody
molecule.
Thus, the invention encompasses an antibody that shares at least 85%, more
preferably, at
least 90%, yet more preferably, at least 91 %, even more preferably, at least
94%, yet more
preferably, at least 95%, more preferably, at least 97%, even more preferably,
at least 98%,
yet more preferably, at least 99%, and most preferably, 100% identity, with
the sequence from
FRI through FR3 of an antibody selected from the group consisting of 3.1.1,
4.1.1, 4.8.1,
4.10.2, 4.13.1, 4.14.3, 6.1.1, 11.2.1 (CP-675,206), 11.6.1, 11.7.1, 12.3.1.1,
2.9.1.1,
ipilimumab, and DP-50.
The antibody can further comprise CDR regions in its light chain derived from
the
A27 or the 012 gene or it may comprise the CDR regions of an antibody selected
from the
group consisting of 3.1.1, 4.1.1, 4.8.1, 4.10.2, 4.13.1, 4.14.3, 6.1.1, 11.2.1
(CP-675,206),
11.6.1, 11.7.1, 12.3.1.1, 2.9.1.1, ipilimumab.
In other embodiments of the invention, the antibody inhibits binding between
CTLA4
and B7-1, B7-2, or both. Preferably, the antibody can inhibit binding with B7-
1 with an IC50 of
about 100 nM or lower, more preferably, about 10 nM or lower, for example
about 5 nM or
lower, yet more preferably, about 2 nM or lower, or even more preferably, for
example, about
1 nM or lower. Likewise, the antibody can inhibit binding with B7-2 with an
IC50 of about 100


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 38-

nM or lower, more preferably, 10 nM or lower, for example, even more
preferably, about 5 nM
or lower, yet more preferably, about 2 nM or lower, or even more preferably,
about 1 nM or
lower.
Further, in another embodiment, the anti-CTLA4 antibody has a binding affinity
for
CTLA4 of about 10-8, or greater affinity, more preferably, about 10-9 or
greater affinity, more
preferably, about 10-10 or greater affinity, and even more preferably, about
10-11 or greater
affinity.
The anti-CTLA4 antibody includes an antibody that competes for binding with an
antibody having heavy and light chain amino acid sequences of an antibody
selected from the
group consisting of 4.1.1, 6.1.1, 11.2.1 (CP-675,206), 4.13.1 and 4.14.3.
Further, the anti-
CTLA4 antibody can compete for binding with antibody ipilimumab.
In another embodiment, the antibody preferably cross-competes with an antibody
having a heavy and light chain sequence, a variable heavy and a variable light
chain
sequence, and/or the heavy and light CDR sequences of antibody 4.1.1, 4.13.1,
4.14.3, 6.1.1.
or 11.2.1 (CP-675,206). For example, the antibody can bind to the epitope to
which an
antibody that has heavy and light chain amino acid sequences, variable
sequences and/or
CDR sequences, of an antibody selected from the group consisting of 4.1.1,
4.13.1, 4.14.3,
6.1.1, or 11.2.1 (CP-675,206) binds. In another embodiment, the antibody cross-
competes
with an antibody having heavy and light chain sequences, or antigen-binding
sequences, of
MDX-D010.
In another embodiment, the invention is practiced using an anti-CTLA4 antibody
that
comprises a heavy chain comprising the amino acid sequences of CDR-1, CDR-2,
and CDR-
3, and a light chain comprising the amino acid sequences of CDR-1, CDR-2, and
CDR-3, of,
an antibody selected from the group consisting of 3.1.1, 4.1.1, 4.8.1, 4.10.2,
4.13.1, 4.14.3,
6.1.1, 11.2.1 (CP-675,206), 11.6.1, 11.7.1, 12.3.1.1, and 12.9.1.1, or
sequences having
changes from the CDR sequences selected from the group consisting of
conservative
changes, wherein the conservative changes are selected from the group
consisting of
replacement of nonpolar residues by other nonpolar residues, replacement of
polar charged
residues other polar uncharged residues, replacement of polar charged residues
by other
polar charged residues, and substitution of structurally similar residues; non-
conservative
substitutions, wherein the non-conservative substitutions are selected from
the group
consisting of substitution of polar charged residue for polar uncharged
residues and
substitution of nonpolar residues for polar residues, additions and deletions.
In a further embodiment of the invention, the antibody contains fewer than 10,
7, 5, or
3 amino acid changes from the germline sequence in the framework or CDR
regions. In
another embodiment, the antibody contains fewer than 5 amino acid changes in
the


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 39-

framework regions and fewer than 10 changes in the CDR regions. In one
preferred
embodiment, the antibody contains fewer than 3 amino acid changes in the
framework
regions and fewer than 7 changes in the CDR regions. In a preferred
embodiment, the
changes in the framework regions are conservative and those in the CDR regions
are somatic
mutations.
In another embodiment, the antibody shares at least 80%, more preferably, at
least
85%, even more preferably, at least 90%, yet more preferably, at least 94%,
preferably, at
least 95%, more preferably, at least 99%, sequence(e.g., amino acid, nucleic
acid, or both)
identity or sequence similarity over the heavy and light chain full-length
sequences, or over
the heavy or the light chain, separately, with the sequences of antibody
3.1.1, 4.1.1, 4.8.1,
4.10.2, 4.13.1, 4.14.3, 6.1.1, 11.2.1 (CP-675,206), 11.6.1, 11.7.1, 12.3.1.1,
12.9.1.1,
ipilimumab. Even more preferably, the antibody shares 100% sequence identity
or sequence
similarity over the heavy chain and the light chain, or with the heavy chain
or the light chain,
separately, of an antibody selected from antibody 3.1.1, 4.1.1, 4.8.1, 4.10.2,
4.13.1, 4.14.3,
6.1.1, 11.2.1 (CP-675,206), 11.6.1, 11.7.1, 12.3.1.1, 12.9.1.1, ipilimumab.
In yet another embodiment, the antibody has heavy and light chain regions
having the
amino acid sequences of ipilimumab (previously known as MDX-01 0).
In another embodiment, the antibody shares at least 80%, more preferably, at
least
85%, even more preferably, at least 90%, yet more preferably, at least 94%,
more preferably,
at least 95%, even more preferably, at least 99%, sequence identity or
sequence similarity
over the heavy and light chain full-length sequences, or over the heavy or the
light chain,
separately, with the sequences of germline V, A27, germline V,r 012, and
germline DP50
(which is an allele of the VH 3-33 gene locus). Even more preferably, the
antibody shares
100% sequence identity or sequence similarity over the heavy chain sequence of
germline
DP50 and/or with the light chain sequence of germline A27, or germline 012.
In one embodiment, the antibody shares at least 80%, more preferably, at least
85%,
even more preferably, at least 90%, yet more preferably, at least 94%,
preferably, at least
95%, more preferably, at least 99%, sequence(e.g., amino acid, nucleic acid,
or both) identity
or sequence similarity over the heavy and light chain variable region
sequences, or over the
heavy or the light chain variable region sequence, separately, with the
sequences of antibody
3.1.1, 4.1.1, 4.8.1, 4.10.2, 4.13.1, 4.14.3, 6.1.1, 11.2.1 (CP-675,206),
11.6.1, 11.7.1, 12.3.1.1,
12.9.1.1, ipilimumab. Even more preferably, the antibody shares 100% sequence
identity or
sequence similarity over the heavy chain and the light chain variable region
sequences, or
with the heavy chain or the light chain sequence, separately, of an antibody
selected from
antibody 3.1.1, 4.1.1, 4.8.1, 4.10.2, 4. 13.1, 4.14.3, 6.1.1, 11.2.1 (CP-
675,206), 11.6.1, 11.7.1,
12.3.1.1, 12.9.1.1, ipilimumab.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 40-

In another embodiment, the antibody shares at least 80%, more preferably, at
least
85%, even more preferably, at least 90%, yet more preferably, at least 94%,
more preferably,
at least 95%, even more preferably, at least 99%, sequence identity or
sequence similarity
over heavy chain variable region sequence with the heavy chain variable
sequence of heavy
germline DP50 (which is an allele of the VH 3-33 gene locus) or with the light
chain variable
sequence of germline Vx A27, or germline V,,012. Even more preferably, the
antibody heavy
chain region sequence shares 100% sequence identity or sequence similarity
with the
sequence of germline DP50 or with the light chain sequence of germline A27, or
germline
012.
In one embodiment of the present invention, the antibody shares at least 80%,
more
preferably, at least 85%, even more preferably, at least 90%, yet more
preferably, at least
95%, more preferably, at least 99%, sequence identity or sequence similarity
with the heavy
chain, the light chain, or both, sequences from FRI through FR4 with the FR1
through FR4
region sequences of antibody 3.1.1, 4.1.1, 4.8.1, 4.10.2, 4.13.1, 4.14.3,
6.1.1, 11.2.1 (CP-
675,206), 11.6.1, 11.7.1, 12.3.1.1, 12.9.1.1, ipilimumab. Even more
preferably, the antibody
shares 100% sequence identity or sequence similarity over the heavy, light, or
both,
sequences from FRI through FR4 with antibody 3.1.1, 4.1.1, 4.8.1, 4.10.2,
4.13.1, 4.14.3,
6.1.1, 11.6.1, 11.7.1, 12.3.1.1, 12.9.1.1, CP-675,206, and ipilimumab.
In another embodiment of the present invention, the antibody shares at least
80%,
more preferably, at least 85%, even more preferably, at least 90%, yet more
preferably, at
least 95%, more preferably, at least 99%, and most preferably, about 100%,
sequence
identity or sequence similarity with the heavy chain sequences from FRI
through FR3 with
the FRI through FR3 region sequences of germline DP50.
In yet another embodiment of the present invention, the antibody shares at
least 80%,
more preferably, at least 85%, even more preferably, at least 90%, yet more
preferably, at
least 95%, more preferably, at least 99%, and most preferably, about 100%,
sequence
identity or sequence similarity with the light chain sequences from FRI
through FR4 with the
FRI through FR4 region sequences of germline VK A27, or germline V1e 012.
In one embodiment of the present invention, the antibody shares at least 80%,
more
preferably, at least 85%, even more preferably, at least 90%, yet more
preferably, at least
95%, more preferably, at least 99%, sequence identity or sequence similarity
with the heavy
chain, the light chain, or both, CDR-1, CDR-2 and CDR-3 sequences of antibody
3.1.1, 4.1.1,
4.8.1, 4.10.2, 4.13.1, 4.14.3, 6.1.1, 11.2.1 (CP-675,206), 11.6.1, 11.7.1,
12.3.1.1, 12.9.1.1,
ipilimumab. Even more preferably, the antibody shares 100% sequence identity
or sequence
similarity over the heavy, light, or both, CDR-1, CDR-2 and CDR-3 sequences
with antibody


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-41-
3.1.1, 4.1.1, 4.8.1, 4.10.2, 4.13.1, 4.14.3, 6.1.1, 11.6.1, 11.7.1, 12.3.1.1,
12.9.1.1, CP-
675,206, and ipilimumab.
In another embodiment of the present invention, the antibody shares at least
80%,
more preferably, at least 85%, even more preferably, at least 90%, yet more
preferably, at
least 95%, more preferably, at least 99%, and most preferably, about 100%,
sequence
identity or sequence similarity with the heavy chain CDR-1 and CDR-2 sequences
with the
CDR-1 and CDR-2 sequences of germline DP50.
In yet another embodiment of the present invention, the antibody shares at
least 80%,
more preferably, at least 85%, even more preferably, at least 90%, yet more
preferably, at
least 95%, more preferably, at least 99%, and most preferably, about 100%,
sequence
identity or sequence similarity with the light chain CDR-1, CDR-2 and CDR-3
sequences with
the CDR-1, CDR-2 and CDR-3 sequences of germline V,, A27, or germline V, 012.
Examples of antibodies employable in the present invention, and methods of
producing
them, are described in, among others, U.S. Patent Application No. 09/472,087,
now issued as
U.S. Patent No. 6,682,736; Int. Appl. No. PCT/US00/23356 (published March 1,
2001, as WO
01/14424) (e.g., antibody ipilimumab, also known as MDX-010, Medarex,
Princeton, NJ); Int.
Appl. No. PCT/US99/28739 (published June 8, 2000, as WO 00/32231); U.S. Pat.
Nos.
5,811,097, 5,855,887, 6,051,227, and 6,207,156; each of which is incorporated
by reference
herein. While information on the amino and nucleic acid sequences relating to
these antibodies
is provided herein, further information can be found in U.S. Patent No.
6,682,736, as well as
WO 00/37504; the sequences set forth in those applications are hereby
incorporated herein by
reference.
Certain uses for these antibodies to treat various cancers were discussed in
U.S.
Patent Application No. 10/153,382, now published as U.S. Patent Application
Publication No.
2003/0086930, which is incorporated by reference as if set forth in its
entirety herein.
Characteristics of human anti-CTLA4 antibodies useful in the methods of the
invention
are extensively discussed in, e.g., U.S. Patent No. 6,682,736, and include
antibodies having
amino acid sequences of an antibody such as, but not limited to, antibody
3.1.1, 4.1.1, 4.8.1,
4.10.2, 4.13.1, 4.14.3, 6.1.1, 11.6.1, 11.7.1, 12.3.1.1, 12.9.1.1, CP-675,206,
and ipilimumab.
The invention also relates to methods using antibodies comprising the amino
acid sequences of
the CDRs of the heavy and light chains of these antibodies, as well as those
comprising
changes in the CDR regions, as described in the above-cited applications and
patent. The
invention also concerns antibodies comprising the variable regions of the
heavy and light chains
of those antibodies. In another embodiment, the antibody is selected from an
antibody
comprising the full length, variable region, or CDR, amino acid sequences of
the heavy and light


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 42-

chains of antibodies 3.1.1, 4.1.1, 4.8.1, 4.10.2, 4.13.1, 4.14.3, 6.1.1,
11.6.1, 11.7.1, 12.3.1.1,
and 12.9.1.1, CP-675,206, and ipilimumab.
While the anti-CTLA4 antibodies discussed previously herein may be preferred,
the
skilled artisan, based upon the disclosure provided herein, would appreciate
that the invention
encompasses a wide variety of anti-CTLA4 antibodies and is not limited to
these particular
antibodies. More particularly, while human antibodies are preferred, the
invention is in no
way limited to human antibodies; rather, the invention encompasses useful
antibodies
regardless of species origin, and includes, among others, chimeric, humanized
and/or
primatized antibodies. Also, although the antibodies exemplified herein were
obtained using
a transgenic mammal, e.g., a mouse comprising a human immune repertoire, the
skilled
artisan, based upon the disclosure provided herein, would understand that the
present
invention is not limited to an antibody produced by this or by any other
particular method.
Instead, the invention includes an anti-CTLA4 antibody produced by any method,
including,
but not limited to, a method known in the art (e.g:, screening phage display
libraries, and the
like) or to be developed in the future for producing an anti-CTLA4 antibody of
the invention.
Based upon the extensive disclosure provided herein and in, e.g., U.S. Patent
No. 6,682,736,
to Hanson et al., and U.S. Pat. App. Pub. No. 2002/0088014, one skilled in the
art can readily
produce and identify an antibody useful for treatment of prostate cancer in
combination with a
hormonal therapeutic agent using the novel methods disclosed herein.
The present invention encompasses human antibodies produced using a transgenic
non-human mammal, f.e., XenoMouseT"' (Abgenix, Inc., Fremont, CA) as disclosed
in the
U.S. 6,682,736, to Hanson et al.
Another transgenic mouse system for production of "human" antibodies is
referred to as
"HuMAb-MouseTM" (Medarex, Princeton, NJ), which contains human immunoglobulin
gene
miniloci that encodes unrearranged human heavy (mu and gamma) and kappa light
chain
immunoglobulin sequences, together with targeted mutations that inactivate the
endogenous
mu and kappa chain loci (Lonberg et al. Nature 368:856-859 (1994), and U.S.
Pat. No.
5,770,429).
However, the invention uses human anti-CTLA4 antibodies produced using any
transgenic mammal such as, but not limited to, the Kirin TC MouseTm (Kirin
Beer Kabushiki
Kaisha, Tokyo, Japan) as described in, e.g., Tomizuka et al., Proc Natl Acad
Sci USA 97:722
(2000); Kuroiwa et al., Nature Biotechnol 18:1086 (2000); U.S. Patent
Application Publication
No. 2004/0120948, to Mikayama et al.; and the HuMAb-MouseT"" (Medarex,
Princeton, NJ)
and XenoMouseT"' (Abgenix, Inc., Fremont, CA), supra. Thus, the invention
encompasses
using an anti-CTLA4 antibody produced using any transgenic or other non-human
animal.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 43-

In another embodiment, the antibodies employed in methods of the invention are
not
fully human, but "humanized". In particular, murine antibodies or antibodies
from other species
can be "humanized" or "primatized" using techniques well known in the art.
See, e.g., Winter
and Harris Immunol. Today 14:43-46 (1993), Wright et al. Crit. Reviews in
Immuno/.12:125-168
(1992), and US Patent No. 4,816,567, to Cabilly et al, and Mage and Lamoyi in
Monoclonal
Antibody Production Techniques and Applications pp. 79-97, Marcel Dekker,
Inc., New York,
NY (1987). Thus, humanized, chimeric antibodies, anti-CTLA4 antibodies derived
from any
species (including single chain antibodies obtained from camelids as described
in, e.g., U.S.
Pat. Nos. 5,759,808 and 6,765,087, to Casterman and Hamers), as well as any
human
antibody, can be combined with a therapeutic agent to practice the novel
methods disclosed
herein.
As will be appreciated based upon the disclosure provided herein, antibodies
for use in
the invention can be obtained from a transgenic non-human mammal, and
hybridomas derived
therefrom, but can also be expressed in cell lines other than hybridomas.
Mammalian cell lines available as hosts for expression are well known in the
art and
include many immortalized cell lines available from the American Type Culture
Collection
(ATCC), including but not limited to Chinese hamster ovary (CHO) cells, NSO,
Sp2, HEK, HeLa
cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), and human
hepatocellular
carcinoma cells (e.g., Hep G2). Non-mammalian prokaryotic and eukaryotic cells
can also be
employed, including bacterial, yeast, insect, and plant cells.
Nucleic acid molecules encoding an anti-CTLA4 antibody, and expression vectors
comprising these nucleic acid molecules, may be used for transfection of a
suitable
mammalian, plant, bacterial or yeast host cell. Transformation may be by any
known method
for introducing polynucleotides into a host cell. Methods for introduction of
heterologous
polynucleotides into mammalian cells are well known in the art and include
dextran-mediated
transfection, calcium phosphate precipitation, polybrene-mediated
transfection, protoplast
fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes,
and direct
microinjection of the DNA into nuclei. In addition, nucleic acid molecules may
be introduced
into mammalian cells by viral vectors. Methods of transforming plant cells are
well known in
the art, including, e.g., Agrobacterium-mediated transformation, biolistic
transformation, direct
injection, electroporation and viral transformation. Methods of transforming
bacterial and
yeast cells are also well known in the art.
An expression vector may also be delivered to an expression system using DNA
biolistics, wherein the plasmid is precipitated onto microscopic particles,
preferably gold, and
the particles are propelled into a target cell or expression system. DNA
biolistics techniques
are well-known the art and devices, e.g., a "gene gun", are commercially
available for delivery


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 44-

of the microparticles in to a cell (e.g., Helios Gene Gun, Bio-Rad Labs.,
Hercules, CA) and
into the skin (PMED Device, PowderMed Ltd., Oxford, UK).
Various expression systems can be used as well known in the art, such as, but
not
limited to, those described in, e.g., Sambrook and Russell, Molecular Cloning,
A Laboratory
Approach, Cold Spring Harbor Press, Cold Spring Harbor, NY (2001), and Ausubel
et al.,
Current Protocols in Molecular Biology, John Wiley & Sons, NY (2002). These
expression
systems include dihydrofolate reductase (DHFR)-based systems, among many
others. The
glutamine synthetase system of expression is discussed in whole or part in
connection with
European Patents No. 0216846131, No. 0256055B1, and No. 0323997B1, and
European Patent
Application No. EP89303964. In one embodiment, the antibody used is made in
NSO cells
using a glutamine synthetase system (GS-NSO). In another embodiment, the
antibody is made
in CHO cells using a DHFR system. Both systems are well-known in the art and
are described
in, among others, Barnes et al. Biotech & Bioengineering 73:261-270 (2001),
and references
cited therein.
Site directed mutagenesis of the antibody CH2 domain to eliminate
glycosylation may
be preferred in order to prevent changes in either the immunogenicity,
pharmacokinetic, and/or
effector functions resulting from non-human glycosylation. Further, the
antibody can be
deglycosylated by enzymatic (see, e.g., Thotakura et al. Meth. Enzymol.
138:350 (1987)) and/or
chemical methods (see, e.g., Hakimuddin et al., Arch. Biochem. Biophys. 259:52
(1987)).
Further, the invention encompasses using an anti-CTLA4 antibody comprising an
altered glycosylation pattern. The skilled artisan would appreciate, based
upon the disclosure
provided herein, that an anti-CTLA4 antibody can be modified to comprise
additional, fewer, or
different glycosylations sites compared with the naturally-occurring antibody.
Such
modifications are described in, e.g., U.S. Patent Application Publication Nos.
2003/0207336,
and 2003/0157108, and International Patent Publication Nos. WO 01/81405 and
00/24893.
Additionally, the invention comprises using an anti-CTLA4 antibody regardless
of the
glycoform, if any, present on the antibody. Moreover, methods for extensively
remodeling the
glycoform present on a glycoprotein are well-known in the art and include,
e.g., those described
in International Patent Publication Nos. WO 03/031464, WO 98/58964, and WO
99/22764, and
US Patent Application Publication Nos. 2004/0063911, 2004/0132640,
2004/0142856,
2004/0072290, and US Patent No. 6,602,684 to Umana et al.
Further, the invention encompasses using an anti-CTLA4 antibody with any art-
known
covalent and non-covalent modification, including, but not limited to, linking
the polypeptide to
one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol,
polypropylene glycol,
or polyoxyalkylenes, in the manner set forth in, for example, U.S. Patent
Application Publication


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 45-

Nos. 2003/0207346 and 2004/0132640, and U.S. Pat. Nos. 4,640,835; 4,496,689;
4,301,144;
4,670,417; 4,791,192; 4,179,337.
Additionally, the invention encompasses using an anti-CTLA4 antibody, or
antigen-
binding portion thereof, chimeric protein comprising, e.g., a human serum
albumin polypeptide,
or fragment thereof. Whether the chimeric protein is produced using
recombinant methods by,
e.g., cloning of a chimeric nucleic acid encoding the chimeric protein, or by
chemical linkage of
the two peptide portions, the skilled artisan would understand once armed with
the teachings
provided herein that such chimeric proteins are well-known in the art and can
confer desirable
biological properties such as, but not limited to, increased stability and
serum half-life to the
antibody of the invention and such molecules are therefore included herein.
Antibodies that are generated for use in the invention need not initially
possess a
particular desired isotype. Rather, the antibody as generated can possess any
isotype and can
be isotype switched thereafter using conventional techniques. These include
direct
recombinant techniques (see, e.g., U.S. Patent 4,816,397), and cell-cell
fusion techniques (see
e.g., U.S. Patent No. 5,916,771).
The effector function of the antibodies of the invention may be changed by
isotype
switching to an IgG1, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM for various
therapeutic uses.
Furthermore, dependence on complement for cell killing can be avoided through
the use of
bispecifics, immunotoxins, or radiolabels, for example.
Although antibody 4.1.1, 4.13.1 and 11.2.1 (CP-675,206) are IgG2 antibodies
and the
sequences of the variable regions of the antibodies are provided herein
(Figures 1-3), and in
the applications and patents referenced and incorporated herein, it is
understood that the full-
length sequences of these antibodies are encompassed herein, as well as the
use of any
antibody comprising the sequences set forth in SEQ ID NQs:1-36, and further
comprising any
constant region, regardless of isotype as more fully discussed elsewhere
herein. Likewise,
any antibody comprising the full-length sequence of ipilimumab, or any portion
thereof,
including a sequence encoding an antigen-binding portion of ipilimumab, can be
used
according to the methods of the invention.
Thus, the skilled artisan, once provided with the teachings provided herein,
would
readily appreciate that the anti-CTLA4 antibody-therapeutic agent combination
of the
invention can comprise a wide plethora of anti-CTLA4 antibodies.
Further, one skilled in the art, based upon the disclosure provided herein,
would
understand that the invention is not limited to administration of only a
single antibody; rather,
the invention encompasses administering at least one anti-CTLA4 antibody,
e.g., one of 4.1.1,
4.13.1, or 11.2.1 (CP-675,206), in combination with a therapeutic agent.
Further, any


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 46-

combination of anti-CTLA4 antibodies can be combined with at least one
therapeutic agent
and the present invention encompasses any such combination and permutation
thereof.

IV. Therapeutic Acients
The present invention relates to combinations comprising anti-CTLA4 antibody
and at
least one therapeutic agent, which may be further combined with additional
agents and/or
therapeutic modalities, e.g., chemotherapy, surgery, radiotherapy,
transplantation, and the
like, to treat cancer. That is, the patient may be subjected to additional
chemotherapy with
agents well-known, such as, but not limited to, growth factor inhibitors,
biological response
modifiers, alkylating agents, intercalating antibiotics, vinca alkaloids,
immunomodulators,
taxanes, platinum compounds, signal transduction inhibitors, selective
estrogen receptor
modulators (SERMs), such as, but not limited to, lasofoxifene, and
angiogenesis inhibitors.
Co-administration of the antibody with an additional therapeutic agent
(combination
therapy) encompasses administering a pharmaceutical composition comprising
both the anti-
CTLA4 antibody and one or more additional therapeutic agents, and
administering two or
more separate pharmaceutical compositions, one comprising the anti-CTLA4
antibody and
the other(s) comprising the additional therapeutic agent(s). Further, although
co-
administration or combination (conjoint) therapy generally mean that the
antibody and
additional therapeutic agents are administered at the same time as one
another, it also
encompasses simultaneous, sequential or separate dosing of the individual
components of
the treatment. Additionally, where an antibody is administered intravenously
and the anti-
cancer agent is administered orally (e.g., imatinib mesylate, gemcitabine,
capecitabine,
temozolomide, and the like), it is understood that their combination is
preferably administered
as two separate pharmaceutical compositions.
Therapeutic agents are numerous and have been described in, for instance, U.S.
Patent Application Publication No. 200410005318, No. 2003/0086930, No.
2002/0086014, and
International Publication No. WO 03/086459, all of which are incorporated by
reference
herein, among many others. Such therapeutic agents include, but are not
limited to,
topoisomerase I inhibitors; other antibodies (rituximab, bevacizumab,
trastuzumab, anti-IGF
1R antibody [e.g., CP-751,871], anti-CD40 antibody [e.g., CP-870,893], and the
like);
chemotherapeutic agents such as, but not limited to, imatinib (GLEEVEC),
SU11248
(SUTENT; sunitinib), SU12662, SU14813; BAY 43-9006, AG-013736 (axitinib),
immunomodulators, including toll-like receptor agonists (e.g., TLR-9 agonist;
such as, but not
limited to, CPG-7909, also referred to as PF03512676 or PROMUNE), and other
immunomodulators, e.g., indoleamine-2,3,-dioxygenase (IDO) inhibitors;
selective estrogen
receptor modulators (SERMs; e.g., lasofoxifene); taxanes; vinca alkaloids;
temozolomide;


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 47-

angiogenesis inhibitors; EGFR inhibitors; VEGF inhibitors; erbB2 receptor
inhibitors; anti-
proliferative agents (e.g., farnesyl protein transferase inhibitors, and avR3
inhibitors, av(35
inhibitors, p53 inhibitors, and the like); immunomodulators; biological
response modifiers;
cytokines; tumor vaccines; tumor-specific antigens; heat shock protein-based
tumor vaccines;
dendritic and stem cell therapies; alkylating agents; folate antagonists;
pyrimidine antagonists;
anthracycline antibiotics; platinum compounds; immune costimulatory molecules
(e.g., CD4,
CD25, PD-1, B7-H3, 4-1BB, OX40, ICOS, CD30, HLA-DR, MHCII, and LFA), and
agonist
antibodies thereto; among many others.
In one embodiment, the methods of the invention may be further combined with
transplantation, e.g., stem cell transplantation, to provide a therapeutic
benefit to a patient
afflicted with breast cancer. Stem cell transplantation may be performed
according to the
methods known in the art and may be allogeneic or autologous stem cell
transplantation.
Additionally, one skilled in the art would appreciate, based upon the
disclosure provided
herein, that transplantation encompasses adoptive transfer of lymphocytes,
either autologous
or obtained from an HLA-matched donor. Where the method comprises stem cell
transplant,
the first dose of the antibody-AI therapy agent combination can be
administered after the
immune system of the mammal has recovered from transplantation, for example,
in the period
of from one to 12 months post transplantation. In certain embodiments, the
first dose is
administered in the period of from one to three, or one to four months post
transplantation.
Transplantation methods are described many treatises, including Appelbaum in
Harrison's
Principles of Internal Medicine, Chapter 14, Braunwald et al., Eds., 15th ed.,
McGraw-Hill
Professional (2001), which is hereby incorporated herein by reference.
As pointed out previously herein, there are many chemotherapeutic agents
currently
available for the treatment of tumors that are suitable for use in the
combination therapy of the
present invention. For example, alkylating agents are a class of drugs that
alkylate DNA,
restricting uncoiling and replication of strands. A preferred alkylating agent
for use in the
methods of the present invention is cyclophosphamide (CYTOXAN). In one
embodiment, CP-
675,206 is administered with low-dose cyclophosphamide. Without wishing to be
bound by
any particular theory, this is because such dose may mediate depletion of
Tregs and because
CTLA4 blockade appears not to affect Tregs such that these two anti-tumor
mechanisms may
provide a synergistic therapeutic effect.
Folate antagonists bind to dihydrofolate reductase (DHFR) and interfere with
pyrimidine (thymidine) synthesis. Methotrexate and pemetrexed (ALIMTA) are
folate
antagonists suitable for use in the methods of the present invention. In
addition to DHFR,
pemetrexed also inhibits thymidylate synthase and glycinamide ribonucleotide
formyl
transferase, two other folate-dependant enzymes involved in thymidine
synthesis.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 48-

Pyrimidine antagonists inhibit enzymes involved in pyrimidine synthesis. As
pyrimidine analogs, they also interfere with DNA production by competing with
normal
nucleotides for incorporation into the DNA molecule. Pyrimidine antagonists
suitable for use
in the methods of the present invention include 5-fluorouracil (5-FU);
capecitabine (XELODA),
a prodrug of 5'-deoxy-5-fluorouridine (5'-FDUR), which is enzymatically
converted to 5-FU in
vivo; and gemcitabine (GEMZAR).
Anthracycline antibiotics inhibit the uncoiling of DNA by intercalation
between DNA
strands. Anthracycline antibiotics include doxorubicin hydrochloride
(ADRIAMYCIN),
epirubicin hydrochloride (ELLENCE, PHARMORUBICIN), daunorubicin (CERUBIDINE,
DAUNOXOME), and idarubicin hydrochloride (IDAMYCIN PFS, ZAVEDOS). Preferred
anthracyclines for use with the present invention include doxorubicin and
epirubicin.
Platinum compounds exert their anti-neoplastic effect by intercalation and
intracalation between DNA strands, which inhibits uncoiling of the DNA.
Platinum compounds
useful in the methods of the present invention include cisplatin (PLATINOL),
oxaliplatin
(ELOXATIN), and carboplatin (PARAPLATIN).
Taxanes promote assembly of microtubules while inhibiting their disassembly
into
tubulin, thereby blocking a cell's ability to break down the mitotic spindle
during mitosis. They
have demonstrated significant activity against many solid tumors as single
agent therapy and
in combination with other chemotherapy agents. One embodiment of the
combination therapy
of the present invention includes the use of one or more taxanes in
combination with an IGF-
I R antibody. Suitable taxanes for use in combination with the IGF-1 R
antibody include
docetaxel (TAXOTERE) and paclitaxel (TAXOL).
Taxane-derivatives, which may be active in cells resistant to doxorubicin,
vinblastine,
paclitaxel, docetaxel, and the like, include XRP-9981 (Sanofi Aventis), and
are encompassed
in the invention.
Vinca alkaloids, like taxanes, are "spindle poisons," acting on the
microtubuies that
form the mitotic spindle. They inhibit mitosis by interfering with microtubule
assembly,
keeping the spindle from being formed. Vinca alkaloids include vindesine
(ELDISINE),
vinblastine sulfate (VELBAN), vincristine sulfate (ONCOVIN) and vinorelbine
tartrate
(NAVELBINE). A preferred vinca alkaloid for use in the methods of the present
invention is
vinorelbine.
BMS-247550 (ixabepilone) promotes tubulin polymerization and microtubule
stabilization, thereby arresting cells in the G2-M phase and inducing tumor
cell apoptosis.
This agent demonstrates activity against taxane-resistant cells.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 49-

Analogs of rapamycin, which bind and inhibit the mammalian target of rapamycin
(mTOR), are also useful and include, among others, CCI-779 (temsirolimus;
Wyeth) and
RAD-001 (everolimus, CERTICAN; Novartis).
The camptothecin analogs act through inhibition of topoisomerase I, an enzyme
critical for DNA replication and packaging. Levels of topoisomerase I are
higher in tumor cells
than in normal tissue. A camptothecin analog useful in the methods of the
present invention
is irinotecan (CAMPTOSAR). Topoisomerase I inhibitors useful in the
embodiments of the
present invention include 9-aminocamptothecin, belotecan, BN-80915 (Roche),
camptothecin,
diflomotecan, edotecarin, exatecan (Daiichi), gimatecan, 10-
hydroxycamptothecin, lurtotecan,
Orathecin (rubitecan, Supergen), SN-38, topotecan, and combinations thereof.
Camptothecin derivatives are of particular interest in the combination
embodiments of
the invention and include camptothecin, 10-hydroxycamptothecin, 9-
aminocamptothecin,
irinotecan, SN-38, edotecarin, topotecan and combinations thereof.
A particularly preferred topoisomerase I inhibitor is irinotecan (CAMPTOSAR)
Topoisomerase II inhibitors useful in the combination embodiments of the
present
invention include aclarubicin, adriamycin, amonafide, amrubicin, annamycin,
daunorubicin,
doxorubicin, elsamitrucin, epirubicin, etoposide, idarubicin, galarubicin,
hydroxycarbamide,
nemorubicin, novantrone (mitoxantrone), pirarubicin, pixantrone, procarbazine,
rebeccamycin, sobuzoxane, tafluposide, valrubicin, and Zinecard (dexrazoxane).
Particularly preferred toposimerase II inhibitors include epirubicin
(Ellence), doxorubicin,
daunorubicin, idarubicin and etoposide.
Alkylating agents that may be used in the embodiments of the invention
include, but
are not limited to, nitrogen mustard N-oxide, cyclophosphamide, AMD-473,
altretamine, AP-
5280, apaziquone, brostallicin, bendamustine, busulfan, carboquone,
carmustine,
chlorambucil, dacarbazine, estramustine, fotemustine, glufosfamide,
ifosfamide, KW-2170,
lomustine, mafosfamide, mechlorethamine, melphalan, mitobronitol, mitolactol,
mitomycin C,
mitoxatrone, nimustine, ranimustine, temozolomide, thiotepa, and platinum-
coordinated
alkylating compounds such as cisplatin, carboplatin (PARAPLATIN), eptaplatin,
lobaplatin,
nedaplatin, oxaliplatin (ELOXATIN, Sanofi), streptozocin, satraplatin and
combinations
thereof.
Antimetabolites that may be used in combination therapy with CTLA4 antibodies,
optionally with one or more other agents include, but are not limited to
dihydrofolate reductase
inhibitors (such as methotrexate and NeuTrexin (trimetresate glucuronate)),
purine antagonists
(such as 6-mercaptopurine riboside, mercaptopurine, 6-thioguanine, cladribine,
clofarabine
(Clolar), fludarabine, nelarabine, and raltitrexed), pyrimidine antagonists
(such as 5-
fluorouracil (5-FU), pemetrexed disodium (Alimta, LY231514, MTA), capecitabine
(Xeloda),


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 50-

cytosine arabinoside, gemcitabine (Gemzar, Eli Lilly), Tegafur (UFT Orzel or
Uforal and
including TS-1 combination of tegafur, gimestat and otostat), doxifluridine,
carmofur,
cytarabine (including ocfosfate, phosphate stearate, sustained release and
liposomal forms),
enocitabine, 5-azacitidine (Vidaza), decitabine, and ethynylcytidine) and
other
antimetabolites such as eflornithine, hydroxyurea, leucovorin, nolatrexed
(Thymitaq), triapine,
trimetrexate,or for example, one of the preferred anti-metabolites disclosed
in European
Patent Application No. 239362 such as N-(5-[N-(3,4-dihydro-2-methyl-4-
oxoquinazolin-6-
ylmethyl)-N-methylamino]-2-thenoyl)-L-glutamic acid and combinations thereof.
In another embodiment the anti-cancer agent is a poly(ADP-ribose) polymerase-1
(PARP-1) inhibitor such as AG-014699, ABT-472, INO-1001, KU-0687 and GPI
18180.
Microtubulin inhibitors that may be used in combination therapy with CTLA4
antibodies, optionally with one or more other agents include, but are not
limited to ABI-007,
Albendazole, Batabulin, CPH-82, EPO 906 (Novartis), discodermolide (XAA-296),
vinfunine
and ZD-6126 (AstraZeneca).
Antibiotics that may be used in combination therapy with CTLA4 antibodies,
optionally
with one or more other agent including, but are not limited to, intercalating
antibiotics such as
actinomycin D, bleomycin, mitomycin C, neocarzinostatin (Zinostatin),
peplomycin, and
combinations thereof.
Plant derived anti-tumor substances (also known as spindle inhibitors) that
may be
used in combination therapy with CP-675,206, but are not limited to, mitotic
inhibitors, for
example vinblastine, vincristine, vindesine, vinorelbine (NAVELBINE),
docetaxel
(TAXOTERE), Ortataxel, paclitaxel (including Taxoprexin a DHA/paciltaxel
conjugate) and
combinations thereof.
Platinum-coordinated compounds include but are not limited to, cisplatin,
carboplatin,
nedaplatin, oxaliplatin (ELOXATIN), Satraplatin (JM-216), and combinations
thereof.
Other agents include alitretinoin, I-asparaginase, AVE-8062 (Aventis),
calcitriol
(Vitamin D derivative), Canfosfamide (Telcyta, TLK-286), Cotara (1311 chTNT
1/b), DMXAA
(Antisoma), exisulind, ibandronic acid, miltefosine, NBI-3001 (IL-4),
pegaspargase, RSR13
(efaproxiral), Targretin (bexarotene), tazarotne (Vitamin A derivative),
Tesmilifene (DPPE),
Theratope, tretinoin, Trizaone (tirapazamine), Xcytrin (motexafin gadolinium)
and Xyotax
(polyglutamate paclitaxel), and combinations thereof.
In another embodiment of the present invention statins may be used in
combination
with CP-675,206. Statins (HMG-CoA reductase inhibitors) may be selected from
the group
consisting of Atorvastatin (Lipitor, Pfizer Inc.), Provastatin (Pravachol,
Bristol-Myers Squibb),
Lovastatin (Mevacor, Merck Inc.), Simvastatin (Zocor, Merck Inc.), Fluvastatin
(Lescol,


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-51-
Novartis), Cerivastatin (Baycol, Bayer), Rosuvastatin (Crestor, AstraZeneca),
Lovostatin and
Niacin (Advicor, Kos Pharmaceuticals), derivatives and combinations thereof.
In a preferred embodiment the statin is selected from the group consisting of
atovorstatin and lovastatin, derivatives and combinations thereof.
Other agents useful as anti-tumor agents include CADUET, Lipitor and
torcetrapib.
In certain embodiments of the invention, the above described methods are
combined
with a cancer vaccine described, e.g., in Rosenberg, S., Development of Cancer
Vaccines,
ASCO Educational Book Spring: 60-62 (2000); Logothetis, C. ASCO Educational
Book
Spring: 300-302 (2000); Khayat, D. 2000, ASCO Educational Book Spring: 414-
428; Foon, K.
2000, ASCO Educational Book Spring: 730-738; see also Restifo, N. and Sznol,
M., in, 1997,
in Cancer: Principles and Practice of Oncology, pp. 3023-3043, Fifth Edition,
DeVita, V. et al.,
eds. (1997), including a vaccine using autologous or allogeneic tumor cells.
Cellular vaccines
have been shown to be effective especially when the tumor cells are transduced
to express
GM-CSF, which is a potent activator of antigen presentation for tumor
vaccination (Dranoff et
al. (1993) Proc. Natl. Acad. Sci U.S.A. 90 (80: 3539-43).
Numerous tumor-specific antigens have been identified to date. The study of
gene
expression and large scale gene expression patterns in various tumors has led
to the
definition of so called tumor specific antigens (Rosenberg, SA Immunity 10:281-
287 (1999)).
In many cases, these tumor specific antigens are differentiation antigens
expressed in the
tumors and in the cell from which the tumor arose, for example melanocyte
antigens gp 100,
MAGE antigens, Trp-2. The tumor antigen may also include the protein
telomerase, which is
required for the synthesis of telomeres of chromosomes and which is expressed
in more than
85% of human cancers and in only a limited number of somatic tissues (Kim, N
et al. Science
266:2011-2013 (1994)). These somatic tissues may be protected from immune
attack by
various means. More importantly, many of these antigens can be shown to be the
targets of
tumor specific T cells found in the host, and such tumor-specific antigens can
be combined
with an anti-CTLA4 antibody to treat cancer by stimulating an anti-tumor
response in the host.
More particularly, CTLA4 inhibition can render these otherwise non-immunogenic
proteins
immunogenic thereby mediating an immune response to the tumor cell expressing
them.
That is, these proteins are normally viewed by the immune system as self
antigens and are
therefore tolerogenic but such tolerization can be overcome by anti-CTLA4
antibodies.
Tumor-specific antigen can also be "neo-antigens" expressed in cancer cells
because
of somatic mutations that alter protein sequence or create fusion proteins
between two
unrelated sequences (i.e. bcr-abl in the Philadelphia chromosome mutation for
CML), or
idiotype from B cell tumors. Other tumor vaccines can include the proteins
from viruses
implicated in human cancers such a Human Papilloma Viruses (HPV), Hepatitis
Viruses (HBV


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 52-

and HCV) and Kaposi's Herpes Sarcoma Virus (KHSV). Regardless of the source of
the
tumor-specific antigen, where it is expressed by a tumor cell, co-
administration of the antigen
with an anti-CTLA4 antibody can render the tumor cell subject to immune attack
thus
providing a therapeutic benefit.
Another form of tumor-specific antigen which can be used in conjunction with
CTLA4
antibody administration is a purified heat shock protein (HSP) isolated from
the tumor tissue.
These heat shock proteins contain fragments of proteins from the tumor cells
and are highly
efficient at delivery to antigen presenting cells for eliciting tumor immunity
(Suot, R &
Srivastava, P Science 269:1585-1588 (1995); Tamura, Y. et al. Science 278:117-
120 (1997).
Thus, administration of an anti-CTLA4 antibody in combination with a tumor HSP
can provide
an important benefit by mediating an immune response to the tumor. HSP-based
tumor
vaccines, and methods for producing the same, include, e.g., ONCOPHAGE (HSPPC-
96),
and AG-858 (HSPPC-70), both from Antigenics (Lexington, MA), among others.
Another therapeutic agent that can be combined with an anti-CTLA4 antibody is
a
dendritic cell (DC). DCs are potent antigen-presenting cells that can be used
to prime
antigen-specific responses. DCs can be produced ex vivo and can be loaded with
various
protein and peptide antigens, as well as with tumor cell extracts (Nestle, F.
et al. Nature
Medicine 4:328-332 (1998)). Where the DCs are loaded with tumor cell extracts,
the precise
nature of the tumor antigen need not be established; rather, the DC cell
processes the tumor
extract and presents the antigen(s) in the context of MHC. Thus, the nature of
the antigen
need not be elucidated and the ability of the DCs to present it can be
exploited. DCs can also
be transduced by genetic means to express desired tumor antigens as well. DCs
have also
been fused directly to tumor cells for the purposes of immunization (Kugler,
A. et al. Nature
Medicine 6:332-336 (2000)). As a method of tumor vaccination, DC immunization
can be
effectively combined with an anti-CTLA4 antibody to mediate a more potent anti-
tumor
response. See also Oh et al., Cancer Res. 64:2610-2618 (2004) (TARP epitopes
and breast
cancer); Kontani et al., Int J Molec Med 12:493-502 (2003) (dendritic cell
vaccine targeting
MUC1 mucin); Holmberg & Sandmaier Expert Rev Vaccines 3:269-277
(2004)(vaccination for
breast or ovarian cancer).
As suggested by data disclosed herein, an immune response to a vaccine-
specific
antigen may be enhanced by coadministration of anti-CTLA4 antibody. Therefore,
the
present invention includes a method of treating cancer comprising
administering to a patient
in need thereof a therapeutically effective amount of an anti-CTLA4 antibody
and a
therapeutically effective amount of at least one therapeutic agent where the
agent comprises
a vaccine and/or antigen, or combination of more than one antigen, or a cell
presenting an
antigen and/or expressing a cytokine, e.g., GM-CSF.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 53-

That is, useful vaccines may be, without limitation, those comprised of breast
cancer-
associated antigens (e.g., HER-2/neu, mammaglobin, prostate and breast tumor-
associated
protein [TARP], MUC1, CEA, sialyl-Tn and other carbohydrate antigens), other
tumor cancer-
associated antigens (e.g.; p53, telomerase), anti-idiotype antibodies such as
11 D10, as well
as vaccines comprising GM-CSF (see, e.g., GVAX, Cell Genesys, Inc., Lapuleucel-
T
[APC8024] and Sipuleucel-T [APC8015], Dendreon Corp.; and TVAX, Geron Corp.),
DNA and
cell-based vaccines, dendritic cell vaccines (reviewed in Banchereau &
Palucka, Nature Revs.
lmmunol. 5:296-306 (2005)), recombinant viral (e.g., vaccinia virus) vaccines,
and heat shock
protein (HSP) vaccines (e.g., vitespen [ONCOPHAGE], Antigenics Inc.).
In one embodiment, the present invention encompasses a combination of CP-
675,206 and a tumor antigen. Tumor antigens or tumor-associated antigens
(TAAs) include,
e.g., cancer-germ cell (CG) antigens (MAGE, NY-ESO-1), mutational antigens
(MUM-1, p53,
CDK-4), over-expressed self-antigens (p53, HER2/NEU), viral antigens
(fromPapilloma Virus,
Epstein-Barr Virus), tumor proteins derived from non-primary open reading
frame mRNA
sequences (NY-ESOI, LAGE1), Melan A, MART-1, MAGE-1, MAGE-3, BAGE, GAGE-1,
GAGE-2, tyrosinase, gp100, gp75, HER-21neu, c-erb-B2, CEA, PSA, MUC-1, CA-125,
Sialyl-
Tn (STn), STn-KLH (THERATOPE, Biomira Inc.), TAG-72, KSA (17-1A), PSMA, p53
(point
mutated and/or overexpressed), RAS (point mutated), EGF-R, VEGF, GD2, GM2,
GD3, Anti-
Id, CD20, CD19, CD22, CD36, Aberrant class II, B1, CD25 30 (IL-2R) (anti-TAC),
or HPV.
TAAs are discussed, for example, in Palena et al., Adv. Cancer Res. 95:115-145
(2006), and
other art-recognized treatises, and include use of gp100 from a non-human
species, e.g.,
mouse gplOO, to increase or enhance an anti-tumor response.
In one embodiment, the TAA comprises at least one characteristic likely to be
related
to generating or enhancing an anti-tumor immune response. Such characteristics
may
include, but are not limited to, prevalence, specificity, immunogenicity, and
necessity for cell
viability or growth. More particularly, prevalence means the antigen is
present in most
patients with a certain type of cancer. In another embodiment, the more types
of cancer cells
that express the antigen compared with expression of the antigen by normal
cells the more
desirable the prevalence would be. In yet another embodiment, the TAA is
specific for the
cancer cells in that the expression of the TAA is significantly greater on a
cancer cell
compared with the level of expression of the TAA on an otherwise identical but
non-cancer
cell, more preferably, the TAA is not detectably expressed on a normal cell
but is expressed
on most, if not all, cancer cells in a patient. Most preferably, the TAA is
unique to a tumor cell
and is not expressed in a normal cell.
In yet another embodiment, the TAA is immunogenic, e.g., it induces a
detectable
immune response to a tumor cell. More specifically, the higher the magnitude
of the


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 54-

response, the more preferred the TAA. Even more preferably, the TAA induces,
increases
and/or prolongs a cellular immune response.
In another embodiment, the TAA is vital to cell growth or survival. That is,
the TAA is
needed by a tumor cell to survive, grow and/or proliferate. Even more
preferably, the TAA is
vital to a tumor cell such that affecting or inhibiting a biological activity
of the TAA prevents or
inhibits cell survival or proliferation. Even more preferably, the TAA is
vital such that inhibiting
or reducing a biological activity of the antigen inhibits development or
selection of a tumor cell
variant not expressing the antigen or a variant cell expressing a mutant
antigen not affected
by the anti-tumor immune response to the original TAA. That is, inducing an
immune
response to the antigen prevents or inhibits cell survival such that a variant
antigen that
avoids the immune response is not selected.
In one embodiment, CP-675,206 is administered to a patient (e.g., a melanoma
patient) previously vaccinated using a tumor vaccine (e.g., a MART-1 antigen),
thereby
inducing, enhancing and/or prolonging an immune response to the vaccine,
thereby providing
a therapeutic benefit to the patient. In one aspect, CP-675,206 can be
administered with at
least one additional therapeutic agent, such as, but not limited to, a
chemotherapeutic agent,
an immunomodulatory agent (a TLR-9 agonist), an immune enhancing antibody
(e.g., an
agonist anti-CD40 antibody), among many additional agents.
In another aspect, a vaccines is be administered prior to, or subsequent to,
administration of the antibody-therapeutic agent combination, and when
chemotherapy is part
of the regimen, a vaccine may be administered prior to chemotherapy. In
certain
embodiments, the antibody-therapeutic agent combination of the invention may
also be
administered prior to chemotherapy. In yet other embodiments, the treatment
can be
combined with stem cell transplant. That is, the antibody-chemotherapeutic
agent
combination can be administered before or after stem cell transplant. A
vaccine may also be
administered before or after stem cell transplantation and, in certain
embodiments,
concomitantly with the antibody.
Stem cell transplantation may be performed according to the methods known in
the
art. Some such methods are described in Appelbaum in Harrison's Principles of
Internal
Medicine, Chapter 14, Braunwald et a/,, Eds., 15th ed., McGraw-Hill
Professional (2001),
which is hereby incorporated herein by reference. Thus, the methods of the
present invention
relate to the treatment of cancer in a mammal who has undergone stem cell
transplantation,
which methods comprise administering to the mammal an amount of a human anti-
CTLA4
antibody in combination with a therapeutic agent, which combination is
effective in treating the
cancer in further combination with stem cell transplantation.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 55-

Where the method comprises stem cell transplant, the first dose of the
antibody-
therapeutic agent combination can be administered after the immune system of
the mammal
has recovered from transplantation, for example, in the period of from one to
12 months post
transplantation. In certain embodiments, the first dose is administered in the
period of from
one to three, or one to four months post transplantation. The patient may
undergo stem cell
transplantation and preparatory treatment(s).
Where an anti-CTLA4 antibody is combined with a standard cancer treatment,
such
as, inter alia, chemotherapeutic regimes, it may be possible to reduce the
dose of
chemotherapeutic reagent administered (Mokyr, M. et al. Cancer Research 58:
5301-5304
(1998)). This is because combined use of an anti-CTLA4 antibody and
chemotherapy can
mediate cell death that is a consequence of the cytotoxic action of most
chemotherapeutic
compounds. Such cell death likely results in increased levels of tumor-
specific antigen in the
antigen presentation pathway, and the anti-CTLA4 antibody mediates an
increased immune
response thereto. Other combination therapies that can result in synergy with
anti-CTLA4
enhancement of the immune response through cell death release of tumor
antigens are
radiation, surgery, and hormone deprivation, among many others. Each of these
protocols,
and others described elsewhere herein, creates a source of tumor antigen in
the host.
Likewise, angiogenesis and signal inhibitors can also be combined with an anti-
CTLA4
antibody to increase an immune response to a tumor cell since inhibition of
angiogenesis or
signal transduction can lead to tumor cell death which may feed tumor antigen
into host
antigen presentation pathways. Therefore, the combination therapies disclosed
herein can
provide an increased source of tumor-specific antigens thereby providing an
increased
immune response to the tumor which, in turn, provides a therapeutic benefit to
the patient.
The present invention, as noted previously, encompasses a combination of an
anti-
CTLA4 antibody and at least one signal transduction inhibitor, such as agents
that can inhibit
EGFR (epidermal growth factor receptor) responses (e.g., EGFR antibodies, EGF
antibodies,
and molecules that are EGFR inhibitors); VEGF (vascular endothelial growth
factor) inhibitors
(e.g., VEGF receptors and molecules that can inhibit VEGF); and erbB2 receptor
(HER2)
inhibitors (e.g., small molecules or antibodies that bind to the erbB2
receptor, where
exemplary antibodies include trastuzumab and pertuzumab, which is a HER
dimerization
inhibitor (HDI)).
EGFR inhibitors are described in, for example in International Patent
Publication Nos.
WO 95/19970, WO 98/14451, WO 98/02434, and U.S. Patent No. 5,747,498, and such
substances can be used in the present invention as described herein. EGFR-
inhibiting agents
include, but are not limited to, the monoclonal antibodies C225, anti-EGFR
22Mab (ImClone
Systems Inc., New York, NY), and ABX-EGF (Abgenix Inc., remont, CA), the
compounds ZD-


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 56-

1839 (AstraZeneca), BIBX-1382 (Boehringer Ingelheim), MDX-447 (Medarex,Inc.,
Annandale,
NJ), and OLX-1 03 (Merck & Co., Whitehouse Station, NJ), TP-38 (IVAX), EGFR
fusion protein,
EGF-vaccine, anti-EGFR immunoliposomes (Hermes Biosciences Inc.), VRCTC-310
(Ventech
Research) and EGF fusion toxin (Seragen Inc., Hopkinton, MA). These and other
EGFR-
inhibiting agents can be used in the present invention in combination with
anti-CTLA4 antibody
to treat various cancers.
Compounds directed at inhibition of epidermal growth factor receptor (EGFR)
tyrosine
kinase (TK) represent a relatively new class of antineoplastic drugs that are
useful in the
method of the present invention. Many human cancers express members of the
EGFR family
on the cell surface. When a ligand binds to EGFR, it sets off a cascade of
cellular reactions
that result in increased cell division and influence other aspects of cancer
development and
progression, including angiogenesis, metastatic spread, and inhibition of
apoptosis. EGFR-
TK inhibitors may selectively target one of the members of the,EGFR family
(EGFR (also
known as HER1 or ErbB-1), HER2/neu (also known as ErbB-2), HER3 (also known as
ErbB-
3), or HER4 (also known as ErbB-4)), or may target two or more of them. EGFR-
TK inhibitors
suitable for use in the present invention include gefitinib (IRESSA),
erlotinib (TARCEVA), CI-
1033 (Pfizer), GW2016 (GlaxoSmithKline), EKB-569 (Wyeth), PKI-166 (Novartis),
CP-
724,714 (Pfizer), CI-1033 (canertinib, Pfizer Inc), and BIBX-1382 (Boeringer-
Ingelheim).
Additional EGFR-TK inhibitors are described in U.S. Patent No. 6,890,924.
VEGF inhibitors, for example SU-5416, SU-6668, SU-11248, SU-12662, SU-14813
(Sugen/Pfizer), as well as AG-013736 (Pfizer) and CP-547,632 (Pfizer, NY) can
also be
employed in combination with the anti-CTLA4 antibody. VEGF inhibitors are
described, for
example, in International Patent Application No. PCT/IB99/00797 (filed May 3,
1999),
International Patent Publication Nos. WO 95/21613; WO 97/22596 (published June
26, 1997);
WO 97/32856 (published Sept. 12, 1997); WO 98/02437 and WO 98/02438 (both
published
Jan. 22, 1998); WO 98/50356; WO 98/54093 (published Dec. 3, 1998); WO 99/10349
(published March 4, 1999); WO 99/16755 (published April 8, 1999); WO 99/24440;
WO
99/61422; U.S. Patent Nos. 5,792,783, 5,834,504; 5,883,113; 5,886,020;
6,177,401; 6,235,764;
6,316,429; 6,395,734; 6,492,383; 6,534,524; and 6,653,308. Other examples of
specific
VEGF inhibitors useful in the present invention are vandetanib (Zactima),
sorafenib
(Bayer/Onyx), AEE788 (Novartis), AZD-2171, VEGF Trap (Regeneron/Aventis),
vatalinib
(also known as PTK-787, ZK-222584; Novartis/Schering AG as described in U.S.
Pat. No.
6,258,812), MACUGEN (pegaptanib octasodium, NX-1838, EYE-001; Pfizer
Inc./Gilead/Eyetech), IM862 (Cytran Inc., Kirkland, WA); neovastat (Aeterna),
IMC-1 C11
ImClone antibody, bevacizumab (AVASTIN, Genentech, Inc., San Francisco, CA);
cetuximab


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 57-

(ERBITUX, ImClone); and ANGIOZYME, a synthetic ribozyme that cleaves mRNA
producing
VEGF1 from Ribozyme (Boulder, CO) and Chiron (Emeryville, CA).
ErbB2 receptor inhibitors, such as GW-282974, GW-572016 (lapatinib) (Glaxo
Wellcome plc), and the monoclonal antibodies AR-209 (Aronex Pharmaceuticals
Inc.,
Woodlands, TX), mapatumumab (HGS-ETR1, Human Genome Sciences, Inc.) an agonist
of
TRAIL Receptor 1), trastuzumab (HERCEPTIN; Genentech, Inc., San Francisco,
CA),
pertuzumab (OMNITARG; 2C4; Genentech, a HER2 dimerization inhibitor HDI)); TAK-
165
(Takeda), GW-572016 (lapatinib, GlaxoSmithKline), pelitinib (EKB-569 Wyeth),
BMS-599626,
PKI-166 (Novartis), dHER2 (HER2 Vaccine, Corixa and GlaxoSmithKline), Osidem
(IDM-1),
APC8024 (HER2 Vaccine, Dendreon), anti-HER2/neu bispecific antibody (Decof
Cancer
Center), B7.her2.IgG3 (Agensys), AS HER2 (Research Institute for Rad Biology &
Medicine),
trifunctional bispecific antibodies (University of Munich), mAB AR-209 (Aronex
Pharmaceuticals Inc), and 2B-1 (Chiron), can be combined with the anti-CTLA4
antibody to
provide effective combination therapy. Other erbB2 receptor inhibitors are
described in, for
example, International Patent Publication Nos. WO 95/19970 (published July 27,
1995); WO
97/13760 (published April 17, 1997); WO 98/02434 (published January 22, 1998);
WO
98/02437 (published January 22, 1998); WO 99/35132 (published July 15, 1999);
WO
99/35146 (published July 15, 1999); U.S. Patent Nos. 5,587,458, and 5,877,305.
ErbB2
receptor inhibitors useful in the present invention are also described in
EP1029853 (published
August 23, 2000) and in International Patent Publication No. WO 00/44728,
(published August
3, 2000). ErbB2 receptor inhibitors useful in the present invention are also
described in
United States Patent Nos. 6,465,449, and 6,284,764, and International
Application No. WO
2001/98277 each of which are herein incorporated by reference in their
entirety. The erbB2
receptor inhibitor compounds and substances described in the aforementioned
PCT
applications, U.S. patents, and U.S. provisional applications, as well as
other compounds and
substances that inhibit the erbB2 receptor, can be used with the anti-CTLA4
antibody in
combination therapy in accordance with the present invention.
Various other compounds, such as styrene derivatives, have also been shown to
possess tyrosine kinase inhibitory properties, and some of tyrosine kinase
inhibitors have
been identified as erbB2 receptor inhibitors. Other erbB2 Inhibitors are
described in
European patent publications EP 566,226 Al (published October 20, 1993), EP
602,851 Al
(published June 22, 1994), EP 635,507 Al (published January 25, 1995), EP
635,498 Al
(published January 25, 1995), and EP 520,722 Al (published December 30, 1992).
These
publications refer to certain bicyclic derivatives, in particular quinazoline
derivatives
possessing anti-cancer properties that result from their tyrosine kinase
inhibitory properties.
Also, WO 92/20642 (published November 26, 1992), refers to certain bis-mono
and bicyclic


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 58-

aryl and heteroaryl compounds as tyrosine kinase inhibitors that are useful in
inhibiting
abnormal cell proliferation. WO 96/16960 (published June 6, 1996), WO 96/09294
(published
March 6, 1996), WO 97/30034 (published August 21, 1997), WO 98/02434
(published
January 22, 1998), WO 98/02437 (published January 22, 1998), and WO 98/02438
(published January 22, 1998), also refer to substituted bicyclic
heteroaromatic derivatives as
tyrosine kinase inhibitors that are useful for the same purpose. Other patent
applications that
refer to anti-cancer compounds are World Patent Application WO 00/44728
(published August
3, 2000), EP 1029853A1 (published August 23, 2000), and WO 01/98277 (published
December 12, 2001), all of which are incorporated herein by reference in their
entirety.
BAY 43-9006 (Onyx Pharmaceuticals) is an inhibitor of both the RAF/MEK/ERK
signaling pathway and the VEGFR-2/PDGFR-(3 signaling cascade. RAF kinase is an
enzyme
in the RAS pathway and mutations in the RAS gene are associated with
approximately 20%
of all human cancers, including 90% of pancreatic cancer, 50% of colon cancer
and about
30% of non-small cell lung cancer (NSCLC), and BRAF, a specific RAF kinase,
has been
demonstrated to be mutated in about two-thirds of melanomas and some
colorectal cancer
(CRC) and other solid tumors. In addition, VEGFR and PDGFR-R play key roles in
angiogenesis such that their inhibition decreases angiogenesis. Thus, the
present invention
encompasses administering a combination comprising an anti-CTLA4 antibody and
BAY 43-
9006 to treat cancer.
PDGFR inhibitors include but not limited to those disclosed in Int. Pat. Pub.
No. WO
01/40217, published June 7, 2001 and Int. Pat. Pub. No. WO 2004/020431,
published March
11, 2004, the contents of which are incorporated in their entirety for all
purposes.
Preferred PDGFR inhibitors include Pfizer's CP-673,451 and CP-868,596 and
their
pharmaceutically acceptable salts.
TIE-2 inhibitors include GlaxoSmithKline's benzimidazoles and pyridines
including
GW-697465A such as described in Int. Pat. Pub. Nos. WO 02/044156 published
June 6,
2002, WO 03/066601 published August 14, 2003, WO 03/074515 published September
12,
2003, WO 03/022852 published March 20, 2003 and WO 01/37835 published May 31,
2001.
Other TIE-2 inhibitors include Regeneron's biologicals such as those described
in Int. Pat.
Pub. No. WO 09/611269 published April 18, 1996, Amgen's AMG-386, and Abbott's
pyrrolopyrimidines such as A-422885 and BSF-466895 described in Int. Pat. Pub.
Nos. WO
09/955335, WO 09/917770, WO 00/075139, WO 00/027822, WO 00/017203 and WO
00/017202.
Use of an angiogenesis inhibitor in combination with an anti-CTLA4 antibody
has
been discussed previously elsewhere herein. Moreover, an angiogenesis
inhibitor includes,
but is not limited to, bevacizumab (AVASTIN) which is a humanized antibody to
VEGF.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 59-

Bevacizumab can be used in combination with 5FU, and is indicated as a first-
line treatment
of patients with metastatic carcinoma of the colon or rectum. Agents that
directly target
angiogenic factors or their receptors offer the prospect for greater activity
in receptor-
competent hematologic malignancies by interrupting autocrine receptor
signaling.
Bevacizumab produces sustained neutralization of circulating VEGF and may be
useful for
treatment of myelodysplastic syndrome (MDS), lymphoma, acute myeloid leukemia
(AML),
and solid tumors. Thus, an anti-CTLA4 antibody and bevacizumab combination
therapy, in
further combination with 5FU and additional chemotherapeutic agents, for
treatment of CRC,
and other malignancies, is encompassed by the present invention.
In another embodiment of the present invention the anti-angiogenesis agent is
a
protein kinase C f3 such as enzastaurin, midostaurin, perifosine,
staurosporine derivative
(such as RO 318425, R0317549, R0318830 or RO 318220 (Roche)), teprenone
(Selbex)
and UCN-01 (Kyowa Hakko).
The receptor tyrosine kinase inhibitors (RTKI) represent a class of synthetic,
small
molecule inhibitors of angiogenic receptor signaling. The first receptor
antagonist to enter
clinical testing in hematologic malignancies is SU5416 (Sugen), which impairs
ligand-induced
autophosphorylation of the VEGFR-1 and VEGFR-2 receptors and c-Kit. SU5416
inhibits
VEGF-induced clonogenic response in leukemia cell lines and promotes apoptosis
in
myeloblasts from AML patients. Other RTKIs, including SU11248 (sunitinib),
SU12662,
SU14813, (Pfizer Inc.), vatalanib (also known as PTK787/ZK222584; Novartis),
and AG-
013736 (Pfizer), can be used in combination with an anti-CTLA4 antibody to
treat AML and
other receptor-competent hematologic malignancies. CP-675,206 may be combined
with
sunitinib (SU-1 1248), among other RTKIs. Thus, the present invention
encompasses a
combination of an anti-CTLA4 antibody and at least one antiangiogenic and/or
signal
transduction inhibitor, including, e.g., SU-11248, SU-12662, SU-14813, AG-
013736, as well
as other angiogenesis and signal transduction inhibitors that are well-known
in the art or
developed in the future.
As noted previously, the combination therapy methods of the invention can be
used
with other agents useful in treating abnormal cell growth or cancer,
including, but not limited
to other agents capable of enhancing antitumor immune responses, such as
additional,
different, CTLA4 antibodies, and other agents also capable of blocking CTLA4;
and anti-
proliferative agents such as farnesyl protein transferase inhibitors, and avP3
inhibitors, such
as the av(33 antibody VITAXIN, avR5 inhibitors, p53 inhibitors, and the like.
Where the antibody of the invention is administered in combination with
another
immunomodulatory agent, the immunomodulatory agent can be selected for example
from the
group consisting of a dendritic cell activator such as CD40 ligand and anti-
CD40 agonist


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 60-

antibodies, as well as enhancers of antigen presentation, enhancers of T-cell
tropism,
inhibitors of tumor-related immunosuppressive factors, such as TGF-(3
(transforming growth
factor beta), and IL-10. Preferred anti-CD40 agonist antibodies encompass
antibodies
disclosed in International Patent Application No. PCT/US02/36107, filed
November 8, 2002,
now published as International Patent Publication No. WO 03/040170, and U.S.
Patent
Application No. 10/292,088, filed November 8, 2002, now published as U.S.
Patent
Publication No. US2003/0211100, including, but not limited to, an antibody
having the heavy
and light chain amino acid 'sequence of antibody 3.1.1, 3.1.1.H-A78T, 3.1.1H-
A78T-V88A-
V97A, 3.1.1 L-L4M-L83V, 3.1.1 H-A78T-V88A-V97A/3.1.1 L-L4M-L83V, 7.1.2,
10.8.3, 15.1.1,
21.2.1, 21.4.1, 22.1.1, 22.1.1 H-C 109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1 H-D
16E, 23.29.1,
and 24.2.1.
The present treatment regimens may also be combined with antibodies or other
ligands that inhibit tumor growth by binding to IGF-1 R (insulin-like growth
factor 1 receptor).
Specific anti-IGF-1 R antibodies that can be used in the present invention
include those
described in International Patent Application No. PCT/USOI/51113, filed
12/20/01, and
published as International Patent Publication No. W002/053596, International
Patent
Application No. PCT/IB2004/002555, filed August 3, 2004, and published as
International
Patent Publication No. WO 2005/016967. Preferred anti-IGFR-1 R antibodies
encompass an
antibody having the heavy and light chain amino acid sequence of, e.g.,
antibody 2.12.1,
2.13.2, 2.14.3, 3.1.1, 4.9.2 and 4.17.3.
Ligands that inhibit signaling via the IGF-1 R also encompass small molecules,
and
other ligands including, inter alia, somavert (PEGVISOMANT), which is a growth
hormone
analog that inhibits IGF-1 signaling. PEGVISOMANT is conjugated with
polyethylene glycol
and can be used, among other things, to treat acromegaly. PEGVISOMANT can be
co-
administered with anti-CTLA4 antibody to treat cancer in that the combination
can inhibit
tumor growth. Thus, PEGVISOMANT, similarly with anti-IGF-1 R antibodies, can
be used in
combination with an anti-CTLA4 antibody to treat cancer as disclosed herein.
Additional exemplary therapeutic and/or prophylactic antibodies include, but
are not
limited to, SYNAGIS (Medlmmune, MD) which is a humanized anti-respiratory
syncytial virus
(RSV) monoclonal antibody; REMICADE (infliximab) (Centocor, PA) which is a
chimeric anti-
TNFa monoclonal antibody; REOPRO (abciximab) (Centocor) which is an anti-
glycoprotein
Ilb/Illa receptor; ZENAPAX (daclizumab) (Roche Pharmaceuticals, Switzerland)
which is a
humanized anti-CD25 monoclonal antibody; AVASTIN (bevacizumab); matuzumab
(Merck
AG), nimotuzumab, panitumumab, EGFR inhibitor antibody (Amgen/Abgenix);
ERBITUX
(cetuximab). Other examples are a humanized anti-CD18 F(ab')2 (Genentech);
CDP860
which is a humanized anti CD18 F(ab')2 (Celltech, UK); PR0542 which is an anti-
HIV gp120


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-61-
antibody fused with CD4 (Progenics/Genzyme Transgenics); OSTAVIR which is a
human anti
Hepatitis B virus antibody (Protein Design Lab/Novartis); PROTOVIR which is a
humanized
anti-CMV IgG1 antibody (Protein Design Lab/Novartis); MAK-195 (SEGARD) which
is a
murine-anti-TNF-a F(ab')2 (Knoll Pharma/BASF); IC14 which is an anti-CD14
antibody (ICOS
Pharm); a humanized anti-VEGF IgGI antibody (Genentech); OVAREX which is a
murine
anti-CA 125 antibody (Altarex); PANOREX which is a murine anti-17-IA cell
surface antigen
IgG2a antibody (Glaxo SmithKline/Centocor); BEC2 which is a murine 5 anti-
idiotype (GD3
epitope) IgG antibody (ImClone System); IMC-C225 which is a chimeric anti-EGFR
IgG
antibody (ImClone System); VITAXIN which is a humanized anti-aV(33 integrin
antibody
(Applied Molecular Evolution/Medlmmune); Campath 1 H/LDP-03 which is a
humanized anti
CD52 IgGI antibody (Leukosite); Smart M195 which is a humanized anti-CD33 IgG
antibody
(Protein Design Lab/Kanebo); rituximab (RITUXAN) which is a chimeric anti-CD20
IgG1
antibody (Biogen IDEC/Genentech, Roche/Zettyaku); BEXXAR (131-1-tositumomab);
belimumab (LymphoStat-B); HuMax-CD20 (HuMax, Genmab); R 1594
(Roche/Genentech);
TRU-015 (Trubion Pharmaceuticals); ocrelizumab (PRO 70769); LYMPHOCIDE which
is a
humanized anti-CD22 IgG antibody (Immunomedics); Smart ID10 which is a
humanized anti-
HLA antibody (Protein Design Lab); ONCOLYM (Lym-1) is a radiolabelled murine
anti-HLA
diagnostic reagent antibody (Techniclone); ABX-IL'8 is a human anti-IL8
antibody (Abgenix);
anti-CD11a is a humanized IgG1 antibody (Genentech/Xoma); ICM3 is a humanized
anti-
ICAM3 antibody (ICOS Pharm); IDEC-114 is a primatized anti-CD80 antibody (IDEC
Pharm/Mitsubishi); SGN14, chimeric anti-CD40 antibody (Seattle Genetics);
SGN40,
humanized anti-CD40 antibody (Seattle Genetics); CHIR-12.12, antagonist anti-
CD40:
antibody (Chiron); ISF-154 (Ad-CD154, Tragen); toralizumab; ABI-793
(Novartis); IDEC-131
is a humanized anti-CD40L antibody (IDEC/Eisai); IDEC-151 is a primatized anti-
CD4
antibody (IDEC); IDEC-152 is a primatized anti-CD23 antibody (IDEC/Seikagaku);
SMART
anti-CD3 is a humanized anti-CD3 IgG (Protein Design Lab); 5G1.1 is a
humanized anti-
complement factor 5 (C5) antibody (Alexion Pharm); adalimumab is a human anti-
TNF-a
antibody (HUMIRA, CAT/Abbott); CDP870 is a humanized anti-TNF-a Fab fragment
(Celltech); MDX-CD4 is a human anti-CD4 IgG antibody (Medarex/Eisai/Genmab);
CDP571 is
a humanized anti-TNF-a IgG4 antibody (Celltech); LDP-02 is a humanized anti-
a4R7
antibody (LeukoSite/Genentech); OrthoClone OKT4A is a humanized anti-CD4 IgG
antibody
(Ortho Biotech); ANTOVA is a humanized anti-CD40L IgG antibody (Biogen);
ANTEGREN is
a humanized anti VLA-4 IgG antibody (Elan); MDX-33 is a human anti-CD64 (FcyR)
antibody
30 (Medarex/Centeon); SCH55700 is a humanized anti-IL-5 IgG4 antibody
(Celltech/Schering); SB-240563 and SB-240683 are humanized anti-IL-5 and IL-4
antibodies,
respectively (SmithKline Beecham); rhuMab-E25 is a humanized anti-IgE IgGI
antibody


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 62-

(Genentech/Norvartis/Tanox Biosystems); ABX-CBL is a murine anti CD-147 IgM
antibody
(Abgenix); BTI- 322 is a rat anti-CD2 IgG antibody (Medimmune/Bio
Transplants);
Orthoclone/OKT3 is a murine anti-CD3 IgG2a antibody (Ortho Biotech); SIMULECT
is a
chimeric anti-CD25 IgG1 antibody (Novartis Pharm); LDP-01 is a humanized anti-
(32-integrin
IgG antibody (LeukoSite); Anti-LFA-1 is a murine anti CD18 F(ab')a (Pasteur-
Merieux/Immunotech); CAT-152 is a human anti-TGF-(32 antibody (Cambridge Ab
Tech); and
CORSEVIN M is a chimeric anti-Factor VII antibody (Centocor). The above-listed
immunoreactive reagents, as well as any other immunoreactive reagents, may be
administered according to any regimen known to those of skill in the art,
including the
regimens recommended by the suppliers of the immunoreactive reagents.
In another embodiment the agent used in conjunction with CP-675,206 is a
hepatocyte growth factor receptor (HGFR or c-MET) antagonist.
In another embodiment of the invention, CP-675,206 or ipilimumab is
administered
with an antibody to MAdCAM to treat cancer.
The nucleotide sequence encoding an antibody or other immunoreactive reagent
may
be obtained from any information available to those of skill in the art (i.e.,
from Genbank, the
literature, or by routine cloning). If a clone containing a nucleic acid
encoding a particular
antibody or an epitope-binding fragment thereof or other immunoreactive
reagent is not
available, but the sequence of the antibody molecule or epitope-binding
fragment thereof or
other immunoreactive reagent is known, a nucleic acid encoding the
immunoglobulin, or other
immunoreactive reagent may be chemically synthesized or obtained from a
suitable source
(e.g., an antibody cDNA library, or a cDNA library generated from, or nucleic
acid, preferably
poly A+ RNA, isolated from any tissue or cells expressing the antibody, such
as hybridoma
cells selected to express an antibody) by any method, including, e.g., PCR
amplification using
synthetic primers hybridizable to the 3' and 5' ends of the sequence or by
cloning using an
oligonucleotide probe specific for the particular gene sequence to identify,
e.g. a cDNA clone
from a cDNA library that encodes the antibody.
The anti-CTLA4 antibody, as well as any other second antibody administered
therewith, may also be administered with a cytokine such as, e.g., interleukin
(e.g., IL-l a, IL-
1(3, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-
15, IL-18), interferon (e.g.,
IFNa, IFN(3, IFNy), tumor necrosis factor (e.g., TNFa, TNF(3), G-CSF, GM-CSF,
TGF-(3, SLC,
endothelial monocyte activating protein-2 (EMAP2), MIP-3a, MIP-3(3, or an MHC
gene, such
as HLA-B7.
In one embodiment, CTLA4 blockade is co-administered with IL-15 or an agonist
thereof, to increase, induce or prolong an immune response. This is because
studies
demonstrated that while CTLA4 blockade induced rejection of engrafted islet
cells, inhibition


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 63-

of IL-15/IL-15R processes inhibited activation of CD8+ T cells thereby
increasing
immunogenic tolerance and preventing graft rejection. That is, anti-CTLA4
blockade leads to
graft rejection and IL-15-blockade had the opposite effect. See Ferrari-Lacraz
et al.,
Transplantation 82:1510-1517 (2006). Thus, where decreasing immunogenic
tolerance is
desired, e.g., where inducing an anti-tumor immune response in a patient is
desired, co-
administration of a CTLA4 antagonist in combination with IL-15, or an agonist
of IL-15/IL-15R,
may enhance a therapeutic anti-tumor response, and may provide a greater
immune
response than either agent alone, and more preferably, may provide a
synergistic effect
greater than the additive effect of each agent combined. Thus, in an
embodiment of the
invention, combination therapy comprising an anti-CTLA4 antibody and an
agonist of the IL-
15/IL-15R signaling pathway (e.g., IL-15, an agonist anti-IL15 antibody, an
agonist anti-IL-15R
antibody, among others), is provided to induce, increase and/or prolong an
immune response.
Additional exemplary cytokines include other members of the TNF family,
including,
but not limited to, TNF-a-related apoptosis-inducing ligand (TRAIL), TNF-a-
related activation-
induced cytokine (TRANCE), TNF-a-related weak inducer of apoptosis (TWEAK),
CD40
ligand (CD40L), LT-a, LT-(3, OX40L, FasL, CD27L, CD30L, 4-1 BBL, APRIL, LIGHT,
TL1,
TNFSF16, TNFSF17, and AITR-L, or a functional portion thereof. See, e.g., Kwon
et ai.,
Curr. Opin. Immunol. 11:340-345 (1999) for a general review of the TNF family.
In one embodiment, interferons and numerous other immune enhancing agents that
may be used in combination therapy with CP-675,206 include, but are not
limited to interferon
alpha, interferon alpha-2a, interferon, alpha-2b, interferon beta, interferon
gamma-la,
interferon gamma-1 b(Actimmune), or interferon gamma-n1, PEG Intron A, and
combinations
thereof. Other agents include interieukin 2 agonists (such as aldesieukin, BAY-
50-4798,
Ceplene (histamine dihydrochloride), EMD-273063, MVA-HPV-IL2, HVA-Muc-1-IL2,
interleukin
2, teceleukin and Virulizin), Ampligen, Canvaxin, CeaVac (CEA), denileukin,
filgrastim,
Gastrimmune (G17DT), gemtuzumab ozogamicin, Glutoxim (BAM-002), GMK vaccine
(Progenics), Hsp 90 inhibitors (such as HspE7 from Stressgen, AG-858, KOS-953,
MVJ-1-1
and STA-4783), imiquimod, krestin (polysaccharide K), lentinan, Melacine
(Corixa), MelVax
(mitumomab), molgramostim, Oncophage (HSPPC-96), OncoVAX (including OncoVAX-CL
and OncoVAX-Pr), oregovomab, sargramostim, sizofiran, tasonermin, TheraCys,
thymalfasin,
pemtumomab (Y-muHMFG1), picibanil, Provenge (Dendreon), ubenimex, WF-10
(Immunokine), Z-100 (Ancer-20 from Zeria), Lenalidomide (REVIMID, Celegene),
thalomid
(Thalidomide), and combinations thereof.
The present invention includes coadministration of an anti-CTLA4 antibody, or
combination of such antibodies, with at least one T cell costimulatory
molecule (also referred
to herein as a "costim"), including, but not limited to, CD4, CD25, and the
like. Costimulatory


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 64-

molecules further include, e.g., PD-1, B7-H3, OX40, ICOS, CD30, HLA-DR, MHCII,
and LFA,
or agonist antibodies thereto. These and other costims are well-known in the
art and have
been well characterized as described in, e.g., Schwartz et al., Nature 410:604-
608 (2001);
Schwartz et al., Nature lmmunol. 3:427-434 (2002); and Zhang et al., Immunity
20:337-347
(2004).
In one alternative embodiment of the invention, CP-675,206 or ipilimumab is
administered with an antibody to human 4-IBB to treat cancer. Suitable anti-4-
1BB
antibodies are described in, e.g., lnt. Appl. No. PCT/US2004/033587 (published
April 21,
2005, as WO 2005/035584). One such antibody is BMS66513.
In another embodiment of the invention, CP-675,206 or ipilimumab is
administered
with an antibody to human mucosal addressin cell adhesion molecule (MAdCAM) to
treat
cancer. Suitable anti-MAdCAM antibodies are described in, e.g., Int. Appl. No.
PCT/US2005/000370 (published July 28, 2005, as WO 2005/067620). Preferably,
the anti-
MAdCAM antibody is an antibody comprising a heavy and light chain variable
region amino
acid sequence of antibody 1.7.2, 1.8.2, 6.14.2, 6.22.2, 6.34.2, 6.67.1,
6.73.2, 6.77.1, 7.16.6,
7.20.5, 7.26.4, and 9.8.2.
In addition to combination of anti-CTLA4 and immunostimulatory compounds to
increase an anti-tumor immune response, it would be understood that certain
immunosuppressive molecules may inhibit or decrease an anti-tumor response.
Accordingly,
in another embodiment of the invention, anti-CTLA antibody is administered in
combination
with an inhibitor of an immunosuppressive molecule. Such immunosuppressive
molecules
include, but are not limited to, IDO, TGF-R, PD-1, among others. Thus, a
combination of anti-
CTLA4 blockade and inhibition of another immunoregulatory pathway is
encompassed in the
present invention, thereby further enhancing the anti-tumor response provided
by anti-CTLA4
blockade.
The treatment regimens described herein may be combined with anti-angiogenesis
agents, such as MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix-
metalloproteinase 9) inhibitors, and COX-II (cyclooxygenase II) inhibitors,
can be used in
conjunction with the antibody in the method of the invention. Examples of
useful COX-II
inhibitors include CELEBREX (celecoxib), parecoxib, deracoxib, ABT-963, COX-
189
(Lumiracoxib), BMS 347070, RS 57067, NS-398, Bextra (valdecoxib), Vioxx
(rofecoxib), SD-
8381, 4-methyl-2-(3,4-dimethylphenyl)-1-(4-sulfamoyl-phenyl)-1 H-pyrrole, 2-(4-
ethoxyphenyl)-
4-methyl-1-(4-sulfamoylphenyl)-1 H-pyrrole, T-614, JTE-522, S-2474, SVT-2016,
CT-3, SC-
58125 and Arcoxia (etoricoxib). Additonally, COX-II inhibitors are disclosed
in U.S. Patent
Application Nos. 10/801,446 and 10/801,429, the contents of which are
incorporated in their
entirety for all purposes.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 65-

In one embodiment the agent is celecoxib as disclosed in U.S. Patent No.
5,466,823,
the contents of which are incorporated by reference in its entirety for all
purposes. In another
embodiment the agent is deracoxib as disclosed in U.S. Patent No. 5,521,207,
the contents of
which are incorporated by reference in its entirety for all purposes.
Other useful anti-angiogenic inhibitors used in conjunction with CP-675,206
include
aspirin, and non-steroidal anti-inflammatory drugs (NSAIDs) which
nonselectively inhibit the
enzymes that make prostagiandins (cyclooxygenase I and II), resulting in lower
levels of
prostaglandins. Such agents include, but are not limited to, Aposyn
(exisulind), Salsalate
(Amigesic), Diflunisal (Dolobid), Ibuprofen (Motrin), Ketoprofen (Orudis),
Nabumetone
(Relafen), Piroxicam (Feldene), Naproxen (Aleve, Naprosyn), Diclofenac
(Voltaren),
Indomethacin (Indocin), Sulindac (Clinoril), Tolmetin (Tolectin), Etodolac
(Lodine), Ketorolac
(Toradol), Oxaprozin (Daypro) and combinations thereof.
Preferred nonselective cyclooxygenase inhibitors include ibuprofen (Motrin),
nuprin,
naproxen (Aleve), indomethacin (Indocin), nabumetone (Relafen) and
combinations thereof.
Other anti-angiogenic compounds include acitretin, angiostatin, aplidine,
cilengtide,
COL-3, combretastatin A-4, endostatin, fenretinide, halofuginone, Panzem (2-
methoxyestradiol), PF03446962 (ALK-1 inhibitor), rebimastat, removab,
Revlimid,
squalamine, thalidomide, ukrain, Vitaxin (alpha-v/beta-3 integrin), and
zoledronic acid.
Examples of useful matrix metalloproteinase inhibitors are described in
International
Patent Publication Nos. WO 96/33172; WO 96/27583; WO 98/07697, WO 98/03516, WO
98/34918, WO 98/34915, WO 98/33768, WO 98/30566, WO 90/05719, WO 99/52910, WO
99/52889, WO 99/29667, European Patent Application Nos. 780386 (published June
25,
1997), 97304971 (filed July 8, 1997), 99308617 (filed October 29, 1999),
606046 (published
July 13, 1994), 931788 (published July 28, 1999), 99302232 (filed March 25,
1999),
International Application PCT/IB98/01113 (filed July 21, 1998), Great Britain
patent application
number 9912961 (filed June 3, 1999), United States Provisional Patent
Application No.
60/148,464 (filed August 12, 1999), and U.S. Patent Nos. 5,863,949, and
5,861,510.
Preferred MMP-2 and MMP-9 inhibitors are those that have little or no activity
inhibiting
MMP-1. More preferred are those that selectively inhibit MMP-2 and/or MMP-9
relative to the
other matrix-metalloproteinases (i.e., MMP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP-
7, MMP-
8, MMP-10, MMP-11, MMP-12, and MMP-13).
Some specific examples of MMP inhibitors useful in the present invention are
AG-3340,
RO 32-3555, RS 13-0830, ABT-510 (Abbott), ABT 518 (Abbott), Apratastat
(Amgen), AZD
8955 (AstraZeneca), Neovostat (AE-941), COL 3 (CollaGenex Pharmaceuticals),
doxycycline
hyclate, MPC 2130 (Myriad) and PCK 3145 (Procyon), and the compounds recited
in the
following list:


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 66-
3-[[4-(4-fluoro-phenoxy)-benzenesulfonyl]-(1-hydroxycarbamoyl-cyclopentyl)-am
ino]-
propionic acid;
3-exo-3-[4-(4-fluoro-phenoxy)-benzenesulfonylam ino]-8-oxa-bicyclo[3.2.1
]octane-3-
carboxylic acid hydroxyamide;
(2R, 3R) 1-[4-(2-chloro-4-fluoro-benzyloxy)-benzenesulfonyi]-3-hydroxy-3-
methyl-
piperidine-2-carboxylic acid hydroxyamide;
4-[4-(4-fluoro-phenoxy)-benzenesulfonylamino]-tetrahydro-pyran-4-carboxylic
acid
hydroxyamide;
3-[[4-(4-fluoro-phenoxy)-benzenesulfonyl]-(1-hydroxycarbamoyl-cyclobutyl)-am
ino]-
propionic acid;
4-[4-(4-chloro-phenoxy)-benzenesulfonylamino]-tetrahydro-pyran-4-carboxylic
acid
hydroxyamide;
(R) 3-[4-(4-chloro-phenoxy)-benzenesulfonylamino]-tetrahydro-pyran-3-
carboxylic
acid hydroxyamide;
(2R, 3R) 1-[4-(4-fluoro-2-methyl-benzyloxy)-benzenesulfonyl]-3-hydroxy-3-
methyl-
piperidine-2-carboxylic acid hydroxyamide;
3-[[4-(4-fluoro-phenoxy)-benzenesulfonyl]-(1-hydroxycarbamoyl-l-methyl-ethyl)-
amino]-propionic acid;
3-[[4-(4-fluoro-phenoxy)-benzenesu Ifonyl]-(4-hyd roxycarbamoyl-tetrahydro-
pyran-4-
yl)-amino]-propionic acid;
3-exo-3-[4-(4-chloro-phenoxy)-benzenesulfonylamino]-8-oxa-bicyclo[3.2.1
]octane-3-
carboxylic acid hydroxyamide;
3-endo-3-[4-(4-fluoro-phenoxy)-benzenesulfonylam ino]-8-oxa-bicyclo[3.2.1
]octane-3-
carboxylic acid hydroxyamide; and
(R) 3-[4-(4-fluoro-phenoxy)-benzenesulfonylamino]-tetrahydro-furan-3-
carboxylic acid
hydroxyamide;
and pharmaceutically acceptable salts and solvates of said compounds.
In another embodiment, the agent is a signal transduction inhibitor. Such
inhibitors
include small molecules, antibodies, and antisense molecules, and encompass
tyrosine
kinase inhibitors, serine/threonine kinase inhibitors. More specifically,
signal transduction
inhibitors include farnesyl protein transferase inhibitors, EGF inhibitors,
ErbB-1 (EGFR), ErbB-
2, pan erb, IGF1 R inhibitors, MEK, c-Kit inhibitors, FLT-3 inhibitors, K-Ras
inhibitors, P13
kinase inhibitors, JAK inhibitors, STAT inhibitors, Raf kinase inhibitors, Akt
inhibitors, mTOR
inhibitor, P70S6 kinase inhibitors and inhibitors of the WNT pathway and so
called multi-
targeted kinase inhibitors


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 67-

In another embodiment, the signal transduction inhibitor is a farnesyl protein
transferase inhibitor. Farnesyl protein transferase inhibitors include the
compounds disclosed
and claimed in United States Patent 6,194,438, issued February 27, 2002;
United States
Patent 6,258,824, issued July 10, 2001; United States Patent 6,586,447, issued
July 1, 2003;
United States Patent 6,071,935, issued June 6, 2000; and United States Patent
6,150,377,
issued November 21, 2000. Other farnesyl protein transferase inhibitors
include AZD-3409
(AstraZeneca), BMS-214662 (Bristol-Myers Squibb), Lonafarnib (Sarasar) and RPR-
115135
(Sanofi-Aventis). Each of the foregoing patent applications and provisional
patent
applications is herein incorporated by reference in their entirety.
In another embodiment the signal transduction inhibitor is a GARF inhibitor.
Preferred
GARF inhibitors (glycinamide ribonucleotide formyltransferse inhibitors)
include Pfizer's AG-
2037 (pelitrexol) and its pharmaceutically acceptable salts. GARF inhibitors
useful in the
practice of the present invention are disclosed in US Patent No. 5,608,082,
which is
incorporated in its entirety for all purposes.
In another embodiment the signal transduction inhibitor is a MEK inhibitor.
MEK
inhibitors include Pfizer's MEK1/2 inhibitor PD325901, Array Biopharm's MEK
inhibitor ARRY-
142886, and combinations thereof.
In another embodiment the anti-cancer signal transduction inhibitor is an mTOR
inhibitor. mTOR inhibitors include everolimus (RAD001, Novartis), zotarolimus,
temsirolimus
(CCI-779, Wyeth), AP 23573 (Ariad), AP23675, Ap23841, TAFA 93, rapamycin
(sirolimus), and
combinations thereof.
In another embodiment the anti-cancer signal transduction inhibitor is an
Aurora 2
inhibitor such as VX-680 and derivatives thereof (Vertex), R 763 and
derivatives thereof (Rigel),
and ZM 447439 and AZD 1152 (AstraZeneca), or a Checkpoint kinase 1/2
inhibitors such as
XL844 (Exilixis).
In another embodiment the anti-cancer signal transduction inhibitor is an Akt
inhibitor
(Protein Kinase B) such as API-2, perifosine and RX-0201.
Additionally, other targeted anti-cancer agents include the raf inhibitors
sorafenib (BAY-
43-9006, Bayer/Onyx), GV-1002, ISIS-2503, LE-AON and GI-4000, BMS 184476, CCI
779,
DTIC, ISIS 2503, ONYX 015, and flavopyridol.
The invention also relates to methods comprising CP-675,206 and cell cycle
inhibitors
such as the CDK2 inhibitors ABT-751 (Abbott), AZD-5438 (AstraZeneca),
alvocidib (flavopiridol,
Aventis), BMS-387,032 (SNS 032 Bristol Myers), EM-1421 (Erimos), indisulam
(Esai), seliciclib
(Cyclacel), BIO 112 (Onc Bio), UCN-01 (Kyowa Hakko), and AT7519 (Astex
Therapeutics) and
Pfizer's multitargeted CDK inhibitors PD0332991 and AG24322.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 68-

The invention also relates to the use of CP-675,206 together with telomerase
inhibitors such as transgenic B lymphocyte immunotherapy (Cosmo Bioscience),
GRN 163L
(Geron), GV1001 (Pharmexa), RO 254020 (and derivatives thereof), and
diazaphilonic acid.
The invention also relates to the use of CP-675,206 with hormonal, anti-
hormonal,
anti-androgenal therapeutic agents such as anti-estrogens including, but not
limited to
toremifene, raloxifene, anti-androgens such as finasteride, mifepristone,
ABARELIX (Praecis),
TRELSTAR, ATAMESTANE (Biomed-777), ATRASENTAN (Xinlay), Bosentan,
doxercalciferol,
and combinations thereof.
The invention also contemplates the use of CP-675,206 together with gene
silencing
agents or gene activating agents such as histone deacetylase (HDAC) inhibitors
such as
suberolanilide hydroxamic acid (SAHA, Merck Inc./Aton Pharmaceuticals),
depsipeptide
(FR901228 or FK228), G2M-777, MS-275, pivaloyloxymethyl butyrate and PXD-101.
The invention also contemplates the use of CP-675,206 with gene therapeutic
agents
such as ADVEXIN (ING 201), TNFerade (GeneVec, a compound which express
TNFalpha in
response to radiotherapy), RB94 (Baylor College of Medicine).
The invention also contemplates CP-675,206 together with ribonucleases such as
Onconase (ranpirnase).
The invention also contemplates CP-675,206 with antisense oligonucleotides
such as
bcl-2 antisense inhibitor Genasense (Oblimersen, Genta).
The invention also contemplates CP-675,206 together with proteosomics such as
PS-
341 (MLN-341) and VELCADE (bortezomib).
The invention also contemplates CP-675,206 together with anti-vascular agents
such
as Combretastatin A4P (Oxigene).
The invention also contemplates combination of CP-675,206 with traditional
cytotoxic
agents including DNA binding agents, mitotic inhibitors, alkylating agents,
anti-metabolites,
intercalating antibiotics, topoisomerase inhibitors and microtubulin
inhibitors.
In an embodiment of the invention, an anti-CTLA4 antibody is administered to a
patient receiving a chemotherapeutic agent for treatment of cancer. Such
chemotherapeutic
agents are known in the art and include, but are not limited to: methotrexate,
taxol,
mercaptopurine, thioguanine, hydroxyurea, cytarabine, cyclophosphamide,
ifosfamide,
nitrosoureas, cisplatin, carboplatin, mitomycin, dacarbazine, procarbizine,
etoposides,
camptothecins, bleomycin, doxorubicin, idarubicin, daunorubicin, dactinomycin,
plicamycin,
mitoxantrone, asparaginase, vinblastine, vincristine, vinorelbine, paclitaxel,
and docetaxel,
doxorubicin, epirubicin, 5-fluorouracil, taxanes such as docetaxel and
paclitaxel, leucovorin,
levamisole, irinotecan, estramustine, etoposide, nitrosoureas such as
carmustine and
lomustine, L-asparaginase, topotecan, nitrogen mustards, cytoxan, etoposide,
BCNU, vinca


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 69-

alkaloids, platinum compounds, mitomycin, gemcitabine, hexamethylmelamine,
temsirolimus
(CCI-779); lapatinib (GW 572016); RAD-001 (everolimus); XRP-9881; ixabepilone
(BMS-
247550); pertuzumab (OMNITARG, 2C4); topotecan, tyrosine kinase inhibitors,
tyrphostins,
imatinib mesylate (GLEEVEC), herbimycin A, genistein, erbstatin, and
lavendustin A.
In other embodiments, suitable chemotherapeutics include, but are not limited
to,
alkylating agents: nitrogen mustards (e.g., cyclophosphamide, ifosfamide,
trofosfamide,
chlorambucil); nitrosoureas (e.g., carmustine (BCNU), lomustine (CCNU));
alkylsulphonates
(e.g., busulfan, treosulfan); triazenes (e.g., dacarbazine); Platinum
containing compounds
(e.g., cisplatin, carboplatin, aroplatin, oxaliplatin); Plant Alkaloids: Vinca
alkaloids (e.g.,
vincristine, vinblastine, vindesine, vinorelbine); Taxoids (e.g., paclitaxel,
docetaxel; DNA
Topoisomerase Inhibitors: epipodophyllins (e.g., etoposide, teniposide,
topotecan, 9-
aminocamptothecin, camptothecin, crisnatol); mitomycins (e.g., mitomycin C,
anti-
metabolites); anti-folates: DHFR inhibitors (e.g., methotrexate, trimetrexate)
IMP
dehydrogenase Inhibitors (e.g., mycophenolic acid, tiazofurin, ribavirin,
EICAR); Ribonuclotide
reductase Inhibitors (e.g., hydroxyurea, deferoxamine); pyrimidine analogs:
uracil analogs
(e.g., 5-fluorouracil, floxuridine, doxifluridine, ratitrexed); cytosine
analogs (e.g., cytarabine
(ara C), cytosine arabinoside, fludarabine); purine analogs (e.g.,
mercaptopurine,
thioguanine); DNA antimetabolites (e.g., 3-HP, 2'-deoxy-5-fluorouridine, 5-HP,
alpha-TGDR,
aphidicolin glycinate, ara-C, 5-aza-2'-deoxycytidine, beta-TGDR,
cyclocytidine, guanazole,
inosine glycodialdehyde, macebecin II, pyrazoloimidazole); Hormonal therapies:
Receptor
antagonists: Anti-estrogen (e.g., tamoxifen, raloxifene, megestrol); aromatase
inhibitors (e.g.,
exemestane, anastrozole, letrozole); GnRH antagonists (e.g., abarelix,
histrelin); selective
estrogen receptor modulators (SERMs) (e.g., lasofoxifene); LH-RH agonists
(e.g., goserelin,
tryptorelin, buserelin, leuprolide acetate); Anti-androgens (e.g., flutamide,
bicalutamide,
nilutamide, megestrol, cyproterone); Retinoids/Deltoids cis-retinoic acid;
vitamin A derivative
(e.g., all-trans retinoic acid (ATRA-IV)); vitamin D3 analogs (e.g., EB 1089,
CB 1093, KH
1060); Photodynamic therapies (e.g., vertoporfin (BPD-MA), phthalocyanine,
photosensitizer
Pc4, demethoxy-hypocrellin A (2BA-2-DMHA); Cytokines, e.g., IL-1 a, IL-1(3, IL-
2, IL-3, IL-4,
IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-15, IL-18, IFNa, IFNR,
IFNy, TNFa, TNF(3, G-
CSF, GM-CSF, TGF-R, SLC, EMAP2, MIP-3a, MIP-3(3, HLA-B7, other members of the
TNF
family (e.g., TRAIL, TRANCE, TWEAK, CD40L, LT-a, LT-(3, OX40L, CD40L, FasL,
CD27L,
CD30L, 4-1BBL, APRIL, LIGHT, TL1, TNFSF16, TNFSF17, and AITR-L, or a
functional
portion thereof); Angiogenesis Inhibitors: angiostatin (plasminogen fragment),
antiangiogenic
antithrombin III, angiozyme, ABT-627, Bay 12-9566, benefin, bevacizumab, BMS-
275291,
cartilage-derived inhibitor (CDI), CAI, CD59 complement fragment, CEP-7055,
Col 3,
combretastatin A-4, endostatin (collagen XVIII fragment), fibronectin
fragment, Gro-beta,


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 70-

halofuginone, heparinases, heparin hexasaccharide fragment, HMV833, human
chorionic
gonadotropin (hCG), IM-862, interferon alpha/beta/gamma, interferon inducible
protein (IP-
10), interleukin-12, Kringle 5 (plasminogen fragment), marimastat,
metalloproteinase
inhibitors (TIMPs), 2-methoxyestradiol, MMI 270 (CGS 27023A), MoAb IMC-1C11,
neovastat
(Aeterna), NM-3, panzem, PI-88, placental ribonuclease inhibitor, plasminogen
activator
inhibitor, platelet factor-4 (PF4), prinomastat, prolactin 16kD fragment,
proliferin-related
protein (PRP), PTK 787/ZK 222594, retinoids, solimastat, squalamine, SS 3304,
SU 5416,
SU6668, SU11248, SU12662, SU14813, BAY 43-9006, AG-013736, tetrahydrocortisol-
S,
tetrathiomolybdate, thalidomide, thrombospondin-1 (TSP-1), TNP-470,
transforming growth
factor-beta (TGF-b), vasculostatin, vasostatin (calreticulin fragment),
ZD6126, ZD 6474,
farnesyl transferase inhibitors (FTI), bisphosphonates; Antimitotic agents
(e.g., allocolchicine,
halichondrin B, coichicine, coichicine derivative, dolstatin 10, maytansine,
rhizoxin,
thiocolchicine, trityl Cysteine); Other agents: isoprenylation inhibitors;
dopaminergic
neurotoxins (e.g., 1-methyl-4-phenylpyridinium ion); cell cycle inhibitors
(e.g., staurosporine):
actinomycins (e.g., actinomycin D, dactinomycin); bleomycins (e.g., bleomycin
A2, bleomycin
B2, peplomycin); anthracyclines (e.g., daunorubicin, doxorubicin (adriamycin),
idarubicin,
epirubicin, pirarubicin, zorubicin, mitoxantrone); mTOR inhibitors (e.g.,
temsirolimus,
everolimus); MDR inhibitors (e.g., verapamil); Ca2+ ATPase inhibitors (e.g.,
thapsigargin);
toll-like receptor agonists (e.g., CpG-7909, also known as PF03512676 or
PROMUNE; Coley
Pharm; reviewed in Krieg, 1998, In: Applied Oligonucleotide Technology, pp.
431-448, C.A.
Stein and A.M. Krieg, (Eds.), John Wiley and Sons, Inc., New York, NY);
costimulatory
molecules (e.g., CD4, CD25, PD-1, B7-H3, 4-1BB, OX40, ICOS, CD30, HLA-DR,
MHCII, and
LFA, and agonist antibodies thereto); among many other agents known in the
art.
Additional anti-cancer agents that may be used in the methods of the present
invention include, but are not limited to: acivicin; aclarubicin; acodazole
hydrochloride;
acronine; adozelesin; aidesleukin; altretamine; ambomycin; ametantrone
acetate; amifostine
trihydrate; aminoglutethimide; amsacrine; anastrozole; anthramycin; arsenic
trioxide;
asparaginase; asperlin; azacitidine; azetepa; azotomycin; Bacillus Calmette-
Guerin;
batimastat; benzodepa; bevacizumab; bicalutamide; bisantrene hydrochloride;
bisnafide
dimesylate; bizelesin; bleomycin sulfate; bortezomib; brequinar sodium;
bropirimine; busulfan;
cactinomycin; calusterone; capecitabine; caracemide; carbetimer; carboplatin;
carmustine;
carubicin hydrochloride; carzelesin; cedefingol; chlorambucil; cirolemycin;
cisplatin;
chlorambucil; cladribine; clodronate; crisnatol mesylate; cyclophosphamide;
cytarabine;
dacarbazine; dactinomycin; darbepoietin; daunorubicin hydrochloride;
decitabine;
dexormaplatin; dexrazoxane; dezaguanine; dezaguanine mesylate; diaziquone;
diethylstilbestrol; docetaxel; doxorubicin; doxorubicin hydrochloride;
droloxifene; droloxifene


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-71-
citrate; dromostanolone propionate; duazomycin; farmorubicin; edatrexate;
eflornithine
hydrochloride; elsamitrucin; enloplatin; enpromate; epipropidine; epirubicin
hydrochloride;
erbulozole; erlotinib; erythropoietin; esorubicin hydrochloride; estramustine;
estramustine
phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine;
everolimus;
exemestane; fadrozole hydrochloride; fazarabine; fenretinide; filgastrim (G-
CSF); floxuridine;
fludarabine phosphate; fludrocortisone; fluorouracil; fluoxyrnesterone;
flurocitabine;
fosquidone; fostriecin sodium; fulvestrant; gefitinib; gemcitabine;
gemcitabine hydrochloride;
gemtuzumab; goserelin; hydroxyurea; ibritumomab tiuxetan; idarubicin
hydrochloride;
ifosfamide; ilmofosine; imatinib; interieukin II (including recombinant
interieukin II, or rIL2),
interferon alfa-2a; interferon alfa-2b; interferon alfa-n1; interferon alfa-
n3; interferon beta-1a;
interferon gamma-1 b; iproplatin; irinotecan hydrochloride; ixabepilone;
ketoconazole;
lanreotide acetate; lapatinib; letrozole; leucovorin; leuprolide acetate;
levamisole; liarozole
hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride;
masoprocol;
maytansine; mechlorethamine hydrochloride; medroxyprogesterone; megestrol
acetate;
melengestrol acetate; melphalan; menogaril; mercaptopurine; mesna;
methotrexate;
methotrexate sodium; metoprine; meturedepa; mitindomide; mitocarcin;
mitocromin; mitogillin;
mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride;
mycophenolic acid;
nocodazole; nogalamycin; octreotide; ormaplatin; oxaliplatin; oxisuran;
paclitaxel;
pamidronate; pegaspargase; PEG-L-asparaginase; PEG-filgastrim; peliomycin;
pentamustine;
pentostatin; peplomycin sulfate; perfosfamide; pertuzumab; pipobroman;
piposulfan;
piroxantrone hydrochloride; plicamycin; plomestane; porfimer 'sodium;
porfimer; porfiromycin;
prednimustine; pemetrexed; procarbazine hydrochloride; puromycin; puromycin
hydrochloride; pyrazofurin; raltitrexed; riboprine; rituximab; rogietimide;
safingol; safingol
hydrochloride; semustine; simtrazene; somavert (PEGVISOMANT); sparfosate
sodium;
sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin;
streptonigrin;
streptozocin; sulofenur; sunitinib; streptozocin; talisomycin; tecogalan
sodium; tegafur;
teloxantrone hydrochloride; temoporfin; temozolomide; temsirolimus;
teniposide; teroxirone;
testolactone; thalidomide; thiamiprine; thioguanine; thiotepa; tiazofurin;
tirapazamine;
topotecan; toremifene citrate; trastuzumab; tretinoin; trestolone acetate;
triciribine phosphate;
trimetrexate; trimetrexate glucuronate; triptorelin; topotecan; tubulozole
hydrochloride; uracil
mustard; uredepa; vapreotide; verteporfin; vinblastine sulfate; vincristine
sulfate; vindesine;
vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine
sulfate; vinorelbine
tartrate; vinrosidine sulfate; vinzolidine sulfate; vorozole; zeniplatin;
zinostatin; zolendronate;
zorubicin hydrochloride.
Other anti-cancer drugs that can be used include, but are not limited to: 20-
epi-1,25
dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene;
adecypenol;


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 72-

adozelesin; aldesieukin; ALL-TK antagonists; altretamine; ambamustine; amidox;
amifostine;
aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole;
andrographolide;
angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-
dorsalizing morphogenetic
protein-1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston;
antisense
oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis
regulators;
apurinic acid; ara-CDP-DL-PTBA; arginine deaminase; asulacrine; atamestane;
atrimustine;
axinastatin 1; axinastatin 2; axinastatin 3; azasetron; azatoxin; azatyrosine;
baccatin III
derivatives; balanol; batimastat; BCR/ABL antagonists; benzochlorins;
benzoylstaurosporine;
beta lactam derivatives; beta-alethine; betaclamycin B; betulinic acid; bFGF
inhibitor;
bicalutamide; bisantrene; bisaziridinyispermine; bisnafide; bistratene A;
bizelesin; breflate;
bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C;
camptothecin
derivatives; canarypox IL-2; capecitabine; carboxamide-amino-triazole;
carboxyamidotriazole;
CaRest M3; CARN 700; cartilage derived inhibitor; carzelesin; casein kinase
inhibitors
(ICOS); castanospermine; cecropin B; cetrorelix; chlorlns; chloroquinoxaline
sulfonamide;
cicaprost; cis-porphyrin; cladribine; clomifene analogues; clotrimazole;
collismycin A;
collismycin B; combretastatin A4; combretastatin analogue; conagenin;
crambescidin 816;
crisnatol; cryptophycin 8; cryptophycin A derivatives; curacin A;
cyclopentanthraquinones;
cycloplatam; cypemycin; cytarabine ocfosfate; cytolytic factor; cytostatin;
dacliximab;
decitabine; dehydrodidemnin B; deslorelin; dexamethasone; dexifosfamide;
dexrazoxane;
dexverapamil; diaziquone; didemnin B; didox; diethyinorspermine; dihydro-5-
azacytidine;
dihydrotaxol, 9-; dioxamycin; diphenyl spiromustine; docetaxel; docosanol;
dolasetron;
doxifluridine; droloxifene; dronabinol; duocarmycin SA; ebselen; ecomustine;
edelfosine;
edrecolomab; eflornithine; elemene; emitefur; epirubicin; epristeride;
estramustine analogue;
estrogen agonists; estrogen antagonists; etanidazole; etoposide phosphate;
exemestane;
fadrozole; fazarabine; fenretinide; filgrastim; finasteride; flavopiridol;
flezelastine; fluasterone;
fludarabine; fluorodaunorunicin hydrochloride; forfenimex; formestane;
fostriecin; fotemustine;
gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase
inhibitors;
gemcitabine; glutathione inhibitors; hepsulfam; heregulin; hexamethylene
bisacetamide;
hypericin; ibandronic acid; idarubicin; idoxifene; idramantone; ilmofosine;
ilomastat;
imidazoacridones; imiquimod; immunostimulant peptides; insulin-like growth
factor-1 receptor
inhibitor; interferon agonists; interferons; interleukins; iobenguane;
iododoxorubicin;
ipomeanol, 4-; iroplact; irsogladine; isobengazole; isohomohalicondrin B;
itasetron;
jasplakinolide; kahaialide F; lamellarin-N triacetate; lanreotide; leinamycin;
lenograstim;
lentinan sulfate; leptolstatin; letrozole; leukemia inhibiting factor;
leukocyte alpha interferon;
leuprolide+estrogen+progesterone; leuprorelin; levamisole; liarozole; linear
polyamine
analogue; lipophilic disaccharide peptide; lipophilic platinum compounds;
lissoclinamide 7;


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 73-

lobaplatin; lombricine; lometrexol; lonidamine; losoxantrone; lovastatin;
loxoribine; lurtotecan;
lutetium texaphyrin; lysofylline; lytic peptides; maitansine; mannostatin A;
marimastat;
masoprocol; maspin; matrilysin inhibitors; matrix metalloproteinase
inhibitors; menogaril;
merbarone; meterelin; methioninase; metoclopramide; MIF inhibitor;
mifepristone; miltefosine;
mirimostim; mismatched double stranded RNA; mitoguazone; mitolactol; mitomycin
analogues; mitonafide; mitotoxin fibroblast growth factor-saporin;
mitoxantrone; mofarotene;
molgramostim; monoclonal antibody, human chorionic gonadotrophin;
monophosphoryl lipid
A+myobacterium cell wall sk; mopidamol; multiple drug resistance gene
inhibitor; multiple
tumor suppressor 1-based therapy; mustard anti-cancer agent; mycaperoxide B;
mycobacterial cell wall extract; myriaporone; N-acetyldinaline; N-substituted
benzamides;
nafarelin; nagrestip; naloxone+pentazocine; napavin; naphterpin; nartograstim;
nedaplatin;
nemorubicin; neridronic acid; neutral endopeptidase; nilutamide; nisamycin;
nitric oxide
modulators; nitroxide antioxidant; nitrullyn; 06-benzylguanine; octreotide;
okicenone;
oligonucleotides; onapristone; ondansetron; ondansetron; oracin; oral cytokine
inducer;
ormaplatin; osaterone; oxaliplatin; oxaunomycin; paclitaxel; paclitaxel
analogues; paclitaxel
derivatives; palauamine; palmitoylrhizoxin; pamidr,onic acid; panaxytriol;
panomifene;
parabactin; pazelliptine; pegaspargase; peldesine; pentosan polysulfate
sodium; pentostatin;
pentrozole; perflubron; perfosfamide; perillyl alcohol; phenazinomycin;
phenylacetate;
phosphatase inhibitors; picibanil; pilocarpine hydrochloride; pirarubicin;
piritrexim; placetin A;
placetin B; plasminogen activator inhibitor; platinum complex; platinum
compounds; platinum-
triamine complex; porflmer sodium; porfiromycin; prednisone; propyl bis-
acridone;
prostaglandin J2; proteasome inhibitors; protein A-based immune modulator;
protein kinase C
inhibitor; protein kinase C inhibitors, microalgal; protein tyrosine
phosphatase inhibitors;
purine nucleoside phosphorylase inhibitors; purpurins; pyrazoloacridine;
pyridoxylated
hemoglobin polyoxyethylene conjugate; raf antagonists; raltitrexed;
ramosetron; ras famesyl
protein transferase inhibitors; ras inhibitors; ras-GAP inhibitor;
retelliptine demethylated;
rhenium Re186 etidronate; rhizoxin; ribozymes; RII retinamide; rogietimide;
rohitukine;
romurtide; roquinimex; rubiginone 131; ruboxyl; safingol; saintopin; SarCNU;
sarcophytol A;
sargramostim; Sdi 1 mimetics; semustine; senescence derived inhibitor 1; sense
oligonucleotides; signal transduction inhibitors; signal transduction
modulators; single chain
antigen binding protein; sizofuran; sobuzoxane; sodium borocaptate; sodium
phenylacetate;
solverol; somatomedin binding protein; sonermin; sparfosic acid; spicamycin D;
spiromustine;
splenopentin; spongistatin 1; squalamine; stem cell inhibitor; stem-cell
division inhibitors;
stipiamide; stromelysin inhibitors; sulfinosine; superactive vasoactive
intestinal peptide
antagonist; suradista; suramin; swainsonine; synthetic glycosaminoglycans;
tallimustine;
tamoxifen methiodide; tauromustine; tazarotene; tecogalan sodium; tegafur;
tellurapyrylium;


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 74-

telomerase inhibitors; temoporfin; temozolomide; teniposide;
tetrachlorodecaoxide;
tetrazomine; thaliblastine; thiocoraline; thrombopoietin; thrombopoietin
mimetic; thymalfasin;
thymopoietin receptor agonist; thymotrinan; thyroid stimulating hormone; tin
ethyl
etiopurpurin; tirapazamine; titanocene bichloride; topsentin; toremifene;
totipotent stem cell
factor; translation inhibitors; tretinoin; triacetyluridine; triciribine;
trimetrexate; triptorelin;
tropisetron; turosteride; tyrosine kinase inhibitors; tyrphostins; UBC
inhibitors; ubenimex;
urogenital sinus-derived growth inhibitory factor; urokinase receptor
antagonists; vapreotide;
variolin B; vector system, erythrocyte gene therapy; velaresol; veramine;
verdins; verteporfin;
vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeniplatin; zilascorb;
and zinostatin
stimalamer.
In another specific embodiment, a combination comprising an anti-CTLA4
antibody
and a second therapeutic agent is administered to a subject receiving
radiation therapy for
treatment of cancer. For radiation treatment, the radiation can be gamma rays
or X-rays. The
methods encompass treatment of cancer comprising radiation therapy, such as
external-
beam radiation therapy, interstitial implantation of radioisotopes (1-125,
palladium, iridium),
radioisotopes such as strontium-89, thoracic radiation therapy,
intraperitoneal P-32 radiation
therapy, and/or total abdominal and pelvic radiation therapy. In preferred
embodiments, the
radiation treatment is administered as external beam radiation or teletherapy
wherein the
radiation is directed from a remote source. In various preferred embodiments,
the radiation
treatment is administered as internal therapy or brachytherapy wherein a
radiaoactive source
is placed inside the body close to cancer cells or a tumor mass. Radiation
therapy can be
administered in accordance to well-known radiotherapy methods for treatment of
cancer. The
dose and regimen for radiotherapy can be readily determined by one skilled in
the art and is
based on the stage of the disease, and other factors well-known in the art.
For a general
overview of radiation therapy, see Hellman, In: Principles of Cancer
Management: Radiation
Therapy, Chapter 16, DeVita et al., eds., 6th edition, 2001, J. B. Lippencott
Company,
Philadelphia.
Co-administration of the antibody with at least one therapeutic agent
(combination
therapy) encompasses administering a pharmaceutical composition comprising
both the anti-
CTLA4 antibody and one or more therapeutic agent(s), and administering two or
more
separate pharmaceutical compositions, one comprising the anti-CTLA4 antibody
and the
other(s) comprising the therapeutic agent(s). Further, although co-
administration or
combination (conjoint) therapy generally mean that the antibody and additional
therapeutic
agents are administered at the same time as one another, it also encompasses
simultaneous,
sequential or separate dosing of the individual components of the treatment.
Additionally,
where an antibody is administered intravenously and the therapeutic agent is
administered


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 75-

orally (e.g., exemestane), it is understood that their combination is
preferably administered as
two separate pharmaceutical compositions.
The present invention also encompasses the administration of other therapeutic
agents in addition to the antibody and first therapeutic agent, either
concurrently with one or
more of those components, 'or sequentially before and/or after. Such
therapeutic agents
include cancer vaccines, anti-vascular agents, anti-proliferative agents,
antiagionesis agents,
signal transduction inhibitors, immunomodulatory agents, cytokines, and
palliative agents to
provide supportive care, such as, but not limited to, analgesics, anti-emetic
agents, anti-
diarrheal agents, and steroids. Preferred anti-emetic agents include
ondansetron
hydrochloride, granisetron hydrochloride, and metoclopramide. Preferred anti-
diarrheal
agents include diphenoxylate and atropine (LOMOTIL), loperamide (IMMODIUM),
octreotide
(SANDOSTATIN), oisalazine (DIPENTUM), and mesalamine (ASACOL). Preferred
steroids
include the non-absorbable steroid budesonide (ENTOCORT), and the steroids for
systemic
administration dexametasone (DECADRON) and prednisone (METICORTEN).
Each administration may vary in its duration from a rapid administration to a
continuous perfusion. As a result, for the purposes of the present invention,
the combinations
are not exclusively limited to those that are obtained by physical association
of the
constituents, but also to those that permit a separate administration, which
can be
simultaneous or spaced out over a period of time. The compositions according
to the
invention are preferably compositions which can be administered parentally.
However, these
compositions may be administered orally or intraperitoneally in the case of
localized regional
therapies.
As will be appreciated by one of skill in the art, the choice of therapeutic
agents to be
used in combination with anti-CTLA4 antibodies, and the timing of their use,
will be
determined in part by the type and stage of the cancer that is being treated.
The methods of the present invention are suitable for use both as first line
therapy
and second line therapy. Treatment of both early stage and advanced
(metastatic) cancers
are within the scope of the present invention.

V. Pharmaceutical Compositions
The invention encompasses the preparation and use of pharmaceutical
compositions
comprising a human anti-CTLA4 antibody of the invention as an active
ingredient in
combination with a therapeutic agent, e.g., a chemotherapeutic agent, another
antibody, an
immunostimulatory agent, a signal transduction inhibitor, an angiogenesis
inhibitor,
preferably, the therapeutic agent is a chemotherapeutic agent. Such a
pharmaceutical
composition may consist of each active ingredient alone, as a combination of
at least one


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 76-

active ingredient (e.g., an effective dose of an anti-CTLA4, an effective does
of a therapeutic
agent) in a form suitable for administration to a subject, or the
pharmaceutical composition
may comprise the active ingredient and one or more pharmaceutically acceptable
carriers,
one or more additional (active and/or inactive) ingredients, or some
combination of these.
In one embodiment, the antibody is administered parenterally (e.g.,
intravenously) in
an aqueous solution while the therapeutic agent (e.g., a chemotherapeutic
agent, a signal
transduction inhibitor, an immunomodulator, an angiogenesis inhibitor, and the
like) is
administered orally in pill/capsule form. However, the skilled artisan would
understand, based
upon the disclosure provided herein, that the invention is not limited to
these, or any other,
formulations, doses, routes of administration, and the like. Rather, the
invention
encompasses any formulation or method of administering an antibody in
combination with a
therapeutic agent, including, but not limited to, administering each agent
separately in a
different formulation via a different route of administration, and
administering the antibody and
the therapeutic agent in a single composition (e.g. where the therapeutic
agent is a protein,
such as, another antibody, a cytokine, a costim, and the like), in an aqueous
composition
administered, inter alia, intravenously), among many others. Thus, the
following discussion
describes various formulations for practicing the methods of the invention
comprising
administration of any anti-CTLA4 antibody in combination with any therapeutic
agent, but the
invention is not limited to these formulations, but comprises any formulation
as can be readily
determined by one skilled in the art once armed with the teachings provided
herein for use in
the methods of the invention.
In certain embodiments, the antibodies may be present in a neutral form
(including
zwitter ionic forms) or as a positively or negatively-charged species. In some
embodiments,
the antibodies may be complexed with a counterion to form a pharmaceutically
acceptable
salt.
The terms "pharmaceutically acceptable salt" refer to a complex comprising one
or
more antibodies and one or more counterions, where the counterions are derived
from
pharmaceutically acceptable inorganic and organic acids and bases.
Pharmaceutically acceptable inorganic bases include metallic ions. More
preferred
metallic ions include, but are not limited to, appropriate alkali metal salts,
alkaline earth metal
salts and other physiological acceptable metal ions. Salts derived from
inorganic bases
include aluminum, ammonium, calcium, cobalt, nickel, molybdenum, vanadium,
manganese,
chromium, selenium, tin, copper, ferric, ferrous, lithium, magnesium, manganic
salts,
manganous, potassium, rubidium, sodium, and zinc, and in their usual valences.
Pharmaceutically acceptable acid addition salts of the antibodies of the
present
invention can be prepared from the following acids, including, without
limitation formic, acetic,


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 77-

acetamidobenzoic, adipic, ascorbic, boric, propionic, benzoic, camphoric,
carbonic, cyclamic,
dehydrocholic, malonic, edetic, ethylsulfuric, fendizoic, metaphosphoric,
succinic, glycolic,
gluconic, lactic, malic, tartaric, tannic, citric, nitric, ascorbic,
glucuronic, maleic, folic, fumaric,
propionic, pyruvic, aspartic, glutamic, benzoic, hydrochloric, hydrobromic,
hydroiodic, lysine,
isocitric, trifluoroacetic, pamoic, propionic, anthranilic, mesylic, orotic,
oxalic, oxalacetic, oleic,
stearic, salicylic, aminosalicylic, silicate, p-hydroxybenzoic, nicotinic,
phenylacetic, mandelic,
embonic, sulfonic, methanesulfonic, phosphoric, phosphonic, ethanesulfonic,
ethanedisulfonic, ammonium, benzenesulfonic, pantothenic, naphthalenesulfonic,
toluenesulfonic, 2-hydroxyethanesulfonic, sulfanilic, sulfuric, nitric,
nitrous, sulfuric acid
monomethyl ester, cyclohexylaminosulfonic, R-hydroxybutyric, glycine,
glycylglycine, glutamic,
cacodylate, diaminohexanoic, camphorsulfonic, gluconic, thiocyanic,
oxoglutaric, pyridoxal 5-
phosphate, chlorophenoxyacetic, undecanoic, N-acetyl-L-aspartic, galactaric
and galacturonic
acids.
Pharmaceutically acceptable organic bases include trimethylamine,
diethylamine, N,
N'-dibenzylethylenediamine, chloroprocaine, choline, dibenzylamine,
diethanolamine,
ethylenediamine, megiumine (N-methylglucamine), procaine, cyclic amines,
quaternary
ammonium cations, arginine, betaine, caffeine, clemizole, 2-ethylaminoethanol,
2-
diethylaminoethanol, 2-dimethylaminoethanol, ethanediamine, butylamine,
ethanolamine,
ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, ethylglucamine,
glucamine,
glucosamine, histidine, hydrabamine, imidazole, isopropylamine,
methylglucamine,
morpholine, piperazine, pyridine, pyridoxine, neodymium, piperidine, polyamine
resins,
procaine, purines, theobromine, triethylamine, tripropylamine,
triethanolamine, tromethamine,
methylamine, taurine, cholate, 6-amino-2-methyl-2-heptanol, 2-amino-2-methyl-
1,3-
propanediol, 2-amino-2-methyl-l-propanol, aliphatic mono- and dicarboxylic
acids, phenyl-
substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic
and aromatic
sulfonic acids, strontium, tricine, hydrazine, phenylcyclohexylamine, 2-(N-
morpholino)ethanesulfonic acid, bis(2-hydroxyethyl)amino-
tris(hydroxymethyl)methane, N-(2-
acetamido)-2-aminoethanesulfonic acid, 1,4-piperazinediethanesulfonic acid, 3-
morpholino-2-
hydroxypropanesulfonic acid, 1,3-bis[tris(hydroxymethyl)methylamino]propane, 4-

morpholinepropanesulfonic acid, 4-(2-hydroxyethyl)piperazine-l-ethanesulfonic
acid, 2-[(2-
hydroxy-1,1-bis(hydroxymethyl)ethyl)amino]ethanesulfonic acid, N,N-bis(2-
hydroxyethyl)-2-
aminoethanesulfonic acid, 4-(N-morpholino)butanesulfonic acid, 3-(N,N-bis[2-
hydroxyethyl]amino)-2-hydroxypropanesulfonic acid, 2-hydroxy-3-
[tris(hydroxymethyl)methylamino]-1-propanesulfonic acid, 4-(2-
hydroxyethyl)piperazine-l-(2-
hydroxypropanesulfonic acid), piperazine-1,4-bis(2-hydroxypropanesulfonic
acid) dihydrate,
4-(2-hydroxyethyl)-1-piperazinepropanesulfonic acid, N,N-bis(2-
hydroxyethyl)glycine, N-(2-


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 78-
hydroxyethyl)piperazine-N'-(4-butanesulfonic acid), N-
[tris(hydroxymethyl)methyl]-3-
aminopropanesulfonic acid, N-tris(Hydroxymethyl)methyl-4-aminobutanesulfonic
acid, N-(1,1-
dimethyl-2-hydroxyethyl)-3-amino-2-hydroxypropanesulfonic acid, 2-
(cyclohexylamino)ethanesulfonic acid, 3-(cyclohexylamino)-2-hydroxy-l-
propanesulfonic acid,
3-(cyclohexylamino)-1-propanesulfonic acid, N-(2-acetamido)iminodiacetic acid,
4-
(cyclohexylamino)-1-butanesulfonic acid, N-[tris(hydroxymethyl)methyl]glycine,
2-amino-2-
(hydroxymethyl)-1,3-propanediol, and trometamol.
The anti-CTLA4 antibody used in the methods of the invention can be
incorporated into
pharmaceutical compositions suitable for administration to a subject.
Typically, the
pharmaceutical composition comprises the antibody and a pharmaceutically
acceptable carrier.
As used herein, "pharmaceutically acceptable carrier" includes any and all
solvents, dispersion
media, coatings, antibacterial and antifungal agents, isotonic and absorption
delaying agents,
and the like that are physiologically compatible. Examples of pharmaceutically
acceptable
carriers include one or more of water, saline, phosphate buffered saline,
dextrose, glycerol,
ethanol and the like, as well as combinations thereof. In many cases, it will
be preferable to
include isotonic agents, for example, sugars, polyalcohols such as mannitol,
sorbitol, or sodium
chloride in the composition. Pharmaceutically acceptable substances such as
wetting or minor
amounts of auxiliary substances such as wetting or emulsifying agents,
preservatives or buffers,
which enhance the shelf life or effectiveness of the antibody or antibody
portion.
The antibodies and therapeutic agents used in the present invention may be in
a variety
of forms. These include, for example, liquid, semi solid and solid dosage
forms, such as liquid
solutions (e.g., injectable and infusible solutions), dispersions or
suspensions, tablets, pills,
powders, liposomes and suppositories. The preferred form depends on the
therapeutic agent,
the intended mode of administration and the therapeutic application. Typical
preferred
compositions are in the form of injectable or infusible solutions, such as
compositions similar to
those used for passive immunization of humans with other antibodies. The
preferred mode of
administration is parenteral (e.g., intravenous, subcutaneous,
intraperitoneal, intramuscular). In
a preferred embodiment, the antibody is administered by intravenous infusion
or injection. In
another preferred embodiment, the antibody is administered by intramuscular or
subcutaneous
injection.
Therapeutic compositions typically must be sterile and stable under the
conditions of
manufacture and storage. The composition can be formulated as a solution,
microemulsion,
dispersion, liposome, or other ordered structure suitable to high drug
concentration. Sterile
injectable solutions can be prepared by incorporating the antibody in the
required amount in an
appropriate solvent with one or a combination of ingredients enumerated above,
as required,
followed by filtered sterilization. Generally, dispersions are prepared by
incorporating the active


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 79-

compound into a sterile vehicle that contains a basic dispersion medium and
the required other
ingredients from those enumerated above. In the case of sterile powders for
the preparation of
sterile injectable solutions, the preferred methods of preparation are vacuum
drying and freeze
drying that yields a powder of the active ingrpdient pius any additional
desired ingredient from a
previously sterile filtered solution thereof. The proper fluidity of a
solution can be maintained, for
example, by the use of a coating such as lecithin, by the maintenance of the
required particle
size in the case of dispersion and by the use of surfactants. Prolonged
absorption of injectable
compositions can be brought about by including in the composition an agent
that delays
absorption, for example, monostearate salts and gelatin.
The antibodies can be administered by a variety of methods known in the art,
including,
without limitation, oral, parenteral, mucosal, by-inhalation, topical, buccal,
nasal, and rectal. For
many therapeutic applications, the preferred route/mode of administration is
subcutaneous,
intramuscular, intravenous or infusion. Non-needle injection may be employed,
if desired. As
will be appreciated by the skilled artisan, the route and/or mode of
administration will vary
depending upon the desired results.
In certain embodiments, the antibody may be prepared with a carrier that will
protect
the compound against rapid release, such as a controlled release formulation,
including
implants, transdermal patches, and microencapsulated delivery systems.
Biodegradable,
biocompatible polymers can be used, such as ethylene vinyl acetate,
polyanhydrides,
polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many
methods for the
preparation of such formulations are patented or generally known to those
skilled in the art. See,
e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson,
ed., Marcel
Dekker, Inc., New York (1978).
Transdermal delivery of biological molecules, including antibodies, cytokines,
and the
like, includes microneedles to create micron-sized temporary transport
pathways in the skin
through which large molecules can be transported. See, e.g., Banga, In:
Therapeutic peptides
and proteins: Formulation, processing and delivery systems, 2"d ed., Taylor &
Francis (Eds.),
2006; Martanto et al., J. Controlled Release 112:357-361 (2006). In one
embodiment,
microneedies include those composed of silicon, metal or a polymer. In another
embodiment,
maltose microneedles which dissolve in the skin may be used to administered
the compositions
of the invention, including anti-CTLA4 antibody, anti-CD40, anti-IGF-1 R
antibody, rituximab,
trastuzumab, IL-15, and the like.
In yet another embodiment, nucleic acid, including nucleic acids encoding a
protein,
preferably, anti-CTLA4 antibody (e.g., CP-675,206, ipilimumab, and the like),
or an ODN, can
be delivered transdermally using biolistics to provide expression of the
antibody or delivery of
the ODN in a micromilieu where antigen presenting cells (e.g., dendritic
cells) and/or T


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 80-

regulatory cells are present (e.g., Tregs in lymph nodes). Gold microparticles
comprising
nucleic acids encoding anti-CTLA4 antibody and/or comprising an ODN or nucleic
acids
encoding a protein of interest, are propelled through the skin using, for
example, a "gene gun"
(PMED Device, e.g., model ND-10, PowderMed Ltd., Oxford, UK). See, e.g., US
Patent Nos.
6,194,389; 6,168,587; 5,478,744. In one embodiment, a synthetic
oligodeoxynucleotide (ODN)
toll-like receptor 9 agonist (e.g., PF03512676) may be administered using
biolistics such that a
microparticle comprising the nucleic acid encoding the antibody and a
microparticle comprising
the ODN, or a microparticle comprising both, are both delivered transdermally.
In another
embodiment, a nucleic acid encoding a TAA (e.g., gp100, tyrosinase, MAGE, MART-
1, NY-
ESO-1, and the like) is delivered transdermally using biolistics such that the
TAA is expressed
in and presented by APCs transfected with the nucleic acid. In yet another
embodiment,
various combinations of an antigen (e.g., TAA), an antibody (anti-CTLA4, anti-
CD40, anti-CD20,
anti-HER2, etc.), a cytokine (e.g., GM-CSF, IL-15, TNFa, among others), an ODN
(e.g.,
PF03512676), a costimulatory molecule (e.g., PD-1, OX40, 4-1 BB, etc.), and
the like, may be
delivered using biolistics to induce, enhance or prolong and anti-tumor
response in a patient.
Dosage regimens may be adjusted to provide the optimum desired 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. It is especially advantageous to formulate
parenteral compositions in
dosage unit form for ease of administration and uniformity of dosage. Dosage
unit form as used
herein refers to physically discrete units suited as uhitary dosages for the
mammalian subjects
to be treated; each unit containing a predetermined quantity of active
compound calculated to
produce the desired therapeutic effect in association with the required
pharmaceutical carrier.
The specification for the dosage unit forms of the invention are dictated by
and directly
dependent on (a) the unique characteristics of the antibody and the particular
therapeutic or
prophylactic effect to be achieved, and (b) the limitations inherent in the
art of compounding
such an active compound for the treatment of sensitivity in individuals.
It is to be noted that dosage values may vary with the type and severity of
the condition
to be alleviated, and may include single or multiple doses. It is to be
further understood that for
any particular subject, specific dosage regimens should be adjusted over time
according to the
individual need and the professional judgment of the person administering or
supervising the
administration of the compositions, and that dosage ranges set forth herein
are exemplary only
and are not intended to limit the scope or practice of the claimed
composition.
In one embodiment, the antibody is administered in a formulation as a sterile
aqueous
solution having a pH that ranges from about 5.0 to about 6.5 and comprising
from about 1
mg/mi to about 200 mg/mi of antibody, from about I millimolar to about 100
millimolar of


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 81-

histidine buffer, from about 0.01 mg/m) to about 10 mg/mI of polysorbate 80,
from about 100
millimolar to about 400 millimolar of trehalose, and from about 0.01
millimolar to about 1.0
millimolar of disodium EDTA dihydrate. Exemplary formulations encompass, but
are not limited
to, such formulations described in PCT/US2006/007551 filed March 2, 2006, now
published as
WO 2006/096488 on Sept. 14, 2006, and PCT/US2006/007555 filed March 2, 2006,
now
published as WO 2006/096491 on Sept. 14, 2006.
In another embodiment, the antibody is administered in an intravenous
formulation as a
sterile aqueous solution containing 5 mg/m, or more preferably, about 10
mg/mI, or yet more
preferably, about 15 mg/mI, or even more preferably, about 20 mg/ml of
antibody, with sodium
acetate, polysorbate 80, and sodium chloride at a pH ranging from about 5 to
6. Preferably, the
intravenous formulation is a sterile aqueous solution containing 5 or 10 mg/ml
of antibody, with
mM sodium acetate, 0.2 mg/mI polysorbate 80, and 140 mM sodium chloride at pH
5.5.
Further, a solution comprising an anti-CTLA4 antibody can comprise, among many
other
compounds, histidine, mannitol, sucrose, trehalose, glycine, poly(ethylene)
glycol, EDTA,
15 methionine, and any combination thereof, and many other compounds known in
the relevant
art.
The compositions of the present invention optionally may further comprise a
pharmaceutically acceptable antioxidant in addition to a chelating agent.
Suitable
antioxidants include, but are not limited to, methionine, sodium thiosulfate,
catalase, and
20 platinum. For example, the composition may contain methionine in a
concentration that
ranges from 1 mM to about 100 mM, and in particular, is about 27 mM.
In one embodiment, part of the dose is administered by an intraveneous bolus
and the
rest by infusion of the antibody formulation. For example, a 0.01 mg/kg
intravenous injection of
the antibody may be given as a bolus, and the rest of a predetermined antibody
dose may be
administered by intravenous injection. A predetermined dose of the antibody
may be
administered, for example, over a period of an hour and a half to two hours to
five hours.
With regard to a therapeutic agent, where the agent is, e.g., a small
molecule, it can
be present in a pharmaceutical composition in the form of a physiologically
acceptable ester
or salt, such as in combination with a physiologically acceptable cation or
anion, as is well
known in the art.
The formulations of the pharmaceutical compositions described herein may be
prepared by any method known or hereafter developed in the art of
pharmacology. In
general, such preparatory methods include the step of bringing the active
ingredient into
association with a carrier or one or more other accessory ingredients, and
then, if necessary
or desirable, shaping or packaging the product into a desired single- or multi-
dose unit.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 82-

Pharmaceutical compositions that are useful in the methods of the invention
may be
prepared, packaged, or sold in formulations suitable for oral, rectal,
vaginal, parenteral,
topical, pulmonary, intranasal, buccal, ophthalmic, or another route of
administration. Other
contemplated formulations include projected nanoparticies, liposomal
preparations, resealed
erythrocytes containing the active ingredient, and immunologically-based
formulations.
A pharmaceutical composition of the invention may be prepared, packaged, or
sold in
bulk, as a single unit dose, or as a plurality of single unit doses. As used
herein, a "unit dose"
is discrete amount of the pharmaceutical composition comprising a
predetermined amount of
the active ingredient. The amount of the active ingredient is generally equal
to the dosage of
the active ingredient which would be administered to a subject or a convenient
fraction of
such a dosage such as, for example, one-half or one-third of such a dosage.
The relative amounts of the active ingredient, the pharmaceutically acceptable
carrier,
and any additional ingredients in a pharmaceutical composition of the
invention will vary,
depending upon the identity, size, and condition of the subject treated and
further depending
upon the route by which the composition is to be administered. By way of
example, the
composition may comprise between 0.1% and 100% (w/w) active ingredient.
In addition to the active ingredient, a pharmaceutical composition of the
invention
may further comprise one or more additional pharmaceutically active
therapeutic agents.
Particularly contemplated additional agents include anti-emetics, anti-
diarrheals,
chemotherapeutic agents, cytokines, and the like.
Controlled- or sustained-release formulations of a pharmaceutical composition
of the
invention may be made using conventional technology.
A formulation of a pharmaceutical composition of the invention suitable for
oral
administration may be prepared, packaged, or sold in the form of a discrete
solid dose unit
including, but not limited to, a tablet, a hard or soft capsule, a cachet, a
troche, or a lozenge,
each containing a predetermined amount of the active ingredient. Other
formulations suitable
for oral administration include, but are not limited to, a powdered or
granular formulation, an
aqueous or oily suspension, an aqueous or oily solution, or an emulsion.
As used herein, an "oily" liquid is one which comprises a carbon-containing
liquid
molecule and which exhibits a less polar character than water.
A tablet comprising the active ingredient may, for example, be made by
compressing
or molding the active ingredient, optionally with one or more additional
ingredients.
Compressed tablets may be prepared by compressing, in a suitable device, the
active
ingredient in a free-flowing form such as a powder or granular preparation,
optionally mixed
with one or more of a binder, a lubricant, an excipient, a surface active
agent, and a
dispersing agent. Molded tablets may be made by molding, in a suitable device,
a mixture of


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 83-

the active ingredient, a pharmaceutically acceptable carrier, and at least
sufficient liquid to
moisten the mixture.
Pharmaceutically acceptable excipients used in the manufacture of tablets
include,
but are not limited to, inert diluents, granulating and disintegrating agents,
binding agents, and
lubricating agents. Known dispersing agents include, but are not limited to,
potato starch and
sodium starch glycolate. Known surface active agents include, but are not
limited to, sodium
lauryl sulphate. Known diluents include, but are not limited to, calcium
carbonate, sodium
carbonate, lactose, microcrystalline cellulose, calcium phosphate, calcium
hydrogen
phosphate, and sodium phosphate. Known granulating and disintegrating agents
include, but
are not limited to, corn starch and alginic acid. Known binding agents
include, but are not
limited to, gelatin, acacia, pre-gelatinized maize starch,
polyvinylpyrrolidone, and
hydroxypropyl methylcellulose. Known lubricating agents include, but are not
limited to,
magnesium stearate, stearic acid, silica, and talc.
Tablets may be non-coated or they may be coated using known methods to achieve
delayed disintegration in the gastrointestinal tract of a subject, thereby
providing sustained
release and absorption of the active ingredient. By way of example, a material
such as
glyceryl monostearate or glyceryl distearate may be used to coat tablets.
Further by way of
example, tablets may be coated using methods described in U.S. Patents numbers
4,256,108; 4,160,452; and 4,265,874 to form osmotically-controlled release
tablets. Tablets
may further comprise a sweetening agent, a flavoring agent, a coloring agent,
a preservative,
or some combination of these in order to provide pharmaceutically elegant and
palatable
preparation.
Hard capsules comprising the active ingredient may be made using a
physiologically
degradable composition, such as gelatin. Such hard capsules comprise the
active ingredient,
and may further comprise additional ingredients including, for example, an
inert solid diluent
such as calcium carbonate, calcium phosphate, or kaolin.
Soft gelatin capsules comprising the active ingredient may be made using a
physiologically degradable composition, such as gelatin. Such soft capsules
comprise the
active ingredient, which may be mixed with water or an oil medium such as
peanut oil, liquid
paraffin, or olive oil.
Liquid formulations of a pharmaceutical composition of the invention which are
suitable for oral administration may be prepared, packaged, and sold either in
liquid form or in
the form of a dry product intended for reconstitution with water or another
suitable vehicle
prior to use.
Liquid suspensions may be prepared using conventional methods to achieve
suspension of the active ingredient in an aqueous or oily vehicle. Aqueous
vehicles include,


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 84-

for example, water and isotonic saline. Oily vehicles include, for example,
almond oil, oily
esters, ethyl alcohol, vegetable oils such as arachis, olive, sesame, or
coconut oil,
fractionated vegetable oils, and mineral oils such as liquid paraffin. Liquid
suspensions may
further comprise one or more additional ingredients including, but not limited
to, suspending
agents, dispersing or wetting agents, emulsifying agents, demulcents,
preservatives, buffers,
salts, flavorings, coloring agents, and sweetening agents. Oily suspensions
may further
comprise a thickening agent. Known suspending agents include, but are not
limited to,
sorbitol syrup, hydrogenated edible fats, sodium alginate,
polyvinylpyrrolidone, gum
tragacanth, gum acacia, and cellulose derivatives such as sodium
'carboxymethylcellulose,
methylcellulose, hydroxypropyl methylcellulose. Known dispersing or wetting
agents include,
but are not limited to, naturally-occurring phosphatides such as lecithin,
condensation
products of an alkylene oxide with a fatty acid, with a long chain aliphatic
alcohol, with a
partial ester derived from a fatty acid and a hexitol, or with a partial ester
derived from a fatty
acid and a hexitol anhydride (e.g., polyoxyethylene stearate,
heptadecaethyleneoxycetanol,
polyoxyethylene sorbitol monooleate, and polyoxyethylene sorbitan monooleate,
respectively). Known emulsifying agents include, but are not limited to,
lecithin and acacia.
Known preservatives include, but are not limited to, methyl, ethyl, or n-
propyl-para-hydroxybenzoates, ascorbic acid, and sorbic acid. Known sweetening
agents
include, for example, glycerol, propylene glycol, sorbitol, sucrose, and
saccharin. Known
thickening agents for oily suspensions include, for example, beeswax, hard
paraffin, and cetyl
alcohol.
Liquid solutions of the active ingredient in aqueous or oily solvents may be
prepared
in substantially the same manner as liquid suspensions, the primary difference
being that the
active ingredient is dissolved, rather than suspended in the solvent. Liquid
solutions of the
pharmaceutical composition of the invention may comprise each of the
components described
with regard to liquid suspensions, it being understood that suspending agents
will not
necessarily aid dissolution of the active ingredient in the solvent. Aqueous
solvents include,
for example, water and isotonic saline. Oily solvents include, for example,
almond oil, oily
esters, ethyl alcohol, vegetable oils such as arachis, olive, sesame, or
coconut oil,
fractionated vegetable oils, and mineral oils such as liquid paraffin.
Powdered and granular formulations of a pharmaceutical preparation of the
invention
may be prepared using known methods. Such formulations may be administered
directly to a
subject, used, for example, to form tablets, to fill capsules, or to prepare
an aqueous or oily
suspension or solution by addition of an aqueous or oily vehicle thereto. Each
of these
formulations may further comprise one or more of dispersing or wetting agent,
a suspending


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 85-

agent, and a preservative. Additional excipients, such as fillers and
sweetening, flavoring, or
coloring agents, may also be included in these formulations.
A pharmaceutical composition of the invention may also be prepared, packaged,
or
sold in the form of oil-in-water emulsion or a water-in-oil emulsion. The oily
phase may be a
vegetable oil such as olive or arachis oil, a mineral oil such as liquid
paraffin, or a combination
of these. Such compositions may further comprise one or more emulsifying
agents such as
naturally occurring gums such as gum acacia or gum tragacanth, naturally-
occurring
phosphatides such as soybean or lecithin phosphatide, esters or partial esters
derived from
combinations of fatty acids and hexitol anhydrides such as sorbitan
monooleate, and
condensation products of such partial esters with ethylene oxide such as
polyoxyethylene
sorbitan monooleate. These emulsions may also contain additional ingredients
including, for
example, sweetening or flavoring agents.
A pharmaceutical composition of the invention may be prepared, packaged, or
sold in
a formulation suitable for rectal administration. Such a composition may be in
the form of, for
example, a suppository, a retention enema preparation, and a solution for
rectal or colonic
irrigation.
Suppository formulations may be made by combining the active ingredient with a
non-irritating pharmaceutically acceptable excipient which is solid at
ordinary room
temperature (i.e., about 20 C) and which is liquid at the rectal temperature
of the subject (i.e.,
about 37 C in a healthy human). Suitable pharmaceutically acceptable
excipients include, but
are not limited to, cocoa butter, polyethylene glycols, and various
glycerides. Suppository
formulations may further comprise various additional ingredients including,
but not limited to,
antioxidants and preservatives.
Retention enema preparations or solutions for rectal or colonic irrigation may
be
made by combining the active ingredient with a pharmaceutically acceptable
liquid carrier. As
is well known in the art, enema preparations may be administered using, and
may be
packaged within, a delivery device adapted to the rectal anatomy of the
subject. Enema
preparations may further comprise various additional ingredients including,
but not limited to,
antioxidants and preservatives.
A pharmaceutical composition of the invention may be prepared, packaged, or
sold in
a formulation suitable for vaginal administration. Such a composition may be
in the form of,
for example, a suppository, an impregnated or coated vaginally-insertable
material such as a
tampon, a douche preparation, or gel or cream or a solution for vaginal
irrigation.
Methods for impregnating or coating a material with a chemical composition are
known in the art, and include, but are not limited to methods of depositing or
binding a
chemical composition onto a surface, methods of incorporating a chemical
composition into


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 86-

the structure of a material during the synthesis of the material (i.e. such as
with a
physiologically degradable material), and methods of absorbing an aqueous or
oily solution or
suspension into an absorbent material, with or without subsequent drying.
Douche preparations or solutions for vaginal irrigation may be made by
combining the
active ingredient with a pharmaceutically acceptable liquid carrier. As is
well known in the art,
douche preparations may be administered using, and may be packaged within, a
delivery
device adapted to the vaginal anatomy of the subject. Douche preparations may
further
comprise various additional ingredients including, but not limited to,
antioxidants, antibiotics,
antifungal agents, and preservatives.
As used herein, "parenteral administration" of a pharmaceutical composition
includes
any route of administration characterized by physical breaching of a tissue of
a subject and
administration of the pharmaceutical composition through the breach in the
tissue. Parenteral
administration thus includes, but is not limited to, administration of a
pharmaceutical
composition by injection of the composition, by application of the composition
through a
surgical incision, by application of the composition through a tissue-
penetrating non-surgical
wound, and the like. In particular, parenteral administration is contemplated
to include, but is
not limited to, subcutaneous, intraperitoneal, intramuscular, intrasternal
injection, and kidney
dialytic infusion techniques.
Formulations of a pharmaceutical composition suitable for parenteral
administration
comprise the active ingredient combined with a pharmaceutically acceptable
carrier, such as
sterile water or sterile isotonic saline. Such formulations may be prepared,
packaged, or sold
in a form suitable for bolus administration or for continuous administration.
Injectable
formulations may be prepared, packaged, or sold in unit dosage form, such as
in ampules or
in multi-dose containers containing a preservative. Formulations for
parenteral administration
include, but are not limited to, suspensions, solutions, emulsions in oily or
aqueous vehicles,
pastes, and implantable sustained-release or biodegradable formulations as
discussed below.
Such formulations may further comprise one or more additional ingredients
including, but not
limited to, suspending, stabilizing, or dispersing agents. In one embodiment
of a formulation
for parenteral administration, the active ingredient is provided in dry (i.e.
powder or granular)
form for reconstitution with a suitable vehicle (e.g., sterile pyrogen-free
water) prior to
parenteral administration of the reconstituted composition.
A composition of the present invention can be administered by a variety of
methods
known in the art. The route and/or mode of administration vary depending upon
the desired
results. The active compounds can be prepared with carriers that protect the
compound
against rapid release, such as a controlled release formulation, including
implants,
transdermal patches, and microencapsulated delivery systems. Biodegradable,
biocompatible


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 87-

polymers can be used, such as ethylene vinyl acetate, polyanhydrides,
polyglycolic acid,
collagen, polyorthoesters, and polylactic acid. Many methods for the
preparation of such
formulations are described by e.g., Sustained and Controlled Release Drug
Delivery Systems,
J. R. Robinson, ed., Marcel Dekker, Inc., New York, (1978). Pharmaceutical
compositions are
preferably manufactured under GMP conditions.
The pharmaceutical compositions may be prepared, packaged, or sold in the form
of
a sterile injectable aqueous or oily suspension or solution. This suspension
or solution may be
formulated according to the known art, and may comprise, in addition to the
active ingredient,
additional ingredients such as the dispersing agents, wetting agents, or
suspending agents
described herein. Such sterile injectable formulations may be prepared using a
non-toxic
parenterally-acceptable diluent or solvent, such as water or 1,3-butane diol,
for example.
Other acceptable diluents and solvents include, but are not limited to,
Ringer's solution,
isotonic sodium chloride solution, and fixed oils such as synthetic mono- or
di-glycerides.
Other parentally-administrable formulations which are useful include those
which comprise
the active ingredient in microcrystalline form, in a liposomal preparation, or
as a component of
a biodegradable polymer systems. Compositions for sustained release or
implantation may
comprise pharmaceutically acceptable polymeric or hydrophobic materials such
as an
emulsion, an ion exchange resin, a sparingly soluble polymer, or a sparingly
soluble salt.
Formulations suitable for topical administration include, but are not limited
to, liquid or
semi-liquid preparations such as liniments, lotions, oil-in-water or water-in-
oil emulsions such
as creams, ointments or pastes, and solutions or suspensions. Topically-
administrable
formulations may, for example, comprise from about 1% to about 10% (w/w)
active ingredient,
although the concentration of the active ingredient may be as high as the
solubility limit of the
active ingredient in the solvent. Formulations for topical administration may
further comprise
one or more of the additional ingredients described herein.
A pharmaceutical composition of the invention may be prepared, packaged, or
sold in
a formulation suitable for pulmonary administration via the buccal cavity.
Such a formulation
may comprise dry particles which comprise the active ingredient and which have
a diameter
in the range from about 0.5 to about 7 nanometers, and preferably from about 1
to about 6
nanometers. Such compositions are conveniently in the form of dry powders for
administration using a device comprising a dry powder reservoir to which a
stream of
propellant may be directed to disperse the powder or using a self-propelling
solvent/powder-dispensing container such as a device comprising the active
ingredient
dissolved or suspended in a low-boiling propellant in a sealed container.
Preferably, such
powders comprise particles wherein at least 98% of the particles by weight
have a diameter
greater than 0.5 nanometers and at least 95% of the particles by number have a
diameter


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 88-

less than 7 nanometers. More preferably, at least 95% of the particles by
weight have a
diameter greater than 1 nanometer and at least 90% of the particles by number
have a
diameter less than 6 nanometers. Dry powder compositions preferably include a
solid fine
powder diluent such as sugar and are conveniently provided in a unit dose
form.
Low boiling propellants generally include liquid propellants having a boiling
point of
below 65 F at atmospheric pressure. Generally the propellant may constitute 50
to 99.9%
(w/w) of the composition, and the active ingredient may constitute 0.1 to 20%
(w/w) of the
composition. The propellant may further comprise additional ingredients such
as a liquid non-
ionic or solid anionic surfactant or a solid diluent (preferably having a
particle size of the same
order as particles comprising the active ingredient).
Pharmaceutical compositions of the invention formulated for pulmonary delivery
may
also provide the active ingredient in the form of droplets of a solution or
suspension. Such
formulations may be prepared, packaged, or sold as aqueous or dilute alcoholic
solutions or
suspensions, optionally sterile, comprising the active ingredient, and may
conveniently be
administered using any nebulization or atomization device. Such formulations
may further
comprise one or more additional ingredients including, but not limited to, a
flavoring agent
such as saccharin sodium, a volatile oil, a buffering agent, a surface active
agent, or a
preservative such as methylhydroxybenzoate. The droplets provided by this
route of
administration preferably have an average diameter in the range from about 0.1
to about 200
nanometers.
The formulations described herein as being useful for pulmonary delivery are
also
useful for intranasal delivery of a pharmaceutical composition of the
invention.
Another formulation suitable for intranasal administration is a coarse powder
comprising the active ingredient and having an average particle from about 0.2
to 500
micrometers. Such a formulation is administered in the manner in which snuff
is taken, i.e.,
by rapid inhalation through the nasal passage from a container of the powder
held close to
the nares.
Formulations suitable for nasal administration may, for example, comprise from
about
as little as 0.1% (w/w) and as much as 100% (w/w) of the active ingredient,
and may further
comprise one or more of the additional ingredients described herein.
A pharmaceutical composition of the invention may be prepared, packaged, or
sold in
a formulation suitable for buccal administration. Such formulations may, for
example, be in
the form of tablets or lozenges made using conventional methods, and may, for
example, 0.1
to 20% (w/w) active ingredient, the balance comprising an orally dissolvable
or degradable
composition and, optionally, one or more of the additional ingredients
described herein.
Alternately, formulations suitable for buccal administration may comprise a
powder or an


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 89-

aerosolized or atomized solution or suspension comprising the active
ingredient. Such
powdered, aerosolized, or aerosolized formulations, when dispersed, preferably
have an
average particle or droplet size in the range from about 0.1 to about 200
nanometers, and
may further comprise one or more of the additional ingredients described
herein.
A pharmaceutical composition of the invention may be prepared, packaged, or
sold in
a formulation suitable for ophthalmic administration. Such formulations may,
for example, be
in the form of eye drops including, for example, a 0.1-1.0% (w/w) solution or
suspension of
the active ingredient in an aqueous or oily liquid carrier. Such drops may
further comprise
buffering agents, salts, or one or more other of the additional ingredients
described herein.
Other ophthalmalmically-administrable formulations which are useful include
those which
comprise the active ingredient in microcrystalline form or in a liposomal
preparation.
As used herein, "additional ingredients" include, but are not limited to, one
or more of
the following: excipients; surface active agents; dispersing agents; inert
diluents; granulating
and disintegrating agents; binding agents; lubricating agents; sweetening
agents; flavoring
agents; coloring agents; preservatives; physiologically degradable
compositions such as
gelatin; aqueous vehicles and solvents; oily vehicles and solvents; suspending
agents;
dispersing or wetting agents; emulsifying agents, demulcents; buffers; salts;
thickening
agents; fillers; emulsifying agents; antioxidants; antibiotics; antifungal
agents; stabilizing
agents; and pharmaceutically acceptable polymeric or hydrophobic materials.
Other
"additional ingredients" which may be included in the pharmaceutical
compositions of the
invention are known in the art and described, for example in Remington's
Pharmaceutical
Sciences, Genaro, ed., Mack Publishing Co., Easton, PA (1985), which is
incorporated
herein by reference.
In one embodiment of the invention, the composition comprising CP-675,206
comprises a sterile solution comprising 20 mM histidine buffer, pH 5.5, 84
mg/mI trehalose
dehydrate, 0.2 mg/mI polysorbate 80, and 0.1 mg/mI disodium EDTA hydrate. In
one aspect,
CP-675,206 is packaged in clear glass vials with a rubber stopper and an
aluminum seal. In
another aspect, the vial contains about 20 mg/mI of CP-675,206 with a nominal
fill of about
400 mg per vial.
The anti-CTLA4/therapeutic agent active ingredient combination of the
invention, can
be administered to an animal, preferably a human. The precise dosage of each
active
ingredient administered will vary depending upon any number of factors,
including but not
limited to, the type of animal and type of disease state being treated, the
age of the animal
and the route of administration.
The anti-CTLA4 antibody may be administered to an animal as frequently as
several
times daily, or it may be administered less frequently, such as once a day,
once a week, once


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 90-

every two weeks, once a month, or even less frequently, such as once every
several months
or even once a year or less. The frequency of the dose will be readily
apparent to the skilled
artisan and will depend upon any number of factors, such as, but not limited
to, the type and
severity of the disease being treated, the type and age of the animal, etc.
The therapeutic agent, preferably, a chemotherapeutic agent, may be
administered to
an animal as frequently as several times daily, or it may be administered less
frequently, such
as once a day, once a week, once every two weeks, once a month, or even less
frequently,
such as once every several months or even once a year or less. The frequency
of the dose
will be readily apparent to the skilled artisan and will depend upon any
number of factors,
such as, but not limited to, the type and severity of the disease being
treated, the type and
age of the animal, etc.
The antibody and therapeutic agent can be co-administered in that they can be
administered separately, on different dates or at different times of the day,
as well as
simultaneously or on the same date. Co-administration thus encompasses any
temporal
combination of administration of the antibody and the therapeutic agent such
that
administration of the two agents mediates a therapeutic benefit to the patient
that is
detectably greater than administration of either agent in the absence of the
other.
An antibody-therapeutic agent combination of the invention may be co-
administered
with numerous other compounds (other antihormonal therapy agents, cytokines,
chemotherapeutic and/or antiviral drugs, among many others). Alternatively,
the
compound(s) may be administered an hour, a day, a week, a month, or even more,
in
advance of the antibody-therapeutic agent combination, or any permutation
thereof. Further,
the compound(s) may be administered an hour, a day, a week, or even more
preferably, after
administration of radiation, stem cell transplant, or administration of any
therapeutic agent
(e.g., cytokine, chemotherapeutic compound, and the like), or any permutation
thereof. The
frequency and administration regimen will be readily apparent to the skilled
artisan and will
depend upon any number of factors such as, but not limited to, the type and
severity of the
disease being treated, the age and health status of the animal, the identity
of the compound
or compounds being administered, the route of administration of the various
compounds, and
the like.
VI. Kits
The invention includes various kits which comprise a therapeutically effective
amount
of a human anti-CTLA4 antibody of the invention and a therapeutically
effective amount of a
therapeutic agent, along with an applicator and instructional materials which
describe use of
the combination to perform the methods of the invention. Although exemplary
kits are


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-91-
described below, the contents of other useful kits will be apparent to the
skilled artisan in light
of the present disclosure. Each of these kits is included within the
invention.
The invention includes a kit for treatment of breast cancer in a patient in
need thereof.
The kit includes a human anti-CTLA4 antibody of the invention and at least one
therapeutic
agent. The inhibitor encompasses, but is not limited to, a chemotherapeutic,
an antibody, a
cytokine, a vaccine, an immunomodulator, among many others. The kit further
comprises an
applicator, including, but not limited to, a syringe, for administration of
the components of the
kit to a patient. Further, the kit comprises an instructional material setting
forth the pertinent
information for the use of the kit to treat breast cancer in the patient.
More preferably, the kit comprises at least one anti-CTLA4 antibody selected
from an
antibody having the heavy and light chain amino acid sequence of antibody
4.1.1, 4.8.1,
4.10.2, 4.13.1, 4.14.3, 6.1.1, 11.2.1 (CP-675,206), 11.6.1, 11.7.1., 12.3.1.1,
12.9.1.1, and
ipilimumab, even more preferably, the antibody is an antibody having the heavy
and light
chain amino acid sequence of antibody 4.13.1, 11.2.1 (CP-675,206), and
ipilimumab (MDX-
010). Preferably, the antibody is an antibody having the heavy and light
sequence of 11.2.1
(CP-675,206). Even more preferably, the anti-CTLA4 antibody is CP-675,206 or
ipilimumab.
Most preferably, the antibody is CP-675,206.
The kit can comprise any number of additional therapeutic agents for treatment
of
cancer. Such agents are set forth previously and include chemotherapeutic
compounds,
cancer vaccines, signal transduction inhibitors, agents useful in treating
abnormal cell growth
or cancer, antibodies or other ligands that inhibit tumor growth by binding to
IGF-1 R, and
cytokines, among many others.
The invention also relates to an article of manufacture (e.g., dosage form
adapted for
i.v. administration) comprising a human anti-CTLA4 antibody in the amount
effective to treat
cancer (e.g., more than 10 mg/kg, at least 15 mg/kg, or 15 mg/kg) and a
therapeutically
effective amount of a second therapeutic agent. In certain embodiments, the
article of
manufacture comprises a container or containers comprising a human anti-CTLA4
antibody, the
therapeutic agent, and a label and/or instructions for use to treat cancer.
In one embodiment, the therapeutic agent is at least one agent selected from a
hormonal therapy agent, a taxane, a heat shock protein, a topoisomerase
inhibitor, a
hormonal therapy agent (e.g., leuprolide), a chemotherapeutic compounds, a
vinca alkaloid, a
platinum compound, a cancer vaccine, a tumor-specific antigen, an angiogenesis
inhibitor, a
signal transduction inhibitor, agents useful in treating abnormal cell growth
or cancer,
antibodies or other ligands that inhibit tumor growth by binding to IGF-1 R,
and cytokines,
among others.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 92-

The kit can comprise any number of additional therapeutic agents for treatment
of
cancer. Such agents are set forth previously and include chemotherapeutic
compounds,
cancer vaccines, signal transduction inhibitors, agents useful in treating
abnormal cell growth
or cancer, antibodies or other ligands that inhibit tumor growth by binding to
IGF-1 R, and
cytokines, and palliative care agents (e.g., anti-diarrheals, anti-emetics,
etc.) among many
others.
In another embodiment of the invention, the kit is for treatment of breast
cancer. The
kit comprises an anti-CTLA4 antibody and a taxane. In one aspect, the taxane
is docetaxel or
paclitaxel. In another aspect, the treatment is first line therapy for the
treatment of patients
with locally advanced or metastatic triple receptor negative breast cancer.
In one embodiment, the kit is for treatment of ovarian cancer. In one aspect,
the kit
comprises an anti-CTLA4 antibody and a therapeutic agent, where the
therapeutic agent is
paclitaxel. In another aspect, the kit further comprises carboplatin. In yet
another aspect, the
kit is for first-line treatment of advanced carcinoma of the ovary. In another
aspect, the kit is
for second line treatment of a patient who has progressed following prior
paclitaxel-based
therapy.
In an embodiment of the invention, the kit is for treatment of non-small cell
lung
cancer. In one embodiment, the kit comprises an anti-CTLA4 antibody and a
chemotherapeutic agent, wherein the therapeutic agent is selected from the
group consisting
of a platin (e.g., carboplatin (PARAPLATIN), paclitaxel (TAXOL)), docetaxel
(TAXOTERE),
sunitinib (SU11248), eriotinib (TARCEVA), bevacizumab (AVASTIN), pemetrexed
(ALIMTA),
and PF03512676 (CpG-7909).
In one embodiment, kit is for first line therapy of locally advanced Stage
IIIb or
metastatic Stage IV NSCLC where the kit comprises an anti-CTLA4 antibody and a
platin. In
another embodiment, the kit comprises carboplatin and paclitaxel. In another
embodiment,
the kit comprises the antibody and bevacizumab. In a further embodiment, the
kit comprises
the antibody and PF03512676. In another embodiment of the invention, the kit
comprises the
antibody and sunitinib.
In one embodiment, the kit is for second line treatment of Stage IIIb or
metastatic
Stage IV NSCLC after failure of platinum-based therapy. In one embodiment, the
kit
comprises an anti-CTLA4 antibody and docetaxel. In another embodiment, the kit
comprises
the antibody and erlotinib. In yet another embodiment, the kit comprises the
antibody and
pemetrexed. In yet another embodiment, the kit comprises an anti-CTLA4
antibody.
In another embodiment of the invention, the kit is for third line treatment of
locally
advanced Stage Illb or metastatic Stage IV NSCLC. The kit comprises an
effective amount of


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 93-

an anti-CTLA4 antibody. In one aspect, the kit is for treatment of cancer
after failure of prior
platinum-based chemotherapy and EGFR inhibition-based therapy.
In one embodiment, the kit is for treatment of pancreatic cancer. The kit
comprises
an anti-CTLA4 antibody and a therapeutic agent, wherein the agent is
gemcitabine. In one
aspect, the treatment is first line therapy for locally advanced, non-
resectable Stage II or
Stage III, or metastatic Stage IV adenocarcinoma of the pancreas.
In one embodiment, the kit is for treatment of melanoma and comprises an
effective
amount of an anti-CTLA4 antibody and an effective amount of IFNa. In one
aspect, the
treatment is adjuvant therapy for Stage II/III melanoma.
In an embodiment of the present invention, the kit is for treatment of
colorectal
carcinoma. In one aspect, the kit comprises an effective amount of an anti-
CTLA4 antibody
and an effective amount of each of fluorouracil, leucovorin and oxaliplatin
(FOLFOX). In
another aspect, the kit comprises an effective amount of an anti-CTLA4
antibody, fluorouracil,
Ieucovorin and irinotecan (FOLFIRI). In one aspect, the treatment is first
line therapy for
metastatic CRC. In another aspect, the therapy is adjuvant therapy for Stage
III colon cancer
in a patient who has undergone resection of a primary tumor.
In another embodiment, the kit is for treatment of CRC and comprises an
effective
amount of an anti-CTLA4 antibody and an effective amount of capecitabine
(XELODA). In
one aspect, the treatment is first line therapy for a patient intolerant of
therapy comprising
oxaliplatin (ELOXATIN) or irinotecan (CAMPTOSAR).

The invention is further described in detail by reference to the following
experimental
examples. These examples are provided for purposes of illustration only, and
are not
intended to be limiting unless otherwise specified. Thus, the invention should
in no way be
construed as being limited to the following examples, but rather, should be
construed to
encompass any and all variations which become evident as a result of the
teaching provided
herein.
EXAMPLES
EXAMPLE 1:
CP-675,206 in Combination with rituximab for First-line Treatment of Indolent
Non-Hodcikin's Lymphoma
Patients having indolent non-Hodgkin's lymphoma are treated with CP-675,206
and
rituximab. Indolent Non-Hodgkin's Lymphomas (NHLs) are slow growing forms of
lymphoma
and encompass what were previously referred to as low grade and some
categories of
intermediate grade NHL in the Working Formulation. Non-Hodgkin's lymphoma
(NHL) with at


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 94-

least one lesion that can be accurately measured in two dimensions and whose
size is > 2 cm
x 1 cm by conventional CT scan or >,1 cm x 1 cm by spiral CT scan are given
standard
rituximab (RITUXAN) antibody therapy per established protocols.
The patient is further administered a single IV infusion (100 mUhr) of CP-
675,206 at
a dose of about 3 mg/kg. Prophylactic anti-emetics and anti-diarrheals are
given as
appropriate. The treatment is repeated after 28 days with escalation of CP-
675,206 dose,
that is, 6 mg/kg, 10 mg/kg and 15 mg/kg, every 28 days thereafter for maximum
of 12 cycles
in the absence of intolerable toxicity or disease progression. Preferably, CP-
675,206 is
administered at at least about 10 mg/kg every three weeks for four cycles and
then every
three months thereafter.
Rituximab is administered via intravenous infusion at about 375 mg/m2 once
weekly
for about four or eight doses. More preferably, rituximab is administered on
day 1 of each
cycle of chemotherapy.
Preferably, the patient is premedicated with antihistamine (HI) at least one
half hour
prior to infusion of either antibody. Premedication is recommended but not
required.
Doses are escalated using an accelerated titration design utilizing a dose-
doubling
schema with 3-6 subjects per cohort. Within each new cohort there is no
required waiting
period between subjects. Subsequent cohorts may not be opened until the first
subject at the
current dose level has been observed for 21 days and subsequent subjects have
been
observed for 14 days.
Rituximab and anti-CTLA4 antibody are administered either sequentially or
simultaneously either once, or repeatedly, as determined.
Following combination treatment with rituximab and anti-CTLA4 antibody, anti-
CTLA4
antibody is administered as single agent (SA) therapy as described supra for
combination
therapy.
CP-675,206 is provided in 20 ml clear glass vials with a rubber stopper and an
aluminum seal. Each vial contains 20 mg/mi (with a nominal fill of 400
mg/vial) of CP-
675,206, in a sterile aqueous solution comprising 20 mM histidine buffer, PH
5.5, 84 mg/mI
trehalose dehydrate, 0.2 mg/mI polysorbate 80, and 0.1 mg/mI disodium EDTA
dehydrate.
For all patients, Eastern Cooperative Oncology Group performance status (ECOG
PS), vital signs, and body weight are assessed pre-dose, and vital signs can
be repeated
post-dose, as clinically indicated. A physical examination (including
ophthalmologic
assessment and signs of autoimmunity) is performed on Day 1. Samples for
hematology
panel (hematocrit, RBC count, WBC count, differential), chemistry (Alkaline
Phosphatase,
calcium, chloride, GGT, LDH, magnesium, phosphorus, random glucose, sodium,
urea, uric
acid), urinalysis (blood, protein), others (activated partial thromboplastin
time [APTT],


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 95-

prothrombin time (PT), autoantibody panel, C reactive protein, TSH, T3, T4,
amylase, lipase,
serum C3, C4, serum Ig level), are obtained.
Baseline human anti-human antibody (HAHA) titer is determined and
pharmacokinetic (PK) specimen is obtained pre-dose.
The following endpoints are measured: PK parameters, HAHA, response rate and
time to progression. Time to progression and overall survival are calculated
using the
Kaplan-Meier product limit method.
EXAMPLE 2:
CP-675,206 in Combination with rituximab for Second-line Treatment of
Aggressive Non-Hodgkin's Lymphoma
Patients having aggressive, CHOP-R refractory NHL with at least one lesion
that can
be accurately measured in two dimensions and whose size is _ 2 cm x 1 cm by
conventional
CT scan or ? 1 cm x 1 cm by spiral CT scan are given standard rituximab
antibody therapy
per established protocols.
The patient is further administered a single IV infusion (100 mL/hr) of CP-
675,206 as
described herein at a dose of about 3 mg/kg. Prophylactic anti-emetics and
anti-diarrheals
are given as appropriate. The treatment is repeated after 28 days with
escalation of the anti-
CTLA4 antibody dose, that is, 6 mg/kg, 10 mg/kg and 15 mg/kg, every 28 days
thereafter for
maximum of 12 cycles in the absence of intolerable toxicity or disease
progression.
Preferably, CP-675,206 is administered at at least about 10 mg/kg every three
weeks for four
cycles and then every three months thereafter. The patient is administered
rituximab
according to an art-recognized dosing regimen.
Preferably, the patient is premedicated with antihistamine (H1) at least one
half hour
prior to infusion of anti-CTLA4. Premedication is recommended but not
required.
Doses are escalated using an accelerated titration design utilizing a dose-
doubling
schema with 3-6 subjects per cohort. Within each new cohort there is no
required waiting
period between subjects. Subsequent cohorts may not be opened until the first
subject at the
current dose level has been observed for 21 days and subsequent subjects have
been
observed for 14 days.
Rituximab (e.g., RITUXAN) and the anti-CTLA4 antibody are administered either
sequentially or simultaneously either once, or repeatedly, as determined.
Following combination treatment with RITUXAN and the anti-CTLA4 antibody, the
CTLA4 antibody is administered as described supra.
CP-675,206 is provided in 20 ml clear glass vials with a rubber stopper and an
aluminum seal. Each vial contains 20 mg/mI (with a nominal fill of 400
mg/vial) of CP-


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 96-

675,206, in a sterile aqueous solution comprising 20 mM histidine buffer, PH
5.5, 84 mg/mI
trehalose dehydrate, 0.2 mg/mI polysorbate 80, and 0.1 mg/mI disodium EDTA
dehydrate.
For all patients, ECOG performance status, vital signs, and body weight are
assessed
pre-dose, and vital signs can be repeated post-dose, as clinically indicated.
A physical
examination (including ophthalmologic assessment and signs of autoimmunity) is
performed
on Day 1. Samples for hematology panel (hematocrit, RBC count, WBC count,
differential),
chemistry (Alkaline Phosphatase, calcium, chloride, GGT, LDH, magnesium,
phosphorus,
random glucose, sodium, urea, uric acid), urinalysis (blood, protein), others
(activated partial
thromboplastin time [APTT], prothrombin time (PT), autoantibody panel, C
reactive protein,
TSH, T3, T4, amylase, lipase, serum C3, C4, serum Ig level), are obtained.
Baseline human anti-human antibody (HAHA) titer is determined and
pharmacokinetic (PK) specimen is obtained pre-dose.
The following endpoints are measured: PK parameters, HAHA, response rate and
time to progression. Time to progression and overall survival are calculated
using the
Kaplan-Meier product limit method.
EXAMPLE 3:
CP-675,206 in Combination with 5-fluorouracil or capecitabine for Adiuvant or
First-line Treatment of Colorectal Carcinoma
Patients having colorectal carcinoma (CRC) and intolerant of oxaliplatin
(ELOXATIN)
or irinotecan (CAMPTOSAR) with at least one lesion that can be accurately
measured in two
dimensions and whose size is _ 2 cm x 1 cm by conventional CT scan or _ 1 cm x
1 cm by
spiral CT scan are given standard chemotherapy using 5FU (fluorouracil) or
XELODA
(capecitabine) per established protocols.
The patient is further administered a single IV infusion (100 mUhr) of CP-
675,206 as
described herein at a dose of about 3 mg/kg. Prophylactic anti-emetics and
anti-diarrheals
are given as appropriate. The treatment is repeated after 28 days with
escalation of the anti-
CTLA4 antibody dose, that is, 6 mg/kg, 10 mg/kg and 15 mg/kg, every 28 days
thereafter for
maximum of 12 cycles in the absence of intolerable toxicity or disease
progression.
Preferably, CP-675,206 is administered at at least about 10 mg/kg every three
weeks for four
cycles and then every three months thereafter.
Preferably, the patient is premedicated with antihistamine (H1) at least one
half hour
prior to infusion of anti-CTLA4. Premedication is recommended but not
required.
Doses are escalated using an accelerated titration design utilizing a dose-
doubling
schema with 3-6 subjects per cohort. Within each new cohort there is no
required waiting
period between subjects. Subsequent cohorts may not be opened until the first
subject at the


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 97-

current dose level has been observed for 21 days and subsequent subjects have
been
observed for 14 days.
Capecitabine for second line therapy or 5-fluorouracil for adjuvant therapy is
administered sequentially or simultaneously with CP-675,206 either once, or
repeatedly, as
determined. Capecitabine and fluorouracil are administered according to well-
known
protocols, for examples, those disclosed in the FDA-approved label for each
compound.
CP-675,206 is provided in 20 ml clear glass vials with a rubber stopper and an
aluminum seal. Each vial contains 20 mg/mI (with a nominal fill of 400
mg/vial) of CP-
675,206, in a sterile aqueous solution comprising 20 mM histidine buffer, PH
5.5, 84 mg/mI
trehalose dehydrate, 0.2 mg/mI polysorbate 80, and 0.1 mg/mI disodium EDTA
dehydrate.
For all patients, ECOG performance status, vital signs, and body weight are
assessed
pre-dose, and vital signs can be repeated post-dose, as clinically indicated.
A physical
examination (including ophthalmologic assessment and signs of autoimmunity) is
performed
on Day 1. Samples for hematology panel (hematocrit, RBC count, WBC count,
differential),
chemistry (Alkaline Phosphatase, calcium, chloride, GGT, LDH, magnesium,
phosphorus,
random glucose, sodium, urea, uric acid), urinalysis (blood, protein), others
(activated partial
thromboplastin time [APTT], prothrombin time (PT), autoantibody panel, C
reactive protein,
TSH, T3, T4, amylase, lipase, serum C3, C4, serum lg level), are obtained.
Baseline human anti-human antibody (HAHA) titer is determined and
pharmacokinetic (PK) specimen is obtained pre-dose.
The following endpoints are measured: PK parameters, HAHA, response rate and
time to progression. Time to progression and overall survival are calculated
using the
Kaplan-Meier product limit method.
EXAMPLE 4:
CP-675,206 in Combination with FOLFOX for Adiuvant or First Line Treatment of
Colorectal Carcinoma
Following surgery/radiotherapy patients having colorectal carcinoma (CRC) with
at
least one lesion that can be accurately measured in two dimensions and whose
size is ? 2 cm
x 1 cm by conventional CT scan or _ 1 cm x 1 cm by spiral CT scan are given
standard
chemotherapy using FOLFOX (fluorouracil, leucovorin and oxaliplatin) per
established
protocols.
The patient is further administered a single IV infusion (100 mL/hr) of anti-
CTLA4
antibodies as described herein at a dose of about 3 mg/kg. Prophylactic anti-
emetics and
anti-diarrheals are given as appropriate. The treatment is repeated after 28
days with
escalation of the anti-CTLA4 antibody dose, that is, 6 mg/kg, 10 mg/kg and 15
mg/kg, every
28 days thereafter for maximum of 12 cycles in the absence of intolerable
toxicity or disease


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 98-

progression. Preferably, CP-675,206 is administered at at least about 10 mg/kg
every three
weeks for four cycles and then every three months thereafter.
FOLFOX or FOLFIRI therapy is administered according to standard protocols.
Briefly, for FOLFOX4, each chemotherapy cycle comprises a two hour infusion of
85 mg/m2
oxaliplatin (ELOXATIN, Sanofi) on day 1 followed by a two hour infusion of 200
mg/m2
leucovorin on days 1 and 2, followed by a bolus of 400 mg/m2 of fluorouracil
on days 1 and 2
and then a twenty-two hour infusion of 600 mg/ma of fluorouracil administered
over two
consecutive days (referred to as the De Gramont schedule). Chemotherapy is
repeated
every two weeks.
In an exemplary regimen, FOLFIRI is administered as a ninety minute infusion
of 180
mg/m2 irinotecan on day 1, a two hour infusion of folinic acid (leucovorin) at
400 mg/m2 on
day 1, and 400-500 mg/m2 fluorouracil IV bolus after leucovorin on day 1,
followed by
fluorouracil at 2400-3000 mg/m2 by continuous i.v. over forty-six hours
starting on day one.
The chemotherapy cycle is repeated every two weeks.
Preferably, the patient is premedicated with antihistamine (HI) at least one
half hour
prior to infusion of anti-CTLA4. Premedication is recommended but not
required.
Doses are escalated using an accelerated titration design utilizing a dose-
doubling
schema with 3-6 subjects per cohort. Within each new cohort there is no
required waiting
period between subjects. Subsequent cohorts may not be opened until the first
subject at the
current dose level has been observed for 21 days and subsequent subjects have
been
observed for 14 days.
FOLFOX therapy is administered sequentially or simultaneously with anti-CTLA4
antibody either once, or repeatedly, as determined.
CP-675,206 is provided in 20 ml clear glass vials with a rubber stopper and an
aluminum seal. Each vial contains 20 mg/mI (with a nominal fill of 400
mg/vial) of CP-
675,206, in a sterile aqueous solution comprising 20 mM histidine buffer, PH
5.5, 84 mg/ml
trehalose dehydrate, 0.2 mg/mi polysorbate 80, and 0.1 mg/ml disodium EDTA
dehydrate.
For all patients, ECOG performance status, vital signs, and body weight are
assessed
pre-dose, and vital signs can be repeated post-dose, as clinically indicated.
A physical
examination (including ophthalmologic assessment and signs of autoimmunity) is
performed
on Day 1. Samples for hematology panel (hematocrit, RBC count, WBC count,
differential),
chemistry (Alkaline Phosphatase, calcium, chloride, GGT, LDH, magnesium,
phosphorus,
random glucose, sodium, urea, uric acid), urinalysis (blood, protein), others
(activated partial
thromboplastin time [APTT], prothrombin time (PT), autoantibody panel, C
reactive protein,
TSH, T3, T4, amylase, lipase, serum C3, C4, serum Ig level), are obtained.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 99-

Baseline human anti-human antibody (HAHA) titer is determined and
pharmacokinetic (PK) specimen is obtained pre-dose.
The following endpoints are measured: PK parameters, HAHA, response rate and
time to progression. Time to progression and overall survival are calculated
using the
Kaplan-Meier product limit method.
EXAMPLE 5:
CP-675,206 and Gemcitabine for Treatment of Pancreatic Cancer
Following surgery/radiotherapy, if any, patients having pancreatic cancer with
at least
one lesion that can be accurately measured in two dimensions and whose size is
? 2 cm x 1
cm by conventional CT scan or _ 1 cm x 1 cm by spiral CT scan are given
standard
chemotherapy comprising gemcitabine (e.g., GEMZAR) per established protocols.
The patient is further administered a single IV infusion of anti-CTLA4
antibodies as
described herein at a dose of at least about 10 mg/kg, preferably, at about 15
mg/kg.
Prophylactic anti-emetics and anti-diarrheals are given as appropriate. The
treatment is
repeated after 28 days with escalation of the anti-CTLA4 antibody dose, if
desired, every 28
days thereafter for maximum of 12 cycles in the absence of intolerable
toxicity or disease
progression. Preferably, CP-675,206 is administered at at least about 10 mg/kg
every three
weeks for four cycles and then every three months thereafter.
Gemcitabine is administered according to standard protocols. More
particularly,
gemcitabine (GEMZAR, Lilly) is administered by intravenous infusion at a dose
of 1000
mg/mz over 30 minutes once weekly for up to 7 weeks (or until toxicity
necessitates reducing
or holding a dose) followed by a week of rest from treatment. Subsequent
cycles should
consist of infusions once weekly for 3 consecutive weeks out of every 4 weeks
. CP-675,206
is administered after the initial gemcitabine cycle (seven weeks on and one
week rest) and
once every three weeks thereafter.
Preferably, the patient is premedicated with antihistamine (H1) at least one
half hour
prior to infusion of anti-CTLA4. Premedication is recommended but not
required.
Where appropriate, antibody dose is escalated using an accelerated titration
design
utilizing a dose-doubling schema with 3-6 subjects per cohort. Within each new
cohort there
is no required waiting period between subjects. Subsequent cohorts may not be
opened until
the first subject at the current dose level has been observed for 21 days and
subsequent
subjects have been observed for 14 days.
CP-675,206 is provided in 20 ml clear glass vials with a rubber stopper and an
aluminum seal. Each vial contains 20 mg/mI (with a nominal fill of 400
mg/vial) of CP-
675,206, in a sterile aqueous solution comprising 20 mM histidine buffer, PH
5.5, 84 mg/mi
trehalose dehydrate, 0.2 mg/mi polysorbate 80, and 0.1 mg/ml disodium EDTA
dehydrate.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-100-
For all patients, ECOG performance status, vital signs, and body weight are
assessed
pre-dose, and vital signs can be repeated post-dose, as clinically indicated.
A physical
examination (including ophthalmologic assessment and signs of autoimmunity) is
performed
on Day 1. Samples for hematology panel (hematocrit, RBC count, WBC count,
differential),
chemistry (Alkaline Phosphatase, calcium, chloride, GGT, LDH, magnesium,
phosphorus,
random glucose, sodium, urea, uric acid), urinalysis (blood, protein), others
(activated partial
thromboplastin time [APTT], prothrombin time (PT), autoantibody panel, C
reactive protein,
TSH, T3, T4, amylase, lipase, serum C3, C4, serum Ig level), are obtained.
Baseline human anti-human antibody (HAHA) titer is determined and
pharmacokinetic (PK) specimen is obtained pre-dose.
The following endpoints are measured: PK parameters, HAHA, response rate and
time to progression. Time to progression and overall survival are calculated
using the
Kaplan-Meier product limit method.
EXAMPLE 6:
CP-675,206 in Combination with imatinib mesylate for First-line Treatment of
Chronic Myeloid Leukemia
Patients having chronic myeloid leukemia (CML) are given standard chemotherapy
using imatinib mesylate (GLEEVEC) per established protocols.
The patient is further administered a single IV infusion (100 mL/hr) of anti-
CTLA4
antibodies as described herein at a dose of about 3 mg/kg. Prophylactic anti-
emetics and
anti-diarrheals are given as appropriate. The treatment is repeated after 28
days with
escalation of the anti-CTLA4 antibody dose, that is, 6 mg/kg, 10 mg/kg and 15
mg/kg, every
28 days thereafter for maximum of 12 cycles in the absence of intolerable
toxicity or disease
progression. Preferably, CP-675,206 is administered at at least about 10 mg/kg
every three
weeks for four cycles and then every three months thereafter.
Imatinib mesylate (GLEEVEC, Novartis) is administered daily at about 400
mg/day for
patients in chronic phase CML and 600 mg/day for adult patients in accelerated
phase or
blast crisis. Imatinib treatment may be continued as long as there is no
evidence of
progressive disease or unacceptable toxicity.
Preferably, the patient is premedicated with antihistamine (H1) at least one
half hour
prior to infusion of anti-CTLA4. Premedication is recommended but not
required.
Doses are escalated using an accelerated titration design utilizing a dose-
doubling
schema with 3-6 subjects per cohort. Within each new cohort there is no
required waiting
period between subjects. Subsequent cohorts may not be opened until the first
subject at the
current dose level has been observed for 21 days and subsequent subjects have
been
observed for 14 days.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 101-

Imatinib mesylate is administered sequentially or simultaneously with anti-
CTLA4
antibody either once, or repeatedly, as determined.
CP-675,206 is provided in 20 ml clear glass vials with a rubber stopper and an
aluminum seal. Each vial contains 20 mg/mI (with a nominal fill of 400
mg/vial) of CP-
675,206, in a sterile aqueous solution comprising 20 mM histidine buffer, PH
5.5, 84 mg/mI
trehalose dehydrate, 0.2 mg/mi polysorbate 80, and 0.1 mg/mI disodium EDTA
dehydrate.
For all patients, ECOG performance status, vital signs, and body weight are
assessed
pre-dose, and vital signs can be repeated post-dose, as clinically indicated.
A physical
examination (including ophthalmologic assessment and signs of autoimmunity) is
performed
on Day 1. Samples for hematology panel (hematocrit, RBC count, WBC count,
differential),
chemistry (Alkaline Phosphatase, calcium, chloride, GGT, LDH, magnesium,
phosphorus,
random glucose, sodium, urea, uric acid), urinalysis (blood, protein), others
(activated partial
thromboplastin time [APTT], prothrombin time (PT), autoantibody panel, C
reactive protein,
TSH, T3, T4, amylase, lipase, serum C3, C4, serum Ig level), are obtained.
Baseline human anti-human antibody (HAHA) titer is determined and
pharmacokinetic (PK) specimen is obtained pre-dose.
The following endpoints are measured: PK parameters, HAHA, response rate and
time to progression. Time to progression and overall survival are calculated
using the
Kaplan-Meier product limit method.
EXAMPLE 7:
CP-675,206 in Combination with Imatinib Mesylate (GLEEVEC) for First-line
Treatment of Chronic Lymghocytic Leukemia
Patients having chronic lymphocytic leukemia (CLL) are given standard
chemotherapy using imatinib mesylate (GLEEVEC) per established protocols.
The patient is further administered a single IV infusion (100 mL/hr) of anti-
CTLA4
antibodies as described herein at a dose of about 3 mg/kg. Prophylactic anti-
emetics and
anti-diarrheals are given as appropriate. The treatment is repeated after 28
days with
escalation of the anti-CTLA4 antibody dose, that is, 6 mg/kg, 10 mg/kg and 15
mg/kg, every
28 days thereafter for maximum of 12 cycles in the absence of intolerable
toxicity or disease
progression. Preferably, CP-675,206 is administered at at least about 10 mg/kg
every three
weeks for four cycles and then every three months thereafter. lmatinib
mesylate (GLEEVEC,
Novartis) is administered daily at about 400 mg/day or 600 mg/day, and may be
continued as
long as there is no evidence of progressive disease or unacceptable toxicity.
Preferably, the patient is premedicated with antihistamine (H1) at least one
half hour
prior to infusion of anti-CTLA4. Premedication is recommended but not
required.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-102-
Doses are escalated using an accelerated titration design utilizing a dose-
doubling
schema with 3-6 subjects per cohort. Within each new cohort there is no
required waiting
period between subjects. Subsequent cohorts may not be opened until the first
subject at the
current dose level has been observed for 21 days and subsequent subjects have
been
observed for 14 days.
GLEEVEC is administered sequentially or simultaneously with anti-CTLA4
antibody
either once, or repeatedly, as determined.
CP-675,206 is provided in 20 ml clear glass vials with a rubber stopper and an
aluminum seal. Each vial contains 20 mg/mi (with a nominal fill of 400
mg/vial) of CP-
675,206, in a sterile aqueous solution comprising 20 mM histidine buffer, PH
5.5, 84 mg/mI
trehalose dehydrate, 0.2 mg/mi polysorbate 80, and 0.1 mg/mi disodium EDTA
dehydrate.
For all patients, ECOG performance status, vital signs, and body weight are
assessed
pre-dose, and vital signs can be repeated post-dose, as clinically indicated.
A physical
examination (including ophthalmologic assessment and signs of autoimmunity) is
performed
on Day 1. Samples for hematology panel (hematocrit, RBC count, WBC count,
differential),
chemistry (Alkaline Phosphatase, calcium, chloride, GGT, LDH, magnesium,
phosphorus,
random giucose, sodium, urea, uric acid), urinalysis (blood, protein), others
(activated partial
thromboplastin time [APTT], prothrombin time (PT), autoantibody panel, C
reactive protein,
TSH, T3, T4, amylase, lipase, serum C3, C4, serum Ig level), are obtained.
Baseline human anti-human antibody (HAHA) titer is determined and
pharmacokinetic (PK) specimen is obtained pre-dose.
The following endpoints are measured: PK parameters, HAHA, response rate and
time to progression. Time to progression and overall survival are calculated
using the
Kaplan-Meier product limit method.
EXAMPLE 8:
CP-675,206 in Combination with carbopiatin and paclitaxel for First-line
Treatment of Advanced Ovarian Carcinoma
Patients having advanced carcinoma of the ovary are given standard
chemotherapy
using carboplatin and paclitaxel per established protocols.
The patient is further administered a single IV infusion (100 mL/hr) of anti-
CTLA4
antibodies as described herein at a dose of about 3 mg/kg. Prophylactic anti-
emetics and
anti-diarrheals are given as appropriate. The treatment is repeated after 28
days with
escalation of the anti-CTLA4 antibody dose, that is, 6 mg/kg, 10 mg/kg and 15
mg/kg, every
28 days thereafter for maximum of 12 cycles in the absence of intolerable
toxicity or disease
progression. Preferably, CP-675,206 is administered at at least about 10 mg/kg
every three
weeks for four cycles and then every three months thereafter.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-103-
In an exemplary regimen, paclitaxel is administered every three weeks at 175
mg/m2
by i.v. infusion over ninety minutes followed by carboplatin (area under the
time-concentration
curve [AUC] of 5) administered i.v. over thirty to sixty minutes.
Preferably, the patient is premedicated with antihistamine (HI) at least one
half hour
prior to infusion of anti-CTLA4. Premedication is recommended but not
required.
Doses are escalated using an accelerated titration design utilizing a dose-
doubling
schema with 3-6 subjects per cohort. Within each new cohort there is no
required waiting
period between subjects. Subsequent cohorts may not be opened until the first
subject at the
current dose level has been observed for 21 days and subsequent subjects have
been
observed for 14 days.
Carboplatin and paclitaxel combination is administered sequentially or
simultaneously
with anti-CTLA4 antibody either once, or repeatedly, as determined.
CP-675,206 is provided in 20 ml clear glass vials with a rubber stopper and an
aluminum seal. Each vial contains 20 mg/ml (with a nominal fill of 400
mg/vial) of CP-
675,206, in a sterile aqueous solution comprising 20 mM histidine buffer, PH
5.5, 84 mg/mi
trehalose dehydrate, 0.2 mg/mI polysorbate 80, and 0.1 mg/ml disodium EDTA
dehydrate.
For all patients, ECOG performance status, vital signs, and body weight are
assessed
pre-dose, and vital signs can be repeated post-dose, as clinically indicated.
A physical
examination (including ophthalmologic assessment and signs of autoimmunity) is
performed
on Day 1. Samples for hematology panel (hematocrit, RBC count, WBC count,
differential),
chemistry (Alkaline Phosphatase, calcium, chloride, GGT, LDH, magnesium,
phosphorus,
random glucose, sodium, urea, uric acid), urinalysis (blood, protein), others
(activated partial
thromboplastin time [APTT], prothrombin time (PT), autoantibody panel, C
reactive protein,
TSH, T3, T4, amylase, lipase, serum C3, C4, serum Ig level), are obtained.
Baseline human anti-human antibody (HAHA) titer is determined and
pharmacokinetic (PK) specimen is obtained pre-dose.
The following endpoints are measured: PK parameters, HAHA, response rate and
time to progression. Time to progression and overall survival are calculated
using the
Kaplan-Meier product limit method.
EXAMPLE 9:
CP-675,206 in Combination with carboplatin and paclitaxel and either
bevacizumab. PF03512676, or sunitinib, for First-line Treatment of Non-Small
Cell Lung
Cancer
Patients having locally advanced Stage Illb or mestatatic Stage IV NSCLC are
given
standard chemotherapy using carboplatin and paclitaxel per established
protocols.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-104-
The patient is further administered a single IV infusion (100 mL/hr) of anti-
CTLA4
antibodies as described herein at a dose of about 15 mg/kg. Prophylactic anti-
emetics and
anti-diarrheals are given as appropriate. The treatment is repeated after 28
days for
approximately 12 cycles in the absence of intolerable toxicity or disease
progression.
Preferably, CP-675,206 is administered at at least about 10 mg/kg every three
weeks for four
cycles and then every three months thereafter.
The patient is further administered either bevacizumab (AVASTIN), PF03512676,
or
sunitinib per established protocols. Bevacizumab may be administered at about
5 mg/kg
every fourteen days by i.v. infusion until disease progression is detected.
Sunitinib may be
administered as one 50 mg oral dose taken daily, on a schedule of four weeks
on treatment
followed by two weeks off. Sunitinib may be taken with or without food. Dose
increase or
reduction of 12.5 mg increments is recommended based on individual safety and
tolerability.
An exemplary carboplatin and paclitaxel dosing regimen is.as provided supra.
Preferably, the patient is premedicated with antihistamine (H1) at least one
half hour
prior to infusion of anti-CTLA4. Premedication is recommended but not
required.
Carboplatin and paclitaxel combination is administered sequentially or
simultaneously
with anti-CTLA4 antibody either once, or repeatedly, as determined. Similarly,
bevacizumab,
sunitinib, or PF03512676 is administered sequentially or simultaneously with
anti-CTLA4-
carboplatin-paclitaxel, either once, or repeatedly, as indicated.
CP-675,206 is provided in 20 ml clear glass vials with a rubber stopper and an
aluminum seal. Each vial contains 20 mg/mI (with a nominal fill of 400
mg/vial) of CP-
675,206, in a sterile aqueous solution comprising 20 mM histidine buffer, PH
5.5, 84 mg/mI
trehalose dehydrate, 0.2 mg/mI polysorbate 80, and 0.1 mg/mI disodium EDTA
dehydrate.
For all patients, ECOG performance status, vital signs, and body weight are
assessed
pre-dose, and vital signs can be repeated post-dose, as clinically indicated.
A physical
examination (including ophthalmologic assessment and signs of autoimmunity) is
performed
on Day 1. Samples for hematology panel (hematocrit, RBC count, WBC count,
differential),
chemistry (Alkaline Phosphatase, calcium, chloride, GGT, LDH, magnesium,
phosphorus,
random glucose, sodium, urea, uric acid), urinalysis (blood, protein), others
(activated partial
thromboplastin time [APTT], prothrombin time (PT), autoantibody panel, C
reactive protein,
TSH, T3, T4, amylase, lipase, serum C3, C4, serum Ig level), are obtained.
Baseline human anti-human antibody (HAHA) titer is determined and
pharmacokinetic (PK) specimen is obtained pre-dose.
The following endpoints are measured: PK parameters, HAHA, response rate and
time to progression. Time to progression and overall survival are calculated
using the
Kaplan-Meier product limit method.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-105-
EXAMPLE 10:
CP-675,206 in Combination with either docetaxel, erlotinib or pemetrexed, for
Second Line Treatment of Non-Small Cell Lung Cancer
Patients having locally advanced Stage Illb or mestatatic Stage IV NSCLC, and
which
have previously failed platinum-based chemotherapy, are given standard
chemotherapy using
one of either docetaxel (TAXOTERE), erlotinib (TARCEVA), or pemetrexed
(ALIMTA) per
established protocols.
The patient is further administered a single IV infusion (100 mL/hr) of anti-
CTLA4
antibodies as described herein at a dose of about 15 mg/kg. Prophylactic anti-
emetics and
anti-diarrheals are given as appropriate. The treatment is repeated after 28
days for
approximately 12 cycles in the absence of intolerable toxicity or disease
progression.
Preferably, CP-675,206 is administered at at least about 10 mg/kg every three
weeks for four
cycles and then every three months thereafter.
Docetaxel (TAXOTERE, Sanofi-Aventis) is administered at about 75 mg/m2 by i.v.
infusion over one hour every three weeks. Erlotinib (TARCEVA, OSI Pharms.) is
administered as a daily dose of about 150 mg taken at least one hour or two
hours after
ingestion of food. Treatment should continue until disease progression or
unacceptable
toxicity occurs. Pemetrexed (ALIMTA, Lilly) is administered at a dose of about
500 mg/ma
administered as an i.v. infusion over 10 minutes on day 1 of each 21-day
cycle.
Preferably, the patient is premedicated with antihistamine (HI) at least one
half hour
prior to infusion of anti-CTLA4. Premedication is recommended but not
required.
Docetaxel, erlotinib or pemetrexed is administered sequentially or
simultaneously with
anti-CTLA4 antibody either once, or repeatedly, as determined.
CP-675,206 is provided in 20 ml clear glass vials with a rubber stopper and an
aluminum seal. Each vial contains 20 mg/mI (with a nominal fill of 400
mg/vial) of CP-
675,206, in a sterile aqueous solution comprising 20 mM histidine buffer, PH
5.5, 84 mg/ml
trehalose dehydrate, 0.2 mg/mi polysorbate 80, and 0.1 mg/mI disodium EDTA
dehydrate.
For all patients, ECOG performance status, vital signs, and body weight are
assessed
pre-dose, and vital signs can be repeated post-dose, as clinically indicated.
A physical
examination (including ophthalmologic assessment and signs of autoimmunity) is
performed
on Day 1. Samples for hematology panel (hematocrit, RBC count, WBC count,
differential),
chemistry (Alkaline Phosphatase, calcium, chloride, GGT, LDH, magnesium,
phosphorus,
random glucose, sodium, urea, uric acid), urinalysis (blood, protein), others
(activated partial
thromboplastin time [APTT], prothrombin time (PT), autoantibody panel, C
reactive protein,
TSH, T3, T4, amylase, lipase, serum C3, C4, serum Ig level), are obtained.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 106-

Baseline human anti-human antibody (HAHA) titer is determined and
pharmacokinetic (PK) specimen is obtained pre-dose.
The following endpoints are measured: PK parameters, HAHA, response rate and
time to progression. Time to progression and overall survival are calculated
using the
Kaplan-Meier product limit method.
EXAMPLE 11:
CP-675,206 in Combination with piatinum-based chemotherapy for Treatment of
Non-Small Cell Lung Cancer
Patients having advanced Stage Illb (with effusion) or mestatatic Stage IV
NSCLC,
and which have previously responded or remained stable after a platinum-based
chemotherapy first line regimen, are given CP-675,206 at least about three
weeks but not
more than about six weeks after the lose of first line platinum-based therapy.
The patient is further administered a single IV infusion (100 mL/hr) of anti-
CTLA4
antibodies as described herein at a dose of about 15 mg/kg. Prophylactic anti-
emetics and
anti-diarrheals are given as appropriate. The treatment is repeated after 28
days for
approximately 12 cycles in the absence of intolerable toxicity or disease
progression. More
preferably, CP-675,206 is administered at at least about 10 mg/kg every three
weeks for four
cycles and then every three months thereafter.
Preferably, the patient is premedicated with antihistamine (H1) at least one
half hour
prior to infusion of anti-CTLA4. Premedication is recommended but not
required.
Docetaxel, eriotinib or pemetrexed is administered sequentially or
simultaneously with
anti-CTLA4 antibody either once, or repeatedly, as determined.
CP-675,206 is provided in 20 ml clear glass vials with a rubber stopper and an
aluminum seal. Each vial contains 20 mg/mI (with a nominal fill of 400
mg/vial) of CP-
675,206, in a sterile aqueous solution comprising 20 mM histidine buffer, PH
5.5, 84 mg/mI
trehalose dehydrate, 0.2 mg/mI polysorbate 80, and 0.1 mg/mI disodium EDTA
dehydrate.
For all patients, ECOG performance status, vital signs, and body weight are
assessed
pre-dose, and vital signs can be repeated post-dose, as clinically indicated.
A physical
examination (including ophthalmologic assessment and signs of autoimmunity) is
performed
on Day 1. Samples for hematology panel (hematocrit, RBC count, WBC count,
differential),
chemistry (Alkaline Phosphatase, calcium, chloride, GGT, LDH, magnesium,
phosphorus,
random glucose, sodium, urea, uric acid), urinalysis (blood, protein), others
(activated partial
thromboplastin time [APTT], prothrombin time (PT), autoantibody panel, C
reactive protein,
TSH, T3, T4, amylase, lipase, serum C3, C4, serum Ig level), are obtained.
Baseline human anti-human antibody (HAHA) titer is determined and
pharmacokinetic (PK) specimen is obtained pre-dose.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-107-
The following endpoints are measured: PK parameters, HAHA, response rate and
time to progression. Time to progression and overall survival are calculated
using the
Kaplan-Meier product limit method.

EXAMPLE 12:
Natural history of diarrhea associated with CTLA4 blockade by anti-CTLA4
antibody
Diarrhea resulting from immune activation has been associated with CTLA4
blockade. For example in patients with stage IV melanoma receiving ipilimumab
(MDX-01 0), a
number of patients developed grade 3/4 autoimmune enterocolitis and severe
diarrhea (Attia
et al., 2005). In a single-dose phase I trial of CP-675,206 at doses up to 15
mg/kg in patients
with solid tumors (n = 39), 9 instances of diarrhea were reported including 3
grade 3 events
(Ribas et al., 2005). The incidence and severity of diarrhea was assessed in
patients
receiving CP-675,206 in a large phase I/II study.
An open-label phase I/II trial of CP-675,206 was conducted in patients with
stage III
(unresectable) or stage IV melanoma and an ECOG PS <_ 1. Diarrhea was assessed
in
patients treated at the phase II doses: 10 mg/kg once per month (Q1 M) in
phase I (n = 22), or
10 mg/kg Q1 M (n = 44) or 15 mg/kg once every 3 months (Q3M) (n = 45) in phase
II.
Medians of 3.5 doses (range, 1 to 18) at 10 mg/kg Q1 M(10Q1 M) in phase 1, 3
doses
(range, 1 to 26) at 10 mg/kg Q1 M in phase II, and 1 dose (range, I to 9) at
15 mg/kg Q3M
(15Q3M) were administered with 100% dose compliance. Treatment-related
diarrhea was
reported by 43 (39%) of 111 patients, and grade 3 diarrhea occurred in 14
(13%) patients.
One patient had grade 4 colitis resulting in a colectomy. Diarrhea (all
grades) occurred with
similar frequency in each dose group; however, grade 3 treatment-related
diarrhea occurred
in 8% of patients treated with 15 mg/kg Q3M compared with 18% of patients
treated with 10
mg/kg QIM in phase I and 14% of patients treated with 10 mg/kg QIM in phase
II. Among 9
patients with an objective response, 8 experienced diarrhea (3 of which were
grade 3). The
majority of cases (65%) were mild to moderate in severity with a median time
to onset of 51
days (range, 1 to 583 days) and resolution of 8 days (range, 1 to 182 days).
More than half of
patients who reported serious events of diarrhea were treated with steroids.
Diarrhea associated with CP-675,206 was primarily mild to moderate in
severity,
transient, and manageable. In addition, 15 mg/kg Q3M may be better tolerated
than 10 mg/kg
Q1 M.

EXAMPLE 13:
Survival of patients with metastatic melanoma treated with anti-CTLA4
monoclonal
antibody CP-675,206 in a phase I/II study


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 108-

CP-675,206 has demonstrated clinical activity in patients with metastatic
melanoma.
Prolonged survival was observed in a prior single-dose phase I study, even in
patients who
did not achieve objective tumor responses.
A multidose phase I/II trial was conducted in patients (N = 119) with
histologically
confirmed stage Illc (unresectable) or stage IV recurrent metastatic melanoma
and ECOG PS
s 1. The study consisted of a phase I, open-label, multidose study (3, 6, and
10 mg/kg) and a
phase I expansion cohort for HLA-A2.1 + patients (10 mg/kg monthly [10Q1 M]),
followed by a
phase II open-label study of 2 dosing regimens: 10 mg/kg Q1 M and 15 mg/kg
every 3 months
(15Q3M). The primary endpoint was safety in phase I, immune monitoring in the
expansion
cohort, and response in phase II. Survival was analyzed as a secondary
endpoint.
In the phase I study, Kaplan-Meier estimates of median overall survival were
17.6
months for all dose groups combined (n = 28). In the phase II study, median
survival was 10.3
months in the 10 mg/kg arm and 11.0 months in the 15 mg/kg arm. Survival
outcomes were
favorable, compared with historical median survival of 7 months, independent
of whether
patients achieved an objective response.
Kaplan-Meier estimate* of
median survival, months (95%
Regimen Enrolled, n Died, n (%) CI)
Phase I 28 18 (64.3) 17.6 (8, 25.2)
3 mg/kg 3 3(100.0) 8.0 (4.4, 17.8)
6 mg/kg 3 3(100.0) 7.5 (4.4, 19.5)
10 mg/kg 8 6(75.0) 19.3 (6.01, NE)

10 mg/kg expansion 14 6(42.9) 25.1 (9.9, NE)
Phase II

10 mg/kg, Q1 M 44 29 (65.8) 10.3 (6.6, 13.2)
15 mg/kg, Q3M 45 32 (71.1) 11.0 (8.1, 16.7)
*Patients censored when last known to be alive.

NE = Cannot be estimated.

Patients participating in a multiple dose study of CP-675,206 showed a
survival time
that was greater than expected on historic controls.


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-109-
EXAMPLE 14:
Comparison of dose and dosing schedule of CP-675,206 anti-CTLA4 monoclonal
antibody in a Phase II clinical trial in patients with advanced melanoma
A two-stage, two-arm phase II trial was conducted to assess the optimal dosing
regimen for pivotal clinical trial testing of CP-675,206.
Eligible patients had measurable melanoma (stage Illc or IV) progressing on or
after
prior therapy with ECOG PS <_ 1. In stage 1, 18 patients per arm were
randomized to either 10
mg/kg once per month (10Q1 M) or 15 mg/kg every 3 months (15Q3M). If 3 or more
patients
in either arm had complete response (CR) or partial response (PR), then 25
more patients
were entered to that arm. Primary endpoint was objective tumor response (OR),
and
secondary endpoints were safety and survival.
Eighty-nine (89) patients received at least 1 dose (44 at 10Q1 M, 45 at
15Q3M), with
both study arms moving to stage 2. Ninety-six percent of patients had stage IV
disease,. and
57% had elevated LDH. There were no significant differences in age, sex,
stage, or baseline
LDH levels between study groups. A median of 3 doses (range, 1 to 26) at 10Q1
M and 1
dose (range, 1 to 9) at 15Q3M were administered with 100% compliance. Dose
delays
occurred in 30% of patients treated at 10Q1 M and 16% at 15Q3M. Two (2)
patients at 10
Q1 M and five (5) patients at 15 Q3M continued on study beyond 12 months.
Six patients at 10Q1 M were discontinued due to the following toxicity:
three due to diarrhea/colitis (one patient requiring colectomy); one due to
Grave's
ophthalmopathy; one due to pancreatitis; and one due to hypersensitivity
reaction. Two
patients at 15Q3M were discontinued: one due to colitis and pancreatitis, and
the other due to
diarrhea (P = 0.14). There were no toxic deaths. The dose of 15Q3M was
associated with
lower incidence of grade 3 or 4 adverse events (AEs), that is, 31 % compared
with 41 % at 10
Q1M;P=0.42.
Responses by investigator assessment were 1 CR and 3 PRs at 10 Q1 M, and 1 CR
and 2 PRs at 15 Q3M, including responses in skin, lymph nodes (LN), bone,
liver, lung, and
adrenal glands. Only 1 patient with PR at 10Q1 M relapsed, and the remaining
responses
were ongoing (18+ to 28+ months). Median survival was 10.3 months at 10Q1 M
and 11.0
months at 15Q3M (P = NS).
The 15 mg/kg every three months (15Q3M) regimen was chosen for further
clinical
testing based on comparable antitumor efficacy and a trend to improved
feasibility and safety
compared with 10 mg/kg every month (10Q1 M).


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
-110-
EXAMPLE 15:
Increased anti-virus titers in combination immunization of Rhesus monkeys
Combination therapy comprising administration of a vaccine and anti-CTLA4
antibody
mediated an increased antiviral immune response.
Rhesus macaques were immunized with FLUZONE subvirion 1998-99 formula (NDC
49281-362-11 Influenza Virus Vaccine, Trivalent Types A and B; Aventis Pasteur
Inc.
(includes Connaught Labs., Pasteur Merieux and Pasteur Merieux Connaught)).
The 1998-
99 vaccine contained hemagglutinin A (HA) from three strains: A/Beijing/262/95
(H1 N1),
A/Sydney/5/97 (H3N2), and B/Harbin/07/94 (a B/Beijing/184/93-like strain). The
monkeys
were immunized intramuscularly (IM) with FLUZONE on day 0 (week 0) and again 4
weeks
later (week 4). Antibody was administered according to the following schedule:
Group 1 was
administered anti-CTLA4 antibody CP-642,570 (5 mg/kg) intravenously (IV) on
week 0;
Group 2 was administered an irrelevant control antibody (anti-KLH antibody; 5
mg/kg IV) on
week 0; Group 3 was immunized IM with FLUZONE on week 0 and reimmunized IM
with
FLUZONE on week 4 and administered anti-CTLA4 antibody (5 mg/kg IV) on week 4;
Group
4 was immunized IM with FLUZONE on week 0 and reimmunized IM with FLUZONE on
week
4 and administered an irrelevant control antibody (anti-KLH antibody at 5
mg/kg IV) on week
4; and Group 5 was immunized IM with FLUZONE on week 0 and reimmunized IM with
FLUZONE on week 4 but no antibody was administered. The animals were followed
for eight
weeks after the initial immunization with FLUZONE.
The anti-CTLA4 antibody administered was human anti-human CTLA4 designated
CP-642,570 (also referred to herein as antibody 4.1.1).
Serum was collected at I hour and 24 hours after anti-CTLA4 administration and
every two weeks thereafter. The following parameters were assessed: anti-CTLA4
serum
levels, IgG titers to FLUZONE, level of Neopterin, and level of 2-5 adenylate
synthetase.
Plasma levels of anti-CTLA4 antibody confirmed exposure, i.e., plasma levels
were
approximately 75 Ng/mI at 1 hour, 40-50 pg/mi at 24 hours and, for the most
part, not
detectable by two weeks (Figure 4). One animal in Group 3 had undetectable
levels of
antibody, possibly due to a missed injection or a switched tube.
The animals were assessed for various markers of immune activation. Neopterin
levels were variable (Figure 5). That is, an initial spike in value was
detected in the anti-
CTLA4 group two weeks post-immunization and treatment. Four weeks after
immunization,
the basal levels dropped and no differences were observed between the groups,
including
groups 3 and 4.
Whole blood was obtained and cell pellets were collected and assayed for 2-5
adenylate synthetase. Groups I and 2 were compared (Figure 6). The data
indicate that


CA 02647282 2008-09-24
WO 2007/113648 PCT/IB2007/000860
- 111-

there was a detectable increase in the anti-CTLA4 group (Group 1) compared
with the control
group.
An increase in anti-FLUZONE IgG level was detected at six weeks in animals
treated
with anti-CTLA4 at the time of the second immunization (Group 3) (Figure 7).
No effect on
anti-FLUZONE titer was observed in any of the other groups of animals. The
anti-FLUZONE
IgG titers for each individual animal is depicted in Figure 8 for prebleed
(week -1) (+), week 0
(open triangle), week 2 (dark triangle), week 4 (open square), week 6 (shaded
circle), and
week 8 (open circle). The data points are grouped according to test group,
i.e., Group 1, 2, 3,
4, and 5. Values shown in boxes indicate repeat titers from the same sample.
Figure 9 shows the anti-FLUZONE serum IgG titers at week 6 in Rhesus monkeys.
Individual animal results were grouped according to test group and indicate
that Group 3
animals (administered anti-CTLA4 antibody on week 4) demonstrated increased
anti-
FLUZONE IgG (flu-IgG) titers compared with the other animals. One animal
exhibited an
increased flu-IgG titer but anti-CTLA4 exposure (by plasma level assay) could
not be
confirmed. While some spikes in anti-flu antibody titers were observed in
control groups, the
titers were lower in magnitude.
In summary, an increase in flu-IgG and 2-5 adenylate synthetase was observed
in
animals administered anti-CTLA4 antibody suggesting that anti-CTLA4
administered to virally
infected patients, to patients immunized with viral antigens, or a portion
thereof, and/or when
combined with anti-viral treatment enhanced therapeutic benefit.
The disclosures of each and every patent, patent application, and publication
cited
herein are hereby incorporated herein by reference in their entirety.
While the invention has been disclosed with reference to specific embodiments,
it is
apparent that other embodiments and variations of this invention may be
devised by others
skilled in the art without departing from the true spirit and scope of the
invention. The
appended claims are intended to be construed to include all such embodiments
and
equivalent variations.


CA 02647282 2008-09-24

111a
SEQUENCE LISTING IN ELECTRONIC FORM

In accordance with Section 111(1) of the Patent Rules, this description
contains a sequence listing in electronic form in ASCII text format
(file: 50054-196 Seq 16-SEP-08 vl.txt).

A copy of the sequence listing in electronic form is available from the
Canadian Intellectual Property Office.

The sequences in the sequence listing in electronic form are reproduced
in the following table.

SEQUENCE TABLE
<110> PFIZER PRODUCTS INC.
Gomez-Navarro, Jesus
Gladue, Ronald G.

<120> CTLA4 Antibody Combination Therapy
<130> PC32630A

<140> app no. to be assigned
<141> 2007-03-07

<150> US 60/789,662
<151> 2006-04-05
<160> 36

<170> PatentIn version 3.4
<210> 1
<211> 1392
<212> DNA
<213> Homo sapiens
<400> 1
atggagtttg ggctgagctg ggttttcctc gttgctcttt taagaggtgt ccagtgtcag 60
gtgcagctgg tggagtctgg gggaggcgtg gtccagcctg ggaggtccct gagactctcc 120
tgtgtagcgt ctggattcac cttcagtagc catggcatgc actgggtccg ccaggctcca 180
ggcaaggggc tggagtgggt ggcagttata tggtatgatg gaagaaataa atactatgca 240
gactccgtga agggccgatt caccatctcc agagacaatt ccaagaacac gctgtttctg 300
caaatgaaca gcctgagagc cgaggacacg gctgtgtatt actgtgcgag aggaggtcac 360
ttcggtcctt ttgactactg gggccaggga accctggtca ccgtctcctc agcctccacc 420
aagggcccat cggtcttccc cctggcgccc tgctccagga gcacctccga gagcacagcg 480
gccctgggct gcctggtcaa ggactacttc cccgaaccgg tgacggtgtc gtggaactca 540
ggcgctctga ccagcggcgt gcacaccttc ccagctgtcc tacagtcctc aggactctac 600
tccctcagca gcgtggtgac cgtgccctcc agcaacttcg gcacccagac ctacacctgc 660
aacgtagatc acaagcccag caacaccaag gtggacaaga cagttgagcg caaatgttgt 720
gtcgagtgcc caccgtgccc agcaccacct gtggcaggac cgtcagtctt cctcttcccc 780
ccaaaaccca aggacaccct catgatctcc cggacccctg aggtcacgtg cgtggtggtg 840
gacgtgagcc acgaagaccc cgaggtccag ttcaactggt acgtggacgg cgtggaggtg 900
cataatgcca agacaaagcc acgggaggag cagttcaaca gcacgttccg tgtggtcagc 960
gtcctcaccg ttgtgcacca ggactggctg aacggcaagg agtacaagtg caaggtctcc 1020


CA 02647282 2008-09-24

111b
aacaaaggcc tcccagcccc catcgagaaa accatctcca aaaccaaagg gcagccccga 1080
gaaccacagg tgtacaccct gcccccatcc cgggaggaga tgaccaagaa ccaggtcagc 1140
ctgacctgcc tggtcaaagg cttctacccc agcgacatcg ccgtggagtg ggagagcaat 1200
gggcagccgg agaacaacta caagaccaca cctcccatgc tggactccga cggctccttc 1260
ttcctctaca gcaagctcac cgtggacaag agcaggtggc agcaggggaa cgtcttctca 1320
tgctccgtga tgcatgaggc tctgcacaac cactacacgc agaagagcct ctccctgtct 1380
ccgggtaaat ga 1392
<210> 2
<211> 444
<212> PRT
<213> Homo sapiens
<400> 2
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Val Val Gln Pro Gly Arg
1 5 10 15
Ser Leu Arg Leu Ser Cys Val Ala Ser Gly Phe Thr Phe Ser Ser His
20 25 30
Gly Met His Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Val Ile Trp Tyr Asp Gly Arg Asn Lys Tyr Tyr Ala Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Phe
65 70 75 80
Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly His Phe Gly Pro Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro
115 120 125
Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly
130 135 140
Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn
145 150 155 160
Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln
165 170 175
Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser
180 185 190
Asn Phe Gly Thr Gln Thr Tyr Thr Cys Asn Val Asp His Lys Pro Ser
195 200 205
Asn Thr Lys Val Asp Lys Thr Val Glu Arg Lys Cys Cys Val Glu Cys
210 215 220
Pro Pro Cys Pro Ala Pro Pro Val Ala Gly Pro Ser Val Phe Leu Phe
225 230 235 240
Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val
245 250 255
Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Gln Phe
260 265 270
Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro
275 280 285
Arg Glu Glu Gln Phe Asn Ser Thr Phe Arg Val Val Ser Val Leu Thr
290 295 300
Val Val His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val
305 310 315 320
Ser Asn Lys Gly Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Thr
325 330 335
Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg
340 345 350


CA 02647282 2008-09-24

111c
Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly
355 360 365
Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro
370 375 380
Glu Asn Asn Tyr Lys Thr Thr Pro Pro Met Leu Asp Ser Asp Gly Ser
385 390 395 400
Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln
405 410 415
Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His
420 425 430
Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys
435 440
<210> 3
<211> 118
<212> PRT
<213> Homo sapiens
<400> 3
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Val Val Gln Pro Gly Arg
1 5 10 15
Ser Leu Arg Leu Ser Cys Val Ala Ser Gly Phe Thr Phe Ser Ser His
20 25 30
Gly Met His Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Val Ile Trp Tyr Asp Gly Arg Asn Lys Tyr Tyr Ala Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Phe
65 70 75 80
Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly His Phe Gly Pro Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 4
<211> 10
<212> PRT
<213> Homo sapiens
<400> 4
Gly Phe Thr Phe Ser Ser His Gly Met His
1 5 10
<210> 5
<211> 15
<212> PRT
<213> Homo sapiens
<400> 5
Val Ile Trp Tyr Asp Gly Arg Asn Lys Tyr Tyr Ala Asp Ser Val
1 5 10 15
<210> 6
<211> 9


CA 02647282 2008-09-24

111d
<212> PRT
<213> Homo sapiens
<400> 6
Gly Gly His Phe Gly Pro Phe Asp Tyr
1 5

<210> 7
<211> 708
<212> DNA
<213> Homo sapiens
<400> 7
atggaaaccc cagcgcagct tctcttcctc ctgctactct ggctcccaga taccaccgga 60
gaaattgtgt tgacgcagtc tccaggcacc ctgtctttgt ctccagggga aagagccacc 120
ctctcctgca gggccagtca gagtattagc agcagcttct tagcctggta ccagcagaga 180
cctggccagg ctcccaggct cctcatctat ggtgcatcca gcagggccac tggcatccca 240
gacaggttca.gtggcagtgg gtctgggaca gacttcactc tcaccatcag cagactggag 300
cctgaagatt ttgcagtgta ttactgtcag cagtatggta cctcaccctg gacgttcggc 360
caagggacca aggtggaaat caaacgaact gtggctgcac catctgtctt catcttcccg 420
ccatctgatg agcagttgaa atctggaact gcctctgttg tgtgcctgct gaataacttc 480
tatcccagag aggccaaagt acagtggaag gtggataacg ccctccaatc gggtaactcc 540
caggagagtg tcacagagca ggacagcaag gacagcacct acagcctcag cagcaccctg 600
acgctgagca aagcagacta cgagaaacac aaagtctacg cctgcgaagt cacccatcag 660
ggcctgagct cgcccgtcac aaagagcttc aacaggggag agtgttag 708
<210> 8
<211> 215
<212> PRT
<213> Homo sapiens
<400> 8
Glu Ile Val Leu Thr Gln Ser Pro Gly Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Ile Ser Ser Ser
20 25 30
Phe Leu Ala Trp Tyr Gin Gin Arg Pro Gly Gln Ala Pro Arg Leu Leu
35 40 45
Ile Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser
50 55 60
Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu
65 70 75 80
Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Tyr Gly Thr Ser Pro
85 90 95
Trp Thr Phe Gly Gln Gly Thr Lys Val Glu Ile Lys Arg Thr Val Ala
100 105 110
Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu Lys Ser
115 120 125
Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro Arg Glu
130 135 140
Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly Asn Ser
145 150 155 160
Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr Ser Leu
165 170 175
Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His Lys Val
180 185 190


CA 02647282 2008-09-24

111e
Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val Thr Lys
195 200 205
Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 9
<211> 108
<212> PRT
<213> Homo sapiens
<400> 9
Glu Ile Val Leu Thr Gln Ser Pro Gly Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Ile Ser Ser Ser
20 25 30
Phe Leu Ala Trp Tyr Gln Gln Arg Pro Gly Gln Ala Pro Arg Leu Leu
35 40 45
Ile Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser
50 55 60
Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu
65 70 75 80
Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Tyr Gly Thr Ser Pro
85 90 95
Trp Thr Phe Gly Gln Gly Thr Lys Val Glu Ile Lys
100 105
<210> 10
<211> 12
<212> PRT
<213> Homo sapiens
<400> 10
Arg Ala Ser Gln Ser Ile Ser Ser Ser Phe Leu Ala
1 5 10
<210> il
<211> 7
<212> PRT
<213> Homo sapiens
<400> 11
Gly Ala Ser Ser Arg Ala Thr
1 5
<210> 12
<211> 9
<212> PRT
<213> Homo sapiens
<400> 12
Gln Gln Tyr Gly Thr Ser Pro Trp Thr
1 5

<210> 13
<211> 1335


CA 02647282 2008-09-24

ilif
<212> DNA
<213> Homo sapiens
<400> 13
caggtgcagc tggtggagtc tgggggaggc gtggtccagc ctgggaggtc cctgagactc 60
tcctgtgcag cgtctggatt caccttcagt agtcatggca tccactgggt ccgccaggct 120
ccaggcaagg ggctggagtg ggtggcagtt atatggtatg atggaagaaa taaagactat 180
gcagactccg tgaagggccg attcaccatc tccagagaca attccaagaa cacgctgtat 240
ttgcaaatga acagcctgag agccgaggac acggctgtgt attactgtgc gagagtggcc 300
ccactggggc cacttgacta ctggggccag ggaaccctgg tcaccgtctc ctcagcctcc 360
accaagggcc catcggtctt ccccctggcg ccctgctcca ggagcacctc cgagagcaca 420
gcggccctgg gctgcctggt caaggactac ttccccgaac cggtgacggt gtcgtggaac 480
tcaggcgctc tgaccagcgg cgtgcacacc ttcccagctg tcctacagtc ctcaggactc 540
tactccctca gcagcgtggt gaccgtgccc tccagcaact tcggcaccca gacctacacc 600
tgcaacgtag atcacaagcc cagcaacacc aaggtggaca agacagttga gcgcaaatgt 660
tgtgtcgagt gcccaccgtg cccagcacca cctgtggcag gaccgtcagt cttcctcttc 720
cccccaaaac ccaaggacac cctcatgatc tcccggaccc ctgaggtcac gtgcgtggtg 780
gtggacgtga gccacgaaga ccccgaggtc cagttcaact ggtacgtgga cggcgtggag 840
gtgcataatg ccaagacaaa gccacgggag gagcagttca acagcacgtt ccgtgtggtc 900
agcgtcctca'ccgttgtgca ccaggactgg ctgaacggca aggagtacaa gtgcaaggtc 960
tccaacaaag gcctcccagc ccccatcgag aaaaccatct ccaaaaccaa agggcagccc 1020
cgagaaccac aggtgtacac cctgccccca tcccgggagg agatgaccaa gaaccaggtc 1080
agcctgacct gcctggtcaa aggcttctac cccagcgaca tcgccgtgga gtgggagagc 1140
aatgggcagc cggagaacaa ctacaagacc acacctccca tgctggactc cgacggctcc 1200
ttcttcctct acagcaagct caccgtggac aagagcaggt ggcagcaggg gaacgtcttc 1260
tcatgctccg tgatgcatga ggctctgcac aaccactaca cgcagaagag cctctccctg 1320
tctccgggta aatga 1335
<210> 14
<211> 444
<212> PRT
<213> Homo sapiens
<400> 14
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Val Val Gln Pro Gly Arg
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser His
20 25 30
Gly Ile His Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Val Ile Trp Tyr Asp Gly Arg Asn Lys Asp Tyr Ala Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr
65 70 75 80
Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Val Ala Pro Leu Gly Pro Leu Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro
115 120 125
Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly
130 135 140
Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn
145 150 155 160
Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln
165 170 175
Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser
180 185 190


CA 02647282 2008-09-24

111g
Asn Phe Gly Thr Gln Thr Tyr Thr Cys Asn Val Asp His Lys Pro Ser
195 200 205
Asn Thr Lys Val Asp Lys Thr Val Glu Arg Lys Cys Cys Val Glu Cys
210 215 220
Pro Pro Cys Pro Ala Pro Pro Val Ala Gly Pro Ser Val Phe Leu Phe
225 230 235 240
Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val
245 250 255
Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Gln Phe
260 265 270
Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro
275 280 285
Arg Glu Glu Gln Phe Asn Ser Thr Phe Arg Val Val Ser Val Leu Thr
290 295 300
Val Val His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val
305 310 315 320
Ser Asn Lys Gly Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Thr
325 330 335
Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg
340 345 350
Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly
355 360 365
Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro
370 375 380
Glu Asn Asri Tyr Lys Thr Thr Pro Pro Met Leu Asp Ser Asp Gly Ser
385 390 395 400
Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln
405 410 415
Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His
420 425 430
Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys
435 440
<210> 15
<211> 118
<212> PRT
<213> Homo sapiens
<400> 15
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Val Val Gln Pro Gly Arg
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser His
20 25 30
Gly Ile His Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Val Ile Trp Tyr Asp Gly Arg Asn Lys Asp Tyr Ala Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr
65 70 75 80
Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Val Ala Pro Leu Gly Pro Leu Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 16
<211> 10


CA 02647282 2008-09-24

111h
<212> PRT
<213> Homo sapiens
<400> 16
Gly Phe Thr Phe Ser Ser His Gly Ile His
1 5 10
<210> 17
<211> 15
<212> PRT
<213> Homo sapiens
<400> 17
Val Ile Trp Tyr Asp Gly Arg Asn Lys Asp Tyr Ala Asp Ser Val
1 5 10 15
<210> 18
<211> 9
<212> PRT
<213> Homo sapiens
<400> 18
Val Ala Pro Leu Gly Pro Leu Asp Tyr
1 5

<210> 19
<211> 645
<212> DNA
<213> Homo sapiens
<400> 19
gaaattgtgt tgacgcagtc tccaggcacc ctgtctttgt ctccagggga aagagccacc 60
ctctcctgca gggccagtca gagtgtcagc agctacttag cctggtacca gcagaaacct 120
ggccaggctc ccaggctcct catctatggt gcatccagca gggccactgg catcccagac 180
aggttcagtg gcagtgggtc tgggacagac ttcactctca ccatcagcag actggagcct 240
gaggattttg cagtgtatta ctgtcaacag tatggtaggt caccattcac tttcggccct 300
gggaccaaag tagatatcaa gcgaactgtg gctgcaccat ctgtcttcat cttcccgcca 360
tctgatgagc agttgaaatc tggaactgcc tctgttgtgt gcctgctgaa taacttctat 420
cccagagagg ccaaagtaca gtggaaggtg gataacgccc tccaatcggg taactcccag 480
gagagtgtca cagagcagga cagcaaggac agcacctaca gcctcagcag caccctgacg 540
ctgagcaaag cagactacga gaaacacaaa gtctacgcct gcgaagtcac ccatcagggc 600
ctgagctcgc ccgtcacaaa gagcttcaac aggggagagt gttag 645
<210> 20
<211> 214
<212> PRT
<213> Homo sapiens
<400> 20
Glu Ile Val Leu Thr Gln Ser Pro Gly Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Val Ser Ser Tyr
20 25 30
Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu Ile
35 40 45


CA 02647282 2008-09-24

111i
Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu Pro
65 70 75 80
Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Tyr Gly Arg Ser Pro Phe
85 90 95
Thr Phe Gly Pro Gly Thr Lys Val Asp Ile Lys Arg Thr Val Ala Ala
100 105 110
Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Giu Gln Leu Lys Ser Gly
115 120 125
Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro Arg Glu Ala
130 135 140
Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly Asn Ser Gln
145 150 155 160
Glu Ser Val. Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr Ser Leu Ser
165 170 175
Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His Lys Val Tyr
180 185 190
Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val Thr Lys Ser
195 200 205
Phe Asn Arg Gly Glu Cys
210
<210> 21
<211> 107
<212> PRT
<213> Homo sapiens
<400> 21
Glu Ile Val Leu Thr Gln Ser Pro Gly Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Val Ser Ser Tyr
. 20 25 30
Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu Ile
35 40 45
Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu Pro
65 70 75 80
Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Tyr Gly Arg Ser Pro Phe
85 90 95
Thr Phe Gly Pro Gly Thr Lys Val Asp Ile Lys
100 105
<210> 22
<211> 11
<212> PRT
<213> Homo sapiens
<400> 22
Arg Ala Ser. Gln Ser Val Ser Ser Tyr Leu Ala
1 5 10
<210> 23
<211> 7
<212> PRT
<213> Homo sapiens


CA 02647282 2008-09-24

111j
<400> 23
Gly Ala Ser Ser Arg Ala Thr
1 5
<210> 24
<211> 9
<212> PRT
<213> Homo sapiens
<400> 24
Gln Gln Tyr Gly Arg Ser Pro Phe Thr
1 5

<210> 25
<211> 1413
<212> DNA
<213> Homo sapiens
<400> 25
atggagtttg ggctgagctg ggttttcctc gttgctcttt taagaggtgt ccagtgtcag 60
gtgcagctgg tggagtctgg gggaggcgtg gtccagcctg ggaggtccct gagactctcc 120
tgtgcagcgt ctggattcac cttcagtagc tatggcatgc actgggtccg ccaggctcca 180
ggcaaggggc tggagtgggt ggcagttata tggtatgatg gaagtaataa atactatgca 240
gactccgtga agggccgatt caccatctcc agagacaatt ccaagaacac gctgtatctg 300
caaatgaaca gcctgagagc cgaggacacg gctgtgtatt actgtgcgag agatccgagg 360
ggagctaccc tttactacta ctactacggt atggacgtct ggggccaagg gaccacggtc 420
accgtctcct cagcctccac caagggccca tcggtcttcc ccctggcgcc ctgctccagg 480
agcacctccg agagcacagc ggccctgggc tgcctggtca aggactactt ccccgaaccg 540
gtgacggtgt cgtggaactc aggcgctctg accagcggcg tgcacacctt cccagctgtc 600
ctacagtcct caggactcta ctccctcagc agcgtggtga ccgtgccctc cagcaacttc 660
ggcacccaga cctacacctg caacgtagat cacaagccca gcaacaccaa ggtggacaag 720
acagttgagc gcaaatgttg tgtcgagtgc ccaccgtgcc cagcaccacc tgtggcagga 780
ccgtcagtct tcctcttccc cccaaaaccc aaggacaccc tcatgatctc ccggacccct 840
gaggtcacgt gcgtggtggt ggacgtgagc cacgaagacc ccgaggtcca gttcaactgg 900
tacgtggacg gcgtggaggt gcataatgcc aagacaaagc cacgggagga gcagttcaac 960
agcacgttcc gtgtggtcag cgtcctcacc gttgtgcacc aggactggct gaacggcaag 1020
gagtacaagt-gcaaggtctc caacaaaggc ctcccagccc ccatcgagaa aaccatctcc 1080
aaaaccaaag ggcagccccg agaaccacag gtgtacaccc tgcccccatc ccgggaggag 1140
atgaccaaga accaggtcag cctgacctgc ctggtcaaag gcttctaccc cagcgacatc 1200
gccgtggagt gggagagcaa tgggcagccg gagaacaact acaagaccac acctcccatg 1260
ctggactccg acggctcctt cttcctctac agcaagctca ccgtggacaa gagcaggtgg 1320
cagcagggga acgtcttctc atgctccgtg atgcatgagg ctctgcacaa ccactacacg 1380
cagaagagcc tctccctgtc tccgggtaaa tga 1413
<210> 26
<211> 451
<212> PRT
<213> Homo sapiens
<400> 26
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Val Val Gln Pro Gly Arg
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr
20 25 30
Gly Met His Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45


CA 02647282 2008-09-24

111k
Ala Val Ile Trp Tyr Asp Gly Ser Asn Lys Tyr Tyr Ala Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr
65 70 75 80
Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Asp Pro Arg Gly Ala Thr Leu Tyr Tyr Tyr Tyr Tyr Gly Met
100 105 110
Asp Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser Ser Ala Ser Thr
115 120 125
Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser
130 135 140
Glu Ser Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu
145 150 155 160
Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His
165 170 175
Thr Phe Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser
180 185 190
Val Val Thr Val Pro Ser Ser Asn Phe Gly Thr Gln Thr Tyr Thr Cys
195 200 205
Asn Val Asp His Lys Pro Ser Asn Thr Lys Val Asp Lys Thr Val Glu
210 215 220
Arg Lys Cys Cys Val Glu Cys Pro Pro Cys Pro Ala Pro Pro Val Ala
225 230 235 240
Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met
245 250 255
Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His
260 265 270
Glu Asp Pro Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val
275 280 285
His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Phe
290 295 300
Arg Val Val Ser Val Leu Thr Val Val His Gln Asp Trp Leu Asn Gly
305 310 315 320
Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ala Pro Ile
325 330 335
Glu Lys Thr Ile Ser Lys Thr Lys Gly Gln Pro Arg Glu Pro Gln Val
340 345 350
Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn Gln Val Ser
355 360 365
Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu
370 375 380
Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro
385 390 395 400
Met Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val
405 410 415
Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met
420 425 430
His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser
435 440 445
Pro Gly Lys
450
<210> 27
<211> 125
<212> PRT
<213> Homo sapiens


CA 02647282 2008-09-24
1111
<400> 27
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Val Val Gln Pro Gly Arg
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr
20 25 30
Gly Met His Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Val Ile Trp Tyr Asp Gly Ser Asn Lys Tyr Tyr Ala Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr
65 70 75 80
Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Asp Pro Arg Gly Ala Thr Leu Tyr Tyr Tyr Tyr Tyr Gly Met
100 105 110
Asp Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser Ser
115 120 125
<210> 28
<211> 10
<212> PRT
<213> Homo sapiens
<400> 28
Gly Phe Thr Phe Ser Ser Tyr Gly Met His
1 5 10
<210> 29
<211> 15
<212> PRT
<213> Homo sapiens
<400> 29
Val Ile Trp Tyr Asp Gly Ser Asn Lys Tyr Tyr Ala Asp Ser Val
1 5 10 15
<210> 30
<211> 16
<212> PRT
<213> Homo sapiens
<400> 30
Asp Pro Arg Gly Ala Thr Leu Tyr Tyr Tyr Tyr Tyr Gly Met Asp Val
1 5 10 15
<210> 31
<211> 714
<212> DNA
<213> Homo sapiens
<400> 31
atggacatga gggtccccgc tcagctcctg gggctcctgc tactctggct ccgaggtgcc 60
agatgtgaca tccagatgac ccagtctcca tcctccctgt ctgcatctgt aggagacaga 120
gtcaccatca cttgccgggc aagtcagagc attaacagct atttagattg gtatcagcag 180
aaaccaggga aagcccctaa actcctgatc tatgctgcat ccagtttgca aagtggggtc 240
ccatcaaggt tcagtggcag tggatctggg acagatttca ctctcaccat cagcagtctg 300


= CA 02647282 2008-09-24

111m
caacctgaag attttgcaac ttactactgt caacagtatt acagtactcc attcactttc 360
ggccctggga ccaaagtgga aatcaaacga actgtggctg caccatctgt cttcatcttc 420
ccgccatctg atgagcagtt gaaatctgga actgcctctg ttgtgtgcct gctgaataac 480
ttctatccca gagaggccaa agtacagtgg aaggtggata acgccctcca atcgggtaac 540
tcccaggaga gtgtcacaga gcaggacagc aaggacagca cctacagcct cagcagcacc 600
ctgacgctga.gcaaagcaga ctacgagaaa cacaaagtct acgcctgcga agtcacccat 660
cagggcctga gctcgcccgt cacaaagagc ttcaacaggg gagagtgtta gtga 714
<210> 32
<211> 214
<212> PRT
<213> Homo sapiens
<400> 32
Asp Ile Gln Met Thr G1n Ser Pro Ser Ser Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Ser Ile Asn Ser Tyr
20 25 30
Leu Asp Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile
35 40 45
Tyr Ala Ala Ser Ser Leu Gln Ser Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln Tyr Tyr Ser Thr Pro Phe
85 90 95
Thr Phe G1y Pro Gly Thr Lys Val Glu I1e Lys Arg Thr Val Ala Ala
100 105 110
Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu Lys Ser Gly
115 120 125
Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro Arg Glu Ala
130 135 140
Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly Asn Ser Gln
145 150 155 160
Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr Ser Leu Ser
165 170 175
Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His Lys Val Tyr
180 185 190
Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val Thr Lys Ser
195 200 205
Phe Asn Arg Gly Glu Cys
210
<210> 33
<211> 107
<212> PRT
<213> Homo sapiens
<400> 33
Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Ser Ile Asn Ser Tyr
20 25 30
Leu Asp Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile
35 40 45
Tyr Ala Ala Ser Ser Leu Gln Ser Gly Va1 Pro Ser Arg Phe Ser Gly
50 55 60


CA 02647282 2008-09-24

111n
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln Tyr Tyr Ser Thr Pro Phe
85 90 95
Thr Phe Gly Pro Gly Thr Lys Val Glu Ile Lys
100 105
<210> 34
<211> 11
<212> PRT.
<213> Homo sapiens
<400> 34
Arg Ala Ser Gln Ser Ile Asn Ser Tyr Leu Asp
1 5 10
<210> 35
<211> 7
<212> PRT
<213> Homo sapiens
<400> 35
Ala Ala Ser Ser Leu Gln Ser
1 5
<210> 36
<211> 9
<212> PRT
<213> Homo sapiens
<400> 36
Gln Gln Tyr Tyr Ser Thr Pro Phe Thr
1 5

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2007-03-26
(87) PCT Publication Date 2007-10-11
(85) National Entry 2008-09-24
Examination Requested 2008-09-24
Dead Application 2011-03-28

Abandonment History

Abandonment Date Reason Reinstatement Date
2010-03-26 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Request for Examination $800.00 2008-09-24
Registration of a document - section 124 $100.00 2008-09-24
Application Fee $400.00 2008-09-24
Maintenance Fee - Application - New Act 2 2009-03-26 $100.00 2008-09-24
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
PFIZER PRODUCTS INC.
Past Owners on Record
GOMEZ-NAVARRO, JESUS
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2008-09-25 125 7,345
Representative Drawing 2009-02-02 1 45
Cover Page 2009-02-02 1 77
Abstract 2008-09-24 1 95
Claims 2008-09-24 3 147
Drawings 2008-09-24 12 434
Description 2008-09-24 111 6,820
PCT 2008-09-24 7 205
Assignment 2008-09-24 3 126
Prosecution-Amendment 2008-09-24 16 580

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :