Language selection

Search

Patent 2752553 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 2752553
(54) English Title: ANTI-CD20 ANTIBODIES AND FUSION PROTEINS THEREOF AND METHODS OF USE
(54) French Title: ANTICORPS ANTI-CD20 ET PROTEINES HYBRIDES DESDITS ANTICORPS, ET METHODES D'UTILISATION
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07K 16/46 (2006.01)
  • A61K 47/68 (2017.01)
  • A61K 39/395 (2006.01)
  • A61K 51/10 (2006.01)
  • C07K 16/28 (2006.01)
  • C07K 19/00 (2006.01)
  • C12N 15/13 (2006.01)
(72) Inventors :
  • HANSEN, HANS (United States of America)
  • QU, ZHENGXING (United States of America)
  • GOLDENBERG, DAVID M. (United States of America)
(73) Owners :
  • IMMUNOMEDICS, INC. (United States of America)
(71) Applicants :
  • IMMUNOMEDICS, INC. (United States of America)
(74) Agent: BERESKIN & PARR LLP/S.E.N.C.R.L.,S.R.L.
(74) Associate agent:
(45) Issued:
(22) Filed Date: 2003-02-14
(41) Open to Public Inspection: 2003-08-21
Examination requested: 2011-09-13
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): No

(30) Application Priority Data:
Application No. Country/Territory Date
60/356,132 United States of America 2002-02-14
60/416,232 United States of America 2002-10-07

Abstracts

English Abstract




The present invention provides humanized, chimeric and human anti-CD20
antibodies and CD 20 antibody fusion proteins that bind to a human B cell
marker, referred to as CD20, which is useful for the treatment and diagnosis
of B-cell disorders, such as B-cell malignancies and autoimmune diseases, and
methods of treatment and diagnosis.


Claims

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





Claims:

1. A chimeric, humanized or human monoclonal antibody that binds to
CD20, said antibody comprising a light chain variable region CDR1 with a
sequence
selected from the group consisting of RASSSVSYIH (SEQ ID NO:23),
RASSSLSFMH (SEQ ID NO:24) and RASSSVSYMH (SEQ ID NO:25); CDR2 with a
sequence ATSNLAS (SEQ ID NO:26); and CDR3 with a sequence selected from the
group consisting of QQWTSNPPT (SEQ ID NO:27), HQWSSNPLT (SEQ ID NO:28)
and QQSFSNPPT (SEQ ID NO:29); and said antibody comprising a heavy chain
variable region CDR1 with a sequence SYNMH (SEQ ID NO:30); CDR2 with a
sequence AIYPGNGDTSYNQKFKG (SEQ ID NO:31); and CDR3 with a sequence
selected from the group consisting of SHYGSNYVDYFDV (SEQ ID NO-34) and
VVYYSNSYWYFDV (SEQ ID NO:35), or an antigen-binding fragment thereof
selected from the group consisting of F(ab')2, Fab', Fab, Fv and sFv.

2. A chimeric, humanized or human monoclonal antibody that binds to
CD20, said antibody comprising a light chain variable region CDR1 with a
sequence
selected from the group consisting of RASSSLSFMH (SEQ ID NO:24) and
RASSSVSYMH (SEQ ID NO:25); CDR2 with a sequence ATSNLAS (SEQ ID
NO:26); and CDR3 with a sequence selected from the group consisting of
QQWTSNPPT (SEQ ID NO:27) and HQWSSNPLT (SEQ ID NO:28); and said
antibody comprising a heavy chain variable region CDR1 with a sequence SYNMH
(SEQ ID NO:30); CDR2 with a sequence AIYPGNGDTSYNQKFKG (SEQ ID NO:31);
and CDR3 is STYYGGDWYFDV (SEQ ID NO:32), or an antigen-binding fragment
thereof selected from the group consisting of F(ab')2, Fab', Fab, Fv and sFv.

3. A chimeric, humanized or human monoclonal antibody that binds to
CD20, said antibody comprising a light chain variable region CDR1 with a
sequence
selected from the group consisting of RASSSVSYIH (SEQ ID NO:23),
RASSSLSFMH (SEQ ID NO:24) and RASSSVSYMH (SEQ ID NO:25); CDR2 with a
sequence ATSNLAS (SEQ ID NO:26); and CDR3 with a sequence HQWSSNPLT
(SEQ ID NO:28); and said antibody comprising a heavy chain variable region
CDR1
with a sequence SYNMH (SEQ ID NO:30); CDR2 with a sequence
AIYPGNGDTSYNQKFKG (SEQ ID NO:31); and CDR3 is STYYGGDWYFDV (SEQ
ID NO:32), or an antigen-binding fragment thereof selected from the group
consisting
of F(ab')2, Fab', Fab, Fv and sFv.

68




4. A chimeric, humanized or human monoclonal antibody that binds to
CD20, said antibody comprising a light chain variable region CDR1 with a
sequence
selected from the group consisting of RASSSLSFMH (SEQ ID NO:24) and
RASSSVSYMH (SEQ ID NO:25); CDR2 with a sequence ATSNLAS (SEQ ID
NO:26); and CDR3 with a sequence selected from the group consisting of
QQWTSNPPT (SEQ ID NO:27), HQWSSNPLT (SEQ ID NO-28) and QQSFSNPPT
(SEQ ID NO:29); and said antibody comprising a heavy chain variable region
CDR1
with a sequence SYNMH (SEQ ID NO:30); CDR2 with a sequence
AIYPGNGDTSYNQKFKG (SEQ ID NO:31); and CDR3 with a sequence selected
from the group consisting of STYYGGDWYFDV (SEQ ID NO:32),
SHYGSNYVDYFDV (SEQ ID NO:34) and VVYYSNSYWYFDV (SEQ ID NO-35), or
an antigen-binding fragment thereof selected from the group consisting of
F(ab')2,
Fab', Fab, Fv and sFv.

5. An immunoconjugate comprising a chimeric, humanized or human
monoclonal antibody that binds to CD20 according to any one of claims 1 to 4,
to
which a therapeutic agent is attached.

6. An immunoconjugate according to claim 5, wherein the therapeutic
agent is a therapeutic radionuclide.

7. An immunoconjugate according to claim 6, wherein the therapeutic
radionuclide is selected from the group consisting of 32P, 33P, 47Sc, 64Cu,
67Cu, 67Ga,
90Y, 111Ag 111In 1251, 1311142Pr 153Sm 161Tb, 166Dy 166 Ho 177Lu 186Re, 188 Re
189Re
212Pb 212Bi, 213Bi, 211At 223Ra and 225Ac.

8. An immunoconjugate according to claim 6, wherein the therapeutic
radionuclide is a radionuclide of iodine.

9. An immunoconjugate according to claim 5, wherein the therapeutic
agent is a drug.

10. An immunoconjugate according to claim 9, wherein the drug is selected
from the group consisting of vinca alkaloids, anthracyclines,
epidophyllotoxin,
taxanes, antimetabolites, alkylating agents, antikinase agents, antibiotics,
Cox-2
inhibitors, antimitotic, antiangiogenic and apoptotic agents, nitrogen
mustards, alkyl
sulfonates, nitrosoureas, triazenes, folic acid analogs, COX-2 inhibitors,
pyrimidine
analogs, purine analogs, platinum coordination complexes and hormones.

69




11. An immunoconjugate according to claim 5, wherein the therapeutic
agent is a toxin.

12. A composition comprising a chimeric, humanized or human monoclonal
antibody as claimed in claim 1, and: (i) a second antibody that targets an
antigen
other than CD20, and/or (ii) an immunmodulator.

13. A composition comprising a chimeric, humanized or human monoclonal
antibody as claimed in claim 1, and a second antibody that targets an antigen
other
than CD20.

14. A composition as claimed in claim 12 or claim 13, wherein the second
antibody targets an antigen selected from the group consisting of CD4, CD5,
CD8,
CD14, CD15, CD19, CD21, CD22, CD23, CD25, CD33, CD37, CD38, CD40,
CD40L, CD46, CD52, CD54, CD74, CD80, CD126, B7, MUC1, 1a, HM1.24, or HLA-
DR, tenascin, VEGF, PIGF, an oncogene, and an oncogene product.

15. A composition according to claim 12 or 13, wherein the second
antibody targets CD22.

16. A composition according to claim 12 or 13, wherein the second
antibody targets CD19.

17. A composition according to claim 12 or 13, wherein the second
antibody targets CD74.

18. A composition according to claim 12 or 13, wherein the second
antibody targets HLA-DR.

19. A composition comprising a chimeric, humanized or human monoclonal
antibody as claimed in claim 1, and an immunomodulator.

20. A composition as claimed in claim 19, wherein the immunomodulator is
selected from the group consisting of a cytokine, a stem cell growth factor, a

lymphotoxin, a hematopoietic factor, a colony stimulating factor (CSF), an
interferon
(IFN), erythropoietin and thrombopoietin.

21. An antibody fusion protein, comprising at least two monoclonal
antibodies, wherein at least one of said antibodies is the antibody according
to any
one of claims 1 to 4.






22. The antibody fusion protein according to claim 21, comprising at least
one monoclonal antibody that targets an antigen other than CD20.

23. The antibody fusion protein according to claim 21, comprising at least
one monoclonal antibody that targets CD22.

24. A nucleic acid molecule comprising an isolated DNA sequence
encoding the monoclonal antibody as claimed in any one of claims 1 to 4.

25. A nucleic acid molecule comprising an isolated DNA sequence
encoding the antibody fusion protein as claimed in any one of claims 21 to 23.

26. An expression vector comprising the DNA sequence of claim 24.
27. An expression vector comprising the DNA sequence of claim 25.
28. A host cell transformed with the DNA sequence of claim 24.

29. A host cell transformed with the DNA sequence of claim 25.

30. A pdHL2 expression vector comprising the DNA sequence of claim 24.
31. A pdHL2 expression vector comprising the DNA sequence of claim 25.
32. A use of a therapeutically effective amount of the antibody as claimed
in any one of claims 1 to 4, or immunoconjugate as claimed in any one of
claims 5 to
11, or composition as claimed in any one of claims 12 to 20, or antibody
fusion
protein as claimed in any one of claims 21 to 23, for treating a B-cell
lymphoma or
leukemia in a subject.

33. The use according to claim 32, wherein said antibody is used
parenterally.

34. The use according to claim 33, wherein said use is intravenous.
35. The use according to claim 33, wherein said use is subcutaneous.

36. The use according to claim 33, comprising using a dose of about 1-20
mg/kg.

37. The use according to claim 33, comprising using a dose of about 1
mg/kg.

38. The use according to claim 37, wherein said dose is used weekly.
39. The use according to claim 38, wherein from 4 to 8 doses are used.
71



40. The use according to claim 37, wherein said dose is used every other
week.

41. The use according to claim 37, wherein said dose is used every 2 to 3
weeks, and is repeated for a total of at least 3 dosages.

42. The use according to claim 37, wherein said dose is used every 2 to 3
weeks for 2-3 months.

43. The use according to claim 37, wherein the interval between doses is
at least two weeks.

44. The use according to claim 37, wherein said doses are used for up to 3
months.

45. A use of a therapeutically effective amount of the antibody as claimed
in any one of claims 1 to 4, or immunoconjugate as claimed in any one of
claims 5 to
11, or composition as claimed in any one of claims 12 to 20, or antibody
fusion
protein as claimed in any one of claims 21 to 23, for treating an autoimmune
disease
in a subject, wherein said autoimmune disease is mediated by B-cells.

46. The use according to claim 45, wherein said antibody is used
parenterally.

47. The use according to claim 46, wherein said use is intravenous.
48. The use according to claim 46, wherein said use is subcutaneous.

49. The use according to claim 46, comprising using a dose of about 1-20
mg/kg.

50. The use according to claim 46, comprising using a dose of about 1
mg/kg.

51. The use according to claim 50, wherein said dose is used weekly.
52. The use according to claim 51, wherein from 4 to 8 doses are used.

53. The use according to claim 50, wherein said dose is used every other
week.

54. The use according to claim 50, wherein said dose is used every 2 to 3
weeks, and is repeated for a total of at least 3 dosages.

72




55. The use according to claim 50, wherein said dose is used every 2 to 3
weeks for 2-3 months.

56. The use according to claim 50, wherein the interval between doses is
at least two weeks.

57. The use according to claim 50, wherein said doses are used for up to 3
months.

58. The antibody fusion protein according to claim 21, comprising at least
one monoclonal antibody that targets CD37.

59. The antibody fusion protein according to claim 21, comprising at least
one monoclonal antibody that targets CD74.

60. The antibody fusion protein according to claim 21, comprising at least
one monoclonal antibody that targets CD19.

61. The antibody fusion protein according to claim 21, comprising at least
one monoclonal antibody that targets CD52.

62. The antibody fusion protein according to claim 21, comprising at least
one monoclonal antibody that targets HLA-DR.

63. The antibody fusion protein according to claim 21, comprising at least
one monoclonal antibody that targets MUC-1.

64. A composition comprising the chimeric, human or humanized antibody
according to any one of claims 1 to 4 and at least one monoclonal antibody
that
targets an antigen other than CD20.

65. A composition comprising the chimeric, human or humanized antibody
according to any one of claims 1 to 4 and at least one monoclonal antibody
that
targets CD22.

66. A composition comprising the chimeric, human or humanized antibody
according to any one of claims 1 to 4 and at least one monoclonal antibody
that
targets CD37.

67. A composition comprising the chimeric, human or humanized antibody
according to any one of claims 1 to 4 and at least one monoclonal antibody
that
targets CD74.

73




68. A composition comprising the chimeric, human or humanized antibody
according to any one of claims 1 to 4 and at least one monoclonal antibody
that
targets CD19.

69. A composition comprising the chimeric, human or humanized antibody
according to any one of claims 1 to 4 and at least one monoclonal antibody
that
targets CD52.

70. A composition comprising the chimeric, human or humanized antibody
according to any one of claims 1 to 4 and at least one monoclonal antibody
that
targets HLA-DR.

71. A composition comprising the chimeric, human or humanized antibody
according to any one of claims 1 to 4 and at least one monoclonal antibody
that
targets MUC-1.

72. A composition comprising the chimeric, human or humanized antibody
according to any one of claims 1 to 4 packaged for subcutaneous administration
and
a pharmaceutically acceptable excipient.

73. The composition according to claim 72, comprising dosage instructions
which specify administration over a period of less than about 3 hours.

74. The composition according to claim 73, wherein said dosage
instructions specify administration of from 25 to 50 mg of the dose within the
first 15-
30 minutes, and specify that the remainder of the dose is to be given over a
period of
2-3 hours.

75. The composition comprising the chimeric, human or humanized
antibody according to any one of claims 1 to 4, packaged in doses of about 1
mg/kg
and a pharmaceutically acceptable excipient.

74

Description

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



CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665

Anti-CD20 Antibodies and Fusion Proteins Thereof and
Methods of Use

BACKGROUND OF THE INVENTION
1. Field of the Invention
The present invention relates to humanized, chimeric and human anti-CD20
antibodies, particularly monoclonal antibodies (mAbs) therapeutic and
diagnostic
conjugates of humanized, chimeric and human anti-CD20 antibodies and methods
of
treating B cell lymphomas and leukemias and various autoimmune diseases using
humanized, chimeric and human anti-CD20 antibodies. The present invention
relates
to antibody fusion proteins or fragments thereof comprising at least two anti-
CD20
mAbs or fragments thereof or at least one anti-CD20 MAb or fragment thereof
and at
least one second MAb or fragment thereof, other than the antiCD20 MAb or
fragment
thereof. The humanized, chimeric and human anti-CD20 mAbs, fragments thereof,
antibody fusion proteins thereof or fragments thereof may be administered
alone, as a
therapeutic conjugate or in combination with a therapeutic immunoconjugate,
with
other naked antibodies, or with therapeutic agents or as a diagnostic
conjugate. The
present invention relates to DNA sequences encoding humanized, chimeric and
human anti-CD20 antibodies, and antibody fusion proteins, vectors and host
cells
containing the DNA sequences, and methods of making the humanized, chimeric
and
human anti-C620 antibodies.
2. Background
The immune system of vertebrates consists of a number of organs and cell
types which have evolved to accurately recognize foreign antigens,
specifically bind
to, and eliminate/destroy such foreign antigens. Lymphocytes, amongst others,
are
critical to the immune system. Lymphocytes are divided into two major sub-
populations, T cells and B cells. Although inter-dependent, T cells are
largely
responsible for cell-mediated immunity and B cells are largely responsible for
antibody production (humoral immunity).


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
In humans, each B cell can produce an enormous number of antibody
molecules. Such antibody production typically ceases (or substantially
decreases)
when a foreign antigen has been neutralized. Occasionally, however,
proliferation of a
particular B cell will continue unabated and may result in a cancer known as a
B cell
lymphoma. B-cell lymphomas, such as the B-cell subtype of non-Hodgkin's
lymphoma, are significant contributors to cancer mortality. The response of B-
cell
malignancies to various forms of treatment is mixed. For example, in cases in
which
adequate clinical staging of non-Hodgkin's lymphoma is possible, field
radiation
therapy can provide satisfactory treatment. Still, about one-half of the
patients die
from the disease. Devesa et al., J Nat'l Cancer Inst. 79:701 (1987).
The majority of chronic lymphocytic leukemias are of B-cell lineage.
Freedman, Hematol. Oncol. Clin. North Ain. 4:405 (1990). This type of B-cell
malignancy is the most common leukemia in the Western world. Goodman et al.,
Leukemia and Lymphoma 22:1 (1996). The natural history of chronic lymphocytic
leukemia falls into several phases. In the early phase, chronic lymphocytic
leukemia
is an indolent disease, characterized by the accumulation of small mature
functionally-incompetent malignant B-cells having a lengthened life span.
Eventually, the doubling time of the malignant B-cells decreases and patients
become
increasingly symptomatic. While treatment can provide symptomatic relief, the
overall survival of the patients is only minimally affected. The late stages
of chronic
lymphocytic leukemia are characterized by significant anemia and/or
thrombocytopenia. At this point, the median survival is less than two years.
Foon et
al., Annals Int. Medicine 113:525 (1990). Due to the very low rate of cellular
proliferation, chronic lymphocytic leukemia is resistant to cytotoxic drug
treatment.
Traditional methods of treating B-cell malignancies, including chemotherapy
and radiotherapy, have limited utility due to toxic side effects. The use of
monoclonal
antibodies to direct radionuclides, toxins, or other therapeutic agents offers
the
possibility that such agents can be delivered selectively to tumor sites, thus
limiting
toxicity to normal tissues. Also, the presence of B-cell antigens on these B-
cell
malignancies makes them optimal targets for therapy with unconjugated B-cell
antibodies, such as against CD19, CD20, CD21, CD23, and CD22 markers on B-
cells.
MA-DR and other antigens may serve as targets for normal and malignant B-
cells,
although they are also expressed on other cell types. Further, certain MUC1,
MUC2,

2


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
MUC3, and MUC4 antigens, preferably MUCI, are also expressed in different
hematopoietic malignancies, including B-cell tumors expressing CD20 and other
B-
cell markers. Still other antigen targets, such as those associated with the
vascular
endothelium of tumors, including tenascin, vascular endothelium growth factor
(VEGF), and placental growth factor (P1GF), as well as other categories of
antigens
associated with B-cell malignancies, such as oncogene products, are also
suitable
targets for said complementary antibodies for use in the present invention.
B cells comprise cell surface proteins which can be utilized as markers for
differentiation and identification. One such human B-cell marker is the human
B
lymphocyte-restricted differentiation antigen Bp35, referred to as CD20. CD20
is
expressed during early pre-B cell development and remains until plasma cell
differentiation. CD20 is expressed on both normal B cells and malignant B
cells
whose abnormal growth can lead to B-cell lymphomas. Antibodies against the
CD20
antigen have been investigated for the therapy of B-cell lymphomas. For
example, a
chimeric anti-CD20 antibody, designated as "IDEC-C2B8," has activity against B-
cell
lymphomas when provided as unconjugated antibodies at repeated injections of
doses
exceeding 500 mg per injection. Maloney et al., Blood 84:2457 (1994); Longo,
Curr.
Opin. Oncol. 8:353 (1996). About 50 percent of non-Hodgkin's patients, having
the
low-grade indolent form, treated with this regimen showed responses.
Therapeutic
responses have also been obtained using 1311-labeled B 1 anti-CD20 murine
monoclonal antibody when provided as repeated doses exceeding 600 mg per
injection. Kaminski et al., N. Engl. J Med. 329:459 (1993); Press et al., N.
Engl. J.
Med. 329:1219 (1993); Press et al., Lancet 346:336 (1995). However, these
antibodies, whether provided as unconjugated forms or radiolabeled forms, have
not
shown high rates of objective and durable responses in patients with the more
prevalent and lethal form of B-cell lymphoma, the intermediate or aggressive
type.
Therefore, a need exists to develop an immunotherapy for B-cell malignancies
that
achieves a therapeutic response of significant duration.
Additional studies targeting CD20 surface antigen have been demonstrated
using an anti-CD20 marine monoclonal antibody, IF5, which was administered by
continuous intravenous infusion to B cell lymphoma patients. Extremely high
levels
(>2 grams) of 1F5 were reportedly required to deplete circulating tumor cells,
and the
results were described as being "transient." Press et al.,
"Monoclonal*Antibody IF5
3


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
(Anti-CD-20) Serotherapy of Human B-Cell Lymphomas." Blood 69/2:584-591
(1987). However, a potential problem with this approach is that non-human
monoclonal antibodies (e.g., murine monoclonal antibodies) typically lack
human
effector functionality, i.e., they are unable to mediate complement-dependent
lysis or
lyse human target cells through antibody-dependent cellular toxicity or Fc-
receptor
mediated phagocytosis. Furthermore, non-human monoclonal antibodies can be
recognized by the human host as a foreign protein and, therefore, repeated
injections
of such foreign antibodies can lead to the induction of immune responses
leading to
harmful hypersensitivity reactions. For murine-based monoclonal antibodies,
this is
often referred to as a Human Anti-Mouse Antibody (HAMA) response.
The use of chimeric antibodies is more preferred because they do not elicit as
strong a HAMA response as murine antibodies. Chimeric antibodies are
antibodies
which comprise portions from two or more different species. For example, Liu,
A. Y.
et al, "Production of a Mouse-Human Chimeric Monoclonal Antibody to CD20 with
Potent Fc-Dependent Biologic Activity" J Inunun. 139/10:3521-3526 (1987),
describe a mouselhuman chimeric antibody directed against the CD20 antigen.
See
also, PCT Publication No. WO 88/04936. However, no information is provided as
to
the ability, efficacy or practicality of using such chimeric antibodies for
the treatment
of B cell disorders in the reference. It is noted that in vitro functional
assays (e.g.,
complement-dependent lysis (CDC); antibody dependent cellular cytotoxicity
(ADCC), etc.) cannot inherently predict the in vivo capability of a chimeric
antibody
to destroy or deplete target cells expressing the specific antigen. See, for
example,
Robinson, R. D. et al., "Chimeric mouse-human anti-carcinoma antibodies that
mediate different anti-tumor cell biological activities," Hum. Antibod
Hybridomas
2:84-93 (1991) (chimeric mouse-human antibody having undetectable ADCC
activity). Therefore, the potential therapeutic efficacy of a chimeric
antibody can only
truly be assessed by in vivo experimentation, preferably in the species of
interest for
the specific therapy.
One approach that has improved the ability of murine monoclonal antibodies
to be effective in the treatment of B-cell disorders has been to conjugate a
radioactive
label or chemotherapeutic agent to the antibody, such that the label or agent
is
localized at the tumor site. For example, the above-referenced 1F5 antibody
and other
B-cell antibodies have been labeled with 131I and were reportedly evaluated
for

4


CA 02752553 2011-09-13

WO 03/068821 PCTIGB03/00665
biodistribution in two patients. See Eary, J. F. et al., "Imaging and
Treatment of B-
Cell Lymphoma" J. Nuc. Med. 31/8:1257-1268 (1990); see also, Press, O. W. et
al.,
"Treatment of Refractory Non-Hodgkin's Lymphoma with Radiolabeled MB-1 (Anti-
CD37) Antibody" J. Clin. Onc. 7/8:1027-1038 (1989) (indication that one
patient
treated with 1311-labeled IF-5 achieved a partial response); Goldenberg, D. M.
et al.,
"Targeting, Dosimetry and Radioimmunotherapy of B-Cell Lymphomas with 131I-
Labeled LL2 Monoclonal Antibody" J. Clin. Oncol. 9/4:548-564 (1991) (three of
eight patients receiving multiple injections reported to have developed a HAMA
response to this CD22 murine antibody); Appelbaum, F. R. "Radiolabeled
Monoclonal Antibodies in the Treatment of Non-Hodgkin's Lymphoma" Hem./Oncol.
Clinics of N. A. 5/5:1013-1025 (1991) (review article); Press, O. W. et al.
"Radiolabeled-Antibody Therapy of B-Cell Lymphoma with Autologous Bone
Marrow Support." Nev, England Journal of Medicine 329/17: 1219-12223 (1993)
(1311-labeled anti-CD20 antibody IF5 and B1); and Kaminski, M. G. et al
"Radioimmunotherapy of B-Cell Lymphoma with [1311] Anti-B 1 (Anti-CD20)
Antibody". NEJM32917:459 (1993) (131I-labeled anti-CD20 antibody B1;
hereinafter
"Kaminski"); PCT published application WO 92/07466 (antibodies conjugated to
chemotherapeutic agents such as doxorubicin or mitomycin). However, these
approaches have not eliminated the obstacles associated with using murine
antibodies,
despite the fact that many patients with lymphoma who have received prior
aggressive
cytotoxic chemotherapy are immune suppressed, thus having lower HAMA rates
than
lymphoma patients who have not been heavily pretreated.
Autoimmune diseases are a class of diseases associated with B-cell disorders.
Examples include immune-mediated thrombocytopenias, such as acute idiopathic
thrombocytopenic purpura and chronic idiopathic thrombocytopenic purpura,
myasthenia gravis, lupus nephritis, lupus erythematosus, and rheumatoid
arthritis.
The most common treatments are corticosteroids and cytotoxic drugs, which can
be
very toxic. These drugs also suppress the entire immune system, can result in
serious
infection, and have adverse affects on the bone marrow, liver and kidneys.
Other
therapeutics that have been used to treat Class III autoimmune diseases to
date have
been directed against T-cells and macrophages. There is a need for more
effective
methods of treating autoimmune diseases, particularly Class III autoimmune
diseases.


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
To address the many issues related to B-cell disorders and their treatment,
the
present invention provides humanized, chimeric and human anti-CD20 monoclonal
antibodies with the same complementarity determining regions (CDRs) that bind
to
the CD20 antigen of the present invention used alone, conjugated to a
therapeutic
agent or in combination with other treatment modalities, for the treatment of
B cell
lymphomas and leukemias and autoimmune disorders in humans and other mammals
without the adverse responses associated with using murine antibodies.

SUMMARY OF THE INVENTION
Accordingly, the present invention provides humanized, chimeric and human
anti-CD20 antibodies that bind to a human B cell marker, referred to as CD20,
which
is useful for the treatment and diagnosis of B-cell disorders, such as B-cell
malignancies and autoinmune diseases.
The present invention further provides methods of treatment of mammalian
subjects, such as humans or domestic animals, with one or more humanized,
chimeric
and human CD20 antibodies, alone, as an antibody fusion protein, as a
therapeutic
conjugate alone or as part of an antibody fusion protein, in combination, or
as a
multimodal therapy, with other antibodies, other therapeutic agents or
immunomodulators or as an immunoconjugate linked to at least one therapeutic
agent,
therapeutic radionuclide or immunomodulator. These humanized, chimeric and
human CD20 antibodies can also be used as a diagnostic imaging agent alone, in
combination with other diagnostic imaging agents, and/or in conjunction with
therapeutic applications.
The present invention additionally is directed to anti-CD20 mAbs or fragments
thereof that contain specific murine CDRs or a combination of murine CDRs from
more than one murine or chimeric anti-CD20 MAb that have specificity for CD20.
These mAbs can be humanized, chimeric or human anti-CD20 mAbs.
The present invention is also directed to antibody fusion proteins comprising
at least two anti-CD2OmAbs or fragments thereof or a first MAb comprising an
anti-
CD20mAbs or fragments thereof and a second MAb.
The present invention is further directed to a therapeutic or diagnostic
conjugates of the anti-CD20 mAbs or fragments thereof or antibody fusion
proteins of
the anti-CD20 mAbs or other mAbs or fragments thereof bound to at least one
6


CA 02752553 2011-09-13

therapeutic agent or at least one diagnostic agent. Antibody fusion proteins
with multiple
therapeutic agents of the same or different type are encompassed by the
present invention.
The present invention is additionally directed to a method of using the anti-
CD20
mAbs or fragments thereof or antibody fusion proteins thereof or fragments
thereof for
therapy, either alone, in combination with each other, as the antibody
component of a
therapeutic immunoconjugate with one or more therapeutic agents or each
administered in
combination with one or more therapeutic agents or with an immunoconjugate
with one or
more therapeutic agents.
The present invention further is directed to a method of using the anti-CD20
mAbs
or fragments thereof or antibody fusion proteins thereof or fragments thereof
as a diagnostic
bound to one or more diagnostic agents.
The present invention additionally is directed to a method of pretargeting a
cell in a
patients suffering from a B-cell lymphoma or leukemia or an autoimmune disease
using an
antibody fusion protein or fragment thereof of the present invention.

BRIEF DESCRIPTION OF THE FIGURES
Figure 1 discloses the V gene sequences cloned by RT-PCR from a hybridoma cell
line producing a murine anti-CD20, and the deduced amino acid sequences of the
variable
light (Figure IA) (SEQ ID NOS: 1-2) and heavy chain (Figure 1B) (SEQ ID NOS: 3-
4) of
the A20 antibody, designated as A2OVk and A20VH, respectively. Underlined
arrows
indicate the sequences of the PCR primers used for cloning. The putative CDR
region
sequences, as defined by the Kabat numbering scheme, are shown in bold and
underlined.
Amino acid sequences are given as single-letter codes below the corresponding
nucleotide
sequence. The Kabat numbering scheme was used for amino acid residues. Amino
acid
residues numbered by a letter represent the insertion residue according to
Kabat, and have
the same number as that of the previous residue. For example, residues 82,
82A, 82B and
82C in Figure 1 B are indicated as 82 A, B, and C, respectively.
Figure 2 discloses the Vk, the variable light chain, and the VH, the variable
heavy chain, sequences of cA20, a chimeric anti-CD20 antibody. The CDR region
sequences are shown in bold and underlined. The amino acid residues and the
nucleotides are numbered sequentially and same numbering system is used for
7


CA 02752553 2011-09-13

humanized V sequences. The light chain variable region is shown in Fig. 2A
(SEQ ID NOS: 5-
6) and the heavy chain variable region is shown in Fig. 2B (SEQ ID NOS: 7-8).
The
numbering system is the same as for Figure 1. The restriction sites used for
constructing cA20
are underlined.
Figure 3 shows a comparison of the binding affinities of the chimeric A20
(cA20), and
murine A20, (A20), in a cell surface competitive binding assay against 12SI-
Tabled A20.
Increasing concentrations of cA20 blocked the binding of radiolabeled A20 to
Raji cells (as
depicted by closed circles) in a comparable manner as that of murine A20
(depicted by closed
diamonds).
Figure 4 compares the amino acid sequences of the variable heavy chain (VH)
and
variable light chain (Vk) of human antibodies, and chimeric and humanzied anti-
CD20
antibodies. Figure 4A compares the amino acid sequences of the variable heavy
chain (VH) of
the human antibodies, EU (SEQ ID NO: 9) and NEWM (FR4 only) (SEQ ID NO: 55),
the
chimeric antibody, (cA20VH) (SEQ ID NO: 8) and two humanized antibodies,
(hA20VHI and
hA20VH2) (SEQ ID NOS: 10-11) and Figure 4B compares the amino acid sequences
of the
variable light chain (Vk) of the human antibody, (REIVk) (SEQ ID NO: 12), a
chimeric
antibody, (cA20Vk) (SEQ ID NO: 6), and a humanized antibody, (hA20Vk) (SEQ ID
NO: 2).
Dots indicate that the residues in A20 are identical to the corresponding
residue in the human
antibody. The CDRs are identified as a boxed region. The Kabat numbering
scheme was used
to number the amino acid residues.
Figure 5 discloses the nucleotide sequences of hA20 light chain V genes,
(hA20Vk)
(Figure 5A) (SEQ ID NOS: 13-14), and heavy chain V genes, hA20VH1 (Figure 513)
(SEQ ID
NOS: 15-16) and hA20VH2 (Figure 5C) (SEQ ID NOS: 17-18), as well as the
adjacent
flanking sequences of the VKpBR2 (Figure 5A) and VHpBS2 (Figures 5B and 5C)
staging
vectors, respectively. The non-translated nucleotide sequences are shown in
lowercase. The
restriction sites used for subcloning are underlined and indicated. The
secretion signal peptide
sequence is indicated by a double underline. Numbering of Vk and VH amino acid
residues is
same as that in Figure 2.
Figure 6 shows the results of a cell surface competitive binding assay to
compare
the binding activity of two humanized A20 antibodies, (hA20-1 and hA20-2),
with that of
A20, cA20 and a chimeric anti-CD20 MAb, c2B8. Figure 6A shows hA20-1 (closed
triangles) and hA20-2 (closed circles) and the murine anti-CD20 antibody, A20
(closed
squares) competed equally well for the binding of 1251-A20 to Raji cells.
Figure 6B shows
hA20-1 (closed circles), cA20 (closed squares) and c2B8 (closed diamonds)
competed
equally well for the binding of 125 1-c2138 to Raji cells.

8


CA 02752553 2011-09-13

Figure 7 discloses the constant region of a human IgGI (CH-hinge) (Figure
7A) (SEQ ID NOS 19-20) and the constant region of a human kappa chain (Ck)
(Figure 7B) (SEQ ID NOS: 21-22).
Figure 8 is a competitive cell surface binding assay. Ag-binding specificity
and affinity studies of humanized anti-CD20 Abs (cA20, hA20, and c1F5,
purified by
affinity chromatography on a Protein A column) were evaluated by a cell
surface
competitive binding assay with murine 2B8 and rituximab (IDEC Pharmaceuticals
Corp., San Diego, CA). Figure 8 (A) is a comparison of the binding activities
of cA20
(square), hA20-1 (triangle) and hA20-1 (circle) with that of m2B8 (diamond);
figure 8
(B) compares of the binding activities of cA20 (square), clF5 (triangle) and
rituximab
(diamond).

Figure 9 is a study comparing the binding activities of hA20 with other anti-
CD20 Abs, including rituximab and murine 131, by a cell surface competitive
binding
assay. A constant amount (100,000 cpm, _10 iCi/1g) of 1251-labeled rituximab
was
incubated with Raji cells in the presence of varying concentrations (0.2-700
nM) of
competing Abs, hA20 (triangle), mBl (Downward triangle) or rituximab (square)
at
4 C fort-2 h.

Figure 10 depicts the cytotoxic effect of crosslinked hA20 and other CD20
Abs on cultured lymphoma cells. Total cell and viable cell cell populations
were
measured by (A) trypan blue staining and cell counting or (B) MTT assay.

Figure 11 is a graph of in vivo therapy studies with various anti-CD20 and
other Abs. Raji cells administered i.v. to SCID mice, to create a Raji
lymphoma
model of disseminated disease.

Figure 12 is a graph depicting in vivo therapy with hA20 and hLL2. Raji cells
administered i.v. to SCID mice, to create a Raji lymphoma model of
disseminated
disease.

DETAILED DESCRIPTION OF THE INVENTION
1. Overview

As discussed above, anti-CD20 antibodies that are unconjugated or labeled
with a therapeutic radionuclide, have failed to provide high rates of
objective and
lasting responses in patients with intermediate or aggressive forms of B-cell
lymphoma. The present invention provides a humanized, a chimeric and a human
anti-CD20 antibody and antibody fusion proteins thereof useful for treatment
of
9


CA 02752553 2011-09-13

mammalian subjects, humans and domestic animals, alone, as a conjugate or
administered in
combination with other therapeutic agents, including other naked antibodies
and antibody
therapeutic conjugates.
The anti-CD20 mAbs of the present invention contain specific murine CDRs or a
combination of murine CDRs from more than one murine or chimeric anti-CD20 MAb
that
have specificity for the CD20 antigen. The anti-CD20 mAbs of the present
invention are
humanized, chimeric or human mAbs and they contain the amino acids of the CDRs
of a
murine anti-CD20 MAb and retain substantially the B-cell and B-cell lymphoma
and
leukemia cell targeting of the murine anti-CD20 MAb. The CDRs of the light
chain variable
region of the anti-CD20 MAb comprises CDRI comprising amino acids RASSSVSYIH
(SEQ ID NO: 23), RASSSLSFMH (SEQ ID NO: 24) or RASSSVSYMH (SEQ ID NO: 25);
CDR2 comprising amino acids ATSNLAS (SEQ ID NO: 26); and CDR3 comprising amino
acids QQWTSNPPT (SEQ ID NO: 27), HQWSSNPLT (SEQ ID NO: 28) or QQSFSNPPT
(SEQ ID NO: 29); and the CDRs of the heavy chain variable region of the anti-
CD20 MAb
comprises CDRI comprising amino acids SYNMH (SEQ ID NO: 30); CDR2 comprising
amino acids AIYPGNGDTSYNQKFKG (SEQ ID NO: 31) and CDR3 comprising amino
acids STYYGGDWYFDV (SEQ ID NO: 32), STYYGGDWYFNV (SEQ ID NO: 33),
SHYGSNYVDYFDV (SEQ ID NO: 34) or VVYYSNSYWYFDV (SEQ ID NO: 35).
In one embodiment, the humanized and chimeric MAb or fragment thereof does not
contain the CDR3 of the heavy chain variable region comprising STYYGGDWYFNV
(SEQ
ID NO: 33). More preferably, CDRI of the light chain variable region does not
comprise
RASSSLSFMH (SEQ ID NO: 24) when the CDR3 of the light chain variable region
comprises HQWSSNPLT (SEQ ID NO: 28) and the CDR3 of the heavy chain variable
region comprises SHYGSNYVDYFDV (SEQ ID NO: 34). In another embodiment, the
CDR3 of the light chain variable region does not comprise HQWSSNPLT (SEQ ID
NO: 28)
when CDRI of the light chain variable region comprises RASSSLSFMH (SEQ ID NO:
24)
and when CDR3 of the heavy chain variable region comprises SHYGSNYVDYFDV (SEQ
ID NO: 34). In a further embodiment, the CDR3 of the heavy chain variable
region does not
comprise SHYGSNYVDYFDV (SEQ ID NO: 34) when the CDRI of the light chain
variable
region comprises RASSSLSFMH (SEQ ID NO: 24), and the CDR3 of the light chain
variable
region comprises HQWSSNPLT (SEQ ID NO: 28). In another embodiment, the CDR1
of the light chain variable region does not comprise RASSSVSYMH (SEQ ID NO:
25) when
the CDR3 of the light chain variable region comprises QQSFSNPPT (SEQ ID NO:
29) and
the CDR3 of the heavy chain variable region comprises VVYYSNSYWYFDV (SEQ ID
NO:
35).
Further, in another embodiment, the anti-CD20 monoclonal antibody (MAb) or
fragment thereof does not contain CDR3 of the light chain variable region of
amino


CA 02752553 2011-09-13

acids QQSFSNPPT (SEQ ID NO: 29) when CDRI of the light chain variable region
comprises RASSSVSYMH (SEQ ID NO: 25) and the CDR3 of the heavy chain variable
region comprises VVYYSNSYWYFDV (SEQ ID NO: 35). Additionally, the anti-CD20
MAb does not contain CDR3 of the heavy chain variable region with amino acids
VVYYSNSYWYFDV (SEQ ID NO: 35) when the CDR1 of the light chain variable
region comprises RASSSVSYMH (SEQ ID NO: 25) and the CDR3 of the light chain
variable region comprises QQSFSNPPT (SEQ ID NO: 29).
In a preferred embodiment, the humanized anti-CD20 (hCD20) monoclonal
antibody or antigen-binding fragment thereof comprising the complementarity
determining regions (CDRs) of at least one murine anti-CD20 MAb variable
region and
the framework regions (FRs) of at least one human MAb variable region, wherein
said
humanized anti-CD20 MAb or fragment thereof retains substantially the B-cell
and B-
cell lymphoma and leukemia cell targeting of said murine anti-CD20 MAb. The
humanized antibody's variable region may comprise a light chain variable
region, a
heavy chain variable region or a both light and heavy chain variable regions.
The
humanized antibody or fragment thereof may further comprise light and heavy
chain
constant regions of at least one human antibody.
The humanized anti-CD20 MAb or fragment thereof of the present invention
comprises the CDRs of a murine anti-CD20 MAb and the framework (FR) regions of
the
light and heavy chain variable regions of a human antibody, while retaining
substantially
the B-cell, and B-cell lymphoma and leukemia cell targeting of the parent
murine anti-
CD20 MAb, and wherein the CDRs of the light chain variable region of the
murine anti-
CD20 MAb comprises CDRI comprising amino acids RASSSVSYIH (SEQ ID NO: 23),
CDR2 comprising amino acids ATSNLAS (SEQ ID NO: 26) and CDR3 comprising
QQWTSNPPT (SEQ ID NO: 27) and the CDRs of the heavy chain variable region of
murine anti-CD20 MAb comprises CDRI comprising amino acids SYNMH (SEQ ID
NO: 30), CDR2 comprising amino acids AIYPGNGDTSYN QKFKG (SEQ ID NO: 31)
and CDR3 comprising amino acids STYYGGDWYFDV (SEQ ID NO: 32). But the
humanized anti-CD20 MAb or fragment thereof may further contain in the FRs of
the
light and heavy chain variable regions of the antibody at least one amino acid
from the
corresponding FRs of the murine MAb. The humanized MAbs may further contain
the
light and heavy chain constant regions of a human antibody. Specifically, the
humanized
anti-CD20 MAb or fragment thereof contains at least one amino acid residue 1,
5, 27, 30,
38, 48, 67, 68, 70, 95, 115 and 116 of the murine heavy chain variable region
of Fig. 4A,
designated as hA20VH1 or hA20VH2 and of at least one amino acid
11


CA 02752553 2011-09-13

residue 4, 21, 35, 38, 45, 46, 59, 99, 104 and 106 of the murine light chain
variable
region Fig. 4B, designated hA20Vk. One or more of the murine amino acid
sequences
can be maintained in the human FR regions of the light and heavy variable
chains if
necessary to maintain proper binding or to enhance binding to the CD20
antigen. More
preferably the humanized anti-CD20 MAb or fragment thereof of the present
invention
comprises the hA20Vk of Figure 4B and the hA2VHI of Figure 4A. Most
preferably,
the humanized anti-CD20 MAb or fragment thereof of the present invention
comprises
the hA20Vk of Figure 4B and the hA2VH2 of Figure 4A. This latter sequence
contains
more human amino acid sequences in the FRs of the VH2 chain than the VHI, and
thus
is more humanized.
The preferred chimeric anti-CD20 (cCD20) MAb or fragment thereof of the
present invention comprises the CDRs of a murine anti-CD20 MAb and the FR
regions
of the light and heavy chain variable regions of the murine anti-CD 20 MAb,
i.e., the Fvs
of the parental murine MAb, and the light and heavy chain constant regions of
a human
antibody, wherein the chimeric anti-CD20 MAb or fragment thereof retains
substantially
the B-cell, and B-cell lymphoma and leukemia cell targeting of the murine anti-
CD20
MAb, wherein the CDRs of the light chain variable region of the chimeric anti-
CD20
MAb comprise CDRI comprising amino acids RASSSVSYIH (SEQ ID NO: 23),
RASSSLSFMH (SEQ ID NO: 24) or RASSSVSYMH (SEQ ID NO: 25); CDR2
comprising amino acids ATSNLAS (SEQ ID NO: 26); and CDR3 comprising amino
acids QQWTSNPPT (SEQ ID NO: 27), HQWSSNPLT (SEQ ID NO: 28) or
QQSFSNPPT (SEQ ID NO: 29); and the CDRs of the heavy chain variable region of
the
chimeric anti-CD20 MAb comprise CDRI comprising amino acids SYNMH (SEQ ID
NO: 30); CDR2 comprising amino acids AIYPGNGDTSYNQKFKG (SEQ ID NO: 31)
and CDR3 comprising STYYGGDWYFDV (SEQ ID NO: 32), STYYGGDWYFNV
(SEQ ID NO: 33), SHYGSNYVDYFDV (SEQ ID NO: 34) or VVYYSNSYWYFDV
(SEQ ID NO: 35) with the following provisos,
(a) wherein the CDR3 of the heavy chain variable region does not comprise
STYYGGDWYFNV (SEQ ID NO: 33), when the CDRI of the light chain variable
region comprises amino acids RASSSVSYIH (SEQ ID NO: 23), CDR2 of the light
chain
variable region comprises amino acids ATSNLAS (SEQ ID NO: 26), CDR3 of the
light
chain variable region comprises amino acids QQWTSNPPT (SEQ ID NO: 27), CDRI of
the heavy chain variable region comprises amino acids SYNMH (SEQ ID NO: 30),
and
CDR2 of the light chain variable region comprises amino acids
AIYPGNGDTSYNQKFKG (SEQ ID NO: 35);

12


CA 02752553 2011-09-13

(b) wherein the CDR3 of the heavy chain variable region does not comprise
SHYGSNYVDYFDV (SEQ ID NO: 34), when the CDRI of the light chain variable
region comprises amino acids RASSSLSFMH (SEQ ID NO: 24), CDR2 of the light
chain variable region comprises amino acids ATSNLAS (SEQ ID NO: 26), CDR3 of
the
light chain variable region comprises amino acids HQWSSNPLT (SEQ ID NO: 28),
CDR] of the heavy chain variable region comprises amino acids SYNMH (SEQ ID
NO:
30), and CDR2 of the light chain variable region comprises amino acids
AIYPGNGDTSYNQKFKG(SEQ ID NO: 31) ; and
(c) wherein the CDR3 of the heavy chain variable region does not comprise
VVYYSNSYWYFDV (SEQ ID NO: 35), when the CDRI of the light chain variable
region comprises amino acids RASSSVSYMH (SEQ ID NO: 25), CDR2 of the light
chain variable region comprises amino acids ATSNLAS (SEQ ID NO: 26), CDR3 of
the
light chain variable region comprises amino acids QQSFSNPPT (SEQ ID NO: 29),
CDRI of the heavy chain variable region comprises amino acids SYNMH (SEQ ID
NO:
30), and CDR2 of the light chain variable region comprises amino acids
AIYPGNGDTSYNQKFKG (SEQ ID NO: 31).
More preferably the chimeric anti-CD20 MAb or fragment thereof comprising
the complementarity-determining regions (CDRs) of a murine anti-CD20 MAb and
the
framework (FR) regions of the light and heavy chain variable regions of the
murine anti-
CD20 MAb and the light and heavy chain constant regions of a human antibody,
wherein
the chimeric anti-CD20 MAb or fragment thereof retains substantially the B-
cell, and B-
cell lymphoma and leukemia cell targeting of the murine anti-CD20 MAb, wherein
the
CDRs of the light chain variable region of the chimeric anti-CD20 MAb
comprises the
CDRs shown in Figs. 4B and 4A, respectively, designated cA20Vk and cA20VH.
Most
preferably, the chimeric anti-CD20 MAb or fragment thereof comprises the light
and
heavy chain variable regions of murine anti-CD20 MAb shown in Figs. 4B and 4A,
respectively, designated cA20Vk and cA20 VH.
The present invention also encompasses a human anti-CD20 MAb or fragment
thereof comprising the light and heavy chain variable and constant regions of
a human
antibody, wherein said human CD20 MAb retains substantially the B-cell, and B-
cell
lymphoma and leukemia cell targeting and cell binding characteristics of a
murine anti-
CD20 MAb, wherein the CDRs of the light chain variable region of the human
anti-
CD20 MAb comprises the same CDRs as set forth above for the chimeric and
humanized anti-CD20 mAbs and as shown in Figs. 4A and 4B.

13


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
The present invention is also intended to encompass antibody fusion proteins
or
fragments thereof comprising at- least two anti-CD20 mAbs or fragments
thereof, as
described above. The antibody fusion protein or fragment thereof of the
present
invention is also intended to encompass an antibody fusion protein or fragment
thereof
comprising at least one first anti-CD20 MAb or fragment thereof as described
above and
at least one second MAb or fragment thereof, other than the antiCD20 IviAb or
fragment
described above. More preferably this second MAb is a MAb reactive with CD4,
CD5,
CD8, CD14, CD15, CD19, CD21, CD22, CD23, CD25, CD33, CD37, CD38, CD40,
CD40L, CD46, CD52, CD54, CD74, CD80, CD126, B7, MUC1, MUC2, MUC3,
MUC4, Ia, HM1.24, HLA-DR, tenascin, VEGF, P1GF, an oncogene, oncogene
product, or a combination thereof, and even an anti-CD20 MAb that is different
than
the anti-CD20 MAb described herein. The antibody fusion proteins of the
present
invention may be composed of one CD20 MAb and one or more of the second mAbs
to provide specificity to different antigens, and are described in more detail
below.
The humanized, chimeric and 'human anti-CD20 antibody may possess
enhanced affinity binding with the epitope, as well as antitumor and anti-B-
cell
activity, as a result of CDR mutation and manipulation of the CDR and other
sequences in the variable region to obtain a superior therapeutic agent for
the
treatment of B-cell disorders, including B-cell lymphomas and leukemias and
autoimmune diseases. Modification to the. binding specificity, affinity or
avidity of an,
antibody is known and described in WO 98/44001, as affinity maturation, and
this
application summarizes methods of modification.

It may also be desirable to modify the antibodies of the present invention to
improve effector function, e.g., so as to enhance antigen-dependent cell-
mediated
cytotoxicity (ADCC) and/or complement dependent cytotoxicity (CDC) of the
antagonist. One or more amino acid substitutions or the introduction of
cysteine in
the Fc region may be made, thereby improving internalization capability and/or
increased complement-mediated cell killing and ADCC.' See Caron et al., J Ex.
Med. 176:1191-1195 (1991) and Shopes, B.J. Immunol. 148:2918-2022 (1992)
An antibody fusion protein may be
prepared that has dual Fc regions with both enhanced complement lysis and ADCC
capabilities.

14


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
The present invention is also directed to DNA sequences comprising a nucleic
acid encoding a MAb or fragment thereof selected from the group consisting
(a) an anti-CD20 MAb or fragment thereof as described herein,
(b) an antibody fusion protein or fragment thereof comprising at least two of
the anti-CD20 mAbs or fragments thereof,
(c) an antibody fusion protein or fragment thereof comprising at least one'
first MAb or fragment thereof comprising the anti-CD20 MAb or fragment thereof
as
described herein and at least one second MAb or fragment thereof, other than
the
antiCD20 MAb or fragment thereof, and
(d) an antibody fusion protein or fragment thereof comprising at least one
first MAb or fragment thereof comprising the anti-CD20 MAb or fragment thereof
and
at least one second MAb or fragment thereof, wherein the second MAb is a MAb
reactive with CD4, CD5, CD8, CD14, CD15, CD19, CD21, CD22, CD23, CD25,
CD33, CD37, CD38, CD40, CD40L, CD46, CD52, CD54, CD74, CD80, CD126, B7,
MUC1, MUC2, MUC3, MUC4, Ia, HMl ?4, HLA-DR, tenascin, VEGF, PIGF, an
oncogene, oncogene product, or a combination thereof.
Also encompassed by the present invention are expression vectors comprising
the DNA sequences. These vectors contain the light and heavy chain constant
regions
and the hinge region of the human immunoglobulin, in the case of vectors for
use in
preparing the humanized, chimeric and human anti-CD20 mAbs or antibody fusion
proteins thereof or fragments thereof. These vectors additionally contain,
where
required, promoters that express the mAbs in the selected host cell,
immunoglobulin
enhances and signal or leader sequences. Vectors that are .particularly useful
in'the
present invention are pdHL2 or GS, particularly when used to express. a
chimeric,
humanized or human antibodies, such as gigs, where the vector codes for the
heavy and
light chain constant regions and hinge region of IgGI. More preferably, the
light and
heavy chain constant regions and hinge region are from a human EU myeloma
immunoglobulin, where optionally at least one of the amino acid in the
allotype positions
is changed to that found in a different IgGl allotype, and wherein optionally
amino acid
253 of the heavy chain of EU based on the EU number system may be replaced
with
alanine. See Edelman et al., Proc. Natl. Acad. Sci USA 63: 78-85 (1969);



CA 02752553 2011-09-13

WO 03/068821 PCTIGB03/00665
Host cells containing the DNA sequences encoding the anti-CD20 mAbs or
fragments thereof or antibody fusion proteins or fragments thereof of the
present
invention or host cells containing the vectors that contain these DNA
sequences are
encompassed by the present invention. Particularly useful host cells are
mammalian
cells, more specifically lymphocytic cells, such as myeloma cells, discussed
in more
detail below.

Also encompassed by the present invention is the method of expressing the
antiCD20 MAb or fragment thereof or antibody fusion protein or fragment
thereof
comprising: (a) transfecting a mammalian cell with a DNA sequence of encoding
the
anti-CD20 mAbs or fragments thereof or antibody fusion proteins or fragments
thereof,
and (b) culturing the cell transfected with the DNA sequence that secretes the
anti-M." 0
or fragment thereof or antibody fusion protein or fragment thereof. Known
techniques
may be used that include a selection marker on the vector so that host cells
that express
the mAbs and the marker can be easily selected.
The present invention particularly encompasses B4ymphoma cell and leukemia
cell targeting diagnostic or therapeutic conjugates comprising an antibody
component
comprising an anti-CD20 MAb or fragment thereof or an antibody fusion protein
or
fragment thereof of the present invention that binds to the B-lymphoma or
leukemia cell,
that is bound to at least one diagnostic or at least one therapeutic agent.
The diagnostic conjugate comprises the antibody component comprising an anti-
CD20 MAb or fragment thereof or an antibody fusion protein or fragment thereof
wherein the diagnostic agent comprises at least one photoactive diagnostic
agent, and
more preferably wherein the label is a radioactive label with an energy
between 60 and
4,000 keV or a non-radioactive label. The radioactive label is preferably a
gamma-,
beta-, and positron-emitting isotope and is selected from the group consisting
of 1251,
1311, 1231 1241, 86Y, 186Re, 188Re, 62Cu, 64Cu, 1111n, 67G a' 6SGa, 99mTc,
94mTC, 18F, 11 C,
13N,110, 76Br and combinations thereof.

The diagnostic conjugate of the present invention also utilizes a diagnostic
agent,
such as a contrast agent, for example, such as manganese, iron or gadolinium.
The therapeutic conjugate of the present invention comprises an antibody
component comprising an antibody fusion protein or fragment thereof, wherein
each of
said mAbs or fragments thereof are bound to at least one therapeutic agent.
The
therapeutic conjugate of preferably is selected from the group consisting of a
radioactive
16


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
label, an inmlunomodulator, a hormone, a photoactive therapeutic agent, a
cytotoxic
agent, which may be a drug or a toxin, and a combination thereof. The drugs
useful in
the present invention are those drugs that possess the pharmaceutical property
selected
from the group consisting of antimitotic, antikinase, alkylating,
antimetabolite, antibiotic,
alkaloid, antiangiogenic, apoptotic agents and combinations thereof More
specifically,
these drugs are selected from the group consisting of nitrogen mustards,
ethylenimine
derivatives, alkyl sulfonates, nitrosoureas, triazenes, folic acid analogs,
COX-2
inhibitors, pyrimidine analogs, purine analogs, antibiotics, enzymes,
epipodophyllotoxins, platinum coordination complexes, vinca alkaloids,
substituted
ureas, methyl hydrazine derivatives, adrenocortical suppressants, antagonists,
endostatin, taxols, camptothecins, anthracyclines, taxanes, and their analogs,
and a
combination thereof. The toxins encompassed by the present invention are
selected
from the group consisting of ricin, abrin, alpha toxin, saporin, ribonuclease
(RNase),
e.g., onconase, DNase I, Staphylococcal enterotoxin-A, pokeweed antiviral
protein,
gelonin, diphtherin toxin, Pseudornonas exotoxin, and Pseudornonas endotoxin.
Useful therapeutic conjugates of the present invention are immunomodulators
selected from the group consisting of a cytokine, a stem cell growth factor, a
lymphotoxin, a hematopoietic factor, a colony stimulating factor (CSF), an
interferon
(IFN), erythropoietin, thrombopoietin and a combination thereof. Specifically
useful
are lymphotoxins such as tumor necrosis factor (INF), hematopoietic factors,
such as
interleukin (IL), colony stimulating factor, such as granulocyte-colony
stimulating
factor (G-CSF) or granulocyte macrophage-colony stimulating factor (GM-CSF)),
interferon, such as interferons-a, -(3 or -y, and stem cell growth factor,
such as
designated "SI factor". More specifically, immunomodulator, such as IL-1, IL-
2; IL-
3, IL-6, IL-10, IL-12, IL-18, IL-21 interferon-y, TNF-a or a combination
thereof are
useful in the present invention.
Particularly useful therapeutic conjugates comprise one or more radioactive
labels that have an energy between 60 and 700 keV. Such radioactive labels are
selected from the group consisting of 22 Ac, 67Ga, 90Y, , 111ce, 1311, 1251,
186Re, 18SRe,
177Lu, 32P, , 64Cu, 67Cu, 212Bi, 213Bi, 211At and combinations thereof Other
useful
therapeutic conjugates are photoactive therapeutic agent, such as a chromogen
or -dye.
17


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
Other useful therapeutic conjugates comprise oligonucleotides, especially
antisense oligonucleotides that preferably are directed against oncogenes and
oncogene products of B-cell malignancies, such as bcl-2.
The present invention particularly encompasses methods of treating a B-cell
lymphoma or leukemia cell disease or an autoimmune disease in a subject, such
as a
mammal, including humans, domestic or companion pets, such as dogs and cats,
comprising administering to the subject a therapeutically effective amount of
an anti-
CD20 MAb or a fragment thereof of the present invention, formulated in a
pharmaceutically acceptable vehicle. This therapy utilizes a "naked antibody"
that does
not have a therapeutic agent bound to it. The administration of the "naked
anti-CD20
antibody" can be supplemented by administering to the subject concurrently or
sequentially a therapeutically effective amount of another "naked antibody"
that binds to
or is reactive with another antigen on the surface of the target cell or that
has other
functions, such as effector functions in the Fc portion of the MAb, that is
therapeutic and
which is discussed herein. Preferred additional mAbs are at least one
humanized,
chimeric, human or murine (in the case of non-human animals) MAb selected from
the
group consisting of a MAb reactive with CD4, CD5, CD8, CD14, CD15, CD19, CD20,
CD21, CD22, CD23, CD25, CD33, CD37, CD38, CD40, CD40L, CD46, CD52,
CD54, CD74, CD80, CD126, B7, MUC1, Ia, HM1.24, and HLA-DR, tenascin,
VEGF, P1GF, an oncogene, oncogene product, or a combination thereof,
formulated in
a pharmaceutically acceptable vehicle.
Both the naked anti-CD20 therapy alone or in combination with other naked
mAbs as discussed above can be further supplemented with the administration,
either
concurrently or sequentially, of a therapeutically effective amount of at
least one
therapeutic agent, formulated in a pharmaceutically acceptable vehicle. As
discussed
herein the therapeutic agent may comprises a cytotoxic agent, a radioactive
label, an
immunomodulator, a hormone, an enzyme, an oligonucleotide, a photoactive
therapeutic
agent or a combination thereof, formulated in a pharmaceutically acceptable
vehicle.
In another therapeutic method, both the naked anti-CD20 therapy alone or in
combination with other naked mAbs, as discussed above, can be further
supplemented
with the administration, either concurrently or sequentially, of a
therapeutically effective
amount of at least one therapeutic conjugate, described herein and formulated
in a
pharmaceutically acceptable vehicle. The antibody component of the therapeutic
18


CA 02752553 2011-09-13

WO 031068821 PCT/GB03/00665
conjugate comprises at least one humanized, chimeric, human or murine (for non-

human subjects) MAb selected from the group consisting of a MAb reactive with
CD4,
CD5, CD8, CD14, CD15, CD19, CD20, CD21, CD22, CD23, CD25, CD33, CD37,
CD38, CD40, CD40L, CD46, CD52, CD54, CD74, CDSO, CD126, B7, MUC1,
MUC2, MUC3, MUC4, Ia, HMI.24, and HLA-DR, tenascin, VEGF, PIGF, an
oncogene, oncogene product, or a combination thereof, formulated in a
pharmaceutically acceptable vehicle. As discussed herein the therapeutic agent
may
comprise a cytotoxic agent, a radioactive label, an immunomodulator, a
hormone, a
photoactive therapeutic agent or a combination thereof, formulated in a
pharmaceutically
acceptable vehicle.
As described herein the present invention particularly encompasses a method of
treating a B-cell lymphoma or leukemia or an autoimmune disease in a subject
comprising administering to a subject a therapeutically effective amount of an
antibody
fusion protein or fragment thereof comprising at least two anti-CD20 mAbs or
fragments
thereof of the present invention or comprising at least one anti-CD20 MAb or
fragment
thereof of the present invention and at least one additional MAb, preferably
selected
from the group consisting of mAbs reactive with CD4, CD5, CD8, CD14, CD15,
CD19, CD20, CD21, CD22, CD23, CD25, CD33, CD37, CD38, CD40, CD40L,
CD46, CD52, CD54, CD74, CD80, CD126, B7, MUC1, MUC2, MUC3, MUC4, Ia,
HM1.24, and HLA-DR, tenascin, VEGF, P1GF, an oncogene, oncogene product, or a
combination thereof, formulated in a pharmaceutically acceptable vehicle.
This therapeutic method can further be supplemented with the administration to
the subject concurrently or sequentially of a therapeutically effective amount
of at least
one therapeutic agent, formulated in a pharmaceutically acceptable vehicle,
wherein the
therapeutic agent is preferably a cytotoxic agent, a radioactive label, an
immunomodulator, a hormone, a photoactive therapeutic agent or a combination
thereof,
formulated in a pharmaceutically acceptable vehicle.
Further, the antibody fusion proteins can be administered to a subject
concurrently or sequentially a therapeutically effective amount of a
therapeutic conjugate
comprising at least one MAb bound to at least one therapeutic agent, wherein
said MAb
component of the conjugate preferably comprises at least one humanized,
chimeric,
human or murine (for non-human subjects) MAb selected from the group
consisting of a
MAb reactive with CD4, CD5, CD8, CD14, CD15, CD19, CD20, CD21, CD22,
19


CA 02752553 2011-09-13

WO (13/068821 PCT/GB03/00665
CD23, CD25, CD33, CD37, CD38, CD40, CD40L, CD46, CD52, CD54, CD74,
CD80, CD126, B7, MUC1, MUC2, MUC3, MUC4, Ia, HM1.24, and HLA-DR,
tenascin, VEGF, P1GF, an oncogene, oncogene product, or a combination thereof,
formulated in a pharmaceutically acceptable vehicle. The antibody fusion
protein itself
can be conjugated to a therapeutic agent and thus provides a vehicle to attach
more
than one therapeutic agent to an antibody component and these therapeutic
agents can
be a combination of different recited agents or combinations of the same
agents, such
as two different therapeutic radioactive labels. Also encompassed by the
present
invention is a method of diagnosing a B-cell lymphoma or leukemia in a subject
comprising administering to the subject, such as a mammal, including humans
and
domestic and companion pets, such as dogs, cats, rabbits, guinea pigs, a
diagnostic
conjugate comprising an anti-CD20 MAb or fragment thereof or an antibody
fusion
protein or fragment thereof of the present invention that binds to the
lymphoma or
leukemia cell, wherein the anti-CD20 MAb or fragment thereof or antibody
fusion
protein or fragment thereof is bound to at least one diagnostic agent,
formulated in a
pharmaceutically acceptable vehicle. The useful diagnostic agents are
described herein.
2. Definitions
In the description that follows, a number of terms are used and the following
definitions are provided to facilitate understanding of the present invention.
An antibody, as described herein, refers to a full-length (i.e., naturally
occurring or formed by normal immunoglobulin gene fragment recombinatorial
processes) immunoglobulin molecule (e.g., an IgG antibody) or an
immunologically
active (i.e., specifically binding) portion of an immunoglobulin molecule,
like an
antibody fragment.
An antibody fragment is a portion of an antibody such as F(ab')2, F(ab)2,
Fab',
Fab, Fv, scFv and the like. Regardless of structure, an antibody fragment
binds with
the same antigen that is recognized by the intact antibody. For example, an
anti-
CD20 monoclonal antibody fragment binds with an epitope of CD20. The term
"antibody fragment" also includes any synthetic or genetically engineered
protein that
acts like an antibody by binding to a specific antigen to form a complex. For
example, antibody fragments include isolated fragments consisting of the
variable
regions, such as the "Fv" fragments consisting of the variable regions of the
heavy
and light chains, recombinant single chain polypeptide molecules in which
light and


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
heavy variable regions are connected by a peptide linker ("scFv proteins"),
and
minimal recognition units consisting of the amino acid residues that mimic the
hypervariable region.
A naked antibody is generally an entire antibody which is not.conjugated to a
therapeutic agent. This is so because the Fc portion of the antibody molecule
provides effector functions, such as complement fixation and ADCC (antibody
dependent cell cytotoxicity), which set mechanisms into action that may result
in cell
lysis. However, it is possible that the Fc portion is not required for
therapeutic
function , with other mechanisms, such as apoptosis, coming into play. Naked
antibodies include both polyclonal and monoclonal antibodies, as well as
certain
recombinant antibodies, such as chimeric, humanized or human antibodies.
A chimeric antibody is a recombinant protein that contains the variable
domains including the complementarity determining regions (CDRs) of an
antibody
derived from one species, preferably a rodent antibody, while the constant
domains of
the antibody molecule is derived from those of a human antibody. For
veterinary
applications, the constant domains of the chimeric antibody may be derived
from that
of other species, such as a cat or dog.
A humanized antibody is a recombinant protein in which the CDRs from an
antibody from one species; e.g., a rodent antibody, is transferred from the
heavy and
light variable chains of the rodent antibody into human heavy and light
variable
domains. The constant domains of the antibody molecule is derived from those
of a
human antibody.

A human antibody is an antibody obtained from transgenic mice that have
been "engineered" to produce specific human antibodies in response to
antigenic
challenge. In this technique, elements of the human heavy and light chain
locus are
introduced into strains of mice derived from embryonic stem cell lines that
contain
targeted disruptions of the endogenous heavy chain and light chain loci. The
transgenic mice can synthesize human antibodies specific for human antigens,
and the
mice can be used to produce human antibody-secreting hybridomas. Methods for
obtaining human antibodies from transgenic mice are described by Green et al.,
Nature Genet. 7:13 (1994), Lonberg et al., Nature 368:856 (1994), and Taylor
et al.,
Int. Immun. 6:579 (1994). A fully human antibody also can be constructed by
genetic
or chromosomal transfection methods, as well as phage display technology, all
of

21


CA 02752553 2011-09-13

WO 03/068821 PCTIGB03/00665
which are known in the art. See for example, McCafferty et al., Nature 348:552-
553
(1990) for the production of human antibodies and fragments thereof in vitro,
from
immunoglobulin variable domain gene repertoires from uni munized donors.. In
this
technique, antibody variable domain genes are cloned in-frame into either a
major or
minor coat protein gene of a filamentous bacteriophage, and displayed as
functional
-antibody fragments on the surface of the phage particle. Because the
filamentous
particle contains a single-stranded DNA copy of the phage genome, selections
based
on the functional properties of the antibody also result in selection of the
gene
encoding the antibody exhibiting those properties. In this way, the phage
mimics
some of the properties of the B cell. Phage display can be performed in a
Variety of
formats, for their review, see e.g. Johnson and Chiswell, Current Opinion in
Structural Biology 3:5564-571 (1993).
Human antibodies may also be generated by in vitro activated B. cells. See
U.S. Patent Nos. 5,567,610 and 5,229,275.

A therapeutic agent is a molecule or atom which is administered separately,
concurrently or sequentially with an antibody moiety or conjugated to an
antibody
moiety, i.e., antibody or antibody fragment, or a subfragment, and is useful
in the
treatment of a disease. Examples of therapeutic agents include antibodies,
antibody
fragments, drugs, toxins, nucleases, hormones, immunomodulators, chelators,
boron
compounds, photoactive agents or dyes and radioisotopes.
A diagnostic agent is a molecule or atom which is administered conjugated to
an antibody moiety, i.e., antibody or antibody fragment, or subfragment, and
is useful
in diagnosing a disease by locating the cells containing the antigen. Useful
diagnostic
agents include, but are not limited to, radioisotopes, dyes (such as with the
biotin-
streptavidin complex), contrast agents, fluorescent compounds or molecules and
enhancing agents (e.g. paramagnetic ions) for magnetic resonance imaging
(MR1).
U.S. Patent No. 6,331,175 describes MRI technique and the preparation of
antibodies
conjugated to a MRI enhancing agents

Preferably, the diagnostic agents are selected from the group consisting of
radioisotopes, enhancing agents for use in magnetic resonance imaging, and
fluorescent compounds. In order to load an antibody component with radioactive
metals or paramagnetic ions, it may be necessary to react it with a reagent
having a
22


CA 02752553 2011-09-13

WO 031168821 PCT/GB03/00665
long tail to which are attached a multiplicity of chelating groups for binding
the ions.
Such a tail can be a polymer such as a polylysine, polysaccharide, or other
derivatized
or derivatizable chain having pendant groups to which can be bound chelating
groups
such as, e.g., ethylenediaminetetraacetic acid (EDTA),
diethylenetriaminepentaacetic
acid (DTPA), porphyrins, polyamines, crown ethers, bis-thiosenricarbazones,
polyoximes, and like groups known to be useful for this purpose. Chelates are
coupled
to the peptide antigens using standard chemistries. The chelate is normally
linked to
the antibody by a group which enables formation of a bond to the molecule with
minimal loss of immunoreactivity and minimal aggregation and/or internal
cross=
linking. other, more unusual, methods and reagents for conjugating chelates to
antibodies are disclosed in U.S. Patent 4,824,659 to Hawthorne, entitled
"Antibody
Conjugates", issued April 25, 1989.
Particularly useful metal-chelate combinations include 2-
benzyl-DTPA and its monomethyl and cyclohexyl analogs, used with diagnostic
isotopes in the general energy range of 60 to 4,4000 keV, such as J2sI~ 131!
1231 1241
~ ,
62Cu, 64Cu,'8F, r 111n, 67Ga, 68Ga, 99mTc, 94mTc, ti "C, r3N, rs0, 76Br , for
radio-imaging.
The same chelates, when complexed with non-radioactive metals, such as
manganese,
iron and gadolinium are useful for MRI, when used along with the antibodies of
the
invention. Macrocyclic chelates such as NOTA, DOTA, and TETA are of use with a
variety of metals and radiometals, most particularly with radionuclides of
gallium,
yttrium and copper, respectively. Such metal-chelate complexes can be made
very
stable by tailoring the ring size to the metal of interest. Other ring-type
chelates such
as macrocyclic polyethers, which are of interest for stably binding nuclides,
such as
223Rd for RAIT are encompassed by the invention.
An immunoconiugate is a conjugate of an antibody component with a
therapeutic or diagnostic agent. The diagnostic agent can comprise a
radioactive or
non-radioactive label, a contrast agent (such as for magnetic resonance
imaging,
computed tomography or ultrasound), and the radioactive label can be a gamma-,
beta-, alpha-, Auger electron-, or positron-emitting isotope.
An expression vector is a DNA molecules comprising a gene that is expressed
in a host cell. Typically, gene expression is placed under the control of
certain
regulatory elements, including constitutive or inducible promoters, tissue-
specific

23


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
regulatory elements and enhancers. Such a gene is said to be "operably linked
to" the
regulatory elements.
A recombinant host may be any prokaryotic or eukaryotic cell that contains
either a cloning vector or expression vector. This term also includes those
prokaryotic
or eukaryotic cells, as well as an transgenic animal, that have been
genetically
engineered to contain the cloned gene(s) in the chromosome or genome of the
host
cell or cells of the host cells. Suitable mammalian host cells include myeloma
cells,
such as SP2/0 cells, and NSO cells, as well as Chinese Hamster Ovary (CHO)
cells,
hybridoma cell lines and other mammalian host cell useful for expressing
antibodies.
Also particularly useful to express mAbs and other fusion proteins, is a human
cell
line, PER.C6 disclosed in WO 0063403 A2, which produces 2 to 200-fold more
recombinant protein as compared to conventional mammalian cell lines, such as
CHO, COS, Vero, Hela, BHK and SP2- cell lines. Special transgenic animals with
a
modified immune system are particularly useful for making fully human
antibodies.
As used herein, the term antibody fusion protein is a recombinantly produced
antigen-binding molecule in which two or more of the same or different single-
chain
antibody or antibody fragment segments with the same or different
specificities are
linked. Valency of the fusion protein indicates how many binding arms or sites
the
fusion protein has to a single antigen or epitope; i.e., monovalent, bivalent,
trivalent or
mutlivalent. The multivalency of the antibody fusion protein means that it can
take
advantage of multiple interactions in binding to an antigen, thus increasing
the avidity
of binding to the antigen. Specificity indicates how many antigens or epitopes
an
antibody fusion protein is able to bind; i.e., monospecific, bispecific,
trispecific,
multispecific. Using these definitions, a natural antibody, e.g., an IgG, is
bivalent
because it has two binding arms but is monospecific because it binds to one
epitope.
Monospecific, multivalent fusion proteins have more than one binding site for
an
epitope but only binds with one epitope, for example a diabody with two
binding site
reactive with the same antigen. The fusion protein may comprise a single
antibody
component, a multivalent or multispecific combination of different antibody
components or multiple copies of the same antibody component. The fusion
protein
may additionally comprise an antibody or an antibody fragment and a
therapeutic
agent. Examples of therapeutic agents suitable for such fusion proteins
include
immunomodulators ("antibody-immunomodulator fusion protein") and toxins
24


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
("antibody-toxin fusion protein"). One preferred toxin comprises a
ribonuclease
(RNase), preferably a recombinant RNase.
A multispecific antibody is an antibody that can bind simultaneously to at
least
two targets that are of different structure, e.g., two different antigens, two
different
epitopes on the same antigen, or a hapten and/or an antigen or epitope. One
specificity would be for a B-cell, T-cell, myeloid-, plasma-, and mast-cell
antigen or
epitope. Another specificity could be to a different antigen on the same cell
type,
such as CD20, CD19, CD21, CD23, CD46, CD80, HLA-DR, CD74, MUC1, and
CD22 on B-cells. Multispecific, multivalent antibodies are constructs that
have more
than one binding site, and the binding sites are of different specificity. For
example, a
diabody, where one binding site reacts with one antigen and the other with the
other
antigen.
A bispecific antibody is an antibody that can bind simultaneously to two
targets which are of different structure. Bispecific antibodies (bsAb) and
bispecific
antibody fragments (bsFab) have at least one arm that specifically binds to,
for
example, a B-cell, T-cell, myeloid-, plasma-, and mast-cell antigen or epitope
and at
least one other arm that specifically binds to atargetable conjugate that
bears a
therapeutic or diagnostic agent. A variety of bispecific fusion proteins can
be
produced using molecular engineering. In one form, the bispecific fusion
protein is
monovalent, consisting of, for example, a scFv with a single binding site for
one
antigen and a Fab fragment with a single binding site for a second antigen. In
another
form, the bispecific fusion protein is divalent, consisting of, for example,
an IgG with
a binding site for one antigen and two scFv with two binding sites for a
second
antigen.
Caninized or felinized antibodies are recombinant proteins in which rodent (or
another species) complementarity determining regions of a monoclonal antibody
have
been transferred from heavy and light variable chains of rodent (or another
species)
immunoglobulin into a dog or cat, respectively, immunoglobulin variable
domain.
Domestic animals include large animals such as horses, cattle, sheep, goats,
llamas, alpacas, and pigs, as well as companion animals. In a preferred
embodiment,
the domestic animal is a horse.

Companion animals include animals kept as pets. These are primarily dogs
and cats, although small rodents, such as guinea pigs, hamsters, rats, and
ferrets, are


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
also included, as are subhuman primates such as monkeys. In a preferred
embodiment the companion animal is a dog or a cat.

3. Preparation of Monoclonal Antibodies including Chimeric, Humanized
and Human Antibodies
Monoclonal antibodies (MAbs) are a homogeneous population of antibodies to
a particular antigen and the antibody comprises only one type of antigen
binding site
and binds to only one epitope on an antigenic determinant. Rodent monoclonal
antibodies to specific antigens may be obtained by methods known to those
skilled in
the art. See, for example, Kohler and Milstein, Nature 256: 495 (1975), and
Coligan
et al. (eds.), CURRENT PROTOCOLS IN IMMUNOLOGY, VOL. 1, pages 2.5.1-
2.6.7 (John Wiley & Sons 1991) [hereinafter "Coligan"]. Briefly, monoclonal
antibodies can be obtained by injecting mice with a composition comprising an
antigen, verifying the presence of antibody production by removing a serum
sample,
removing the spleen to obtain B-lymphocytes, fusing the B-lymphocytes with
myeloma cells to produce hybridomas, cloning the hybridomas, selecting
positive
clones which produce antibodies to the antigen, culturing the clones that
produce
antibodies to the antigen, and isolating the antibodies from the hybridoma
cultures.
MAbs can be isolated and purified from hybridoma cultures by a variety of
well-established techniques. Such isolation techniques include affinity
chromatography with Protein-A Sepharose, size-exclusion chromatography, and
ion-
exchange chromatography. See, for example, Coligan at pages 2.7.1-2.7.12 and
pages
2.9.1-2.9.3. Also, see Baines et al., "Purification of Immunoglobulin G
(IgG)," in
METHODS IN MOLECULAR BIOLOGY, VOL. 10, pages 79-104 (The Humana
Press, Inc. 1992).
After the initial raising of antibodies to the immunogen, the antibodies can
be
sequenced and subsequently prepared by recombinant techniques. Humanization
and
chimerization of murine antibodies and antibody fragments are well known to
those
skilled in the art. For example, humanized monoclonal antibodies are produced
by
transferring mouse complementary determining regions from heavy and light
variable
chains of the mouse immunoglobulin into a human variable domain, and then,
substituting human residues in the framework regions of the murine
counterparts. The

26


CA 02752553 2011-09-13

WO 03/068821 PCT/G B03/00665
use of antibody components derived from humanized monoclonal antibodies
obviates
potential problems associated with the immunogenicity of murine constant
regions.
General techniques for cloning murine immunoglobulin variable domains are
described, for example, by the publication of Orlandi et aL, Proc. Nat'! Acad.
Sci.
USA 86: 3833 (1989). Techniques
for constructing chimeric antibodies are well known to those of skill in the-
art. As an
example, Leung et al., Hybridoma 13:469 (1994), describe how they produced an
LL2 chimera by combining DNA sequences encoding the V. and VH domains of LL2
monoclonal antibody, an anti-CD22 antibody, with respective human x and IgGi.
constant region domains. This publication also provides the nucleotide
sequences of
the LL2 light and heavy chain variable regions, V,; and VH, respectively.
Techniques
for producing humanized MAbs are described, for example, by Jones et al.,
Nature
321: 522 (1986), Riechmann et al., Nature 332: 323 (1988), Verhoeyen et al.,
Science
239:153 4 (1988), Carter et al., Proc. Nat'l Acad. Sci. USA 89: 4285 (1992),
Sandhu,
Crit. Rev. Biotech. 12: 437 (1992), and Singer et al., J. Iinmun. 150:2844
(1993).-

A chimeric antibody is a recombinant protein that contains the variable
domains -including the CDRs derived from one species of animal, such as a
rodent
antibody, while the remainder of the antibody molecule; i.e., the constant
domains, is
derived from a human antibody. Accordingly, a chimeric monoclonal antibody can
also be humanized by replacing the sequences of the murine FR in the variable
domains
of the chimeric MAb with one or more different human FR. Specifically, mouse
CDRs
are transferred from heavy and light variable chains of the mouse
immunoglobulin
into the corresponding variable domains of a human antibody. As simply
transferring
mouse CDRs into human FRs often results in a reduction or even loss of
antibody
affinity, additional modification might be required in order to restore the
original affinity
of the marine antibody. This can be accomplished by the replacement of one or
more
some human residues in the FR regions with their murine counterparts to obtain
an
antibody that possesses good binding affinity to its epitope. See, for
example, Tempest
et al., Biotechnology 9:266 (1991) and Verhoeyen et al., Science 239: 1534
(1988).
Further, the affinity of humanized, chimeric and human MAbs to a specific
epitope can
be increased by mutagenesis of the CDRs, so that a lower dose of antibody may
be as

27


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
effective as a higher dose of a lower affinity MAb prior to mutagenesis. See
for
example, W00029584A1
Another method for producing the antibodies of the present invention is by
production in the milk of transgenic livestock. See, e.g., Colman, A.,
Biochem. Soc.
Symp., 63: 141-147;1998; U.S. Patent 5,827,690.
Two DNA constructs are prepared which contain,
respectively, DNA segments encoding paired immunoglobulin heavy and light
chains.
The DNA segments are cloned into expression vectors which contain a promoter
sequence that is preferentially expressed in mammary epithelial cells.
Examples
include, but are not limited to, promoters from rabbit, cow and sheep casein
genes, the
cow a-lactoglobulin gene, the sheep P-lactoglobulin gene and the mouse whey
acid
protein gene. Preferably, the inserted fragment is flanked on its 3' side by
cognate
genomic sequences from a mammary-specific gene. This provides a
polyadenylation
site and transcript-stabilizing sequences. The expression cassettes are co-
injected into
the pronuclei of fertilized, mammalian eggs, which are then implanted into the
uterus
of a recipient female and allowed to gestate. After birth, the progeny are
screened for
the presence of both transgenes by Southern analysis. In order for the
antibody to be
present, both heavy and light chain genes must be expressed concurrently in
the same
cell. Milk from transgenic females is analyzed for the presence and
functionality of
the antibody or antibody fragment using standard immunological methods known
in
the art. The antibody can be purified from the milk using standard methods
known in
the art.
A fully human antibody of the present invention, i.e., human anti-CD20 MAbs
or other human antibodies, such as anti-CD22, anti-CD 19, anti-CD23, or anti-
CD21
MAbs for combination therapy with. humanized, chimeric or human anti-CD20,
antibodies, can be obtained from a transgenic non-human animal. See, e.g.,
Mendez
et al., Nature Genetics, 15: 146-156 (1997); U.S. Patent No. 5,633,425,
For example, a human antibody can be
recovered from a transgenic mouse possessing human immunoglobulin loci. The
mouse humoral immune system is humanized by inactivating the endogenous
immunoglobulin genes and introducing human immunoglobulin loci. The human
immunoglobulin loci are exceedingly complex and comprise a large number of
discrete segments which together occupy almost 0.2% of the human genome. To
28


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
ensure that transgenic mice are capable of producing adequate repertoires of
antibodies, large portions of human heavy- and light-chain loci must be
introduced
into the mouse genome. This is accomplished in a stepwise process beginning
with
the formation of yeast artificial chromosomes (YACs) containing either human
heavy-
or light-chain immunoglobulin loci in germline configuration. Since each
insert is
approximately 1 Mb in size, YAC construction requires homologous recombination
of
overlapping fragments of the immunoglobulin loci. The two YACs, one containing
the heavy-chain loci and one containing the light-chain loci, are introduced
separately
into mice via fusion of YAC-containing yeast spheroblasts with mouse embryonic
stem cells. Embryonic stem cell clones are then microinjected into mouse
blastocysts.
Resulting chimeric males are screened for their ability to transmit the YAC
through
their germline and are bred with mice deficient in murine antibody production.
Breeding the two transgenic strains, one containing the human heavy-chain loci
and
the other containing the human light-chain loci, creates progeny which produce
human antibodies in response to immunization.
Further recent methods for producing bispecific mAbs include engineered
recombinant mAbs which have additional cysteine residues so that they
crosslink more
strongly than the more common immunoglobulin isotypes. See, e.g., FitzGerald
et al.,
Protein Eng. 10(10):1221-1225, 1997. Another approach is to engineer
recombinant
fusion proteins linking two or more different single-chain antibody or
antibody fragment
segments with the needed dual specificities. See, e.g., Coloma et al., Nature
Biotech.
15:159-163, 1997. A variety of bispecific fusion proteins can be produced
using
molecular engineering. In one form, the bispecific fusion protein is
monovalent,
consisting of, for example, a scFv with a single binding site for one antigen
and a Fab
fragment with a single binding site for a second antigen. In another form, the
bispecific fusion protein is divalent, consisting of, for example, an IgG with
two
binding sites for one antigen and two scFv with two binding sites for a second
antigen.
Bispecific fusion proteins linking two or more different single-chain
antibodies
or antibody fragments are produced in similar manner. Recombinant methods can
be
used to produce a variety of fusion proteins. For example a fusion protein
comprising a Fab fragment derived from a humanized monoclonal anti-CD20
antibody and a scFv derived from a murine anti-diDTPA can be produced. A
flexible
29


CA 02752553 2011-09-13

linker, such as GGGS (SEQ ID NO: 36) connects the scFv to the constant region
of the
heavy chain of the anti-CD20 antibody. Alternatively, the scFv can be
connected to the
constant region of the light chain of another humanized antibody. Appropriate
linker
sequences necessary for the in-frame connection of the heavy chain Fd to the
scFv are
introduced into the VL and VK domains through PCR reactions. The DNA fragment
encoding the scFv is then ligated into a staging vector containing a DNA
sequence
encoding the CHI domain. The resulting scFv-CHI construct is excised and
ligated into
a vector containing a DNA sequence encoding the VH region of an anti-CD20
antibody.
The resulting vector can be used to transfect an appropriate host cell, such
as a
mammalian cell for the expression of the bispecific fusion protein.

4. Production of Antibody Fragments
Antibody fragments which recognize specific epitopes can be generated by
known techniques. The antibody fragments are antigen binding portions of an
antibody,
such as F(ab')2, Fab', Fab, Fv, sFv and the like. Other antibody fragments
include, but are
not limited to: the F(ab)'2 fragments which can be produced by pepsin
digestion of the
antibody molecule and the Fab' fragments, which can be generated by reducing
disulfide
bridges of the F(ab)'2 fragments. Alternatively, Fab' expression libraries can
be
constructed (Huse et al., 1989, Science, 246:1274-1281) to allow rapid and
easy
identification of monoclonal Fab' fragments with the desired specificity. The
present
invention encompasses antibodies and antibody fragments.
A single chain Fv molecule (scFv) comprises a. VL domain and a VH domain.
The VL and VH domains associate to form a target binding site. These two
domains are
further covalently linked by a peptide linker (L). A scFv molecule is denoted
as either
VL-L-VH if the VL domain is the N-terminal part of the scFv molecule, or as VH-
L-VL
if the VH domain is the N-terminal part of the scFv molecule. Methods for
making scFv
molecules and designing suitable peptide linkers are described in US Patent
No.
4,704,692, US Patent No. 4,946,778, R. Raag and M. Whitlow, "Single Chain
Fvs."
FASEB Vol 9:73-80 (1995) and R.E. Bird and B.W. Walker, "Single Chain Antibody
Variable Regions," TIBTECH, Vol 9: 132-137 (1991).

An antibody fragment can be prepared by proteolytic hydrolysis of the full
length antibody or by expression in E. coli or another host of the DNA coding
for the


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
fragment. An antibody fragment can be obtained by pepsin or papain digestion
of full
length antibodies by conventional methods. For example, an antibody fragment
can
be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S
fragment denoted F(ab')2. This fragment can be further cleaved using a thiol
reducing
agent, and optionally a blocking group for the sulfhydryl groups resulting
from
cleavage of disulfide linkages, to produce 3.5S Fab' monovalent fragments.
Alternatively, an enzymatic cleavage using papain produces two monovalent Fab
fragments and an Fc fragment directly. These methods are described, for
example, by
Goldenberg, U.S. Patent Nos. 4,036,945 and 4,331,647 and references contained
thereinõ Also,
see Nisonoff et al., Arch Biochem. Biophys. 89: 230 (1960); Porter, Biochem. J
73:
119 (1959), Edelman et al., in METHODS IN. ENZYMOLOGY VOL. 1, page 422
(Academic Press 1967), and Coligan at pages 2.8.1-2.8.10 and 2.10.-2.10.4.
Another form of an antibody fragment is a peptide coding for a single
eomplementarity-determining region (CDR). A CDR is a segment of the variable
region of an antibody that is complementary in structure to the epitope to
which the
antibody binds and is more variable than the rest of the variable-region.
Accordingly,
a CDR is sometimes referred to as hypervariable region. A variable region
comprises
three CDRs. CDR peptides can be obtained by constructing genes encoding the
CDR.
of an antibody of interest. Such genes are prepared, for example, by using the
polymerase chain reaction to synthesize the variable region from RNA of
antibody-
producing cells. See, for example, Larrick et al., Methods: A Companion to
Methods
in Enzv,nology 2: 106 (1991); Courtenay-Luck, "Genetic Manipulation of
Monoclonal
Antibodies," in MONOCLONAL ANTIBODIES: PRODUCTION, ENGINEERING
AND CLINICAL APPLICATION, Ritter et al. (eds.), pages 166-179 (Cambridge
University Press 1995); and Ward et al., "Genetic Manipulation and Expression
of
.Antibodies," in MONOCLONAL ANTIBODIES: PRINCIPLES AND
APPLICATIONS, Birch et al., (eds.), pages 137-185 (Wiley-Liss, Inc: 1995).
Other methods of cleaving antibodies, such as separation of heavy chains to
form monovalent light-heavy chain fragments, further cleavage of fragments, or
other
enzymatic, chemical or genetic techniques may also be used, so long as the
fragments
bind to the antigen that is recognized by the intact antibody.

31


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
5. Aultispecific and multivalent antibodies
The anti-CD20 antibodies, as well as other antibodies with different
specificities for use in combination therapy, described herein, can also be
made as
multispecific antibodies (comprising at least one binding site to a CD20
epitope or
antigen and at least one binding site to another epitope on CD20 or another
antigen)
and multivalent antibodies (comprising multiple binding sites to the same
epitope or
antigen). Multivalent target binding proteins are described in US Serial No.
09/911,610 (Leung et al.).
The present invention provides a bispecific antibody or antibody fragment
having at least a binding region that specifically binds a targeted cell
marker and at
least one other binding region that specifically binds a targetable conjugate.
The
targetable conjugate comprises a carrier portion which comprises or bears at
least one
epitope recognized by at least one binding region of the bispecific antibody
or
antibody fragment.
A variety of recombinant methods can be used to produce bispecific antibodies
and antibody fragments as described above.
An anti-CD20 multivalent antibody is also contemplated in the present
invention. This multivalent target binding protein is constructed by
association of a
first and a second polypeptide. The first polypeptide comprises a first single
chain Fv
molecule covalently linked to a first immunoglobulin-like domain which
preferably is
an immunoglobulin light chain variable region domain. The second polypeptide
comprises a second single chain Fv molecule covalently linked to it second
immunogglobulin-like domain which preferably is an immunoglobulin heavy chain
variable region domain. Each of the first and second single chain Fv molecules
forms
a target binding site, and the first and second immunoglobulin-like domains
associate
to form a third target binding site.
A single chain Fv molecule with the VL-L-VH configuration, wherein L is a
linker, may associate with another single chain Fv molecule with the VH-L-VL
configuration to form a bivalent dimer. In this case, the VL domain of the
first scFv
and the VH domain of the second scFv molecule associate to form one target
binding
site, while the VH domain of the first scFv and the VL domain of the second
scFv
associate to form the other target binding site.

32


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
Another embodiment of the present invention is a CD20 bispecific, trivalent
targeting protein comprising two heterologous polypeptide chains associated
non-
covalently to form three binding sites, two of which have affinity for one
target and a
third which has affinity for a hapten that can be made and attached to a
carrier for a
diagnostic and/or therapeutic agent. Preferably, the binding protein has two
CD20
binding sites and one CD22 binding site. The bispecific, trivalent targeting
agents
have two different scFvs, one scFv contains two VH domains from one antibody
connected by a short linker to the VL domain of another antibody and the
second scFv
contains two VL domains from the first antibody connected by a short linker to
the VH
domain of the other antibody. The methods for generating multivalent,
multispecific
agents from VH and VL domains provide that individual chains synthesized from
a
DNA plasmid in a host organism are composed entirely of VH domains (the VH-
chain)
or entirely of VL domains (the VL-chain) in such a way that any agent of
multivalency
and multispecificity can be produced by non-covalent association of one VH-
chain
with one VL-chain. For example, forming a trivalent, trispecific agent, the VH-
chain
will consist of the amino acid sequences of three VH domains, each from an
antibody
of different specificity, joined by peptide linkers of variable lengths, and
the VL-chain
will consist of complementary VL domains, joined by peptide linkers similar to
those
used for the VH-chain. Since the VH and VL domains of antibodies associate in
an
anti-parallel fashion, the preferred method in this invention has the VL
domains in the
VL-chain arranged in the reverse order of the VH domains in the VH-chain.

6. Diabodies, Triabodies and Tetrabodies

The anti-CD20 antibodies of the present invention can also be used to prepare
functional bispecific single-chain antibodies (bscAb), also called diabodies,
and can
be produced in mammalian cells using recombinant methods. See, e.g., Mack et
al.,
Proc. Natl. Acad. Sci., 92: 7021-7025, 1995. For example, bscAb are
produced by joining two single-chain Fv fragments via a glycine-serine linker
using
recombinant methods. The V light-chain (VL) and V heavy-chain (VH) domains of
two antibodies of interest are isolated using standard PCR methods. The VL and
VH
cDNA's obtained from each hybridoma are then joined to form a single-chain

fragment in a two-step fusion PCR. The first PCR step introduces the (Gly4-
Serl)3
linker, and the second step joins the VL and VH amplicons. Each single chain
33


CA 02752553 2011-09-13

WO 03/068821 PCT/CB03/00665
molecule is then cloned into a bacterial expression vector. Following
amplification,
one of the single-chain molecules is excised and sub-cloned into the other
vector,
containing the second single-chain molecule of interest. The resulting bscAb
fragment is subcloned into an eukaryotic expression vector. Functional protein
expression can be obtained by transfecting the vector into chinese hamster
ovary cells.
Bispecific fusion proteins are prepared in a similar manner. Bispecific single-
chain
antibodies and bispecific fusion proteins are included within the scope of the
present
invention.
For example, a humanized, chimeric or human anti-CD20 monoclonal
.antibody can be used to produce antigen specific diabodies, triabodies, and
tetrabodies. The monospecific diabodies, triabodies, and tetrabodies bind
selectively
to targeted antigens and as the number of binding sites on the molecule
increases, the
affinity for the target cell increases and a longer residence time is observed
at the
desired location. For diabodies, the two chains comprising the VH polypeptide
of the
humanized CD20 MAb connected to the VK polypeptide of the humanized CD20
1\4Ab by a five amino acid residue linker are utilized. - Each chain forms one
half of
the humanized CD20 diabody. In the case of triabodies, the three chains
comprising
VH polypeptide of the humanized CD20 MAb connected to the VK polypeptide of
the
humanized CD20 MAb by no linker are utilized. Each chain forms one third of
the
hCD2.0 triabody.
The ultimate use of the bispecific diabodies described herein is for pre-
targeting CD20 positive tumors for subsequent specific delivery of diagnostic
or
therapeutic agents. These diabodies bind selectively to targeted antigens
allowing for
increased affinity and a longer residence time at the desired location.
Moreover, non-
antigen bound diabodies are cleared from the body quickly and exposure of
normal
tissues is minimized. Bispecific antibody point mutations. for enhancing the
rate of
clearance are known.

Bispecific diabodies for affinity enhancement are disclosed in US
Application Nos. 101270,07.1 (Rossi et al.), 101270,073 (Rossi et al.) and
10/328,190
(Rossi et al.). The
diagnostic and therapeutic agents can include isotopes, drugs, toxins,
cytokines,
hormones, growth factors, conjugates, radionuclides, and metals. For example,

34


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
gadolinium metal is used for magnetic resonance imaging (MRI). Examples of
radionuclides are 225Ac, 18F, 68Ga, 67Ga, 90Y, 86 Y, 111In11311' 1251, 1231,
99m.Tc, 94mTC,
186Re, 1S8Re, 177Lu, 62Cu, 64Cu4 67Cu, 212Bi, 213Bi, 32p, 11C, 13N, 150, 76Br,
and 211At.

Other radionuclides are also available as diagnostic and therapeutic agents,
especially
those in the energy range of 60 to 4,000 keV.
More recently, a tetravalent tandem diabody (termed tandab) with dual
specificity has also been reported (Cochlovius et al:, Cancer Research (2000)
60:
4336-4341). The bispecific tandab is a dimer of two identical polypeptides,
each
containing four variable domains of two different antibodies (VHI, VL1, VH2,
VL2)
linked in an orientation to facilitate the formation of two potential binding
sites for
each of the two different specificities upon self-association.

7. Conjugated multivalent and multispecific anti-CD20 antibodies
In another embodiment of the instant invention is a conjugated multivalent
anti-
CD20 antibody. Additional amino acid residues may be added to either the N- or
C-
terminus of the first or the second polypeptide. The additional amino acid
residues may
comprise a peptide tag, a signal peptide, a cytokine, an enzyme (for example,
a pro-drug
activating enzyme), a hormone, a peptide toxin, such as pseudomonas extoxin, a
peptide
drug, a cytotoxic protein or other functional proteins. As used herein, a
functional
protein is a protein which has a biological function.
In one embodiment, drugs, toxins, radioactive compounds, enzymes,
hormones, cytotoxic proteins, chelates, cytokines and other functional agents
may be
conjugated to the multivalent target binding protein, preferably through
covalent
attachments to the side chains of the amino acid residues of the multivalent
target
binding protein, for example amine, carboxyl, phenyl, thiol or hydroxyl
groups.
Various conventional linkers may be used for this purpose, for example,
diisocyanates, diisothiocyanates, bis(hydroxysuccinimide) esters,
carbodiimides,
maleimide-hydroxysuccinimide esters, glutaraldehyde and the like. Conjugation
of
agents to the multivalent protein preferably does not significantly affect the
protein's
binding specificity or affinity to its target. As used herein, a functional
agent is an
agent which has a biological function. A preferred functional agent is a
cytotoxic
agent.



CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
In still other embodiments, bispecific antibody-directed delivery of
therapeutics or prodrug polymers to in vivo targets can be combined with
bispecific
antibody delivery of radionuclides, such that combination chemotherapy and
radioinununotherapy is achieved. Each therapy can be conjugated to the
targetable
conjugate and administered simultaneously, or the nuclide can be given as part
of a
first targetable conjugate and the drug given in a later step as part of a
second
targetable conjugate.
In another embodiment, cytotoxic agents may be conjugated to a polymeric
carrier, and the polymeric carrier may subsequently be conjugated to the
multivalent
target binding protein. For this method, see Ryser et al., Proc. Nail. Acad.
Sci. USA,
75:3867-3870, 1978, US Patent No. 4,699,784 and US Patent No. 4,046,722.
Conjugation preferably does not significantly
affect the binding specificity or affinity of the multivalent binding protein.

8. Humanized, Chimeric and Human Antibodies Use for Treatment and
Diagnosis
Humanized, chimeric and human monoclonal antibodies, i.e., anti-CD20 MAbs
and other MAbs described herein, in accordance with this invention are
suitable for
use in therapeutic methods and diagnostic methods. Accordingly, the present
invention
contemplates the administration of the humanized, chimeric and human
antibodies of the
present invention alone as a naked antibody or administered as a multimodal
therapy,
temporally according to a dosing regimen, but not conjugated to, a therapeutic
agent.
The efficacy of the naked anti-CD20 MAbs can be enhanced by supplementing
naked
antibodies with one or more other naked antibodies, i.e., MAbs to specific
antigens, such
as CD4, CD5, CD8, CD14, CD15, CD19, CD21, CD22, CD23, CD25, CD33, CD37,
CD38, CD40, CD40L, CD46, CD52, CD54, CD74, CD80, CD126, B7, MUM, Ia,
HM1 24, or HLA-DR, tenascin, VEGF, P1GF, an oncogene, an oncogene product, or
a
combination thereofwith one or more immunoconjugates of anti-CD20, or
antibodies to
theses recited antigens, conjugated with therapeutic agents, including drugs,
toxins,
immunomodulators, hormones, therapeutic radionuclides, etc., with one or more
therapeutic agents, including drugs, toxins, immunomodulators, hormones,
therapeutic
radionuclides, etc., administered concurrently or sequentially or according to
a
prescribed dosing regimen, with the MAbs. Preferred B-cell antigens include
those
36


CA 02752553 2011-09-13

WO 03/068821 PCT/6B03/00665
equivalent to human CD19, CD20, CD21, CD22., CD23, CD46, CD52, CD74, CD80,
and CD5 antigens. Preferred T-cell antigens include those equivalent to human
CD4,
CD8 and CD25 (the IL-2 receptor) antigens. An equivalent to HLA-DR antigen can
be used in treatment of both B-cell and T-cell disorders. Particularly
preferred B-cell
antigens are those equivalent to human CD19, CD22, CD21, CD23, CD74, CD80, and
HLA-DR antigens. Particularly preferred T-cell antigens are those equivalent
to
human CD4, CD8 and CD25 antigens. CD46 is an antigen on the surface of cancer
cells that block complement-dependent lysis (CDC).

Further, the present invention contemplates the administration of an
immunoconjugate for diagnostic and therapeutic uses in B cell lymphomas and
other
disease or disorders. An immunoconjugate, as described herein, is a molecule
comprising an antibody component and a therapeutic or diagnostic agent,
including a
peptide which may bear the diagnostic or therapeutic agent. An immunoconjugate
retains the immunoreactivity of the antibody component, i.e., the antibody
moiety has
about the same or slightly reduced ability to bind the cognate antigen after
conjugation
as before conjugation.
A wide variety of diagnostic and therapeutic agents can be advantageously
conjugated to the antibodies of the invention. The therapeutic agents recited
here are
those agents that also are useful for administration separately with the naked
antibody as
described above. Therapeutic agents include, for example, chemotherapeutic
drugs such
as vinca alkaloids, anthracyclines, epidophyllotoxin, taxanes,
antimetabolites, alkylating
agents, antikinase agents, antibiotics, Cox-2 inhibitors, antimitotics,
antiangiogenic and
apoptotoic agents, particularly doxorubicin, methotrexate, taxol, CPT-1 1,
camptothecans, and others from these and other classes of anticancer agents ,
and the
like. Other useful cancer chemotherapeutic drugs for the preparation of
immunoconjugates and antibody fusion proteins include nitrogen mustards, alkyl
sulfonates, nitrosoureas, triazenes, folic acid analogs, COX-2 inhibitors,
pyrimidine
analogs, purine analogs, platinum coordination complexes, hormones, and the
like.
Suitable chemotherapeutic agents are described in REMINGTON'S
PHARMACEUTICAL SCIENCES, 19th Ed. (Mack Publishing Co. 1995), and in
GOODMAN AND GILMAN'S THE PHARMACOLOGICAL BASIS OF
THERAPEUTICS, 7th Ed. (MacMillan Publishing Co. 1985), as well as revised
37


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
editions of these publications. Other suitable chemotherapeutic agents, such
as
experimental drugs, are known to those of skill in the art.
Additionally, a chelator such as DTPA, DOTA, TETA, or NOTA or a suitable
peptide, to which a detectable label, such as a fluorescent molecule, or
cytotoxic agent,
such as a heavy metal or radionuclide, can be conjugated. For example, a
therapeutically useful immunoconjugate can be obtained by conjugating a
photoactive
agent or dye to an antibody composite. Fluorescent compositions, such as
fluorochrome, and other chromogens, or dyes, such as porphyrins sensitive to
visible
light, have been used to detect and to treat lesions by .directing the
suitable light to the
lesion. In therapy, this has been termed photoradiation, phototherapy, or
photodynarnic therapy (Joni et al. (eds.), PHOTODYNAMIC THERAPY OF
TUMORS AND OTHER DISEASES (Libreria Progetto 1985); van den Bergh, Chem.
Britain 22:430 (1986)). Moreover, monoclonal antibodies have been coupled with
photoactivated dyes for achieving phototherapy. Mew et al., J. Immunol.
130:1473
(1983); idem., Cancer Res. 45:4380 (1985); Oseroff et al., Proc. Natl. Acad
Sci. USA
83:8744 (1986); idem., Photochem. Photobiol. 46:83 (1987); Hasan et al., Prog.
Clin.
Biol. Res. 288:471 (1989); Tatsuta et al., Lasers Surg. Med 9:422 (1989);
Pelegrin et
al., Cancer 67:2529 (1991). However, these earlier studies did not include use
of
endoscopic therapy applications, especially with the use of antibody fragments
or
subfragments. Thus, the present invention contemplates the 'therapeutic use of
immunoconjugates comprising photoactive agents or dyes.
Also contemplated by the present invention are the use of radioactive and non-
radioactive agents as diagnostic agents. A suitable non-radioactive diagnostic
agent is
a contrast agent suitable for magnetic resonance imaging, computed tomography
or
ultrasound. Magnetic imaging agents include, for example, non-radioactive
metals,
such as manganese, iron and gadolinium, complexed with metal-chelate
combinations
that include 2 benzyl-DTPA and its monomethyl and cyclohexyl analogs, when
used
along with the antibodies of the invention. See U.S. Serial No. 09/921,290
filed on
October 10, 2001.

Furthermore, a radiolabeled antibody or immunoconjugate may comprise a y-
emitting radioisotope or a positron-emitter useful for diagnostic imaging.
Suitable
radioisotopes, particularly in the energy range of 60 to 4,000keV, include
1311, 1231,
1241, 86Y662 Cu, 640u, 111In' 670a, 68Ga, 99mTC, 94mTc, 18F, 11C, 13N, 150
75Br, and the
38


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
like. See for example, U.S. Patent Application entitled "Labeling Targeting
Agents
with Gallium-68"- Inventors G.L.Griffiths and. W.J. McBride, (U.S. Provisional
Application No. 60/342,104), which discloses positron emitters, such as 18F,
6SGa,
94mTc. and the like, for imaging purposes and which is incorporated in its
entirety by
reference. Particularly useful therapeutic radionuclides include, but are not
limited to,
32P, 33P, 47SC, 64C111 67Cu, 67Ga, 90Y, 1114, 1111n, 1251, 1311, 142Pr, 153Sm,
161Th, 166Dy,
166Ho, 177Lu, 186Re, I88Re, 189Re, 212Pb, 212Bi, 213Bi, 211At, 223Rd and
225Ac.

Particularly useful diagnostic/detection radionuclides include, but are not
limited to,
18F, 52Fe, 62Cu, 64Cu, 67CU, 67Ga, 68Ga, 86Y, 89Zr, 94mTC, 94TC, 99mTe, 111in,
1231, 1241,
1251, 1311, 154-158Gd, 32P, 90Y, 18SRe, and 175Lu.

A toxin, such as Pseudomonas exotoxin, may also be complexed to or form the
therapeutic agent portion of an antibody fusion protein of an anti-CD20
antibody of the
present invention. Other toxins suitably employed in the preparation of such
conjugates or other fusion proteins, include ricin, abrin, ribonuclease
(RNase), DNase
I, Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin,
diphtherin
toxin, Pseudomonas exotoxin, and Pseudomonas endotoxin. See, for
example,.Pastan
et al., Cell 47:641 (1986), and Goldenberg, CA -A Cancer Journal for
Clinicians
44:43 (1994). Additional toxins suitable for use in the present invention are
known to
those of skill in the art and are disclosed in U.S. Patent 6,077,499.

An immunomodulator, such as a cytokine may also be conjugated to, or form
the therapeutic agent portion of an antibody fusion protein or be administered
with the
humanized anti-CD20 antibodies of the present invention. Suitable cytokines
for the
present invention include, but are not limited to, interferon and
interleukins, as
described below.
An oligonucleotide, such the antisense molecules inhibiting bel-2 expression
that are described in U.S. 5,734,033 (Reed),
may also be conjugated to, or form the therapeutic agent portion of an
antibody
fusion protein or be administered with the humanized anti-CD20 antibodies of
the
present invention.

39


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
9. Preparation of Immunoconjugates
Any of the antibodies or antibody fusion proteins of the present invention can
be conjugated with one or more therapeutic or diagnostic agents. Generally,
one
therapeutic or diagnostic agent is attached to each antibody or antibody
fragment but
more than one therapeutic agent or diagnostic agent can be attached to the
same
antibody or antibody fragment. The antibody fusion proteins of the present
invention
comprise two or more antibodies or fragments thereof and each of the
antibodies that
composes this fusion protein can contain a therapeutic agent or diagnostic
agent.
Additionally, one or more of the antibodies of the antibody fusion protein can
have
more than one therapeutic of diagnostic agent attached. Further, the
therapeutic
agents do not need to be the same but can be different therapeutic agents. For
example, one can attach a drug and a radioisotope to the same fusion protein.
Particularly, an IgG can be radiolabeled with 13'1 and attached to a drug. The
1311 can
be incorporated into the tyrosine of the IgG and the drug attached to the
epsilon amino
group of the IgG lysines. Both therapeutic and diagnostic agents also can be
attached
to reduced SH groups and to the carbohydrate side chains.
Radionuclides suitable for treating a disease tissue substantially decay by
beta-
particle emission and include, but are not limited to: 32P, 33P, 47Sc, 59Fe,
64Cu, 67Cu,
75Se, 77As, 89Sr, 90Y, 99MO, 105Rh, 109Pd, 111Ag, 1251, 1311, 142Pr, 143Pr,
149Pm, 153Sm,
161-Tb, 16611o, 169Er, 177Lu, 186Re, 188Re, 189Re, 1941r, 198Au, 199Au, 211Pb,
212Pb and 213Bi.

Maximum decay energies of useful beta-particle-emitting nuclides are
preferably 20-
5,000 keV, more preferably 100-4,000 keV, and most preferably 500-2,500 keV.
Also preferred are radionuclides that substantially decay with Auger-emitting
particles. For example, 58Co, 67Ga, SOmBr, 99mTC, 103mpjl, 109p, 1 "In, 119Sb,
1251, 161Ho,
159mOs and '921r. Decay energies of useful Auger-particle-emitting nuclides
are
preferably < 1,000 keV, more preferably < 100 keV, and most preferably < 70
keV.
Also preferred are radionuclides that substantially decay with generation of
alpha-
223
particles. Such radionuclides include, but are not limited to: 1s2Dy, 211m,
212Bi,
Ra,
2191 215po, 211Bi, 22'Ac, 221Fr, 217At, 213Bi.and 255Fm. Decay energies of
useful alpha-
particle-emitting radionuclides are preferably 2,000-10,000 keV, more
preferably
3,000-8,000 keV, and most preferably 4,000-7,000 keV.
Radionuclides useful as diagnostic agents utilizing gamma-ray detection
include, but are not limited to: 51Cr, 57Co, 58Co, 59Fe, 67Cu, 67Ga, 75Se,
97Ru, 99mTc,


CA 02752553 2011-09-13

WO 03/068821 PCT/CB03/00665
111111, 114mIn, 1231, 1251, 1311, 169, 197Hg, and 201T1. Decay energies of
useful gamma-
ray emitting radionuclides are preferably 20-2000 ke'V, more preferably 60-600
keV,
and most preferably 100-300 keV.

.Radionuclides useful for positron emission tomography include, but are not
limited to: '8F, 51 , 52m , 52Fe, 55C0, 62Cu, 64Cu, 65 , 72M, 75Br, 76Br,
s2mRb,
83Sr, 86Y, "Zr, 94mTc, 110In11201, and 124L Total decay energies of useful
positron-
emitting radionuclides are preferably < 2,000 keV, more preferably under 1,000
keV,
and most preferably < 700 keV.
Bispecific antibodies of the present invention are useful in pretargeting
methods
and provide a preferred way to deliver two therapeutic agents or two
diagnostic agents to
a subject. U.S. Serial Nos. 09/382,186 and 09/337,756 discloses a method of
pretargeting using a bispecific antibody, in which the bispecific antibody is
labeled with
125I and delivered to a subject, followed by a divalent peptide labeled with
99mTc.

Pretargeting methods are also
described in US Serial Nos. 09/823,746 (Hansen et al.) and 10/150,654
(Goldenberg et
al.),
.

The delivery results in excellent tumor/normal tissue ratios for 125I and
99mTc, thus showing the utility of two diagnostic radioisotopes. Any
combination of
known therapeutic agents or diagnostic agents can be used to label the
antibodies and
antibody fusion proteins. The binding specificity of the antibody component of
the MAb
conjugate, the efficacy of the therapeutic agent or diagnostic agent and the
effector
activity of the Fc portion of the antibody can be determined by standard
testing of the
conjugates.
The invention is directed to a method for pretargeting a cell in a patients
suffering from a B-cell lymphoma or leukemia or an autoimmune disease
comprising-
(i) administering an antibody fusion protein or fragment thereof that is
multispecific having at least one arm that specifically binds the cell and at
least one
other arm that specifically binds a targetable conjugate; (ii) optionally,
administering
to the patient a clearing composition, and allowing the composition to clear
non-
antigen bound antibody fusion protein or fragment thereof from circulation;
and (iii)
administering to the patient a targetable conjugate comprising a carrier
portion which
41


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
comprises or bears at least one epitope recognizable by at least one other arm
of the
antibody fusion protein or fragment thereof, and is conjugated at least one
first
therapeutic or diagnostic agent. The antibody fusion protein of the present
invention
should be multispecific antibody. In a preferred embodiment the antibody is a
bispecific
antibody, and can be a diabody. The first therapeutic agent is selected from
the group
consisting of a radioactive label, an immunomodulator, a hormone, a
photoactive
therapeutic agent, a cytotoxic agent, an oligonucleotide and a combination
thereof and
wherein the first diagnostic agent is at least one of a radioactive label, a
photoactive
diagnostic agent or a non-radioactive label.
The antibody fusion protein or fragment thereof also may be conjugated to a
second
therapeutic, such as at least one radioactive label, an immunomodulator, a
hormone, a
photoactive therapeutic agent, a cytotoxic agent, an oligonucleotide and a
combination
thereof or may be conjugated the second diagnostic agent, such as at least one
of a
radioactive label, a photoactive diagnostic agent or a non-radioactive label.
In one
embodiment, the first and second therapeutic agent or diagnostic agent are the
same.
A therapeutic or diagnostic agent can be attached at the hinge region of a
reduced antibody component via disulfide bond formation. As an alternative,
such
peptides can be attached to the antibody component using a heterobifunctional
cross-
linker, such as N-succinyl 3-(2-pyridyldithio)propionate (SPDP). Yu et al.,
Int. J
Cancer 56: 244 (1994). General techniques for such conjugation are well-known
in
the art. See, for example, Wong, CHEMISTRY OF PROTEIN CONJUGATION
AND CROSS-LINKING (CRC Press 1991); Upeslacis et al., "Modification of
Antibodies by Chemical Methods," in MONOCLONAL ANTIBODIES:
PRINCIPLES AND APPLICATIONS, Birch et al. (eds.), pages 187-230 (Wiley-Liss,
Inc. 1995); Price, "Production and Characterization of Synthetic Peptide-
Derived
Antibodies," in MONOCLONAL ANTIBODIES: PRODUCTION, ENGINEERING
AND CLINICAL APPLICATION, Ritter et al. (eds.), pages 60-84 (Cambridge
University Press 1995). Alternatively, the therapeutic or diagnostic agent can
be
conjugated via a carbohydrate moiety in the Fc region of the antibody. The
carbohydrate group can be used to increase the loading of the same peptide
that is
bound to a thiol group, or the carbohydrate moiety can be used to bind a
different
peptide.

42


CA 02752553 2011-09-13

WO 03/068821 PCT/CB03/00665
Methods for conjugating peptides to antibody components via an antibody
carbohydrate moiety are well-known to those of skill in the art. See, for
example,
Shih et al., Int. J. Cancer 41: 832 (1988); Shih et al., Int. J Cancer 46:
1101 (1990);
and Shih et al., U.S. Patent No. 5,057,313.
The general method involves reacting an antibody component
having an oxidized carbohydrate portion with a carrier polymer that has at
least one
free amine function and that is loaded with a plurality of peptide. This
reaction results
in an initial Schiff base (imine) linkage, which can be stabilized by
reduction to a
secondary amine to form the final conjugate.
The Fe region is absent if the antibody used as the antibody component of the
immunoconjugate is an antibody fragment. However, it is possible to introduce
a
carbohydrate moiety into the light chain variable region of a full length
antibody or
antibody fragment. See, for example, Leung et al., J..Immunol. 154: 5919
(1995);
Hansen et al., U.S. Patent No. 5,443,953 (1995), Leung et al., U.S. patent No.
6,254,868. The .
engineered carbohydrate moiety is used to attach the therapeutic or diagnostic
agent.
10._ Pharmaceutically Acceptable Excipients
The humanized, chimeric and human anti-CD20 mAbs to be delivered to a
subject can consist of the MAb alone, immunoconjugate, fusion protein, or can
comprise one -or more pharmaceutically suitable excipients, one or. more
additional
ingredients, or some combination of these.
The immunoconjugate or naked antibody of the present invention can be
formulated according to known methods to prepare pharmaceutically useful
compositions, whereby the immunoconjugate or naked antibody are combined in a
mixture with a pharmaceutically suitable excipient. Sterile phosphate-buffered
saline
is one example of a pharmaceutically suitable excipient. Other suitable
excipients are.
well-known to those in the art. See, for example, Ansel et al., PHARMACEUTICAL
DOSAGE FORMS AND DRUG DELIVERY SYSTEMS, 5th Edition (Lea & Febiger
1990), and Gennaro (ed.), REMINGTON'S PHARMACEUTICAL SCIENCES, 1 Sth
Edition (Mack Publishing Company 1990), and revised editions thereof.
The immunoconjugate or naked antibody of the present invention can be
formulated for intravenous administration via, for example, bolus injection or
43


CA 02752553 2011-09-13

WO 03/068821 PCT/603/00665
continuous infusion. Preferably, the antibody of the present invention is
infused over
a period of less than about 4 hours, and more preferably, over a period of
less than
about 3 hours. For example, the first 25-50 mg could be infused within 30
minutes,
preferably even 15 min, and the remainder infused over the next 2-3 hrs.
Formulations
for injection can be presented in unit dosage form, e.g., in ampules or in
multi-dose
containers, with an added preservative. The compositions can take such forms
as
suspensions, solutions or emulsions in oily or aqueous vehicles, and can
contain
formulatory agents such as suspending, stabilizing and/or dispersing agents.
Alternatively, the active ingredient can be in powder form for constitution
with a
suitable vehicle, e.g., sterile pyrogen-free water, before use.
Additional pharmaceutical methods may be employed to control the duration
of action of the therapeutic or diagnostic conjugate or naked antibody.
Control
release preparations can be prepared through the use of polymers to complex or
adsorb the immunoconjugate or naked antibody. For example, biocompatible
polymers include matrices of poly(ethylene-co-vinyl acetate) and matrices of a
polyanhydride copolymer of a stearic acid dimer and sebacic acid. Sherwood et
al.,
Bio/Technology 10: 1446 (1992). The rate of release of an immunoconjugate or
antibody from such a matrix depends upon the molecular weight of the
immunoconjugate or antibody, the amount of immunoconjugate, antibody within
the
matrix, and the size of dispersed. particles. Saltzman et al., Biophys. J. 55:
163
(1989); Sherwood et al., supra. Other solid dosage forms are described in
Ansel et
al., PHARMACEUTICAL DOSAGE FORMS AND DRUG DELIVERY SYSTEMS,
5th Edition (Lea & Febiger 1990), and Gennaro (ed.), REMINGTON'S
PHARMACEUTICAL SCIENCES, 18th Edition (Mack Publishing Company- 1990),
and revised editions thereof.
The immunoconjugate, antibody fusion proteins, or naked antibody may also
be administered to a mammal subcutaneously or even by other parenteral routes.
Moreover, the administration may be by continuous infusion or by single or
multiple
boluses. Preferably, the antibody of the present invention us infused over a
period of
less than about 4 hours, and more preferably, over a period of less than about
3 hours.
This is preferably performed by infusing slowly at first. For example, a dose
of 25 to
50 mg is infused within 15-30 minutes and the remainder of the dose is infused
over a
period of up to 2-3 hrs. In general, the dosage of an administered
immunoconjugate,

44


CA 02752553 2011-09-13

WO (13/068821 PCT/CB03/00665
fusion protein or naked antibody for humans will vary depending upon such
factors as
the patient's age, weight, height, sex, general medical condition and previous
medical
history. Typically, it is desirable to provide the recipient with a dosage of
immunoconjugate, antibody fusion protein or naked antibody that is in the
range of
from about 1 mg/kg to 20 mg/kg as a single intravenous infusion, although a
lower or
higher dosage also may be administered as circumstances dictate. Therefore, 1-
20
mg/kg for a 70 kg patient, for example, is a dose of 70-1,400 mg, or 41-824
mg/m2 for
a 1.7-m patient. This dosage may be repeated as needed, for example, once per
week
for 4-10 weeks, preferably once per week for 8 weeks, and more preferably,
once per
week for 4 weeks. It may also be given less frequently, such as every other
week for
several months. More specifically, an antibody of the present invention, such
as
naked anti-CD20, may be administered as one dosage every 2 or 3 weeks,
repeated for
a total of at least 3 dosages. Also preferred, the antibodies of the present
invention
may be administered once per week for 4-8 weeks. In other words, if the dosage
is
lowered to approximately 200-300 mg/m2 (which is 340 mg per dosage for a 1.7-m
patient, or 4.9 mg/kg for a 70 kg patient), it may be administered once weekly
for 4 to
8 weeks. Alternatively, the dosage schedule may be decreased, namely every 2
or 3
weeks for 2-3 months; for example, if the dosage is 300-500 mg/m' (i.e., 510-
850 mg
for a 1.7-m patient, or 7.3-12 mg/kg for a 70 kg patient). The dosing schedule
can
optionally be repeated at other intervals and dosage may be given through
various
parenteral routes, with appropriate adjustment of the dose and schedule.

For purposes of therapy, the immunoconjugate, fusion protein, or naked
antibody is administered to a mammal in a therapeutically effective amount. A
suitable subject for the present invention are usually a human, although a non-
human
animal subject is also contemplated. An antibody preparation is said to be
administered in a "therapeutically effective amount' 'if the amount
administered is
physiologically significant. An agent is physiologically significant if its
presence
results in a detectable change in the physiology of a recipient mammal. In
particular,
an antibody preparation of the present invention is physiologically
significant if its
presence invokes an antitumor response or mitigates the signs and symptoms of
an
autoimmune disease state. A physiologically significant effect could also be
the
evocation of a humoral and/or cellular immune response in the recipient
mammal.



CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
11. Methods of Treatment
The present invention contemplates the use of naked anti-CD20 antibodies of
the present invention as the primary composition for treatment of B cell
disorders and
other diseases. In particular, the compositions described herein are
particularly useful
for treatment of various autoimmune as well as indolent forms of B-cell
lymphomas,
aggressive forms of B-cell lymphomas, chronic lymphatic leukemias, acute
lymphatic
leukemias, and Waldenstrom's macroglobulinernia. For example, the humanized
anti-CD20 antibody components and immunoconjugates can be used to treat both
indolent and aggressive forms of non-Hodgkin's lymphoma.
The compositions for treatment contain at least one humanized, chimeric or
human monoclonal anti-CD20 antibody alone or in combination with other
antibodies, such as other humanized, chimeric, or human antibodies,
therapeutic
agents or immunomodulators. In particular, combination therapy with a fully
human
antibody is also contemplated and is produced by the methods as set forth
above.
Naked or conjugated antibodies to the same or different epitope or antigen
may be also be combined with one or more of the antibodies of the present
invention.
For example, a humanized, chimeric or human naked anti-CD20 antibody may be
combined with another naked humanized, naked chimeric or naked human anti-
CD20,
a humanized, chimeric or human naked anti-CD20 antibody may be combined with
an
anti-CD20 immunoconjugate, a naked anti-CD20 antibody may be combined with an
anti-CD22 radioconjugate or an anti-CD22 naked antibody may be combined with a
humanized, chimeric or human anti-CD20 antibody conjugated to an isotope, or
one
or more chemotherapeutic agents, cytokines, toxins or a combination thereof. A
fusion protein of a humanized, chimeric or human CD20 antibody and a toxin or
immunomodulator, or a fusion protein of at least two different B-cell
antibodies (e.g.,
a CD20 and a CD22 MAb) may also be used in this invention. Many different
antibody combinations, targeting at least two different antigens associated
with B-cell
disorders, as listed already above, may be constructed, either as naked
antibodies or as
partly naked and partly conjugated with a therapeutic agent or
immunomodulator, or
merely in combination with another therapeutic agents, such as a cytotoxic
drug or
with radiation.

As used herein, the term "immunomodulator" includes cytokines, stem cell
growth factors, lymphotoxins, such as tumor necrosis factor (TNF), and
46


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
hematopoietic factors, such as interleukins (e.g., interleukin-1 (IL-1), IL-2,
IL-3, IL-6,
IL-10, IL-12, IL-21 and IL-18), colony stimulating factors (e.g., granulocyte-
colony
stimulating factor (G-CSF) and granulocyte macrophage-colony stimulating
factor
(GM-CSF)), interferons (e.g., interferons-a, -(3 and -y), the stem cell growth
factor
designated "S1 factor," erythropoietin and thrombopoietin. Examples of
suitable
immunomodulator moieties include IL-2, IL-6, IL-10, IL-12, IL-18, IL-21,
interferon-
y, TNF-a, and the like. Alternatively, subjects can receive naked anti-CD20
antibodies and a separately administered cytokine, which can be administered
before,
concurrently or after administration of the naked anti-CD20 antibodies. As
discussed
supra, the anti-CD20 antibody may also be conjugated to the immunomodulator.
The
immunomodulator may also be conjugated to a hybrid antibody consisting of one
or
more antibodies binding to different antigens.
Multimodal therapies of the present invention further include immunotherapy
with naked anti-CD20 antibodies supplemented with administration of anti-CD22,
anti-CD 19, anti-CD21, anti-CD74, anti-CD80, anti-CD23, anti-CD46 or lILA DR
(including the invariant chain) antibodies in the form of naked antibodies,
fusion
proteins, or as immunoconjugates. The naked anti-CD20 antibodies or fragments
thereof may also be supplemented with naked antibodies against a MUCI antigen
that
is expressed on certain B-cells. These antibodies include polyclonal,
monoclonal,
chimeric, human or humanized antibodies that recognize at least one epitope on
these
antigenic determinants. Anti-CD 19 and anti-CD22 antibodies are known to those
of
skill in the art. See, for example, Ghetie et al., Cancer Res. 48:2610 (1988);
Hekman
.et al., Cancer Inzmunol. Iinmunother. 32:364 (1991); Longo, Curr. Opin.
Oncol.
8:353 (1996) and U.S. Patent Nos. 5,798,554 and 6,187,287

In another form of multimodal therapy, subjects' receive naked anti-CD20
antibodies, and/or immunoconjugates, in ' conjunction with standard cancer
chemotherapy. For example, "CVB" (1.5 g/m2 cyclophosphamide, -200-400 mg/m2
etoposide, and 150-200 mg/rn2 carmustine) is a regimen used to treat non-
Hodgkin's
lymphoma. Patti et al., Eur. J. Haematol. 51: 18 (1993). Other suitable
combination
chemotherapeutic regimens are well-known to those of skill in the art. See,
for
example, Freedman et al., "Non-Hodgkin's Lymphomas," in CANCER MEDICINE,
VOLUME 2, 3rd Edition, Holland et al. (eds.), pages 2028-2068 (Lea & Febiger
47


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
1993). As an illustration, first generation chemotherapeutic regimens for
treatment of
intermediate-grade non-Hodgkin's lymphoma (NHL) include C-MOPP
(cyclophosphamide, vincristine, procarbazine and prednisone) and CHOP
(cyclophosphamide, doxorubicin, vincristine, and prednisone). A useful second
generation chemotherapeutic regimen is m-BACOD (methotrexate, bleomycin,
doxorubicin, cyclophosphamide, vincristine, dexamethasone and leucovorin),
while a
suitable third generation regimen is MACOP-B (methotrexate, doxorubicin,
cyclophosphamide, vincristine, prednisone, bleomycin and leucovorin).
Additional
useful drugs include phenyl butyrate and brostatin-1. In a preferred
multimodal
therapy, both chemotherapeutic drugs and cytokines are co-administered with an
antibody, immunoconjugate or fusion protein according to the present
invention. The
cytokines, chemotherapeutic drugs and antibody or immunoconjugate can be
administered in any order, or together.
In a preferred embodiment, NHL or the autoimmune disease is treated with 4
weekly infusions of the humanized anti-CD20 antibody at a does of 200-400
mg/m22
weekly for 4 consecutive weeks (iv over 2-6 hours), repeated as needed 'over
the next
months/yrs. Preferably, the humanized anti-CD-20 antibody is administered at a
dose
of 200-300 mg/m2 once every other week or every third week, for 4 to 8
injections.
Also preferred, NHL is treated with 4 weekly infusions as above, or injections
less
frequently as above, but combined with epratuzumAb (anti-CD22 humanized
antibody) on the same days, at a dose of 360 mg/m2, given as iv infusion over
1 hour,
either before, during or after the anti-CD20 monoclonal antibody infusion. Or,
the
antibodies used in combination therapy may also be infused in alternative
sequences,
such that they are alternated on different weeks, resulting in each being
given every
other week for a total injection sequence for each of 4 to 8 or more doses.
These
dosage schedules can then be repeated at different intervals, such as every 3-
6
months, depending on the patient's clinical status and response to each
therapy
regimen. Still preferred, NHL is treated with 4 weekly infusions, or less
frequent
infusions, of the anti-CD20 antibody as above, combined with one or more
injections
of CD22 MAb radiolabeled with a therapeutic isotope such as yttrium-90 (at a
total
dose of Y-90 between 5 and 35 mCi/meter-square as one or more injections over
a
period of weeks or months). US Serial No. 09/590,284 (Goldenberg et al.)
discloses

48


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
immunotherapy of autoimmune disorders using an anti-CD22 antibody.

In addition, a therapeutic composition of the present invention can contain a,
mixture or hybrid molecules of monoclonal naked anti-CD20 antibodies directed
to
different, non-blocking CD20 epitopes. Accordingly, the present invention
contemplates therapeutic compositions comprising a mixture of monoclonal anti-
CD20 antibodies that bind at least two CD20 epitopes. Additionally, the
therapeutic
composition described herein may contain a mixture of anti-CD20 antibodies
with
varying CDR sequences.
Although naked anti-CD20 antibodies are the primary therapeutic
compositions for treatment of B cell lymphoma and autoimmune diseases, the
efficacy of such antibody therapy can be enhanced by supplementing the naked
antibodies, with supplemental agents, such as immunomodulators, like
interferons,
including IFNI, IFNb and IFNy, interleukins including IL-1, IL-2, IL-6, IL-12,
IL-15,
IL-18, IL-21, and cytokines including G-CSF and GM-CSF. Accordingly, the CD20
antibodies can be combined not only with antibodies and cytokines, either as
mixtures
(given separately or in some predetermined dosing regiment) or as conjugates
or
fusion proteins to the anti-CD20 antibody, but also can be given as a
combination
with drugs. For example, the anti-CD20 antibody may be combined with CHOP as a
4-drug chemotherapy regimen. Additionally, a naked anti-CD20 antibody may be
combined with a naked anti-CD22 antibodies and CHOP or fludarabine as a drug
combination for NHL therapy. Immunotherapy of B-cell malignancies using an
anti-
CD22 antibody is described in US Patent No. 6,183,744 (Goldenberg et al.) and
US
Serial No. 09/307,816 (Goldenberg et al.).
The supplemental therapeutic compositions can be administered
before) concurrently or after administration of the anti-CD20 antibodies.
As discussed supra, the antibodies of the present invention can be used for
treating B cell lymphoma and leukemia, and other B cell diseases or disorders.
For
example, anti-CD20 antibodies can be used to treat B-cell related autoimmune
diseases, including Class III autoimmune diseases such as immune-mediated
thrombocytopenias, such as acute idiopathic thrombocytopenic purpura and
chronic
idiopathic thrombocytopenic purpura, dermatomyositis, Sjogren's syndrome,
multiple
sclerosis, Sydenham's chorea, myasthenia gravis, systemic lupus erythematosus,
49


CA 02752553 2011-09-13

WO 03/068821 PCTIGBO3/00665
lupus nephritis, rheumatic fever, rheumatoid arthritis, polyglandular
syndromes,
bullous pemphigoid, diabetes mellitus, Henoch-Schonlein purpura, post-
streptococcal
nephritis, erythema nodosum, Takayasu's arteritis, Addison's disease,
rheumatoid
arthritis, sarcoidosis, ulcerative colitis, erythema multiforme, IgA
nephropathy,
polyarteritis nodosa, ankylosing spondylitis, Goodpasture's syndrome,
thromboangitis
ubiterans, , primary biliary cirrhosis, Hashimoto's thyroiditis,
thyrotoxicosis,
scleroderma, chronic active hepatitis, polymyositis/dermatomyositis,
polychondritis,
pamphigus vulgaris, Wegener's granulomatosis, membranous nephropathy,
amyotrophic lateral sclerosis, tabes dorsalis, giant cell
arteritis/polymyalgia,
pernicious anemia, rapidly progressive glomerulonephritis and fibrosing
alveolitis.
Anti-CD20 antibodies may also induce apoptosis in cells expressing the CD20
antigen. Evidence of this induction is supported in the literature. For
example, it was
demonstrated that apoptosis could be induced using lymphoid cells that have Fc-

receptors reactive with the IgGI-Fc of CD20 MAbs that crosslinked. See Shan et
al.,
Cancer Inzmunol. Immunother. 48(12):673-683 (2000). Further, it was reported
that
aggregates of a chimeric CD20 MAb, i.e., homopolymers, induced apoptosis. See
Ghetie et al., Blood 97(5): 1392-1398 (2000) and Ghetie et al., Proc. Natl.
Acad. Sci
USA 94(14): 7509-7514 (1997).
Antibodies specific to the CD20 surface antigen of B cells can be injected
into
a mammalian subject, which then bind to the CD20 cell surface antigen of both
normal and malignant B cells. A mammalian subject includes humans and domestic
animals, including pets, such as dogs and cats. The anti-CD20 mAbs of the
present
invention, i.e., humanized, chimeric, human, c aninized and felinized, and
even murine
anti-CD20 mAbs, can be used to treat the non-human mammalian subjects when
there
is a species crossreactivity for the CD20 antigen. See Examples 10 and 11,
below.
The murine mAbs, which are immunogenic in humans, are usually less immunogenic
in non-human mammalian subjects. The anti-CD20 antibody bound to the CD20
surface antigen leads to the destruction and depletion of neoplastic B cells.
Because
both normal and malignant B cells express the CD20 antigen, the anti-CD20
antibody
will result in B cell death. However, only normal B cells will repopulate and
the
malignant B cells will be eradicated or significantly reduced. Additionally,
chemical
agents or radioactive labels having the potential to destroy the tumor can be



CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
conjugated to the anti-CD20 antibody such that the agent is specifically
targeted to the
neoplastic B cells.

12. Expression Vectors
The DNA sequence encoding a humanized, chimeric or human anti-CD20
MAb can be recombinantly engineered into a variety of known host vectors that
provide for replication of the nucleic acid. These vectors can be designed,
using
known methods, to contain the elements necessary for directing transcription,
translation, or both, of the nucleic acid in a cell to which it is delivered.
Known
methodology can be used to generate expression constructs the have a protein-
coding
sequence operably linked with appropriate transcriptional/translational
control signals.
These methods include in vitro recombinant DNA techniques and synthetic
techniques. For example, see Sambrook et al., 1989, MOLECULAR CLONING: A
LABORATORY MANUAL, Cold Spring Harbor Laboratory (New York); Ausubel et
al., 1997, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley &
Sons (New York). Also provided for in this invention is the delivery of a
polynucleotide not associated with a vector.
Vectors suitable for use in the instant invention can be viral or non-viral.
Particular examples of viral vectors include adenovirus, AAV, herpes simplex
virus,
lentivirus, and retrovirus vectors. An example of a non-viral vector is a
plasmid. In a
preferred embodiment, the vector is a plasmid.
An expression vector, as described herein, is a polynucleotide comprising a
gene
that is expressed in a host cell. Typically, gene expression is placed under
the control of
certain regulatory elements, including constitutive or inducible promoters,
tissue-specific
regulatory elements, and enhancers. Such a gene is said to be "operably linked
to" the
regulatory elements.
Preferably, the expression vector of the instant invention comprises the DNA
sequence encoding a humanized, chimeric or human anti-CD20 MAb, which includes
both the heavy and the light chain variable and constant regions. However, two
expression vectors may be used, with one comprising the heavy chain variable
and
constant regions and the other comprising the light chain variable and
constant regions.
Still preferred, the expression vector further comprises a promoter. Because
any strong
promoter can be used, a DNA sequence encoding a secretion signal peptide, a
genomic

51


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
sequence encoding a human IgG l heavy chain constant region, an Ig enhancer
element
and at least one DNA sequence encoding a selection marker.
Also contemplated herein is a method for expressing a humanized anti-CD20
MAb, comprising (i) linearizing at least one expression vector comprising a
DNA
sequence encoding a humanized, chimeric, or human anti-CD20 MAb, (ii)
transfecting
mammalian cells with at least one of said linearized vector, (iii) selecting
transfected
cells which express a marker gene, and (iv) identifying the cells secreting
the humanized
anti-CD20 MAb from the transfected cells.

13. Methods of Making Anti-CD20 Antibodies
In general, the VK (variable light chain) and Vii (variable heavy chain)
sequences
for an anti-CD20 MAb can be obtained. by a variety of molecular cloning
procedures,
such as RT PCR, 5'-RACE, and cDNA library screening. Specifically, the V genes
of an
anti-CD20 MAb can be cloned by PCR amplification from a cell that expresses a
murine
or chimeric. anti-CD20 MAb, sequenced. To confirm their authenticity, the
cloned VL
and VH genes can be expressed in cell culture as a chimeric Ab as described by
Orlandi
et al., (Proc. Natl. Acad. Sci., USA, 86: 3833 (1989)),
Based on the V gene sequences, a humanized anti-CD20 MAb can then be designed
and
constructed as described by Leung et al. (Mol. ImmunoL, 32: 1413 (1995)),
which is
incorporated by reference. cDNA can be - prepared from any known hybridoma
line or
transfected cell line producing a murine or chimeric anti-CD20 MAb by general
molecular cloning techniques (Sambrook et al., Molecular Cloning, A laboratory
manual,. 2" d Ed (1989)). The Vx sequence for the MAb may be amplified using
the
primers VKIBACK and VKIFOR (Orlandi et aL, 1989) or the- extended primer set
described by Leung et al. (BioTechniques, 15: 286 (1993));`,
while VH sequences can be amplified using the primmer pair
VHHIBACK/VHIFOR (Orlandi et al., 1989 above), or the primers annealing to the
constant region of murine IgG described by Leung et al. (Hybridoma, 13:469
(1994)),
The PCR reaction mixtures containing 10 l of the
first strand cDNA product, 10 pl of IOX PCR buffer [500 mM KC1, 100 mM Tris-
HC1
(pH 8.3), 15 mM MgCI2, and 0.01% (w/v).gelatin] (Perkin Elmer Cetus, Norwalk,
CT),
250 pM of each dNTP, 200 nM of the primers, and 5 units of Taq DNA polymerase
(Perlin Elmer Cetus) can be subjected to 30 cycles of PCR Each PCR cycle
preferably
52


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
consists of denaturation at 94 C for I min, annealing at 50 *C for 1.5 min,
and
polymerization at 72'C for 1.5 min. Amplified Vi: and VH fragments can be
purified on
2% agarose (BioRad, Richmond, CA). Similarly, the humanized V genes can be
constructed by a combination of long oligonucleotide template syntheses and
PCR
amplification as described by Leung et al. (Mol. linmunol., 32: 1413 (1995)).
See
Example 3 for a method for the synthesis of an oligo A. and an oligo B on an
automated
RNA/DNA synthesizer (Applied Biosystems, foster City, CA) for use in
constructing
humanized V genes.
PCR products for Vic can be subcloned into a staging vector, such as a pBR327-
based staging vector, VKpBR, that contains an Ig promoter, a signal peptide
sequence
and convenient restriction sites to facilitate in-frame ligation of the Vic
PCR products.
PCR,products for VH can be subcloned into a similar staging vector, such as
the
pBluescript-based VHpBS. Individual clones containing the respective PCR
products
may be sequenced by, for example, the method of Sanger et al. (Proc. NatL Acad
Sci.,
USA, 74: 5463 (1977)),,
The DNA sequences described herein are to, be taken as including all alleles,
mutants and variants thereof, whether occurring naturally or induced.
The expression cassettes containing the Vic and VH, together with the promoter
and signal peptide sequences can be excised from VKpBR and VHpBS,
respectively, by
double restriction digestion as HindIII-BamHI fragments. The Vic and VH
expression
cassettes can then be ligated into appropriate expression vectors, such as pKh
and pG1 g,
respectively (Leung et al., Hybridoma, 13:469 (1994)). The- expression vectors
can be
co-transfected into an appropriate cell, e.g., myeloma Sp2/0-Ag14 (ATCC, VA),
colonies selected for hygromycin resistance, and supernatant fluids monitored
for
production of a chimeric or humanized anti-CD20 MAb by, for example, an ELISA
assay, as described below. Alternately, the Vic and VH expression cassettes
can be
assembled in the modified staging vectors, VKpBR2 and VHpBS2, excised as
Xba1/BamHI and X holBamHI fragments, respectively, and subcloned into a.single
expression vector, such as pdHL2, as described by Gilles et al. (J. ImmunoL
Methods
125:191 (1989) and also shown in Losman et al.,. Cancer, 80:2660 (1997)) for
the
expression in Sp2/0-Ag14 cells. Another vector that is useful in the present
invention is
the GS vector, as described in Barnes et al., Cytotechnology 32:109-123
(2000), which is
preferably expressed in the NSO cell line and CHO cells. Other appropriate
mammalian
53


CA 02752553 2011-09-13

WO 03/1168821 PCT/GB03/00665
expression systems are described in Werner et al., Arzneim.-ForschiDrug Res.
48(II),
Nr. 8, 870-880 (1998).

Co-transfection and assay for antibody secreting clones by ELISA, can be
carried
out as follows. About 10 g of VKpKh (light chain expression vector) and 20
jig of
VHpG1g (heavy chain expression vector) can be used for the transfection of 5 X
106
SP2/0 myeloma cells by electroporation (BioRad, Richmond, CA) according to Co
et al.,
J ImmunoL, 148: 1149 (1992). Following
transfection, cells may be grown in 96-well microliter plates in complete HSFM
medium
(Life Technologies, Inc., Grand Island, NY) at 37 C, 5%CO2. The selection
process can
be initiated after two days by the addition of hygromycin selection medium
(Calbiochem, San Diego, CA) at a final concentration of 500 units/ml of
hygromycin..
Colonies typically emerge 2-3 weeks post-electroporation. The cultures can
then be
expanded for further analysis.
Transfectoma clones that are positive for the secretion of chimeric or
humanized
heavy chain can be identified by ELISA assay. Briefly, supernatant samples
(100 l)
from transfectoma cultures, are added in triplicate to ELISA microtiter plates
precoated
with goat anti-human (GAM)-IgG, F(ab')2 fragment-specific antibody (Jackson
ImmunoResearch, West Grove, PA). Plates are incubated for 1 h at room
temperature.
Unbound proteins are removed by washing three times with wash buffer (PBS,
containing 0.05% polysorbate 20). Horseradish peroxidase (HRP) conjugated GAH-
IgG, Fc fragment-specific antibodies (Jackson ImmunoResearch) are added to the
wells,.
(100 l of antibody stock diluted x 104, supplemented with the unconjugated
antibody to
a final concentration of 1.0 g/ml). Following an incubation of 1 h, the
plates are
washed, typically three times. A reaction solution, [100 l, containing 167 pg
of
orthophenylene-diamine (OPD) (Sigma, St. Louis, MO), 0.025% hydrogen peroxide
in
PBS], is added to the wells. Color is allowed to develop in the dark for 30
minutes. The
reaction is stopped by the addition of 50 pl of 4 N HCl solution into each
well before
measuring absorbance at 490 nm in an automated ELISA reader (Bio-Tek
instruments,
Winooski, VT). Bound chimeric antibodies are than determined relative to an
irrelevant
chimeric antibody standard (obtainable from Scotgen, Ltd., Edinburg,
Scotland).
Antibodies can be isolated from cell culture media as follows. Transfectoma
cultures are adapted to serum-free medium. For production of chimeric
antibody, cells
are grown as a 500 ml culture in roller bottles using HSFM. Cultures are
centrifuged
54


CA 02752553 2011-09-13

and the supernatant filtered through a 0. 2 membrane. The filtered medium is
passed
through a protein A column (1 x 3 cm) at a flow rate of Iml/min. The resin is
then
washed with about 10 column volumes of PBS and protein A-bound antibody is
eluted
from the column with 0.1 M glycine buffer (pH 3.5) containing 10 mM EDTA.
Fractions
of 1.0 ml are collected in tubes containing 10 l of 3 M Tris (pH 8.6), and
protein
concentrations determined from the absorbance at 280/260nm. Peak fractions are
pooled,
dialyzed against PBS, and the antibody concentrated, for example, with the
Centricon 30
(Amicon, Beverly, MA). The antibody concentration is determined by ELISA, as
before,
and its concentration adjusted to about 1 mg/ml using PBS. Sodium azide, 0.01%
(w/v),
is conveniently added to the sample as preservative.
The following are the nucleotide sequences of the primers used to prepare the
anti-CD20 antibodies:
hA20VKA
5'-CATCTCTGAG CGCATCTGTT GGAGATAGGG TCACTATGAC
TTGTAGGGCC AGCTCAAGTG TAAGTTACAT CCACTGGTTC CAGCAGAAAC
CAGGGAAAGC ACCTAAACCC TGGATTTATG-3'
(SEQ ID NO: 37)
hA20VKB
5'-GGTGTCCCTG TCCGATTCTC TGGCAGCGGA TCTGGGACAG
ATTACACTTT CACCATCAGC TCTCTTCAAC CAGAAGACAT TGCAACATAT
TATTGTCAGC AGTGGACTAG TAACCCACCC ACGTTCGGTG-3'
(SEQ ID NO: 38)
hA20VKA-Backward
5'-CAGCTGACCC AGTCTCCATC ATCTCTGAGC GCATCTGTTG-3'
(SEQ ID NO: 39)

hA2OVKA-Forward
5'-AGGTTCGAAG TGGCATAAAT CCAGGGTTTA GGTGCT-3'
(SEQ ID NO: 40)

hA20VKB Backward
5'-CACTTCGAAC CTGGCTTCTG GTGTCCCTGT CCGATTCTC-3'
(SEQ ID NO: 41)



CA 02752553 2011-09-13
hA20VKB Forward
5'-ACGTTAGATC TCCAGCTTGG TCCCTCCACC GAACGTGGGT GGGTTA-3'
(SEQ ID NO: 42)

hA20VHA
5'-CTGAAGTCAA GAAACCTGGG TCATCGGTGA AGGTCTCCTG
CAAGGCTTCT GGCTACACCT TTACTAGTTA CAATATGCAC TGGGTCAAGC
AGGCACCTGG ACAGGGTCTG GAATGGATTG (3-3' (SEQ ID NO: 43)
hA20VHB
5'-ATCAGAAGTT CAAGGGTAAA GCCACACTGA CTGCCGACGA
ATCCACCAAT ACAGCCTACA TGGAGCTGAG CAGCCTCAGG TCTGAGGACA
CGGCATTTTA TTACTGTGCA AGATCGACTT ACTACGGCGG TGACTGGTAC
TTCGATGTCT G-3' (SEQ ID NO: 44)

hA20VHA Backward
5'-CAGCTGCAGC AATCAGGGGC TGAAGTCAAG AAACCTGGG-3'
(SEQ ID NO: 45)

hA20VHA Forward
5'-TTCCGGGATA AATAGCTCCA ATCCATTCCAA GACCCTG-3'
(SEQ ID NO: 46)

hA20VHB Backward
5'-ATCCCGGAAA TGGTGATACT TCCTACAATC AGAAGTTCAA
GGGTAAAGCC A-3' (SEQ ID NO: 47)

hA20VHB Forward
5'-GGAGACGGTG ACCGTGGTGC CTTGGCCCCA GACATCGAAG TACCAG-3'
(SEQ ID NO: 48)

hA20VH2A
5'-CTGAAGTCAA GAAACCTGGG TCATCAGTGA AGGTCTCCTG
CAAGGCTTCT GGCTACACCT TTAGTAGTTA CAATATGCAC TGGGTCAGAC
AGGCACCTGG ACAGGGTCTG GAATGGATGG G-3' (SEQ ID NO: 49)

56


CA 02752553 2011-09-13
hA20VH2B
5'-ATCAGAAGTT CAAGGGTAGA GCCACAATAA CTGCCGACGA
ATCCACCAAT ACAGCCTACA TGGAGCTGAG CAGCCTGAGG
TCTGAGGACA CGGCATTTTA TTTTTGTGCA AGATCGACTT
ACTACGGCGG TGACTGGTAC TTCGATGTCT G-3' (SEQ ID NO: 50)
hA20VH2A Forward
5'-TTCCGGGATA AATAGCTCCC ATCCATTCCA GACCCTG-3'
(SEQ ID NO: 51)

hA20VH2B Backward
5'-ATCCCGGAAA TGGTGATACT TCCTACAATC AGAAGTTCAA
GGGTAGAGCC A-3' (SEQ ID NO: 52)

The invention is further described by reference to the following examples,
which
are provided for illustration only. The invention is not limited to the
examples but rather
includes all variations that are evident from the teachings provided herein.

EXAMPLES
Example 1. Construction of a humanized anti-CD20 antibody
The VH and Vic genes of A20, an anti-CD20 antibody, was obtained by RTPCR
using the primer pairsVH1BACK/VHIFOR and VKIBACKIVKIFOR, respectively
Orlandi et al., (Proc. Natl. Acad. Sci., USA, 86: 3833 (1989) ). Multiple
independent
clones were sequenced to eliminate possible errors resulting from the PCR
reaction. The
cloned murine VH and VK sequences as the final PCR product were designated
A2OVk
(FigurelA) and A20VH (Figure 1B), respectively. A chimeric A20 (cA20) antibody
was
constructed and expressed in Sp2/0 cell. The Vk and VH of sequences of cA20
are
shown in Figure 2. The cA20 antibody bound to Raji cell and competed with
radiolabeled A20 purified from the hybridoma cell culture supernatant (Figure
3). This
result confirmed the authenticity of the cloned V genes.
A single light chain and two heavy chain variable region sequences encoding
the
humanized anti-hCD20 (hA20) antibody were designed and constructed. Human REI
framework sequences were used for VK (FigurelA), and a combination of EU and
NEWM framework sequences were used for VH (Figure 1B). There are a number
57


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
of amino acid changes in each chain outside of the CDR regions when compared
to
the starting human antibody frameworks. The heavy chain of hA20, hA20VHI,
contains nine changes, while hA20VH2 contains three changes from the human EU
frameworks (Figure 4A). hA20VH2 is preferred because it contains more amino
acids
from the human antibody framework region than hA20VH1. The light chain of
hA20,
hA20Vx, contains seven amino acid changes from the REI framework (Figure 4B).
Example 2. Method of hA20 antibody construction
Each variable chain was constructed in two parts, a 5'- and 3'-half,
designated
as "A" and "B" respectively. Each half was produced by PCR amplification of a
single strand synthetic oligonucleotide template with two short flanking
primers,
using Taq polymerase. The amplified fragments were first cloned into the pCR4
TA
cloning vector from Invitrogen (Carlsbad, CA) and subjected to DNA sequencing.
The templates and primer pairs are listed as follows:
Template Primers
VKA VkA-Backward/VkA-Forward
VKB VkB-BackwardNkB-Forward
VHIA VHA-Backward/VH1A-Forward
VH1B VH1B-Backward/VHB-Forward
VH2A VHA-Backward/VH2A-Forward
VH2B VH7B-Backward/VHB-Forward
Light chain
For constructing the full-length DNA of the humanized Vic sequence, oligo
hA20VKA (120 mer) and hA2OVKB (130 mer) were synthesized on an automated.
RNA/DNA synthesizer (Applied Biosystems). hA20VKA and B represent the nt 26-
145 and 166-195, respectively, of the hA20 Vx. (See Fig. 5A) Oligo hA20VKA and
B were cleaved from the support and deprotected by treatment with concentrated
ammonium hydroxide. After samples were vacuum-tried and resuspended in 100 l
of
water, incomplete oligomers (less than 100-mer) were removed by centrifugation
through a ChormaSpin-100 column (Clontech, Palo Alto, CA). All flanking
primers
were prepared similarly, except ChromaSpin-30 columns were used to remove
synthesis by-products. 1 l of ChromaSpin column purified hA20VKA was PCR
amplified in a reaction volume of 100 l containing 10 l of l OX PCR buffer
[500 mM
58


CA 02752553 2011-09-13

WO 03/068821 ACT/GB03/00665
KCI, 100 mM Tris-HCI (pH 8.3),15 mM MgCl,, and 0.01% (w/v) gelatin] (Perkin
Elmer Cetus, Norwalk, CT), 250 M of each dNTP, 200 nM of VITA-Backward and
VkA-Forward, and 5 units of Taq DNA polymerise (Perkin Elmer Cetus). This
reaction
mixture was subjected to 30 cycles of PCR reaction consisting of denaturation
at 94 C
for 1 min, annealing at 50 ~C for 1.5 min, and polymerization at 72 'C for 1.5
min.
hA20VKB was PCR-amplified by the primer pair W13-Backward and W-B-Forward.
under similar condition. The amplified VKA and VKA fragments were purified on
2%
agarose (BioRad, Richmond, CA). Unique restriction sites were designed at the
ends
of each fragment to facilitate joining through DNA ligation. The amplified
VKA'
fragment contained a PvuII restriction site, CAGCTG, at its 5'-end and a BstBI
restriction site, TTCGAA, at the 3'-end. The amplified VKB fragment contained
a
BstBI restriction site at its 5'-end and a Bglll restriction site, AGATCT, at
the 3'-end.
Assembly of the full-length Vic chain was accomplished by restriction enzyme
digestion of each fragment with the appropriate 5'=- and 3'-enzymes and
ligation into
the VKpBR2 vector previously digested with Pvui and Boll (Bell digested end is
compatible with that of BgIII). The resulting ligated product contains the A
fragment
ligated to the Pvu1I site, the B fragment ligated to the Boll site, and the A
and B
fragments joined together at the BstBI site (Figure 5A). VKpBR2 is a modified
staging vector of VKpBR (Leung et al., Hybridoma, 13:469 (1994)), into which a
Xbal
restriction site was introduced at 14 bases upstream of the translation
initiation codon.
Upon confirmation of a correct open reading frame by DNA sequencing, the
intact
chain was removed from VKpBR2 as a Xbal to BamHI fragment and ligated into the
pdHL2 expression vector. The vector containing only Vic sequence was
designated as
hA2OVicpdHL2. pdHL2 contains the expression cassettes for both human IgGl Cl,
C2, C3, and hinge regions (Fig. 7A) and the human kappa chain Ck (Fig. 7B)
under
the control of IgH enhancer and MT, promoter, as well as a mouse dhfr gene,
controlled by a weak SV40 promotor, as a marker for selection of transfectants
and
co-amplification of the trans-genes (Gillies et al., J. bnmunol. Methods
125:191 (1989);
Losman et al., Cancer 80:2660 (1997)). By replacing the Vic and VH segments of
pdHL2, different chimeric or humanized Abs can be expressed.

59


CA 02752553 2011-09-13
Heavy chain
For the construction of hA20VH1, oligo VH1A (121 mer) and VH1B (151 mer),
representing the nt 23-143 and 179-329, respectively, (See Fig.5B) were
synthesized as
described above. Similarly, for hA20VH2, oligo VH2A and VH2B were prepared.
These
oligos were PCR-amplified by their respective primer pairs as listed in
Example 2. The
same construction method as done for VK was carried out for both types of VH,
with the
following modifications: the 5'-end restriction site of the A fragments was
Pstl
(CTGCAG) and the 3'-end restriction site of B fragments was BstEII (GGTCACC).
These fragments were joined together upon ligation into the VHpBS2 vector at a
common Neil site (CCCGG), resulting in full-length VH sequences (Figure 5B
and5C)
and confirmed by DNA sequencing. VHpBS2 is a modified staging vector of VHpBS
(Leung et al., Hybridoma, 13:469 (1994)), into which a XhoI restriction site
was
introduced at 16 bases upstream of the translation initiation codon. The
assembled VH
genes were subcloned as Xhol-BamHI restriction fragments into the expression
vector
containing the Vie sequence, hA20VxpdHL2. Since the heavy chain region of
pdHL2
lacks a BamHI restriction site, this ligation required use of the HNB linker
to provide a
bridge between the BamHI site of the variable chain and the HindIII site
present in the
pdHL2 vector. The resulting expression vectors were designated as hA20-lpdHL2
and
hA20-2pdHL2.
HNB linker 5'-AGCTTGCGGCCGC-3' (SEQ ID NO: 30)
3'-ACGCCGGCGCTAG-5' (SEQ ID NO:31)
Example 3. Transfection and Expression of hA20 Antibodies
Approximately 30 i.g of the expression vectors for hA20 were linearized by
digestion with Sall and transfected into Sp2/0-Ag14 cells by electroporation
(450V and
25 F). The transfected cells were plated into 96-well plates for 2 days and
then selected
for drug-resistance by adding MTX into the medium at a final concentration of
0.025
M. MTX-resistant colonies emerged in the wells 2-3 weeks. Supernatants from
colonies surviving selection were screened for human Ab secretion by ELISA
assay.
Briefly, 100 l supernatants were added into the wells of a microtiter plate
precoated
with GAH-IgG, F(ab')2 fragment-specific Ab and incubated for 1 h at room
temperature.
Unbound proteins were removed by washing three times with wash buffer (PBS
containing 0.05% polysorbate 20). HRP-conjugated GAH-IgG, Fc fragment-


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
specific Ab was added to the wells. Following an incubation of 1 h, the plate
was
washed. The bound HRP-conjugated Ab was revealed by reading A490nm after the
addition of a substrate solution containing 4 mM OPD and 0.04% Ii202. Positive
cell
clones were expanded and hA20-1 and hA20-2 were purified from cell culture
supernatant by affinity chromatography on a Protein A column.

Example 4. Binding Activity of Anti-CD20 Antibodies
A competition cell-binding assay was carried out to assess the
immunoreactivity of hA20 relative to the parent CAM and the anti-CD20 Ab c2B8.
A
constant amount of 125I-labeled murine A20 or c2B8 (100,000 cpm, -10 Ci/ g)
was
incubated with Raji cell in the presence of varying concentrations (0.2-700
nM) of
hA20-1, -2, murine A20, cA20, or c2B8 at 4 C for 1-2 h. Unbound Abs were
removed by washing the cells in PBS. The radioactivity associated with cells
was
determined after washing. As shown in Figure 6, both humanized A20 mAbs, hA20-
1
and hA20-2, exhibited comparable binding activities as A20, the murine anti-
CD20
MAb, CAN, and c2B8, a chimeric anti-CD20 MAb, when competing with binding of
125I-A20 or 125I-c2B8 to Raji cells.

By direct binding of radiolabeled Mabs to Raji cells and Scatchard plot
anlaysis, the dissociation constants were measured to be 2.9 and 4.2 nm for
cA20 and
hA20, respectively, in comparison to 3.9 nM for C2B8. In vitro crosslinking
experiments, using a goat anti- human IgG, Fc fragment specific antibody to
complex
with the antibodies showed similar killing of Raji NHL cells between cA20 and
hA20,
as well as C2B8.

Example 5. Treatment of a Patient with Relapsed Intermediate-Grade Non-
Hodgkin's Lymphoma
A patient with intermediate grade non-Hodgkin's lymphoma has failed prior
aggressive chemotherapy, consisting of CHOP x 6, which led to a complete
remission
for four months, another course of CHOP x 6, resulting in progression, D-MOPP
x 2,
resulting in stable disease for three months, and CVB with peripheral stem
cell
transplantation, which led to a partial remission for five months. The patient
presents
with recurrent lymphoma in a neck lymph node, measurable by computerized
tomography and palpation.

61


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
The patient is infused within 3 hrs with 450 mg of humanized CD20
monoclonal antibody A20 on days 0, 14, 28, and 42 with no serious adverse
effects
noted either during or immediately after the infusions. Eight weeks later,
palpation
of the neck node enlargement shows a measurable decrease of about 50%. Follow-
up
measurements made at twenty weeks post therapy show no evidence of the disease
in
the neck, and nowhere else, as confirmed by computed tomography studies of the
body. Since new disease is not detected elsewhere, the patient is considered
to be in
complete remission. Follow-up studies every 10-12 weeks confirms a complete
remission for at least ten months post therapy.

Example 6. Treatment of a patient with chronic idiopathic thrombocytopenia
purpura

A 45-year-old female with chronic idiopathic thrombocytopenia purpura has
been treated with prednisone, gamma globulins, and high dose dexamethasone,
but
the disease progresses. She undergoes splenectomy, which fails to stabilize
the
disease. Her platelet count falls to less than 30,000/microliter, and
hemorrhagic
events increase in frequency. The patient is then treated with the humanized
CD20
A20 MAb, 500 mg intravenously on the first week, followed by a dose of 250 mg
given once every other week for a total of 4 injections. Ten weeks after the
last dose
of A20 a marked increase in platelet number is observed, to
150,000/microliter, and
the hemorrhagic events disappear. Five months after the last antibody infusion
the
disease is in remission.

Example 7. Treatment of a patient with progressive rheumatoid arthritis
A 70 year old female, with severe progressive rheumatoid arthritis of the
finger joints, wrists, and elbows, has failed therapy with methotrexate, and
obtains
only minor relief when placed on Enbrel therapy.. The patient is then treated
with
A20 humanized CD20 MAb, 300 mg intravenously each week, for a period of four
weeks. After 3 months, a 40% improvement in measures of disease activity is
observed, which is maintained for 5 months. The patient is again treated with
A20, at
the same dose and frequency. The patient continues to improve, and 6 months
after
the second A20 MAb therapy, a 60% improvement is observed. No human anti-A20
antibodies are observed at any time during, or after the A20 therapy. Although
62


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
normal B-cells are depleted from the blood, no infectious complications, or
other
drug-related severe toxicity is observed.

Example 8. Treatment of a patient with myasthenia gravis
A 65 year old male has failed all conventional therapy for myasthenia gravis,
and is admitted to a neurological intensive therapy unit. The patient was
stabilized
by plasma exchange, and given intravenous immunoglobulin to reduce the titer
of
antiacetylcholine receptor antibody. The patient remained bedridden, and was
then
treated with A20 humanized CD20 MAb, 400 mg intravenously once every other.
week, for a period of ten weeks. One week after the last dose of A20, no blood
B-
cells were detectable, and a significant drop in the titer of the anti-
acetylcholine
antibody was observed. Four months after the last A20 MAb dose the patient was
mobile, and was released from the hospital.

Example 9. Treatment of a Dog with Aggressive Non-Hodgkin's B-cell
Lymphoma in Lymph Nodes and Bone Marrow
A 65-pound, 7-year old male Golden Retriever is diagnosed with diffuse large
cell aggressive lymphoma. The dog is placed on combination chemotherapy with
vincristine, cyclophosphamide, prednisolone, and doxorubicin, with good
response.
However, the dog subsequently is characterized as having progressive
lymphadenopathy, and seven months after this is found to have extensive
lymphoma
infiltration of bone marrow, extensive lymphoadenopathy of neck, chest,
abdomen,
pelvis, and hepatosplenomegaly.

The dog is given therapy with 1 F5 chimeric monoclonal antibody. The dog is
infused intravenously with 120 mg of 1F5 antibody, and the treatment is
repeated
weekly for 4 weeks following this initial treatment. Four months after the
final dose
of 1F5, a computerized tomography scan of the patient shows no evidence of
lymphoma, and all signs and symptoms of the disease were not evident.

Example 10. Treatment of -a Dog with Relapsed Intermediate-Grade Non-
Hodgkin's Lymphoma

A 78-pound, 9-year old, German Shepherd dog with intermediate grade non-
Hodgkin's lymphoma receives chemotherapy, which initially leads to a complete
63


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
remission for five months, followed by another course of chemotherapy which
results
in stable disease for six months. The dog then presents with recurrent
lymphoma in
the chest and in a neck lymph node, both measurable by computerized tomography
and palpation, respectively.

The patient is infused with a 90Y-labeled immunoconjugate of L243 (HLA-
DR) monoclonal antibody weekly for two weeks, at a radiation dose of 8 mCi in
50
mg of antibody protein, in combination with the A20 humanized CD20 antibody at
a
dose of 100 mg per each weekly infusion. Three weeks later, palpation of the
neck
node enlargement shows a measurable decrease, while a repeat computerized
tomography scan of the chest shows a marked reduction in tumor. Follow-up
measurements made at ten weeks post therapy show evidence of the disease in
the
neck or the chest being reduced by a about 60 percent. Since new disease is
not
detected elsewhere, the patient is considered to be in partial remission.
Follow-up
studies every 10-12 weeks confirms a partial remission for at least 7 months
post
therapy.

Example 11. Treatment of a Cat with Relapsed Lymphoma
A 10-pound, 12-year-old, domestic short hair presents with enlargement of a
single submandibular lymph node. After excision, there is recurrence of the
lesion at
6 months. The lesion is again excised, but then reappears 6 months later. The
cat is
then given weekly treatments for 4 weeks with an 131I-labeled immunoconjugate
of
anti-CD20 B 1 monoclonal antibody, at a radiation dose of 15 mCi in 45 mg
antibody
protein. The treatment is repeated 3 months later. When examined 3 months
after the
last treatment, a marked decrease can be palpated. No recurrence of the
disease is
observed for over one year.

Example 12. Evalulation of chimeric and humanized anti-CD20 Mabs in human
NHL cells in culture or xenografted in SCID mice

The properties of a chimeric (cA20) and humanized (hA20) CD20 antibody
was assessed in NHL cell lines. The results demonstrate that cA20 and hA20
behave
similarly to Rituximab, staining more than 99% of Raji, Ramos, RL, Daudi and
Su-
DHL-6 cells and reacting with approximately 5% of lymphocytes (expected % B_
cells). In all B-cell lines, specific growth inhibition was seen with the
Mabs, but the
64


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
level o f inhibition varied between the cell lines, with Su-DBL being the most
sensitive. In the absence of cross-linking, murine anti-CD20, cA20, hA20 and
rituximab all yielded between 77 and 90% inhibition. With cross-linking,
inhibition
of proliferataion ranged from 94-98%. Rituximab, cA20, and hA20 were also
similar
in their ability to induce apoptosis in Raji cells in the presence of a cross-
linking
second monoclonal antibody.
Also, SCID mice were injected intravenously with 2.5X106 Raji cells on day
0. Injections of murine, chimeric and humanized anti-CD20 antibodies, and the
cA20
F(ab')2 fragment were initiated on day-1 with 100 I]g/injection of intact
antibody, or
67 Eg/injection F(ab')2 fragment, five times per week for two weeks, the twice
weekly for three weeks. In one study, control mice died of disseminated
disease with
a median survivial time of 15 days post tumor innoculation, but median
survival was
extended to 38 days for cA20, 22.5 days for hA20, and 21 days for murine anti-
CD20
treated mice (all statistically significant by log-rank analysis (p< 0.005)).
In another
study, control mice died of disseminated disease manifested with CNS paralysis
with
a median survival time of 16.Sdays post tumor innoculation, but median
survival was
extended to 105days for cA20, 70 days for hA20, and 98 days for rituximab
treated
mice (all statistically significant extensions by log-rank analysis
(p<0.0001), Figure
11).

Example 13. Competitive cell surface binding assay.
Ag-binding specificity and affinity studies of humanized anti-CD20 Abs
(cA20, hA20, and c 1 F5), purified by affinity chromatography on a Protein A
column)
were evaluated by a cell surface competitive binding assay with murine 2B8 and
rituximab (IDEC Pharmaceuticals Corp., San Diego, CA) (Figure 8). Briefly, a
constant amount (100,000 cpm, -10 iCi/ g) of 125I-labeled (A) m2B8 or (B)
rituximab
was incubated with Raji cells in the presence of varying concentrations (0.2-
700 nM)
of competing Abs (cA20, hA20, m2B8, clF5, or rituximab) at 4 C for 1-2 h.
Unbound Abs were removed by washing the cells with PBS. Radioactivity
associated
with the cells was determined after washing. Figure 8 (A) is a comparison of
the
binding activities of cA20 (square), hA20-1 (triangle) and hA20-1 (circle)
with that of
m2B8 (diamond); Figure 8 (B) Compares the binding activities of cA20 (square),
c 1 F5 (triangle) and rituximab (diamond).



CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
In another study, the binding activities of hA20 with other anti-CD20 Abs,
rituximab and murine B 1 were compared by a cell surface competitive binding
assay
(Figure 9). Briefly, a constant amount (100,000 cpm, -10 iCi/ig) of 125I-
labeled
rituximab was incubated with Raji cells in the presence of varying
concentrations
(0.2-700 nM) of competing Abs, hA20 (triangle), mB 1 (Downward triangle) or
rituximab (square) at 4 C for 1-2 h. Unbound Abs were removed by washing the
cells
with PBS. Radioactivity associated with the cells was determined after
washing. The
IC50 values for these three Abs were calculated to be 6.8, 34, and 5,
respectively.
Example 14. Cytotoxic effect of crosslinked hA20 and other CD20 Abs on
cultured lymphoma cells.
Raji cells were treated with various CD20 Abs in the presence of a crosslinker
(an anti-human IgG, Fc fragment specific antibody;) to complex the CD20
antibodies
(Figure 10). A final concentration of 5 ig/ml of hA20, cA20, rituximab, or a
positive
control Ab, hLL1, was incubated with Raji cells, with 20 ig/ml of the
crosslinker
(red), without the crosslinker (orange), or with an anti-mouse IgG, Fc
fragment
specific antibody (blue) for 48 h. Total cell and viable cell populations were
measured
by (A) trypan blue staining and cell counting or (B) MIT assay (B). The data
show a
similar effect of hA2O and rituximab on Raji NHL cell survival, and that the
cytotoxic
effect is dependent on the specific crosslinking of the antibodies.

Example 15. In vivo therapy with hA20 and hLL2.
Raji cells wer administered i.v. to 60 SCID mice, at 2.5 x 106 cells/100
l/mouse (Figure 12). MAbs were administered i.p. on days 1 to 11, followed by
MAb injections twice per week, for approximately 3 weeks. The body weight of
the
animals was measured weekly until the study was terminated. The animals were
examined daily for paralysis of the hind legs. When paralysis occured, the
animals
were sacrificed and necropsied for visual inspection of disseminated tumor
nodules
(specifically in lungs, kidneys, and ovaries). Control mice treated with a
control
humanized IgGI Ab, hMN-14 (an anti-CEA antibody), died of disseminated disease
manifested with CNS paralysis. The median survival time was 13 days post tumor
i.v. inoculation. Median survival in the group treated with hA20 was extended
to
about 25 days. This value represents median survival increase of approximately
2 fold
66


CA 02752553 2011-09-13

WO 03/068821 PCT/GB03/00665
for hA20. Although the group treated with hLL2 alone showed the same median
survival time compared to the control mice, treatment with combination of hA20
and
hLL2 increased the median survival time of the mice to approximately 30 days.

67

Representative Drawing

Sorry, the representative drawing for patent document number 2752553 was not found.

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
(22) Filed 2003-02-14
(41) Open to Public Inspection 2003-08-21
Examination Requested 2011-09-13
Dead Application 2019-08-02

Abandonment History

Abandonment Date Reason Reinstatement Date
2018-08-02 R30(2) - Failure to Respond
2019-02-14 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Request for Examination $800.00 2011-09-13
Application Fee $400.00 2011-09-13
Maintenance Fee - Application - New Act 2 2005-02-14 $100.00 2011-09-13
Maintenance Fee - Application - New Act 3 2006-02-14 $100.00 2011-09-13
Maintenance Fee - Application - New Act 4 2007-02-14 $100.00 2011-09-13
Maintenance Fee - Application - New Act 5 2008-02-14 $200.00 2011-09-13
Maintenance Fee - Application - New Act 6 2009-02-16 $200.00 2011-09-13
Maintenance Fee - Application - New Act 7 2010-02-15 $200.00 2011-09-13
Maintenance Fee - Application - New Act 8 2011-02-14 $200.00 2011-09-13
Maintenance Fee - Application - New Act 9 2012-02-14 $200.00 2012-01-19
Maintenance Fee - Application - New Act 10 2013-02-14 $250.00 2013-01-21
Maintenance Fee - Application - New Act 11 2014-02-14 $250.00 2014-01-21
Maintenance Fee - Application - New Act 12 2015-02-16 $250.00 2015-01-21
Maintenance Fee - Application - New Act 13 2016-02-15 $250.00 2016-01-20
Maintenance Fee - Application - New Act 14 2017-02-14 $250.00 2017-01-19
Maintenance Fee - Application - New Act 15 2018-02-14 $450.00 2018-01-19
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
IMMUNOMEDICS, INC.
Past Owners on Record
None
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 2011-09-13 67 3,929
Claims 2011-09-13 7 288
Drawings 2011-09-13 19 508
Abstract 2011-09-13 1 9
Cover Page 2011-11-04 1 30
Abstract 2011-11-08 1 9
Claims 2015-04-08 7 286
Description 2016-04-06 67 3,928
Amendment 2017-06-28 18 793
Claims 2017-06-28 7 261
Examiner Requisition 2018-02-02 9 1,108
Correspondence 2011-10-03 1 39
Assignment 2011-09-13 6 167
Prosecution-Amendment 2013-12-10 1 42
Prosecution-Amendment 2014-10-10 3 235
Prosecution-Amendment 2015-04-08 9 394
Examiner Requisition 2015-10-09 4 277
Amendment 2016-04-06 3 156
Examiner Requisition 2016-12-28 4 257

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 :