Sélection de la langue

Search

Sommaire du brevet 2316089 

Énoncé de désistement de responsabilité concernant l'information provenant de tiers

Une partie des informations de ce site Web a été fournie par des sources externes. Le gouvernement du Canada n'assume aucune responsabilité concernant la précision, l'actualité ou la fiabilité des informations fournies par les sources externes. Les utilisateurs qui désirent employer cette information devraient consulter directement la source des informations. Le contenu fourni par les sources externes n'est pas assujetti aux exigences sur les langues officielles, la protection des renseignements personnels et l'accessibilité.

Disponibilité de l'Abrégé et des Revendications

L'apparition de différences dans le texte et l'image des Revendications et de l'Abrégé dépend du moment auquel le document est publié. Les textes des Revendications et de l'Abrégé sont affichés :

  • lorsque la demande peut être examinée par le public;
  • lorsque le brevet est émis (délivrance).
(12) Demande de brevet: (11) CA 2316089
(54) Titre français: NOUVELLES CELLULES ET NOUVELLES MOLECULES PARTICIPANT A LA REGULATION IMMUNITAIRE
(54) Titre anglais: NOVEL CELLS AND MOLECULES INVOLVED IN IMMUNE REGULATION
Statut: Réputée abandonnée et au-delà du délai pour le rétablissement - en attente de la réponse à l’avis de communication rejetée
Données bibliographiques
(51) Classification internationale des brevets (CIB):
  • C12N 5/0783 (2010.01)
  • A61K 31/7088 (2006.01)
  • A61K 35/17 (2015.01)
  • A61K 38/17 (2006.01)
  • A61K 39/395 (2006.01)
  • A61P 35/00 (2006.01)
  • A61P 37/06 (2006.01)
  • A61P 37/08 (2006.01)
  • C07K 14/725 (2006.01)
  • C07K 16/28 (2006.01)
  • C07K 19/00 (2006.01)
  • C12N 5/10 (2006.01)
  • C12N 15/12 (2006.01)
(72) Inventeurs :
  • ZHANG, LI (Canada)
  • ZHANG, ZHU XU (Canada)
  • YOUNG, KEVIN (Canada)
(73) Titulaires :
  • LI ZHANG
  • ZHU XU ZHANG
  • KEVIN YOUNG
(71) Demandeurs :
  • LI ZHANG (Canada)
  • ZHU XU ZHANG (Canada)
  • KEVIN YOUNG (Canada)
(74) Agent: SMART & BIGGAR LP
(74) Co-agent:
(45) Délivré:
(22) Date de dépôt: 2000-08-24
(41) Mise à la disponibilité du public: 2001-04-08
Licence disponible: S.O.
Cédé au domaine public: S.O.
(25) Langue des documents déposés: Anglais

Traité de coopération en matière de brevets (PCT): Non

(30) Données de priorité de la demande:
Numéro de la demande Pays / territoire Date
60/158,132 (Etats-Unis d'Amérique) 1999-10-08
60/226,573 (Etats-Unis d'Amérique) 2000-08-21

Abrégés

Abrégé anglais


Novel cells and molecules involved in immune regulation are
disclosed. The novel cells are regulatory T cells having the phenotype
CD3+.alpha..beta.-TcR+CD4-CD8-CD44-CD28-NK1.1-. The regulatory cells
express
high levels of Ly-6A and osteopontin while non-regulatory cells do not.
Inhibiting Ly-6A or osteopontin inhibits the suppressive properties of the
cells.

Revendications

Note : Les revendications sont présentées dans la langue officielle dans laquelle elles ont été soumises.


-81-
We Claim:
1. An isolated regulatory T cell having a phenotype
CD3+.alpha..beta.-TcR+CD4-CD8-CD44-CD28-NK1.1-.
2. An isolated regulatory T cell according to claim 1 having the
phenotype CD3+.alpha..beta.TCR+CD4-CD8-CD25+CD28-CD30+CD44-NK1.1-.
3. A use of a regulatory T cell according to claim 1 or 2 to suppress
an immune response.
4. A use of a regulatory T cell according to claim 1 or 2 to prevent
graft rejection.
5. A use of a regulatory T cell according to claim 1 or 2 to treat an
autoimmune disease.
6. A use of a regulatory T cell according to claim 1 or 2 to treat an
allergy.
7. A use of a regulatory T cell according to claim 1 or 2 to treat
cancer.
8. An antibody that binds to a cell according to claim 1 or 2.
9. A use of an antibody that stimulates or induces a regulatory T
cell according to claim 1 or 2 to suppress an immune response.
10. A use of an antibody that stimulates or induces a regulatory T
cell according to claim 1 or 2 to prevent graft rejection.

-82-
11. A use of an antibody that stimulates or induces a regulatory T
cell according to claim 1 or 2 to treat an autoimmune disease.
12. A use of an antibody that stimulates or induces a regulatory T
cell according to claim 1 or 2 to treat an allergy.
13. A use of an antibody that stimulates or induces a regulatory T
cell according to claim 1 or 2 to treat cancer.
14. A method of expanding a population of regulatory T cells
having the phenotype CD3+.alpha..beta.-TcR+CD4-CD8-CD44-CD28-NK1.1- in vitro
comprising:
(a) obtaining a sample comprising the regulatory T cells or
precursors thereof;
(b) stimulating the cells with antigen; and
(c) culturing the cells under conditions suitable for the
expansion of the regulatory T cells.
15. A method according to claim 14 wherein the sample is blood or
bone marrow.
16. A method according to claim 14 wherein the antigen is allogenic
lymphocytes mismatched at one MHC class I locus.
17. A method according to claim 14 wherein the antigen is an
autoantigen.
18. A method according to claim 14 wherein the antigen is an
allergen.

-83-
19. A method according to any one of claims 14 to 18 wherein the cells
are cultured in the presence of IL-2 and IL-4.
20. A method according to any one of claims 14 to 19 wherein the
sample is depleted of CD4+ and CD8+ T cells prior to step (b).
21. A use of a regulatory T cell prepared according to the method of
any one of claims 14 to 20 to suppress an immune response.
22. A use of a regulatory T cell prepared according to the method of
any one of claims 14 to 20 to prevent graft rejection.
23. A use of a regulatory T cell prepared according to the method of
any one of claims 14 to 20 to treat an autoimmune disease.
24. A use of a regulatory T cell prepared according to the method of
any one of claims 14 to 20 to treat an allergy.
25. A use of a regulatory T cell prepared according to the method of
any one of claims 14 to 20 to treat cancer.
26. A use of an Ly-6A protein or a nucleic acid encoding an Ly-6A
protein to suppress an immune response.
27. A use of an Ly-6A protein or a nucleic acid encoding an Ly-6A
protein to prevent graft rejection.
28. A use of an Ly-6A protein or a nucleic acid encoding an Ly-6A
protein to treat an autoimmune disease.
29. A use of an Ly-6A protein or a nucleic acid encoding an Ly-6A
protein to treat an allergy.

-84-
30. A use of an Ly-6A protein or a nucleic acid encoding an Ly-6A
protein to treat cancer.
31. A use according to any one of claims 26 to 30 wherein the Ly-6A
protein is a soluble fusion protein.
32. A use of an osteopontin protein or a nucleic acid encoding
osteopontin to suppress an immune response.
33. A use of an osteopontin protein or a nucleic acid encoding
osteopontin to prevent graft rejection.
34. A use of an osteopontin protein or a nucleic acid encoding
osteopontin to treat an autoimmune disease.
35. A use of an osteopontin protein or a nucleic acid encoding
osteopontin to treat an allergy.
36. A use of an osteopontin protein or a nucleic acid encoding
osteopontin to treat cancer.
37. A use according to any one of claims 26 to 30 wherein the
osteopontin protein is a soluble fusion protein.
38. A use of an agent that inhibits a regulatory cell according to claim
1 or 2, Ly-6A or osteopontin to enhance or induce an immune response.
39. A use according to claim 38 wherein the agent is an antibody that
binds the regulatory T cells.

-85-
40. A use according to claim 39 wherein the antibody induces
apoptosis of the regulatory T cells.
41. A use according to claim 38 wherein the agent is an antibody that
binds the Ly-6A protein.
42. A use according to claim 38 wherein the agent is an antisense
oligonucleotide that is complimentary to a nucleic acid sequence from an
Ly-6A gene.
43. A use according to claim 38 wherein the agent is an antisense
oligonucleotide that is complimentary to a nucleic acid sequence from an
osteopontin gene.
44. A use according to claim 38 wherein the agent inhibits the
interaction between the FasL on the regulatory T cells and Fas on target
cells.
45. A use according to claim 44 wherein the agent is soluble Fas.
46. A use according to claim 44 wherein the agent is a Fas-Fc fusion
protein.
47. A use according to claim 38 wherein the agent that inhibits the
regulatory cells is selected from cyclosporin A, IL-10, Anti-IFN.gamma.
antibodies
or anti-TCR antibodies.
48. A use of a regulatory T cell according to claim 1 or 2 to prevent or
treat graft verus host disease.

-86-
49. A use of an antibody that stimulates or induces a regulatory T cell
according to claim 1 or 2 to prevent or treat graft versus host disease.
50. A use of a regulatory T cell prepared according to the method of
any one of claims 14 to 20 to prevent or treat graft versus host disease.
51. A use of an Ly-6A protein or a nucleic acid encoding an Ly-6A
protein to prevent or treat graft versus host disease.
52. A use of an osteopontin protein or a nucleic acid encoding
osteopontin to prevent or treat graft versus host disease.

Description

Note : Les descriptions sont présentées dans la langue officielle dans laquelle elles ont été soumises.


CA 02316089 2000-08-24
-1-
B&P File No. 10723-14/MG
Title: Novel Cells and Molecules Involved in Immune Regulation
FIELD OF THE INVENTION
The present invention relates to methods and compositions for
modulating an immune response. The invention includes novel cells
and molecules that are useful in suppressing an immune response.
BACKGROUND OF THE INVENTION
Transplantation is the current therapeutic modality for patients
with end-stage organ failure. Joint efforts of clinicians and immunologists
have now made it possible for 80-95% of patients to live with a functional
allograft for one year post-transplantation. Nonetheless, it is important to
realize that despite vast improvements in 1 year graft survival rates,
long-term graft survival has remained relatively unchanged over the past
30 years (7, 8).
Long-term immunosuppression with drugs, such as cyclosporin, is
the most commonly used approach for enhancing allograft survival.
However, the ability of immunosuppressive drugs currently in use to
prevent long-term graft rejection is much less robust (8, 9). The advantage
of cyclosporin treatment diminishes beyond 1-year as evidenced by only a
small increase (1-year) in half-lives of graft survival (8). Even with
continuous immunosuppressive treatment, half of the patients receiving
renal transplants lose their grafts within 5 years (10). In addition, as
immunosuppressive drugs inhibit immune responses non-specifically,
their use results in a high incidence of infectious and malignant
complications in these recipients (11). Hence, the induction of
donor-specific tolerance in the absence of expensive, toxic and non-specific
immunosuppressive therapy has been a goal of clinicians for many years.
Numerous studies indicate that under appropriate conditions,
pre-exposure of an adult individual to a specific antigen (Ag) may lead to
tolerance to that Ag rather than immunization (12, 2, 3, 13-21).
Intravenous injection of lymphoid cells carrying specific transplantation
Ags identical to the graft donor, including donor specific transfusion (DST)

CA 02316089 2000-08-24
_2_
of blood or bone marrow, is one such approach used to induce tolerance to
alloantigens both in man and mice (13-17, 22-39). Although the overall
effect of random pre-transplantation transfusions on short-term (1 year or
less) graft survival remains controversial, especially after the use of
cyclosporin (40), the significant enhancement of long-term graft survival
by pre-transplant DST has been observed in many studies (22, 24-39).
Unlike immunosuppressive drugs that non-specifically inhibit immune
responses, DST reduces the specific immune response to the donor Ag
without impairing the immune response to other Ags, thereby avoiding
the complications of infection and malignancy generated by using
immunosuppressive drugs. Despite the fact that the beneficial effect of
DST on long-term allograft survival as a phenomenon has been observed
for nearly 30 years, the mechanisms whereby the introduction of donor
cells lead to tolerance are still not fully understood.
SUMMARY OF THE INVENTION
The present inventors have isolated novel regulatory T cells that
have many important roles in immune regulation including inducing
antigen specific tolerance, preventing graft versus host disease and treating
cancer. The phenotype of a novel regulatory T cells are CD3+a(3-TcR+CD4-
CD8-CD44-CD28-NKl.l-. The cells do not express IL-2, IL-4, IL-10 and IL-13
but do express IFN-y and TGF-[3 mRNA after activation. Treatment of the
regulatory T cells with IL -10, cyclosporin A, anti-IFN-y or anti-TCR
antibodies abrogates suppression by the cells.
Accordingly, the present invention provides an isolated regulatory
T cell having the phenotype CD3+a(3-TcR+CD4-CDS-CD44-CD28-NK1.1-.
More specifically, the regulatory T cells have the phenotype
CD3+a(3TCR+CD4-CD8-CD25+CD28-CD30+CD44-NK1.1-.
The present invention also includes methods for stimulating or
expanding the regulatory T cells both in vitro and in vivo.
The present invention also includes the use of the novel
regulatory cells to suppress an immune response. Accordingly, the present

CA 02316089 2000-08-24
-3-
invention provides a method of suppressing an immune response
comprising administering an effective amount of a regulatory T cell
having the phenotype CD3+a(3-TcR+CD4-CD8-CD44-CD28-NK1.1- to an
animal in need of such treatment.
In one embodiment, the present invention provides a method of
inducing immune tolerance in a recipient animal comprising
administering an effective amount of a regulatory T cell having the
phenotype CD3+a(3-TcR+CD4-CD8'CD44-CD28-NK1.1- to an animal in need
of such treatment. In a particular embodiment, the novel regulatory cells
may be used to prevent graft rejection.
In another embodiment, the present invention provides a method
of preventing or treating graft versus host disease comprising
administering an effective amount of a regulatory T cell having the
phenotype CD3+a(3-TcR+CD4-CD8'CD44-CD28-NK1.1- to an animal in need
of such treatment.
In a further embodiment, the present invention provides a
method of preventing or treating an autoimmune disease comprising
administering an effective amount of a regulatory T cell having the
phenotype CD3+a(3-TcR+CD4-CD8-CD44-CD28-NK1.1- to an animal in need
of such treatment.
The present invention also includes the use of the novel
regulatory cells to treat cancer. Accordingly, the present invention
provides a method of preventing or treating cancer comprising
administering an effective amount of a regulatory T cell having the
phenotype CD3+a[3-TcR+CD4-CD8-CD44-CD28-NK1.1- to an animal in need
of such treatment.
The present invention also includes antibodies to the novel
regulatory cells of the invention and the use of the antibodies in
stimulating or inhibiting the regulatory T cells. As such, the antibodies are
useful in therapies to suppress or enhance an immune response.

CA 02316089 2000-08-24
-4-
The present inventors have also found that the proteins Ly-6A
and osteopontin are highly expressed on the above described regulatory T
cells but not on non-regulatory cells. Further, they have also shown that
blocking Ly-6A or osteopontin abolishes suppression by these cells.
Accordingly, the present invention provides a method of
suppressing an immune response comprising administering an effective
amount of an Ly-6A protein, a nucleic acid sequence encoding an Ly-6A
protein, osteopontin or a nucleic acid sequence encoding osteopontin to an
animal in need of such treatment.
In one embodiment, the present invention provides a method of
inducing immune tolerance in a recipient animal comprising
administering an effective amount of an Ly-6A protein, a nucleic acid
sequence encoding an Ly-6A protein, osteopontin or a nucleic acid
sequence encoding osteopontin to the recipient animal. In a particular
embodiment, the Ly-6A or osteopontin may be used to prevent graft
rejection.
In another embodiment, the present invention provides a method
of preventing or treating graft versus host disease comprising
administering an effective amount of an Ly-6A protein, a nucleic acid
sequence encoding an Ly-6A protein, osteopontin or a nucleic acid
sequence encoding osteopontin to an animal.
In a further embodiment, the present invention provides a
method of preventing or treating an autoimmune disease comprising
administering an effective amount of an Ly-6A protein, a nucleic acid
sequence encoding an Ly-6A protein, osteopontin or a nucleic acid
sequence encoding osteopontin to an animal having, suspected of having,
or susceptible to having an autoimmune disease.
In yet another embodiment the present invention provides a
method of preventing or treating cancer comprising administering an
effective amount of an Ly-6A protein, a nucleic acid sequence encoding an
Ly-6A protein, osteopontin or a nucleic acid sequence encoding
osteopontin to an animal.

CA 02316089 2000-08-24
-5-
The invention also includes pharmaceutical compositions
containing the novel regulatory cells, antibodies to the novel cells, Ly-6A
proteins, nucleic acids encoding Ly-6A protein, osteopontin or a nucleic
acid sequence encoding osteopontin for use in inducing tolerance in
transplantation or autoimmune disease or in treating cancer.
As stated above, the novel regulatory cells and the proteins Ly-6A
and osteopontin are associated with immune suppression. Consequently,
inhibiting the regulatory cells or proteins may also be useful in preventing
immune suppression.
Therefore, in another aspect, the present invention provides a
method of preventing immune suppression comprising administering an
effective amount of an agent that inhibits a regulatory T cell having the
phenotype CD3+a[3-TcR+CD4-CD8-CD44-CD28-NK1.1- or Ly-6A or
osteopontin to an animal in need thereof. In a preferred embodiment, the
agent is an antibody that binds the regulatory cells or an antibody that
binds Ly-6A or osteopontin or an antisense oligonucleotide that inhibits
the expression of Ly-6A or osteopontin.
The invention also includes pharmaceutical compositions
containing the above inhibitors for use in inducing or augmenting an
immune response.
Other features and advantages of the present invention will
become apparent from the following detailed description. It should be
understood, however, that the detailed description and the specific
examples while indicating preferred embodiments of the invention are
given by way of illustration only, since various changes and modifications
within the spirit and scope of the invention will become apparent to those
skilled in the art from this detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS
The invention will now be described in relation to the drawings in
which:
Figure 1(a) is a schematic illustration of the 2C T cell.

CA 02316089 2000-08-24
-6-
Figure 1(b) is a schematic illustration of breeding 2CF1 and 2CF1-Tg-
animals.
Figure 1(c) is a schematic illustration of the experimental model.
Figure 2: (a) Spleen cells from DST-treated tolerant mice
specifically enhance Ld+ skin allograft survival when transferred to naive
syngeneic mice. (2Cxdm2)Fl mice were given DST from (B6xBALB/c)Fl or
left as controls. One week later, all of the mice were given skin grafts from
both (B6xBALB/c)F1 and C3H. At three weeks after skin grafting, all
DST-treated mice accepted (B6xBALB/c)Fl grafts but rejected C3H grafts
(tolerant), whereas non-DST-treated mice rejected both grafts
(non-tolerant). (B6xdm2)F1 naive mice were adoptively transferred with
viable splenocytes (5x10~/mouse) collected from either tolerant (t, n=8) or
non-tolerant ( ~, n=5) mice. The next day, each mouse received skin grafts
from both (B6xBALB/c)F1 (dash lines) and C3H (solid lines) mice.
Survival of the skin grafts was monitored and scored for 120 days.
(b) Inhibition of anti-Ld MLR by 1B2+DN T cells. 1B2+CD8+ cells (1x103
cells/well) from naive (2Cxdm2)Fl mice were used as responder cells and
stimulated by 3x105 cells/well irradiated (20 Gy) splenocytes from
(B6xBALB/c)Fl mice. Purified 1B2+CD8+ (white), 1B2+CD4+ (grey) or
1B2+DN (black) cells from tolerant mice 120 days after skin grafting were
added into the MLR cultures as putative regulatory cells at various ratios
as indicated. Cells were cultured in aMEM supplemented with 10% FCS,
U/ml of rIL-2 and rIL-4. Proliferation was measured by [3H]-TdR
incorporation. The cultures to which no putative regulatory cells were
25 added were used as controls. The results represent 3 independent
experiments each with 5 replicates.
Figure 3: Dose dependent inhibition of syngeneic CD8+ T cells i n
vitro by DN T cells.
(a) 1B2+DN T cell clones are able to inhibit proliferation of CD8+ T cells
30 carrying the same TCR specificity. Purified naive CD8+ T cells from
(2Cxdm2)Fl mice were used as responders (1x103/well) and stimulated by

CA 02316089 2000-08-24
irradiated (B6xBALB/c)F1 splenocytes. Varying numbers of 1B2+DN clones
generated from tolerant (TN02, ~ and TN12, ~) and naive (CN04, X) mice
were added to the MLR cultures as putative regulatory cells, and a
1B2+CD8+ T cell clone (C02, 0) was used as a control. Cell proliferation was
measured by [3H]-TdR incorporation. The data are expressed as percent
inhibition of proliferation compared to the controls to which no putative
regulatory T cells were added. The experiments were repeated 4 times and
the results represent the 5 other DN clones and 2 other 1B2+CD8+ T cell
clones (not shown).
(b) Inhibition of 1B2+CD8+ T cell mediated cytotoxicity by 1B2+DN cells.
Varying numbers (as indicated) of 1B2+CD8+ cells from the spleen of naive
(2Cxdm2)Fl mice were used as effector cells and stimulated by irradiated
Ld+ spleen cells from (B6xBALB/c)Fl mice. 1B2+DN T cell clone (TN12)
were added at a 5:1 ratio 24 hours later. Killing of 104 P815 target cells per
well by 1B2+CD8+ T cells (~), 1B2+DN T cell clones (~), and 1B2+CD8+ plus
1B2+DN T cells (~) was measured by using standard 5lCr release assay at 90
hours after the addition of the DN T cell clones. The data are expressed as
percent specific killing of P815 target cells, and represents 6 replicates.
(c) Specific prolongation of Ld+ skin allograft survival in non-transgenic
mice after infusion of 1B2+DN T cells. Naive (B6xdm2)Fl mice were
infused with either 1x10 (dotted line, n=6) or 2x10 (solid line, n=6) cells
from a 1B2+DN clone TN12. Three mice were injected with 2x10 cells
from a 1B2+CD8+ clone C02 (dashed line) as controls. Each mouse was
given skin grafts from both (B6xBALB/c)Fl and C3H (not shown) 1 day
after injection of T cell clones. Skin graft survival was monitored for 120
days.
(d) Suppression of syngeneic CD8+ T cells by CD3+DN T cells in normal
mice.
CD8+ and DN T cells were purified from spleens of (B6xdm2)Fl (~) and
MRL/+ (~, H-2K) mice. (B6xdm2) T cells were stimulated with irradiated
C3H splenocytes and MRL/+ T cells were stimulated with irradiated

CA 02316089 2000-08-24
_g_
(B6xBALB/c)F1 splenocytes. CD8+ T cells were used as responders
(2,000/well), and varying numbers of syngeneic DN T cells were added to
the corresponding MLR cultures as putative regulatory cells. Cell
proliferation was measured by [3H]-TdR incorporation. The data are
expressed as percent inhibition of proliferation compared to the controls to
which no DN T cells were added. The data represent 2 experiments each
with 5 replicate cultures.
Figure 4: (a) Regulatory T cells express a unique combination of
cell surface markers. Regulatory and non-regulatory T cell clones (i-vi) as
well as fresh spleen cells from tolerant mice 120 days after skin grafting
(vii-ix) were stained with mAbs specific for the a(3-TCR (1B2), y8-TCR, CD4,
CDB, CD25, CD28, CD30, CD44, CD45, CD62L, CD69, LFA-1, and NK1.1. The
relative levels of expression of 1B2 and CDB, CD25, CD28, CD30, and CD44
on DN regulatory T cells (gray) and 1B2+CD8+ non-regulatory T cells
(white) are shown. The negative control is shown in Black (iii). Data
represent 8 regulatory, 3 non-regulatory T cell clones and spleen T cells
from 5 tolerant animals. The observed pattern of expression remained the
same before and after stimulation.
(b) Expression of cytokines, Fas and Fast mRNA in 1B2+DN and 1B2+CD8+
T cells. 1B2+CD8+ clone (C02, lane 1), 1B2+DN clones (TN12, TN01, TN11
lanes 2-4, respectively) and purified splenic 1B2+CD8+ T cells (lane 5) were
activated by irradiated Ld+ spleen cells. At 4, 10, 20 and 90 hrs after
activation, RNA samples were collected from viable cells.
Semi-quantitative analysis of the mRNA levels of IL-2, IL-4, IL-10, IL-13,
IFN-y, TNF-a, TGF-(3, Fas, Fast and GAPDH was performed by RT-PCR.
Results shown here are cells obtained 10 hrs after activation. Similar
results were obtained for other time points (not shown).
(c) Direct cell contact is required for DN T cell-mediated suppression.
Naive 1B2+CD8+ cells (4x103 cells/well) were stimulated with irradiated Ld+
spleen cells (1.2x106 cells/well) in a 24-well tissue culture plate to which
4x104 cells/well 1B2+DN T cell clones were added (left panels). In addition,

CA 02316089 2000-08-24
-9-
the anti-Ld MLR were set up in a transwell culture system (right panel).
The data are shown as percent inhibition of proliferation compared with
controls to which no DN clones were added. Open and solid bars represent
two different DN clones (TN12 and TN02) tested. The results represent 2
independent experiments each with 5 replicate cultures.
Figure 5: DN T cells induce suppression by killing activated CD8+
T cells through the Fas/FasL pathway.
(a) Regulatory and non-regulatory T cell clones mediate cytotoxicity
through different pathways. 1B2+CD8+ or 1B2+DN T cells were stimulated
independently by Ld+ spleen cells as described in the methods and used as
effector cells. Con A activated (B6x2C)Fl syngeneic (Ld-, Fas+, hatched bars)
and (B6xBALB/c)Fl allogeneic (Ld+, Fas+, open bars) T cells (>95% CD8+) as
well as allogeneic P815 tumour cells (Ld+, Fas-, solid bars) were labeled with
either 0.lmCi/ml of 5lCr for 1 hr or 10~,Ci/ml of [3H]-TdR overnight and
used as target cells. After co-culture with the effector cells at 37°C
for 4 hrs
at E:T 30:1 for perform-dependant killing or 18 hours at E:T 10:1 for
Fas-dependant cytotoxicity, % specific lysis of target cells was calculated.
The data are expressed as mean percent killing of 5 replicate cultures from
3 independent experiments.
(b) Fas Fc fusion protein (Fas-Fc) blocks DN T cell-mediated cytotoxicity.
DN T cell clones (TN12, open bars and CN04, solid bars) were activated by
Ld+ splenocytes and used as effector cells. 1B2+CD8+ T cells from
(2Cxdm2)Fl mice were stimulated by (B6xBALB/c)Fl spleen cells. Activated
1B2+CD8+ T cells were collected, labeled with 10~,Ci/ml of [3H]-TdR at
37°C
overnight, and used as targets at 10,000/well. Fresh irradiated
(B6xBALB/c)F1 splenocytes were added during the cytotoxicity assay. Lysis
of [3H]-TdR-labeled primary activated 1B2+CD8+Fas+ targets was measured
at E:T ration 5:1 in the presence of varying doses of Fas-Fc as indicated.
JAM assay was used to determine Fas-dependant cytotoxicity. Mean
percent inhibition of specific lysis of 1B2+CD8+Fas+ target cells compared to

CA 02316089 2000-08-24
-10-
killing detected in the absence of Fas-Fc is shown. The data represent 3
replicate cultures from 2 independent experiments.
(c) DN T cells are able to lyse activated CD8+ cells from normal, but not
Fas-mutant lpr mice. Primary CD8+ T cells were purified from the spleen
of MRL/+ (p), MRL/lpr (D), (B6xdm2)Fl (n), and B6/lpr (~) mice. Primary
a(3-TCR+CD3+DN T cells were purified from both (B6xdm2)F1 (dash lines)
and MRL/lpr mice (solid lines). The CD8+ and DN T cells from
(B6xdm2)Fl and B6/Ipr mice were stimulated by irradiated (20 Gy) C3H
(H-2K) splenocytes, and CD8+ and DN T cells from MRL/+ and MRL/lpr
mice were stimulated by irradiated (B6xBALB/c)F1 cells. Activated
CD3+DN T cells were collected and used as effector cells at E:T ratios as
indicated. Fresh irradiated (B6xBALB/c)Fl splenocytes were added during
the cytotoxicity assay. Specific lysis of target cells was determined as
described in the methods. The data are expressed as mean percent killing
of 3 replicate cultures from 2 independent experiments.
(d) DN T cells do not induce bystander killing in activated CD8+ T cells
that express a different TCR specificity. Spleen cells from mice expressing
a(3TCR-transgene specific for the LCMV-gp were stimulated in vitro with
peptide p33 (peptide p33-41, a gift from Dr. P. Ohashi). Female T3.70+CD8+
anti-male HY transgenic T cells (anti-HY) were stimulated with irradiated
male B6 spleen cells. C3H (H-2K) T cells were stimulated with irradiated
SJL (H-2S) spleen cells. 1B2+CD8+ T cells were stimulated by irradiated
(B6xBALB/c)Fl spleen cells. Activated CD8+ T cells from the above cultures
were collected, labeled with lO~Ci/ml of [3H]-TdR at 37°C overnight,
and
used as targets. 1B2+DN T cells were stimulated independently by
(B6xBALB/c)Fl spleen cells, then co-cultured with various target cells at
37°C for 18 hrs at varying E:T ratios as indicated. Fresh, appropriate,
irradiated spleen cells were added during cytotoxicity assays. Percentage
specific lysis of anti-LCMV p33 (black), anti-HY (hatched), anti-H-2S (gray),
and anti-Ld (white) target cells was determined. The data are expressed as

CA 02316089 2000-08-24
-11-
mean percent killing of 3 replicate cultures from 2-3 independent
experiments.
Figure 6: TCR -Ld interaction is critical for DN T cell-mediated
suppression.
(a) Kinetics of Ld expression on the surface of 1B2+DN and 1B2+CD8+ T cells
after stimulation with Ld+ spleen cells. Primary 1B2+CD8+ T cells and
1B2+DN clones were cultured separately with irradiated (B6xBALB/c)Fi
splenocytes in the presence of IL-2 and IL-4 (30 U/ml). At various time
points after culture, cells were harvested and triple stained with mAbs 1B2,
anti-CD8 and anti-Ld (30-5-7s). The expression of Ld on 1B2+ and 1B2+CD8+
T cells was analyzed by using a flow cytometer. Changes in Ld mean
fluorescence intensity (MFI) over time after activation of 1B2+DN (dash
line) and 1B2+CD8+ T cells (solid line) are shown. The results represent 2
independent experiments.
(b) Histograms show Ld expression on 1B2+DN (left) and 1B2+CD8+ (right)
T cells after 2 days stimulation by Ld+ spleen cells (gray). Markers indicate
the percentage of cells that express Ld in each subset of cells compared to
the corresponding controls (white).
(c) Blocking either Ld on DN T cells or 1B2 on CD8+ T cells abrogates DN T
cell-mediated cytotoxicity. 1B2+DN T cells were purified following 2 days
of stimulation with irradiated Ld+ splenocytes, and used as effector cells
(1x104/well). 1B2+CD8+ T cells were activated with irradiated Ld+
splenocytes for 4 days, labeled for 12 hours with [3H]-TdR, and used as
target cells (1x103/well). The DN cells were either left untreated (solid
line) or preincubated with anti-Ld mAb (20~.g/ml, dashed line) for 1 hour
before addition to the cultures. Labeled CD8+ T cells were either left
untreated (~), or preincubated with 1B2 (~t) or irrelevant isotype matched
control (8) mAb (100~g/ml) for 1 hour and then washed prior to being
used as targets. The effector and target cells were co-cultured for 18 hours
in the presence of rIL-2 and rIL-4 (30u/ml) and specific lysis of target cells

CA 02316089 2000-08-24
-12-
was determined using the JAM assay. The results represent 5 replicate
cultures.
Figure 7 is a schematic diagram showing 1B2+DN T cell
suppression of 1B2+CD8+ T cells.
Figure 8: Ly-6A is important for DN T cell mediated suppression.
(a) High expression of Ly-6A on the surface of DN regulatory but not non-
regulatory T cells. TN12 DN cells, non-regulatory CD8+ C02 cells, and the
TN12 DN T cells which were incubated with IL-10 and lost suppressive
function as described in Figure 15a were stained with anti-Ly-6A mAb,
followed by goat-anti-mouse-FITC secondary antibody. Data were analyzed
by flow cytometer.
(b) Pre-treatment with Ly-6A antisense oligo nucleotide reverse TN12 DN
T cell clone mediated suppression. The nucleotide (5'-
AGTGTGAGAAGTGTCCAT-3'), which is an antisense sequence to Ly-6A
mRNA, was use to inhibit Ly-6A expression. TN12 DN regulatory T cell
clones were pretreated with 5mM antisense oligo nucleotide at 37°C for
24
hours or left untreated as control. The anti-Ld MLR was set up to which
untreated and pretreated TN12 cells were added at various ratios as
putative regulatory cells. Cell proliferation was measured by [3H]-TdR
incorporation. The data are expressed as percent inhibition of
proliferation compared to the control to which no putative regulatory cells
were added.
(c) Ly-6A knockout DN T cells fail to kill activated syngeneic CD8+ T cells.
DN and CD8+ T cells were purified from the spleen of normal B6 (open
bars) and B6-Ly-6A knockout (closed bars) mice (both are H-2b), and
stimulated by irradiated splenocytes from BALB/c mice (H-2d) separately.
Activated CD8+ T cells from B6 or B6-Ly-6A knockout mice were labelled
with 10~,Ci/ml of [3H]-TdR and used as targets. After co-culture with the
activated DN T cells, specific lysis of the CD8+ target cells by DN T cells
was
calculated using the equation: % Specific killing = (S-E)/S x 100, where E

CA 02316089 2000-08-24
-13-
(experimental) is cpm of retained DNA in the presence of DN effector cells,
and S (spontaneous) is cpm of retained DNA in the absence of DN T cells.
Figure 9: (a) Expansion of donor-derived DN T cells and
elimination of anti-host CD8+ T cells following infusion of anti-Ld spleen
cells. ScidFl mice (Ld+) were injected with either 3x10 spleen cells from
anti-Ld 2CF1 mice alone (left) or in combination with 105 A20 lymphoma
cells (right). Mice were sacrificed at various time points as indicated, and
the total number of 1B2+DN and 1B2+CD8+ T cells in the spleen of the
recipients was monitored by FACS.
(b) Elimination of lymphoma cells by ScidFl mice after infusion of one-
class I locus mismatched allogeneic lymphocytes. ScidFl mice were either
infused with 105 A20 tumor cells along with 3x10 spleen cells from 2CF1
mice (n=12) or A20 alone (n=11). Survival of the recipient mice was
monitored for over 150 days.
(c) No GVHD after infusion of one-class I locus mismatched allogeneic
lymphocytes. ScidFl mice were infused with 3x10 allogeneic spleen cells
from either 2CF1 (n=42) or B6 (n=5) mice. Survival and body weight of the
recipients was monitored for more than 150 days.
Figure 10: Prevention of death of lymphoma without causing
GVHD by fusion of one-class I locus mismatched allogeneic lymphocytes.
(a) (B6xBALB/c)Fl mice were lethally irradiated (8.5 Gy) and reconstituted
with 4x10 splenocytes from (B6xdm2)Fl mice, together or in the absence of
A20 lymphoma cells. As controls, (B6xBALB/c)Fl mice were infused with
A20 alone, or lethally irradiated without reconstitution. Survival and
body weight were monitored for more than 150 days.
(b) (B6xBALB/c)Fl mice were lethally irradiated (8.5 Gy) and reconstituted
with 4x10 splenocytes either from B6 (semi-allogeneic, left panel) or 2CF1
(Ld mismatched, right panel) mice. Liver histology is shown at 100 days
after infusion of allogeneic cells.
Figure 11: Reconstituted mice obtained immunity to other
alloantigens. ScidFl mice were reconstituted with 2CF1 spleen cells or left

CA 02316089 2000-08-24
-14-
as controls. 3 weeks later both groups were given skin grafts from
syngeneic (B6xBALB/c)Fl, or third-party allogeneic (B6xC3H)Fl mice. Skin
graft survival was monitored.
Figure 12: Infusion of in vitro generated DN regulatory T cells
leads to elimination of lymphoma without causing GVHD.
(a) DN regulatory T cells prevent death caused by lymphoma.
(B6xBALB/c)F1 mice were injected i.v. with 5x106 L12.2 or CN04 DN
regulatory T cells together with a lethal dose (105 cells/mouse) of A20 B
lymphoma cells. As a control, (B6xBALB/c~1 mice were infused with 105
A20 cells alone. Survival and general health of the recipients was
monitored.
(b) Gross picture of two mouse livers: (B6xBALB/c)Fl mouse was injected
with 105 A20 B lymphoma cells. Four weeks after tumor inoculation, the
liver was harvested (left). The liver is greatly enlarged, has a bulging
surface and rounded edges. Note also it has a diffuse pale fan appearance
instead of the normal deep brown color. The liver on the right panel is
from another (B6xBALB/c)F1 mouse that received the same number of
A20 cells plus 5x106 DN regulatory T cells. It is normal in size, shape and
color.
c) Histopathology of livers of (B6xBALB/c)Fl mice (Hematoxylin and eosin
stain x300). (B6xBALB/c)Fl mice were injected with 105 A20 B lymphoma
cells in the absence (left panel) or presence (right panel) of 5x106 DN
regulatory T cells. Four weeks after tumor inoculation, the livers were
harvested and samples were stained with Hematoxylin and eosin. The left
panel shows a typical large cell lymphoma. There is very diffuse
infiltration by large malignant tumor cells which completely replaces the
normal liver architecture. Most of the tumor cells are mononuclear,
pleomorphic and have irregular hyperchromatic nuclei. Many of the
smaller irregular cells are undergoing necrosis. Mitoses and atypical
mitoses are frequent. Occasional tumor giant cells are present. The right
panel shows that the liver histology is normal. The hepatocytes, liver cell

CA 02316089 2000-08-24
-15-
cords, portal and venous structures are all normal. There is neither
evidence of malignancy nor graft versus host disease.
Figure 13:
(a) Specific recognition of cell surface molecules on DN regulatory T cells
by the monoclonal antibodies (mAbs). The DN regulatory T cell clone
TN12.2 and CD8+ non-regulatory T cell clone (C02) were stained with the
mAb P3C2/G1, followed by an anti-rat secondary PE labelled mAb. Cells
were analyzed using a flow cytometer. The data shown is representative of
25 monoclonal antibodies that the inventors have generated so far.
(b) mAb generated against DN T cell clones can also specifically bind to
primary activated DN regulatory T cells. DN regulatory T cells and CD8+ T
cells purified from the spleen of B6 mice were activated and stained with
mAb P3C2/G1, and analyzed as described in (a).
(c) Induction of death in apoptosis-resistant DN regulatory T cells.
Apoptosis-resistant TN12 DN regulatory T cells were incubated for various
time periods with P3C2/G1 mAb. After incubation, the DN T cells were
stained using Annexin V, a marker for early apoptosis and analyzed using
a flow cytometer. Data shown is at 20 hours after incubation with the
monoclonal antibody.
(d) mAbs are able to reverse suppression of 1B2+CD8+ T cells by DN T cells.
Suppression assays were set up in 96 well plates as described in Figure 2(b).
The DN T cell clone TN12.2 was used as suppressor cells, and 1B2+CD8+ T
cells from the spleen of (2Cxdm2)Fl mice were used as responders. In
addition, 100u1 of supernatant from P3C2/A2 or control P3G3/D1 mAb
hybridomas was added to each well. As controls, supernatant was added to
DN T cells clones in the absence of 1B2+CD8+ responder cells. Proliferation
was measured by 3H-[TdR] incorporation. Data shown is the proliferation
of 1B2+CD8+ responder and 1B2+DN suppressor T cells above the control
DN T cells incubated in the absence of responder cells.
Figure 14: (a) In vitro generated DN regulatory T cell lines are
resistant to TCR cross-linking induced apoptosis. 1B2+DN T cell clones
(TN02, TN12 and CN04), 1B2+CDS+ T cell clone (T01) and primary activated

CA 02316089 2000-08-24
-16-
1B2+CD8+ T cells (2CF1) were cross-linked with plate-bond 1B2 mAb (65
~,g/ml). At various time points after TCR-cross-linking, the percentage of
dead cells in each culture was determined by trypan blue staining.
(b) IL-10 increases susceptibility of DN T cells to apoptosis. TN12 DN T
cells were cross-linked in the absence (open bars) or presence of IL-10 (100
ng/ml, black bars). Percentage cell death was determine as described in (a).
Figure 15: IL-10, CsA, anti-TCR and anti-IFN~y antibodies abrogate
DN regulatory T cell mediated suppression.
(a) TN12 DN regulatory T cells were pre-incubated with 100ng/ml rIL-10
for 4 days and their ability to suppress anti-Ld MLR was compared with
non-IL-10 treated and 1B2+CD8+ non-regulatory T cells.
(b) Cyclosporin A, anti-TCR and anti-IFN-y mAbs abrogate suppressive
function of DN T cells. 1B2+DN regulatory TN12 cells were co-cultured
with 1B2+CD8+ T cells at 10:1 ratio in the presence of IL-2, IL-4 and
irradiated Ld+ splenocytes for 3.5 days in the absence (control) or presence
of one of the following reagents: CsA, 1B2 mAb or anti-IFN-'y mAb at the
concentrations as indicated.
Figure 16: Expression and function of Osteopontin (OPN) in DN
regulatory T cells.
(a) High expression of osteopontin mRNA on regulatory T cells. The
expression of osteopontin mRNA in DN regulatory T cells (TN12), IL-10-
treated TN12 cells which lost suppressive function (TN12-IL-10) and non-
regulatory CD8+ T cells (C02) was compared by using Northern blot
analysis. The expression of house keeping gene GAPDH from each cell
type was used as a control for equal loading.
(b) Expression of Osteopontin protein on DN regulatory T cells. D N
regulatory T cells (CN04) were stained by either purified mouse anti-rat
OPN mAb MPIIIB10 (DSHB, The University of Iowa) followed by FITC-
labelled anti-mouse monoclonal antibody or secondary monoclonal
antibody alone as a control. Data were analysed using a Flow cytometer.

CA 02316089 2000-08-24
-17-
(c) Inhibition of DN T cell-mediated lysis of activated syngeneic CDS+ T
cells by anti-OPN mAb. Activated Ld specific CD8+ T cells were labelled
with [3H]-TdR and used as targets. DN regulatory T cells (CN04) were
either left untreated as a control or preincubated with various
concentrations of anti-OPN mAb as indicated for 1 hour at 37°C prior to
being used as effector cells. The DN T cells were co-cultured with the CD8+
target cells at a 5:1 ratio for 18 hrs. Specific lysis of CD8+ T cells was
measured as previously described. Percentage inhibition of DNT cell
mediated killing by anti-OPN was compared with the cultures to which no
antibody was added.
Figure 17: (a) Induction of long-term xenograft tolerance by
pretransplant donor-specific transfusion (DST) and short-term CD4
depletion. B6 mice were infused with 4x10 viable spleen cells from Lewis
rats on day -7 and injected i.p. with depleting anti-CD4 mAb on days -2, 0,
3 (DST+anti-CD4, n=6). Control B6 mice were left untreated (no treatment,
n=6). On day 0, all the B6 mice were transplanted with Lewis heart. Graft
survival was monitored by daily palpation.
(b) Increase of DN and decrease of CD4+ and CD8+ T cells in accepted
xenogeneic heart grafts. B6 mice were treated as described in (a). At 20
days after transplantation, heart grafts were harvested and graft infiltrating
cells were triple-stained with anti-CD3, anti-CD4 and anti-CD8 mAbs.
Percentages of CD4+, CD8+ and CD3+CD4-CD8- T cells were analyzed by
using a flow cytometer. Data shown are the infiltrating cells pooled from 2
heart xenografts.
Figure 18:1B2+DN T cells infiltrate accepted skin allografts
(a) Pretransplant infusion of one MHC class I locus (Ld) mismatched
donor lymphocytes leads to permanent acceptance of donor specific skin
allografts. Anti-Ld TCR transgenic 2CF1 mice were given a donor specific
transfusion (DST) of 4x10 viable Ld+ splenocytes from (B6xBALB/c)F1
mice. One week later, the 2CF1 mice were given skin grafts from both
(B6xBALB/c)Fl (DST, n=20) and 3rd-party control C3H (DST + C3H, n=20)

CA 02316089 2000-08-24
-18-
mice. Some 2CF1 mice were transplanted with (B6xBALB/c)Fl skin graft
without DST (no DST). Graft survival was monitored for more than 120
days.
(b) Increase of 1B2+DN T cells in accepted skin allografts. The 2CF1 mice
were treated as described in (a). At 7 days after transplantation, the C3H
skin grafts were all rejecting, but the (B6xBALB/c)F1 grafts were accepted.
The skin accepted grafts were harvested, and the infiltrating cells were
triple-stained for the anti-Ld transgenic TCR (1B2), CD4 and CD8 and
analyzed by using a flow cytorneter. Data shown here represent results
from 5 mice analyzed.
(c) Bar graft shows the % of 1B2+CD8+ and 1B2+DN T cells in DST treated
accepted and non-DST treated rejecting skin allografts pooled from 5 mice
at 7 days after transplantation.
Figure 19: Increase of IL-4 and IFNJy in accepted skin allografts.
2CF1 mice were treated as in Figure 18. After 7 days the skin from accepted
(B6xBALB/c)F1 grafts and rejecting C3H grafts were harvested, and stained
for the anti-Ld transgenic TCR (1B2), CD4, CD8 and various cytokines as
indicated. The cells were analyzed by using a flow cytometer. The markers
indicate the percentage of cells producing each cytokines. Data shown here
are the pooled results from 5 mice.
DETAILED DESCRIPTION OF THE INVENTION
I. Novel Regulatov T cells
The present inventors have isolated novel regulatory T cells that
have important functions in immune regulation. The novel regulatory
cells are distinguished from previously described regulatory cells as they
possess a unique phenotype and express a unique array of cytokines. In
particular, the novel cells are CD3+a(3-TCR+CD4-CD8-CD25+CD28-CD30+
CD44-NK1.1-. These cell surface markers distinguish the cells from any
previously described T cell subset such as activated helper, cytotoxic or
memory T cells. The novel regulatory cells are also distinguished from
bone marrow derived CD4-CD8- T cells which express NK1.1 and from

CA 02316089 2000-08-24
-19-
CD4-CD8- T cells described by others. The novel regulatory cells do not
express IL-2, IL-4, IL-10 and IL-13 but do express IFN-y and TGF-(3 mRNA
after activation which distinguish them from Thl, Th2 or Th3/Trl cells.
Accordingly, the present invention provides an isolated immune
regulatory T cell having the phenotype CD3+a~3-TcR+CD4-CD8-CD44-CD28-
NK1.1-. The regulatory cells having the phenotype CD3+a(3-TcR+CD4-CD8-
CD44-CD28-NK1.1- are sometimes referred to herein as "the novel
regulatory cells", "the regulatory cells of the invention", "the DN
regulatory T cells" or "the regulatory T cells". Further, the term "a
regulatory T cell" includes one or more of the regulatory T cells of the
invention.
The inventors have also shown that suppression by the novel
regulatory cells requires cell contact and is not mediated through a soluble
factor released by the cells. The novel cells also require signals through
both the T cell receptor (TCR) and Fas/FasL to mediate suppression. The
suppressive properties of the novel regulatory cells can be abolished by IL-
10, cyclosporin A (CsA), anti-IFN-y and anti-TCR antibodies (Figure 15b).
Accordingly, IL-10, CsA, anti-IFN-'y and anti-TCR antibodies can be used to
inhibit the function of the regulatory cells.
The present invention also includes the generation of the novel
regulatory T cells in vitro. The cells may be isolated from normal animals,
for example by isolating lymphocytes, labelling T cells and sorting for cells
containing the desired phenotype using a FACS sorter. The inventors
have demonstrated that the novel regulatory cells generally require IL-2,
IL-4 to proliferate and to suppress. The inventors have further developed
methods to activate and expand antigen-specific regulatory T cells in vitro
by stimulating the novel regulatory T cells with one class I mismatched
allogeneic lymphocytes in the presence of IL-2 and IL-4. Accordingly, IL-2
and IL-4 can be used to increase the number and improve the function of
the novel regulatory cells.

CA 02316089 2000-08-24
-20-
Accordingly, the present invention provides a method of
expanding a population of regulatory T cells having the phenotype
CD3+a~i-TcR+CD4-CD8-CD44-CD28-NK1.1- in vitro comprising:
(a) obtaining a sample comprising the regulatory T cells or
precursors thereof;
(b) stimulating the cells with antigen; and
(c) culturing the cells under conditions suitable for the
expansion of the regulatory T cells.
The sample may be any sample that contains the regulatory T cells
or precursors of the regulatory T cells including, but not limited to, blood,
bone marrow, lymphoid tissue, epithelia, thymus, liver, spleen, cancerous
tissues, lymph node tissue, infected tissue, fetal tissue and fractions or
enriched portions thereof. The sample is preferably blood including
peripheral blood or fractions thereof, including buffy coat cells,
mononuclear cells and low density mononuclear cells (LDMNC). The
regulatory cells may be obtained from a sample of blood using techniques
known in the art such as density gradient centrifugation.
Prior to stimulating the sample or fraction thereof (such as
LDMNC) with antigen, the sample or fraction thereof may be depleted of
other cell types such as B cells, NK cells and CD4+ or CD8+ T cells. The
sample may be depleted of certain cell types using techniques known in
the art. In one embodiment, the cells of a particular phenotype may be
depleted by culturing the starting sample or fraction thereof with an
antibody cocktail containing antibodies specific for markers on the cells to
be depleted. Preferably, the antibodies in the cocktail are tetrameric
antibody complexes as described in United States Patent No. 4,868,109 to
Lansdorp.
The antigen can be any antigen depending on the desired antigen
specificity of the regulatory T cells. For example, the antigen may be a
donor specific antigen or cells containing a donor specific antigen when
the cells are used to prevent graft rejection, an autoantigen when the cells

CA 02316089 2000-08-24
-21-
are used to treat autoimmune disease, an allergen when the cells are used
to treat an allergy or a tumor antigen when the cells are used to treat
cancer. The antigens can be used in any form including purified peptides,
soluble proteins, plasmid expressing cDNA encoding specific antigens, cell
lines expressing specific antigens (EBV transformed cell lines, dendritic
cells, fibroblasts transfected with specific antigens such as foreign MHC
molecules), molecules that cause autoimmune diseases, allergy, and
tumor antigens.
The cells are cultured with the antigen "under conditions suitable
for the expansion of the regulatory T cells" which means in an appropriate
culture medium and for a suitable period of time to allow for the
expansion of the regulatory T cells. The appropriate culture medium is
any media that supports the expansion of the regulatory T lymphocytes
and it preferably contains IL-2 and IL-4. By "expansion" it is meant that the
number of the regulatory cells after step (c) is higher than the number of
regulatory cells in the initial sample.
The inventors have demonstrated that the regulatory T cells are
resistant to activation induced cell death. IL-10 can convert the apoptosis-
resistant DN regulatory T cells to apoptosis sensitive phenotype. In
addition, certain antibodies to the novel regulatory cells may also induce
apoptosis in the cells. Accordingly, IL-10 and/or antibodies that bind the
novel regulatory cells can be used to reduce the number of DN regulatory
T cells and can be used in therapies where immune suppression is not
desired.
The present invention also includes the use of the novel cells to
suppress an immune response. Accordingly, the present invention
provides a method of suppressing an immune response comprising
administering an effective amount of a regulatory T cell having the
phenotype CD3+a(3-TcR+CD4-CD8-CD44'CD28-NKl.1- to an animal in need
of such treatment.

CA 02316089 2000-08-24
-22-
The present inventors have demonstrated that injection of the
regulatory T cells into animals can significantly enhance skin allograft
survival in a dose-dependant and antigen-specific manner. Accordingly,
in one embodiment, the present invention provides a method of
preventing graft rejection in a recipient animal comprising administering
an effective amount of a regulatory T cell having the phenotype
CD3+a~3-TcR+CD4-CD8-CD44-CD28-NK1.1- to an animal in need of such
treatment. The term "graft" includes organs, tissues and cells. The graft
donor may be from alto- or xeno donors.
The present inventors have also shown that the regulatory T cells
activated in vitro by a specific antigen can specifically kill the CD8+ T
cells
that are activated by the same antigen but not those activated by different
antigen. Accordingly, the cells can be used to specifically suppress an
immune response caused by CD8+ T cells.
The present inventors have further identified the methods to
increase the number and function of the novel DN regulatory T cells
within allografts by infusion of one MHC class I locus mismatched
allogeneic lymphocytes, which in turn leads to permanent acceptance of
the allografts (Figure 18). The inventors have demonstrated that the
numbers of both regulatory T cells and IL-2 and IL-4 producing cells within
accepted allografts are significantly increased (Figure 19). Accordingly,
graft
survival could be enhanced by increasing local IL-2 and IL-4 levels by
either injection of rIL-2 and rIL-4 or increase IL-2/4 producing cells locally
in conjunction with local injection of regulating T cells.
The inventors have also shown that injection of the regulatory T
cells that are generated in vitro into animals can prevent death caused by
injection of lethal dose of tumor cells in the absence of GVHD.
Accordingly, in vitro cultured regulatory T cells can be used in the
treatment of leukaemia, lymphomas and other malignant diseases. The
inventors have identified the methods to increase the number of the
novel regulatory T cells in immunodeficient recipients by infusion of one

CA 02316089 2000-08-24
-23-
MHC class I locus mismatched donors, which lead to elimination of tumor
cells in the absence of GVHD.
The inventors have identified novel mechanisms by which the
regulatory T cells prevent GVHD and promote anti-tumor response. After
bone marrow transplantation mature donor T cells (CD4+ and CD8+) will
recognize alto MHC expressed on the host, be activated and express high
level of Fas. These activated donor T cells will destroy target cells and
tissues that express the host alloantigens and cause GVHD. T'he inventors
have demonstrated that the novel regulatory T cells constitutively express
a high level of Fas ligand. Upon encountering antigen-presenting cells
(APC), the regulatory T cells can "steal" host alloantigens from the surface
of APC through the anti-host TCR, and turn themselves into killer cells.
Because the regulatory T cells express the "stolen" host alloantigens on
their surface, they can attract the activated anti-host cytotoxic T cells.
Once
the anti-host cytotoxic T cells recognised the alloantigens on the regulatory
T cells, the latter will send death signals through Fas ligand to the former.
Unlike the anti-host cytotoxic T cells, which kill target cells through
perform-mediated pathway, the killing mediated by the regulatory T cells
requires direct cell-cell contact and depends on Fas-Fast interaction. Most
host tissues, although they express MHC class I molecules, do not express
Fas, and will not be destroyed directly by the regulatory T cells. Therefore
the regulatory T cell themselves do not cause GVHD. On the other hand,
the tumor cells, such as B cell lymphoma, express both recipients MHC
class I and Fas. The regulatory T cells can recognize the MHC class I
expressed on tumor cells through their specific TCR, and send death
signals to tumor cells through Fas ligand to cause death of tumor cells and
prevent death caused by lymphoma.
One dose donor-specific transfusion (DST) of one MHC class I
locus mismatched donor lymphocytes before transplantation leads to
permanent acceptance of skin allograft that are mismatched for the same
MHC class I plus multiple minor histocompatibility antigens in both
transgenic and normal mice. (Skin graft is the most stringent test for

CA 02316089 2000-08-24
-24-
allograft survival due to its strong immunogeneicity). The immune
system of recipients becomes tolerant only to the graft donor and remains
competent to recognition and response to other antigens such as tumor
antigens and viruses. Similarly, infusion of viable spleen cells from
donors mismatched for one MHC class I locus with recipients with or
without multiple minor histocompatibility antigen mismatches does not
cause graft versus host disease (GVHD) in the recipients. The infused
donor lymphocytes can effectively mediate anti-leukemia responses.
When one class I mismatched graft donor is not available, recipient cells
(such as dendritic cells) transfected with one HLA alloantigen can be used
to replace DST.
The inventors have demonstrated that pre-transplant DST in
conjunction with several injections of mAb to selectively deplete CD4+ T
cells leads to a long term survival of rat cardiac xenografts in mice (Figure
17a). Because the recipients are non-thymactomized, new anti-donor T
cells generated from the thymus must have been inactivated by regulatory
T cells in the recipient in order to maintain the xenografts. CD4+ T cells
have been speculated to play a role in allograft tolerance. As CD4+ T cells
are physically depleted in this model, it is unlikely that CD4+ T cells are
responsible for the xenograft survival. The inventors have demonstrated
that number of DN T cells in accepted allografts is significantly increased
in DST and anti-CD4 treated animals (Figure 17b) and that DN T cells can
enhance skin allograft survival by specific killing of anti-donor CD8+ T
cells. Depletion of CD4+ T cells leads to an increase of the proportion of
DN T cells in the recipient. Therefore it is plausible that the xenograft
tolerance seen after treatment with DST and anti-CD4 depleting mAb is
due to enhancement of the number and function of donor-specific DN
regulatory T cells.
II. Antibodies to the Re ulator;~ Cells
The inventors have prepared monoclonal antibodies (mAbs)
generated by immunization of animals with regulatory T cells of the
invention. The inventors have so far generated 25 mAbs that can

CA 02316089 2000-08-24
-25-
specifically bind to the surface of regulatory T cells. Some of the mAbs can
convert apoptosis-resistant regulatory T cells into apoptosis-sensitive
phenotype. Some mAbs can enhance growth of regulatory T cells and
some can abolish the suppressive function of regulatory T cells.
Accordingly, the present invention includes use of these mAbs for
isolation and purification of regulatory T cells. The present invention also
includes the use of these mAbs for induction of apoptosis in tumor cells,
and for up- or down-regulation of survival and function of regulatory T
cells. The invention also includes the use of these mAbs and their
therapeutic modifications in prevention and treatment of diseases such as
graft rejection, autoimmune diseases, malignant diseases, allergy and
AIDS which are discussed in greater detail below. Modifications of mAbs
include generation of recombinant mAbs fused with human
immunoglobulin Fc portion, conjugate mAbs with enzymes, isotopes etc.
III. Genes Expressed in Novel Re~ulatorv Cells
The present inventors have isolated several genes that are
expressed in the novel regulatory cells of the invention but are not
expressed in non-regulatory cells. The inventors have shown that
treatment of the novel cells with IL-10 switches the cells from a regulatory
to a non-regulatory phenotype. As a result, it was predicted that IL-10 may
regulate the expression of certain molecules necessary for suppression by
the novel regulatory cells. Consequently, using PCR-selected cDNA
subtraction, the inventors have identified many genes that are
differentially expressed in regulatory cells but not non-regulatory cells.
The inventors have generated cDNA libraries from both regulatory and
non-regulatory T cells. These libraries can be used together with the
regulatory specific mAbs to identify the molecules involved in control of
the function of regulatory T cells.
a) Ly-6A
The inventors have demonstrated that Ly-6A is highly expressed
on the novel regulatory cells. Ly-6A is a glycosyl phosphatidylinositol
(GPI)-anchored cell surface molecule expressed on most peripheral

CA 02316089 2000-08-24
-26-
lymphocytes, thymocytes and other cells. Incubating the novel regulatory
cells with IL-10 (which converts the regulatory phenotype into a non-
regulatory one as discussed above) reduces the expression of Ly-6A.
Further, blocking Ly-6A expression with an antisense oligonucleotide
abolishes suppression by the novel regulatory cells. The regulatory cells
obtained from Ly-6A knockout mice can not kill activated CD8+ T cells.
These results suggest that Ly-6A may act to down regulate lymphocyte
responses. Consequently, administering Ly-6A may be used to enhance
immune tolerance or suppression and inhibiting Ly-6A may be used to
enhance an immune response.
The term "Ly-6A protein" as used herein includes the full length
Ly-6A protein as well as fragments or portions of the protein. Preferred
fragments or portions of the protein are those that are sufficient to
suppress an immune response. The Ly-6A protein also includes fragments
that can be used to prepare antibodies.
The Ly-6A protein may be prepared as a soluble fusion protein.
The fusion protein may contain the extracellular domain of Ly-6A linked
to an immunoglobulin (Ig) Fc Region. The Ly-6A fusion may be prepared
using techniques known in the art. Generally, a DNA sequence encoding
the extracellular domain of Ly-6A is linked to a DNA sequence encoding
the Fc of the Ig and expressed in an appropriate expression system where
the Ly-6A - FcIg fusion protein is produced. The Ly-6A protein may be
obtained from known sources or prepared using recombinant DNA
techniques. The protein may have any of the known published sequences
for Ly-6A. The protein may also be modified to contain amino acid
substitutions, insertions and/or deletions that do not alter the
immunosuppressive properties of the protein. Conserved amino acid
substitutions involve replacing one or more amino acids of the Ly-6A
amino acid sequence with amino acids of similar charge, size, and/or
hydrophobicity characteristics. When only conserved substitutions are
made the resulting analog should be functionally equivalent to the Ly-6A
protein. Non-conserved substitutions involve replacing one or more

CA 02316089 2000-08-24
-27-
amino acids of the Ly-6A amino acid sequence with one or more amino
acids which possess dissimilar charge, size, and/or hydrophobicity
characteristics.
The Ly-6A protein may be modified to make it more
therapeutically effective or suitable. For example, the Ly-6A protein may
be cyclized as cyclization allows a peptide to assume a more favourable
conformation. Cyclization of the Ly-6A peptides may be achieved using
techniques known in the art. In particular, disulphide bonds may be
formed between two appropriately spaced components having free
sulfhydryl groups. The bonds may be formed between side chains of
amino acids, non-amino acid components or a combination of the two. In
addition, the Ly-6A protein or peptides of the present invention may be
converted into pharmaceutical salts by reacting with inorganic acids
including hydrochloric acid, sulphuric acid, hydrobromic acid, phosphoric
acid, etc., or organic acids including formic acid, acetic acid, propionic
acid,
glycolic acid, lactic acid, pyruvic acid, oxalic acid, succinic acid, malic
acid,
tartaric acid, citric acid, benzoic acid, salicylic acid, benzenesulphonic
acid,
and tolunesulphonic acids.
b) Osteopontin
The inventors have demonstrated that osteopontin is highly
expressed on regulatory but not on non-regulatory T cells and incubation
of anti-osteopontin antibody can reverse the suppressive function of the
regulatory T cells. Consequently, administering osteopontin may be used
to enhance immune tolerance or suppression and inhibiting osteopontin
may be used to enhance an immune response.
The term "osteopontin protein" as used herein includes the full
length osteopontin protein as well as fragments or portions of the protein.
Preferred fragments or portions of the protein are those that are sufficient
to suppress an immune response. The osteopontin protein also includes
fragments that can be used to prepare antibodies.
The osteopontin protein may be prepared as a soluble fusion
protein. The fusion protein may contain the extracellular domain of

CA 02316089 2000-08-24
_28_
osteopontin linked to an immunoglobulin (Ig) Fc Region. The
osteopontin fusion may be prepared using techniques known in the art.
Generally, a DNA sequence encoding the extracellular domain of
osteopontin is linked to a DNA sequence encoding the Fc of the Ig and
expressed in an appropriate expression system where the osteopontin -
FcIg fusion protein is produced. The osteopontin protein may be obtained
from known sources or prepared using recombinant DNA techniques.
The protein may have any of the known published sequences for
osteopontin. The protein may also be modified to contain amino acid
substitutions, insertions and/or deletions that do not alter the
immunosuppressive properties of the protein. Conserved amino acid
substitutions involve replacing one or more amino acids of the
osteopontin amino acid sequence with amino acids of similar charge, size,
and/or hydrophobicity characteristics. When only conserved substitutions
are made the resulting analog should be functionally equivalent to the
osteopontin protein. Non-conserved substitutions involve replacing one
or more amino acids of the osteopontin amino acid sequence with one or
more amino acids which possess dissimilar charge, size, and/or
hydrophobicity characteristics.
The osteopontin protein may be modified to make it more
therapeutically effective or suitable. For example, the osteopontin protein
may be cyclized as cyclization allows a peptide to assume a more
favourable conformation. Cyclization of the osteopontin peptides may be
achieved using techniques known in the art. In particular, disulphide
bonds may be formed between two appropriately spaced components
having free sulfhydryl groups. The bonds may be formed between side
chains of amino acids, non-amino acid components or a combination of
the two. In addition, the osteopontin protein or peptides of the present
invention may be converted into pharmaceutical salts by reacting with
inorganic acids including hydrochloric acid, sulphuric acid, hydrobromic
acid, phosphoric acid, etc., or organic acids including formic acid, acetic
acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid,

CA 02316089 2000-08-24
-29-
succinic acid, malic acid, tartaric acid, citric acid, benzoic acid, salicylic
acid,
benzenesulphonic acid, and tolunesulphonic acids.
IV. Ap,~lications
a) Inducing Immune Suppression
The present invention includes the use of the novel regulatory
cells, antibodies to the novel regulatory cells as well as Ly-6A and
osteopontin to induce immune suppression.
Accordingly, in one aspect, the present invention provides a
method of suppressing an immune response comprising administering an
effective amount of a regulatory T cell having the phenotype
CD3+a[3-TcR+CD4-CD8-CD44-CD28-NK1.1- to an animal in need of such
treatment.
In a preferred embodiment, the regulatory T cells are prepared in
vitro as described above and injected into the animal. In another
embodiment, the regulatory T cells are directly induced or stimulated in
vivo, for example using IL-2 and IL-4 or antibodies to the cells and/or by
stimulating the cells with antigen or one MHC Class I locus mismatched
allogenic lymphocytes.
In another aspect, the present invention provides a method of
suppressing an immune response comprising administering an effective
amount of an antibody that stimulates a regulatory T cell having the
phenotype CD3+a~i-TcR+CD4-CD8-CD44-CD28'NK1.1- to an animal in need
of such treatment.
In a further aspect, the present invention provides a method of
suppressing an immune response comprising administering an effective
amount of an Ly-6A protein or a nucleic acid sequence encoding an Ly-6A
protein to an animal in need of such treatment.
In yet another aspect, the present invention provides a method of
suppressing an immune response comprising administering an effective
amount of an osteopontin protein or a nucleic acid sequence encoding an
osteopontin protein to an animal in need of such treatment.

CA 02316089 2000-08-24
-30-
Administration of an "effective amount" of the active agent (i.e.,
regulatory T cells, antibodies, osteopontin or Ly-6A protein or nucleic acid
of the present invention) is defined as an amount effective, at dosages and
for periods of time necessary to achieve the desired result. The effective
amount of the active agent may vary according to factors such as the
disease state, age, sex, and weight of the animal. Dosage regima may be
adjusted to provide the optimum therapeutic response. For example,
several divided doses may be administered daily or the dose may be
proportionally reduced as indicated by the exigencies of the therapeutic
situation.
The term "animal" as used herein includes all members of the
animal kingdom including humans.
In one embodiment, the present invention provides a method of
inducing specific immune tolerance in a recipient animal comprising
administering an effective amount of the regulatory T cells, antibodies to
the regulatory cells, Ly-6A protein or a nucleic acid sequence encoding an
Ly-6A protein or osteopontin or a nucleic acid sequence encoding
osteopontin to the recipient animal.
The term "inducing immune tolerance" means rendering the
immune system unresponsive to a particular antigen without inducing a
prolonged generalized immune deficiency. The term "antigen" means a
substance that is capable of inducing an immune response. In the case of
autoimmune disease, immune tolerance means rendering the immune
system unresponsive to an auto-antigen that the host is recognizing as
foreign, thus causing an autoimmune response. In the case of allergy,
immune tolerance means rendering the immune system unresponsive to
an allergen that generally causes an immune response in the host. In the
case of transplantation, immune tolerance means rendering the immune
system unresponsive to the antigens on the transplant. An alloantigen
refers to an antigen found only in some members of a species, such as
blood group antigens. A xenoantigen refers to an antigen that is present in
members of one species but not members of another. Correspondingly, an

CA 02316089 2000-08-24
-31-
allograft is a graft between members of the same species and a xenograft is
a graft between members of a different species.
The recipient can be any member of the animal kingdom
including rodents, pigs, cats, dogs, ruminants, non-human primates and
preferably humans.
In a preferred embodiment, the method of the invention is used
to prevent rejection of a transplanted graft. The graft may be organs,
tissues, cells or the like. The organ, tissue or cells to be transplanted can
be
from the same species as the recipient (allograft) or can be from another
species (xenograft). The graft can be any tissue, organ or cell including
heart, liver, kidney, lung, pancreas, pancreatic islets, brain tissue, cornea,
bone, intestine, skin and hematopoietic cells.
As an example, for prevention or treatment of alto- and xeno-graft
rejection, donor-specific regulatory T cells can be generated in the
following way. Before organ transplantation the regulatory T cells can be
purified from blood or bone marrow of the recipient. The purified
regulatory T cells can be stimulated with donor antigens in various forms
including irradiated donor lymphocytes or recipient cells (e.g., DC or
fibroblasts) that have been genetically manipulated to express one donor
MHC antigen. Suitable concentrations of IL-2 and IL-4 will be added to the
culture. The donor-specific regulatory T cells generated in such way can
be injected back into the recipients before transplantation with or without
co-injection of rIL-2 and rIL-4. Because these regulatory T cells can
specifically kill the recipient CD8+ T cells that are activated by the same
donor as the inventors have demonstrated, the regulatory T cells should
be able to protect the graft from being attacked by anti-donor cytotoxic T
cells.
The method of the invention may be also used to prevent graft
versus host disease wherein the immune cells in the transplant mount an
immune attack on the recipient's organs and tissues. This can occur when
the tissue to be transplanted contains immune cells such as when bone

CA 02316089 2000-08-24
-32-
marrow or lymphoid tissue is transplanted when treating leukemias,
aplastic anemias and enzyme or immune deficiencies, for example.
Accordingly, in another embodiment, the present invention
provides a method of preventing or inhibiting graft versus host disease in
a recipient animal receiving an organ or tissue transplant comprising
administering an effective amount of the regulatory T cells, antibodies to
the regulatory T cells, an Ly-6A protein or a nucleic acid sequence encoding
an Ly-6A protein or osteopontin or a nucleic acid sequence encoding
osteopontin to the organ or tissue prior to the transplantation in the
recipient animal.
As an example, to prevent graft versus host diseases caused by
allogeneic bone marrow transplantation, regulatory T cells can be purified
from blood or bone marrow of graft donor and expanded in vitro. The
activated regulatory T cells can be injected back into the recipient before
bone marrow transplantation from the same donor. The donor cytotoxic T
cells that are activated by host alloantigens will be killed by the donor
regulatory T cells therefore to prevent GVHD. Donor cells that are
activated by third party antigens, including viruses and bacteria, will not be
killed and thus can fight off infections.
The method of the present invention may also be used to treat or
prevent autoimmune disease. In an autoimmune disease, the immune
system of the host fails to recognize a particular antigen as "self" and an
immune reaction is mounted against the host's tissues expressing the
antigen. Normally, the immune system is tolerant to its own host's
tissues and autoimmunity can be thought of as a breakdown in the
immune tolerance system.
Accordingly, in a further embodiment, the present invention
provides a method of preventing or treating an autoimmune disease
comprising administering an effective amount of the regulatory T cells,
antibodies to the regulatory T cells, Ly-6A protein or a nucleic acid
sequence encoding an Ly-6A protein or osteopontin or a nucleic acid

CA 02316089 2000-08-24
-33-
sequence encoding osteopontin to an animal having, suspected of having,
or susceptible to having an autoimmune disease.
Autoimmune diseases that may be treated or prevented according
to the present invention include, but are not limited to, type 1 insulin
dependent diabetes mellitus, adult respiratory distress syndrome,
inflammatory bowel disease, dermatitis, meningitis, thrombotic
thrombocytopenic purpura, Sjogren's syndrome, encephalitis, uveitic,
leukocyte adhesion deficiency, rheumatoid arthritis, rheumatic fever,
Reiter's syndrome, psoriatic arthritis, progressive systemic sclerosis,
primary biniary cirrhosis, pemphigus, pemphigoid, necrotizing vasculitis,
myasthenia gravis, multiple sclerosis, lupus erythematosus, polymyositis,
sarcoidosis, granulomatosis, vasculitis, pernicious anemia, CNS
inflammatory disorder, antigen-antibody complex mediated diseases,
autoimmune haemolytic anemia, Hashimoto's thyroiditis, Graves disease,
habitual spontaneous abortions, Reynard's syndrome, glomerulonephritis,
dermatomyositis, chronic active hepatitis, celiac disease, autoimmune
complications of AIDS, atrophic gastritis, ankylosing spondylitis and
Addison's disease.
As an example for the treatment of autoimmune diseases, the
regulatory T cells can be purified from a patient and stimulated with
antigens that are known to be involved in the induction/progression of
autoimmune diseases, such as collagen in arthritis, myelin in multiple
sclerosis, etc. The antigen-specific regulatory T cells generated in vitro
will
be injected back to the patient. The regulatory T cells will be able to
specifically kill activated CD8+ and/or CD4+ T cells that cause autoimmune
diseases.
As stated previously, the method of the present invention may
also be used to treat or prevent an allergic reaction. In an allergic
reaction,
the immune system mounts an attack against a generally harmless,
innocuous antigen or allergen. Allergies that may be prevented or treated
using the methods of the invention include, but are not limited to, hay
fever, asthma, atopic eczema as well as allergies to poison oak and ivy,

CA 02316089 2000-08-24
house dust mites, bee pollen, nuts, shellfish, penicillin and numerous
others.
Accordingly, in a further embodiment, the present invention
provides a method of preventing or treating an allergy comprising
administering an effective amount of the regulatory T cells, antibodies, Ly-
6A protein or a nucleic acid sequence encoding an Ly-6A protein or
osteopontin or a nucleic acid sequence encoding osteopontin to an animal
having or suspected of having an allergy.
In case of allergic diseases whereby the antigens are known (such
as milk products, peanuts, pollens, etc), antigen-specific DN T cells will be
purified from patients and stimulated in vitro with corresponding
allergens, and inject back to the patients.
b) Preventing Immune Suppression
The present invention also includes methods of preventing
immune suppression by administering an agent that inhibits the
regulatory T cells, Ly-6A or osteopontin.
There are a large number of situations whereby it is desirable to
prevent immune suppression including, but not limited to, the treatment
of infections, cancer and Acquired Immune Deficiency Syndrome.
In one aspect, the present invention provides a method of
preventing immune suppression comprising administering an effective
amount of an agent that inhibits the activation and/or the function of the
regulatory T cells to an animal in need thereof. Agents that inhibit the
regulatory cells include CSA, IL-10, anti-IFN~y and anti-TCR antibodies in
addition to antibodies to the regulatory cells.
In another aspect, the present invention provides a method of
preventing immune suppression comprising administering an effective
amount of an agent that inhibits Ly-6A to an animal in need thereof.
In a further aspect, the present invention provides a method of
preventing immune suppression comprising administering an effective
amount of an agent that inhibits osteopontin to an animal in need thereof.

CA 02316089 2000-08-24
-35-
In one embodiment, the present invention provides a method of
preventing immune suppression comprising administering an effective
amount of an agent that binds the regulatory T cells, Ly-6A or osteopontin
to an animal in need thereof.
In one embodiment, the agent that binds the regulatory T cells, Ly-
6A or osteopontin is an antibody. Antibodies to the regulatory T cells are
described above. In particular, the inventors have shown that some
antibodies to the regulatory T cells can induce apoptosis and are therefore
useful in suppressing the regulatory T cells.
Antibodies to Ly-6A or osteopontin may be prepared using
techniques known in the art such as those described by Kohler and
Milstein, Nature 256, 495 (1975) and in U.S. Patent Nos. RE 32,011;
4,902,614; 4,543,439; and 4,411,993, which are incorporated herein by
reference. (See also Monoclonal Antibodies, Hybridomas: A New
Dimension in Biological Analyses, Plenum Press, Kennett, McKearn, and
Bechtol (eds.), 1980, and Antibodies: A Laboratory Manual, Harlow and
Lane (eds.), Cold Spring Harbor Laboratory Press, 1988, which are also
incorporated herein by reference). Within the context of the present
invention, antibodies are understood to include monoclonal antibodies,
polyclonal antibodies, antibody fragments (e.g., Fab, and F(ab')2) and
recombinantly produced binding partners.
In another embodiment, the agent that inhibits the regulatory T
cells is an agent that interferes with the fas-fast interaction between the
regulatory T cells and their target. In a specific embodiment, the agent may
be a soluble Fas fusion protein (such as Fas-Fc) which binds to the Fast on
the regulatory T cells and inhibits their function.
In another embodiment, the Ly-6A or osteopontin inhibitor is an
antisense oligonucleotide that inhibits the expression of Ly-6A or
osteopontin. Antisense oligonucleotides that are complimentary to a
nucleic acid sequence from an Ly-6A gene or an osteopontin gene can be
used in the methods of the present invention to inhibit Ly-6A or

CA 02316089 2000-08-24
-36-
osteopontin. The present inventors have prepared antisense
oligonucleotides to Ly-6A which are described in Example 2.
Consequently, the present invention provides a method of
preventing immune suppression comprising administering an effective
amount of an antisense oligonucleotide that is complimentary to a nucleic
acid sequence from an Ly-6A gene to an animal in need thereof.
The present invention also provides, a method of preventing
immune suppression comprising administering an effective amount of an
antisense oligonucleotide that is complimentary to a nucleic acid sequence
from an osteopontin gene to an animal in need thereof.
The term antisense oligonucleotide as used herein means a
nucleotide sequence that is complimentary to its target.
The term "oligonucleotide" refers to an oligomer or polymer of
nucleotide or nucleoside monomers consisting of naturally occurring
bases, sugars, and intersugar (backbone) linkages. The term also includes
modified or substituted oligomers comprising non-naturally occurring
monomers or portions thereof, which function similarly. Such modified
or substituted oligonucleotides may be preferred over naturally occurring
forms because of properties such as enhanced cellular uptake, or increased
stability in the presence of nucleases. The term also includes chimeric
oligonucleotides which contain two or more chemically distinct regions.
For example, chimeric oligonucleotides may contain at least one region of
modified nucleotides that confer beneficial properties (e.g. increased
nuclease resistance, increased uptake into cells), or two or more
oligonucleotides of the invention may be joined to form a chimeric
oligonucleotide.
The antisense oligonucleotides of the present invention may be
ribonucleic or deoxyribonucleic acids and may contain naturally occurring
bases including adenine, guanine, cytosine, thymidine and uracil. The
oligonucleotides may also contain modified bases such as xanthine,
hypoxanthine, 2-aminoadenine, 6-methyl, 2-propyl and other alkyl
adenines, 5-halo uracil, 5-halo cytosine, 6-aza uracil, 6-aza cytosine and

CA 02316089 2000-08-24
-37-
6-aza thymine, pseudo uracil, 4-thiouracil, 8-halo adenine,
8-aminoadenine, 8-thiol adenine, 8-thiolalkyl adenines, 8-hydroxyl
adenine and other 8-substituted adenines, 8-halo guanines, 8-amino
guanine, 8-thiol guanine, 8-thiolalkyl guanines, 8-hydroxyl guanine and
other 8-substituted guanines, other aza and deaza uracils, thymidines,
cytosines, adenines, or guanines, 5-trifluoromethyl uracil and 5-trifluoro
cytosine.
Other antisense oligonucleotides of the invention may contain
modified phosphorous, oxygen heteroatoms in the phosphate backbone,
short chain alkyl or cycloalkyl intersugar linkages or short chain
heteroatomic or heterocyclic intersugar linkages. For example, the
antisense oligonucleotides may contain phosphorothioates,
phosphotriesters, methyl phosphonates, and phosphorodithioates. In an
embodiment of the invention there are phosphorothioate bonds links
between the four to six 3'-terminus bases. In another embodiment
phosphorothioate bonds link all the nucleotides.
The antisense oligonucleotides of the invention may also
comprise nucleotide analogs that may be better suited as therapeutic or
experimental reagents. An example of an oligonucleotide analogue is a
peptide nucleic acid (PNA) wherein the deoxyribose (or ribose) phosphate
backbone in the DNA (or RNA), is replaced with a polyamide backbone
which is similar to that found in peptides (P.E. Nielsen, et al Science 1991,
254, 1497). PNA analogues have been shown to be resistant to degradation
by enzymes and to have extended lives in vivo and in vitro. PNAs also
bind stronger to a complimentary DNA sequence due to the lack of charge
repulsion between the PNA strand and the DNA strand. Other
oligonucleotides may contain nucleotides containing polymer backbones,
cyclic backbones, or acyclic backbones. For example, the nucleotides may
have morpholino backbone structures (U.S. Pat. No. 5,034,506).
Oligonucleotides may also contain groups such as reporter groups, a group
for improving the pharmacokinetic properties of an oligonucleotide, or a
group for improving the pharmacodynamic properties of an antisense

CA 02316089 2000-08-24
_3g_
oligonucleotide. Antisense oligonucleotides may also have sugar
mimetics.
The antisense nucleic acid molecules may be constructed using
chemical synthesis and enzymatic ligation reactions using procedures
known in the art. The antisense nucleic acid molecules of the invention
or a fragment thereof, may be chemically synthesized using naturally
occurring nucleotides or variously modified nucleotides designed to
increase the biological stability of the molecules or to increase the physical
stability of the duplex formed with mRNA or the native gene e.g.
phosphorothioate derivatives and acridine substituted nucleotides. The
antisense sequences may be produced biologically using an expression
vector introduced into cells in the form of a recombinant plasmid,
phagemid or attenuated virus in which antisense sequences are produced
under the control of a high efficiency regulatory region, the activity of
which may be determined by the cell type into which the vector is
introduced.
c) Preventing or Treating Cancer
As hereinbefore mentioned, the inventors have demonstrated
that injection of the regulatory cells can prevent death caused by tumor
cells.
Accordingly, the present invention provides a method of
preventing or treating cancer comprising administering an effective
amount of a regulatory T cell having the phenotype CD3+a~3-TcR+CD4-
CD8-CD44-CD28-NK1.1- to an animal in need of such treatment.
The method may be used to treat any cancer or malignant disease
including, but not limited to, leukemias, lymphomas (Hodgkins and non-
Hodgkins), plasmacytomas, histiocytomas, melanomas, adenomas,
sarcomas, carcinomas of solid tissues, hypoxic tumours, squamous cell
carcinomas, genitourinary cancers such as cervical and bladder cancer,
hematopoietic cancers, head and neck cancers, and nervous system
cancers.

CA 02316089 2000-08-24
-39-
Because the regulatory T cells express high level of Fas ligand and
are able to recognize the host HLA alloantigens expressed on tumor cells
through their TCR, they are able to kill tumor cells that express both host
MHC molecules and Fas as the inventors have demonstrated. In the case
that tumor cells do not express alto MHC antigen or Fas, the regulatory T
cells will be co-injected with mature donor CDS+ T cells. As the regulatory
T cells can kill CD8+ T cells that are activated by the same alloantigen,
activated donor CD8+ T cells that can specifically recognize host antigen
and cause GVHD will be killed by the regulatory T cells though the
mechanisms the inventors have demonstrated. However, the donor CD8+
T cells that are activated by tumor antigens will not be affected by
regulatory T cells. Therefore tumor cells can be eliminated by co-injected
tumor-specific CD8+ T cells through a perform-dependant pathway. For
hematopoietic malignancies, the regulatory T cells will be injected
intravenously, for solid tumors, regulatory T cells will be injected both
intravenously and at the site of tumor after surgical removal of the tumor.
The present invention also provides a method of preventing or
treating cancer comprising inducing or stimulating an effective amount of
a regulatory T cell having the phenotype CD3+a(3-TcR+CD4-CD8-CD44
CD28-NK1.1- in an animal in need of such treatment. In one embodiment,
the regulatory T cells may be induced with IL-2 and IL-4 or by an antibody
that stimulates the regulatory cells.
(d) Compositions
The invention also includes pharmaceutical compositions
containing the regulatory T cells, the antibodies to the T cells, Ly-6A or
osteopontin proteins or nucleic acids for use in immune suppression or
treating cancer as well as pharmaceutical compositions containing
inhibitors of these for use in preventing immune suppression.
Such pharmaceutical compositions can be for intralesional,
intravenous, topical, rectal, parenteral, local, inhalant or subcutaneous,
intradermal, intramuscular, intrathecal, transperitoneal, oral, and

CA 02316089 2000-08-24
_4p_
intracerebral use. The composition can be in liquid, solid or semisolid
form, for example pills, tablets, creams, gelatin capsules, capsules,
suppositories, soft gelatin capsules, gels, membranes, tubelets, solutions or
suspensions.
The pharmaceutical compositions of the invention can be
intended for administration to humans or animals. Dosages to be
administered depend on individual needs, on the desired effect and on the
chosen route of administration.
The pharmaceutical compositions can be prepared by per se
known methods for the preparation of pharmaceutically acceptable
compositions which can be administered to patients, and such that an
effective quantity of the active substance is combined in a mixture with a
pharmaceutically acceptable vehicle. Suitable vehicles are described, for
example, in Remington's Pharmaceutical Sciences (Remington's
Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., USA
1985).
On this basis, the pharmaceutical compositions include, albeit not
exclusively, the active compound or substance in association with one or
more pharmaceutically acceptable vehicles or diluents, and contained in
buffered solutions with a suitable pH and iso-osmotic with the
physiological fluids. The pharmaceutical compositions may additionally
contain other agents such as immunosuppressive drugs or antibodies to
enhance immune tolerance or immunostimulatory agents to enhance the
immune response through the novel antigen specific regulatory T cells.
In one embodiment, the pharmaceutical composition for use in
inducing immune tolerance comprises an effective amount of the
regulatory T cells, an antibody against the regulatory T cells, an Ly-6A
protein or osteopontin in admixture with a pharmaceutically acceptable
diluent or carrier. The Ly-6A protein or osteopontin is preferably prepared
as an immunoadhesion molecule in soluble form which can be
administered to the patient. In the case of tissue or organ transplantation,
the composition preferably contains Ly-6A proteins or osteopontin in

CA 02316089 2000-08-24
-41-
soluble form which may be injected intravenously or perfused directly at
the site of the transplantation.
In another embodiment, the pharmaceutical composition for use
in inducing immune tolerance comprises an effective amount of a nucleic
acid molecule encoding an Ly-6A protein or osteopontin in admixture
with a pharmaceutically acceptable diluent or carrier.
The nucleic acid molecules of the invention encoding an Ly-6A
protein may be used in gene therapy to induce immune tolerance.
Recombinant molecules comprising a nucleic acid sequence encoding an
Ly-6A protein, or fragment thereof, may be directly introduced into cells or
tissues in vivo using delivery vehicles such as retroviral vectors,
adenoviral vectors and DNA virus vectors. They may also be introduced
into cells in vivo using physical techniques such as microinjection and
electroporation or chemical methods such as coprecipitation and
incorporation of DNA into liposomes. Recombinant molecules may also
be delivered in the form of an aerosol or by lavage. The nucleic acid
molecules of the invention may also be applied extracellularly such as by
direct injection into cells. The nucleic acid molecules encoding Ly-6A or
osteopontin are preferably prepared as a fusion with a nucleic acid
molecule encoding an immunoglobulin (Ig) Fc region. As such, the Ly-6A
or osteopontin protein will be expressed in vivo as a soluble fusion
protein.
In another aspect, the pharmaceutical composition for use in
preventing immune suppression comprises an effective amount of an
agent that inhibits the regulatory T cells, Ly-6A, or osteopontin in
admixture with a pharmaceutically acceptable diluent or carrier. Such
compositions may be administered as a vaccine either alone or in
combination with other active agents or antigens. When used in
combination, the Ly-6A inhibitors may act like an adjuvant by potentiating
the immune response to the antigen in the vaccine.
In one embodiment, the pharmaceutical composition for use in
preventing immune suppression comprises an effective amount of an

CA 02316089 2000-08-24
-42-
antibody to the regulatory cells or Ly-6A or osteopontin in admixture with
a pharmaceutically acceptable diluent or carrier. The antibodies may be
delivered intravenously.
In another embodiment, the pharmaceutical composition for use
in preventing immune suppression comprises an effective amount of an
antisense oligonucleotide nucleic acid complimentary to a nucleic acid
sequence from an Ly-6A gene or osteopontin in admixture with a
pharmaceutically acceptable diluent or carrier. The oligonucleotide
molecules may be administered as described above for the compositions
containing Ly-6A or osteopontin nucleic acid sequences.
The following non-limiting examples are illustrative of the
present invention:
EXAMPLES
Example 1
Double Negative Regulator3r T cells
Induction of indefinite donor-specific allograft survival by
pre-transplantation DST.
The suggested mechanisms whereby the introduction of donor
cells leads to tolerance include clonal deletion, clonal anergy, suppression
by regulatory cells, regulation of cell surface molecules or cytokines
expression, and promotion of microchimerism, (24, 38, 46, 47). Although
informative, these studies are not definitive. The major obstacle to
understanding the mechanism of donor specific transfusion (DST)
-induced tolerance is the lack of a specific cell markers) for detecting
donor-specific antigen-reactive cells in vivo. This limitation precludes
tracing the cellular and molecular events occurring within patients or
normal (i.e. non-transgenic) animals after encountering donor antigen
(Ag).
In order to obtain a system in which a T cell response to a defined
allogeneic MHC Ag can be monitored in vivo, 2C anti-Ld T cell receptor
(TCR) transgenic mice were used. 2C mice carry functionally re-arranged
TCR a- (one copy) and ~i-chain (eight copies) transgenes derived from a

CA 02316089 2000-08-24
-43-
cytotoxic T cell clone 2C, which is alloreactive for class I MHC Ag Ld. The
specificity for Ld requires the transgenic a- and ~3-chains as well as the CD8
co-receptor (48, 49). The 2C clonotypic TCR (both a- and ~i-chains) is
recognized by the mAb 1B2 (50). As schematically shown in Figures 1, 2C
mice on C57BL/6 (B6) background (H-2b~b, anti-Ld TCR+) were bred with
dm2 mice (a BALB/c Ld loss mutant, H-2Dd+, Kd+, I-Ed+~ I-Ad+~ Ld-), The
2CF1 mice (H-2b~d, Ld-, either 1B2+ or 1B2-) were used as recipients.
(B6xBALB/c)Fl mice (BYJFl, H-2b~d, Ld+) mismatched only for Ld with the
2CF1 mice were used as lymphocyte and graft donors. As a result, the only
immune response in this system is derived from recipient cells reacting to
Ld Ag expressed on the donor. The 2CF1 mice were intravenously injected
with viable Ld+ BYJFl lymphocytes followed by transplantation of skin
grafts obtained from both BYJFl (donor-specific graft) and 3rd-party
allogeneic control mice. All 3rd-party allogeneic grafts were rejected
between 11-13 days, whereas skin grafts from BYJF1 mice survived
indefinitely (>100 days, defined as tolerant mice) (16, 17). Interestingly,
injection of cyclosporin A (CsA) either on the day of DST, or skin grafting,
abolished DST-induced tolerance (17). These findings clearly indicate the
possibility of the induction of donor-specific long-term allograft tolerance
by DST in the absence of any immunosuppressive drugs.
Regulatory T cells are involved in DST-induced tolerance.
Down-regulation of immune responses to self or allogeneic Ags by
regulatory T cells has been demonstrated in a number of in vivo models of
autoimmunity and transplantation (1, 2, 4-6, 41-44). Waldmann et al have
shown that the injection of non-depleting CD4 specific mAb plus DST
could induce long-term survival of cardiac allografts (2, 21). Lymphocytes
from tolerant mice were showed to suppress naive syngeneic T cells in
vivo and to induce their tolerance to the same Ags (2, 21). Orally feeding
animals with Ags can induce Ag-specific tolerance (3, 19, 51). Weiner's
group isolated TGF-(3 producing CD4+ T cell clones from these tolerant
mice that were able to protect naive mice from developing autoimmune

CA 02316089 2000-08-24
disease induced by the same Ag (3). A similar finding has been reported by
Groux et al (5).
In the inventors' model, DST led to an elimination of the majority
(~60% of total) of donor-specific T cells in the periphery, and a significant
increase of IL-4 in the sera of recipients (16). Since none of the recipients
was thymectomized, and the anti-donor T cells isolated from tolerant mice
were fully functional upon Ag re-stimulation in vivo (13-15), the
inventors concluded that the function of thymic emigrant anti-Ld T cells
must have been inhibited, as Ld+ skin grafts were maintained. Indeed, the
inventors found that the spleen cells from tolerant mice were able to
specifically inhibit the anti-Ld response mediated by syngeneic naive T cells
in vivo (16), and enhance Ld+ skin allograft survival after adoptive transfer
into naive syngeneic mice (Figure 2a). These results suggest an important
role of regulatory T cells in DST-induced Ag-specific tolerance.
The regulatory T cells reported by others are CD4+ (3, 5, 41). To
determine which subset of T cells from tolerant 2CF1 mice was responsible
for the inhibition of the anti-Ld response, 1B2+CD8+, 1B2+CD4+ and
1B2+CD4-CD8- (double negative, DN) T cells were purified from the spleen
of tolerant mice, and tested for their regulatory function in vitro. A
dose-dependent inhibition of proliferation of naive anti-Ld T cells was
observed only in cultures to which 1B2+DN cells from tolerant mice were
added. No inhibition was seen when 1B2+CD8+ or 1B2+CD4+ T cells from
tolerant mice were added to the MLR (Figure 2b). These results
demonstrate that mature DN, but not CD4+ or CD8+, T cells are responsible
for inhibiting the anti-Ld response mediated by naive T cells in this model.
Generation of DN regulatory T cell clones.
The majority of a(3 T cells in the periphery of mice or humans
express the CD4 or CD8 co-receptor. About 1-5% of peripheral T cells are
DN (52). Although extensive studies have been done on CD4+ or CD8+ T
cells, little is known about the function and homeostasis of DN T cells.
This paucity of information is largely due to the relatively small number

CA 02316089 2000-08-24
-45-
of DN T cells in the periphery, which make such studies more difficult. In
order to characterize the DN regulatory T cells and understand the
mechanisms of suppression, the inventors generated panels of 1B2+D N
and 1B2+CD8+ T cell clones from the spleen of tolerant mice. Of the 38
clones generated to date, 8 1B2+DN and 4 1B2+CD8+ T cell clones grew
successfully. All of the 8 1B2+DN T cell clones displayed a dose-dependent
inhibition of anti-Ld response of naive T cells, whereas only 1 of 4
1B2+CD8+ T cell clones showed moderate suppressive function (table 1).
1B2+CD4-CD8- T cells from the spleen of tolerant mice are able to inhibit
anti-Ld responses in vitro and in vivo
To confirm the existence of antigen-specific regulatory cells in
DST-treated animals, the inventors first investigated whether the spleen
cells from tolerant animals can inhibit anti-Ld responses in vivo. Naive
(B6xdm2)Fl mice were adoptively transferred with syngeneic splenocytes
from either DST-treated (tolerant) mice, or mice that did not receive DST
and rejected Ld+ skin grafts within 2 weeks (non-tolerant), and were
subsequently given skin grafts from both Ld+ ((B6xBALB/c)F1,
antigen-specific) and third-party (C3H, H-2K) mice. As shown in Figure 2a,
all animals receiving spleen cells from non-tolerant mice acutely
eliminated the Ld+ skin allografts (median survival time (MST) =11 days).
However, the survival of Ld+ skin allografts was prolonged significantly in
the recipients infused with splenocytes from tolerant mice (MST = 33 days,
n=8, p<0.05). The 3rd-party skin allografts were rejected with similar
kinetics (MST = 11 days) by all of the recipients. These data demonstrate
that the spleen cells from tolerant mice are able to transfer tolerance to
naive syngeneic animals and specifically enhance Ld+ skin allograft
survival.
The vast majority of peripheral T cells in (2Cxdm2)Fl mice express
transgenic TCR specific for MHC class I Ld. The transgenic TCR can be
detected by the clonotypic monoclonal antibody (mAb) 1B2. To determine
which subset of T cells from tolerant (2Cxdm2)Fl mice was responsible for

CA 02316089 2000-08-24
-46-
the inhibition of the anti-Ld response mediated by naive T cells, 1B2+CDS+,
1B2+CD4+ and 1B2+CD4-CD8- (double negative, DN) T cells were purified
from the spleen of tolerant mice 120 days after skin grafting, and tested for
their regulatory function in vitro. A dose-dependent inhibition of
proliferation of naive anti-Ld T cells in mixed lymphocyte reactions (MLR)
was observed only in cultures to which 1B2+DN T cells from tolerant mice
were added (Figure 2b). No inhibition was seen when 1B2+CD8+ or
1B2+CD4+ T cells from tolerant mice were added to the MLR. These results
demonstrate that the mature DN, but not CD4+ or CD8+ T cells are
responsible for inhibiting the anti-Ld response mediated by naive T cells in
this model.
DN T cells obtained from both transgenic and normal mice can inhibit
allogeneic immune responses mediated by T cells of the same TCR
specificity.
In order to characterize the DN regulatory T cells and understand
the mechanism of suppression, the inventors generated panels of 1B2+DN
and 1B2+CD8+ T cell clones from the spleens of both naive and DST-treated
tolerant (2Cxdm2)Fl mice. Regardless of the origin, the DN T cells grew
only when stimulated by Ld+ cells in the presence of exogenous IL-2 and
IL-4. All 8 1B2+DN T cell clones that grew successfully displayed a
dose-dependent inhibition of proliferation of naive anti-Ld T cells,
whereas 3 of 4 1B2+CD8+ T cell clones showed no suppression. The
representative results are shown in Figure 3a. In addition to suppression
of proliferation, the cytotoxicity mediated by naive anti-Ld T cells was also
significantly impaired (Figure 3b).
To confirm that the DN regulatory T cells obtained from the TCR
transgenic mice have a physiological relevance, the inventors first
investigated whether the DN T cell clones generated from transgenic mice
are able to enhance Ld-specific skin allograft survival in non-transgenic
mice. Naive unmanipulated (B6xdm2)F1 mice were infused with the
1B2+DN suppressive clone TN12 and then given Ld+ and 3rd-party (C3H)
skin allografts. As controls, some mice were infused with the

CA 02316089 2000-08-24
-47-
non-suppressive 1B2+CD8+ T cell clone C02. All the recipients infused
with the non-suppressive C02 cells rejected Ld+ skin allografts within 14
days as seen in non-injected mice. In contrast, the survival of Ld+ skin
grafts was prolonged in the recipients of suppressive 1B2+DN T cell clones
in a dose-dependant manner (MST= 43.5 days, p<0.01). Two out of 6
recipients accepted the Ld+ skin allografts indefinitely (>120 days) (Figure
3c). The 3rd-party skin allografts were all acutely rejected (MST=12 days)
regardless of the types of cells infused (not shown). These results indicate
that 1B2+DN T cell clones can specifically enhance Ld+ skin graft survival
in normal mice.
Next, the inventors studied whether DN T cells in the spleen of
normal (non-transgenic) mice also have regulatory function.
a(3-TCR+CD3+DN T cells were purified from normal (B6xdm2)F1 and
(MRL/+, H-2k) mice, and their ability to suppress syngeneic CD8+ cells was
examined. As seen in 1B2+DN T cell clones, a dose-dependent inhibition
of proliferation of syngeneic CD8+ T cells by activated CD3+DN T cells was
also observed when the same culture conditions (i.e., alloantigen plus IL-2
and IL-4) were used (Figure 3d). This finding suggests that peripheral DN
T cells from both transgenic and normal mice are able to function as
regulatory T cells, obviating concerns that the DN regulatory T cells
observed in the TCR transgenic animals are a non-physiologically relevant
oddity. Moreover, the DN regulatory T cell clones can be generated from
both tolerant and naive animals (Figure 3a), suggesting that the DN
regulatory T cell precursors may exist in the spleen of normal mice. The
role of DST may be to promote the activation/function of DN T cells.
DN regulatory T cells express a unique set of cell surface markers.
To further characterize the antigen specific regulatory T cells, the
expression of cell surface markers was compared between regulatory and
non-regulatory T cells. Regulatory clones obtained from both tolerant and
naive animals express equivalent levels of a~i-TCR, CD25, LFA-1, CD69,
CD45, CD62L, and CTLA-4 when compared with non-regulatory 1B2+CD8+

CA 02316089 2000-08-24
-48-
T cell clones or primary activated 1B2+CD8+ T cells. They are negative for
TCR and NK1.1. Interestingly, unlike 1B2+CD8+ T cells or clones, none of
the DN regulatory T cell clones express CD28 or CD44 at any time point
after activation. The only molecule that is expressed on 1B2+DN but not
on 1B2+CD8+ T cell clones observed to date is CD30 (Figure 4, i-vi). A
similar pattern of cell surface markers was observed when DN T cells
collected from tolerant mice 120 days after skin grafting were studied
(Figure 4,vii-ix). These results demonstrate that DN regulatory T cells do
express a unique combination of cell surface markers (i.e.,
a(3-TCR+CD4-CD8-CD25+CD28-CD30+CD44-) which makes them
distinguishable from any previously described T cell subset (e.g., activated
helper, cytotoxic or memory T cells). These mature, peripheral DN T cells
also differ from bone marrow derived DN natural suppressor T cells
which express NK1.1 and DN T cell clones described by others. These
findings may explain why earlier attempts to identify a single unique
marker on CD4+ or CD8+ suppressor T cells failed.
Suppression mediated by DN T cells requires direct cell-cell contact
To delineate the mechanisms of suppression, the inventors first
examined whether suppression was mediated by secreting soluble
suppressive factors such as TGF-~i or IL-10 as reported by others.
Semi-quantitative reverse-transcription polymerase chain reaction
(RT-PCR) revealed that both regulatory and non-regulatory T cell clones
express equivalent levels of IFN-y, TGF-(3, and TNF- transcripts, and none
of them express IL-2, IL-4, or IL-13 mRNA at any time after activation.
Interestingly, IL-10 was only expressed in primary activated 1B2+CD8+ T
cells and 1B2+CD8+ clones, but not in any DN regulatory T cell clones
(Figure 4b). These findings indicate that our DN regulatory T cells possess
a unique array of cytokines that differ from Thl, Th2 or Th3/Tr1 cells, and
that IL-10 is unlikely a suppressive factor produced by DN regulatory T
cells as seen in CD4+ regulatory cells. They also imply that suppression
may be mediated by means other than secretion of suppressive cytokines.

CA 02316089 2000-08-24
-49-
Consistent with this notion, supernatant collected from regulatory T cell
clones after stimulation with irradiated Ld+ spleen cells was not able to
inhibit the naive anti-Ld response in vitro (not shown). When naive
anti-Ld T cells were co-cultured with regulatory T cells in a transwell
system to prevent direct cell-cell contact but maintain diffusion of secreted
soluble factors, no suppression was observed (Figure 4c). These results
support the conclusion that suppression by DN regulatory T cells requires
cell contact. In addition, the inventors also demonstrated that suppression
is not likely due to competition for either APC, or growth factors, as
reported by others. Specifically, increasing the number of APC, or the
concentrations of IL-2/IL-4 did not reverse the suppression (not shown).
DN T cells are able to kill activated CD8+ T cells through the Fas-dependant
pathway.
Next, the inventors addressed whether suppression is mediated
through direct killing of 1B2+CD8+ T cells, and if so, which pathway is used
by DN T cells. Two major pathways are involved in T cell mediated
cytotoxicity: one perform-dependant, the other Fas-dependant. To study
the ability of DN T cells to kill anti-Ld T cells through these pathways,
three different target cells were used in cytotoxicity assays: Concanavalin A
(Con A) activated spleen cells from both (B6xBALB/c)F1 (La+, Fas+) and
1B2+CD8+ T cells (Ld-, Fas+) as well as a mastocytoma cell line P815 (Ld+,
Fas-). Interestingly, although both activated regulatory and non-regulatory
T cells express similar levels of TCR (Figure 4) and Fas ligand (FasL), they
lysed target cells by using different cytolytic pathways. As seen in most
cytotoxic CD8+ T cells, the non-regulatory 1B2+CD8+ T cells lysed Ld+
allogeneic, but not Ld- syngeneic target cells regardless of Fas expression
through the perform-dependant pathway (Figure 5a, left panel). In
contrast, 1B2+DN regulatory T cells killed neither Ld+ allogeneic nor
syngeneic T cells through the perform-dependant pathway (Figure 4a,
middle panel). Furthermore these regulatory T cells did not kill Fas-
allogeneic cells, but they were able to kill both Fas+ anti-Ld and Fas+ Ld+
CD8+ T cells in an 18-hour killing assay (Figure 5a, right panel). These

CA 02316089 2000-08-24
-50-
results indicate the possibility that Fas-Fast interactions were involved in
DN T cell-mediated killing, which is consistent with report by others34,
To verify the involvement of Fas-Fast interaction in DN T
cell-mediated killing, the DN T cells were incubated with varying doses of
Fas-Fc fusion protein prior to and during the cytotoxicity assays. As shown
in Figure 5b, the ability of DN T cells to kill activated 1B2+CD8+ T cells was
blocked in a dose-dependant manner by Fas-Fc fusion protein. This
finding indicates that blocking Fast on the DN T cell abolishes DN T
cell-mediated cytotoxicity. To further determine the importance of Fas in
DN T cell-mediated cytotoxicity, the ability of purified a(3-TCR+CD3+DN T
cells from (B6xdm2)Fl and MRL/lpr mice to kill CD8+ cells from wild-type
and Fas mutant lpr mice was determined. As shown in Figure 5c, DN T
cells from both normal and lpr mice were able to kill activated CD8+ T cells
that express wild-type Fas, and the cytotoxicity of DN T cells collected from
Ipr mice was more pronounced than those from normal mice. However,
in both strains of mice, the cytotoxicity was significantly reduced when
activated CD8+ cells from Fas mutant lpr mice were used as target cells.
Taken together, these results demonstrate that DN regulatory T clones and
cells are able to kill activated CD8+ T cells, and that Fas/FasL interactions
are important for the cytotoxicity to take place.
1B2+DN T cell mediated cytotoxicity is not due to bystander killing.
Our in vivo skin grafting results indicate that DN T cell
suppression is antigen specific, which is consistent with the in vitro
finding that activated Fas+ Ld+ T cells are killed by anti-Ld DN T cells.
However, the finding that syngeneic Ld- 1B2+CD8+ T cells were also killed
suggests the possibility that DN T cells may mediate a non-specific
bystander killing through Fas/FasL interactions, as seen in some CD4+ and
CD8+ T cells. In order to determine the antigen specificity of DN T cell
mediated cytotoxicity, 1B2+DN T cells were stimulated by Ld+ cells and used
as effector cells. Activated Ld- Fas+CD8+ T cells with different antigen
specificities including a) 1B2+CD8+ T cells (anti-Ld); b) female anti-male HY

CA 02316089 2000-08-24
-51-
TCR transgenic T cells (anti-HY); c) anti-lymphocytic choriomeningitis
virus glycoprotein TCR transgenic T cells (anti-LCMV-gp); d) C3H anti-SJL
non-transgenic T cells (anti-H-2S) were used as targets. Although the target
cells are all Ld-, and express a similar level of Fas following activation,
only
those 1B2+CD8+ T cells that carry the same TCR specificity as the DN T cells
were killed. None of the CD8+Fas+ T cells that express a TCR with different
antigen specificity from DN T cells were lysed (Figure 5d). These data
demonstrate that 1) 1B2+DN T cells do not lyse Fas+ T cells through
bystander killing. 2) Sharing TCR-specificity between DN and target T cells
is required for cytotoxicity to take place.
TCR-Ld interaction is critical for 1B2+DN T cell-mediated killing.
The finding that both Ld+ and anti-Ld T cells, but not other
activated Ld- CD8+ T cells are killed by 1B2+DN T cells suggests that a
specific TCR-Ld interaction may be involved in the mechanism of
suppression. In order for suppression to be mediated by the anti-Ld TCR,
either the target or effector cell must express Ld. Recent studies have
indicated that T cells are able to acquire MHC from APC. To study whether
either Ld- target or regulatory T cells could acquire Ld during their
activation, 1B2+DN and 1B2+CD8+ cells were independently incubated with
irradiated Ld+ APC, and their ability to pick-up and express Ld was
monitored at different time points by triple staining of cultured cells with
1B2, anti-CDB, and anti-Ld mAbs. As shown in Figure 6a, expression of Ld
on both 1B2+DN and 1B2+CD8+ T cells was observed within minutes after
incubation with Ld+ spleen cells. By 12 hours, there was no detectable Ld
on the surface of 1B2+CD8+ T cells. In contrast, Ld expression was observed
on 24% of the 1B2+DN T cells, even at 48 hours (Figure 6b). These results
demonstrated that both 1B2+DN and 1B2+CD8+ T cells are able to acquire Ld
from APC, and express it on their surface. However, the expression of Ld
on 1B2+CD8+ T cells was transient as seen by others, whereas, the Ld
expression on DN T cells persisted for at least 2 days.
Next, the inventors investigated whether Ld expressed on DN T
cells was critical for lysis of syngeneic anti-Ld CD8+ T cells by blocking

CA 02316089 2000-08-24
-52-
studies. When Ld molecules on DN T cells were neutralized by
pre-incubation with anti-Ld mAb prior to being used as suppressor cells,
their ability to kill 1B2+CD8+ T cells was eradicated. Likewise, when the
TCR on 1B2+CD8+ cells was blocked by pre-incubation with 1B2 mAb before
being used as target cells, DN T cell-mediated cytotoxicity was also
abolished (Figure 6c). Thus, blocking either Ld on the DN T cells or TCR
on anti-Ld CD8+ T cells abrogates DN T cell-mediated lysis of syngeneic
CD8+ T cells. Together, these data clearly demonstrate that lysis of
syngeneic CD8+ T cells by DN T cells requires a specific interaction between
TCR and Ld alloantigen along with Fas/FasL interaction. These findings
not only explain why suppression seen in our model is antigen-specific in
vitro and in vivo, but also provide a novel model for understanding the
mechanism of regulatory T cell-mediated suppression.
Discussion
In this Example the inventors have identified and characterized a
novel subset of antigen-specific regulatory T cells from the spleen of both
transgenic and non-transgenic mice. The antigen-specific regulatory T
cells express a unique combination of cell surface markers
(a/3-TCR+CD4-CD8-CD25+CD28-CD30+CD44-NK1.1-) and array of cytokines
(express IFN-y, TGF-(3, and TNF-a, but not IL-2, IL-4, IL-10 and IL-13),
which make them distinguishable from any previously reported T cell
subsets. The inventors demonstrated that 1B2+DN T cells and clones
obtained from both naive and tolerant mice can specifically suppress
anti-Ld responses in vitro and enhance donor specific skin allograft
survival. In addition, since DN regulatory T cells can be obtained by either
pre-transplantation DST or in vitro stimulation of naive cells with
alloantigen in the presence of exogenous IL-2/IL-4, it suggests that the DN
regulatory T cell precursors may pre-exist in normal individuals. Once the
proper conditions are provided, the DN T cells will activate, proliferate,
and specifically inhibit CD8+ T cells activated by the same alloantigen, and
thus prevent rejection of specific allografts. The role of DST in the

CA 02316089 2000-08-24
-53-
induction of donor-specific tolerance may be to promote the activation and
expansion of DN regulatory T cells in vivo by providing antigen
stimulation and IL-2/IL-4. The following results support this notion: 1)
Both antigen and exogenous IL-2 and IL-4 are required for the DN T cells to
survive and proliferate in vitro. 2) The level of IL-4 in the sera of DST
recipients increased significantly. 3) The number of DN T cells in the
accepted Ld+ skin allografts and cardiac xenographts of DST-treated tolerant
animals was significantly increased (Figure 17 and 18).
Down-regulation of specific immune responses by regulatory T
cells in vitro and in vivo has been observed in numerous cases. However,
the mechanism by which regulatory T cells mediate antigen-specific
suppression remains unclear. Various mechanisms such as competition
with antigen-specific T cells for APC or growth factors, and the production
of suppressive cytokines have been proposed. The inventors
demonstrated, in their model, that DN regulatory T cell mediated
suppression requires direct contact with activated CD8+ T cells, suggesting
that suppression is not simply due to secretion of inhibitory cytokines or
soluble factors. Since DN T cell mediated suppression can not be abolished
by the addition of an excessive number of APC or IL-2/IL-4, it indicates that
suppression is neither due to competition for the surface area on APC nor
growth factors with CD8+ cells. Moreover, the inventors demonstrate that
DN T cells are able to kill activated CD8+ T cells through the Fas/FasL
pathway. This killing can be inhibited by Fas-Fc fusion protein, and the
ability of DN T cells to kill syngeneic CD8+ T cells from Fas-mutant lpr
mice was significantly impaired. Together, these results demonstrate that
Fas/FasL interactions are involved, at least partially, in DN T
cell-mediated suppression.
The fact that DN T cells are able to kill both activated Ld+
allogeneic and anti-Ld syngeneic T cells, but not other CD8+ T cells
activated by 3rd party antigens suggests the involvement of specific antigen
recognition during suppression. Huang et al reported recently that naive
1B2+CD8+ T cells were able to acquire Ld peptide-MHC clusters from APC

CA 02316089 2000-08-24
through TCR-mediated endocytosis. The Ld expression on the surface of
1B2+CD8+ cells could be detected within minutes to hours of interaction
with APC. During this process the 1B2+CD8+ T cells were sensitized to
peptide-specific lysis by neighbouring 1B2+CDS+ T cells. The 1B2+CD8+ T
cells therefore killed each other (fratricide), leading to down-regulation of
immune response. In our study, Ld expression on 1B2+CD8+ cells was also
detected within the first few hours after interaction with Ld+ APC. If
suppression were due to fratricide between CD8+ T cells, a significant
reduction of CD8+ T cells would be observed within the first 24 hours of
encountering the antigen. However, our results from kinetic studies
indicated no significant death of 1B2+CD8+ T cells within the first 66 hours
after encountering Ld+ spleen cells (not shown). In addition, the finding
that 1B2+CD8+ T cells did not suppress an anti-Ld response does not
support the hypothesis that suppression of CD8+ T cells in our model is
due to fratricide.
Alternatively, the inventors hypothesized that DN T cells may
acquire Ld from APC and present it to anti-Ld CD8+ T cells. The following
results from our studies support this hypothesis. First, the inventors
demonstrate that 1B2+DN T cells are able to acquire Ld from APC and
express it on their surface for at least 48 hours, in contrast to its
transient (<
min) expression on 1B2+CD8+ T cells. Second, DN T cells lost their
ability to kill activated 1B2+CD8+ T cells when pre-incubated with anti-Ld
mAb. Third, preoccupying the anti-Ld TCR on CD8+ T cells with 1B2 mAb
prevented DN T cell mediated killing. Fourth, DN T cells that do not have
25 Ld on their surface (e.g., after 4 days activation) are unable to kill
activated
syngeneic CD8+ T cells. Together, these results demonstrate that DN T
cells are able to "borrow" alloantigen from APC and turn themselves into
killer APC which express both specific alloantigen and Fast. Once
activated Fas+ syngeneic CD8+ T cells recognize the alloantigen expressed
30 on the surface of DN T cells, the latter will send a death signal through
their Fast to the CD8+ T cells. Activated T cells that express Fas but cannot
interact specifically with DN T cells through antigen-TCR binding will not

CA 02316089 2000-08-24
-55-
be affected. CD8+ T cells that express Ld can specifically interact with the
TCR of DN T cells and are therefore also killed. The mechanism of
suppression utilized by DN T cells in this model (Figure 7) is clearly
different from any previously proposed models including veto cells8.
Whether the APC that activate DN T cells and "lend" alloantigen to DN T
cells are the same as those that present antigen to CD8+ T cells, and
whether other co-stimulatory molecules are involved in the interactions
between DN T cells and APC are currently under study. The possibility of
"fratricide" of DN T cells as observed in CD8+ T cells is also under
investigation.
Taken together, the inventors have identified a novel subset of
regulatory T cells which down-regulate specific immune responses i n
vitro and in vivo. The inventors have also identified a novel mechanism
by which DN regulatory T cells mediate antigen-specific suppression.
These findings illustrate how donor-specific transplantation tolerance can
be achieved and explain how tolerance to self-antigens can be maintained
in the periphery. More importantly, the findings provide novel
therapeutic modalities for the prevention and treatment of graft rejection
and autoimmune diseases.
Methods for Example 1
Mice. C57BL/6 (B6, H-2b), (B6xBALB/c)Fl (H-2b~d), BALB/c-H-2-dm2 (dm2,
a BALB/c Ld loss mutant), C3H (H-2k), SJL (H-2S), B6/lpr (H-2b), MRL/+
(H-2k) and MRL/lpr (H-2k) mice were purchased from Jackson Laboratories
(Bar Harbor, ME). Anti-HY TCR transgenic mice were obtained from Dr.
H.S. Teh and anti-LCMV-gp transgenic mice is a gift of Dr. P. Ohashi. A
breeding stock of 2C transgenic mice was kindly provided by Dr. D. Y. Loh.
A large fraction of T cells in the periphery of the 2C mice express a
transgenic TCR reactive against Ld class I MHC. These T cells can be
detected by a clonotypic mAb 1B2 and are predominantly CD8+. The
specificity for Ld requires both transgenic a- and (3-chains. 2C transgenic
mice were first back-crossed onto B6 mice for more than 10 generations to

CA 02316089 2000-08-24
-56-
obtain the transgene on B6 background and then bred with dm2 mice. The
subsequent anti-Ld transgenic TCR+ (B62Cxdm2)F1 and transgenic TCR-
(B6xdm2)Fl mice (both H-2b~d, Ld-) were used for in vivo and in vitro
studies. All mice were maintained in the specific pathogen free animal
colony at the Ontario Cancer Institute.
DST, skin grafting and adoptive transfer of lymphocytes. (2Cxdm2)Fl mice
were either infused with viable spleen cells collected from (B6xBALB/c)Fi
mice (DST-treated) or left non-injected (control). One week later, all
(2Cxdm2)Fl mice were given skin grafts from both (B6xBALB/c)F1 and C3H
as previously reported. At 3 weeks after skin grafting, when all the
recipients in the control group rejected both (B6xBALB/c)Fl and C3H skin
grafts, and the DST-treated mice had accepted all (B6xBALB/c)Fl grafts and
rejected C3H grafts, the spleen cells were collected from either DST-treated
(tolerant) or control (non-tolerant) mice and adoptively transferred (5x10
cells/mouse) into unmanipulated naive syngeneic mice. The latter were
given skin grafts from both (B6xBALB/c)Fl and C3H mice one day after
injection of lymphocytes.
Cell sorting. Splenocytes were collected from tolerant mice (21 or 120 days
after skin grafting) or normal naive mice. Cells were stained with
FITC-labelled clonotypic mAb 1B2 (hybridoma was kindly provided by Dr.
H. Eilson, MIT) or FITC-conjugated anti-CD3 mAb, along with
PE-conjugated anti-CD4 mAb and Cy-Chrome conjugated anti-CD8 mAb
(all from PharMingen). The 1B2+/CD3+CD8+, 1B2+/CD3+CD4+ and
1B2+/CD3+CD8-CD4- T cells were sorted by using a cell sorter (Coulter Epics
V, Hialeah, FL) and the purity and viability of the cells after sorting were
more than 95%.
Generation and maintenance of 1B2+ T cell clones. Spleen cells were
collected from naive or tolerant (B62Cxdm2)F1 mice 120 days after
transplantation of (B6xBALB/c)Fl skin allografts. These cells were used to
generate T cell clones using standard cloning and subcloning procedures.
To maintain the T cell clones, 5x104 cells were cultured in a 24-well plate

CA 02316089 2000-08-24
-57-
containing 5x105 irradiated Ld+ cells in an a-Minimum Essential Media
(a-MEM) supplemented with 10% FCS and 30U/ml rIL-2 and 30U/ml
rIL-4. The cells were incubated at 37°C with 5% C02. The T cell clones
were stimulated in the above manner every 3-4 days, and used as
suppressor cells. From the initial 38 clones generated, 8 1B2+CD4-CD8-
clones and 4 1B2+CD4-CD8+ clones were grown successfully.
Cell Surface Marker Staining. T cell clones at various time points after
activation were stained with fluorescence-conjugated mAb specifically
recognizing the a(3-TCR (1B2), CD4, CDB, CD25, CD28, CD44, CD30, CD62L,
CD69, LFA-1, y8-TCR, and NK1.1 (PharMingen). Spleen cells from both
naive and tolerant mice were collected, triple stained with 1B2-Red 670,
CD8-FITC or CD4/CD8-FITC (both from PharMingen) and one of the
following PE-conjugated antibodies (anti-CD28 or anti-CD44, both from
PharMingen). Data were acquired and analyzed on an EPICS XL-MCL flow
cytometer (COULTER CORPORATION, Miami, FL).
MLR and suppression Assays. MLR: Naive splenic CD8+ T cells (1000
cells/well) were co-cultured in 96-well plates with irradiated (20 Gy)
sex-matched splenocytes (3x105 cells/well) from (B6xBALB/c)Fl mice in
a-MEM supplemented with 10% FCS and 30U/ml of rIL-2 and rIL-4 as
sources of growth factor. Various numbers of purified putative suppressor
T cells were added to MLR. After a 3-day incubation, 1Ci of [3H]-TdR was
added to each well. Eighteen hours later, cells were harvested and counted
in a beta counter. Cultures to which no putative suppressor cells were
added were used as controls. Suppression assays were also performed
using a transwell culture system (Costar). 1B2+DN T cell clones (TN02 or
TN012) were added to the top chambers (4x104 cells/well) together with
irradiated Ld+ stimulator cells. The top and bottom chambers were
separated by a 0.4-m membrane which allows soluble factors, but not cells,
to pass through. After 3 days of culture, the proliferation of 1B2+CD8+ cells
in the bottom chambers was measured by [3H]-TdR incorporation.

CA 02316089 2000-08-24
-58-
Cytotoxicity assays. Target cell death resulting from co-culture with DN T
cells was measured as previously reported. Briefly, DN T cells were
stimulated by irradiated allogeneic splenocytes for 2-3 days in the presence
of IL-2/IL-4. Viable cells were harvested and seeded into 96-well microtiter
plates as effector cells. Target cells were stimulated with appropriate
antigens in vitro for 2-3 days. Activated CD8+ T cells were collected,
labeled with 0.lmCi/ml of 5lCr for 1 hr or 10~,Ci/ml of [3H]-TdR at
37°C
overnight and used as targets. After co-culture with the effector cells at
37°C for either 4 or 18 hours (to measure perform-dependant and
Fas-dependant cytotoxicity, respectively) in the presence or absence of
irradiated allogeneic splenocytes, the cells were harvested and counted in a
gamma or beta counter. Specific cell lysis was calculated using the
equation: % Specific killing = (S-E)/S x 100, where E (experimental) is cpm
of retained DNA in the presence of effector cells, and S (spontaneous) is
cpm of retained DNA in the absence of effector cells.
RT-PCR analysis: 1B2+CD8+ or 1B2+DN T cells were activated by either
irradiated Ld+ spleen cells or plate-bound 1B2 antibody. Before activation,
and 4, 10, 20, and 90 hrs after activation, viable cells were collected for
RNA extraction using TriZol reagent (GIBCO, BRL). cDNA was prepared
from RNA with 0.5mg/ml pd(N)6 Random Hexamer Primer (Pharmacia)
and 300 units of murine MLV reverse transcriptase (GIBCO BRL). 2~,1 of
the cDNA mixture was used in a PCR reaction with 10 pmol of forward
and reverse primers and 2.5U of Taq DNA polymerase (Gibcol BRL). The
sequences of the specific sense and anti-sense oligonucleotide primer pairs
of 5' and 3' were the same as the inventors previously reported. Samples
were amplified through 35 cycles at an annealing temperature of 59°C in
a
PCR Thermal Cycler (MJ Research, Watertown, MA). The products were
separated on a 1.5% agarose gel by electrophoresis stained with ethidium
bromide.

CA 02316089 2000-08-24
-59-
Example 2
Identification and characterization of the genes that are crucial for DN T
cell-mediated immunosuRyression
The inventors have demonstrated that 1B2+ DN T cells and clones
from tolerant mice can specifically inhibit anti-Ld responses in vitro and in
vivo. Since agents such as IL-10 and CsA abrogate the suppressive
function of the DN T cells, it provides an excellent model to study the
differentially expressed molecules in regulatory and non-regulatory T cells.
Identification and characterization of the molecules responsible for
suppression will not only provide insight into mechanisms) leading to
Ag-specific suppression, but also may lead to the development of novel
therapeutic modalities.
The inventors have demonstrated that pre-treatment with IL-10
switches DN T cell clone (TN12) from a regulatory to a non-regulatory
phenotype (Figure 15a). This finding suggests that IL-10 may regulate the
expression of certain molecules crucial for DN T cell-mediated
suppression. Using PCR-Selected cDNA subtraction technique (83), the
inventors have, in a forward reaction, identified 14 genes that are
differentially expressed in regulatory and non-regulatory T cells. One of
the molecules highly expressed in the DN regulatory T cells encodes Ly-6A
(Figure 8a).
To determine the function of Ly-6A in DN T cell-mediated
immunosuppression.
Ly-6A (Seal-1 or TAP) is a glycosyl phosphatidylinositol
(GPI)-anchored cell surface molecule expressed on most peripheral
lymphocytes, thymocytes, and a variety of other cells (84-88). Ly-6A is
involved in cell adhesion and T cell activation (89-92). The consequence
of Ly-6A stimulation, however, may either enhance or inhibit T cell
activity (89, 93-96). The mechanisms underlying the differential roles of
Ly-6A on T cell activity is not known and the ligand for Ly-6A remains to
be identified.

CA 02316089 2000-08-24
-60-
The inventors have demonstrated that the suppression mediated
by DN T cells requires direct cell-cell contact with CD8+ T cells (Figure 4c).
Furthermore, these DN T cells do not express detectable levels of CD4,
CDB, CD28, or CTLA-4 (80), indicating that the classical co-stimulatory
molecules are not regulating DN T cell function. Interestingly, after
incubation with rIL-10, which converts the regulatory T cell phenotype
into a non-regulatory one, the expression of Ly-6A is significantly reduced
(Figure 8a). Moreover, the inventors data indicate that blocking the
expression of Ly-6A on TN12 clone using anti-sense oligonucleotides
abolished suppression (Figure 8b) and DN T cells from Ly-6A knockout
mice do not have suppressive function (Figure 8c).
Construction of a Ly-6A fusion protein and determine the function of the
fusion protein.
The results shown in Figure 8 demonstrate an important role of
Ly-6A in DN T cell mediated suppression. Therefore, the inventors will
generate a Ly-6A-Fc fusion protein for the following purposes: to study the
potential of the fusion protein in modulating the DN T cell mediated
suppression, and to identify and isolate the counterligand(s). Briefly, the
cDNA encoding the extracellular region of Ly-6A is amplified by PCR, and
inserted into a plasmid expressing mouse Fc~2a obtained from Dr. T Strom
(100) to create a fusion gene of Ly-6A and Fc~2a. The fusion gene will then
be cut from the original plasmid and cloned into the eukaryotic expression
vector pBK/CMV (Stratagene). Upon sequence confirmation, the
construct will be transfected into CHO cells by electroporation. Transfected
cells will be selected with 6418, and subcloned. High producing clones will
be selected by screening culture supernatants in ELISA using Ly-6A specific
mAb as the capture antibody, and Alkaline Phosphatase-coupled anti-Fc~2a
as the detection antibody. Western blot analysis will be used to confirm
the size and specificity of the Ly-6A-Fc using anti-Ly-6A and anti-Fc~2a
mAbs (both from PharMingen).

CA 02316089 2000-08-24
-61-
The ability of the Ly-6A-Fc protein to modulate the DN T cell
mediated suppression will first be determined in vitro. Anti-Ld MLRs will
be set-up to which DN regulatory T cell clones will be added at a 5:1 ratio.
Varying doses (for example, 1-50~,g/ml) of Ly-6A-Fc protein will be either
preincubated with the DN T cells, the stimulators or added directly into
the cultures at various time points. Proliferation and the number of CD8+
and DN T cells in the culture will be determined as described above. If the
addition of Ly-6A-Fc to the culture alters the DN-mediated Ag-specific
suppression in vitro, its role in allograft rejection will also be determined
in vivo. 2CF1-Tg- mice will be divided into 4 groups (10 mice per group).
One group of mice will be injected with DN regulatory T cells, one with
Ly-6A-Fc (for example, 50-100 g/mouse/day for 7 days), one injected with
both DN T cells and Ly-6A-Fc protein, and an additional control group will
be treated with saline. All mice will be transplanted with skin grafts from
both BYJF1 and C3H as described previously. The results from these
experiments will indicate whether Ly-6A-Fc has any role in preventing
allograft rejection.
Significance
Long-term graft rejection and complications arising from
immunosuppressive therapy remain major obstacles in clinical
transplantation. The solution to these problems requires a better
understanding of the mechanisms leading to donor-specific tolerance.
This application utilises DST-induced long-term donor-specific allograft
acceptance in mice as a model to identify the mechanisms of tolerance,
particularly the mechanisms of antigen-specific suppression mediated by
regulatory T cells. The results of these studies will provide significant
insights into the cellular and molecular mechanisms involved in the
induction and maintenance of Ag-specific tolerance. If the molecules
responsible for Ag-specific suppression can be identified, it could provide
the possibility for the substitution of DST with treatment at the molecular
level. Such findings will not only greatly broaden the application of DST

CA 02316089 2000-08-24
-62-
in clinical transplantation, but could also be applied for the treatment of
autoimmune disease.
Example 3
The dual function of re u~ lator3r T cells in anti-l3imphoma and~reventing
graft versus host disease
Transplantation of allogeneic lymphocytes could be a potent
therapeutic modality for the treatment of leukemia/lymphoma if graft
versus host disease (GVHD) could be prevented while graft versus
leukemia (GVL) effects could be maintained. Recent studies demonstrate
that pre-transplant donor-specific transfusion (DST) of one MHC molecule
mismatched lymphocytes leads to permanent acceptance of the skin or
cardiac allografts of the lymphocyte donor origin (Example 1). Because all
third party allografts were promptly rejected by the recipient, it indicates
that the tolerance is Ag-specific, and the recipients retained their immune
responses to other alloantigens. The inventors have recently analysed the
mechanism underlying DST-induced donor-specific tolerance, and
demonstrated that a novel subset of regulatory T cells play an important
role in the induction of antigen-specific tolerance (Example 1). The
regulatory cells can specifically lyse target cells that either express the
same
TCR or the specific antigen that can be recognized by the TCR, but not 3rd
party controls (Figure 5a and 5b). Preincubation of DN T cells with either
anti-TCR mAb (Figure 6c and 15b) or Fas-Fc fusion protein (Figure 5b)
abolish DN T cell mediated cytotoxicity. These findings suggest the
possibility of induction of tolerance to host MHC antigens while retain
anti-leukemia effect by infusion of one MHC mismatched allogeneic
lymphocytes, and that DN T cells may play an important role in down-
regulation of anti-host response and promoting anti-lymphoma effect.
Results
1. Absence of GVHD after infusion of non-fractionated one class I
mismatched allogeneic splenocytes.
To study whether encounter with one class I alloantigen in vivo
leads to tolerance of infused donor lymphocytes to the host rather than

CA 02316089 2000-08-24
-63-
causing GvHD, non-irradiated or sublethally irradiated (2Gy) ScidFl mice
were injected intravenously with 4x10 viable spleen cells from 2CF1 mice.
In this system, the donor and recipient are mismatched for one class I
locus Ld. The expected immune response would be 1B2+CD8+ cells from
the lymphocyte donor reacting to Ld expressed on the recipient mice. As
shown in Figure 9c, all the control C.B-17 ScidFl mice injected with B6
lymphocytes (n=5) developed an acute GvHD as evidenced by hunched
posture, ruffled fur, sever diarrhoea, weight loss and died within 2 weeks
after infusion of fully mismatched allogeneic lymphocytes. Histology
study revealed typical acute GvHD. In contrast, none of the ScidFl mice
(n=42) that received Ld mismatched allogeneic lymphocytes lost weight
(Figure 9c, insert) or showed any clinical signs of GvHD. They remained
healthy for the period of study (>150 days) (Figure 9c). No difference was
observed between irradiated and non-irradiated mice.
To exclude the possibility of mild GvHD, mice were sacrificed for
pathohistologic evaluation of GvHD between 100 and 150 days after
infusion of allogeneic lymphocytes. Tissue samples from each of the four
major sites of GvHD involvement (hepatic parenchyma, biliary system,
small intestine and skin) were harvested from recipient mice and
evaluated for lymphocytic infiltration and GvHD according to Fowler et al
{54}. No evidence for acute or chronic GvHD was observed. No difference
in the histology of liver, skin or small intestine could be observed between
normal and 2CF1-injected ScidFl mice.
To study whether the introduction of naive anti-host T cells could
break the tolerance and cause GvHD, three ScidFl mice were given a
second injection of naive 2CF1 allogeneic cells 6 weeks after the first
infusion, and GvHD was monitored for 180 days. Again, no signs of
GVHD were observed (not shown). These results are consistent with
observations by others and demonstrate that, although injection of fully-
or semi-allogeneic splenic cells resulted in the development of GVHD in

CA 02316089 2000-08-24
the recipients, infusion of one class I antigen (Ld) mismatched
unfractioned spleen cells does not cause GVHD in the recipients.
2. Obtaining anti-lymphoma effect in Scid~ mice
Next, the inventors investigated whether the ScidFl mice
reconstituted with Ld mismatched 2CF1 spleen cells can mediate an anti-
lymphoma response. ScidFl mice were i.v. injected with syngeneic A20
lymphoma cells (105/mouse) with (treated) or without (control) co-
injection of with 4x10 naive 2CF1 splenocytes on the same day. Both
groups of mice were monitored for the development of lymphoma. As
shown in Figure 9b all the ScidF1 mice that were injected with tumour
cells alone died of lymphoma between 28-42 days (median survival time
32 days, n=11) after injection of tumor cells. Those that died were
autopsied and all were found to have lymphoma. Histopathologic studies
revealed a very diffused infiltration in the liver by tumour cells.
Malignant cells were also found in the spleen and lymph nodes, but not in
lung, heart or kidneys (not shown). In striking contrast, all the ScidFl mice
(n=12) that received the same number of A20 tumour cells plus a single Ld
mismatched 2CF1 lymphocytes on the day of tumour cell inoculation
remained healthy for the period of study (>150 days) (Figure 9b). A normal
histological appearance was observed in all the major organs that had been
examined. Neither tumour infiltration nor GVHD was found in any
organ of any animals. Therefore, a profound anti-tumour ability had been
established in immunodeficient mice without causing GVHD. These
findings demonstrate that a single infusion of non-fractionated mature
anti-Ld allogeneic splenocytes can establish anti-lymphoma response in
immunodeficient mice without causing GvHD.
3. Reconstituted mice were tolerant to Ld while obtaining immunity to
other alloantigens.
The data from the above studies suggest that the infused anti
donor cells may have developed tolerance to host but not tumor antigens.
To further confirm that the infused donor cells developed tolerance only

CA 02316089 2000-08-24
-65-
to Ld, and still retained their capability to respond to other antigens, the
inventors studied the ability of the 2CF1 reconstituted ScidFl mice to reject
skin allografts. ScidFl mice were either infused with 2CF1 cells (treated) or
left untreated as controls. 3 weeks later, mice of both groups were given
skin grafts from (B6xC3H)F1 (semi-allogeneic to the recipient and
lymphocyte donor), and (B6xBALB/c)Fl (syngeneic to the recipient and
mismatched for Ld with the lymphocyte donor). Skin graft survival was
monitored as the inventors previously described. As shown in Figure 11,
all (B6xBALB/c)Fl skin grafts were permanently accepted by both untreated
and 2CF1-treated ScidFl mice. However, the (B6xC3H)F1 skin allografts,
while accepted by non-treated ScidFl mice, were rejected by all 2CFl-treated
ScidFl mice (Figure 11). These results are consistent with our previous
observations in skin graft model (27,28) and demonstrated that infusion of
non-fractionated viable anti-Ld spleen cells into Ld+ hosts leads to specific
tolerance of infused lymphocytes to Ld yet retain their ability to respond to
other alloantigens and tumor antigens.
4. Infusion of Ld mismatched donor lymphocytes provoke GVL in the
absence of GVHD in non-transgenic mice.
The above observations are based on a transgenic-scid model. This
model, although very useful for monitoring the fate and functional
properties of anti-host T cells and delineating mechanisms involved in
GVHD and GVL, might not represent pathophysiological responses in
normal mice. Therefore, it is important to validate the above findings in
non-transgenic mice. To this end, (B6xdm2)Fl mice (Db~d, Kb/d~ Lb/dm2)
were used as lymphocyte donors, and lethally (8.5 Gy) irradiated
(B6xBALB/c)Fl mice (Db~d, ICb/d, Lb/d) were used as recipients. In this
setting, there is also only one class I Ag (Ld) mismatch between donor and
recipient. This mimics the system used in the above studies but avoids
using both transgenic and scid mice. Lethally irradiated (B6xBALB/c)Fl
mice (n=16) were infused intravenously with a single dose (4x10~/mouse)
of viable splenic cells from sex-matched (B6xdm2)Fl mice. Half of them

CA 02316089 2000-08-24
-66-
(n=8) were also injected with 105 A20 syngeneic lymphoma cells. In
addition, some non-irradiated (B6xBALB/c)Fl mice were infused with A20
cells alone, and some were irradiated without the infusion of any cells.
Survival and GVHD were monitored. All lethally irradiated
(B6xBALB/c)F1 mice not given any treatment died within 1 week after
irradiation. Non-irradiated (B6xBALB/c)Fl mice died between 5-6 weeks
after infusion of A20 syngeneic lymphoma cells. Interestingly, 8/8 mice
infused with splenic cells from (B6xdm2)F1 mice survived over 180 days
without weight loss or any clinical sign of GVHD (Figure 10a, insert).
Histology studies at various time points after infusion of allogeneic
lymphocytes indicated no signs of GVHD in liver (Figure 10b, right panel),
skin and small intestine (not shown). Six of the 8 mice that were infused
with both (B6xdm2)Fl spleen cells and A20 tumor cells enjoyed tumour-
free survival for over 180 days (Figure 10a). These results confirm the
findings in transgenic-scid model, and demonstrate that infusion of Ld
mismatched spleen cells helps immune-incompetent animals to reject
tumour in the absence of GvHD without the need for any non-specific
immunosuppressive drugs.
5. Significant increase in 1B2+DN T cells in recipients after infusion of
2CF1 cells.
In order to identify the cells that are able to suppress anti-host
response and / or mediate anti-lymphoma and to understand the
underlying mechanisms, the inventors monitored the fate of infused
donor T lymphocytes in vivo. ScidFl mice were injected with 2CF1 spleen
cells alone or together with A20 cells. At various time points after
infusion, the spleen and lymph nodes were harvested from ScidFl
recipients, and the number of CD8+, CD4+, CD3+CD8- and NK1.1+ cells
were stained with appropriate mAbs and analyzed by using a flow
cytometer. A vigorous expansion followed by a massive apoptotic cell
death of 1B2+CD8+ cells was observed within the first few days after the
infusion of 2CF1 cells. At 3 weeks after injection, the majority of 1B2+CD8+

CA 02316089 2000-08-24
-67-
T cells was deleted from the periphery (Figure 9, left). On the other hand,
no significant change was observed in the number of 1B2+CD4+ T cells or
NK cells measured at 1, 3 and 8 weeks (not shown). These results are
consistent with previous reports. Notably, the number of 1B2+DN T cells
steadily increased more than 30-fold at 8 weeks after infusion. This
population of cells remained elevated for the period of study (>120 days)
(Figure 9a, left). Similar kinetic changes were observed in ScidF1 mice
infused with both 2CF1 and A20 cells, although proliferation of 1B2+CD8+
cells at the early stage was less robust and the increase in the number of
1B2+DN T cells was more vigorous (Figure 9a, right).
6. Specific suppression of anti-host T cells in vivo by previously infused
donor derived cells.
Accumulating evidence suggests that regulatory T cells play an
important role in prevention of autoimmune diseases and allograft
rejection. To investigate whether regulatory T cells also play a role in
preventing GvHD, male (B6xBALB/c)F1 mice were infused with male 2CFi
cells. Eight weeks after the first infusion, the recipient mice were given a
second injection of naive splenocytes from both male 2CF1 and female
anti-HY TCR transgenic mice. Some male naive (B6xBALB/c)F1 mice
were injected with the same number of 2CF1 and anti-HY cells as controls.
At 1 and 4 days after injection, the spleen and lymph nodes were
harvested, cells were stained with mAbs specific for 1B2, T3.70, and CDB,
and analyzed using flow cytometer. Both the percentage and total number
of anti-Ld and anti-HY CD8+ T cells in the spleen and lymph nodes of mice
that were given only one injection were similar to those that received two
injections. On day 4, however, the number of 1B2+CD8+ T cells, but not
anti-HY T cells, was significantly lower in the mice received 2 injections
than those received only one injection, suggesting proliferation of newly
infused naive anti-Ld T cells were specifically suppressed in vivo by
previously infused 2CF1 cells. Interestingly, the numbers of T3.70+CD8+ T
cells were comparable between the two groups both on day 1 and 4. These

CA 02316089 2000-08-24
-68-
data demonstrate that the donor-derived cells from previous injection are
able to specifically inhibit anti-host T cells but not a 3rd party T cells in
vivo.
7. 1B2+DN T cells induce apoptosis in anti-host 1B2+CD8+ but not anti-HY
T cells.
Extensive studies have been performed to delineate the role of
mature CD4+ and CDS+ donor T cells in GvH and GvL responses. By
contrast, little is done to reveal the function of mature DN T cells in this
context. As the majority of T cells in 2CF1-reconstituted mice are DN T
cells, it is possible that these donor-derived DN T cells are responsible for
inhibition of newly infused naive anti-host T cells and/or anti-lymphoma
effect. To test this hypothesis, 1B2+DN T cells were purified from 2CF1
reconstituted mice and used as effector cells. Their ability to kill activated
anti-host T cells were examined in vitro. The cytotoxicity was seen only
when 1B2+CD8+ T cells were used as target cells. No killing was seen when
a 3rd party anti-HY T cells were used as targets. These data provided direct
evidence that DN T cells can specifically kill activated 1B2+CD8+ T cells,
and suggest that DN T cells may prevent GVHD by killing activated anti-
host T cells.
8. DN regulatory T cells are able to kill lymphoma cells.
The inventors have shown that injection of 2CF1 and A20 cells
lead to a more vigorous increase in the number of DN T cells in ScidFi
mice than injection of 2CF1 cells alone (Figure 9a, right panel). The
inventors also demonstrated that the DN T cells are the major subset of T
cells in these mice. To determine whether DN T cells are directly
involved in eliminating A20 tumor cells, DN T cells were purified from
2CF1 reconstituted mice at 8 weeks after infusion, and their ability to lyse
A20 cells was determined in vitro. DN T cells specifically lysed A20 cells
but not 3rd party tumor cells. In order to investigate the role of DN T cells
in GvL, the inventors generated a panel of Ld-specific 1B2+DN T cell lines
from the spleen of 2CF1 mice. As seen in purified 1B2+DN T cells, these

CA 02316089 2000-08-24
-69-
1B2+DN T cell lines are also able to lyse A20 B cell lymphoma, but not 3rd
party tumor cells in vitro. To further evaluate the ability of DN T cell
lines in GvL and GvHD, a group of (B6xBALB/c)F1 mice were infused
with various doses of 1B2+DN T cell lines together with A20 cells. All the
mice that received A20 cells alone died between 32-38 days (n=10).
Interestingly, mice that infused with a lethal dose of A20 cells plus
5x106/mouse of either CN04 (n=10) or TN12 (n=5) 1B2+DN T cells lived
healthily over 100 days free from lymphoma and GVHD (Figure 12a).
Figure 12b shows the livers of two mice injected with A20 lymphoma cells
in the absence (left) or presence (right) of the DN regulatory cells. The
liver from the mouse that received the regulatory cells appears normal.
This is confirmed in Figure 12c which shows the histophathology of the
livers. These results demonstrate that allogeneic DN T cells generated i n
vitro are able to mediate anti-lymphoma response without causing GVHD.
Example 4
Induction of donor-specific transplantation tolerance by pretransnlant
infusion of one class I locus mismatched viable cells to enhance the
function of antigen-specific DN regulator3~ T cells in recipients
The differences in major histocompatibility complex (MHC),
human leukocyte antigens (HLA) in human, between donor and recipient
are the major mechanism of graft rejection. Therefore, in clinic bone
marrow and organ (such as kidney) transplantation, completely match for
6 HLA loci is sought. However, since HLA are highly polymorphic, it is
difficult to find a donor with all the 6 HLA loci matched with the recipient.
Moreover, even with a complete match for HLA, graft rejection still takes
place due to the incompatibility of minor histocompatibility antigens
between donor and recipient. The inventors have demonstrated in mice,
that donor-specific transfusion (DST) of one MHC molecule mismatched
lymphocytes before transplantation leads to permanent acceptance of skin
allografts of the lymphocyte donor origin. Because all third party allografts
were promptly rejected by the recipient, it suggests that the tolerance

CA 02316089 2000-08-24
-70-
induced by DST is Ag-specific, and the recipients retained their immune
responses to other alloantigens.
Furthermore, the inventors have demonstrated that tolerance
induced by one MHC locus mismatched DST can help to overcome the
rejection caused by multiple minor histocompatibility mismatches in both
transgenic and normal mice. Recently, the inventors have demonstrated
that the same strategies also successfully prevent cardiac allograft rejection
and graft versus host disease (Figure 9c and l0a-b). Although most of our
studies were done by using an MHC class I Ld mismatched model, similar
results have been reported by other groups that when one of other class I
loci or class II loci mismatches is used. DST can also enhance donor
specific cardiac survival. When transplanted patients were analysed
retrospectively according to the number of HLA mismatches between
donor and recipient, it was found that both graft and patient survival rate
is similar between patients who received one HLA mismatched grafts and
those that received HLA completely matched grafts. Taken together, these
findings suggest that instead of searching for HLA completely matched
donors, using one HLA locus mismatched graft donors can significantly
shorten the waiting time for transplantation and reduce mortality of
patients.
The inventors have demonstrated that the dose and time interval
between DST and transplantation is important for the outcome of the
transplant. Our results also indicate that promoting activation and
proliferation of DN T cells in recipients is one of the important
mechanisms of DST-induced donor-specific transplantation tolerance.
Furthermore, the inventors demonstrate that the survival and function of
DN T cells depend on exogenous IL-2 and IL-4. Therefore, co-injection of
rIL-2 and rIL-4 may further improve the activity of DN T cells. When
living donors are used (in cases of bone marrow, kidney and liver
transplantation), infusion of one HLA locus mismatched viable donor
lymphocytes 1-7days before transplantation will lead to specific tolerance to
the graft donor without impairing the immune responses of the recipient

CA 02316089 2000-08-24
-71-
to other antigens such as viruses. The inventors claim the method of
selecting graft donors mismatched for one HLA locus with recipients as
the best donor- recipient combination, and give one infusion of viable
lymphocyte mismatched for one HLA locus together with infusion of rIL-2
and rIL-4 between 1-7 days before transplantation without using any
immunosuppressive drugs.
The inventors have demonstrated that infusion of one class I
mismatched viable lymphocytes can induce permanent donor-specific
allograft survival. The underlying mechanism of DST-induced tolerance
is that recipient DN regulatory T cells are activated and expanded after
infusion of one class I molecule mismatched lymphocytes. As one class I
mismatched donors are still difficult to find, and infusion of cells should
be done at least one day prior transplantation, this therapy can only be
applied to very limited number of living donors. To overcome these
disadvantages, the inventors can transfect recipient cells with one HLA
antigen and use these cells to induce specific tolerance to that particular
HLA alloantigen.
Example 5
Monoclonal antibodies that can specifically bind to the surface and regulate
apo~tosis and function of regulator~r T cells
Accumulating evidence indicate that regulatory T cells play a very
important role in autoimmune diseases and allograft rejection. So far,
however, there is no antibody commercially available which can
exclusively recognize regulatory T cells. This is largely due to the inability
to isolate and clone regulatory T cells. As described in Example 1, the
inventors have identified and cloned novel DN regulatory T cells from
mouse spleen cells. In order to generate mAbs that can specifically
recognize regulatory T cells, the inventors used our DN regulatory T cell
clones to immunize rats and have produced 1,200 hybridomas. The
inventors also established an immunofluorescent assay by which 500-800
supernatants produced by hybridomas can be screened in one day. So far
the inventors have identified about 50 hybridomas of which the

CA 02316089 2000-08-24
-72-
supernatants can specifically recognize surface molecules on DN
regulatory, but not on CD8+ non-regulatory T cells (Figure 13a). Of the 6
hybridomas that have been cloned and subcloned, 25 positive clones are
able to produce mAbs that can exclusively recognize DN regulatory T cells
by using both a fluorescence plate reader and the flow cytometric analyses.
The inventors have demonstrated previously that DN regulatory
T cells also exist in normal animals (Example 1). Our preliminary results
show that some of the mAbs can also bind to DN T cell lines generated
from normal B6 mice and B6-lpr mice (Figure 13b). To study the effect of
these mAbs on the survival and function of DN regulatory T cells in vitro,
the inventors have performed the following experiments. To evaluate the
role of mAb in enhancing or abrogating the immunosuppressive function
of DN regulatory T cells the following experiment was performed.
Hybridoma supernatants were added to our convention suppression assays
as described elsewhere and percent suppression of proliferation of
1B2+CD8+ T cells by 1B2+DN T cells was compared with the cultures to
which no mAb were added. As shown in Figure 13d some of the mAb
supernatants can abolish the suppression of syngeneic CD8+ T cells
mediated by DN T cells. The inventors have demonstrated that the DN
regulatory T cells are resistant to activation induced apoptosis (Figure 14a).
To further determine whether the reversing of suppressive function of
DN regulatory T cells is due to the induction of apoptosis in DN T cells,
DN T cell clones were incubated in the presence or absence of TCR cross-
linking. At various time points after incubation, cells were collected and
stained with Annexin V a marker for early apoptosis. The number of
apoptotic DN T cells in each culture was determined FACS. As shown in
Figure 13c, some of the mAbs can convert our apoptosis resistant DN
regulatory T cell clones into apoptosis sensitive phenotype regardless of
cross-linking of TCR. This data indicate that some of the mAbs can bind to
surface molecules expressed on apoptosis-resistant cells and induce death
of these cells.

CA 02316089 2000-08-24
-73-
Example 6
Molecules that are highly expressed in DN T cells and important for the
function and survival of DN T cells
The inventors have demonstrated that pre-treatment with IL-10
switches DN T cell clone (TN12) from a regulatory to a non-regulatory
phenotype (Figure 15a). This finding suggests that IL-10 may regulate the
expression of certain molecules crucial for DN T cell-mediated
suppression. As described in Example 2, the inventors have identified
genes that are differentially expressed in regulatory and non-regulatory T
cells. In addition to the Ly-6A molecule discussed in Example 2, the
inventors have now determined that osteopontin is also highly expressed
on the regulatory cells. Northern blot analysis has confirmed a higher
level of expression of Eta-1 in the regulatory TN12 cells compared to C02
non-regulatory T cells and IL-10-treated TN12 cells (Figure 16a). A high
level expression of the Eta-1 gene product, Osteopontin, on DN regulatory
T cells has also been confirmed by flow cytometer (Figure 16b). The Eta-1
gene was originally found in early activated T cells after infection (R.P
Singh, J Exp Med, 1990,171:1931-6. R, Patarca, J Exp. Med, 1989, 170:145).
The inventors demonstrate that blocking of Osteopontin on DN T
cells with mAb can reverse the function of DN T cells (Figure 16c).
Human Ig Fc will be fused with the proteins that are specifically expressed
on DN regulatory T cells such as Ly-6A and Osteopontin. The Fc fusion
proteins can be used to identify their counter ligands.
Taken together, these novel cells and molecules provide new
revenues to specifically inhibit or enhance immune responses and novel
therapeutic modalities for the prevention and treatment of multiple
human diseases.
While the present invention has been described with reference to
what are presently considered to be the preferred examples, it is to be
understood that the invention is not limited to the disclosed examples. To
the contrary, the invention is intended to cover various modifications and

CA 02316089 2000-08-24
-74-
equivalent arrangements included within the spirit and scope of the
appended claims.
All publications, patents and patent applications are herein
incorporated by reference in their entirety to the same extent as if each
individual publication, patent or patent application was specifically and
individually indicated to be incorporated by reference in its entirety.

CA 02316089 2000-08-24
-75-
~
~C
~ + + + + + + + + +
, , , , , , , , , , , , ,
GIJ
A , , , , , , , , , , , ,
A
U
O
A + + + + + + + + + + + +
N
+ + + + + + + + + + + +
U
v
N
c~ +
, ~ ~ ~ ~ ~ + + + + +
A4 U
~
v
00
A , , , , , , , , + + + A +
O U
.,..,
~,
V ~ U
C~ ~ A + + + + + + + + + + + + +
'b
O ~ U
b
, , ~ ~ ~ ~ + + + + + w
U
C
A
, , , , , , , , , , ,
U 3
~
o b
N .
+ + + + + + + + + + + + +
~. Q
N N ~ N M ~' ,t~.-~ , ~ ~ V7
O
O ....O O O O O -~ ._. N t~-,0 ~ y ~
;t ~ ~ Z z z o 0 0
E- F-U CUU U U U F- U E-~- U a E- ~ ~'

CA 02316089 2000-08-24
-76-
References: .
1. F. Lancaster, Y. L. Chui, J. R. Batchelor, Nature 315, 336-337 (1985).
2. S. Qin et al., Science 259, 974-976 (1993).
3. Y. Chen, V. K. Kuchroo, J. Inobe, D. A. Hafler, H. L. Weiner,
Science 265, 1237-1240 (1994).
4. A. G. Baxter, S. J. Kinder, K. J. L. Hammond, R. Scollay, D. I.
Godfrey, Diabetes 46, 572-582 (1997).
5. H. Groux et al., Nature 389, 737-742 (1997).
6. A. Bushell, M. Niimi, P. J. Morris, K. J. Wood, J.Immunol. 162,
1359-1366 (1999).
7. D. W. Gjertson, Clinical transplants (UCLA Tissue Typing
Laboratory, Los Angeles,Califorina, 1991), pp. 225-235.
8. D. W. Gjertson, J. M. Cecka, P. I. Terasaki, Transplantation 60,
1384-1388 (1995).
9. G. Thiel et al., Transplant.Proc. 26, 2493-2498 (1994).
10. G. Opelz, Transplant.Proc. 24, 2342-2355 (1992).
11. J. J. Fung, A. W. Thomson, A. Pinna, R. R. Selby, T. E. Starzl,
Transplant.Proc. 24, 2372-2374 (1992).
12. A. M. Mowat, Immunol.Today 93-98 (1987).
13. L. Zhang, D. R. Martin, W. P. Fung-Leung, H. S. Teh, R. G. Miller,
J.Immunol. 148, 3740-3745 (1992).
14. L. Zhang et al., J.Immunol. 152, 2222-2228 (1994).
15. L. Zhang, W. P. FungLeung, R. G. Miller, J.1 m m a n o I. 155,
3464-3471 (1995).
16. L. M. Yang, B. DuTemple, Q. Khan, L. Zhang, Blood 91, 324-330
(1998).
17. L. M. Yang, B. DuTemple, R. M. Gorczynski, G. A. Levy, L. Zhang,
Tansplantation 67, 1404-1410 (1999).
18. A. Miller, O. Lider, A. B. Roberts, M. B. Sporn, H. L. Weiner,
Proc.Natl.Acad.Sci.USA 89, 421-425 (1992).
19. Y. Chen, J. Inobe, H. L. Weiner, J.Immunol. 155, 910-916 (1995).
20. H. L. Weiner et al., Annu.lZev.lmmunol. 12, 809-837 (1994).

CA 02316089 2000-08-24
21. S. P. Cobbold, E. Adams, S. E. Marshall, J. D. Davies, H.
Waldmann, Immunol.Rev. 5-33 (1996).
22. C. B. Anderson et al., Transplant.Proc. 271, 991-994 (1995).
23. D. Saitovitch, A. Bushell, D. W. Mabbs, P. J. Morris, K. J. Wood,
Transplantation 61, 1642-1647 (1996).
24. C. B. Anderson and D. C. Brennan, Transplant.Rev. 9, 49-62 (1995).
25. O. Jr. Salvatierra et al., Transplantation 40, 654-659 (1985).
26. O. Salvatierra et al., Transplant.Proc. XIX, 160-166 (1987).
27. W. H. Barber et al., Transplantation 51, 70-75 (1991).
28. E. van Twuyver et al., New Engl. j.Med. 325, 1210-1213 (1991).
29. E. L. Lagaaij et al., New EngI.J.Med. 701-705 (1989).
30. J. J. van Rood and F. H. J. Claas, Science 248, 1388-1393 (1990).
31. D. Middleton, J. Martin, J. Douglas, M. McClelland,
Transplantation 58, 845-848 (1994).
32. S. moue et al., Transplant.Proc. 28, 1220-1221 (1996).
33. L. P. de Waal and E. van Twuyver, Crit.Rev.Immunol. 10, 417-425
(1991).
34. S. S. Hori et al., J.Immunol. 143, 1447-1447 (1990).
35. E. van Twuyver et al., Transplantation 48, 844-847 (1989).
36. E. van Twuyver, W. M. Kast, R. J. D. Mooijaart, C. J. M. Melief, L.
P. de Waal, Eur. j.lmmunol. 20, 441-444 (1990).
37. H. W. Sollinger, M. Kalayoglu, F. O. Belzer, Ann.Surg. 315-321
(1986).
38. D. C. Brennan and M. W. Flye, Am.J.Kidney Dis. 26, 701-715 (1995).
39. W. M. Kast et al., Eur.J.lmmunol. 18, 2105-2108 (1988).
40. G. Lundgren et al., Lancet 2, 66-69 (1986).
41. Han.H.S., H. S. Jun, T. Utsugi, J. W. Yoon, J Autoimmune 9,
331-339 (1996).
42. S. Cobbold and H. Waldmann, Curr.Opin.lmmunol. 10, 518-524
(1998).
43. F. L. Ierino, K. Yamada, T. Hatch, J. Rembert, D. Sachs, J.Immunol
162, 550-559 (1999).

CA 02316089 2000-08-24
_78_
44. J.-G. Chai et al., Eur J Immunol 29, 686-692 (99 A.D.).
45. R. Quigley, K. J. Wood, P. J. Morris, j.lmmunol. 142, 463-470 (1988).
46. B. Charlton, H. J. Auchincloss, C. G. Fathman,
Annu.Rev.lmmunol. 12, 207-234 (1994).
47. J. Sprent, H. Kosaka, E. K. Gao, C. D. Surh, S. R. Webb,
Immunol.Rev. 133, 151-176 (1993).
48. W. C. Sha et al., Nature 335, 271-274 (1988).
49. W. C. Sha et al., Nature 336, 73-76 (1988).
50. D. M. Kranz, S. Tonegawa, H. N. Eisen, Proc.Natl.Acad.Sci.USA 81,
7922-7926 (1984).
51. Y. Chen et al., Nature 376, 177-180 (1995).
52. I. N. Crispe, Curr.Opin.Immunol. 6, 438-441 (1994).
53. P. S. Linsley, J. Bradshaw, M. Urnes, L. Grosmaire, J. A. Ledbetter, J
Immunol 150, 3161-3169 (1993).
54. X. Zhang, S. Sun, I. Hwang, D. F. Tough, J. Sprent, Immunity 8,
591-599 (1998).
55. C. Tanchot et al., Immunity 8, 581-590 (1998).
56. S. Strober, V. Palathumpat, R. Schwadron, B. Hertel-Wulff,
J.Immunol. 138, 699 (1987).
57. V. S. Abraham, D. H. Sachs, M. Sykes, J.Immunol. 148, 3746-3752
(1998).
58. S. Strober et al., Immunol.Rev. 149, 217-230 (1996).
59. F. Erard, M.-T. Wild, J. A. Garcia-Sanz, G. Le Gross, Science 260,
1802-1805 (1993).
60. E. Maggi et al., J.Exp.Med. 180, 489-495 (1994).
61. M. Sykes, K. A. Hoyles, M. L. Romick, D. H. Sachs, Cell.lmmunol.
129, 478 (1990).
62. B. M. Hall, Transplantation 51, 1141-1151 (1991).
63. P. L. Mottram, W. R. Han, L. J. Purcell, I. F. C. McKenzie, W. W.
Hannock, Transplantation 59, 559-565 (1995).
64. R. Bacchetta et al., J.Exp.Med. 179, 493-502 (1994).
65. L. Qin et al., J.Immunol. 156, 2316-2323 (1996).

CA 02316089 2000-08-24
-79-
66. S. Qian et al., Transplantation 62, 1709-1714 (1996).
67. X. X. Zheng et al., The Journal of Immunology 154, 5590-5600
(1995).
68. M. S. Lee, L. Wogensen, J. Shizuru, M. B. Oldstone, N. Sarvetnick,
J.Clin.lnvest 93, 1332-1338 (1994).
69. W. Li et al., Transplantation 66, 1587-1596 (1998).
70. L. Zhang, Eur.J.lmmunol. 26, 2208-2214 (1996).
71. Q. Khan, J. M. Penninger, L. M. Yang, L. E. K. I. Marra, L. Zhang, J
Immunol 162, 5860-5867 (1999).
72. L. S. Taams et al., Eur J Immunol 28, 2902-2912 (1998).
73. G. Lombardi, S. Sidhu, R. Batchelor, R. Lechler, Science 264,
1587-1589 (1994).
74. H. T. Lau, M. Yu, A. Fontana, C. J. Stoeckert Jr., Science 273, 109-112
(1996).
75. T. S. Griffith, T. Brunner, S. M. Fletcher, D. R. Green, T. A.
Ferguson, Science 270, 1189-1192 (1995).
76. D. Bellgrau et al., Nature 377, 630-632 (1995).
77. L. Zhang and R. G. Miller, Transplantation 56, 918-921 (1993).
78. M. Kopf et al., Nature 362, 245-248 (1993).
79. L. Zhang, R. G. Miller, J. Zhang, J.Exp.Med. 183, 2065-2073 (1996).
80. L. M. Yang, Z. X. Zhang, K. Young, B. DuTemple, L. Zhang, Nat.
Med. (under 2nd review), (1999).
81. G. S. Korbutt, J. F. Elliott, R. V. Rajotte, Diabetes 46, 317-322 (1997).
82. J. F. George et al., Nat.Med. 4, 333-335 (1998).
83. L. Diatchenko et al., Proc.Natl.Acad.Sci.U.S.A 93, 6025-6030 (1996).
84. A. J. Feeney and R. Riblet, Immunogenetics 37, 217-221 (1993).
85. K. P. Leclair et al., Proc.Natl.Acad.Sci.U.S.A 84, 1638-1643 (1987).
86. E. M. Shevach and P. E. Korty, Immunol.Today 10, 195-200 (1989).
87. H. Reiser et al., Cell 47, 365-370 (1986).
88. E. T. Yeh, H. Reiser, A. Bamezai, K. L. Rock, Cell 52, 665-674 (1988).
89. T. R. Malek, G. Ortega, C. Chan, R. A. Kroczek, E. M. Shevach,
J.Exp.Med. 164, 709-722 (1986).

CA 02316089 2000-08-24
-80-
90. Flood Patrick, Dougherty Joseph, Ron Yacov, J.Exp.Med 172,
115-120 (1990).
91. Sang-Kyou Lee, Bing Su, Stephen E.Maher, Alfred L.M.Bothwell,
EMBO journal. 13, 2167-2176 (1994).
92. Anill Banezai and Kenneth L.Rock, J.Immunol. 92, 4294-4298
(1995).
93. E. K. Codias, J. E. Butter, T. J. Fleming, T. R. Malek, J. I m m a n o 1.
145,1407-1414 (1990).
94. W. L. Stanford et al., J.Exp.Med 186, 705-717 (1997).
95. E. T. Yeh, H. Reiser, B. Benacerraf, K. L. Rock, J.Immunol. 137,
1232-1238 (1986).
96. J. J. Sussman et al., Nature 334, 625-628 (1988).
97. E. Suri-Payer, A. Z. Amar, A. M. Thornton, E. M. Shevach,
j.lmmunol. 160, 1212-1218 (1998).
98. S. Sakaguchi, T. Takahashi, Y. Nishizuka, J.Exp.Med. 156,
1577-1586 (1982).
99. S. Sakaguchi, T. Takahashi, Y. Nishizuka, J.Exp.Med. 156,
1565-1576 (1982).
100. S. J. Davis and P. A. van der Merwe, ImmunoLToday 17, 177-187
(1996).
101. A. O. Hueber et al., Curr.Biol. 7, 705-708 (1997).
102. A. W. O'Regan et al., J.Immunol. 162, 1024-1031 (1999).
103. G. F. Weber, S. Ashkar, M. J. Glimcher, H. Cantor, Science 271,
509-512 (1996).
104. L. S. Davis, S. S. Patel, J. P. Atkinson, P. E. Lipsky, J.Immunol. 141,
2246-2252 (1988).
105. P. E. Korty, C. Brando, E. M. Shevach, j.lmmunol. 146, 4092-4098
(1991).

Dessin représentatif

Désolé, le dessin représentatif concernant le document de brevet no 2316089 est introuvable.

États administratifs

2024-08-01 : Dans le cadre de la transition vers les Brevets de nouvelle génération (BNG), la base de données sur les brevets canadiens (BDBC) contient désormais un Historique d'événement plus détaillé, qui reproduit le Journal des événements de notre nouvelle solution interne.

Veuillez noter que les événements débutant par « Inactive : » se réfèrent à des événements qui ne sont plus utilisés dans notre nouvelle solution interne.

Pour une meilleure compréhension de l'état de la demande ou brevet qui figure sur cette page, la rubrique Mise en garde , et les descriptions de Brevet , Historique d'événement , Taxes périodiques et Historique des paiements devraient être consultées.

Historique d'événement

Description Date
Inactive : CIB désactivée 2015-03-14
Inactive : CIB du SCB 2015-01-17
Inactive : CIB expirée 2015-01-01
Inactive : CIB enlevée 2014-11-03
Inactive : CIB enlevée 2014-11-03
Inactive : CIB attribuée 2014-11-03
Inactive : CIB enlevée 2014-11-03
Inactive : CIB attribuée 2014-11-03
Inactive : CIB enlevée 2014-11-03
Inactive : CIB enlevée 2014-11-03
Inactive : CIB attribuée 2014-11-03
Inactive : CIB attribuée 2014-11-03
Inactive : CIB enlevée 2014-11-03
Inactive : CIB enlevée 2014-11-03
Inactive : CIB désactivée 2013-11-12
Inactive : CIB attribuée 2013-01-07
Inactive : CIB enlevée 2013-01-07
Inactive : CIB attribuée 2013-01-07
Inactive : CIB en 1re position 2013-01-07
Inactive : CIB enlevée 2013-01-07
Inactive : CIB expirée 2010-01-01
Inactive : CIB de MCD 2006-03-12
Inactive : CIB de MCD 2006-03-12
Inactive : CIB de MCD 2006-03-12
Inactive : CIB de MCD 2006-03-12
Inactive : CIB de MCD 2006-03-12
Inactive : CIB de MCD 2006-03-12
Inactive : CIB de MCD 2006-03-12
Inactive : CIB de MCD 2006-03-12
Demande non rétablie avant l'échéance 2004-08-24
Le délai pour l'annulation est expiré 2004-08-24
Réputée abandonnée - omission de répondre à un avis sur les taxes pour le maintien en état 2003-08-25
Inactive : Grandeur de l'entité changée 2002-08-26
Inactive : Page couverture publiée 2001-04-08
Demande publiée (accessible au public) 2001-04-08
Inactive : CIB en 1re position 2000-10-05
Inactive : CIB attribuée 2000-10-04
Inactive : CIB en 1re position 2000-10-04
Inactive : CIB attribuée 2000-10-04
Inactive : CIB attribuée 2000-10-04
Inactive : CIB attribuée 2000-10-04
Inactive : CIB attribuée 2000-10-04
Inactive : CIB attribuée 2000-10-04
Inactive : CIB attribuée 2000-10-04
Inactive : CIB attribuée 2000-10-04
Demande de priorité reçue 2000-09-29
Lettre envoyée 2000-09-19
Inactive : Certificat de dépôt - Sans RE (Anglais) 2000-09-15
Exigences de dépôt - jugé conforme 2000-09-15
Demande reçue - nationale ordinaire 2000-09-07

Historique d'abandonnement

Date d'abandonnement Raison Date de rétablissement
2003-08-25

Taxes périodiques

Le dernier paiement a été reçu le 2002-08-07

Avis : Si le paiement en totalité n'a pas été reçu au plus tard à la date indiquée, une taxe supplémentaire peut être imposée, soit une des taxes suivantes :

  • taxe de rétablissement ;
  • taxe pour paiement en souffrance ; ou
  • taxe additionnelle pour le renversement d'une péremption réputée.

Les taxes sur les brevets sont ajustées au 1er janvier de chaque année. Les montants ci-dessus sont les montants actuels s'ils sont reçus au plus tard le 31 décembre de l'année en cours.
Veuillez vous référer à la page web des taxes sur les brevets de l'OPIC pour voir tous les montants actuels des taxes.

Historique des taxes

Type de taxes Anniversaire Échéance Date payée
Taxe pour le dépôt - petite 2000-08-24
TM (demande, 2e anniv.) - générale 02 2002-08-26 2002-08-07
Titulaires au dossier

Les titulaires actuels et antérieures au dossier sont affichés en ordre alphabétique.

Titulaires actuels au dossier
LI ZHANG
ZHU XU ZHANG
KEVIN YOUNG
Titulaires antérieures au dossier
S.O.
Les propriétaires antérieurs qui ne figurent pas dans la liste des « Propriétaires au dossier » apparaîtront dans d'autres documents au dossier.
Documents

Pour visionner les fichiers sélectionnés, entrer le code reCAPTCHA :



Pour visualiser une image, cliquer sur un lien dans la colonne description du document. Pour télécharger l'image (les images), cliquer l'une ou plusieurs cases à cocher dans la première colonne et ensuite cliquer sur le bouton "Télécharger sélection en format PDF (archive Zip)" ou le bouton "Télécharger sélection (en un fichier PDF fusionné)".

Liste des documents de brevet publiés et non publiés sur la BDBC .

Si vous avez des difficultés à accéder au contenu, veuillez communiquer avec le Centre de services à la clientèle au 1-866-997-1936, ou envoyer un courriel au Centre de service à la clientèle de l'OPIC.


Description du
Document 
Date
(aaaa-mm-jj) 
Nombre de pages   Taille de l'image (Ko) 
Description 2000-08-23 80 4 181
Revendications 2000-08-23 6 156
Dessins 2000-08-23 19 409
Abrégé 2000-08-23 1 12
Certificat de dépôt (anglais) 2000-09-14 1 163
Rappel de taxe de maintien due 2002-04-24 1 111
Courtoisie - Lettre d'abandon (taxe de maintien en état) 2003-09-21 1 176
Correspondance 2000-09-14 1 8
Correspondance 2000-09-28 1 33
Taxes 2002-08-06 1 40