Tumour
immunosurveillance : the spontaneous recognition and elimination of
tumours by T cells.
In the 1950s, it was shown that the immune system could recognize and
respond to tumor-associated
antigensref.
However, reports in the 1970s that nude
mice
do not demonstrate an increase in tumor incidence were considered by some
to contradict the hypothesis of tumor immunosurveillanceref.
The hypothesis was rejuvenated by Robert Schreiber (St.Louis, USA) who
reported an increase in the incidence of chemically induced tumors, and
wider spectrum of tumors deficient in the tumor suppressor p53, in mice
defective in responsiveness to IFN-gref1,
ref2,
ref3.
Those studies also could explain results obtained using nude mice that,
though deficient in lymphocytes, have IFN-g-producing
innate immune cells, such as natural killer (NK) cells. Papers from Mark
Smyth (East Melbourne, Australia) have provided further evidence for immune
surveillance and have shown that perforin-mediated killing is an important
mechanism in tumor immunityref1,
ref2,
ref3.
Perforin functions as a tumor suppressor in mice that have a variety of
molecular genetic defects, including v-abl transgenic mice and mice
deficient in mlh1 (a DNA-mismatch repair gene) and p53, that,
when crossed with perforin-deficient
mice, produce offspring that have increased plasmacytomas and lymphomasref.
An important issue is whether these mouse models relate to human diseases.
Notably, some patients with lymphoma
may have mutations in the gene encoding perforinref.
Metastatic cancers is a systemic disease that is expected to be monitored
by ...
killer cells
(e.g. wild-type mice reject transplants of MHC class-I-defient spontaneous
B-cell
lymphomas
by NK cells and gd T cellsref)
expression of NKG2D
ligands confers an effective barrier to tumour formation as they are
targeted by NK cells
and gd T lymphocytes
: Upregulation of the inducible gene products MICA (human) and Rae-1 (mouse)
may promote tumor surveillance and autoimmunity by engaging the activating
receptor NKG2D on NK cells and T cells. Nevertheless, sustained expression
of MICA by tumors can also elicit NKG2D downregulation, perhaps indicating
'immunoevasion'. Investigating this paradox, we report here that constitutive
Rae-1e transgene expression in normal epithelium
elicited local and systemic NKG2D downregulation, generalized but reversible
defects in NK cell-mediated cytotoxicity and mild CD8+ T cell defects.
The extent of NKG2D downregulation correlated well with the incidence and
progression of cutaneous carcinogenesis, emphasizing the utility of NKG2D
as a marker of tumor resistance. Thus, NKG2D engagement is a natural mediator
of immunosurveillance, which can be compromised by locally sustained ligand
expression but potentially restored by innate immune activationref.
If other stem cells or tissues upregulate expression of NKG2D ligands after
transplantation, NKG2D may contribute to allogeneic bone marrow graft rejection
in immunocompetent hostsref.
down-regulation of MHC class I expression : NK
cells
inhibitory receptors are no longer activated
Th1-cell immunity
that promotes CTL
response
: the importance of perforin
in tumour surveiilance is clear from studies of perforin-deficient mice,
which spontaneously develop B-cell lymphomas. Granzymes
A and B
are not essential for CTL- or NK-cell-mediated rejection of spontaneous
or experimental tumours.
MHC class I processing pathway :
cross-presentation
of tumor antigens : heat shock proteins (HSPs) family
members released in the interstitium from necrotic cancer cells carry donor
Ags for endocytosis and cross-presentation by APCs
TAP-independent pathways are useful for TAP-deficient
cells, e.g. in those tumors where there is a down-regulation of TAP (see
below)
extralymphatic tumor cells that lack conventional
second signals efficiently induce CD8+ T cells if they reach
draining secondary
lymphoid organs
and if helper T cells are availableref.
when autologous dendritic cells that had taken up
apoptotic B lymphoma cells and induced to a fully mature state were used
to stimulate nonadherent PBMCs from healthy donors, they induced strong
cytotoxicity against B lymphoma cells in a HLA-A0201-restricted manner.
The cells failed to induce cytotoxicity, however, when purified T cells
were used as effector cells. Depletion of CD56+ cells, but not
CD14+ or CD19+ cells, abrogated the cytotoxicity
of nonadherent cells, showing that the help was provided by natural killer
cells. Further, when natural killer cells were present in the cultures,
a strong and persistent production of IL-18,
but not IL-12 and IL-15, was observed. Blocking IL-18 significantly reduced
the cytotoxicity of nonadherent cells against B lymphoma cellsref
IFNs contribute to tumor suppression by inducing p53
expression and boosting its response to stress signals, leading to an enhancement
of cancer cell apoptosis
TNF and TNFR superfamilies : anti-tumor mechanisms could be classified
into direct actions onto tumor cells and indirect effects through immune
or non-immune components of tumor-bearing hostref
E.g. :
signs of a Th1 immune response
within colorectal cancers
are associated with the absence of pathological evidence of early metastatic
invasion (venous emboli and lymphatic and perineural invasion) and with
prolonged survivalref
Many primary tumours originate at mucosal sites in which they
are first encountered by the mucosal immune system : most experiments carried
out with animal models available at present, and expecially with transplantable
tumour that grow in subcutaneous sites, do not shed light on this subject.
There may be fundamental differences between the development of tumours
injected into a mouse or induced by chemical carcinogens compared to naturally
occurring spontaneous tumors in humans. For example, the act of injection
itself and the presence of dead tumour cells as well as tissue-culture
contaminants, such as fetal bovine serum in the inoculum, may induce local
inflammatory reactions. In tumours induced by painting chemical carcinogens
onto the skin of highly susceptible mice, Rae-1b and H-60 expression may
be up-regulated, resulting in their rejection by gd
T lymphocytes.
In addition, whereas carcinogenesis in humans may be a slow process that
occurs over several years, mouse experiments use high doses of carcinogens,
which cause transformation in just weeks or months. At these doses, carcinogens
may cause massive and rapid mutagenesis, resulting in greatly increased
number of neoantigens that may alert the immune system.
Although
a model explains why established tumors grow and do not undergo immune-mediated
rejection is that tumors function as immunologically normal tissue, as
evidence exists that in some models and under some conditions the immune
system can play an active role in suppressing the growth of very early
tumours (immune attack), in this paradigm, when tumour do successfully
grow, they are thought of as having escaped from this immunosurveillance
due to natural selection of tumour immune "escape" or "evasion"
phenotypes as viewed in a Darwinian light (cancer immunoediting).
The central engine for generating immunoresistant tumour cells variants
is the genomic instability and dysregulation that is characteristic of
the transformed genome. Tumor-induced immunosuppressionref
generates ...
STAT3
is costitutively activated in many tumour cells, inhibiting the production
of pro-inflammatory mediators including TNF-a,
IL-6,
CCL5
/ RANTES
and CXCL10
: blocking STAT3 signalling with specific inhibitors leads to DC activation
(mediated by induction of STAT3 in the DCs themselves) and could have direct
anti-tumour effects. Further transformed cells directly block DC maturation
by secreting inhibitory facotrs that are induced by Stat3 activity, diverse
depending on the tumour type : VEGF from B16 tumours, IL-10 from Src-transformed
fibroblasts, ...ref
a new model of tumour angiogenesis in which DEFB29
recruits CD11c+ immature DCs that have the potential to adopt
endothelial characteristics (endothelial-cell markers, such as CD34 and
VE-cadherin) in the presence of VEGF via VEGFR2,
without proliferation (ruling out the involvement of a stem-cell population),
increasing tumour growth through vasculogenesis. This model is also applicable
to human ovarian tumours, which express endogenous b-defensins.
By promoting the endothelial specialization of immature DCs, the tumour
might not only ensure a sufficient blood supply but also prevent these
DCs from initiating an immune response against tumour antigens. In this
case, therapeutic targeting of this pathway could both reduce tumour growth
and promote immune attack. Interestingly, tumour-infiltrating CD11c+ cells
could present antigen to T cells when removed from the tumour milieu in
vitro, showing that these cells can function as endothelial-like cells
or DCs depending on the environmentref
B16 melanoma cells actively modulate the interaction between DCs derived
from bone marrow precursors and NK/LAK cells propagated from the spleen
of C57BL/6 mice. DCs increased in a dose-dependent manner the ability of
NK/LAK cells to kill melanoma cells and to produce cytokines. This activatory
cross-talk entailed the production of IL-18 by DCs and of IFN-g
by NK/LAK cells. Melanoma cells were not a passive target of NK activity;
they regulated the outcome of the interaction between DCs and NK/LAK cells,
inhibiting the in vitro production of cytokines as effectively as
the genetic deletion of IL-18 or IFN-gref
loss
or down-regulation of MHC class I antigens in tumor cells : anyway
loss of H-2 expression in TAP1-/- or LMP-2-/- mice
does not increase the onset or incidence of a variety of spontaneous tumors.
On the other hand, some recurring tumors lose cell-surface MHC class I
both in mice with pre-existing immunity induced by immunization with B7-1-transfected
tumor cells and in humans after partial responses to T cell-based immunotherapy
total loss of HLA class I expression is not uncommon in many tumors,
including melanoma,
colorectal
carcinoma
and prostate
adenocarcinoma.
In breast carcinoma,
the frequency of total HLA class I loss is > 50%. The mechanisms than underlie
total loss of HLA class I include
mutations in one copy of the b2-microglobulin
gene in association with LOH involving the second allele : the loss of
b2-microglobulin
has been observedin patients experiencing objective partial responses after
T-cell based immunotherapy, although a causal relationship in clinical
scenarios is difficult to demonstrate unequivocally
defects in MHC genes and in the antigen processing and transport pathway
selective loss of HLA class I haplotype, locus or allele. HLA haplotype
loss can be due to LOH on chromosome 6. Several mechanisms are involved
in locus down-regulation, which is more frequent with HLA-B than HLA-A
antigens. In melanomas, c-Myc oncogene overexpression correlates with selective
HLA-B locus down-regulation. Loss of transcription factor binding to locus-specific
regulatory elements can induce HLA-B locus down-regulation in colorectal
carcinoma
cells. In melanoma,
the gene products of the HLA-C locus are often expressed poorly or not
at all. The defects underlying HLA class I allele-specific loss include
mutations in the genes encoding HLA class I heavy chain.
in leukaemic
cells,
downregulation of HLA-B expression is detected in most of the samples,
but is restricted to HLA-Bw6 alleles, which - in contrast to HLA-Bw4 alleles
- are not thought to be involved in inhibiting the attack by NK cells,
so that selective HLA-Bw6 donregulation could prevent attack by CTLs while
avoiding NK-cell killingref
Explanations for why tumor cells that have lost HLA class I are not
destroyed by NK cells :
loss or downregulation of NKG2D
ligands expression (see below)
lack of expression of costimulatory molecules CD80
/ B7-1,
CD86
/ B7-2 / B7.2,
CD40
and CD70
by tumors may also hinder optimal NK cell activation via CD28
and CD27
costimulatory pathways
immunosuppressant cytokines such as TGF-b1
or MIF
produced by tumor cells
direct cytotoxicity of tumor cells by iNKT
cells
is common to both humans and mice, and that the high expression level of
CD1d
may be a predictor whether the tumor is a good target of iNKT cellsref.
lack of appropriate costimulation
lack of expression of costimulatory molecules by
tumor cells may lead to T cell anergy and suboptimal activation of NK cells.
In an experimental setting, insertion of genes encoding CD80
/ B7-1,
CD86
/ B7-2 / B7.2
or both into tumors generally increases the immunogenicity of those tumors
but does not necessarily lead to regression
constitutive activation of STAT3,
a common oncogenic signaling pathway, suppresses tumor expression of proinflammatory
mediatorsref
most human cancer cells express PD-L1
/ B7-H1,
impairing of T cell function and survival. Patients harboring high intratumoral
expression levels of B7-H1, contributed by renal
cell carcinoma (RCC)
cells alone, lymphocytes alone, or tumor and/or lymphocytes combined, exhibit
aggressive tumors and are 4.5 times more likely to die from their cancer
than patients exhibiting low levels of B7-H1 expression (RR 4.53)ref
other undefined soluble factors or cell-surface molecules expressed by
tumour cells
LIGHT
stimulates tumour rejection via recruitment and costimulation of naive
CTLs inside tumoursref
using an experimental system utilizing the adoptive transfer of transgenic
T cells into naive recipients, the population of Gr-1+
immature dendritic cells (iDC) from tumor-bearing mice was able to induce
CD8+ T cell tolerance. These iDC accumulate in large numbers
in spleens, lymph nodes, and tumor tissues of tumor-bearing mice and are
comprised of precursors of myeloid cells. Neither iDC from control mice
nor progeny of tumor-derived iDC, including tumor-derived CD11c+
DCs, were able to render T cells nonresponsive. iDC are able to take up
soluble protein in vivo, process it, and present antigenic epitopes
on their surface and induce Ag-specific T cell anergy. Thus, this is a
first demonstration that in tumor-bearing mice CD8+ T cell tolerance
is induced primarily by iDC that may have direct implications for cancer
immunotherapyref.
immunosuppressive cytokines that tumours produce
VEGF (secreted by gastric carcinoma,
lung, head and neck, and breast cancers) inhibits DC differntiation and
maturation through suppression of NF-kB
in HSCs.
IL-10
inhibits differentiation of DCs from stem cell precursors and compromises
maturation and the functional status of DCs. It inhibits antigen presentation,
IL-12
production and induction of Th1
responses in vivo. IL-10 also enhances spontaneous DC apoptosis
as well as susceptibility to autologous NK cell lysis. IL-10 may protect
tumor cells from CTLs by down-regulation of HLA classes I and II and ICAM-1.
The loss of
HLA class I expression could also be due to IL-10-mediated down-regulation
of TAP1 and TAP2 proteins in tumor cells. PGE2
is expressed as a result of enhanced expression of COX-2
in multiple human tumours : PGE2 increases the production of
IL-10 by lymphocytes and macrophages and inhibits IL-12 production by macrophages.
TGF-b1
may also be released by cells dying apoptotically. It inhibits the activation,
proliferation, and activity of lymphocytes in vivo.
Tumors may not necessarily produce these cytokines as escape mechanisms
: the hypothesized immunosuppressive functions may be mere side-effects
of the angiogenic and growth factor functions of these cytokines.
recently, a number of endogenous molecules have been reported to be ligands
of TLRs.
It has been suggested that the release of these putative endogenous ligands
of TLRs during cancer progression may cause chronic inflammation leading
to the recruitment of myeloid suppressor cells and down-regulation of T-cell
and natural killer (NK) cell receptor z chain
resulting in T and NK cell dysfunction. However, the reported putative
endogenous TLR ligands may have been contaminated with PAMPsref.
loss of tumor antigens and immunodominance
: decreased expression of melanoma-melanocyte
differentiation antigens (MDAs)
is associated with disease progression. In one study, cells were MART-1+
in 100% of stage I lesions but only 75% of stage IV lesions. Decreased
antigen expression has also been found in residual tumors after peptide
vaccination. With a gp100
peptide (209-2M) vaccine, there was a decrease in gp100 expression in tumors
after treatment (47% versus 32%), whereas expression of MART1 was unchanged
(54%). In addition, the amount of tumor antigen expressed may also be important
for recognition. Again, these are correlative studies and fully controlled
human studies can be difficult or impossible to come by. It seems likely,
however, that as T cell-based tumor immunotherapy becomes stronger, escape
mechanisms such as antigen loss are likely to become more prominent. The
exact mechanisms that control the down-regulation of tumor antigens are
not known in most cases; however, the propagation of such antigen loss
variants may be facilitated by epitope
immunodominance.
The theory of immunodominance, as it relates to tumor escape, predicts
that one of the ways that antigen-loss variants within a tumor are shielded
from immune pressure is that the parental tumor cells that carry the immunodominant
epitope serve as a red flag for immune attack, thereby diverting attention
fromm the tumor variants. Once the parental cells are eliminated, a new
hierarchy is established among the variant subpopulations, and formerly
immunorecessive epitopes become dominant. A tumor variant that has lost
the restricting HLA class I allele while retaining the immunodominant antigen
could cross-present this antigen to CD8+ CTLs by DCs and maintain
an immunodominant response to a "phantom" target at the expense of more
appropriate and effective responses to other antigens.
defective death receptor signaling render tumor
cells resistant to death receptor-mediated apoptosis
no signalling from the death-inducing signalling complex (DISC) due to
FADD mutation, X-linked inhibitor of apoptosis protein (XIAP) inhibition
of caspase-3 and low second mitochondria-derived activator of caspase (SMAC)
/ direct IAP binding protein with low pI (Diablo) release
low expression of death receptors by post-transcriptional regulation
AICD in human melanoma epitope-specific primary CTL involves selective
mitochondrio-nuclear translocation of AIF without cytochrome c release,
caspase-3 and caspase-8 activation, and results from large-scale DNA fragmentation.
The JNK inhibitor, SP600125, blocks the mitochondrio-nuclear translocation
of AIF and prevents AICD in these CTL. These findings suggest that the
AICD in human melanoma epitope specific primary CTL is mediated by mitochondrial
AIF release and JNK is involved in regulation of this death processref.
expanded CD4+CD25+
Treg lymphocytes
in response to the increased presentation of autoantigens during tumour
growth. Their depletion plus injection of an anti-CTLA-4
antibody
enhanced reactivity to a known TAA.
tumor tissue and ascites from patients with ovarian cancer contain high
levels of cells with all the hallmarks of regulatory T cells. These cells
migrate into the tumor microenvironment in a process mediated by the chemokine
CCL22,
and are capable of suppressing antitumor responsesref
the AE17 murine mesothelioma model was used for this study. Both intra-tumoural
Treg cells and those in the periphery of tumour-bearing mice
were identified by flow cytometry. The effect on tumour growth of intra-tumoural
depletion of Treg cells using the PC61 anti-CD25 mAb was then
examined. CD4+ Treg cells co-expressing both the
CD25 cell surface marker and the transcription factor Foxp3 were identified
within murine mesotheliomas. These intra-tumoural Treg cells
increase significantly as a percentage of total CD4+ T cells
within the tumour as it grows. Depletion of intra-tumoural Treg
cells with anti-CD25 mAb injected directly into the tumours can cause significantly
reduced tumour growth. Localised, intra-tumoural depletion of Treg
cells is a new, clinically relevant treatment option for established tumoursref.
intratumoral Treg cells inhibit the proliferation and granule
production of activated autologous infiltrating CD8+ T cells.
Degranulation and subsequent cytotoxic activity of infiltrating CD8+
T cells exposed to lymphoma B cells is completely attenuated by the presence
of intratumoral Treg cells. Furthermore increased numbers of
intratumoral Treg cells correlates with the number of CD8+
T cells in biopsy specimens from patients with B-cell NHL, supporting the
in vitro findings that intratumoral Treg cells inhibit proliferation
of infiltrating CD8+ T cells. Taken together, these data indicate
that human lymphoma B cells are sensitive to autologous CTL-mediated cell
death but are protected by the inhibitory function of intratumoral Treg
cellsref.
an increased frequency of Treg was demonstrated in both peripheral
blood and mesenteric lymph nodes of patients with colorectal cancer (CRC)
compared with either healthy controls or patients with inflammatory bowel
disease (IBD). Depletion of Treg from peripheral blood mononuclear
cells (PBMC) of CRC patients unmasked CD4+ T cell responses,
as observed by IFN-g release, to the tumor associated
antigen 5T4, whereas no effect was observed in a healthy age-matched control
groupref
improper activation of macrophages and polymorphonuclear cells induces
a state of oxidative stress in cancer patients that markedly suppresses
the function of T cells.
the presence of leukocytes in human tumors was first noted in neoplastic
tissues by Rudolf Virchow in 1863 (Virchow R. Aetiologie der neoplastischen
Geschwulste/Pathogenie der neoplastischen Geschwulste. In: Hirschwald VvA,
ed. Die Krankhaften Geschwustle. Berlin, Germany;1863: 57-101; Virchow
R. Reizung und Reizbarkeit. Arch Pathol Anat Klin Med. 1858;14: 1-63).
He suggested that the "lymphoreticular infiltrate" reflected an important
interaction between cancer cells and cells involved in chronic inflammation.
tumor-associated macrophages (TAMs) have been the subject of investigation
in a number of different tumors, including breast, prostate, ovarian, and
cervical cancers, where a clear correlation with inferior survival has
been seen with increased TAM contentref.
Curiously, in other tumors such as gastric and lung cancer, the results
have been contradictoryref.
These disparate findings may be due to specific functional characteristics
of macrophages in some tumors and may be context dependent. Nonetheless,
in the majority of studies increased TAM density has been associated with
both metastatic spread and diminished survivalref.
Lymphoma-associated
macrophage (LAM) counts are correlated with poor prognosis in follicular
lymphoma patients. Distinct role of macrophages in different tumor microenvironmentsref
proliferation arrest of T lymphocytes when exposed to tryptophan
shortage, which can be provoked by indoleamine
2,3-dioxygenase (IDO),
an enzyme that is expressed in placenta and catalyzes tryptophan degradation.
Most human tumors constitutively express IDO causing a lack of accumulation
of specific T cells at the tumor site.
tumour cells often express insufficient levels
of
NKG2D
ligands to stimulate tumour-cell rejection, either because expression of
the ligands is not sufficiently upregulted early in the development of
the tumour, or because tumour cells with lower levels of ligand expression
are selected by the immune system in vivo as the tumour evolves
MICA is overexpressed and shed by MMPs
as a soluble form from the cell surface of many
carcinoma cells into the circulation, causing systemic endocytosis and
degradation of the activating receptor NKG2D,
and hence its down-regulation in T lymphocytes (and possible other
immune system cells).
However, the existence of tumours that evade NKG2D does not detract from
the possibility that many other tumours are successfully eliminated by
NKG2D-mediated immune activation, because such evasion mechanisms are unlikely
to be universally effective. Notably, rejection of tumour cells that expressed
NKG2D ligands confers resistance to subsequent tumour cells that lack NKG2D
ligands, inducing long-lasting immunity at least in some cases, indicating
the promise of NKG2D ligands as adjuvants in tumour
vaccines.
How NKG2D ligands expressed by tumour cells enhance priming of tumour antigen-specific
CD8+ T lymphocytes remains to be established. Depletion of NK
lymphocytes before vaccination failed to prevent T-cell priming, indicating
that NK-cell-derived cytokines or the formation of tumour debris as a result
of NK-cell-mediated tumour-cell lysis are not required for this process,
although they might contribute to it. An alternative possibility is that
NKG2D ligands expressed by the tumour cells directly costimulate tumour-antigen-specific
CD8+ T lymphocytes. It is also possible that the activation
of APCs that express NKG2D is increased by the ligand-expressing tumour
cells, leading to more effective processing and/or presentation of tumour
antigens.
galectin-1 (Gal1)
is expressed by many different tumour types (e.g. mouse melanoma cells)
and increased expression correlates with tumour aggressiveness and metastasis.
It induces apoptosis in T lymphocytes. Blocking Gal1 production by melanoma
cells not only restores the ability of the host immune system to initiate
a tumor-specific primary Th1 response, it also provides resistance
to subsequent challenge with Gal1-expressing tumour cellsref.
cancer immunosurveillance is known to control certain virus-associated
tumours but its role in the control of spontaneous tumours is a matter
of some controversy. If immunosurveillance is involved, any immunogenic
tumours that do eventually grow are likely to be escape variants, selected
for low immunogenicity by T cells. By generating a transgenic mouse model
of sporadic cancer based on rare spontaneous activation of a dormant oncogene,
it has been shown that sporadic immunogenic tumours do not escape their
recognition but induce T-cell tolerance. In this model, tumours derive
slowly from single cells and express a TSA, thus reflecting physiological
tumour development as closely as possible. Whereas vaccinated mice remain
tumour-free throughout their lifetime, naive mice always develop
a progressively growing tumour. Despite specific recognition by T cells,
the tumours do not lose their intrinsic immunogenicity and are rejected
after transplantation in T-cell-competent recipients. Furthermore, in the
primary host tumour-induced tolerance is associated with the expansion
of non-functional T cells. Together, these data argue against immunosurveillance
of spontaneous cancerref1,
ref2,
ref3
expression of the heterodimeric cytokine IL-23,
but not of its close relative IL-12,
is increased in human tumours. Expression of these cytokines antagonistically
regulates local inflammatory responses in the tumour microenvironment and
infiltration of intra-epithelial lymphocytes. Whereas IL-12 promotes infiltration
of cytotoxic T cells, IL-23 promotes inflammatory responses such as upregulation
of the matrix metalloprotease MMP9,
and increases angiogenesis but reduces CD8 T-cell infiltration. Genetic
deletion or antibody-mediated elimination of IL-23 leads to increased infiltration
of cytotoxic T cells into the transformed tissue, rendering a protective
effect against chemically induced carcinogenesis. Finally, transplanted
tumours are growth-restricted in hosts depleted for IL-23-/-or
in IL-23R-/- mice. Although many strategies for immune therapy
of cancer attempt to stimulate an immune response against solid tumours,
infiltration of effector cells into the tumour tissue often appears to
be a critical hurdle. IL-23 is an important molecular link between tumour-promoting
pro-inflammatory processes and the failure of the adaptive immune surveillance
to infiltrate tumoursref.
A2A adenosine receptor (A2AR)
has been shown to be a critical and nonredundant negative regulator of
immune cells in protecting normal tissues from inflammatory damage. A2AR
also protects cancerous tissues by inhibiting incoming antitumor T lymphocytes.
Genetic deletion of A2AR in the host resulted in rejection of established
immunogenic tumors in approximately 60% of A2AR-deficient mice
with no rejection observed in control WT mice. The use of antagonists,
including caffeine, or targeting the A2 receptors by siRNA pretreatment
of T cells improved the inhibition of tumor growth, destruction of metastases,
and prevention of neovascularization by antitumor T cells. Effects of A2AR
are T cell autonomous. The inhibition of antitumor T cells via their A2AR
in the adenosine-rich tumor microenvironment may explain the paradoxical
coexistence of tumors and antitumor immune cells in some cancer patients
(the "Hellstrom paradox"). The hypoxia-->adenosine-->A2AR
pathway should be targeted as a cancer immunotherapy strategy to prevent
the inhibition of antitumor T cells in the tumor microenvironment. The
same strategy may prevent the premature termination of immune response
and improve the vaccine-induced development of antitumor and antiviral
T cells. The observations of autoimmunity during melanoma rejection in
A2AR-deficient mice suggest that A2AR in T cells
is also important in preventing autoimmunity. Thus, although using the
hypoxia-->adenosine-->A2AR pathway inhibitors may improve antitumor
immunity, the recruitment of this pathway by selective drugs is expected
to attenuate the autoimmune tissue damageref.
tumor-expressed albumin, unlike normal serum albumin purified from
blood, is capable of inhibiting T-cell activation, proliferation, and function
in both in vitro and in vivo settings. Tumor-expressed albumin
does not appear to affect antigen processing or presentation by professional
antigen-presenting cells. The activity appears to lie in relatively small,
lipid-like moieties that are presumably cargo for tumor-expressed albumin,
and that activity can be removed from the albumin by lipid removal or treatment
with lipaseref
lymphocyte apoptosis within the tumor environment : CD70,
a cytokine overexpressed in RCC, promotes lymphocyte apoptosis through
interaction with its receptor CD27 and with the intracellular receptor-binding
protein SIVA. Apoptosis increased after cocultivating lymphocytes with
the RCC cell lines A498 and CAKI2. The addition of recombinant soluble
CD70 to both native lymphocytes and a T-cell cell line resulted in increased
lymphocyte apoptosis as well. Furthermore, induced apoptosis could be partially
blocked with anti-CD27 and anti-CD70 antibodiesref.
inhibition of NK lymphocytes
: CD4+CD25+ Treg
lymphocytes
that suppress T cell-mediated immune responses may also regulate other
arms of an effective immune response. In particular,Treg directly
inhibit NKG2D-mediated NK cell cytotoxicity in vitro and in vivo,
effectively suppressing NK cell-mediated tumor rejection. In vitro,
Treg were shown to inhibit NKG2D-mediated cytolysis largely
by a TGF-b-dependent mechanism and independently
of IL-10. Adoptively transferred Treg suppressed NK cell antimetastatic
function in RAG-1-deficient mice. Depletion of Treg before NK
cell activation via NKG2D and the activating IL-12 cytokine, dramatically
enhanced NK cell-mediated suppression of tumor growth and metastases. There
is at least one mechanism by which Treg can suppress NK cell
antitumor activity and highlight the effectiveness of combining Treg
inhibition with subsequent NK cell activation to promote strong innate
antitumor immunityref.
TGF-b1
associated with other blood cells may be involved in limiting NK cell activation
: blocking endogenous TGF-b resulted in significant
NK cell IFN-g production under suboptimal stimulation
conditionsref.
NK cells can induce DC maturation, a differentiation process whereby DCs
respond to a environmental stimulus and acquire the ability of eliciting
adaptive immunity. Conversely, maturing DCs promote NK functions in
vivo and in vitro. This interplay has important consequences
on the immune response to pathogens and possibly to neoplastic cells. B16
melanoma cells actively modulate the interaction between DCs derived from
bone marrow precursors and NK/LAK cells propagated from the spleen of C57BL/6
mice. DCs increased in a dose-dependent manner the ability of NK/LAK cells
to kill melanoma cells and to produce cytokines. This activatory cross-talk
entailed the production of IL-18 by DCs and of IFN-g
by NK/LAK cells. Melanoma cells were not a passive target of NK activity;
they regulated the outcome of the interaction between DCs and NK/LAK cells,
inhibiting the in vitro production of cytokines as effectively as the genetic
deletion of IL-18 or IFN-g. Interference with
the NK/DC interaction possibly represents a mechanism used by growing tumors
to evade the immune responseref
activation of NKT
lymphocytes
inhibit effective CTL-mediated tumor rejection by IL-13
via the IL-4R-STAT6 pathway. CD4+DX5+ NKT suppressor cells regulate the
growth of UV-induced skin cancers and mediate antigen-specific immune suppression.
apoptosis of inflammatory cells within 3-5 days upon binding of
their CD95 / Fas / Apo-1
to CD178 / FasL
expressed on certain tumor cells (tumour counter-attack)
resistance to apoptosis
by cancer cells : cancer cells enhance their ability to withstand an
attack by cytotoxic immune effector cells via acquisition of specific genetic
alterations that interfere with the shared mitochondrial death signaling
pathway entrained by granzyme
B,
IFN-g,
and Apo2L/TRAIL,
3 key mediators of immunologic cell-mediated cytotoxicity. The coexistence
of specific mitochondrial signaling defects (either deletion of Bax, overexpression
of Bcl-xL, or deletion of Smac) with expression of XIAP decreases
the sensitivity of cancer cells to IFN-g/Apo2L/TRAIL-
or granzyme B-induced apoptosis, lymphocyte-mediated cytotoxicity in
vitro, and adoptive cellular immunotherapy in vivo. Conversely,
negating XIAP expression or function in tumor cells with defective mitochondrial
signaling enables direct activation of caspase-3/-7 by granzyme B or Apo2L/TRAIL,
and restores their susceptibility to immunologic cytotoxicityref
resistance to attack by complement
by cancer cells (a major limitation in the use of monoclonal
antibodies
as monotherapies) : expression of the complement-regulatory proteins CD55
/ decay accelerating factor (DAF),
CD46
/ MCP
and CD59 / MIRL
are deregulated in cancer with tumors showing loss of one or more inhibitors
and strong overexpression of others. However, tumor sensitivity to complement
can be restored by co-administration of antibodies that bind to the functional
domains of complement-regulatory proteins. Overexpression of complement-regulatory
proteins on tumors also makes them potential targets for cancer vaccines.
However, these vaccines have to be carefully designed to induce immune
responses that recognize inhibitors overexpressed on tumors and that do
not detect the levels expressed by normal cellsref.
antibody-mediated immunologic enhancement
: both B and T cell immune responses in situ are fully compatible
with progressive growth of small inocula either as ascites or as solid
subcutaneous tumors of a number of carcinogen-induced, immunogenic, murine
tumors in their syngeneic hosts. Tumor cell, after 2 weeks of growth in
vivo, become resistant to the induced killer cells. The modulating
agent bound to these resistant tumor cells appears to be anti-tumor antibody
directed against epitopes are uniquely expressed on tumor cells (oncotopes).
These observations are compatible with data first reported 35 years ago
suggesting that anti-tumor antibody promoted or enhanced tumor growthref
unknown pathways : CCL-2 / MCP-1,
osteopontin, and midkine
from oncogenic TC-1 cells producing E6 and E7 oncoproteins confer resistance
to anti-tumor immunity induced via DNA vaccination anti-E7ref
defect in the lymphocyte-vessel wall interactions : the angiogenic
nature of endothelial cells (EC) lining tumor blood vessels may account
for such anergy. In this study, we examined whether mechanisms other than
down-regulation of adhesion molecules could be involved, particularly signaling
pathways dependent on the caveolae platforms. To mimic the influence of
the tumor microenvironment, EC were exposed to TNF-a
and the proangiogenic VEGF. A dramatic inhibition of lymphocyte adhesion
on activated EC following either short or long VEGF pretreatments. VEGF
did not influence the abundance of major adhesion molecules, but was associated
with a defect in ICAM-1 and VCAM-1 clustering at the EC surface. Overexpression
of the caveolar structural protein, caveolin-1, overcame the VEGF-mediated
inhibition of adhesion and restored ICAM-1 clustering. Conversely, EC transduction
with a caveolin-1 small interfering RNA reduced the TNF-alpha-dependent
increase in adhesion. VEGF-induced NO production by the endothelial NO
synthase as the main target of the changes in caveolin-1 abundance. The
NO synthase inhibitor N-nitro-l-arginine methyl ester could reverse
the inhibitory effects of VEGF on lymphocyte adhesion and EC cytoskeleton
rearrangement. Symmetrically, a NO donor was shown to prevent the ICAM
clustering-mediated lymphocyte adhesion, thereby recapitulating the effects
of VEGF. In conclusion, this study provides new insights on the mechanisms
leading to the tumor EC anergy vs immune cells and opens new perspectives
for the use of antiangiogenic strategies as adjuvant approaches to cancer
immunotherapyref
Later in the natural history of a tumor, during the progressive growth
phase, tumors may become more immune-activating for a variety of reasons
:
tumour can damage or disrupt surrounding tissue or trigger a stress response
when they outstrip their oxygen and nutrient supplies. These processes
can cause pH imbalance that results from metabolic disturbance, generation
of ROS, up-regulation of stress protective factors - such as hsps - and
death by necrosis or apoptosis
as tumours grow progressively, dysregulated genetic and epigenetic events
lead to the expression of large numbers of neoantigens (one study estimates
that the average malignancy contained > 10,000 mutations)
All these factors may act as danger signals.
Some observations contradict the idea that the immune system spontaneously
mounts lethal attacks against tumour cells. If cycle of immune pressure
and tumor escape were operative during tumour development, one might expect
to observe (either by examination or by imaging modalities), progressive
tumour growth that was interspersed with one or more periods of contraction
: however, solid tumours generally do not have growth curves with evidence
of significant drops or depressions and simply grow larger and larger.
On the other hand, it is also possible that selection is taking place at
a cellular level, expecially during early tumor development. Ongoing selection
of tumor variants on a microscopic or cellular level would not necessarily
result in gross or macroscopic changes, such as those easily seen histologically
or with commonly used imaging modalities. Similarly, a lack of inflammation
at the site of solid tumors may be interpreted by some to cast further
doubt on the hypotheses of spontaneous immune attack and immune escape.
Although immue cells can be observed in or around tumors, spontaneous local
inflammation is generally not seen clinically or histologically : again,
however, it is possible that this inflammation is occurring chronically
and on a microscopic level. Thus, there is little direct evidence in mice
or humans to support the concept of tumour shaping or sculpting by the
unmanipulated immune system once tumours are vascularized and established
: nevertheless these hypothesized processes may well turn out to be important
upon further investigation. On the other hand the setting of partially
successful antitumour immunotherapy
is one scenario that might be expected to result in tumor escape.
The immune tolerance hypothesis suppose that endogenous tumours
can induce the tolerance of T cells that are specific for neoantigens.
A fundamental corollary is that the residual repertoire of tumour-antigen-specific
T cells will be :
of low affinity for peptide-MHC complex, associated generally with the
presence of undesirable residues at crucial anchor positions in the antigen
specific for epitopes that are presented at low density
in a mouse experimental model, the immune-privileged state of the eye permits
the outgrowth of xenogeneic uveal
melanoma
cells, but does not protect these tumors against adoptive
immunotherapy
with highly potent anti-tumor CTLref
Little is known about the fate of given tumor-specific CTL clones in cancer
patients. Studies in patients with favorable outcomes may be very informative.
In this longitudinal study, we tracked, quantified, and characterized functionally
defined antigen-specific T-cell clones ex vivo, in peripheral blood
and at tumor sites, in two long-term melanoma survivors. MAGE-A10-specific
CD8+ T-cell clones with high avidity to antigenic peptide and
tumor lytic capabilities persisted in peripheral blood over > 10 years,
with quantitative variations correlating with the clinical course. These
clones were also found in emerging metastases, and, in one patient, circulating
clonal T cells displayed a fully differentiated effector phenotype at the
time of relapse. Longevity, tumor homing, differentiation phenotype, and
quantitative adaptation to the disease phases suggest the contribution
of the tracked tumor-reactive clones in the tumor control of these long-term
metastatic survivor patients. Focusing research on patients with favorable
outcomes may help to identify parameters that are crucial for an efficient
antitumor response and to optimize cancer immunotherapyref
Cancer immunotherapy : a major
goal of cancer immunotherapy has been to generate a large number of highly
avid, tumor-specific T cells that can last in vivo for a long time and
resist tolerizationref.
Existing methods focus on reshaping the normal T cell repertoire and fall
into 2 categories:
methods relying on massive expansion of the few antigen-specific T cells,
which is usually a demanding task and not always successful.
active immunotherapy, which involves activating the effectors in the host
immune system to inhibit cancer cell growth and reject tumor (e.g., cancer
vaccination)ref1,
ref2
passive immunotherapy, which directly provides the host with effectors
to react against cancer (e.g., adoptive transfer of in vitro expanded
or modified anti-tumor T cells)ref1,
ref2,
ref3,
ref4,
ref5,
ref6.
Antigen-specific T cells expanded from an individual's T cell repertoire
cannot be readily transferred to others because of the risk of severe graft-versus-host
diseaseref
donor lymphocyte infusions (DLIs)
creating autologous T cells of a desired specificity with T
cell receptor (TCR)
gene transfer (instructive immunotherapy) :
conventional TCR transgenic animals can be made via pronuclear injection
of prerearranged TCR genesref.
However, this method offers no opportunity for therapeutic application
in humans
retrovirus-mediated expression of TCR genes in peripheral T cells
could endow them with the desired specificityref1,
ref2.
However, such T cells present certain challenges: they are activated mature
cells that have already expressed an endogenous TCR of the unknown specificity;
their effector function may be restricted by the conditions under which
they are activated in vitro; and it is unclear how long they persist
in vivoref.
retrovirus-mediated expression of TCR genes in hematopoietic
stem cells (HSCs)
: HSCs are attractive targets for TCR gene transfer because of their roles
as T cell progenitors, enormous regeneration capacity, and availability
for modification in humansref.
A method to impart the desired anti-tumor specificities (anti-chicken OVA)
to the RAG1–/– mouse T cell repertoire by delivering tumor-specific
TCR genes into HSCs via MIG retrovirusref,
followed by adaptive transfer to generate a continuous stream of anti-tumor
T cells in host mice, has been describedref.
OVA is the well characterized target antigen for both a CD8 TCR, OT1 (recognizing
OVAp257–269, designated as OVAp1)ref,
and a CD4 TCR, OT2 (recognizing OVAp329–337, designated as OVAp2)ref,
offering us the opportunity to study both the CD8 cytotoxic and CD4 helper
arms of antigen-specific T cell immunity. Coexpression of the 2 TCR subunits
from the same vector has proved critical because no antigen-specific T
cells were detected when a 2-vector system reported previously to infect
WT HSCs was usedref
: the coexpression was achieved by linking cDNAs encoding TCR a
and b chains with an internal ribosome entry
site and inserting this into the retroviral vector under control of the
viral LTR promote. The vectors expressing OT1 or OT2 TCR were designated
MOT1 or MOT2. In addition to using HSCs as the target for gene transfer,
this approach has other features that facilitate clinical application:
single-vector gene delivery, small genetic elements (TCR a
and b cDNAs), and a convenient viral promoter
to drive the expression of transgenes. The recipients were allowed to reconstitute
their T cell compartments for 7 weeks before analysis. Some 3–7% of the
total BM cells were transduced, including long-term HSCs that expressed
c-kit and Sca-1ref.
The percentage of transduced HSCs remained constant to 8-month posttransfer
and persisted through a secondary BM transfer, indicating that they maintained
their stem cell features of longevity and self-renewal. Therefore, this
genetic method is sufficiently robust to ensure the maintenance of a transduced
HSC population for the lifetime of the host. 25% of the total peripheral
CD8 T cells in C57BL/6J(B6) female mice/MOT1 expressed OT1 TCR and 8% of
the total peripheral CD4 T cells in B6/MOT2 mice expressed OT2 TCR, with
no leakage into the other T cell compartment. In numerous experiments,
an average of 20% (ranging from 15% to 30%) of the total peripheral CD8
T cells in B6/MOT1 mice carrying the OT1 TCR specificity was achieved,
and an average of 8% (ranging from 5% to 10%) of the total peripheral CD4
T cells carrying the OT2 TCR specificity. This high percentage of antigen-specific
T cells remained constant for 8 months posttransfer. Therefore, retrovirus-mediated
expression of TCR cDNAs in WT HSCs can stably generate a significant population
of T cells with the desired features and specificity. In natural conditions,
this percentage can be achieved only by clonal expansion of a few high-affinity
T cell clones after strong immune stimulationref.
The RAG1–/– background offered a clean system for our intended
study, because of the lack of endogenous TCR expressionref.
Surprisingly, TCR a and b
chain
expression patterns in the thymocytes of RAG1/MOT2 mice closely resembled
the b chain expression in RAG1/OT2 Tg mice that
is under control of the endogenous TCR promoter and enhancersref.
The mature OT2 T cells recovered from the periphery of the 2 kinds of mice
were comparable, although there was more heterogeneity of TCR expression
in the OT2 T cells from RAG1/MOT2 mice. A similar magnitude of antigenic
response was observed when the animals were immunized with OVAp2 antigen.
T cells harvested from mice displayed the canonical naïve phenotype
of CD8 T cells: CD25–CD69–CD62LhighCD44low.
Upon stimulation with OVA in vitro, these T cells responded vigorously
as measured by proliferation and IFN-g production,
with a magnitude comparable to the response of transgenic T cells obtained
from conventional TCR transgenic miceref
(but we have no comparison to functionality of normally developed OVA-specific
T cells). The activated T cells exhibited the typical effector cytotoxic
T cells phenotype: CD25+CD69+CD62LlowCD44high.
The ability of these OT1 T cells to generate and maintain CD8 T cell memory
by adoptive transfer of in vitro-activated OT1 cytotoxic T cell
to host mice was then tested because continuous generation of naïve
OT1 T cells makes it difficult to directly analyze memory formation in
B6/MOT1 mice. Sixteen weeks after transfer, long-lived OT1 T cells were
detected in the hosts, which displayed the memory phenotype (CD25–CD69–CD62LhighCD44high).
These cells showed a stronger and faster response to antigen stimulation
when compared with naïve OT1 T cells. Furthermore, they proliferated
upon cytokine stimulation, which is a unique feature of memory T cellsref.
The therapeutic potential of this method was tested in the E.G7 mouse tumor
modelref.
OVA serves as the tumor antigen in this model, allowing us to study either
anti-tumor cytotoxic T cell immunity in B6/MOT1 mice, anti-tumor helper
T cell immunity in B6/MOT2 mice, or the combination of both arms in B6/MOT1+MOT2
mice (B6 mice receiving both MOT1- and MOT2-transduced WT HSCs)ref.
Suppression of syngenic tumor growth was studied by using the following
protocol: B6/MOT1, B6/MOT2, and B6/MOT1+MOT2 mice were allowed to reconstitute
their T cell compartment over 8–10 weeks after receiving retroviral transduced
HSCs. E.G7 tumor cells were then injected s.c., and EL.4 tumor cells were
used as a controlref.
To evaluate the effects of peptide immunization, half of the experimental
groups were immunized by using DCs loaded with OVAp1 (DC/OVAp1) 4 days
after tumor injection. Four mice were used in each treatment group, and
the experiments were performed three times. Compared with the B6 control,
significant tumor suppression was observed in B6/MOT1 mice, with complete
suppression in 50% of the mice. No recurrence of tumors was observed as
long as the experiments ran (up to 200 days). For the other 50%, tumor
growth was suppressed up to 18 days but eventually progressed. E.G7 tumor
cells recovered from these mice were still recognized by naïve OT1
T cells , excluding the possibility of "epitope escape" by OVA antigen
mutation. The OT1 cells recovered from the tumor-bearing mice could no
longer respond to OVAp1 antigen stimulation in vitro, as measured
by IFN-g production. These anergic OT1 T cells
displayed a surface phenotype that resembled naïve T cells but with
higher CD44 expression, indicating that they had encountered tumor antigen.
Interestingly, total T cells still responded to anti-CD3 stimulation, suggesting
the existence of an active antigen-specific tumor tolerogenic mechanismref.
Similar results were obtained for B6/MOT2 tumor-bearing mice, suggesting
that tolerogenic mechanisms act on both anti-tumor cytotoxic and helper
T cells. DC-based peptide immunization enhanced OT1 T cell functionref
and thoroughly suppressed tumor growth in all of the B6/MOT1 mice. Interestingly,
significant tumor suppression was also observed in B6/MOT2 mice despite
the fact that unlike OT1 T cells OT2 T cells cannot directly recognize
E.G7 tumor cells because of their lack of MHC II expression. The possible
mechanism in this case would be cross-presentation of tumor antigens by
the host-derived antigen-presenting cellsref.
Even more strikingly, when B6/MOT2 mice were immunized with DC/OVAp1 complete
suppression of tumor growth was observed in all of the mice (some tumors
grew up to a barely detectable size but soon regressed). This series of
experiments indicated that the CD4 anti-tumor T cells could provide essential
help for the endogenous cytotoxic T cell to mount an anti-tumor response,
regardless of whether they can recognize the tumor directly or not. As
expected, a combinatorial tumor suppression effect was observed in B6/MOT1+MOT2
mice, as exhibited by a slower tumor growth in mice even when the tumor
broke through. DC/OVAp1 immunization totally suppressed tumor growth in
all of the mice. Importantly, peptide immunization did not work for B6
control mice, and in all experimental groups challenged with control tumor
EL.4, the tumors grew up at a similar rate regardless of immunization,
confirming that suppression of E.G7 tumor growth was tumor antigen-specific
and mediated by the engineered anti-tumor CD8 and/or CD4 T cells. The idea
of combining the 2 arms of anti-tumor T cell immunity was further extended
with the aim of eradicating an established large vascularized solid tumor.
Physiologically, tumors achieve growth by initially overcoming the host's
anti-tumor immuno-surveillance and then constantly tolerizing the anti-tumor
T cells in the hostref.
A protocol was devised to mimic these processes: mice in which E.G7 tumors
grew despite the presence of anti-tumor CD8 (B6/MOT1) or both anti-tumor
CD8 and CD4 T cells (B6/MOT1+MOT2) were immunized with one dose of DCs
loaded with both OVAp1 and OVAp2 (denoted as DC/OVAp1+2), at the time when
their tumors reached 30 mm2 [18 days after tumor challenge,
tumors extant for this duration and with this size are considered to be
long-term established tumorsref].
The tumors in B6/MOT1 mice receiving immunization were suppressed for 10
days but then grew, reaching 400 mm2 in 50 days. In contrast, tumors in
B6/MOT1+MOT2 mice shrank after the immunization and were not longer detectable
within 12 days; no tumor recurrence was observed for as long as the experiments
ran (>200 days). These results demonstrated that established solid vascularized
tumors could be eradicated by providing both the CD8 and CD4 arms of anti-tumor
T cell immunity combined with appropriate peptide immunization to boost
the responses of both arms. This method can be easily extended to generate
T cells targeting multiple epitopes of the antigen, thus countering "epitope
escape" by antigen mutationsref.
It might also be extended to engineering the BcR on B cells, allowing the
cells to secrete antibodies of predefined specificity. Therefore, instructive
immunotherapy has the potential to establish in vivo a lifelong
"complete" adaptive immunity (including CD8 cytotoxic T cell, CD4 helper
T cell, and B cell arms) to target any pathogens where antigens and the
cognate TCR/B cell receptors are knownref1,
ref2.
In addition, this method could be used for rapid and efficient analysis
of such problems as the development and life history of antigen-specific
T cells and in vivo anti-tumor T cell response and tolerance, providing
a useful tool for basic T cell studies.
Research groups :
Departments of Internal Medicine and Obstetrics and Gynecology, University
of Arkansas, Little Rock, AR : Hermonat PL, Liu Y,
Department of Microbiology and Immunology, Texas Tech University, Lubbock,
TX; USA : Wan W, Tedesco A, Chiriva-Internati M
Scientific Direction, Istituto Clinico Humanitas, Milan; : Grizzi F
Foundation “M. Rodriguez”, Milan; Italy : Grizzi F
Immunovirology and Biotherapy Unit and Experimental Oncology 1, CRO, IRCCS
Aviano : Guidoboni M, Caggiari L, De Re V, Gasparotto G, Marzotto A, Martorelli
D, Baiocchi M, Dolcetti R
Human Tumor Immunobiology Unit, INT,Milano; Italy : Anichini A
Dipartimento di Scienze Oncologiche e Chirurgiche, Università di
Padova : Bobisse S, Tisato V, Mandruzzato S, Zoso A, Indraccolo S, Zanovello
P, Rosato A
H San Raffaele Scientific Institute, Milano; Italy : Casorati G
Flow Cytometry and Cell Therapy Unit, Medical Oncology, University and
IRCCS “S. Matteo”, Pavia, Italy : Ferrari S, Rovati B, Collovà E,
Grasso D, Sagrada P, Danova M, Riccardi A