Está en la página 1de 20

Cancer immunity A9

Jan W. Gratama, Reno Debets and Ralph A. Willemsen

clinical application of immunotherapy to treat can-


Introduction: cancer immunity from a cer has been rekindled in the last two decades by
historical perspective the revival of the immunosurveillance theory, the dis-
covery and structural characterisation of TUMOR-
Almost 100 years ago, Ehrlich and coworkers ASSOCIATED ANTIGENS, our progress in understanding
observed the presence of infiltrates of mononuclear the molecular pathways required for the induction
cells around or inside tumor lesions [1].This finding and maintenance of immune responses, and
led them to propose that tumors could be recog- methodological advances to generate specific
nized and inhibited by the ‘magic bullets’ of the immunological probes in the form of tumor-specific
immune system. At the end of the 19th century, stud- cytotoxic T LYMPHOCYTES (CTL) and monoclonal
ies were initiated that aimed to actively immunize ANTIBODIES (mAb). Results obtained until now have
cancer patients against their own cancerous tissue. revealed [4]:
During the subsequent decades, cancer patients • the increased susceptibility of immunodeficient
were nonspecifically immune-stimulated with rela- patients to cancer as compared to immunocompe-
tively crude leukocyte extracts such as transfer fac- tent individuals, supporting the concept of
tor, immune-RNA, bacterial extracts such as bacillus immunosurveillance against cancer [5–7];
Calmette-Guérain, Coley’s toxin or levamisole. These • the molecular characterization of a wide range of
studies were initated in spite of the fact that little was various types of TUMOR-ASSOCIATED ANTIGENS on can-
known about the various components of the cer cells, as detailed below [8];
immune system that could react against cancer and • the feasibility of using TUMOR-ASSOCIATED ANTIGENS
even less was known about the structures on cancer as vaccines and DENDRITIC CELLS to induce tumor
cells that can be recognized by the immune system. rejection in tumor-bearing animals and patients
The discrimination between ‘self’ and ‘nonself’ by [9–14];
the immune system – the 1960 Nobel Prize-winning • the efficacy of INTERLEUKIN (IL)-2 and adoptively
concept of Burnet and Medawar – has been pivotal transferred T LYMPHOCYTES, expanded from tumor-
for modern tumor immunology. Subsequently, infiltrating LYMPHOCYTES (TIL), in some patients
Thomas and Burnet developed the ‘immune surveil- with metastatic cancer, in particular melanoma
lance’ theory. The core of this theory is that the and renal cell carcinoma [15–18];
immune system protects the host from cancer by • the immunosuppressive capacities of tumor cells,
detecting and destroying newly-formed cancer cells, i.e., to secrete immunosuppressive CYTOKINES such
recognized as nonself [2, 3]. This immune surveil- as transforming growth factor (TGF)-β [19] and IL-
lance theory, including its critics and the various 10 [20] and to inhibit LEUKOCYTES with antitumor
experiments supporting and disproving it,has strong- capacities.
ly influenced tumor immunological research during
the past 40 years. Here, we discuss the key elements involved in the
Ever since,the existence of immunity against can- generation of antitumor responses: the cellular and
cer has been abundantly demonstrated in animal humoral components of the immune system,and the
(mostly murine) models and in man. Interest in the target ANTIGENS on the tumor cells.With this basis, we
130 Cancer immunity

review the various immunotherapeutic approaches CYTES respond to antigen by secreting their own anti-
to cancer and put the envisaged future develop- gen receptors as Ab after having differentiated into
ments into the perspective of current experiences. plasma cells. Ab interact with effector mechanisms
via binding of their constant part (Fc) to comple-
ment, Fc-receptor-bearing phagocytes and Fc-recep-
Key players in the immune responses tor-bearing cytotoxic (NK and T) LYMPHOCYTES.
The major T LYMPHOCYTE SUBSETS are characterized
against cancer by expression of the differentiation markers CD4 or
CD8. Whilst Ab mostly react with intact proteins or
Both innate and adaptive components of the carbohydrates,T cells do so with peptides expressed
immune system interact to generate antigen-specific on the cellular surface via presentation by scaffolds,
immune responses.As detailed in the previous chap- i. e., molecules of the MAJOR HISTOCOMPATIBILITY COM-
ters, the INNATE IMMUNE SYSTEM constitutes the body’s PLEX (MHC), on ANTIGEN-PRESENTING CELLS (APC). One
first line of defense against ‘foreign invaders’. INNATE of the most potent types of APC is the dendritic cell
IMMUNITY involves a large number of different cell (DC).DC pick up ANTIGENS in the skin or other periph-
populations such as epithelial cells, MONOCYTES, eral tissues, and migrate to the T-cell zones of LYM-
MACROPHAGES, DENDRITIC CELLS, POLYMORPHONUCLEAR PHOID ORGANS where they stimulate naïve CD4+ and
LEUKOCYTES or GRANULOCYTES, and some lymphocyte CD8+ T cells.
subsets that are at the interface between INNATE and Most CD4+ T cells are ‘T-helper (Th) cells’ and rec-
ADAPTIVE IMMUNITY (natural killer [NK] LYMPHOCYTES, ognize antigen in the form of 15 to 25-mer peptides
clusters of differentiation [CD]5+ B LYMPHOCYTES and presented by class II MHC molecules. These mole-
T cell-receptor [TCR]-γδ+ T LYMPHOCYTES). The INNATE cules present peptides that are mainly derived from
IMMUNE SYSTEM also comprises a variety of humoral the extracellular compartment, as opposed to pep-
factors such as CYTOKINES,chemokines,enzymes such tides presented by class I MHC molecules that cap-
as lysozyme, metal-binding proteins, integral mem- ture endogenously processed peptides (see below;
brane ion transporters, complex carbohydrates and [21]). CD4+ T cells are the major regulators of most
complement. The phagocytic cells (MACROPHAGES, immune responses. They augment immune respons-
GRANULOCYTES) and the COMPLEMENT SYSTEM constitute es by secreting CYTOKINES that stimulate cytotoxic
effector mechanisms by which the ‘invaders’ can be CD8+ T cells (so-called Th1-type CYTOKINES such as
destroyed. The production of CYTOKINES and INTERFERON [IFN]-γ), or B cells to mount Ab responses
CHEMOKINES acts in concert with antigen presentation (so-called Th2-type CYTOKINES such as IL-4 and IL-5).
by DENDRITIC CELLS and MONOCYTES to initiate adaptive CD8+ cytotoxic T LYMPHOCYTES (CTL) react with 8
immune responses. to 10-mer peptides presented on the cellular surface
ADAPTIVE IMMUNITY makes use of a unique mecha- by the scaffolds formed by their class I MHC mole-
nism whereby genetic mutations occurring in two cules. These peptides contain 2 to 3 so-called
specialised cell populations, B and T LYMPHOCYTES, ‘anchoring residues’ that fit into specific pockets of
produce numerous molecular ‘shapes’ that are the class I MHC molecule [22]. CD8+ T cells destroy
expressed as ANTIBODIES (Ab) and TCR. Figure 1 pro- their target cells, after attachment of (1) their TCR to
vides a simplified overview of how the effector com- the appropriate MHC-peptide complex, and (2) their
ponents of the adaptive immune system (T cells and accessory molecules (such as CD8 and CD28) to the
Ab) are regulated and eliminate their targets. corresponding ligands, by perforating their mem-
Antigen-specific immunity is generated when Ab branes with enzymes (i.e., perforin and granzymes)
and TCR are expressed and upregulated through the or by triggering a process of self-destruction (termed
formation and release of CYTOKINES and chemokines. apoptosis). In this way, CD8+ T cells can move from
Thus, ADAPTIVE IMMUNITY involves a wide range of anti- one tumor cell to another expressing the same MHC-
gen receptors expressed on the surface of T and B peptide complexes, and thus can mount a very spe-
LYMPHOCYTES to detect ‘foreign’ molecules. B LYMPHO- cific and robust antitumor response.
Key players in the immune responses against cancer 131

FIGURE 1
Key players of the immune system in mounting an antitumor response. A ‘professional’ antigen-presenting cell (APC)
presents antigenic peptides (depicted as diamonds) to a helper T lymphocyte (HTL) via its Class II MHC molecules (CL-
II), and to a cytotoxic T lymphocyte (CTL) via its Class I MHC molecules (CL-I). The T-helper cells may also recognize
antigen on a tumor cell itself if the tumor cells express Class II MHC molecules (right side of figure); similarly, a CTL
may also recognize antigen on a tumor cell itself if the tumor cells express Class I MHC molecules (upper left). The T-
helper cells recognize antigen through their T-cell receptors for antigen (TCR) which are supported in this task by CD4
coreceptor molecules. By the same token, CTL recognize antigen by their TCR supported by CD8 coreceptor molecules.
Other important accessory molecules and their receptors, such as CD28 on T cells and CD80/CD86 on APC, as well
as downregulatory T cells are not shown here (see text for details). The T-helper cells support CTL activation and pro-
liferation by secreting so-called T-helper 1 cytokines (Th1; e.g., IL-2 and IFN-γ). On the other hand, T-helper cells stim-
ulate B lymphocytes through Th2 cytokines (e.g., IL-4 and IL-5). B cells recognize soluble antigen through their B-cell
receptor for antigen; upon activation, B cells differentiate into plasma cells which secrete antibodies specific for that
antigen. Upon engagement of their TCR and accessory molecules, CD8 and also CD4+ T cells can destroy tumor cells
by secreting granzymes, perforins and cytokines such as TNF-α, or by upregulation of CD95 (Fas ligand) on tumor cells.
The remnants of destroyed tumor cells can be taken up by APC, processed and presented to T cells; in addition, they
can be specifically recognized by Ag and eliminated via Fc-receptor-expressing phagocytes.

In the setting of ADOPTIVE IMMUNOTHERAPY of can- [25].Thus, CD4+ T-cell help results in de novo genera-
cer (see below), the administration of autologous tion of tumor-specific CD8+ CTL and concomitant
CD4+ T helper cells concurrently with CD8+ CTL has tumor destruction [26]. The exact requirement for
been shown to prevent exhaustion of the infused CD4+ T-cell help during priming may depend on the
CD8+ CTL [23, 24]. CD4+ T cells activate DENDRITIC nature of the stimulus, but the requirement for CD4+
CELLS through crosslinking of CD40, which thus pro- T cells during memory responses is beyond dispute
vide enhanced antigen presentation and co-stimula- [27].We propose that CD4+ T cells have a critical role
tion, which leads to priming of CD8+ CTL function in anti-tumor immunity that goes beyond the mere
132 Cancer immunity

induction and maintenance of tumor-specific CD8+ pools derived from tumor cells have been transient-
CTL. Specifically, CD4+ T cells activate CD8+ CTL ly transfected into cell lines expressing the relevant
already present at the site of the tumor and enable class I MHC molecules. These transfected cell lines
other CD8+ CTL to migrate into the tumor, induce an were then assayed for their ability to specifically
anti-tumor Ab response, and otherwise contribute to stimulate cytokine production by class I MHC-
tumor regression through production of IFN-γ which matched, tumor-reactive T-cell CLONEs. The subse-
inhibits tumor-induced angiogenesis [23, 28]. quent transfection of individual cDNAs or cosmids
CYTOKINES produced by CD4+ T-helper cells also from pools that specifically stimulated the T cell
regulate MACROPHAGES by stopping their migration CLONEs allowed the identification of individual TAA.
after their engagement,allowing them to accumulate The first human TAA that was identified in this way
at a particular site.Thus, more efficient PHAGOCYTOSIS by screening a large genomic library from a human
is stimulated,so that increasing numbers of ‘invaders’ leukocyte antigen (HLA)-A*0101+ melanoma patient
are efficiently destroyed. CD4+ T cells amplify them- was termed melanoma antigen (MAGE)-1 [33]. To
selves by secreting CYTOKINES and interacting with identify the MAGE-1 peptide presented by HLA-
them; IL-2 is a key example for this ‘autocrine loop.’ A*0101, selected 9-mer peptides were synthesized
This action enhances the T-helper cell response and and screened for their capacity to specifically stimu-
thus the entire immune system’s response to foreign late the MAGE-1-specific cytotoxic T-cell CLONE isolat-
antigens. ed from this patient [34]. Ever since, the use of this
A minor subset of CD4+ T cells, characterized by ‘reverse immunology’ approach has allowed the
coexpression of CD25 and in particular the glucocor- identification of a large number of TAA (see below
ticoid-induced TUMOR NECROSIS FACTOR receptor, are and Table 1). The definition of class I MHC peptide-
essential negative regulators of immune function binding motifs, i.e., the nature and position of pock-
[29]. These T-regulatory cells can suppress both ets along the peptide-binding groove of the MHC
innate and adaptive immune responses. They are molecule that bind the anchoring residues of the
capable of robust Ag-driven proliferation as observed peptides, has allowed the computer-assisted screen-
for all other adaptive immune responses.Importantly, ing of large numbers of potentially immunogenic
the presence of tumor-specific CD4+ T-regulatory peptides, including many TAA [22].
cells at tumor sites may inhibit T-cell responses The identification of class II MHC-binding pep-
against cancer (see below; [30]). tides has thus far met with more problems than that
of class I MHC-binding peptides due to (1) the vari-
able length of the peptides that can be bound by
Expression of targets for the immune class II molecules, and (2) the availability of less
information on peptide-anchoring residues for class
system by cancer cells II MHC molecules. To date, there is no strategy avail-
able for the identification of class II MHC-restricted
Tumor-associated antigens (TAA) recognized antigen that matches the successes obtained in the
by T lymphocytes identification of class I MHC-restricted antigens.

Clinical immunotherapeutic studies of cancer per-


formed using in vitro expanded T LYMPHOCYTES isolat- Antigens recognized by antibodies (Ab)
ed from tumors (see below) have demonstrated that
CD8+ T cells can specifically lyse, in an MHC-restrict- In general, ANTIGENS recognized by Ab are mostly
ed fashion, autologous tumor cells or tumors arising intact proteins or carbohydrates,and are not present-
from different tissues, but not normal cells [31, 32]. ed by MHC molecules.Therefore, such ANTIGENS can-
Class I MHC molecules are expressed on the surface not be recognized by T LYMPHOCYTES via their TCR
of most nucleated cells. To identify these MHC- which only recognize MHC-presented peptides.
restricted tumor antigens, cDNA or DNA cosmid Many of these TAA are merely overexpressed by
Expression of targets for the immune system by cancer cells 133

tumor cells in comparison to their normal counter- nized by T cells (MART)-1 [40],glycoprotein (gp) 100
parts. Although these TAA are not ‘tumor-specific’ in [41] and tyrosinase [42] in melanoma, as well as
the strict immunological sense, they can effectively prostate-specific antigen (PSA; [43]) and prostate-
be used to selectively target the immune system to specific membrane antigen (PSMA; [44]) in prostate
tumors resulting in clinical antitumor effects [35]. cancer. These ANTIGENS can elicit ‘spontaneous’ CD8+
Using monoclonal Ab, tissue-specific ANTIGENS have CTL responses in patients with melanoma and
been identified that,although not tumor-specific,can healthy individuals.In melanoma,increased frequen-
serve as target structures for immunotherapeutic cies of these CTL are seen, but these are functionally
interventions (see below). inactive (i.e., anergic) [51]. A point of concern with
A highly significant development in the study of the use of these ANTIGENS as targets for cancer
the humoral response to cancer is the serological immunotherapy is that normal cells expressing these
identification of ANTIGENS by recombinant expression ANTIGENS also will be destroyed,as illustrated in a clin-
cloning (SEREX). This technique uses diluted sera ical trial of melanoma patients treated with ex vivo
from patients with cancer to screen tumor cDNA expanded TIL specific for MART-1 and gp100 [15].
libraries expressed in Escherichia coli in order to The second category comprises the ‘cancer-
identify ANTIGENS that have elicited high-titer IgG Abs testis antigens,’ so-called for their expression by his-
[36].The first application of SEREX revealed the New tologically different tumors and, among normal tis-
York-esophagus 1 (NY-ESO-1) antigen using serum sues, by spermatogonia and spermatocytes of the
from a patient with squamous cell carcinoma of that testis and occasionally by placental trophoblasts.
organ [37]. Ever since, a multitude of ANTIGENS from The expression of cancer/testis ANTIGENS by tumors
tumors of many histologies, including melanoma results from reactivation of genes that are normally
and colon,lung and renal cancers have been discov- silent in adult tissues but are transcriptionally acti-
ered using SEREX.The function of many of the newly vated in some tumors. Examples of cancer-testis
identified proteins is still unknown. Importantly, ANTIGENS are the MAGE [48], B antigen (BAGE) [49]
screening for high-titer IgG TAA-specific Ab should and G antigen (GAGE) [50] families of antigens, as
identify patients with CD4+ T-cell responses to that well as NY-ESO-1 [37]. These ANTIGENS are less
TAA as such a response is required for Ig class immunogenic than differentiation antigens, and do
switching. Indeed, Class II MHC-restricted TAA-specif- not elicit ‘spontaneous’ CTL responses. In this con-
ic CD4+ T cells, as well as class I MHC-restricted CD8+ text it is interesting that members of the MAGE-A
T-cell responses to the same tumor-derived proteins group are expressed by medullary thymic epithelial
as identified by the high-titer IgG TAA-specific Ab, cells, where central tolerance for these ANTIGENS
have been detected in some of these cases [38, 39]. may be induced [52].
The third group is constituted by ANTIGENS that are
overexpressed by tumors arising from a variety of tis-
Classification of tumor-associated antigens sues, and that are also expressed, albeit at much
(TAA) lower levels, by normal tissues. This high level of
expression by tumor cells as revealed by microarray
The identified TAA can be grouped into several gen- or differential display analyses has been instrumen-
eral categories [8]. Table 1 provides an overview of tal for the identification of many of these ANTIGENS.
these categories, examples of TAA within each cate- Examples are carcinoembryonic antigen (CEA) on
gory, expression patterns of the listed TAA and infor- gastrointestinal,breast and lung cancers [53],human
mation as to whether cellular and humoral immune epidermal receptor-2/neurological (Her-2/neu) on
responses against these TAA have been reported. melanoma,ovarian and breast cancers [54],and car-
The first group is formed by tissue-specific differ- boxy-anhydrase 9 on renal cell carcinoma [55].
entiation antigens. These ANTIGENS are expressed by Several mAb have been raised against TAA from this
normal and neoplastic cells of the same lineage. group which are currently used for cancer immuno-
Examples are melanoma antigen (Melan)-A recog- therapy (see below).
134 Cancer immunity

TABLE 1. EXAMPLES OF TUMOR-ASSOCIATED ANTIGENS1

Tumor-associated Observed type of Tissue distribution by malignant cells Refs.3


antigen immune response and their normal counterparts2
T-cell- Antibody-
mediated mediated

Tissue-specific differentiation antigens


Melan-A/MART-1 + – Malignant: melanoma [40]
Normal: melanocytes
gp100 + – Malignant: melanoma [41]
Normal: melanocytes
Tyrosinase + + Malignant: melanoma [42, 45]
Normal: melanocytes
PSA + + Malignant: prostate adenocarcinoma [43, 46]
Normal: epithelial cells of the prostate gland
PSMA + + Malignant: prostate adenocarcinoma [44, 47]
Normal: epithelial cells of the prostate gland

Cancer-testis antigens4
MAGE family + + Malignant: melanoma, breast, colon, H&N, [36, 48]
(N)SCLC, sarcoma, thyroid5
BAGE + + Malignant: melanoma, bladder, breast, H&N, [45, 49]
NSCLC, sarcoma
GAGE family + – Malignant: melanoma, bladder, breast, [50]
colon, esophagus, leukemias, lymphomas,
mesothelioma, (N)SCLC, sarcoma, seminoma
NY-ESO-1 + + Malignant: melanoma, bladder, breast, [37–39]
H&N, liver, (N)SCLC, lymphoma, ovary, prostate,
sarcoma, thyroid

Overexpressed antigens
CEA + +6 Malignant: colon, breast, gastric,(N)SCLC, [53, 56]
pancreas, rectum
Normal: embryonic gastro-intestinal tissues
Her-2/neu + + Malignant: breast, gastric, melanoma, ovary, [57, 58]
pancreas
Normal: epithelial cells
Carboxyanhydrase 9 + + Malignant: renal cell carcinoma [55, 59]
Normal: bile duct epithelium, gastric mucosal cells

Malignancy-associated fusion proteins4


BCR-ABL + + Malignant: chronic myelogenous leukemia [63–65]
PML-RARα –7 – Malignant: acute promyelocytic leukemia [66]
Expression of targets for the immune system by cancer cells 135

TABLE 1 (continued)

Tumor-associated Observed type of Tissue distribution by malignant cells Refs.3


antigen immune response and their normal counterparts2
T-cell- Antibody-
mediated mediated

Malignancy-associated point mutations of normal genes4


p53 _ + Malignant: colon, breast, ovary [58, 67, 68]
β-catenin + – Malignant: melanoma [69]

Viral proteins expressed by malignant cells4


EBV LMP-1 + + Malignant: EBV+ Hodgkin’s lymphoma, [73, 74]
nasopharyngeal carcinoma
HPV E6 & E7 + + Malignant: HPV (serotypes 16 and 18)+ [75, 76]
cervical carcinoma

Malignancy-associated minor histocompatibility genes


HA-1 + – Malignant: hematopoietic malignancies, some [80]
solid tumors
Normal: hematopoietic cells
BCL2A1 + – Malignant: hematopoietic malignancies, some [81]
solid tumors
Normal: hematopoietic cells

1 Ref. [8] provides a comprehensive listing of TAA as published until 1 August 2000. Information in this table is an
excerpt of this listing, extended with information from some more recent publications.
2 Abbreviations used in this column: (N)SCLC, (non) small-cell lung cancer; GI, gastrointestinal; H&N, squamous cell
carcinoma occurring in the head and neck region.
3 References providing evidence for cellular or humoral immune reactivity against the relevant TAA are provided.
4 This group of TAA is not expressed by normal tissues.
5 Expression patterns of MAGE-A1 are shown. The MAGE family includes the MAGE-A, B and C groups; within each
group, multiple proteins have been identified [48].
6 Humoral responses against CEA have only been observed by vaccination [56].
7 Cellular immune responses against PML-RARα have only been observed in vitro [67].

A fourth group of truly tumor-specific ANTIGENS The candidate ANTIGENS are derived from the region
arises from the fusion of distant genes resulting from immediately surrounding and containing the fusion
translocation of chromosomes in tumor cells. site.Although these ANTIGENS may represent the most
Therefore,the resulting fusion proteins are highly dis- specific targets for immunotherapy, their use for this
ease-specific (e.g.,breakpoint cluster region-Abelson purpose is limited, as only a few epitopes have been
[BCR-ABL] in chronic myelogenous leukemia [60] observed to bind efficiently to a small subset of class
and promyelocytic leukemia-retinoic acid receptor α I molecules and to be naturally processed from the
[PML-RARα] in acute promyelocytic leukemia [61]). protein [62].
136 Cancer immunity

Another group of unique tumor ANTIGENS arises patient’s hematopoietic system is replaced by that of
from point mutations of normal genes whose molec- an HLA-matched, but mHAg-mismatched STEM CELL
ular changes often accompany neoplastic transfor- donor with the capability to recognize the mHAg
mation or tumor progression. These mutational anti- expressed by the tumor as foreign. In this setting,
gens are only expressed by tumor cells and not by mHAg, being allo-antigens, are not subject to self-tol-
their normal counterparts.In addition to their limited erance and likely to be more immunogenic than the
immunogenicity for T-cell responses (similar to above-described MHC-restricted tumor-associated
fusion proteins), their therapeutic utility is limited self antigens.
further because induced immune responses are
restricted to those individual tumors expressing
these antigens. Examples are mutated forms of the Immunotherapy of cancer
p53 tumor suppressor protein in a variety of tumors
[58, 67, 68], as well as mutated β-catenin in Cytokines
melanoma [69].
Virus-encoded proteins have been identified as CYTOKINES are endogenous molecules that affect the
sources of TAA in tumors that emerge from virally immune response and include, e.g., IL-1, IFN-α and
transformed cells.These proteins typically contribute TUMOR NECROSIS FACTOR (TNF)-α (with proinflamma-
to the malignant transformation. Examples of virally tory properties), IL-2, IL-12 and IFN-γ (with T-cell
encoded TAA are the latent membrane protein potentiating properties), IL-8 and macrophage
(LMP)-1 of Epstein-Barr virus (EBV) that is expressed inflammatory protein-1 (with chemotactic proper-
by nasopharyngeal carcinoma and EBV+ Hodgkin’s ties) and granulocyte-macrophage colony-stimulat-
lymphoma [70], and the human papilloma virus ing factor (GM-CSF, with classical growth-stimulating
(HPV)-encoded E6 and E7 proteins expressed by properties). Recombinant CYTOKINES that have been
HPV+ cervical carcinoma [71, 72]. As these ANTIGENS most widely used against cancer are IFN-α, IL-2 and
are not expressed by normal cells,their usefulness as TNF-α.IFN-α has multiple effects on (tumor) cell pro-
targets for immunotherapy of cancer appears to be liferation, angiogenesis, immune function and
straightforward. expression of genes coding for Class I MHC mole-
Last but not least (see below), so-called ‘minor cules, tumor ANTIGENS and adhesion molecules. Its
histocompatibility antigens’ (mHAg) can act as TAA antitumor effects have been well documented in
and be instrumental in the rejection of malignant hairy cell leukemia [82], Karposi’s sarcoma [83] and
cells after allogeneic hematopoietic STEM CELL trans- metastatic renal cell carcinoma [84,85] whilst its effi-
plantation (SCT) [77]. mHAg are Class I MHC-pre- cacy as adjuvant systemic therapy for stage II-III
sented peptides from polymorphic, naturally melanoma is still questionable [86].
processed intracellular proteins that can be encod- IL-2 has never been shown to have direct antitu-
ed by mitochondrial DNA, the Y chromosome or mor activity, but has been used for immuno-stimula-
autosomal chromosomes [78]. mHAg-specific CD8+ tion in metastatic renal cell carcinoma and malig-
T cells have been isolated from SCT recipients, par- nant melanoma. Some of these patients obtained
ticularly those having developed graft-versus-host longstanding partial (10–15%) or complete (~5%)
disease [79]. Twelve mHAg have now been traced regression of their tumors; the latter were frequently
back to the proteins from which they originate; inter- of long duration (i.e., >7 years), suggesting immuno-
estingly,half of these proteins are expressed by malig- logical eradication of cancer [87].The diverse multi-
nant cells and/or appear to have a role in malignant system toxicity observed with high-dose i.v. IL-2 ther-
cell transformation [77]. Therefore, these mHAg can apy has prompted major efforts to develop effective
be classified as TAA. As mHAg are expressed by nor- regimens using lower doses of IL-2. Whilst low-dose
mal hematopoietic cells, their use as targets for IL-2 regimens in metastatic melanoma do not appear
immunotherapy of cancer is limited to the setting of to be effective, response rates similar to those
allogeneic STEM CELL transplantation, in which the obtained with high-dose IL-2 therapy have been
Immunotherapy of cancer 137

observed in renal cell carcinoma patients [88]. A immunoglobulins with the desired specificities, sub-
variety of combination regimens has been tested, types and affinities for clinical use [96]. Examples of
especially with low-dose IL-2 and IFN-α for renal cell humanized mAb currently in clinical use against
carcinoma.For example,the combination of IL-2,IFN- cancer are shown in Table 2. The mechanism of
α and the cytotoxic drug 5-fluorouracil has yielded action of mAb against tumors is complex and highly
response rates of 2%, 16% and 39% in 3 major studies dependent on the nature of the target molecule.The
[89–91]. This large variation in response rates indi- effects of Ab can be enhanced by combination with
cates that there is no convincing evidence that com- cytoreductive treatment or CYTOKINES.Ab can activate
bination therapy is superior to that with IL-2 alone. effector functions by their Fc portions: antibody-
TNF-α is a pleiotropic agent with direct and indi- dependent cellular cytotoxicity (ADCC, in which the
rect antitumor effects, and is a mediator of septic mAb interact via their Fc portions with the Fc recep-
shock [92]. Hence, dose-limiting toxicity of systemic tors on phagocytic cells and NK LYMPHOCYTES) and
TNF-α is already encountered at concentrations 10- complement-dependent cytotoxicity (in which the
50 times lower than needed for antitumor effects in Fc receptors bind components of the complement
murine models [93].However,the surgical technique cascade).For example,ADCC appears to be the main
of isolated limb perfusion allows the application of effector mechanism for rituximab to eliminate B
therapeutically effective concentrations of TNF-α; cells [106]. In addition, Ab may also induce apopto-
response rates between 64% and 100% have been sis, cell cycle arrest, inhibition of cell proliferation as
reported when TNF-α was combined with the cyto- well as angiogenesis and metastatic spread.A case in
toxic drug melphalan for the treatment of in-transit point is trastuzumab whose mechanism of action
melanoma metastases (reviewed in [94]). The com- includes, besides activation of the immune system,
bination of TNF-α and melphalan was similarly effec- downregulation of the constitutive growth-signalling
tive to treat unresectable soft-tissue sarcoma, where properties of the Her-2 receptor network on cancer
limb amputation could be avoided in 64% to 90% of cells [107]. Finally, anticancer mAb may be covalent-
the patients [94]. Animal studies have revealed that ly linked to drugs in order to selectively deliver these
the combined use of TNF-α with melphalan rather to tumors with the aim to improve antitumor effica-
than the individual compounds is critical to destroy cy and reduce the systemic toxicity of therapy. An
tumors;TNF-α targets the tumor vasculature,allowing example of such an immunoconjugate is gemtuzum-
strongly enhanced accumulation of melphalan in ab ozogamicin, a humanized CD33 mAb conjugated
the tumor leading to tumor vessel destruction and with calicheamicin: a highly potent antitumor antibi-
metabolic shut-down of the tumor [95]. otic that cleaves double-stranded DNA. This com-
pound has been found effective in the treatment of
relapsed acute myelogenous leukemia [102].
Monoclonal antibodies (mAb)

The early clinical applications of murine mAb Cancer vaccination


against cancer in the 80s were hampered by the
development of human antimurine Ab (HAMA) The purpose of active antigen-specific immunothera-
responses that rendered these mAb ineffective by py, i.e., vaccination, against cancer is to induce poly-
neutralization and shortened their in vivo survival. clonal immune responses against TAA, with the
However, the subsequently developed technology to added benefits of (i) achieving more stable levels of
‘humanize’ murine mAb in the 90s to avoid the gen- these immune responses, and (ii) inducing immuno-
eration of HAMA revived the interest to treat cancer logical memory to obtain long-term immunosurveil-
with mAb. The genetic assembly of human lance against TAA-expressing cancer cells [108].
IMMUNOGLOBULIN framework regions with the COM- Early vaccination efforts made use of whole tumor
PLEMENTARITY DETERMINING REGIONS of murine Ab spe- cells or tumor lysates of autologous or allogeneic ori-
cific for human cancers allowed the creation of gin.These efforts were mainly directed at melanoma
138 Cancer immunity

TABLE 2. MONOCLONAL ANTITUMOR ANTIBODIES IN CLINICAL USE

Name Target Ag1 Clinical use2 Refs.

Alemtuzumab CD52 (Refractory) B-CLL, T-PLL [97, 98]


Bevacizumab VEGF Colorectal cancer (combined with [99]
cytoreductive therapy)
Cetuximab EGFR Head and neck cancer (combined with [100]
local radiotherapy)
Edrecolomab Ep-CAM Colorectal cancer [101]
Gemtuzumabozo-gamicin CD33 AML in first relapse [102]
Rituximab CD20 Various subgroups of B-NHL, B-CLL [103, 104]
Trastuzumab Her-2/neu Breast cancer [105]

1 Abbreviations used in this column: EGFR, epidermal growth factor receptor; Ep-CAM, epithelial cell adhesion mol-
ecule; Her-2/neu, human epidermal receptor-2/neurological; VEGF, vascular endothelial growth factor.
2 AML, acute myelogenous leukemia; B-CLL, B-chronic lymphocytic leukemia; B-NHL, B-non-Hodgkin’s lymphoma;
T-PLL, T-prolymphocytic leukemia.

and yielded little evidence of consistent efficacy. For native, unmodified peptides remained intact. An
example, long-term follow-up of patients with example is the melanoma protein gp100-derived
metastatic melanoma, randomized to observation peptide ITDQVPFSV, which is presented by HLA-
versus immunization with an allogeneic melanoma A*0201. CD8+ T cells specific for this peptide have
oncolysate, showed no significant differences in been isolated from tumors, but the MHC binding
overall or disease-free survival between both groups algorithm [22] predicts that methionine or leucine
[109]. Meanwhile, the definition of TAA (see above) rather than threonine at position 2 would have been
and the advent of sensitive techniques to detect TAA- optimal for binding to HLA-A*0201. Indeed, a variant
specific T-cell responses [110] had greatly augment- peptide with M substituted for T showed better
ed the ability to perform immunological assessments immunogenicity in vitro and in vivo [117].
in patients, but the few clinical responses to vaccina- Fully matured DC can be used in vitro and in vivo
tion hindered the ability to interpret these in vitro to generate T-cell reponses against TAA after they
assessments. have been loaded with TAA. Myeloid DC, derived
Preclinical experiments indicated that a variety from cultures of MONOCYTES or CD34+ hematopoietic
of methods could be used to vaccinate against TAA, precursor cells, have mostly been used in clinical tri-
such as the use of minimal determinant peptides als. Loading can be done with longer peptides con-
[111], proteins [112], viral oncolysates [113], TAA- taining both Class I and Class II MHC-presented epi-
encoding DNA [114], heat shock proteins [115], topes, recombinant protein or tumor lysates. In all
whole tumor cells [116], and DCs [9, 11–15]. The cases, the MHC molecules will be endogenously
modification and optimization of Class I MHC-bind- loaded with TAA after proteolytic processing. Of
ing ‘anchor’ residues in some minimal determinant interest is that vaccination with DC loaded with a
peptides was found to improve their immunogenici- MHC class II-restricted peptide derived from the
ty in vitro and in vivo, whilst their ability to generate MAGE-C2 antigen rapidly induced strong and pep-
CD8+ T cells that crossreact with and recognize the tide-specific TH1 RESPONSES in patients with metastatic
Immunotherapy of cancer 139

melanoma [118]. Also, DNA and RNA encoding TAA instrumental for the identification of TAA (see
can be used to express these ANTIGENS in DC. Early above). Treatment of melanoma patients with poly-
clinical trials have shown the feasibility of DC vacci- clonal autologous TIL plus IL-2 yielded complete or
nation, its lack of toxicity and its antitumor potential. partial responses in approximately one-third of the
Still, many parameters of the preparation of this type patients [27]. Alternatively, CD8+ CTL CLONEs isolated
of vaccine must be better defined to prove its antitu- and expanded from cultured peripheral blood
mor efficacy in larger clinical trials [119]. mononuclear cells were used and shown to be effec-
Alternatively, vaccination with cultured tumor tive for prevention of herpesvirus reactivations [127]
cells that have been genetically modified to produce and treatment of melanoma [128].When this form of
an immunostimulatory cytokine (e.g., IL-2, GM-CSF) treatment was preceded by cytoreductive therapy to
or to express an accessory molecule for T cells (e.g., induce lymphopenia, an extensive and persistent
CD80/CD86) on their surface, has been shown to clonal T-cell repopulation was observed.These T-cells
improve antitumor immune reactivity in animal had the original TAA specificity of the TIL, and were
models.The use of GM-CSF yielded the most consis- able to migrate to the tumor sites and induce partial
tent data in this respect [120]. Clinical application antitumor responses in almost half of the patients
requires that patients must have their tumor excised [15]. Such studies clearly indicate the promise of
in order to establish cell lines which then have to be adoptive cellular immunotherapy for cancer, provid-
transduced with a highly efficient retroviral vector ed that the tumor-specific T cells persist for pro-
encoding GM-CSF. Initial clinical trials showed some- longed periods of time in order to exert their antitu-
times significant in vitro but infrequent clinical anti- mor effects, and do not destroy normal tissues result-
tumor responses [121, 122]. ing in severe adverse events. For the long-term per-
sistence of adoptively transferred CD8+ T cells, CD4+
T-cell help is needed (see above and [129]), which
Adoptive autologous cellular immunotherapy currently is provided in surrogate form by recombi-
nant CYTOKINES such as IL-2.
Based on promising animal data, initial approaches However, adoptive transfer of tumor-specific CTL
to adoptive cellular immunotherapy of cancer have for cancer treatment can be severely hampered by
made use of ex vivo IL-2-activated peripheral blood the difficulty of reproducibly isolating and expand-
mononuclear cells that exhibited so-called lym- ing such LYMPHOCYTES. As an alternative, polyclonally
phokine-activated killer (LAK) activity [123]. LAK activated T cells have been ‘retargeted’, initially by
activity was defined as the capability to kill fresh and using bispecific mAb [130] and later by transfer of
cultured tumor cells, but not normal cells, in an genes encoding tumor-specific receptors [131].
MHC-unrestricted fashion. Patients with advanced Bispecific mAb can be directed towards both a TAA
melanoma and renal cell cancer were treated with and CD3, a part of the TCR complex. By coating poly-
high-dose IL-2 and up to 2 × 1011 autologous LYMPHO- clonal CTL with such bispecific mAbs, their tumor-
CYTES with LAK activity. Although complete and par- cell killing potential can be targeted to tumor cells
tial tumor regressions were seen in up to one-third of bearing that TAA.This principle has been tested clin-
patients [124], subsequent randomized studies ically for the locoregional treatment of advanced
attributed these responses to IL-2, whilst no evidence ovarian cancer. In spite of objective antitumor
for a benefit from LAK cells was demonstrated [125, responses at the site of treatment (i.e., intraperi-
126]. toneally) in one-third of patients, this approach
As an alternative to LAK as a source of antitumor proved to be complicated by limited accessibility of
activity,TIL were isolated, expanded ex vivo using IL- solid tumors to the therapy, dissociation of bi-specif-
2 to very high numbers (i.e., up to 1011) and rein- ic mAb from CTL, development of human anti-
fused into the patients. Such TIL cultures from mouse ANTIBODY responses and the limited capacity
melanoma lesions frequently yielded CTL CLONEs of mAb-sensitized T LYMPHOCYTES to recycle their
with HLA-restricted tumor recognition,which proved tumoricidal capacity [130]. The transfer of genes
140 Cancer immunity

encoding TAA-specific receptors into T cells (i.e., post-SCT, patients received additional immunosup-
genetic retargeting) became feasible as an alterna- pression to allow engraftment and suppress GVHD.
tive to bispecific mAb-mediated retargeting after the During this time most patients showed growth of
development of highly efficient protocols for retro- their tumors. After full engraftment, immunosuppres-
virus-mediated gene transfer into polyclonally acti- sion was tapered (and to some patients CYTOKINES or
vated T cells and for rapid expansion of the trans- donor lymphocyte infusions were given) until GVHD
duced cells, all under ‘good manufacturing practice’ was sufficiently suppressed or stabilised at a low
(GMP) conditions [131]. Autologous T LYMPHOCYTES grade. Among 33 patients treated in this way, 15
genetically retargeted to carboxyanhydrase 9 by a showed evidence of regression of their renal tumors
single-chain Ab-type receptor specific for this TAA [136]. However, the GVT activity with these regimens
[55] are currently being investigated clinically in is often associated with severe and life-threatening
patients with metastatic renal cell cancer [132]. GVHD,which may lead to a treatment-related mortal-
ity between 10% and 20%. The characterization of
antigenic targets with limited tissue distribution, per-
Allogeneic hematopoietic stem cell mitting donor LYMPHOCYTES to preferentially target
transplantation (SCT) malignant cells and not critical normal tissues, cou-
pled with methods to select and/or generate T cells
High-dose chemoradiotherapy followed by rescue with such specificities, should provide a much-need-
from the resulting ablation of hematopoietic func- ed improvement to this approach.
tion with SCT from an HLA-matched donor has
become standard therapy for many hematological
malignancies. One problem with this treatment is Conclusions and future developments
graft-versus-host disease (GVHD) due to donor-
derived T cells recognizing mismatched mHAg It has now been firmly established that the human
expressed by normal tissues of the host.As the malig- immune system is capable of recognizing and elimi-
nant cells that survive chemoradiotherapy are also of nating spontaneously arising tumors, although can-
host origin, patients who develop GVHD have less cer cells are generally less immunogenic than micro-
frequent recurrence of the original disease due to bial pathogens such as bacteria, fungi and viruses.
the associated graft-versus-tumor (GVT) effect.T cells Nevertheless, immunotherapeutic modalities, such
mediate this antitumor activity,because (1) infusions as CYTOKINES and mAb, already have become compo-
of T cells from the SCT donor to treat leukemic nents of several standard treament regimens of
relapse after SCT sometimes result in complete human malignancies. Many vaccines have advanced
remissions, and (2) the complete remissions through preliminary testing to efficacy trials, and
observed after so-called non-myeloablative SCT must have shown little toxicity but also limited effects in
result from GVT effects because the reduced-intensi- patients with established tumors. Tumors frequently
ty cytoreductive therapy cannot eliminate all resid- interfere with the development and function of
ual disease [133].This concept has been extended to immune responses. Thus, one of the challenges for
the treatment of solid tumors.Whilst preliminary data cancer immunotherapy is to use advances in cellular
from several groups provide evidence for a GVT and molecular immunology to develop strategies
effect in renal cell cancer, no such effect has been that effectively and safely augment antitumor
observed in advanced melanoma and only anecdot- responses.With our increasing understanding of the
al data are available on other solid tumors [134,135]. requirements for immune cell activation, homing
Several significant clinical antitumor responses were and accumulation at tumor sites, and for the disrup-
seen in patients with metastatic renal cell cancer tion of regulatory mechanisms that inhibit immuno-
undergoing nonmyeloablative SCT from an HLA- logical anticancer responses at the sites of the
matched sibling after preparation with cyclophos- tumors, our abilities to design and engineer
phamide and fludarabine. During the first 100 days immunotherapeutic approaches with antitumor
Summary 141

capacities beyond what can be elicited from the nor- tively and safely augment antitumor responses.These
mal immune system should advance. advances have enabled the characterization of so-
Initial clinical trials of adoptive transfer of large called TUMOR-ASSOCIATED ANTIGENS (TAA) which can
numbers of autologous tumor-reactive T LYMPHOCYTES function as targets for the immune system.These TAA
have shown promising antitumor effects, in particu- include peptides that can be recognized by T LYM-
lar against melanoma. The ability to transfer genes PHOCYTES as they are presented by class I or II HLA
with high efficiency into polyclonally activated T molecules, or intact proteins or carbohydrates that
cells has raised wide interest in the genetic retarget- are not presented by HLA molecules and are recog-
ing of T LYMPHOCYTES against tumors with the poten- nized by ANTIBODIES.TAA can be classified into (1) tis-
tial to circumvent the complex and cumbersome sue-specific differentiation antigens, (2) ‘cancer-
procedures to isolate and expand ‘spontaneous’ testis’ antigens, (3) normally occurring antigens that
tumor-specific T cells.However,these approaches are are overexpressed, (4) fusion proteins, (5) mutation-
based on end-stage T cells with probably limited sur- al antigens, (6) virally encoded antigens and (7)
vival times upon administration.The transfer of TAA- minor histocompatibility antigens. Various forms of
specific receptors to hematopoietic STEM CELLs (HSC) adoptive tumor immunotherapy have been devel-
in order to retarget their progeny against cancer oped, including (combinations of) CYTOKINES, mono-
would appear to be a solution for this problem, but clonal antibodies, and autologous cellular immuno-
the observation of oncogenic events after transfer of therapy – initially using nonspecifi-cally expanded
a growth factor receptor-encoding gene into HSC LYMPHOCYTES,later with expanded TAA-specific T cells
[137] has tempered the enthusiasm for this derived from tumor infiltrates. The active forms of
approach. Meanwhile, transplantation of allogeneic tumor immunotherapy include vaccination and allo-
HSC (following pretreatment with a reduced-intensi- geneic hematopoietic STEM CELL (HSC) transplanta-
ty cytoreductive regimen) has emerged as a promis- tion. The latter is currently a standard treatment for
ing approach to provide cancer patients with long- hematological malignancies but still experimental
term immunosurveillance against their tumors,albeit for solid tumors. As HSC can be a life-long source of
currently with significant toxicity [136]. The expres- antitumor T cells, we believe that this approach has
sion of so-called mHAg, constituting TAA, by tumor the potential to become a very powerful
cells is of key importance for the concept of allo- immunotherapeutic modality of cancer.
immunotherapy of cancer. The complexes of MHC
and allo-TAA are probably more immunogenic than
those of MHC and self-TAA that are the targets of clas- Selected readings
sical, autologous T-cell-based immunotherapy [77].
This characteristic, combined with the fact that a Zinkernagel RM, Doherty PC (1997) The discovery of MHC
long-term source of antitumor immunosurveillance restriction. Immunol Today 18: 14–17
in the form of HSC is provided, has the potential to Carter P (2001) Improving the efficacy of antibody-based
make alloimmunotherapy a very powerful form of cancer therapies. Nat Rev Cancer 1: 118–129
immunotherapy of cancer. Eggermont AM, de Wilt JH, ten Hagen TL (2003) Current
uses of isolated limb perfusion in the clinic and a
model system for new strategies. Lancet Oncol 4:
Summary 429–437
Berzofsky JA,Terabe M, Oh S, Belyakov IM, Ahlers JD, Janik
The human immune system is capable of recogniz- JE, Morris JC (2004) Progress on new vaccine strate-
ing and eliminating spontaneously arising tumors. gies for the immunotherapy and prevention of cancer.
However, tumors frequently interfere with the devel- J Clin Invest 113: 1515–1525
opment and function of immune responses.With the Childs RW, Barrett J (2004) Nonmyeloablative allogeneic
recent advances in cellular and molecular immunol- immunotherapy for solid tumors. Annu Rev Med 55:
ogy, strategies are now being developed that effec- 459–475
142 Cancer immunity

References Coleman D, Niedzwiecki D, Gilboa E,Vieweg J (2003)


Immunological and clinical responses in metastatic
1 Ehrlich P (1909) Über den jetzigen Stand der Karzi- renal cancer patients vaccinated with tumor RNA-
nomforschung. Ned Tijdschr Geneesk 53: 273–290 transfected dendritic cells. Cancer Res 63: 2127–2133
2 Thomas L (1959) Mechanisms involved in tissue dam- 14 O’Rourke MG, Johnson M, Lanagan C, See J,Yang J, Bell
age by the endotoxins of gram negative bacteria. In: JR, Slater GJ, Kerr BM, Crowe B, Purdie DM et al (2003)
HS Lawrence (ed): Cellular and Humoral Aspects of Durable complete clinical responses in a phase I/II
the Hypersensitive States. Hoeber-Harper, New York, trial using an autologous melanoma cell/dendritic
451–468 cell vaccine. Cancer Immunol Immunother 52: 387–395
3 Burnet FM (1970) The concept of immunological sur- 15 Dudley ME,Wunderlich JR, Robbins PF,Yang JC, Hwu P,
veillance. Prog Exp Tumor Res 13: 1–27 Schwartzentruber DJ,Topalian SL, Sherry R, Restifo NP,
4 Vakkila J, Lotze MT (2003) Cellular immunotherapy of Hubicki AM et al (2002) Cancer regression and
cancer. Blood Ther Med 3: 84–90 autoimmunity in patients after clonal repopulation
5 Pollock BH, Jenson HB, Leach CT, McClain KL, Hutchi- with antitumor lymphocytes. Science 298: 850–854
son RE, Garzarella L, Joshi VV, Parmley RT, Murphy SB 16 Freedman RS, Kudelka AP, Kavanagh JJ, Verschraegen
(2003) Risk factors for pediatric human immunodefi- C,Edwards CL,Nash M,Levy L,Atkinson EN,Zhang HZ,
ciency virus-related malignancy. JAMA 289: 2393–2399 Melichar B et al (2000) Clinical and biological effects
6 Yang L,Yamagata N,Yadav R, Brandon S, Courtney RL, of intraperitoneal injections of recombinant interfer-
Morrow JD, Shyr Y, Boothby M, Joyce S, Carbone DP et on-gamma and recombinant interleukin 2 with or
al (2003) Cancer-associated immunodeficiency and without tumor-infiltrating lymphocytes in patients with
dendritic cell abnormalities mediated by the prosta- ovarian or peritoneal carcinoma. Clin Cancer Res 6:
glandin EP2 receptor. J Clin Invest 111: 727–735 2268–2278
7 Euvrard S, Kanitakis J, Claudy A (2003) Skin cancers 17 Figlin RA, Thompson JA, Bukowski RM,Vogelzang NJ,
after organ transplantation. N Engl J Med 348: 1681- Novick AC, Lange P, Steinberg GD, Belldegrun AS
1691 (1999) Multicenter, randomized, phase III trial of
8 Renkvist N, Castelli C, Robbins PF, Parmiani G (2001) A CD8(+) tumor-infiltrating lymphocytes in combination
listing of human tumor antigens recognized by T cells. with recombinant interleukin-2 in metastatic renal
Cancer Immunol Immunother 50: 3–15 cell carcinoma. J Clin Oncol 17: 2521–2529
9 Tanaka H, Shimizu K, Hayashi T, Shu S (2002) Thera- 18 Rosenberg SA, Yannelli JR, Yang JC, Topalian SL,
peutic immune response induced by electrofusion of Schwartzentruber DJ, Weber JS, Parkinson DR, Seipp
dendritic and tumor cells. Cell Immunol 220: 1-12 CA, Einhorn JH,White DE (1994) Treatment of patients
10 Muller MR, Grunebach F, Nencioni A, Brossart P (2003) with metastatic melanoma with autologous tumor-
Transfection of dendritic cells with RNA induces CD4- infiltrating lymphocytes and interleukin 2. J Natl Can-
and CD8-mediated T cell immunity against breast car- cer Inst 86: 1159–1166
cinomas and reveals the immunodominance of pre- 19 Gorelik L, Flavell RA (2001) Immune-mediated eradi-
sented T cell epitopes. J Immunol 170: 5892–5896 cation of tumors through the blockade of transform-
11 Cui Y, Kelleher E, Straley E, Fuchs E, Gorski K, Levitsky ing growth factor-beta signaling in T cells. Nat Med 7:
H, Borrello I, Civin CI, Schoenberger SP, Cheng L et al 1118–1122
(2003) Immunotherapy of established tumors using 20 Chia CS, Ban K, Ithnin H, Singh H, Krishnan R, Mokhtar
bone marrow transplantation with antigen gene-mod- S, Malihan N, Seow HF (2002) Expression of inter-
ified hematopoietic stem cells. Nat Med 9: 952–958 leukin-18, INTERFERON-gamma and interleukin-10 in
12 Kokhaei P, Rezvany MR, Virving L, Choudhury A, Rab- hepatocellular carcinoma. Immunol Lett 84: 163–172
bani H, Osterborg A, Mellstedt H (2003) dendritic cells 21 Pieters J (2000) MHC class II-restricted antigen pro-
loaded with apoptotic tumour cells induce a stronger cessing and presentation. Adv Immunol 75: 159-208
T-cell response than dendritic cell-tumour hybrids in 22 Falk K, Rotzschke O, Stevanovic S, Jung G, Rammensee
B-CLL. Leukemia 17: 894–899 HG (1991) Allele-specific motifs revealed by sequenc-
13 Su Z, Dannull J, Heiser A,Yancey D, Pruitt S, Madden J,
References 143

ing of self-peptides eluted from MHC molecules. 35 Carter P (2001) Improving the efficacy of antibody-
Nature 351: 290–296 based cancer therapies. Nat Rev Cancer 1: 118–129
23 Hunziker L, Klenerman P, Zinkernagel RM, Ehl S (2002) 36 Sahin U,Tureci O, Schmitt H, Cochlovius B, Johannes T,
Exhaustion of cytotoxic T cells during adoptive Schmits R, Stenner F, Luo G, Schobert I, Pfreundschuh
immunotherapy of virus carrier mice can be prevent- M (1995) Human neoplasms elicit multiple specific
ed by B cells or CD4+ T cells.Eur J Immunol 2: 374–382 immune responses in the autologous host. Proc Natl
24 Marzo AL, Kinnear BF, Lake RA, Frelinger JJ, Collins EJ, Acad Sci USA 92: 11810–11813
Robinson BW, Scott B (2000) Tumor-specific CD4+ T 37 Chen YT, Scanlan MJ, Sahin U,Tureci O, Gure AO,Tsang
cells have a major “post-licensing” role in CTL mediat- S, Williamson B, Stockert E, Pfreundschuh M, Old LJ
ed anti-tumor immunity. J Immunol 165: 6047–6055 (1997) A testicular antigen aberrantly expressed in
25 Schoenberger SP,Toes RE, van der Voort EI, Offringa R, human cancers detected by autologous antibody
Melief CJ (1998) T-cell help for cytotoxic T lympho- screening. Proc Natl Acad Sci USA 94: 1914–1918
cytes is mediated by CD40-CD40L interactions. Nature 38 Jager E, Jager D, Karbach J, Chen YT, Ritter G, Nagata Y,
393: 480–483 Gnjatic S, Stockert E,Arand M, Old LJ et al (2000) Iden-
26 Surman DR, Dudley ME, Overwijk WW, Restifo NP tification of NY-ESO-1 epitopes presented by human
(2000) Cutting edge: CD4+ T cell control of CD8+ T cell histocompatibility antigen (HLA)-DRB4*0101-0103
reactivity to a model tumor antigen. J Immunol 164: and recognized by CD4(+) T lymphocytes of patients
562–565 with NY-ESO-1-expressing melanoma. J Exp Med 191:
27 Bevan MJ (2004) Helping the CD8(+) T-cell response. 625–630
Nat Rev Immunol 4: 595–602 39 Wang RF,Johnston SL,Zeng G,Topalian SL,Schwartzen-
28 Ibe S, Qin Z, Schuler T, Preiss S, Blankenstein T (2001) truber DJ, Rosenberg SA (1998) A breast and
Tumor rejection by disturbing tumor stroma cell inter- melanoma-shared tumor antigen: T cell responses to
actions. J Exp Med 194: 1549–1559 antigenic peptides translated from different open
29 Wang HY, Lee DA, Peng G, Guo Z, Li Y, Kiniwa Y, Shevach reading frames. J Immunol 161: 3598–3606
EM,Wang RF (2004) Tumor-specific human CD4+ regu- 40 Kawakami Y, Eliyahu S, Delgado CH, Robbins PF, Sak-
latory T cells and their ligands: implications for aguchi K, Appella E, Yannelli JR, Adema GJ, Miki T,
immunotherapy. Immunity 20: 107–118 Rosenberg SA (1994) Identification of a human
30 Gavin M, Rudensky A (2003) Control of immune melanoma antigen recognized by tumor-infiltrating
homeostasis by naturally arising regulatory CD4+ T lymphocytes associated with in vivo tumor rejection.
cells. Curr Opin Immunol 15: 690–696 Proc Natl Acad Sci USA 91: 6458–6462
31 Zinkernagel RM, Doherty PC (1997) The discovery of 41 Bakker AB, Schreurs MW, de Boer AJ, Kawakami Y,
MHC restriction. Immunol Today 18: 14–17 Rosenberg SA, Adema GJ, Figdor CG (1994)
32 Carrel S, Johnson JP (1993) Immunologic recognition Melanocyte lineage-specific antigen gp100 is recog-
of malignant melanoma by autologous T lympho- nized by melanoma-derived tumor-infiltrating lympho-
cytes. Curr Opin Oncol 5: 383–389 cytes. J Exp Med 179: 1005–1009
33 Van der Bruggen P, Traversari C, Chomez P, Lurquin C, 42 Brichard V, Van Pel A, Wolfel T, Wolfel C, De Plaen E,
De Plaen E,Van den Eynde B, Knuth A, Boon T (1991) Lethe B, Coulie P, Boon T (1993) The tyrosinase gene
A gene encoding an antigen recognized by cytolytic T codes for an antigen recognized by autologous
lymphocytes on a human melanoma. Science 254: cytolytic T lymphocytes on HLA-A2 melanomas. J Exp
1643–1647 Med 178: 489–495
34 Traversari C, van der Bruggen P, Luescher IF, Lurquin C, 43 Epstein JI (1993) PSA and PAP as immunohistochemi-
Chomez P, Van Pel A, De Plaen E, Amar-Costesec A, cal markers in prostate cancer. Urol Clin North Am 20:
Boon T (1992) A nonapeptide encoded by human 757–770
gene MAGE-1 is recognized on HLA-A1 by cytolytic T 44 Israeli RS, Powell CT, Corr JG, Fair WR, Heston WD
lymphocytes directed against tumor antigen MZ2-E. J (1994) Expression of the prostate-specific membrane
Exp Med 176: 1453–1457 antigen. Cancer Res 54: 1807–1811
45 Cancer Immunome Database. URL: http://www2.licr.
144 Cancer immunity

org/CancerImmunomeDB/ (Accessed September Jonas U, Zwartendijk J,Warnaar SO (1986) Monoclon-


2004) al ANTIBODY G 250 recognizes a determinant present
46 Harada M, Kobayashi K, Matsueda S, Nakagawa M, in renal-cell carcinoma and absent from normal kid-
Noguchi M, Itoh K (2003) Prostate-specific antigen- ney. Int J Cancer 38: 489–494
derived epitopes capable of inducing cellular and 56 Berinstein NL (2002) Carcinoembryonic antigen as a
humoral responses in HLA-A24+ prostate cancer target for therapeutic anticancer vaccines: a review. J
patients. Prostate 57: 152–159 Clin Oncol 20: 2197–2207
47 Kobayashi K,Noguchi M,Itoh K,Harada M (2003) Iden- 57 Fisk B, Blevins TL, Wharton JT, Ioannides CG (1995)
tification of a prostate-specific membrane antigen- Identification of an immunodominant peptide of HER-
derived peptide capable of eliciting both cellular and 2/neu protooncogene recognized by ovarian tumor-
humoral immune responses in HLA-A24+ prostate can- specific cytotoxic T-lymphocyte lines. J Exp Med 181:
cer patients. Cancer Sci 94: 622–627 2109–2117
48 De Plaen E,Arden K,Traversari C, Gaforio JJ, Szikora JP, 58 Scanlan MJ, Gout I, Gordon CM,Williamson B, Stockert
De Smet C, Brasseur F, van der Bruggen P, Lethe B, E, Gure AO, Jager D, Chen YT, Mackay A, O’Hare MJ et al
Lurquin C et al (1994) Structure, chromosomal local- (2001) Humoral immunity to human breast cancer:
ization, and expression of 12 genes of the MAGE fami- antigen definition and quantitative analysis of mRNA
ly. Immunogenetics 40: 360–369 expression. Cancer Immun 1: 4
49 Boël P,Wildmann C, Sensi ML, Brasseur R, Renauld JC, 59 Vissers JL, De Vries IJ, Engelen LP, Scharenborg NM,
Coulie P, Boon T, van der Bruggen P (1995) BAGE: a Molkenboer J, Figdor CG, Oosterwijk E, Adema GJ
new gene encoding an antigen recognized on human (2002) Renal cell carcinoma-associated antigen G250
melanomas by cytolytic T lymphocytes. Immunity 2: encodes a naturally processed epitope presented by
167–175 human leukocyte antigen-DR molecules to CD4(+) T
50 Van den Eynde B, Peeters O, De Backer O, Gaugler B, lymphocytes. Int J Cancer 100: 441–444
Lucas S, Boon T (1995) A new family of genes coding 60 Kurzrock R, Gutterman JU,Talpaz M (1988) The molec-
for an antigen recognized by autologous cytolytic T ular genetics of Philadelphia chromosome-positive
lymphocytes on a human melanoma. J Exp Med 182: leukemias. N Engl J Med 319: 990–998
689–698 61 Ferrucci PF, Grignani F, Pearson M, Fagioli M, Nicoletti I,
51 Zippelius A, Batard P, Rubio-Godoy V, Bioley G, Lienard Pelicci PG (1997) Cell death induction by the acute
D, Lejeune F, Rimoldi D, Guillaume P, Meidenbauer N, promyelocytic leukemia-specific PML/RARalpha
Mackensen A et al (2004) Effector function of human fusion protein.Proc Natl Acad Sci USA 94: 10901–10906
tumor-specific CD8 T cells in melanoma lesions: a 62 Dermime S, Bertazzoli C, Marchesi E, Ravagnani F,
state of local functional tolerance. Cancer Res 64: Blaser K, Corneo GM, Pogliani E, Parmiani G, Gamba-
2865–2873 corti-Passerini C (1996) Lack of T-cell-mediated recog-
52 Gotter J, Brors B, Hergenhahn M, Kyewski B (2004) nition of the fusion region of the pml/RAR-alpha
Medullary epithelial cells of the human thymus hybrid protein by lymphocytes of acute promyelocyt-
express a highly diverse selection of tissue-specific ic leukemia patients. Clin Cancer Res 2: 593–600
genes colocalized in chromosomal clusters.J Exp Med 63 Clark RE, Dodi IA, Hill SC, Lill JR, Aubert G, Macintyre
199: 155–166 AR, Rojas J, Bourdon A, Bonner PL,Wang L et al (2001)
53 Tsang KY, Zaremba S, Nieroda CA, Zhu MZ, Hamilton Direct evidence that leukemic cells present HLA-asso-
JM, Schlom J (1995) Generation of human cytotoxic T ciated immunogenic peptides derived from the BCR-
cells specific for human carcinoembryonic antigen ABL b3a2 fusion protein. Blood 98: 2887–2893
epitopes from patients immunized with recombinant 64 Yasukawa M,Ohminami H,Kojima K,Hato T,Hasegawa
vaccinia-CEA vaccine. J Natl Cancer Inst 87: 982–990 A, Takahashi T, Hirai H, Fujita S (2001) HLA class II-
54 Menard S, Pupa SM, Campiglio M, Tagliabue E (2003) restricted antigen presentation of endogenous bcr-abl
Biologic and therapeutic role of HER2 in cancer. fusion protein by chronic myelogenous leukemia-
Oncogene 22: 6570–6578 derived dendritic cells to CD4(+) T lymphocytes.
55 Oosterwijk E, Ruiter DJ, Hoedemaeker PJ, Pauwels EK, Blood 98: 1498–1505
References 145

65 Talpaz M, Qiu X, Cheng K, Cortes JE, Kantarjian H, human papillomavirus type 16 in infected women.
Kurzrock R (2000) Autoantibodies to Abl and Bcr pro- Clin Diagn Lab Immunol 9: 877–882
teins. Leukemia 14: 1661–1666 76 Baay MF, Duk JM, Burger MP, de Bruijn HW, Stolz E, Her-
66 Gambacorti-Passerini C, Grignani F, Arienti F, Pandolfi brink P (1999) Humoral immune response against
PP, Pelicci PG, Parmiani G (1993) Human CD4 lympho- proteins E6 and E7 in cervical carcinoma patients pos-
cytes specifically recognize a peptide representing the itive for human papilloma virus type 16 during treat-
fusion region of the hybrid protein pml/RAR alpha ment and follow-up. Eur J Clin Microbiol Infect Dis 18:
present in acute promyelocytic leukemia cells. Blood 126–132
81: 1369–1375 77 Spierings E, Wieles B, Goulmy E (2004) Minor histo-
67 Scanlan MJ,Chen YT,Williamson B,Gure AO,Stockert E, compatibility antigens big in tumour therapy. Trends
Gordan JD, Tureci O, Sahin U, Pfreundschuh M, Old LJ Immunol 25: 56–60
(1998) Characterization of human colon cancer anti- 78 Goulmy E (1997) Minor histocompatibility antigens:
gens recognized by autologous antibodies. Int J Can- from T cell recognition to peptide identification. Hum
cer 76: 652–658 Immunol 54: 8–14
68 Stone B,Schummer M,Paley PJ,Thompson L,Stewart J, 79 Mutis T, Gillespie G, Schrama E, Falkenburg JH, Moss P,
Ford M, Crawford M, Urban N, O’Briant K, Nelson BH Goulmy E (1999) Tetrameric HLA class I minor histo-
(2003) Serologic analysis of ovarian tumor antigens compatibility antigen peptide complexes demonstrate
reveals a bias toward antigens encoded on 17q. Int J minor histocompatibility antigen-specific cytotoxic T
Cancer 104: 73–84 lymphocytes in patients with graft-versus-host disease.
69 Robbins PF, El-Gamil M, Li YF, Kawakami Y, Loftus D, Nat Med 5: 839–842
Appella E, Rosenberg SA (1996) A mutated beta- 80 Den Haan JM, Meadows LM, Wang W, Pool J, Blokland
catenin gene encodes a melanoma-specific antigen E, Bishop TL, Reinhardus C, Shabanowitz J, Offringa R,
recognized by tumor infiltrating lymphocytes. J Exp Hunt DF et al (1998) The minor histocompatibility
Med 183: 1185–1192 antigen HA-1: a diallelic gene with a single amino acid
70 Li HP, Chang YS (2003) Epstein-Barr virus latent mem- polymorphism. Science 279: 1054–1057
brane protein 1: structure and functions. J Biomed Sci 81 Akatsuka Y, Nishida T, Kondo E, Miyazaki M,Taji H, Iida
10: 490–504 H, Tsujimura K, Yazaki M, Naoe T, Morishima Y et al
71 Munger K, Basile JR, Duensing S, Eichten A, Gonzalez (2003) Identification of a polymorphic gene, BCL2A1,
SL, Grace M, Zacny VL (2001) Biological activities and encoding two novel hematopoietic lineage-specific
molecular targets of the human papillomavirus E7 minor histocompatibility antigens. J Exp Med 197:
oncoprotein. Oncogene 20: 7888–7898 1489–1500
72 Mantovani F, Banks L (2001) The human papillo- 82 Grever M, Kopecky K, Foucar MK, Head D, Bennett JM,
mavirus E6 protein and its contribution to malignant Hutchison RE, Corbett WE, Cassileth PA, Habermann T,
progression. Oncogene 20: 7874–7887 Golomb H et al (1995) Randomized comparison of
73 Subklewe M, Chahroudi A, Bickham K, Larsson M, pentostatin versus interferon alfa-2a in previously
Kurilla MG,Bhardwaj N,Steinman RM (1999) Presenta- untreated patients with hairy cell leukemia: an inter-
tion of Epstein-Barr virus latency antigens to CD8(+), group study. J Clin Oncol 13: 974–982
interferon-gamma-secreting, T lymphocytes. Eur J 83 Krown SE (2001) Management of Kaposi sarcoma: the
Immunol 29: 3995– 4001 role of interferon and thalidomide. Curr Opin Oncol
74 Xu J, Ahmad A, D’Addario M, Knafo L, Jones JF, Prasad 13: 374–381
U,Dolcetti R,Vaccher E,Menezes J (2000) Analysis and 84 Medical Research Council Renal Cancer Collabora-
significance of anti-latent membrane protein-1 anti- tors (1999) interferon alpha and survival in metastatic
bodies in the sera of patients with EBV-associated dis- renal carcinoma: early results of a randomised con-
eases. J Immunol 164: 2815–2822 trolled trial. Lancet 353: 14–17
75 Nakagawa M,Viscidi R,Deshmukh I,Costa MD,Palefsky 85 Pyrhonen S, Salminen E, Ruutu M, Lehtonen T, Nurmi
JM, Farhat S, Moscicki AB (2002) Time course of M, Tammela T, Juusela H, Rintala E, Hietanen P, Kel-
humoral and cell-mediated immune responses to lokumpu-Lehtinen PL (1999) Prospective randomized
146 Cancer immunity

trial of interferon alfa-2a plus vinblastine versus vin- 95 De Wilt JH, ten Hagen TL, de Boeck G, van Tiel ST, de
blastine alone in patients with advanced renal cell Bruijn EA, Eggermont AM (2000) Tumour necrosis fac-
cancer. J Clin Oncol 17: 2859–2867 tor alpha increases melphalan concentration in
86 Eggermont AM, Punt CJ (2003) Does adjuvant sys- tumour tissue after isolated limb perfusion.Br J Cancer
temic therapy with interferon-alpha for stage II-III 82: 1000–1003
melanoma prolong survival? Am J Clin Dermatol 4: 96 Presta LG, Chen H, O’Connor SJ, Chisholm V, Meng YG,
531–536 Krummen L, Winkler M, Ferrara N (1997) Humaniza-
87 Rosenberg SA,Yang JC,White DE, Steinberg SM (1998) tion of an anti-vascular endothelial growth factor
Durability of complete responses in patients with monoclonal antibody for the therapy of solid tumors
metastatic cancer treated with high-dose interleukin- and other disorders. Cancer Res 57: 4593–4599
2: identification of the antigens mediating response. 97 Keating MJ, Flinn I, Jain V, Binet JL, Hillmen P, Byrd J,
Ann Surg 228: 307–319 Albitar M, Brettman L, Santabarbara P, Wacker B et al
88 Tourani JM, Lucas V, Mayeur D, Dufour B, DiPalma M, (2002) Therapeutic role of alemtuzumab (Cam-path-
Boaziz C, Grise P,Varette C, Pavlovitch JM, Pujade-Lau- 1H) in patients who have failed fludarabine: results of
raine E et al (1996) Subcutaneous recombinant inter- a large international study. Blood 99: 3554–3561
leukin-2 (rIL-2) in out-patients with metastatic renal 98 Dearden CE,Matutes E,Catovsky D (2002) Alemtuzum-
cell carcinoma. Results of a multicenter SCAPP1 trial. ab in T-cell malignancies. Med Oncol 19 (Suppl):
Ann Oncol 7: 525–528 S27–S32
89 Ravaud A, Audhuy B, Gomez F, Escudier B, Lesimple T, 99 Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T,
Chevreau C,Douillard JY,Caty A,Geoffrois L,Ferrero JM Hainsworth J, Heim W, Berlin J, Baron A, Griffing S,
et al (1998) Subcutaneous interleukin-2, interferon Holmgren E et al (2004) Bevacizumab plus irinotecan,
alfa-2a, and continuous infusion of fluorouracil in fluorouracil, and leucovorin for metastatic colorectal
metastatic renal cell carcinoma: a multicenter phase II cancer. N Engl J Med 350: 2335–2342
trial. Groupe Francais d’Immunotherapie. J Clin Oncol 100 Robert F, Ezekiel MP, Spencer SA, Meredith RF, Bonner
16: 2728–2732 JA, Khazaeli MB, Saleh MN, Carey D, LoBuglio AF,
90 Dutcher JP, Atkins M, Fisher R, Weiss G, Margolin K, Wheeler RH et al (2001) Phase I study of anti-epider-
Aronson F, Sosman J, Lotze M, Gordon M, Logan T et al mal growth factor receptor antibody cetuximab in
(1997) Interleukin-2-based therapy for metastatic combination with radiation therapy in patients with
renal cell cancer: the Cytokine Working Group experi- advanced head and neck cancer. J Clin Oncol 19:
ence, 1989–1997. Cancer J Sci Am 3 (Suppl 1): S73–S78 3234–3243
91 Lopez Hanninen E, Kirchner H, Atzpodien J (1996) 101 Riethmuller G, Holz E, Schlimok G, Schmiegel W, Raab
Interleukin-2 based home therapy of metastatic renal R,Hoffken K,Gruber R,Funke I,Pichlmaier H,Hirche H
cell carcinoma: risks and benefits in 215 consecutive et al (1998) Monoclonal ANTIBODY therapy for resect-
single institution patients. J Urol 155: 19–25 ed Dukes’ C colorectal cancer: seven year outcome of
92 Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, a multicenter randomized trial. J Clin Oncol 16:
Williamson B (1975) An endotoxin-induced serum 1788–1794
factor that causes necrosis of tumors. Proc Natl Acad 102 Sievers EL, Larson RA, Stadtmauer EA, Estey E, Lowen-
Sci USA 72: 3666–3670 berg B, Dombret H, Karanes C, Theobald M, Bennett
93 Asher A, Mule JJ, Reichert CM, Shiloni E, Rosenberg SA JM, Sherman ML et al [Mylotarg Study Group] (2001)
(1987) Studies on the anti-tumor efficacy of systemi- Efficacy and safety of gemtuzumab ozogamicin in
cally administered recombinant tumor necrosis factor patients with CD33-positive acute myeloid leukemia in
against several murine tumors in vivo. J Immunol 138: first relapse. J Clin Oncol 19: 3244–3254
963–974 103 McLaughlin P, Grillo-Lopez AJ, Link BK, Levy R, Czucz-
94 Eggermont AM, de Wilt JH, ten Hagen TL (2003) Cur- man MS, Williams ME, Heyman MR, Bence-Bruckler I,
rent uses of isolated limb perfusion in the clinic and a White CA, Cabanillas F et al (1998) Rituximab
model system for new strategies. Lancet Oncol 4: chimeric anti-CD20 monoclonal antibody therapy for
429–437 relapsed indolent lymphoma: half of patients respond
References 147

to a four-dose treatment program. J Clin Oncol 16: Huang L, Shu S, Gordon D, Chang AE (1993) Direct
2825–2833 gene transfer with DNA-liposome complexes in
104 Lin TS, Lucas MS, Byrd JC (2003) Rituximab in B-cell melanoma: expression, biologic activity, and lack of
chronic lymphocytic leukemia. Semin Oncol 30: toxicity in humans. Proc Natl Acad Sci USA 90:
483–492 11307–11311
105 Cobleigh MA,Vogel CL,Tripathy D, Robert NJ, Scholl S, 115 Castelli C, Rivoltini L, Rini F, Belli F, Testori A, Maio M,
Fehrenbacher L,Wolter JM, Paton V, Shak S, Lieberman Mazzaferro V,Coppa J,Srivastava PK,Parmiani G (2004)
G et al (1999) Multinational study of the efficacy and Heat shock proteins: biological functions and clinical
safety of humanized anti-HER2 monoclonal antibody application as personalized vaccines for human can-
in women who have HER2-overexpressing metastatic cer. Cancer Immunol Immunother 53: 227–233
breast cancer that has progressed after chemotherapy 116 Ward S, Casey D, Labarthe MC,Whelan M, Dalgleish A,
for metastatic disease. J Clin Oncol 17: 2639–2648 Pandha H,Todryk S (2002) Immunotherapeutic poten-
106 Uchida J, Hamaguchi Y, Oliver JA, Ravetch JV, Poe JC, tial of whole tumour cells. Cancer Immunol Immuno-
Haas KM, Tedder TF (2004) The innate mononuclear ther 51: 351–357
phagocyte network depletes B lymphocytes through 117 Parkhurst MR, Salgaller ML, Southwood S, Robbins PF,
Fc receptor-dependent mechanisms during anti-CD20 Sette A, Rosenberg SA, Kawakami Y (1996) Improved
antibody immunotherapy. J Exp Med 199: 1659–1669 induction of melanoma-reactive CTL with peptides
107 Baselga J, Albanell J (2001) Mechanism of action of from the melanoma antigen gp100 modified at HLA-
anti-HER2 monoclonal antibodies. Ann Oncol 12 A*0201-binding residues. J Immunol 157: 2539–2548
(Suppl 1): S35–S41 118 Schuler-Thurner B, Schultz ES, Berger TG, Weinlich G,
108 Berzofsky JA, Terabe M, Oh S, Belyakov IM, Ahlers JD, Ebner S, Woerl P, Bender A, Feuerstein B, Fritsch PO,
Janik JE, Morris JC (2004) Progress on new vaccine Romani N et al (2002) Rapid induction of tumor-spe-
strategies for the immunotherapy and prevention of cific type 1 T helper cells in metastatic melanoma
cancer. J Clin Invest 113: 1515–1525 patients by vaccination with mature, cryopreserved,
109 Wallack MK, Sivanandham M, Balch CM, Urist MM, peptide-loaded monocyte-derived dendritic cells. J
Bland KI, Murray D, Robinson WA, Flaherty L, Richards Exp Med 195: 1279–1288
JM,Bartolucci AA et al (1998) Surgical adjuvant active 119 Figdor CG, de Vries IJ, Lesterhuis WJ, Melief CJ (2004)
specific immunotherapy for patients with stage III Dendritic cell immunotherapy: mapping the way. Nat
melanoma: the final analysis of data from a phase III, Med 10: 475–480
randomized, double-blind, multicenter vaccinia 120 Dranoff G (2003) GM-CSF-secreting melanoma vac-
melanoma oncolysate trial. J Am Coll Surg 187: 69–77 cines. Oncogene 22: 3188–3192
110 Romero P, Cerottini JC, Speiser DE (2004) Monitoring 121 Soiffer R, Lynch T, Mihm M, Jung K, Rhuda C,
tumor antigen specific T-cell responses in cancer Schmollinger JC, Hodi FS, Liebster L, Lam P, Mentzer S
patients and phase I clinical trials of peptide-based et al (1998) Vaccination with irradiated autologous
vaccination. Cancer Immunol Immunother 53: 249–255 melanoma cells engineered to secrete human granu-
111 Scheibenbogen C, Letsch A, Schmittel A, Asemissen locyte-macrophage colony-stimulating factor gener-
AM,Thiel E, Keilholz U (2003) Rational peptide-based ates potent antitumor immunity in patients with
tumour vaccine development and T cell monitoring. metastatic melanoma. Proc Natl Acad Sci USA 95:
Semin Cancer Biol 13: 423–429 13141–13146
112 Disis ML, Cheever MA (1996) Oncogenic proteins as 122 Antonia SJ, Seigne J, Diaz J, Muro-Cacho C, Extermann
tumor antigens. Curr Opin Immunol 8: 637–642 M, Farmelo MJ, Friberg M, Alsarraj M, Mahany JJ, Pow-
113 Kim EM, Sivanandham M, Stavropoulos CI, Bartolucci Sang J et al (2002) Phase I trial of a B7-1 (CD80) gene
AA,Wallack MK (2001) Overview analysis of adjuvant modified autologous tumor cell vaccine in combina-
therapies for melanoma – a special reference to tion with systemic interleukin-2 in patients with
results from vaccinia melanoma oncolysate adjuvant metastatic renal cell carcinoma. J Urol 167: 1995–2000
therapy trials. Surg Oncol 10: 53–59 123 Grimm EA, Mazumder A, Zhang HZ, Rosenberg SA
114 Nabel GJ, Nabel EG,Yang ZY, Fox BA, Plautz GE, Gao X, (1982) Lymphokine-activated killer cell phenomenon.
148 Cancer immunity

Lysis of natural killer-resistant fresh solid tumor cells CH et al (1995) Regression of advanced ovarian carci-
by interleukin 2 activated autologous human periph- noma by intraperitoneal treatment with autologous T
eral blood lymphocytes. J Exp Med 155: 1823–1841 lymphocytes retargeted by a bispecific monoclonal
124 Kruit WH, Goey SH, Lamers CH, Gratama JW,Visser B, antibody. J Natl Cancer Inst 87: 1463–1469
Schmitz PI, Eggermont AM, Bolhuis RL, Stoter G (1997) 131 Lamers CH, Willemsen RA, Luider BA, Debets R, Bol-
High-dose regimen of interleukin-2 and interferon- huis RL (2002) Protocol for gene transduction and
alpha in combination with lymphokine-activated expansion of human T lymphocytes for clinical
killer cells in patients with metastatic renal cell can- immuno-gene therapy of cancer. Cancer Gene Ther 9:
cer. J Immunother 20: 312–320 613–623
125 Rosenberg SA, Lotze MT, Yang JC, Topalian SL, Chang 132 Lamers CHJ, Sleijfer S, Willemsen RA, Debets R, Kruit
AE, Schwartzentruber DJ, Aebersold P, Leitman S, Line- WHJ, Gratama JW, Stoter G (2004) Adoptive immuno-
han WM, Seipp CA et al (1993) Prospective random- gene therapy of cancer with single chain antibody
ized trial of high-dose interleukin-2 alone or in con- [scFv(Ig)] gene modified T lymphocytes. J Biol Reg
junction with lymphokine-activated killer cells for the Homeost Agents 18: 134–140
treatment of patients with advanced cancer. J Natl 133 Slavin S, Morecki S,Weiss L, Shapira MY, Resnick I, Or R
Cancer Inst 85: 622-632 (2004) Nonmyeloablative stem cell transplantation:
126 Law TM, Motzer RJ, Mazumdar M, Sell KW, Walther PJ, reduced-intensity conditioning for cancer immuno-
O’Connell M, Khan A, Vlamis V, Vogelzang NJ, Bajorin therapy – from bench to patient bedside. Semin Oncol
DF (1995) Phase III randomized trial of interleukin-2 31: 4–21
with or without lymphokine-activated killer cells in 134 Childs R, Chernoff A, Contentin N, Bahceci E, Schrump
the treatment of patients with advanced renal cell car- D,Leitman S,Read EJ,Tisdale J,Dunbar C,Linehan WM
cinoma. Cancer 76: 824–832 et al (2000) Regression of metastatic renal cell carci-
127 Walter EA, Greenberg PD, Gilbert MJ, Finch RJ, Watan- noma after nonmyeloablative allogeneic peripheral-
abe KS, Thomas ED, Riddell SR (1995) Reconstitution blood stem-cell transplantation. N Engl J Med 343:
of cellular immunity against cytomegalovirus in recip- 750–758
ients of allogeneic bone marrow by transfer of T-cell 135 Hentschke P,Barkholt L,Uzunel M,Mattsson J,Wersall P,
clones from the donor. N Engl J Med 333: 1038–1044 Pisa P, Martola J,Albiin N,Wernerson A, Soderberg M et
128 Yee C,Thompson JA, Byrd D, Riddell SR, Roche P, Celis al (2003) Low-intensity conditioning and hematopoi-
E, Greenberg PD (2002) Adoptive T cell therapy using etic stem cell transplantation in patients with renal
antigen-specific CD8+ T cell clones for the treatment of and colon carcinoma. Bone Marrow Transplant 31:
patients with metastatic melanoma: in vivo persist- 253–261
ence, migration, and antitumor effect of transferred T 136 Childs RW, Barrett J (2004) Nonmyeloablative allo-
cells. Proc Natl Acad Sci USA 99: 16168–16173 geneic immunotherapy for solid tumors. Annu Rev
129 Rooney CM, Smith CA, Ng CY, Loftin SK, Sixbey JW, Gan Med 55: 459–475
Y, Srivastava DK, Bowman LC, Krance RA, Brenner MK 137 Hacein-Bey-Abina S, Von Kalle C, Schmidt M, McCor-
et al (1998) Infusion of cytotoxic T cells for the preven- mack MP, Wulffraat N, Leboulch P, Lim A, Osborne CS,
tion and treatment of Epstein-Barr virus-induced lym- Pawliuk R, Morillon E et al (2003) LMO2-associated
phoma in allogeneic transplant recipients. Blood 92: clonal T cell proliferation in two patients after gene
1549–1555 therapy for SCID-X1. Science 302: 415–419
130 Canevari S, Stoter G, Arienti F, Bolis G, Colnaghi MI, Di
Re EM, Eggermont AM, Goey SH, Gratama JW, Lamers

También podría gustarte