Anda di halaman 1dari 13

Advanced Drug Delivery Reviews 56 (2004) 1219 – 1231

www.elsevier.com/locate/addr

Folate-mediated targeting of T cells to tumors


Edward J. Roy a, Ute Gawlick a, Brent A. Orr a, David M. Kranz b,*
a
Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
b
Department of Biochemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, IL 61801, USA
Received 30 August 2003; accepted 5 January 2004

Abstract

Recently various strategies have been developed to exploit in a clinical setting the well established finding that T cells can
specifically recognize and destroy tumor cells. Several independent approaches to the targeting of T cells against cancer have
been explored, including the use of bispecific antibodies (anti-T cell/anti-tumor cell) to redirect T cells, vaccines to induce
tumor-reactive T cells, and adoptive transfer of ex vivo activated, tumor-reactive T cells. In this review, we focus on studies in
which high-affinity folate receptors (FRs) on tumor cells have served as targets for redirecting or enhancing the effectiveness of
activated T cells. Bispecific antibody conjugates of folate and antibodies to the T cell receptor (TCR) complex can generate
tumor-reactive T cell responses. The development of folate/antibody conjugates specific for the T cell co-stimulatory molecule
CD28 could yield activated T cells that recognize endogenous peptide – major histocompatibility complex (MHC) antigens on
tumor cells. Finally, we discuss a less investigated area in which high-affinity FRs on macrophages, or other antigen presenting
cells (APCs), may provide opportunities in the design of tumor-antigen-specific vaccines.
D 2004 Elsevier B.V. All rights reserved.

Keywords: Immunotherapy; T cell responses; Folate/antibody conjugates; Antigen presenting cells; Macrophages

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1220
2. T Cell immunotherapy of cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1220
2.1. Bispecific antibodies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1221
2.2. T cell vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1222
2.3. Adoptive T cell therapies. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1222
3. Folate-mediated targeting of T cells to cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1222
3.1. Tumor models. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1223
3.2. Bispecific folate/antibody conjugates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1223
3.3. Optimal T cell targeting: cytokine co-treatments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1225
4. Potential strategies for eliciting effector and memory T cells against tumors . . . . . . . . . . . . . . . . . . . . . . . . 1225

* Corresponding author. Tel.: +1-217-244-2821; fax: +1-217-244-5858.


E-mail address: d-kranz@uiuc.edu (D.M. Kranz).

0169-409X/$ - see front matter D 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2004.01.006
1220 E.J. Roy et al. / Advanced Drug Delivery Reviews 56 (2004) 1219–1231

4.1. Co-stimulatory molecules . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1225


4.2. Antigen presenting cells: folate-mediated uptake . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1226
5. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1228
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1228
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1228

1. Introduction cells against cancers. While it is beyond the scope of


the present review to cover these in detail, an over-
Early studies of Gorer showed that mice could view of the general approaches is presented in Section
reject very large tumor burdens that are not histocom- 2. In Section 3, we summarize how the high-affinity
patible [1], although the nature of the immune re- folate receptors (FRs) found on some tumor cells have
sponse had not yet been uncovered. In hindsight, these been used to specifically focus the activity of T cells
findings revealed the tremendous potential of T cells against the tumors. These studies include the genera-
in the elimination of cancers. The molecular basis of tion of folate/antibody conjugates against epitopes of
the T cell recognition process began to be elucidated the TCR/CD3 complex. Tumor models that have been
by many investigators, notably with the discoveries by developed to study these effects include syngeneic
Zinkernagel and Doherty [2] that T cells recognize a tumors in immunocompetent mice, FR + human
foreign antigen only when it is associated with a self tumors transplanted into immunodeficient mice, and
protein encoded by the major histocompatibility com- FR+ choroid plexus tumors that arise endogenously in
plex (MHC). Subsequent work by Boon and others SV40 transgenic mice. As with many clinical
showed that specific tumor-antigen peptides, bound to approaches to generating T cell anti-tumor responses,
a product of the MHC, could be isolated and shown to the use of cytokines, especially IL-2 and IL-12, to
stimulate cytotoxic T lymphocyte (CTL) activity optimize T cell activity have been tested. Finally, in
[3,4]. There are now many peptides that have been Section 4 we describe two strategies that are aimed at
associated with different types of cancers, and that eliciting longer term immune responses against can-
have formed the basis for specific tumor vaccines in cer, both involving folate-mediated induction of ef-
pre-clinical and clinical trials [5]. fector and/or memory T cells that are specific for
The cell surface molecules involved in T cell tumor antigens.
immunity have been the topics of study by many
laboratories [6], yielding valuable information about
the basic mechanisms of T cell biology and guiding 2. T Cell immunotherapy of cancer
the development of many strategies for eliciting T
cell activity against tumors. Among the key T cell The normal interactions between a naı̈ve T cell
molecules are the ah T cell receptor (TCR) that and an APC that lead to T cell activation are shown
recognizes the peptide/MHC antigen and that asso- in Fig. 1A APCs take up foreign proteins or tumor
ciates with subunits of the signaling complex called cells and display processed peptides bound to a
CD3 (Fig. 1). Co-stimulatory T cell molecules such protein of the MHC. The heterodimeric ah TCRs
as CD28 are essential in driving T cells through the on a small fraction of T cells are capable of binding
proliferative and differentiation pathways required to these specific peptide/MHC products. Ligation of
for effective targeting. The ligands for the co-stim- the TCR/CD3 complex in addition to ligation of co-
ulatory molecules, and the peptide/MHC antigens stimulatory molecules such as CD28 with its ligand
themselves, are normally presented to the T cells B7 leads to T cell activation and proliferation [8]. T
by ‘‘professional’’ antigen presenting cells (APCs) helper cells that express the class II MHC-specific
[7]. APCs include B cells, macrophages, and, in accessory molecule CD4 and CTLs that express the
particular, dendritic cells. class I MHC-specific accessory molecule CD8 are
The field of T cell-based therapies has expanded activated by the corresponding peptide/MHC com-
over the past 20 years to include a variety of strategies plexes. Our discussion here will focus largely on
aimed at stimulating and redirecting the activity of T CTLs, as most folate-mediated studies have been
E.J. Roy et al. / Advanced Drug Delivery Reviews 56 (2004) 1219–1231 1221

Fig. 1. Activation and effector phases of CTL function. (A) A naı̈ve T cell is activated by two signals; one through the TCR/CD3 complex and
one through the co-stimulatory molecule CD28. The naı̈ve T cell expresses an ah TCR that, in association with the accessory molecule CD8,
has sufficient affinity for a specific peptide MHC complex (pMHC2) that is expressed by an APC. CD28 binds to its ligands, members of the B7
family, on the APC. (B) The activated CTL from panel A is capable of recognizing and lysing a tumor cell that expresses pMHC2, without
participation of CD28.

conducted with CTLs. Once activated, CTLs can 2.1. Bispecific antibodies
migrate to the site of a tumor where they recognize
and lyse tumor cells that express the antigenic Initial studies showed that covalent attachment of
peptide/MHC (Fig. 1B). Recognition of peptide/ an anti-TCR antibody to the surface of a tumor cell
MHC by CTLs is facilitated by the action of CD8 that lacked the appropriate pep/MHC allowed the
that enhances binding to the class I product and tumor cell to be lysed [11]. These studies soon led to
recruits other signaling molecules such as the kinase the development of bispecific anti-TCR/anti-tumor
p56lck [9,10]. Three general approaches to stimulate cell antibodies that also redirected the activity of
and/or redirect the activity of T cells are discussed activated CTLs against the tumor [12,13]. In the
below. decade following, many different tumor antigens
1222 E.J. Roy et al. / Advanced Drug Delivery Reviews 56 (2004) 1219–1231

were examined in studies with bispecific antibodies set of factors that are involved in the potential success
directed against the ah TCR or the CD3 subunits of T cell vaccine approaches include the existence of
that form the TCR/CD3 complex [14,15]. The mech- an adequate T cell repertoire in a patient (e.g. immu-
anisms for activating T cells typically involved either nosuppression could exist due to previous therapies,
the use of activating anti-CD3 antibodies, or alter- factors released by the tumor, or the age of the
natively, the use of bacterial superantigens such as patient), the nature of the antigen (e.g. defined pep-
Staphyloccocal enterotoxin B that stimulate large tides, proteins, whole tumor cell preparations, or
fractions of T cells [16]. Antibodies to CD3 allowed mRNA), the optimal state of dendritic cell maturation
recruitment of all T cells, whereas antibodies specific (where dendritic cells are used ex vivo), the route of
for particular variable regions of the TCR would administration, and delivery schedule. Considerable
recruit only subpopulations of T cells that bear that pre-clinical and clinical work will be needed to begin
variable region. to resolve these issues.
In principle, activating agents must in some way
allow the engagement of the co-stimulatory molecule 2.3. Adoptive T cell therapies
CD28 by its ligands of the B7 family (e.g. through
binding of the Fc region of anti-CD3 antibodies to Fc Two recent reports on the treatment of melanoma
receptors (FcR) on APCs or binding of superantigens patients with ex vivo activated T cells have encour-
to class II MHC on APCs). Efforts to mimic these aged additional efforts towards adoptive T cell thera-
interactions led to the characterization of bispecific pies [23,24]. A disadvantage of adoptive therapy is
antibody conjugates that contain an anti-CD28 anti- the tedious and time-consuming need for culturing of
body and an anti-tumor cell antibody [17,18]. This each patient’s T cells. A further issue is whether the T
approach seeks to convert a tumor cell into an APC cell repertoire of a patient has been ‘tolerized’ against
thereby allowing the tumor to act as both the initiator tumor antigens to the extent that T cells with adequate
and the target of T cell responses. TCR avidity and activity can not be expanded in vitro
[25 –27]. A possible strategy under these circumstan-
2.2. T cell vaccines ces is to combine adoptive T cell therapy with gene
therapy using TCRs isolated and/or evolved in vitro
There are currently many clinical trials that involve for specificity or higher affinity against a peptide
the use of antigenic peptides, tumor cells, or tumor antigen [10,28 – 30]. This approach has been applied
mRNA to stimulate T cell responses [19 – 21]. These with some success in animal models, using TCR
vaccines are designed to elicit internalization and genes isolated from viral or tumor-specific T cells
subsequent presentation of the administered tumor [31,32].
antigens by professional APCs (especially dendritic An alternative approach to the use of TCR genes
cells, the most potent APCs in the immune system) has been to express a chimeric gene fusion that
and thereby to generate effector and memory T cells encodes an antibody-binding region (VH – VL) and a
against tumors. Many studies have begun to elucidate T cell signaling region such as the CD3 ~ subunit
the numerous factors involved in T cell vaccines, as [33 –35]. While such T cells (called ‘‘T-bodies’’) have
reviewed most recently by Finn [21]. Attempts to been engineered with a number of different antibodies
exploit folate-mediated T cell activity will be depen- to demonstrate redirected activity of T cells, it remains
dent on the knowledge that is gained in these areas. to be seen if the approach yields various other T cell
For example, some efforts now include the ex vivo functions in vivo (e.g. T cell migration, persistence,
isolation and activation of dendritic cells that can and memory) that are important in tumor targeting.
process and present tumor peptides along with co-
stimulatory ligands for optimal T cell activation.
Nussenzweig, Steinman and colleagues have explored 3. Folate-mediated targeting of T cells to cancer
strategies to enhance the uptake of antigen using
antigen/antibody conjugates that bind to dendritic cell The purpose of this review is to focus on those
surface molecules such as DEC205 [22]. The complex studies that have involved targeting of T cells against
E.J. Roy et al. / Advanced Drug Delivery Reviews 56 (2004) 1219–1231 1223

tumors that express the high-affinity FR. The expres- the FR have been described and could in principle
sion of FR on tissues and tumors [36 – 39] is the topic also be used as syngeneic models of T cell targeting
of other articles in this issue, and will thus not be [45,46].
discussed further here. We have used an endogenously arising FR+ tumor
model that involves the SV40 large T antigen trans-
3.1. Tumor models genic line of mice called SV11 [47,48]. These mice
develop endogenous brain tumors and ultimately
Several different tumor models have been used to succumb to the tumor around 100 days of life. Tumors
test the efficacy of T cell targeting to FR+ tumors. derived from the choroid plexus express very high
Animal models of bispecific antibody targeting levels of the FR [49], similar to human choroid plexus
against human tumors have typically relied on the carcinomas and ependymomas. MRI can be used to
transplantation of the human tumor lines into immu- monitor the status of tumors, thus providing a non-
nodeficient SCID or nude mice [40,41]. In these invasive method for comparing various targeting
cases, in vitro activated human T cells are also regimens. Because tumors arise endogenously, SV11
adoptively transferred to provide a source of effector mice represent the scenario encountered clinically, in
cells. We have described a murine model that allows which an individual’s T cells are tolerant of the tumor
activation of an endogenous T cell population and [50]. Studies of adoptively transferred T cells, derived
transplantation of human tumors, an approach that is from syngeneic C57Bl/6 mice that are not transgenic
desirable in translation of therapies to human cancer. for SV40 large T antigen, showed the potential of T
In our model, the human FR+ tumor line KB was cell therapies, as the lifespan of treated SV11 mice
transplanted into 2C TCR/RAG / mice that bear a could be more than doubled [51]. SV11 mice are
single monoclonal population of T cells [42]. 2C tolerized against the SV40 large T antigen, and hence
TCR/RAG / mice express TCR transgenes for a they may lack the T cell repertoire capable of eliciting
and h chains from a mouse CTL clone called 2C and strong anti-SV40 responses. However, SV11 mice
knockout of the recombination-activation gene-1 that may contain T cells with TCRs that have lower
is required for gene rearrangement in B and T cells affinities for particular SV40 peptide epitopes and
[43]. The CD8+ monoclonal T cell population can be these T cells may be activated against the tumor upon
activated in vivo with the agonist peptide recognized sufficient stimulation [51].
by CTL 2C called SIYR, but their TCR does not
recognize human tumor cells. However, the 2C TCR/ 3.2. Bispecific folate/antibody conjugates
RAG / model is limited by the absence of a number
of cell types (B cells, CD4+ helper T cells, and CD4+ Among the first tumor antigens targeted with the
regulatory T cells) that are known to affect the bispecific antibody approach was an ovarian carcino-
immune response against tumors. ma antigen recognized by two monoclonal antibodies
A syngeneic model of FR+ tumors has involved the called Mov18 and Mov19 [36,40,52,53]. The tumor
BALB/c tumor M109, a spontaneous lung carcinoma antigen recognized by these antibodies was a folate
[44]. In this tumor model, an indirect approach was binding protein now known to bind folate with
used to target tumor cells, involving the induction of relatively high-affinity (KD f 1 nM). Mezzanzanica
an anti-hapten antibody response in BALB/c mice, et al. showed that Mov18 specific antigen was stable
followed by transplantation of M109, and administra- on the cell surface and was present for a sufficient
tion of folate/hapten conjugates. While the rejection of time to allow for redirected lysis of ovarian carcino-
M109 required the presence of anti-hapten antibodies, mas by T cells [40]. Several clinical trials for patients
it is not clear if T cells were required as a component with ovarian carcinoma were only marginally success-
of the effector response. Nevertheless, this syngeneic ful, limited at least in part by the induction of human
tumor model could be used to evaluate the various anti-mouse antibody (HAMA) responses [54]. More
folate-mediated targeting strategies that require im- recent pre-clinical studies have used chimeric versions
munocompetent mice, as described below. Other of the bispecific antibody in an attempt to avoid
BALB/c tumor lines that express various levels of HAMA responses [55].
1224 E.J. Roy et al. / Advanced Drug Delivery Reviews 56 (2004) 1219–1231

As an alternative to the bispecific antibody ap- CD3 antibodies were shown to retarget the lysis of FR+
proach, we explored the use of bispecific folate con- tumor cell lines (Fig. 2A) [46]. Furthermore, these
jugates in which folate was coupled to antibodies conjugates were capable of modest improvements in
specific for TCRs. Folate/antibody conjugates offer survival when SV11 mice were treated with the super-
the advantages of having a smaller size compared to antigen followed by folate/anti-CD3 IgG [57].
traditional bispecific antibodies, a factor that has been Subsequent studies showed that the smallest bis-
correlated with increased tumor penetration [56]. The pecific antibody yet produced, folate-coupled to a
folate/antibody conjugate also should exhibit a de- single-chain Fv antibody ( f 25 kDa), was also very
creased probability of generating anti-Ig immune efficient in mediating T cell lysis of FR+ tumor cells
responses. Conjugates of folate and anti-TCR or anti- [58]. Folate/scFv and folate/Fab conjugates were

Fig. 2. Folate-mediated activities of CTLs. (A) A conjugate of folate and an anti-TCR/CD3 antibody can mediate the lysis of FR+ tumor cells by
an activated CTL. (B) In principle, a conjugate of folate and an anti-CD28 antibody could convert a FR+ tumor cell into an ‘APC’. In this
scenario, the CTL is activated by engagement of the TCR/CD3 complex with a specific peptide-MHC tumor antigen and by engagement of the
CD28 with the folate/anti-CD28 antibody bound to FR on the tumor cell surface.
E.J. Roy et al. / Advanced Drug Delivery Reviews 56 (2004) 1219–1231 1225

equally effective in promoting tumor rejection in the and therefore may allow more sustained T cell activity
KB tumor model in TCR/RAG mice [42]. This study [63,64]. It is too early to tell whether a standardized
also showed the importance of pre-activating the and perhaps alternating regimen of IL2 and/or IL-15
endogenous T cell population prior to treatment with could improve T cell therapeutic approaches. IL-2 and
folate conjugates. The advantage of smaller conju- a-IFN have been used in combination with folate
gates stems from their ability to penetrate into solid conjugated haptens to promote anti-tumor immune
tumors, but their most notable disadvantage is a very responses mediated by antibody-dependent cellular
short serum lifetime (minutes). While intact IgG anti- cytotoxicity (ADCC) [44]. To the best of our knowl-
bodies have considerably longer serum lifetimes edge, IL-15 has not been tested with folate-mediated
(days), the Fc region can invoke FcR mediated effects approaches.
involving excessive cytokine release, as was original- IL-12 has also received considerable attention as a
ly observed with the anti-CD3 antibody OKT3 in possible adjuvant for tumor vaccines since it acts as a
humans. These side-effects can be minimized by using T cell potentiating agent [65,66]. IL-12 is secreted by
a mutated Fc region that has reduced binding to the APCs and TH cells and it acts on CD8+ T cells to
FcR [59]. generate longer term effector cells and possibly,
The issues that remain to be resolved for this memory T cells [65,67]. Studies with SV11 mice have
approach hold true for all bispecific antibody shown that IL-12 alone, without folate/antibody con-
approaches, including those that have been tested jugates, induces extensive T cell infiltration into
clinically. Most importantly, the success will depend choroids plexus tumors [68]. Unfortunately, attempts
on the degree to which a sustained and robust T cell to combine IL-12 treatments with bispecific folate/
response can be achieved (see below). Their poten- anti-TCR antibody to boost the strength of the T cell
tial will also depend on the level of FR expressed response have led to lethal effects (see below). IL-12
on tumors and the extent to which FR-negative treatment of cancer patients in a phase II trial unex-
tumor cells can thrive and thereby escape redirected pectedly had severe toxicity [69], but it is not clear if
T cell activity. An early report suggested that the FR the lethality that we observed in mice is related
may enhance tumor growth and therefore may be mechanistically [70]. Nevertheless, studies described
associated with selective pressure to maintain the below indicate that IL-12 could be a very important
target [60]. component of T cell therapy that uses FR-targeted co-
stimulation, with minimal toxicity.
3.3. Optimal T cell targeting: cytokine co-treatments

In the past decade it has become clear that various 4. Potential strategies for eliciting effector and
cytokines play key roles in the induction and mainte- memory T cells against tumors
nance of T cells. It is still unclear how these cytokines
can best be used clinically to generate a powerful T 4.1. Co-stimulatory molecules
cell response against tumors. Perhaps the most rele-
vant cytokines for purposes of a CD8+ T cell response The approaches with bispecific antibodies de-
are IL-2, IL-12, and IL-15. IL-2 has long been known scribed above rely on the FR as the target for cross-
to be critical factor for T cell proliferation and it has linking the TCR/CD3 complex on the T cell. In order
been used clinically in many immunotherapeutic regi- to induce long-term immunity against a tumor, it
mens [61,62]. Early studies showed that IL-2 at high would be useful to generate T cells that recognize
doses caused significant side-effects but the most tumor-associated pepMHC complexes. Self-reactive,
recent trials with adoptive transferred T cells have low-avidity T cells that have escaped clonal deletion
used IL-2 at lower doses to help alleviate the adverse during thymic education can become CTL effector
responses [62]. It has been suggested that IL-2 may cells upon sufficient stimulation [71], and peripherally
not be optimal in that it promotes activation-induced anergized CTLs can be recovered by repetitive in vitro
cell death of self-reactive CD8+ T cells, whereas the stimulation [72]. While the activation of self-reactive
cytokine IL-15 can prevent apoptosis of CD8+ T cells CTLs has been demonstrated, it appears that T cell
1226 E.J. Roy et al. / Advanced Drug Delivery Reviews 56 (2004) 1219–1231

activation is often transient and anergy prevails upon 4.2. Antigen presenting cells: folate-mediated uptake
subsequent stimulation [73]. As indicated, the activa-
tion of naı̈ve T cells is highly dependent on signaling As indicated above, during the course of our
through the CD28/B7 co-stimulatory pathway in ad- studies with IL-12 and folate/anti-TCR antibody con-
dition to the TCR/pepMHC interaction. Because tu- jugates we observed that animals treated with a
mor cells typically lack B7 molecules, optimal combination of these agents were highly susceptible
stimulation requires processing of tumor antigens to lethal reactions that occurred within 48 h of treat-
through APCs. Stimulation of the TCR without ment. The toxicity was observed with both SV11 and
CD28 can in some cases lead to peripheral T cell normal C57Bl/6 mice, indicating that the reaction was
anergy against the peptide-MHC antigen (i.e. on the independent of FR+ tumors. Although the basis of this
tumor). toxicity has not been further resolved, we suspect that
The crucial role of CD28 in the activation of low- a toxic-shock like syndrome may be responsible. In
avidity potentially tumor-reactive T cells has made it a this scenario, the folate/anti-TCR conjugates might
target for eliciting an anti-tumor T cell response. In bind to FR+ macrophages that have been activated
this context, various strategies have emerged: (1) the through an IL-12-dependent mechanism (e.g. IL-12
development of bispecific antibody conjugates which induces g-IFN, which is known to induce FRs on
simultaneously crosslink CD28 on T cells to tumor macrophages [79]). IL-12 may also act through the
antigens (Fig. 2B), (2) transfection of tumor cells with synergistic activation of T cells. The observed effect
the B7 gene [74], and (3) transfection of T cells with a may be analogous to that of bacterial superantigens,
secreted bispecific protein composed of B7 and an which induce toxic-shock by binding to class II MHC
anti-tumor single-chain antibody [75]. The advantage expressed on APCs such as macrophages and to Vh
of the first approach is that it does not require ex vivo regions of the TCR on T cells. This cross-linking leads
manipulation the patient’s tumor or T cells. The to the massive release of T cell cytokines and subse-
objective of these strategies is to convert tumor cells quent organ failure.
into competent APCs that are capable of activating Several observations suggest that the high-affinity
anti-tumor T cell responses against tumor pepMHC FRs on activated macrophages (or dendritic cells, if
antigens. Under the optimal conditions, the approach they are expressed on these cells also) could provide
might also promote the generation of memory T cells a useful mechanism for the delivery of T cell-specific
that could target re-occurring cancers. antigens or whole tumor cells (Fig. 3): (1) Low and
Human clinical trials of ovarian cancer using his colleagues have previously shown that the high-
bispecific antibody to CD3 and FR showed an in- affinity FR could be exploited as a target for the
crease in tumor redirected lysis when combined with a endocytosis of folate-coupled macromolecules, in
bispecific antibody to CD28 and FR [76]. There is order to facilitate their uptake [80 –85]; (2) macro-
some evidence that long-term immunity is influenced phages are known to express the ligands (B7.1,
by the strength of the initial T cell response as induced B7.2) for co-stimulation of T cells and Nussenzweig,
by bispecific agents [77]. The induction of optimal Steinman and colleagues have shown that receptor-
memory cells may require both co-stimulatory signals mediated uptake of protein antigens by dendritic
and IL-12 or other cytokines. Under normal condi- cells may provide a strategy for creating optimized
tions, T cell activation and memory induction appear vaccines [22]; (3) activated macrophages express
to involve the action of IL-12 secreted by activated FRs [79] at levels that are even diagnostic for
APCs upon CD40/CD40L interaction, the so-called inflammatory reactions [86], and hence the FR may
third signal [78]. Our lab has investigated the role of be an ideal candidate for facilitating the endocytosis
CD28 co-stimulation against FR+ SV11 tumors by of T cell antigens by APCs; and (4) T cells reside in
testing various folate/anti-CD28 antibody conjugates regions where these cells might be able to present
alone and in combination with IL-12 (manuscript in antigen.
preparation). Again, the advantage of using folate as As pointed out by Lu and Low, it is possible that
the targeting agent is that it can be readily coupled to antibody-mediated uptake of tumor cells by activated
antibodies and tested in vitro and in vivo. macrophages may also have been involved in the
E.J. Roy et al. / Advanced Drug Delivery Reviews 56 (2004) 1219–1231 1227

Fig. 3. Possible strategy for generating vaccines using FR-mediated uptake of antigens. As macrophages express the high-affinity FR, it may be
possible to facilitate their uptake and processing of specific antigens to T cells by conjugation of known antigens or whole tumor cells to folate.
The enhanced levels of the specific peptide/MHC (pMHC2) could activate T cells that attack tumor cells expressing pMHC2.

effects observed in their study, using folate/hapten tion properties of FcR on dendritic cells and macro-
conjugate [44,87]. In principle, antibody-mediated phages. In contrast, it is unclear if folate-mediated
uptake of tumor cells by APCs could be more effec- uptake of antigens by macrophages will be as efficient
tive than folate-mediated uptake of antigens, as the as FcRs and it is not known if dendritic cells bear
former relies on well-known binding and internaliza- high-affinity FRs.
1228 E.J. Roy et al. / Advanced Drug Delivery Reviews 56 (2004) 1219–1231

5. Concluding remarks cells to tumors. The work was supported by grants


from the NIH.
The destruction of tumor cells by T cells is now
regarded as a desirable goal in the design of cancer
References
immunotherapeutics. There are several distinct
approaches toward T cell immunotherapies that are [1] P.A. Gorer, The genetic and antigenic basis of tumor trans-
in various stages of clinical and pre-clinical studies. plantation, J. Pathol. Bacteriol. 44 (1937) 691 – 697.
At this time it is not known what the optimal approach [2] R.M. Zinkernagel, P.C. Doherty, Immunological surveil-
will be and in fact it is quite possible that different lance against altered self components by sensitised T lym-
approaches will be required, depending on the type of phocytes in lymphocytic choriomeningitis, Nature 251
(1974) 547 – 548.
cancer and the patient. While it is still unclear whether [3] T. Boon, L.J. Old, Cancer tumor antigens, Curr. Opin. Immu-
T cell therapies that involve folate-mediated targeting nol. 9 (1997) 681 – 683.
will provide measurable advantages, the principles of [4] S.A. Rosenberg, A new era for cancer immunotherapy based
high-affinity interactions with FRs and the use of on the genes that encode cancer antigens, Immunity 10 (1999)
281 – 287.
small molecule (folate) as conjugates facilitates the
[5] S.A. Rosenberg, Progress in human tumour immunology and
exploration of their use in this platform. Accordingly, immunotherapy, Nature 411 (2001) 380 – 384.
folate conjugated to an anti-T cell antibody redirects [6] S.J. Davis, S. Ikemizu, E.J. Evans, L. Fugger, T.R. Bakker,
the activity of a T cell against a FR positive tumor. P.A. van der Merwe, The nature of molecular recognition by T
Whereas antibodies against the TCR complex can cells, Nat. Immunol. 4 (2003) 217 – 224.
directly mediate lysis of the tumor by an activated [7] P. Cresswell, A. Lanzavecchia, Antigen processing and recog-
nition, Curr. Opin. Immunol. 13 (2001) 11 – 12.
cytotoxic T cell, antibodies against co-stimulatory [8] D.J. Lenschow, T.L. Walunas, J.A. Bluestone, CD28/B7 sys-
molecules provide an opportunity to induce T cells tem of T cell costimulation, Ann. Rev. Immunol. 14 (1996)
with tumor specificity (i.e. tumor-specific pepMHC 233 – 258.
that are recognized by the TCR). T cells stimulated in [9] A. Arcaro, C. Gregoire, T.R. Bakker, L. Baldi, M. Jordan, L.
this manner, in combination with the appropriate Goffin, N. Boucheron, F. Wurm, P.A. van der Merwe, B.
Malissen, I.F. Luescher, CD8beta endows CD8 with efficient
cytokine regimen, could develop into longer-term coreceptor function by coupling T cell receptor/CD3 to raft-
memory cells that provide greater protection against associated CD8/p56(lck) complexes, J. Exp. Med. 194 (2001)
the cancer. 1485 – 1495.
In another approach, FRs expressed on activated [10] P.D. Holler, D.M. Kranz, Quantitative analysis of the contri-
bution of TCR/pepMHC affinity and CD8 to T cell activation,
macrophages could be targeted with folate/antigen
Immunity 18 (2003) 255 – 264.
complexes. Activated macrophages serve as APCs, [11] D.M. Kranz, S. Tonegawa, H.N. Eisen, Attachment of an anti-
with appropriate co-stimulatory ligands for the acti- receptor antibody to nontarget cells renders them susceptible
vation of resting T cells. Hence, it may be possible to to lysis by a clone of cytotoxic T lymphocytes, Proc. Natl.
enhance an inflammatory T cell reaction against tumor Acad. Sci. USA 81 (1984) 7922 – 7926.
antigens, thereby stimulating a potent anti-tumor re- [12] M.A. Liu, D.M. Kranz, J.T. Kurnick, L.S. Boyle, R. Levy,
H.N. Eisen, Heteroantibody duplexes target cells for lysis by
sponse. An advantage of this approach is that it does cytotoxic T lymphocytes, Proc. Natl. Acad. Sci. USA 82
not rely on the expression of high-affinity FRs by a (1985) 8648 – 8652.
tumor. While both of these approaches will require [13] U.D. Staerz, O. Kanagawa, M.J. Bevan, Hybrid antibodies can
significant additional pre-clinical work, the ability to target sites for attack by T cells, Nature 314 (1985) 628 – 631.
readily conjugate folate to proteins of interest will [14] M.W. Fanger, D.L. Drakeman, Bispecific antibodies, Drug
News Perspect. 8 (1995) 133 – 137.
facilitate studies to explore their potential in the area [15] D.M. Segal, G.J. Weiner, L.M. Weiner, Bispecific antibodies
of T cell therapeutics. in cancer therapy, Curr. Opin. Immunol. 11 (1999) 558 – 562.
[16] J.A. Bluestone, D.S. Bruce, K.A. Newell, Activation of T cells
in vivo using anti-CD3 and staphylococcal enterotoxins, Int. J.
Acknowledgements Cancer Suppl. 7 (1992) 39 – 41.
[17] B.J. Kroesen, A. Bakker, R.A. van Lier, H.T. The, L. de Leij,
Bispecific antibodymediated target cell-specific costimulation
We thank other members of the lab for their efforts of resting T cells via CD5 and CD28, Cancer Res. 55 (1995)
on projects involving folate-mediated targeting of T 4409 – 4415.
E.J. Roy et al. / Advanced Drug Delivery Reviews 56 (2004) 1219–1231 1229

[18] C. Renner, W.F. Jung, U. Sahin, R. Vanlier, M. Pfreundschuh, Melief, C. Huber, H.J. Stauss, M. Theobald, Circumventing
The role of lymphocyte subsets and adhesion molecules in T tolerance to a human MDM2-derived tumor antigen by
cell-dependent cytotoxicity mediated by CD3 and CD28 bis- TCR gene transfer, Nat. Immunol. 2 (2001) 962 – 970.
pecific monoclonal antibodies, Eur. J. Immunol. 25 (1995) [33] G. Gross, T. Waks, Z. Eshhar, Expression of immunoglobulin-
2027 – 2033. T-cell receptor chimeric molecules as functional receptors with
[19] P.G. Coulie, P. Bruggen, T-cell responses of vaccinated cancer antibody-type specificity, Proc. Natl. Acad. Sci. USA 86
patients, Curr. Opin. Immunol. 15 (2003) 131 – 137. (1989) 10024 – 10028.
[20] G. Schuler, B. Schuler-Thurner, R.M. Steinman, The use of [34] P. Hwu, J.C. Yang, R. Cowherd, J. Treisman, G.E. Shafer, Z.
dendritic cells in cancer immunotherapy, Curr. Opin. Immu- Eshhar, S.A. Rosenberg, In vivo antitumor activity of T cells
nol. 15 (2003) 138 – 147. redirected with chimeric antibody/T-cell receptor genes, Can-
[21] O.J. Finn, Cancer vaccines: between the idea and reality, Nat. cer Res. 55 (1995) 3369 – 3373.
Rev. Immunol. 3 (2003) 630 – 641. [35] J.H. Pinthus, T. Waks, K. Kaufman-Francis, D.G. Schindler,
[22] D. Hawiger, K. Inaba, Y. Dorsett, M. Guo, K. Mahnke, M. A. Harmelin, H. Kanety, J. Ramon, Z. Eshhar, Immuno-gene
Rivera, J.V. Ravetch, R.M. Steinman, M.C. Nussenzweig, therapy of established prostate tumors using chimeric recep-
Dendritic cells induce peripheral T cell unresponsiveness un- tor-redirected human lymphocytes, Cancer Res. 63 (2003)
der steady state conditions in vivo, J. Exp. Med. 194 (2001) 2470 – 2476.
769 – 779. [36] L.R. Coney, A. Tomassetti, L. Carayannopoulos, V. Frasca,
[23] M.E. Dudley, J.R. Wunderlich, P.F. Robbins, J.C. Yang, P. B.A. Kamen, M.I. Colnaghi, V.R. Zurawski Jr., Cloning of a
Hwu, D.J. Schwartzentruber, S.L. Topalian, R. Sherry, N.P. tumor-associated antigen: MOv18 and MOv19 antibodies rec-
Restifo, A.M. Hubicki, M.R. Robinson, M. Raffeld, P. ognize a folate-binding protein, Cancer Res. 51 (1991)
Duray, C.A. Seipp, L. Rogers-Freezer, K.E. Morton, S.A. 6125 – 6132.
Mavroukakis, D.E. White, S.A. Rosenberg, Cancer regres- [37] S.D. Weitman, R.H. Lark, L.R. Coney, D.W. Fort, V. Frasca,
sion and autoimmunity in patients after clonal repopulation V.R. Zurawski Jr., B.A. Kamen, Distribution of the folate
with antitumor lymphocytes, Science 298 (2002) 850 – 854. receptor GP38 in normal and malignant cell lines and tissues,
[24] C. Yee, J.A. Thompson, D. Byrd, S.R. Riddell, P. Roche, E. Cancer Res. 52 (1992) 3396 – 3401.
Celis, P.D. Greenberg, Adoptive T cell therapy using antigen- [38] J.F. Ross, P.K. Chaudhuri, M. Ratnam, Differential regulation
specific CD8+ T cell clones for the treatment of patients with of folate receptor isoforms in normal and malignant tissues in
metastatic melanoma: in vivo persistence, migration, and anti- vivo and in established cell lines. Physiologic and clinical
tumor effect of transferred T cells, Proc. Natl. Acad. Sci. USA implications, Cancer 73 (1994) 2432 – 2443.
99 (2002) 16168 – 16173. [39] A.C. Antony, Folate receptors, Annu. Rev. Nutr. 16 (1996)
[25] D.E. Speiser, R. Miranda, A. Zakarian, M.F. Bachmann, K. 501 – 521.
McKall-Faienza, B. Odermatt, D. Hanahan, R.M. Zinkernagel, [40] D. Mezzanzanica, M.A. Garrido, D.S. Neblock, P.E. Daddona,
P.S. Ohashi, Self antigens expressed by solid tumors do not S.M. Andrew, V.R. Zurawski Jr., D.M. Segal, J.R. Wunder-
efficiently stimulate naive or activated T cells: implications for lich, Human T-lymphocytes targeted against an established
immunotherapy, J. Exp. Med. 186 (1997) 645 – 653. human ovarian carcinoma with a bispecific F(abV)2 antibody
[26] M. Theobald, J. Biggs, J. Hernandez, J. Lustgarten, C. prolong host survival in a murine xenograft model, Cancer
Labadie, L.A. Sherman, Tolerance to p53 by A2.1-restricted Res. 51 (1991) 5716 – 5721.
cytotoxic T lymphocytes, J. Exp. Med. 185 (1997) 833 – 841. [41] O.C. Boerman, J.G. Tibben, L.F. Massuger, R.A. Claessens,
[27] L.A. Sherman, D.J. Morgan, C.T. Nugent, F.J. Hernandez, F.H. Corstens, Tumour targeting of the anti-ovarian carcino-
H.T. Kreuwel, A. Murtaza, A. Ko, J. Biggs, Self-tolerance ma  anti-CD3/TCR bispesific monoclonal antibody OC/TR
and the composition of T cell repertoire, Immunol. Res. 21 and its parental MOv18 antibody in experimental ovarian can-
(2000) 305 – 313. cer, Anticancer Res. 15 (1995) 2169 – 2174.
[28] H.W. Kessels, M.D. van Den Boom, H. Spits, E. Hooijberg, [42] L.A. Rund, B.K. Cho, T.C. Manning, P.D. Holler, E.J. Roy,
T.N. Schumacher, Changing T cell specificity by retroviral T D.M. Kranz, Bispecific agents target endogenous murine T
cell receptor display, Proc. Natl. Acad. Sci. USA 97 (2000) cells against human tumor xenografts, Int. J. Cancer 83
14578 – 14583. (1999) 141 – 149.
[29] P.D. Holler, P.O. Holman, E.V. Shusta, S. O’Herrin, K.D. [43] T.C. Manning, L.A. Rund, M.M. Gruber, F. Fallarino, T.F.
Wittrup, D.M. Kranz, In vitro evolution of a T cell recep- Gajewski, D.M. Kranz, Antigen recognition and allogeneic
tor with high affinity for peptide/MHC, Proc. Natl. Acad. tumor rejection in CD8+ TCR transgenic/RAG / mice, J.
Sci. USA 97 (2000) 5387 – 5392. Immunol. 159 (1997) 4665 – 4675.
[30] T.N. Schumacher, T-cell-receptor gene therapy, Nat. Rev. [44] Y. Lu, P.S. Low, Folate targeting of haptens to cancer cell
Immunol. 2 (2002) 512 – 519. surfaces mediates immunotherapy of syngeneic murine
[31] H.W. Kessels, M.C. Wolkers, M.D. van den Boom, M.A. van tumors, Cancer Immunol. Immunother. 51 (2002) 153 – 162.
der Valk, T.N. Schumacher, Immunotherapy through TCR [45] K.E. Brigle, M.J. Spinella, E.H. Westin, I.D. Goldman, In-
gene transfer, Nat. Immunol. 2 (2001) 957 – 961. creased expression and characterization of two distinct folate
[32] T. Stanislawski, R.H. Voss, C. Lotz, E. Sadovnikova, R.A. binding proteins in murine erythroleukemia cells, Biochem.
Willemsen, J. Kuball, T. Ruppert, R.L. Bolhuis, C.J. Pharmacol. 47 (1994) 337 – 345.
1230 E.J. Roy et al. / Advanced Drug Delivery Reviews 56 (2004) 1219–1231

[46] D.M. Kranz, T.A. Patrick, K.E. Brigle, M.J. Spinella, E.J. J.R. Thistlethwaite Jr., Phase I trial of a humanized, Fc recep-
Roy, Conjugates of folate and anti-T cell receptor antibod- tor nonbinding OKT3 antibody, huOKT3gamma1(Ala-Ala) in
ies specifically target folate-receptor-positive tumor cells for the treatment of acute renal allograft rejection, Transplantation
lysis, Proc. Natl. Acad. Sci. USA 92 (1995) 9057 – 9061. 68 (1999) 608 – 616.
[47] T. Van Dyke, C. Finlay, A.J. Levine, A comparison of several [60] F. Bottero, A. Tomassetti, S. Canevari, S. Miotti, S. Menard,
lines of transgenic mice containing the SV early genes, Cold M.I. Colnaghi, Gene transfection and expression of the ovar-
Spring Harbor Symp. Quant. Biol. 50 (1985) 671 – 678. ian carcinoma marker folate binding protein on NIH/3T3 cells
[48] T.A. Van Dyke, C. Finlay, D. Miller, J. Marks, G. Lozano, A.J. increases cell growth in vitro and in vivo, Cancer Res. 53
Levine, Relationship between simian virus 40 large tumor (1993) 5791 – 5796.
antigen expression and tumor formation in transgenic mice, [61] S.A. Rosenberg, Immunotherapy of cancer using interleukin
J. Virol. 61 (1987) 2029 – 2032. 2: current status and future prospects, Immunol. Today 9
[49] T.A. Patrick, D.M. Kranz, T.A. Van Dyke, E.J. Roy, Folate (1988) 58 – 62.
receptors as potential therapeutic targets in choroid plexus [62] W.W. Overwijk, M.R. Theoret, N.P. Restifo, The future of
tumors of SV40 transgenic mice, J. Neuro-Oncol. 32 (1997) interleukin-2: enhancing therapeutic anticancer vaccines, Can-
111 – 123. cer J. Sci. Am. 6 (Suppl. 1) (2000) S76 – 80.
[50] T.D. Schell, L.M. Mylin, I. Georgoff, A.K. Teresky, A.J. Lev- [63] T.A. Waldmann, S. Dubois, Y. Tagaya, Contrasting roles of
ine, S.S. Tevethia, Cytotoxic T-lymphocyte epitope immuno- IL-2 and IL-15 in the life and death of lymphocytes: implica-
dominance in the control of choroid plexus tumors in simian tions for immunotherapy, Immunity 14 (2001) 105 – 110.
virus 40 large T antigen transgenic mice, J. Virol. 73 (1999) [64] T.A. Waldmann, Immunotherapy: past, present and future,
5981 – 5993. Nat. Med. 9 (2003) 269 – 277.
[51] T.D. Schell, S.S. Tevethia, Control of advanced choroid plex- [65] G. Trinchieri, Interleukin-12 and the regulation of innate re-
us tumors in SV40 T antigen transgenic mice following prim- sistance and adaptive immunity, Nat. Rev. Immunol. 3 (2003)
ing of donor CD8(+) T lymphocytes by the endogenous tumor 133 – 146.
antigen, J. Immunol. 167 (2001) 6947 – 6956. [66] J.E. Portielje, J.W. Gratama, H.H. van Ojik, G. Stoter, W.H.
[52] S. Miotti, S. Canevari, S. Menard, D. Mezzanzanica, G. Porro, Kruit, IL-12: a promising adjuvant for cancer vaccination,
S.M. Pupa, M. Regazzoni, E. Tabliabue, M.I. Colnaghi, Char- Cancer Immunol. Immunother. 52 (2003) 133 – 144.
acterization of human ovarian carcinoma-associated antigens [67] R. Mortarini, A. Borri, G. Tragni, I. Bersani, C. Vegetti, E.
defined by novel monoclonal antibodies with tumor-restricted Bajetta, S. Pilotti, V. Cerundolo, A. Anichini, Peripheral burst
specificity, Int. J. Cancer 39 (1987) 297 – 303. of tumor-specific cytotoxic T lymphocytes and infiltration of
[53] S. Ferrini, A. Cambiaggi, C. Cantoni, S. Canevari, D. Mez- metastatic lesions by memory CD8+ T cells in melanoma
zanzanica, M.I. Colnaghi, L. Moretta, Targeting of T or NK patients receiving interleukin 12, Cancer Res. 60 (2000)
lymphocytes against tumor cells by bispecific monoclonal 3559 – 3568.
antibodies: role of different triggering molecules, Int. J. Can- [68] E.J. Roy, U. Gawlick, B.A. Orr, L.A. Rund, A.G. Webb, D.M.
cer 7 (1992) 15 – 18. Kranz, IL-12 treatment of endogenously arising murine brain
[54] C.H. Lamers, J.W. Gratama, S.O. Warnaar, G. Stoter, R.L. tumors, J. Immunol. 165 (2000) 7293 – 7299.
Bolhuis, Inhibition of bispecific monoclonal antibody (bsAb)- [69] J. Cohen, IL-12 deaths: explanation and a puzzle, Science 270
targeted cytolysis by human anti-mouse antibodies in ovarian (1995) 908.
carcinoma patients treated with bsAb-targeted activated T lym- [70] J.P. Leonard, M.L. Sherman, G.L. Fisher, L.J. Buchanan, G.
phocytes, Int. J. Cancer 60 (1995) 450 – 457. Larsen, M.B. Atkins, J.A. Sosman, J.P. Dutcher, N.J. Vogel-
[55] R.M. Luiten, S.O. Warnaar, D. Sanborn, C.H. Lamers, R.L. zang, J.L. Ryan, Effects of single-dose interleukin-12 expo-
Bolhuis, S.V. Litvinov, V.R. Zurawski Jr., L.R. Coney, Chime- sure on interleukin-12-associated toxicity and interferon-
ric bispecific OC/TR monoclonal antibody mediates lysis of gamma production, Blood 90 (1997) 2541 – 2548.
tumor cells expressing the folate-binding protein (MOv18) and [71] K.E. de Visser, T.A. Cordaro, H.W. Kessels, F.H. Tirion, T.N.
displays decreased immunogenicity in patients, J. Immunother. Schumacher, A.M. Kruisbeek, Low-avidity self-specific T
20 (1997) 496 – 504. cells display a pronounced expansion defect that can be over-
[56] T. Yokota, D.E. Milenic, M. Whitlow, J. Schlom, Rapid tumor come by altered peptide ligands, J. Immunol. 167 (2001)
penetration of a single chain Fv and comparison with other 3818 – 3828.
immunoglobulin forms, Cancer Res. 52 (1992) 3402 – 3408. [72] C. Ohlen, M. Kalos, D.J. Hong, A.C. Shur, P.D. Greenberg,
[57] T.A. Patrick, D.M. Kranz, J.F. Zachary, E.J. Roy, Intracerebral Expression of a tolerizing tumor antigen in peripheral tissue
bispecific ligand-antibody conjugate increases survival of ani- does not preclude recovery of high-affinity CD8+ T cells or
mals bearing endogenously arising brain tumors, Int. J. Cancer CTL immunotherapy of tumors expressing the antigen,
78 (1998) 470 – 479. J. Immunol. 166 (2001) 2863 – 2870.
[58] B.K. Cho, E.J. Roy, T.A. Patrick, D.M. Kranz, Single-chain [73] P.P. Lee, C. Yee, P.A. Savage, L. Fong, D. Brockstedt, J.S.
Fv/folate conjugates mediate efficient lysis of folate-receptor- Weber, D. Johnson, S. Swetter, J. Thompson, P.D. Greenberg,
positive tumor cells, Bioconjug. Chem. 8 (1997) 338 – 346. M. Roederer, M.M. Davis, Characterization of circulating T
[59] E.S. Woodle, D. Xu, R.A. Zivin, J. Auger, J. Charette, R. cells specific for tumor-associated antigens in melanoma
O’Laughlin, D. Peace, L.K. Jollife, T. Haverty, J.A. Bluestone, patients, Nat. Med. 5 (1999) 677 – 685.
E.J. Roy et al. / Advanced Drug Delivery Reviews 56 (2004) 1219–1231 1231

[74] S.E. Townsend, J.P. Allison, Tumor rejection after direct cos- [80] C.P. Leamon, P.S. Low, Delivery of macromolecules into liv-
timulation of CD8+ T cells by B7-transfected melanoma cells, ing cells: a method that exploits folate receptor endocytosis,
Science 259 (1993) 368 – 370. Proc. Natl. Acad. Sci. USA 88 (1991) 5572 – 5576.
[75] B. Blanco, P. Holliger, L. Alvarez-Vallina, Autocrine costimu- [81] C.P. Leamon, I. Pastan, P.S. Low, Cytotoxicity of folate-Pseu-
lation: tumor-specific CD28-mediated costimulation of T cells domonas exotoxin conjugates toward tumor cells. Contribu-
by in situ production of a bifunctional B7-anti-CEA diabody tion of translocation domain, J. Biol. Chem. 268 (1993)
fusion protein, Cancer Gene Ther. 9 (2002) 275 – 281. 24847 – 24854.
[76] A. Mazzoni, D. Mezzanzanica, G.R. Jung, H. Wolf, M.I. [82] C.P. Leamon, P.S. Low, Selective targeting of malignant cells
Colnaghi, H. Wolf, S. Canevari, CD3-CD28 costimulation with cytotoxin – folate conjugates, J. Drug Target. 2 (1994)
as a means to avoiding T cell preactivation in bispecific 101 – 112.
monoclonal antibody-based treatment of ovarian carcinoma, [83] R.J. Lee, P.S. Low, Delivery of liposomes into cultured KB
Cancer Res. 56 (1996) 5443 – 5449. cells via folate receptormediated endocytosis, J. Biol. Chem.
[77] L. Grosse-Hovest, M. Brandl, M. Dohlsten, T. Kalland, W. 269 (1994) 3198 – 3204.
Wilmanns, G. Jung, Tumor growth inhibition with bispe- [84] R.J. Lee, P.S. Low, Folate-mediated tumor cell targeting of
cific antibody fragments in a syngeneic mouse melanoma liposome-entrapped doxorubicin in vitro, Biochim. Biophys.
model: the role of targeted T-cell co-stimulation via CD28, Acta 1233 (1995) 134 – 144.
Int. J. Cancer 80 (1999) 138 – 144. [85] S. Wang, R.J. Lee, G. Cauchon, D.G. Gorenstein, P.S. Low,
[78] M. Cella, D. Scheidegger, K. Palmer-Lehmann, P. Lane, A. Delivery of antisense oligodeoxyribonucleotides against the
Lanzavecchia, G. Alber, Ligation of CD40 on dendritic human epidermal growth factor receptor into cultured KB
cells triggers production of high levels of interleukin-12 cells with liposomes conjugated to folate via polyethylene
and enhances T cell stimulatory capacity: T – T help via glycol, Proc. Natl. Acad. Sci. USA 92 (1995) 3312 – 3318.
APC activation, J. Exp. Med. 184 (1996) 747 – 752. [86] M.J. Turk, G.J. Breur, W.R. Widmer, C.M. Paulos, L.C. Xu,
[79] N. Nakashima-Matsushita, T. Homma, S. Yu, T. Matsuda, N. L.A. Grote, P.S. Low, Folate-targeted imaging of activated
Sunahara, T. Nakamura, M. Tsukano, M. Ratnam, T. Mat- macrophages in rats with adjuvant-induced arthritis, Arthritis
suyama, Selective expression of folate receptor beta and its Rheum. 46 (2002) 1947 – 1955.
possible role in methotrexate transport in synovial macrophages [87] Y. Lu, P.S. Low, Targeted immunotherapy of cancer: develop-
from patients with rheumatoid arthritis, Arthritis Rheum. 42 ment of antibody-induced cellular immunity, J. Pharm. Phar-
(1999) 1609 – 1616. macol. 55 (2003) 163 – 167.

Anda mungkin juga menyukai