You are on page 1of 23

Critical Reviews in Clinical Laboratory Sciences, 2009; 46(4): 167–189

REVIEW ARTICLE

Cancer immunotherapy
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

Constantin N. Baxevanis, Sonia A. Perez, and Michael Papamichail


Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
Referee: Professor Guido Forni, Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of
Torino, Torino, Italy

Abstract
Recent scientific advances have expanded our understanding of the immune system and its response to
malignant cells. The clinical goal of tumour immunotherapy is to provide either passive or active immu-
nity against malignancies by harnessing the immune system to target tumours. Monoclonal antibodies,
cytokines, cellular immunotherapy, and vaccines have increasingly become successful therapeutic agents
for the treatment of solid and haematological cancers in preclinical models, clinical trials, and practice. In
this article, we review recent advances in the immunotherapy of cancer, focusing on new strategies and
future perspectives as well as on clinical trials attempting to enhance the efficacy of immunotherapeutic
modalities and translate this knowledge into effective cancer therapies.
Keywords:  Adoptive transfer; cancer immunotherapy; cancer vaccines; chemotherapy; cytokines;
immunomodulation; monoclonal antibodies; NK cells; NKT cells; regulatory cells
For personal use only.

Abbreviations:  AICD, activation-induced cell death; AML, acute myeloid leukemia; BiTEs, bispecific
T-cell engager molecules; CAR, chimeric antigen receptor; CEA, carcinoembryonic antigen; CTL,
cytotoxic T lymphocyte; CTLA, cytotoxic T lymphocyte antigen; DC, dendritic cells; DTH, delayed-type
hypersensitivity; EpCAM, epithelial cell adhesion molecule; FLIP, flice inhibitory protein; GC, glucocorticoid;
GvHD, graft versus host disease; HC, hydrocortisone; HLA, human leucocyte antigen; IDO, indoleamine-
2,3-dioxygenase; IFN, interferon; IL, interleukin; KIR, killer cell immunoglobulin-like receptors; mAb,
monoclonal antibody; MHC, major histocompatibility complex; MUC1, mucin-1; NK, natural killer cell; NKT,
natural killer T cell; NSCLC, n
­ on-small-cell lung cancer; PADRE, pan-DR epitope; PD, programmed death;
TAA, tumour associated antigen; TCR, T cell receptor; TGF, transforming growth factor; Th, T helper cell; TLR,
toll-like receptor; TRAIL, tumour necrosis factor-inducing ligand; Tregs, T regulatory cells; VEGF, vascular
endothelial growth factor; PBMC, peripheral blood mononuclear cells; PSA, prostate specific antigen; rV,
recombinant vaccinia virus; scFv, single-chain antibody fragments.

Introduction renal cell carcinoma.1 Monoclonal antibodies have


become increasingly used therapeutic agents for the
Recent scientific advances have expanded our treatment of various types of malignancies. Many are
­understanding of the immune system and its response now being tested as components of adjuvant or first-
to malignant cells. These breakthroughs provided for line therapies to assess their efficacy in improving or
rapid progress in the field of cancer immunology and prolonging survival.2 Genetic constructs displaying
immunotherapy. The clinical goal of tumour immu- antibody-like specificity represent promising tools
notherapy is to provide either passive or active immu- for therapeutic interventions.3,4 Vaccine therapy rep-
nity against malignancies by harnessing the immune resent attempts to activate the immune system to
system to target tumours. recognize cancer cells and eliminate them from the
Cytokine therapy, applied in the form of exoge- body. Technological advances have allowed the con-
nously administered recombinant interferon-gamma struction of various types of vaccines such as DNA
(IFN) or interleukin-2 (IL-2), was the first type of or RNA vaccines, recombinant viruses expressing
immunotherapy to be applied in patients with vari- tumour genes, genetically engineered tumour cells
ous types of cancer, mostly including melanoma and or dendritic cells (DC), and synthetic long-peptides

Address for correspondence:  Professor M. Papamichail, Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras
Avenue, Athens 11522, Greece. E-mail: papamichail@ciic.gr
(Received 29 December 2008; revised 12 February 2009; accepted 26 March 2009)
ISSN 1040-8363 print/ISSN 1549-781X online © 2009 Informa UK Ltd
DOI: 10.1080/10408360902937809 http://www.informahealthcare.com/lab
168   C. N. Baxevanis et al.

representing a plethora of tumour immunogenic IL-2 was a second exogenous cytokine to dem-
epitopes.5 All these innovative approaches have onstrate anti-tumour activity against melanoma
considerably improved the field of cancer vaccines. and was approved for the treatment of adults with
Vaccines will certainly have an impact on the inci- advanced metastatic melanoma. IL-2 plays a central
dence of certain cancers, and research is ongoing role in immune regulation and T-cell proliferation.17
into therapeutic vaccines for various cancers, thereby High-dose bolus intravenous recombinant IL-2 was
opening up the possibility of administering vacci- shown to have anti-tumour effects when administered
nations to patients during disease-free intervals to with or without lymphokine-activated killer cells in
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

prevent recurrences. Combining immunotherapy phase II clinical trials involving advanced metastatic
with conventional therapies may prove to be the most melanoma patients.17,18 In these studies, an objective
effective strategy for patient benefit.6 Moreover, vacci- response rate of 16% (median response duration,
nation against cancer-specific antigens can sensitize 8.9 months; range, 4 to 106+) with a durable response
the tumour to subsequent chemotherapeutic treat- rate of 4% was demonstrated.18–21 IL-2 has been also
ment.7 Adoptive-cell (T, NK, NKT) transfer therapy approved for the treatment of renal and kidney carci-
has developed into an effective treatment, mainly for noma as well as haematological malignancies.
patients with metastatic melanoma.8 Current applica-
tion of this therapy relies on the ex vivo generation of
highly active, highly avid tumour-reactive lymphocyte Cytokines with high potential in treating
cultures from endogenous tumour-infiltrating lym- cancer
phocytes9 or on the genetic engineering of cells using
antigen receptor genes to express de novo tumour IL-7 is a 25 kDa T-cell growth factor that costimu-
antigen recognition.3,10 lates T cell receptor (TCR) signalling.22–24 IL-7 is
In this article, we review recent advances and per- required for T-cell development and T-cell survival
spectives in the immunotherapy of cancer, focusing in the ­periphery.22 It signals via the IL-7 receptor (R),
For personal use only.

on new strategies to enhance the efficacy of immu- comprising c and IL-7R.24,25 Expression of IL-7R
notherapeutic modalities as well as on clinical trials marks cells destined to become memory cells during
attempting to translate this knowledge into effective evolution of the immune response.26–28 In preclinical
cancer therapies. studies, IL-7 was demonstrated to act as a vaccine
adjuvant exerting profound effects on subdominant
responses.29 It has also been shown to enhance CD4+
FDA approved cytokines for cancer and CD8+ effector and CD8+ memory populations
immunotherapy and to support tumour protection following vaccina-
tion with DC.28 IL-7 is essential for the augmented
IFN, was the first cytokine to demonstrate anti-tumour anti-tumour effects during lymphopenia.27,28 Two
activity in patients with advanced melanoma. IFN-2b, phase I/II trials utilizing recombinant IL-7 have been
a type I IFN, is a highly pleiotropic cytokine with immu- completed in patients with cancer.30,31 There was no
noregulatory, antiproliferative, differentiation-induc- toxicity while dramatic increases in total body CD4+
ing, apoptotic, and antiangiogenic properties in mul- and CD8+ T cells were observed with no selective
tiple malignancies,1 and objective tumour response increase in Tregs. There appeared to be a preferential
rates of approximately 20% were observed in phase expansion of naïve T cells with an increased T-cell
I/II trials for metastatic disease.11,12 In 1995, recom- repertoire diversity. IL-7 may be considered a vaccine
binant IFN-2b became the first immunotherapy adjuvant to enhance the efficacy of tumour vaccines
approved for adjuvant treatment of stage IIB/III targeting self/tumour antigens for which an estab-
melanoma. The value of adjuvant high-dose IFN-2b lished self-tolerance exists.
for the treatment of high-risk melanoma has been eval- IL-15 is a cytokine and T-cell growth factor that is
uated in three US cooperative group studies.13–15 These produced by macrophages and DC and acts on CD8+
studies showed that high-dose IFN-2b significantly T cells, CD4+ T cells, and NK cells.32 It signals through
reduced the risk of recurrence; two of three studies IL-15R, which presents IL-15 in trans to IL-2/15 
also demonstrated a significant improvement in over- and the  chain receptor.33,34 IL-15 has various func-
all survival rates compared with observation or the tions, of which the most prominent are (i) inhibition of
GM2 ganglioside conjugated to keyhole limpet hemo- antigen-induced cell death (AICD) of T cells (in con-
cyanin mixed with QS-21 adjuvant vaccine.16 IFN-2b trast to IL-2, which promotes AICD),34–36 (ii) reversal of
has been also approved for the treatment of renal and T-cell anergy,34–36 (iii) promotion of induction of long-
kidney carcinoma, follicular lymphoma, hairy cell lived CD8+ T cells with potent anti-tumour efficacy,37
leukemia, and chronic myelogenous leukemia. (iv) substitution of CD4+ T-cell help in cytotoxic T
Cancer immunotherapy   169

lymphocyte (CTL) induction,35 (v) activation of NK of neutralizing human anti-mouse antibodies.62 These
cells,38 and (vi) induction of tumour regression in limitations were overcome by generating chimeric or
mice.39–41 IL-15 may potentially be applicable as a humanized mAbs.63 Moreover, growing knowledge of
vaccine adjuvant to induce longer-lived, more effica- key cellular pathways in tumour induction and pro-
cious CD8+ T cells, as a single agent to overcome T-cell gression provided the platform for appreciating the
anergy, as a T-cell growth factor in conjunction with role of humoral immunity in cancer defence, thus
adoptive T-cell immunotherapy, and as a systemic or paving the way for an increasing proportion of new
intratumoural cytokine therapy for cancer. drugs entering clinical trials.
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

IL-12 is a 70 kDa glycoprotein that binds IL-12R on Some molecules, such as trastuzumab, rituximab,
NK, T cells, DC, and macrophages.42 It promotes IFN alemtuzumab, cetuximab, are now widely used in
release by IL-12R-expressing T, NK, and NKT cells clinical practice.2,64,65 These antibodies are now being
and induces T helper 1 (Th1) polarization as well as tested in different indications alone or in combina-
proliferation of IFN expressing T cells.43,44 IL-12 also tion with standard chemotherapy. They are also being
displays anti-angiogenic activity and plays an impor- developed for the treatment of inflammatory diseases
tant role in anti-cancer development and immunity (rituximab). Numerous other antibodies are currently
in animal models.45–47 Phase I and II clinical data48–53 in pre-clinical and clinical development phases for
have demonstrated low efficacy when recombinant several malignancies, including renal carcinoma,
IL-12 was administered as monotherapy in patients melanoma, lymphomas, leukaemia, and breast, ovar-
with melanoma and renal cell carcinoma. Because of ian, and colorectal cancer.2,61 An alternative approach
its potency to induce robust IFN production by TCR- is to conjugate the mAb to a toxin,66 a cytotoxic agent,
activated T cells, IL-12 may induce more impressive or a radioisotope (Table 1).67 In other cases, these anti-
effects when co-administered with a vaccine. Its use as bodies aim to modify the tumour microenvironment
an adjuvant may both polarize Th1 responses and aug- through inhibition of angiogenesis or by enhancing
ment CD8 responses in any antigen-specific strategy. host immune responses against cancer.
For personal use only.

IL-21 belongs to the -chain family of cytokines If the molecule targeted by the antibodies is clearly
(together with IL-2, IL-4, IL-7, IL-9, and IL-15) and it identified, most often the precise mechanism of
is primarily produced by CD4+ T cells.54,55 It induces action of these immunoglobulins is understood. They
differentiation of pro-inflammatory CD4+ Th17 cells, can have direct effects in inducing apoptosis or pro-
stimulates memory and naïve CD8+ T-cell expansion grammed cell death,15 but they can also block growth
synergistically with IL-7 or IL-15, and improves degree factor receptors, efficiently arresting proliferation of
of expansion and affinity of antigen-specific CTL clones tumour cells.64,68 Indirect effects include (i) recruit-
generated in vitro.54–57 Its effects on NK cells include ing cells that exert cytotoxicity, such as monocytes,
augmentation/differentiation and anti-tumour macrophages, and, most important, T and NK lym-
activity.56–58 IL-21 also inhibits maturation, activation, phocytes (an immune pathway known as antibody-
and cytokine production of immature DC.59,60 In pre- dependent cell-mediated cytotoxicity)62,68 and (ii)
clinical models in vivo, IL-21 has been demonstrated binding complement, which leads to toxicity known
to induce tumour rejection, prevention of metastases, as complement-dependent cytotoxicity.68 mAbs may
and immune memory.59,60 Because of its effects on also act as regulators of immune responses in vivo.
CD8+ T cells, IL-21 may be essential for in vitro gen- We have recently demonstrated that breast cancer
eration and expansion of tumour antigen-specific CTL patients on trastuzumab exhibited decreased levels
lines/clones to be used in adoptive immunotherapy. of serum HER-2/neu, which correlated with a drop in
Moreover, it can be used as a systemic immunomodu- the numbers of peripheral T regulatory cells (Tregs)
lator combined with monoclonal antibody therapy for and objective clinical responses.69 This novel func-
potentiating NK cell-mediated ADCC. tion of mAbs may have important implications for the
down-regulation of tumour-associated peripheral
and local immune suppression mechanisms.
Monoclonal antibodies for cancer treatment

The development of hybridoma technology by Kohler Immunomodulatory mAbs for the treatment of
and Milstein61 led to the production of monoclonal cancer
antibodies (mAbs), which have considerably modified
the field of clinical oncology. The poor performance of Immunostimulatory mAbs directed to immune recep-
mAbs in early clinical settings was attributed to short tors have emerged as a new and promising strategy
antibody half-life and the immunogenicity of the to fight cancer. In general, mAbs can be designed
mouse protein in humans (resulting in the production to bind molecules on the surface of lymphocytes or
170   C. N. Baxevanis et al.

antigen-presenting cells to provide activating signals temozolomide in patients with advanced relapsed or
(e.g. CD28, CD137, CD40, and OX40).70,71 On the other refractory melanoma.82
hand, mAbs can also be used to block the action Phase I studies of ipilimumab were performed
of surface receptors that normally downregulate in patients with prostate cancer,83 melanoma,84 and
immune responses [cytotoxic ­T-lymphocyte–associated ovarian cancer.85 In these studies, patients after a
antigen 4 (CTLA-4) and PD-1/B7-H1]. In com- single administration of ipilimumab achieved some
bined regimes of immunotherapy, these mAbs are clinical efficacy, as manifested by incomplete reduc-
expected to improve therapeutic immunizations against tion of tumour size and extensive tumour necro-
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

tumours as observed in preclinical studies. sis with leukocyte infiltration. In phase II studies,
Anti-4-1BB (agonistic anti-CD137) mAb has been repeated administrations with ipilimumab allowed
successfully tested as an anti-cancer molecule in pre- more patients to achieve objective responses.86 The
clinical studies.70 4-1BB is a member of the tumour combination of ipilimumab with chemotherapeu-
necrosis factor/nerve growth factor family of receptors tics [dacarbazine87 or docetaxel88], with IL-289 or with
and has a natural ligand (4-1BBL) that is expressed on melanoma-associated peptide vaccines90 improved
activated T lymphocytes as well as on NK cells and the rate of complete responses in patients compared
DC.72 This mAb, which acts against CD137, has the with the monotherapy arms, demonstrating that com-
ability to stimulate potent anti-tumour responses73 bining anti-CTLA-4 therapy (i) with cytotoxic therapy,
and, paradoxically, ameliorates autoimmune mani- such as chemotherapeutics, may create an enhanced
festations in mice.73 environment for immunomodulatory treatment
Therapy with mAbs against CTLA-4, which block by potentially releasing endogenous antigens from
the inhibitory action of CTLA-4 on T cells, is capable tumours and/or altering the immune milieu, and
of inducing anti-tumour responses in mice as well (ii) with treatments that have different mechanisms
as in humans, but is accompanied by adverse events of action, it may be possible to produce additive or
in the form of autoimmune reactions.74 Anti-CTLA-4 synergistic effects.
For personal use only.

mAb was developed to bind to the CTLA-4 on T cells


and thereby prevent the inhibitory cascade triggered
by CTLA-4 binding to B7.75–77 In a murine model, anti- Genetic constructs specifically targeting
CTLA-4 mAb has demonstrated anti-tumour activity tumour-associated antigens
in moderately antigenic and highly immunogenic
tumours.75 Anti-CTLA-4 mAb alone is not sufficient Chimeric antigen receptors
to alter the growth of poorly immunogenic tumours,
such as MC38.78,79 Preclinical investigations have been Chimeric antigen receptors (CARs) represent a tech-
performed using anti-CTLA-4 mAb in combination nological improvement in the field of cancer immu-
with cancer vaccines to increase the magnitude of notherapy for retargeting the functional program
the T-cell response and the avidity of antigen-specific of immune lymphocytes towards specific molecu-
T cells.77 Based on the strength of these preclinical lar targets expressed on the cell surface of tumour
data, clinical trials have been initiated with two fully cells. CARs are genetic constructs consisting of an
human anti-CTLA4 mAbs. antigen-recognizing antibody molecule linked to a
Tremelimumab induced durable objective T-lymphocyte signalling domain that can be modi-
responses with low-grade toxicities when used as fied to include co-stimulatory signals that improve the
second-line monotherapy in a phase I study with activation of CAR-expressing cells.3The main advan-
melanoma patients treated with single, escalating tage of CARs is their ability to redirect T-lymphocyte
doses.80 Subsequently, in a phase II trial, patients specificity and their killing/effector activity towards
with advanced melanoma randomly were assigned a selected target in a non-major histocompatibility
to receive either  10 mg/kg tremelimumab monthly complex (MHC)-restricted fashion, exploiting the
or 15 mg/kg tremelimumab every three months.81 antigen-binding properties of mAbs.3 Ligand binding
Although the response rate was not significantly dif- by the chimeric receptor triggers phosphorylation of
ferent between the two arms, the 15 mg/kg every the immunoglobulin tyrosine activation motifs in the
three months regimen was associated with a lower cytoplasmic region of the molecule and this activates a
incidence of grade 3/4 adverse events. Consequently, signalling cascade that is required for the induction of
the 15 mg/kg every three months dosing regimen cytotoxicity, cytokine secretion, and proliferation.91,92
was selected for further study and is currently being CAR-expressing cells, by recognizing tumour antigens
investigated for single-agent anti-tumour activity in a in a non-MHC-restricted manner, are not affected by
larger phase II open-label trial and in a randomized down-regulation of HLA (human leucocyte antigen)
comparative phase III study against dacarbazine or class I antigens and by defects in the antigen-processing
Cancer immunotherapy   171

machinery of tumour cells.3 Moreover, this technol- function by folding into a specific three-dimen-
ogy enables the generation of large numbers of lym- sional structure that dictates high-affinity binding
phocytes with specificity for their autologous tumours to the targeted protein.4,111 Because they inhibit the
to be used in adoptive cellular immunotherapy.92–94 activity of existing proteins directly, aptamers are
CAR technology has been successfully applied to more similar to mAb or small molecule drugs than
enhance tumour recognition by primary T cells,95–97 to antisense compounds, and this property greatly
and several studies have demonstrated specific kill- increases the number of clinical indications that are
ing of tumour target cells in vitro and in vivo following potentially treatable by nucleic acid-based com-
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

redirection of T and NK cell lines98–100 or primary NK pounds.111 Aptamers exhibit specificity and avidity
cells101,102 with chimeric receptors. comparable to or exceeding that of mAbs and can
Several clinical trials using lymphocytes engineered be generated against most targets.4 Unlike mAbs,
to express CARs have been implemented; however, so aptamers can be synthesized in a chemical process
far, no positive clinical responses have been noted.103,104 and hence offer significant advantages in terms of
One major obstacle to this form of therapy is the pres- reduced production cost and simpler regulatory
ence of high serum levels of soluble monovalent anti- approval processes.4,112
gen, which upon binding to grafted cells would render In many instances, aptamers have been shown
them anergic. Thus, such modalities of therapy would to inhibit the function of their targets, presumably
be restricted to patients with low levels of soluble by blocking binding of the cognate ligand.4,113 An
tumour antigen. The antigenic diversity in tumour-cell aptamer targeting vascular endothelial growth factor
populations may also hamper positive clinical results. (VEGF) became the first aptamer approved for human
The use of lymphocytes grafted with CARs recognizing therapy to treat age-related macular degeneration,
more than one antigen may circumvent this problem. arguably a milestone in the application of aptamer
Preclinical studies have demonstrated the capacity of technology.114 Modulation of immune responses
T cells expressing bispecific CARs to lyse tumour cells in vivo by using aptamers was first described by
For personal use only.

expressing both carcinoembryonic antigen (CEA) and Santulli-Marotto et  al.,115 who generated aptamers
TAG-72103 or HIV-infected cells expressing wild-type that bind and inhibit the function of murine CTLA-4,
and mutant envelope proteins.105 thereby enhancing tumour immunity. Bivalent and
Another limitation of CARs is their potential immu- multivalent configurations of other immunomodulat-
nogenicity. Since single-chain antibody fragments ing aptamers binding to 4-1BB or OX40 costimulated
(scFv) are most often derived from a mAb of murine T-cell activation in vitro and promoted tumour rejec-
origin, this might trigger a host immune response and tion in vivo.112,116
therefore accelerate T-cell clearance. Humanizing Aptamers offer important advantages over antibod-
the mouse portions of CARs or by using previously ies to manipulate the immune system for therapeutic
humanized scFvs106,107 may offer a solution to this purposes. Antibodies are cell-based products requir-
problem. Another aspect that should be taken into ing a complex and regulatory approval process that in
consideration when utilizing lymphocytes grafted many cases provide a serious obstacle to their availa-
with CARs in clinical trials is that T cells of cancer bility for clinical application. Because aptamers can be
patients may possess abnormalities in their TCR- chemically synthesized, the manufacturing and regu-
mediated signalling.108,109 Thus, the question to be latory approval processes should be much simpler and
addressed is whether potential downstream defects less costly. The use of ­immune-modulating aptamers
in the signalling cascade will impair signal transduc- as adjuvants to cancer takes advantage of the inherent
tion initiated by the chimeric receptor(s). lack of immunogenicity of aptamers.4 Antibodies are
unlikely to be suitable for this application. Thus, for the
field of therapeutic aptamers to reach its full potential,
Aptamers translational researchers and biotechnology compa-
nies must take advantage of the unique properties of
The idea of using nucleic acid molecules as thera- aptamers to address important, unexplored clinical
peutic agents was initially entered into praxis with issues.
the development of antisense strategies. Antisense
compounds are single-stranded nucleic acids
that disrupt the synthesis of a targeted protein by Bispecific scFv antibodies
hybridizing in a sequence-dependent manner to
the mRNAs that encode it.110 The mechanism of Bispecific T-cell engager molecules (BiTEs) consti-
inhibition by single-stranded nucleic acid aptamers tute a class of two flexibly linked scFvs that have the
is different in that these directly inhibit a protein’s potential to redirect tumour-resident and circulating
172   C. N. Baxevanis et al.

T cells to lyse tumour cells.10 One scFv interacts with observed in a small minority of patients, the overall
epitopes on tumour cells, whereas the other scFv results have been disappointing.130 The negative expe-
binds an epitope on the TCR/CD3 complex of T cells. rience with anti-tumour vaccines may suggest that
BiTEs have the potency and efficacy to target tumour development of novel vaccine strategies for cancer are
cells at low T-cell numbers without the need for T-cell needed to improve recognition, immune response,
co-stimulation. BiTEs have been demonstrated to effector functions, and trafficking of T cells induced
induce lytic synapses between human T cells and a by vaccination. These goals may be achieved by con-
MHC class I-negative, tumour cell line, resulting in current administration of novel immunotherapeutics
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

potent target-cell lysis.117 This shows that certain T-cell with an immunopotentiating profile.
recognition molecules on target cells are dispensable
for synapse formation and BiTE activity and suggests
that BiTE-activated polyclonal T cells may ignore Blocking tumour-induced immune suppression
major immune evasion mechanisms of tumour cells
in vivo, such as loss of MHC class I expression. The Mabs capable of neutralizing the biologic activity of
concepts underlying BiTE technology have been suppressive factors released by the tumour, such as
successfully validated in preclinical studies with two anti-VEGF antibody bevacizumab, which, in addi-
different target antigens, namely CD19 and epithelial tion to its effect on tumour vasculature may decrease
cell-adhesion molecule (EpCAM).118–121 An EphA2- VEGF-induced inhibition of DC and T-cell function,70
specific bispecific single-chain antibody (bscAb), mAbs to transforming growth factor (TGF)-, and
linked to anti-CD3 bscAb, was recently designed to IL-10 are also potential means to reverse immune
target epitopes that are uniquely exposed on malig- escape induced by tumour-released molecules. In
nant cells.122 bscEphA2xCD3 had the specificity and addition, nonsteroidal anti-inflammatory drugs,
potency to redirect low numbers of T cells for lysis of including the specific cyclooxygenase-2 inhibitor
EphA2-expressing tumour cells that possessed few celecoxib, could be a means of inhibiting cyclooxyge-
For personal use only.

available sites for bscEphA2xCD3, both in cell cultures nase-2 that leads to prostaglandin E2 production.131
and animal models. A CD19/CD3-directed bscAb
(referred to as MEDI-538, MT103, or bscCD19xCD3)
is currently being investigated in the clinic as a poten- Removal or blocking the function of cells with
tial therapy for B-cell malignancies. suppressive activity

The immunosuppressive microenvironment of a


The concept of anti-tumour vaccination tumour reduces the effect of therapeutic vaccines, both
during the induction of immunity and in the effector
Tumour vaccines have been designed to either phase of the response. One way to improve the induc-
increase immune recognition of tumour cells or to tion phase is to block the negative regulators of the
enhance the anti-tumour immune response through activation of effector T cells.132 Antibodies against one
lymphocyte activation.123 Vaccines have utilized such molecule, CTLA-4, are being evaluated in clini-
mixtures of antigens or single antigens specific to cal trials.133 CTLA-4 is expressed on activated T cells,
the target. Preclinical studies with vaccines showed where it serves as an inhibitor of activation. Blocking
evidence of complete tumour regression and/or the activity of CTLA-4 allows greater expansion of all
prolonged stabilization of tumour growth.124 In early T-cell populations, presumably including those with
clinical trials, vaccines prepared from whole tumour anti-tumour reactivity (see above). The depletion of
cells were associated with limited activity, presumably immunosuppressive cells from the tumour environ-
due to the already biased nature of the host immune ment is also mandatory for the development of robust
response to specific tumour-associated antigens.125 anti-tumour immunity. Denileukin diftitox (DAB/IL2;
The discovery and cloning of a number of tumour- Ontak) is a recombinant DNA-derived cytotoxic pro-
associated antigens spurred interest in active immu- tein composed of diphtheria toxin fragments A and B
nization for cancer immunotherapy. Attempts at and the full-length IL-2 molecule. DAB/IL2 binds to
immunotherapy of cancer have included therapeutic CD25 (the IL-2R chain) and, following internalization,
vaccination with recombinant viral vectors encoding inhibits protein synthesis, causing cell death within
tumour-associated antigens,126 recombinant proteins hours.134 DAB/IL2 is FDA approved for the treatment
with appropriate adjuvants,127 antigen-loaded DC,128 of patients with persistent or recurrent cutaneous T cell
DNA encoding tumour-associated antigens,5 and syn- lymphoma whose malignant cells express CD25. When
thetic peptides.129 However, apart from in melanoma, administered to patients with melanoma, this protein
in which impressive clinical responses have been depletes the blood of regulatory T cells. In most patients
Cancer immunotherapy   173

(90%), this treatment has resulted in the production of untreatable. This is broadly true even in instances
melanoma-specific CD8 T cells.135 The function of the in which conventional anticancer treatments have
immunosuppressive enzyme indoleamine 2,­3-dioxyge- been used to generate a scenario of minimal resid-
nase (IDO) can be inhibited by ­1-methyl-tryptophan,136 ual disease in which to administer immunotherapy.
and clinical trials testing its effects in patients with can- Therapeutic vaccination in the adjuvant setting is
cer are planned. This reagent would have the potential rarely capable of eradicating residual tumour cells
of synergizing with immunotherapy by inhibiting that are present after surgery and eliciting a complete
immune suppression by IDO. response. This inability probably reflects the fact that
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

debulking tumours removes only one component of


the systemic immune suppression that they estab-
Induction of sensitivity to apoptosis lish, namely the intratumoural suppressive milieu.
This indicates that the suppressive T cells/DC that
Modulation of tumour necrosis factor-inducing ligand have already escaped the tumour may be active in
(TRAIL) receptors, which are preferentially expressed the lymph nodes and that the immunosuppressive
by cancer cells, may synergize with vaccination ther- cytokine profile present in the peripheral blood of
apy and other targeted therapy approaches. Histone the patient is capable of limiting vaccine efficacy
deacetylase inhibitors have been shown to up-regulate even in the absence of the immunosuppressive
the TRAIL death receptor DR5, leading to sensitization tumour microenvironment.
of cancer cells to TRAIL-induced death,137 and therefore Such considerations have led to the hypothesis
would be candidate drugs to use in combination with that immunization with a therapeutic cancer vaccine
TRAIL-ligand constructs, TRAIL ­receptor-activating at the earliest stages of carcinogenesis—before local
antibodies, and vaccine-induced immune effector and systemic immunosuppressive environments
cells. Furthermore, the demethylating agent decitab- are established by the tumour—will yield the best
ine has been shown to sensitize melanoma cell lines immune responses to vaccination and therefore con-
For personal use only.

to the cytotoxic effects of IFNs by inducing the expres- fer excellent protection against the progression of can-
sion of proapoptotic and growth-inhibitory genes.138 cer development. An example of a currently available
The antiapoptotic members of the Bcl-2 family are vaccine that may benefit from early administration
potential targets for immunosensitizing drugs. Bcl-2 is resulting from improved screening methods, includes
also up-regulated in many cancers,139 and inactivating a breast cancer vaccine consisting of a HER-2/neu
its function may result in a proapoptotic cancer cell peptide (E75) mixed with received granulocyte-mac-
inflammatory milieu facilitating the cytotoxic effect rophage colony-stimulating factor (GM-CSF) that has
of vaccine-induced immune cells. ABT-737 is a small- been successful in preventing disease recurrence in
molecule inhibitor of the anti-apoptotic proteins node-positive patients with minimal disease bur-
Bcl-2, Bcl-XL, and Bcl-w. Mechanistic studies reveal den.143 In addition, Holmes et al.144 performed a phase
that ABT-737 does not directly initiate the apoptotic I clinical trial of the novel Ii-Key/HER-2(776-790)
process but enhances the effects of death signals, hybrid preventive vaccine in disease-free, node-neg-
displaying synergistic cytotoxicity with chemothera- ative breast cancer patients. In this study, the Ii-Key
peutics and radiation.140 Apoptotic pathways may modified vaccine showed enhanced immunological
also be targeted by altering transcription of regula- responses in the absence of an immunoadjuvant. In a
tors of apoptosis. Inhibition of nuclear factor-B in randomized, double-blind study, patients with stage
melanoma sensitizes cells to TRAIL-induced (but not II breast cancer, with no evidence of disease and at
Fas or TNF-) apoptosis.141 An additional approach low risk for recurrence, received injections with oxi-
would be the use of the proteasome inhibitor bort- dized mannan-MUC1 to immunize against MUC1 and
ezomib, which is known to inhibit nuclear factor-B prevent cancer recurrence/metastases.145 Immunized
and down-regulate the antiapoptotic molecule flice patients had no recurrences (0/16) for more than five
inhibitory protein (FLIP).142 years after treatment, whereas the recurrence rate
in patients receiving placebo was 27% (4/15). In the
majority of vaccinated patients, both humoral and
Novel approaches for improving clinical cellular immunity to MUC1 was evident.
results with cancer vaccines

Preventive vaccination Therapeutic vaccines combined with chemotherapy

Compromised immunity in patients with advanced The combination of active immunization with
cancer is largely unavoidable and, currently, standard treatments is provocative because of the
174   C. N. Baxevanis et al.

immunosuppressive effects of most standard treat- to the chemotherapy (61.9%) that immediately
ments. The in vivo preclinical data by Machiels et al.146 followed vaccination. Clinical response to subse-
addressing the issue of chemotherapy and cancer quent chemotherapy was closely associated with
vaccines indicated that taxane therapy (employing induction of immunologic response to vaccination.
taxol) may in fact increase T-cell precursors rather Furthermore, Wheeler et al.151 analyzed survival and
than deplete them. Later on, Arlen et al.147 designed progression times in 25 vaccinated (13 with and 12
one phase II study of patients with metastatic without subsequent chemotherapy) and 13 non-vac-
androgen-independent prostate cancer randomized cinated de novo glioblastoma (GBM) patients receiv-
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

to receive vaccine alone or vaccine with low-dose ing chemotherapy. Vaccinated patients receiving
docetaxel. The vaccination regimen was composed subsequent chemotherapy exhibited significantly
of (a) recombinant vaccinia virus (rV) that expresses longer times to tumour recurrence after chemother-
the prostate-specific antigen gene (rV-PSA) admixed apy relative to their own previous recurrence times
with (b) rV that expresses the B7.1 costimulatory gene as well as significantly longer post-chemotherapy
(rV-B7.1) and (c) sequential booster vaccinations with recurrence times and survival relative to patients
recombinant fowlpox virus (rF-) containing the PSA receiving isolated vaccination or chemotherapy.
gene (rF-PSA). Patients received GM-CSF with each These data suggest that vaccination against
vaccination. cancer-specific antigens can sensitize the tumour
The median increase in T-cell precursors to PSA was against subsequent chemotherapeutic treatment.
equal in both arms following three months of therapy. Although the mechanisms that underlie such a
In addition, immune responses to other prostate can- synergistic effect have not been elucidated yet, it is
cer-associated tumour antigens were also detected speculated that the vaccination-induced increase
post-vaccination. Eleven patients who progressed on in the frequency of primed T cells may constitute
vaccine alone crossed over to receive docetaxel at the a major advantage by the time tumour microen-
time of progression. Median progression-free survival vironment is modified by cytotoxic drugs [e.g. dis-
For personal use only.

on docetaxel was 6.1 months after receiving vaccine ruption of tumour stroma, decreased suppressive
compared with 3.7 months with the same regimen in activity, increased sensitivity of tumour cells to
a historical control. This was the first clinical trial to granzymes and up-regulation of tumour-associated
show that docetaxel can be administered safely with antigens11].
immunotherapy without inhibiting vaccine specific
T-cell responses. Based on their findings, the authors
hypothesized that patients previously vaccinated Adjuvants and multiple peptide vaccines, including
with an anticancer vaccine may respond longer T-helper epitopes
to docetaxel compared with a historical control of
patients receiving docetaxel alone. Over the years, various vaccines containing exactly
In another phase I study,148 17 patients with different fitting MHC class I-binding peptides have been
types of advanced cancer were immunized with anti- tested for their therapeutic efficacy against both
gen cytochrome P450 1B1 (CYP1B1), which is overex- virally and non-virally induced cancers.123 However,
pressed on almost all human tumours.149 Six patients the observed immunological and clinical responses
developed immunity to CYP1B1, three of whom devel- were rather poor. Therefore, ways to develop more
oped disease stabilization. All but one of 11 patients powerful cancer vaccines should be thoroughly
who did not develop immunity to CYP1B1 progressed explored. Preclinical studies demonstrate that
and did not respond to salvage therapy. Five patients tumour-specific CD4+ Th cells critically contribute
who developed immunity to CYP1B1 required salvage to the development and efficacy of anti-tumour
therapy for progressive metastatic disease and showed responses.152–154 The effectiveness of these Th cells
marked response to their next treatment regimen, probably lies in their capacity to deliver essential
which for most lasted longer than one year. activation signals to DC, needed for an optimal
Antonia et al.150 vaccinated 29 patients with exten- priming of tumour-specific CTLs.153,154 In addition,
sive stage small cell lung cancer with a DC-based p53 antigen-specific CD4+ T cells may provide CTL with
vaccine. Objective clinical response to vaccination essential growth stimuli during the effector phase.43
as well as subsequent chemotherapy was evaluated. In line with this notion, several studies show that
p53-specific T-cell responses to vaccination were effective CTL priming can be induced by the inclu-
observed in 57.1% of patients. One patient showed sion of Th epitopes in peptide vaccines.152,155
a clinical response to vaccination, whereas most of Another strategy to potentiate vaccines is the use
the patients had disease progression. However, they of molecularly defined strong DC-activating adju-
observed a high rate of objective clinical responses vants, such as oligodeoxynucleotide (ODN)-CpG,
Cancer immunotherapy   175

monophosphoryl lipid A (MPL), anti-CD40 Ab, and Clinical trials using polyepitope
GM-CSF.156–158 An increase in the length of the peptide long-peptide vaccines
used for vaccination strongly affects the magnitude of
the induced CD8+ T-cell response. Vaccination with The necessity of CD4 T-cell help to generate and sustain
long peptides22–45 amino acids in length) containing MHC class I-restricted CD8 T-cell responses has led
either a human papillomavirus (HPV)-derived, an not only to the use of MHC class II-restricted epitopes
adenovirus-derived or a modified ovalbumin-derived derived from the same protein,168,169 but also to uni-
CTL epitope resulted in more robust and effective CTL versal, nonspecific MHC class II-restricted epitopes,
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

responses than vaccination with the minimal CTL such as PADRE, in clinical vaccination trials.170,171 Two
peptide.152,159 Even under conditions that precluded clinical studies172,173 determined whether HER-2/neu–
T-cell help, a single vaccination with a long-peptide specific CD8 T-cell immunity could be elicited using
vaccine induced significantly higher CD8+ T-cell HER-2/neu–derived MHC class II helper peptides,
responses in MHC class II knockout mice and in CD40 which contained encompassed HLA-A2–binding
knockout mice than a minimal CTL peptide.152,159 motifs. Nineteen HLA-A2 patients with HER-2/neu–
A re-evaluation of the early reports on the success- overexpressing cancers received a vaccine prepara-
ful use of peptide vaccines in animal models reveals tion consisting of putative HER-2/neu helper peptides
that most of the vaccines used were actually longer p369–384, p688–703, and p971–984. Contained within
than the exact minimal CTL peptide epitope.160–163 these sequences are the HLA-A2–binding motifs p369–
Physical linking of Th and CTL epitopes or conjugation 377, p689–697, and p971–979. Following vaccination,
of toll-like receptors (TLR) ligands to CTL peptides the mean peptide-specific T-cell precursor frequency
increases the size of the ultimate peptide used and to the HLA-A2 peptides increased in the majority of
as such might have improved the CTL response. The patients. In addition, the peptide-specific T cells were
mechanism that explains the difference between long able to lyse tumours. The responses were long lived
and minimal CTL peptides lies in the fact that injec- and detectable for more than one year after the final
For personal use only.

tion of short peptides leads to the exogenous load- vaccination in patients. These results demonstrated
ing in vivo of MHC class I molecules on all cells that that HER-2/neu MHC class II epitopes containing
have such molecules, including T cells and B cells.164 encompassed MHC class I epitopes are able to induce
Moreover, these B and T cells circulate and, therefore, long-lasting HER-2–specific IFN-–producing CD8 T
also arrive in lymph nodes throughout the body in the cells. In addition, the generation of protein-specific
absence of immunostimulatory adjuvants. This una- immunity after peptide immunization was associated
voidably leads to immunological tolerance because T with epitope spreading reflecting the initiation of an
and B cells, in contrast to properly activated DC, lack endogenous immune response.
the co-stimulatory surface molecules required for A polyepitope peptide vaccine, named IDM-
appropriate effector CTL generation. 2101, has been evaluated for safety and induction of
Another advantage of long-peptide vaccines over immune responses in phase I trials in colon cancer
minimal CTL-peptide vaccines is the increased dura- and metastatic non-small-cell lung cancer (NSCLC)
tion of in vivo epitope presentation in the antigen- patients174 and for clinical efficacy in phase II tri-
draining lymph node,164 previously shown to be impor- als in NSCLC patients.175 The IDM-2101 vaccine was
tant for clonal expansion165 and for IFN production designed to induce CTLs against five tumour-asso-
by effector T cells.166 Such improved in vivo presenta- ciated antigens (TAAs) frequently overexpressed in
tion and concomitant induction of T-cell expansion NSCLC (i.e. carcinoembryonic antigen, p53, HER-
by an extended CTL peptide epitope is particularly 2/neu, and melanoma antigens [MAGE] 2 and 3).
apparent if the CTL epitope concerned displays IDM-2101 is composed of 10 synthetic peptides from
weaker MHC class I binding.164 Another advantage of these TAAs, nine of the peptides representing CTL
long-peptide vaccines is that they include a plethora epitopes. Each CTL epitope is restricted by HLA-A2.1
of immunogenic epitopes and thus are capable of and at least one other member of the HLA-A2 super-
eliciting T-cell responses against many different HLA family of major histocompatibility complex class I
class I and class II alleles.167 This will certainly prevent molecules, providing coverage of approximately 45%
escape of tumours from immunosurveillance through of the general population. The tenth synthetic peptide
mutation of T-cell epitopes and down-regulation of is the pan-DR epitope PADRE. In the phase I studies,
certain HLA alleles. Therefore, injection of long-pep- no significant adverse effects were observed, and a
tide vaccines will ensure pluralism in the anti-tumour strong CTL response in the majority of patients was
response in which several tumour-specific CD8+ and noticed.174
CD4+ T cells will contribute to more efficient tumour In the phase II study, patients achieved initial sta-
killing. ble disease and overall survival in comparison with
176   C. N. Baxevanis et al.

historical patients who received conventional treat- showed partial regression (>50% reduction in size)
ment.175 A novel melanoma vaccine comprising six of the lesions with marked relief of symptoms. These
melanoma-associated peptides as antigenic targets results are encouraging because further improvement
for melanoma reactive helper T cells was evaluated can be expected from co-formulation of this vaccine
for safety and immunogenicity in a phase I/II trial.176 with a potent TLR ligand.
Vaccination with the helper peptide vaccine was well We are currently utilizing peptide HER-2/neu
tolerated. Proliferation assays revealed induction of (776–790), chemically linked to a four amino-acid
T-cell responses to the melanoma helper peptides long peptide from the regulatory region of the MHC
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

in 81% of patients. Among patients with measurable class II invariant chain (Ii-Key),152 for enhanced
disease, objective clinical responses were observed in immunogenicity, in a phase I trial with HER-2/neu+
two patients (12%), with response durations of 1 and prostate cancer patients. Only grade I toxicity has
3.9 years. Durable stable disease was observed in two been observed in 10% of the patients, whereas 80% of
additional patients for periods of 1.8 and 4.6 years. those (24 of 30) have responded with increased immu-
Welters et  al.177 initiated a phase I/II clinical vac- nological responses to the vaccine in vivo (measured
cination study in end-stage cervical cancer patients as delayed-type hypersensitivity (DTH)) and in vitro
with a vaccine consisting of a set of 13 overlapping (as determined by the IFN-based ELISPOT assay).
peptides, 27–35 amino acids in length, of the complete We are observing similar results using this Ii-Key
sequence of the HPV16 E6 and E7 oncogenic proteins. conjugated HER-2/neu peptide in an ongoing phase
The vaccine, in Montanide adjuvant, was well toler- II study with HER-2/neu+ node-positive and high risk
ated, and one out of 35 patients had a complete and node-negative breast cancer patients for prevention of
lasting disease regression. Five additional patients recurrences. Although our peptide vaccine is injected
achieved ­stable disease with salvage chemotherapy admixed with GM-CSF as an adjuvant, strong DTH
after vaccination lasting up to two years, but, on aver- responses in some of the patients also can be detected
age, all other patients died within five months. This in the absence of GM-CSF, demonstrating the high
For personal use only.

indicated that therapeutic vaccination in this cate- effectiveness of the Ii-Key/peptide formulation.
gory of patients has only limited therapeutic action by
itself, but might exert much better therapeutic activity
in conjunction with conventional chemotherapy or Adoptive cellular immunotherapy
radiation therapy.
At the same time, six patients who had been sur- Adoptive transfer of autologous tumour-specific T
gically treated for HPV16-positive cervical cancer cells, as initially reported in the late 1980s, has been
were subcutaneously vaccinated with the same set of shown to reproducibly mediate the destruction of
overlapping long peptides. Vaccine-induced HPV16 bulky tumours in a respectable percentage of the
E6-specific T-cell responses were detected in six out patients treated.179 This approach is mainly based on
of six patients and HPV16 E7-specific T-cell responses growing tumour-infiltrating lymphocytes ex vivo from
in five out of six patients. The responses were broad, surgically excised tumour specimens of patients and
comprised both HPV16-specific CD4+ and CD8+ then adoptively transferring them into the patient
T-cells, and could be detected up to 12 months after along with bolus high-dose IL-2.8 This type of immu-
the last vaccination. The vaccine-induced responses notherapy has the advantage that one can administer
were dominated by effector-type CD4+CD25+FOXP3− extremely high numbers of non-modified or geneti-
type 1 cytokine IFN-producing T cells (Th cells), cally engineered lymphocytes with high avidity for
but also included the expansion of T cells with a tumour antigens.9 Furthermore, adoptive cellular
CD4+CD25+FOXP3+ phenotype (Tregs). The pres- immunotherapy (ACT) can include both CD4+ and
ence and vaccine-induced increase of HPV16-specific CD8+ T cells capable of recognizing tumour antigens
Tregs indicate that strategies to eliminate or disarm and synergistically acting in the anti-tumour immune
these cells should be considered for immunothera- response.180,181 These cells are capable of massive
peutic strategies against HPV-induced cancers. expansion in both mice and humans. Another advan-
The same group conducted a phase II vaccina- tage of ACT is that the adoptively transferred T cells
tion study178 in patients with HPV16-positive vulvar are capable of trafficking to virtually every somatic
intraepithelial neoplasia grade III, a premalignant site.182 However, these promises have not translated
condition. Patients were vaccinated four times with into clinical successes for cancer patients. Even
the same vaccine, which consisted of 13 overlapping where tumour regression or complete responses were
long peptides of the E6 and E7 oncoproteins of HPV16. achieved, there was usually relapse of the disease.183–
Of the patients analyzed, 25% achieved complete 185
Increasing numbers of reports over recent years
regression of all lesions, and a number of patients have highlighted potential immunosuppressive
Cancer immunotherapy   177

mechanisms that act on tumours and systemically from the gut and the systemic liberation of bacterial-
in cancer patients to block effective anti-tumour derived lipopolysaccharide. This TLR 4 agonist, then,
immune responses.186 One method to overcome the acts to activate the innate immune system, leading
immunosuppressive environment induced by the to increased levels of proinflammatory cytokines,
tumours is to use lymphodepleting preparative regi- activated DC, and enhanced anti-tumour function of
mens to provide an altered environment for trans- adoptively transferred T cells.
ferred cells.187,188 The third mechanism by which lymphodeple-
tion is thought to augment ACT is through the
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

depletion of immune cells that act to suppress


Augmentation of adoptive immunotherapy through immune responses in vivo.193 Tregs are a subset
lymphodepletion of CD4+ T  cells that are known to inhibit both the
activation and proliferation of cytotoxic and helper
Adoptive transfer of ex vivo expanded anti-tumour T cells, and they are likely to play a major role in
T cells after a nonmyeloablative lymphodepleting the induction of functional immune tolerance to
preparative regimen with chemotherapeutic rea- tumours often observed in cancer patients.194 Other
gents is the most effective treatment for patients with regulatory CD4+ T-cell subsets likely targeted by
metastatic melanoma. When anti-tumour T cells are lymphodepletion include Tr1 and Th3 cells, which
given after lymphodepletion, usually 50% of patients produce the immunosuppressive cytokines IL-10
refractory to other treatments experience objective and TGF-ß, respectively.195,196 Increased numbers of
clinical responses with this treatment.187,189 Not all Tregs in cancer patients have been correlated with
patients’ tumours yield anti-tumour T cells, however, an unfavourable prognosis, arguing that elimination
and only about one-third of the patients experience of these cells may result in an improved efficacy of
durable tumour regressions. Thus, there are signifi- adoptive immunotherapy.197 A novel mechanism
cant efforts to continue to improve current therapies. that promotes anti-tumour immunity was recently
For personal use only.

Preclinical data show that increasing the intensity of suggested by Shvets et al.,198 who found that in irra-
lymphodepletion to a myeloablative preparative regi- diated mice undergoing homeostatic proliferation,
men that requires hematopoietic stem cell transplant T cells displayed severely decreased expression of
might further improve the effectiveness of adoptive inhibitory molecules PD-1 and CTLA-A4, which
cell transfer.190,191 Nevertheless, this type of immuno- rendered them insensitive to tumour-induced sup-
therapy, besides melanoma, should also be applied to pressor mechanisms. In addition, these cells could
other types of cancer. not be converted to Tregs since they were refractory
to Foxp3 expression upon treatment with TGF.
Although lymphodepletion is effective at eliminat-
How does lymphodepletion improve adoptive T-cell ing regulatory T cells in vivo, recent studies have
therapy? shown that these cells repopulate the periphery very
rapidly, highlighting the transient and possibly lim-
While the exact mechanisms by which lymphodeple- iting nature of this approach.199
tion regimens lead to improved responses to adoptive The improved clinical results from lymphodeple-
T-cell transfer remain to be defined, three ­non-mutually tion in the context of ACT, combined with the recent
exclusive models have been proposed. In one scenario, discovery and characterization of diverse regulatory
prior depletion of lymphocytes may create “space” for immune cell subsets, provided new strategies for
the adoptively transferred cells within the lymphocyte tumour immunotherapy. These are based on the very
compartment. In this model, homeostatic mechanisms important point that immunoregulatory mechanisms
result in increased proliferation and enhanced survival that suppress anti-tumour responses need to be iden-
of the transferred T cells, largely through increased tified and addressed in a more efficacious manner. It
access to endogenous common-chain cytokines, such probably will not matter how many effector T cells we
as IL-2, IL-7, and IL-15.23,192 can generate or how efficiently they can kill tumour
Myeloablative lymphodepletion regimens involv- cells in vitro if they are being totally suppressed in vivo.
ing total body irradiation also induce superior anti-tu- Thus, it is becoming apparent that by understanding
mour responses when combined with adoptive T-cell the regulatory complexicity of the immune system,
transfer, as shown in both human cancer patients one can begin to design truly rational treatment strate-
and in animal models.192 Radiation induces damage gies designed to shift the balance in favour of immune
to the gut, leading to the translocation of microbes activation.
178   C. N. Baxevanis et al.

Cancer immunotherapy using adoptive transfer of NKT cells increased in two out of three cases, and
in vitro-activated NKT cells augmentation of IFN--producing cells in PBMCs was
detected by an ELISPOT assay. Intracellular cytokine
Lipid antigens bind to CD1 molecules with their staining showed the majority of IFN- producing cells
alkyl chains buried in hydrophobic pockets and detected after activated NKT cell administration to be
expose their polar lipid headgroup whose fine struc- CD3- CD56+ NK cells, but not CD3+ cells. Therefore,
ture is recognized by the TCR of CD1-restricted NKT -GalCer-activated V24 NKT cells appeared to sub-
cells.200–202 CD1-restricted T cells carry out effector, sequently stimulate NK cells to produce IFN-.215
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

helper, and adjuvant-like functions and interact Clinical trials aiming at endogenous activation
with other cell types, including macrophages, DC, of NKT cells have also been reported. In a phase
NK cells, T cells, and B cells, thereby contributing to I study, Giaccone et  al.217 administered KRN7000
both innate and adaptive immune responses.203–205 intravenously in 24 patients with solid tumours.
Insights gained from mice and humans now deline- No dose-limiting toxicity was observed over a wide
ate the extensive range of diseases in which CD1- range of doses (50–4800 µg/m2). Immunomonitoring
restricted T cells play important roles and reveal demonstrated that NKT cells (CD3+V24+Vß11+)
differences in the role of CD1a, CD1b, and CD1c typically disappeared from the blood within 24 hours
in contrast to CD1d.206 Invariant TCR alpha chains of KRN7000 injection. Additional biological effects
(V24JQ gene segment in humans and V14J281 included increased serum cytokine levels (TNF- and
in mice), self-lipid reactivity, and rapid effector GM-CSF) in five of 24 patients and a transient decrease
responses uncover a subset of CD1d-restricted T in peripheral blood NK cell numbers and cytotoxic-
cells (NKT cells) having unique effector functions ity in seven of 24 patients. Importantly, the observed
without a counterpart among MHC-restricted T biological effects depended on pre-treatment NKT-
cells. cell numbers rather than on the dose of KRN7000.
Changes in V24+Vb11+ NKT cell number and Pre-treatment NKT-cell numbers were significantly
For personal use only.

function are associated with human autoimmune lower in patients compared with healthy controls
diseases and cancer.206–209 Restoration of this cor- (P = 0.0001). No clinical responses were recorded,
responding NKT cell population in mice or in vivo and seven patients experienced stable disease for a
activation with alpha-galactosylceramide (-GalCer; median duration of 123 days.
KRN7000) can prevent or reduce tumour growth and n preclinical studies, several laboratories have
autoimmunity.210–212 Although the therapeutic value demonstrated that -GalCer loaded onto DC is more
of these NKT cells in man remains to be determined, efficient than soluble -GalCer in stimulating activa-
large numbers of functional antigen-specific NKT tion and expansion of endogenous anti-tumour NKT
cells can be expanded in vitro. V24+V11+ human cells.218,219 In a phase I study,220 -GalCer-pulsed DC
NKT cells can be expanded continuously by repeated were intravenously administered in 11 lung cancer
stimulation with KRN7000, from unfractionated patients at escalating doses. No severe adverse events
donor peripheral blood mononuclear cells (PBMC), were observed during this study in any patient. After
in the presence of IL-2 for more than two months the first and second injection of -GalCer-pulsed
with a potential yield of >1012 cells.213 Expanded NKT DC, a dramatic increase in peripheral blood Va24
cells retain their CD4+ or CD4- phenotype after res- NKT cells was observed in one case, and significant
timulation and are functional as shown by cytokine responses were seen in two cases. No patient was
secretion, killing of antigen-pulsed target cells, and found to meet the criteria for partial or complete
activation of NK cell cytotoxicity.213 n vitro expanded responses, whereas two cases remained unchanged
V24 NKT cells show substantial cytotoxicity against for more than a year with good quality of life.
tumour target cells.213,214 Nieda et  al.221 designed a phase I study to treat
Adoptive transfer of in vitro activated NKT cells patients having metastatic malignancies with
was performed in patients with lung cancer who -GalCer-pulsed CD1d-expressing DC. The results
were refractory to standard treatment.215 Intravenous showed that this therapy had substantial, rapid, and
injection of activated V24 NKT cells was car- highly reproducible specific effects on V24+V11+
ried out to test immunological, safety, and clinical NKT cells and provided the first human in vivo evi-
responses. Transferred V24 NKT cells, co-cultured dence that V24+V11+ NKT cell stimulation leads
with -GalCer and IL-2 for two or three weeks, mostly to activation of both innate and acquired immu-
expressed either double negative or CD8+ NKT cells, nity, resulting in modulation of NK, T-, and B-cell
which have been reported to produce Th1-type numbers and increased serum IFN. Uchida et al.222
cytokines.216 After two administrations of activated conducted a phase I study with -GalCer-pulsed
V24 NKT cells, the circulating V24 antigen presenting cells (APCs) administered to the
Cancer immunotherapy   179

nasal submucosa of 11 patients with head and neck IL-21) as part of vaccination strategies has been suc-
cancer and evaluated the safety and feasibility of such cessfully tested in preclinical cancer models.224,236,237
a treatment. During the clinical study period, no seri- Human NK cells discriminate between differ-
ous adverse events were observed. Following the first ent allelic forms of MHC molecules via killer cell
and second administration of a-GalCer-pulsed APCs, ­immunoglobulin-like receptors (KIRs), which are
an increased number of NKT cells were observed in clonally distributed, and each cell in the repertoire
four patients, and enhanced natural killer activity was bears at least one receptor that is specific for self-MHC
detected in the peripheral blood of eight patients. class I molecules.238 Consequently, when faced with
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

Although the objective anti-tumour response mismatched allogeneic targets, NK cells in the reper-
rate remains low at present, continuous refine- toire will sense the missing expression of self-MHC
ment to augment the cytolytic activity is needed, class I alleles and will mediate alloreactions.239,240
without losing the safety profile that is the marked Donor versus recipient NK cell alloreactivity derives
advantage of cancer immunotherapy. Considering from a mismatch between donor NK clones (carrying
the mechanisms of the anti-tumour effect via NKT specific inhibitory receptors for self-MHC class I mol-
cells, the development of a combination therapy ecules) and MHC class I ligands on recipient cells.
seems quite reasonable, since the NKT cells show an When faced with mismatched allogeneic targets, these
adjuvant effect and modulate other immune effec- donor NK clones sense the missing expression of self-
tor cells. Combinations with mAb-based immuno- HLA class I alleles and mediate alloreactions.241,242
therapy may be interesting additional approaches Transplantation from NK alloreactive haploidenti-
to investigate. As previously mentioned, other rea- cal donors controls acute myeloid leukemia (AML)
gents activating distinct cell populations that pro- relapse and improves engraftment without causing
mote adaptive immunity, such as peptide vaccine graft versus host disease (GvHD).243 The effectiveness
to activate tumour antigen-specific CD8+ T cells, of NK-cell alloreactivity, as revealed in the haploi-
might also be useful for activating tumour-specific dentical transplant setting, has led to the establish-
For personal use only.

immunity in combination with NKT cell-activation ment of specific criteria for donor selection that have
immunotherapy. enhanced survival rates of leukaemia patients,243–245
These results will encourage extending the use of
mismatched transplants to more leukaemia patients
Clinical application of NK cells without a matched donor. Other developments
might include designing protocols that will facilitate
NK cells kill many tumour cell lines and may also exploitation of NK-cell alloreactivity in the setting of
play a critical role in anti-tumour immunity.223,224 unrelated donor transplantation. Perspectives from
For tumours with low-to-undetectable MHC mol- murine models suggest that alloreactive NK cells
ecule expression, NK cells may be the predominant might become part of the conditioning regimen, as
immune effector mechanism of defence.225 NK cells they may potentiate the graft versus leukaemia effect
can also kill tumour cells that express normal levels of of the transplant,243–246 help engraftment, and protect
MHC class I through interaction between activation against T-cell–mediated GvHD. They might also be
of natural cytotoxicity receptors and specific MHC- infused post-transplant to help prevent or control
like ligands on target cells.226,227 In addition to direct leukaemia relapse.
cytotoxic activity against malignant cells, NK cells are Studies are currently seeking to apply donor allore-
believed to play a crucial role in the early phase of active NK cells beyond the field of transplantation as
adaptive immunity engagement against both infec- adoptive immunotherapy for cancer. One can even
tious agents and tumour cells, thus providing a key envisage ways to overcome autologous NK-cell inhi-
link between innate and adaptive immunity.228,229 In bition by self-HLA molecules to direct autologous
particular, NK cell-mediated tumour killing appears NK cells against tumours, thus obviating the need for
to elicit an efficient T-cell response, likely by inducing immune suppression and/or allogeneic transplanta-
DC maturation.230,231 IL-2, a powerful stimulator of NK tion. A study of relapsing acute myeloid leukemia
cell cytotoxic activity, has been successfully combined patients has demonstrated that the infusion of NK cells
with different vaccination regimens in humans,232–234 from haploidentical donors is safe. Cells expanded
but the relative contribution to the anti-tumour effect in vivo after lymphopenia was induced by high-dose
is difficult to determine and has been rarely investi- cyclophosphamide and fludarabine. Interestingly,
gated.235 Moreover, the toxicity associated with the remissions were observed when potentially alloreactive
systemic administration of IL-2 has hampered the KIR-ligand mismatched donors were used.247 Another
clinical utilization of this cytokine. The administra- report shows that NK-cell overexpression of activating
tion of other NK cell activators (e.g. IL-12, IL-15, IL-18, receptors overcomes the inhibitory signals delivered
180   C. N. Baxevanis et al.

through receptors for HLA molecules and enhances Conclusion


NK killing of NK-resistant leukaemia targets.101
The limiting factor in clinical protocols utilizing A plethora of knowledge has been obtained from pre-
NK cells for adoptive transfer remains the number of clinical studies and clinical trials on the capacity of
appropriately activated NK that can be obtained.248 the immune system to develop anti-cancer responses
Usually the donor undergoes a four-to-five hour leu- and the multiple pathways through which immuno-
kapheresis, which is a protracted and cumbersome therapeutic modalities can boost a patient’s immune
procedure. We have recently reported249 novel data system to fight its own tumour. Further elucidating
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

on the effect of glucocorticoids (GCs), along with the molecular mechanisms underlying interactions
stimulatory signals from IL-15 or IL-2, on NK cell between tumours and the immune system effectors
expansion and function. Although GCs have been will re-enforce our investigational capacity to devise
reported to exert an inhibitory effect on NK cells, novel immunotherapeutic modalities, which will
we have demonstrated that NK cells activated with prove to be more efficacious. It is also obvious that
IL-15 (or IL-2) in the presence of hydrocortisone the mechanisms underlying anti-tumour responses
(HC) not only retain their cytotoxic activity, but are need to be better delineated in pre-clinical models
also protected from apoptosis, and their prolifera- and clinical trials. There are important conclusions to
tive rate is significantly elevated. HC plus IL-15 is be drawn from our current knowledge, and thus the
capable of inducing up to a 400-fold expansion of future of cancer immunotherapy is likely to include
human NK cells within 20 days of culture compared the following strategies.
to about a 50-fold with IL-15 alone, thus making Increasing bodies of evidence suggest that cyto-
feasible their large-scale production from a limited toxic drugs positively influence immune responses
volume of peripheral blood ( 200 mL of peripheral against tumours. This “side” effect on tumour immu-
blood could give rise to more than 109 NK cells nology modifies the clinical response to chemother-
using this protocol) and their potential application apy. An emerging modified modality suggests that
For personal use only.

in clinical protocols. Furthermore, expanding NK therapeutic vaccination increases clinical responses


cells in the presence of HC, thus rendering them to subsequent salvage chemotherapy. Thus, the
pre-conditioned to the presence of GCs, is advanta- combination of vaccination and chemotherapy may
geous, since their intravenous injection into a recip- provide substantial clinical benefits for patients with
ient exposes them to a microenvironment where advanced cancer. Chemotherapy may also improve
active cortisol is naturally present at doses similar immune-mediated destruction of tumours by facili-
to those used in vitro. tating the induction of an immune response against
Autologous NK cells, activated and expanded in tumour stroma.
vitro in the presence of IL-15 and HC, have been Targeting tumour stroma as an immunotherapeutic
found to be effective in vivo in a lung metastasis strategy may be very effective. Radiotherapy can also
mouse model. These NK cells retained their ability be combined with vaccination strategies to improve
to efficiently infiltrate tumour-bearing lung tissue their effectiveness so long as attention is paid to dos-
and to persist for more than three days after NK cell ages and the timing of each treatment.
infusion. Furthermore, survival of animals with pre- Therapeutic vaccination in late disease is at
established lung tumours, treated with NK cells acti- present very ineffective, although we might be able
vated and expanded with IL-15 and HC, was more to see clinical responses in the face of multiepitope
than six-fold extended compared to untreated ani- vaccines (synthetic long peptides and naturally
mals (unpublished data). We have initiated a phase occurring polypeptides) incorporating various CTL
I clinical study in patients with advanced NSCLC, and Th epitopes. Polypeptides will be exlusively
who are adoptively transferred with allogeneic NK processed by DC, which will then trigger robust
cells expanded ex vivo with IL-2 or IL-15 and HC, anti-tumour responses. This effect can be further
in order to take advantage of both, i.e. the novel NK enhanced by the use of DC-activating molecules
cell-expansion protocol to obtain large numbers such as TLR ligands.
of appropriate effector cells and the KIR mismatch Targeted immunotherapies with anti-tumour
between donor and recipient. The selection of lung mAbs either as monotherapy or in combination with
cancer is based on the observation that the majority chemotherapy or vaccines have shown great promise
of intravenously administered NK cells accumulate in advanced cancers. mAbs exert their effects either by
in the lung and are retained at the tumour site.250 This inducing tumour cell apoptosis/necrosis or by form-
protocol has been applied so far in 15 patients with ing immune complexes with soluble tumour antigens,
no toxic effects. which, in this way, will be processed and presented by
Cancer immunotherapy   181

DC more quickly (i.e. through Fc receptor phagocyto- altering such mechanisms. This progress in cancer
sis). Moreover, mAbs may act to potentiate anti-tumour immunology will in the near future introduce elegant
immunity by depleting regulatory T cells, triggering combinatorial treatments involving targeting drugs
costimulatory molecules, or blocking coinhibition. with novel immunotherapeutic strategies aiming at
As a general option, immunotherapy intervention immunostimulation with simultaneous elimination of
requires tumour debulking and, therefore, should tumour-induced suppressive mechanisms. These new
be combined with surgery and chemotherapy. therapies in cancer are expected to induce improved
Immunotherapeutic approaches should be tested in clinical responses and, eventually, cancer prevention.
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

patients whose tumours have been resected and who


are receiving adjuvant chemotherapy, but are at high Declaration of interest: The authors report no con-
risk for recurrence. Tumour debulking will greatly flicts of interest. The authors alone are responsible for
diminish immunosuppressive mechanisms that may the content of this paper.
otherwise be induced by the cancer.
Last, and most important, there is growing evidence
pointing to the value of vaccinating at early stages of References
cancer progression. Such preventive vaccines, when
1. Kirkwood J. Cancer immunotherapy: the interferon-alpha
applied in patients with cancer at early stages, when experience. Semin Oncol 2002; 29: 18–26.
the tumour load is low enough to establish a local and 2. Boyiadzis M, Foon KA. Approved monoclonal antibodies for
systemic immunosuppressive network, may induce cancer therapy. Expert Opin Biol Ther 2008; 8: 1151–1158.
efficient anti-tumour responses and enhanced clini- 3. Baxevanis CN, Papamichail M. Targeting of tumor cells by
cal benefit. Advances in early cancer detection meth- lymphocytes engineered to express chimeric receptor genes.
Cancer Immunol Immunother 2004; 53: 893–903.
ods and the development of efficacious cancer vac- 4. Nimjee SM, Rusconi CP, Sullenger BA. Aptamers: an emerg-
cines will allow vaccination of patients with various ing class of therapeutics. Annu Rev Med 2005; 56: 555–583.
types of cancer at early stages of disease, when they 5. van der Burg SH, Bijker MS, Welters MJ, Offringa R, Melief CJ.
For personal use only.

still having intact immune systems. This may become Improved peptide vaccine strategies, creating synthetic arti-
the most promising strategy for preventing tumour ficial infections to maximize immune efficacy. Adv Drug
Deliv Rev 2006; 58: 916–930.
progression. Currently, realization of preventive vac- 6. Zitvogel L, Apetoh L, Ghiringhelli F, Kroemer G.
cination will be feasible in patients with certain types Immunological aspects of cancer chemotherapy. Nat Rev
of cancer (e.g. breast, prostate, ovary, and colon) who Immunol 2008; 8: 59–73.
are free of disease after treatment with conventional 7. Ramakrishnan R, Antonia S, Gabrilovich DI. Combined
therapy, but at high risk of recurrence. It is prob- modality immunotherapy and chemotherapy: a new perspec-
tive. Cancer Immunol Immunother 2008; 57: 1523–1529.
able that an effective preventive peptide-vaccination 8. Rosenberg SA, Packard BS, Aebersold PM, Solomon D,
modality will require polyepitope vaccines, combined Topalian SL, Toy ST, Simon P, Lotze MT, Yang JC, Seipp CA
with strong adjuvants (e.g. TLR agonists) and immu- et al. Use of tumor-infiltrating lymphocytes and interleukin-2
nomodulatory mAbs. Phase I/II studies assessing in the immunotherapy of patients with metastatic melanoma.
preventive combined immunotherapy will definitely A preliminary report. N Engl J Med 1988; 319: 1676–1680.
9. Morgan RA, Dudley ME, Wunderlich JR, Hughes MS,
provide the platform for designing more efficacious Yang JC, Sherry RM, Royal RE, Topalian SL, Kammula US,
protocols for cancer therapy. Restifo NP, Zheng Z, Nahvi A, de Vries CR, Rogers-
However, autoimmunity is a possible side effect Freezer LJ, Mavroukakis SA, Rosenberg SA. Cancer regres-
of cancer immunotherapy in those instances when sion in patients after transfer of genetically engineered
intense immunotherapy is necessary, and thus the ­lymphocytes. Science 2006; 314: 126–129.
10. Wolf E, Hofmeister R, Kufer P, Schlereth B, Baeuerle PA.
incidence of autoimmunity in early or especially in BiTEs: bispecific antibody constructs with unique anti-tu-
advanced cancer should be evaluated in the short or mor activity. Drug Discov Today 2005; 10: 1237–1244.
long term following immunotherapy. 11. Kirkwood JM, Ernstoff M. Melanoma: therapeutic options
A large body of evidence has been gathered about with recombinant interferons. Semin Oncol 1985; 12: 7–12.
the potential of the immune system to control tumour 12. Kirkwood JM, Ernstoff MS, Davis CA, Reiss M, Ferraresi R,
Rudnick SA. Comparison of intramuscular and intravenous
promotion and the various pathways through which recombinant alpha-2 interferon in melanoma and other
immunotherapy can induce robust immune responses cancers. Ann Intern Med 1985; 103: 32–36.
for fighting cancer. Such knowledge has stimulated the 13. Kirkwood JM, Strawderman MH, Ernstoff MS, Smith TJ,
invention of novel immunotherapeutic modalities, Borden EC, Blum RH. Interferon alfa-2b adjuvant therapy
including therapeutic antibodies, vaccines, and cell- of high-risk resected cutaneous melanoma: the Eastern
Cooperative Oncology Group Trial EST 1684. J Clin Oncol
based treatments. Moreover, improvements in our 1996; 14: 7–17.
understanding of antiapoptotic and immune escape 14. Kirkwood JM, Ibrahim JG, Sondak VK, Richards J, Flaherty LE,
mechanisms have resulted in the discovery of biologi- Ernstoff MS, Smith TJ, Rao U, Steele M, Blum RH. High- and
cally active drugs capable of specifically targeting and low-dose interferon alfa-2b in high-risk melanoma: first
182   C. N. Baxevanis et al.

analysis of intergroup trial E1690/S9111/C9190. J Clin Oncol 29. Melchionda F, Fry TJ, Milliron MJ, McKirdy MA, Tagaya Y,
2000; 18: 2444–2458. Mackall CL. Adjuvant IL-7 or IL-15 overcomes immunodo-
15. Kirkwood JM, Ibrahim JG, Sosman JA, Sondak VK, minance and improves survival of the CD8+ memory cell
Agarwala  SS, Ernstoff MS, Rao U. High-dose interferon pool. J Clin Invest 2005; 115: 1177–1187.
alfa-2b significantly prolongs relapse-free and overall 30. Rosenberg SA, Sportes C, Ahmadzadeh M, Fry TJ, Ngo LT,
survival compared with the GM2-KLH/QS-21 vaccine in Schwarz SL, Stetler-Stevenson M, Morton KE, Mavroukakis
patients with resected stage IIB-III melanoma: results of SA, Morre M, Buffet R, Mackall CL, Gress RE. IL-7 admin-
intergroup trial E1694/S9512/C509801. J Clin Oncol 2001; istration to humans leads to expansion of CD8+ and CD4+
19: 2370–2380. cells but a relative decrease of CD4+ T-regulatory cells.
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

16. Kirkwood JM, Bender C, Agarwala S, Tarhini A, Shipe- J Immunother 2006; 29: 313–319.
Spotloe J, Smelko B, Donnelly S, Stover L. Mechanisms and 31. Sportes C, Hakim FT, Memon SA, Zhang H, Chua KS,
management of toxicities associated with high-dose inter- Brown MR, Fleisher TA, Krumlauf MC, Babb RR, Chow CK,
feron alfa-2b therapy. J Clin Oncol 2002;. 20: 3703–3718. Fry TJ, Engels J, Buffet R, Morre M, Amato RJ, Venzon DJ,
17. Smith KA. Interleukin-2: inception, impact, and implica- Korngold R, Pecora A, Gress RE, Mackall CL. Administration
tions. Science 1988; 240: 1169–1176. of rhIL-7 in humans increases in vivo TCR repertoire diver-
18. Atkins MB, Kunkel L, Sznol M, Rosenberg SA. High-dose sity by preferential expansion of naive T cell subsets. J Exp
recombinant interleukin-2 therapy in patients with meta- Med 2008; 205: 1701–1714.
static melanoma: long-term survival update. Cancer J Sci 32. Burton JD, Bamford RN, Peters C, Grant AJ, Kurys G,
Am 2000; 6(Suppl 1): S11–S14. Goldman CK, Brennan J, Roessler E, Waldmann TA. A lym-
19. Atkins MB, Lotze MT, Dutcher JP, Fisher RI, Weiss G, phokine, provisionally designated interleukin T and produced
Margolin  K, Abrams J, Sznol M, Parkinson D, Hawkins M, by a human adult T-cell leukemia line, stimulates T-cell pro-
Paradise C, Kunkel L, Rosenberg SA. High-dose recom- liferation and the induction of lymphokine-activated killer
binant interleukin 2 therapy for patients with metastatic cells. Proc Natl Acad Sci USA 1994; 91: 4935–4939.
melanoma: analysis of 270 patients treated between 1985 33. Burkett PR, Koka R, Chien M, Chai S, Boone DL, Ma A.
and 1993. J Clin Oncol 1999; 17: 2105–2116. Coordinate expression and trans presentation of inter-
20. Dutcher JP, Creekmore S, Weiss GR, Margolin K, leukin (IL)-15Ralpha and IL-15 supports natural killer cell
Markowitz AB, Roper M, Parkinson D, Ciobanu N, Fisher RI, and memory CD8+ T cell homeostasis. J Exp Med 2004; 200:
Boldt DH et  al. A phase II study of interleukin-2 and lym- 825–834.
phokine-activated killer cells in patients with metastatic 34. Dubois S, Mariner J, Waldmann TA, Tagaya Y. IL-15Ralpha
For personal use only.

malignant melanoma. J Clin Oncol 1989; 7: 477–485. recycles and presents IL-15 in trans to neighboring cells.
21. Parkinson DR, Abrams JS, Wiernik PH, Rayner AA, Immunity 2002; 17: 537–547.
Margolin KA, Van Echo DA, Sznol M, Dutcher JP, Aronson FR, 35. Tan JT, Ernst B, Kieper WC, LeRoy E, Sprent J, Surh CD.
Doroshow JH et  al. Interleukin-2 therapy in patients with Interleukin (IL)-15 and IL-7 jointly regulate homeostatic
metastatic malignant melanoma: a phase II study. J Clin proliferation of memory phenotype CD8+ cells but are not
Oncol 1990; 8: 1650–1656. required for memory phenotype CD4+ cells. J Exp Med 2002;
22. Schluns KS, Kieper WC, Jameson SC, Lefrancois L. 195: 1523–1532.
Interleukin-7 mediates the homeostasis of naive and mem- 36. Becker TC, Wherry EJ, Boone D, Murali-Krishna K, Antia R,
ory CD8 T cells in vivo. Nat Immunol 2000; 1: 426–432. Ma A, Ahmed R. Interleukin 15 is required for proliferative
23. Fry TJ, Mackall CL. Interleukin-7: master regulator of renewal of virus-specific memory CD8 T cells. J Exp Med
peripheral T-cell homeostasis? Trends Immunol 2001; 22: 2002; 195: 1541–1548.
564–571. 37. Klebanoff CA, Gattinoni L, Torabi-Parizi P, Kerstann K,
24. Geiselhart LA, Humphries CA, Gregorio TA, Mou S, Cardones AR, Finkelstein SE, Palmer DC, Antony PA,
Subleski  J, Komschlies KL. IL-7 administration alters the Hwang ST, Rosenberg SA, Waldmann TA, Restifo NP. Central
CD4:CD8 ratio, increases T cell numbers, and increases T memory self/tumor-reactive CD8+ T cells confer superior
cell function in the absence of activation. J Immunol 2001; antitumor immunity compared with effector memory T
166: 3019–3027. cells. Proc Natl Acad Sci USA 2005; 102: 9571–9576.
25. Fry TJ, Moniuszko M, Creekmore S, Donohue SJ, Douek DC, 38. Sotiriadou NN, Perez SA, Gritzapis AD, Mahaira LG,
Giardina S, Hecht TT, Hill BJ, Komschlies K, Tomaszewski J, Salagianni M, Baxevanis CN, Papamichail M. Beneficial
Franchini G, Mackall CL. IL-7 therapy dramatically alters effect of short-term exposure of human NK cells to IL15/IL12
peripheral T-cell homeostasis in normal and SIV-infected and IL15/IL18 on cell apoptosis and function. Cell Immunol
nonhuman primates. Blood 2003; 101: 2294–2299. 2005; 234: 67–75.
26. Storek J, Gillespy T, 3rd, Lu H, Joseph A, Dawson MA, 39. Kobayashi H, Dubois S, Sato N, Sabzevari H, Sakai Y,
Gough  M, Morris J, Hackman RC, Horn PA, Sale GE, Waldmann TA, Tagaya Y. Role of trans-cellular IL-15 pres-
Andrews RG, Maloney DG, Kiem HP. Interleukin-7 improves entation in the activation of NK cell-mediated killing, which
CD4 T-cell reconstitution after autologous CD34 cell trans- leads to enhanced tumor immunosurveillance. Blood 2005;
plantation in monkeys. Blood 2003; 101: 4209–4218. 105: 721–727.
27. Fry TJ, Mackall CL. The many faces of IL-7: from lymphopoi- 40. Klebanoff CA, Finkelstein SE, Surman DR, Lichtman MK,
esis to peripheral T cell maintenance. J Immunol 2005; 174: Gattinoni L, Theoret MR, Grewal N, Spiess PJ, Antony  PA,
6571–6576. Palmer DC, Tagaya Y, Rosenberg SA, Waldmann TA,
28. Gattinoni L, Finkelstein SE, Klebanoff CA, Antony PA, Restifo NP. IL-15 enhances the in vivo antitumor activity of
Palmer  DC, Spiess PJ, Hwang LN, Yu Z, Wrzesinski C, tumor-reactive CD8+ T cells. Proc Natl Acad Sci USA 2004;
Heimann DM, Surh CD, Rosenberg SA, Restifo NP. Removal 101: 1969–1974.
of homeostatic cytokine sinks by lymphodepletion enhances 41. Teague RM, Sather BD, Sacks JA, Huang MZ, Dossett  ML,
the efficacy of adoptively transferred tumor-specific CD8+ T Morimoto J, Tan X, Sutton SE, Cooke MP, Ohlen C,
cells. J Exp Med 2005 202: 907–912. Greenberg PD. Interleukin-15 rescues tolerant CD8+ T cells
Cancer immunotherapy   183

for use in adoptive immunotherapy of established tumors. 58. Perez SA, Mahaira LG, Sotiropoulou PA, Gritzapis AD,
Nat Med 2006; 12: 335–341. Iliopoulou EG, Niarchos DK, Cacoullos NT, Kavalakis YG,
42. Yadav D, Sarvetnick N. Cytokines and autoimmunity: redun- Antsaklis AI, Sotiriadou NN, Baxevanis CN, Papamichail M.
dancy defines their complex nature. Curr Opin Immunol Effect of IL-21 on NK cells derived from different umbilical
2003; 15: 697–703. cord blood populations. Int Immunol 2006; 18: 49–58.
43. Kennedy R, Celis E. Multiple roles for CD4+ T cells in 59. Chen Z, Laurence A, O’Shea JJ. Signal transduction path-
anti-tumor immune responses. Immunol Rev 2008; 222: ways and transcriptional regulation in the control of Th17
129–144. differentiation. Semin Immunol 2007; 19: 400–408.
44. Baxevanis CN, Gritzapis AD, Papamichail M. In vivo anti- 60. Davis ID, Skak K, Smyth MJ, Kristjansen PE, Miller DM,
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

tumor activity of NKT cells activated by the combination of Sivakumar PV. Interleukin-21 signaling: functions in cancer
IL-12 and IL-18. J Immunol 2003; 171: 2953–2959. and autoimmunity. Clin Cancer Res 2007; 13: 6926–6932.
45. Smyth MJ, Thia KY, Street SE, Cretney E, Trapani JA, 61. Köhler G, Milstein C. Continuous cultures of fused cells
Taniguchi M, Kawano T, Pelikan SB, Crowe NY, Godfrey DI. secreting antibody of predefined specificity. Nature.
Differential tumor surveillance by natural killer (NK) and 1975;256(5517):495–497.
NKT cells. J Exp Med 2000; 191: 661–668. 62. Iannello A, Ahmad A. Role of antibody-dependent cell-me-
46. Noguchi Y, Jungbluth A, Richards EC, Old LJ. Effect of inter- diated cytotoxicity in the efficacy of therapeutic anti-cancer
leukin 12 on tumor induction by 3-methylcholanthrene. monoclonal antibodies. Cancer Metastasis Rev 2005; 24:
Proc Natl Acad Sci USA 1996; 93: 11798–11801. 487–499.
47. Nanni P, Nicoletti G, De Giovanni C, Landuzzi L, Di Carlo E, 63. Clynes R. Antitumor antibodies in the treatment of cancer:
Cavallo F, Pupa SM, Rossi I, Colombo MP, Ricci C, Astolfi A, Fc receptors link opsonic antibody with cellular immunity.
Musiani P, Forni G, Lollini PL. Combined allogeneic tumor Hematol Oncol Clin North Am. 2006;20(3):585–612.
cell vaccination and systemic interleukin 12 prevents mam- 64. Strome SE, Sausville EA, Mann D. A mechanistic perspective
mary carcinogenesis in HER-2/neu transgenic mice. J Exp of monoclonal antibodies in cancer therapy beyond target-
Med 2001; 194: 1195–1205. related effects. Oncologist 2007; 12: 1084–1095.
48. Colombo MP, Trinchieri G. Interleukin-12 in anti-tumor 65. Castillo J, Winer E, Quesenberry P. Newer monoclonal anti-
immunity and immunotherapy. Cytokine Growth Factor Rev bodies for hematological malignancies. Exp Hematol 2008;
2002; 13: 155–168. 36: 755–768.
49. Voest EE, Kenyon BM, O’Reilly MS, Truitt G, D’Amato  RJ, 66. Kreitman RJ. Immunotoxins for targeted cancer therapy.
Folkman J. Inhibition of angiogenesis in vivo by Aaps J 2006; 8: E532–E551.
For personal use only.

­interleukin 12. J Natl Cancer Inst 1995; 87: 581–586. 67. Reff ME, Heard C. A review of modifications to recombinant
50. Cebon J, Jager E, Shackleton MJ, Gibbs P, Davis ID, antibodies: attempt to increase efficacy in oncology applica-
Hopkins  W, Gibbs S, Chen Q, Karbach J, Jackson H, tions. Crit Rev Oncol Hematol 2001; 40: 25–35.
MacGregor DP, Sturrock S, Vaughan H, Maraskovsky E, 68. Weiner GJ. Monoclonal antibody mechanisms of action in
Neumann A, Hoffman E, Sherman ML, Knuth A. Two phase I cancer. Immunol Res 2007; 39: 271–278.
studies of low dose recombinant human IL-12 with Melan-A 69. Perez SA, Karamouzis MV, Skarlos DV, Ardavanis A,
and influenza peptides in subjects with advanced malignant Sotiriadou NN, Iliopoulou EG, Salagianni ML, Orphanos G,
melanoma. Cancer Immun 2003; 3: 7. Baxevanis CN, Rigatos G, Papamichail M. CD4+CD25+ regu-
51. Peterson AC, Harlin H, Gajewski TF. Immunization with latory T-cell frequency in HER-2/neu (HER)-positive and
Melan-A peptide-pulsed peripheral blood mononuclear HER-negative advanced-stage breast cancer patients. Clin
cells plus recombinant human interleukin-12 induces clini- Cancer Res 2007; 13: 2714–2721.
cal activity and T-cell responses in advanced melanoma. 70. Melero I, Hervas-Stubbs S, Glennie M, Pardoll DM, Chen L.
J Clin Oncol 2003; 21: 2342–2348. Immunostimulatory monoclonal antibodies for cancer ther-
52. Hamid O, Solomon JC, Scotland R, Garcia M, Sian S, Ye W, apy. Nat Rev Cancer 2007; 7: 95–106.
Groshen SL, Weber JS. Alum with interleukin-12 augments 71. Baxevanis CN, Perez SA, Papamichail M. Combinatorial
immunity to a melanoma peptide vaccine: correlation with treatments including vaccines, chemotherapy and mono-
time to relapse in patients with resected high-risk disease. clonal antibodies for cancer therapy. Cancer Immunol
Clin Cancer Res 2007; 13: 215–222. Immunother 2009; 58: 317–324.
53. Gollob JA, Mier JW, Veenstra K, McDermott DF, Clancy D, 72. Goodwin RG, Din WS, Davis-Smith T, Anderson DM,
Clancy M, Atkins MB. Phase I trial of twice-weekly intra- Gimpel  SD, Sato TA, Maliszewski CR, Brannan CI,
venous interleukin 12 in patients with metastatic renal cell Copeland NG, Jenkins NA et al. Molecular cloning of a ligand
cancer or malignant melanoma: ability to maintain IFN- for the inducible T cell gene 4-1BB: a member of an emerg-
gamma induction is associated with clinical response. Clin ing family of cytokines with homology to tumor necrosis fac-
Cancer Res 2000; 6: 1678–1692. tor. Eur J Immunol 1993; 23: 2631–2641.
54. Spolski R, Leonard WJ. The Yin and Yang of interleukin-21 73. Sica G, Chen L. Modulation of the immune response through
in allergy, autoimmunity and cancer. Curr Opin Immunol 4-1BB. Adv Exp Med Biol 2000; 465: 355–362.
2008; 20: 295–301. 74. Teft WA, Kirchhof MG, Madrenas J. A molecular perspective
55. Leonard WJ, Zeng R, Spolski R. Interleukin 21: a cytokine/ of CTLA-4 function. Annu Rev Immunol 2006; 24: 65–97.
cytokine receptor system that has come of age. J Leukoc Biol 75. Egen JG, Kuhns MS, Allison JP. CTLA-4: new insights into its
2008; 84: 348–356. biological function and use in tumor immunotherapy. Nat
56. Wilk E, Kalippke K, Buyny S, Schmidt RE, Jacobs R. New Immunol 2002; 3: 611–618.
aspects of NK cell subset identification and inference of 76. Peggs KS, Quezada SA, Korman AJ, Allison JP. Principles
NK cells’ regulatory capacity by assessing functional and and use of anti-CTLA4 antibody in human cancer immuno-
genomic profiles. Immunobiology 2008; 213: 271–283. therapy. Curr Opin Immunol 2006; 18: 206–213.
57. Ouyang W, Kolls JK, Zheng Y. The biological functions of T 77. Hodge JW, Chakraborty M, Kudo-Saito C, Garnett CT,
helper 17 cell effector cytokines in inflammation. Immunity Schlom J. Multiple costimulatory modalities enhance CTL
2008; 28: 454–467. avidity. J Immunol 2005; 174: 5994–6004.
184   C. N. Baxevanis et al.

78. Leach DR, Krummel MF, Allison JP. Enhancement of anti- 91. Hombach A, Wieczarkowiecz A, Marquardt T, Heuser C,
tumor immunity by CTLA-4 blockade. Science 1996; 271: Usai L, Pohl C, Seliger B, Abken H. Tumor-specific T cell
1734–1736. activation by recombinant immunoreceptors: CD3 zeta sig-
79. Hodge JW, Schlom J. Comparative studies of a retrovirus ver- naling and CD28 costimulation are simultaneously required
sus a poxvirus vector in whole tumor-cell vaccines. Cancer for efficient IL-2 secretion and can be integrated into one
Res 1999; 59: 5106–5111. combined CD28/CD3 zeta signaling receptor molecule.
80. Ribas A, Camacho LH, Lopez-Berestein G, Pavlov D, J Immunol 2001; 167: 6123–6131.
Bulanhagui CA, Millham R, Comin-Anduix B, Reuben JM, 92. Gritzapis AD, Mamalaki A, Kretsovali A, Papamatheakis  J,
Seja E, Parker CA, Sharma A, Glaspy JA, Gomez-Navarro J. Belimezi M, Perez SA, Baxevanis CN, Papamichail M.
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

Antitumor activity in melanoma and anti-self responses in a Redirecting mouse T hybridoma against human breast and
phase I trial with the anti-cytotoxic T lymphocyte-associated ovarian carcinomas: in vivo activity against HER-2/neu
antigen 4 monoclonal antibody CP-675,206. J Clin Oncol 2005; expressing cancer cells. Br J Cancer 2003; 88: 1292–1300.
23: 8968–8977. 93. Mamalaki A, Gritzapis AD, Kretsovali A, Belimezi M,
81. Ribas A, Hanson DC, Noe DA, Millham R, Guyot DJ, Papamatheakis J, Perez SA, Papamichail M, Baxevanis CN.
Bernstein SH, Canniff PC, Sharma A, Gomez-Navarro J. In vitro and in vivo antitumor activity of a mouse CTL hybri-
Tremelimumab (CP-675,206), a cytotoxic T lymphocyte doma expressing chimeric receptors bearing the single chain
associated antigen 4 blocking monoclonal antibody in clini- Fv from HER-2/neu- specific antibody and the gamma-chain
cal development for patients with cancer. Oncologist 2007; from Fc(epsilon) RI. Cancer Immunol Immunother 2003; 52:
12: 873–883. 513–522.
82. Taniguchi T, Matsui H, Fujita T, Takaoka C, Kashima N, 94. Eshhar Z. The T-body approach: redirecting T cells with anti-
Yoshimoto R, Hamuro J. Structure and expression of a cloned body specificity. Handb Exp Pharmacol 2008; 181: 329–342.
cDNA for human interleukin-2. Nature 1983; 302: 305–310. 95. Kershaw MH, Jackson JT, Haynes NM, Teng MW, Moeller M,
83. Small EJ, Tchekmedyian NS, Rini BI, Fong L, Lowy I, Hayakawa Y, Street SE, Cameron R, Tanner JE, Trapani JA,
Allison JP. A pilot trial of CTLA-4 blockade with human anti- Smyth MJ, Darcy PK. Gene-engineered T cells as a superior
CTLA-4 in patients with hormone-refractory prostate can- adjuvant therapy for metastatic cancer. J Immunol 2004; 173:
cer. Clin Cancer Res 2007; 13: 1810–1815. 2143–2150.
84. Tchekmedyian NS, Glaspy JA, Korman AJ. MDX-010 (human 96. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar  Z,
anti-CTLA4): a phase I trial in malignant melanoma. Proc Mavroukakis SA, White DE, Wunderlich JR, Canevari  S,
Am Soc Clin Oncol 2002; 21(abstr 56): 15a. Rogers-Freezer L, Chen CC, Yang JC, Rosenberg SA, Hwu P. A
For personal use only.

85. Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, phase I study on adoptive immunotherapy using gene-mod-
Seiden MV, Davis T, Henry-Spires R, MacRae S, Willman A, ified T cells for ovarian cancer. Clin Cancer Res 2006; 12:
Padera R, Jaklitsch MT, Shankar S, Chen TC, Korman  A, 6106–6115.
Allison JP, Dranoff G. Biologic activity of cytotoxic T lym- 97. Pinthus JH, Waks T, Malina V, Kaufman-Francis K,
phocyte-associated antigen 4 antibody blockade in pre- Harmelin  A, Aizenberg I, Kanety H, Ramon J, Eshhar Z.
viously vaccinated metastatic melanoma and ovarian Adoptive immunotherapy of prostate cancer bone lesions
carcinoma patients. Proc Natl Acad Sci USA 2003; 100: using redirected effector lymphocytes. J Clin Invest 2004;
4712–4717. 114: 1774–1781.
86. Maker AV, Yang JC, Sherry RM, Topalian SL, Kammula US, 98. Uherek C, Tonn T, Uherek B, Becker S, Schnierle B,
Royal RE, Hughes M, Yellin MJ, Haworth LR, Levy C, Allen T, Klingemann HG, Wels W. Retargeting of natural killer-cell
Mavroukakis SA, Attia P, Rosenberg SA. Intrapatient dose cytolytic activity to ErbB2-expressing cancer cells results in
escalation of anti-CTLA-4 antibody in patients with meta- efficient and selective tumor cell destruction. Blood 2002;
static melanoma. J Immunother 2006; 29: 455–463. 100: 1265–1273.
87. Hersh E, Weber J, Powderley J. Disease control and long term 99. Schirrmann T, Pecher G. Tumor-specific targeting of a cell
survival in chemotherapy naive patients with advanced line with natural killer cell activity by asialoglycoprotein
melanoma treated with ipilimumab (MDX-010) with or with- receptor gene transfer. Cancer Immunol Immunother 2001;
out dacarbazine. J Clin Oncol 2008; 26(abstr 9022): 488s. 50: 549–556.
88. Small EJ, Higano C, Tchekmedyian NS. Randomized phase II 100. Muller T, Uherek C, Maki G, Chow KU, Schimpf A, Klingemann
study comparing 4 monthly doses of ipilimumab (MDX-010) HG, Tonn T, Wels WS. Expression of a CD20-specific chimeric
as a single agent or in combination with a single dose of antigen receptor enhances cytotoxic activity of NK cells and
docetaxel in patients with hormone refractory prostate can- overcomes NK-resistance of lymphoma and leukemia cells.
cer. J Clin Oncol 2006; 24(abstr 4609): 243s. Cancer Immunol Immunother 2008; 57: 411–423.
89. Maker AV, Phan GQ, Attia P, Yang JC, Sherry RM, 101. Imai C, Iwamoto S, Campana D. Genetic modification of
Topalian SL, Kammula US, Royal RE, Haworth LR, Levy C, primary natural killer cells overcomes inhibitory signals and
Kleiner D, Mavroukakis SA, Yellin M, Rosenberg SA. induces specific killing of leukemic cells. Blood 2005; 106:
Tumor regression and autoimmunity in patients treated 376–383.
with cytotoxic T lymphocyte-associated antigen 4 block- 102. Pegram HJ, Jackson JT, Smyth MJ, Kershaw MH, Darcy PK.
ade and interleukin 2: a phase I/II study. Ann Surg Oncol Adoptive transfer of gene-modified primary NK cells can
2005; 12: 1005–1016. specifically inhibit tumor progression in vivo. J Immunol
90. Attia P, Phan GQ, Maker AV, Robinson MR, Quezado MM, 2008; 181: 3449–3455.
Yang JC, Sherry RM, Topalian SL, Kammula US, Royal RE, 103. Alvarez-Vallina L. Genetic approaches for antigen-selective
Restifo NP, Haworth LR, Levy C, Mavroukakis SA, Nichol G, cell therapy. Curr Gene Ther 2001; 1: 385–397.
Yellin MJ, Rosenberg SA. Autoimmunity correlates with 104. Ren-Heidenreich L, Lum LG. Life or death of T cells with
tumor regression in patients with metastatic melanoma antigen-specific receptors—using T cells for cancer adop-
treated with anti-cytotoxic T-lymphocyte antigen-4. J Clin tive immunotherapy/gene therapy. Curr Gene Ther 2001; 1:
Oncol 2005; 23: 6043–6053. 253–255.
Cancer immunotherapy   185

105. Patel SD, Moskalenko M, Smith D, Maske B, Finer MH, Eradication of tumors from a human colon cancer cell line
McArthur JG. Impact of chimeric immune receptor extracel- and from ovarian cancer metastases in immunodeficient
lular protein domains on T cell function. Gene Ther 1999; 6: mice by a single-chain Ep-CAM-/CD3-bispecific antibody
412–419. construct. Cancer Res 2005; 65: 2882–2889.
106. Hombach A, Schneider C, Sent D, Koch D, Willemsen RA, 122. Hammond SA, Lutterbuese R, Roff S, Lutterbuese P,
Diehl V, Kruis W, Bolhuis RL, Pohl C, Abken H. An entirely Schlereth B, Bruckheimer E, Kinch MS, Coats S, Baeuerle PA,
humanized CD3 zeta-chain signaling receptor that directs Kufer P, Kiener PA. Selective targeting and potent control of
peripheral blood t cells to specific lysis of carcinoembry- tumor growth using an EphA2/CD3-Bispecific single-chain
onic antigen-positive tumor cells. Int J Cancer 2000; 88: antibody construct. Cancer Res 2007; 67: 3927–3935.
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

115–120. 123. Ribas A. Update on immunotherapy for melanoma. J Natl


107. Hoogenboom HR, Marks JD, Griffiths AD, Winter G. Building Compr Canc Netw 2006; 4: 687–694.
antibodies from their genes. Immunol Rev 1992; 130: 41–68. 124. Bijker MS, Melief CJ, Offringa R, van der Burg SH. Design and
108. Levey DL, Srivastava PK. Alterations in T cells of cancer-bear- development of synthetic peptide vaccines: past, present
ers: whence specificity? Immunol Today 1996; 17: 365–368. and future. Expert Rev Vaccines 2007; 6: 591–603.
109. Zier K, Gansbacher B, Salvadori S. Preventing abnormali- 125. Kanodia S, Da Silva DM, Kast WM. Recent advances in strat-
ties in signal transduction of T cells in cancer: the promise egies for immunotherapy of human papillomavirus-induced
of cytokine gene therapy. Immunol Today 1996; 17: 39–45. lesions. Int J Cancer 2008; 122: 247–259.
110. Melisi D, Troiani T, Damiano V, Tortora G, Ciardiello F. 126. Pol S, Michel ML. Therapeutic vaccination in chronic
Therapeutic integration of signal transduction targeting hepatitis B virus carriers. Expert Rev Vaccines 2006; 5:
agents and conventional anti-cancer treatments. Endocr 707–716.
Relat Cancer 2004; 11: 51–68. 127. Gilboa E. DC-based cancer vaccines. J Clin Invest 2007; 117:
111. Dollins CM, Nair S, Sullenger BA. Aptamers in immuno- 1195–1203.
therapy. Hum Gene Ther 2008; 19: 443–450. 128. Cavallo F, Offringa R, van der Burg SH, Forni G, Melief CJ.
112. McNamara JO, Kolonias D, Pastor F, Mittler RS, Chen L, Vaccination for treatment and prevention of cancer in ani-
Giangrande PH, Sullenger B, Gilboa E. Multivalent 4-1BB mal models. Adv Immunol 2006; 90: 175–213.
binding aptamers costimulate CD8+ T cells and inhibit 129. Melief CJ, van der Burg SH. Immunotherapy of established
tumor growth in mice. J Clin Invest 2008; 118: 376–386. (pre)malignant disease by synthetic long peptide vaccines.
113. White RR, Sullenger BA, Rusconi CP. Developing aptamers Nat Rev Cancer 2008; 8: 351–360.
into therapeutics. J Clin Invest 2000; 106: 929–934. 130. Kim CJ, Dessureault S, Gabrilovich D, Reintgen DS,
For personal use only.

114. Ng EW, Shima DT, Calias P, Cunningham ET, Jr., Guyer DR, Slingluff  CL Jr. Immunotherapy for melanoma. Cancer
Adamis AP. Pegaptanib, a targeted anti-VEGF aptamer Control 2002; 9: 22–30.
for ocular vascular disease. Nat Rev Drug Discov 2006; 5: 131. Steinman RM, Mellman I. Immunotherapy: bewitched,
123–132. bothered, and bewildered no more. Science 2004; 305:
115. Santulli-Marotto S, Nair SK, Rusconi C, Sullenger B, Gilboa E. 197–200.
Multivalent RNA aptamers that inhibit CTLA-4 and enhance 132. Sutmuller RP, van Duivenvoorde LM, van Elsas A,
tumor immunity. Cancer Res 2003; 63: 7483–7489. Schumacher TN, Wildenberg ME, Allison JP, Toes RE,
116. Dollins CM, Nair S, Boczkowski D, Lee J, Layzer JM, Gilboa E, Offringa R, Melief CJ. Synergism of cytotoxic T lymphocyte-
Sullenger BA. Assembling OX40 aptamers on a molecular associated antigen 4 blockade and depletion of CD25(+) reg-
scaffold to create a receptor-activating aptamer. Chem Biol ulatory T cells in antitumor therapy reveals alternative path-
2008; 15: 675–682. ways for suppression of autoreactive cytotoxic T lymphocyte
117. Offner S, Hofmeister R, Romaniuk A, Kufer P, Baeuerle PA. responses. J Exp Med 2001; 194: 823–832.
Induction of regular cytolytic T cell synapses by bispecific 133. Kirkwood JM, Tarhini AA, Panelli MC, Moschos SJ, Zarour
single-chain antibody constructs on MHC class I-negative HM, Butterfield LH, Gogas HJ. Next generation of immuno-
tumor cells. Mol Immunol 2006; 43: 763–771. therapy for melanoma. J Clin Oncol 2008; 26: 3445–3455.
118. Dreier T, Baeuerle PA, Fichtner I, Grun M, Schlereth B, 134. Mahnke K, Schonfeld K, Fondel S, Ring S, Karakhanova S,
Lorenczewski G, Kufer P, Lutterbuse R, Riethmuller G, Wiedemeyer K, Bedke T, Johnson TS, Storn V, Schallenberg S,
Gjorstrup P, Bargou RC. T cell costimulus-independent and Enk AH. Depletion of CD4+CD25+ human regulatory T
very efficacious inhibition of tumor growth in mice bear- cells in vivo: kinetics of Treg depletion and alterations in
ing subcutaneous or leukemic human B cell lymphoma immune functions in vivo and in vitro. Int J Cancer 2007;
xenografts by a CD19-/CD3- bispecific single-chain anti- 120: 2723–2733.
body construct. J Immunol 2003; 170: 4397–4402. 135. Rasku MA, Clem AL, Telang S, Taft B, Gettings K, Gragg H,
119. Hoffmann P, Hofmeister R, Brischwein K, Brandl C, Cramer D, Lear SC, McMasters KM, Miller DM, Chesney J.
Crommer S, Bargou R, Itin C, Prang N, Baeuerle PA. Serial Transient T cell depletion causes regression of melanoma
killing of tumor cells by cytotoxic T cells redirected with a metastases. J Transl Med 2008; 6: 12.
CD19-/CD3-bispecific single-chain antibody construct. Int J 136. Munn DH, Mellor AL. IDO and tolerance to tumors. Trends
Cancer 2005; 115: 98–104. Mol Med 2004; 10: 15–18.
120. Brischwein K, Schlereth B, Guller B, Steiger C, Wolf A, 137. Insinga A, Monestiroli S, Ronzoni S, Gelmetti V, Marchesi F,
Lutterbuese R, Offner S, Locher M, Urbig T, Raum T, Viale A, Altucci L, Nervi C, Minucci S, Pelicci PG. Inhibitors
Kleindienst P, Wimberger P, Kimmig R, Fichtner I, Kufer P, of histone deacetylases induce tumor-selective apoptosis
Hofmeister R, da Silva AJ, Baeuerle PA. MT110: a novel bis- through activation of the death receptor pathway. Nat Med
pecific single-chain antibody construct with high efficacy 2005; 11: 71–76.
in eradicating established tumors. Mol Immunol 2006; 43: 138. Gollob JA, Sciambi CJ. Decitabine up-regulates S100A2
1129–1143. expression and synergizes with IFN-gamma to kill uveal
121. Schlereth B, Fichtner I, Lorenczewski G, Kleindienst P, melanoma cells. Clin Cancer Res 2007; 13: 5219–5225.
Brischwein K, da Silva A, Kufer P, Lutterbuese R, Junghahn I, 139. Vlaykova T, Talve L, Hahka-Kemppinen M, Hernberg M,
Kasimir-Bauer S, Wimberger P, Kimmig R, Baeuerle PA. Muhonen T, Collan Y, Pyrhonen S. Immunohistochemically
186   C. N. Baxevanis et al.

detectable bcl-2 expression in metastatic melanoma: asso- 151. Wheeler CJ, Das A, Liu G, Yu JS, Black KL. Clinical respon-
ciation with survival and treatment response. Oncology siveness of glioblastoma multiforme to chemotherapy after
2002; 62: 259–268. vaccination. Clin Cancer Res 2004; 10: 5316–5326.
140. Maiuri MC, Le Toumelin G, Criollo A, Rain JC, Gautier F, 152. Sotiriadou NN, Kallinteris NL, Gritzapis AD, Voutsas IF,
Juin P, Tasdemir E, Pierron G, Troulinaki K, Tavernarakis N, Papamichail M, von Hofe E, Humphreys RE, Pavlis T,
Hickman JA, Geneste O, Kroemer G. Functional and physi- Perez SA, Baxevanis CN. Ii-Key/HER-2/neu(776-790) hybrid
cal interaction between Bcl-X(L) and a BH3-like domain in peptides induce more effective immunological responses
Beclin-1. Embo J 2007; 26: 2527–2539. over the native peptide in lymphocyte cultures from patients
141. Franco AV, Zhang XD, Van Berkel E, Sanders JE, Zhang XY, with HER-2/neu+ tumors. Cancer Immunol Immunother
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

Thomas WD, Nguyen T, Hersey P. The role of NF-kappa  B 2007; 56: 601–613.
in TNF-related apoptosis-inducing ligand (TRAIL)- 153. Bennett SR, Carbone FR, Karamalis F, Flavell RA, Miller JF,
induced apoptosis of melanoma cells. J Immunol 2001; 166: Heath WR. Help for cytotoxic-T-cell responses is mediated
5337–5345. by CD40 signalling. Nature 1998; 393: 478–480.
142. Sayers TJ, Brooks AD, Koh CY, Ma W, Seki N, Raziuddin A, 154. Schoenberger SP, Toes RE, van der Voort EI, Offringa R,
Blazar BR, Zhang X, Elliott PJ, Murphy WJ. The protea- Melief CJ. T-cell help for cytotoxic T lymphocytes is mediated
some inhibitor PS-341 sensitizes neoplastic cells to TRAIL- by CD40-CD40L interactions. Nature 1998; 393: 480–483.
mediated apoptosis by reducing levels of c-FLIP. Blood 2003; 155. Scheibenbogen C, Schadendorf D, Bechrakis NE,
102: 303–310. Nagorsen  D, Hofmann U, Servetopoulou F, Letsch A,
143. Peoples GE, Gurney JM, Hueman MT, Woll MM, Ryan GB, Philipp  A, Foerster  MH, Schmittel A, Thiel E, Keilholz U.
Storrer CE, Fisher C, Shriver CD, Ioannides CG, Ponniah S. Effects of granulocyte-macrophage colony-stimulating fac-
Clinical trial results of a HER2/neu (E75) vaccine to prevent tor and foreign helper protein as immunologic adjuvants on
recurrence in high-risk breast cancer patients. J Clin Oncol the T-cell response to vaccination with tyrosinase peptides.
2005; 23: 7536–7545. Int J Cancer 2003; 104: 188–194.
144. Holmes JP, Benavides LC, Gates JD, Carmichael MG, 156. Salucci V, Mennuni C, Calvaruso F, Cerino R, Neuner P,
Hueman  MT, Mittendorf EA, Murray JL, Amin A, Craig  D, Ciliberto G, La Monica N, Scarselli E. CD8+ T-cell tolerance
von Hofe E, Ponniah S, Peoples GE. Results of the first can be broken by an adenoviral vaccine while CD4+ T-cell
phase I clinical trial of the novel II-key hybrid preventive tolerance is broken by additional co-administration of a
HER-2/neu peptide (AE37) vaccine. J Clin Oncol 2008; 26: Toll-like receptor ligand. Scand J Immunol 2006; 63: 35–41.
3426–3433. 157. Ahonen CL, Doxsee CL, McGurran SM, Riter TR, Wade WF,
For personal use only.

145. Apostolopoulos V, Pietersz GA, Tsibanis A, Tsikkinis A, Barth RJ, Vasilakos JP, Noelle RJ, Kedl RM. Combined TLR
Drakaki H, Loveland BE, Piddlesden SJ, Plebanski M, and CD40 triggering induces potent CD8+ T cell expansion
Pouniotis DS, Alexis MN, McKenzie IF, Vassilaros S. Pilot with variable dependence on type I IFN. J Exp Med 2004;
phase III immunotherapy study in early-stage breast cancer 199: 775–784.
patients using oxidized mannan-MUC1 [ISRCTN71711835]. 158. Schwarz K, Storni T, Manolova V, Didierlaurent A, Sirard JC,
Breast Cancer Res 2006; 8: R27. Rothlisberger P, Bachmann MF. Role of Toll-like receptors
146. Machiels JP, Reilly RT, Emens LA, Ercolini AM, Lei RY, in costimulating cytotoxic T cell responses. Eur J Immunol
Weintraub D, Okoye FI, Jaffee EM. Cyclophosphamide, 2003; 33: 1465–1470.
doxorubicin, and paclitaxel enhance the antitumor immune 159. Zwaveling S, Ferreira Mota SC, Nouta J, Johnson M,
response of granulocyte/macrophage-colony stimulating Lipford  GB, Offringa R, van der Burg SH, Melief CJ.
factor-secreting whole-cell vaccines in HER-2/neu tolerized Established human papillomavirus type 16-expressing
mice. Cancer Res 2001; 61: 3689–3697. tumors are effectively eradicated following vaccination with
147. Arlen PM, Gulley JL, Parker C, Skarupa L, Pazdur M, long peptides. J Immunol 2002; 169: 350–358.
Panicali  D, Beetham P, Tsang KY, Grosenbach DW, 160. Kast WM, Brandt RM, Melief CJ. Strict peptide length is
Feldman J, Steinberg SM, Jones E, Chen C, Marte J, Schlom J, not required for the induction of cytotoxic T lymphocyte-
Dahut  W. A randomized phase II study of concurrent mediated antiviral protection by peptide vaccination. Eur J
docetaxel plus vaccine versus vaccine alone in metastatic Immunol 1993; 23: 1189–1192.
androgen-independent prostate cancer. Clin Cancer Res 161. Gao XM, Zheng B, Liew FY, Brett S, Tite J. Priming of influ-
2006; 12: 1260–1269. enza virus-specific cytotoxic T lymphocytes vivo by short
148. Gribben JG, Ryan DP, Boyajian R, Urban RG, Hedley  ML, synthetic peptides. J Immunol 1991; 147: 3268–3273.
Beach K, Nealon P, Matulonis U, Campos S, Gilligan  TD, 162. Minev BR, McFarland BJ, Spiess PJ, Rosenberg SA, Restifo NP.
Richardson PG, Marshall B, Neuberg D, Nadler LM. Insertion signal sequence fused to minimal peptides elicits
Unexpected association between induction of immunity to specific CD8+ T-cell responses and prolongs survival of thy-
the universal tumor antigen CYP1B1 and response to next moma-bearing mice. Cancer Res 1994; 54: 4155–4161.
therapy. Clin Cancer Res 2005; 11: 4430–4436. 163. Reinholdsson-Ljunggren G, Ramqvist T, Ahrlund-Richter L,
149. Maecker B, Sherr DH, Vonderheide RH, von Bergwelt- Dalianis T. Immunization against polyoma tumors with syn-
Baildon MS, Hirano N, Anderson KS, Xia Z, Butler MO, thetic peptides derived from the sequences of middle- and
Wucherpfennig KW, O’Hara C, Cole G, Kwak SS, Ramstedt U, large-T antigens. Int J Cancer 1992; 50: 142–146.
Tomlinson AJ, Chicz RM, Nadler LM, Schultze JL. The 164. Bijker MS, van den Eeden SJ, Franken KL, Melief CJ, van der
shared tumor-associated antigen cytochrome P450 1B1 is Burg SH, Offringa R. Superior induction of anti-tumor CTL
recognized by specific cytotoxic T cells. Blood 2003; 102: immunity by extended peptide vaccines involves prolonged,
3287–3294. DC-focused antigen presentation. Eur J Immunol 2008; 38:
150. Antonia SJ, Mirza N, Fricke I, Chiappori A, Thompson  P, 1033–1042.
Williams N, Bepler G, Simon G, Janssen W, Lee JH, Menander K, 165. Lefrancois L, Marzo A, Williams K. Sustained response initi-
Chada S, Gabrilovich DI. Combination of p53 cancer vaccine ation is required for T cell clonal expansion but not for effec-
with chemotherapy in patients with extensive stage small cell tor or memory development in vivo. J Immunol 2003; 171:
lung cancer. Clin Cancer Res 2006; 12: 878–887. 2832–2839.
Cancer immunotherapy   187

166. Obst R, van Santen HM, Melamed R, Kamphorst AO, papillomavirus 16 in end-stage cervical cancer patients
Benoist  C, Mathis D. Sustained antigen presentation shows low toxicity and robust immunogenicity. Clin Cancer
can promote an immunogenic T cell response, like den- Res 2008; 14: 169–177.
dritic cell activation. Proc Natl Acad Sci USA 2007; 104: 179. Rosenberg SA, Dudley ME. Cancer regression in patients
15460–15465. with metastatic melanoma after the transfer of autologous
167. Offringa R, van der Burg SH, Ossendorp F, Toes RE, Melief CJ. antitumor lymphocytes. Proc Natl Acad Sci USA 2004; 101:
Design and evaluation of antigen-specific vaccination strat- 14639–14645.
egies against cancer. Curr Opin Immunol 2000; 12: 576–582. 180. Surman DR, Dudley ME, Overwijk WW, Restifo NP. Cutting
168. Widmann C, Romero P, Maryanski JL, Corradin G, edge: CD4+ T cell control of CD8+ T cell reactivity to a model
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

Valmori D. T helper epitopes enhance the cytotoxic response tumor antigen. J Immunol 2000; 164: 562–565.
of mice immunized with MHC class I-restricted malaria 181. Touloukian CE, Leitner WW, Topalian SL, Li YF, Robbins PF,
peptides. J Immunol Methods 1992; 155: 95–99. Rosenberg SA, Restifo NP. Identification of a MHC class
169. Fayolle C, Deriaud E, Leclerc C. In vivo induction of cyto- II-restricted human gp100 epitope using DR4-IE transgenic
toxic T cell response by a free synthetic peptide requires mice. J Immunol 2000; 164: 3535–3542.
CD4+ T cell help. J Immunol 1991; 147: 4069–4073. 182. Palmer DC, Balasubramaniam S, Hanada K, Wrzesinski  C,
170. Weber JS, Hua FL, Spears L, Marty V, Kuniyoshi C, Celis E. A Yu  Z, Farid S, Theoret MR, Hwang LN, Klebanoff CA,
phase I trial of an HLA-A1 restricted MAGE-3 epitope peptide Gattinoni L, Goldstein AL, Yang JC, Restifo NP. Vaccine-
with incomplete Freund’s adjuvant in patients with resected stimulated, adoptively transferred CD8+ T cells traffic indis-
high-risk melanoma. J Immunother 1999; 22: 431–440. criminately and ubiquitously while mediating specific tumor
171. Ressing ME, van Driel WJ, Brandt RM, Kenter GG, de Jong JH, destruction. J Immunol 2004; 173: 7209–7216.
Bauknecht T, Fleuren GJ, Hoogerhout P, Offringa R, Sette A, 183. Leen AM, Rooney CM, Foster AE. Improving T cell therapy
Celis E, Grey H, Trimbos BJ, Kast WM, Melief CJ. Detection of for cancer. Annu Rev Immunol 2007; 25: 243–265.
T helper responses, but not of human papillomavirus-spe- 184. Greenberg PD, Cheever MA, Fefer A. Eradication of dis-
cific cytotoxic T lymphocyte responses, after peptide vac- seminated murine leukemia by chemoimmunotherapy with
cination of patients with cervical carcinoma. J Immunother cyclophosphamide and adoptively transferred immune
2000; 23: 255–266. syngeneic Lyt-1 + 2- lymphocytes. J Exp Med 1981; 154:
172. Disis ML, Gooley TA, Rinn K, Davis D, Piepkorn M, 952–963.
Cheever MA, Knutson KL, Schiffman K. Generation of T-cell 185. Greenberg PD, Cheever MA, Treatment of disseminated
immunity to the HER-2/neu protein after active immuniza- leukemia with cyclophosphamide and immune cells: tumor
For personal use only.

tion with HER-2/neu peptide-based vaccines. J Clin Oncol immunity reflects long-term persistence of tumor-specific
2002; 20: 2624–2632. donor T cells. J Immunol 1984; 133: 3401–3407.
173. Knutson KL, Schiffman K, Disis ML. Immunization with a 186. Croci DO, Zacarias Fluck MF, Rico MJ, Matar P, Rabinovich GA,
HER-2/neu helper peptide vaccine generates HER-2/neu Scharovsky OG. Dynamic cross-talk between tumor and
CD8 T-cell immunity in cancer patients. J Clin Invest 2001; immune cells in orchestrating the immunosuppressive
107: 477–484. network at the tumor microenvironment. Cancer Immunol
174. Beebe M, Qin M, Moi M, Wu S, Heiati H, Walker L, Newman M, Immunother 2007; 56: 1687–1700.
Fikes J, Ishioka GY. Formulation and Characterization 187. Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL,
of a Ten-peptide Single-vial Vaccine, EP-2101, Designed Restifo NP, Royal RE, Kammula U, White DE, Mavroukakis SA,
to Induce Cytotoxic T-lymphocyte Responses for Cancer Rogers LJ, Gracia GJ, Jones SA, Mangiameli DP, Pelletier MM,
Immunotherapy. Hum Vaccin. 2008; 4(3):37–45. Gea-Banacloche J, Robinson MR, Berman DM, Filie AC,
175. Barve M, Bender J, Senzer N, Cunningham C, Greco FA, Abati A, Rosenberg SA. Adoptive cell transfer therapy fol-
McCune D, Steis R, Khong H, Richards D, Stephenson J, lowing non-myeloablative but lymphodepleting chemo-
Ganesa P, Nemunaitis J, Ishioka G, Pappen B, Nemunaitis M, therapy for the treatment of patients with refractory meta-
Morse M, Mills B, Maples PB, Sherman J, Nemunaitis  JJ. static melanoma. J Clin Oncol 2005; 23: 2346–2357.
Induction of immune responses and clinical efficacy 188. Rapoport AP, Stadtmauer EA, Aqui N, Badros A, Cotte J,
in a phase II trial of IDM-2101, a 10-epitope cytotoxic Chrisley L, Veloso E, Zheng Z, Westphal S, Mair R, Chi N,
T-lymphocyte vaccine, in metastatic non-small-cell lung Ratterree B, Pochran MF, Natt S, Hinkle J, Sickles C, Sohal A,
cancer. J Clin Oncol 2008; 26: 4418–4425. Ruehle K, Lynch C, Zhang L, Porter DL, Luger S, Guo C,
176. Slingluff CL, Jr., Petroni GR, Olson W, Czarkowski A, Fang  HB, Blackwelder W, Hankey K, Mann D, Edelman R,
Grosh  WW, Smolkin M, Chianese-Bullock KA, Neese  PY, Frasch C, Levine BL, Cross A, June CH. Restoration of immu-
Deacon DH, Nail C, Merrill P, Fink R, Patterson JW, Rehm PK. nity in lymphopenic individuals with cancer by vaccination
Helper T-cell responses and clinical activity of a melanoma and adoptive T-cell transfer. Nat Med 2005; 11: 1230–1237.
vaccine with multiple peptides from MAGE and melanocytic 189. Gattinoni L, Powell DJ Jr, Rosenberg SA, Restifo NP. Adoptive
differentiation antigens. J Clin Oncol 2008; 26: 4973–4980. immunotherapy for cancer: building on success. Nat Rev
177. Welters MJ, Kenter GG, Piersma SJ, Vloon AP, Lowik MJ, Immunol 2006; 6: 383–393.
Berends-van der Meer DM, Drijfhout JW, Valentijn AR, 190. Wrzesinski C, Paulos CM, Gattinoni L, Palmer DC, Kaiser A,
Wafelman AR, Oostendorp J, Fleuren GJ, Offringa R, Yu Z, Rosenberg SA, Restifo NP. Hematopoietic stem cells
Melief  CJ, van der Burg SH. Induction of tumor-specific promote the expansion and function of adoptively trans-
CD4+ and CD8+ T-cell immunity in cervical cancer patients ferred antitumor CD8 T cells. J Clin Invest 2007; 1172:
by a human papillomavirus type 16 E6 and E7 long peptides 492–501.
vaccine. Clin Cancer Res 2008; 14: 178–187. 191. Muranski P, Boni A, Wrzesinski C, Citrin DE, Rosenberg SA,
178. Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends- Childs R, Restifo NP. Increased intensity lymphodepletion
van der Meer DM, Vloon AP, Drijfhout JW, Wafelman AR, and adoptive immunotherapy—how far can we go? Nat Clin
Oostendorp J, Fleuren GJ, Offringa R, van der Burg SH, Pract Oncol 2006; 3: 668–681.
Melief CJ. Phase I immunotherapeutic trial with long pep- 192. Paulos CM, Kaiser A, Wrzesinski C, Hinrichs CS, Cassard L,
tides spanning the E6 and E7 sequences of high-risk human Boni A, Muranski P, Sanchez-Perez L, Palmer DC, Yu Z,
188   C. N. Baxevanis et al.

Antony PA, Gattinoni L, Rosenberg SA, Restifo NP. Toll-like 208. Tahir SM, Cheng O, Shaulov A, Koezuka Y, Bubley GJ,
receptors in tumor immunotherapy. Clin Cancer Res 2007; Wilson SB, Balk SP, Exley MA. Loss of IFN-gamma produc-
13: 5280–5289. tion by invariant NK T cellsps in advanced cancer. J Immunol
193. Lizee G, Radvanyi LG, Overwijk WW, Hwu P. 2001; 167: 4046–4050.
Immunosuppression in melanoma immunotherapy: poten- 209. Taniguchi M, Harada M, Kojo S, Nakayama T, Wakao H.
tial opportunities for intervention. Clin Cancer Res 2006; 12: The regulatory role of Valpha14 NKT cells in innate and
2359s–2365s. acquired immune response. Annu Rev Immunol 2003; 21:
194. Sakaguchi S. Naturally arising Foxp3-expressing CD25+CD4+ 483–513.
regulatory T cells in immunological tolerance to self and 210. Linsen L, Somers V, Stinissen P. Immunoregulation of
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

non-self. Nat Immunol 2005; 6: 345–352. autoimmunity by natural killer T cells. Hum Immunol 2005;
195. Roncarolo MG, Bacchetta R, Bordignon C, Narula S, 66: 1193–1202.
Levings MK. Type 1 T regulatory cells. Immunol Rev 2001; 211. Yamamura T, Sakuishi K, Illes Z, Miyake S. Understanding
182: 68–79. the behavior of invariant NKT cells in autoimmune diseases.
196. Weiner HL. Induction and mechanism of action of trans- J Neuroimmunol 2007; 191: 8–15.
forming growth factor-beta-secreting Th3 regulatory cells. 212. Molling JW, Moreno M, de Groot J, van der Vliet HJ,
Immunol Rev 2001; 182: 207–214. von Blomberg BM, van den Eertwegh AJ, Scheper RJ,
197. Lizee G, Radvanyi LG, Overwijk WW, Hwu P. Improving anti- Bontkes HJ. Chronically stimulated mouse invariant NKT
tumor immune responses by circumventing immunoregu- cell lines have a preserved capacity to enhance protection
latory cells and mechanisms. Clin Cancer Res 2006; 12: against experimental tumor metastases. Immunol Lett
4794–4803. 2008; 118: 36–43.
198. Shvets A, Chakrabarti R, Gonzalez-Quintial R, Baccala R, 213. Rogers PR, Matsumoto A, Naidenko O, Kronenberg M,
Theofilopoulos AN, Prud’homme GJ. Impaired negative reg- Mikayama T, Kato S. Expansion of human Valpha24+ NKT
ulation of homeostatically proliferating T cells. Blood 2008; cells by repeated stimulation with KRN7000. J Immunol
133: 622–625. Methods 2004; 285: 197–214.
199. Powell DJ, Jr, de Vries CR, Allen T, Ahmadzadeh M. 214. Nishi N, van der Vliet HJ, Koezuka Y, von Blomberg BM,
Rosenberg  SA. Inability to mediate prolonged reduction Scheper RJ, Pinedo HM, Giaccone G. Synergistic effect of
of regulatory T Cells after transfer of autologous CD25- KRN7000 with interleukin-15, -7, and -2 on the expansion of
depleted PBMC and interleukin-2 after lymphodepleting human V alpha 24+V beta 11+ T cells in vitro. Hum Immunol
chemotherapy. J Immunother 2007; 30: 438–447. 2000; 61: 357–365.
For personal use only.

200. Kawano T, Cui J, Koezuka Y, Toura I, Kaneko Y, Motoki  K, 215. Motohashi S, Ishikawa A, Ishikawa E, Otsuji M, Iizasa T,
Ueno H, Nakagawa R, Sato H, Kondo E, Koseki H, Hanaoka H, Shimizu N, Horiguchi S, Okamoto Y, Fujii S,
Taniguchi M. CD1d-restricted and TCR-mediated activation Taniguchi M, Fujisawa T, Nakayama T. A phase I study of
of valpha14 NKT cells by glycosylceramides. Science 1997; in vitro expanded natural killer T cells in patients with
278: 1626–1629. advanced and recurrent non-small cell lung cancer. Clin
201. Brossay L, Chioda M, Burdin N, Koezuka Y, Casorati G, Cancer Res 2006; 12: 6079–6086.
Dellabona P, Kronenberg M. CD1d-mediated recognition 216. Lee PT, Benlagha K, Teyton L, Bendelac A. Distinct func-
of an alpha-galactosylceramide by natural killer T cells is tional lineages of human V(alpha)24 natural killer T cells.
highly conserved through mammalian evolution. J Exp Med J Exp Med 2002; 195: 637–641.
1998; 188: 1521–1528. 217. Giaccone G, Punt CJ, Ando Y, Ruijter R, Nishi N, Peters M,
202. Spada FM, Koezuka Y, Porcelli SA. CD1d-restricted recogni- von Blomberg BM, Scheper RJ, van der Vliet HJ, van den
tion of synthetic glycolipid antigens by human natural killer Eertwegh AJ, Roelvink M, Beijnen J, Zwierzina H, Pinedo HM.
T cells. J Exp Med 1998; 188: 1529–1534. A phase I study of the natural killer T-cell ligand alpha-ga-
203. Fujii S, Shimizu K, Smith C, Bonifaz L, Steinman RM. lactosylceramide (KRN7000) in patients with solid tumors.
Activation of natural killer T cells by alpha-galactosylcera- Clin Cancer Res 2002; 8: 3702–3709.
mide rapidly induces the full maturation of dendritic cells in 218. Toura I, Kawano T, Akutsu Y, Nakayama T, Ochiai T,
vivo and thereby acts as an adjuvant for combined CD4 and Taniguchi M. Cutting edge: inhibition of experimental tumor
CD8 T cell immunity to a coadministered protein. J Exp Med metastasis by dendritic cells pulsed with alpha-galactosyl-
2003; 198: 267–279. ceramide. J Immunol 1999; 163: 2387–2391.
204. Fujii S, Liu K, Smith C, Bonito AJ, Steinman RM. The linkage 219. Fujii S, Shimizu K, Kronenberg M, Steinman RM. Prolonged
of innate to adaptive immunity via maturing dendritic cells IFN-gamma-producing NKT response induced with alpha-
in vivo requires CD40 ligation in addition to antigen pres- galactosylceramide-loaded DCs. Nat Immunol 2002; 3:
entation and CD80/86 costimulation. J Exp Med 2004; 199: 867–874.
1607–1618. 220. Ishikawa A, Motohashi S, Ishikawa E, Fuchida H,
205. Hayakawa Y, Takeda K, Yagita H, Kakuta S, Iwakura Y, Van Higashino K, Otsuji M, Iizasa T, Nakayama T, Taniguchi M,
Kaer L, Saiki I, Okumura K. Critical contribution of IFN- Fujisawa T. A phase I study of alpha-galactosylceramide
gamma and NK cells, but not perforin-mediated cytotoxic- (KRN7000)-pulsed dendritic cells in patients with advanced
ity, to anti-metastatic effect of alpha-galactosylceramide. and recurrent non-small cell lung cancer. Clin Cancer Res
Eur J Immunol 2001; 31: 1720–1727. 2005; 11: 1910–1917.
206. Brigl M, Brenner MB. CD1: antigen presentation and T cell 221. Nieda M, Okai M, Tazbirkova A, Lin H, Yamaura A, Ide K,
function. Annu Rev Immunol 2004; 22: 817–890. Abraham R, Juji T, Macfarlane DJ, Nicol AJ. Therapeutic
207. Motohashi S, Kobayashi S, Ito T, Magara KK, Mikuni O, activation of Valpha24+Vbeta11+ NKT cells in human sub-
Kamada N, Iizasa T, Nakayama T, Fujisawa T, Taniguchi M. jects results in highly coordinated secondary activation of
Preserved IFN-alpha production of circulating Valpha24 acquired and innate immunity. Blood 2004; 103: 383–389.
NKT cells in primary lung cancer patients. Int J Cancer 2002; 222. Uchida T, Horiguchi S, Tanaka Y, Yamamoto H, Kunii N,
102: 159–165. Motohashi S, Taniguchi M, Nakayama T, Okamoto Y. Phase I
Cancer immunotherapy   189

study of alpha-galactosylceramide-pulsed antigen present- 236. Berzofsky JA, Ahlers JD, Belyakov IM. Strategies for designing
ing cells administration to the nasal submucosa in unresect- and optimizing new generation vaccines. Nat Rev Immunol
able or recurrent head and neck cancer. Cancer Immunol 2001; 1: 209–219.
Immunother 2008; 57: 337–345. 237. Lee P, Wang F, Kuniyoshi J, Rubio V, Stuges T, Groshen S,
223. Cheng WF, Hung CF, Lin KY, Ling M, Juang J, He L, Lin CT, Gee C, Lau R, Jeffery G, Margolin K, Marty V, Weber J. Effects
Wu TC. CD8+ T cells, NK cells and IFN-gamma are important of interleukin-12 on the immune response to a multipeptide
for control of tumor with downregulated MHC class I expres- vaccine for resected metastatic melanoma. Clin Oncol 2001;
sion by DNA vaccination. Gene Ther 2003; 10: 1311–1320. 19: 3836–3847.
224. Ma HL, Whitters MJ, Konz RF, Senices M, Young DA, 238. Moretta L, Moretta A, Unravelling natural killer cell func-
Critical Reviews in Clinical Laboratory Sciences Downloaded from informahealthcare.com by University of Notre Dame Australia on 06/14/13

Grusby MJ, Collins M, Dunussi-Joannopoulos K. IL-21 acti- tion: triggering and inhibitory human NK receptors. Embo
vates both innate and adaptive immunity to generate potent J 2004; 23: 255–259.
antitumor responses that require perforin but are independ- 239. Moretta L, Ciccone E, Moretta A, Hoglund P, Ohlen C,
ent of IFN-gamma. J Immunol 2003; 171: 608–615. Karre  K. Allorecognition by NK cells: nonself or no self?
225. Cerwenka A, Baron JL, Lanier LL. Ectopic expression of Immunol Today 1992; 13: 300–306.
retinoic acid early inducible-1 gene (RAE-1) permits natural 240. Karre K. NK cells, MHC class I molecules and the missing
killer cell-mediated rejection of a MHC class I-bearing tumor self. Scand J Immunol 2002; 55: 221–228.
in vivo. Proc Natl Acad Sci USA 2001; 98: 11521–11526. 241. Moretta A, Bottino C, Pende D, Tripodi G, Tambussi  G,
226. Moretta A, Bottino C, Vitale M, Pende D, Cantoni C, Viale  O, Orengo A, Barbaresi M, Merli A, Ciccone E,
Mingari MC, Biassoni R, Moretta L. Activating receptors and Moretta  L. Identification of four subsets of human CD3-
coreceptors involved in human natural killer cell-mediated CD16+ natural killer (NK) cells by the expression of clon-
cytolysis. Annu Rev Immunol 2001; 19: 197–223. ally distributed functional surface molecules: correlation
227. Wu J, Lanier LL. Natural killer cells and cancer. Adv Cancer between subset assignment of NK clones and ability to
Res 2003; 90: 127–156. mediate specific alloantigen recognition. J Exp Med 1990;
228. Mocikat R, Braumuller H, Gumy A, Egeter O, Ziegler H, 172: 1589–1598.
Reusch U, Bubeck A, Louis J, Mailhammer R, Riethmuller G, 242. Colonna M, Brooks EG, Falco M, Ferrara GB, Strominger JL.
Koszinowski U, Rocken M. Natural killer cells activated by Generation of allospecific natural killer cells by stimula-
MHC class I(low) targets prime dendritic cells to induce pro- tion across a polymorphism of HLA-C. Science 1993; 260:
tective CD8 T cell responses. Immunity 2003; 19: 561–569. 1121–1124.
229. Diefenbach A, Raulet DH. The innate immune response 243. Ruggeri L, Capanni M, Tosti A, Urbani E, Posati S, Aversa F,
For personal use only.

to tumors and its role in the induction of T-cell immunity. Martelli MF, Velardi A. Innate immunity against hematologi-
Immunol Rev 2002; 188: 9–21. cal malignancies. Cytotherapy 2002; 4: 343–346.
230. Kelly JM, Darcy PK, Markby JL, Godfrey DI, Takeda K, 244. van der Meer A, Schaap NP, Schattenberg AV, van
Yagita H, Smyth MJ. Induction of tumor-specific T cell mem- Cranenbroek B, Tijssen HJ, Joosten I. KIR2DS5 is associated
ory by NK cell-mediated tumor rejection. Nat Immunol with leukemia free survival after HLA identical stem cell
2002; 3: 83–90. transplantation in chronic myeloid leukemia patients. Mol
231. Moretta A. Natural killer cells and dendritic cells: rendez- Immunol 2008; 45: 3631–3638.
vous in abused tissues. Nat Rev Immunol 2002; 2: 957–964. 245. Verheyden S, Demanet C. NK cell receptors and their ligands
232. Lotem M, Shiloni E, Pappo I, Drize O, Hamburger T, in leukemia. Leukemia 2008; 22: 249–257.
Weitzen  R, Isacson R, Kaduri L, Merims S, Frankenburg S, 246. Ruggeri L, Capanni M, Martelli MF, Velardi A. Cellular ther-
Peretz T. Interleukin-2 improves tumour response to DNP- apy: exploiting NK cell alloreactivity in transplantation. Curr
modified autologous vaccine for the treatment of metastatic Opin Hematol 2001; 8: 355–359.
malignant melanoma. Br J Cancer 2004; 90: 773–780. 247. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA,
233. Osanto S, Schiphorst PP, Weijl NI, Dijkstra N, Van Wees A, Yun GH, Fautsch SK, McKenna D, Le C, Defor TE, Burns LJ,
Brouwenstein N, Vaessen N, Van Krieken JH, Hermans J, Orchard PJ, Blazar BR, Wagner JE, Slungaard A, Weisdorf DJ,
Cleton FJ, Schrier PI. Vaccination of melanoma patients with Okazaki IJ, McGlave PB. Successful adoptive transfer and in
an allogeneic, genetically modified interleukin 2-producing vivo expansion of human haploidentical NK cells in patients
melanoma cell line. Hum Gene Ther 2000; 11: 739–750. with cancer. Blood 2005; 105: 3051–3057.
234. Rosenberg SA, Yang JC, Schwartzentruber DJ, Hwu P, 248. Arai S, Klingemann HG. Natural killer cells: can they be use-
Marincola FM, Topalian SL, Restifo NP, Dudley ME, ful as adoptive immunotherapy for cancer? Expert Opin Biol
Schwarz  SL, Spiess PJ, Wunderlich JR, Parkhurst MR, Ther 2005; 5: 163–172.
Kawakami Y, Seipp CA, Einhorn JH, White DE. Immunologic 249. Perez SA, Mahaira LG, Demirtzoglou FJ, Sotiropoulou PA,
and therapeutic evaluation of a synthetic peptide vaccine Ioannidis P, Iliopoulou EG, Gritzapis AD, Sotiriadou NN,
for the treatment of patients with metastatic melanoma. Nat Baxevanis CN, Papamichail M. A potential role for hydrocor-
Med 1998; 4: 321–327. tisone in the positive regulation of IL-15-activated NK-cell
235. Moller P, Moller H, Sun Y, Dorbic T, Henz BM, Wittig B, proliferation and survival. Blood 2005; 106: 158–166.
Schadendorf D. Increased non-major histocompatibility 250. Albertsson PA, Basse PH, Hokland M, Goldfarb RH,
complex-restricted lytic activity in melanoma patients vac- Nagelkerke JF, Nannmark U, Kuppen PJ. NK cells and the
cinated with cytokine gene-transfected autologous tumor tumour microenvironment: implications for NK-cell function
cells. Cancer Gene Ther 2000; 7: 976–984. and anti-tumour activity. Trends Immunol 2003; 24: 603–609.

You might also like