ArticlePDF AvailableLiterature Review

Current State and Perspectives of Dendritic Cell Vaccination in Cancer Immunotherapy

Authors:
  • Hautarztpraxis Glattbrugg

Abstract

Recent progress in the approach towards immunotherapy of cancer consists in molecular definition of tumor antigens, new tools for phenotypical and functional characterization of tumor-specific effector cells and clinical use of novel adjuvants for optimal stimulation of a cancer-specific immune response such as dendritic cells. In spite of these advances and immunological as well as clinical responses in selected patients, mechanisms involved in dendritic-cell-based cancer immunotherapy are still poorly understood. Therefore, a standardized study design and small pilot trials are needed to explore open scientific questions in future clinical trials. This review focuses on the different parameters of dendritic cell biology relevant to cancer immunotherapy and on innovative approaches to hopefully enhance the efficacy of dendritic cell vaccination.
Fax +41 61 306 12 34
E-Mail karger@karger.ch
www.karger.com
Review
Skin Pharmacol Physiol 2006;19:124–131
DOI: 10.1159/000092592
Current State and Perspectives of
Dendritic Cell Vaccination in Cancer
Immunotherapy
A. Farkas
a, b
C. Conrad
a
G. Tonel
a
Z. Borbenyi
c
L. Kemeny
b
A. Dobozy
b
F.O. Nestle
a
a
Department of Dermatology, University Hospital of Zurich, Zurich , Switzerland; b
Department of
Dermatology and Allergology, University of Szeged, and c
2nd Department of Medicine and
Cardiology Center, Medical Faculty, Albert Szent-Györgyi Medical and Pharmaceutical Center,
University of Szeged, Szeged , Hungary
Introduction
In the past decades, many immunotherapeutic ap-
proaches for cancer have been developed. Recent scien-
tifi c progress included the discovery of tumor-associated
antigens (TAAs), new tools for monitoring an anticancer
immune response and application of novel adjuvants for
amplifi cation of the immune response such as dendritic
cells (DCs) [1] . DCs are potent antigen-presenting cells
recognizing microorganisms, secreting proinfl ammatory
cytokines and stimulating primary T-cell immune re-
sponses [2] . DC-based vaccines have shown effi cacy in
animal models and are now being tested in the clinic.
There are many parameters which should be taken into
account in DC vaccination studies including optimal DC
source and type, optimal maturation status, optimal cell
dose, optimal antigen preparation, optimal route, opti-
mal timing and optimal means of assessing immune and
clinical responses [3] . Furthermore, possible immune es-
cape mechanisms of tumors affecting DCs should also be
investigated [4]
.
Key Words
Cancer Immunotherapy Dendritic cell vaccination
Abstract
Recent progress in the approach towards immunother-
apy of cancer consists in molecular defi nition of tumor
antigens, new tools for phenotypical and functional char-
acterization of tumor-specifi c effector cells and clinical
use of novel adjuvants for optimal stimulation of a can-
cer-specifi c immune response such as dendritic cells. In
spite of these advances and immunological as well as
clinical responses in selected patients, mechanisms in-
volved in dendritic-cell-based cancer immunotherapy
are still poorly understood. Therefore, a standardized
study design and small pilot trials are needed to explore
open scientifi c questions in future clinical trials. This re-
view focuses on the different parameters of dendritic cell
biology relevant to cancer immunotherapy and on inno-
vative approaches to hopefully enhance the effi cacy of
dendritic cell vaccination.
Copyright © 2006 S. Karger AG, Basel
Received: May 2, 2005
Accepted after revision: June 23, 2005
Published online: April 6, 2006
Arpad Farkas, MD, PhD
Department of Dermatology, University Hospital of Zurich
Gloriastrasse 31, CH–8091 Zurich (Switzerland)
Tel. +41 44 255 3977, Fax +41 44 255 4418
E-Mail arpad.farkas@usz.ch or farkas_arpad@yahoo.com
© 2006 S. Karger AG, Basel
1660–5527/06/0193–0124$23.50/0
Accessible online at:
www.karger.com/spp
Dendritic-Cell-Based Cancer
Immunotherapy
Skin Pharmacol Physiol 2006;19:124–131 125
Source, Type of DCs and Their in vitro
Manipulation for Clinical Applications
The fi rst DC studies utilized whole blood leukapher-
esis products and gradient-based methods to enrich DC
precursors. Vaccination with these cells induced some
immunological and clinical responses in non-Hodgkin’s
lymphoma [5] , multiple myeloma [6] and prostate cancer
[7] . The potential problem with peripherial blood DC iso-
lation is the low yield. There are current attempts to mo-
bilize DCs using systemic fms -related tyrosine kinase 3
(FLT3) ligand [8] . The expansion of DCs from bone mar-
row precursors is also under investigation. CD34+ pro-
genitor-derived DC preparations lead to the induction of
T-cell responses and clinical responses in metastatic mel-
anoma [9] or multiple myeloma [10] .
Although DC numbers in peripherial blood are very
small, it is possible to generate acceptable numbers of cells
in vitro. The common way to generate DC vaccines is to
culture monocytes in the presence of interleukin-4 (IL-4)
and granulocyte macrophage colony-stimulating factor
(GM-CSF) for 5–7 days which results in the generation of
immature monocyte-derived DCs (moDCs). Immature
moDCs may be loaded with antigen and given to the pa-
tient or they may be subjected to a maturation step for
2–3 days. Usually, the maturation cocktail contains IL-4,
GM-CSF, in combination with IL-1
, IL-6, tumor necrosis
factor-
and prostaglandin E
2
(PGE
2
), referred to as the
gold standard maturation cocktail [11] . These standard
moDCs are homogenous, have high viability, migrate well
and induce cytotoxic T-lymphocyte (CTL) responses.
There is evidence that less time is required for DC differ-
entiation in vivo [12, 13] and there are studies which show
that fully functional DCs can be generated within 48 h
from monocytes in vitro, so-called ‘fast DC’ [14] . Most of
the clinical studies indicate that mature DCs are superior
to immature DCs in stimulating T cells [15, 16] , but there
are some clinical trials which clearly show immune re-
sponses and selected clinical responses using immature
DCs [17, 18] . Furthermore, some data indicate that ‘semi-
mature’ moDCs can prime strong CD4 T-cell responses
with a T-helper 1 (Th1)-type cytokine profi le in cancer pa-
tients [19] . The production of bioactive IL-12 by DCs is
crucial for the development of naïve T-cell precursors into
Th1-dominant cells, which will ensure the generation of
CTLs [20] . Thus, the IL-12 production of the ex-vivo-gen-
erated DCs provides information on their functional ma-
turity. There is a list of agents that inhibit IL-12 production
while leaving other aspects of the DC maturation process
unaffected, one of them being PGE
2
[21] . Indeed it has
been shown that DCs matured in the presence of the gold
standard maturation cocktail produce low levels of IL-12
[22] . DCs that fail to produce suffi cient IL-12 might prime
Th2 responses [23] ; therefore, some studies focus on en-
hancing the effi cacy of DC-based immunotherapy by
transduction of the IL-12 gene
into moDCs [24] . Not only
the maturation signals but also the kinetics of DC activa-
tion are very important in infl uencing their IL-12 produc-
tion. DCs produce IL-12 within 8–16 h after IL-12-induc-
ing stimuli, and later become resistant to further stimula-
tion. Thus, only properly activated DCs can effi ciently
prime Th1 responses, whereas so-called ‘exhausted’ DCs
may prime Th2 or nonpolarized T-cell responses [23] .
Many other protocols have been investigated for the
generation of DCs. For example, moDCs can also be gen-
erated in the presence of IL-13 or IL-15 instead of IL-4,
and for DC maturation, simply tumor necrosis factor-
without additional cytokines has been used [11] . Other
protocols may contain CD40 ligand, FLT3 ligand, stem
cell factor [25] , transforming growth factor-
[25] , lipo-
polysaccharide, poly-I:C, interferon-
(IFN-
), IFN-
,
and there are methods where leukemic cells were used as
DC sources [11] .
For clinical purposes, the development of DC vaccines
requires standardized methods that comply with Good
Manufacturing Practice (GMP) guidelines. Cytokines
and many Toll-like receptor ligands are now available in
GMP quality. Fetal calf serum (FCS) was part of initial
DC protocols, but DCs are now mostly generated in the
absence of FCS using autologous plasma [26] or in serum-
free cultures [27] in order to avoid unwanted loading with
xenoantigens being part of FCS [28] . A future approach
to the safe clinical use of DCs might be closed culture
system. The investigation of such systems is under way
in clinical trials [29–35] . Cryopreservation of DCs might
be an advantage to obtain standardized DC preparations
during vaccination studies. Many freezing and thawing
methods have been described in the literature [36–40] ,
and it seems that cryopreservation does not cause signif-
icant changes in the phenotype or function of DCs, indi-
cating that cryopreserved aliquots of DCs are suitable for
clinical application [41] .
Antigen Preparation and Loading
Tumors express many antigens which are potential tar-
gets for immunotherapy. DCs must obtain tumor anti-
gens to initiate antigen-specifi c immune responses. The
selection of tumor antigens and the way of loading ex-
Farkas /Conrad /Tonel /Borbenyi /Kemeny /
Dobozy /Nestle
Skin Pharmacol Physiol 2006;19:124–131
126
vivo-generated DCs is an important step to develop an
effi cient cancer vaccine. Antigen presentation strategies
use peptides, proteins, tumor lysates, tumor-derived
RNA or genetically modifi ed recombinant viruses for de-
livery of DNA. Other trials use exosomes plus peptides,
apoptotic bodies, DC/tumor fusions, tumoral idiotype or
-galactosylceramide. The majority of vaccinations were
carried out with tumor lysate or peptide-loaded DCs, but
there is a tendency to use RNA-loaded DCs [11] .
Whole tumor cell preparations aim to deliver a wide
range of tumor antigens to DCs. For this purpose, whole
apoptotic [42–44] or necrotic tumor [45, 46] cells, entire
tumor lysates [47–49] , and DC/tumor cell fusion prod-
ucts [50–52] have been used. Identifi cation of specifi c
TAAs is not necessary when we use whole tumor cell prep-
arations, but this strategy depends on the availability of
suffi cient tumor tissue from the patient. In the case of
unfractionated tumor material, there is a potential risk of
inducing autoimmunity. One of the strategies with whole
tumor cells is the use of hybridomas created by the fusion
of DCs with tumor cells. The standardization of hybrid
generation, using electrofusion or polyethylene glycol, is
not easy to achieve. To address this problem, a new meth-
od was developed to generate hybrid cell vaccines, based
on gene transfer of a viral fusogenic membrane glycopro-
tein into tumor cells. These DC/tumor cell hybrids can
be rapidly isolated, are highly potent in vitro in antigen
presentation assays, target lymph nodes in vivo and are
powerful immunogens against metastatic disease in mice
[53] . There are clinical trials published with DC/tumor
fusions in patients with solid tumors [51, 54, 55] and he-
matological malignancies [56–58] . Recently, investiga-
tors have used allogeneic DCs to generate DC/tumor fu-
sion vaccines. In this case, DCs are generated from nor-
mal donors which may have better functional properties
[59, 60] . On the other hand, T-cell responses to alloge-
neic DC/tumor fusions are dependent on tumor MHC
class I molecule expression, which is often inconsistent.
In a recent report, 24 patients underwent serial vaccina-
tion with
allogeneic DC/tumor hybrids. Two patients ex-
perienced a partial response, and 8 patients
demonstrated
stable disease [61] .
There is no need for tumor tissue if peptide combina-
tions are used and more accurate peptide-specifi c T-cell
response monitoring is possible [62] . As the induced im-
mune response is largely epitope-specifi c, no cross-reac-
tivity to normal tissue antigens is expected. For this meth-
od, it is a prerequisite to know the tumor epitopes, patient
HLA type and amino acid sequence of relevant peptides.
To avoid the generation of tumor cell escape variants, a
broad spectrum of tumor antigens can be used. Another
possibility is to use an unfractionated mixture of peptides
eluted from tumor cells to target a broad repertoire of tu-
mor antigens [63, 64] .
Purifi ed or recombinant proteins can also be a choice
for loading DCs [5] . Loading DCs with soluble protein
antigens circumvents the need of peptide epitopes bind-
ing to specifi c MHC surface molecules. The potential to
induce a broad repertoire of antigen-specifi c T cells might
be advantageous and should minimize immune escape.
As the immature phenotype of DCs is associated with ef-
cient protein uptake, the costimulatory molecule expres-
sion should be carefully monitored before loading [65] .
During a study of patients with
hormone-refractory pros-
tate cancer, autologous human DCs were exposed to a
recombinant fusion protein, linking full-length human
prostatic
acid phosphatase and GM-CSF and then in-
fused intravenously. The postvaccination
proliferative
responses that did develop were specifi c for the
immuniz-
ing antigen. Delayed time to disease progression also
cor-
related with development of a proliferative response to
prostatic
acid phosphatase [66] .
The RNA-based strategy to load DCs with tumor cell
antigens is receiving more attention [11, 67] . Even when
the amount of tumor material is limited, suffi cient
amounts of total RNA can be prepared and transfected
into DCs resulting in MHC class I-restricted antigen pre-
sentation [37, 68] . However, obtaining effective MHC
class II presentation is a challenge. Tumor-specifi c RNA
can be enriched by subtractive hybridization with normal
cell RNA; thus, the risk of autoimmunity can be decreased
[69] . Recently, it has been demonstrated that not only
immature DCs but alternatively mature DCs can be di-
rectly transfected with RNAs [70] . In one comparative
study, it was demonstrated that DCs loaded with tumor
mRNA were more potent in inducing T-cell responses
than DCs loaded with apoptotic cancer cells [44] .
Genetically modifi ed recombinant viruses can be used
to transfect genes of interest into DCs [71, 72] . However,
the potential adverse effects such as cell toxicity and the
induction of antivirus immune responses have so far lim-
ited the use of such vaccines [73, 74] . It has also been
shown that viral transduction of DCs may downregulate
their costimulatory molecule expression [73] .
Biodegradable poly(lactide-co-glycolide) microspheres
(PLGA-MS) are under investigation as delivery tools for
synthetic peptide antigen loading of human moDCs. Im-
mature moDCs readily take up PLGA-MS and present
epitopes from encapsulated proteins or peptides both on
MHC class I and class II. These DCs have good migra-
Dendritic-Cell-Based Cancer
Immunotherapy
Skin Pharmacol Physiol 2006;19:124–131 127
tion, cytokine secretion, survival and allostimulatory
properties and their antigen presentation capacity is pro-
longed [75] . However, PLGA polymers undergo bulk hy-
drolysis, resulting in a dramatic drop in pH value that can
signifi cantly degrade encapsulated DNA molecules, and
their DNA release rate is diffi cult to regulate. To solve
this problem, two alternative polyortho esters (POEs)
have recently been tested, and in contrast to the bulk deg-
radation seen in PLGA polymers, POEs exhibit surface-
restricted hydrolysis, causing a milder pH shift, less like-
ly to result in DNA degradation [76] .
Delivery of DCs
The route of vaccine administration is a very impor-
tant issue [77] . Various vaccination routes have been uti-
lized such as intravenous, intradermal, subcutaneous or
intranodal. A study which compared the intravenous, in-
tradermal and intranodal routes in melanoma patients
has suggested that the intravenous administration may
not work as well and that intranodal administration as
rst applied by our group [47] seems to result in superior
T-cell function [78] . Recent studies in mice suggest that
the number of DCs migrating from the skin to draining
lymph nodes after gene gun use is hundred times higher
than estimated earlier and these DCs persist for 2 weeks
[79] . Myeloid DC migration is controlled by the chemo-
kine receptor CCR7 and its ligands [80] . PGE
2
, which is
in most cases part of the maturation cocktail, induces a
very weak bioactive IL-12 production, but is important
for functional CCR7 expression of DCs [13, 81] . Cur-
rently, alternative efforts are being employed to facilitate
DC migration following intradermal injection such as the
distribution of DCs to more than one areas of the body
and pretreatment of the skin with proinfl ammatory cyto-
kines or Toll-like receptor ligands [82, 83] .
The direct injection of DCs into the lymph nodes cir-
cumvents the skin migration problem [49] . Intranodal
injection might destroy the fi ne architecture of the lymph
node; therefore, intralymphatic injection of DCs might
be an alternative.
Ex-vivo-derived DCs can be directly injected into the
tumor site [84] . In one early clinical study, patients with
metastatic melanoma and breast cancer were treated with
intratumoral DC vaccination and regression of the in-
jected tumors was seen in 6 out of 10 patients [85] . There
are attempts to enhance the effi cacy of intratumoral vac-
cination by engineering proinfl ammatory cytokine-ex-
pressing DCs [86–88] .
In vivo DC Manipulation
In vivo targeting of DCs would be an ideal situation
to circumvent demanding and time-consuming in vitro
cell manipulation. Several DC-associated C-type lectin
receptors such as DEC205 and DC-SIGN have been
shown to endocytose antigens coupled to antibodies di-
rected against them [89, 90] . The antigen DEC205 com-
plex leads to both CD4 and CD8 T-cell responses indicat-
ing cross-presentation of endocytosed antigens [90] , but
only the simultaneous delivery of a DC maturation stim-
ulus via CD40,
together with an antigen coupled to
DEC205, results in immunostimulation [90] . The ma-
nipulation of CD40 on DCs not only helps to overcome
peripheral T-cell tolerance [91] but also enhances their
costimulatory molecule expression, cytokine secretion
and induces anti-apoptotic signals [92] .
The combination of FLT3
ligand with a DC activator,
immunostimulatory DNA,
and a tumor antigen to acti-
vate and load DCs in vivo showed that DCs can be acti-
vated, and potent antitumor
immunity can be induced in
mice [93, 94] .
Epidermal DCs or Langerhans cells (LCs) migrate to
draining lymph nodes and undergo maturation. LC-tar-
geted delivery of gene products has been accomplished by
plasmid DNA encoding TAAs fused to GM-CSF or che-
mokines [95] . Migratory LCs might also be entrapped by
subcutaneous implantation of an ethylene vinyl acetate
polymer rod releasing macrophage infl ammatory protein
3
and hapten. The entrapped LCs may be in situ antigen
loaded by a second ethylene vinyl acetate polymer rod re-
leasing TAAs. This technology induced tumor-specifi c im-
munity without the use of ex vivo DC manipulation [96] .
Another immunization strategy has recently been de-
scribed which uses a plasmid DNA expressing HIV pro-
teins. The DNA is formulated to a mannosylated particle
to target antigen-presenting cells and to protect the DNA
from intracellular degradation. This topically applied vac-
cination represents an effective in situ DC-based immuni-
zation possibility providing new therapeutic options [97] .
Future Challenges and Conclusions
Until now, a large number of DC vaccination studies
have been performed [98] ; however, the comparison of
these trials is diffi cult because of the lack of standardiza-
tion. It will be important to standardize the parameters
of DC vaccination and to test the many different vari-
ables in defi nitive trials with clear endpoints.
Farkas /Conrad /Tonel /Borbenyi /Kemeny /
Dobozy /Nestle
Skin Pharmacol Physiol 2006;19:124–131
128
Closed culture systems may simplify the handling pro-
cedures and will circumvent the use of expensive GMP
facilities. The efforts focusing on the in vivo application
of DCs will also hopefully facilitate the laborious and
time-consuming in vitro cell manipulation. The optimal
DC type for vaccination remains to be determined in the
future; therefore, other DC types should be tested for ther-
apeutic use such as plasmacytoid DCs (pDC) [99] and
IFN-
-derived DCs (IFN-DC) [100] . pDCs are known to
infi ltrate tumor tissues [101] and they have the capacity
to produce high amounts of type I IFNs [99] , which may
induce the maturation of local myeloid DCs. Highly ac-
tive IFN-DCs can be generated from monocytes in the
presence of IFN-
and GM-CSF [100] and may be useful
in future vaccination studies (e.g. chronic myelogenous
leukemia) [102] .
Preferably, vaccination against multiple tumor anti-
gens and TAAs expressed by the tumor and also by the
stroma should be used. Many tumors such as malignant
melanoma are characterized by high genetic diversity and
instability; therefore, it would be desirable to analyze the
immunogenic profi le of the primary tumor and of the
metastatic lesions with the help of microarrays to design
and choose the specifi c vaccine [103] .
It will be important to remove regulatory T cells or to
block their function, e.g. with the help of anti-CTLA-4
antibody [104, 105] . Clinical studies have just started ex-
ploiting this option. Other combination possibilities need
to be investigated such as DC vaccination with concurrent
cytokine administration, with chemotherapy and in com-
bination with therapies targeting the tumor stroma [106] .
The stage of cancer determines the effi cacy of immu-
notherapy. Until now, end-stage patients have been treat-
ed with a weak immune system; therefore, it is suggested
that in future studies DC vaccination should be extended
for treatment of patients with low tumor burden.
In conclusion, future controlled clinical trials will
hopefully provide new insight into immune activation,
tumor regression and tumor immune escape during DC
vaccination.
Acknowledgements
We apologize that many excellent papers could not be cited due
to space limitations. Arpad Farkas is on the leave from the Depart-
ment of Dermatology and Allergology in Szeged, Hungary, and was
supported by an International Union against Cancer (UICC) fel-
lowship.
References
1 Banchereau J, Steinman RM: Dendritic cells
and the control of immunity. Nature 1998; 392:
245–252.
2 Banchereau J, Briere F, Caux C, Davoust J,
Lebecque S, Liu YJ, Pulendran B, Palucka K:
Immunobiology of dendritic cells. Annu Rev
Immunol 2000; 18: 767–811.
3 Nestle FO, Farkas A, Conrad C: Dendritic-cell-
based therapeutic vaccination against cancer.
Curr Opin Immunol 2005; 17: 163–169.
4 Gabrilovich D: Mechanisms and functional
signifi cance of tumour-induced dendritic-cell
defects. Nat Rev Immunol 2004; 4: 941–952.
5 Hsu FJ, Benike C, Fagnoni F, Liles TM, Czer-
winski D, Taidi B, Engleman EG, Levy R: Vac-
cination of patients with B-cell lymphoma us-
ing autologous antigen-pulsed dendritic cells.
Nat Med 1996; 2: 52–58.
6 Reichardt VL, Okada CY, Liso A, Benike CJ,
Stockerl-Goldstein KE, Engleman EG, Blume
KG, Levy R: Idiotype vaccination using den-
dritic cells after autologous peripheral blood
stem cell transplantation for multiple myeloma
– A feasibility study. Blood 1999; 93: 2411–
2419.
7 Burch PA, Breen JK, Buckner JC, Gastineau
DA, Kaur JA, Laus RL, Padley DJ, Peshwa
MV, Pitot HC, Richardson RL, Smits BJ, So-
papan P, Strang G, Valone FH, Vuk-Pavlovic
S: Priming tissue-specifi c cellular immunity in
a phase I trial of autologous dendritic cells for
prostate cancer. Clin Cancer Res 2000; 6:
2175–2182.
8 Maraskovsky E, Daro E, Roux E, Teepe M,
Maliszewski CR, Hoek J, Caron D, Lebsack
ME, McKenna HJ: In vivo generation of hu-
man dendritic cell subsets by Flt3 ligand.
Blood 2000; 96: 878–884.
9 Palucka AK, Dhodapkar MV, Paczesny S,
Ueno H, Fay J, Banchereau J: Boosting vacci-
nations with peptide-pulsed CD34+ progeni-
tor-derived dendritic cells can expand long-
lived melanoma peptide-specifi c CD8+ T cells
in patients with metastatic melanoma. J Im-
munother 2005; 28: 158–168.
10 Titzer S, Christensen O, Manzke O, Tesch H,
Wolf J, Emmerich B, Carsten C, Diehl V,
Bohlen H: Vaccination of multiple myeloma
patients with idiotype-pulsed dendritic cells:
immunological and clinical aspects. Br J Hae-
matol 2000; 108: 805–816.
11 Figdor CG, de Vries IJ, Lesterhuis WJ, Melief
CJ: Dendritic cell immunotherapy: mapping
the way. Nat Med 2004; 10: 475–480.
12 Shortman K, Liu YJ: Mouse and human den-
dritic cell subtypes. Nat Rev Immunol 2002; 2:
151–161.
13 Luft T, Jefford M, Luetjens P, Toy T, Hochrein
H, Masterman KA, Maliszewski C, Shortman
K, Cebon J, Maraskovsky E: Functionally dis-
tinct dendritic cell (DC) populations induced
by physiologic stimuli: prostaglandin E(2) reg-
ulates the migratory capacity of specifi c DC
subsets. Blood 2002; 100: 1362–1372.
14 Obermaier B, Dauer M, Herten J, Schad K,
Endres S, Eigler A: Development of a new pro-
tocol for 2-day generation of mature dendritic
cells from human monocytes. Biol Proced On-
line 2003; 5: 197–203.
15
Adams S, O’Neill D, Bhardwaj N: Maturation
matters: importance of maturation for antitu-
mor immunity of dendritic cell vaccines. J Clin
Oncol 2004; 22: 3834–3835; author reply 3835.
16 Jonuleit H, Giesecke-Tuettenberg A, Tuting T,
Thurner-Schuler B, Stuge TB, Paragnik L,
Kandemir A, Lee PP, Schuler G, Knop J, Enk
AH: A comparison of two types of dendritic
cell as adjuvants for the induction of melano-
ma-specifi c T-cell responses in humans follow-
ing intranodal injection. Int J Cancer 2001; 93:
243–251.
Dendritic-Cell-Based Cancer
Immunotherapy
Skin Pharmacol Physiol 2006;19:124–131 129
17 Smithers M, O’Connell K, MacFadyen S,
Chambers M, Greenwood K, Boyce A, Abdul-
Jabbar I, Barker K, Grimmett K, Walpole E,
Thomas R: Clinical response after intradermal
immature dendritic cell vaccination in meta-
static melanoma is associated with immune
response to particulate antigen. Cancer Immu-
nol Immunother 2003; 52: 41–52.
18 Su Z, Dannull J, Heiser A, Yancey D, Pruitt S,
Madden J, Coleman D, Niedzwiecki D, Gilboa
E, Vieweg J: Immunological and clinical re-
sponses in metastatic renal cancer patients vac-
cinated with tumor RNA-transfected dendritic
cells. Cancer Res 2003; 63: 2127–2133.
19 Gilliet M, Kleinhans M, Lantelme E, Schaden-
dorf D, Burg G, Nestle FO: Intranodal injec-
tion of semimature monocyte-derived dendrit-
ic cells induces T helper type 1 responses to
protein neoantigen. Blood 2003; 102: 36–42.
20 Trinchieri G: Interleukin-12 and the regulation
of innate resistance and adaptive immunity.
Nat Rev Immunol 2003; 3: 133–146.
21 Kalinski P, Vieira PL, Schuitemaker JH, de
Jong EC, Kapsenberg ML: Prostaglandin E(2)
is a selective inducer of interleukin-12 p40 (IL-
12p40) production and an inhibitor of bioac-
tive IL-12p70 heterodimer. Blood 2001; 97:
3466–3469.
22 Lee AW, Truong T, Bickham K, Fonteneau JF,
Larsson M, Da Silva I, Somersan S, Thomas
EK, Bhardwaj N: A clinical grade cocktail of
cytokines and PGE2 results in uniform matu-
ration of human monocyte-derived dendritic
cells: implications for immunotherapy. Vac-
cine 2002; 20(suppl 4):A8–A22.
23 Langenkamp A, Messi M, Lanzavecchia A,
Sallusto F: Kinetics of dendritic cell activation:
impact on priming of TH1, TH2 and nonpolar-
ized T cells. Nat Immunol 2000; 1: 311–316.
24 Nishimura N, Nishioka Y, Shinohara T, Sone
S: Enhanced effi ciency by centrifugal manipu-
lation of adenovirus-mediated interleukin 12
gene transduction into human monocyte-de-
rived dendritic cells. Hum Gene Ther 2001; 12:
333–346.
25 Zou GM, Tam YK: Cytokines in the genera-
tion and maturation of dendritic cells: recent
advances. Eur Cytokine Netw 2002; 13: 186–
199.
26 Moldenhauer A, Nociari MM, Dias S, Lalezari
P, Moore MA: Optimized culture conditions
for the generation of dendritic cells from pe-
ripheral blood monocytes. Vox Sang 2003; 84:
228–236.
27 Akiyama Y, Tanosaki R, Inoue N, Shimada M,
Hotate Y, Yamamoto A, Yamazaki N, Ka-
washima I, Nukaya I, Takesako K, Maruyama
K, Takaue Y, Yamaguchi K: Clinical response
in Japanese metastatic melanoma patients
treated with peptide cocktail-pulsed dendritic
cells. J Transl Med 2005; 3: 4.
28 Toldbod HE, Agger R, Bolund L, Hokland M:
Potent infl uence of bovine serum proteins in
experimental dendritic cell-based vaccination
protocols. Scand J Immunol 2003; 58: 43–50.
29 Buchler T, Kovarova L, Musilova R, Bourkova
L, Ocadlikova D, Bulikova A, Hanak L, Mi-
chalek J, Hajek R: Generation of dendritic cells
using cell culture bags – Description of a meth-
od and review of literature. Hematology 2004;
9: 199–205.
30 Sorg RV, Ozcan Z, Brefort T, Fischer J,
Ackermann R, Muller M, Wernet P: Clinical-
scale generation of dendritic cells in a closed
system. J Immunother 2003; 26: 374–383.
31 Berger TG, Feuerstein B, Strasser E, Hirsch U,
Schreiner D, Schuler G, Schuler-Thurner B:
Large-scale generation of mature monocyte-
derived dendritic cells for clinical application
in cell factories. J Immunol Methods 2002;
268: 131–140.
32 Tuyaerts S, Noppe SM, Corthals J, Breckpot
K, Heirman C, De Greef C, Van Riet I, Thiele-
mans K: Generation of large numbers of den-
dritic cells in a closed system using Cell Facto-
ries. J Immunol Methods 2002; 264: 135–151.
33 Pullarkat V, Lau R, Lee SM, Bender JG, Weber
JS: Large-scale monocyte enrichment coupled
with a closed culture system for the generation
of human dendritic cells. J Immunol Methods
2002; 267: 173–183.
34 Celluzzi CM, Welbon C: Dendritic cell culture:
a simple closed culture system using Ficoll,
monocytes, and a table-top centrifuge. J Hema-
tother Stem Cell Res 2003; 12: 575–585.
35 Berger TG, Strasser E, Smith R, Carste C,
Schuler-Thurner B, Kaempgen E, Schuler G:
Effi cient elutriation of monocytes within a
closed system (Elutra) for clinical-scale genera-
tion of dendritic cells. J Immunol Methods
2005; 298: 61–72.
36 Celluzzi CM, Welbon C: A simple cryopreser-
vation method for dendritic cells and cells used
in their derivation and functional assessment.
Transfusion 2003; 43: 488–494.
37 Mu LJ, Lazarova P, Gaudernack G, Saeboe-
Larssen S, Kvalheim G: Development of a clin-
ical grade procedure for generation of mRNA
transfected dendritic cells from purifi ed frozen
CD34(+) blood progenitor cells. Int J Immuno-
pathol Pharmacol 2004; 17: 255–263.
38 Hori S, Heike Y, Takei M, Maruyama M,
Inoue Y, Lee JJ, Kim HJ, Harada Y, Kawai H,
Shimosaka A, Kami M, Tanosaki MD, Waka-
sugi H, Saito S, Takaue Y, Kakizoe T: Freeze-
thawing procedures have no infl uence on the
phenotypic and functional development of
dendritic cells generated from peripheral blood
CD14+ monocytes. J Immunother 2004; 27:
27–35.
39 Westermann J, Korner IJ, Kopp J, Kurz S,
Zenke M, Dorken B, Pezzutto A: Cryopreser-
vation of mature monocyte-derived human
dendritic cells for vaccination: infl uence on
phenotype and functional properties. Cancer
Immunol Immunother 2003; 52: 194–198.
40 de Vries IJ, Eggert AA, Scharenborg NM, Vis-
sers JL, Lesterhuis WJ, Boerman OC, Punt CJ,
Adema GJ, Figdor CG: Phenotypical and func-
tional characterization of clinical grade den-
dritic cells. J Immunother 2002; 25: 429–438.
41 Feuerstein B, Berger TG, Maczek C, Roder C,
Schreiner D, Hirsch U, Haendle I, Leisgang W,
Glaser A, Kuss O, Diepgen TL, Schuler G,
Schuler-Thurner B: A method for the produc-
tion of cryopreserved aliquots of antigen-pre-
loaded, mature dendritic cells ready for clinical
use. J Immunol Methods 2000; 245: 15–29.
42 Goldszmid RS, Idoyaga J, Bravo AI, Steinman
R, Mordoh J, Wainstok R: Dendritic cells
charged with apoptotic tumor cells induce
long-lived protective CD4+ and CD8+ T cell
immunity against B16 melanoma. J Immunol
2003; 171: 5940–5947.
43 Albert ML, Pearce SF, Francisco LM, Sauter
B, Roy P, Silverstein RL, Bhardwaj N: Imma-
ture dendritic cells phagocytose apoptotic cells
via alphavbeta5 and CD36, and cross-present
antigens to cytotoxic T lymphocytes. J Exp
Med 1998; 188: 1359–1368.
44 Jarnjak-Jankovic S, Pettersen RD, Saeboe-
Larssen S, Wesenberg F, Gaudernack G: Eval-
uation of dendritic cells loaded with apoptotic
cancer cells or expressing tumour mRNA as
potential cancer vaccines against leukemia.
BMC Cancer 2005; 5: 20.
45 Sauter B, Albert ML, Francisco L, Larsson M,
Somersan S, Bhardwaj N: Consequences of cell
death: exposure to necrotic tumor cells, but not
primary tissue cells or apoptotic cells, induces
the maturation of immunostimulatory den-
dritic cells. J Exp Med 2000; 191: 423–434.
46 Chang GC, Lan HC, Juang SH, Wu YC, Lee
HC, Hung YM, Yang HY, Whang-Peng J, Liu
KJ: A pilot clinical trial of vaccination with
dendritic cells pulsed with autologous tumor
cells derived from malignant pleural effusion
in patients with late-stage lung carcinoma.
Cancer 2005; 103: 763–771.
47 Nestle FO, Alijagic S, Gilliet M, Sun Y, Grab-
be S, Dummer R, Burg G, Schadendorf D: Vac-
cination of melanoma patients with peptide –
Or tumor lysate-pulsed dendritic cells. Nat
Med 1998; 4: 328–332.
48 De Vleeschouwer S, Arredouani M, Ade M,
Cadot P, Vermassen E, Ceuppens JL, Van
Gool SW: Uptake and presentation of malig-
nant glioma tumor cell lysates by monocyte-
derived dendritic cells. Cancer Immunol Im-
munother 2005; 54: 372–382.
49 Maier T, Tun-Kyi A, Tassis A, Jungius KP,
Burg G, Dummer R, Nestle FO: Vaccination
of patients with cutaneous T-cell lymphoma
using intranodal injection of autologous tu-
mor-lysate-pulsed dendritic cells. Blood 2003;
102: 2338–2344.
50 Walden P: Hybrid cell vaccination for cancer
immunotherapy. Adv Exp Med Biol 2000; 465:
347–354.
51 Trefzer U, Herberth G, Wohlan K, Milling A,
Thiemann M, Sherev T, Sparbier K, Sterry W,
Walden P: Vaccination with hybrids of tumor
and dendritic cells induces tumor-specifi c T-
cell and clinical responses in melanoma stage
III and IV patients. Int J Cancer 2004; 110:
730–740.
Farkas /Conrad /Tonel /Borbenyi /Kemeny /
Dobozy /Nestle
Skin Pharmacol Physiol 2006;19:124–131
130
52 Trefzer U, Weingart G, Chen Y, Herberth G,
Adrian K, Winter H, Audring H, Guo Y, Ster-
ry W, Walden P: Hybrid cell vaccination for
cancer immune therapy: fi rst clinical trial with
metastatic melanoma. Int J Cancer 2000; 85:
618–626.
53 Phan V, Errington F, Cheong SC, Kottke T,
Gough M, Altmann S, Brandenburger A, Em-
ery S, Strome S, Bateman A, Bonnotte B,
Melcher A, Vile R: A new genetic method to
generate and isolate small, short-lived but
highly potent dendritic cell-tumor cell hybrid
vaccines. Nat Med 2003; 9: 1215–1219.
54 Haenssle HA, Krause SW, Emmert S, Zutt M,
Kretschmer L, Schmidberger H, Andreesen R,
Soruri A: Hybrid cell vaccination in metastatic
melanoma: clinical and immunologic results of
a phase I/II study. J Immunother 2004; 27:
147–155.
55 Avigan D, Vasir B, Gong J, Borges V, Wu Z,
Uhl L, Atkins M, Mier J, McDermott D, Smith
T, Giallambardo N, Stone C, Schadt K, Dol-
goff J, Tetreault JC, Villarroel M, Kufe D:
Fusion cell vaccination of patients with meta-
static breast and renal cancer induces immu-
nological and clinical responses. Clin Cancer
Res 2004; 10: 4699–4708.
56 Wahl U, Nossner E, Kronenberger K, Gangnus
R, Pohla H, Staege MS, Kolb HJ, Hallek M,
Mocikat R: Vaccination against B-cell chronic
lymphocytic leukemia with trioma cells: pre-
clinical evaluation. Clin Cancer Res 2003; 9:
4240–4246.
57 Galea-Lauri J, Darling D, Mufti G, Harrison
P, Farzaneh F: Eliciting cytotoxic T lympho-
cytes against acute myeloid leukemia-derived
antigens: evaluation of dendritic cell-leukemia
cell hybrids and other antigen-loading strate-
gies for dendritic cell-based vaccination.
Cancer Immunol Immunother 2002; 51: 299–
310.
58 Ossenkoppele GJ, Stam AG, Westers TM, de
Gruijl TD, Janssen JJ, van de Loosdrecht AA,
Scheper RJ: Vaccination of chronic myeloid
leukemia patients with autologous in vitro cul-
tured leukemic dendritic cells. Leukemia 2003;
17: 1424–1426.
59 Neves AR, Ensina LF, Anselmo LB, Leite KR,
Buzaid AC, Camara-Lopes LH, Barbuto JA:
Dendritic cells derived from metastatic cancer
patients vaccinated with allogeneic dendritic
cell-autologous tumor cell hybrids express
more CD86 and induce higher levels of inter-
feron-gamma in mixed lymphocyte reactions.
Cancer Immunol Immunother 2005; 54: 61–
66.
60
Suzuki T, Fukuhara T, Tanaka M, Nakamu-
ra A, Akiyama K, Sakakibara T, Koinuma D,
Kikuchi T, Tazawa R, Maemondo M, Hagi-
wara K, Saijo Y, Nukiwa T: Vaccination of
dendritic cells loaded with interleukin-12-
secreting cancer cells augments in vivo anti-
tumor immunity: characteristics of syngeneic
and allogeneic antigen-presenting cell cancer
hybrid cells. Clin Cancer Res 2005; 11: 58–
66.
61 Avigan D, George D, Kantoff P, Figlin R, Kufe
D, Olencki T, Vasconcelles M, Vasir B, Xu Y,
Bukowski RM: Interim safety and effi cacy re-
sults from a phase I/II study of vaccination
with electrofused allogeneic dendritic cells/au-
tologous tumor-derived cells in patients with
stage IV renal cell carcinoma. J Clin Oncol
ASCO Annu Meet Proc, New Orleans, 2004,
vol 22, p 169.
62 Romero P, Cerottini JC, Speiser DE: Monitor-
ing tumor antigen specifi c T-cell responses in
cancer patients and phase I clinical trials of
peptide-based vaccination. Cancer Immunol
Immunother 2004; 53: 249–255.
63 Zitvogel L, Mayordomo JI, Tjandrawan T, De-
Leo AB, Clarke MR, Lotze MT, Storkus WJ:
Therapy of murine tumors with tumor peptide-
pulsed dendritic cells: dependence on T cells,
B7 costimulation, and T helper cell 1-associ-
ated cytokines. J Exp Med 1996; 183: 87–97.
64 Santin AD, Bellone S, Ravaggi A, Pecorelli S,
Cannon MJ, Parham GP: Induction of ovarian
tumor-specifi c CD8+ cytotoxic T lymphocytes
by acid-eluted peptide-pulsed autologous den-
dritic cells. Obstet Gynecol 2000; 96: 422–430.
65 Zhou Y, Bosch ML, Salgaller ML: Current
methods for loading dendritic cells with tumor
antigen for the induction of antitumor immu-
nity. J Immunother 2002; 25: 289–303.
66 Small EJ, Fratesi P, Reese DM, Strang G, Laus
R, Peshwa MV, Valone FH: Immunotherapy
of hormone-refractory prostate cancer with an-
tigen-loaded dendritic cells. J Clin Oncol 2000;
18: 3894–3903.
67 Jarnjak-Jankovic S, Pettersen RD, Saeboe-
Larssen S, Wesenberg F, Olafsen MR, Gauder-
nack G: Preclinical evaluation of autologous
dendritic cells transfected with mRNA or load-
ed with apoptotic cells for immunotherapy of
high-risk neuroblastoma. Cancer Gene Ther,
in press.
68 Nair S, Boczkowski D: RNA-transfected den-
dritic cells. Expert Rev Vaccines 2002; 1: 507–
513.
69 Boczkowski D, Nair SK, Snyder D, Gilboa E:
Dendritic cells pulsed with RNA are potent an-
tigen-presenting cells in vitro and in vivo. J
Exp Med 1996; 184: 465–472.
70 Schaft N, Dorrie J, Thumann P, Beck VE,
Muller I, Schultz ES, Kampgen E, Dieckmann
D, Schuler G: Generation of an optimized
polyvalent monocyte-derived dendritic cell
vaccine by transfecting defi ned RNAs after
rather than before maturation. J Immunol
2005; 174: 3087–3097.
71 Di Nicola M, Carlo-Stella C, Mortarini R, Bal-
dassari P, Guidetti A, Gallino GF, Del Vecchio
M, Ravagnani F, Magni M, Chaplin P, Casci-
nelli N, Parmiani G, Gianni AM, Anichini A:
Boosting T cell-mediated immunity to tyrosi-
nase by vaccinia virus-transduced, CD34(+)-
derived dendritic cell vaccination: a phase I
trial in metastatic melanoma. Clin Cancer Res
2004; 10: 5381–5390.
72 Dullaers M, Breckpot K, Van Meirvenne S,
Bonehill A, Tuyaerts S, Michiels A, Straetman
L, Heirman C, De Greef C, Van Der Bruggen
P, Thielemans K: Side-by-side comparison of
lentivirally transduced and MRNA-electro-
porated dendritic cells: implications for cancer
immunotherapy protocols. Mol Ther 2004; 10:
768–779.
73 Jenne L, Schuler G, Steinkasserer A: Viral vec-
tors for dendritic cell-based immunotherapy.
Trends Immunol 2001; 22: 102–107.
74 Kirk CJ, Mule JJ: Gene-modifi ed dendritic
cells for use in tumor vaccines. Hum Gene
Ther 2000; 11: 797–806.
75 Waeckerle-Men Y, Groettrup M: PLGA mi-
crospheres for improved antigen delivery to
dendritic cells as cellular vaccines. Adv Drug
Deliv Rev 2005; 57: 475–482.
76 Wang C, Ge Q, Ting D, Nguyen D, Shen HR,
Chen J, Eisen HN, Heller J, Langer R, Putnam
D: Molecularly engineered poly(ortho ester)
microspheres for enhanced delivery of DNA
vaccines. Nat Mater 2004; 3: 190–196.
77 Barratt-Boyes SM, Figdor CG: Current issues
in delivering DCs for immunotherapy. Cyto-
therapy 2004; 6: 105–110.
78 Bedrosian I, Mick R, Xu S, Nisenbaum H,
Faries M, Zhang P, Cohen PA, Koski G,
Czerniecki BJ: Intranodal administration of
peptide-pulsed mature dendritic cell vaccines
results in superior CD8+ T-cell function in
melanoma patients. J Clin Oncol 2003; 21:
3826–3835.
79 Garg S, Oran A, Wajchman J, Sasaki S, Maris
CH, Kapp JA, Jacob J: Genetic tagging shows
increased frequency and longevity of antigen-
presenting, skin-derived dendritic cells in vivo.
Nat Immunol 2003; 4: 907–912.
80 Randolph GJ: Dendritic cell migration to
lymph nodes: cytokines, chemokines, and lipid
mediators. Semin Immunol 2001; 13: 267–
274.
81 Scandella E, Men Y, Legler DF, Gillessen S,
Prikler L, Ludewig B, Groettrup M: CCL19/
CCL21-triggered signal transduction and mi-
gration of dendritic cells requires prostaglan-
din E
2
. Blood 2004; 103: 1595–1601.
82 Nair S, McLaughlin C, Weizer A, Su Z, Bocz-
kowski D, Dannull J, Vieweg J, Gilboa E: In-
jection of immature dendritic cells into adju-
vant-treated skin obviates the need for ex vivo
maturation. J Immunol 2003; 171: 6275–
6282.
83 Martin-Fontecha A, Sebastiani S, Hopken UE,
Uguccioni M, Lipp M, Lanzavecchia A, Sal-
lusto F: Regulation of dendritic cell migration
to the draining lymph node: impact on T lym-
phocyte traffi c and priming. J Exp Med 2003;
198: 615–621.
84 Crittenden MR, Thanarajasingam U, Vile RG,
Gough MJ: Intratumoral immunotherapy: us-
ing the tumour against itself. Immunology
2005; 114: 11–22.
85 Triozzi PL, Khurram R, Aldrich WA, Walker
MJ, Kim JA, Jaynes S: Intratumoral injection
of dendritic cells derived in vitro in patients
with metastatic cancer. Cancer 2000; 89: 2646–
2654.
Dendritic-Cell-Based Cancer
Immunotherapy
Skin Pharmacol Physiol 2006;19:124–131 131
86 Satoh Y, Esche C, Gambotto A, Shurin GV,
Yurkovetsky ZR, Robbins PD, Watkins SC,
Todo S, Herberman RB, Lotze MT, Shurin
MR: Local administration of IL-12-transfected
dendritic cells induces antitumor immune re-
sponses to colon adenocarcinoma in the liver
in mice. J Exp Ther Oncol 2002; 2: 337–349.
87 Nishioka Y, Hirao M, Robbins PD, Lotze MT,
Tahara H: Induction of systemic and therapeu-
tic antitumor immunity using intratumoral in-
jection of dendritic cells genetically modifi ed
to express interleukin 12. Cancer Res 1999; 59:
4035–4041.
88 Miller PW, Sharma S, Stolina M, Butterfi eld
LH, Luo J, Lin Y, Dohadwala M, Batra RK,
Wu L, Economou JS, Dubinett SM: Intratu-
moral administration of adenoviral interleu-
kin 7 gene-modifi ed dendritic cells augments
specifi c antitumor immunity and achieves tu-
mor eradication. Hum Gene Ther 2000; 11:
53–65.
89 Engering A, Geijtenbeek TB, van Kooyk Y:
Immune escape through C-type lectins on den-
dritic cells. Trends Immunol 2002; 23: 480–
485.
90 Bonifaz L, Bonnyay D, Mahnke K, Rivera M,
Nussenzweig MC, Steinman RM: Effi cient tar-
geting of protein antigen to the dendritic cell
receptor DEC-205 in the steady state leads to
antigen presentation on major histocompati-
bility complex class I products and peripheral
CD8+ T cell tolerance. J Exp Med 2002; 196:
1627–1638.
91 Diehl L, den Boer AT, Schoenberger SP, van
der Voort EI, Schumacher TN, Melief CJ, Of-
fringa R, Toes RE: CD40 activation in vivo
overcomes peptide-induced peripheral cyto-
toxic T-lymphocyte tolerance and augments
anti-tumor vaccine effi cacy. Nat Med 1999; 5:
774–779.
92 Grewal IS, Flavell RA: CD40 and CD154 in
cell-mediated immunity. Annu Rev Immu-
nol 1998; 16: 111–135.
93 Merad M, Sugie T, Engleman EG, Fong L: In
vivo manipulation of dendritic cells to induce
therapeutic immunity. Blood 2002; 99: 1676–
1682.
94
Okano F, Merad M, Furumoto K, Engleman
EG: In vivo manipulation of dendritic cells
overcomes tolerance to unmodifi ed tumor-as-
sociated self antigens and induces potent anti-
tumor immunity. J Immunol 2005; 174: 2645–
2652.
95 Biragyn A, Tani K, Grimm MC, Weeks S,
Kwak LW: Genetic fusion of chemokines to
a self tumor antigen induces protective, T-cell
dependent antitumor immunity. Nat Bio-
technol 1999; 17: 253–258.
96 Kumamoto T, Huang EK, Paek HJ, Morita
A, Matsue H, Valentini RF, Takashima A:
Induction of tumor-specifi c protective immu-
nity by in situ Langerhans cell vaccine. Nat
Biotechnol 2002; 20: 64–69.
97 Lori F, Trocio J, Bakare N, Kelly LM, Liszie-
wicz J: DermaVir, a novel HIV immunisation
technology. Vaccine 2005; 23: 2030–2034.
98 Ridgway D: The fi rst 1,000 dendritic cell vac-
cines. Cancer Invest 2003; 21: 873–886.
99 Colonna M, Trinchieri G, Liu YJ: Plasmacy-
toid dendritic cells in immunity. Nat Immu-
nol 2004; 5: 1219–1226.
100 Mohty M, Vialle-Castellano A, Nunes JA, Is-
nardon D, Olive D, Gaugler B: IFN-alpha
skews monocyte differentiation into Toll-like
receptor 7-expressing dendritic cells with po-
tent functional activities. J Immunol 2003;
171: 3385–3393.
101 Salio M, Cella M, Vermi W, Facchetti F, Pal-
mowski MJ, Smith CL, Shepherd D, Colonna
M, Cerundolo V: Plasmacytoid dendritic cells
prime IFN-gamma-secreting melanoma-spe-
cifi c CD8 lymphocytes and are found in pri-
mary melanoma lesions. Eur J Immunol
2003; 33: 1052–1062.
102 Gabriele L, Borghi P, Rozera C, Sestili P, An-
dreotti M, Guarini A, Montefusco E, Foa R,
Belardelli F: IFN-alpha promotes the rapid
differentiation of monocytes from patients
with chronic myeloid leukemia into activated
dendritic cells tuned to undergo full matura-
tion after LPS treatment. Blood 2004; 103:
980–987.
103 Vogl A, Sartorius U, Vogt T, Roesch A,
Landthaler M, Stolz W, Becker B: Gene ex-
pression profi le changes between melanoma
metastases and their daughter cell lines: im-
plication for vaccination protocols. J Invest
Dermatol 2005; 124: 401–404.
104 Sutmuller RP, Offringa R, Melief CJ: Reviv-
al of the regulatory T cell: new targets for drug
development. Drug Discov Today 2004; 9:
310–316.
105 Phan GQ, Yang JC, Sherry RM, Hwu P, To-
palian SL, Schwartzentruber DJ, Restifo NP,
Haworth LR, Seipp CA, Freezer LJ, Morton
KE, Mavroukakis SA, Duray PH, Steinberg
SM, Allison JP, Davis TA, Rosenberg SA:
Cancer regression and autoimmunity in-
duced by cytotoxic T lymphocyte-associated
antigen 4 blockade in patients with metastat-
ic melanoma. Proc Natl Acad Sci USA 2003;
100: 8372–8377.
106 Nair S, Boczkowski D, Moeller B, Dewhirst
M, Vieweg J, Gilboa E: Synergy between tu-
mor immunotherapy and antiangiogenic
therapy. Blood 2003; 102: 964–971.
... As the predominant antigen presenting cell, application of dendritic cell in cancer vaccines is a promising option for therapeutic vaccine development. Genetic modification of dendritic cells to express tumor antigens and/or immunomodulatory cytokines can improve their capacity to stimulate a strong antitumor immune response and subsequent tumor destruction (Arnouk 2012;Farkas et al. 2006). Herein, genetically engineered therapeutic cancer vaccines entering pharmaceutical market have been reviewed. ...
Article
Full-text available
Sulfur mustard (SM), also known as mustard gas, is a chemical weapon which by now has been used in many wars. The most concerning SM toxic effect is probable carcinogenicity. In this study, the genetic and epigenetic mechanisms of SM carcinogenicity, by focusing on treatment of SM-associated malignancies, particularly gene therapeutics, cancer vaccines, and epigenetic medications, have been criticized. The required data were collected through an organized search on valid scientific databases. For SM carcinogenicity due to acute or chronic exposure, the entire original and review articles were evaluated. In addition, studies on the therapeutic effects of available genetic and epigenetic medications were included. Currently, four gene therapeutics, two cancer vaccines with genetic bases, and seven epigenetic medications are available for cancer treatment. Genetic and epigenetic cancer treatments including Gendicine, Imlygic, Provenge, Cimavax-EGF, Azacitidine, Vorinostat, Romidepsin, and Belinostat will yield outstanding benefits for SM-exposed patients who suffer from cancer.
... As alluded to, DC-based vaccination and immunization strategies, based on the application of ex vivo antigen-loaded or genetically engineered autologous DCs to tune T-cell responses have meanwhile evolved into a viable therapeutic option for cancer. 92 This success has inspired several groups to explore the potential of DC-based vaccination for atherosclerosis. Studies in animal models so far are at least encouraging and support the notion that DC-based vaccination and immunization hold promise for therapeutic immunomodulation of atherosclerosis. ...
Article
Cardiovascular diseases (CVDs) are one of the leading causes of mortality worldwide.1 It has been so for decades, notwithstanding a wide array of – mostly preventive – treatment modalities targeting known risk factors, such as hyperlipidemia, type 2 diabetes mellitus, hypertension, or obesity. Recent technical and conceptual advances have unveiled important contributions of the immune system in the pathophysiology of a variety of CVDs such as atherosclerosis, ischemic stroke, chronic heart failure, and other myocardial conditions like myocardial ischemia and reperfusion, viral myocarditis, and cardiac transplantation.2–4 In many of these disorders, so-called danger-associated molecular patterns (DAMPs), released from necrotic tissue and dying cells, can lead to the activation of certain immune cell populations such as monocytes/ macrophages, granulocytes, and T cells, thus aggravating ongoing inflammatory processes at the lesion site. Dendritic cells (DCs) are key modulators of immunity, pivotal in directing innate and adaptive immune responses against microbial, viral, but also modified self-antigens present at the sites of injury. Given the tissue trauma underlying various CVDs, it is not surprising that recent observations have allocated a regulatory role for DCs in CVD-associated immune responses. Interestingly, nondiseased arteries of young individuals were seen to host a network of resident vascular DCs (CD1a+ S100+ lag+ CD31− CD83− CD86−),5 representing a phenotype related to Langerhans cells in the skin. In agreement, monocyte-derived CD11c+ CD68+ dendritic cells could be detected in the atherosclerosis-prone lesser curvature and aortic sinus in inbred atherosclerosis-susceptible (C57Bl/6), but not resistant mouse strains (balb/c).6 Murine vascular resident DCs express an immature phenotype with low expression of costimulatory molecules, and are present in the subendothelial space with occasional probing into the vascular lumen. DCs have …
... É provável que, nestas condições, respondam de forma anómala, o que, entre outros efeitos, pode provocar a autodestruição dos linfócitos 10 . É portanto necessário prosseguir os estudos e melhorar as técnicas, para que a produção de vacinas terapêuticas a partir de cultura de células dendríticas possa vir a ser, com sucesso, uma rotina no tratamento do doente oncológico 37 . Em conclusão, por tudo o que foi exposto, podemos afirmar que o desenvolvimento de modelos experimentais utilizando a cultura de células tem dado um importante contributo para a melhor compreensão da oncogénese, nomeadamente no cancro do pulmão, permitindo um diagnóstico precoce e a instituição de terapêuticas dirigidas e mais eficazes. ...
Article
Full-text available
In the beginning of the 20th century, tissue culture was started with the aim of studying the behaviour of animal cells in normal and stress conditions.The cell study at molecular level depends on their capacity of growing and how they can be manipulated in laboratory. In vitro cell culture allows us the possibility of studying biological key processes, such as growth, differentiation and cell death, and also to do genetic manipulations essential to the knowledge of structure and genes function.Human stem cells culture provides strategies to circumvent other models’ deficiencies.It seems that cancer stem cells remain quiescent until activation by appropriated micro-environmental stimulation.Several studies reveal that different cancer types could be due to stem cell malignant transformations. Removal of these cells is essential to the development of more effective cancer therapies for advanced disease. On the other hand, dendritic cells modified in culture may be used as a therapeutic vaccine in order to induce tumour withraw
... At present we show that the use of oxLDL-pulsed DCs resulted in a 92% or 87% reduction in lesion size in the carotid arteries compared to the saline or the mDCs-treated control groups, respectively. Ex-vivo generated and antigen-loaded DCs have been used in vaccination protocols in many animal models 34,35 and have been used to improve immunity in cancer 36 and HIV-infected patients. 37,38 Since the initial study of Palinski 39 , a number of studies have shown the effectiveness of immunization with oxLDL or apoB100 peptides as an immunotherapy for atherosclerosis in experimental animal models leading to a reduction in atherosclerosis ranging from 40 to 70% in mice and rabbits. ...
Article
Full-text available
Abstract Background. Modification of lipoproteins plays an important role in the development,of atherosclerosis. Oxidatively modified low-density lipoprotein (oxLDL) has a number of pro-inflammatory effects, whereas immunization,with various forms of oxLDL is able to reduce,atherosclerosis. The uptake of modified LDL by dendritic cells (DCs) and the presentation of epitopes thereof may form an important step in the immunomodulatoryeffects of LDL. In this study we transferred oxLDL-pulsed DCs to LDLr, mice ,every other day before induction of atherosclerosis by Western-type diet feeding. Transfer of oxLDL-pulsed DCs resulted in a 92% and 87% reduction in carotid artery lesion size compared to the PBS-treated or mature DCs-treated groups, respectively (P< 0.001). Lesion size in the aortic valves was not affected but we observed,at both sites an increase in plaque stability. The reduction in atherosclerosis was accompanied,by a 3.8 fold increase (P< 0.05) in Cu-oxLDL specific IgG levels whereas the levels of anti-malondialdehyde-LDL (MDA-LDL) specific IgG and IgM were not significantly affected. In addition, we also showed that the sera of mice treated with oxLDL-pulsed DCs reduced the formation of foam cells as compared to sera from PBS or mDCs- treated mice. Conclusions. Weconclude,that oxLDL-pulsed DCs induce an enhanced production of anti-oxLDL IgG
... Dendritic cells (DCs) initiate primary and memory immune responses as well as activate innate immunity and therefore, play a pivotal role in immunotherapy [1]. Accounting for only 0.02-0.2% of the total white blood cells, the number of DCs that can be isolated from peripheral blood is limited [2]. ...
Article
Full-text available
Dendritic cells (DCs) play a key role in innate and adaptive immunity but the access to sufficient amount of DCs for basic and translational research has been limited. We established a novel ex vivo system to develop and expand DCs from hematopoietic stem/progenitor cells (HPCs). Both human and mouse HPCs were expanded first in feeder culture supplemented with c-Kit ligand (KL, stem cell factor, steel factor or CD117 ligand), Flt3 ligand (fms-like tyrosine kinase 3, Flt3L, FL), thrombopoietin (TPO), IL-3, IL-6, and basic fibroblast growth factor (bFGF), and then in a second feeder culture ectopically expressing all above growth factors plus GM-CSF and IL-15. In the dual culture system, CD34+ HPCs differentiated toward DC progenitors (DCPs), which expanded more than five orders of magnitude. The DCPs showed myeloid DC surface phenotype with up-regulation of transcription factors PU.1 and Id2, and DC-related factors homeostatic chemokine ligand 17 (CCL17) and beta-chemokine receptor 6 (CCR6). Multiplex ELISA array and cDNA microarray analyses revealed that the DCPs shared some features of IL-4 and IL-15 DCs but displayed a pronounced proinflammatory phenotype. DCP-derived DCs showed antigen-uptake and immune activation functions analogous to that of the peripheral blood-derived DCs. Furthermore, bone marrow HPC-derived DCP vaccines of tumor-bearing mice suppressed tumor growth in vivo. This novel approach of generating DCP-DCs, which are different from known IL-4 and IL-15 DCs, overcomes both quantitative and qualitative limitations in obtaining functional autologous DCs from a small number of HPCs with great translational potential.
... 6,16,17 We have used autologous plasma in our cultures, which will allow the elimination of the xenogenic components, an important aspect of GMP protocols. 15,23 Lineage analyses of the expanded cells showed that the majority of them belonged to the myeloid lineage and there was a preferential lineage commitment towards CD14+ cells. CD34+ cells differentiate along two independent pathways that consist of CD1a+ cells and CD14+ cells that give rise to Langerhans DCs and interstitial DCs, respectively. ...
Article
Full-text available
Advances in the past two decades in dendritic cell (DC) biology paved the way to exploit them as a promising tool in cancer immunotherapy. The prerequisite for DC vaccine preparations is large-scale in vitro generations of homogeneous, mature, and functional DCs. Frequent improvements are being made in the existing in vitro DC production protocols to achieve this goal. In our previous study we reported a large-scale generation of mature, functional DCs from umbilical cord blood (UCB) CD34+ cells. Here we report that this method can be used for the efficient generation of DCs from UCB mononuclear cells (MNCs) and thus the hematopoietic stem cell isolation step is not essential. MNCs or CD34+ cells isolated from the same cord blood (CB) samples were used for the generation of DCs. DCs were characterized for morphology, phenotype, and functional assays including antigen uptake, chemotaxis, and mixed leukocyte reaction. Similarly DCs generated from the MNCs of same fresh and frozen CB units were compared. The morphologic, phenotypic, and functional characterization of the DCs generated from various sets show that they were comparable in nature irrespective of the starting population used. We conclude that the CD34+ isolation step is not essential for the generation of mature, functional DCs and thus can be eliminated. More importantly, we show that DCs can be generated with equal efficiency from the MNCs of frozen CB units. Our culture method will be useful for exploiting the potential of UCB as an additional source for allogeneic DCs in the clinical settings.
Article
Full-text available
Lentivectors are potential vaccine delivery vehicles because they can efficiently transduce a variety of non-dividing cells, including antigen-presenting cells, and do not cause expression of extra viral proteins. To improve safety while retaining efficiency, a dendritic cell (DC)-specific lentivector was constructed by pseudotyping the vector with an engineered viral glycoprotein derived from Sindbis virus. We assessed the level of anti-tumor immunity conferred by this engineered lentivector encoding the melanoma antigen gp100 in a mouse model. Footpad injection of the engineered lentivectors results in the best antigen-specific immune response as compared with subcutaneous and intraperitoneal injections. A single prime vaccination of the engineered lentivectors can elicit a high frequency (up to 10%) of gp100-specific CD8(+) T cells in peripheral blood 3 weeks after the vaccination and this response will be maintained at around 5% for up to 8 weeks. We found that these engineered lentivectors elicited relatively low levels of anti-vector neutralizing antibody responses. Importantly, direct injection of this engineered lentivector inhibited the growth of aggressive B16 murine melanoma. These data suggest that DC-specific lentivectors can be a novel and alternative vaccine carrier with the potential to deliver effective anti-tumor immunity for cancer immunotherapy.
Article
This study seeks to assess the use of labeling with micron-sized iron oxide (MPIO) particles for the detection and quantification of the migration of dendritic cells (DCs) using cellular magnetic resonance imaging (MRI). DCs were labeled with red fluorescent MPIO particles for detection by cellular MRI and a green fluorescent membrane dye (PKH67) for histological detection. MPIO-labeled DCs or unlabeled control DCs were injected into mice footpads at two doses (0.1 × 10(6) and 1 × 10(6)). Images were acquired at 3 Tesla before DC injection and 2, 3, and 7 days post-DC injection. Labeling DCs with MPIO particles did not affect viability, but it did alter markers of DC activation and maturation. MRI and fluorescence microscopy allowed for the detection of MPIO-labeled DCs within the draining popliteal nodes after their injection into the footpad. This paper presents the first report of the successful use of fluorescent MPIO particles to label and track DC migration.
Article
Full-text available
Dendritic cells (DCs) are antigen-presenting cells with a unique ability to induce primary immune responses. DCs capture and transfer information from the outside world to the cells of the adaptive immune system. DCs are not only critical for the induction of primary immune responses, but may also be important for the induction of immunological tolerance, as well as for the regulation of the type of T cell–mediated immune response. Although our understanding of DC biology is still in its infancy, we are now beginning to use DC-based immunotherapy protocols to elicit immunity against cancer and infectious diseases.
Article
The idiotype (Id) determinant on the multiple myeloma (MM) protein can be regarded as a tumor-specific marker. Immunotherapy directed at the MM Id may stem the progression of this disease. We report here on the first 12 MM patients treated at our institution with high-dose therapy and peripheral blood stem cell transplantation (PBSCT) followed by Id immunizations. MM patients received PBSCT to eradicate the majority of the disease. PBSCT produced a complete response in 2 patients, a partial response in 9 patients and stable disease in 1 patient. Three to 7 months after high-dose therapy, patients received a series of monthly immunizations that consisted of two intravenous infusions of Id-pulsed autologous dendritic cells (DC) followed by five subcutaneous boosts of Id/keyhole limpet hemocyanin (KLH) administered with adjuvant. Between 1 and 11 × 106 DC were obtained by leukapheresis in all patients even after PBSCT. The administration of Id-pulsed DC and Id/KLH vaccines were well tolerated with patients experiencing only minor and transient side effects. Two of 12 patients developed an Id-specific, cellular proliferative immune response and one of three patients studied developed a transient but Id-specific cytotoxic T-cell (CTL) response. Eleven of the 12 patients generated strong KLH-specific cellular proliferative immune responses showing the patients’ immunocompetence at the time of vaccination. The two patients who developed a cellular Id-specific immune response remain in complete remission. Of the 12 treated patients, 9 are currently alive after autologous transplantation with a minimum follow-up of 16 months, 2 patients died because of recurrent MM and 1 patient succumbed to acute leukemia. These studies show that patients make strong anti-KLH responses despite recent high-dose therapy and that DC-based Id vaccination is feasible after PBSCT and can induce Id-specific T-cell responses. Further vaccine development is necessary to increase the proportion of patients that make Id-specific immune responses. The clinical benefits of Id vaccination in MM remain to be determined.
Article
Dendritic cells (DCs) represent a family of ontogenically distinct leukocytes involved in immune response regulation. The ability of DCs to stimulate T-cell immunity has led to their use as vectors for immunotherapy vaccines. However, it is unclear whether and to what degree in vitro–generated DCs are representative of DCs that develop in vivo. Treatment of mice with human Flt3 ligand (FL) dramatically increases the number of DCs. We report here that administration of FL to healthy human volunteers increased the number of circulating CD11c+ IL-3Rlow DC (mean 44-fold) and CD11c− IL-3Rhigh DC precursors (mean 12-fold). Moreover, the CD11c+ DCs were efficient stimulators of T cells in vitro. Thus, FL can expand the number of circulating, functionally competent human DCs in vivo.
Article
Immunization with defined tumor antigens is currently limited to a small number of cancers where candidates for tumor rejection antigens have been identified. In this study we investigated whether pulsing dendritic cells (DC) with tumor-derived RNA is an effective way to induce CTL and tumor immunity. DC pulsed with in vitro synthesized chicken ovalbumin (OVA) RNA were more effective than OVA peptide-pulsed DC in stimulating primary, OVA-specific CTL responses in vitro. DC pulsed with unfractionated RNA (total or polyA+) from OVA-expressing tumor cells were as effective as DC pulsed with OVA peptide at stimulating CTL responses. Induction of OVA-specific CTL was abrogated when polyA+ RNA from OVA-expressing cells was treated with an OVA-specific antisense oligodeoxynucleotide and RNase H, showing that sensitization of DC was indeed mediated by OVA RNA. Mice vaccinated with DC pulsed with RNA from OVA-expressing tumor cells were protected against a challenge with OVA-expressing tumor cells. In the poorly immunogenic, highly metastatic, B16/F10.9 tumor model a dramatic reduction in lung metastases was observed in mice vaccinated with DC pulsed with tumor-derived RNA (total or polyA+, but not polyA- RNA). The finding that RNA transcribed in vitro from cDNA cloned in a bacterial plasmid was highly effective in sensitizing DC shows that amplification of the antigenic content from a small number of tumor cells is feasible, thus expanding the potential use of RNA-pulsed DC-based vaccines for patients bearing very small, possibly microscopic, tumors.
Article
Hybrid cell vaccination with cell fusion products (CFPs) of autologous tumor cells and mature allogenic MHC II bearing dendritic cells has been described to induce cytotoxic T lymphocyte (CTL)-mediated immune responses. The aim of this study was to assess safety, antitumor activity, and immune responses of a CFP-vaccine in patients with disseminated malignant melanoma. In a phase I/II study, we treated 11 patients by monthly intracutaneous or subcutaneous application of a CFP vaccine generated by electrofusion of autologous melanoma cells with mature allogenic dendritic cells. In addition, patients received subcutaneous low-dose interleukin-2 injections for 6 days after each vaccination. No serious adverse effects were observed. Ten patients showed progressive disease and one patient had a short-lasting stable disease. None of the patients developed a positive delayed-type hypersensitivity reaction against irradiated autologous melanoma cells. In 2 patients, who were monitored in more detail, we found no evidence of induction of a specific antimelanoma T-cell response by analyzing the proliferation, cytokine secretion, and cytotoxicity of their T cells toward autologous melanoma cells. No unequivocal beneficial effects of the used CFP vaccine could be demonstrated.
Article
Interleukin-12 p70 (IL-12p70) heterodimer, composed of p35 and p40 subunits, is a major Th1-driving cytokine, promoting cell-mediated immunity. in contrast, IL-12p40 homodimer, secreted by APC in the absence of p35 expression, and free p40 monomer do not mediate IL-12 activity but act as IL-12 antagonists. Here it is reported that prostaglandin E-2 (PGE(2)), an inflammatory mediator with a previously known Th2-driving function, dose-dependently enhances the IL-12p40 mRNA expression and the secretion of IL-12p40 protein in human tumor necrosis factor-alpha (TNF alpha)-stimulated immature dendritic cells (DCs). This effect is selective and is not accompanied by the induction of IL-12p35 expression or by secretion of IL-12p70 heterodimer, Inability of TNF alpha /PGE(2) to induce IL-12p70 was not compensated by interferon gamma (IFN gamma), which strongly enhanced the lipopolysaccharide (LPS)-induced IL-12p70 production. In addition to the selective induction of IL-12p40 in TNF alpha -stimulated Dos, PGE(2) inhibited the production of IL-12p70 and IL-12p40 in DCs stimulated with LPS or CD40 ligand, These data suggest an additional level of the Th2-promoting activity of PGE(2), via selective induction of IL-12p40, Selective induction of IL-12p40 and suppression of bioactive IL-12p70 may have negative impact on anticancer vaccination with PGE(2)-matured DCs, (Blood.2001;97:3466-3469) (C) 2001 by The American Society of Hematology.
Article
The outcome of antigen recognition by naive CD8+ cytotoxic T lymphocytes (CTLs) in the periphery is orchestrated by CD4+ T-helper cells, and can either lead to priming or tolerization. The presence of T-helper cells favors the induction of CTL immunity, whereas the absence of T-helper cells can result in CTL tolerance. The action of T helper cells in CTL priming is mediated by CD40−CD40 ligand interactions. We demonstrate here that triggering of CD40 in vivo can considerably enhance the efficacy of peptide-based anti-tumor vaccines. The combination of a tolerogenic peptide vaccine containing a minimal essential CTL epitope with an activating antibody against CD40 converts tolerization into strong CTL priming. Moreover, CD40 ligation can provide an already protective tumor-specific peptide vaccine with the capacity to induce therapeutic CTL immunity in tumor-bearing mice. These findings indicate that the CD40−CD40 ligand pair can act as a 'switch', determining whether naive peripheral CTLs are primed or tolerized, and support the clinical use of CD40-stimulating agents as components of anti-cancer vaccines.
Article
Hybrid cell vaccination is a new cancer immune therapy approach that aims at recruiting T cell help for the induction of tumour specific cytolytic immunity. The vaccines are generated by fusion of the patients' tumour cells with allogeneic MHC class II bearing cells to combine the tumour's antigenicity with the immunogenicity of allogeneic MHC molecules. Safety and anti-tumour activity of this treatment were assessed in a clinical trial that has yielded one complete and one partial remission, and 5 cases of stable disease among 16 patients with advanced stage metastatic melanoma. As evidenced by histology, the vaccination induced T cell relocation into tumour nodules. Stable disease could be maintained by repeated booster injections for more than 24 months in some patients. The side effects were minor. Occasional occurrences of vitiligo spots after vaccination were indicative of a restricted therapy induced auto-immune reactivity. The results suggest that hybrid cell vaccination is a safe cancer immune therapy potentially effective for induction of acute anti-tumour response as well as long-term maintenance. Int. J. Cancer 85:618–626, 2000. © 2000 Wiley-Liss, Inc.