You are on page 1of 14

Indian Journal of Experimental Biology

Vol 45, June, 2007, pp 491-504

Review Article

Dendritic cells and antigen trapping technology ⎯ A revolution in


vaccine/immunotherapy strategy
Chiranjib Pal1, Dipyaman Ganguly2, Kausik Paul3 & Srijib Pal4
1
Department of Zoology, Brahmananda Keshab Chandra College, 111/2, B.T. Road, Bonhooghly, Kolkata 700 108, India
2
Graduate School of Biomedical Sciences, University of Texas at Houston, Health Science Centre, Houston, TX, USA
3
Indian Institute of Chemical Biology, Kolkata 700 032, India
4
Sanjiivani Medical Institute, Kolkata700 108, India

Vaccines based on dendritic cells—the immune system’s key responders to foreign invaders— grabbed the spotlight of
this decade. Scientists have devised a dozen different ways to make dendritic cell vaccines. They have linked dendritic cells
with all kinds of antigens, including peptides derived from gene mutations, tumor/pathogen RNA, viral vectors, and with
whole pathogen/tumor lysate. And they are adding cytokines such as granulocyte macrophage colony stimulating factor or
interleukin 4 during dendritic cell growth or maturation or at the site of vaccination to try to boost response. We are still
learning the best way to generate the dendritic cells, load them with the antigen and send them to the right place in the body,
and use of the biological stage of development of dendritic cells that is best suited to stimulate a response. In the present
review attempts have been made to present a comprehensive synopsis of the history, development and ramifications of
evolving knowledge on dendritic cell biology and the prospects for being developed as a rational immunotherapeutic tool.
Further clinical studies are warranted.

Keywords: Cancer, Dendritic cell (DC), Infectious diseases, Immunotherapy, Viral Diseases, Vaccine

Host defense relies on a concerted action of both antigens to the cells of adaptive immune system. Apart
antigen (Ag)-nonspecific innate immunity and Ag- from being highly immunogenic, DC have been shown
specific adaptive immunity. Immunology has long to play an important role in peripheral tolerance, as
been focused on antigens and lymphocytes, but the well as for the regulation of the type of T cell mediated
mere presence of these two parties do not always lead immune responses and for the long lasting
to a perfect immune response. A third party, the immunological memory. This review for the most part
dendritic cells (DC) are found to be the key initiator of it concentrates on the developing strategies of
and modulator of the primary immune response after immunotherapy by using the dendritic cells.
the discovery of a novel cell type in peripheral
lymphoid organs of mice by Steinman in 19731. Heterogeneity of dendritic cell subsets
Mice
Evolutionary pressure has led to the development of
At least two distinct pathways of DC development
adaptive immunity, the key features of which are, (a)
have been identified in mice. Myeloid precursors give
the ability to rearrange genes of the immunoglobin
rise to myeloid DC in presence of granulocyte
family, permitting creation of large diversity of Ag-
macrophage colony stimulating factor2,3. CD8α+
specific clones and, (b) immunological memory. Yet
lymphoid DC can also arise from lymphoid
this highly sophisticated and potent system needs to
precursors4-8. Transfer of a population of purified
be instructed and regulated by Ag-presenting cells.
lymphoid precursors into irradiated host results
Dendritic cells are a fraction of matured and
terminally differentiated antigen presenting cells development of DC that express CD8α. Lymphoid
found to be generated from different progenitors with DC are localized in the T cell rich areas of the
myeloid or lymphoid commitments, with a unique periarteriolar lymphatic sheaths (PALS) in spleen and
ability to induce primary immune response. DC lymph nodes6, 9-11. In contrast myeloid DC are in the
captures and transfer information from the exogenous marginal zone bridging channels of the spleen9-11.
Both subsets express high levels of CD11c, MHC II
_______________ and the costimulatory molecules CD86 and CD40.
*Telephone: 033-2577 2486 (O),
E-mail: chiranjibpal@yahoo.co.in, Lymphoid DC make higher level of IL-12 and less
cpcu.immunology@gmail.com
492 INDIAN J EXP BIOL, JUNE 2007

phagocytic than the myeloid DC9. responses, DC also participate in the presentation of
Human self-antigen during the course of central and
DC heterogeneity in humans is reflected in all the peripheral T cell tolerance induction via various
four parameters of anatomical localization, mechanisms, including activation of T regulatory
progenitors, functional properties and type of immune (Treg) cells, induction of T cell anergy, and TH1/TH2
response induced12-15. polarization. The exact mechanisms involved in the
(a) Anatomical localization⎯ Depending on decision of a DC to become immunogenic or
anatomical localization the DC are classified as the tolerogenic have not been elucidated, however,
skin epidermal langerhans cells (LC), dermal DC upcoming scientific reports suggests that DC function
(interstitial DC), splenic marginal DC, T-zone is dependent on their maturation stage16.
interdigitating cells, germinal-center DC, thymic DC,
liver DC and blood DC. Recruitment and maturation: The phenotypic
(b) Precursor populations⎯ For instance, in nuances
human at least two subsets of DC precursors circulate Newly generated DC precursors migrate,
in the blood, CD14+CD11c+ monocytes and lineage- presumably through blood stream, from the bone
negative LINnegCD11c-IL3Rα+ precursors DC. The marrow to non-lymphoid tissues, where they
LINnegCD11c+ cells may represent a third precursor, eventually become resident cells. Circulating DC
although these cells are more committed because they precursors accumulate rapidly (within an hour) at the
can spontaneously differentiate into DC when put into sites of antigen deposition in response to the
culture. production of chemokines upon local inflammation as
(c) Function⎯ Precursors of DC correlate with the demonstrated in bronchial epithelium after Ag
functional diversity evidenced from in vitro inhalation17-19. CD34+ hematopoietic cell derived
generation of different subtypes of human DC. CD34+ immature DC expresses CCR6, whose ligand, MIP3α,
precursors from umbilical cord blood and adult bone
marrow cultured in presence of GM-CSF and TNF-α
give rise to functional CD1a+CD14+ Langerhans cells.
CD34+Lin-CD10+ lymphoid restricted precursors
isolated from bone marrow and cultured with
cytokines give rise to DC equivalent to murine
lymphoid-derived DC. Blood monocytes in presence
of cytokines GM-CSF and IL-4, for growth and TNF-
α, for maturation give rise to mature DC type 1
(DC1). CD45RAhiCD11cloIL3Rhi plasmacytoid
precursors from blood, tonsil and thymus in presence
of IL-3 give rise to CD11clo mature DC type 2 (DC2)
(Fig. 1). Precursor DC1 cells produce TNF-α and IL-
6, whereas the precursor DC2 cells rapidly produce
IFN-α and IFN-β in response to the appropriate
microbial stimuli. DC1 cells are found to prime T
cells to produce a TH1 response which is associated
with high production of IL-12 in response to the
appropriate stimuli. In contrast DC2 induce the TH2
response.
(d) Outcome of immune response⎯ The final Fig. 1 ⎯ Pathways of human dendritic cell (DC) development.
outcome of immune response refers to the induction These pathways were deduced from the culture studies in different
of tolerance or immunity. Dendritic cells are experimental conditions67-79. Some DC lineages require
professional APC that play a key role in initiating exogenous stimuli for full activation. Other DC lineages probably
remain at a precursor state in an uninfected individual, because
effector T cell responses in secondary lymphoid they require stimulation by microbial products to produce mature
organs. Equally important to the initiation of effector DC. IPC, Interferron producing Cell; pDC, Precursor of DC;
CLA, Cutaneous lymphocyte associated antigen.
PAL et. al.: DENDRITIC CELL BASED VACCINATION & IMMUOTHERAPY 493

appears to be the most powerful chemokine guiding


their migration20, 21.
Immature DC are very efficient in Ag capture and
can use several pathways ─ (a) macropinocytosis, (b)
receptor-mediated endocytosis via C-type lectin
receptors (mannose receptor, DEC-205) or Fcγ
receptor type I (CD64) and II (CD32), (c)
phagocytosis of particles such as latex bead, apoptotic
and necrotic cell fragments (involving CD36 and
αvβ3 or αvβ5 integrins), viruses, and bacteria
including mycobacteria, as well as intracellular
parasites such as Leishmania major22.
The antigen or pathogen induces the immature DC
to undergo phenotypic and functional changes that
culminate in the complete transition from the Ag-
capturing cell to APC. DC maturation is intimately
linked with their migration from the peripheral tissue
to the draining lymphoid organs, and therefore these
are the two key events in the life span of DC.
Numerous factors induce and regulate DC maturation
(Fig. 2). The maturation process is associated with
several coordinated cellular events such as (a) loss of
endocytic/phagocytic receptors, (b) upregulation of
costimulatory molecules CD40, CD80, CD83, CD86,
(c) shift in lysosomal compartments with
downregulation of CD68 and upregulation of DC-
lysosome associated membrane protein (DC-LAMP),
(d) changes in MHC II compartments and (e) change
in morphology.
Mature DC downregulate CCR6, the receptor for
MIP3α, consequently escape from local gradient of
chemokines, upregulate the chemokine receptor CCR7
and accordingly acquire responsiveness to MIP3β
(ELC, Exodous 3) and 6Ckine [secondary lymphoid
tissue chemokine (SLC), Exodus2]. Consequently,
maturing DC leave the inflamed tissue and enter the
lymph stream, potentially directed by 6 Ckine that is
expressed on lymphatic vessel. Maturing DC entering
the draining lymph node are driven into the paracortical
areas in response to the production of MIP3β and Fig. 2 ⎯ Maturation of DC. The factors inducing progression
6Ckine by cells spread over the T cell zone. from one stage to another and the properties of each
differentiation/maturation stage.

The decision-makers: Activating the T cells to immunotherapies can be enhanced by the inclusion of
shape the immune response ligands that bind to these cell membranes. The
The ability to prime naïve T cells constitutes a enhanced versatility of new vector systems allows the
unique and critical function of DC both in vitro and in combinatorial construction of immunotherapies that
vivo. Because virtually all signals to T cells begin at contain elements that target several points in the
the cell membrane, the efficacy of vaccines and other pathway of T cell activation23.
At the simplest level, T cell activation requires two
494 INDIAN J EXP BIOL, JUNE 2007

signals. Signal 1 is delivered through the T-cell breakthrough came in mid 90’s when Hsu et. al.
receptor (TCR) after engagement by a peptide-MHC successfully vaccinated patients with B-cell lymphoma
complex. Signal 2, the costimulatory signal, was using autologous antigen-pulsed dendritic cells25 and in
thought originally to be coming from CD80 (B7-1) case of inactivated influenza viruses, when presented
interacting with CD28 on T cell. The costimulatory on dendritic cells in vitro, were shown to elicit human
molecules/signals so far identified, fall into three cytolytic CD8+ T cell responses26.
families ─ the B7 family, the tumor-necrosis factor The accessibility of DC in the immune system,
(TNF) receptor and cytokines. The B7 family together with its central role in so many disease
currently comprises six members with defined processes, makes it highly appropriate for therapeutic
costimulatory activity. The founding members CD80 manipulations. Until recently, immunotherapies tested
(B7-1) and CD86 (B7-2), bind to the activating clinically have been fairly crude, failing to take
receptor of T cell, CD28, and a counter regulatory advantage of the molecular pathways that regulate
inhibitory receptor, cytotoxic T-lymphocyte antigen 4 immunity. The elucidation of specific molecules that
(CTLA4). Newer B7 family members, B7-H1/PDL1 induce DC proliferation and maturation has provided
and B7-DC/PDL2, have been reported to have both immunologists with an important tool for the
costimulatory and inhibitory effect. Their inhibitory engineered vaccines with enhanced therapeutic
activity is probably mediated through the potency. Recent studies have also shed light on the
programmed cell death 1 (PD1) receptor, which pathogenic roles of DC in various diseases such as
contains an immunoreceptor tyrosine-based inhibitory autoimmunity, allergy, transplantation, infection and
motif (ITIM). B7RP-1, which binds to the activating cancer. This review discusses the ways in which
receptor inducible costimulatory molecule (ICOS), molecular and cell biological insights into immune
seems to be particularly important in T cell-B cell regulation are being applied to design strategies for
interaction and antibody production. The receptor for both quantitative and qualitative improvement of
B7H3 has not been identified yet. TNF receptor immunotherapy for diseases.
families are upregulated on T cell activation,
indicating that the role of this family might be to Engineered dendritic cells: Way to customized
amplify responses once T cell activation has occurred. vaccines
T cells can also interact with DC via CD40 ligand- The most common target of active immunotherapy
CD40 signaling pathway leading to increased strategies is the enhancement or modulation of the
expression of CD80/CD86 and cytokine release (IL-1, function of antigen-presenting cells (APC). This
TNF, Chemokines and IL-12). Engagement of strategy is based on the concept that the quantitative
RANK, a member of TNFR family by its ligand and qualitative characteristics of a T cell response to
(RANKL/TRANCE) expressed on activated T cells, an antigen depend on the signals that the T cells
stimulates the secretion of cytokines like IL-1, IL-6 receive from the APC. DC are the most potent type of
and IL-12 by DC. Finally cytokines such as IL-12 are APC and virtually all stages of DC development and
very important in determining the direction of T cell function can be modulated by engineered vaccines.
development i.e., TH1 versus TH2-as a consequence of GM-CSF and other cytokines, such as FLT3 ligand
T cell stimulation. Once these complex interactions and IL-4 that are mitogenic or co-mitogenic, induce
and signaling outcomes are defined, it should be an intermediate stage of DC differentiation that is
possible to build multiple costimulatory signals into characterized by the efficient uptake and processing
vaccines in an appropriate combinatorial fashion. of antigen27. DC maturation is dependent on binding
of two classes of receptors present on DC, the Toll
Prospects of DC based vaccination and like receptor (TLR) and Tumor necrosis factor
immunotherapy: Experimental successes and receptor (TNFR) families. TLRs are pattern
clinical experiences recognition receptors, which bind common chemical
DC have been chosen as one of the key factors for moieties that are expressed by pathogens and known
vaccine research from late 1980s when it was found as pathogen associated molecular patterns (PAMPs)
that human T cell shows proliferative responses to such as lipopolysaccharide (LPS) and unmethylated
particulate microbial antigens and are supported by cell CpG DNA sequences. DC have been shown to be
populations enriched for dendritic cells24. The responsible for the capacity of CpG
PAL et. al.: DENDRITIC CELL BASED VACCINATION & IMMUOTHERAPY 495

oligodeoxynucleotides to act as an adjuvant for AdhTERT-transfected DC were able to generate CTL


vaccine against Leishmania major in mice28. responses and these CTLs primed against hTERT
The discovery of specific receptors on DC that are exhibited killing of telomerase positive but not
responsible for receptor mediated endocytosis has telomerase negative tumor lines of diverse tissue
enabled the modification of antigens, so that they can origins. This study revealed the fact that hTERT gene,
be more efficiently bound to these DC uptake particularly as delivered via the recombinant
receptors. Indeed, antigen-GM-CSF fusion protein is adenovirus, may be useful as a vaccine to induce
an example of potential DC targeting29 because GM- specific T-cell-mediated tumor immunity in cancer
CSF not only stimulates DC proliferation, but might patients34. Use of adenovirus and retrovirus as vector
also act to target the antigen to endosomal for the transduction of DC during vaccine engineering
compartments by binding to the GM-CSF receptor. has been favoured in several recent studies.
Fusion of antigen and immunoglobin Fc region Immunization of retrovirus-transfected DC induces
enhanced the Fc receptor (FcR)-mediated antigen specific cytotoxic T lymphocytes for two distinct
uptake by APC. DNA vaccine encoding an E7-HSP70 malarial peptides presented by Kd molecule35. DC
fusion protein was 30-fold more effective than the transfected with an adenovirus vector encoding IL-12
wild type E7 protein in generating T cell response30. is a potent vaccine for invasive pulmonary
The two best characterized endogenous DC aspergillosis, a fungal infection36. DC are uniquely
maturation factors of the TNF family are TNF-α and able to decode the fungus-associated information and
CD40 ligand (CD40L). In vivo ligation of CD40 translate it in qualitatively different T helper (TH)
enhances priming against the endogenous tumor immune responses, in vitro and in vivo37. DC sense
antigen and promotes CD8+ T cell effector function fungi in a morphotype-specific manner, through the
in SV40 T antigen transgenic mice31. engagement of distinct recognition receptors
The ability to culture DC ex vivo has led to ultimately affecting cytokine production and
numerous studies using ex vivo antigen loaded DC as costimulation. Adoptive transfer of different types of
vaccine against tumor and infectious diseases. The DC activates protective and non-protective TH cells as
incorporation of GM-CSF, IL-12 or its encoding gene well as regulatory T cells and affects the outcome of
into recombinant protein, DNA or viral vaccines has the infections. DC transfected with fungal RNA also
also been shown to significantly enhance restore antifungal resistance in hematopoietic
immunization. Even Dendritic cells engineered to transplantation. Thus, the remarkable functional
express the FLT3 ligand stimulate type I immune plasticity of DC in response to fungi can be exploited
response, and induce enhanced cytotoxic T and for the deliberate targeting of cells and pathways of
natural killer cell cytotoxicities and anti-tumor cell-mediated immunity in response to DC based
immunity32. fungal vaccines37.
Human telomerase reverse transcriptase (hTERT)
is the catalytic component of a functional telomerase Use of DC in vaccination strategies against
complex, which is important in maintaining cell infections
immortality33. In most normal human adult cells, the Infectious disease is one of the leading causes of
expression of telomerase is very low and/or transient. human death. Particularly in the developing world,
In contrast, almost 90% of human tumors express a malaria, infectious diarrhea, tuberculosis,
relatively high level of telomerase implying the leishmaniasis and many more illness are still existing
possibility of using hTERT as a universal candidate and contributing to significant suffering as well as
tumor antigen. Human monocyte-derived dendritic mortality. For a time infectious diseases were thought
cells (DC) lack telomerase activity. Similar to other to be under control but at present appearance of AIDS
normal somatic cells, DC express the RNA (hTR) and other infectious diseases including leishmaniasis
component but not the catalytic component, hTERT. is the major threat for our survival. Ahuja et al.
Telomerase activity could be reconstituted using successfully transferred adoptively the auotologous
either lipid-mediated transfection of the hTERT DC engineered to secrete IL-12 (IL-12 transfected)
plasmid DNA or transfection of an E1, E3-deleted and pulsed ex vivo with soluble protozoan parasite L.
adenoviral vector containing the hTERT gene. donovani antigens (SLDA) in a mouse model.
496 INDIAN J EXP BIOL, JUNE 2007

Following vaccination it protects the mice from marginal effect is seen at 50 mg/kg body weight42.
infection as evidenced from parasite load in liver and Treatment with Sb (50 mg/kg body weight) alone
spleen and also from cytokine profile38. By current reduced parasite burden by 39.6±13.5% (P<0.05) and
estimates, leishmaniasis affects people in 88 51.7 ± 13.3% (P<0.01) in spleen and liver,
countries, with 350 million at risk of contracting the respectively. Strikingly, complete clearance of
disease and approximately 2 million new cases each parasite burden from spleen and liver was achieved
year (www.who.int/emc/diseases/leish/leisdis1.html). when infected mice received both SLDA-pulsed DC
The devastating impact of this disease is exemplified and Sb but not in all other experimental groups. The
by the epidemic of visceral leishmaniasis that repetitive in vitro stimulation by SLDA-pulsed DC
occurred in the 1990s in Sudan. Of all the parasitic has confirmed the fact that CD4+IFNγ+ T cells
diseases, leishmaniasis is considered the most likely effectively emerged in higher percentage, when the
to succumb to vaccination. There are concerns about splenocytes were taken from L. donovani infected
whether the same vaccine will work for all mice (13.62%) compared to uninfected mice (6.1%).
leishmaniasis. New discovery of vaccines against Accumulation of IFN-γ mRNA has been detected in
leishmaniasis still draws the attention of science. lymph node cells of L. donovani infected mice
There are concerns about the cost-effective administrated with SLDA-pulsed DC, but the
production of vaccines against visceral leishmaniasis expression of IFN-γ mRNA was not further enhanced
because the leishmaniasis is the primary threat for the when infected mice received combined therapy with
developing and under developed tropical countries. SLDA-pulsed DC plus Sb. Our data on combined
The present authors, being the members of Dr. therapy may have the following explanations. Sb may
Bandyopadhyay’s group for the first time, tried to have potentiated the antileishmanial activity of DC-
formulate an anti-leishmanial immunotherapy strategy based therapy induced IFN-γ by enhancing IFN-γ
that is really useful for poor countries. The signaling. Alternatively, Sb, by its ability to potentiate
experimental strategy was successfully developed in the production of reactive oxygen species43 and to
murine model. The cheaper DC-based induce programmed cell death in Leishmania, may
immunotherapy combined with antimony based provide additional antileishmanial activity over DC-
chemotherapy cures established murine visceral based therapy induced IFN-γ.
leishmaniasis39. Bone marrow derived DC40 Toxoplasma gondii can cause severe sequelae in
differentiated after treatment with GM-CSF and IL-4 the fetus when the mother acquires the infection
and pulsed with SLDA in the presence of TNF-α during pregnancy, as well as life-threatening
produced significant amounts of IL-12 (276 ± 40 neuropathy in immunocompromised patients, in
pg/ml vs 76 ± 22 pg/ml by unpulsed DC), and their particular those with AIDS. T. gondii antigen-pulsed-
administration in infected mice significantly reduced dendritic cell-derived exosomes induce a protective
splenic and hepatic parasite burden (by 96.6 ± 3.5 and immune response against T. gondii infection.
90.9 ± 4.2% respectively, compared with PBS-treated Adoptive transfer of T. gondii-pulsed DC-derived
infected controls (P<0.0005 for each comparison). exosomes to the spleen elicited a strong systemic
This difference in two organs may be attributed to the Th1-modulated Toxoplasma-specific immune
DC subtypes and/or CCR7-mediated migration of DC, response in vivo, and conferred good protection
which is dependent on the expression of CCL21 and against infection. These findings support the
CCL19 chemokines by stromal cells within the T cell possibility that DC-derived exosomes can be used for
areas of secondary lymphoid organs41. The parasite T. gondii immunoprophylaxis and for immuno-
burden in both the organs also decreased significantly prophylaxis against many other pathogens44.
following administration of unpulsed DC (P<0.05). Burkholderia pseudomallei, the causative agent of
SLDA-pulsed or unpulsed Mφ reduced parasite melioidosis, is a gram-negative bacterium which can
burden in both organs only marginally, and the cause either chronic infections or acute lethal sepsis in
differences were not statistically significant. In the infected individuals. The disease is endemic in
mouse model of visceral leishmaniasis, the optimum Southeast Asia and northern Australia. Upon virulent
antileishmanial effect by Sb is achieved by a high challenge with B. pseudomallei strains K96243,
dose of Sb (~500 mg/kg body weight), and only a NCTC 4845 or 576, animals immunized with
PAL et. al.: DENDRITIC CELL BASED VACCINATION & IMMUOTHERAPY 497

dendritic cells that were pulsed with heat-killed antigens were able to stimulate T cells to produce
K96243 and matured in the presence of CpG 1826 various cytokines and proliferate in HCV Ag-
showed significant levels of protection45. dependent manner49-52. Relative to mice inoculated
with non-transfected dendritic cells, splenocytes
DC in anti-viral immunotherapy derived from mice immunized with either hepatitis
Control of a viral infection in vivo requires a rapid C virus Core-transfected or non-structural 5-
and efficient cytotoxic T lymphocyte response. transfected dendritic cells showed 3 to 5-fold
Lentivirus mediated introduction of antigen in DC greater antigen-specific cytotoxic T lymphocyte
confers a protective antiviral immunity in vivo in a activity. These findings suggest that vaccination
lymphocytic choriomeningitis virus model. Therefore, with protein-transfected dendritic cells may
lentiviral vectors may be excellent vaccine candidates constitute an important antiviral strategy for
for viral infections46. Induction of protective hepatitis C virus53.
immunity to bacteria Listeria monocytogenes was Immunization with Ad-S-transfected [Recombinant
observed in case of DC retrovirally transfected with a adenovirus expressing hepatitis B surface antigen
cytotoxic T lymphocyte epitope minigene47. In a (HBsAg) (Ad-S)] DC effectively induced HBsAg-
preliminary investigation on therapeutic DC based specific CTLs in vivo and down-regulated the
vaccine, 18 chronically HIV-1 infected and untreated circulating HBsAg and HBV DNA in HBV transgenic
individuals showing stable viral loads at least for 6 mice. Furthermore, these efficacies were stronger than
months were immunized with autologous monocyte that of HBsAg-pulsed DC and plasmid DNA. Thus,
derived DC loaded with autologous aldrithiol-2- DC transfected with recombinant adenovirus may be a
inactivated HIV-1. Plasma viral load levels were promising candidate for an effective CTL-based
decreased by 80% (median) over the first 112 day therapeutic vaccine against HBV54. The DC from
following immunization. Prolonged suppression of chronic hepatitis B patients had a lower expression of
viral load of more than 90% was seen in 8 individuals costimulatory molecules CD80, CD86 and impaired
for the last 1 year. The suppression of viral load was allogeneic mixed lymphocyte reaction capacity
positively correlated with HIV-1 specific IL-2 or IFN- compared to those from normal controls. However,
γ expressing CD4+ T cells and with HIV-1 gag the impaired DC function could be restored partially
specific perforin expressing CD8+ effector cells, by a cytokine cocktail supplementation in vitro.
suggesting that a robust virus-specific CD4+ T helper Mature DC loaded with HBsAg or HBcAg showed a
(TH1) response is required for inducing and greater capacity for autologous T cell proliferation
maintaining virus specific CD8+ effector to contain and antigen-specific IFN-γ production than immature
HIV-1 in vivo. The results suggest that inactivated DC. Moreover, as a DC-loading antigen, HBcAg was
whole virus pulsed DC vaccines could be a promising more immunogenic than HBsAg. The impaired
strategy for treating people with chronic HIV-1 function of DC in patients with CHB may be restored
infection48. by supplementation in vitro with a cocktail of
A majority of hepatitis C virus (HCV)-infected cytokines, and therapeutic DC vaccines might be
individuals becomes chronically infected, which effective to treat CHB infection in humans55.
can result in liver cirrhosis and hepatocellular
carcinoma. Patients with chronic HCV are unable to DC in anti-tumor immunotherapy
prime and maintain vigorous T-cell responses. The supremacy of DC based anti-tumor vaccination
Hepatitis C virus (HCV) specific T cell responses strategies can be judged clearly from a single
have been suggested to play a significant role in injection of CD34+ progenitor-derived dendritic cell
viral clearance. Recombinant adenoviral vectors vaccine leading to induction of T-cell immunity in
containing HCV derived Core and NS3 genes were patients with stage IV melanoma56. Treatment of
used to endogenously express HCV Core and NS3 malignant glioma is difficult and discouraging. Even
proteins in human DC. These HCV-antigen after resection and maximal adjuvant therapy, the
expressing DC were used to prime and stimulate prognosis remains poor. Yu et. al.57 successfully
autologous T cells obtained from uninfected healthy proved the idea that vaccination of malignant glioma
donors. The DC expressing HCV Core or NS3 patients with peptide-pulsed DC elicits systemic
498 INDIAN J EXP BIOL, JUNE 2007

Advances in DC technology suggest an opportunity


for using this potent antigen presentation system to
deliver myeloma Id to the autologous host to elicit
anti-tumor immune responses. Murine dendritic cells
pulsed with an anti-idiotype antibody induce antigen-
specific protective anti-tumor immunity 60.
Heat-shock protein-based anticancer immuno-
therapy is an idea mooted in early 90’s. But the use of
dendritic cells for this purpose is just new and novel.
Heat-treated CT-26 (HSCT) cells showed a higher
HSP70 protein expression. Pulsing with heat shocked
CT-26 cell lysate elevated the co-stimulatory and
MHC-II molecule expression on bone marrow derived
DC as well as IL-12 p70 secretion. The frequency of
IFN-γ secreting CTLs induced by HSCT-26 DC was
significantly more than that induced by non heat
treated CT-26 lysate pulsed-DC. Heat-shocked tumor
cell lysate-pulsed DC can evoke anti-tumor immune
response in vivo effectively and serve as a novel DC-
based tumor vaccine 61. The mice vaccinated with
HSP105-pulsed BM-DC were markedly protected
from the growth of subcutaneous tumors,
accompanied by a massive infiltration of both CD4+
T cells and CD8+ T cells into the tumors. Both CD4+
T cells and CD8+ T cells play a crucial role in anti-
tumor immunity. In this study both CD4+ T cells and
CD8+ T cells specific to HSP105 were induced by
Fig. 3 ⎯ Ways of DC-based vaccination. Immature DC are
efficient in phagocytosis compared to matured DC. Peptide eluted stimulation with HSP105-pulsed DC. As a result,
from cell lysate of tumors or other pathogens like Leishmania sp., vaccination of mice with BM-DC pulsed with
Toxoplasma sp. are successfully tried for vaccination. RNA/DNA HSP105 could elicit a stronger tumor rejection
from bacteria, virus or fungi are good agent for DC pulsed compared to DNA vaccination62.
vaccination. Immature DC engulfs apoptic bodies more efficiently
and proved as best adjuvant against tumor cells. TNF-α, CD40L,
DC fused with tumor cells may also be effective
IL-10, IL-6 etc. are used as maturation stimuli to mature the DC for immune response induction. A pilot clinical trial
before administration. using dendritomas, purified hybrids from the fusion of
dendritic cells and tumor cells combined with a low
cytotoxicity and intracranial T-cell infiltration. The
dose of IL-2 was conducted in metastatic melanoma
same group demonstrated that an active
patients in order to determine its safety and potential
immunotherapy strategy with tumor lysate pulsed
immunological and clinical responses. Eight of nine
autologous DC could be possible for generating
patients demonstrated immunologic reactions by
antigen-specific cytotoxicity in phase I study in brain
tumor patients58. Results of such a phase I study increased IFN-γ expressing T cells63.
utilizing monocyte-derived DC pulsed with tumor DC can capture antigen from apoptotic tumor cells
and induce MHC class I and class II restricted
RNA in children and young adults with brain cancer
showed that DC-RNA vaccines (Fig. 3) were both responses. Dendritic cells loaded with apoptotic tumor
safe and feasible in children with tumors of the central cells induce stronger T cell responses than dendritic
cell-tumor hybrids in B-CLL. But endocytosed
nervous system with a single leukapheresis59.
apoptotic tumor cells induced a significantly stronger
Immunoglobin produced by myeloma cells is T cell response than DC hybrids and as such should
clone-specific and carries an idiotype which may be a be a better candidate for vaccine production64.
suitable tumor-specific antigen for immune targeting. Autologous DC transfected with total renal tumor
PAL et. al.: DENDRITIC CELL BASED VACCINATION & IMMUOTHERAPY 499

Fig. 4 ⎯ Attempts at antitumor vaccination using the dendritic cells over the past few years80–108. Most successful attempts have been
made in case of neuroblastoma and multiple myeloma.

RNA have been shown to be potent stimulators of placebo controlled trial suggest that in men with
CTLs and anti-tumor immunity in vitro. A Phase I Gleason ≤7 tumors, Provenge® delays disease
trial revealed that the tumor-related mortality of the progression, delays the onset of disease related pain
study subjects was unexpectedly low with only 3 of and results in a survival advantage when compared to
10 patients dying from disease after a mean follow-up patients who had been randomized to placebo66.
of 19.8 months. These data provide a scientific
rationale for continued clinical investigation of this Epilogue
polyvalent vaccine strategy in the treatment of The quest for the ideal immunotherapeutic
metastatic renal cell carcinoma65 and potentially, for approach has for long been the desired objective in
other cancers (Fig. 4). the treatment of cancer and other infectious diseases
(Table 1). DC used as an adjuvant in the
Provenge®: DC vaccine from bench to bedside
immunotherapy for patients with malignant glioma
Provenge® is a therapeutic vaccine composed of
seems to be promising. As with DC vaccination trials
autologous antigen presenting cells cultured ex vivo
for other malignancies, DC immunotherapy of glioma
with a recombinant fusion antigen consisting of
proved to be biologically safe and without major side
prostatic acid phosphatase (PAP) linked to
effects. Nevertheless, its efficacy remains to be more
granulocyte macrophage colony stimulating factor.
closely examined in randomized and controlled
The drug is in the final stage of clinical evaluation in
clinical trials. The development of advanced methods
men with androgen-independent prostate cancer.
for genetic manipulation and transduction of DC prior
Results from a completed phase 3, double-blind
to vaccination in humans may considerably increase
500 INDIAN J EXP BIOL, JUNE 2007

Table 1 ⎯ Major attempts at DC based vaccination: Strategy, experiment and success


Attempts taken Source of antigen Strategy Brief result Ref.
Therapeutic against L.donovani antigen IL-12 transfected DC Live parasites were not detected in the 36
experimental visceral (SLDA) liver of mice vaccinated with IL-12-
leishmaniasis transfected, SLDA-pulsed DC.
Therapeutic against L.donovani antigen Bone marrow derived DC Complete clearance of parasitemia from 37
experimental visceral (SLDA) in combination with SAG spleen and liver with Th1 biased
leishmaniasis response.
Prophylactic against T.gondii antigen DC derived exosome Toxoplasma specific Th1 response in 38
T.gondii vivo, good protection against infection.
Therapeutic against Autologous adrithiol- Autologous monocyte Suppression of viral load, generation of 42
patients infected with 2 inactivated HIV-1 derived DC HIV-1 specific IL-2+CD4+, IFN-γ+CD4+
HIV-1 T cells and HIV-1 gag specific perforin+
CD8+ T cells.
Immunization against Recombinant- Bone marrow derived DC HBsAg-Ad-S transfected DC effectively 48
HBV in transgenic mice HBsAg-Ad-S (BM DC) induced Tc cells and HBsAg-specific
transduced DC CTL in vivo and down-regulated the
circulating HBsAg.
Therapeutic against Surface peptides of Autologous PBMC Development of systemic cytotoxicity in 51,52
malignant glioma patient autologous glioma derived DC four of seven tested patients. Robust
cells intratumoral cytotoxic and memory T-
cell infiltration was detected in two of
four patients.
Immunization syngenic DCs pulsed with heat Bone marrow derived DC HSCT-26 DC elevated the co-stimulatory 55
BALB/c mice shocked colon cancer (BM DC) and MHC-II molecule expression of
(CT-26) cell lysate BMDC as well as IL-12p70 secretion.
(HSCT-26 DC) The IFN-γ secreting CTL induced by
HSCT-26 DC were significantly more
than those induced by CT-26 DC.
Therapeutic against DC-Tumor cell Autologous DC Eight of nine patients demonstrated 57
metastatic melanoma in hybrid immunologic reactions by increased IFN-
patient (Dendritomas) γ expressing T cells.
Therapeutic against renal Renal tumor RNA- Autologous DC In six of seven evaluable subjects, 59
tumor in patient transfected DC expansion of tumor-specific T cells was
detected after immunization.
Provenge® Recombinant Autologous DC Provenge® delays disease progression and 66
Therapeutic vaccine prostatic acid results in a survival advantage when
against prostate cancer phosphatase (PAP) compared to patients who had been
linked to GMCSF randomized to placebo.

the clinical benefits from this type of biological versus host diseases. The investigations in these
treatment. new fields are still in a nascent state of
Dendritic cells, being recognized to have the development. Nevertheless, scads of cerebration
unique capability of initiating a strong primary have already gone into it and the literature reporting
immune response, has long been identified to be an the myriad stories of success and pitfalls have
adept machinery for vaccination against different enriched the understanding in a great way.
nonself pathogenic antigens and modified-self
tumor antigens. Pathogen-specific immunotherapies Acknowledgement
or tumor-targeting vaccination strategies can be far Thanks are due to Dr. Santu Bandyopadhyay for
more strengthened by use of a potent natural constant inspiration and suggestions and UGC, New
adjuvant like dendritic cells. Moreover, Delhi for financial assistance.
characterization of the tolerogenic properties of few
dendritic cell subsets can open up new avenues in References
the form of tolerizing-immunotherapy against 1 Steinman R M & Cohn Z A, Identification of a novel cell
autoimmune disorders or post-transplantation graft type in peripheral lymphoid organs of mice. I. Morphology,
PAL et. al.: DENDRITIC CELL BASED VACCINATION & IMMUOTHERAPY 501

quantitation, tissue distribution, J Exp Med, 137 (1973) hematopoietic progenitors from human cord blood
1142. differentiate along two independent dendritic cell pathways
2 Inaba K, Inaba M, Romani N, Aya H, Deguchi M, Ikehara in response to granulocyte-macrophage colony- stimulating
S, Muramatsu S & Steinman R M, Generation of large factor plus tumor necrosis factor a: II. Functional analysis,
numbers of dendritic cells from mouse bone marrow Blood, 90 (1997) 1458.
cultures supplemented with granulocyte/macrophage 16 Verginis P, Li H S & Carayanniotis G, Tolerogenic
colony-stimulating factor, J Exp Med, 176 (1992) 1693. Semimature Dendritic Cells Suppress Experimental
3 Scheicher C, Mehlig M, Zecher R & Reske K, Dendritic Autoimmune Thyroiditis by Activation of Thyroglobulin-
cells from mouse bone marrow: in vitro differentiation Specific CD4+CD25+ T Cells, J Immunol, 174 (2005) 7433.
using low doses of recombinant granulocyte-macrophage 17 McWilliam A S, Napoli S, Marsh A M, Pemper F L, Nelson
colony-stimulating factor, J Immunol Methods, 154 (1992) D J, Pimm C L, Stumbles P A, Wells T N & Holt P G,
253. Dendritic cells are recruited into the airway epithelium
4 Ardavin C, Wu L, Li C L & Shortman K, Thymic dendritic during the inflammatory response to a broad spectrum of
cells and T cells develop simultaneously in the thymus from stimuli, J Exp Med, 184 (1996) 2429.
a common precursor population, Nature, 362 (1993) 761. 18 McWilliam A S, Nelson D, Thomas J A & Holt P G, Rapid
5 Saunders D, Lucas K, Ismaili J, Wu L, Maraskovsky E, dendritic cell recruitment is a hallmark of the acute
Dunn A & Shortman K, Dendritic cell development in inflammatory response at mucosal surfaces, J Exp Med, 179
culture from thymic precursor cells in the absence of (1994) 1331.
granulocyte/macrophage colony-stimulating factor, J Exp 19 Grayson M H, Lung dendritic cells and the inflammatory
Med, 184 (1996) 2185. response, Annul Allergy Asthma Immunol, 96 (2006) 643.
6 Steinman R M, Pack M & Inaba K, Dendritic cells in the T- 20 Dieu M C, Vanbervliet B, Vicari A, Bridon J M, Oldham E,
cell areas of lymphoid organs, Immunol Rev, 156 (1997) 25. Ait-Yahia S, Briere F, Zlotnik A, Lebecque S & Caux C,
7 Vremec D, Zorbas M, Scollay R, Saunders D J, Selective recruitment of immature and mature dendritic
Ardavin C F, Wu L & Shortman K, The surface phenotype cells by distinct chemokines expressed in different anatomic
of dendritic cells purified from mouse thymus and spleen: sites, J Exp Med, 188 (1998) 373.
Investigation of the CD8 expression by a subpopulation of 21 Power C A, Church D J, Meyer A, Alouani S, Proudfoot A
dendritic cells, J Exp Med, 176 (1992) 47. E, Clark-Lewis I, Sozzani S, Mantovani A & Wells T N,
8 Wu L, Li C L & Shortman K, Thymic dendritic cell Cloning and characterization of a specific receptor for the
precursors: Relationship to the T lymphocyte lineage and novel CC chemokine MIP-3alpha from lung dendritic cells,
phenotype of the dendritic cell progeny, J Exp Med, 184 J Exp Med, 186 (1997) 825.
(1996) 903. 22 Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu
9 Pulendran B, Lingappa J, Kennedy M K, Smith J, Teepe M, Y, Pulendran B & Palucka K, Immunobiology of Dendritic
Rudensky A, Maliszewski C R & Maraskovsky E, Cells, Annul Rev Immunol, 18 (2000) 767.
Developmental pathways of dendritic cells in vivo: Distinct 23 Pardoll D M, Spinning Molecular Immunology in to
function, phenotype, and localization of dendritic cell Successful Immunotherapy, Nature Reviews, 2 (2002) 227.
subsets in FLT3 ligand-treated mice, J Immunol, 159 (1997) 24 Mittal A & Nath I, Human T cell proliferative responses to
2222. particulate microbial antigens are supported by populations
10 De Smedt T, Pajak B, Muraille E, Lespagnard L, Heinen E, enriched in dendritic cells, Clin Exp Immunol, 69 (1987)
De Baetselier P, Urbain J, Leo O & Moser M, Regulation of 611.
dendritic cell numbers and maturation by 25 Hsu F J, Benike C, Fagnoni F, Liles T M, Czerwinski D,
lipopolysaccharide in vivo, J Exp Med, 184 (1996) 1413. Taidi B, Engleman E G & Levy R., Vaccination of patients
11 Leenen P J, Radosevic K, Voerman J S, Salomon B, van with B-cell lymphoma using autologous antigen-pulsed
Rooijen N, Klatzmann D & van Ewijk W, Heterogeneity of dendritic cells, Nat Med, 2 (1996) 52.
mouse spleen dendritic cells: in vivo phagocytic activity, 26 Bender A, Bui L K, Feldman M A, Larsson M & Bhardwaj
expression of macrophage markers, and subpopulation N, Inactivated influenza virus, when presented on dendritic
turnover, J Immunol, 160 (1998) 2166. cells, elicits human CD8+ cytolytic T cell responses, J Exp
12 Romani N, Gruner S, Brang D, Kampgen E, Lenz A, Med, 182 (1995) 1663.
Trockenbacher B, Konwalinka G, Fritsch P O, Steinman R 27 Maraskovsky E, Brasel K, Teepe M, Roux E R, Lyman S D,
M & Schuler G, Proliferating dendritic cell progenitors in Shortman K & McKenna H J, Dramatic increase in the
human blood, J Exp Med, 180 (1994) 83. numbers of functionally mature dendritic cells in Flt3-
13 Zhou L J & Tedder T F, CD14+ blood monocytes can Ligand treated mice: multiple dendritic cell sub-populations
differentiate into functionally mature CD83+ dendritic cells, identified, J Exp Med, 184 (1996) 1953.
Proc Natl Acad Sci USA, 93 (1996) 2588. 28 Shah J A, Darrah P A, Ambrozak D R, Turon T N, Mendez
14 Nestle F O, Zheng X G, Thompson C B, Turka L A & S, Kirman J, Wu C Y, Glaichenhaus N & Seder R A,
Nickoloff B J, Characterization of dermal dendritic cells Dendritic cells are responsible for the capacity of CpG
obtained from normal human skin reveals phenotypic and oligodeoxynucleotides to act as an adjuvant for protective
functionally distinctive subsets. J Immunol, 151 (1993) vaccine immunity against Leishmania major in mice, J Exp
6535. Erratum. J Immunol, 152 (1994) 376. Med, 198 (2003) 281.
15 Caux C, Massacrier C, Vanbervliet B, Dubois B, Durand I, 29 Mohamadzadeh M & Luftig R, Dendritic cells: In the
Cella M, Lanzavecchia A & Banchereau J, CD34+ forefront of immunopathogenesis and vaccine development
502 INDIAN J EXP BIOL, JUNE 2007

– A review, Journal of Immune Based Therapies and Phosphoinositide 3-Kinase and Mitogen-Activated Protein
Vaccines, 2 (2004) 1. Kinase Activation in Leishmania donovani-Infected
30 Chen C H, Wang T L, Hung C F, Yang Y, Young R A, Macrophages, Antimicrob Agents Chemother, 50 (2006)
Pardoll D M & Wu T C, Enhancement of DNA vaccine 1788.
potency by linkage of antigen gene to an HSP70 gene, 44 Aline F, Bout D, Amigorena S, Roingeard P & Dimier-
Cancer Res, 15 (2000) 1035. Poisson I, Toxoplasma gondii antigen-pulsed-dendritic cell-
31 Staveley-O'Carroll K, Schell T D, Jimenez M, Mylin L M, derived exosomes induce a protective immune response
Tevethia M J, Schoenberger S P & Tevethia S S, In vivo against T. gondii infection, Infect Immun, 72 (2004) 4127.
ligation of CD40 enhances priming against the endogenous 45 Elvin S J, Healey G D, Westwood A, Knight S C, Eyles J E
tumor antigen and promotes CD8+ T cell effector function in & Williamson E D, Protection against heterologous
SV40 T antigen transgenic mice, J Immunol, 171 (2003) 697. Burkholderia pseudomallei strains by dendritic cell
32 Liu Y, Huang H, Chen Z, Zong L & Xiang J, Dendritic cells immunization, Infect Immun, 74 (2006) 1706.
engineered to express the Flt3 ligand stimulate type I 46 Zarei S, Abraham S, Arrighi J F, Haller O, Calzascia T,
immune response, and induce enhanced cytoxic T and Walker P R, Kundig T M, Hauser C & Piguet V, Lentiviral
natural killer cell cytotoxicities and antitumor immunity, J transduction of dendritic cells confers protective antiviral
Gene Med , 5 (2003) 668. immunity in vivo, J Virol, 78 (2004) 7843.
33 Rhyu M S, Telomeres, Telomeres and immortality, J Na 47 Nakamura Y, Suda T, Nagata T, Aoshi T, Uchijima M,
Cancer Inst, 87 (1995) 884. Yoshida A, Chida K, Koide Y & Nakamura H, Induction of
34 Frolkis M, Fischer M B, Wang Z, Lebkowski J S, Chiu C P protective immunity to Listeria monocytogenes with dendritic
& Majumdar A S, Dendritic cells reconstituted with human cells retrovirally transfected with a cytotoxic T lymphocyte
telomerase gene induce potent cytotoxic T-cell response epitope minigene, Infect Immun, 71 (2003) 1748.
against different types of tumors, Cancer Gene Ther, 10 48 Lu W, Arraes L C, Ferreira W T & Andrieu J M,
(2003) 239. Therapeutic dendritic-cell vaccine for chronic HIV-1
35 Hetttihewa L M, Immunization of retrovirus-transfected infection, Nat Med, 10 (2004) 1359.
dendritic cells induces specific cytotoxic T lymphocytes for 49 Li W, Krishnadas D K, Li J, Tyrrell D L & Agrawal B,
two distinct malarial peptides presented by Kd molecule, Int Induction of primary human T cell responses against
Immunopharmacol, 3 (2003) 1401. hepatitis C virus-derived antigens NS3 or core by
36 Shao C, Qu J, He L, Zhang Y, Wang J, Zhou H, Wang Y & autologous dendritic cells expressing hepatitis C virus
Liu X, Dendritic cells transfected with an adenovirus vector antigens: Potential for vaccine and immunotherapy, J
encoding interleukin-12 are a potent vaccine for invasive Immunol, 176 (2006) 6065.
pulmonary aspergillosis, Genes Immun, 6 (2005) 103. 50 Li W, Li J, Tyrrell D L & Agrawal B, Expression of
37 Bozza S, Montagnoli C, Gaziano R, Rossi G, Nkwanyuo G, hepatitis C virus-derived core or NS3 antigens in human
Bellocchio S & Romani L, Dendritic cell-based vaccination dendritic cells leads to induction of pro-inflammatory
against opportunistic fungi, Vaccine, 22 (2004) 857. cytokines and normal T-cell stimulation capabilities, J Gen
38 Ahuja S S, Reddick R L, Sato N, Montalbo E, Kostecki V, Virol, 87 (2006) 61.
Zhao W, Dolan M J, Melby P C & Ahuja S K, Dendritic 51 Yu H, Huang H, Xiang J, Babiuk L A & van Drunen Littel-
cell (DC)-based anti-infective strategies: DC engineered to van den Hurk S, Dendritic cells pulsed with hepatitis C
secrete IL-12 are a potent vaccine in a murine model of an virus NS3 protein induce immune responses and protection
intracellular infection, J Immunol, 163 (1999) 3890. from infection with recombinant vaccinia virus expressing
39 Ghosh M, Pal C, Ray M, Maitra S, Mandal L & NS3, J Gen Virol, 87 (2006) 1.
Bandyopadhyay S, Dendritic Cell-Based Immunotherapy 52 Encke J, Findeklee J, Geib J, Pfaff E & Stremmel W,
Combined with Antimony-Based Chemotherapy Cures Prophylactic and therapeutic vaccination with dendritic cells
Established Murine Visceral Leishmaniasis, J Immunol, 170 against hepatitis C virus infection, Clin Exp Immunol, 142
(2003) 5625. (2005) 362.
40 Labeur, M S, Roters B, Pers B, Mehling A, Luger T A, 53 Kuzushita N, Gregory S H, Monti N A, Carlson R, Gehring
Schwarz T, & Grabbe S, Generation of tumor immunity by S & Wands J R, Vaccination with protein-transfected
bone marrow-derived dendritic cells correlates with dendritic cells elicits a sustained response to hepatitis C
dendritic cell maturation stage, J Immunol, 162 (1999) 168. viral antigens, Gastroenterology,130 (2006) 453.
41 Luther S A, Tang H L, Hyman P L, Farr A G & Cyster J G, 54 Huang Y, Chen Z, Jia H, Wu W, Zhong S & Zhou C,
Co-expression of the chemokines ELC and SLC by T cell Induction of Tc1 response and enhanced cytotoxic T
zone stromal cells and deletion of the ELC gene in the lymphocyte activity in mice by dendritic cells transfected
plt/plt mice, Proc Natl Acad Sci, USA, 97 (2000) 12694. with adenovirus expressing HBsAg, Clin Immunol, 119
42 Murray H W, Montelibano C, Peterson R & Sypek J P, (2006) 280.
Interleukin-12 regulates the response to chemotherapy in 55 Duan X Z, He H X & Zhuang H, Restoration in vitro of
experimental visceral leishmaniasis, J Infect Dis, 182 (2000) impaired T-cell responses in patients with chronic hepatitis
1497. B by autologous dendritic cells loaded with hepatitis B virus
43 Mookerjee Basu J, Mookerjee A, Sen P, Bhaumik S, Sen P, proteins (R2), J Gastroenterol Hepatol, 21 (2006) 970.
Banerjee S, Naskar K, Choudhuri S K, Saha B, Raha S & 56 Palucka A K, Dhodapkar M V, Paczesny S, Burkeholder S,
Roy S, Sodium Antimony Gluconate Induces Generation of Wittkowski K M, Steinman R M, Fay J & Banchereau J,
Reactive Oxygen Species and Nitric Oxide via Single injection of CD34+ progenitor-derived dendritic cell
PAL et. al.: DENDRITIC CELL BASED VACCINATION & IMMUOTHERAPY 503

vaccine can lead to induction of T-cell immunity in patients 70 Young J W, Szabolcs P & Moore M A S, Identification of
with stage IV melanoma, J Immunother, 26 (2003) 432. dendritic cell colony-forming units among normal human
57 Yu J S, Wheeler C J, Zeltzer P M, Ying H, Finger D N, Lee CD34+ bone marrow progenitors that are expanded by c-kit
P K, Yong W H, Incardona F, Thompson R C, Riedinger M ligand and yield pure dendritic cell colonies in the presence
S, Zhang W, Prins R M & Black K L, Vaccination of of granulocyte/macrophage colony-stimulating factor and
malignant glioma patients with peptide-pulsed dendritic tumor necrosis factor α, J Exp Med, 182 (1995)1111.
cells elicits systemic cytotoxicity and intracranial T-cell 71 Galy A, Travis M, Cen D & Chen B, Human T B, natural
infiltration, Cancer Res, 61 (2001) 842. killer and dendritic cells arise from a common bone marrow
58 Yu J S, Liu G, Ying H, Yong W H, Black K L & Wheeler C progenitor cell subset, Immunity, 3 (1995) 459.
J, Vaccination with tumor lysate-pulsed dendritic cells 72 Sallusto F & Lanzavecchia A, Efficient presentation of
elicits antigen-specific, cytotoxic T-cells in patients with soluble antigen by cultured human dendritic cells is
malignant glioma, Cancer Res, 64 (2004) 4973. maintained by granulocyte/macrophage colony-stimulating
59 Caruso D A, Orme L M, Neale A M, RaDCliff F J, Amor G factor plus interleukin 4 and downregulated by tumor
M, Maixner W, Downie P, Hassall T E, Tang M L & necrosis factor α, J Exp Med, 179 (1994) 1109.
Ashley D M, Results of a phase 1 study utilizing monocyte- 73 Bender A, Sapp M, Schuler G, Steinman R M & Bhardwaj
derived dendritic cells pulsed with tumor RNA in children N, Improved methods for the generation of dendritic cells
and young adults with brain cancer, Neurooncol, 6 (2004) from nonproliferating progenitors in human blood, J
236. Immunol Methods, 196 (1996) 121.
60 Saha A, Chatterjee S K, Foon K A, Primus F J & 74 Romani N, Reider D, Heuer M, Ebner S, Kampgen E, Eibl
Bhattacharya Chatterjee M, Murine dendritic cells pulsed B, Niederwieser D & Schuler G, Generation of mature
with an anti-idiotype antibody induce antigen-specific dendritic cells from human blood. An improved method
protective antitumor immunity, Cancer Res, 63 (2003) with special regard to clinical applicability, J Immunol
2844. Methods, 196 (1996) 137.
61 Qiu J, Li G W, Sui Y F, Song H P, Si S Y & Ge W, Heat- 75 Randolph G J, Beaulieu S, Lebecque S, Steinman R M &
shocked tumor cell lysate-pulsed dendritic cells induce Muller W A, Differentiation of monocytes into dendritic
effective anti-tumor immune response in vivo, World J cells in a model of transendothelial trafficking, Science, 282
Gastroenterol, 12 (2006) 473. (1998) 480.
62 Yokomine K, Nakatsura T, Minohara M, Kira J, Kubo T, 76 Rissoan M C, Soumelis V, Kadowaki N, Grouard G, Briere F,
Sasaki Y & Nishimura Y, Immunization with heat shock de Waal M R & Liu Y J, Reciprocal control of T helper cell
protein 105-pulsed dendritic cells leads to tumor rejection in and dendritic cell differentiation, Science, 283 (1999) 1183.
mice, Biochem Biophys Res Commun, 343 (2006) 269. 77 Grouard G, Rissoan M C, Filgueira L, Durand I,
63 Wei Y, Sticca R P, Holmes L M, Burgin K E, Li J, Banchereau J & Liu Y J, The enigmatic plasmacytoid T
Williamson J, Evans L, Smith S J, Stephenson J J & cells develop into dendritic cells with interleukin (IL)-3 and
Wagner T E, Dendritoma vaccination combined with low CD40-ligand, J Exp Med, 185 (1997) 1101.
dose interleukin-2 in metastatic melanoma patients induced 78 Res P C, Couwenberg F, Vyth-Dreese F A & Spits H,
immunological and clinical responses, Int J Oncol, 3 (2006) Expression of pTα mRNA in a committed dendritic cell
585. precursor in the human thymus, Blood, 94 (1999) 2647.
64 Kokhaei P, Rezvany M R, Virving L, Choudhury A, 79 Spits H, Couwenberg F, Bakker A Q, Weijer K &
Rabbani H, Osterborg A & Mellstedt H, Dendritic cells Uittenbogaart C H, Id2 and Id3 inhibit development of
loaded with apoptotic tumour cells induce a stronger T-cell CD34+ stem cells into predendritic cell (pre-DC)2 but not
response than dendritic cell-tumour hybrids in B-CLL, into pre-DC1. Evidence for a lymphoid origin of pre-DC2, J
Leukemia, 17 (2003) 894. Exp Med, 192 (2000) 1775.
65 Su Z, Dannull J, Heiser A, Yancey D, Pruitt S, Madden J, 80 Chang A E, Redman B G, Whitfield J R, Nickoloff B J,
Coleman D, Niedzwiecki D, Gilboa E & Vieweg J, Braun T M, Lee P P, Geiger J D & Mule J J, A phase I trial
Immunological and clinical responses in metastatic renal of tumor lysate pulsed dendritic cells in the treatment of
cancer patients vaccinated with tumor RNA-transfected advanced cancer, Clin Cancer Res, 8 (2002) 1021.
dendritic cells, Cancer Res, 63 (2003) 2127. 81 Geiger J, Hutchinson R, Hohenkirk L F, McKenna E, Chang
66 Rini B I, Technology evaluation: APC-8015.Dendreon, A & Mule J, Treatment of solid tumours in children with
Curr Opin Mol Ther, 4 (2002) 76. tumour-lysate pulsed dendritic cells, Lancet, 356 (2000) 1163.
67 Caux C & Banchereau J, in vitro Regulation of Dendritic 82 Morse M A, Mosca P J, Hobeika A, Clay T, Fisher M, Peter
Cell Development and Function (eds Whetton A & Gordon St. M, Williams R, Cumming R I, Caron D & Lyerly H K,
J, Plenum Press, London, 1996) 263. Immunotherapy with Flt3-ligand (Flt3L) mobilized
68 Strunk D, Egger C, Leitner G, Hanau D & Stingl, G, A skin dendritic cells (DC) loaded with carcinoembryonic antigen
homing molecule defines the Langerhans cell progenitor in (CEA) peptide (CAP-1) in patients with metastatic
human peripheral blood, J Exp Med, 185 (1997) 1131. malignancies expressing CEA, Proc Am Soc Clin Oncol, 19
69 Strobl H, Riedl E, Scheinecker C, Bello-Fernandez C, Pickl (2000) 469.
W F, Rappersberger K, Majdic O & Knapp W, TGF-beta 1 83 Morse M A, Coleman R E, Akabani G, Niehaus N,
promotes in vitro development of dendritic cells from Coleman D & Lyerly H K, Migration of human dendritic
CD34+ hemopoietic progenitors, J Immunol,157 (1996) cells after injection in patients with metastatic malignancies,
1499. Cancer Res, 59 (1999) 56.
504 INDIAN J EXP BIOL, JUNE 2007

84 Fong L, Hou Y, Benike C, Yuen A, Fisher G A, Davis M M free generated dendritic cells, Blood, 96 (2000) 63.
& Engleman E C, Vaccination with increasing doses of Flt3 97 Wen Y J, Ling M, Bailey-Wood R & Lim S H, Idiotype
ligand expanded dendritic cells loaded with a protein-pulsed adherent peripheral blood mononuclear cell-
carcinoembyonic antigen derived epitope, Proc Am Assoc derived dendritic cells prime immune system in multiple
Cancer Res, 43 (2002)143. myeloma, Clin Cancer Res, 4 (1998) 957.
85 Sandanaga N, Nagashima H, Mashino K, Tahara K, 98 Ishisaka T, Nevin B, Valone F H, Kothari S S & Peshwa M
Yamaguchi H, Ohta M, Fujii T, Tanaka F, Inoue H, V, Comparison of cryopreserved versus fresh formulation of
Takesako K, Akiyoshi T & Mori M, Dendritic cell idiotype-loaded autologous dendritic cell vaccine, Blood, 98
vaccination with mage peptide is a novel therapeutic (2001) 309.
approach for gastrointestinal carcinomas, Clin Cancer Res, 99 Yi Q, Desikan R, Barlogie B & Munsh N, Optimizing
7 (2001) 2277. dendritic cell-based immunotherapy in multiple myeloma,
86 Stift A, Dubsky P, Friedt J P, Bachleitner T, Schueller G, Blood, 98 (2001) 374.
Tadlbauer K, Jakesz R & Gnant M F, A pilot study for a 100 Brossart P, Wirths S, Stuhler G, Reichardt V L, Kanz L &
phase I trial of active immunotherapy using tumor lysate Brugger W, Induction of cytotoxic T-lymphocyte responses
pulsed autologous dendritic cells in patients with incurable in vivo after vaccinations with peptide-pulsed dendritic
malignancies, Proc Am Soc Clin Oncol, 19 (2000) 464. cells, Blood, 96 (2001) 3102.
87 Schott M, Feldkamp J, Lettmann M, Simon D, Scherbaum 101 Aoyama T, Fujii Y, Hasumi K, Yamaguchi S, Terai M,
W A & Seissler J, Dendritic cell immunotherapy in a Yasuda R, Arai T, Nathan F E, Mastrangelo M J & Sato T,
neuroendocrine pancreas carcinoma, Clin Endocrin, 55 Active specific immunotherapy with tumor-antigen pulsed
(2001) 271. dendritic cells (DC) and CD-40 ligand expressing activated
88 Gjertsen M K, Bakka A, Breivik J, Saeterdal I, Gedde-Dahl T cells (AT). Proc Am Soc Clin Oncol, 20 (2001) 221.
T 3rd, Stokke K T, Solheim B G, Egge T S, Soreide O, 102 Holtl L, Rieser C, Papesh C, Ramoner R, Herold M,
Thorsby E & Gaudernack G, Ex vivo peptide vaccination in Klocker H, Radmayr C, Stenzl A, Bartsch G & Thurner M,
patients with advanced pancreatic cancer: Results of a phase Cellular and immune responses in patients with metastatic
I/II study, Int J Cancer, 65 (1996) 450–453. renal cell carcinoma after vaccination with antigen pulsed
89 Cull G, Durant L, Haynes A & Russell N, Generation of dendritic cells, J Urol, 161 (1999) 777.
idiotype-specific immune responses following vaccination 103 Cajal R, Maordomo J I, Yubero A, Lasierra P, Isla D,
with autologous paraprotein pulsed dendritic cells in Blasco C, Fuertes M A, Palomera R, Sousa R, Guermes A,
myeloma, Blood, 92 (2000) 275. Escudero P, Saenz A, Garcia M, Larrad L & Tres A,
90 Lacy M Q, Wettstein P, Gertz M A, Gastineau D A, Greipp Immunological and clinical effects of immunotherapy with
P R, Fonseca R, Lust J, Witzig T, Rajkumar S V, Valone F dendritic cells pulsed with tumor lysates in patients with
& Kyle R A, Dendritic cell-based idiotype vaccination in advanced cancer. A pilot trial, Proc Am Soc Clin Oncol, 19
post transplant multiple myeloma, Blood, 96 (2000) 374. (2000) 454.
91 Lim S H & Bailey-Wood R, Idiotype protein pulsed 104 Geiger J D, Hutchinson R.J, Hohenkirk L F, McKenna, E A,
dendritic cell vaccination in multiple myeloma, Int J Yanik G A, Levine J E, Chang A E, Braun T M & Mule J J,
Cancer, 83 (1999) 215. Vaccination of pediatric solid tumor patients with tumor
92 Liso A, Stockerl-Goldstein K E, Aufferman S, Benike C J, lysate-pulsed dendritic cells can expand specific T cells and
Reichardt V, van Beckhoven A, Rajapaksa R, Engleman E mediate tumor regression, Cancer Res, 61 (2001) 8513.
G, Blume K G & Levy R, Idiotype vaccination using 105 Hinkel A, Gitlitz B, Mulders P, Moldawer N, Tso C, Kaboo
dendritic cells afer autologous peripheral blood progenitor R, Belldegrun A & Figlin R, Dendritic cell (DC) therapy for
cell transplantation for multiple myeloma, Blood, 94 (1999) metastatic renal cell carcinoma (mRCC)—A translational
715. phase I clinical trial, Proc Am Soc Clin Oncol, 17 (1998)
93 MacKenzie M, Peshwa M V, Wun T, Wolf J, Mason J, 432.
Caggiano V, Redfern C, Strang G & Valone F, 106 Marten A, Renoth S, Heinicke T, von Lilienfeld-Toal M,
Immunotherapy of advanced refractory multiple myeloma Schmidt-Wolf IGH, Allogeneic dendritic cells fused with
with idiotype-pulsed dendritic cells (mylovenge), Blood, 96 tumor cells: Preclinical results and outcome of a first
(2000) 166. clinical phase I/II trial in patients with metastatic renal cell
94 Titzer S, Christensen O, Manzke O, Tesch H, Wolf J, carcinoma, Blood, 98 (2001) 40.
Emmerich B, Carsten C, Diehl V & Bohlen H, Vaccination 107 Kugler A, Stuhler G, Walden P, Zoller G, Zobywalski A,
of multiple myeloma patients with idiotype-pulsed dendritic Brossart P, Trefzer U, Ullrich S, Muller C A, Becker V,
cells: Immunological and clinical aspects, Brit J Haematol, Gross A J, Hemmerlein B, Kanz L, Muller G A & Ringert R
108 (2000) 805. H, Regression of human metastatic renal cell carcinoma
95 Maecker H T, Auffermann-Gretzinger S, Nomura L E, Liso after vaccination with tumor cell dendritic cell hybrids, Nat
A, Czerwinski D K & Levy R, Detection of CD4 T-cell Med, 6 (2000) 332.
responses to a tumor vaccine by cytokine flow cytometry, 108 Midgley R, Kerr D J, Mirza N, Milner A, Hodgkin E,
Clin Cancer Res, 7; Suppl 3 (2001) 902. Barker S & Adams D, A phase II dendritic cell (DC)
96 Reichardt V L, Wirths S, Milazzo C, Brossart P, Stuhler G, immunotherapy trial for hepatocellular carcinoma (HC),
Einsele, H, Brugger W & Kanz L, Specific immunotherapy Proc Am Soc Clin Oncol, 21 (2002) 23.
of multiple myeloma patients using idiotype pulsed serum

You might also like