Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Aug 31.
Published in final edited form as: Curr Opin Investig Drugs. 2010 Dec;11(12):1399–1408.

New approaches to the development of adenoviral dendritic cell vaccines in melanoma

Lisa H Butterfield 1,, Lazar Vujanovic 1
PMCID: PMC3758558  NIHMSID: NIHMS497264  PMID: 21154122

Abstract

Considerable research in the field of immunotherapy for melanoma has demonstrated that this tumor type can be responsive to therapeutic immune activation strategies. In early clinical trials, vaccine strategies using dendritic cells (DCs) and adenovirus (Ad) vectors (AdVs) were safe and immunogenic, and induced clinical responses in a minority of patients. Research from the past several years has yielded an improved mechanistic understanding of DC biology, AdV effects on DCs and the crosstalk that occurs between antigen-loaded DCs and specific lymphocyte subsets. This knowledge base is being combined with technological advances in cytokine delivery, AdV design and in vivo DC targeting. These developments are leading to novel AdV-transduced DC-based therapeutic modalities that may further advance melanoma immunotherapy. Interactions between AdVs and DCs, initial clinical trial results, and new developments in DC engineering and in AdV biology are reviewed.

Keywords: Adenovirus, cancer vaccine, costimulation, cytokine, dendritic cell, melanoma

Introduction

It has been estimated that there will be 68,130 new cases of invasive melanoma and 8700 deaths from this disease in the US during 2010 [1]. The incidence of melanoma has increased significantly during the past several decades from an annual incidence of 6% in the 1970s, and is now increasing at a rate of 3% annually. While the 5-year overall survival for this disease is 91%, after melanoma has spread regionally, the 5-year survival decreases to 62%, and with spreading to a distant site, the 5-year survival rate is only 15% [1]. The median age at presentation of melanoma is 47 to 48 years and, until recently, no therapy had demonstrated an increase in overall survival in patients with unresectable metastatic disease. Support for immune-based approaches that have potential to impact on disease significantly was provided in a recent clinical trial by Hodi et al [2], in which ipilimumab (Bristol-Myers Squibb/Medarex) was demonstrated to improve survival in patients with metastatic melanoma.

Immunotherapy approaches have mainly been used in the treatment of melanoma because this disease is often viewed as an ‘immunogenic’ tumor type. Several immunotherapy approaches have progressed into clinical testing, including cancer vaccines, the adoptive transfer of tumor-specific T-cells, antibodies and cytokines. This review focuses on cancer vaccines for melanoma in which dendritic cells (DCs) and adenovirus (Ad) vectors (AdV) are used to promote antitumor immunity. Significant progress has been made in developing engineered DCs carrying AdV-encoded tumor antigens, cytokines and costimulatory molecules. Moreover, novel vector backbones and new DC-targeting approaches are progressing toward clinical testing.

Dendritic cells

DCs are the key physiological stimulators of naïve and memory lymphocytes [35]. The presentation of antigen epitopes by a sufficiently matured DC occurs in optimal immunostimulatory conditions, with high levels of MHC class I and II, costimulatory (CD80, CD86 and CD40) and adhesion (CD54) molecule expression, and optimal amounts of cytokines (IL-12p70) and chemokines (CCL22/MDC, CXCL8/IL-8, CCL19/MIP-3β) produced [6]; these signals stimulate the immune system to induce a protective type 1 cytotoxic response against the antigen. This process is in contrast to antigen presentation that occurs in the tumor microenvironment, which lacks adequate costimulation or the production of appropriate cytokines, resulting in the downregulation of immune responses [7]. In animal models, DCs pulsed with tumor antigen-derived peptide epitopes have been demonstrated to induce antitumor responses [810]. Moreover, this DC-based cancer vaccine strategy has generated clinically relevant responses (including complete clinical responses) in patients with low grade lymphoma, multiple myeloma, advanced malignant melanoma or prostate cancer [1117].

Immunotherapy clinical trials in patients with metastatic cancer [18], and with metastatic melanoma in particular [19], have demonstrated that DC-based trials result overall in a 7% objective response rate and a 9.5% tumor regression rate, respectively. These early trials provide strong support to DC immunogenicity and their potential for clinical use. However, research with DCs has identified a high degree of plasticity, depending on culture conditions and maturation treatments [20]. Defined culture additives and maturation cocktails of cytokines and TLR agonists can potently modulate the biology of the resultant DCs; standardized cells produced under defined culture conditions can better instruct the lymphocytes that they interact with to become cytotoxic, T-helper type 1 (Th1), Th2 or regulatory in function [20].

Adenovirus vectors

AdVs are an established gene delivery vehicle with an extensive safety record. Ads are well characterized [21], and several classes of replication-defective vectors have been developed and tested. Although the presence of pre-existing neutralizing antibodies to this vector (from environmental exposure) may be a concern, in mice pre-immunized with AdV (to induce circulating neutralizing antibodies) and subsequently immunized with AdV-transduced DCs (AdV/DCs), the same high level of antitumor immunity was induced, regardless of neutralizing antibody levels [22,23]. Conversely, systemic delivery (ie, intravenous, intramuscular or subcutaneous) of virus was strongly inhibited by pre-existing anti-AdV immunity, and the resultant level of in vivo infection of host DCs was reduced, as were T-cell responses [24].

Adenovirus vector infection and signaling

The entry of AdVs into DCs requires two sets of interactions between the virus and the host cell. First, AdVs from the Ad2 and Ad5 serotypes attach to cells by the binding of the knob domain of the fiber protein to the coxsackie-Ad receptor (CAR) present on the host cell [25,26]. This binding is followed by a secondary interaction in which the penton base protein engages the αvβ3 and αvβ5 integrins, stimulating cell signaling and resulting in entry of the AdV into the DCs [26,27]. Following internalization of the AdV, the virus localizes initially in early endosomes and then in late endosomes; the virus later escapes to the cytoplasm, where transgene expression occurs [28]. The linear viral dsDNA genome is then transported to the nucleus, but is maintained extrachromosomally. Although DCs express low levels of CAR, the cells do express high levels of integrins, as well as the heparin-sensitive receptor that is recognized by a distinct segment (ie, the shaft) of the AdV fiber, allowing DCs to be transduced efficiently with AdVs [29,30]. High AdV multiplicities of infection yield > 95% transduction efficiency, and result in high levels of transgene expression and no cytopathic effects [31]. TLRs do not appear to be involved in AdV-mediated signaling in human DCs; signaling is instead induced by a nucleic acid-sensing mechanism that recognizes the viral dsDNA in the cytoplasm and induces IFN regulatory factor 3 (IRF3) activation [32]. In addition, myeloid DCs also activate PKR and PI3K signaling pathways in response to AdV infection, but the activation mechanism remains unclear [33,34].

Adenovirus vector-induced changes in dendritic cell phenotype and function

AdV transduction has a profound impact on human DC biology and function. Immature monocyte-derived DCs transduced with high levels of AdV (multiplicity of infection of ≥ 500 PFU/cell) become more mature, as demonstrated by: (i) increased levels of CCR7, CD83, CD86 and HLA-DR expression; (ii) increased production of IL-8, TNF, IL-15, IL-12p70, IFNα and IFNβ; (iii) decreased secretion of IL-10 and IL-13; and (iv) reduced antigen uptake via macropinocytosis [28,3540]. AdV-infected DCs also secrete elevated levels of chemokines, such as CCL2/MCP-1, CCL3/MIP-1α, CCL4/MIP-1β, CCL5/RANTES, CCL19/MIP-3β, CXCL8/IL-8, CXCL9/MIG and CXCL10/IP-10, allowing the cells to recruit several different lymphocyte subsets [Vujanovic L, Butterfield LH: unpublished data]. In addition, AdV infection induces specific changes to the antigen presenting machinery in human DCs, including increased expression levels of the peptide transporters TAP-1 and TAP-2, and the HLA class I peptide-loading chaperone ERp57 [39]. These data identify functional and molecular changes in AdV/DCs that explain why these cells are efficient at stimulating antigen-specific, IFNγ -producing T-cells [28,3540]. E1-deleted, replication-deficient AdV/DCs also stimulate T-cells that recognize AdV epitopes (which are recall antigens for most individuals) [4143], perhaps because of the leakiness of AdV gene expression or because of the processing of preformed viral proteins transferred at transduction [42].

Antigen-engineered dendritic cells

The successful engineering of DCs with recombinant AdV, and the clear superiority of this approach to traditional physical methods of transfection, such as with calcium phosphate and lipids, was demonstrated in 1997 [31]. Since this time, AdVs have been used as vectors to deliver antigens, cytokines, costimulatory molecules and other transgenes to DCs. Other methods have been used for the transfer of genes to DCs [44], including, most recently, lentivirus vectors [45,46] and optimized physical methods such as electroporation [47]. Tumor antigen-engineered AdV/DCs are capable of processing and presenting antigen epitopes in the context of both MHC class I and II molecules. The continuous display of multiple peptide epitopes is restricted by the patient’s own MHC alleles, and has the advantage of generating polyclonal T-cell responses. Broad and potent activation of multiple CD8+ and CD4+ T-cell specificities [48,49] can be induced by AdV/DCs, and the responses obtained are superior to protein-loaded DCs [50]. While murine models have suggested CD8+ T-cells are the most important antitumor effectors, considerable data also support the significance of CD4+ helper T-cells and the activation of innate immunity to cooperatively induce an effective antitumor response [51]. The provision of antigen-specific mediated by CD4+ helper T-cells may improve the function of CD8+ effector CTLs. For example, antigen-specific activation of CD4+ helper T-cells resulted in the potent activation of CD8+ T-cells, which are then capable of expansion, differentiation and secondary rounds of proliferation [52].

DCs transduced with specific antigen genes process and present antigenic peptides for at least 10 days, while DCs pulsed with the immunodominant peptides present at 50% of the maximal level by day 2 post-pulsing; the maximal CD8+ MART-1-specific T-cell response is superior for the AdV/DCs [53]. Many comparisons of exogenous peptide pulsing versus antigen transfection in DCs have been conducted, and support the superiority of DC transfection with full-length antigen genes for optimal peptide-specific T-cell activation [5457]. Plasmid DNA transfection can be effective when using efficient delivery methods [58], including nucleofection, but in a direct comparison of nucleofection versus transduction with a fiber-modified Ad5/35 AdV, the fiber-modified Ad5/35 AdV was superior [59]. An additional improvement in antigen transduction may be the engineering of DCs to express multiple antigens, resulting in the stimulation of multi-antigen CD8+ and CD4+ T-cell responses [Butterfield LH, Vujanovic L, Kirkwood JM: unpublished data]. This strategy would theoretically allow for the more effective treatment of tumor lesions by also promoting determinant spreading, which has been positively correlated with the survival of patients with cancer [15,16,60]. The stimulation of T-cell responses with multiple antigens may be critical to combat the formation of tumor variants that exhibit antigen- and antigen processing machinery-loss [6167].

Clinical trials of adenovirus vector-transduced dendritic cells

AdV/DCs have progressed to clinical testing [68,69]. In an AdV/DC clinical trial in which DCs were transduced with both AdVMART-1 (MART-1 is a protein antigen that is present on melanocytes) and AdVgp100 (gp100 is a melanoma-associated antigen), 1 of 17 evaluable patients had a complete response [69] and 3 out of 12 patients with vitiligo were reported [68]. In a second trial testing AdVMART-1/DCs in patients (n = 23) with late-stage melanoma [70], the vaccine was safe and immunogenic, and led to activation and expansion of the desired MART-1-specific CD8+ and CD4+ T-cells. In addition, NK cells were activated in a subset of patients and expansion of regulatory T-cells was not detected. Clinical responses for the patients (n = 14) receiving all three vaccines were 1 patient with a suspected complete response, 4 with stable disease (27 to 42 months in duration), and 1 who became eligible for resection and subsequently remained without disease (> 58 months) [70]. These trials demonstrated that ‘first-generation’ AdV/DC vaccines were safe, immunogenic and exhibited clinical activity, and that the use of such vaccines was feasible.

Innate immune activation mediated by adenovirus vector-transduced dendritic cells

The ability of AdV/DCs to generate robust antigen-specific protective antitumor CD4+ and CD8+ T-cell responses has been well characterized; however, the role of NK cells in this response is less clear. The effectiveness of AdV/DC-based vaccines may, ultimately, depend on the ability of AdV/DCs to crosstalk with NK cells and to activate, polarize and bridge innate and adaptive immunity [7174]. DCs and NK cells are essential components of the innate immune system that interact and reciprocally regulate each other, and induce an enhanced polarization of type 1 cytokine secretion [7376]. Activated NK cells induce increases in the expression of maturation markers and the secretion of IL-12p70 in DCs [7376]. Reciprocally, matured DCs induce the expression of activation markers, enhance IFNγ secretion, perforin-mediated tumoricidal activity and proliferation in NK cells, and stimulate the ability of such cells to control in vivo viral infections and tumor growth [77]. This early cellular crosstalk is believed to be an essential regulatory immune mechanism that bridges innate and adaptive immune functions, and defines the quality and magnitude of antiviral and antitumor immune responses.

The ability of human AdV/DCs to activate NK cells was tested directly in vitro. AdV/DCs encoding Escherichia coli β-galactosidase (LacZ; a biologically inert protein) activated resting NK cells effectively, without enhancing AdV/DC susceptibility to NK cell-mediated killing, as measured by induced increases in CD69 expression (ie, a NK cell activation marker), IFNγ secretion, cell proliferation and killing of tumor cell targets in vitro and in vivo [40]. AdV/DC-mediated activation of NK cells required cell-to-cell contact, and was mediated by transmembrane TNF and trans-presented IL-15 present on the surface of DCs [40]. AdV/DCs also recruited both major subsets of NK cells effectively via CXCL8/IL-8 and CXCL10/IP-10 secretion that, coupled with their ability to activate NK cells, makes them potent immune activators of not only adaptive, but also of innate immune responses [Vujanovic L, Butterfield LH: unpublished data].

Cytokine-engineered dendritic cells

The maturation of DCs with cocktails of reagents results in cytokine production. However, in order for reproducible, constitutive and high levels of production of specific cytokines to be obtained from DCs, AdV transduction has been used. Several cytokines and chemokines known to be important mediators of antitumor immune responses have been tested in gene therapy protocols through the use of ex vivo gene-engineered DCs. Some of the cytokines and chemokines tested include IL-12, IL-15, IL-18, TNF and CCL21/6Ckine [7382].

IL-12p70 and IL-18 as immunotherapeutic agents

IL-12 and IL-18 are two cytokines that are of particular interest to tumor immunologists because of their ability to promote synergistically IFNγ secretion from and proliferation of CD4+ effector T-cells and NK cells [78,79,83,84]. IL-12 is a heterodimeric, multifunctional cytokine with properties that bridge innate and adaptive immunity. This cytokine acts as a key regulator of cell-mediated immune responses, and induces type 1 immunity by stimulating NK cells and T-cells to produce IFNγ, to proliferate and to exhibit cytolytic activity [85]. In addition, IL-12 inhibits angiogenesis by promoting IFNγ-inducible genes and lymphocyte-endothelial cell crosstalk [8688]. While type 1 immune responses have been associated with spontaneous or therapy-induced regression of tumor lesions [89,90], tumor-infiltrating lymphocytes isolated from patients with progressive lesions have been generally reported to exhibit dominant Th2-type (ie, secrete IL-4 and IL-5) or regulatory (Th3)-type (ie, secrete IL-10 or TGFβ1) responses [8991]. Therefore, the type 1 immunity-skewing properties of IL-12 have provided the rationale for exploiting this cytokine as an anticancer agent.

IL-18 is a member of the IL-1 cytokine superfamily that has an important role in regulating immune responses. IL-18 is produced by APCs (DCs and monocytes), as well as by the pituitary gland, keratinocytes and osteoblasts, and adrenal cortical, intestinal epithelial and Kupffer cells (ie, phagocytes lining the hepatic sinusoids) [92]. While IL-18 was characterized initially as an IFNγ-inducing factor [93], later studies demonstrated that IL-18 is a unique cytokine that is capable of inducing either Th1 or Th2 polarization, depending on the type and context of stimuli, the cytokine priming milieu and underlying genetic influences [94]. Furthermore, IL-18 displays chemoattractant properties for both myeloid- and plasmacytoid-derived DCs [95,96]. IL-12p70 induces T-cell surface expression of the IL-18 receptor (IL-18R) by naïve T-cells [97], while IL-18 potentiates the differentiation of Th1 cells initiated by IL-12 [98].

IL-12p70 has been tested using various administration methods for its ability to enhance Th1-type responses, and has achieved considerable immunostimulatory results in vitro and in vivo [85]. Furthermore, IL-12 combined with other immunotherapy approaches, particularly when administered with IL-18, has demonstrated improved immunostimulatory results compared with IL-12 alone. However, in clinical trials, systemically administered recombinant IL-12p70 (rIL-12p70) displayed unacceptable toxicities, including fever/chills, fatigue, nausea, vomiting, headache, anemia, neutropenia, lymphopenia, hyperglycemia, thrombocytopenia, hypoalbuminemia and even death [99104]. Studies in mice demonstrated that these toxicities were mainly caused by IFNγ overproduction by NK cells [105]. Moreover, IL-12-mediated toxicity was particularly exacerbated with the coadministration of recombinant IL-18 (rIL-18) [106]. Studies in mice also demonstrated that simultaneous administration of rIL-12 and rIL-18 led to severe systemic inflammation, as a result of IFNγ secretion by NK cells, and 100% mortality, and these effects were STAT4-dependent [106].

IL-12p70 and IL-18 gene therapy delivered intratumorally through ex vivo gene-engineered dendritic cells

One possible approach to prevent systemic toxicity and to minimize toxic effects is to use gene transfer methods to limit IL-12p70 and IL-18 production within the tumor environment. This strategy has demonstrated increased efficacy and safety profiles in several studies and trials [107109]. For example, in a phase I clinical trial in which recombinant Ad-transduced, IL-12p70 gene-modified autologous DCs were delivered intratumorally to colorectal, hepatic or pancreatic carcinomas, treatment was well tolerated with limited toxicities observed (lymphopenia and fever occurred in the majority of patients) [108]. However, objective clinical responses in the treated patients were limited. Mouse models have also demonstrated that the intratumoral delivery of AdV/DCs encoding IL-12p70 (under the control of a constitutive promoter), particularly when co-transduced with an AdV encoding IL-18, promoted enhanced Th1-type immunity and determinant spreading in the curative antitumor CTL repertoire, and acute tumor rejection [82,110,111]. While DC-based IL-12p70 gene therapy displayed potent efficacy, the sustained high levels of IL-12p70 and IL-18 could potentially cause unwanted toxicities, as well as cytokine-dependent limitations in DC migration after intratumoral administration [81,82,110,111].

The RheoSwitch therapeutic system: A novel method of controlling transgene expression in adenovirus vector-transduced dendritic cells

To circumvent concerns regarding sustained high levels of IL-12p70 with gene therapy approaches, the INXN-3001 plus INXN-1001 combination regimen (Intrexon) that incorporated a novel AdV encoding a conditional IL-12p70 expression system, which used the RheoSwitch therapeutic system (RTS) for the regulatory control of long-term therapeutic gene expression, was developed and tested in a subcutaneous B16 melanoma mouse model [112]. The RTS approach incorporated the rAd.RheoIL12 AdV in which the mouse IL-12p70 subunit genes were placed under the control of an inducible promoter that was activated by a diacylhydrazine small-molecule ligand [113]. Using this system, transgene expression was controlled effectively in transduced DCs (DC.RheoIL12) after intratumoral injection by regulating administration of the ligand. DC.RheoIL12 cells secreted significant amounts of IL-12p70 in a tightly regulated manner in vitro, and in vivo the treatment benefit of DC.RheoIL12 delivered into B16 lesions was strictly ligand-dependent [113]. The unique safety feature of DC.RheoIL12 was accentuated when this treatment was combined with IL-2: TNFα-associated toxicity was ameliorated by halting ligand administration [113]. The use of the RTS approach can delay the activation of IL-12p70 production by injected DCs, potentially allowing the cells to acquire tumor antigens and to migrate to a tissue-draining lymph node, as well as providing a mechanism to inhibit IL-12p70 production if toxicity is observed.

Engineering dendritic cells to express high levels of costimulatory molecules

Costimulatory molecules expressed on the surface of DCs are considered to provide ‘signal 2’ (where ‘signal 1’ is the MHC/antigen complex), which is required for proper activation of adaptive immunity. The lack of ‘signal 2’ induces CD4+ and CD8+ T-cell anergy. Mature DCs express high levels of the costimulatory CD80, CD86 and CD40 molecules, which are critical for immunity in most models studied. The CD40 ligand (CD40L) is expressed on activated T-cells, and the CD40-CD40L signaling pathway promotes DC and T-cell crosstalk to facilitate full maturation of DCs. CD40L in several forms has been used to activate DCs in vitro, but an alternative strategy is to engineer DCs to express CD40L directly, which allows for strong signaling within DCs. This strategy has been used to activate DCs to secrete high levels of IL-12p70, migrate toward CCL19/MIP-3β and activate MART-1-specific T-cells [114]. Such DCs could gather antigen in vivo following intratumoral injection, or be co-transduced with an antigen-encoding AdV [114]. CD40L–engineered DCs were observed to be strong IL-12p70 producers, to express high levels of CD80, CD86, CD83 and IL-10, and to lead to reduced T-cell activation [115]. As noted, DCs are exquisitely sensitive to environmental signals, and seemingly minor methodological changes can impact the resultant DC-lymphocyte crosstalk [20].

Adenovirus vector-transduced dendritic cells engineered to secrete chemokines

CCL21/6Ckine is constitutively expressed in human lymph nodes, spleen and appendix and attracts CCR7-positive lymphocytes and DCs to lymph nodes [116]. In vitro, DCs transduced with AdV-encoded CCL21/6Ckine expressed high levels of this chemokine, resulting in the migration of lymphocytes to the DCs [117]. Such chemokine-engineered DCs have been tested with and without antigen loading in a B16 melanoma mouse model model [118], and were observed to induce the migration of CD8+ and CD4+ T-cells, as well as DCs. This strategy is now being tested clinically in patients with metastatic melanoma [119].

Novel adenoviral vector strategies for engineering dendritic cells

There are many innovative approaches in preclinical development that use combinations of AdVs and DCs in various disease models, including melanoma models. However, new strategies are required to improve viability, efficacy and targeting of AdV/DC vaccines.

Alternative adenovirus vector serotypes

Given the prevalence of pre-existing immunity to the commonly used Ad5 vectors, different Ad serotypes have been tested for use with DCs in order to determine their transduction efficiency, immunogenicity and downstream effects on DC and T-cells. In one in vitro study, chimeric Ad5/Ad35 vectors in which the fiber region of Ad5 was substituted with the Ad35 region were tested [120]. These vectors exhibited increased efficiency with respect to DC transduction and subsequent T-cell activation, while maintaining positive effects on DC maturation [120]. In a second study, Ad5/35 and Ad35 vectors were compared in human CD46 transgenic mouse-derived DCs. Ad35 serotype vectors promoted a stronger immune response compared with Ad5 vectors [121], suggesting that antitumor immunity might be enhanced for Ad35-based approaches. A study of neutralizing anti-Ad antibodies in humans in Africa and in the US demonstrated the presence of high levels of circulating antibodies to Ad5, but lower levels to Ad35, indicating that, as expected, Ad5 exposure is more prevalent [122]. While this observation clearly impacts systemic AdV delivery, the impact of neutralizing antibodies on AdV/DCs may be minimal [22,23]. Ad3 vectors that use CD80 and CD86 costimulatory molecules as surface receptors have also been tested [123]; the use of these costimulatory molecules potentially makes these vectors naturally tropic for B-cells and DCs when administered in vivo.

Efficient targeting of adenovirus vectors to dendritic cells

Another approach to improve the transduction efficiency of DCs is to use CD40 for the specific targeting of AdVs to DCs. For example, the use of a bispecific antibody (specific for CD40 and the fiber-knob of AdV) improved transduction efficiency and human DC maturation in vitro [29]. This approach was tested in a subsequent study in human skin explants, and demonstrated increased efficiency and potential as an in vivo DC targeting strategy [124]. In addition, the targeting of AdVMART-1 to both myeloid and plasmacytoid DCs in human lymph node cell suspensions and subsequent activation of MART-1-specific T-cells was demonstrated [125]. Other targeting approaches that can improve in vitro transduction efficiency and that may be useful for in vivo targeting include the use of the FcγR1 (CD64) via a bispecific antibody approach [126], or modifying the CAR with a CAR-TAT (HIV TAT protein transduction domain) adapter molecule [127].

Using adenovirus vectors for improved dendritic cell-targeting to tumors and improved antigen release

Notably, AdV CCL19/MIP-3β can induce not only lymphocyte, but also DC migration toward the engineered cells [128]. An AdV encoding CCL5/RANTES has been tested in murine tumor models [128] and, in addition to increasing the levels of DC migration toward tumors, macrophages, NK cells and CD8+ T-cells, the engineered cells also migrated toward tumors. Furthermore, oncolytic AdVs, which replicate selectively in tumors and which can lead to tumor lysis, are also being used for the in vivo release of tumor antigens that can then be taken up by host DCs, leading to the activation of adaptive immunity [129]. The oncolytic AdV strategy has been combined with immune modulatory molecules (eg, IL-12p70 and 4-1BBL) to promote DC infiltration and antitumor immunity, as evaluated in a B16-F10 tumor model in mice [130].

Conclusion

Initial clinical trials of antigen-engineered AdV/DC vaccines in patients with melanoma have demonstrated that these agents are safe and immunogenic, and can promote potent and durable clinical responses in a minority of patients. Moreover, novel strategies involving multiple antigens, cytokines and chemokines, regulatable promoters and improved DC targeting are being translated to patients. These new approaches are based on improved knowledge of DC biology, AdV effects on DCs and lymphocyte activation.

There are major opportunities in the area of DC-based vaccines. First, there is a need to implement immunological monitoring in clinical trials in order to understand the effects of AdVs, full-length antigens, and cytokines or chemokines encoded by the vector on the diverse lymphocyte populations being activated. Second, it is important for specific DC culture and maturation conditions to be combined rationally with AdVs in order to optimize the antigen presentation and lymphocyte activation abilities of these cells. Third, the most cost-effective and broadly applicable approach would be to use a virus that is capable of specific in vivo targeting of DCs; therefore, current approaches are in need of further development. Ultimately, these studies are expected to provide immunotherapy approaches for the treatment of melanoma that exhibit greater efficacy. These strategies may lead to stand-alone therapies that are useful in early-stage disease, or may be used as part of a combination strategy that includes the adoptive transfer of lymphocytes, antibodies or other types of interventions in more advanced disease.

Acknowledgements

Research described in this article was supported, in part, by the NIH grant P50 CA121973-Project 2 awarded to Lisa Butterfield and the University of Pittsburgh Cancer Institute.

References

•• of outstanding interest

• of special interest

  • 1.Cancer Facts & Figures. American Cancer Society. 2010 www.cancer.org/research/cancerfactsfigures/index.
  • 2.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–723. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392(6673):245–252. doi: 10.1038/32588. [DOI] [PubMed] [Google Scholar]
  • 4.Steinman RM, Young JW. Signals arising from antigen-presenting cells. Curr Opin Immunol. 1991;3(3):361–372. doi: 10.1016/0952-7915(91)90039-4. [DOI] [PubMed] [Google Scholar]
  • 5.Steinman RM, Pack M, Inaba K. Dendritic cell development and maturation. Adv Exp Med Biol. 1997;417:1–6. doi: 10.1007/978-1-4757-9966-8_1. [DOI] [PubMed] [Google Scholar]
  • 6.McColl SR. Chemokines and dendritic cells: A crucial alliance. Immunol Cell Biol. 2002;80(5):489–496. doi: 10.1046/j.1440-1711.2002.01113.x. [DOI] [PubMed] [Google Scholar]
  • 7.LaSalle JM, Hafler DA. T cell anergy. FASEB J. 1994;8(9):601–608. doi: 10.1096/fasebj.8.9.8005388. [DOI] [PubMed] [Google Scholar]
  • 8.Zitvogel L, Mayordomo JI, Tjandrawan T, DeLeo 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-associated cytokines. J Exp Med. 1996;183(1):87–97. doi: 10.1084/jem.183.1.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Porgador A, Gilboa E. Bone marrow-generated dendritic cells pulsed with a class I-restricted peptide are potent inducers of cytotoxic T lymphocytes. J Exp Med. 1995;182(1):255–260. doi: 10.1084/jem.182.1.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Flamand V, Sornasse T, Thielemans K, Demanet C, Bakkus M, Bazin H, Tielemans F, Leo O, Urbain J, Moser M. Murine dendritic cells pulsed in vitro with tumor antigen induce tumor resistance in vivo . Eur J Immunol. 1994;24(3):605–610. doi: 10.1002/eji.1830240317. [DOI] [PubMed] [Google Scholar]
  • 11.Timmerman JM, Levy R. Dendritic cell vaccines for cancer immunotherapy. Annu Rev Med. 1999;50:507–529. doi: 10.1146/annurev.med.50.1.507. [DOI] [PubMed] [Google Scholar]
  • 12.Hsu FJ, Benike C, Fagnoni F, Liles TM, Czerwinski D, Taidi B, Engleman EG, Levy R. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat Med. 1996;2(1):52–58. doi: 10.1038/nm0196-52. [DOI] [PubMed] [Google Scholar]
  • 13.Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, Burg G, Schadendorf D. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998;4(3):328–332. doi: 10.1038/nm0398-328. [DOI] [PubMed] [Google Scholar]
  • 14.Thurner B, Haendle I, Röder C, Dieckmann D, Keikavoussi P, Jonuleit H, Bender A, Maczek C, Schreiner D, von den Driesch P, Bröcker EB, et al. Vaccination with MAGE-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma. J Exp Med. 1999;190(11):1669–1678. doi: 10.1084/jem.190.11.1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Butterfield LH, Ribas A, Dissette VB, Amarnani SN, Vu HT, Oseguera D, Wang HJ, Elashoff RM, McBride WH, Mukherji B, Cochran AJ, et al. Determinant spreading associated with clinical response in dendritic cell-based immunotherapy for malignant melanoma. Clin Cancer Res. 2003;9(3):998–1008. [PubMed] [Google Scholar]
  • 16.Ribas A, Glaspy JA, Lee Y, Dissette VB, Seja E, Vu HT, Tchekmedyian NS, Oseguera D, Comin-Anduix B, Wargo JA, Amarnani SN, et al. Role of dendritic cell phenotype, determinant spreading, and negative costimulatory blockade in dendritic cell-based melanoma immunotherapy. J Immunother. 2004;27(5):354–367. doi: 10.1097/00002371-200409000-00004. [DOI] [PubMed] [Google Scholar]
  • 17.Engell-Noerregaard L, Hansen TH, Andersen MH, Thor Straten P, Svane IM. Review of clinical studies on dendritic cell-based vaccination of patients with malignant melanoma: Assessment of correlation between clinical response and vaccine parameters. Cancer Immunol Immunother. 2009;58(1):1–14. doi: 10.1007/s00262-008-0568-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: Moving beyond current vaccines. Nat Med. 2004;10(9):909–915. doi: 10.1038/nm1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Banchereau J, Ueno H, Dhodapkar M, Connolly J, Finholt JP, Klechevsky E, Blanck JP, Johnston DA, Palucka AK, Fay J. Immune and clinical outcomes in patients with stage IV melanoma vaccinated with peptide-pulsed dendritic cells derived from CD34+ progenitors and activated with type I interferon. J Immunother. 2005;28(5):505–516. doi: 10.1097/01.cji.0000171292.79663.cb. [DOI] [PubMed] [Google Scholar]
  • 20.Mailliard RB, Wankowicz-Kalinska A, Cai Q, Wesa A, Hilkens CM, Kapsenberg ML, Kirkwood JM, Storkus WJ, Kalinski P. α-Type-1 polarized dendritic cells: A novel immunization tool with optimized CTL-inducing activity. Cancer Res. 2004;64(17):5934–5937. doi: 10.1158/0008-5472.CAN-04-1261. [DOI] [PubMed] [Google Scholar]
  • 21.Campos SK, Barry MA. Current advances and future challenges in adenoviral vector biology and targeting. Curr Gene Ther. 2007;7(3):189–204. doi: 10.2174/156652307780859062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ribas A, Butterfield LH, McBride WH, Dissette VB, Koh A, Vollmer CM, Hu B, Chen AY, Glaspy JA, Economou JS. Characterization of antitumor immunization to a defined melanoma antigen using genetically engineered murine dendritic cells. Cancer Gene Ther. 1999;6(6):523–536. doi: 10.1038/sj.cgt.7700076. [DOI] [PubMed] [Google Scholar]
  • 23.Ribas A, Butterfield LH, McBride WH, Jilani SM, Bui LA, Vollmer CM, Lau R, Dissette VB, Hu B, Chen AY, Glaspy JA, et al. Genetic immunization for the melanoma antigen MART-1/Melan-A using recombinant adenovirus-transduced murine dendritic cells. Cancer Res. 1997;57(14):2865–2869. [PubMed] [Google Scholar]
  • 24.Lindsay RW, Darrah PA, Quinn KM, Wille-Reece U, Mattei LM, Iwasaki A, Kasturi SP, Pulendran B, Gall JG, Spies AG, Seder RA. CD8+ T cell responses following replication-defective adenovirus serotype 5 immunization are dependent on CD11c+ dendritic cells but show redundancy in their requirement of TLR and nucleotide-binding oligomerization domain-like receptor signaling. J Immunol. 2010;185(3):1513–1521. doi: 10.4049/jimmunol.1000338. [DOI] [PubMed] [Google Scholar]
  • 25.Roelvink PW, Mi Lee G, Einfeld DA, Kovesdi I, Wickham TJ. Identification of a conserved receptor-binding site on the fiber proteins of CAR-recognizing Adenoviridae . Science. 1999;286(5444):1568–1571. doi: 10.1126/science.286.5444.1568. [DOI] [PubMed] [Google Scholar]
  • 26.Nemerow GR, Stewart PL. Role of αv integrins in adenovirus cell entry and gene delivery. Microbiol Mol Biol Rev. 1999;63(3):725–734. doi: 10.1128/mmbr.63.3.725-734.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wickham TJ, Mathias P, Cheresh DA, Nemerow GR. Integrins αvβ3 and αvβ5 promote adenovirus internalization but not virus attachment. Cell. 1993;73(2):309–319. doi: 10.1016/0092-8674(93)90231-e. [DOI] [PubMed] [Google Scholar]
  • 28.Perreau M, Mennechet F, Serratrice N, Glasgow JN, Curiel DT, Wodrich H, Kremer EJ. Contrasting effects of human, canine, and hybrid adenovirus vectors on the phenotypical and functional maturation of human dendritic cells: Implications for clinical efficacy. J Virol. 2007;81(7):3272–3284. doi: 10.1128/JVI.01530-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tillman BW, de Gruijl TD, Luykx-de Bakker SA, Scheper RJ, Pinedo HM, Curiel TJ, Gerritsen WR, Curiel DT. Maturation of dendritic cells accompanies high-efficiency gene transfer by a CD40-targeted adenoviral vector. J Immunol. 1999;162(11):6378–6383. [PubMed] [Google Scholar]
  • 30.Cheng C, Gall JG, Kong WP, Sheets RL, Gomez PL, King CR, Nabel GJ. Mechanism of Ad5 vaccine immunity and toxicity: Fiber shaft targeting of dendritic cells. PLoS Pathog. 2007;3(2):e25. doi: 10.1371/journal.ppat.0030025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Arthur JF, Butterfield LH, Roth MD, Bui LA, Kiertscher SM, Lau R, Dubinett S, Glaspy J, McBride WH, Economou JS. A comparison of gene transfer methods in human dendritic cells. Cancer Gene Ther. 1997;4(1):17–25. [PubMed] [Google Scholar]
  • 32. Nociari M, Ocheretina O, Schoggins JW, Falck-Pedersen E. Sensing infection by adenovirus: Toll-like receptor-independent viral DNA recognition signals activation of the interferon regulatory factor 3 master regulator. J Virol. 2007;81(8):4145–4157. doi: 10.1128/JVI.02685-06.• AdV infection rapidly activates innate immunity, which subsequently stimulates inflammatory antiviral responses and adaptive immunity. The Ad sensor, the ligand and the triggered signaling response were previously undefined. This study demonstrated that infection by AdV stimulated an IRF3-mediated IFN and proinflammatory response that was mediated through a TLR-independent DNA-sensing mechanism.
  • 33.Zhu J, Huang X, Yang Y. Innate immune response to adenoviral vectors is mediated by both Toll-like receptor-dependent and -independent pathways. J Virol. 2007;81(7):3170–3180. doi: 10.1128/JVI.02192-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Tan PH, Beutelspacher SC, Xue SA, Wang YH, Mitchell P, McAlister JC, Larkin DF, McClure MO, Stauss HJ, Ritter MA, Lombardi G, et al. Modulation of human dendritic cell function following transduction with viral vectors: Implications for gene therapy. Blood. 2005;105(10):3824–3832. doi: 10.1182/blood-2004-10-3880. [DOI] [PubMed] [Google Scholar]
  • 35.Butterfield LH, Jilani SM, Chakraborty NG, Bui LA, Ribas A, Dissette VB, Lau R, Gamradt SC, Glaspy JA, McBride WH, Mukherji B, et al. Generation of melanoma-specific cytotoxic T lymphocytes by dendritic cells transduced with a MART-1 adenovirus. J Immunol. 1998;161(10):5607–5613. [PubMed] [Google Scholar]
  • 36.Diao J, Smythe JA, Smyth C, Rowe PB, Alexander IE. Human PBMC-derived dendritic cells transduced with an adenovirus vector induce cytotoxic T-lymphocyte responses against a vector-encoded antigen in vitro . Gene Ther. 1999;6(5):845–853. doi: 10.1038/sj.gt.3300899. [DOI] [PubMed] [Google Scholar]
  • 37.Lundqvist A, Choudhury A, Nagata T, Andersson T, Quinn G, Fong T, Maitland N, Pettersson S, Paulie S, Pisa P. Recombinant adenovirus vector activates and protects human monocyte-derived dendritic cells from apoptosis. Hum Gene Ther. 2002;13(13):1541–1549. doi: 10.1089/10430340260201635. [DOI] [PubMed] [Google Scholar]
  • 38.Schumacher L, Ribas A, Dissette VB, McBride WH, Mukherji B, Economou JS, Butterfield LH. Human dendritic cell maturation by adenovirus transduction enhances tumor antigen-specific T-cell responses. J Immunother. 2004;27(3):191–200. doi: 10.1097/00002371-200405000-00003. [DOI] [PubMed] [Google Scholar]
  • 39.Vujanovic L, Whiteside TL, Potter DM, Chu J, Ferrone S, Butterfield LH. Regulation of antigen presentation machinery in human dendritic cells by recombinant adenovirus. Cancer Immunol Immunother. 2009;58(1):121–133. doi: 10.1007/s00262-008-0533-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Vujanovic L, Szymkowski DE, Alber S, Watkins SC, Vujanovic NL, Butterfield LH. Virally infected and matured human dendritic cells activate natural killer cells via cooperative activity of plasma membrane-bound TNF and IL-15. Blood. 2010;116(4):575–583. doi: 10.1182/blood-2009-08-240325.• Although AdV/DCs have been investigated for their ability to stimulate antigen-specific CD4+ and CD8+ T-cell responses, little is known about their interaction with NK cells. This study demonstrated that AdV/DCs effectively activated NK cells by cell-to-cell contact through the cooperative activities of membrane-bound TNF and IL-15.
  • 41.Smith CA, Woodruff LS, Kitchingman GR, Rooney CM. Adenovirus-pulsed dendritic cells stimulate human virus-specific T-cell responses in vitro . J Virol. 1996;70(10):6733–6740. doi: 10.1128/jvi.70.10.6733-6740.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Roth MD, Cheng Q, Harui A, Basak SK, Mitani K, Low TA, Kiertscher SM. Helper-dependent adenoviral vectors efficiently express transgenes in human dendritic cells but still stimulate antiviral immune responses. J Immunol. 2002;169(8):4651–4656. doi: 10.4049/jimmunol.169.8.4651. [DOI] [PubMed] [Google Scholar]
  • 43.Gahéry-Ségard H, Molinier-Frenkel V, Le Boulaire C, Saulnier P, Opolon P, Lengagne R, Gautier E, Le Cesne A, Zitvogel L, Venet A, Schatz C, et al. Phase I trial of recombinant adenovirus gene transfer in lung cancer. Longitudinal study of the immune responses to transgene and viral products. J Clin Invest. 1997;100(9):2218–2226. doi: 10.1172/JCI119759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Meng WS, Butterfield LH. Activation of antigen-presenting cells by DNA delivery vectors. Expert Opin Biol Ther. 2005;5(8):1019–1028. doi: 10.1517/14712598.5.8.1019. [DOI] [PubMed] [Google Scholar]
  • 45.Grabski E, Waibler Z, Schüle S, Kloke BP, Sender LY, Panitz S, Cichutek K, Schweizer M, Kalinke U. Comparative analysis of transduced primary human dendritic cells generated by the use of three different lentiviral vector systems. Mol Biotechnol. 2010 doi: 10.1007/s12033-010-9340-z. [DOI] [PubMed] [Google Scholar]
  • 46.Negri DR, Bona R, Michelini Z, Leone P, Macchia I, Klotman ME, Salvatore M, Cara A. Transduction of human antigen-presenting cells with integrase-defective lentiviral vector enables functional expansion of primed antigen-specific CD8+ T cells. Hum Gene Ther. 2010;21(8):1029–1035. doi: 10.1089/hum.2009.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mobergslien A, Sioud M. Optimized protocols for siRNA delivery into monocytes and dendritic cells. Methods Mol Biol. 2010;629:71–85. doi: 10.1007/978-1-60761-657-3_5. [DOI] [PubMed] [Google Scholar]
  • 48.Liu Y, Daley S, Evdokimova VN, Zdobinski DD, Potter DM, Butterfield LH. Hierarchy of α fetoprotein (AFP)-specific T cell responses in subjects with AFP-positive hepatocellular cancer. J Immunol. 2006;177(1):712–721. doi: 10.4049/jimmunol.177.1.712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Perez-Diez A, Butterfield LH, Li L, Chakraborty NG, Economou JS, Mukherji B. Generation of CD8+ and CD4+ T-cell response to dendritic cells genetically engineered to express the MART-1/Melan-A gene. Cancer Res. 1998;58(23):5305–5309. [PubMed] [Google Scholar]
  • 50.Evdokimova VN, Liu Y, Potter DM, Butterfield LH. AFP-specific CD4+ helper T-cell responses in healthy donors and HCC patients. J Immunother. 2007;30(4):425–437. doi: 10.1097/CJI.0b013e31802fd8e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Castellino F, Germain RN. Cooperation between CD4+ and CD8+ T cells: When, where, and how. Annu Rev Immunol. 2006;24:519–540. doi: 10.1146/annurev.immunol.23.021704.115825. [DOI] [PubMed] [Google Scholar]
  • 52.van Stipdonk MJ, Lemmens EE, Schoenberger SP. Naïve CTLs require a single brief period of antigenic stimulation for clonal expansion and differentiation. Nat Immunol. 2001;2(5):423–429. doi: 10.1038/87730. [DOI] [PubMed] [Google Scholar]
  • 53.Mehrotra S, Chhabra A, Chakraborty A, Chattopadhyay S, Slowik M, Stevens R, Zengou R, Mathias C, Butterfield LH, Dorsky DI, Economou JS, et al. Antigen presentation by MART-1 adenovirus-transduced interleukin-10-polarized human monocyte-derived dendritic cells. Immunology. 2004;113(4):472–481. doi: 10.1111/j.1365-2567.2004.01978.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.He Y, Zhang J, Mi Z, Robbins P, Falo LD., Jr Immunization with lentiviral vector-transduced dendritic cells induces strong and long-lasting T cell responses and therapeutic immunity. J Immunol. 2005;174(6):3808–3817. doi: 10.4049/jimmunol.174.6.3808. [DOI] [PubMed] [Google Scholar]
  • 55.Metharom P, Ellem KA, Wei MQ. Gene transfer to dendritic cells induced a protective immunity against melanoma. Cell Mol Immunol. 2005;2(4):281–288. [PubMed] [Google Scholar]
  • 56.Nakamura M, Iwahashi M, Nakamori M, Ueda K, Ojima T, Naka T, Ishida K, Yamaue H. Dendritic cells transduced with tumor-associated antigen gene elicit potent therapeutic antitumor immunity: Comparison with immunodominant peptide-pulsed DCs. Oncology. 2005;68(2–3):163–170. doi: 10.1159/000086770. [DOI] [PubMed] [Google Scholar]
  • 57.Yuan J, Latouche JB, Reagan JL, Heller G, Riviere I, Sadelain M, Young JW. Langerhans cells derived from genetically modified human CD34+ hemopoietic progenitors are more potent than peptide-pulsed Langerhans cells for inducing antigen-specific CD8+ cytolytic T lymphocyte responses. J Immunol. 2005;174(2):758–766. doi: 10.4049/jimmunol.174.2.758. [DOI] [PubMed] [Google Scholar]
  • 58.Curti A, Pandolfi S, Aluigi M, Isidori A, Alessandrini I, Chiodoni C, Testoni N, Colombo MP, Baccarani M, Lemoli RM. Interleukin-12 production by leukemia-derived dendritic cells counteracts the inhibitory effect of leukemic microenvironment on T cells. Exp Hematol. 2005;33(12):1521–1530. doi: 10.1016/j.exphem.2005.08.005. [DOI] [PubMed] [Google Scholar]
  • 59.van Leeuwen EB, Cloosen S, Senden-Gijsbers BL, Germeraad WT, Bos GM. Transduction with a fiber-modified adenoviral vector is superior to non-viral nucleofection for expressing tumor-associated Ag mucin-1 in human DC. Cytotherapy. 2006;8(1):36–46. doi: 10.1080/14653240500508166. [DOI] [PubMed] [Google Scholar]
  • 60.Ranieri E, Kierstead LS, Zarour H, Kirkwood JM, Lotze MT, Whiteside T, Storkus WJ. Dendritic cell/peptide cancer vaccines: Clinical responsiveness and epitope spreading. Immunol Invest. 2000;29(2):121–125. doi: 10.3109/08820130009062294. [DOI] [PubMed] [Google Scholar]
  • 61.Jäger E, Ringhoffer M, Altmannsberger M, Arand M, Karbach J, Jäger D, Oesch F, Knuth A. Immunoselection in vivo: Independent loss of MHC class I and melanocyte differentiation antigen expression in metastatic melanoma. Int J Cancer. 1997;71(2):142–147. doi: 10.1002/(sici)1097-0215(19970410)71:2<142::aid-ijc3>3.0.co;2-0. [DOI] [PubMed] [Google Scholar]
  • 62.Hicklin DJ, Dellaratta DV, Kishore R, Liang B, Kageshita T, Ferrone S. β2-microglobulin gene mutations in human melanoma cells: Molecular characterization and implications for immune surveillance. Melanoma Res. 1997;7(Suppl 2):S67–S74. [PubMed] [Google Scholar]
  • 63.Kageshita T, Hirai S, Ono T, Hicklin DJ, Ferrone S. Down-regulation of HLA class I antigen-processing molecules in malignant melanoma: Association with disease progression. Am J Pathol. 1999;154(3):745–754. doi: 10.1016/S0002-9440(10)65321-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kamarashev J, Ferrone S, Seifert B, Boni R, Nestle FO, Burg G, Dummer R. TAP1 down-regulation in primary melanoma lesions: An independent marker of poor prognosis. Int J Cancer. 2001;95(1):23–28. doi: 10.1002/1097-0215(20010120)95:1<23::aid-ijc1004>3.0.co;2-4. [DOI] [PubMed] [Google Scholar]
  • 65.Seliger B, Ritz U, Abele R, Bock M, Tampe R, Sutter G, Drexler I, Huber C, Ferrone S. Immune escape of melanoma: First evidence of structural alterations in two distinct components of the MHC class I antigen processing pathway. Cancer Res. 2001;61(24):8647–8650. [PubMed] [Google Scholar]
  • 66.Yamshchikov GV, Mullins DW, Chang CC, Ogino T, Thompson L, Presley J, Galavotti H, Aquila W, Deacon D, Ross W, Patterson JW, et al. Sequential immune escape and shifting of T cell responses in a long-term survivor of melanoma. J Immunol. 2005;174(11):6863–6871. doi: 10.4049/jimmunol.174.11.6863. [DOI] [PubMed] [Google Scholar]
  • 67.Jäger E, Maeurer M, Höhn H, Karbach J, Jäger D, Zidianakis Z, Bakhshandeh-Bath A, Orth J, Neukirch C, Necker A, Reichert TE, et al. Clonal expansion of Melan A-specific cytotoxic T lymphocytes in a melanoma patient responding to continued immunization with melanoma-associated peptides. Int J Cancer. 2000;86(4):538–547. doi: 10.1002/(sici)1097-0215(20000515)86:4<538::aid-ijc16>3.0.co;2-g. [DOI] [PubMed] [Google Scholar]
  • 68. Tsao H, Millman P, Linette GP, Hodi FS, Sober AJ, Goldberg MA, Haluska FG. Hypopigmentation associated with an adenovirus-mediated gp100/MART-1-transduced dendritic cell vaccine for metastatic melanoma. Arch Dermatol. 2002;138(6):799–802. doi: 10.1001/archderm.138.6.799.• First clinical trial report of Ad-transduced DCs, demonstrating the feasibility, safety and immune effects of the approach in patients with melanoma.
  • 69.Haluska F, Linette G, Jonash S, Hodi S, Longerich S, Yang S, Webb I, Stowell C, Kaplan J, Roberts B, Goldberg M. Immunologic gene therapy of melanoma: Phase I study of therapy with dendritic cells transduced with recombinant adenoviruses encoding melanoma antigens. Proc Am Soc Clin Oncol. 2000;19:453a. [Google Scholar]
  • 70.Butterfield LH, Comin-Anduix B, Vujanovic L, Lee Y, Dissette VB, Yang JQ, Vu HT, Seja E, Oseguera DK, Potter DM, Glaspy JA, et al. Adenovirus MART-1-engineered autologous dendritic cell vaccine for metastatic melanoma. J Immunother. 2008;31(3):294–309. doi: 10.1097/CJI.0b013e31816a8910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Wargo JA, Schumacher LY, Comin-Anduix B, Dissette VB, Glaspy JA, McBride WH, Butterfield LH, Economou JS, Ribas A. Natural killer cells play a critical role in the immune response following immunization with melanoma-antigen-engineered dendritic cells. Cancer Gene Ther. 2005;12(6):516–527. doi: 10.1038/sj.cgt.7700818. [DOI] [PubMed] [Google Scholar]
  • 72.Hart DN. Dendritic cells: Unique leukocyte populations which control the primary immune response. Blood. 1997;90(9):3245–3287. [PubMed] [Google Scholar]
  • 73.Fernandez NC, Lozier A, Flament C, Ricciardi-Castagnoli P, Bellet D, Suter M, Perricaudet M, Tursz T, Maraskovsky E, Zitvogel L. Dendritic cells directly trigger NK cell functions: Cross-talk relevant in innate anti-tumor immune responsesin vivo . Nat Med. 1999;5(4):405–411. doi: 10.1038/7403. [DOI] [PubMed] [Google Scholar]
  • 74.Gerosa F, Baldani-Guerra B, Nisii C, Marchesini V, Carra G, Trinchieri G. Reciprocal activating interaction between natural killer cells and dendritic cells. J Exp Med. 2002;195(3):327–333. doi: 10.1084/jem.20010938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Piccioli D, Sbrana S, Melandri E, Valiante NM. Contact-dependent stimulation and inhibition of dendritic cells by natural killer cells. J Exp Med. 2002;195(3):335–341. doi: 10.1084/jem.20010934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ferlazzo G, Tsang ML, Moretta L, Melioli G, Steinman RM, Munz C. Human dendritic cells activate resting natural killer (NK) cells and are recognized via the NKp30 receptor by activated NK cells. J Exp Med. 2002;195(3):343–351. doi: 10.1084/jem.20011149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Zamai L, Ponti C, Mirandola P, Gobbi G, Papa S, Galeotti L, Cocco L, Vitale M. NK cells and cancer. J Immunol. 2007;178(7):4011–4016. doi: 10.4049/jimmunol.178.7.4011. [DOI] [PubMed] [Google Scholar]
  • 78.Tourkova IL, Yurkovetsky ZR, Gambotto A, Makarenkova VP, Perez L, Balkir L, Robbins PD, Shurin MR, Shurin GV. Increased function and survival of IL-15-transduced human dendritic cells are mediated by up-regulation of IL-15Rα and Bcl-2. J Leukoc Biol. 2002;72(5):1037–1045. [PubMed] [Google Scholar]
  • 79.Kianmanesh A, Hackett NR, Lee JM, Kikuchi T, Korst RJ, Crystal RG. Intratumoral administration of low doses of an adenovirus vector encoding tumor necrosis factor α together with naïve dendritic cells elicits significant suppression of tumor growth without toxicity. Hum Gene Ther. 2001;12(17):2035–2049. doi: 10.1089/10430340152677395. [DOI] [PubMed] [Google Scholar]
  • 80.Baratelli F, Takedatsu H, Hazra S, Peebles K, Luo J, Kurimoto PS, Zeng G, Batra RK, Sharma S, Dubinett SM, Lee JM. Pre-clinical characterization of GMP grade CCL21-gene modified dendritic cells for application in a phase I trial in non-small cell lung cancer. J Transl Med. 2008;6:38. doi: 10.1186/1479-5876-6-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Vujanovic L, Ranieri E, Gambotto A, Olson WC, Kirkwood JM, Storkus WJ. IL-12p70 and IL-18 gene-modified dendritic cells loaded with tumor antigen-derived peptides or recombinant protein effectively stimulate specific type-1 CD4+ T-cell responses from normal donors and melanoma patientsin vitro . Cancer Gene Ther. 2006;13(8):798–805. doi: 10.1038/sj.cgt.7700964. [DOI] [PubMed] [Google Scholar]
  • 82.Tatsumi T, Huang J, Gooding WE, Gambotto A, Robbins PD, Vujanovic NL, Alber SM, Watkins SC, Okada H, Storkus WJ. Intratumoral delivery of dendritic cells engineered to secrete both interleukin (IL)-12 and IL-18 effectively treats local and distant disease in association with broadly reactive Tc1-type immunity. Cancer Res. 2003;63(19):6378–6386. [PubMed] [Google Scholar]
  • 83.Ahn HJ, Maruo S, Tomura M, Mu J, Hamaoka T, Nakanishi K, Clark S, Kurimoto M, Okamura H, Fujiwara H. A mechanism underlying synergy between IL-12 and IFN-γ-inducing factor in enhanced production of IFN-γ. J Immunol. 1997;159(5):2125–2131. [PubMed] [Google Scholar]
  • 84.Stoll S, Jonuleit H, Schmitt E, Muller G, Yamauchi H, Kurimoto M, Knop J, Enk AH. Production of functional IL-18 by different subtypes of murine and human dendritic cells (DC): DC-derived IL-18 enhances IL-12-dependent Th1 development. Eur J Immunol. 1998;28(10):3231–3239. doi: 10.1002/(SICI)1521-4141(199810)28:10<3231::AID-IMMU3231>3.0.CO;2-Q. [DOI] [PubMed] [Google Scholar]
  • 85.Del Vecchio M, Bajetta E, Canova S, Lotze MT, Wesa A, Parmiani G, Anichini A. Interleukin-12: Biological properties and clinical application. Clin Cancer Res. 2007;13(16):4677–4685. doi: 10.1158/1078-0432.CCR-07-0776. [DOI] [PubMed] [Google Scholar]
  • 86.Qin Z, Blankenstein T. CD4+ T cell-mediated tumor rejection involves inhibition of angiogenesis that is dependent on IFN receptor expression by nonhematopoietic cells. Immunity. 2000;12(6):677–686. doi: 10.1016/s1074-7613(00)80218-6. [DOI] [PubMed] [Google Scholar]
  • 87.Qin Z, Schwartzkopff J, Pradera F, Kammertoens T, Seliger B, Pircher H, Blankenstein T. A critical requirement of interferon γ -mediated angiostasis for tumor rejection by CD8+ T cells. Cancer Res. 2003;63(14):4095–4100. [PubMed] [Google Scholar]
  • 88.Appay V. The physiological role of cytotoxic CD4+ T-cells:The holy grail? Clin Exp Immunol. 2004;138(1):10–13. doi: 10.1111/j.1365-2249.2004.02605.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lowes MA, Bishop GA, Crotty K, Barnetson RS, Halliday GM. T helper 1 cytokine mRNA is increased in spontaneously regressing primary melanoma. J Invest Dermatol. 1997;108(6):914–919. doi: 10.1111/1523-1747.ep12292705. [DOI] [PubMed] [Google Scholar]
  • 90.Schwaab T, Heaney JA, Schned AR, Harris RD, Cole BF, Noelle RJ, Phillips DM, Stempkowski L, Ernstoff MS. A randomized phase II trial comparing two different sequence combinations of autologous vaccine and human recombinant interferon γ and human recombinant interferon α2B therapy in patients with metastatic renal cell carcinoma: Clinical outcome and analysis of immunological parameters. J Urol. 2000;163(4):1322–1327. doi: 10.1016/s0022-5347(05)67771-3. [DOI] [PubMed] [Google Scholar]
  • 91.Wittke F, Hoffmann R, Buer J, Dallmann I, Oevermann K, Sel S, Wandert T, Ganser A, Atzpodien J. Interleukin 10 (IL-10): An immunosuppressive factor and independent predictor in patients with metastatic renal cell carcinoma. Br J Cancer. 1999;79(7–8):1182–1184. doi: 10.1038/sj.bjc.6690189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Okamura H, Kashiwamura S, Tsutsui H, Yoshimoto T, Nakanishi K. Regulation of interferon- production by IL-12 and IL-18. Curr Opin Immunol. 1998;10(3):259–264. doi: 10.1016/s0952-7915(98)80163-5. [DOI] [PubMed] [Google Scholar]
  • 93.Okamura H, Tsutsi H, Komatsu T, Yutsudo M, Hakura A, Tanimoto T, Torigoe K, Okura T, Nukada Y, Hattori K, et al. Cloning of a new cytokine that induces IFN- γ production by T cells. Nature. 1995;378(6552):88–91. doi: 10.1038/378088a0. [DOI] [PubMed] [Google Scholar]
  • 94.Reddy P. Interleukin-18: Recent advances. Curr Opin Hematol. 2004;11(6):405–410. doi: 10.1097/01.moh.0000141926.95319.42. [DOI] [PubMed] [Google Scholar]
  • 95.Gutzmer R, Langer K, Mommert S, Wittmann M, Kapp A, Werfel T. Human dendritic cells express the IL-18R and are chemoattracted to IL-18. J Immunol. 2003;171(12):6363–6371. doi: 10.4049/jimmunol.171.12.6363. [DOI] [PubMed] [Google Scholar]
  • 96.Kaser A, Kaser S, Kaneider NC, Enrich B, Wiedermann CJ, Tilg H. Interleukin-18 attracts plasmacytoid dendritic cells (DC2s) and promotes Th1 induction by DC2s through IL-18 receptor expression. Blood. 2004;103(2):648–655. doi: 10.1182/blood-2002-07-2322. [DOI] [PubMed] [Google Scholar]
  • 97.Yoshimoto T, Takeda K, Tanaka T, Ohkusu K, Kashiwamura S, Okamura H, Akira S, Nakanishi K. IL-12 up-regulates IL-18 receptor expression on T cells, Th1 cells, and B cells: Synergism with IL-18 for IFN-γ production. J Immunol. 1998;161(7):3400–3407. [PubMed] [Google Scholar]
  • 98.Robinson D, Shibuya K, Mui A, Zonin F, Murphy E, Sana T, Hartley SB, Menon S, Kastelein R, Bazan F, O’Garra A. IGIF does not drive Th1 development but synergizes with IL-12 for interferon-γ production and activates IRAK and NFκB. Immunity. 1997;7(4):571–581. doi: 10.1016/s1074-7613(00)80378-7. [DOI] [PubMed] [Google Scholar]
  • 99.Lenzi R, Rosenblum M, Verschraegen C, Kudelka AP, Kavanagh JJ, Hicks ME, Lang EA, Nash MA, Levy LB, Garcia ME, Platsoucas CD, et al. Phase I study of intraperitoneal recombinant human interleukin 12 in patients with Mullerian carcinoma, gastrointestinal primary malignancies, and mesothelioma. Clin Cancer Res. 2002;8(12):3686–3695. [PubMed] [Google Scholar]
  • 100.van Herpen CM, Huijbens R, Looman M, De Vries J, Marres H, Van De Ven J, Hermsen R, Adema GJ, De Mulder PH. Pharmacokinetics and immunological aspects of a phase Ib study with intratumoral administration of recombinant human interleukin-12 in patients with head and neck squamous cell carcinoma: A decrease of T-bet in peripheral blood mononuclear cells. Clin Cancer Res. 2003;9(8):2950–2956. [PubMed] [Google Scholar]
  • 101.Lee P, Wang F, Kuniyoshi J, Rubio V, Stuges T, Groshen S, Gee C, Lau R, Jeffery G, Margolin K, Marty V, et al. Effects of interleukin-12 on the immune response to a multipeptide vaccine for resected metastatic melanoma. J Clin Oncol. 2001;19(18):3836–3847. doi: 10.1200/JCO.2001.19.18.3836. [DOI] [PubMed] [Google Scholar]
  • 102.van Herpen CM, Looman M, Zonneveld M, Scharenborg N, de Wilde PC, van de Locht L, Merkx MA, Adema GJ, de Mulder PH. Intratumoral administration of recombinant human interleukin 12 in head and neck squamous cell carcinoma patients elicits a T-helper 1 profile in the locoregional lymph nodes. Clin Cancer Res. 2004;10(8):2626–2635. doi: 10.1158/1078-0432.ccr-03-0304. [DOI] [PubMed] [Google Scholar]
  • 103.Atkins MB, Robertson MJ, Gordon M, Lotze MT, DeCoste M, DuBois JS, Ritz J, Sandler AB, Edington HD, Garzone PD, Mier JW, et al. Phase I evaluation of intravenous recombinant human interleukin 12 in patients with advanced malignancies. Clin Cancer Res. 1997;3(3):409–417. [Google Scholar]
  • 104.Younes A, Pro B, Robertson MJ, Flinn IW, Romaguera JE, Hagemeister F, Dang NH, Fiumara P, Loyer EM, Cabanillas FF, McLaughlin PW, et al. Phase II clinical trial of interleukin-12 in patients with relapsed and refractory non-Hodgkin’s lymphoma and Hodgkin’s disease. Clin Cancer Res. 2004;10(16):5432–5438. doi: 10.1158/1078-0432.CCR-04-0540. [DOI] [PubMed] [Google Scholar]
  • 105.Mazzolini G, Narvaiza I, Perez-Diez A, Rodriguez-Calvillo M, Qian C, Sangro B, Ruiz J, Prieto J, Melero I. Genetic heterogeneity in the toxicity to systemic adenoviral gene transfer of interleukin-12. Gene Ther. 2001;8(4):259–267. doi: 10.1038/sj.gt.3301387. [DOI] [PubMed] [Google Scholar]
  • 106.Carson WE, Dierksheide JE, Jabbour S, Anghelina M, Bouchard P, Ku G, Yu H, Baumann H, Shah MH, Cooper MA, Durbin J, et al. Coadministration of interleukin-18 and interleukin-12 induces a fatal inflammatory response in mice: Critical role of natural killer cell interferon-γ production and STAT-mediated signal transduction. Blood. 2000;96(4):1465–1473. [PubMed] [Google Scholar]
  • 107.Sangro B, Mazzolini G, Ruiz J, Herraiz M, Quiroga J, Herrero I, Benito A, Larrache J, Pueyo J, Subtil JC, Olague C, et al. Phase I trial of intratumoral injection of an adenovirus encoding interleukin-12 for advanced digestive tumors. J Clin Oncol. 2004;22(8):1389–1397. doi: 10.1200/JCO.2004.04.059. [DOI] [PubMed] [Google Scholar]
  • 108.Mazzolini G, Alfaro C, Sangro B, Feijoo E, Ruiz J, Benito A, Tirapu I, Arina A, Sola J, Herraiz M, Lucena F, et al. Intratumoral injection of dendritic cells engineered to secrete interleukin-12 by recombinant adenovirus in patients with metastatic gastrointestinal carcinomas. J Clin Oncol. 2005;23(5):999–1010. doi: 10.1200/JCO.2005.00.463. [DOI] [PubMed] [Google Scholar]
  • 109.Mazzolini G, Prieto J, Melero I. Gene therapy of cancer with interleukin-12. Curr Pharm Des. 2003;9(24):1981–1991. doi: 10.2174/1381612033454261. [DOI] [PubMed] [Google Scholar]
  • 110.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 responses to colon adenocarcinoma in the liver in mice. J Exp Ther Oncol. 2002;2(6):337–349. doi: 10.1046/j.1359-4117.2002.01050.x. [DOI] [PubMed] [Google Scholar]
  • 111.Tatsumi T, Takehara T, Yamaguchi S, Sasakawa A, Miyagi T, Jinushi M, Sakamori R, Kohga K, Uemura A, Ohkawa K, Storkus WJ, et al. Injection of IL-12 gene-transduced dendritic cells into mouse liver tumor lesions activates both innate and acquired immunity. Gene Ther. 2007;14(11):863–871. doi: 10.1038/sj.gt.3302941. [DOI] [PubMed] [Google Scholar]
  • 112. Komita H, Zhao X, Katakam AK, Kumar P, Kawabe M, Okada H, Braughler JM, Storkus WJ. Conditional interleukin-12 gene therapy promotes safe and effective antitumor immunity. Cancer Gene Ther. 2009;16(12):883–891. doi: 10.1038/cgt.2009.33.• Describes a regulatory element encoded by an AdV that is responsive to a soluble, orally available ligand. This advance allows transgene expression to be induced or repressed by administering a small-molecule compound. The regulated IL-12 Ad, transduced into DCs, is in clinical testing for melanoma.
  • 113.Kumar PKA. RheoSwitch system: A highly sensitive ecdysone receptor-based gene regulation system induced by synthetic small-molecule ligands. In: Friedmann T, Rossi JJ, editors. Gene Transfer: Delivery and Expression of DNA and RNA. Cold Spring Harbor, NY, USA: Cold Spring Harbor Laboratory Press; 2007. pp. 643–651. [Google Scholar]
  • 114.Knippertz I, Hesse A, Schunder T, Kampgen E, Brenner MK, Schuler G, Steinkasserer A, Nettelbeck DM. Generation of human dendritic cells that simultaneously secrete IL-12 and have migratory capacity by adenoviral gene transfer of hCD40L in combination with IFN-γ. J Immunother. 2009;32(5):524–538. doi: 10.1097/CJI.0b013e3181a28422. [DOI] [PubMed] [Google Scholar]
  • 115.Tuettenberg A, Fondel S, Steinbrink K, Enk AH, Jonuleit H. CD40 signalling induces IL-10-producing, tolerogenic dendritic cells. Exp Dermatol. 19(1):44–53. doi: 10.1111/j.1600-0625.2009.00975.x. [DOI] [PubMed] [Google Scholar]
  • 116.Rossi D, Zlotnik A. The biology of chemokines and their receptors. Annu Rev Immunol. 2000;18:217–242. doi: 10.1146/annurev.immunol.18.1.217. [DOI] [PubMed] [Google Scholar]
  • 117.Riedl K, Baratelli F, Batra RK, Yang SC, Luo J, Escuadro B, Figlin R, Strieter R, Sharma S, Dubinett S. Overexpression of CCL-21/secondary lymphoid tissue chemokine in human dendritic cells augments chemotactic activities for lymphocytes and antigen presenting cells. Mol Cancer. 2003;2:35. doi: 10.1186/1476-4598-2-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kirk CJ, Hartigan-O’Connor D, Mule JJ. The dynamics of the T-cell antitumor response: Chemokine-secreting dendritic cells can prime tumor-reactive T cells extranodally. Cancer Res. 2001;61(24):8794–8802. [PubMed] [Google Scholar]
  • 119.Mule JJ. Dendritic cell-based vaccines for pancreatic cancer and melanoma. Ann N Y Acad Sci. 2009;1174:33–40. doi: 10.1111/j.1749-6632.2009.04936.x. [DOI] [PubMed] [Google Scholar]
  • 120.Rea D, Havenga MJ, van Den Assem M, Sutmuller RP, Lemckert A, Hoeben RC, Bout A, Melief CJ, Offringa R. Highly efficient transduction of human monocyte-derived dendritic cells with subgroup B fiber-modified adenovirus vectors enhances transgene-encoded antigen presentation to cytotoxic T cells. J Immunol. 2001;166(8):5236–5244. doi: 10.4049/jimmunol.166.8.5236. [DOI] [PubMed] [Google Scholar]
  • 121.Sakurai F, Nakashima K, Yamaguchi T, Ichinose T, Kawabata K, Hayakawa T, Mizuguchi H. Adenovirus serotype 35 vector-induced innate immune responses in dendritic cells derived from wild-type and human CD46-transgenic mice: Comparison with a fiber-substituted Ad vector containing fiber proteins of Ad serotype 35. J Control Release. 2010 doi: 10.1016/j.jconrel.2010.08.025. [DOI] [PubMed] [Google Scholar]
  • 122.Nwanegbo E, Vardas E, Gao W, Whittle H, Sun H, Rowe D, Robbins PD, Gambotto A. Prevalence of neutralizing antibodies to adenoviral serotypes 5 and 35 in the adult populations of The Gambia, South Africa, and the United States. Clin Diagn Lab Immunol. 2004;11(2):351–357. doi: 10.1128/CDLI.11.2.351-357.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Short JJ, Pereboev AV, Kawakami Y, Vasu C, Holterman MJ, Curiel DT. Adenovirus serotype 3 utilizes CD80 (B7.1) and CD86 (B7.2) as cellular attachment receptors. Virology. 2004;322(2):349–359. doi: 10.1016/j.virol.2004.02.016. [DOI] [PubMed] [Google Scholar]
  • 124. de Gruijl TD, Luykx-de Bakker SA, Tillman BW, van den Eertwegh AJ, Buter J, Lougheed SM, van der Bij GJ, Safer AM, Haisma HJ, Curiel DT, Scheper RJ, et al. Prolonged maturation and enhanced transduction of dendritic cells migrated from human skin explants after in situdelivery of CD40-targeted adenoviral vectors. J Immunol. 2002;169(9):5322–5331. doi: 10.4049/jimmunol.169.9.5322.• Describes an important and complex in vitro model of human skin in which Ad was targeted via CD40 to DCs in situ, and the resultant DC activation and migration, thereby progressing this in vivo DC-targeting approach closer to clinical testing.
  • 125.Hangalapura BN, Oosterhoff D, Aggarwal S, Wijnands PG, van de Ven R, Santegoets SJ, van den Tol MP, Hooijberg E, Pereboev A, van den Eertwegh AJ, Curiel DT, et al. Selective transduction of dendritic cells in human lymph nodes and superior induction of high-avidity melanoma-reactive cytotoxic T cells by a CD40-targeted adenovirus. J Immunother. 2010;33(7):706–715. doi: 10.1097/CJI.0b013e3181eccbd4. [DOI] [PubMed] [Google Scholar]
  • 126.Sapinoro R, Maguire CA, Burgess A, Dewhurst S. Enhanced transduction of dendritic cells by FcγRI-targeted adenovirus vectors. J Gene Med. 2007;9(12):1033–1045. doi: 10.1002/jgm.1112. [DOI] [PubMed] [Google Scholar]
  • 127.Kim HS, Kim CH, Park MY, Park JS, Park HM, Sohn HJ, Kim HJ, Kim SG, Oh ST, Kim TG. Efficient co-transduction of adenoviral vectors encoding carcinoembryonic antigen and survivin into dendritic cells by the CAR-TAT adaptor molecule enhance anti-tumor immunity in a murine colorectal cancer model. Immunol Lett. 2010;131(1):73–80. doi: 10.1016/j.imlet.2010.03.001. [DOI] [PubMed] [Google Scholar]
  • 128.Lapteva N, Aldrich M, Weksberg D, Rollins L, Goltsova T, Chen SY, Huang XF. Targeting the intratumoral dendritic cells by the oncolytic adenoviral vaccine expressing RANTES elicits potent antitumor immunity. J Immunother. 2009;32(2):145–156. doi: 10.1097/CJI.0b013e318193d31e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Sarkar D, Su ZZ, Park ES, Vozhilla N, Dent P, Curiel DT, Fisher PB. A cancer terminator virus eradicates both primary and distant human melanomas. Cancer Gene Ther. 2008;15(5):293–302. doi: 10.1038/cgt.2008.14. [DOI] [PubMed] [Google Scholar]
  • 130.Huang JH, Zhang SN, Choi KJ, Choi IK, Kim JH, Lee MG, Kim H, Yun CO. Therapeutic and tumor-specific immunity induced by combination of dendritic cells and oncolytic adenovirus expressing IL-12 and 4-1BBL. Mol Ther. 2010;18(2):264–274. doi: 10.1038/mt.2009.205. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES