associate editor: michael g. rosenblum dendritic...

23
1521-0081/67/4/731753$25.00 http://dx.doi.org/10.1124/pr.114.009456 PHARMACOLOGICAL REVIEWS Pharmacol Rev 67:731753, October 2015 Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics ASSOCIATE EDITOR: MICHAEL G. ROSENBLUM Dendritic Cells as Pharmacological Tools for Cancer Immunotherapy s Sébastien Anguille, Evelien L. Smits, Christian Bryant, Heleen H. Van Acker, Herman Goossens, Eva Lion, Phillip D. Fromm, Derek N. Hart, Viggo F. Van Tendeloo, and Zwi N. Berneman Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F., D.N.H.) Abstract..................................................................................... 732 I. Cancer Immunotherapy ..................................................................... 732 II. Dendritic CellBased Cancer Immunotherapy ................................................ 734 A. Immunobiological Aspects of Dendritic CellBased Cancer Immunotherapy ............... 734 B. Pharmacological Aspects of Dendritic CellBased Cancer Immunotherapy................. 736 III. Toward the Delivery of Effective Dendritic CellBased Cancer Immunotherapy: The Acute Myeloid Leukemia Showcase ................................................................ 739 A. Using Dendritic Cells with Optimal Immunocompetence (Condition Number 1) ........... 740 1. Source of DC Precursor Cell........................................................... 740 2. Culture Time......................................................................... 740 3. Culture Media........................................................................ 740 4. Differentiation Protocol............................................................... 740 5. Maturation Protocol................................................................... 740 6. Choice of Target Antigen.............................................................. 742 7. Source of Antigen(s)................................................................... 742 8. Antigen-Loading Method.............................................................. 743 9. Designer Dendritic Cells.............................................................. 744 10. Route of Administration............................................................... 744 B. Enhancing the Strength of the Antitumor Immune Effector Cells (Condition Number 2) . . . 744 1. Enhancing T-CellMediated Antitumor Immunity..................................... 744 a. Cytotoxic T-lymphocyte antigen4.................................................. 745 b. Programmed death-1. ............................................................. 745 c. B and T lymphocyte attenuator.................................................... 745 d. Immunosuppressive cells. ......................................................... 745 2. Enhancing Natural Killer CellMediated Antitumor Immunity......................... 745 C. Making Tumor Cells Susceptible to Immune Attack (Condition Number 3) ................ 746 1. Targeting Programmed Death-1....................................................... 746 2. Targeting Indoleamine 2,3-Dioxygenase............................................... 746 3. Targeting CD200. .................................................................... 746 This work was supported by grants of the Research Foundation Flanders (FWO Vlaanderen, Grant G039914N); the Methusalem program of the Flemish Government attributed to Goossens; the Belgian Foundation against Cancer (Stichting tegen Kanker); the Belgian Public Utility Foundation VOCATIO; and the Belgian Hercules Foundation. S.A. is a former PhD fellow of the Research Foundation Flanders and a former holder of an Emmanuel van der Schueren Fellowship granted by the Flemish League against Cancer (Vlaamse Liga tegen Kanker). S.A. was awarded a 2012 Endeavour Research Fellowship by the Department of Industry, Innovation, Science, Research and Tertiary Education of the Australian Government. Address correspondence to: Dr. Sébastien Anguille, Antwerp University Hospital, Center for Cell Therapy & Regenerative Medicine (U111), Wilrijkstraat 10, 2650 Edegem, Belgium. E-mail: [email protected] dx.doi.org/10.1124/pr.114.009456. s This article has supplemental material available at pharmrev.aspetjournals.org. 731 by guest on February 1, 2019 Downloaded from

Upload: dangnhi

Post on 12-Dec-2018

213 views

Category:

Documents


0 download

TRANSCRIPT

1521-0081/67/4/731–753$25.00 http://dx.doi.org/10.1124/pr.114.009456PHARMACOLOGICAL REVIEWS Pharmacol Rev 67:731–753, October 2015Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics

ASSOCIATE EDITOR: MICHAEL G. ROSENBLUM

Dendritic Cells as Pharmacological Tools for CancerImmunotherapys

Sébastien Anguille, Evelien L. Smits, Christian Bryant, Heleen H. Van Acker, Herman Goossens, Eva Lion, Phillip D. Fromm,Derek N. Hart, Viggo F. Van Tendeloo, and Zwi N. Berneman

Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, TumorImmunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological

Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital,Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of

Sydney, Sydney, New South Wales, Australia (C.B., P.D.F., D.N.H.)

Abstract. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 732I. Cancer Immunotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 732II. Dendritic Cell–Based Cancer Immunotherapy. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 734

A. Immunobiological Aspects of Dendritic Cell–Based Cancer Immunotherapy . . . . . . . . . . . . . . . 734B. Pharmacological Aspects of Dendritic Cell–Based Cancer Immunotherapy. . . . . . . . . . . . . . . . . 736

III. Toward the Delivery of Effective Dendritic Cell–Based Cancer Immunotherapy: The AcuteMyeloid Leukemia Showcase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 739A. Using Dendritic Cells with Optimal Immunocompetence (Condition Number 1) . . . . . . . . . . . 740

1. Source of DC Precursor Cell.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7402. Culture Time. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7403. Culture Media. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7404. Differentiation Protocol. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7405. Maturation Protocol.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7406. Choice of Target Antigen. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7427. Source of Antigen(s).. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7428. Antigen-Loading Method. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7439. Designer Dendritic Cells. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 74410. Route of Administration.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 744

B. Enhancing the Strength of the Antitumor Immune Effector Cells (Condition Number 2) . . . 7441. Enhancing T-Cell–Mediated Antitumor Immunity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 744

a. Cytotoxic T-lymphocyte antigen‐4. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 745b. Programmed death-1. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 745c. B and T lymphocyte attenuator. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 745d. Immunosuppressive cells. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 745

2. Enhancing Natural Killer Cell–Mediated Antitumor Immunity.. . . . . . . . . . . . . . . . . . . . . . . . 745C. Making Tumor Cells Susceptible to Immune Attack (Condition Number 3) . . . . . . . . . . . . . . . . 746

1. Targeting Programmed Death-1. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7462. Targeting Indoleamine 2,3-Dioxygenase. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7463. Targeting CD200. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 746

This work was supported by grants of the Research Foundation Flanders (FWO Vlaanderen, Grant G039914N); the Methusalem programof the Flemish Government attributed to Goossens; the Belgian Foundation against Cancer (Stichting tegen Kanker); the Belgian PublicUtility Foundation VOCATIO; and the Belgian Hercules Foundation. S.A. is a former PhD fellow of the Research Foundation Flanders anda former holder of an Emmanuel van der Schueren Fellowship granted by the Flemish League against Cancer (Vlaamse Liga tegen Kanker).S.A. was awarded a 2012 Endeavour Research Fellowship by the Department of Industry, Innovation, Science, Research and TertiaryEducation of the Australian Government.

Address correspondence to: Dr. Sébastien Anguille, Antwerp University Hospital, Center for Cell Therapy & Regenerative Medicine(U111), Wilrijkstraat 10, 2650 Edegem, Belgium. E-mail: [email protected]

dx.doi.org/10.1124/pr.114.009456.s This article has supplemental material available at pharmrev.aspetjournals.org.

731

by guest on February 1, 2019

Dow

nloaded from

4. Mobilizing Acute Myeloid Leukemia Cells from Immune-Privileged Bone MarrowNiches. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 746

5. Undoing Tumor Cells’ Cloak of Invisibility.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7476. Reducing Tumor Burden. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 747

IV. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 748References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 748

Abstract——Although the earliest—rudimentary—attempts at exploiting the immune system for cancertherapy can be traced back to the late 18th Century,it was not until the past decade that cancerimmunotherapeutics have truly entered mainstreamclinical practice. Given their potential to stimulate bothadaptive and innate antitumor immune responses,dendritic cells (DCs) have come under intensescrutiny in recent years as pharmacological tools forcancer immunotherapy. Conceptually, the clinicaleffectiveness of this form of active immunotherapyrelies on the completion of three critical steps: 1) theDCs used as immunotherapeutic vehicles must properlyactivate the antitumor immune effector cells of thehost, 2) these immune effector cells must be receptive to

stimulation by the DCs and be competent to mediatetheir antitumor effects, which 3) requires overcomingthe various immune-inhibitorymechanisms used by thetumor cells. In this review, following a brief overviewof the pivotal milestones in the history of cancerimmunotherapy, we will introduce the reader tothe basic immunobiological and pharmacologicalprinciples of active cancer immunotherapy usingDCs. We will then discuss how current research istrying to define the optimal parameters for each ofthe above steps to realize the full clinical potential ofDC therapeutics. Given its high suitability forimmune interventions, acute myeloid leukemia waschosen here to showcase the latest research trendsdriving the field of DC-based cancer immunotherapy.

I. Cancer Immunotherapy

Although the idea of exploiting the immune systemfor cancer therapy has been around for more than twocenturies, immunotherapy has only recently becomea mainstream cancer treatment (Rosenberg, 1999).This journey culminated in immunotherapy beingnominated by the journal Science as Breakthrough ofthe Year for 2013 (Couzin-Frankel, 2013). The timelinepresented in Fig. 1 provides a nonexhaustive overviewof the pivotal milestones in the history of cancerimmunotherapy. The first known attempt to immunizeagainst cancer was made in 1777 by the surgeon to theDuke of Kent, James Nooth, who injected himself withmalignant tissue to prevent the development of cancer(Rosenberg, 1999; Ichim, 2005). Cancer immunother-apy formally started in 1891 (Ichim, 2005) (Fig. 1),when William Coley —regarded by many as the Fatherof Immunotherapy— described the regression of cancerafter injection of a bacterial preparation and postu-lated that this was due to the induction of antitumorimmunity (Coley, 1891). In 1909, Paul Ehrlich hypoth-esized that the immune system could protect the hostagainst developing cancers, thereby laying the founda-tion for the “cancer immune surveillance” theory(Ehrlich, 1909). It took another 50 years, however,before the concept of cancer immune surveillance was

developed by Lewis Thomas (Thomas, 1959) and FrankMacFarlane Burnet (Burnet, 1967). Thomas’ andBurnet’s views on the critical role of the immunesystem in preventing the development of cancer weresupported by a series of mouse tumor transplantationexperiments conducted during the mid-1940s to early-1960s (Fig. 1), which led to the hypothesis that tumorcells express specific “antigens” that permit theirrecognition and elimination by the immune system(Gross, 1943; Foley, 1953; Baldwin, 1955; Prehn andMain, 1957; Klein et al., 1960). In the followingdecades, the interactions between the immune systemand cancer cells were defined, resulting in the centraltenet of cancer immunotherapy: the finding that CD8+

effector T cells (also called cytotoxic T lymphocytes[CTLs]) are capable of selectively killing tumor cellsafter recognition of specific antigens expressed on thetumor cell surface in association with major histocom-patibility complex (MHC) molecules (Zinkernagel andDoherty, 1974a,b; Coulie et al., 2014). More recently,other immune components, such as natural killer (NK)cells, have been shown to contribute to the host defenseagainst cancer (Marcus et al., 2014).

Most currently available cancer immunotherapeuticsaim to harness the T-cell (adaptive) immune responseagainst cancer (Coulie et al., 2014). These can broadlybe divided into passive and active therapies (Mellman

ABBREVIATIONS: ACT, adoptive cell transfer; AML, acute myeloid leukemia; APC, antigen-presenting cell; BTLA, B and T lymphocyteattenuator; CTL, cytotoxic T lymphocyte; CTLA-4, cytotoxic T-lymphocyte antigen‐4; CXCR-4, C-X-C chemokine receptor type-4; DC, dendriticcell; GM-CSF, granulocyte-macrophage colony-stimulating factor; HMGB1, high mobility group box 1; HOCl, hypochlorous acid; HSP, heat-shock protein; IDO, indoleamine 2,3-dioxygenase; IFN, interferon; IL, interleukin; LC, Langerhans cell; mAb, monoclonal antibody; mDC,myeloid dendritic cell; MDSC, myeloid-derived suppressor cell; MHC, major histocompatibility complex; moDC, monocyte-derived dendriticcell; NK, natural killer; PBMC, peripheral blood mononuclear cell; PD-1, programmed death-1; pDC, plasmacytoid dendritic cell; PD-L1/2,programmed death-ligand 1 or 2; PGE2, prostaglandin E2; poly(I:C), polyinosinic:polycytidylic acid; TCR, T-cell receptor; TH, T helper type;TLR, Toll-like receptor; TNF, tumor necrosis factor; Treg, regulatory T cell; WT1, Wilms’ tumor 1.

732 Anguille et al.

et al., 2011; Topalian et al., 2011). Passive immuno-therapies have been in clinical practice for some timein the form of T-cell immunomodulatory monoclonalantibodies (mAbs) and adoptive cell transfer (ACT)(Mellman et al., 2011). Ipilimumab (Yervoy; Bristol-Myers Squibb, New Brunswick, NJ) is the prototype ofthe rapidly growing family of T-cell–potentiatingmAbs. It targets cytotoxic T-lymphocyte antigen‐4(CTLA-4), which serves as an immune checkpointmolecule on T cells (i.e., a molecule involved in thenegative regulation of T-cell function) (Hodi et al.,2010; Pardoll, 2012). ACT involves the transfusion ofallogeneic or autologous tumor-reactive T cells (and/orother antitumor immune effector cells) (June, 2007).The past decade has witnessed several major advance-ments in the field of ACT, including the use of T cellsgenetically engineered to express T-cell receptors(TCRs) or chimeric antigen receptors to enable target-ing of predefined, clinically relevant tumor antigens(Park et al., 2011; Restifo et al., 2012). For detailedinformation on mAb-mediated immune checkpointblockade and ACT, we refer the reader to otheroverview articles on these topics (Pardoll, 2012;Kershaw et al., 2013).This review will focus on active immunotherapy

using tumor antigen–loaded dendritic cells (DCs) foranticancer “vaccination.” Although it is increasinglyevident that at least part of its pharmacologicalmechanism-of-action relies on activation of innateantitumor effectors (in particular NK cells) (Lionet al., 2012a; Van Elssen et al., 2014; Van Ackeret al., 2015), the basic concept behind DC therapy is toexploit the intrinsic antigen-presenting properties of

DCs to elicit a potent tumor antigen-specific T-cell–driven immune response (Anguille et al., 2014; Dattaet al., 2014). In contrast to the two aforementionedpassive cancer immunotherapy strategies, DC therapyaspires to “train” the host’s immune system to recognizeand attack cancer cells and, in so doing, enable thegeneration of immunologic memory (Rescigno et al.,2007; Melero et al., 2014). As shown in the timeline, theDC story began more than four decades ago in 1973(Fig. 1). In that year, Zanvil Cohn and Ralph Steinman(who was awarded the 2011 Nobel Prize in Medicineand Physiology for his seminal work on DCs) describeda rare population of mouse splenocytes with a distinc-tive tree-like morphology (Steinman and Cohn, 1973).The cells were therefore named dendritic cells, after theGreek word for tree (dendreon) (Steinman, 2007).Interest in the therapeutic exploitation of DCs grewafter it became apparent that DCs, in their role ashighly specialized antigen-presenting cells (APCs), areinstrumental in the development of adaptive antitumorimmunity (Palucka and Banchereau, 2012; Mellman,2013). The first clinical trial of DC-based vaccinetherapy was published in 1996 (Hsu et al., 1996) (Fig. 1).Since then, numerous clinical studies have beenconducted across a wide range of cancer types, in-cluding phase III clinical trials in melanoma, prostatecancer, glioblastoma, and renal cell carcinoma (Anguilleet al., 2014). These studies provided ample evidencethat DC therapy is safe and capable of mountingprotective antitumor immunity without major toxicity(Anguille et al., 2014). Until recently, the clinicalbenefit of DC-based cancer immunotherapy wasdeemed modest. However, this field was reinvigorated

Fig. 1. Milestones in the history of cancer immunotherapy. The timeline depicts some of the pivotal milestones in the history of cancerimmunotherapy, with a particular focus on dendritic cell (DC)-based cancer immunotherapy. References: 1(Rosenberg, 1999); 2(Coley, 1891); 3(Ehrlich,1909); 4–8(Gross, 1943; Foley, 1953; Baldwin, 1955; Prehn and Main, 1957; Klein et al., 1960); 9(Thomas et al., 1957); 10, 11(Thomas, 1959; Burnet, 1967);12(Coulie et al., 2014); 13(Steinman and Cohn, 1973); 14, 15(Zinkernagel and Doherty, 1974a,b); 16(Kiessling et al., 1975); 17(van der Bruggen et al., 1991);18(Hsu et al., 1996); 19(Kantoff et al., 2010); 20(Pardoll, 2012). *Allogeneic hematopoietic stem-cell transplantation, which is nowadays widely used inthe treatment of hematologic malignancies including acute myeloid leukemia, is considered as a form of immunotherapy because its primarymechanism-of-action relies on the transfusion of lymphocytes endowed with antitumor activity (the so-called graft-versus-leukemia or graft-versus-tumor activity) (Topalian et al., 2011). #Ipilimumab (Yervoy; Bristol-Myers Squibb), an mAb directed against CTLA-4, was the first-in-class immunecheckpoint mAb to be approved by the U.S. Food and Drug Administration (FDA) and by the European Medicines Agency for patients with metastaticmelanoma (in 2011). Nivolumab (Opdivo; Bristol-Myers Squibb/Ono Pharmaceutical) and pembrolizumab (Keytruda; Merck, Kenilworth, NJ), twomAbs directed against the immune checkpoint molecule PD-1, have gained recent approval by the Japanese health authorities and the FDA,respectively, for use in patients with advanced melanoma (Galluzzi et al., 2014).

Dendritic Cell–Based Cancer Immunotherapy 733

after the demonstration of survival gains in DC-treatedcancer patients in a number of trials (Anguille et al.,2014; Cao et al., 2014; Wang et al., 2014). The foremostexample is sipuleucel-T (Provenge; Dendreon Corpora-tion, Seattle, WA), the first and currently the only DC-based immunotherapeutic that received approval fromthe U.S. Food and Drug Administration. Despitelimited therapeutic benefit based on the assessment ofthe prostate-specific antigen tumor marker response,sipuleucel-T was found to improve overall survival by4.1 months compared with placebo (25.8 versus 21.7months) in a phase III clinical trial for metastatichormone-refractory prostate cancer (Kantoff et al.,2010). The field of DC-based cancer immunotherapycontinues to evolve rapidly (Anguille et al., 2014).Current research efforts are driven by a constant desireto optimize this form of immunotherapy and to increaseits antitumor activity (Datta et al., 2014). There arenow a diverse range of DC vaccine products withimproved therapeutic activity (Anguille et al., 2014).The ever-expanding portfolio of immune checkpointmAbs and other cancer (immunotherapy) drugs has setthe stage for combination therapies, which will likelyfurther improve the efficacy of DC-based cancerimmunization (Anguille et al., 2014). Following a briefintroduction into the basics of the biology of DCs(section II.A) and the various DC vaccine formulationscurrently used for immunotherapy (section II.B), wewill review the different avenues that are currentlybeing pursued to maximize the immunologic potencyand therapeutic activity of this exciting and promisingimmunotherapeutic modality (section III).

II. Dendritic Cell–Based Cancer Immunotherapy

A. Immunobiological Aspects of Dendritic Cell–BasedCancer Immunotherapy

Dendritic cells are a rare population of APCs thatoriginate from hematopoietic progenitor cells in thebone marrow (Collin et al., 2013). They are specializedin antigen uptake, processing, and presentation toT cells (MacDonald et al., 2002). DCs can be foundthroughout the body, in particular at sites of potentialantigenic encounter (e.g., the skin) (Benteyn et al.,2015). The taxonomy of the DC system is complex andwill be only briefly discussed here. The reader isdirected to MacDonald et al. (2002), Ziegler-Heitbrocket al. (2010), Collin et al. (2013), Merad et al. (2013),Cohn and Delamarre (2014), and Mody et al. (2015), fora comprehensive review of the DC nomenclature. Thehuman DC family consists of several distinct subsets,which differ from each other with respect to theirlocalization, cell-surface phenotype, and functionalspecialization (Collin et al., 2013). For example, thehuman skin hosts two main types of DCs: epidermalLangerhans cells (LCs) and dermal DCs (Mody et al.,2015). Langerhans cells are characterized by expression

of the cell-surface markers CD1a and CD207/Langerinand are functionally endowed with a potent CTL-stimulatory activity, especially when compared withCD14+ dermal DCs (Ueno et al., 2010; Banchereauet al., 2012). Blood DCs, which constitute less than 1%of the total peripheral blood mononuclear cell (PBMC)fraction (Jongbloed et al., 2010), are usually dividedinto two main subsets based on their hematopoieticcell-lineage origin (MacDonald et al., 2002): myeloidDCs (mDCs; also termed conventional DCs; myeloidlineage) and plasmacytoid DCs (pDCs; lymphoidlineage). The myeloid DC population is characterizedby the phenotypic expression of CD11c and containstwo distinct subsets (MacDonald et al., 2002; Collin et al.,2013): CD1c/BDCA-1+ mDCs and CD141/BDCA-3+

mDCs. Plasmacytoid DCs can be phenotypicallydiscriminated from mDCs based on the lack of CD11cexpression and based on their positivity for CD123,CD303/BDCA-2, and CD304/BDCA-4 (MacDonaldet al., 2002; Collin et al., 2013). The functionaldifferences between these individual DC subsets, inparticular the subroles they play in antitumor immu-nity, remain to be fully elucidated. CD141+ mDCs havebeen postulated to be superior over other DCs subsetsat (tumor) antigen cross-presentation (Jongbloed et al.,2010) [i.e., the ability of APCs to present exogenousantigens—which are normally directed into the MHCclass II pathway—via MHC class I molecules to CD8+

T cells (Nierkens et al., 2013; Cohn and Delamarre,2014)], although more recent studies have shown thatall blood DC subsets have fairly similar cross-presentation capacities (Cohn et al., 2013; Seguraet al., 2013; Tel et al., 2013b).

Dendritic cells can also be subdivided into two maintypes based on their developmental stage: immatureDCs and mature DCs (Datta et al., 2014; Benteynet al., 2015). Immature DCs continuously sample theperipheral tissues for foreign antigens. They are highlyspecialized in uptake of these antigens and areequipped with a wide range of phagocytic andendocytic receptors (e.g., C-type lectins, such as themannose receptor) to fulfill this function (Sallustoet al., 1995). Immature DCs have rather low expressionof MHC and T-cell costimulatory molecules on theircell surface and a limited capacity for secretingproinflammatory cytokines (Benteyn et al., 2015). Asa result, they are largely ineffective at stimulatingT-cell responses (Benteyn et al., 2015). Moreover,immature DCs can lead to T-cell tolerance by inducingT-cell deletion/anergy and by promoting the generationof T cells with intrinsic immunosuppressive properties[also called regulatory T (Treg) cells] (Mahnke et al.,2002). When antigen uptake occurs in the presence ofinflammatory cytokines or danger signals (such asthose provided by infectious organisms or dying cancercells), immature DCs will undergo a maturation pro-cess that is characterized by decreased capacity for

734 Anguille et al.

antigen uptake, increased expression of MHC and T-cell costimulatory molecules (such as CD40, CD70,CD80, and CD86), and enhanced production of proin-flammatory cytokines and chemokines (Benteyn et al.,2015). DCs express different families of patternrecognition receptors that enable them to sense suchdanger signals; among these, the family of the so-calledToll-like receptors (TLRs) is perhaps the best charac-terized (Medzhitov, 2001). As will be discussed later inthis review, TLR ligands are being increasinglyadopted in DC vaccine manufacturing protocols tostimulate DC maturation. Examples are polyinosinic:polycytidylic acid [poly(I:C), a TLR3 agonist] (Ammiet al., 2015), picibanil (OK-432, a TLR4 agonist)(Kitawaki et al., 2011a), and resiquimod (R-848, a dualTLR7/8 agonist) (Smits et al., 2008). The conversion ofimmature DCs into mature DCs is also accompaniedby an upregulation of chemokine receptors, such as theC:C chemokine receptor type-7 (Sallusto et al., 1998).

This enables the DCs to migrate via the afferentlymphatics to the T-cell–rich areas of the lymph nodes(De Vries et al., 2003; Wimmers et al., 2014) (seeSupplemental Video for an animation illustrating theprocess of DC migration to the lymph nodes).

In the lymph nodes, DCs will present the antigensthey captured via MHC molecules to T cells, whichenter the T-cell–rich areas via so-called high-endothelial venules (Topalian et al., 2011; Vasaturoet al., 2013) (Supplemental Video). Effective stimula-tion of antigen-specific T cells is a hallmark feature ofmature DCs and requires the delivery of three signals(Kalinski and Okada, 2010; Kirkwood et al., 2012) (Fig.2, inset, and Supplemental Video):

1. Signal 1: Presentation of the antigen in thecontext of MHC molecules to a compatible TCR.Antigenic peptides bound to MHC class IImolecules are presented to CD4+ T cells [which

Fig. 2. The clinical success of DC-based therapies against AML and, by extension, other cancer types relies on three key conditions. Improvements inthe therapeutic efficacy of DC vaccine strategies against AML will require 1) a further enhancement of the immunogenicity of the DC products toensure optimal stimulation of the immune effector cells (such as T and NK cells); 2) augmenting the strength of the antileukemic immune response,e.g., by overcoming Treg-mediated immunosuppression; and 3) increasing the responsiveness of AML cells to immune attack by making these cellsvisible to the immune system and by overcoming their immune escape mechanisms. The figure inset shows a close-up of the immunological synapsebetween DCs and T cells. The outcome of DC–T-cell interaction, i.e., T-cell activation or inhibition, is determined by the balance between variouspositive (+) and negative (2) signals delivered via distinct cell surface receptors and their ligands, cytokines (e.g., IL-10 and IL-12) and enzymes (e.g.,IDO). Targeting immune checkpoints, such as CTLA-4, PD-1 or its ligands, and BTLA, can help to tip the balance in favor of protective anti-tumorimmunity. HVEM, herpes virus entry mediator; TNF(R)SF, TNF (receptor) super family; SOCS-1, suppressor of cytokine signaling-1.

Dendritic Cell–Based Cancer Immunotherapy 735

can differentiate into T helper type 1 (TH1), type2 (TH2), or type 17 (TH17) subsets] (Datta et al.,2014), whereas those bound to MHC class Imolecules are recognized by CD8+ CTLs (Cohnand Delamarre, 2014) (Supplemental Video).

2. Signal 2: Delivery of appropriate costimulatorysignaling. The outcome of this process is de-termined by the balance between the differentactivating (positive) and inhibiting (negative)signals that the T cells receive from the DCs(Vasaturo et al., 2013). These signals are de-livered through interaction of costimulatory andcoinhibitory molecules expressed on the DCsurface with their respective counter-receptorson the T cells (Vasaturo et al., 2013) (Supple-mental Video). As shown in Fig. 2, multiplecostimulatory and coinhibitory receptor/ligandpairs have been identified. Most of these mole-cules are either immunoglobulin superfamilymembers or tumor necrosis factor (TNF) andTNF receptor superfamily members (Murphyet al., 2006). CD80, CD86, and programmeddeath-ligand 1 or 2 (PD-L1/2) are examples ofimmunoglobulin superfamily members expressedon the DC surface, whereas CD28, CTLA-4,programmed death (PD)-1, and B andT lymphocyte attenuator (BTLA) belong to thesame family but are expressed on the T cells(Greaves and Gribben, 2013). CD40, CD70,OX40L, 4-1BBL, and herpes virus entry media-tor, which are expressed on the DC surface, andCD40L, CD27, OX40, and 4-1BB, which areexpressed on the T cells, are all examples ofTNF (receptor) superfamily members (Vinay andKwon, 2009) (Fig. 2, inset).

3. Signal 3: Production of T-cell–stimulatory cyto-kines. The third signal or “polarization” signal,involves the secretion of proinflammatory cyto-kines by DCs, including interferons (IFNs) andinterleukin-12 (IL-12) (Kalinski and Okada,2010; Vasaturo et al., 2013). These cytokinesstimulate the expansion, effector function, andmemory development of CTLs (Curtsinger andMescher, 2010). IL-12, which can be producedafter interaction of CD40 with CD40L, also playsa decisive role in determining the type of CD4+

T-cell immunity (Kalinski and Okada, 2010); itpolarizes the differentiation of naive CD4+ T cellsinto TH1 cells, which in turn provide “help” to theCTLs (Kalinski and Okada, 2010; Wimmerset al., 2014).

Recently, the addition of a fourth signal to the above“three-signal theory” has been proposed. This signalcovers all DC-related factors that enable the activatedCTLs to home to the tumor site (Kalinski and Okada,2010). Once arrived at the tumor site, these CTLs will

look for tumor cells that express their cognate antigenin the form of peptide fragments bound to MHC class Imolecules (Anguille et al., 2013). After interaction ofthe MHC/antigen complex with the TCR, CTLs will betriggered to release cytotoxic effector molecules (suchas perforins and granzymes), resulting in lysis of therecognized tumor cells (Trapani and Smyth, 2002;Anguille et al., 2013).

B. Pharmacological Aspects of Dendritic Cell–BasedCancer Immunotherapy

Over the past few decades, a great deal of researchhas been committed to translating the above immuno-biological principles into clinically applicable pharma-ceutical approaches. From the pharmacological point ofview, DC-based immunotherapy is regarded as a vacci-nation strategy whereby tumor antigen-loaded DCs areadministered to the patient with the intent of elicitingprotective and specific CTL immunity toward thetumor cells (Cohn and Delamarre, 2014). From theregulatory point of view, DC vaccines are classified as“human cell, tissue, or cellular or tissue-based prod-ucts” (U.S. Food and Drug Administration terminology)or as “advanced therapy medicinal products” (EuropeanMedicines Agency terminology), which are a specialgroup of therapeutics including gene therapies,tissue-engineered products, and somatic cell therapies(Maciulaitis et al., 2012). Sipuleucel-T is the first, andso far only, DC-based advanced therapy medicinalproduct that received regulatory approval for thetreatment of metastatic, hormone-refractory prostatecancer (Melero et al., 2014). The manufacturing of thisautologous blood cell product starts with a so-calledleukapheresis procedure to obtain large numbers ofautologous PBMCs. These PBMCs, which containAPCs including, but not restricted to, DCs, are thencultured ex vivo for 36–44 hours in the presence ofPA2024, a recombinant fusion protein consisting of thetumor antigen prostatic acid phosphatase and theimmunostimulatory cytokine granulocyte-macrophagecolony-stimulating factor (GM-CSF). After this simul-taneous antigen loading and activation step, the cellsare washed and suspended for intravenous reinfusioninto the patient (Kantoff et al., 2010). In addition tothis transfusion-like protocol, DCs can also be isolateddirectly from the blood and used for immunotherapeu-tic purposes (Bol et al., 2013; Wimmers et al., 2014).The use of native blood DCs has, however, beenhampered by the relatively low frequency of DCs inperipheral blood (as discussed earlier in this section,DCs represent only 1% of the total PBMC fraction) (Bolet al., 2013; Wimmers et al., 2014). Hence, only fewstudies exist that have attempted the clinical trans-lation of this strategy. One example involves thecollection of large numbers of PBMCs (through leuka-pheresis) and subsequent isolation of blood DCs usinga mAb against CMRF-56, an antigen that is expressed

736 Anguille et al.

on the DC surface after a short overnight in vitroincubation (Lopez et al., 2003; Radford et al., 2005).The CMRF-56 isolation platform can be used to obtaina blood DC-enriched PBMC fraction that containsroughly one-third of DCs including both CD1c+ andCD141+ mDC subsets as well as pDCs (Vari et al.,2008). Directly isolated, highly purified native bloodCD1c+ DCs (Schreibelt et al., 2014) as well as pDCs(Tel et al., 2013a) have been used to vaccinatemelanoma patients. Just recently, a similar strategywas adopted by Prue et al. (2015) using CD1c+ mDCsdirectly isolated by immunomagnetic selection fromleukapheresis products of prostate cancer patients.Despite the overall limited experience, these studiesconfirm the technical feasibility of using naturallycirculating DCs as vaccine vehicles for cancer immu-notherapy (Wimmers et al., 2014).Nevertheless, rather than using primary DCs, most

clinical experience so far has been obtained with DCscultured ex vivo starting from precursor cells that areavailable in much greater quantities than naturallycirculating blood DCs (Anguille et al., 2012c). Celltypes frequently used as starting material for DC

vaccine manufacturing are monocytes (Anguille et al.,2012c) and CD34+ hematopoietic progenitor cellsderived from bone marrow (Lardon et al., 1997),peripheral blood (Mu et al., 2004), or cord blood (Chenet al., 2013). Monocyte-derived DCs (moDCs) are by farthe most commonly used DC type in DC vaccineformulations (Anguille et al., 2014; Chiang et al.,2015). This type of DC vaccine is usually produced byisolating CD14+ monocytes from leukapheresis prod-ucts and by culturing them in vitro for 5–6 days intoimmature DCs with GM-CSF and IL-4 (Tacken et al.,2007) (Fig. 3; Supplemental Video). Because immatureDCs are considered to be poor stimulators of immunity,the next step in DC vaccine manufacturing is to triggertheir activation/maturation by exposing the immatureDCs for another 2 days to a proinflammatory cytokinecocktail composed of TNF-a, IL-1b, IL-6, and prosta-glandin E2 (PGE2), commonly referred to as the“Jonuleit” cytokine cocktail (Jonuleit et al., 1997)(Fig. 3). To complete the DC vaccine production process,DCs must be loaded with disease-relevant tumorantigen(s), a step that can be performed either at the im-mature or at themature stage (Tacken et al., 2007) (Fig. 3).

Fig. 3. The gold-standard protocol for DC vaccine manufacturing. DC-based immunotherapeutics are classically prepared using an in vitro cultureprocedure, in which 1) peripheral blood monocytes are allowed to differentiate into immature DCs in the presence of GM-CSF and IL-4 for 5–6 days andthen 2) exposed for another 2 days to a proinflammatory cytokine cocktail to induce DC maturation. Before administration to the patient, DCs are firstloaded with tumor antigen “cargo”; this antigen-loading step can be performed either at the immature or mature DC stage. Each of these phases in theDC manufacturing process can have a major impact on the immunostimulatory potency of the final DC product. Therefore, much ongoing research isdevoted to modifying and optimizing the culture conditions for DC generation to ensure that DCs with the “best possible” immunostimulatorycapacities are being used for immunotherapy and, ultimately, to maximize the chances of therapeutic success. Examples of such modifications arelisted in the text box insets.

Dendritic Cell–Based Cancer Immunotherapy 737

Although being considered as the “gold-standard”DCs, these so-called IL-4 moDCs might be suboptimalfor inducing TH1-mediated anticancer T-cell immu-nity because they resemble in vivo occurring CD14+

dermal DCs known to stimulate humoral immunity(Palucka et al., 2010; Palucka and Banchereau, 2012).As discussed above, LCs, presumably via a mechanismthat involves IL-15, are far more superior than dermalDCs (Banchereau et al., 2012) and moDCs (Romanoet al., 2012; Hutten et al., 2014) in terms of theircapacity to stimulate antigen-specific CTL immunity.This explains the impetus behind the recent efforts todesign new protocols for the generation of DCs withLC-like features (Hutten et al., 2014). Such LC-type,IL-15–expressing DCs can be generated from CD34+

hematopoietic progenitor cells using specific differen-tiation factors (Romano et al., 2011) or from CD14+

monocytes by replacing IL-4 with IL-15 (Mohamadzadehet al., 2001; Anguille et al., 2009, 2013).As may be clear from the previous section on the

immunobiology of DCs, the immunogenicity of DCvaccines will not only depend on the type of DC used inthe vaccine formulation, but also on the functionalstate of the DCs induced by maturation signals(Kirkwood et al., 2012; Datta et al., 2014). The conventionalJonuleit cytokine cocktail implemented in most DCvaccine production protocols has several importantlimitations, which will be discussed in detail in thenext section. Hence, there is currently a great deal ofresearch into developing alternative DC maturationprocedures (Butterfield, 2013; Anguille et al., 2014).Within this context, TLR agonists have come underintense scrutiny given their ability to endow DCs withpotent TH1-polarizing capacity, which is the type ofimmune response that is most desired in cancerimmunotherapy (Kirkwood et al., 2012). Such TLRstimuli can either be administered concomitantly withthe DC vaccine or can be incorporated into the vaccineitself, for example as ex vivo maturation agents(Anguille et al., 2014). Various clinical-grade TLR-based protocols for maturing DCs have been pub-lished in recent years, but, for the sake of brevity, onlytwo will be discussed here: a-type-1–polarized DCsand TriMix DCs. a-Type-1–polarized DCs, whichderive their name from the fact that they are highlyspecialized at inducing TH1 immune responses(Kirkwood et al., 2012), are generated by exposingimmature DCs to a combination of TNF-a, IL-1b, IFN-g,IFN-a, and the TLR3 agonist poly(I:C) (Okada et al.,2011; Hansen et al., 2013). Okada et al. (2011) recentlyreported on the results of a clinical trial of a-type-1–polarized DC vaccinations with concurrent systemicadministration of a TLR3 agonist in patients withrecurrent malignant glioma. Although single arm andearly phase, the study by Okada et al. (2011) hinted ata possible clinical benefit and provided the impetus forother, currently ongoing clinical trials investigating

the clinical efficacy of this particular DC vaccineformulation (Kirkwood et al., 2012; Anguille et al.,2014). Instead of exogenous pulsing with maturation-inducing cytokines, DCs with an activated/maturephenotype can also be obtained using genetic engi-neering techniques such as mRNA electroporation(Benteyn et al., 2015). Thielemans’ group at the FreeUniversity of Brussels (Brussels, Belgium) has pio-neered a method to generate clinical-grade, matureDCs by electroporating immature DCs ex vivo witha mixture of three mRNA molecules (TriMix) coding fora constitutively active TLR4, CD40L, and CD70,respectively (Benteyn et al., 2015). These so-calledTriMix DCs have been used with promising results tovaccinate melanoma patients (Van Lint et al., 2014).

Another pharmacological aspect that deserves to bementioned here relates to the route of administrationof DC vaccines. To encounter T cells and induce tumorantigen-specific T-cell immunity, DCs must berecruited to the lymph nodes (Verdijk et al., 2008).DCs that are injected intravenously are believed to beineffective because they localize in lungs, liver, spleen,and bone marrow, rather than in the lymph nodes(Fong et al., 2001; Mullins et al., 2003). Nevertheless,in a recently published clinical trial (Wilgenhof et al.,2013), signs of clinical benefit have been obtained afterintravenous TriMix DC vaccination, corroboratingearlier studies that already indicated that DCs caninduce immunity even when administered via theintravenous route (Fong et al., 2001; Bedrosian et al.,2003). Most often, DC vaccines are delivered byintradermal injection nearby superficial lymph nodes(Wimmers et al., 2014). After injection into the skin,DCs will use their chemokine receptors (e.g., C:Cchemokine receptor type-7) to migrate from the skinvia the afferent lymphatics to the draining lymphnodes (Wimmers et al., 2014) (Supplemental Video).Importantly, however, trafficking studies revealed thatmost of the intradermally injected DCs die at theinjection site and that only a minority (less than 5%)effectively reach the lymph nodes (Verdijk et al., 2009).To obtain higher numbers of DCs in the lymph nodes,intranodal vaccination has been tested, but its superi-ority over the intradermal administration route hasnot been unequivocally established (Bedrosian et al.,2003; Lesterhuis et al., 2011). Variable results ofintranodal injection might be related to the complexityof the procedure, so that even when performed withultrasound guidance by a highly experienced radiolo-gist, a high portion of the injections will not or onlypartly be delivered intranodally (de Vries et al., 2005;Verdijk et al., 2009; Lesterhuis et al., 2011).

In addition to the administration route, the optimaldose of DC vaccination is another pharmacologicalvariable that still remains to be fully established despitethe numerous phase I clinical trials that have alreadybeen performed (Figdor et al., 2004; Butterfield, 2013).

738 Anguille et al.

Many of these trials did not show any evidence of dose-limiting toxicity (Draube et al., 2011; Butterfield,2013). Maximum tolerated doses are generally notreached in DC vaccine studies, and the highestachievable dose levels are usually defined by thetotal number of DCs that can be produced from oneleukapheresis (i.e., the maximum feasible dose)(Butterfield, 2013; Tel et al., 2013a). A meta-analysis of29 DC vaccine trials in prostate cancer and renal cellcancer, including a total of 906 patients, revealeda positive association between total DC dose andclinical outcome, indicating that appropriate dosageis critical for therapeutic success (Draube et al., 2011).Unfortunately, this meta-analysis does not permit todraw firm conclusions regarding the minimum effec-tive dose required per vaccination. Despite the generalbelief that high numbers of DCs are required tostimulate a potent antitumor immune response invivo, the above-mentioned study by Tel et al. (2013a)using naturally circulating pDCs revealed that clinicalresponses can be obtained even with relatively smallnumbers of DCs (0.3–3 � 106 DCs per vaccine). Extraevidence that higher DC numbers do not necessarilyresult in higher benefit was provided by Aarntzen et al.(2013), who observed a paradoxical increase in DCsreaching the lymph nodes if the number of intrader-mally injected DCs was reduced. Imaging data from thesame group also revealed that antitumor T-cellresponses can be induced with as few as 5 � 105 DCsin the T-cell areas of the draining lymph nodes (Verdijket al., 2009). In line with this, using a computationalmodel, Celli et al. (2012) estimated that less than 100antigen-bearing DCs are sufficient to initiate a T-cellresponse in the lymph node. Overall, these dataindicate that DC vaccines may be effective even atvery low doses.New routes of DC-based cancer therapy are cur-

rently being explored, including in vivo targeting ofDCs (Caminschi et al., 2012; Datta et al., 2014) and theuse of artificial APCs (Eggermont et al., 2014). Thesetopics have been excellently reviewed by others

(Caminschi et al., 2012; Eggermont et al., 2014; Modyet al., 2015) and will not be covered in this review.

III. Toward the Delivery of Effective DendriticCell–Based Cancer Immunotherapy: The Acute

Myeloid Leukemia Showcase

A growing body of evidence indicates that DCvaccination can be of clinical benefit to cancer patients(Draube et al., 2011; Anguille et al., 2014; Cao et al.,2014; Wang et al., 2014), encouraging the furtherdevelopment of this form of immunotherapy. Neverthe-less, there is a general agreement among cancerresearchers that the true clinical potential of DC-based cancer immunotherapy has not been attained yet(Bot et al., 2013; Datta et al., 2014). Conceptually, thegeneration of antitumor immunity by DCs requiresthree sequential steps (Mellman et al., 2011), asschematically represented in Fig. 2:

1. The DCs used for therapy must be immunosti-mulatory and induce the right type of immuneresponse (Anguille et al., 2014).

2. The antitumor immune effector cells of the host,most notably CTLs and NK cells, must becapable of mounting protective immunity (Lizeeet al., 2013).

3. The tumor cells must be susceptible to immuneattack, which requires overcoming the tumorimmune escape mechanisms (Kirkwood et al.,2012).

Hence, the key to unlocking the full clinical potential ofDC-based cancer immunotherapeutics is to establishhow these three conditions can be optimally fulfilled.In this section, we will provide an overview of theresearch efforts that are currently being undertaken toenable successful completion of each of these threesteps. The focus here lies on the optimization of DC-based immunotherapy for AML, which, for the reasonsspecified in Table 1, is a particularly useful tumor type

TABLE 1Why AML is a useful tumor type to optimize and evaluate the efficacy of DC-based immunotherapeutics

Reasons

• AML is an immunogenic malignancy known to be susceptible to lysis by leukemia antigen-specific CD8+ CTLs as well as NK cells (Schurch et al.,2013). The so-called graft-versus-leukemia effect associated with allogeneic hematopoietic stem cell transplantation (HSCT) elegantly illustratesthe therapeutic role of the immune system in AML (Schurch et al., 2013). AML is therefore believed to be particularly amenable toimmunotherapeutic approaches (Anguille et al., 2012c; Martner et al., 2013; Schurch et al., 2013).

• Conventional chemotherapy followed by allogeneic HSCT is associated with significant toxicity and is therefore not feasible for older patients withAML, who represent the majority of AML patients (Anguille et al., 2012c). Their favorable toxicity profile makes immunotherapy approaches, suchas DC-based immunotherapy, worthy of investigation to fill this treatment void (Anguille et al., 2011a).

• DC therapy has already shown to be potentially useful in AML, especially when applied in a minimal residual disease (MRD) setting(i.e., a situation in which a small amount of tumor cells persists after therapy and that is responsible for tumor recurrence) (Van Tendeloo et al.,2010; Anguille et al., 2012c). This corroborates the notion that immunotherapy is most optimally applied as an adjunct therapy at early stages ofcancer when only few immunosuppressive tumor cells are present (Gulley et al., 2011; Anguille et al., 2014).

• Standard response evaluation criteria used in solid malignancies (e.g., RECIST) do not appear to capture the true clinical benefit ofimmunotherapy approaches (Schlom, 2012; Anguille et al., 2014). In AML, numerous surrogate end points for evaluation of treatment efficacy(e.g., molecular MRD markers) have been developed and validated for use in clinical trials, facilitating translational research (Cilloni et al., 2009;Hourigan and Karp, 2013).

Dendritic Cell–Based Cancer Immunotherapy 739

to evaluate the clinical efficacy of DC-based immuno-therapy. Although AML was chosen to showcase howthe DC immunotherapy field is evolving, many of thestrategies that will be discussed here are broadlyapplicable and are currently under active investigationin other tumor types as well.

A. Using Dendritic Cells with OptimalImmunocompetence (Condition Number 1)

The efficacy of any DC-based therapeutic strategydepends, in the first place, on the immunostimulatoryquality of the DCs used (Anguille et al., 2014) (Fig. 2).As discussed earlier, the DC family is very heteroge-neous, consisting of different DC subsets capable ofeliciting different types of immunity. Hence, selectingthe “right” DC subset for immunotherapy is ofparamount importance (Gilboa, 2007; Anguille et al.,2009; Frankenberger and Schendel, 2012; Kirkwoodet al., 2012; Chiang et al., 2015). Much ongoingresearch is concentrated on optimizing the DC productdesign to ensure that DCs with the best possibleimmunogenic characteristics are being used (Anguilleet al., 2014; Datta et al., 2014; Chiang et al., 2015).There are several aspects of the DC manufacturingprocess that determine the immunogenicity of the finalDC therapeutic product, each of which is beingevaluated for possible improvements (Fig. 3) (Figdoret al., 2004; Gilboa, 2007; Tacken et al., 2007; Anguilleet al., 2009; Tesfatsion, 2014).1. Source of DC Precursor Cell. The first aspect

relates to the choice of precursor cell for ex vivo DCpreparation. As discussed above, most clinical trialsperformed hitherto have relied on monocytes as pre-cursor cells for DC vaccine manufacturing (Supple-mental Video) (Gilboa, 2007; Tacken et al., 2007;Chiang et al., 2015). Monocytes can be isolated fromleukapheresis harvests by various techniques, includ-ing centrifugal elutriation, adherence, or immunomag-netic selection using anti-CD14 mAb-coated magneticbeads (Table 2) (Nicolette et al., 2007). Given the factthat moDCs are by far the most frequently used DCtype in clinical trials, we will only address here thecurrent strategies for optimizing moDC culture proto-cols (Fig. 3). Table 2 provides a compilation of theclinical trials of moDC-based vaccination in AML. Foreach clinical trial listed, details on the logisticalaspects related to moDC vaccine manufacturing anda summary of the key clinical findings are provided(Table 2).2. Culture Time. The gold-standard protocol for

moDC generation encompasses 1 week of culture,including ;5 days for differentiation of monocytes intoimmature DCs and ;2 days for converting theseimmature DCs into mature DCs (Fig. 3; SupplementalVideo) (Caux et al., 1992; Anguille et al., 2009). Anumber of reports have demonstrated the feasibility ofreducing the total time of in vitro moDC culture from 7

to ;3 days without affecting the phenotype or functionof the resultant DCs (Czerniecki et al., 1997, 2001;Dauer et al., 2003; Anguille et al., 2009; Beck et al.,2011; Chiang et al., 2011). Moreover, these so-calledFast DCs have been shown to possess a bettercostimulatory phenotype than their 7-day counterparts(Lichtenegger et al., 2012) and have a lower expressionof the T-cell coinhibitory molecule PD-L1 (Frankenbergerand Schendel, 2012). In addition, the use of Fast DCsoffers the advantage of reducing the time-consumingprocess of ex vivo DC manufacturing (Anguille et al.,2009). As a consequence, Fast DCs are increasinglyfinding their way into clinical trials, a trend that isalso observed in the field of DC therapy for AML(Subklewe et al., 2014) (Table 2).

3. Culture Media. The type of medium used for exvivo DC culture is an often overlooked parameter thatmay influence the functional capacity of the culturedDCs (da Silva Simoneti et al., 2014). All aspects of theDC vaccine manufacturing process, including theculture media and reagents used, must comply to goodmanufacturing practice requirements that are enforcedby health regulatory agencies to ensure the quality andsafety of the DC product (Figdor et al., 2004). Differentgood manufacturing practice-grade DC culture mediahave been marketed, each yielding DCs with differentimmune-stimulatory properties (da Silva Simonetiet al., 2014), indicating that the choice of cell culturemedium determines the potency of the final DC vaccineproduct. Serum, which is often added to the culturemedium, is postulated to have a negative influence onDC vaccine functionality in vitro (da Silva Simonetiet al., 2014; Kolanowski et al., 2014), but the in vivoimpact of using serum remains to be studied.

4. Differentiation Protocol. As discussed earlier, themost commonly used cytokines for inducing moDCdifferentiation are GM-CSF and IL-4 (Gilboa, 2007;Anguille et al., 2009) (Fig. 3). The choice of DCdifferentiation regimen is an important determinantof the immunogenic quality of moDCs (Ueno et al.,2010). This is supported by the observation that theability of moDCs to stimulate antitumor immunity canbe dramatically improved by replacing IL-4 with IL-15(Dubsky et al., 2007; Anguille et al., 2009, 2012a) orIFN-a (Papewalis et al., 2008). Such alternativelydifferentiated DCs can also exert direct cytotoxicitytoward AML cells (Papewalis et al., 2008; Anguilleet al., 2012a), further increasing their attractivenessfor clinical exploitation (Roothans et al., 2013; Telet al., 2014).

5. Maturation Protocol. The second step in moDCmanufacturing involves the induction of DC matura-tion, a step that is usually performed by exogenousadministration of a cytokine cocktail composed of TNF-a, IL-1b, IL-6, and PGE2 (Jonuleit et al., 1997; Gilboa,2007) (Fig. 3; Supplemental Video). The major limita-tion to using this particular mixture is the inability of

740 Anguille et al.

TABLE

2Clinicaltrials

ofmoD

C-bas

edva

ccinationin

AML

Cas

erepo

rtsareex

clude

dfrom

this

overview

.

Ref.

Log

istics

NClinical

Resultsa

Sou

rce/Isolation

Method

Culture

Tim

eCulture

Med

ium

Differentiation

Matur

ation

Reg

imen

Targe

tAntige

nAntige

nSou

rce

Loa

dingMethod

Other

Asp

ects

Adm

in.

Rou

te

Lee

etal.,20

04PBSC

harvest/

CD14

+

isolation

8–9da

ysAIM

VGM-C

SF+IL

-4TNF-a

NA

AML

cell

lysa

teb

Pulsing

/SC/IV

2Disea

sepr

ogression

(2)

Kitaw

akiet

al.,

2011

aLeu

kaph

eresis/

Elutriation

3or

6da

ysCellG

roGM-C

SF+IL

-4OK-432

WT1,

hTERT,

etc.

Apo

ptotic

AMLcellsc

Pulsing

adjuva

nt

OK-432

ID4

Disea

sestab

ilization

(2)

↓AML

blas

tcount(1)

Oga

sawaraan

dOta,20

14;

Ota

and

Oga

sawara,

2014

Leu

kaph

eresis/

Adh

eren

ceND

ND

GM-C

SF+IL

-4OK-432

+PGE2

WT1

Pep

tide

Pulsing

adjuva

nt

OK-432

ID7d

↓WT1mRNA

leve

l(2)

DiPersioet

al.,

2009

;Khou

ryet

al.,20

10

Leu

kaph

eresis/

Adh

eren

ceND

ND

Uns

pecified

cytokine

mix

Uns

pecified

cytokine

mix

hTERT

mRNA

Electropo

ration

+LAMP

ID21

DFSof

79%

[12m

]

Van

Ten

deloo

etal.,20

10;

Bernem

anet

al.,20

14

Leu

kaph

eresis/

CD14

+

isolation

8da

ysCellG

ro+1%

hAB

GM-C

SF+IL

-4TNF-a+IL

-1+IL

-6+PGE2

WT1

mRNA

Electropo

ration

+/2LAMP

ID30

Indu

ctionof

CR

(2/3)

Indu

ctionof

CMR

(8/23)

Con

tinued

CR

[63m

](8)

Disea

sestab

ilization

(3)

Kitaw

akiet

al.,

2011

bLeu

kaph

eresis/

Elutriation

ND

ND

GM-C

SF+IL

-4TNF-a+PGE2

WT1

Pep

tide

Pulsing

+ZOL

ID/IV

3Disea

sestab

ilization

(1)

↓AML

blas

tcount(1)

Rosen

blattet

al.,

2014

Leu

kaph

eresis/

Adh

eren

ce7–

10da

ysND

GM-C

SF+IL

-4TNF-a

MUC1,

WT1,

PRAME,etc.

AML

cells

Fus

ionhy

brids

(PEG)

adjuva

nt

GM-C

SF

SC

13Con

tinued

CR

[23m

](9)

Sch

norfeilet

al.,

2014

;Subk

lewe

etal.,20

14

Leu

kaph

eresis/

Adh

eren

ce3da

ysND

GM-C

SF+I

L-4

R-848

+poly(I:

C)+othe

reWT1,

PRAME

mRNA

Electropo

ration

/ID

2fND

(ongo

ing

stud

y)

CD14

+isolation,mag

netic

isolation

ofCD14

mon

ocytes;C(M

)R,complete(m

olecular)

remission

;DFS,diseas

e-free

survival;hAB,hu

man

AB

seru

m;hT

ERT,hu

man

telomeras

ereve

rsetran

scriptas

e;LAMP,inclusion

oflysosome-as

sociated

mem

bran

epr

oteinin

mRNAconstru

ct;M

UC1,

mucin1;

ND,n

oda

ta;P

BSC,p

eripheral

bloo

dstem

cell;P

EG,p

olye

thylen

eglycol;P

RAME,p

referentially

expr

essedan

tige

nin

melan

oma;

ZOL,incombination

withzoledr

onate.

aThenumbe

rin

parentheses

indicatesthenumbe

rof

patien

tsin

whom

thede

sign

ated

clinical

effect

was

observed

.Thenumbe

rin

bracke

tsindicatesthefollow

up-tim

e(inmon

ths).

bPrepa

redby

repe

ated

free

ze-thaw

cycles.

c Prepa

redby

g-irrad

iation

.dIn

cluding2pa

tien

tswithacutelymph

oblastic

leuke

mia.

e IncludingTNF-a,IL

-1b,IF

N-g,PGE2.

f Sch

edulesto

include

20pa

tien

ts.

Dendritic Cell–Based Cancer Immunotherapy 741

the resultant DCs to produce IL-12p70 (Kalinski et al.,2001). The lack of IL-12 secretion can mainly beattributed to the presence of PGE2 (Kalinski et al.,2001). In addition, PGE2 can induce indoleamine 2,3-dioxygenase (IDO) expression in moDCs (Braun et al.,2005), which promotes the activation of Treg cells(Chung et al., 2009) (Fig. 2, inset) and can preventDCs from attracting naive T cells (Muthuswamy et al.,2010). However, because PGE2 is considered manda-tory for licensing DCs to migrate to the lymph nodes, itcannot simply be omitted from the maturation regimen(Scandella et al., 2002). The incorporation of TLRagonists into the DC maturation cocktail may help tocircumvent this problem. TLR-matured DCs have beenshown to combine migratory capacity with high IL-12production, even with PGE2 in the maturation cocktail(Boullart et al., 2008; Anguille et al., 2009; Beck et al.,2011). In addition, TLR maturation stimuli also enableDCs to overcome Treg-mediated immunosuppression(Pasare and Medzhitov, 2003; Lichtenegger et al.,2012) and to activate innate antitumor effectors, inparticular NK cells (Beck et al., 2011; Lichteneggeret al., 2012). It is therefore not surprising to witness anincreased use of TLR-based maturation regimens inclinical trials of DC-based cancer immunotherapy(Galluzzi et al., 2012). In AML, several DC vaccinestudies have already incorporated TLR agonists[i.e., poly(I:C), OK-432, and R-848] to promote DCmaturation (Table 2).6. Choice of Target Antigen. The primary immuno-

logic goal of DC-based cancer therapy is to immunize orvaccinate the patients by inducing a (memory) tumorantigen-specific T-cell immune response via the in-trinsic antigen-presenting ability of DCs (Gilboa,2007). As a consequence, the choice of antigenic targetprobably represents one of the most critical aspects inDC vaccine design (Gilboa, 2007). Since the discoveryof the first cancer antigen by Boon and colleagues in1991 (van der Bruggen et al., 1991) (Fig. 1), more than100 tumor antigens have been identified and theirnumber is still increasing (Cheever et al., 2009).Selecting the right target antigen has thus becomea challenging task (Cheever et al., 2009; Anguille et al.,2012b). To facilitate and rationalize this antigenselection process, the Translational Research WorkingGroup of the National Cancer Institute has proposeda number of criteria for assessing the suitability ofa given tumor antigen for immunotherapeutic target-ing (Cheever et al., 2009). Based on this report,preference should be given to antigens that: i) areoverexpressed in the tumor cell compartment but not(or only minimally) expressed in normal cells andtissues; ii) are consistently overexpressed amongpatients with a given tumor type and, within eachpatient, expressed in most to all tumor cells; iii) play arole in the malignant phenotype; iv) contain both CD8+

and CD4+ T-cell epitopes and are immunologically

relevant (i.e., demonstration of immunity toward theantigen in cancer patients); and v) hold clinicalrelevance (i.e., demonstration of clinical responses inimmunotherapy studies targeting the given antigen)(Cheever et al., 2009; Anguille et al., 2012b). Oneexample of a tumor-associated antigen that fulfils allthese requirements and that is known to be overex-pressed in AML is the Wilms’ tumor 1 (WT1) protein(Anguille et al., 2012b), which has already been shownto be an excellent choice for DC-based immunotherapyin AML (Van Tendeloo et al., 2010; Van Driesscheet al., 2012). An upcoming trend is to target antigensthat are preferentially expressed in the cancer stemcell compartment, to enable eradication of the malig-nant cell population that is responsible for tumorpropagation and/or recurrence (Ruben et al., 2013;Snauwaert et al., 2013).

7. Source of Antigen(s). The best source of antigenicmaterial remains another unresolved issue in DCproduct design (Gilboa, 2007) (Fig. 3). Tumor antigenscan be derived from either “undefined” or “defined”antigen sources (Nicolette et al., 2007). Undefinedantigen preparations contain both known and un-known epitopes (Nicolette et al., 2007); examples usedin the setting of DC therapy for AML are peptideseluted from the tumor cell surface, whole AML cellpreparations for fusion with DCs, necrotic AML celllysates (e.g., prepared by repeated freeze-thaw cycles),apoptotic AML cells (e.g., prepared by g-irradiation),total tumor RNA extracted from the AML cells, andAML cell-derived extracellular vesicles (van der Polet al., 2012) including exosomes (small vesicles ofendosomal origin released from live cells) and apoptoticblebs (large vesicles released from cells undergoingapoptosis) (van den Ancker et al., 2010; Anguille et al.,2012c; Yao et al., 2014). Sources of defined antigens aresynthetic peptides or nucleic acids encoding a specifictumor antigen (van den Ancker et al., 2010; Anguilleet al., 2012c). Several of these undefined and definedantigen sources have already been used in moDC-based vaccine trials in AML (Table 2).

The main advantage of using undefined antigenpreparations is that they cover the full repertoire ofantigens expressed by the tumor cells (Schui et al.,2002; Nicolette et al., 2007; Tesfatsion, 2014). At thesame time, this is also considered to be their majordisadvantage. Indeed, with an undefined antigenicpayload, there is a high likelihood of deliveringimmunologically irrelevant antigens or even self-antigens, which may lead to T-cell tolerance orautoimmunity (Schottker and Schmidt-Wolf, 2011).For example, some concern has been recently raisedover two tumor antigens, survivin and receptor forhyaluronic acid–mediated motility, because of theirexpression in activated T lymphocytes imposing a riskfor T-cell fratricide (Leisegang et al., 2010; Snauwaertet al., 2012). Furthermore, using tumor cells or tumor

742 Anguille et al.

cell elements as antigen source entails the risk ofdelivering other tumor-derived factors that haveimmunosuppressive effects (e.g., IL-10 and transform-ing growth factor-b) (Schui et al., 2002; Schottker andSchmidt-Wolf, 2011; Szczepanski et al., 2011; Rubenet al., 2013; Hong et al., 2014). Nevertheless, severalstudies have shown that an effective CTL immuneresponse can be triggered by DCs charged with eithernecrotic leukemic cell lysates or apoptotic leukemiccells or blebs (Galea-Lauri et al., 2002; Spisek et al.,2002; Weigel et al., 2006; Ruben et al., 2014). Oneplausible explanation for this observation is that thepotential immunosuppressive effects of tumor cells canbe overcome by the various immunostimulatory factorsreleased from tumor cells upon necrotic or apoptoticcell death induction (Chiang et al., 2010). For example,tumor cells undergoing necrosis have been shown torelease heat-shock proteins (HSPs) and high mobilitygroup box 1 (HMGB1), factors that are known topromote the maturation of DCs via a TLR4-dependentmechanism (Chiang et al., 2010). Apoptotic tumor cellscan also release HSPs and HMGB1 and exposecalreticulin on their cell surface. Calreticulin acts asan “eat me signal,” facilitating phagocytic uptake byDCs (Chiang et al., 2010; Guo et al., 2014; de Bruynet al., 2015). Oxidation by hypochlorous acid (HOCl)treatment is a relatively novel strategy to producehighly immunogenic tumor cell preparations for DCantigen loading. In addition to inducing necrotic celldeath (with the related advantage of eliciting therelease of HSPs, HMGB1, and other DC-activatingsignals), exposure of tumor cells to HOCl gives rise tooxidized biomolecules, which can lead to direct DCactivation and promote DC antigen processing and(cross-) presentation. Moreover, it has been proposedthat HOCl reduces the production of immunosuppres-sive cytokines (e.g., IL-10) by tumor cells (Chiang et al.,2010, 2013, 2015). To the best of our knowledge, HOCl-treated leukemic cells have not yet been used asantigen source in moDC-based immunotherapy proto-cols in AML.8. Antigen-Loading Method. The method used for

delivering the antigenic payload to the DCs is anotheraspect that requires careful consideration, because thishas a major impact on the antigen-presenting qualityof the final DC therapeutic product (Gilboa, 2007)(Fig. 3). So far, most DC-based immune interventionsagainst cancer, including AML, have relied on theuse of peptide pulsing or pulsing with lysed orapoptotic tumor cells for antigen loading (Figdoret al., 2004; van den Ancker et al., 2010) (Table 2).Other antigen-loading methods, such as chemical orelectrical fusion of DCs with tumor cells and the use oftumor-derived extracellular vesicles (exosomes andapoptotic blebs) as antigen-delivery carriers, are in-creasingly finding their way into clinical trials(Galluzzi et al., 2012; Ruben et al., 2014; Gu et al., 2015).

Although full-scale comparisons of these variousapproaches are lacking, there are some data favoringthe use of certain antigen-delivery methods overothers. For example, pulsing of moDCs with apoptoticAML cells has a higher antigen-loading efficiencycompared with pulsing with lysates (Galea-Lauriet al., 2002; van den Ancker et al., 2011). Fusion ofDCs with AML cells also seems to be superior toloading with lysed or apoptotic leukemic cells (Galea-Lauri et al., 2002; Klammer et al., 2005), although onereport found no clear difference in functionalitybetween lysate-pulsed DCs and AML-DC fusionhybrids (Weigel et al., 2006). The use of leukemia-derived exosomes and apoptotic blebs also seems to besuperior to pulsing with lysates (Gu et al., 2015) andapoptotic tumor cells (Ruben et al., 2014), respectively.A relatively new method for antigen loading of DCsinvolves the use of antigen-containing synthetic nano-or microparticles (Zhang et al., 2013). This strategy notonly allows for delivering antigen but also for providingDCs with immunostimulatory adjuvants (Zhang et al.,2013). Zhang et al. (2013) recently showed that poly-meric microparticles containing both WT1 and theTLR9 agonist CpG can be used to generate DCs withpotent capacity to stimulate WT1-specific antileukemicimmunity. DCs can also be genetically modified tosynthesize and express predefined tumor antigen(s),e.g., by using lentiviral vectors encoding a given tumorantigen (Sundarasetty et al., 2013) or by electroporat-ing tumor antigen-encoding mRNA into the cells(Smits et al., 2009). The latter technique has gainedincreasing popularity over the past decade (Gilboa andVieweg, 2004) and has also been adopted in moDC-based vaccine trials in patients with AML (Anguilleet al., 2012c) (Table 2). The attractiveness of the mRNAelectroporation strategy lies in its demonstrated in vivoability to induce a broad (i.e., multiepitope) leukemia-specific immune response without the need for takinginto account the human leukocyte antigen constitutionof the patient; this is in contrast with tumor antigenpeptide pulsing, which is restricted to a particularhuman leukocyte antigen (Van Tendeloo et al., 2010).

Although each loading strategy has its own specificadvantages and disadvantages, perhaps the mostdecisive argument for selecting a particular method iswhether it allows for delivering the antigen in thecontext of both MHC class I and II molecules (Gilboaand Vieweg, 2004). Indeed, the simultaneous pre-sentation of MHC class I and class II epitopes torecruit both CD8+ and CD4+ T cells is considered to bea critical determinant for effective and long-lastingantitumor immunity (Wang, 2002) (SupplementalVideo). When DCs are pulsed with lysates or apoptotictumor cells, the antigens will predominantly enter theexogenous, MHC class II–processing pathway; theseantigen-loading methods rely on cross-presentation toenable class I presentation (Decker et al., 2006).

Dendritic Cell–Based Cancer Immunotherapy 743

Although cross-presentation has been shown to occurin vivo (Kitawaki et al., 2011a), this process may notalways be effective (Decker et al., 2006). One way toovercome this is to combine different antigen-loadingmethods (e.g., pulsing with lysates in conjunction withmRNA loading) to allow the antigen(s) to access boththe MHC class I– and II–processing pathways (Deckeret al., 2009; Decker et al., 2006). Another appealingstrategy is to use tumor antigen-encoding mRNA thatis appended with an MHC class II–targeting sequenceof an endosomal/lysosomal protein, such as DC-lysosome-associated membrane protein (Benteynet al., 2013).9. Designer Dendritic Cells. The advent of electro-

poration technology and other genetic engineeringmethods has now also opened up the possibility tofine-tune the phenotype and function of DCs towardthe desired immunostimulatory direction (Smits et al.,2009). DCs with an enhanced immunostimulatoryprofile can be obtained by genetically enforcing theexpression of costimulatory molecules, e.g., OX40L(Yanagita et al., 2004) or the TH1-polarizing cytokineIL-12 (Curti et al., 2005) (Fig. 2, inset). Short-interfering RNA technology has been used effectivelyto silence genes that have a negative impact on theimmunostimulatory potency of DCs, e.g., genes codingfor coinhibitory molecules, such as PD-L1/2 (Hoboet al., 2010, 2013), genes coding for suppressivecytokines, such as IL-10 (Iversen et al., 2009; Iversenand Sioud, 2015), or genes coding for signalingmolecules regulating cytokine expression in DCs, suchas signal transducer and activator of transcription-3(Brady et al., 2013; Sanseverino et al., 2014) andsuppressor of cytokine signaling-1 (Zhou et al., 2006)(Fig. 2, inset).10. Route of Administration. The classic parenteral

routes of drug administration, e.g., intravenous, areconceptually less appropriate for DC vaccine adminis-tration, because DCs administered through theseroutes are not primarily distributed to their main siteof action, the lymph node (Malik et al., 2014). To enabledelivery to the lymph node, DCs are usually adminis-tered by subcutaneous or intradermal injection nearbyperipheral lymph nodes or by intralymphatic or directintranodal injection (Pyzer et al., 2014). A meta-analysis of clinical trials of DC-based immunotherapyin prostate cancer and renal cell cancer revealed thatthe choice of administration route may indeed in-fluence the therapeutic efficacy, with lymph node-targeting administration routes being superior overthe intravenous route (Draube et al., 2011). Neverthe-less, intravenous DC vaccination has been reported toelicit durable clinical responses in melanoma (e.g.,intravenously injected TriMix DCs) (Wilgenhof et al.,2013) and to prolong overall survival in prostate cancer(e.g., sipuleucel-T) (Kantoff et al., 2010), allowing us toconclude that intravenous administration can be an

effective route to deliver DC vaccines. Focusing onAML, most clinical trials of moDC-based vaccinationhave employed the intradermal administration route;only limited experience has been obtained with sub-cutaneous or intravenous DC vaccine administration inthe context of AML (Table 2). One study combinedintradermal and intravenous administration in anattempt to target DCs to both the lymph nodes andthe bone marrow (Kitawaki et al., 2011b), which is theprimary site in AML where high-avidity antileukemiaT cells reside (Melenhorst et al., 2009). However,because of the small sample size of this study, it isimpossible to conclude whether combined intradermal/intravenous administration is superior over intrader-mal administration alone from a clinical standpoint.

B. Enhancing the Strength of the Antitumor ImmuneEffector Cells (Condition Number 2)

For any DC strategy to be effective, it is imperativethat the immune cells of the host are responsive tostimulation and functionally capable of mediatingtumor rejection (Mellman et al., 2011). However, veryoften the functioning of the immune system isdisturbed in cancer patients (Finn, 2012). This obser-vation also applies to patients with AML, whofrequently have a compromised immune status thatprevents them from mounting protective antileukemicimmune responses (Anguille et al., 2011a; Schurchet al., 2013). For instance, patients with AML oftenspontaneously develop leukemia-specific CTLs(Anguille et al., 2012b), but these are every so oftenanergic (Narita et al., 2001; Beatty et al., 2009) ordysfunctional (Le Dieu et al., 2009; Florcken et al.,2013) and thereby fail to recognize and kill theirtargets. Similarly, at the innate immunity level, NKcells of AML patients are often functionally disabled(Lion et al., 2012b). This impaired immune competencecan dramatically hamper the efficacy of DC therapy inpatients with AML. Therefore, to maximize the chanceof treatment success, future DC studies for AML mustnot only focus on enforcing the intrinsic immunosti-mulatory properties of the DCs used in the therapeuticformulation, but also on harnessing the capacity of thepatients’ immune cells to respond to DC stimulationand to properly perform their cytotoxic effector func-tions against AML cells.

1. Enhancing T-Cell–Mediated Antitumor Immunity.The adaptive immune response against cancer can beharnessed by modulating the signals that govern T-cellstimulation or inhibition at the DC/T-cell immunologicsynapse (Vasaturo et al., 2013) (Fig. 2, inset). Althoughin some studies the focus has been on engagingcostimulatory pathways to enhance T-cell function(Houtenbos et al., 2007), most research is aimed attargeting immune checkpoint pathways, such as theB7/CTLA-4 axis and the PD-1/PD-L axis (Vasaturoet al., 2013; Anguille et al., 2014; Datta et al., 2014).

744 Anguille et al.

a. Cytotoxic T-lymphocyte antigen‐4. Combining DCtherapy with CTLA-4 blockade represents an appeal-ing strategy to potentiate DC vaccine-induced T-cellimmunity (Anguille et al., 2014; Datta et al., 2014).Zhong et al. (2006) showed that the concomitant use ofan mAb against CTLA-4 enhances the in vitro capacityof DCs to induce functional CTL responses againstAML cells. The feasibility of using the anti–CTLA-4mAb ipilimumab in patients with hematologic malig-nancies, including AML patients, was recently con-firmed (Bashey et al., 2009). It is important, however,to mention that ipilimumab treatment is associatedwith a high percentage of immune-related adverseevents (Norde et al., 2012). Furthermore, conflictingdata exist regarding the effect of ipilimumab on Treg

cells with some studies showing inhibition of Treg cells(Norde et al., 2012) and other studies showing Treg cellproliferation (Kavanagh et al., 2008).b. Programmed death-1. A similar approach

revolves around the use of DCs in combination withtherapeutic blockade of the PD-1/PD-L coinhibitorypathway (Norde et al., 2012). Blockade of PD-1 onT cells has been shown to promote the capacity of DCsto stimulate leukemia antigen-specific T cells (Hoboet al., 2012). The observed promotion of antileukemicimmunity after interference with the PD-1/PD-Lsignaling pathway is not only due to enhancedactivation of effector T cells but also in part due toreducing Treg cell numbers and attenuating theirimmunosuppressive activity (Ge et al., 2009; Zhouet al., 2010). A phase II clinical trial investigating thecombined use of DC therapy with the anti–PD-1 mAbpidilizumab (CT-011; CureTech, Yavne, Israel) iscurrently underway in remission patients with AML(Rosenblatt et al., 2011). A clinical trial of PD-L1/2–silenced DC vaccination in combination withdonor lymphocyte infusions for the treatment ofpost-transplant leukemia relapse has also beenregistered (CCMO identifier, NL37318) (Norde et al.,2012).c. B and T lymphocyte attenuator. Although CTLA-4

and PD-1 are by far the best characterized T-cellcoinhibitory molecules, other negative regulators of T-cell function, such as BTLA, have been identified(Norde et al., 2012) (Fig. 2, inset). BTLA, which belongsto the same family of coinhibitory receptors as CTLA-4and PD-1, has been implicated in the functionalinhibition of tumor antigen-specific T cells (Nordeet al., 2012). As demonstrated for PD-1, blockade ofBTLA has been shown to enable in vitro expansion ofleukemia antigen-specific T cells upon DC stimulation(Hobo et al., 2012). In contrast to CTLA-4 and PD-1,mAbs targeting BTLA have not yet entered the clinicaltrial arena (Hobo et al., 2012).d. Immunosuppressive cells. Another attractive

way to empower the antitumor effector response isto inhibit immunosuppressive cell populations, such

as Treg cells and myeloid-derived suppressor cells(MDSCs) (Anguille et al., 2014). Treg cells, which occurwith increased frequency in patients with AML, arewell known to have suppressive effects on DCs as wellas on CTLs and NK cells and thus constitute animportant barrier to effective DC-based immunother-apy (Ustun et al., 2011; Anguille et al., 2014; Chianget al., 2015; Vasaturo et al., 2015). In a murine AMLmodel, Delluc et al. (2009) observed that Treg depletionfacilitates the induction of antileukemic immunity bysubsequent DC immunization, providing a strongrationale for implementing Treg-targeted therapy inDC-based immunotherapy programs (Ustun et al.,2011). Depletion or blockade of Treg cells can beachieved with a variety of pharmacological agents,including but not limited to the anti-CD25 mAbdaclizumab (Zinbryta; Biogen Idec, Cambridge, MA/AbbVie, North Chicago, IL), the IL-2 immunotoxindenileukin diftitox (Ontak; Eisai Medical Research),low-dose cyclophosphamide or IDO inhibitors such as1-methyl-tryptophan (Ustun et al., 2011; Anguilleet al., 2014). Several of these agents are available forclinical use and are currently being tested in AMLpatients (Ustun et al., 2011).

MDSCs, a heterogeneous population of immaturemyeloid cells with immunosuppressive properties,have recently drawn increasing attention as potentialtargets for improving the efficacy of DC-based cancerimmunotherapies (Anguille et al., 2014; Vasaturoet al., 2015). Although the precise role and significanceof these cells in AML remains largely elusive (Teagueand Kline, 2013; De Veirman et al., 2014), the numberof MDSCs appears to be inversely correlated withclinical outcome in DC vaccine-treated AML patients(Ogasawara and Ota, 2014), underscoring their poten-tial importance.

2. Enhancing Natural Killer Cell–Mediated Antitu-mor Immunity. Although much focus is being placedon augmenting T-cell immunity, it is important not tolose sight of the important role of NK cells inantitumor immunity (Lion et al., 2012a,b). As dis-cussed above, the efficacy of DC therapy relies in parton the presence of a competent innate immune system(Lion et al., 2012a; Van Elssen et al., 2014). In thiscontext, NK cell activation after DC administration inAML patients has been shown to correlate withfavorable clinical outcome (Van Tendeloo et al.,2010; Lion et al., 2012a). In view of this, it is likelythat the efficacy of DC-based immunotherapy can befurther improved by harnessing the NK-cell stimula-tory potential of DCs or by combining DCs withtherapies that potentiate NK-cell immunity (Anguilleet al., 2011a, 2013, 2014). DCs can be endowed withthe ability to stimulate NK cell cytotoxicity towardAML cells by using IL-15 instead of IL-4 during moDCdifferentiation and/or by using TLR ligands for in-ducing their maturation (Anguille et al., 2013, 2015;

Dendritic Cell–Based Cancer Immunotherapy 745

Okada et al., 2015). DCs genetically engineered toexpress NK cell–activating cytokines, such as IFN-a(Anguille et al., 2011b), can also be used to harnessthe antileukemic activity of NK cells (Willemen et al.,2015). Another conceptually attractive approach is touse these cytokines as adjuvants during DC therapy(Anguille et al., 2014). IL-15, which recently becameavailable for clinical use (Conlon et al., 2015), may beof particular interest in this regard given its potentNK-cell stimulatory properties (Van den Bergh et al.,2015). In addition, IL-15 can sensitize AML cells forNK cell–mediated killing by upregulating activatingNK cell receptors on the AML cell surface (Szczepanskiet al., 2010). Endogenous IL-15 is induced in AMLpatients after lymphodepleting chemotherapy andleads to in vivo expansion of NK cells (Miller et al.,2005; Bachanova et al., 2014). This postchemotherapysurge in IL-15 provides a strong rationale forscheduling the administration of DCs in AMLpatients after the initial chemotherapy (Barrett andLe Blanc, 2010).

C. Making Tumor Cells Susceptible to Immune Attack(Condition Number 3)

Whether DC-induced antitumor immunity will lead tocancer eradication ultimately depends on the strengthof the immune system to overcome the various immuneescape mechanisms used by the tumor cells (Anguilleet al., 2014; Vasaturo et al., 2015) (Fig. 2). AML cellsalso exploit a myriad of mechanisms to defend them-selves against immune attack (Barrett and Le Blanc,2010; Teague and Kline, 2013). A comprehensive reviewon immune escape in AML has been provided elsewhere(Lion et al., 2012b; Teague and Kline, 2013) and willtherefore not be discussed in detail here. In this section,we will only mention some examples of immune escapemechanisms for which treatment solutions are readilyapplicable and that can be targeted to synergisticallyenhance the efficacy of DC-based cancer immunother-apy in AML.1. Targeting Programmed Death-1. One well rec-

ognized mechanism by which AML cells inhibitantileukemic immune responses is through expres-sion of ligands for PD-1 (Zhang et al., 2009; Zhouet al., 2010; Teague and Kline, 2013). As discussed inthe previous section, PD-L1/2 provides an inhibitorysignal to T cells during DC–T-cell interaction (Fig. 2,inset). Because PD-L1 can also be expressed on AMLblasts and can be used by these cells to inhibit CTLfunction directly (Saudemont and Quesnel, 2004) aswell as indirectly via Treg cells (Zhou et al., 2010),conjoint targeting of the PD-1/PD-L pathway can helpto improve the efficacy of DC therapy on three fronts:1) by favoring effective antigen presentation at theDC–T-cell immunologic synapse, 2) by restoring AML-induced suppression of CTL function, and 3) bycounteracting Treg cells.

2. Targeting Indoleamine 2,3-Dioxygenase.Another example of an immunosuppressive moleculethat can be expressed by both DCs and AML cells isIDO (Curti et al., 2007, 2010; Chamuleau et al., 2008;Sorensen et al., 2009; Teague and Kline, 2013) (Fig. 2).In view of its central role in Treg biology, targeting IDOhas been proposed as an attractive strategy to over-come Treg-mediated immune escape in AML (Ustunet al., 2011; Teague and Kline, 2013; Arpinati andCurti, 2014). A clinical trial using the IDO inhibitor 1-methyl tryptophan in combination with DC therapywas recently initiated for patients with metastaticbreast cancer (ClinicalTrials.gov identifier: NCT01042535).It should, however, be taken into account that the adaptiveimmune system can react against IDO-expressing tumorcells by targeting IDO (Sorensen et al., 2009). Indeed,spontaneous T-cell responses against IDO have beendetected in cancer patients, and these IDO-reactiveCTLs have been shown capable of killing IDO-expressing AML blasts (Sorensen et al., 2009). Froma theoretical point of view, IDO inhibitors could thushave a negative impact on antileukemic immunity.Other Treg-targeting strategies may therefore bepreferred.

3. Targeting CD200. The CD200 surface moleculewas recently recognized as a key mediator of immuneescape in AML (Martner et al., 2013). CD200 contrib-utes to AML-induced immunosuppression througha multifaceted mode of action, which includes activeinhibition of TH1 responses (Coles et al., 2012a),induction of Treg cells (Coles et al., 2012b), andsuppression of NK cell function (Coles et al., 2011). Inan in vitro study of AML, Memarian et al. (2013)showed that abrogation of CD200/CD200R interactionenhances the T-cell-stimulatory capacity of DCs. Theseobservations provide a compelling rationale for com-bining DC vaccination with anti-CD200 agents. Sama-lizumab (Alexion Pharmaceuticals, Cheshire, CT), ananti-CD200 mAb, is currently under investigation ina phase I clinical trial in patients with chronic lympho-cytic leukemia and multiple myeloma (Mahadevanet al., 2010), but, to the best of our knowledge, hasnot yet been studied in the context of AML or incombination with DC therapy.

4. Mobilizing Acute Myeloid Leukemia Cells fromImmune-Privileged Bone Marrow Niches. AML cellsare also capable of hiding from immune detection, atleast in part by residing in immune-privileged bonemarrow niches (Barrett and Le Blanc, 2010; Peled andTavor, 2013). One conceptually attractive strategy toincrease their visibility to the immune system is tomobilize AML cells out of these protective bone marrowniches (Zeng et al., 2009; Uy et al., 2012). Antagonistsfor C-X-C chemokine receptor type-4 (CXCR-4) can beused for this purpose as they disrupt the interactionbetween CXCR-4 expressed on AML cells with stromalcell–derived factor-1a expressed on bone marrow

746 Anguille et al.

stromal cells (Zeng et al., 2009; Uy et al., 2012; Zhanget al., 2012; Peled and Tavor, 2013). A recent phase Iand II clinical study in AML patients has confirmedthat plerixafor (Mozobil; Genzyme/Sanofi, Cambridge,MA), a CXCR-4 antagonist used for hematopoietic stemcell mobilization, can disengage AML cells from thebone marrow and increase their sensitivity to chemo-therapy (Uy et al., 2012). Histone deacetylase inhib-itors, which are increasingly being used in thetreatment of AML, may have a similar effect byrepressing CXCR-4 expression in AML cells (Gulet al., 2010; Mandawat et al., 2010).5. Undoing Tumor Cells’ Cloak of Invisibility.

In addition to residing in protective bone marrowniches, AML cells are notorious for creating an“invisibility cloak” that prevents them from beingrecognized by T cells and NK cells (Teague and Kline,2013). Elements of this “invisibility cloak” includedownregulation of leukemia antigen presentation, re-duced expression of MHC and T-cell costimulatorymolecules, or shedding of surface molecules involved inimmune recognition and activation (e.g., NKG2Dligands) (Barrett and Le Blanc, 2010; van Luijn et al.,2010; Lion et al., 2012b; Teague and Kline, 2013).These immune evasion mechanisms can clearly affectthe efficacy of active specific immunotherapyapproaches such as DC therapy. Therefore, strategiesfor potentiating immune recognition and destructionof AML cells may be exploited in combination with

DC-based immunotherapy to increase its therapeuticactivity (Anguille et al., 2014). A number of pharma-cological agents can be used for this purpose, includingcytokines, TLR ligands, hypomethylating agents (e.g.,azacitidine and decitabine), histone deacetylase in-hibitors, and kinase inhibitors (for references, seeTable 3).

6. Reducing Tumor Burden. Finally, AML cellsmay not only evade the immune system by hidingfrom immune recognition but also by being present insuch large numbers that the immune system is simplyoutweighed (Emens and Jaffee, 2005; Drake, 2010;Schlom, 2012). This provides a compelling rationale tofirst reduce the tumor burden with conventionalchemotherapy before applying immunotherapy strate-gies, such as DC therapy. Support for this concept hasbeen obtained in different tumor models (Choudhuryet al., 2006; Drake, 2010; Schlom, 2012), includingAML, where the administration of DCs after standardremission induction chemotherapy (i.e., in the settingof minimal residual disease) has shown encouragingresults (Van Tendeloo et al., 2010). Although chemo-therapy is traditionally being considered to be immu-nosuppressive (e.g., by tilting the balance betweenantitumor immune effector cells and Treg cells in favorof the latter) (Lichtenegger et al., 2014), manychemotherapeutic agents used for AML appear to haveparadoxical immune-potentiating effects (Obeid et al.,2007; Liseth et al., 2010; Wemeau et al., 2010; van de

TABLE 3Examples of nonchemotherapeutic antileukemic agents capable of reinstating the immunogenicity of AML cells

Drug Effects on AML Cell Immunogenicity Effects on DCs (and Other Antitumor Immune Effectors)

Cytokines (e.g., interferon-a) • Immunogenic cell death (Rizza et al., 2015) • ↑ DC activation and function (Anguille et al.,2011b)

• ↑ MHC and/or costimulatory molecule expression(Anguille et al., 2011b)

• ↑ direct antileukemic cytotoxicity of DCs (Telet al., 2014)

• ↑ AML antigen expression expression (Anguilleet al., 2011b)

• ↑ activity of T cells and NK cells (Anguille et al.,2011b)

TLR ligands [e.g., poly(I:C), R-848] • Immunogenic cell death (Smits et al., 2007; Lionet al., 2011)

• ↑ DC activation and function (Smits et al., 2007;Lion et al., 2011)

• ↑ MHC and/or costimulatory molecule expression(Smits et al., 2007)

• ↑ activity of T cells and NK cells (Smits et al.,2010)

Hypomethylating drugs (e.g.,azacitidine, decitabine)

• Immunogenic cell death (Tang et al., 2013) • ↑ DC function (Frikeche et al., 2011)• Derepression of AML antigen expression (e.g.,CTAs, WT1) (Almstedt et al., 2010; Goodyearet al., 2010; Atanackovic et al., 2011; Kwon et al.,2012; Yao et al., 2013)

• ↑ Treg cells (conflicting data) (Goodyear et al.,2012; Gang et al., 2014)

• ↑ MHC and/or costimulatory molecule expression(Wang et al., 2013)

• ↓ activity of NK cells (Schonefeldt et al., 2013;Gang et al., 2014)

• ↑ NKG2D ligand expression (Rohner et al., 2007)Histone deacetylase inhibitors • Immunogenic cell death (Fredly et al., 2011) • ↓ DC function (Falkenberg and Johnstone, 2014)

• ↑ differentiation of AML cells toward a DCphenotype (Moldenhauer et al., 2004)

• ↑ IDO expression in DCs (Reddy et al., 2008)

• Derepression of AML antigen expression (e.g.,CTAs) (Goodyear et al., 2010; Yao et al., 2013)

• ↑ MHC and/or costimulatory molecule expression(Maeda et al., 2000; Yao et al., 2013)

• ↑ NKG2D ligand expression (Rohner et al., 2007;Diermayr et al., 2008; Poggi et al., 2009)

Protein kinase C inhibitor (bryostatin) • ↑ differentiation of AML cells toward a DCphenotype (Roddie et al., 2002)

• ↑ DC activation and function (Do et al., 2004;Ariza et al., 2011)

• ↑ NKG2D ligand expression (Rohner et al., 2007)

CTAs, cancer-testis antigens.

Dendritic Cell–Based Cancer Immunotherapy 747

Ven et al., 2012). For example, cytarabine andanthracyclines (the two standard chemotherapeuticagents in AML) cause AML cells to undergo immuno-genic apoptosis (Wemeau et al., 2010; Fredly et al.,2011), thereby favoring the development of antileuke-mia immunity (Barrett and Le Blanc, 2010; Rezvaniet al., 2012). These observations provide strong supportfor the concept of chemoimmunotherapy in AML. DC-based vaccination strategies are also being increas-ingly applied in the context of allogeneic hematopoieticstem-cell transplantation for AML to boost the graft-versus-leukemia reaction. The reader is referred toPlantinga et al. (2014) for an update on DC-basedimmunotherapy in the postallogeneic hematopoieticstem-cell transplantation setting.

IV. Conclusions

Because they lie at the heart of the immune system’sability to recognize and eliminate cancer cells, DCshave attracted a great deal of interest for antitumorimmunotherapy in recent decades (Palucka andBanchereau, 2012; Anguille et al., 2014). Current effortsin the field of DC-based cancer immunotherapy aregeared toward identifying the key factors responsiblefor therapeutic success/failure to unleash the fullclinical potential of DCs for cancer immunotherapy(Kirkwood et al., 2012; Anguille et al., 2014; Chianget al., 2015). AML can be a good study model for thispurpose, in particular because it allows us to examinethe efficiency of this form of immunotherapy in a lowtumor burden context (i.e., in the setting of minimalresidual disease), which is probably the most appro-priate clinical situation to apply immunotherapy(Gulley et al., 2011). In recent years, many effortshave been made to refine and optimize the protocols forDC manufacturing (condition no. 1), but, at the sametime, we should be aware that therapeutic success islikely not to result from a single intervention. Thefuture of DC therapy lies in combining this form ofactive specific immunotherapy with other therapies toincrease the antitumor activity of the immune effectorcells (condition no. 2) and to overcome the myriad ofimmune escape mechanisms used by cancer cells(condition no. 3) (Anguille et al., 2014; Chiang et al.,2015). The past decade has witnessed the advent ofa new generation of DC vaccines with improved potency.This, together with the ever-expanding portfolio ofimmunomodulatory and immune-potentiating therapiesavailable for combinatorial use, may allow us toconclude that we now stand on the brink of realizingthe true potential of DCs for cancer immunotherapy.

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Anguille,Smits, Bryant, Van Acker, Goossens, Lion, Fromm, Van Tendeloo,Berneman.

ReferencesAarntzen EHJG, Srinivas M, Bonetto F, Cruz LJ, Verdijk P, Schreibelt G, van deRakt M, Lesterhuis WJ, van Riel M, and Punt CJA, et al. (2013) Targeting of111In-labeled dendritic cell human vaccines improved by reducing number of cells.Clin Cancer Res 19:1525–1533.

Almstedt M, Blagitko-Dorfs N, Duque-Afonso J, Karbach J, Pfeifer D, Jäger E,and Lübbert M (2010) The DNA demethylating agent 5-aza-29-deoxycytidineinduces expression of NY-ESO-1 and other cancer/testis antigens in myeloid leu-kemia cells. Leuk Res 34:899–905.

Ammi R, De Waele J, Willemen Y, Van Brussel I, Schrijvers DM, Lion E, and SmitsEL (2015) Poly(I:C) as cancer vaccine adjuvant: knocking on the door of medicalbreakthroughs. Pharmacol Ther 146:120–131.

Anguille S, Lion E, Smits E, Berneman ZN, and van Tendeloo VF (2011a) Dendriticcell vaccine therapy for acute myeloid leukemia: questions and answers. HumVaccin 7:579–584.

Anguille S, Lion E, Tel J, de Vries IJ, Couderé K, Fromm PD, Van Tendeloo VF,Smits EL, and Berneman ZN (2012a) Interleukin-15-induced CD56(+) myeloiddendritic cells combine potent tumor antigen presentation with direct tumoricidalpotential. PLoS ONE 7:e51851.

Anguille S, Lion E, Van den Bergh J, Van Acker HH, Willemen Y, Smits EL, VanTendeloo VF, and Berneman ZN (2013) Interleukin-15 dendritic cells as vaccinecandidates for cancer immunotherapy. Hum Vaccin Immunother 9:1956–1961.

Anguille S, Lion E, Willemen Y, Van Tendeloo VF, Berneman ZN, and Smits EL(2011b) Interferon-a in acute myeloid leukemia: an old drug revisited. Leukemia25:739–748.

Anguille S, Smits EL, Cools N, Goossens H, Berneman ZN, and Van Tendeloo VF(2009) Short-term cultured, interleukin-15 differentiated dendritic cells have po-tent immunostimulatory properties. J Transl Med 7:109.

Anguille S, Smits EL, Lion E, van Tendeloo VF, and Berneman ZN (2014) Clinicaluse of dendritic cells for cancer therapy. Lancet Oncol 15:e257–e267.

Anguille S, Van Acker HH, Van den Bergh J, Willemen Y, Goossens H, Van TendelooVF, Smits EL, Berneman ZN, and Lion E (2015) Interleukin-15 dendritic cellsharness NK cell cytotoxic effector function in a contact- and IL-15-dependentManner. PLoS ONE 10:e0123340.

Anguille S, Van Tendeloo VF, and Berneman ZN (2012b) Leukemia-associatedantigens and their relevance to the immunotherapy of acute myeloid leukemia.Leukemia 26:2186–2196.

Anguille S, Willemen Y, Lion E, Smits EL, and Berneman ZN (2012c) Dendritic cellvaccination in acute myeloid leukemia. Cytotherapy 14:647–656.

Ariza ME, Ramakrishnan R, Singh NP, Chauhan A, Nagarkatti PS, and NagarkattiM (2011) Bryostatin-1, a naturally occurring antineoplastic agent, acts as a Toll-like receptor 4 (TLR-4) ligand and induces unique cytokines and chemokines indendritic cells. J Biol Chem 286:24–34.

Arpinati M and Curti A (2014) Immunotherapy in acute myeloid leukemia. Immu-notherapy 6:95–106.

Atanackovic D, Luetkens T, Kloth B, Fuchs G, Cao Y, Hildebrandt Y, Meyer S,Bartels K, Reinhard H, and Lajmi N, et al. (2011) Cancer-testis antigen expressionand its epigenetic modulation in acute myeloid leukemia. Am J Hematol 86:918–922.

Bachanova V, Cooley S, Defor TE, Verneris MR, Zhang B, McKenna DH, CurtsingerJ, Panoskaltsis-Mortari A, Lewis D, and Hippen K, et al. (2014) Clearance of acutemyeloid leukemia by haploidentical natural killer cells is improved using IL-2diphtheria toxin fusion protein. Blood 123:3855–3863.

Baldwin RW (1955) Immunity to methylcholanthrene-induced tumours in inbred ratsfollowing atrophy and regression of the implanted tumours. Br J Cancer 9:652–657.

Banchereau J, Thompson-Snipes L, Zurawski S, Blanck JP, Cao Y, Clayton S, GorvelJP, Zurawski G, and Klechevsky E (2012) The differential production of cytokinesby human Langerhans cells and dermal CD14(+) DCs controls CTL priming. Blood119:5742–5749.

Barrett AJ and Le Blanc K (2010) Immunotherapy prospects for acute myeloid leu-kaemia. Clin Exp Immunol 161:223–232.

Bashey A, Medina B, Corringham S, Pasek M, Carrier E, Vrooman L, Lowy I,Solomon SR, Morris LE, and Holland HK, et al. (2009) CTLA4 blockade withipilimumab to treat relapse of malignancy after allogeneic hematopoietic celltransplantation. Blood 113:1581–1588.

Beatty GL, Smith JS, Reshef R, Patel KP, Colligon TA, Vance BA, Frey NV, JohnsonFB, Porter DL, and Vonderheide RH (2009) Functional unresponsiveness andreplicative senescence of myeloid leukemia antigen-specific CD8+ T cells after al-logeneic stem cell transplantation. Clin Cancer Res 15:4944–4953.

Beck B, Dörfel D, Lichtenegger FS, Geiger C, Lindner L, Merk M, Schendel DJ,and Subklewe M (2011) Effects of TLR agonists on maturation and function of3-day dendritic cells from AML patients in complete remission. J Transl Med9:151.

Bedrosian I, Mick R, Xu S, Nisenbaum H, Faries M, Zhang P, Cohen PA, Koski G,and Czerniecki BJ (2003) Intranodal administration of peptide-pulsed maturedendritic cell vaccines results in superior CD8+ T-cell function in melanomapatients. J Clin Oncol 21:3826–3835.

Benteyn D, Anguille S, Van Lint S, Heirman C, Van Nuffel AM, Corthals J,Ochsenreither S, Waelput W, Van Beneden K, and Breckpot K, et al. (2013) Designof an optimized Wilms’ tumor 1 (WT1) mRNA construct for enhanced WT1 expressionand improved immunogenicity in vitro and in vivo. Mol Ther Nucleic Acids 2:e134.

Benteyn D, Heirman C, Bonehill A, Thielemans K, and Breckpot K (2015) mRNA-based dendritic cell vaccines. Expert Rev Vaccines 14:161–176.

Berneman ZN, Van de Velde AL, Willemen Y, Anguille S, Saevels K, Germonpré P,Huizing MT, Peeters M, Snoeckx A, and Parizel P, et al. (2014) Vaccination withWT1 mRNA-electroporated dendritic cells: report of clinical outcome in 66 cancerpatients (Abstract). Blood 124:310.

Bol KF, Tel J, de Vries IJ, and Figdor CG (2013) Naturally circulating dendritic cellsto vaccinate cancer patients. OncoImmunology 2:e23431.

748 Anguille et al.

Bot A, Marincola F, and Smith KA (2013) Repositioning therapeutic cancer vaccinesin the dawning era of potent immune interventions. Expert Rev Vaccines 12:1219–1234.

Boullart AC, Aarntzen EH, Verdijk P, Jacobs JF, Schuurhuis DH, Benitez-Ribas D,Schreibelt G, van de Rakt MW, Scharenborg NM, and de Boer A, et al. (2008)Maturation of monocyte-derived dendritic cells with Toll-like receptor 3 and 7/8ligands combined with prostaglandin E2 results in high interleukin-12 productionand cell migration. Cancer Immunol Immunother 57:1589–1597.

Brady MT, Miller A, Sait SN, Ford LA, Minderman H, Wang ES, Lee KP, BaumannH, and Wetzler M (2013) Down-regulation of signal transducer and activator oftranscription 3 improves human acute myeloid leukemia-derived dendritic cellfunction. Leuk Res 37:822–828.

Braun D, Longman RS, and Albert ML (2005) A two-step induction of indoleamine2,3 dioxygenase (IDO) activity during dendritic-cell maturation. Blood 106:2375–2381.

Burnet FM (1967) Immunological aspects of malignant disease. Lancet 1:1171–1174.Butterfield LH (2013) Dendritic cells in cancer immunotherapy clinical trials: are wemaking progress? Front Immunol 4:454.

Caminschi I, Maraskovsky E, and Heath WR (2012) Targeting dendritic cells in vivofor cancer therapy. Front Immunol 3:13.

Cao JX, Zhang XY, Liu JL, Li D, Li JL, Liu YS, Wang M, Xu BL, Wang HB, and WangZX (2014) Clinical efficacy of tumor antigen-pulsed DC treatment for high-gradeglioma patients: evidence from a meta-analysis. PLoS ONE 9:e107173.

Caux C, Dezutter-Dambuyant C, Schmitt D, and Banchereau J (1992) GM-CSF andTNF-alpha cooperate in the generation of dendritic Langerhans cells. Nature 360:258–261.

Celli S, Day M, Müller AJ, Molina-Paris C, Lythe G, and Bousso P (2012) How manydendritic cells are required to initiate a T-cell response? Blood 120:3945–3948.

Chamuleau ME, van de Loosdrecht AA, Hess CJ, Janssen JJ, Zevenbergen A, DelwelR, Valk PJ, Löwenberg B, and Ossenkoppele GJ (2008) High INDO (indoleamine2,3-dioxygenase) mRNA level in blasts of acute myeloid leukemic patients predictspoor clinical outcome. Haematologica 93:1894–1898.

Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, Mellman I,Prindiville SA, Viner JL, and Weiner LM, et al. (2009) The prioritization of cancerantigens: a national cancer institute pilot project for the acceleration of trans-lational research. Clin Cancer Res 15:5323–5337.

Chen H, Jin Y, Chen T, Zhang M, Ma W, Xiong X, and Tao X (2013) The antitumoreffect of human cord blood-derived dendritic cells modified by the livin a gene inlung cancer cell lines. Oncol Rep 29:619–627.

Chiang CL, Balint K, Coukos G, and Kandalaft LE (2015) Potential approaches formore successful dendritic cell-based immunotherapy. Expert Opin Biol Ther 15:569–582.

Chiang CL, Benencia F, and Coukos G (2010) Whole tumor antigen vaccines. SeminImmunol 22:132–143.

Chiang CL, Hagemann AR, Leskowitz R, Mick R, Garrabrant T, Czerniecki BJ,Kandalaft LE, Powell DJ Jr, and Coukos G (2011) Day-4 myeloid dendritic cellspulsed with whole tumor lysate are highly immunogenic and elicit potent anti-tumor responses. PLoS ONE 6:e28732.

Chiang CL, Kandalaft LE, Tanyi J, Hagemann AR, Motz GT, Svoronos N, MontoneK, Mantia-Smaldone GM, Smith L, and Nisenbaum HL, et al. (2013) A dendriticcell vaccine pulsed with autologous hypochlorous acid-oxidized ovarian cancer ly-sate primes effective broad antitumor immunity: from bench to bedside. ClinCancer Res 19:4801–4815.

Choudhury A, Mosolits S, Kokhaei P, Hansson L, Palma M, and Mellstedt H (2006)Clinical results of vaccine therapy for cancer: learning from history for improvingthe future. Adv Cancer Res 95:147–202.

Chung DJ, Rossi M, Romano E, Ghith J, Yuan J, Munn DH, and Young JW (2009)Indoleamine 2,3-dioxygenase-expressing mature human monocyte-derived den-dritic cells expand potent autologous regulatory T cells. Blood 114:555–563.

Cilloni D, Renneville A, Hermitte F, Hills RK, Daly S, Jovanovic JV, Gottardi E, FavaM, Schnittger S, and Weiss T, et al. (2009) Real-time quantitative polymerasechain reaction detection of minimal residual disease by standardized WT1 assay toenhance risk stratification in acute myeloid leukemia: a European LeukemiaNetstudy. J Clin Oncol 27:5195–5201.

Cohn L, Chatterjee B, Esselborn F, Smed-Sörensen A, Nakamura N, Chalouni C, LeeBC, Vandlen R, Keler T, and Lauer P, et al. (2013) Antigen delivery to earlyendosomes eliminates the superiority of human blood BDCA3+ dendritic cells atcross presentation. J Exp Med 210:1049–1063.

Cohn L and Delamarre L (2014) Dendritic cell-targeted vaccines. Front Immunol 5:255.

Coles SJ, Hills RK, Wang EC, Burnett AK, Man S, Darley RL, and Tonks A (2012a)Expression of CD200 on AML blasts directly suppresses memory T-cell function.Leukemia 26:2148–2151.

Coles SJ, Hills RK, Wang EC, Burnett AK, Man S, Darley RL, and Tonks A (2012b)Increased CD200 expression in acute myeloid leukemia is linked with an increasedfrequency of FoxP3+ regulatory T cells. Leukemia 26:2146–2148.

Coles SJ, Wang EC, Man S, Hills RK, Burnett AK, Tonks A, and Darley RL (2011)CD200 expression suppresses natural killer cell function and directly inhibits pa-tient anti-tumor response in acute myeloid leukemia. Leukemia 25:792–799.

Coley WB (1891) Contribution to the knowledge of sarcoma. Ann Surg 14:199–220.Collin M, McGovern N, and Haniffa M (2013) Human dendritic cell subsets. Immu-nology 140:22–30.

Conlon KC, Lugli E, Welles HC, Rosenberg SA, Fojo AT, Morris JC, Fleisher TA,Dubois SP, Perera LP, and Stewart DM, et al. (2015) Redistribution, hyper-proliferation, activation of natural killer cells and CD8 T cells, and cytokine pro-duction during first-in-human clinical trial of recombinant human interleukin-15in patients with cancer. J Clin Oncol 33:74–82.

Coulie PG, Van den Eynde BJ, van der Bruggen P, and Boon T (2014) Tumourantigens recognized by T lymphocytes: at the core of cancer immunotherapy. NatRev Cancer 14:135–146.

Couzin-Frankel J (2013) Breakthrough of the year 2013. Cancer immunotherapy.Science 342:1432–1433.

Curti A, Pandolfi S, Aluigi M, Isidori A, Alessandrini I, Chiodoni C, Testoni N,Colombo MP, Baccarani M, and Lemoli RM (2005) Interleukin-12 production byleukemia-derived dendritic cells counteracts the inhibitory effect of leukemic mi-croenvironment on T cells. Exp Hematol 33:1521–1530.

Curti A, Pandolfi S, Valzasina B, Aluigi M, Isidori A, Ferri E, Salvestrini V, BonannoG, Rutella S, and Durelli I, et al. (2007) Modulation of tryptophan catabolism byhuman leukemic cells results in the conversion of CD25- into CD25+ T regulatorycells. Blood 109:2871–2877.

Curti A, Trabanelli S, Onofri C, Aluigi M, Salvestrini V, Ocadlikova D, Evangelisti C,Rutella S, De Cristofaro R, and Ottaviani E, et al. (2010) Indoleamine 2,3-dioxygenase-expressing leukemic dendritic cells impair a leukemia-specific im-mune response by inducing potent T regulatory cells. Haematologica 95:2022–2030.

Curtsinger JM and Mescher MF (2010) Inflammatory cytokines as a third signal forT cell activation. Curr Opin Immunol 22:333–340.

Czerniecki BJ, Carter C, Rivoltini L, Koski GK, Kim HI, Weng DE, Roros JG, HijaziYM, Xu S, and Rosenberg SA, et al. (1997) Calcium ionophore-treated peripheralblood monocytes and dendritic cells rapidly display characteristics of activateddendritic cells. J Immunol 159:3823–3837.

Czerniecki BJ, Cohen PA, Faries M, Xu S, Roros JG, and Bedrosian I (2001) Diversefunctional activity of CD83+ monocyte-derived dendritic cells and the implicationsfor cancer vaccines. Crit Rev Immunol 21:157–178.

da Silva Simoneti G, Saad ST, and Gilli SC (2014) An efficient protocol for thegeneration of monocyte derived dendritic cells using serum-free media for clinicalapplications in post remission AML patients. Ann Clin Lab Sci 44:180–188.

Datta J, Terhune JH, Lowenfeld L, Cintolo JA, Xu S, Roses RE, and Czerniecki BJ(2014) Optimizing dendritic cell-based approaches for cancer immunotherapy. YaleJ Biol Med 87:491–518.

Dauer M, Obermaier B, Herten J, Haerle C, Pohl K, Rothenfusser S, Schnurr M,Endres S, and Eigler A (2003) Mature dendritic cells derived from human mono-cytes within 48 hours: a novel strategy for dendritic cell differentiation from bloodprecursors. J Immunol 170:4069–4076.

de Bruyn M, Wiersma VR, Helfrich W, Eggleton P, and Bremer E (2015) The ever-expanding immunomodulatory role of calreticulin in cancer immunity. Front Oncol5:35.

De Veirman K, Van Valckenborgh E, Lahmar Q, Geeraerts X, De Bruyne E, Menu E,Van Riet I, Vanderkerken K, and Van Ginderachter JA (2014) Myeloid-derivedsuppressor cells as therapeutic target in hematological malignancies. Front Oncol4:349.

De Vries IJ, Krooshoop DJ, Scharenborg NM, Lesterhuis WJ, Diepstra JH, VanMuijen GN, Strijk SP, Ruers TJ, Boerman OC, and Oyen WJ, et al. (2003) Effectivemigration of antigen-pulsed dendritic cells to lymph nodes in melanoma patients isdetermined by their maturation state. Cancer Res 63:12–17.

de Vries IJ, Lesterhuis WJ, Barentsz JO, Verdijk P, van Krieken JH, Boerman OC,Oyen WJ, Bonenkamp JJ, Boezeman JB, and Adema GJ, et al. (2005) Magneticresonance tracking of dendritic cells in melanoma patients for monitoring of cel-lular therapy. Nat Biotechnol 23:1407–1413.

Decker WK, Xing D, Li S, Robinson SN, Yang H, Steiner D, Komanduri KV,and Shpall EJ (2009) Th-1 polarization is regulated by dendritic-cell comparison ofMHC class I and class II antigens. Blood 113:4213–4223.

Decker WK, Xing D, Li S, Robinson SN, Yang H, Yao X, Segall H, McMannis JD,Komanduri KV, and Champlin RE, et al. (2006) Double loading of dendritic cellMHC class I and MHC class II with an AML antigen repertoire enhances correlatesof T-cell immunity in vitro via amplification of T-cell help. Vaccine 24:3203–3216.

Delluc S, Hachem P, Rusakiewicz S, Gaston A, Marchiol-Fournigault C, Tourneur L,Babchia N, Fradelizi D, Regnault A, and Sang KH, et al. (2009) Dramatic efficacyimprovement of a DC-based vaccine against AML by CD25 T cell depletionallowing the induction of a long-lasting T cell response. Cancer ImmunolImmunother 58:1669–1677.

Diermayr S, Himmelreich H, Durovic B, Mathys-Schneeberger A, Siegler U,Langenkamp U, Hofsteenge J, Gratwohl A, Tichelli A, and Paluszewska M, et al.(2008) NKG2D ligand expression in AML increases in response to HDAC inhibitorvalproic acid and contributes to allorecognition by NK-cell lines with single KIR-HLAclass I specificities. Blood 111:1428–1436.

DiPersio JF, Collins RH Jr, Blum W, Devetten MP, Stiff P, Elias L, Reddy A, SmithJA, and Khoury HJ (2009) Immune responses in AML patients following vacci-nation with GRNVAC1, autologous RNA transfected dendritic cells expressingtelomerase catalytic subunit hTERT (Abstract). Blood 114:262.

Do Y, Hegde VL, Nagarkatti PS, and Nagarkatti M (2004) Bryostatin-1 enhances thematuration and antigen-presenting ability of murine and human dendritic cells.Cancer Res 64:6756–6765.

Drake CG (2010) Prostate cancer as a model for tumour immunotherapy. Nat RevImmunol 10:580–593.

Draube A, Klein-González N, Mattheus S, Brillant C, Hellmich M, Engert A, and vonBergwelt-Baildon M (2011) Dendritic cell based tumor vaccination in prostate andrenal cell cancer: a systematic review and meta-analysis. PLoS ONE 6:e18801.

Dubsky P, Saito H, Leogier M, Dantin C, Connolly JE, Banchereau J, and PaluckaAK (2007) IL-15-induced human DC efficiently prime melanoma-specific naiveCD8+ T cells to differentiate into CTL. Eur J Immunol 37:1678–1690.

Eggermont LJ, Paulis LE, Tel J, and Figdor CG (2014) Towards efficient cancerimmunotherapy: advances in developing artificial antigen-presenting cells. TrendsBiotechnol 32:456–465.

Ehrlich P (1909) Über den jetzigen Stand der Karzinomforschung. Ned TijdschrGeneeskd 5:273–290.

Emens LA and Jaffee EM (2005) Leveraging the activity of tumor vaccines withcytotoxic chemotherapy. Cancer Res 65:8059–8064.

Falkenberg KJ and Johnstone RW (2014) Histone deacetylases and their inhibitors incancer, neurological diseases and immune disorders.Nat Rev Drug Discov 13:673–691.

Dendritic Cell–Based Cancer Immunotherapy 749

Figdor CG, de Vries IJ, Lesterhuis WJ, and Melief CJ (2004) Dendritic cell immu-notherapy: mapping the way. Nat Med 10:475–480.

Finn OJ (2012) Immuno-oncology: understanding the function and dysfunction of theimmune system in cancer. Ann Oncol 23 (Suppl 8):viii6–9.

Flörcken A, van Lessen A, Terwey TH, Dörken B, Arnold R, Pezzutto A,and Westermann J (2013) Anti-leukemia T cells in AML: TNF-a⁺ CD8⁺ T cells mayescape detection and possibly reflect a stage of functional impairment.Hum VaccinImmunother 9:1200–1204.

Foley EJ (1953) Antigenic properties of methylcholanthrene-induced tumors in miceof the strain of origin. Cancer Res 13:835–837.

Fong L, Brockstedt D, Benike C, Wu L, and Engleman EG (2001) Dendritic cellsinjected via different routes induce immunity in cancer patients. J Immunol 166:4254–4259.

Frankenberger B and Schendel DJ (2012) Third generation dendritic cell vaccines fortumor immunotherapy. Eur J Cell Biol 91:53–58.

Fredly H, Ersvær E, Gjertsen BT, and Bruserud O (2011) Immunogenic apoptosis inhuman acute myeloid leukemia (AML): primary human AML cells expose calreti-culin and release heat shock protein (HSP) 70 and HSP90 during apoptosis. OncolRep 25:1549–1556.

Frikeche J, Clavert A, Delaunay J, Brissot E, Grégoire M, Gaugler B, and Mohty M(2011) Impact of the hypomethylating agent 5-azacytidine on dendritic cells func-tion. Exp Hematol 39:1056–1063.

Galea-Lauri J, Darling D, Mufti G, Harrison P, and Farzaneh F (2002) Elicitingcytotoxic T lymphocytes against acute myeloid leukemia-derived antigens: evalu-ation of dendritic cell-leukemia cell hybrids and other antigen-loading strategiesfor dendritic cell-based vaccination. Cancer Immunol Immunother 51:299–310.

Galluzzi L, Kroemer G, and Eggermont A (2014) Novel immune checkpoint blockerapproved for the treatment of advanced melanoma. OncoImmunology 3:e967147.

Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, and Kroemer G (2012) Trial watch: Dendriticcell-based interventions for cancer therapy. OncoImmunology 1:1111–1134.

Gang AO, Frosig TM, Brimnes MK, Lyngaa R, Treppendahl MB, Gronbak K, DufvaIH, Straten PT, and Hadrup SR (2014) 5-Azacytidine treatment sensitizes tumorcells to T-cell mediated cytotoxicity and modulates NK cells in patients with my-eloid malignancies. Blood Cancer J 4:e197.

Ge W, Ma X, Li X, Wang Y, Li C, Meng H, Liu X, Yu Z, You S, and Qiu L (2009) B7-H1up-regulation on dendritic-like leukemia cells suppresses T cell immune functionthrough modulation of IL-10/IL-12 production and generation of Treg cells. LeukRes 33:948–957.

Gilboa E (2007) DC-based cancer vaccines. J Clin Invest 117:1195–1203.Gilboa E and Vieweg J (2004) Cancer immunotherapy with mRNA-transfected den-dritic cells. Immunol Rev 199:251–263.

Goodyear O, Agathanggelou A, Novitzky-Basso I, Siddique S, McSkeane T, Ryan G,Vyas P, Cavenagh J, Stankovic T, and Moss P, et al. (2010) Induction of a CD8+ T-cell response to the MAGE cancer testis antigen by combined treatment withazacitidine and sodium valproate in patients with acute myeloid leukemia andmyelodysplasia. Blood 116:1908–1918.

Goodyear OC, Dennis M, Jilani NY, Loke J, Siddique S, Ryan G, Nunnick J, KhanumR, Raghavan M, and Cook M, et al. (2012) Azacitidine augments expansion ofregulatory T cells after allogeneic stem cell transplantation in patients with acutemyeloid leukemia (AML). Blood 119:3361–3369.

Greaves P and Gribben JG (2013) The role of B7 family molecules in hematologicmalignancy. Blood 121:734–744.

Gross L (1943) Intradermal immunization of C3H mice against a sarcoma thatoriginated in an animal of the same line. Cancer Res 3:326–333.

Gu X, Erb U, Büchler MW, and Zöller M (2015) Improved vaccine efficacy of tumorexosome compared to tumor lysate loaded dendritic cells in mice. Int J Cancer 136:E74–E84.

Gul H, Marquez-Curtis LA, Jahroudi N, Larratt LM, and Janowska-Wieczorek A(2010) Valproic acid exerts differential effects on CXCR4 expression in leukemiccells. Leuk Res 34:235–242.

Gulley JL, Madan RA, and Schlom J (2011) Impact of tumour volume on the potentialefficacy of therapeutic vaccines. Curr Oncol 18:e150–e157.

Guo ZS, Liu Z, and Bartlett DL (2014) Oncolytic immunotherapy: dying the right wayis a key to eliciting potent antitumor immunity. Front Oncol 4:74.

Hansen M, Hjortø GM, Donia M, Met Ö, Larsen NB, Andersen MH, thor Straten P,and Svane IM (2013) Comparison of clinical grade type 1 polarized and standardmatured dendritic cells for cancer immunotherapy. Vaccine 31:639–646.

Hobo W, Maas F, Adisty N, de Witte T, Schaap N, van der Voort R, and Dolstra H(2010) siRNA silencing of PD-L1 and PD-L2 on dendritic cells augments expansionand function of minor histocompatibility antigen-specific CD8+ T cells. Blood 116:4501–4511.

Hobo W, Norde WJ, Schaap N, Fredrix H, Maas F, Schellens K, Falkenburg JH,Korman AJ, Olive D, and van der Voort R, et al. (2012) B and T lymphocyte at-tenuator mediates inhibition of tumor-reactive CD8+ T cells in patients after al-logeneic stem cell transplantation. J Immunol 189:39–49.

Hobo W, Novobrantseva TI, Fredrix H, Wong J, Milstein S, Epstein-Barash H, Liu J,Schaap N, van der Voort R, and Dolstra H (2013) Improving dendritic cell vaccineimmunogenicity by silencing PD-1 ligands using siRNA-lipid nanoparticles com-bined with antigen mRNA electroporation. Cancer Immunol Immunother 62:285–297.

Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, GonzalezR, Robert C, Schadendorf D, and Hassel JC, et al. (2010) Improved survivalwith ipilimumab in patients with metastatic melanoma. N Engl J Med 363:711–723.

Hong CS, Muller L, Boyiadzis M, and Whiteside TL (2014) Isolation and character-ization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS ONE 9:e103310.

Hourigan CS and Karp JE (2013) Minimal residual disease in acute myeloid leu-kaemia. Nat Rev Clin Oncol 10:460–471.

Houtenbos I, Westers TM, Dijkhuis A, de Gruijl TD, Ossenkoppele GJ, and van deLoosdrecht AA (2007) Leukemia-specific T-cell reactivity induced by leukemicdendritic cells is augmented by 4-1BB targeting. Clin Cancer Res 13:307–315.

Hsu FJ, Benike C, Fagnoni F, Liles TM, Czerwinski D, Taidi B, Engleman EG,and Levy R (1996) Vaccination of patients with B-cell lymphoma using autologousantigen-pulsed dendritic cells. Nat Med 2:52–58.

Hutten T, Thordardottir S, Hobo W, Hübel J, van der Waart AB, Cany J, Dolstra H,and Hangalapura BN (2014) Ex vivo generation of interstitial and Langerhanscell-like dendritic cell subset-based vaccines for hematological malignancies.J Immunother 37:267–277.

Ichim CV (2005) Revisiting immunosurveillance and immunostimulation: Implica-tions for cancer immunotherapy. J Transl Med 3:8.

Iversen PO, Semaeva E, Sorensen DR, Wiig H, and Sioud M (2009) Dendritic cellsloaded with tumor antigens and a dual immunostimulatory and anti-interleukin10-specific small interference RNA prime T lymphocytes against leukemic cells.Transl Oncol 2:242–246.

Iversen PO and Sioud M (2015) Engineering therapeutic cancer vaccines that acti-vate antitumor immunity. Methods Mol Biol 1218:263–268.

Jongbloed SL, Kassianos AJ, McDonald KJ, Clark GJ, Ju X, Angel CE, Chen CJ,Dunbar PR, Wadley RB, and Jeet V, et al. (2010) Human CD141+ (BDCA-3)+dendritic cells (DCs) represent a unique myeloid DC subset that cross-presentsnecrotic cell antigens. J Exp Med 207:1247–1260.

Jonuleit H, Kühn U, Müller G, Steinbrink K, Paragnik L, Schmitt E, Knop J,and Enk AH (1997) Pro-inflammatory cytokines and prostaglandins induce mat-uration of potent immunostimulatory dendritic cells under fetal calf serum-freeconditions. Eur J Immunol 27:3135–3142.

June CH (2007) Adoptive T cell therapy for cancer in the clinic. J Clin Invest 117:1466–1476.

Kalinski P and Okada H (2010) Polarized dendritic cells as cancer vaccines: directingeffector-type T cells to tumors. Semin Immunol 22:173–182.

Kali�nski P, Vieira PL, Schuitemaker JH, de Jong EC, and Kapsenberg ML (2001)Prostaglandin E(2) is a selective inducer of interleukin-12 p40 (IL-12p40) pro-duction and an inhibitor of bioactive IL-12p70 heterodimer. Blood 97:3466–3469.

Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH,Ferrari AC, Dreicer R, and Sims RB, et al.; IMPACT Study Investigators (2010)Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl JMed 363:411–422.

Kavanagh B, O’Brien S, Lee D, Hou Y, Weinberg V, Rini B, Allison JP, Small EJ,and Fong L (2008) CTLA4 blockade expands FoxP3+ regulatory and activatedeffector CD4+ T cells in a dose-dependent fashion. Blood 112:1175–1183.

Kershaw MH, Westwood JA, and Darcy PK (2013) Gene-engineered T cells for cancertherapy. Nat Rev Cancer 13:525–541.

Khoury HJ, Collins RH Jr, Blum W, Maness L, Stiff P, Kelsey SM, Reddy A, SmithJA, and DiPersio JF (2010) Prolonged administration of the telomerase vaccineGRNVAC1 is well tolerated and appears to be associated with favorable outcomesin high-risk acute myeloid leukemia (AML) (Abstract). Blood 116:904.

Kiessling R, Klein E, and Wigzell H (1975) “Natural” killer cells in the mouse. I.Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity anddistribution according to genotype. Eur J Immunol 5:112–117.

Kirkwood JM, Butterfield LH, Tarhini AA, Zarour H, Kalinski P, and Ferrone S(2012) Immunotherapy of cancer in 2012. CA Cancer J Clin 62:309–335.

Kitawaki T, Kadowaki N, Fukunaga K, Kasai Y, Maekawa T, Ohmori K, Itoh T,Shimizu A, Kuzushima K, and Kondo T, et al. (2011a) Cross-priming of CD8(+)T cells in vivo by dendritic cells pulsed with autologous apoptotic leukemic cells inimmunotherapy for elderly patients with acute myeloid leukemia. Exp Hematol 39:424–433.

Kitawaki T, Kadowaki N, Fukunaga K, Kasai Y, Maekawa T, Ohmori K, Kondo T,Maekawa R, Takahara M, and Nieda M, et al. (2011b) A phase I/IIa clinical trial ofimmunotherapy for elderly patients with acute myeloid leukaemia using dendriticcells co-pulsed with WT1 peptide and zoledronate. Br J Haematol 153:796–799.

Klammer M, Waterfall M, Samuel K, Turner ML, and Roddie PH (2005) Fusionhybrids of dendritic cells and autologous myeloid blasts as a potential cellularvaccine for acute myeloid leukaemia. Br J Haematol 129:340–349.

Klein G, Sjogren HO, Klein E, and Hellstrom KE (1960) Demonstration of resistanceagainst methylcholanthrene-induced sarcomas in the primary autochthonous host.Cancer Res 20:1561–1572.

Kolanowski ST, Sritharan L, Lissenberg-Thunnissen SN, Van Schijndel GM, VanHam SM, and ten Brinke A (2014) Comparison of media and serum supplemen-tation for generation of monophosphoryl lipid A/interferon-g-matured type I den-dritic cells for immunotherapy. Cytotherapy 16:826–834.

Kwon YR, Son MJ, Kim HJ, and Kim YJ (2012) Reactivation of silenced WT1transgene by hypomethylating agents - implications for in vitro modeling of che-moimmunotherapy. Immune Netw 12:58–65.

Lardon F, Snoeck HW, Berneman ZN, Van Tendeloo VF, Nijs G, Lenjou M,Henckaerts E, Boeckxtaens CJ, Vandenabeele P, and Kestens LL, et al. (1997)Generation of dendritic cells from bone marrow progenitors using GM-CSF, TNF-alpha, and additional cytokines: antagonistic effects of IL-4 and IFN-gamma andselective involvement of TNF-alpha receptor-1. Immunology 91:553–559.

Le Dieu R, Taussig DC, Ramsay AG, Mitter R, Miraki-Moud F, Fatah R, Lee AM,Lister TA, and Gribben JG (2009) Peripheral blood T cells in acute myeloid leu-kemia (AML) patients at diagnosis have abnormal phenotype and genotype andform defective immune synapses with AML blasts. Blood 114:3909–3916.

Lee JJ, Kook H, Park MS, Nam JH, Choi BH, Song WH, Park KS, Lee IK, Chung IJ,and Hwang TJ, et al. (2004) Immunotherapy using autologous monocyte-deriveddendritic cells pulsed with leukemic cell lysates for acute myeloid leukemia relapseafter autologous peripheral blood stem cell transplantation. J Clin Apher 19:66–70.

Leisegang M, Wilde S, Spranger S, Milosevic S, Frankenberger B, Uckert W,and Schendel DJ (2010) MHC-restricted fratricide of human lymphocytesexpressing survivin-specific transgenic T cell receptors. J Clin Invest 120:3869–3877.

750 Anguille et al.

Lesterhuis WJ, de Vries IJ, Schreibelt G, Lambeck AJ, Aarntzen EH, Jacobs JF,Scharenborg NM, van de Rakt MW, de Boer AJ, and Croockewit, et al. (2011) Routeof administration modulates the induction of dendritic cell vaccine-inducedantigen-specific T cells in advanced melanoma patients. Clin Cancer Res 17:5725–5735.

Lichtenegger FS, Lorenz R, Gellhaus K, Hiddemann W, Beck B, and Subklewe M(2014) Impaired NK cells and increased T regulatory cell numbers during cytotoxicmaintenance therapy in AML. Leuk Res 38:964–969.

Lichtenegger FS, Mueller K, Otte B, Beck B, Hiddemann W, Schendel DJ,and Subklewe M (2012) CD86 and IL-12p70 are key players for T helper 1 polar-ization and natural killer cell activation by Toll-like receptor-induced dendriticcells. PLoS ONE 7:e44266.

Lion E, Anguille S, Berneman ZN, Smits EL, and Van Tendeloo VF (2011) Poly(I:C)enhances the susceptibility of leukemic cells to NK cell cytotoxicity and phagocy-tosis by DC. PLoS ONE 6:e20952.

Lion E, Smits EL, Berneman ZN, and Van Tendeloo VF (2012a) NK cells: key tosuccess of DC-based cancer vaccines? Oncologist 17:1256–1270.

Lion E, Willemen Y, Berneman ZN, Van Tendeloo VF, and Smits EL (2012b) Naturalkiller cell immune escape in acute myeloid leukemia. Leukemia 26:2019–2026.

Liseth K, Ersvaer E, Hervig T, and Bruserud Ø (2010) Combination of intensivechemotherapy and anticancer vaccines in the treatment of human malignancies:the hematological experience. J Biomed Biotechnol 2010:692097.

Lizée G, Overwijk WW, Radvanyi L, Gao J, Sharma P, and Hwu P (2013) Harnessingthe power of the immune system to target cancer. Annu Rev Med 64:71–90.

López JA, Bioley G, Turtle CJ, Pinzón-Charry A, Ho CS, Vuckovic S, Crosbie G,Gilleece M, Jackson DC, and Munster D, et al. (2003) Single step enrichment ofblood dendritic cells by positive immunoselection. J Immunol Methods 274:47–61.

MacDonald KP, Munster DJ, Clark GJ, Dzionek A, Schmitz J, and Hart DN (2002)Characterization of human blood dendritic cell subsets. Blood 100:4512–4520.

Maciulaitis R, D’Apote L, Buchanan A, Pioppo L, and Schneider CK (2012) Clinicaldevelopment of advanced therapy medicinal products in Europe: evidence thatregulators must be proactive. Mol Ther 20:479–482.

Maeda T, Towatari M, Kosugi H, and Saito H (2000) Up-regulation of costimulatory/adhesion molecules by histone deacetylase inhibitors in acute myeloid leukemiacells. Blood 96:3847–3856.

Mahadevan D, Lanasa MC, Whelden M, Faas SJ, Ulery TL, Kukreja A, Li L,Bedrosian CL, and Heffner LT (2010) First-in-human phase I dose escalation studyof a humanized anti-CD200 antibody (Samalizumab) in patients with advancedstage B cell chronic lymphocytic leukemia (B-CLL) or multiple myeloma (MM)(Abstract). Blood 116:1024.

Mahnke K, Schmitt E, Bonifaz L, Enk AH, and Jonuleit H (2002) Immature, but notinactive: the tolerogenic function of immature dendritic cells. Immunol Cell Biol80:477–483.

Malik B, Rath G, and Goyal AK (2014) Are the anatomical sites for vaccine admin-istration selected judiciously? Int Immunopharmacol 19:17–26.

Mandawat A, Fiskus W, Buckley KM, Robbins K, Rao R, Balusu R, Navenot JM,Wang ZX, Ustun C, and Chong DG, et al. (2010) Pan-histone deacetylase inhibitorpanobinostat depletes CXCR4 levels and signaling and exerts synergistic anti-myeloid activity in combination with CXCR4 antagonists. Blood 116:5306–5315.

Marcus A, Gowen BG, Thompson TW, Iannello A, Ardolino M, Deng W, Wang L,Shifrin N, and Raulet DH (2014) Recognition of tumors by the innate immunesystem and natural killer cells. Adv Immunol 122:91–128.

Martner A, Thorén FB, Aurelius J, and Hellstrand K (2013) Immunotherapeuticstrategies for relapse control in acute myeloid leukemia. Blood Rev 27:209–216.

Medzhitov R (2001) Toll-like receptors and innate immunity. Nat Rev Immunol 1:135–145.

Melenhorst JJ, Scheinberg P, Chattopadhyay PK, Gostick E, Ladell K, Roederer M,Hensel NF, Douek DC, Barrett AJ, and Price DA (2009) High avidity myeloidleukemia-associated antigen-specific CD8+ T cells preferentially reside in the bonemarrow. Blood 113:2238–2244.

Melero I, Gaudernack G, Gerritsen W, Huber C, Parmiani G, Scholl S, Thatcher N,Wagstaff J, Zielinski C, and Faulkner I, et al. (2014) Therapeutic vaccines forcancer: an overview of clinical trials. Nat Rev Clin Oncol 11:509–524.

Mellman I (2013) Dendritic cells: master regulators of the immune response. CancerImmunol Res 1:145–149.

Mellman I, Coukos G, and Dranoff G (2011) Cancer immunotherapy comes of age.Nature 480:480–489.

Memarian A, Nourizadeh M, Masoumi F, Tabrizi M, Emami AH, Alimoghaddam K,Hadjati J, Mirahmadian M, and Jeddi-Tehrani M (2013) Upregulation of CD200 isassociated with Foxp3+ regulatory T cell expansion and disease progression inacute myeloid leukemia. Tumour Biol 34:531–542.

Merad M, Sathe P, Helft J, Miller J, and Mortha A (2013) The dendritic cell lineage:ontogeny and function of dendritic cells and their subsets in the steady state andthe inflamed setting. Annu Rev Immunol 31:563–604.

Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK,McKenna D, Le C, Defor TE, and Burns LJ, et al. (2005) Successful adoptivetransfer and in vivo expansion of human haploidentical NK cells in patients withcancer. Blood 105:3051–3057.

Mody N, Dubey S, Sharma R, Agrawal U, and Vyas SP (2015) Dendritic cell-basedvaccine research against cancer. Expert Rev Clin Immunol 11:213–232.

Mohamadzadeh M, Berard F, Essert G, Chalouni C, Pulendran B, Davoust J, BridgesG, Palucka AK, and Banchereau J (2001) Interleukin 15 skews monocyte differ-entiation into dendritic cells with features of Langerhans cells. J Exp Med 194:1013–1020.

Moldenhauer A, Frank RC, Pinilla-Ibarz J, Holland G, Boccuni P, Scheinberg DA,Salama A, Seeger K, Moore MA, and Nimer SD (2004) Histone deacetylase in-hibition improves dendritic cell differentiation of leukemic blasts with AML1-containing fusion proteins. J Leukoc Biol 76:623–633.

Mu LJ, Lazarova P, Gaudernack G, Saeboe-Larssen S, and Kvalheim G (2004) De-velopment of a clinical grade procedure for generation of mRNA transfected

dendritic cells from purified frozen CD34(+) blood progenitor cells. Int J Immu-nopathol Pharmacol 17:255–263.

Mullins DW, Sheasley SL, Ream RM, Bullock TN, Fu YX, and Engelhard VH (2003)Route of immunization with peptide-pulsed dendritic cells controls the distributionof memory and effector T cells in lymphoid tissues and determines the pattern ofregional tumor control. J Exp Med 198:1023–1034.

Murphy KM, Nelson CA, and Sedý JR (2006) Balancing co-stimulation and inhibitionwith BTLA and HVEM. Nat Rev Immunol 6:671–681.

Muthuswamy R, Mueller-Berghaus J, Haberkorn U, Reinhart TA, Schadendorf D,and Kalinski P (2010) PGE(2) transiently enhances DC expression of CCR7 butinhibits the ability of DCs to produce CCL19 and attract naive T cells. Blood 116:1454–1459.

Narita M, Takahashi M, Liu A, Nikkuni K, Furukawa T, Toba K, Koyama S, Takai K,Sanada M, and Aizawa Y (2001) Leukemia blast-induced T-cell anergy demon-strated by leukemia-derived dendritic cells in acute myelogenous leukemia. ExpHematol 29:709–719.

Nicolette CA, Healey D, Tcherepanova I, Whelton P, Monesmith T, Coombs L, FinkeLH, Whiteside T, and Miesowicz F (2007) Dendritic cells for active immunotherapy:optimizing design and manufacture in order to develop commercially and clinicallyviable products. Vaccine 25 (Suppl 2):B47–B60.

Nierkens S, Tel J, Janssen E, and Adema GJ (2013) Antigen cross-presentation bydendritic cell subsets: one general or all sergeants? Trends Immunol 34:361–370.

Norde WJ, Hobo W, van der Voort R, and Dolstra H (2012) Coinhibitory molecules inhematologic malignancies: targets for therapeutic intervention. Blood 120:728–736.

Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, Castedo M,Mignot G, Panaretakis T, and Casares N, et al. (2007) Calreticulin exposure dic-tates the immunogenicity of cancer cell death. Nat Med 13:54–61.

Ogasawara M and Ota S (2014) Dendritic cell vaccination in acute leukemia patientsinduces reduction of myeloid-derived suppressor cells: immunological analysis ofa pilot study (Abstract). Blood 124:1113.

Okada H, Kalinski P, Ueda R, Hoji A, Kohanbash G, Donegan TE, Mintz AH, EnghJA, Bartlett DL, and Brown CK, et al. (2011) Induction of CD8+ T-cell responsesagainst novel glioma-associated antigen peptides and clinical activity by vacci-nations with alpha-type 1 polarized dendritic cells and polyinosinic-polycytidylicacid stabilized by lysine and carboxymethylcellulose in patients with recurrentmalignant glioma. J Clin Oncol 29:330–336.

Okada S, Han S, Patel ES, Yang L-J, and Chang L-J (2015) STAT3 signaling con-tributes to the high effector activities of interleukin-15-derived dendritic cells.Immunol Cell Biol 93:461–471.

Ota S and Ogasawara M (2014) Vaccination of acute leukemia patients with WT1peptide-pulsed dendritic cells induces immunological and clinical responses: a pilotstudy (Abstract). Blood 124:2319.

Palucka K and Banchereau J (2012) Cancer immunotherapy via dendritic cells. NatRev Cancer 12:265–277.

Palucka K, Banchereau J, and Mellman I (2010) Designing vaccines based on biologyof human dendritic cell subsets. Immunity 33:464–478.

Papewalis C, Jacobs B, Wuttke M, Ullrich E, Baehring T, Fenk R, Willenberg HS,Schinner S, Cohnen M, and Seissler J, et al. (2008) IFN-alpha skews monocytesinto CD56+-expressing dendritic cells with potent functional activities in vitro andin vivo. J Immunol 180:1462–1470.

Pardoll DM (2012) The blockade of immune checkpoints in cancer immunotherapy.Nat Rev Cancer 12:252–264.

Park TS, Rosenberg SA, and Morgan RA (2011) Treating cancer with geneticallyengineered T cells. Trends Biotechnol 29:550–557.

Pasare C and Medzhitov R (2003) Toll pathway-dependent blockade of CD4+CD25+T cell-mediated suppression by dendritic cells. Science 299:1033–1036.

Peled A and Tavor S (2013) Role of CXCR4 in the pathogenesis of acute myeloidleukemia. Theranostics 3:34–39.

Plantinga M, de Haar C, Nierkens S, and Boelens JJ (2014) Dendritic cell therapy inan allogeneic-hematopoietic cell transplantation setting: an effective strategy to-ward better disease control? Front Immunol 5:218.

Poggi A, Catellani S, Garuti A, Pierri I, Gobbi M, and Zocchi MR (2009) Effective invivo induction of NKG2D ligands in acute myeloid leukaemias by all-trans-retinoicacid or sodium valproate. Leukemia 23:641–648.

Prehn RT and Main JM (1957) Immunity to methylcholanthrene-induced sarcomas.J Natl Cancer Inst 18:769–778.

Prue RL, Vari F, Radford KJ, Tong H, Hardy MY, D’Rozario R, Waterhouse NJ,Rossetti T, Coleman R, and Tracey C, et al. (2015) A phase I clinical trial of CD1c(BDCA-1)+ dendritic cells pulsed with HLA-A*0201 peptides for immunotherapy ofmetastatic hormone refractory prostate cancer. J Immunother 38:71–76.

Pyzer AR, Avigan DE, and Rosenblatt J (2014) Clinical trials of dendritic cell-basedcancer vaccines in hematologic malignancies. Hum Vaccin Immunother 10:3125–3131.

Radford KJ, Turtle CJ, Kassianos AJ, Vuckovic S, Gardiner D, Khalil D, Taylor K,Wright S, Gill D, and Hart DN (2005) Immunoselection of functional CMRF-56+blood dendritic cells from multiple myeloma patients for immunotherapy.J Immunother 28:322–331.

Reddy P, Sun Y, Toubai T, Duran-Struuck R, Clouthier SG, Weisiger E, Maeda Y,Tawara I, Krijanovski O, and Gatza E, et al. (2008) Histone deacetylase inhibitionmodulates indoleamine 2,3-dioxygenase-dependent DC functions and regulatesexperimental graft-versus-host disease in mice. J Clin Invest 118:2562–2573.

Rescigno M, Avogadri F, and Curigliano G (2007) Challenges and prospects of im-munotherapy as cancer treatment. Biochim Biophys Acta 1776:108–123.

Restifo NP, Dudley ME, and Rosenberg SA (2012) Adoptive immunotherapy forcancer: harnessing the T cell response. Nat Rev Immunol 12:269–281.

Rezvani K, Yong AS, Mielke S, Savani BN, Jafarpour B, Eniafe R, Le RQ, Musse L,Boss C, and Childs R, et al. (2012) Lymphodepletion is permissive to the de-velopment of spontaneous T-cell responses to the self-antigen PR1 early after al-logeneic stem cell transplantation and in patients with acute myeloid leukemia

Dendritic Cell–Based Cancer Immunotherapy 751

undergoing WT1 peptide vaccination following chemotherapy. Cancer ImmunolImmunother 61:1125–1136.

Rizza P, Moretti F, Capone I, and Belardelli F (2015) Role of type I interferon ininducing a protective immune response: perspectives for clinical applications. Cy-tokine Growth Factor Rev 26:195–201.

Roddie PH, Horton Y, and Turner ML (2002) Primary acute myeloid leukaemia blastsresistant to cytokine-induced differentiation to dendritic-like leukaemia cells canbe forced to differentiate by the addition of bryostatin-1. Leukemia 16:84–93.

Rohner A, Langenkamp U, Siegler U, Kalberer CP, and Wodnar-Filipowicz A (2007)Differentiation-promoting drugs up-regulate NKG2D ligand expression and en-hance the susceptibility of acute myeloid leukemia cells to natural killer cell-mediated lysis. Leuk Res 31:1393–1402.

Romano E, Cotari JW, Barreira da Silva R, Betts BC, Chung DJ, Avogadri F, FinkMJ, St Angelo ET, Mehrara B, and Heller G, et al. (2012) Human Langerhans cellsuse an IL-15R-a/IL-15/pSTAT5-dependent mechanism to break T-cell toleranceagainst the self-differentiation tumor antigen WT1. Blood 119:5182–5190.

Romano E, Rossi M, Ratzinger G, de Cos MA, Chung DJ, Panageas KS, Wolchok JD,Houghton AN, Chapman PB, and Heller G, et al. (2011) Peptide-loaded Langer-hans cells, despite increased IL15 secretion and T-cell activation in vitro, elicitantitumor T-cell responses comparable to peptide-loaded monocyte-derived den-dritic cells in vivo. Clin Cancer Res 17:1984–1997.

Roothans D, Smits E, Lion E, Tel J, and Anguille S (2013) CD56 marks humandendritic cell subsets with cytotoxic potential. OncoImmunology 2:e23037.

Rosenberg SA (1999) A new era for cancer immunotherapy based on the genes thatencode cancer antigens. Immunity 10:281–287.

Rosenblatt J, Stone RM, Avivi I, Uhl L, Neuberg D, Joyce R, Tzachanis D, Levine JD,Boussiotis VA, and Zwicker J, et al. (2011) Clinical trial evaluating DC/AML fusioncell vaccination alone and in conjunction with PD-1 blockade in AML patients whoachieve a chemotherapy-induced remission. Blood 118:432–433.

Rosenblatt J, Stone RM, Uhl L, Neuberg D, Vasir B, Somaiya P, Joyce R, Levine JD,Boussiotis VA, and Zwicker J, et al. (2014) Clinical trial evaluating DC/AML fusioncell vaccination in AML patients who achieve a chemotherapy-induced remission(Abstract). Biol Blood Marrow Transplant 20:S50.

Ruben JM, van den Ancker W, Bontkes HJ, Westers TM, Hooijberg E, OssenkoppeleGJ, de Gruijl TD, and van de Loosdrecht AA (2014) Apoptotic blebs from leukemiccells as a preferred source of tumor-associated antigen for dendritic cell-basedvaccines. Cancer Immunol Immunother 63:335–345.

Ruben JM, Visser LL, Bontkes HJ, Westers TM, Ossenkoppele GJ, de Gruijl TD,and van de Loosdrecht AA (2013) Targeting the acute myeloid leukemic stem cellcompartment by enhancing tumor cell-based vaccines. Immunotherapy 5:859–868.

Sallusto F, Cella M, Danieli C, and Lanzavecchia A (1995) Dendritic cells use mac-ropinocytosis and the mannose receptor to concentrate macromolecules in themajor histocompatibility complex class II compartment: downregulation by cyto-kines and bacterial products. J Exp Med 182:389–400.

Sallusto F, Schaerli P, Loetscher P, Schaniel C, Lenig D, Mackay CR, Qin S,and Lanzavecchia A (1998) Rapid and coordinated switch in chemokine receptorexpression during dendritic cell maturation. Eur J Immunol 28:2760–2769.

Sanseverino I, Purificato C, Varano B, Conti L, Gessani S, and Gauzzi MC (2014)STAT3-silenced human dendritic cells have an enhanced ability to prime IFNgproduction by both ab and gd T lymphocytes. Immunobiology 219:503–511.

Saudemont A and Quesnel B (2004) In a model of tumor dormancy, long-term per-sistent leukemic cells have increased B7-H1 and B7.1 expression and resist CTL-mediated lysis. Blood 104:2124–2133.

Scandella E, Men Y, Gillessen S, Förster R, and Groettrup M (2002) ProstaglandinE2 is a key factor for CCR7 surface expression and migration of monocyte-deriveddendritic cells. Blood 100:1354–1361.

Schlom J (2012) Therapeutic cancer vaccines: current status and moving forward.J Natl Cancer Inst 104:599–613.

Schnorfeil F, Lichtenegger FS, Geiger C, Köhnke T, Bücklein V, Altmann T, WagnerB, Henschler R, Bigalke I, and Kvalheim G, et al. (2014) Next-generation dendriticcells for immunotherapy of acute myeloid leukemia. J Immunother Cancer 2 (Suppl3):84.

Schönefeldt C, Sockel K, Wehner R, Sopper S, Wolf D, Wermke M, Thiede C,Oelschlägel U, Ehninger G, and Bornhäuser M, et al. (2013) Azacytidine impairsNK cell activity in AML and MDS patients undergoing MRD-based pre-emptivetreatment after allogeneic stem cell transplantation. Blood Cancer J 3:e136.

Schöttker B and Schmidt-Wolf IG (2011) Pulsing with blast cell lysate or blast-derived total RNA reverses the dendritic cell-mediated cytotoxic activity ofcytokine-induced killer cells against allogeneic acute myelogenous leukemia cells.Ger Med Sci 9:Doc18.

Schreibelt G, Westdorp H, Bol KF, Winkels G, Petry K, Gerritsen WR, Figdor CG,and de Vries IJM (2014) Natural human BDCA1+ myeloid dendritic cells induceimmunological and clinical anti-tumor responses in metastatic melanoma patients;12th CIMT Annual Meeting; 2014 May 6–8;Mainz, Germany.

Schui DK, Singh L, Schneider B, Knau A, Hoelzer D, and Weidmann E (2002)Inhibiting effects on the induction of cytotoxic T lymphocytes by dendritic cellspulsed with lysates from acute myeloid leukemia blasts. Leuk Res 26:383–389.

Schürch CM, Riether C, and Ochsenbein AF (2013) Dendritic cell-based immuno-therapy for myeloid leukemias. Front Immunol 4:496.

Segura E, Durand M, and Amigorena S (2013) Similar antigen cross-presentationcapacity and phagocytic functions in all freshly isolated human lymphoid organ-resident dendritic cells. J Exp Med 210:1035–1047.

Smits EL, Anguille S, Cools N, Berneman ZN, and Van Tendeloo VF (2009) Dendriticcell-based cancer gene therapy. Hum Gene Ther 20:1106–1118.

Smits EL, Cools N, Lion E, Van Camp K, Ponsaerts P, Berneman ZN, and VanTendeloo VF (2010) The Toll-like receptor 7/8 agonist resiquimod greatly increasesthe immunostimulatory capacity of human acute myeloid leukemia cells. CancerImmunol Immunother 59:35–46.

Smits EL, Ponsaerts P, Berneman ZN, and Van Tendeloo VF (2008) The use of TLR7 andTLR8 ligands for the enhancement of cancer immunotherapy. Oncologist 13:859–875.

Smits EL, Ponsaerts P, Van de Velde AL, Van Driessche A, Cools N, Lenjou M, NijsG, Van Bockstaele DR, Berneman ZN, and Van Tendeloo VF (2007) Proin-flammatory response of human leukemic cells to dsRNA transfection linked toactivation of dendritic cells. Leukemia 21:1691–1699.

Snauwaert S, Vandekerckhove B, and Kerre T (2013) Can immunotherapy specifi-cally target acute myeloid leukemic stem cells? OncoImmunology 2:e22943.

Snauwaert S, Vanhee S, Goetgeluk G, Verstichel G, Van Caeneghem Y, Velghe I,Philippé J, Berneman ZN, Plum J, and Taghon T, et al. (2012) RHAMM/HMMR(CD168) is not an ideal target antigen for immunotherapy of acute myeloid leu-kemia. Haematologica 97:1539–1547.

Sørensen RB, Berge-Hansen L, Junker N, Hansen CA, Hadrup SR, Schumacher TN,Svane IM, Becker JC, thor Straten P, and Andersen MH (2009) The immunesystem strikes back: cellular immune responses against indoleamine 2,3-dioxygenase.PLoS ONE 4:e6910.

Spisek R, Chevallier P, Morineau N, Milpied N, Avet-Loiseau H, Harousseau JL,Meflah K, and Gregoire M (2002) Induction of leukemia-specific cytotoxic responseby cross-presentation of late-apoptotic leukemic blasts by autologous dendritic cellsof nonleukemic origin. Cancer Res 62:2861–2868.

Steinman RM (2007) Dendritic cells: understanding immunogenicity. Eur J Immunol37 (Suppl 1):S53–S60.

Steinman RM and Cohn ZA (1973) Identification of a novel cell type in peripherallymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J ExpMed 137:1142–1162.

Subklewe M, Geiger C, Lichtenegger FS, Javorovic M, Kvalheim G, Schendel DJ,and Bigalke I (2014) New generation dendritic cell vaccine for immunotherapy ofacute myeloid leukemia. Cancer Immunol Immunother 63:1093–1103.

Sundarasetty BS, Singh VK, Salguero G, Geffers R, Rickmann M, Macke L, BorchersS, Figueiredo C, Schambach A, and Gullberg U, et al. (2013) Lentivirus-induceddendritic cells for immunization against high-risk WT1(+) acute myeloid leukemia.Hum Gene Ther 24:220–237.

Szczepanski MJ, Szajnik M, Welsh A, Foon KA, Whiteside TL, and Boyiadzis M(2010) Interleukin-15 enhances natural killer cell cytotoxicity in patients withacute myeloid leukemia by upregulating the activating NK cell receptors. CancerImmunol Immunother 59:73–79.

Szczepanski MJ, Szajnik M, Welsh A, Whiteside TL, and Boyiadzis M (2011) Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppressnatural killer cell function via membrane-associated transforming growth factor-beta1. Haematologica 96:1302–1309.

Tacken PJ, de Vries IJ, Torensma R, and Figdor CG (2007) Dendritic-cell immuno-therapy: from ex vivo loading to in vivo targeting. Nat Rev Immunol 7:790–802.

Tang H, Tian E, Liu C, Wang Q, and Deng H (2013) Oxidative stress inducesmonocyte necrosis with enrichment of cell-bound albumin and overexpression ofendoplasmic reticulum and mitochondrial chaperones. PLoS ONE 8:e59610.

Teague RM and Kline J (2013) Immune evasion in acute myeloid leukemia: currentconcepts and future directions. J Immunother Cancer 1:13.

Tel J, Aarntzen EH, Baba T, Schreibelt G, Schulte BM, Benitez-Ribas D, BoermanOC, Croockewit S, Oyen WJ, and van Rossum M, et al. (2013a) Natural humanplasmacytoid dendritic cells induce antigen-specific T-cell responses in melanomapatients. Cancer Res 73:1063–1075.

Tel J, Anguille S, Waterborg CE, Smits EL, Figdor CG, and de Vries IJ (2014)Tumoricidal activity of human dendritic cells. Trends Immunol 35:38–46.

Tel J, Schreibelt G, Sittig SP, Mathan TS, Buschow SI, Cruz LJ, Lambeck AJ, FigdorCG, and de Vries IJ (2013b) Human plasmacytoid dendritic cells efficiently cross-present exogenous Ags to CD8+ T cells despite lower Ag uptake than myeloiddendritic cell subsets. Blood 121:459–467.

Tesfatsion DA (2014) Dendritic cell vaccine against leukemia: advances and per-spectives. Immunotherapy 6:485–496.

Thomas ED, Lochte HL Jr, Lu WC, and Ferrebee JW (1957) Intravenous infusion ofbone marrow in patients receiving radiation and chemotherapy.N Engl J Med 257:491–496.

Thomas L (1959) Discussion of cellular and humoral aspects of the hypersensitivitystates, in Cellular and Humoral Aspects of Hypersensitivity (Lawrence HS, ed) pp529–532, Hoeber-Harper, New York.

Topalian SL, Weiner GJ, and Pardoll DM (2011) Cancer immunotherapy comes ofage. J Clin Oncol 29:4828–4836.

Trapani JA and Smyth MJ (2002) Functional significance of the perforin/granzymecell death pathway. Nat Rev Immunol 2:735–747.

Ueno H, Schmitt N, Klechevsky E, Pedroza-Gonzalez A, Matsui T, Zurawski G, Oh S,Fay J, Pascual V, and Banchereau J, et al. (2010) Harnessing human dendritic cellsubsets for medicine. Immunol Rev 234:199–212.

Ustun C, Miller JS, Munn DH, Weisdorf DJ, and Blazar BR (2011) Regulatory T cellsin acute myelogenous leukemia: is it time for immunomodulation? Blood 118:5084–5095.

Uy GL, Rettig MP, Motabi IH, McFarland K, Trinkaus KM, Hladnik LM, Kulkarni S,Abboud CN, Cashen AF, and Stockerl-Goldstein KE, et al. (2012) A phase 1/2 studyof chemosensitization with the CXCR4 antagonist plerixafor in relapsed or re-fractory acute myeloid leukemia. Blood 119:3917–3924.

Van Acker HH, Anguille S, Van Tendeloo VF, and Lion E (2015) Empowering gammadelta T cells with antitumor immunity by dendritic cell-based immunotherapy.OncoImmunology 4:e1021538.

van de Ven R, Reurs AW, Wijnands PG, van Wetering S, Kruisbeek AM, HooijbergE, Scheffer GL, Scheper RJ, and de Gruijl TD (2012) Exposure of CD34+ pre-cursors to cytostatic anthraquinone-derivatives induces rapid dendritic cell dif-ferentiation: implications for cancer immunotherapy. Cancer ImmunolImmunother 61:181–191.

van den Ancker W, van Luijn MM, Ruben JM, Westers TM, Bontkes HJ,Ossenkoppele GJ, de Gruijl TD, and van de Loosdrecht AA (2011) Targeting Toll-like receptor 7/8 enhances uptake of apoptotic leukemic cells by monocyte-deriveddendritic cells but interferes with subsequent cytokine-induced maturation. CancerImmunol Immunother 60:37–47.

752 Anguille et al.

van den Ancker W, van Luijn MM, Westers TM, Bontkes HJ, Ruben JM, de GruijlTD, Ossenkoppele GJ, and van de Loosdrecht AA (2010) Recent advances inantigen-loaded dendritic cell-based strategies for treatment of minimal residualdisease in acute myeloid leukemia. Immunotherapy 2:69–83.

Van den Bergh JM, Van Tendeloo VF, and Smits EL (2015) Interleukin-15: new kidon the block for antitumor combination therapy. Cytokine Growth Factor Rev 26:15–24.

van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den EyndeB, Knuth A, and Boon T (1991) A gene encoding an antigen recognized by cytolyticT lymphocytes on a human melanoma. Science 254:1643–1647.

van der Pol E, Böing AN, Harrison P, Sturk A, and Nieuwland R (2012) Classifica-tion, functions, and clinical relevance of extracellular vesicles. Pharmacol Rev 64:676–705.

Van Driessche A, Berneman ZN, and Van Tendeloo VF (2012) Active specific im-munotherapy targeting the Wilms’ tumor protein 1 (WT1) for patients with he-matological malignancies and solid tumors: lessons from early clinical trials.Oncologist 17:250–259.

Van Elssen CH, Oth T, Germeraad WT, Bos GM, and Vanderlocht J (2014) Naturalkiller cells: the secret weapon in dendritic cell vaccination strategies. Clin CancerRes 20:1095–1103.

Van Lint S, Wilgenhof S, Heirman C, Corthals J, Breckpot K, Bonehill A, Neyns B,and Thielemans K (2014) Optimized dendritic cell-based immunotherapy formelanoma: the TriMix-formula. Cancer Immunol Immunother 63:959–967.

van Luijn MM, van den Ancker W, Chamuleau ME, Ossenkoppele GJ, van Ham SM,and van de Loosdrecht AA (2010) Impaired antigen presentation in neoplasia: basicmechanisms and implications for acute myeloid leukemia. Immunotherapy 2:85–97.

Van Tendeloo VF, Van de Velde A, Van Driessche A, Cools N, Anguille S, Ladell K,Gostick E, Vermeulen K, Pieters K, and Nijs G, et al. (2010) Induction of completeand molecular remissions in acute myeloid leukemia by Wilms’ tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl Acad Sci USA 107:13824–13829.

Vari F, Munster DJ, Hsu JL, Rossetti TR, Mahler SM, Gray PP, Turtle CJ, Prue RL,and Hart DN (2008) Practical blood dendritic cell vaccination for immunotherapyof multiple myeloma. Br J Haematol 143:374–377.

Vasaturo A, Di Blasio S, Peeters DG, de Koning CC, de Vries JM, Figdor CG,and Hato SV (2013) Clinical implications of co-inhibitory molecule expression inthe tumor microenvironment for DC vaccination: a game of stop and go. FrontImmunol 4:417.

Vasaturo A, Verdoes M, de Vries J, Torensma R, and Figdor CG (2015) Restoringimmunosurveillance by dendritic cell vaccines and manipulation of the tumormicroenvironment. Immunobiology 220:243–248.

Verdijk P, Aarntzen EH, Lesterhuis WJ, Boullart AC, Kok E, van RossumMM, StrijkS, Eijckeler F, Bonenkamp JJ, and Jacobs JF, et al. (2009) Limited amounts ofdendritic cells migrate into the T-cell area of lymph nodes but have high immuneactivating potential in melanoma patients. Clin Cancer Res 15:2531–2540.

Verdijk P, Aarntzen EH, Punt CJ, de Vries IJ, and Figdor CG (2008) Maximizingdendritic cell migration in cancer immunotherapy. Expert Opin Biol Ther 8:865–874.

Vinay DS and Kwon BS (2009) TNF superfamily: costimulation and clinical appli-cations. Cell Biol Int 33:453–465.

Wang LX, Mei ZY, Zhou JH, Yao YS, Li YH, Xu YH, Li JX, Gao XN, Zhou MH,and Jiang MM, et al. (2013) Low dose decitabine treatment induces CD80 ex-pression in cancer cells and stimulates tumor specific cytotoxic T lymphocyteresponses. PLoS ONE 8:e62924.

Wang RF (2002) Enhancing antitumor immune responses: intracellular peptide de-livery and identification of MHC class II-restricted tumor antigens. Immunol Rev188:65–80.

Wang X, Zhao HY, Zhang FC, Sun Y, Xiong ZY, and Jiang XB (2014) Dendritic cell-based vaccine for the treatment of malignant glioma: a systematic review. CancerInvest 32:451–457.

Weigel BJ, Panoskaltsis-Mortari A, Diers M, Garcia M, Lees C, Krieg AM, Chen W,and Blazar BR (2006) Dendritic cells pulsed or fused with AML cellular antigen

provide comparable in vivo antitumor protective responses. Exp Hematol 34:1403–1412.

Wemeau M, Kepp O, Tesnière A, Panaretakis T, Flament C, De Botton S, Zitvogel L,Kroemer G, and Chaput N (2010) Calreticulin exposure on malignant blasts pre-dicts a cellular anticancer immune response in patients with acute myeloid leu-kemia. Cell Death Dis 1:e104.

Wilgenhof S, Van Nuffel AM, Benteyn D, Corthals J, Aerts C, Heirman C, Van Riet I,Bonehill A, Thielemans K, and Neyns B (2013) A phase IB study on intravenoussynthetic mRNA electroporated dendritic cell immunotherapy in pretreated ad-vanced melanoma patients. Ann Oncol 24:2686–2693.

Willemen Y, Van den Bergh JM, Lion E, Anguille S, Roelandts VA, Van Acker HH,Heynderickx SD, Stein BM, Peeters M, and Figdor CG, et al. (2015) Engineeringmonocyte-derived dendritic cells to secrete interferon-a enhances their ability topromote adaptive and innate anti-tumor immune effector functions. CancerImmunol Immunother 64:831–842.

Wimmers F, Schreibelt G, Sköld AE, Figdor CG, and De Vries IJ (2014) Paradigmshift in dendritic cell-based immunotherapy: from in vitro generated monocyte-derived DCs to naturally circulating DC subsets. Front Immunol 5:165.

Yanagita S, Hori T, Matsubara Y, Ishikawa T, and Uchiyama T (2004) Retroviraltransduction of acute myeloid leukaemia-derived dendritic cells with OX40 ligandaugments their antigen presenting activity. Br J Haematol 124:454–462.

Yao Y, Wang C, Wei W, Shen C, Deng X, Chen L, Ma L, and Hao S (2014) Dendriticcells pulsed with leukemia cell-derived exosomes more efficiently induce antileu-kemic immunities. PLoS ONE 9:e91463.

Yao Y, Zhou J, Wang L, Gao X, Ning Q, Jiang M, Wang J, Wang L, and Yu L (2013)Increased PRAME-specific CTL killing of acute myeloid leukemia cells by eithera novel histone deacetylase inhibitor chidamide alone or combined treatment withdecitabine. PLoS ONE 8:e70522.

Zeng Z, Shi YX, Samudio IJ, Wang RY, Ling X, Frolova O, Levis M, Rubin JB, NegrinRR, and Estey EH, et al. (2009) Targeting the leukemia microenvironment byCXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy inAML. Blood 113:6215–6224.

Zhang L, Gajewski TF, and Kline J (2009) PD-1/PD-L1 interactions inhibit antitumorimmune responses in a murine acute myeloid leukemia model. Blood 114:1545–1552.

Zhang L, Zhao S, Duan J, Hu Y, Gu N, Xu H, and Yang XD (2013) Enhancement ofDC-mediated anti-leukemic immunity in vitro by WT1 antigen and CpG co-encapsulated in PLGA microparticles. Protein Cell 4:887–889.

Zhang Y, Patel S, Abdelouahab H, Wittner M, Willekens C, Shen S, Betems A, JoulinV, Opolon P, and Bawa O, et al. (2012) CXCR4 inhibitors selectively eliminateCXCR4-expressing human acute myeloid leukemia cells in NOG mouse model. CellDeath Dis 3:e396.

Zhong RK, Loken M, Lane TA, and Ball ED (2006) CTLA-4 blockade by a humanMAb enhances the capacity of AML-derived DC to induce T-cell responses againstAML cells in an autologous culture system. Cytotherapy 8:3–12.

Zhou H, Zhang D, Wang Y, Dai M, Zhang L, Liu W, Liu D, Tan H, and Huang Z (2006)Induction of CML28-specific cytotoxic T cell responses using co-transfected den-dritic cells with CML28 DNA vaccine and SOCS1 small interfering RNA expressionvector. Biochem Biophys Res Commun 347:200–207.

Zhou Q, Munger ME, Highfill SL, Tolar J, Weigel BJ, Riddle M, Sharpe AH, ValleraDA, Azuma M, and Levine BL, et al. (2010) Program death-1 signaling and regu-latory T cells collaborate to resist the function of adoptively transferred cytotoxicT lymphocytes in advanced acute myeloid leukemia. Blood 116:2484–2493.

Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, Hart DN, Leenen PJ, LiuYJ, MacPherson G, and Randolph GJ, et al. (2010) Nomenclature of monocytes anddendritic cells in blood. Blood 116:e74–e80.

Zinkernagel RM and Doherty PC (1974a) Immunological surveillance against alteredself components by sensitised T lymphocytes in lymphocytic choriomeningitis.Nature 251:547–548.

Zinkernagel RM and Doherty PC (1974b) Restriction of in vitro T cell-mediated cy-totoxicity in lymphocytic choriomeningitis within a syngeneic or semiallogeneicsystem. Nature 248:701–702.

Dendritic Cell–Based Cancer Immunotherapy 753