1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52
REVIEW ARTICLE Q1
Q37
Antitumor dendritic cell–based vaccines: lessons from 20 years of clinical trials and future perspectives ~ CONSTANTINO, CELIA ~ JOAO GOMES, AMILCAR FALCAO, MARIA T. CRUZ, and BRUNO M. NEVES COIMBRA AND AVEIRO, PORTUGAL
Dendritic cells (DCs) are versatile elements of the immune system and are best known for their unparalleled ability to initiate and modulate adaptive immune responses. During the past few decades, DCs have been the subject of numerous studies seeking new immunotherapeutic strategies against cancer. Despite the initial enthusiasm, disappointing results from early studies raised some doubts regarding the true clinical value of these approaches. However, our expanding knowledge of DC immunobiology and the definition of the optimal characteristics for antitumor immune responses have allowed a more rational development of DC-based immunotherapies in recent years. Here, after a brief overview of DC immunobiology, we sought to systematize the knowledge provided by 20 years of clinical trials, with a special emphasis on the diversity of approaches used to manipulate DCs and their consequent impact on vaccine effectiveness. We also address how new therapeutic concepts, namely the combination of DC vaccines with other anticancer therapies, are being implemented and are leveraging clinical outcomes. Finally, optimization strategies, new insights, and future perspectives on the field are also highlighted. (Translational Research 2015;-:1–22) Abbreviations: APCs ¼ antigen-presenting cells; BMP4 ¼ bone morphogenetic protein 4; cDCs ¼ classical DCs; CDPs ¼ common DC precursors; CLPs ¼ common lymphoid precursors; CLRs ¼ C-type lectin receptors; CMPs ¼ common myeloid precursors; CTLs ¼ cytotoxic T cells; CTLA-4 ¼ cytotoxic T-lymphocyte–associated protein 4; DC ¼ dendritic cell; Dex ¼ dendritic cell–derived exosomes; GM-CSF ¼ granulocyte-macrophage colony–stimulating factor; HLA ¼ human leukocyte antigen; HSC ¼ hematopoietic stem cells; i.d. ¼ intradermal; IDO ¼ indoleamine2,3-dioxygenase; IL ¼ interleukin; IFN ¼ interferon; i.n. ¼ intranodal; iPSCs ¼ induced pluripotent stem cells; i.t. ¼ intratumoral; i.v. ¼ intravenous; LC ¼ Langerhans cells; mAb ¼ monoclonal antibody; M-CSF ¼ macrophage colony–stimulating factor; MDPs ¼ macrophage and DC precursors; MDSCs ¼ myeloid-derived suppressor cells; MHC ¼ major histocompatibility complex; MoDCs ¼ monocyte-derived DCs; NK ¼ natural killer; PAMPs ¼ pathogen-associated molecular
Q39
From the Faculty of Pharmacy and Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI) and Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), University of Coimbra, Coimbra, Portugal; CNC, Institute for Biomedical Imaging and Life Sciences, University of Coimbra, Coimbra, Portugal; Department of Chemistry and QOPNA, Mass Spectrometry Centre, University of Aveiro, Aveiro, Portugal.
Submitted for publication May 19, 2015; revision submitted July 25, 2015; accepted for publication July 28, 2015. Reprint requests: Bruno M. Neves, Faculty of Pharmacy and Centre for Neuroscience and Cell Biology, University of Coimbra, 3000548 Coimbra, Portugal; e-mail:
[email protected]. 1931-5244/$ - see front matter Ó 2015 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.trsl.2015.07.008
1 REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:06 pm ce
53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106
2
107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160
Translational Research - 2015
Constantino et al
patterns; PAP ¼ prostatic acid phosphatase; PBMCs ¼ peripheral blood mononuclear cells; PD-1 ¼ programmed cell death 1; pDCs ¼ plasmacytoid DCs; PGE2 ¼ prostaglandin E2; PolyI:C ¼ polyinosinic:polycytidylic acid; PKR ¼ protein kinase RNA-activated receptors; RECIST ¼ response evaluation criteria in solid tumors; s.c. ¼ subcutaneous; SCF ¼ stem cell factor; TAAs ¼ tumor-associated antigens; TCR ¼ T-lymphocyte receptor; TGF ¼ transforming growth factor; TLR ¼ toll-like receptor; TRAIL ¼ TNF-related apoptosis-inducing ligand; Tregs ¼ regulatory T cells; WHO ¼ World Health Organization
Q2
INTRODUCTION
Q3
Q4
Q5
Dendritic cells (DCs), initially described by Steinman and Cohn in a series of pioneer studies,1 play a critical role at the interface between the innate and adaptive arms of the immune system. These cells induce primary immune responses, potentiate the effector functions of previously primed T lymphocytes, and orchestrate/ modulate the communication between other innate and adaptive immune cells.2 Although they represent a very small population of leukocytes, DCs are professional antigen-presenting cells (APCs) that are highly efficient in generating robust immune responses and maintaining tolerance to self or harmless foreign antigens. This functional plasticity renders DCs a very attractive tool for the design of immunotherapeutic approaches, namely to boost the immune system during cancer treatment. Since the first pilot study using ex vivo MAGE-1–pulsed DCs for the treatment of melanoma in 1995,3 more than 300 clinical trials on antitumor DC-based treatments have been completed or are currently ongoing (Fig 1, A). Ongoing or previously completed randomized phase 3 clinical trials have been performed across a wide range of malignancies, including melanoma, prostate cancer, malignant glioma, renal cancer, and lymphoma (Fig 1, B). The great expectations created by the early experimental results were not always followed by objective clinical responses, and some skepticism ensued. In fact, according to the World Health Organization criteria or the response evaluation criteria in solid tumors, antitumor DC-based vaccines rarely exceed 15% of the objective responses.4 However, the paradigm of antitumor activity assessment is changing, and overall survival (OS) is now viewed as one of the most relevant outcomes to measure therapeutic benefits.5 In 2010, this notion was reinforced by the Food and Drug Administration (FDA) approval of the first DC-based vaccine, sipuleucel-T (Dendreon, Washington), as a treatment for metastatic hormone-resistant prostate cancer. Despite the fact that only 5% of patients exhibited tumor regression in the phase 3 clinical trial, the vaccine was shown to significantly increase the OS by 22.5%.6 Meanwhile, the FDA and the European Medicines
Agency (EMA) granted a special regulatory framework, such as orphan drug designation, to several other antitumor DC-based vaccines. The ICT-107 vaccine (ImmunoCellular Therapeutics, California) for the Q6 treatment of glioblastoma multiforme and the AV0113 vaccine for malignant glioma from the Austrian biotech company Activartis received the orphan drug designation by the FDA and EMA. In 2012, the EMA also granted orphan drug designation to DCP-001, a vaccine for acute myeloid leukemia produced by the Dutch company DCPrime BV. Finally, antitumor DC-based Q7 vaccines have been tested in several countries as Hospital Exemption therapies, a special regime defined by the regulation on advanced therapy medicinal products. An example of a vaccine within this framework is the DCVax-L from Northwest Biotherapeutics (Bethesda, Maryland) that was recently approved by the German Paul Ehrlich Institute for treatment of glioma. In the present work, the current status of DC immunobiology is briefly reviewed and the characteristics of antitumor DC-based vaccines that have been tested in more than 300 clinical trials for more than the last 2 decades are then summarized. We specially address the practical aspects of DC vaccine production and the influence of the different procedures on clinical effectiveness. Finally, new therapeutic concepts, optimization strategies, and future perspectives on the area are also highlighted. DC IMMUNOBIOLOGY Origin and classification. DCs are a heterogeneous family of specialized APCs that ultimately share the same hematopoietic stem cell precursor. In bone marrow, hematopoietic stem cells give rise to common myeloid precursors and common lymphoid precursors, which then originate intermediate progenitors, such as macrophage and DC precursors. Macrophage and DC precursors further differentiate into common DC precursors that are restricted to the generation of the 2 known DC populations: classical DCs (cDCs), and plasmacytoid DCs (pDCs) (reviewed in Refs 7 and 8). It is now accepted that, independent of the precursor lineage, the differentiation and expansion of specific DC subsets is strongly regulated by different
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:06 pm ce
161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214
Translational Research Volume -, Number -
3
print & web 4C=FPO
215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268
Constantino et al
Fig 1. Twenty years of clinical trials using antitumor DC-based immunotherapies. (A) Clinical trials reporting antitumor DC-based vaccines registered at ClinicalTrial.gov until March 2015. (B) Frequencies distribution of clinical trials among the different malignancies and their actual status. In ‘‘Other’’ category are included bladder cancer, mesothelioma, urothelial carcinoma, and cervical cancer. DC, dendritic cell.
Q8
Q9
hematopoietic cytokines such as Fms-like tyrosine kinase cytokine-3 ligand (Flt3L), macrophage colony– stimulating factor, and granulocyte-macrophage colony–stimulating factor (GM-CSF).8 On the basis of this knowledge, the ‘‘graded commitment’’ model was proposed.9 This model states that the different precursors are not restricted to give rise to one particular type of DC. Rather, there is a gradation of probability ranging from precursors that can give rise to all DC subtypes to those that can only produce the classical or plasmacytoid subsets. The classification of the different human DC subsets has been the subject of intense debate and constant evolution. DCs were for long classified on the basis of phenotypical and functional characteristics. However, given that these characteristics are often overlapping an unequivocal classification has been challenging. Recently, a new classification system based primarily on DC ontogeny and secondarily on their location, function, and phenotype-gained relevance.10 DCs found in lymphoid and nonlymphoid tissues are therefore classified into 2 main groups, cDCs and pDCs. Human skin hosts 3 major subtypes of cDCs differentiated by their expression of CD1a, CD14, and CD141: CD41/CD1ahi/CD142/DC-SIGN2/CD20611/CD162, CD41/CD1alow/CD141/DC-SIGN1/CD2061/CD162, and CD1411/XCR11 DCs.8,11 CD1411/XCR11 DCs have been shown to be extremely efficient in antigen cross-presentation and to produce significant amounts of tumor necrosis factor a (TNF-a), CXCL10, and interleukin 12 (IL-12)p70 on stimulation.12-14 These cells effectively interact with natural killer (NK) cells and CD81 T cells, an event facilitated by the
expression of the XCR1 chemokine receptor. The human epidermis is also populated by other specialized APCs termed Langerhans cells (LCs). LCs are characterized by low CD11c and high CD1a and CD2017 expressions, which distinguish them from cDCs subsets. In addition, unlike any other DC population, LCs derive from embryonic precursors and have the capacity to self-renew locally.15 LCs are capable to cross-present antigens and to evoke tolerogenic or immunogenic responses depending on the microenvironment and type of maturation stimulus.11 In blood and lymphoid and nonlymphoid tissues, 2 cDC populations have been described according to their expression of nonoverlapping markers CD141 and CD1c. CD1411 DCs are primarily present in the blood and lymph nodes, although they can also be found in the lungs and liver. CD1c1 DCs are the predominant cDCs in the blood, and are also found in lymph nodes, spleen, and mucosa. These cells express toll-like receptors (TLRs) 1–10 and some C-type lectin receptors, such as DC-SIGN, DEC-205, DCIR, mannose receptor, Cleac9A, and Dectin 1 and 2.16,17 CD1c1 DC Q10 subpopulation produces IL-8, IL-10, TNF-a, and IL23 on activation, and their major role appears to be related to the modulation of mucosal T-cell responses and immunity toward extracellular pathogens.18 Finally, besides the 2 cDCs subsets, blood and lymphoid tissues also host pDCs, cells phenotypically characterized as major histocompatibility complex (MHC)-II1/ CD11c2/CD1231/CD3031/CD3041. pDCs have an Q11 enormous functional plasticity and are able to polarize T cells into Th1, Th2, regulatory T cells (Tregs), and cytotoxic T cells (CTLs). These cells are primarily
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:06 pm ce
269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322
4
323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376
Translational Research - 2015
Constantino et al
implicated in antiviral responses by producing large amounts of type 1 interferon (IFN-a/b) and by efficiently cross-presenting viral antigens to the CD81 T lymphocytes.20 Antigen recognition, processing, and presentation. DCs sample their microenvironment and capture antigens by receptor-mediated endocytosis, phagocytosis, and macropinocytosis. The antigen-processing mechanisms depend on the origin (endogenous or exogenous) and molecular nature of the antigen (protein or lipid).21 Therefore, 3 processing and presentation mechanisms have been characterized: (1) the exogenous (or endosomal) pathway, where antigenic peptides are generated and coupled to MHC-II molecules in phagolysosomes and then transported to the cell surface and presented to CD41 T cells; (2) the endogenous (or proteasome) pathway, where the intracellular proteins are degraded by the proteasome, and the resulting peptides are coupled to MHC-I molecules in the Golgi and then transported to plasma membrane and presented to CD81 T cells; and (3) mechanisms where lipid antigens are coupled to CD1 family molecules and presented to CD81 T cells, g/d T cells, or NK T cells.22 Exogenous antigens can also be presented to CD81 T cells via MHC-I complexes, a process termed crosspresentation. In cross-presentation, internalized proteins are retrotranslocated into the cytosol and degraded by the proteasome. The resulting antigenic peptides migrate to the ER, where they are coupled to MHC-I molecules and subsequently presented to the CD81 T lymphocytes, inducing either immunogenic responses (cross-priming) or tolerance (cross-tolerance).23 Cross-presentation is, therefore, critical for the priming and activation of CTLs against viruses, tumors, and intracellular bacteria. DC maturation. In the classical view, on contact with a ‘‘danger signal,’’ conventional immature DCs undergo a complex process of morphologic, phenotypic, and functional modifications referred to as maturation. These modifications allow the egress of DCs from the peripheral tissues to the marginal zones in the draining lymph nodes, where they present antigens to naive T lymphocytes. Several stimuli have been shown to trigger DCs maturation, including small reactive chemicals, proinflammatory cytokines, such as IL-1b, IL-6, IFN-g, and TNF-a, and pathogen-associated molecular patterns, such as lipopolysaccharides, bacterial DNA, and double-stranded RNA.24 One hallmark of DC maturation is the upregulation of costimulatory molecules, such as CD40, CD54, CD80, CD83, and CD86, and the shift in the chemokine receptor profile. The expression of costimulatory molecules is of great importance for the adequate stimulation of
T cells during antigen presentation, and the modification of chemokine receptors results in the acquisition of migration.24 Although immature DCs express the CCR1, CCR2, CCR5, CCR6, CXCR1, and CXCR2 receptors, mature DCs upregulate CXCR4 and CCR7 and gain responsiveness to the lymphoid chemokines CCL19 and CCL20.25 The profile of cytokines and che- Q12 mokines produced by the DCs also undergoes profound alterations during maturation, being fundamentally dependent on the DC subset and on the stimulus that triggers the maturation process. The expression of the cytokines TNF-a, IL-10, IL-1a/b, IL-12p70, IFN-g, IL-8, IL-6, and IL-23 is normally increased in mature DCs.26 After contact with the maturation stimulus, the DCs transiently produce chemokines, such as CCL2, CCL3, CCL4, CCL5, CCL8, and CXCL8, which are important for the recruitment of monocytes and neutrophils to the site of infection/inflammation. Later, the Q13 production of the lymphoid chemokines, such as CCL17, CCL18, CCL19, CCL22, and CXCL10 increases, promoting the recruitment of T and B lymphocytes and facilitating the interaction of DCs with these cells.27 DC–T cell interactions. During antigen presentation, DCs provide 3 signals that drive the activation and polarization of T cells into their effector and regulatory populations.28 Signal 1 is provided by the interaction of the T-lymphocyte receptor with the MHC-I or MHC-II– antigen complexes presented by the DCs. In the absence of costimulation, signal 1 is commonly associated with the inactivation of naive T lymphocyte by anergy or deletion, promoting tolerogenic responses. The second signal, costimulation, results from the interaction of the costimulatory molecules expressed by the DCs with the respective ligands on the surface of T cells. Together with signal 1, costimulation promotes T-cell survival and proliferation and stabilizes cytokine production. The increased expression of costimulatory molecules during DC maturation has long been associated with the transition from a tolerogenic to an immunogenic state.29 However, numerous data show that costimulatory molecules associated with immunogenic responses may also be involved in the induction of tolerance. In fact, the binding of the costimulatory molecules CD80 and CD86 to cytotoxic T-lymphocyte antigen 4 (CTLA-4) Q14 acts as a negative regulator of T-cell activation. Additionally, the interaction of CD80 and CD86 with CTLA-4 and CD28 has also been shown to be essential for Treg development, homeostasis, and suppressor activity.30 Similarly, the interaction between 4-1BBL and OX40L expressed on DCs with their receptors, 41BB and OX40, respectively, in T cells can promote immunogenic31 or tolerogenic responses.32 Therefore, Q15 signal 2 is the result of a complex balance between
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:06 pm ce
377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430
Translational Research Volume -, Number -
431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484
Constantino et al
positive and negative costimulation during DC-T cell cross talk. DCs secrete soluble factors such as cytokines and chemokines (signal 3), which are crucial for the differentiation of CD81 T cells into CTLs33 and for the polarization of CD41 T cells into their different effectors (Th1, Th2, and Th17)34 or regulatory subpopulations (Treg, Tr1, and Th3).35 The presence of IL-12p70, IL-15, and IFN-a/b during the presentation of MHCI–antigen complexes to CD81 T cells enhances their expansion, promotes CTL differentiation, and regulates memory T-cell formation.36 Although IL-12p70 appears to be the critical ‘‘signal 3’’ for CD81 T-cell expansion, it was shown that it may negatively impact the development of both primary and secondary antigen-specific memory CD81 T cells.37 In the DC-mediated polarization of CD41 T cells, the Th1 phenotype is induced by IL-12p70, IL-18, and IL-27, whereas Th17 differentiation primarily depends on transforming growth factor b (TGF-b) and IL-6, although other cytokines, such as IL-1b, IL-21, and IL-23, may also be involved. Regarding Tregs polarization, it is now well established that it depends on the DC ontogeny and capacity to produce IL-2, IL-10, and TGF-b as well as on T-lymphocyte intrinsic factors and on the balance between positive and negative costimulatory signals.38 DESIRED PROPERTIES FOR ANTITUMOR IMMUNOTHERAPIES
Nearly 3 decades of experimental and clinical observations have allowed the scientific community to shed light on the optimal characteristics for antitumor immunotherapies. An important milestone was the identification of the key role played by CD81 T cells in immune responses to cancer.39,40 The induction of antigenspecific CD81 T cells and the subsequent generation of CTLs leads to the recognition of MHC-I–antigenic peptide complexes presented on the surface of tumor cells, triggering their destruction. Therefore, desirable antitumor vaccines should expand the numbers of circulating tumor-specific CTLs; improve their cytolytic abilities (increasing the production of granzymes and perforin); increase the CTLs’ avidity for MHC-I molecules on tumor cells and increase the expression of molecules, such as CXCR3 and CD103/CD49a, that favor the migration and persistence of CTLs into the tumor areas.41,42 It is also desirable that the elicited CD81 T cells express high levels of positive costimulatory molecules such as CD137 and low levels of the immune checkpoint proteins CTLA-4 and programmed cell death 1 (PD-1).43 Finally, antitumor vaccines are also expected to modulate the circulating effector and memory CD81 T cells.
5
In addition to the CTL-driven responses, effective immunity against tumors is also strongly dependent on CD41 T cells and NK cells. CD41 T cells, particularly the Th1 subset, directly kill tumor cells,44 activate tumor infiltrating-macrophages and contribute to the differentiation and expansion of antigen-specific CTLs by producing cytokines such as IL-2 and IL-21.45,46 CD41 T cells are also required for the adequate formation of long-term memory CD81 T cells.47 The role of the NK cells in tumor immunosurveillance has gained substantial interest in recent years.48 In response to a balance between inhibitory/stimulatory signals from invariant receptors, NK cells directly kill tumor cells without prior immunization or MHC restriction. Q16 This is accomplished through at least 4 mechanisms: cytoplasmic granule release, effector molecule production, death receptor-induced apoptosis via the TNFrelated apoptosis-inducing ligand and Fas ligand, or antibody-dependent cellular cytotoxicity.49 In addition, NK cells indirectly contribute to tumor elimination by modulating the functions of other immune cells. Activated NK cells potentiate DC maturation and IL-12p70 production and favor immunogenic DC populations by killing immature DCs, whereas sparing fully activated DCs.50 Finally, the cell debris that result from the NK-induced destruction of tumor cells fuel the DCs with antigens, which enhance their cross-presentation to the CD81 T cells.51 However, some care must be taken with the elicitation of Th1 responses and the activation of NK cells in the context of cancer treatment. By producing IFN-g, these cells potently upregulate the expression of PD-1 ligands (PD-L1 and PD-L2) in tumors, which may contribute to tumor immune evasion.52 Other CD41 T-cell subsets, such as Th2 cells and Tregs, also counteract the CTLs antitumor activity. By secreting IL-4, Th2 cells limit the cytolytic activity of CTLs by decreasing the expression of granzymes and perforin,53 whereas Tregs negatively affect CTLs because of the production of IL-10 and TGF-b.54,55 In addition, Tregs can also impair CD81 T-cell expansion by competing with them for the cytokine IL-2.56 RATIONALE FOR ANTITUMOR DC-BASED VACCINES
DCs represent the perfect tool for immunotherapeutic interventions because of their unparalleled capacity for antigen presentation, their ability to modulate other immune players, and their functional plasticity.2 The effectiveness of DC vaccines is the subject of debate, mainly because the clinical outcomes addressed may not be the most adequate. However, there is no doubt that these approaches elicit strong tumor antigenspecific immune responses (CTLs and Th1 cells). A
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:06 pm ce
485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538
6
539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592
Q17
Translational Research - 2015
Constantino et al
meta-analysis of antitumor DC-based vaccines for prostate cancer and renal cell carcinoma revealed that the interventions induced these immune responses in 77% and 61% of the patients, respectively.57 Moreover, in addition to boosting adaptive immunity, the DCs were shown to enhance the antitumor activity of NK cells by increasing their cytolytic abilities and IFN-g production.58 Following this seminal publication, a plethora of other studies revealed the crucial relevance of DC-NK cell cross talk for efficient tumor elimination (reviewed in Lion et al59). In fact, effective DC vaccine-mediated antitumor immunity is at least in part dependent on NK cell activity.51 This was elegantly demonstrated in murine models of melanoma and metastatic lung tumors, where the tumor eradication observed after DC vaccination was completely abrogated in animals that had been depleted of NK cells.60,61 Although only a limited number of DC-based cancer vaccination trials implemented NK cell motorization, the existing data indicate that nearly 50% of patients showed an increased frequency and/or an induction of NK cell activation.59 Therefore, it is now clear that the antitumor responses promoted by DC vaccination rely on their capacity to prime and activate CD81 T cells, to polarize CD41 T cells into Th1 populations, and to cross talk with NK cells.62-65 Another important argument favoring DC-based immunotherapies is their safety profile. Twenty years of observations in numerous phase I and II trials demonstrated that DC vaccines are generally well tolerated and induce minimal toxicity. The most common manifestations are local reactions at the injection sites, such as rash and pruritus, and occasionally systemic effects occur, including fever and malaise.4 Despite the initial concerns regarding the possibility of inducing autoimmunity, the DC-based therapies are rarely associated with severe immunotoxicity reactions. METHODOLOGIES FOR THE PRODUCTION OF DCBASED VACCINES
Q18
The present review is focused on the diversity of approaches used to directly manipulate or target DCs in the context of cancer treatment, with a special emphasis on the impact of the different methodologies on vaccine effectiveness. The information systematized subsequently was collected from www.clinicaltrials.gov (until March 2015) and from key articles on the subject. Table I summarizes the different methodologies/approaches and the respective benefits and drawbacks. The complete list of clinical trials analyzed and their major characteristics can be found in the Supplementary material. Ex vivo DC manipulation. Ex vivo manipulation of DCs is by far the most explored strategy and is used in
approximately 97% of the clinical trials initiated to date (Fig 2, A). This approach requires obtaining DCs or DC precursors from patients, manipulating them (inducing maturation and loading antigens), and reinjecting them into the donor. From the time of the production of this type of DC-based vaccine according to good manufacturing procedures is a time- and costintensive procedure, individual aliquots are normally produced and cryopreserved at the beginning of the treatment and are released along the vaccination schedule. Cryopreservation, when performed under optimal conditions, does not significantly affect the viability, phenotype, or function of the DCs, and the thawed cells are suitable for clinical use.66 Currently, there is no standardized procedure for ex vivo manipulation, which results in a plethora of protocols that differ in the source of DCs, the maturation stimulus, the nature and procedure for antigen loading and, finally, the route of administration. DC source. The DCs used in tumor immunotherapies can be either autologous or allogenic. As shown in Fig 2, B, the use of autologous DCs has been by far the preferred approach. Although allogenic DCs are less frequently used, their usage is supported by experimental evidence demonstrating that allogeneic human leukocyte antigen (HLA) molecules represent potent immunogenic signals and boost antitumor immunity.67,68 Besides, it may be advantageous to use cells from normal donors because DCs and DC precursors from cancer-bearing patients are targeted by tumor immunosuppressive factors that render them functionally aberrant.69,70 The immunosuppressive factors that are usually overproduced by tumor cells include IL10, TGF-b, vascular endothelial growth factor (VEGF), IL-6, and prostaglandin E2 (PGE2), and dysfunctional DCs have been observed in patients with chronic lymphocytic leukemia, melanoma, ovarian, breast, renal, prostate, lung, and head and neck cancers.71-74 Allogenic DCs are commonly obtained by differentiation of peripheral blood mononuclear cells (PBMCs) of unrelated healthy donors. However, at least 2 clinical trials use DC-like cell lines (Fig 2, B). The use of the proprietary dendritic progenitor cell line DCone was tested in acute myeloid Q19 leukemia patients (NCT01373515), and the safety and tolerability of subcutaneous (s.c.) administration of an allogeneic pDC line was tested in patients with melanoma (NCT01863108). Given that circulating DCs represent less than 1% of PBMCs, the cells used in therapeutic cancer vaccines are typically differentiated from autologous leukapheresis-isolated CD141 monocytes or CD341 hematopoietic progenitors. Differentiation from monocytes involves the cell culture for 5–7 days in the
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:06 pm ce
593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646
Translational Research Volume -, Number -
647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700
Constantino et al
7
Table I. Advantages and drawbacks of the different methodologies/approaches used for production of DCbased antitumor vaccines Advantages
Ex vivo DC manipulation Genetic source Autologous
- No risk of graft-vs-host disease
Allogenic
- Allogenic HLA molecules represent potent immunogenic signals67,68
DC source CD14+ monocytes
- Abundant source: CD14+ monocytes represent 10% of PBMCs
CD34+ hematopoietic progenitors Natural occurring DCs iPSC-derived DCs
Antigens and loading procedures Peptide pulsing
Protein pulsing
- CD34+-derived DCs stimulate more effective CTL responses than MoDCs75,76 - Low doses of plasmacytoid DCs effectively induce CD4+ and CD8+ T-cell responses77 - iPSCs may be continuously expanded in vitro153 - DCs can be tailored to a desired phenotype (eg, CD11c+CD141+XCR1+ DCs)154 - Peptides are synthesized and purified at low cost - Enables direct monitoring of T-cell responses - Short peptides are directly loaded into MHC molecules - Synthetic peptides allow for modifications - Full-length proteins may present more epitopes for immune recognition - Prolonged antigen presentation
Tumor lysates
- Encompasses multiple TAAs (even those not yet characterized)
Tumor apoptotic bodies
- Encompasses multiple TAAs (even those not yet characterized)
RNA transfection
- Tumor mRNA may be amplified as needed - Possibility to introduce mRNAs coding for immunostimulatory proteins
Viral transduction
- The vector may intrinsically activate DCs95 - Possibility to introduce genes coding for cytokines and costimulatory molecules94
DC-tumor cell hybrids
- Encompasses multiple TAAs - Antigen presentation is maintained for days
DC maturation protocols TNF-a + IL-1b + IL-6 + PGE2 TNF-a + IL-1b + IFN-a + IFN-g + polyinosinic: polycytidylic acid Routes of administration Intravenous Intradermal/subcutaneous
Drawbacks
- DCs from cancer-bearing patients are frequently dysfunctional69-74 - Survival of injected DCs may be shortened by T-cell–mediated rejection - CD14+-derived DCs (MoDCs) are not as efficient as other DC subsets in eliciting CTL responses - Limited number: represent 0.1% of PBMCs - Limited number: circulating DCs represent less than 1% of PBMCs - Laborious and expensive - Production according to GMP requirements is technically challenging - Limited number of known immunogenic TAAs - HLA restriction - Peptides with low affinity for MHC may be poorly immunogenic - Limited number of known immunogenic TAAs - Propensity of proteins to be targeted for MHC-II - Dependence on the availability of tumor cells - Immunologic responses more complex to monitor - Dependence on the availability of tumor cells - Immunologic responses more complex to monitor - Reagents and procedures to transfect DCs may affect their viability - Limited stability and short lifespan of mRNA - Immune responses to vector antigens may overwhelm those for tumor antigens95 - Viral vectors may perturb DC functions95 - Low expression of IL-12p70 and costimulatory molecules97
- PGE2 enhances CCR7 expression and DC migratory capacities106 - Efficiently induces IL-12p70 - aDC1s elicit potent CTL responses102,109-111
- PGE2 may reduce the capacity of DCs to produce IL-12p70107 - aDC1s may have reduced migratory capacities compared with standard matured DCs
- Induces antigen-specific humoral responses more efficiently than i.d. and i.n. routes117 - Elicit strong specific antitumor Th1 and CTL responses
- Injected DCs preferentially accumulate in the spleen and liver - Just 1%–5% of injected DCs reach the peripheral lymph nodes117,118 (Continued )
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:06 pm ce
701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754
8
755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808
Translational Research - 2015
Constantino et al
Table I. (Continued ) Advantages
Intranodal Intratumoral In vivo DC targeting Cancer antigens fused to monoclonal antibodies targeting DC surface receptors
Drawbacks
- Substantially more DCs reach the T-cell areas in lymph nodes118,120 - May delay and reverse the tolerization of tumor-infiltrating effector T cells124
- Technically exigent - Great variability between patients - Technically exigent - Associated with high morbidity
- Bypasses the expensive and laborious ex vivo DC generation process - Allows targeting specific DC subsets128-130
- Limited to known TAAs - It requires the coadministration of adjuvants130 - Targeted receptor must be unambiguously expressed
Other strategies directly/indirectly involving DCs Nontargeted antigen-based - Easier to produce vaccines - Stable in many storage conditions - Induce CD4+ and CD8+ T-cell responses GM-CSF–secreting tumor cells - Potentiates TAAs presentation by endogenous DCs136 - Consistent induction of cellular and humoral antitumor immune responses137-139
Implantable DC-recruiting scaffolds
Dex
- Bypasses the expensive and laborious ex vivo DC generation process - The approach takes advantage of endogenous DCs - Preclinical studies show induction of specific and effective antitumor immunity141-143 - Dex are able to elicit CD4+ and CD8+ T-cell responses144-147 - Exosomes may carry additional mRNAs and miRNAs to enhance immune responses - Production under GMP conditions is well defined
- Limited to known TAAs - Immune responses may be transient and/ or of low magnitude - Prolonged GM-CSF production by tumor cells may cause immune tolerance140 - Dependence on the availability of tumor cells - Immunologic responses more complex to monitor - The design of biocompatible polymeric scaffolds incorporating functional proteins (chemokines) is technically challenging - Until present the results are only from preclinical studies - Existing clinical data indicate that T-cell responses elicited by Dex are limited149,150 - Requires large quantities of ex vivo generated DCs for exosome production
Abbreviations: CTL, cytotoxic T cell; DC, dendritic cell; Dex, dendritic cell–derived exosomes; GMP, good manufacturing procedures; GM-CSF, granulocyte-macrophage colony–stimulating factor; HLA, human leukocyte antigen; IFN, interferon; IL, interleukin; iPSC, induced pluripotent stem cell; MHC, major histocompatibility complex; MoDC, monocyte-derived dendritic cells; mRNA, messenger RNA; PBMC, peripheral blood mononuclear cell; PGE2, prostaglandin E2; TAA, tumor-associated antigen; TNF-a, tumor necrosis factor a.
presence of GM-CSF and IL-4, producing DCs with an immature phenotype that are commonly referred as monocyte-derived DCs (MoDCs). When CD341 hematopoietic precursors are used, the patients are frequently treated with GM-CSF before leukapheresis to mobilize precursors from the bone marrow. The harvested CD341 cells are then cultured with GM-CSF, TNF-a, Flt3L, TGF-b, and stem cell factor for 11– 12 days. This procedure leads to a heterogeneous mixture of APCs, namely MoDCs, LC-like cells, and other myeloid cells.75 Most clinical trials with a stated DC source use MoDCs (Fig 2, B). The preference for MoDCs instead of CD341-derived DCs is not related to their superior clinical efficacy, but rather to the limited number of CD341 precursors that can be isolated from apheresis products. In fact, CD341-derived DCs, and particularly their LC subset, were shown to stimulate in vitro more effective CTL responses than their counterpart MoDCs independent of the presence
of IL-12p70.75 Moreover, the efficacy of peptideloaded CD341-derived LCs and equivalent MoDCs was directly compared in stage III/IV melanoma patients (NCT00700167), and the results indicate that without exogenous IL-15 administration, the LCbased vaccines stimulate significantly more IFN-g–producing CTLs.76 Sipuleucel-T (Provange), the first FDA-approved cell-based antitumor vaccine, uses a different protocol to obtain DCs. Autologous PBMCs were incubated for 36–48 hours with a recombinant fusion protein consisting of human prostatic acid phosphatase and GM-CSF. GM-CSF targets the prostatic acid phosphatase antigen to APCs, namely DCs, promoting its uptake/processing and simultaneously inducing cellular maturation. The Q20 biologically active components of the vaccine are the CD541-matured APCs.6 Finally, among the clinical trials analyzed, only one uses ex vivo manipulated naturally occurring DCs (NCT01690377) (Fig 2, B). The
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:06 pm ce
809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862
Translational Research Volume -, Number -
9
print & web 4C=FPO
863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916
Constantino et al
Fig 2. Manipulation strategies and types of DCs used in antitumor vaccines. (A) Clinical trials with approaches directly exploring DCs: manipulation ex vivo followed by reinjection and in vivo DC targeting. (B) Frequencies distribution of clinical trials by DC genetic source (autologous or allogenic) and DC type. ‘‘Natural DCs’’ refer to primary cells directly isolated from the donor. ‘‘Not defined’’ indicates that the information regarding the parameter was not found in the clinical trial description or in the literature. DCs, dendritic cells; LCs, Langerhans cells; MoDCs, monocyte-derived dendritic cells.
authors77 of a study evaluated the efficacy of intranodal (i.n.) injection of tumor peptide–loaded pDCs in patients with metastatic melanoma. The results indicate that despite the limited number of pDCs administered, several patients mounted antivaccine CD41 and CD81 T-cell responses, demonstrating the feasibility of the approach. Selection of antigens and loading procedures. The choice of tumor antigens and the loading procedures are important parameters for DC-based vaccine production.78 A limited fraction (approximately 10%) of the tumor-associated antigens (TAAs) appear to be immunogenic, and among these, only a few are effectively associated with tumor rejection.79 TAAs are normally
unique mutated proteins, antigens derived from oncogenic viruses, or shared nonmutated self-antigens, such as tyrosinase, tyrosinase-related proteins, gp100, or MART-1.80 Q21 DCs have been pulsed with isolated/recombinant TAAs (full-length proteins or peptides), transfected with tumor messenger RNA (mRNAs), transduced Q22 with TAA-coding genes, and loaded with tumor cell lysates or apoptotic tumor cells (Fig 3, A). All these procedures were shown to elicit protective and therapeutic anticancer immune responses without inducing specific toxicity.81 Although pulsing DCs with short peptides lead to its direct loading onto MHC molecules at the cell surface, proteins and tumor lysates require
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:06 pm ce
917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970
971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024
Translational Research - 2015
Constantino et al
print & web 4C=FPO
10
Fig 3. Antigen loading procedures and routes of administration of ex vivo manipulated DCs. (A) Frequencies distribution of clinical trials by DC antigen loading strategies. (B) Administration routes for ex vivo manipulated DCs. In some cases, the same trial compares the efficiency of more than one administration route leading to count numbers (322) superior to the total number of respective trials (303). Not defined indicates that the information regarding the parameter was not found in the clinical trial description or in the literature. DCs, dendritic cells; i.d., intradermal; i.l., intralymphatical; i.n., intranodal; i.t., intratumoral; i.v., intravenous; s.c., subcutaneous.
Q23
internalization, processing, and presentation. However, the use of defined peptides is limited to the low number of characterized TAAs and by the fact that some of them are HLA-A1 or HLA-A24 restricted, requiring therefore that the patient’s haplotype must be known and adequate. Regarding the loading of DCs with allogenic/autologous tumor cell lysates or apoptotic bodies, it is a strategy for long known to elicit antitumor immunity.82 Its major advantage relies on the use of whole tumor proteome, encompassing that way multiple TAAs. In terms of effectiveness, several studies indicate that DCs loaded with apoptotic tumor cells elicit stronger immune responses than lysate-loaded or RNApulsed DCs.83,84 The major drawback of using tumor lysates or apoptotic bodies as a source of antigens instead of defined peptides is the dependence on the availability of tumor cells. Moreover, immunologic responses are more complex to monitor, which make
the correlation between the interventions and the clinical outcomes difficult. Another common strategy is to transfect DCs with mRNA extracted from tumor cells or with in vitro synthesized mRNAs that encode particular TAAs. The ability of these loaded DCs to elicit strong antitumor CD41 and CD81 T-cell responses is well documented85-87 and is one of the most commonly used approaches in clinical trials (Fig 3, A). When compared with peptide loading, transfection with whole tumor mRNA avoids the limitation imposed by the use of known TAAs and matched HLA phenotypes. In addition, siRNAs targeting the immunoproteasome components (NCT00672542) or mRNA coding for maturation agents and costimulatory molecules (NCT01066390) could be introduced, enhancing the Q24 generation of TAA-derived peptides and eliminating the need for an additional DC maturation step. The latter
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078
Translational Research Volume -, Number -
1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132
Constantino et al
approach was evaluated in melanoma patients using a vaccine composed of DCs transfected with mRNA encoding the immunostimulatory proteins CD40L, CD70, and constitutively active TLR4 in combination with mRNAs’ encoding tyrosinase, MAGE-A3, MAGE-C2, and gp100 (NCT01066390). The results demonstrated that most patients developed functional TAA-specific CD81 and CD41 T-cell responses, which were detected both at the skin and systemically.88,89 Among the strategies to transfect DCs, electroporation has shown to be the most efficient method, temporarily increasing cell membrane permeability, which facilitates the entry of mRNA without the need for additional reagents.90 DCs have also frequently been genetically engineered to stably express TAAs, such as mucin 1, p53, tyrosinase, melan-A, and gp100.91-93 The use of bacteria or viral vectors to deliver the DNA to DCs is safe, and the possibility of introducing genes encoding cytokines or costimulatory molecules is very appealing.94 Furthermore, the vector may intrinsically activate DCs by triggering endosomal or cytoplasmic sensors, such as TLRs, RIG, and protein kinase RNA-activated receptors, bypassing the need of a separate maturation step.95 Finally, the ex vivo fusion of DCs with tumor cells represents another approach for antigen loading. These cell hybrids, known as ‘‘dendritomes,’’ were shown to induce antitumor immune responses in vivo96-98 and have been tested in multiple clinical trials for more than the past few decades (Fig 3, A). Notably, dendritomes were found to be less effective in eliciting CTL responses than DCs loaded with apoptotic tumor cells,99 an observation that was attributed to the reduced expression of costimulatory molecules and IL-12p70 by the cell hybrids.97 DC maturation protocols. The DC maturation status and cytokine profile are parameters of great importance for the effectiveness of DC-based immunotherapies. Numerous clinical trials have shown a clear superiority of mature DCs over their immature counterparts.57,100 Immature DCs have a reduced ability to migrate from the injection sites to the lymph nodes,100 and can cause the inhibition of CTL functions by inducing IL-10– producing antigen-specific Tregs.101 In addition to the maturation status, the DC cytokine and chemokine profiles, particularly the expression of IL-12p70 and CCR7, are of great relevance for vaccine effectiveness. The cytokine IL-12p70 plays a central role in CD81 T-cell expansion and CTL differentiation.36 Clinically, the higher levels of IL-12p70 produced by DCs used in vaccines for glioma and melanoma patients were found to positively correlate with a more favorable clinical outcome.102,103 The chemokine receptor CCR7 governs the migration of DCs from the
11
peripheral tissues to the draining lymph nodes under either homeostatic or inflammatory conditions. In the context of DC-based immunotherapies, the migration of DCs from the injection sites to the lymph nodes and the subsequent interaction with the T cells is a key feature for effective immunization. A growing body of evidence demonstrates that DCs expressing high levels of CCR7 elicit a more effective antigenspecific immune response in vivo, which lowers the required DC dosage.104,105 The appropriate DC maturation stimulus remains a matter of debate. On the basis of the literature, several maturation protocols have been tested, which consist of individual stimuli or cocktails that combine proinflammatory cytokines, CD40L, and TLR agonists. The maturation cocktail that was used most commonly across the clinical trials analyzed includes TNF-a, IL-1b, IL-6, and PGE2. The use of PGE2 is somehow paradoxical because although it enhances the DC migratory capacities by inducing CCR7 expression,106 it reduces IL-12p70 production.107 Protocols that combine exposure to the TLR3 ligand polyinosinic:polycytidylic acid and PGE2 were proposed to circumvent this problem and explore the exquisite capacity of TLR agonists to induce IL-12p70 expression.108 Another common DC maturation cocktail includes TNF-a, IL-1b, IFN-a, IFN-g and polyinosinic:polycytidylic acid. The resulting DCs, termed aDC1s, efficiently secrete IL-12p70 and were shown to elicit more potent CTL responses than the standard maturated DCs.102,109-111 From our analysis, aDC1-based vaccines were tested in at least 8 clinical trials (Fig 2, B). To increase their immunogenicity, DC vaccines are frequently administered along with adjuvants. The most frequently used adjuvants are GM-CSF, IL-2, IFN-a, and TLR agonists. GM-CSF strongly induces the proliferation of DC precursors and has a stronger chemotactic effect over DCs, promoting their recruitment and maturation. Interferon alpha enhances DC antigen cross-presentation and, therefore, promotes more effective CTL responses.112 Strong preclinical evidence has demonstrated that the cytokine IL-2 can improve DC vaccine efficacy.113 However, subsequent clinical trials have not shown superior antitumor immune responses in regimens combining DCs with IL-2.114 In fact, because of its stimulatory abilities toward Tregs and myeloid-derived suppressor cells (MDSCs), IL-2 can even negatively affect DC-based immunotherapies. In addition to their inclusion in maturation cocktails, TLR agonists are increasingly being used as adjuvants in DC vaccines. In the clinical trials analyzed, the most common are poly-ICLC, rintatolimod (TLR3 ligands), imiquimod (TLR7 ligand), resiquimod (TLR7/8 ligand), and DUK-CPG-001 (TLR9
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186
12
1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240
Q25
Translational Research - 2015
Constantino et al
ligand). Another important question in the maturation protocols is the duration of DC exposure to the stimulus. The contact of DCs with maturation signals (danger signals) in vivo is transient because the cells rapidly leave the site of contact (normally the peripheral tissues) and reach the lymph nodes in 2–4 hours.115 However, most protocols in the literature indicate 24–48 hours of exposure. These time periods may be too long, exhausting the ability of the DCs to produce IL-12p70 and, therefore, decreasing their ability to elicit protective Th1 and CTL responses.116 Overall, we can conclude that the diversity of the maturation procedures used in the production of DCbased vaccines hampers the assessment of the real effects of the maturation stimuli because it is not the only parameter that differs among studies. It would be of great interest to design clinical trials that compare the effectiveness of DCs that have been matured with different cocktails to pursue a certain level of standardization. Routes of administration and cell doses. The route of administration, frequency of injections, and the number of cells injected are relevant aspects to consider in DC-based vaccinations, given that the efficacy of the intervention will strongly depend on the migration of sufficient numbers of DCs to T-cell areas in secondary lymphoid tissues. For more than the last 2 decades of clinical trials, DCs have been administered via different routes: intravenous (i.v.), intradermal (i.d.), s.c., i.n., and directly into the tumor (Fig 3, B). All these routes of administration elicit specific antitumor immune responses, but with different characteristics and efficiencies.116 The i.v. injected DCs are transiently detected in the lungs and then preferentially accumulate in the liver and spleen, with little distribution to the lymph nodes. In a clinical trial with prostate cancer patients, i.v. administration was shown to induce antigen-specific humoral responses more efficiently than the i.d. or i.n. routes.117 Regarding i.d. and s.c. routes, most DCs remain at the injection sites and are cleared by infiltrating macrophages.118 However, the 1%–5% of cells that actually reach the peripheral lymph nodes were shown to be sufficient to induce specific antitumor T cells.117,118 Furthermore, several lines of evidence suggest that the DCs retained at the injection site may act as adjuvants. They recruit, activate, and transfer antigens to resident DCs, namely dermal DCs and LCs, that then more effectively activate CD81 T cells.119 For these reasons and the fact that they are technically less exigent and less expensive, the s.c. and i.d. routes are used most commonly in trials for solid tumor treatments (Fig 3, B). Administration via i.n. route is performed under ultrasound guidance of the needle by an experienced radiologist. The
efficacy of the procedure heavily depends on the correct delivery and can, therefore, present great variability between patients. Surprisingly, although substantially more DCs migrate to the T-cell areas in the lymph nodes on correct i.n. vaccination, the antitumor T-cell responses are comparable or inferior to i.d. administration.118,120 Another route used in a few phase I/II clinical trials is intratumoral administration (Fig 3, B). The approach is technically exigent and associated with a high rate of intervention-associated morbidity. It has been tested in the treatment of several solid tumors, such as melanoma, breast, liver, and pancreatic cancers.121-123 An overall evaluation of the published studies demonstrates that the procedure is safe and could elicit antitumor immunity in at least some of the patients. Recently, using autochthonous prostate cancer animal models, Higham et al124 showed that one advantage of intratumor DC administration relies on the capacity of these cells to delay and reverse the tolerization of tumor-infiltrating effector T cells. As shown in Fig 3, B, currently, there is an increased tendency to administer DC vaccines via multiple routes, such as i.d. 1 s.c. or i.d. 1 i.v., with the aim of inducing more robust and broader immune responses. The minimal number of DCs required to induce an efficient immune response in humans has not yet been established. This is reflected in clinical trials by a plethora of DC doses and administration regimens. Doses ranging from as low as 0.3 3 106 naturally occurring pDCs injected via i.n. route every 2 weeks (NCT01690377) to 3 doses of 200 3 106 MoDCs administered via s.c. route (NCT00704938) can be found among analyzed trials. Verdijk et al determined that 0.5 3 106 DCs reaching the T-cell areas in the lymph nodes could be sufficient to induce de novo immune responses. Considering that a maximum of 4% of DCs administered via i.d/s.c. route migrate and only 80% remain viable after injection, the authors estimated that a minimal dose would consist of 15 3 106 cells.118 Experiments performed in lung cancer and neuroblastoma animal models showed that efficacy of DC vaccines is positively correlated with the dose used. Moreover, in one of the early clinical trials testing sipuleucel-T, the median time to disease progression was found to be 31.7 weeks for patients who received more than 100 3 106 cells per infusion compared with 12.1 weeks for patients who received fewer cells.125 However, the correlation between the number of administered DCs and vaccine effectiveness may vary with the selected administration route. Surprisingly, reducing the number of DCs injected via i.d. route was recently shown to improve their homing to the lymph nodes and, consequently, to potentially enhance vaccine efficacy.126,127 The authors suggested that
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294
Translational Research Volume -, Number -
1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343 1344 1345 1346 1347 1348
Q26
Constantino et al
multiple i.d. injections with small amounts of DCs would be a preferable strategy. In vivo DC targeting. Targeting antigens to DCs in vivo represents an enormous breakthrough for antitumor immunotherapies because it allows researchers to bypass the expensive and laborious ex vivo DC generation process.128 Antigens can be unambiguously targeted to different DC subsets, eliciting antigen-specific CD41 and CD81 T cell-mediated responses (reviewed in Caminschi et al129). The procedure involves the coupling of antigens to monoclonal antibodies (mAbs) that are specific for DC surface molecules, such as Fc receptors, CD40 and C-type lectin receptors, or the use lentiviral vectors with preferential tropism to DCs. Notably, the delivery of antigens via mAb induces tolerance in the absence of adjuvants, and requires the coadministration of DC maturation agents such as TLR3, TLR7/8, or CD40 agonists.130 On the basis of animal experiments, the delivery of antigens to DEC205, CD40, DC-SIGN, DCIR, mannose receptor, Cleac9A, and XCR1 showed promising results in inducing antitumor immunity (CD41 and CD81 T-cell responses, as well as humoral responses).129,131,132 The clinical application of the approach has been slow and has faced significant challenges. Several important factors must be considered in the development of human DC-targeting vaccines, including the biological functions of the targeted DCs, the pattern of expression and functions of the chosen receptors, and the adjuvants coadministered. Among the few in vivo DC-targeting therapies that are being tested in humans, DEC-205 is the most common targeted receptor (Fig 2, A). These vaccines consist of s.c. and i.d. administration of the cancer antigen NYESO-1 fused to a human anti–DEC-205 mAb in combination with the adjuvant poly-ICLC (NCT009 48961), decitabine (NCT01834248), the indoleamine2,3-dioxygenase (IDO) inhibitor INCB024360 (NCT0 2166905), or recombinant Flt3L (NCT02129075). The targeting of human chorionic gonadotropin beta protein to DCs via the mannose receptor is also being clinically evaluated for the treatment of breast, colorectal, pancreatic, bladder, and ovarian cancers. The fusion protein is injected via i.d. and s.c. routes in combination with adjuvants, such as GM-CSF and poly-ICLC (NCT0070 9462), or administered via i.v. route (NCT00648102). Using a different approach, a lentiviral vector encoding NY-ESO-1 and pseudotyped with Sindbis virus envelope proteins that are modified to target DC-SIGN on DCs (IDLV305) is under evaluation for the treatment of NY-ESO-1–positive solid tumors (NCT02122861). Although the clinical application of these nextgeneration DC-based vaccines is still in the early stages and, therefore, has several challenges that still have to
13
be overcome, it represents an exciting and promising new field in antitumor immunotherapy and overall immune modulation. Other DC-based approaches. Apart from ex vivo manipulation and direct in vivo targeting, several other approaches exploit the immunogenic potential of DCs in cancer therapy. These include nontargeted antigenbased vaccines, GM-CSF–secreting tumor cell vaccines, implantable DC-recruiting/activating scaffolds, and DCderived exosomes (Dex). Q27 The nontargeted antigen-based vaccines are composed of peptides, proteins, or tumor nucleic acids. When injected, these antigens are captured and processed by the APCs, namely DCs, eliciting an antigen-specific immune response.131 Initial studies showed that this approach can induce CD41 and CD81 T-cell responses, although with low or no impact on the clinical course of the disease.133 The lack of clinical benefits in these cases was associated to the dominant polarization of CD41 T cells into IL-4– and IL-5–producing Th2 subsets and to the differentiation of tumor-specific Tregs. To overcome these immunosuppressive events and to potentiate antigen capture by the DCs, vaccines have begun to be administered in combination with GM-CSF, adjuvants, and low doses of lymphodepleting agents, such as cyclophosphamide.134,135 In phase II trials, vaccines composed of the MAGE-A3–recombinant protein and the AS15 adjuvant system were shown to cause clinical improvements in both metastatic melanoma (NCT00086866) and resected non-small cell lung cancer (NCT00290355). For the GM-CSF–secreting tumor cell vaccines, irradiated autologous tumor cells or allogenic tumor cell lines have been engineered to secrete GM-CSF and were then injected into patients. These engineered tumor cells strongly attract macrophages, granulocytes, T cells, and DCs, potentiating tumor antigen presentation.136 The clinical evaluation of this vaccination strategy revealed a consistent induction of cellular and humoral antitumor responses in melanoma,137 prostate cancer,138 and pancreatic cancer.139 As a major drawback, several animal models have shown that prolonged GM-CSF production by tumor cells may lead to disease progression as a result of immune tolerance caused by the recruitment of myeloid suppressor cells and the differentiation of tolerogenic DCs.140 A new approach for recruiting endogenous DCs to a source of tumor antigens recently garnered particular interest. Mooney et al141 showed that biocompatible polymers may be designed to incorporate and release a DC chemotactic agent, an adjuvant, and tumor antigens in a controlled manner. The authors tested the implantation of polylactide-co-glycolide scaffolds incorporating GM-CSF, various TLR agonists, and tumor lysates in mice in
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399 1400 1401 1402
14
1403 1404 1405 1406 1407 1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434 1435 1436 1437 1438 1439 1440 1441 1442 1443 1444 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456
Q28
Q29
Translational Research - 2015
Constantino et al
the context of preventive and therapeutic melanoma vaccination. The obtained results demonstrated that the mice receiving the scaffold mounted specific and protective antitumor immunity with an OS of 90% for the preventive vaccine and 33% long-term survival for the therapeutic vaccine.141,142 Using the same rationale, Wang et al143 developed a 2-step strategy to modulate the endogenous DCs in situ. In the first step, the DCs were recruited to an injectable GM-CSF– releasing thermosensitive mPEG2poly(lactic-co-glycolic acid) hydrogel. Then, the recruited DCs were loaded with cancer antigens through the use of viral or nonviral vectors. The strategy was shown to generate robust tumor-specific immune responses in both prophylactic and therapeutic models of murine melanoma.143 These encouraging results in animal models led to the clinical application of the strategy. Currently, an ongoing clinical trial is evaluating the feasibility of this approach for treatment of melanoma (NCT01753089). The matrix of the DC-activating scaffold is composed of poly(lactic-co-glycolic acid) and incorporates an autologous tumor cell lysate, GM-CSF, and CpG oligodeoxynucleotides. This strategy may represent the future of DC-based immunotherapies, given that it overcomes the survival and homing problems of the injected ex vivo–generated DCs and even surpasses the limitation of single antigen immunization observed in the in vivo DC–targeting approach. Dex have received great attention as potential immunotherapeutic agents because of the pioneer studies of Zitvogel et al,144 who showed that they inhibit tumor growth in an MHC- and CD81 T cell-dependent manner. The capacity of Dex to elicit CD81 and CD41 T-cell responses was attributed to the transfer of antigen–MHC-I and MHC-II complexes to the endogenous DCs.145 Similar to DCs, Dex were found to modulate NK cells, promoting their proliferation and activation in IL-15Ra–dependent and NKG2Dmediated processes, respectively.146 Recently, the activity of Dex against tumors was additionally shown to rely on B-cell–mediated mechanisms147 and on their capacity to enhance the immunogenicity of tumor cells.148 Despite the abundant experimental data on the use of Dex for antitumor immunotherapy, their clinical evaluation remains scarce (Fig 2, B). Results from 2 clinical trials on melanoma and non-small cell lung cancer patients demonstrated that Dex could be safely administered, nevertheless inducing only limited CTLs responses.149,150 Finally, although all clinical trials performed to date test antitumor DC-based vaccines in a therapeutic context, numerous preclinical experiments showed that preventive DC vaccination can significantly delay or even prevent the development of several tu-
mors.151,152 This supports the establishment of clinical trials that evaluate the safety and efficacy of DC-based vaccines for cancer prevention in high-risk groups in the near future. LESSONS FROM THE PAST AND FUTURE PERSPECTIVES
It is now clear that the effectiveness of antitumor DCbased vaccines depends on the ability of the DCs to induce antigen-specific CTLs and Th1 cells as well as NK cell cross talk and activation.51,131 These features must be achieved by selecting or targeting the adequate DC subset and by tailoring its maturation status and cytokine/chemokine profile. In accordance Q30 with this notion and based on their remarkable capacity to cross-present antigens, LC-like DCs derived from CD341 progenitors (NCT00700167 and NCT0 1456104) or CD141 monocytes (NCT01189383) are being evaluated in clinical trials for the treatment of melanoma. Similarly, the clinical effectiveness of other DC subsets, such as pDCs and Th1 polarizing-DCs, is under assessment in melanoma (NCT00390338), prostate cancer (NCT00970203), and glioma patients (NCT00766753). The recent characterization of a novel human DC subset (CD11c1CD1411XCR11) with superior antigen cross-presentation and NK cell activation capacities has excited great enthusiasm on its possible use.12,14 However, CD11c1CD1411XCR11 DCs represent less than 0.1% of PMBCs, a major drawback for their clinical application. In fact, a relevant obstacle for the progress of DC-based vaccines has been the lack of sufficient DC sources other than CD141 MoDCs. To overcome this issue, the emerging field of induced pluripotent stem cells (iPSCs) may represent a significant opportunity.153 Recently, Silk et al154 established a protocol for the differentiation of CD11c1CD1411XCR11 DCs by culturing human iPSCs with GM-CSF, stem cell factor, VEGF, and bone morphogenetic protein 4. These iPSC-derived DCs were shown to efficiently cross-present exogenously supplied peptides to naive CD81 T cells, inducing their expansion and activation. Because the iPSCs may be continuously expanded in vitro, they represent an unlimited source of autologous DCs that can be tailored to the desired phenotype, bypassing problems such as limited cell numbers and patient-topatient variability.153 Another fact that became evident is that the DC vaccine-induced antitumor immune effector cells must overcome multiple immunoescape/immunosuppressive mechanisms (reviewed in Topalian et al155). Solid tu- Q31 mors often generate an immunosuppressive microenvironment by producing soluble mediators such as adenosine, IDO, PGE2, TGF-b, and VEGF. These
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 1470 1471 1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482 1483 1484 1485 1486 1487 1488 1489 1490 1491 1492 1493 1494 1495 1496 1497 1498 1499 1500 1501 1502 1503 1504 1505 1506 1507 1508 1509 1510
Translational Research Volume -, Number -
15
print & web 4C=FPO
1511 1512 1513 1514 1515 1516 1517 1518 1519 1520 1521 1522 1523 1524 1525 1526 1527 1528 1529 1530 1531 1532 1533 1534 1535 1536 1537 1538 1539 1540 1541 1542 1543 1544 1545 1546 1547 1548 1549 1550 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560 1561 1562 1563 1564
Constantino et al
Fig 4. Frequency of therapeutic approaches combining DC-based vaccines and other anticancer treatments. (A) DC-based vaccines are increasingly being tested in combination with other treatments such as conventional chemotherapy, radiotherapy, and other immunotherapies. (B) Frequencies distribution of other therapies being tested with DC-based vaccines. Other cell therapies include stem cell transplantation, transgenic CTLs, ex vivo expanded T lymphocytes, and cytokine-induced killer cells. *In some cases, in the same trial, DC-based vaccines are tested in combination with multiple therapies leading to count numbers (149) superior to the total number of respective trials (119). DCs, dendritic cells; CTL, cytotoxic T cells; CTLA-4, cytotoxic T-lymphocyte–associated protein 4; IDO, indoleamine-2,3-dioxygenase; mAb, monoclonal antibody; PD-1, programmed cell death 1; VEGF, vascular endothelial growth factor.
mediators inhibit the proliferation and differentiation of Th1 cells, suppress the activity of CTLs and DCs, and induce the differentiation of Tregs.156-158 Moreover, the tumor microenvironment is also prone to attract immunosuppressive cellular regulators such as circulating Tregs, MDSCs, and tumor-associated macrophages. Finally, immune checkpoints, an inhibitory network that normally maintains self-tolerance and prevents excessive and uncontrolled immune responses, can be co-opted by tumors to evade immune destruction.159,160 The major immune checkpoints that are dysregulated by tumors rely on ligand-receptor interactions, such as CD28–CTLA-4 and PD-1–PD-L1. There was a significant effort to establish strategies to bypass these barriers, and combination therapies emerged as a logical approach to potentiate DC vaccine effectiveness. The analysis of clinical trials for more than the last 2 decades reflects a clear tendency to adopt combination therapies instead of standalone DC vaccination (Fig 4, A). Some of the most common approaches involve the use of general lymphodepleting chemotherapeutic agents such as temozolomide and cyclophosphamide and fludarabine (Fig 4, B). By depleting the immune cells, these drugs eliminate negative regulators such Tregs and MDSCs, promoting a favorable environment for DC-induced expansion of antitumor effector cells in the recovery phase. In a pilot clinical trial, Ridolfi et al161 showed that the administration of low doses of temozolomide to melanoma patients before DC vaccination specifically reduced the CD41CD251Foxp31 Tregs. In turn, cyclophosphamide was shown to enhance antitumor immunity through
mechanisms that rely on Treg elimination and resetting DC homeostasis.162 The combination of DC vaccines with agents that preferentially depletes Tregs by directly targeting their abundantly expressed IL-2 receptor a chain (CD25) has also been tested. The agents most commonly used for this purpose are the anti-CD25 mAb basiliximab (NCT00626483) and daclizumab (NCT00847106) or denileukin diftitox (Ontak), a re- Q32 combinant IL-2 and diphtheria toxin fusion protein (NCT00703105, NCT00056134, and NCT00128622). Although some studies indicate that these combination therapies significantly increase DC vaccine efficacy as a result of increased antitumor-specific CTLs,163,164 others demonstrate that they induce tolerogenic DCs and promote the survival of resting Tregs.165 These paradoxical effects are partially because of CD25 expression not restricted to Tregs. As CD25 is also ex- Q33 pressed on effector T cells and activated NK cells, its blockade may compromise their antitumor activity. Another strategy for enhancing antitumor immunity consists of the combination of DC vaccines with immune checkpoint inhibitors. This approach is being tested in clinical experiments targeting the immune checkpoint receptors PD-1 (NCT01441765, NCT010 67287, NCT01096602, and NCT01753089) and CT LA-4 (NCT00090896 and NCT01753089) or the tryptophan degradation enzyme IDO (NCT01042535). Preliminary data obtained in patients with metastatic melanoma indicate that DC vaccines in combination with the anti–CTLA-4 mAb tremelimumab could be more effective than the same interventions in monotherapy (NCT00090896).166 Recently, combinations with
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
1565 1566 1567 1568 1569 1570 1571 1572 1573 1574 1575 1576 1577 1578 1579 1580 1581 1582 1583 1584 1585 1586 1587 1588 1589 1590 1591 1592 1593 1594 1595 1596 1597 1598 1599 1600 1601 1602 1603 1604 1605 1606 1607 1608 1609 1610 1611 1612 1613 1614 1615 1616 1617 1618
1619 1620 1621 1622 1623 1624 1625 1626 1627 1628 1629 1630 1631 1632 1633 1634 1635 1636 1637 1638 1639 1640 1641 1642 1643 1644 1645 1646 1647 1648 1649 1650 1651 1652 1653 1654 1655 1656 1657 1658 1659 1660 1661 1662 1663 1664 1665 1666 1667 1668 1669 1670 1671 1672
Translational Research - 2015
Constantino et al
web 4C=FPO
16
Fig 5. Evolution of antitumor DC-based immunotherapies. First approaches were based on the administration of ex vivo manipulated DCs in monotherapy regimen, relying only on the capacity of DCs to generate antitumor effector cells (Th1, CTLs, and NK cells). Currently, DC vaccines are frequently combined to therapies aiming to reduce tumor burden such as chemotherapy, radiotherapy, anti-VEGF mAbs, and to therapies seeking to overcome tumor-associated immunosuppression. The later include broad lymphodepleting agents (temozolomide and cyclophosphamide); preferential regulatory T cells depleting agents (anti-CD25 mAbs and denileukin diftitox); blockers of immunosuppressive molecules (IDO inhibitors, anti–CTLA-4 and anti–PD-1 mAbs). Promising approaches to be explored in following years are based on in vivo delivery of antigens to DCs and on the development of implantable devices that will recruit, load, and mature DCs. CTLs, cytotoxic T cells; DCs, dendritic cells; IDO, indoleamine-2,3-dioxygenase; iPSCs, induced pluripotent stem cells; mAb, monoclonal antibody; MoDCs, monocyte-derived dendritic cells; NK, natural killer; PBMCs, peripheral blood mononuclear cells; PD-1, programmed cell death 1; PLGA, poly(lactic-co-glycolic acid); TAAs, tumor-associated antigens; VEGF, vascular endothelial growth factor.
Q34
new anticancer drugs, such as the anti-VEGF mAb bevacizumab (Avastin) (NCT00683241, NCT00913913, and NCT02010606) and the tyrosine kinase inhibitors dasatinib and sunitinib (NCT01876212 and NCT00 678119), were also addressed. Moreover, by exploiting possible synergistic effects, DC-based vaccines have increasingly been used along with conventional radiotherapy and chemotherapy (NCT00639639, NCT01 973322, and NCT00617409). The positive effects of these therapeutic regimens are at least in part because of increased tumor cell apoptosis that fuels the DCs with relevant tumor antigens. From these 2 decades of clinical trials, it became evident that the classical objective responses, such as tumor shrinkage or tumor marker levels, are not adequate endpoints to evaluate the effectiveness of
DC-based antitumor immunotherapies. A systematic review conducted by Anguille et al4 on clinical trials evaluating DC vaccination in melanoma, prostate cancer, renal cell carcinoma, and glioma demonstrated that the clinical benefits in terms of the objective responses are real, but small. Objective responses were only observed in 8.5% of melanoma patients, 7.1% of prostate cancer patients, 11.5% of renal cell carcinoma patients, and 15.6% of glioma patients. In the same study, the authors found that although 2 trials on melanoma did not show survival benefits, all the other 38 studies demonstrated an increase in the median OS, ranging from 20% to 344%.4 These results reinforce the notion that survival is probably the most accurate endpoint to evaluate the therapeutic benefits of antitumor DC-based vaccines. However, clinical trials that
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
1673 1674 1675 1676 1677 1678 1679 1680 1681 1682 1683 1684 1685 1686 1687 1688 1689 1690 1691 1692 1693 1694 1695 1696 1697 1698 1699 1700 1701 1702 1703 1704 1705 1706 1707 1708 1709 1710 1711 1712 1713 1714 1715 1716 1717 1718 1719 1720 1721 1722 1723 1724 1725 1726
Translational Research Volume -, Number -
1727 1728 1729 1730 1731 1732 1733 1734 1735 1736 1737 1738 1739 1740 1741 1742 1743 1744 1745 1746 1747 1748 1749 1750 1751 1752 1753 1754 1755 1756 1757 1758 1759 1760 1761 1762 1763 1764 1765 1766 1767 1768 1769 1770 1771 1772 1773 1774 1775 1776 1777 1778 1779 1780
Constantino et al
monitor OS may be excessively long and costly.131 To surpass this limitation, alternative surrogate endpoints have been proposed. A growing number of studies are adopting immune response criteria, in which the effectiveness of the vaccination is evaluated by the expansion of antigen-specific effector T cells and NK cells.167 Finally, the selection of patients is also a relevant aspect to consider when designing an antitumor DC vaccination clinical trial. Optimally, patients should present a low tumor burden and be at an early stage of the disease, thus increasing the likelihood of developing effective antitumor immunity. CONCLUSIONS
Despite the variable rate of success in inducing clear beneficial outcomes, the first generation of antitumor DC-based vaccines has proven to be safe. Meanwhile, DC immunotherapies have substantially evolved because of our expanding knowledge of DC and tumor biology (Fig 5). The use of specific DC subsets that are tailored to a particular phenotype, the definition of adequate clinical endpoints and, particularly, the combination with other antitumor therapies have paved the way to the development of next-generation DC immunotherapies. Therefore, it is expected that these strategies may reveal the full therapeutic potential of DC-based vaccines for the treatment of malignant diseases in the future. UNCITED REFERENCE
19. ACKNOWLEDGMENTS
Q35
Conflict of Interests: All the authors have read the journal’s policy on the disclosure of potential conflicts of interest and have none to declare. The authors thank FCT/MEC for the financial support to the QOPNA Research Unit (FCT UID/QUI/00062/ 2013), the national funds, and where applicable the FEDER within the PT2020 Partnership Agreement, and also the Portuguese RNEM Network. CNC is funded by FEDER funds through the Operational Program Competitiveness Factors-COMPETE, and the national funds are funded by the FCT-Foundation for Science and Technology under strategic project UID/ NEU/04539/2013. All authors have read the journal’s authorship agreement. Supplementary Data
Supplementary data related to this article can be found at http://dx.doi.org/10.1016/j.trsl.2015.07.008.
17
REFERENCES
1. Steinman RM, Cohn ZA. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J Exp Med 1973;137:1142–62. Available at: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid5213 9237&tool5pmcentrez&rendertype5abstract. Accessed May 8, 2015. 2. Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature 2007;449:419–26. 3. Mukherji B, Chakraborty NG, Yamasaki S, et al. Induction of antigen-specific cytolytic T cells in situ in human melanoma by immunization with synthetic peptide-pulsed autologous antigen presenting cells. Proc Natl Acad Sci U S A 1995;92: 8078–82. Available at: http://www.pubmedcentral.nih.gov/ articlerender.fcgi?artid541290&tool5pmcentrez&rendertype5 abstract. Accessed May 8, 2015. 4. Anguille S, Smits EL, Lion E, van Tendeloo VF, Berneman ZN. Clinical use of dendritic cells for cancer therapy. Lancet Oncol 2014;15:e257–67. 5. Madan RA, Gulley JL, Fojo T, Dahut WL. Therapeutic cancer vaccines in prostate cancer: the paradox of improved survival without changes in time to progression. Oncologist 2010;15: 969–75. 6. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411–22. 7. Poltorak MP, Schraml BU. Fate mapping of dendritic cells. Front Immunol 2015;6:199. 8. Merad M, Sathe P, Helft J, Miller J, Mortha A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol 2013;31:563–604. 9. Naik SH. Demystifying the development of dendritic cell subtypes, a little. Immunol Cell Biol 2008;86:439–52. 10. Guilliams M, Ginhoux F, Jakubzick C, et al. Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol 2014;14:571–8. 11. Malissen B, Tamoutounour S, Henri S. The origins and functions of dendritic cells and macrophages in the skin. Nat Rev Immunol 2014;14:417–28. 12. Bachem A, G€uttler S, Hartung E, et al. Superior antigen crosspresentation and XCR1 expression define human CD11c1 CD1411 cells as homologues of mouse CD81 dendritic cells. J Exp Med 2010;207:1273–81. 13. Jongbloed SL, Kassianos AJ, McDonald KJ, et al. Human CD1411 (BDCA-3)1 dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J Exp Med 2010;207:1247–60. 14. Haniffa M, Shin A, Bigley V, et al. Human tissues contain CD141hi cross-presenting dendritic cells with functional homology to mouse CD1031 nonlymphoid dendritic cells. Immunity 2012;37:60–73. 15. Kanitakis J, Morelon E, Petruzzo P, Badet L, Dubernard J-M. Self-renewal capacity of human epidermal Langerhans cells: observations made on a composite tissue allograft. Exp Dermatol 2011;20:145–6. 16. Geijtenbeek TBH, Gringhuis SI. Signalling through C-type lectin receptors: shaping immune responses. Nat Rev Immunol 2009;9:465–79. 17. Van Vliet SJ, Garcıa-Vallejo JJ, van Kooyk Y. Dendritic cells and C-type lectin receptors: coupling innate to adaptive immune responses. Immunol Cell Biol 2008;86:580–7. 18. Schlitzer A, Ginhoux F. Organization of the mouse and human DC network. Curr Opin Immunol 2014;26:90–9.
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
Q36
1781 1782 1783 1784 1785 1786 1787 1788 1789 1790 1791 1792 1793 1794 1795 1796 1797 1798 1799 1800 1801 1802 1803 1804 1805 1806 1807 1808 1809 1810 1811 1812 1813 1814 1815 1816 1817 1818 1819 1820 1821 1822 1823 1824 1825 1826 1827 1828 1829 1830 1831 1832 1833 1834
18
1835 1836 1837 1838 1839 1840 1841 1842 1843 1844 1845 1846 1847 1848 1849 1850 1851 1852 1853 1854 1855 1856 1857 1858 1859 1860 1861 1862 1863 1864 1865 1866 1867 1868 1869 1870 1871 1872 1873 1874 1875 1876 1877 1878 1879 1880 1881 1882 1883 1884 1885 1886 1887 1888
Translational Research - 2015
Constantino et al
19. Dzionek A, Fuchs A, Schmidt P, et al. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood. J Immunol 2000;165:6037–46. 20. Hoeffel G, Ripoche A-C, Matheoud D, et al. Antigen crosspresentation by human plasmacytoid dendritic cells. Immunity 2007;27:481–92. 21. Vyas JM, Van der Veen AG, Ploegh HL. The known unknowns of antigen processing and presentation. Nat Rev Immunol 2008;8: 607–18. 22. Adams EJ. Lipid presentation by human CD1 molecules and the diverse T cell populations that respond to them. Curr Opin Immunol 2014;26:1–6. 23. Amigorena S, Savina A. Intracellular mechanisms of antigen cross presentation in dendritic cells. Curr Opin Immunol 2010; 22:109–17. 24. Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells. Annu Rev Immunol 2000;18:767–811. 25. Randolph GJ, Ochando J, Partida-Sanchez S. Migration of dendritic cell subsets and their precursors. Annu Rev Immunol 2008; 26:293–316. 26. Blanco P, Palucka AK, Pascual V, Banchereau J. Dendritic cells and cytokines in human inflammatory and autoimmune diseases. Cytokine Growth Factor Rev 2008;19:41–52. 27. Lukacs-Kornek V, Engel D, Tacke F, Kurts C. The role of chemokines and their receptors in dendritic cell biology. Front Biosci 2008;13:2238–52. Available at: http://www.ncbi.nlm.nih.gov/ pubmed/17981706. Accessed June 25, 2015. 28. Reis e Sousa C. Dendritic cells in a mature age. Nat Rev Immunol 2006;6:476–83. 29. Tan JKH, O’Neill HC. Maturation requirements for dendritic cells in T cell stimulation leading to tolerance versus immunity. J Leukoc Biol 2005;78:319–24. 30. Bour-Jordan H, Bluestone JA. Regulating the regulators: costimulatory signals control the homeostasis and function of regulatory T cells. Immunol Rev 2009;229:41–66. 31. Redmond WL, Ruby CE, Weinberg AD. The role of OX40mediated co-stimulation in T-cell activation and survival. Crit Rev Immunol 2009;29:187–201. Available at: http://www. pubmedcentral.nih.gov/articlerender.fcgi?artid53180959&tool 5pmcentrez&rendertype5abstract. Accessed June 25, 2015. 32. Hippen KL, Harker-Murray P, Porter SB, et al. Umbilical cord blood regulatory T-cell expansion and functional effects of tumor necrosis factor receptor family members OX40 and 4-1BB expressed on artificial antigen-presenting cells. Blood 2008; 112:2847–57. 33. Curtsinger JM, Johnson CM, Mescher MF. CD8 T cell clonal expansion and development of effector function require prolonged exposure to antigen, costimulation, and signal 3 cytokine. J Immunol 2003;171:5165–71. Available at: http://www.ncbi. nlm.nih.gov/pubmed/14607916. Accessed June 25, 2015. 34. Kaiko GE, Horvat JC, Beagley KW, Hansbro PM. Immunological decision-making: how does the immune system decide to mount a helper T-cell response? Immunology 2008;123: 326–38. 35. Kastenmuller W, Gasteiger G, Subramanian N, et al. Regulatory T cells selectively control CD81 T cell effector pool size via IL2 restriction. J Immunol 2011;187:3186–97. 36. Williams MA, Bevan MJ. Effector and memory CTL differentiation. Annu Rev Immunol 2007;25:171–92. 37. Pearce EL, Shen H. Generation of CD8 T cell memory is regulated by IL-12. J Immunol 2007;179:2074–81. Available at: http://www.ncbi.nlm.nih.gov/pubmed/17675465. Accessed May 17, 2015.
38. Josefowicz SZ, Lu L-F, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol 2012; 30:531–64. 39. Melief CJ, Kast WM. Cytotoxic T lymphocyte therapy of cancer and tumor escape mechanisms. Semin Cancer Biol 1991;2: 347–54. Available at: http://www.ncbi.nlm.nih.gov/pubmed/ 1773050. Accessed May 8, 2015. 40. Hariharan K, Braslawsky G, Black A, Raychaudhuri S, Hanna N. The induction of cytotoxic T cells and tumor regression by soluble antigen formulation. Cancer Res 1995;55:3486–9. Available at: http://www.ncbi.nlm.nih.gov/pubmed/7627951. Accessed May 8, 2015. 41. Appay V, Douek DC, Price DA. CD81 T cell efficacy in vaccination and disease. Nat Med 2008;14:623–8. 42. Zhang N, Bevan MJ. CD8(1) T cells: foot soldiers of the immune system. Immunity 2011;35:161–8. 43. Quezada SA, Peggs KS. Exploiting CTLA-4, PD-1 and PD-L1 to reactivate the host immune response against cancer. Br J Cancer 2013;108:1560–5. 44. Haabeth OAW, Tveita AA, Fauskanger M, et al. How Do CD4(1) T cells detect and eliminate tumor cells that either lack or express MHC class II molecules? Front Immunol 2014;5:174. 45. Spolski R, Leonard WJ. Interleukin-21: a double-edged sword with therapeutic potential. Nat Rev Drug Discov 2014;13: 379–95. 46. Antony PA, Piccirillo CA, Akpinarli A, et al. CD81 T cell immunity against a tumor/self-antigen is augmented by CD41 T helper cells and hindered by naturally occurring T regulatory cells. J Immunol 2005;174:2591–601. Available at: http:// www.pubmedcentral.nih.gov/articlerender.fcgi?artid51403291 &tool5pmcentrez&rendertype5abstract. Accessed May 8, 2015. 47. Sun JC, Bevan MJ. Defective CD8 T cell memory following acute infection without CD4 T cell help. Science 2003;300: 339–42. 48. Larsen SK, Gao Y, Basse PH. NK cells in the tumor microenvironment. Crit Rev Oncog 2014;19:91–105. Available at: http:// www.pubmedcentral.nih.gov/articlerender.fcgi?artid54062922 &tool5pmcentrez&rendertype5abstract. Accessed May 8, 2015. 49. Cheng M, Chen Y, Xiao W, Sun R, Tian Z. NK cell-based immunotherapy for malignant diseases. Cell Mol Immunol 2013;10: 230–52. 50. Hayakawa Y, Screpanti V, Yagita H, et al. NK cell TRAIL eliminates immature dendritic cells in vivo and limits dendritic cell vaccination efficacy. J Immunol 2004;172:123–9. Available at: http://www.ncbi.nlm.nih.gov/pubmed/14688317. Accessed May 8, 2015. 51. Pampena MB, Levy EM. Natural killer cells as helper cells in dendritic cell cancer vaccines. Front Immunol 2015;6:13. 52. Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol 2008;8:467–77. 53. Sasaki K, Pardee AD, Okada H, Storkus WJ. IL-4 inhibits VLA4 expression on Tc1 cells resulting in poor tumor infiltration and reduced therapy benefit. Eur J Immunol 2008;38:2865–73. 54. Chen M-L, Pittet MJ, Gorelik L, et al. Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity through TGFbeta signals in vivo. Proc Natl Acad Sci U S A 2005;102: 419–24. 55. Jarnicki AG, Lysaght J, Todryk S, Mills KHG. Suppression of antitumor immunity by IL-10 and TGF-beta-producing T cells infiltrating the growing tumor: influence of tumor environment on the induction of CD41 and CD81 regulatory T cells. J Immunol 2006;177:896–904. Available at: http://www.ncbi.nlm. nih.gov/pubmed/16818744. Accessed May 9, 2015.
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
1889 1890 1891 1892 1893 1894 1895 1896 1897 1898 1899 1900 1901 1902 1903 1904 1905 1906 1907 1908 1909 1910 1911 1912 1913 1914 1915 1916 1917 1918 1919 1920 1921 1922 1923 1924 1925 1926 1927 1928 1929 1930 1931 1932 1933 1934 1935 1936 1937 1938 1939 1940 1941 1942
Translational Research Volume -, Number -
1943 1944 1945 1946 1947 1948 1949 1950 1951 1952 1953 1954 1955 1956 1957 1958 1959 1960 1961 1962 1963 1964 1965 1966 1967 1968 1969 1970 1971 1972 1973 1974 1975 1976 1977 1978 1979 1980 1981 1982 1983 1984 1985 1986 1987 1988 1989 1990 1991 1992 1993 1994 1995 1996
Constantino et al
56. McNally A, Hill GR, Sparwasser T, Thomas R, Steptoe RJ. CD41CD251 regulatory T cells control CD81 T-cell effector differentiation by modulating IL-2 homeostasis. Proc Natl Acad Sci U S A 2011;108:7529–34. 57. Draube A, Klein-Gonzalez N, Mattheus S, et al. Dendritic cell based tumor vaccination in prostate and renal cell cancer: a systematic review and meta-analysis. PLoS One 2011;6:e18801. 58. Fernandez NC, Lozier A, Flament C, et al. Dendritic cells directly trigger NK cell functions: cross-talk relevant in innate anti-tumor immune responses in vivo. Nat Med 1999;5:405–11. 59. Lion E, Smits ELJM, Berneman ZN, Van Tendeloo VFI. NK cells: key to success of DC-based cancer vaccines? Oncologist 2012;17:1256–70. 60. Boudreau JE, Bridle BW, Stephenson KB, et al. Recombinant vesicular stomatitis virus transduction of dendritic cells enhances their ability to prime innate and adaptive antitumor immunity. Mol Ther 2009;17:1465–72. 61. Bouwer AL, Saunderson SC, Caldwell FJ, et al. NK cells are required for dendritic cell-based immunotherapy at the time of tumor challenge. J Immunol 2014;192:2514–21. 62. Makkouk A, Weiner GJ. Cancer immunotherapy and breaking immune tolerance: new approaches to an old challenge. Cancer Res 2015;75:5–10. 63. Aarntzen EHJG, De Vries IJM, Lesterhuis WJ, et al. Targeting CD4(1) T-helper cells improves the induction of antitumor responses in dendritic cell-based vaccination. Cancer Res 2013; 73:19–29. 64. Schreibelt G, Bol KF, Aarntzen EH, et al. Importance of helper T-cell activation in dendritic cell-based anticancer immunotherapy. Oncoimmunology 2013;2:e24440. 65. Kalinski P. Dendritic cells in immunotherapy of established cancer: roles of signals 1, 2, 3 and 4. Curr Opin Investig Drugs 2009; 10:526–35. Available at: http://www.pubmedcentral.nih.gov/ articlerender.fcgi?artid52919813&tool5pmcentrez&renderty pe5abstract. Accessed June 26, 2015. 66. Westermann J, K€orner IJ, Kopp J, et al. Cryopreservation of mature monocyte-derived human dendritic cells for vaccination: influence on phenotype and functional properties. Cancer Immunol Immunother 2003;52:194–8. 67. Fl€ orcken A, Kopp J, van Lessen A, et al. Allogeneic partially HLA-matched dendritic cells pulsed with autologous tumor cell lysate as a vaccine in metastatic renal cell cancer: a clinical phase I/II study. Hum Vaccin Immunother 2013;9: 1217–27. 68. Hus I, Roli nski J, Tabarkiewicz J, et al. Allogeneic dendritic cells pulsed with tumor lysates or apoptotic bodies as immunotherapy for patients with early-stage B-cell chronic lymphocytic leukemia. Leukemia 2005;19:1621–7. 69. Gabrilovich D. Mechanisms and functional significance of tumour-induced dendritic-cell defects. Nat Rev Immunol 2004; 4:941–52. 70. Pinzon-Charry A, Maxwell T, Lopez JA. Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol 2005;83:451–61. 71. Orsini E, Guarini A, Chiaretti S, Mauro FR, Foa R. The circulating dendritic cell compartment in patients with chronic lymphocytic leukemia is severely defective and unable to stimulate an effective T-cell response. Cancer Res 2003;63: 4497–506. Available at: http://www.ncbi.nlm.nih.gov/pubmed/ 12907623. Accessed May 9, 2015. 72. Della Bella S, Gennaro M, Vaccari M, et al. Altered maturation of peripheral blood dendritic cells in patients with breast cancer. Br J Cancer 2003;89:1463–72.
19
73. Toriyama K, Wen DR, Paul E, Cochran AJ. Variations in the distribution, frequency, and phenotype of Langerhans cells during the evolution of malignant melanoma of the skin. J Invest Dermatol 1993;100:269S–73S. Available at: http://www.ncbi.nlm. nih.gov/pubmed/7680054. Accessed June 26, 2015. 74. Troy AJ, Summers KL, Davidson PJ, Atkinson CH, Hart DN. Minimal recruitment and activation of dendritic cells within renal cell carcinoma. Clin Cancer Res 1998;4:585–93. Available at: http:// www.ncbi.nlm.nih.gov/pubmed/9533525. Accessed June 26, 2015. 75. Ratzinger G, Baggers J, de Cos MA, et al. Mature human Langerhans cells derived from CD341 hematopoietic progenitors stimulate greater cytolytic T lymphocyte activity in the absence of bioactive IL-12p70, by either single peptide presentation or cross-priming, than do dermal-interstitial or monoc. J Immunol 2004;173:2780–91. Available at: http://www.ncbi.nlm.nih.gov/ pubmed/15294997. Accessed May 9, 2015. 76. Romano E, Rossi M, Ratzinger G, et al. Peptide-loaded Langerhans cells, despite increased IL15 secretion and T-cell activation in vitro, elicit antitumor T-cell responses comparable to peptideloaded monocyte-derived dendritic cells in vivo. Clin Cancer Res 2011;17:1984–97. 77. Tel J, Aarntzen EHJG, Baba T, et al. Natural human plasmacytoid dendritic cells induce antigen-specific T-cell responses in melanoma patients. Cancer Res 2013;73:1063–75. 78. Schnurr M, Chen Q, Shin A, et al. Tumor antigen processing and presentation depend critically on dendritic cell type and the mode of antigen delivery. Blood 2005;105:2465–72. 79. Kroemer G, Zitvogel L. Can the exome and the immunome converge on the design of efficient cancer vaccines? Oncoimmunology 2012;1:579–80. 80. Seremet T, Brasseur F, Coulie PG. Tumor-specific antigens and immunologic adjuvants in cancer immunotherapy. Cancer J 2011;17:325–30. 81. Galluzzi L, Senovilla L, Vacchelli E, et al. Trial watch: dendritic cell-based interventions for cancer therapy. Oncoimmunology 2012;1:1111–34. 82. Albert ML, Pearce SF, Francisco LM, et al. Immature dendritic cells phagocytose apoptotic cells via alphavbeta5 and CD36, and cross-present antigens to cytotoxic T lymphocytes. J Exp Med 1998;188:1359–68. Available at: http://www.pubmedcentral. nih.gov/articlerender.fcgi?artid52212488&tool5pmcentrez& rendertype5abstract. Accessed May 9, 2015. 83. Kokhaei P, Choudhury A, Mahdian R, et al. Apoptotic tumor cells are superior to tumor cell lysate, and tumor cell RNA in induction of autologous T cell response in B-CLL. Leukemia 2004;18:1810–5. 84. Zappasodi R, Pupa SM, Ghedini GC, et al. Improved clinical outcome in indolent B-cell lymphoma patients vaccinated with autologous tumor cells experiencing immunogenic death. Cancer Res 2010;70:9062–72. 85. Boczkowski D, Nair SK, Snyder D, Gilboa E. Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J Exp Med 1996;184:465–72. Available at: http://www. pubmedcentral.nih.gov/articlerender.fcgi?artid52192710&tool5 pmcentrez&rendertype5abstract. Accessed May 9, 2015. 86. Bonehill A, Van Nuffel AMT, Corthals J, et al. Single-step antigen loading and activation of dendritic cells by mRNA electroporation for the purpose of therapeutic vaccination in melanoma patients. Clin Cancer Res 2009;15:3366–75. 87. Wilgenhof S, Van Nuffel AMT, Corthals J, et al. Therapeutic vaccination with an autologous mRNA electroporated dendritic cell vaccine in patients with advanced melanoma. J Immunother 2011;34:448–56.
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016 2017 2018 2019 2020 2021 2022 2023 2024 2025 2026 2027 2028 2029 2030 2031 2032 2033 2034 2035 2036 2037 2038 2039 2040 2041 2042 2043 2044 2045 2046 2047 2048 2049 2050
20
2051 2052 2053 2054 2055 2056 2057 2058 2059 2060 2061 2062 2063 2064 2065 2066 2067 2068 2069 2070 2071 2072 2073 2074 2075 2076 2077 2078 2079 2080 2081 2082 2083 2084 2085 2086 2087 2088 2089 2090 2091 2092 2093 2094 2095 2096 2097 2098 2099 2100 2101 2102 2103 2104
Translational Research - 2015
Constantino et al
88. Benteyn D, Van Nuffel AMT, Wilgenhof S, et al. Characterization of CD81 T-cell responses in the peripheral blood and skin injection sites of melanoma patients treated with mRNA electroporated autologous dendritic cells (TriMixDC-MEL). Biomed Res Int 2013;2013:976383. 89. Van Nuffel AMT, Benteyn D, Wilgenhof S, et al. Dendritic cells loaded with mRNA encoding full-length tumor antigens prime CD41 and CD81 T cells in melanoma patients. Mol Ther 2012;20:1063–74. 90. Gilboa E, Vieweg J. Cancer immunotherapy with mRNAtransfected dendritic cells. Immunol Rev 2004;199:251–63. 91. Di Nicola M, Carlo-Stella C, Mortarini R, et al. Boosting T cellmediated immunity to tyrosinase by vaccinia virus-transduced, CD34(1)-derived dendritic cell vaccination: a phase I trial in metastatic melanoma. Clin Cancer Res 2004;10:5381–90. 92. Steele JC, Rao A, Marsden JR, et al. Phase I/II trial of a dendritic cell vaccine transfected with DNA encoding melan A and gp100 for patients with metastatic melanoma. Gene Ther 2011;18: 584–93. 93. Butterfield LH, Comin-Anduix B, Vujanovic L, et al. Adenovirus MART-1-engineered autologous dendritic cell vaccine for metastatic melanoma. J Immunother 2008;31:294–309. 94. Humrich J, Jenne L. Viral vectors for dendritic cell-based immunotherapy. Curr Top Microbiol Immunol 2003;276:241–59. Available at: http://www.ncbi.nlm.nih.gov/pubmed/12797451. Accessed July 5, 2015. 95. Wang H, Zhang L, Kung SKP. Emerging applications of lentiviral vectors in dendritic cell-based immunotherapy. Immunotherapy 2010;2:685–95. 96. Wang J, Saffold S, Cao X, Krauss J, Chen W. Eliciting T cell immunity against poorly immunogenic tumors by immunization with dendritic cell-tumor fusion vaccines. J Immunol 1998; 161:5516–24. Available at: http://www.ncbi.nlm.nih.gov/ pubmed/9820528. Accessed May 9, 2015. 97. Vasir B, Wu Z, Crawford K, et al. Fusions of dendritic cells with breast carcinoma stimulate the expansion of regulatory T cells while concomitant exposure to IL-12, CpG oligodeoxynucleotides, and anti-CD3/CD28 promotes the expansion of activated tumor reactive cells. J Immunol 2008;181:808–21. Available at: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid5293 8172&tool5pmcentrez&rendertype5abstract. Accessed May 9, 2015. 98. Rosenblatt J, Glotzbecker B, Mills H, et al. PD-1 blockade by CT-011, anti-PD-1 antibody, enhances ex vivo T-cell responses to autologous dendritic cell/myeloma fusion vaccine. J Immunother 2011;34:409–18. 99. Kokhaei P, Rezvany MR, Virving L, et al. Dendritic cells loaded with apoptotic tumour cells induce a stronger T-cell response than dendritic cell-tumour hybrids in B-CLL. Leukemia 2003; 17:894–9. 100. De Vries IJM, Lesterhuis WJ, Scharenborg NM, et al. Maturation of dendritic cells is a prerequisite for inducing immune responses in advanced melanoma patients. Clin Cancer Res 2003;9:5091–100. Available at: http://www.ncbi.nlm.nih.gov/ pubmed/14613986. Accessed May 9, 2015. 101. Dhodapkar MV, Steinman RM, Krasovsky J, Munz C, Bhardwaj N. Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J Exp Med 2001;193:233–8. Available at: http://www.pubmedcentral. nih.gov/articlerender.fcgi?artid52193335&tool5pmcentrez& rendertype5abstract. Accessed May 9, 2015. 102. Okada H, Kalinski P, Ueda R, et al. Induction of CD81 T-cell responses against novel glioma-associated antigen peptides and clin-
103.
104.
105.
106.
107.
108.
109.
110.
111.
112.
113.
114.
115.
116.
117.
ical activity by vaccinations with {alpha}-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in. J Clin Oncol 2011;29:330–6. Carreno BM, Becker-Hapak M, Huang A, et al. IL-12p70producing patient DC vaccine elicits Tc1-polarized immunity. J Clin Invest 2013;123:3383–94. Okada N, Mori N, Koretomo R, et al. Augmentation of the migratory ability of DC-based vaccine into regional lymph nodes by efficient CCR7 gene transduction. Gene Ther 2005;12: 129–39. Gonzalez FE, Ortiz C, Reyes M, et al. Melanoma cell lysate induces CCR7 expression and in vivo migration to draining lymph nodes of therapeutic human dendritic cells. Immunology 2014; 142:396–405. Scandella E, Men Y, Gillessen S, F€orster R, Groettrup M. Prostaglandin E2 is a key factor for CCR7 surface expression and migration of monocyte-derived dendritic cells. Blood 2002; 100:1354–61. Jongmans W, Tiemessen DM, van Vlodrop IJH, Mulders PFA, Oosterwijk E. Th1-polarizing capacity of clinical-grade dendritic cells is triggered by Ribomunyl but is compromised by PGE2: the importance of maturation cocktails. J Immunother 2005;28:480–7. Available at: http://www.ncbi.nlm.nih.gov/ pubmed/16113604. Accessed May 9, 2015. Boullart ACI, Aarntzen EHJG, Verdijk P, et al. Maturation of monocyte-derived dendritic cells with Toll-like receptor 3 and 7/8 ligands combined with prostaglandin E2 results in high interleukin-12 production and cell migration. Cancer Immunol Immunother 2008;57:1589–97. Park M-H, Yang D-H, Kim M-H, et al. Alpha-type 1 polarized dendritic cells loaded with apoptotic allogeneic breast cancer cells can induce potent cytotoxic T lymphocytes against breast cancer. Cancer Res Treat 2011;43:56–66. Akiyama Y, Oshita C, Kume A, et al. a-Type-1 polarized dendritic cell-based vaccination in recurrent high-grade glioma: a phase I clinical trial. BMC Cancer 2012;12:623. Hansen M, Hjortø GM, Donia M, et al. Comparison of clinical grade type 1 polarized and standard matured dendritic cells for cancer immunotherapy. Vaccine 2013;31:639–46. Spadaro F, Lapenta C, Donati S, et al. IFN-a enhances crosspresentation in human dendritic cells by modulating antigen survival, endocytic routing, and processing. Blood 2012;119: 1407–17. Shimizu K, Fields RC, Giedlin M, Mule JJ. Systemic administration of interleukin 2 enhances the therapeutic efficacy of dendritic cell-based tumor vaccines. Proc Natl Acad Sci U S A 1999;96:2268–73. Available at: http://www.pubmedcentral.nih. gov/articlerender.fcgi?artid526772&tool5pmcentrez&render type5abstract. Accessed May 9, 2015. Redman BG, Chang AE, Whitfield J, et al. Phase Ib trial assessing autologous, tumor-pulsed dendritic cells as a vaccine administered with or without IL-2 in patients with metastatic melanoma. J Immunother 2008;31:591–8. Martın-Fontecha A, Lanzavecchia A, Sallusto F. Dendritic cell migration to peripheral lymph nodes. Handb Exp Pharmacol 2009;188:31–49. Simon T, Fonteneau J-F, Gregoire M. Dendritic cell preparation for immunotherapeutic interventions. Immunotherapy 2009;1: 289–302. Fong L, Brockstedt D, Benike C, Wu L, Engleman EG. Dendritic cells injected via different routes induce immunity in cancer patients. J Immunol 2001;166:4254–9. Available at: http://www. ncbi.nlm.nih.gov/pubmed/11238679. Accessed May 9, 2015.
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
Q38
2105 2106 2107 2108 2109 2110 2111 2112 2113 2114 2115 2116 2117 2118 2119 2120 2121 2122 2123 2124 2125 2126 2127 2128 2129 2130 2131 2132 2133 2134 2135 2136 2137 2138 2139 2140 2141 2142 2143 2144 2145 2146 2147 2148 2149 2150 2151 2152 2153 2154 2155 2156 2157 2158
Translational Research Volume -, Number -
2159 2160 2161 2162 2163 2164 2165 2166 2167 2168 2169 2170 2171 2172 2173 2174 2175 2176 2177 2178 2179 2180 2181 2182 2183 2184 2185 2186 2187 2188 2189 2190 2191 2192 2193 2194 2195 2196 2197 2198 2199 2200 2201 2202 2203 2204 2205 2206 2207 2208 2209 2210 2211 2212
Constantino et al
118. Verdijk P, Aarntzen EHJG, Lesterhuis WJ, et al. Limited amounts of dendritic cells migrate into the T-cell area of lymph nodes but have high immune activating potential in melanoma patients. Clin Cancer Res 2009;15:2531–40. 119. Yewdall AW, Drutman SB, Jinwala F, Bahjat KS, Bhardwaj N. CD81 T cell priming by dendritic cell vaccines requires antigen transfer to endogenous antigen presenting cells. PLoS One 2010; 5:e11144. 120. Lesterhuis WJ, de Vries IJM, Schreibelt G, et al. Route of administration modulates the induction of dendritic cell vaccineinduced antigen-specific T cells in advanced melanoma patients. Clin Cancer Res 2011;17:5725–35. 121. Endo H, Saito T, Kenjo A, et al. Phase I trial of preoperative intratumoral injection of immature dendritic cells and OK-432 for resectable pancreatic cancer patients. J Hepatobiliary Pancreat Sci 2012;19:465–75. 122. Guo J, Zhu J, Sheng X, et al. Intratumoral injection of dendritic cells in combination with local hyperthermia induces systemic antitumor effect in patients with advanced melanoma. Int J Cancer 2007;120:2418–25. 123. Chi K-H, Liu S-J, Li C-P, et al. Combination of conformal radiotherapy and intratumoral injection of adoptive dendritic cell immunotherapy in refractory hepatoma. J Immunother 2005; 28:129–35. Available at: http://www.ncbi.nlm.nih.gov/pubmed/ 15725956. Accessed May 9, 2015. 124. Higham EM, Shen C-H, Wittrup KD, Chen J. Cutting edge: delay and reversal of T cell tolerance by intratumoral injection of antigen-loaded dendritic cells in an autochthonous tumor model. J Immunol 2010;184:5954–8. 125. Small EJ, Fratesi P, Reese DM, et al. Immunotherapy of hormone-refractory prostate cancer with antigen-loaded dendritic cells. J Clin Oncol 2000;18:3894–903. Available at: http://www.ncbi.nlm.nih.gov/pubmed/11099318. Accessed May 9, 2015. 126. Aarntzen EHJG, Srinivas M, Bonetto F, et al. Targeting of 111Inlabeled dendritic cell human vaccines improved by reducing number of cells. Clin Cancer Res 2013;19:1525–33. 127. Aarntzen EH, Srinivas M, Schreibelt G, et al. Reducing cell number improves the homing of dendritic cells to lymph nodes upon intradermal vaccination. Oncoimmunology 2013;2:e24661. 128. Tacken PJ, Figdor CG. Targeted antigen delivery and activation of dendritic cells in vivo: steps towards cost effective vaccines. Semin Immunol 2011;23:12–20. 129. Caminschi I, Maraskovsky E, Heath WR. Targeting dendritic cells in vivo for cancer therapy. Front Immunol 2012;3:13. 130. Bonifaz L, Bonnyay D, Mahnke K, Rivera M, Nussenzweig MC, Steinman RM. Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD81 T cell tolerance. J Exp Med 2002; 196:1627–38. Available at: http://www.pubmedcentral.nih.gov/ articlerender.fcgi?artid52196060&tool5pmcentrez&renderty pe5abstract. Accessed May 6, 2015. 131. Palucka K, Banchereau J. Dendritic-cell-based therapeutic cancer vaccines. Immunity 2013;39:38–48. 132. Bonifaz LC, Bonnyay DP, Charalambous A, et al. In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T cell vaccination. J Exp Med 2004;199: 815–24. 133. Leffers N, Lambeck AJA, Gooden MJM, et al. Immunization with a P53 synthetic long peptide vaccine induces P53-specific immune responses in ovarian cancer patients, a phase II trial. Int J Cancer 2009;125:2104–13.
21
134. Walter S, Weinschenk T, Stenzl A, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med 2012; 18:1254–61. 135. Schuster SJ, Neelapu SS, Gause BL, et al. Vaccination with patient-specific tumor-derived antigen in first remission improves disease-free survival in follicular lymphoma. J Clin Oncol 2011;29:2787–94. 136. Gupta R, Emens LA. GM-CSF-secreting vaccines for solid tumors: moving forward. Discov Med 2010;10:52–60. Available at: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid5 3086372&tool5pmcentrez&rendertype5abstract. Accessed June 29, 2015. 137. Kusumoto M, Umeda S, Ikubo A, et al. Phase 1 clinical trial of irradiated autologous melanoma cells adenovirally transduced with human GM-CSF gene. Cancer Immunol Immunother 2001;50:373–81. Available at: http://www.ncbi.nlm.nih.gov/ pubmed/11676397. Accessed May 10, 2015. 138. Higano CS, Corman JM, Smith DC, et al. Phase 1/2 doseescalation study of a GM-CSF-secreting, allogeneic, cellular immunotherapy for metastatic hormone-refractory prostate cancer. Cancer 2008;113:975–84. 139. Laheru D, Lutz E, Burke J, et al. Allogeneic granulocyte macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activation. Clin Cancer Res 2008;14:1455–63. 140. Filipazzi P, Valenti R, Huber V, et al. Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colonystimulation factor-based antitumor vaccine. J Clin Oncol 2007; 25:2546–53. 141. Ali OA, Huebsch N, Cao L, Dranoff G, Mooney DJ. Infectionmimicking materials to program dendritic cells in situ. Nat Mater 2009;8:151–8. 142. Ali OA, Verbeke C, Johnson C, et al. Identification of immune factors regulating antitumor immunity using polymeric vaccines with multiple adjuvants. Cancer Res 2014;74:1670–81. 143. Liu Y, Xiao L, Joo K-I, Hu B, Fang J, Wang P. In situ modulation of dendritic cells by injectable thermosensitive hydrogels for cancer vaccines in mice. Biomacromolecules 2014;15: 3836–45. 144. Zitvogel L, Regnault A, Lozier A, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med 1998;4:594–600. Available at: http://www.ncbi.nlm.nih.gov/pubmed/9585234. Accessed May 10, 2015. 145. Andre F, Chaput N, Schartz NEC, et al. Exosomes as potent cellfree peptide-based vaccine. I. Dendritic cell-derived exosomes transfer functional MHC class I/peptide complexes to dendritic cells. J Immunol 2004;172:2126–36. Available at: http://www. ncbi.nlm.nih.gov/pubmed/14764678. Accessed March 20, 2015. 146. Viaud S, Terme M, Flament C, et al. Dendritic cell-derived exosomes promote natural killer cell activation and proliferation: a role for NKG2D ligands and IL-15Ralpha. PLoS One 2009;4: e4942. 147. N€aslund TI, Gehrmann U, Qazi KR, Karlsson MCI, Gabrielsson S. Dendritic cell-derived exosomes need to activate both T and B cells to induce antitumor immunity. J Immunol 2013;190:2712–9. 148. Romagnoli GG, Zelante BB, Toniolo PA, Migliori IK, Barbuto JAM. Dendritic cell-derived exosomes may be a tool for cancer immunotherapy by converting tumor cells into immunogenic targets. Front Immunol 2014;5:692.
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
2213 2214 2215 2216 2217 2218 2219 2220 2221 2222 2223 2224 2225 2226 2227 2228 2229 2230 2231 2232 2233 2234 2235 2236 2237 2238 2239 2240 2241 2242 2243 2244 2245 2246 2247 2248 2249 2250 2251 2252 2253 2254 2255 2256 2257 2258 2259 2260 2261 2262 2263 2264 2265 2266
22
2267 2268 2269 2270 2271 2272 2273 2274 2275 2276 2277 2278 2279 2280 2281 2282 2283 2284 2285 2286 2287 2288 2289 2290 2291 2292 2293 2294 2295 2296 2297 2298 2299 2300 2301 2302 2303 2304 2305 2306 2307 2308 2309 2310 2311 2312 2313 2314 2315 2316 2317 2318 2319 2320
Translational Research - 2015
Constantino et al
149. Escudier B, Dorval T, Chaput N, et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derivedexosomes: results of the first phase I clinical trial. J Transl Med 2005;3:10. 150. Morse MA, Garst J, Osada T, et al. A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. J Transl Med 2005;3:9. 151. Birkh€auser FD, Koya RC, Neufeld C, et al. Dendritic cell–based immunotherapy in prevention and treatment of renal cell carcinoma. J Immunother 2013;36:102–11. 152. Ahmed MS, Bae Y-S. Dendritic cell-based therapeutic cancer vaccines: past, present and future. Clin Exp Vaccine Res 2014; 3:113–6. 153. Sachamitr P, Hackett S, Fairchild PJ. Induced pluripotent stem cells: challenges and opportunities for cancer immunotherapy. Front Immunol 2014;5:176. 154. Silk KM, Silk JD, Ichiryu N, et al. Cross-presentation of tumour antigens by human induced pluripotent stem cell-derived CD141(1)XCR11 dendritic cells. Gene Ther 2012;19:1035–40. 155. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 2015;27:450–61. 156. Gabrilovich DI, Chen HL, Girgis KR, et al. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med 1996;2: 1096–103. Available at: http://www.ncbi.nlm.nih.gov/pubmed/ 8837607. Accessed June 30, 2015. 157. Pickup M, Novitskiy S, Moses HL. The roles of TGFb in the tumour microenvironment. Nat Rev Cancer 2013;13:788–99. 158. Prendergast GC, Smith C, Thomas S, et al. Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune
159. 160. 161.
162.
163.
164.
165.
166.
167.
escape in cancer. Cancer Immunol Immunother 2014;63: 721–35. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252–64. Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015;348:56–61. Ridolfi L, Petrini M, Granato AM, et al. Low-dose temozolomide before dendritic-cell vaccination reduces (specifically) CD41CD2511Foxp31 regulatory T-cells in advanced melanoma patients. J Transl Med 2013;11:135. Radojcic V, Bezak KB, Skarica M, et al. Cyclophosphamide resets dendritic cell homeostasis and enhances antitumor immunity through effects that extend beyond regulatory T cell elimination. Cancer Immunol Immunother 2010;59: 137–48. Dannull J, Su Z, Rizzieri D, et al. Enhancement of vaccinemediated antitumor immunity in cancer patients after depletion of regulatory T cells. J Clin Invest 2005;115:3623–33. Morse MA, Hobeika AC, Osada T, et al. Depletion of human regulatory T cells specifically enhances antigen-specific immune responses to cancer vaccines. Blood 2008;112:610–8. Baur AS, Lutz MB, Schierer S, et al. Denileukin diftitox (ONTAK) induces a tolerogenic phenotype in dendritic cells and stimulates survival of resting Treg. Blood 2013;122: 2185–94. Ribas A, Comin-Anduix B, Chmielowski B, et al. Dendritic cell vaccination combined with CTLA4 blockade in patients with metastatic melanoma. Clin Cancer Res 2009;15:6267–76. Wolchok JD, Hoos A, O’Day S, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res 2009;15:7412–20.
REV 5.4.0 DTD TRSL944_proof 19 August 2015 6:07 pm ce
2321 2322 2323 2324 2325 2326 2327 2328 2329 2330 2331 2332 2333 2334 2335 2336 2337 2338 2339 2340 2341 2342 2343 2344 2345 2346 2347 2348 2349 2350 2351 2352 2353 2354 2355 2356 2357 2358 2359 2360 2361 2362 2363 2364 2365 2366 2367 2368 2369 2370 2371 2372 2373 2374