1,515
Views
49
CrossRef citations to date
0
Altmetric
Author's View

Trial watch

Immunostimulatory cytokines in cancer therapy

, , , , , , , & show all
Article: e29030 | Received 25 Apr 2014, Accepted 26 Apr 2014, Published online: 03 Jun 2014

Abstract

Tumor-targeting immune responses provide a significant contribution to (when they do not entirely account for) the clinical activity of diverse antineoplastic regimens, encompassing not only a large panel of immunotherapeutic strategies but also conventional cytotoxic molecules, targeted anticancer agents and irradiation. In line with this notion, several approaches have been devised to elicit novel or boost existing anticancer immune responses, including the administration of immunomodulatory cytokines. Such a relatively unspecific intervention suffices to mediate clinical effects in (at least a subset of) patients bearing particularly immunogenic tumors, like melanoma and renal cell carcinoma. More often, however, immunostimulatory cytokines are administered to boost the immunogenic potential of other agents, including (but not limited to) immune checkpoint-blocking antibodies, anticancer vaccines, oncolytic viruses and immunogenic chemotherapeutics. Here, we summarize the latest advances in the clinical development of recombinant cytokines as an immunomodulatory intervention for cancer therapy.

Introduction

The word ‘cytokines’ is commonly employed to refer to a large and heterogeneous group of small and for the most part soluble (glyco)proteins that regulate—in an autocrine, paracrine or endocrine manner—virtually all biological functions, including (but not limited to) proliferative responses, differentiation, chemotaxis, inflammatory reactions, innate and adaptive immunity, and cell death.Citation1-Citation4 The cytokine family nowadays includes more than 140 distinct members, and this number is expected to grow as various cytokine-like molecules are discovered every year.Citation5-Citation7 Several attempts have been made throughout the past 3 decades to classify cytokines based on structural and/or functional considerations, leading to the introduction of relatively unspecific terms like ‘chemokines,’ referring to small cytokines involved in the regulation of chemotaxis, ‘interleukins,’ referring to cytokines that regulate the crosstalk between leukocytes, and ‘colony-stimulating factors,’ referring to cytokines that control hematopoiesis.Citation8-Citation10 Along with the realization of the astonishing pleiotropism of the cytokine system, however, such classifications turned out to be reductionist and relatively imprecise, and thus were abandoned.Citation5,Citation6 This said, terms including interleukins, chemokines and colony-stimulating factors are still largely employed by the scientific community, mainly for historical reasons.

Cytokine signaling is highly pleiotropic, at least in part because (1) virtually all cell types throughout the body produce (one or several) cytokines; (2) the same cytokine can signal via various receptors/receptor isoforms, which are generally characterized by differential binding affinity and expression patterns; (3) cytokines generally participate in signaling cascades that regulate the release of other biologically active molecules, including other cytokines; and (4) the activity of cytokines is heavily influenced by contextual parameters such as local concentration, cell type, receptor isoform and the presence of additional cytokines.Citation5,Citation6,Citation8,Citation9

Besides regulating homeostatic hematopoiesisCitation10 and participating in physiological angiogenesisCitation11,Citation12 cytokines are released in response to a wide array of insults, such as traumatic events, infections, and cancer.Citation13-Citation15 In these settings, cytokines are secreted in discrete waves to coordinate (1) the removal of the pathogenic stimulus, and (2) the re-establishment of tissue homeostasis.Citation16-Citation18 When such an adaptive response fails and the initiating stimulus cannot be removed, however, the continuous secretion of specific cytokines may promote chronic inflammation. Of note, patients affected by a chronic inflammatory response, be it systemic or local, exhibit an increased propensity to develop some neoplasms, including colorectal carcinoma.Citation19-Citation21 Most likely, this originates from the increased production of mutagenic reactive oxygen species at sites of inflammation, as well as from the local secretion of mitogenic cytokines. Altogether, these observations lend further support to the notion that the biological activity of several cytokines exhibits a high degree of context dependency.

As soon as it became clear that tumors do not go unnoticed by the immune system, several approaches have been developed to elicit novel (or reinstate pre-existent) tumor-targeting immune responses. These therapeutic strategies include highly specific interventions, such as dendritic cell (DC)-based, peptide-based and DNA-based anticancer vaccines,Citation22-Citation24 as well as relatively non-specific maneuvers, such as the local or systemic administration of Toll-like receptor (TLR) agonists,Citation25,Citation26 immunostimulatory antibodies,Citation27 or cytokines.Citation5,Citation6 In spite of the fact that cytokines do not actively elicit a tumor-targeting immune response but rather boost the antineoplastic potential of natural, tumor-specific immune effectors, no less than three recombinant cytokines are currently approved by the US Food and Drug Administration (FDA) or equivalent regulatory agencies for use as standalone therapeutic interventions in adult cancer patients. First, interferon (IFN-)α2a (Roferon-A®) is used in subjects with hairy cell leukemia and chronic phase, Philadelphia chromosome-positive chronic myelogenous leukemia (CML), upon minimal pretreatment (within 1 y of diagnosis). Second, IFN-α2b (Intron A®) is employed for the therapy of hairy cell leukemia, AIDS-related Kaposi's sarcoma, follicular lymphoma, multiple myeloma, melanoma, condyloma acuminata and cervical intraepithelial neoplasms. Third, interleukin (IL)-2 (aldesleukin, Proleukin®) is approved for the treatment of metastatic forms of melanoma and renal cell carcinoma (RCC) (source http://www.fda.gov). The use of recombinant granulocyte colony-stimulating factor (G-CSF, also known as filgrastim, lenograstim or Neupogen®) and recombinant granulocyte monocyte colony-stimulating factor (GM-CSF, also known as molgramostim, sargramostim, Leukomax®, Mielogen® or Leukine®) in cancer patients has also been licensed by the US FDA. However, these cytokines are not (yet) harnessed for their ability to boost anticancer immune responses. Rather, they are employed as mitogenic factors, (1) to favor the reconstitution of the immune system in transplanted patients, who are generally subjected to lymphodepleting/lymphoablating regimens, as well as in patients treated with aggressive antimitotic chemotherapy, who are prone to develop febrile neutropenia;Citation28-Citation30 (2) to recruit bone marrow precursors to the peripheral blood in the context of autologous stem cell transplantation;Citation31,Citation32 (3) to prevent the neutropenia-inducing activity of specific chemotherapeutics;Citation33,Citation34 and (4) to favor the replication of quiescent leukemic cells, thus exposing them to the antineoplastic activity of drugs that preferentially target actively proliferating cells.Citation35 Finally, recombinant tumor necrosis factor α (TNFα) is currently approved by multiple regulatory agencies including the European Medicine Agency (EMA), but not by the US FDA, for use in patients with limb-threatening soft tissue sarcoma and melanoma.Citation36-Citation43 In this setting, TNFα is generally co-administered with melphalan (an alkylating agent) to isolated limbs under mild hyperthermic conditions, a safe and relatively simple procedure that has been associated with consistent rates of objective responses.Citation44-Citation46

Owing to their pleiotropic biological activity, cytokines can be associated with clinically relevant side effects, especially when administered systemically. There are 3 major concerns related to the use of cytokines in (cancer) patients: (1) the elicitation of an acute, sepsis-like, potentially lethal systemic reaction characterized by the massive release into the circulation of pyrogenic and cytotoxic cytokines;Citation47-Citation51 (2) the exacerbation of chronic inflammatory foci that may initiate oncogenesis or accelerate tumor progression;Citation19-Citation21 and (3) the activation of a mitogenic program in otherwise poorly proliferating cells, favoring the accumulation of genetic/epigenetic defects and hence increasing the likelihood of malignant transformation.Citation52-Citation54 In fact, some cytokines including multiple members of the platelet-derived growth factor (PDGF) family cannot be employed as therapeutic interventions owing to their excessive mitogenic (and hence potentially oncogenic) potential.Citation5,Citation6,Citation55

In previous issues of OncoImmunology, we discussed the scientific grounds supporting the use of cytokines as experimental immunostimulatory interventions in cancer patients as well as recent studies assessing the authentic clinical value of this regimen.Citation5,Citation6 Here, we present the newest developments in this exciting area of investigation. Of note, studies assessing the clinical profile of cytokines as immunoreconstituting agents, studies involving FDA-approved immunostimulatory cytokines (i.e., IFN-α2a, IFN-α2b and IL-2) employed as “on-label” interventions (see above), as well as studies investigating the antineoplastic activity of potentially oncotoxic cytokines, such as TNFα, will not be discussed here.

Literature Update

Since the submission of our latest Trial Watch dealing with topic (April 2013),Citation5 the results of at least 10 clinical studies evaluating the therapeutic profile of cytokines as off-label immunostimulatory interventions in cancer patients have been published in peer-reviewed scientific journals (source http://www.ncbi.nlm.nih.gov/pubmed).

Dutcher and colleagues, in collaboration with the Eastern Cooperative Oncology Group, tested the ability of recombinant IL-1α to boost the antineoplastic activity of cyclophosphamide (an immunostimulatory alkylating agent)Citation56-Citation60 in patients with advanced solid tumors. In this Phase I clinical study, 3 different IL-1α doses and administration schedules were evaluated. Common side effects included fever, chills, hypotension, nausea/emesis, and elevations in circulating hepatic enzymes. Moreover, the co-administration of IL-1α failed to rescue the neutropenic effects of cyclophosphamide, suggesting that other, comparatively more specific (and hence less toxic) cytokines may be best suitable to provide a hematopoietic support to chemotherapy.Citation61

Vitale and coworkers investigated the therapeutic profile of subcutaneous low-dose IL-2, combined with the somatostatin analog lanreotide,Citation62-Citation64 in 6 patients with symptomatic and advanced medullary thyroid carcinoma. The authors observed that a 6-mo regimen of lanreotide plus low-dose IL-2 was well tolerated by all patients, improved their quality of life and elicited a partial response or disease stabilization in 2 or 3 individuals, respectively.Citation64 Tomov and collaborators reported a case of hepatocellular carcinoma that recurred upon the surgical resection of a primary lesion of 60 mm initially misdiagnosed as hepatic adenoma. Seven new lesions developed upon surgery, and the patient received high-dose IL-2, bacillus Calmette-Guérin (an attenuated variant of Mycobacterium bovis that is currently employed for the treatment of transitional cell carcinomas of the bladder)Citation25,Citation65 and melatonin (a pineal hormone that appears to mediate antineoplastic effects, especially in combination with IL-2).Citation66-Citation68 Of note, the tumor rapidly became undetectable by magnetic resonance imaging and computer tomography, and the patient remained in complete remission for at least 6 y after the confirmed diagnosis of untreatable hepatocellular carcinoma.Citation69

Robertson et al. performed a dose-escalation Phase I study to test the safety and therapeutic profile of recombinant human IL-18 in non-Hodgkin lymphoma (NHL) patients treated with the CD20-targeting mAb rituximab.Citation70-Citation73 Rituximab (375 mg/m2) was administered i.v. once weekly for a total of 4 wks, while escalating doses of IL-18 (1, 3, 10, 20, 30, and 100 mug/kg) were given as a 2 h intravenous infusion weekly for 12 consecutive wks. No dose-limiting toxicities were observed. Common side effects were chills, fever, headache and nausea, while abnormal laboratory findings included transient asymptomatic lymphopenia, hyperglycemia, anemia, hypoalbuminemia as well as temporary elevations in circulating bilirubin and hepatic enzymes. Of note, 5 out of 19 patients experienced objective clinical responses. Altogether, these findings suggest that recombinant human IL-18 is well tolerated at doses at which it may improve the therapeutic profile of rituximab in NHL patients.Citation74

Gorin and colleagues tested the ability of G-CSF to boost the therapeutic profile of the anti-CD52 mAb alemtuzumab, which mostly originates from antibody-dependent cell-mediated cytotoxicity,Citation75,Citation76 in 12 patients with relapsed or refractory acute lymphoblastic leukemia. In the context of this Phase II clinical study, patients received 5 mug/kg G-CSF per day along with 30 mg alemtuzumab 3 times per wk for a total of 12–18 infusions. Fever/chills, skin rash and bronchospasm were the most common side effects. Four patients achieved a complete response, defined as the disappearance of leukemic blasts from the bone marrow. Nonetheless, all patients progressed within a few months and all but one died. These results indicate that alemtuzumab plus G-CSF may induce robust but temporary clinical responses.Citation77

Cheung and coworkers investigated the ability of GM-CSF to improve the response of 79 patients with persistent osteomedullary neuroblastoma to 3F8, a mAb specific for GD2 ganglioside.Citation78-Citation80 Patients were treated with 3F8 plus GM-CSF for up to 24 mo, or until the development of neutralizing anti-3F8 antibodies. In the context of this Phase II clinical trial, toxicities were generally manageable and 38% of patients achieved an objective response as defined by metaiodobenzyl-guanidine scan. Moreover, the 5-y progression-free survival of patients receiving 3F8 plus subcutaneous GM-CSF was 24 ± 6%, which was significantly better than that of patients treated with 3F8 plus intravenous GM-CSF (11 ± 7%).Citation81-Citation83

Zarogoulidis et al. tested whether IFN-α and IFN-γ, administered alone (3 MIUs) or in combination (1.5 plus 1.5 MIUs) 3 times per wk, would improve the activity of carboplatin-, fosfamide- and etoposide-based chemotherapy in a cohort of 164 individuals with small cell lung carcinoma (SCLC). No differences in survival between groups were observed in the context of this Phase II clinical trial when all patients were included in the analysis. However, when only individuals with early disease were considered, IFN-α appeared to provide a survival benefit to SCLC patients treated with chemotherapy.Citation84

Coker and colleagues performed a Phase I dose-escalation study of oral temozolomide, an alkylating agent, combined with subcutaneous pegylated IFN-α2b in 19 patients with refractory or advanced solid tumors. The authors identified the maximum tolerated dose of the combination in 100 mg/m2 temozolomide on days 1–7 and 15–21 plus 1.5 µg/kg IFN-α2b per week on 28-d cycles, and reported that the pharmacokinetics of pegylated IFN-α2b are not altered by the co-administration of temozolomide.Citation85

Eto and collaborators prospectively investigated the predictive value of 11 single nucleotide polymorphisms (SNPs) affecting 8 distinct genes linked to immune responses among 203 RCC patients treated with 3 doses per wk of IFN-α (5 MIUs). The authors reported a response rate of 13.8% (9 complete responses, 19 partial responses), which was not influenced by any of the SNPs analyzed in this study. However, when disease stabilization for >24 wks was included among clinically favorable outcomes, a SNP affecting signal transducer and activator of transcription 3 (STAT3) was statistically associated with clinical responses, confirming previous observations from the same group.Citation86,Citation87

Harmon and coworkers evaluated potential biomarkers of efficacy among 750 treatment-naïve metastatic RCC patients randomized to receive 50 mg/day sunitinib (a multi-targeted receptor tyrosine kinase inhibitor)Citation88-Citation90 on a 4-wk on/2-wk off schedule or 9 MIUs subcutaneous IFN-α 3 times per wk. Circulating IL-8 and VEGF-A levels at baseline were associated with overall survival independent of treatment. However, no independent predictors of IFN-α efficacy were identified by multivariate analysis.Citation91

Among recent translational studies focusing on immunostimulatory cytokines in general, we found of particular interest the works of (1) Guermonprez and colleagues, who discovered a signaling pathway triggered by Plasmodium infection that regulates DC homeostasis and adaptive immune response upon the release of fms-related tyrosine kinase 3 ligand (FLT3L);Citation92 (2) Sim and coworkers, who demonstrated that CD4+CD25+FOXP3+ Tregs accumulating in melanoma patients treated with high-dose IL-2 express inducible T-cell co-stimulator (ICOS) and exhibit an activated phenotype, as indicated by elevated levels of CD39, CD73 and transforming growth factor β1 (TGFβ1);Citation93 (3) Mortha and collaborators, who showed that GM-CSF is required for the establishment of Treg-dependent oral tolerance by intestinal macrophages;Citation94 and (4) West et al., who demonstrated that combining IL-2 with a mAb specific for CD274 mediates synergistic immunostimulatory effects.Citation95 These latter findings confirm and extend previous results indicating that immune checkpoint-blocking agents such as the cytotoxic T lymphocyte-associated protein 4 (CTLA4)-targeting mAb ipilimumab may significantly ameliorate the therapeutic profile of immunostimulatory cytokines.Citation96,Citation97

Update on Ongoing Clinical Trials

When this Trial Watch was being redacted (April 2014), official sources listed no less than 88 clinical trials launched after May 1st, 2013 that would evaluate the efficacy and safety of immunostimulatory cytokines in cancer patients (source http://www.clinicaltrials.gov). In 54 of these studies, IL-2 (14 trials), GM-CSF (4 trials), G-CSF (33 trials) and IFN-α (3 trials) were used as on-label interventions. These studies will not be discussed here. In addition, 34 clinical trials have been launched during the last 12 mo to investigate the immunostimulatory potential of various cytokines in off-label settings ().

Table 1. Clinical trials recently launched to evaluate the safety and efficacy of immunostimulatory cytokines in cancer patients*

Recombinant IL-2 is being tested (1) in combination with the adoptive transfer of natural killer (NK) cells,Citation55,Citation98 in patients with relapsed or refractory acute myeloid leukemia (AML) (NCT01898793), multiple myeloma (MM) (NCT01884688), and v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2 (ERBB2)+ breast or gastric carcinoma (NCT02030561), in the latter setting coupled to the ERBB2-targeting monoclonal antibody (mAb) trastuzumab;Citation99-Citation104 (2) in combination with autologous peripheral blood lymphocytes engineered to express a NY-ESO-1-specific T-cell receptor, alone or coupled to ipilimumabCitation105-Citation107 in individuals affected by diverse solid tumors (NCT01967823; NCT02070406); (3) as an adjuvant to T cells expressing a chimeric antigen receptor specific for folate hydrolase 1 (FOLH1, best known as PSMA),Citation108,Citation109 in prostate cancer patients subjected to non-myeloablative conditioning (NCT01929239); and (4) in combination with G-CSF, GM-CSF and a mAb specific for ganglioside GD2,Citation110 for the treatment of minimal residual disease in children with advanced neuroblastoma treated with aggressive induction chemotherapy and stem cell transplantation (NCT01857934). Moreover, various studies have recently been launched to test the clinical profile of IL-2 variants retargeted to cancer cells by means of tumor-associated antigen (TAA)-specific antibodies or antibody fragments, i.e., IL-2-based immunocytokines.Citation111-Citation113 Thus, (1) the safety and therapeutic potential of a carcinoembryonic antigen (CEA)-directed IL-2 variant (RO6895882) are being assessed in patients with advanced and/or metastatic solid tumors (NCT02004106); (2) a fusion between IL-2 and the Fv fragment of a mAb specific for tenascin C (TNC)Citation114 is being tested, in combination with the microtubular inhibitor paclitaxel, in Merkel cell carcinoma patients (NCT02054884); (3) the therapeutic value of a CD20-retargeted form of IL-2 is being investigated as a standalone intervention in subjects with NHL (NCT01874288); (4) the safety and efficacy of IL-2 fused to the Fv fragment of a mAb specific for the extradomain B of fibronectin (L19),Citation115-Citation117 administered i.t. in combination with L19-TNFα, are being evaluated in melanoma patients (NCT02076633); and (5) the therapeutic profile of L19-IL-2 administered as a standalone agent immediately after stereotactic ablative body radiotherapyCitation118 is being investigated in subjects affected by metastatic non-small cell lung carcinoma (NSCLC) (NCT02086721). Moreover, (1) glycosylated recombinant human IL-7 is being tested as adjuvant to sipuleucel-T (an FDA-approved vaccine based on autologous peripheral blood mononuclear cells)Citation119 in subjects with castration-resistant prostate cancer (NCT01881867); (2) pegylated recombinant human IL-10 administered s.c. is being assessed as a standalone therapeutic intervention in patients with advanced solid tumors (NCT02009449); and (3) the clinical profile of recombinant human IL-15 given i.v. in combination with autologous activated NK cells is under evaluation in children and young adults affected by solid neoplasms (NCT01875601).

The safety and clinical potential of IFN-α2b, invariably administered s.c., are being investigated in cohorts of (1) subjects with gastrointestinal neuroendocrine tumors that failed to respond to somatostatin analogs, who receive IFN-α2b as a standalone agent (NCT01860742); (2) pediatric patients with unresectable of recurrent craniopharyngioma, who are treated with a pegylated variant of IFN-α2b (NCT01964300); (3) individuals with advanced melanoma or RCC, receiving pegylated IFN-α2b in combination with a mAb specific for programmed cell death 1 (PDCD1, best-known as PD-1)Citation120-Citation123 (NCT02089685); (4) patients with chronic myeloid leukemia, who are treated with normal or pegylated IFN-α2b plus tyrosine kinase inhibitors including imatinibCitation124 (NCT01933906; NCT02001818), dasatinibCitation125 (NCT01872442) and nilotinibCitation126 (NCT01866553; NCT02001818); and (5) adults affected by anal intraepithelial neoplasms, who are concurrently vaccinated with a human papillomavirus Type 16 (HPV-16)-targeting peptide-based vaccineCitation24 (NCT01923116). In addition, a not-better defined variant of IFN-α is being tested as standalone intervention for molecular relapse in t(8; 21) AML patients who previously underwent allogeneic stem cell transplantation (NCT02027064); and recombinant IFN-γ is being assessed for its therapeutic efficacy in subjects with soft tissue sarcoma (NCT01957709).

GM-CSF is being intensively investigated for its ability to boost tumor-targeting immune responses elicited by wide panel of immunotherapeutic interventions. In particular, GM-CSF is being tested as an adjuvant to (1) ipilimumab,Citation27,Citation70 in subjects with unresectable, Stage IIIC or IV melanoma (NCT02009397); (2) rituximab,Citation70,Citation71 in individuals affected by follicular B-cell lymphoma (NCT01939730); (3) a peptide vaccine directed against folate receptor 1 (FOLR1, also known as FBP),Citation127,Citation128 in patients with breast and ovarian carcinoma (NCT02019524); (4) SL-071, a multipeptide-based vaccine targeting several epitopes overexpressed by glioma cells,Citation129,Citation130 administered to glioblastoma patients in combination with the TLR7 agonist imiquimodCitation131,Citation132 (NCT02078648); (5) a peptide vaccine specific for Wilms tumor 1 (WT1),Citation133,Citation134 in mesothelioma patients who have completed multimodal therapy (NCT01890980); (6) a tyrosinase (TYR)-targeting peptide vaccine,Citation135-Citation137 in melanoma patients who underwent tumor resection (NCT01989572); (7) an autophagosome-based vaccine derived from allogeneic cancer cells,Citation138-Citation140 administered to NSCLC patients in combination with imiquimod (NCT01909752); and (8) cyclophosphamide, the vascular endothelial growth factor (VEGF)-specific mAb bevacizumabCitation70,Citation71,Citation141,Citation142 and a vaccine based on autologous cancer cells, in patients with glioblastoma multiforme or gliosarcoma (NCT01903330). Finally, the intratumoral administration of recombinant FLT3LCitation143-Citation145 to B-cell lymphoma patients is being tested for its capacity to recruit DCs to neoplastic lesions and hence allow low-dose radiation therapy and a TLR3 agonist to induce clinically relevant anticancer immune responses (NCT01976585). This is a novel application for recombinant human FLT3L (also known as CDX-301), which is being developed as an alternative or a support to G-CSF for the mobilization of hematopoietic cell precursors in bone marrow donors, with promising results (source http://www.celldex.com/). Of note, official sources list NCT01989572 and NCT01939730 as “completed,” yet no results are appear to be available at the moment.

As for the clinical trials listed in our previous Trial Watches dealing with this topic,Citation98,Citation146 the following studies have changed status during the past 12 mo: NCT00589550, NCT00977145, NCT01099631, NCT01099631, NCT01131364, NCT01337544, which appear as ‘terminated’; NCT00784524, NCT01236573 and NCT01490047, which are listed as ‘suspended’; NCT00631371 and NCT00923351, which show as ‘active, not recruiting’ but are associated with results; as well as NCT00601796, NCT00660270, NCT00719264 NCT00891475, NCT01021059, NCT01220648, NCT01404936, NCT01592045, NCT01637532, NCT01639885 and NCT01673217, which have been completed (source http://www.clinicaltrials.gov).

While NCT00977145, NCT00589550, NCT01099631 and NCT01392170 have been terminated owing to slow accrual, NCT01337544 has been halted because the parents of two patients enrolled who died presented a claim against the hospital. The reasons underlying the termination of NCT01131364 are not available. NCT00784524 has been suspended for interim analysis, whereas NCT01236573 has been temporarily halted for final data collection and primary outcome assessment. The reasons behind the suspension of NCT01490047 have not been reported. The results of NCT00660270,Citation147 NCT00719264, NCT00891475, NCT01021059, NCT01220648, NCT01592045, NCT01637532, NCT01639885 and NCT01673217 have not been disseminated yet. Conversely results are available for NCT00601796, NCT00631371, NCT00923351 and NCT01404936. In the context of NCT00601796, testing a GM-CSF-involving cell-based vaccine in combination with cyclophosphamide and all-trans retinoic acid in lung cancer patients, 5 immunological responses were observed among 14 evaluable patients, the median time to progression and median overall survival among 24 treated patients being 2.4 and 8 mo, respectively. Preliminary results from NCT00631371, which is still ongoing, revealed that bevacizumab plus IFN-α is not inferior to bevacizumab plus the mammalian target of rapamycin (mTOR) inhibitor temsirolimus for the treatment of advanced RCC patients, but associated with lower incidence of serious adverse effects (36.6% vs. 44.3%). NCT00923351 is investigating the ability of recombinant IL-7 to boost the therapeutic activity of a DC-based vaccine in patients with Ewing's Sarcoma, rhabdomyosarcoma or neuroblastoma. Preliminary results indicate that IL-7 may indeed promote the immunogenic potential of DCs loaded ex vivo with autologous cancer cell lysates but not increase the toxicity of the procedure. The number of patients enrolled and analyzed so far, however, appears to be excessively low for drawing robust conclusions from this study. NCT01404936 evaluated the combination of IFN-α2a with a multi-agent chemotherapeutic regimen in Hodgkin lymphoma patients. In this setting, 23 out of 30 patients achieved a complete response to treatment, while serious side effects affected only 10% of participants. Additional, randomized and comparatively larger clinical studies are required to validate these findings.

Concluding Remarks

The activation of novel or the reactivation of existing immune responses has been shown to underlie the clinical efficacy not only of an increasingly wide panel of immunotherapeutic interventionsCitation148 but also of multiple radiotherapeutic and chemotherapeutic regimens.Citation56,Citation57,Citation149,Citation150 Along with the realization that the immune system plays a fundamental role in the response of cancer patients to therapy, great interest has gathered around the possibility to harness the immunostimulatory potential of multiple cytokines to drive tumor-targeting immune responses. As discussed above, however, using cytokines as standalone immunostimulatory interventions does not suffice to elicit therapeutically relevant immune responses in a majority of cancer patients, exception made for individuals with melanoma and RCC, which are particularly immunogenic per se. Thus, current efforts focus on the use of immunostimulatory cytokines as adjuvants to other immunotherapeutic paradigms, especially immune checkpoint-blocking mAbs. IL-2 and GM-CSF are perhaps the molecules that have generated the greatest interest in this setting. However, recent preclinical and clinical data indicate that the immunological activity of both IL-2 and GM-CSF may exhibit a significant degree of context dependency. Indeed, high-dose IL-2 has been shown to promote the accumulation of immunosuppressive CD4+CD25+FOXP3+ regulatory T cells (Tregs) in both cancer and HIV-1 patients,Citation93,Citation151-Citation156 while GM-CSF has been involved in the establishment of Treg-mediated oral tolerance by intestinal macrophages.Citation94 These data suggest that selectively targeting IL-2 or GM-CSF to specific immune effectors may further improve their immunostimulatory activity (and hence their clinical profile). So far, immunocytokines have mostly been designed to deliver immunostimulatory signals the tumor microenvironment in a relatively unspecific manner (i.e., they have been developed based on TAA-specific mAb). However, neoplastic lesions contain high amounts of Tregs, myeloid-derived suppressor cells and tumor-associated macrophages,Citation157 implying that such a strategy may promote the unwarranted expansion of immunosuppressive cells. Further studies are required to unveil whether targeting immunocytokines to specific populations of immune effector cells results in optimal immunostimulation.

Abbreviations:
AML=

acute myeloid leukemia

CML=

chronic myelogenous leukemia

DC=

dendritic cell

FDA=

Food and Drug Administration

FLT3L=

fms-related tyrosine kinase 3 ligand

G-CSF=

granulocyte colony-stimulating factor

GM-CSF=

granulocyte macrophage colony-stimulating factor

IFN=

interferon

IL=

interleukin

mAb=

monoclonal antibody

NSCLC=

non-small cell lung carcinoma

NHL=

non-Hodgkin lymphoma

PDGF=

platelet-derived growth factor

RCC=

renal cell carcinoma

SCLC=

small cell lung carcinoma

SNP=

single nucleotide polymorphism

TAA=

tumor-associated antigen

TLR=

Toll-like receptor

TNFα=

tumor necrosis factor α

Treg=

regulatory T cell

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

Authors are supported by the Ligue contre le Cancer (équipe labelisée); Agence National de la Recherche (ANR); Association pour la recherche sur le cancer (ARC); Cancéropôle Ile-de-France; AXA Chair for Longevity Research; Institut National du Cancer (INCa); Fondation Bettencourt-Schueller; Fondation de France; Fondation pour la Recherche Médicale (FRM); the European Commission (ArtForce); the European Research Council (ERC); the LabEx Immuno-Oncology; the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); the SIRIC Cancer Research and Personalized Medicine (CARPEM); and the Paris Alliance of Cancer Research Institutes (PACRI).

References

  • Tato CM, Cua DJ. SnapShot: Cytokines I. Cell 2008; 132:324. e1
  • Tato CM, Cua DJ. SnapShot: cytokines II. Cell 2008; 132:500; PMID: 18267079
  • Tato CM, Cua DJ. SnapShot: cytokines III. Cell 2008; 132:900; PMID: 18329374
  • Tato CM, Cua DJ. SnapShot: Cytokines IV. Cell 2008; 132:e1 - 2; PMID: 18358817
  • Vacchelli E, Eggermont A, Fridman WH, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulatory cytokines. Oncoimmunology 2013; 2:e24850; http://dx.doi.org/10.4161/onci.24850; PMID: 24073369
  • Vacchelli E, Galluzzi L, Eggermont A, Galon J, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Immunostimulatory cytokines. Oncoimmunology 2012; 1:493 - 506; http://dx.doi.org/10.4161/onci.20459; PMID: 22754768
  • Papatriantafyllou M. Cytokines: true to their family name. Nat Rev Immunol 2013; 13:544 - 5; http://dx.doi.org/10.1038/nri3496; PMID: 23827957
  • Borish LC, Steinke JW. 2. Cytokines and chemokines. J Allergy Clin Immunol 2003; 111:Suppl S460 - 75; http://dx.doi.org/10.1067/mai.2003.108; PMID: 12592293
  • Steinke JW, Borish L. 3. Cytokines and chemokines. J Allergy Clin Immunol 2006; 117:Suppl Mini-Primer S441 - 5; http://dx.doi.org/10.1016/j.jaci.2005.07.001; PMID: 16455343
  • Metcalf D. The colony-stimulating factors and cancer. Nat Rev Cancer 2010; 10:425 - 34; http://dx.doi.org/10.1038/nrc2843; PMID: 20495576
  • Neufeld G, Kessler O. Pro-angiogenic cytokines and their role in tumor angiogenesis. Cancer Metastasis Rev 2006; 25:373 - 85; http://dx.doi.org/10.1007/s10555-006-9011-5; PMID: 17006765
  • Benelli R, Lorusso G, Albini A, Noonan DM. Cytokines and chemokines as regulators of angiogenesis in health and disease. Curr Pharm Des 2006; 12:3101 - 15; http://dx.doi.org/10.2174/138161206777947461; PMID: 16918437
  • Ohlsson K, Björk P, Bergenfeldt M, Hageman R, Thompson RC. Interleukin-1 receptor antagonist reduces mortality from endotoxin shock. Nature 1990; 348:550 - 2; http://dx.doi.org/10.1038/348550a0; PMID: 2147233
  • Tracey KJ, Fong Y, Hesse DG, Manogue KR, Lee AT, Kuo GC, Lowry SF, Cerami A. Anti-cachectin/TNF monoclonal antibodies prevent septic shock during lethal bacteraemia. Nature 1987; 330:662 - 4; http://dx.doi.org/10.1038/330662a0; PMID: 3317066
  • Andrews DM, Chow MT, Ma Y, Cotterell CL, Watt SV, Anthony DA, Akira S, Iwakura Y, Trapani JA, Zitvogel L, et al. Homeostatic defects in interleukin 18-deficient mice contribute to protection against the lethal effects of endotoxin. Immunol Cell Biol 2011; 89:739 - 46; http://dx.doi.org/10.1038/icb.2010.168; PMID: 21263463
  • Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol 2005; 6:1191 - 7; http://dx.doi.org/10.1038/ni1276; PMID: 16369558
  • Brenner C, Galluzzi L, Kepp O, Kroemer G. Decoding cell death signals in liver inflammation. J Hepatol 2013; 59:583 - 94; http://dx.doi.org/10.1016/j.jhep.2013.03.033; PMID: 23567086
  • Senovilla L, Galluzzi L, Zitvogel L, Kroemer G. Immunosurveillance as a regulator of tissue homeostasis. Trends Immunol 2013; 34:471 - 81; http://dx.doi.org/10.1016/j.it.2013.06.005; PMID: 23891238
  • Dranoff G. Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer 2004; 4:11 - 22; http://dx.doi.org/10.1038/nrc1252; PMID: 14708024
  • Coussens LM, Werb Z. Inflammation and cancer. Nature 2002; 420:860 - 7; http://dx.doi.org/10.1038/nature01322; PMID: 12490959
  • Coussens LM, Zitvogel L, Palucka AK. Neutralizing tumor-promoting chronic inflammation: a magic bullet?. Science 2013; 339:286 - 91; http://dx.doi.org/10.1126/science.1232227; PMID: 23329041
  • Vacchelli E, Vitale I, Eggermont A, Fridman WH, Fučíková J, Cremer I, Galon J, Tartour E, Zitvogel L, Kroemer G, et al. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2013; 2:e25771; http://dx.doi.org/10.4161/onci.25771; PMID: 24286020
  • Pol J, Bloy N, Obrist F, Eggermont A, Galon J, Hervé Fridman W, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: DNA vaccines for cancer therapy. Oncoimmunology 2014; 3:e28185; http://dx.doi.org/10.4161/onci.28185; PMID: 24800178
  • Aranda F, Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Peptide vaccines in cancer therapy. Oncoimmunology 2013; 2:e26621; http://dx.doi.org/10.4161/onci.26621; PMID: 24498550
  • Vacchelli E, Eggermont A, Sautès-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Toll-like receptor agonists for cancer therapy. Oncoimmunology 2013; 2:e25238; http://dx.doi.org/10.4161/onci.25238; PMID: 24083080
  • Yamazaki T, Hannani D, Poirier-Colame V, Ladoire S, Locher C, Sistigu A, Prada N, Adjemian S, Catani JP, Freudenberg M, et al. Defective immunogenic cell death of HMGB1-deficient tumors: compensatory therapy with TLR4 agonists. Cell Death Differ 2014; 21:69 - 78; http://dx.doi.org/10.1038/cdd.2013.72; PMID: 23811849
  • Aranda F, Vacchelli E, Eggermont A, Galon J, Fridman WH, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulatory monoclonal antibodies in cancer therapy. Oncoimmunology 2014; 3:e27297; http://dx.doi.org/10.4161/onci.27297; PMID: 24701370
  • Arellano M, Lonial S. Clinical uses of GM-CSF, a critical appraisal and update. Biologics 2008; 2:13 - 27; http://dx.doi.org/10.2147/BTT.S1355; PMID: 19707424
  • Khoury HJ, Loberiza FR Jr., Ringdén O, Barrett AJ, Bolwell BJ, Cahn JY, Champlin RE, Gale RP, Hale GA, Urbano-Ispizua A, et al. Impact of posttransplantation G-CSF on outcomes of allogeneic hematopoietic stem cell transplantation. Blood 2006; 107:1712 - 6; http://dx.doi.org/10.1182/blood-2005-07-2661; PMID: 16239431
  • Sebban C, Lefranc A, Perrier L, Moreau P, Espinouse D, Schmidt A, Kammoun L, Ghesquieres H, Ferlay C, Bay JO, et al. A randomised phase II study of the efficacy, safety and cost-effectiveness of pegfilgrastim and filgrastim after autologous stem cell transplant for lymphoma and myeloma (PALM study). Eur J Cancer 2012; 48:713 - 20; http://dx.doi.org/10.1016/j.ejca.2011.12.016; PMID: 22248711
  • Hosing C. Hematopoietic stem cell mobilization with G-CSF. Methods Mol Biol 2012; 904:37 - 47; PMID: 22890920
  • Demirer T, Ayli M, Ozcan M, Gunel N, Haznedar R, Dagli M, Fen T, Genc Y, Dincer S, Arslan O, et al. Mobilization of peripheral blood stem cells with chemotherapy and recombinant human granulocyte colony-stimulating factor (rhG-CSF): a randomized evaluation of different doses of rhG-CSF. Br J Haematol 2002; 116:468 - 74; http://dx.doi.org/10.1046/j.1365-2141.2002.03264.x; PMID: 11841454
  • Naeim A, Henk HJ, Becker L, Chia V, Badre S, Li X, Deeter R. Pegfilgrastim prophylaxis is associated with a lower risk of hospitalization of cancer patients than filgrastim prophylaxis: a retrospective United States claims analysis of granulocyte colony-stimulating factors (G-CSF). BMC Cancer 2013; 13:11; http://dx.doi.org/10.1186/1471-2407-13-11; PMID: 23298389
  • Chan KK, Siu E, Krahn MD, Imrie K, Alibhai SM. Cost-utility analysis of primary prophylaxis versus secondary prophylaxis with granulocyte colony-stimulating factor in elderly patients with diffuse aggressive lymphoma receiving curative-intent chemotherapy. J Clin Oncol 2012; 30:1064 - 71; http://dx.doi.org/10.1200/JCO.2011.36.8647; PMID: 22393098
  • Pabst T, Vellenga E, van Putten W, Schouten HC, Graux C, Vekemans MC, Biemond B, Sonneveld P, Passweg J, Verdonck L, et al, Dutch-Belgian Hemato-Oncology Cooperative Group (HOVON), German AML Study Group (AMLSG), Swiss Collaborative Group for Clinical Cancer Research (SAKK). Favorable effect of priming with granulocyte colony-stimulating factor in remission induction of acute myeloid leukemia restricted to dose escalation of cytarabine. Blood 2012; 119:5367 - 73; http://dx.doi.org/10.1182/blood-2011-11-389841; PMID: 22422824
  • Deroose JP, Eggermont AM, van Geel AN, Burger JW, den Bakker MA, de Wilt JH, Verhoef C. Long-term results of tumor necrosis factor alpha- and melphalan-based isolated limb perfusion in locally advanced extremity soft tissue sarcomas. J Clin Oncol 2011; 29:4036 - 44; http://dx.doi.org/10.1200/JCO.2011.35.6618; PMID: 21931039
  • Deroose JP, Eggermont AM, van Geel AN, de Wilt JH, Burger JW, Verhoef C. 20 years experience of TNF-based isolated limb perfusion for in-transit melanoma metastases: TNF dose matters. Ann Surg Oncol 2012; 19:627 - 35; http://dx.doi.org/10.1245/s10434-011-2030-7; PMID: 21879272
  • Deroose JP, Grünhagen DJ, van Geel AN, de Wilt JH, Eggermont AM, Verhoef C. Long-term outcome of isolated limb perfusion with tumour necrosis factor-α for patients with melanoma in-transit metastases. Br J Surg 2011; 98:1573 - 80; http://dx.doi.org/10.1002/bjs.7621; PMID: 21739427
  • Eggermont AM. The success of TNF alpha in isolated limb perfusion for irresectable extremity soft tissue sarcomas, melanoma and carcinomas: observations in patients and preclinical perfusion models. Gan To Kagaku Ryoho 1996; 23:1357 - 70; PMID: 8854755
  • Eggermont AM, Schraffordt Koops H, Klausner JM, Kroon BB, Schlag PM, Liénard D, van Geel AN, Hoekstra HJ, Meller I, Nieweg OE, et al. Isolated limb perfusion with tumor necrosis factor and melphalan for limb salvage in 186 patients with locally advanced soft tissue extremity sarcomas. The cumulative multicenter European experience. Ann Surg 1996; 224:756 - 64, discussion 764-5; http://dx.doi.org/10.1097/00000658-199612000-00011; PMID: 8968230
  • Eggermont AM, Schraffordt Koops H, Klausner JM, Schlag PM, Kroon BB, Ben-Ari G, Lejeune FJ. Isolated limb perfusion with high-dose tumor necrosis factor-alpha for locally advanced extremity soft tissue sarcomas. Cancer Treat Res 1997; 91:189 - 203; http://dx.doi.org/10.1007/978-1-4615-6121-7_13; PMID: 9286497
  • Eggermont AM, Schraffordt Koops H, Liénard D, Kroon BB, van Geel AN, Hoekstra HJ, Lejeune FJ. Isolated limb perfusion with high-dose tumor necrosis factor-alpha in combination with interferon-gamma and melphalan for nonresectable extremity soft tissue sarcomas: a multicenter trial. J Clin Oncol 1996; 14:2653 - 65; PMID: 8874324
  • Eggermont AM, Suciu S, Testori A, Santinami M, Kruit WH, Marsden J, Punt CJ, Salès F, Dummer R, Robert C, et al. Long-term results of the randomized phase III trial EORTC 18991 of adjuvant therapy with pegylated interferon alfa-2b versus observation in resected stage III melanoma. J Clin Oncol 2012; 30:3810 - 8; http://dx.doi.org/10.1200/JCO.2011.41.3799; PMID: 23008300
  • Grünhagen DJ, Brunstein F, ten Hagen TL, van Geel AN, de Wilt JH, Eggermont AM. TNF-based isolated limb perfusion: a decade of experience with antivascular therapy in the management of locally advanced extremity soft tissue sarcomas. Cancer Treat Res 2004; 120:65 - 79; http://dx.doi.org/10.1007/1-4020-7856-0_4; PMID: 15217218
  • Trabulsi NH, Patakfalvi L, Nassif MO, Turcotte RE, Nichols A, Meguerditchian AN. Hyperthermic isolated limb perfusion for extremity soft tissue sarcomas: systematic review of clinical efficacy and quality assessment of reported trials. J Surg Oncol 2012; 106:921 - 8; http://dx.doi.org/10.1002/jso.23200; PMID: 22806575
  • Grünhagen DJ, de Wilt JH, ten Hagen TL, Eggermont AM. Technology insight: Utility of TNF-alpha-based isolated limb perfusion to avoid amputation of irresectable tumors of the extremities. Nat Clin Pract Oncol 2006; 3:94 - 103; http://dx.doi.org/10.1038/ncponc0426; PMID: 16462850
  • Atkins MB, Lotze MT, Dutcher JP, Fisher RI, Weiss G, Margolin K, Abrams J, Sznol M, Parkinson D, Hawkins M, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 1999; 17:2105 - 16; PMID: 10561265
  • Fyfe GA, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie AC. Long-term response data for 255 patients with metastatic renal cell carcinoma treated with high-dose recombinant interleukin-2 therapy. J Clin Oncol 1996; 14:2410 - 1; PMID: 8708739
  • Fyfe G, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie AC. Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J Clin Oncol 1995; 13:688 - 96; PMID: 7884429
  • Hauschild A. Adjuvant interferon alfa for melanoma: new evidence-based treatment recommendations?. Curr Oncol 2009; 16:3 - 6; http://dx.doi.org/10.3747/co.v16i3.447; PMID: 19526078
  • Anger B, Porzsolt F, Leichtle R, Heinze B, Bartram C, Heimpel H. A phase I/II study of recombinant interferon alpha 2a and hydroxyurea for chronic myelocytic leukemia. Blut 1989; 58:275 - 8; http://dx.doi.org/10.1007/BF00320165; PMID: 2736308
  • Hershman D, Neugut AI, Jacobson JS, Wang J, Tsai WY, McBride R, Bennett CL, Grann VR. Acute myeloid leukemia or myelodysplastic syndrome following use of granulocyte colony-stimulating factors during breast cancer adjuvant chemotherapy. J Natl Cancer Inst 2007; 99:196 - 205; http://dx.doi.org/10.1093/jnci/djk028; PMID: 17284714
  • Di Cosimo S, Ferretti G, Papaldo P, Carlini P, Fabi A, Ruggeri EM, Alimonti A, Nardoni C, Cognetti F. Does the concurrent use of anthracycline and granulocyte colony-stimulating factor influence the risk of secondary leukaemia in breast cancer women?. Ann Oncol 2005; 16:1209 - 10; http://dx.doi.org/10.1093/annonc/mdi201; PMID: 15857847
  • Relling MV, Boyett JM, Blanco JG, Raimondi S, Behm FG, Sandlund JT, Rivera GK, Kun LE, Evans WE, Pui CH. Granulocyte colony-stimulating factor and the risk of secondary myeloid malignancy after etoposide treatment. Blood 2003; 101:3862 - 7; http://dx.doi.org/10.1182/blood-2002-08-2405; PMID: 12531808
  • Aranda F, Vacchelli E, Obrist F, Eggermont A, Fridman WH, Galon J, et al. Trial Watch: Adoptive cell transfer for anticancer immunotherapy. OncoImmunology 2013; 2:e24238; http://dx.doi.org/10.4161/onci.24238
  • Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov 2012; 11:215 - 33; http://dx.doi.org/10.1038/nrd3626; PMID: 22301798
  • Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity 2013; 39:74 - 88; http://dx.doi.org/10.1016/j.immuni.2013.06.014; PMID: 23890065
  • Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 2013; 342:971 - 6; http://dx.doi.org/10.1126/science.1240537; PMID: 24264990
  • Becker JC, Schrama D. The dark side of cyclophosphamide: cyclophosphamide-mediated ablation of regulatory T cells. J Invest Dermatol 2013; 133:1462 - 5; http://dx.doi.org/10.1038/jid.2013.67; PMID: 23673502
  • Connor JP, Cristea MC, Lewis NL, Lewis LD, Komarnitsky PB, Mattiacci MR, Felder M, Stewart S, Harter J, Henslee-Downey J, et al. A phase 1b study of humanized KS-interleukin-2 (huKS-IL2) immunocytokine with cyclophosphamide in patients with EpCAM-positive advanced solid tumors. BMC Cancer 2013; 13:20; http://dx.doi.org/10.1186/1471-2407-13-20; PMID: 23320927
  • Dutcher JP, Neuberg D, Atkins MB, Tester WJ, Wadler S, Stewart JA, Chachoua A, Schuchter LM. Report of a Phase I Evaluation of Dose and Schedule of Interleukin-1 Alpha and Cyclophosphamide in Patients with Advanced Tumors: An Eastern Cooperative Oncology Group Study (PX990) and Review of IL-1-Based Studies of Hematopoietic Reconstitution. J Interferon Cytokine Res 2014; 34:376 - 84; http://dx.doi.org/10.1089/jir.2013.0010; PMID: 24433038
  • Susini C, Buscail L. Rationale for the use of somatostatin analogs as antitumor agents. Ann Oncol 2006; 17:1733 - 42; http://dx.doi.org/10.1093/annonc/mdl105; PMID: 16801334
  • Reubi JC, Schonbrunn A. Illuminating somatostatin analog action at neuroendocrine tumor receptors. Trends Pharmacol Sci 2013; 34:676 - 88; http://dx.doi.org/10.1016/j.tips.2013.10.001; PMID: 24183675
  • Vitale G, Lupoli G, Guarrasi R, Colao A, Dicitore A, Gaudenzi G, Misso G, Castellano M, Addeo R, Facchini G, et al. Interleukin-2 and lanreotide in the treatment of medullary thyroid cancer: in vitro and in vivo studies. J Clin Endocrinol Metab 2013; 98:E1567 - 74; http://dx.doi.org/10.1210/jc.2013-1443; PMID: 23884781
  • Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology 2012; 1:894 - 907; http://dx.doi.org/10.4161/onci.20931; PMID: 23162757
  • Cutando A, López-Valverde A, Arias-Santiago S, DE Vicente J, DE Diego RG. Role of melatonin in cancer treatment. Anticancer Res 2012; 32:2747 - 53; PMID: 22753734
  • Vishwas DK, Mukherjee A, Haldar C. Melatonin improves humoral and cell-mediated immune responses of male golden hamster following stress induced by dexamethasone. J Neuroimmunol 2013; 259:17 - 25; http://dx.doi.org/10.1016/j.jneuroim.2013.03.002; PMID: 23582490
  • Srinivasan V, Pandi-Perumal SR, Brzezinski A, Bhatnagar KP, Cardinali DP. Melatonin, immune function and cancer. Recent Pat Endocr Metab Immune Drug Discov 2011; 5:109 - 23; http://dx.doi.org/10.2174/187221411799015408; PMID: 22074586
  • Tomov B, Popov D, Tomova R, Vladov N, Den Otter W, Krastev Z. Therapeutic response of untreatable hepatocellular carcinoma after application of the immune modulators IL-2, BCG and melatonin. Anticancer Res 2013; 33:4531 - 5; PMID: 24123026
  • Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2013; 2:e22789; http://dx.doi.org/10.4161/onci.22789; PMID: 23482847
  • Vacchelli E, Aranda F, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Tumor-targeting monoclonal antibodies in cancer therapy. Oncoimmunology 2014; 3:e27048; http://dx.doi.org/10.4161/onci.27048; PMID: 24605265
  • Akhtar S, Maghfoor I. Rituximab plus CHOP for diffuse large-B-cell lymphoma. N Engl J Med 2002; 346:1830 - 1, author reply 1830-1; http://dx.doi.org/10.1056/NEJM200206063462317; PMID: 12050349
  • Coiffier B, Lepage E, Briere J, Herbrecht R, Tilly H, Bouabdallah R, Morel P, Van Den Neste E, Salles G, Gaulard P, et al. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med 2002; 346:235 - 42; http://dx.doi.org/10.1056/NEJMoa011795; PMID: 11807147
  • Robertson MJ, Kline J, Struemper H, Koch KM, Bauman JW, Gardner OS, Murray SC, Germaschewski F, Weisenbach J, Jonak Z, et al. A dose-escalation study of recombinant human interleukin-18 in combination with rituximab in patients with non-Hodgkin lymphoma. J Immunother 2013; 36:331 - 41; http://dx.doi.org/10.1097/CJI.0b013e31829d7e2e; PMID: 23799412
  • Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zucman-Rossi J, Zitvogel L, Kroemer G. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2012; 1:28 - 37; http://dx.doi.org/10.4161/onci.1.1.17938; PMID: 22720209
  • Hale G, Bright S, Chumbley G, Hoang T, Metcalf D, Munro AJ, Waldmann H. Removal of T cells from bone marrow for transplantation: a monoclonal antilymphocyte antibody that fixes human complement. Blood 1983; 62:873 - 82; PMID: 6349718
  • Gorin NC, Isnard F, Garderet L, Ikhlef S, Corm S, Quesnel B, Legrand O, Cachanado M, Rousseau A, Laporte JP. Administration of alemtuzumab and G-CSF to adults with relapsed or refractory acute lymphoblastic leukemia: results of a phase II study. Eur J Haematol 2013; 91:315 - 21; PMID: 23738686
  • Kushner BH, Cheung NK. GM-CSF enhances 3F8 monoclonal antibody-dependent cellular cytotoxicity against human melanoma and neuroblastoma. Blood 1989; 73:1936 - 41; PMID: 2653466
  • Heiner JP, Miraldi F, Kallick S, Makley J, Neely J, Smith-Mensah WH, Cheung NK. Localization of GD2-specific monoclonal antibody 3F8 in human osteosarcoma. Cancer Res 1987; 47:5377 - 81; PMID: 3115567
  • Cheung NK, Lazarus H, Miraldi FD, Abramowsky CR, Kallick S, Saarinen UM, Spitzer T, Strandjord SE, Coccia PF, Berger NA. Ganglioside GD2 specific monoclonal antibody 3F8: a phase I study in patients with neuroblastoma and malignant melanoma. J Clin Oncol 1987; 5:1430 - 40; PMID: 3625258
  • Cheung IY, Hsu K, Cheung NK. Activation of peripheral-blood granulocytes is strongly correlated with patient outcome after immunotherapy with anti-GD2 monoclonal antibody and granulocyte-macrophage colony-stimulating factor. J Clin Oncol 2012; 30:426 - 32; http://dx.doi.org/10.1200/JCO.2011.37.6236; PMID: 22203761
  • Cheung NK, Cheung IY, Kramer K, Modak S, Kuk D, Pandit-Taskar N, Chamberlain E, Ostrovnaya I, Kushner BH. Key role for myeloid cells: Phase II results of anti-GD2 antibody 3F8 plus granulocyte-macrophage colony-stimulating factor for chemoresistant osteomedullary neuroblastoma. Int J Cancer 2014; Forthcoming http://dx.doi.org/10.1002/ijc.28851; PMID: 24644014
  • Cheung NK, Guo H, Hu J, Tassev DV, Cheung IY. Humanizing murine IgG3 anti-GD2 antibody m3F8 substantially improves antibody-dependent cell-mediated cytotoxicity while retaining targeting in vivo. Oncoimmunology 2012; 1:477 - 86; http://dx.doi.org/10.4161/onci.19864; PMID: 22754766
  • Zarogoulidis K, Ziogas E, Boutsikou E, Zarogoulidis P, Darwiche K, Kontakiotis T, Tsakiridis K, Porpodis K, Latsios D, Chatzizisi O, et al. Immunomodifiers in combination with conventional chemotherapy in small cell lung cancer: a phase II, randomized study. Drug Des Devel Ther 2013; 7:611 - 7; http://dx.doi.org/10.2147/DDDT.S43184; PMID: 23901264
  • Coker SA, Dandamudi UB, Beelen AP, Crosby NA, Fisher JL, Obrocea M, Ernstoff MS, Lewis LD. A phase I, dose-escalation study of cyclical weekly oral temozolomide and weekly PEG-interferon alpha-2b in patients with refractory or advanced solid tumours. J Chemother 2013; 25:362 - 8; http://dx.doi.org/10.1179/1973947813Y.0000000102; PMID: 24093213
  • Eto M, Kamba T, Miyake H, Fujisawa M, Kamai T, Uemura H, Tsukamoto T, Azuma H, Matsubara A, Nishimura K, et al, Japan Immunotherapy SNPs-Study Group for Kidney Cancer. STAT3 polymorphism can predict the response to interferon-α therapy in patients with metastatic renal cell carcinoma. Eur Urol 2013; 63:745 - 52; http://dx.doi.org/10.1016/j.eururo.2012.09.052; PMID: 23063454
  • Ito N, Eto M, Nakamura E, Takahashi A, Tsukamoto T, Toma H, Nakazawa H, Hirao Y, Uemura H, Kagawa S, et al. STAT3 polymorphism predicts interferon-alfa response in patients with metastatic renal cell carcinoma. J Clin Oncol 2007; 25:2785 - 91; http://dx.doi.org/10.1200/JCO.2006.09.8897; PMID: 17602083
  • Faivre S, Demetri G, Sargent W, Raymond E. Molecular basis for sunitinib efficacy and future clinical development. Nat Rev Drug Discov 2007; 6:734 - 45; http://dx.doi.org/10.1038/nrd2380; PMID: 17690708
  • Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007; 356:115 - 24; http://dx.doi.org/10.1056/NEJMoa065044; PMID: 17215529
  • Nicholaou T, Wong R, Davis ID. Tumour lysis syndrome in a patient with renal-cell carcinoma treated with sunitinib malate. Lancet 2007; 369:1923 - 4; http://dx.doi.org/10.1016/S0140-6736(07)60903-9; PMID: 17560435
  • Harmon CS, DePrimo SE, Figlin RA, Hudes GR, Hutson TE, Michaelson MD, Négrier S, Kim ST, Huang X, Williams JA, et al. Circulating proteins as potential biomarkers of sunitinib and interferon-α efficacy in treatment-naïve patients with metastatic renal cell carcinoma. Cancer Chemother Pharmacol 2014; 73:151 - 61; http://dx.doi.org/10.1007/s00280-013-2333-4; PMID: 24220935
  • Guermonprez P, Helft J, Claser C, Deroubaix S, Karanje H, Gazumyan A, Darasse-Jèze G, Telerman SB, Breton G, Schreiber HA, et al. Inflammatory Flt3l is essential to mobilize dendritic cells and for T cell responses during Plasmodium infection. Nat Med 2013; 19:730 - 8; http://dx.doi.org/10.1038/nm.3197; PMID: 23685841
  • Sim GC, Martin-Orozco N, Jin L, Yang Y, Wu S, Washington E, Sanders D, Lacey C, Wang Y, Vence L, et al. IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients. J Clin Invest 2014; 124:99 - 110; http://dx.doi.org/10.1172/JCI46266; PMID: 24292706
  • Mortha A, Chudnovskiy A, Hashimoto D, Bogunovic M, Spencer SP, Belkaid Y, Merad M. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science 2014; 343:1249288; http://dx.doi.org/10.1126/science.1249288; PMID: 24625929
  • West EE, Jin HT, Rasheed AU, Penaloza-Macmaster P, Ha SJ, Tan WG, Youngblood B, Freeman GJ, Smith KA, Ahmed R. PD-L1 blockade synergizes with IL-2 therapy in reinvigorating exhausted T cells. J Clin Invest 2013; 123:2604 - 15; http://dx.doi.org/10.1172/JCI67008; PMID: 23676462
  • Omori R, Eguchi J, Hiroishi K, Ishii S, Hiraide A, Sakaki M, Doi H, Kajiwara A, Ito T, Kogo M, et al. Effects of interferon-α-transduced tumor cell vaccines and blockade of programmed cell death-1 on the growth of established tumors. Cancer Gene Ther 2012; 19:637 - 43; http://dx.doi.org/10.1038/cgt.2012.42; PMID: 22790963
  • Prieto PA, Yang JC, Sherry RM, Hughes MS, Kammula US, White DE, Levy CL, Rosenberg SA, Phan GQ. CTLA-4 blockade with ipilimumab: long-term follow-up of 177 patients with metastatic melanoma. Clin Cancer Res 2012; 18:2039 - 47; http://dx.doi.org/10.1158/1078-0432.CCR-11-1823; PMID: 22271879
  • Vacchelli E, Eggermont A, Fridman WH, Galon J, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Adoptive cell transfer for anticancer immunotherapy. Oncoimmunology 2013; 2:e24238; http://dx.doi.org/10.4161/onci.24238; PMID: 23762803
  • Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE Jr., Davidson NE, Tan-Chiu E, Martino S, Paik S, Kaufman PA, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med 2005; 353:1673 - 84; http://dx.doi.org/10.1056/NEJMoa052122; PMID: 16236738
  • Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, Gianni L, Baselga J, Bell R, Jackisch C, et al, Herceptin Adjuvant (HERA) Trial Study Team. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med 2005; 353:1659 - 72; http://dx.doi.org/10.1056/NEJMoa052306; PMID: 16236737
  • Mavilio D, Galluzzi L, Lugli E. Novel multifunctional antibody approved for the treatment of breast cancer. Oncoimmunology 2013; 2:e24567; http://dx.doi.org/10.4161/onci.24567; PMID: 23802090
  • Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T, et al, ToGA Trial Investigators. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010; 376:687 - 97; http://dx.doi.org/10.1016/S0140-6736(10)61121-X; PMID: 20728210
  • Gianni L, Eiermann W, Semiglazov V, Manikhas A, Lluch A, Tjulandin S, Zambetti M, Vazquez F, Byakhow M, Lichinitser M, et al. Neoadjuvant chemotherapy with trastuzumab followed by adjuvant trastuzumab versus neoadjuvant chemotherapy alone, in patients with HER2-positive locally advanced breast cancer (the NOAH trial): a randomised controlled superiority trial with a parallel HER2-negative cohort. Lancet 2010; 375:377 - 84; http://dx.doi.org/10.1016/S0140-6736(09)61964-4; PMID: 20113825
  • Smith I, Procter M, Gelber RD, Guillaume S, Feyereislova A, Dowsett M, Goldhirsch A, Untch M, Mariani G, Baselga J, et al, HERA study team. 2-year follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer: a randomised controlled trial. Lancet 2007; 369:29 - 36; http://dx.doi.org/10.1016/S0140-6736(07)60028-2; PMID: 17208639
  • Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010; 363:711 - 23; http://dx.doi.org/10.1056/NEJMoa1003466; PMID: 20525992
  • Mavilio D, Lugli E. Inhibiting the inhibitors: Checkpoints blockade in solid tumors. Oncoimmunology 2013; 2:e26535; http://dx.doi.org/10.4161/onci.26535; PMID: 24244910
  • Robert C, Thomas L, Bondarenko I, O’Day S, M D JW, Garbe C, Lebbe C, Baurain JF, Testori A, Grob JJ, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011; 364:2517 - 26; http://dx.doi.org/10.1056/NEJMoa1104621; PMID: 21639810
  • Kloss CC, Condomines M, Cartellieri M, Bachmann M, Sadelain M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol 2013; 31:71 - 5; http://dx.doi.org/10.1038/nbt.2459; PMID: 23242161
  • Schroten C, Kraaij R, Veldhoven JL, Berrevoets CA, den Bakker MA, Ma Q, Sadelain M, Bangma CH, Willemsen RA, Debets R. T cell activation upon exposure to patient-derived tumor tissue: a functional assay to select patients for adoptive T cell therapy. J Immunol Methods 2010; 359:11 - 20; http://dx.doi.org/10.1016/j.jim.2010.04.006; PMID: 20460126
  • Modak S, Kushner BH, Kramer K, Vickers A, Cheung IY, Cheung NK. Anti-GD2 antibody 3F8 and barley-derived (1 → 3),(1 → 4)-β-D-glucan: A Phase I study in patients with chemoresistant neuroblastoma. Oncoimmunology 2013; 2:e23402; http://dx.doi.org/10.4161/onci.23402; PMID: 23802080
  • Govindan SV, Goldenberg DM. Designing immunoconjugates for cancer therapy. Expert Opin Biol Ther 2012; 12:873 - 90; http://dx.doi.org/10.1517/14712598.2012.685153; PMID: 22679911
  • Albertini MR, Hank JA, Gadbaw B, Kostlevy J, Haldeman J, Schalch H, Gan J, Kim K, Eickhoff J, Gillies SD, et al. Phase II trial of hu14.18-IL2 for patients with metastatic melanoma. Cancer Immunol Immunother 2012; 61:2261 - 71; http://dx.doi.org/10.1007/s00262-012-1286-5; PMID: 22678096
  • Pasche N, Neri D. Immunocytokines: a novel class of potent armed antibodies. Drug Discov Today 2012; 17:583 - 90; http://dx.doi.org/10.1016/j.drudis.2012.01.007; PMID: 22289353
  • Brack SS, Silacci M, Birchler M, Neri D. Tumor-targeting properties of novel antibodies specific to the large isoform of tenascin-C. Clin Cancer Res 2006; 12:3200 - 8; http://dx.doi.org/10.1158/1078-0432.CCR-05-2804; PMID: 16707621
  • Gutbrodt KL, Schliemann C, Giovannoni L, Frey K, Pabst T, Klapper W, Berdel WE, Neri D. Antibody-based delivery of interleukin-2 to neovasculature has potent activity against acute myeloid leukemia. Sci Transl Med 2013; 5:ra118; http://dx.doi.org/10.1126/scitranslmed.3006221; PMID: 24005158
  • Spitaleri G, Berardi R, Pierantoni C, De Pas T, Noberasco C, Libbra C, González-Iglesias R, Giovannoni L, Tasciotti A, Neri D, et al. Phase I/II study of the tumour-targeting human monoclonal antibody-cytokine fusion protein L19-TNF in patients with advanced solid tumours. J Cancer Res Clin Oncol 2013; 139:447 - 55; http://dx.doi.org/10.1007/s00432-012-1327-7; PMID: 23160853
  • Johannsen M, Spitaleri G, Curigliano G, Roigas J, Weikert S, Kempkensteffen C, Roemer A, Kloeters C, Rogalla P, Pecher G, et al. The tumour-targeting human L19-IL2 immunocytokine: preclinical safety studies, phase I clinical trial in patients with solid tumours and expansion into patients with advanced renal cell carcinoma. Eur J Cancer 2010; 46:2926 - 35; http://dx.doi.org/10.1016/j.ejca.2010.07.033; PMID: 20797845
  • Vacchelli E, Vitale I, Tartour E, Eggermont A, Sautès-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Anticancer radioimmunotherapy. Oncoimmunology 2013; 2:e25595; http://dx.doi.org/10.4161/onci.25595; PMID: 24319634
  • Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, et al, IMPACT Study Investigators. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010; 363:411 - 22; http://dx.doi.org/10.1056/NEJMoa1001294; PMID: 20818862
  • PD-1 inhibitors raise survival in NSCLC. Cancer Discov 2014; 4:6.
  • Weintraub K. Drug development: Releasing the brakes. Nature 2013; 504:S6 - 8; http://dx.doi.org/10.1038/504S6a; PMID: 24352363
  • Dranoff G. Immunotherapy at large: Balancing tumor immunity and inflammatory pathology. Nat Med 2013; 19:1100 - 1; http://dx.doi.org/10.1038/nm.3335; PMID: 24013749
  • Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 2013; 369:134 - 44; http://dx.doi.org/10.1056/NEJMoa1305133; PMID: 23724846
  • Chen LL, Gouw L, Sabripour M, Hwu WJ, Benjamin RS. Combining targeted therapy with immunotherapy (interferon-α): Rational, efficacy in gastrointestinal stromal tumor model and implications in other malignancies. Oncoimmunology 2012; 1:773 - 6; http://dx.doi.org/10.4161/onci.19729; PMID: 22934279
  • Yang Y, Lizée G, Hwu P. Strong emerging rationale for combining oncogene-targeted agents with immunotherapy. Oncoimmunology 2013; 2:e22730; http://dx.doi.org/10.4161/onci.22730; PMID: 23524978
  • Feldhahn N, Arutyunyan A, Stoddart S, Zhang B, Schmidhuber S, Yi SJ, Kim YM, Groffen J, Heisterkamp N. Environment-mediated drug resistance in Bcr/Abl-positive acute lymphoblastic leukemia. Oncoimmunology 2012; 1:618 - 29; http://dx.doi.org/10.4161/onci.20249; PMID: 22934254
  • Kim DK, Lee TV, Castilleja A, Anderson BW, Peoples GE, Kudelka AP, Murray JL, Sittisomwong T, Wharton JT, Kim JW, et al. Folate binding protein peptide 191-199 presented on dendritic cells can stimulate CTL from ovarian and breast cancer patients. Anticancer Res 1999; 19:4B 2907 - 16; PMID: 10652572
  • Peoples GE, Anderson BW, Lee TV, Murray JL, Kudelka AP, Wharton JT, Ioannides CG. Vaccine implications of folate binding protein, a novel cytotoxic T lymphocyte-recognized antigen system in epithelial cancers. Clin Cancer Res 1999; 5:4214 - 23; PMID: 10632363
  • Okada H, Kalinski P, Ueda R, Hoji A, Kohanbash G, Donegan TE, Mintz AH, Engh JA, Bartlett DL, Brown CK, et al. Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with alpha-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J Clin Oncol 2011; 29:330 - 6; http://dx.doi.org/10.1200/JCO.2010.30.7744; PMID: 21149657
  • Okada H, Lieberman FS, Walter KA, Lunsford LD, Kondziolka DS, Bejjani GK, Hamilton RL, Torres-Trejo A, Kalinski P, Cai Q, et al. Autologous glioma cell vaccine admixed with interleukin-4 gene transfected fibroblasts in the treatment of patients with malignant gliomas. J Transl Med 2007; 5:67; http://dx.doi.org/10.1186/1479-5876-5-67; PMID: 18093335
  • Holcmann M, Drobits B, Sibilia M. How imiquimod licenses plasmacytoid dendritic cells to kill tumors. Oncoimmunology 2012; 1:1661 - 3; http://dx.doi.org/10.4161/onci.22033; PMID: 23264929
  • Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology 2012; 1:699 - 716; http://dx.doi.org/10.4161/onci.20696; PMID: 22934262
  • Zauderer MG, Krug LM. Novel therapies in phase II and III trials for malignant pleural mesothelioma. J Natl Compr Canc Netw 2012; 10:42 - 7; PMID: 22223868
  • Krug LM, Dao T, Brown AB, Maslak P, Travis W, Bekele S, Korontsvit T, Zakhaleva V, Wolchok J, Yuan J, et al. WT1 peptide vaccinations induce CD4 and CD8 T cell immune responses in patients with mesothelioma and non-small cell lung cancer. Cancer Immunol Immunother 2010; 59:1467 - 79; http://dx.doi.org/10.1007/s00262-010-0871-8; PMID: 20532500
  • Tarhini AA, Leng S, Moschos SJ, Yin Y, Sander C, Lin Y, Gooding WE, Kirkwood JM. Safety and immunogenicity of vaccination with MART-1 (26-35, 27L), gp100 (209-217, 210M), and tyrosinase (368-376, 370D) in adjuvant with PF-3512676 and GM-CSF in metastatic melanoma. J Immunother 2012; 35:359 - 66; http://dx.doi.org/10.1097/CJI.0b013e31825481fe; PMID: 22495394
  • Aarntzen EH, De Vries IJ, Lesterhuis WJ, Schuurhuis D, Jacobs JF, Bol K, Schreibelt G, Mus R, De Wilt JH, Haanen JB, et al. Targeting CD4(+) T-helper cells improves the induction of antitumor responses in dendritic cell-based vaccination. Cancer Res 2013; 73:19 - 29; http://dx.doi.org/10.1158/0008-5472.CAN-12-1127; PMID: 23087058
  • Aarntzen EH, Bol K, Schreibelt G, Jacobs JF, Lesterhuis WJ, Van Rossum MM, Adema GJ, Figdor CG, Punt CJ, De Vries IJ. Skin-test infiltrating lymphocytes early predict clinical outcome of dendritic cell-based vaccination in metastatic melanoma. Cancer Res 2012; 72:6102 - 10; http://dx.doi.org/10.1158/0008-5472.CAN-12-2479; PMID: 23010076
  • Su S, Zhou H, Xue M, Liu JY, Ding L, Cao M, Zhou ZX, Hu HM, Wang LX. Anti-tumor efficacy of a hepatocellular carcinoma vaccine based on dendritic cells combined with tumor-derived autophagosomes in murine models. Asian Pac J Cancer Prev 2013; 14:3109 - 16; http://dx.doi.org/10.7314/APJCP.2013.14.5.3109; PMID: 23803088
  • Li Y, Wang LX, Pang P, Cui Z, Aung S, Haley D, Fox BA, Urba WJ, Hu HM. Tumor-derived autophagosome vaccine: mechanism of cross-presentation and therapeutic efficacy. Clin Cancer Res 2011; 17:7047 - 57; http://dx.doi.org/10.1158/1078-0432.CCR-11-0951; PMID: 22068657
  • Twitty CG, Jensen SM, Hu HM, Fox BA. Tumor-derived autophagosome vaccine: induction of cross-protective immune responses against short-lived proteins through a p62-dependent mechanism. Clin Cancer Res 2011; 17:6467 - 81; http://dx.doi.org/10.1158/1078-0432.CCR-11-0812; PMID: 21810919
  • Muhsin M, Graham J, Kirkpatrick P. Bevacizumab. Nat Rev Drug Discov 2004; 3:995 - 6; http://dx.doi.org/10.1038/nrd1601; PMID: 15645606
  • Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov 2004; 3:391 - 400; http://dx.doi.org/10.1038/nrd1381; PMID: 15136787
  • Lim SN, Kuhn S, Hyde E, Ronchese F. Combined TLR stimulation with Pam3Cys and Poly I: C enhances Flt3-ligand dendritic cell activation for tumor immunotherapy. J Immunother 2012; 35:670 - 9; http://dx.doi.org/10.1097/CJI.0b013e318270e135; PMID: 23090076
  • Hennies CM, Reboulet RA, Garcia Z, Nierkens S, Wolkers MC, Janssen EM. Selective expansion of merocytic dendritic cells and CD8DCs confers anti-tumour effect of Fms-like tyrosine kinase 3-ligand treatment in vivo. Clin Exp Immunol 2011; 163:381 - 91; http://dx.doi.org/10.1111/j.1365-2249.2010.04305.x; PMID: 21235535
  • Curran MA, Allison JP. Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors. Cancer Res 2009; 69:7747 - 55; http://dx.doi.org/10.1158/0008-5472.CAN-08-3289; PMID: 19738077
  • Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology 2012; 1:306 - 15; http://dx.doi.org/10.4161/onci.19549; PMID: 22737606
  • Linehan DC, Tan MC, Strasberg SM, Drebin JA, Hawkins WG, Picus J, Myerson RJ, Malyapa RS, Hull M, Trinkaus K, et al. Adjuvant interferon-based chemoradiation followed by gemcitabine for resected pancreatic adenocarcinoma: a single-institution phase II study. Ann Surg 2008; 248:145 - 51; http://dx.doi.org/10.1097/SLA.0b013e318181e4e9; PMID: 18650621
  • Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy. Science 2013; 342:1432 - 3; http://dx.doi.org/10.1126/science.342.6165.1432; PMID: 24357284
  • Kroemer G, Zitvogel L. Abscopal but desirable: The contribution of immune responses to the efficacy of radiotherapy. Oncoimmunology 2012; 1:407 - 8; http://dx.doi.org/10.4161/onci.20074; PMID: 22754758
  • Galluzzi L, Kepp O, Kroemer G. Immunogenic cell death in radiation therapy. Oncoimmunology 2013; 2:e26536; http://dx.doi.org/10.4161/onci.26536; PMID: 24404424
  • Ahmadzadeh M, Rosenberg SA. IL-2 administration increases CD4+ CD25(hi) Foxp3+ regulatory T cells in cancer patients. Blood 2006; 107:2409 - 14; http://dx.doi.org/10.1182/blood-2005-06-2399; PMID: 16304057
  • Cesana GC, DeRaffele G, Cohen S, Moroziewicz D, Mitcham J, Stoutenburg J, Cheung K, Hesdorffer C, Kim-Schulze S, Kaufman HL. Characterization of CD4+CD25+ regulatory T cells in patients treated with high-dose interleukin-2 for metastatic melanoma or renal cell carcinoma. J Clin Oncol 2006; 24:1169 - 77; http://dx.doi.org/10.1200/JCO.2005.03.6830; PMID: 16505437
  • Wei S, Kryczek I, Edwards RP, Zou L, Szeliga W, Banerjee M, Cost M, Cheng P, Chang A, Redman B, et al. Interleukin-2 administration alters the CD4+FOXP3+ T-cell pool and tumor trafficking in patients with ovarian carcinoma. Cancer Res 2007; 67:7487 - 94; http://dx.doi.org/10.1158/0008-5472.CAN-07-0565; PMID: 17671219
  • Brandenburg S, Takahashi T, de la Rosa M, Janke M, Karsten G, Muzzulini T, Orinska Z, Bulfone-Paus S, Scheffold A. IL-2 induces in vivo suppression by CD4(+)CD25(+)Foxp3(+) regulatory T cells. Eur J Immunol 2008; 38:1643 - 53; http://dx.doi.org/10.1002/eji.200737791; PMID: 18493984
  • Camisaschi C, Filipazzi P, Tazzari M, Casati C, Beretta V, Pilla L, Patuzzo R, Maurichi A, Cova A, Maio M, et al. Effects of cyclophosphamide and IL-2 on regulatory CD4+ T cell frequency and function in melanoma patients vaccinated with HLA-class I peptides: impact on the antigen-specific T cell response. Cancer Immunol Immunother 2013; 62:897 - 908; http://dx.doi.org/10.1007/s00262-013-1397-7; PMID: 23589107
  • Weiss L, Letimier FA, Carriere M, Maiella S, Donkova-Petrini V, Targat B, Benecke A, Rogge L, Levy Y. In vivo expansion of naive and activated CD4+CD25+FOXP3+ regulatory T cell populations in interleukin-2-treated HIV patients. Proc Natl Acad Sci U S A 2010; 107:10632 - 7; http://dx.doi.org/10.1073/pnas.1000027107; PMID: 20498045
  • Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, Sautès-Fridman C, Ma Y, Tartour E, Zitvogel L, et al. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology 2012; 1:1323 - 43; http://dx.doi.org/10.4161/onci.22009; PMID: 23243596

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.