Skip to main content
Oncoimmunology logoLink to Oncoimmunology
. 2013 May 7;2(7):e24850. doi: 10.4161/onci.24850

Trial Watch

Immunostimulatory cytokines

Erika Vacchelli 1,2,3, Alexander Eggermont 2, Wolf Hervé Fridman 4,5,6, Jérôme Galon 6,7,8,9, Laurence Zitvogel 2,10, Guido Kroemer 3,4,6,11,12,†,*, Lorenzo Galluzzi 1,4,11,†,*
PMCID: PMC3782010  PMID: 24073369

Abstract

During the past two decades, the notion that cancer would merely constitute a cell-intrinsic disease has gradually been complemented by a model postulating that the immune system plays a relevant role during all stages of oncogenesis and tumor progression. Along with this conceptual shift, several strategies have been devised to stimulate tumor-specific immune responses, including relatively unselective approaches such as the systemic administration of adjuvants or immunomodulatory cytokines. One year ago, in the July issue of OncoImmunology, we described the main biological features of this large group of proteins and discussed the progress of ongoing clinical studies evaluating their safety and therapeutic potential in cancer patients. Here, we summarize the latest developments in this area of clinical research, focusing on high impact studies that have been published during the last 13 mo and clinical trials launched in the same period to investigate which cytokines can be employed as safe and efficient immunostimulatory interventions against cancer.

Keywords: GM-CSF, IFN, IL-2, TGFβ, TNFα, chemokines

Introduction

More than 130 distinct cytokines have been characterized so far, making up a highly pleiotropic signaling system that can operate in an autocrine, paracrine or endocrine fashion.1-4 Such a pleiotropism at least in part originates from (1) the highly heterogeneous nature of the cytokine family taken as a whole; (2) the fact that cytokines are produced by a wide variety of cell types throughout the body, including immune, epithelial, endothelial and stromal cells; (3) the existence of several receptors and/or receptor isoforms that exhibit different affinity for the same cytokine, which frequently are expressed by distinct cell types; (4) the fact that cytokines are often involved in feedback and feedforward circuitries that control the secretion and release of other biologically active molecules, including additional cytokines; and (5) the fact that the biological activity of a given cytokine is highly context-dependent, that is, it is influenced by several cell-intrinsic and cell-extrinsic variables, first of all the presence of other cytokines.5-7 Thus, taken together cytokines regulate (in a partially redundant and overlapping fashion) biological functions as diverse as proliferation, differentiation, chemotaxis, inflammation, (innate and adaptive) immune responses and cell death.

Besides controlling physiological processes such as hematopoiesis,8 cytokines play a critical role in organismal responses to various danger conditions, including infection by pathogens and oncogenesis. In this context, specific cytokine cascades have evolved to sequentially promote the neutralization/removal of the triggering stimulus, allow for tissue repair, and—along with the restoration of homeostasis—self-extinguish.9 In some cases, however, the initial stimulus cannot be efficiently removed and cytokine production becomes chronic, driving (at least some extent of) tissue damage. Such sites of chronic inflammation are particularly relevant for oncogenesis as they represent abundant sources of potentially mutagenic products (e.g., reactive oxygen species) as well as of mitogenic, anti-apoptotic and angiogenic cytokine cocktails.10-12 Thus, the administration of immunomodulatory cytokines may not only provoke acute toxicities (often mimicking a state of severe infection), but also exert—at least theoretically—bona fide carcinogenic effects. Both these possibilities must be taken under attentive consideration for the development of safe immunotherapeutic regimens based on immunostimulatory cytokines.

Great efforts have been made in this direction during the past two decades, along with the realization that tumors do not merely originate from cell-intrinsic defects but evolve in the context of cell-extrinsic alterations, including (among many others) an ever more permissive immune system.13-15 Thus, dozens—if not hundreds—of clinical studies have investigated whether the local or systemic administration of immunostimulatory cytokines—most often in combination with conventional chemotherapeutics—would constitute a safe and efficient maneuver to (re)activate tumor-specific immune responses. In spite of such an intense wave of clinical investigation, which we have extensively discussed 1 y ago,7 only three recombinant cytokines are nowadays licensed by the FDA or other international regulatory agencies for use as immunostimulatory agents to boost anticancer immunity (in patients who are 18 y of age or older): interferon (IFN)-α2a (also known as Roferon-A®), which is indicated for the therapy of hairy cell leukemia and chronic phase, Philadelphia chromosome-positive chronic myelogenous leukemia (CML), upon minimal pretreatment (within 1 y of diagnosis); IFN-α2b (also known as Intron A®), which is employed for the treatment of follicular lymphoma, hairy cell leukemia, AIDS-related Kaposi's sarcoma, multiple myeloma, melanoma, condyloma acuminata and cervical intraepithelial neoplasms; and interleukin (IL)-2 (also known as aldesleukin or Proleukin®), which is used in patients affected by metastatic melanoma and metastatic renal cell carcinoma (source www.fda.gov). A few other recombinant cytokines, including granulocyte colony-stimulating factor (G-CSF, also known as filgrastim, lenograstim or Neupogen®) and granulocyte monocyte colony-stimulating factor (GM-CSF, also known as molgramostim, sargramostim, Leukomax®, Mielogen® or Leukine®) are currently approved for use in cancer patients, yet are mainly employed (1) to facilitate the reconstitution of the immune system upon transplantation (which is often preceded by lymphodepleting/lymphoablating regimens) or aggressive chemotherapy;16-18 (2) as a prophylactic measure to minimize chemotherapy-induced neutropenia, in particular in the context of gemcitabine- (a nucleoside analog) and cisplatin- (a DNA-damaging agent) based regimens;19,20 (3) as a priming stimulus, to recruit possibly quiescent leukemic cells into the cell cycle and hence increase their chemosensitivity;21 or (4) to mobilize bone marrow precursors for autologous stem cell transplantation.22,23

Along the lines of our monthly Trial Watch series,7,24-37 here we will summarize the latest advances on the use of cytokines as immunostimulatory agents to re(activate) tumor-specific immune responses, focusing on high impact studies that have been published during the last 13 mo and clinical trials that have been initiated in the same period to assess the safety and efficacy of this immunotherapeutic approach. In line with the objective of this Trial Watch, neither studies investigating the use of cytokines as immunoreconstituting agents [upon hematopoietic stem cell transplantation, to prevent chemotherapy-associated neutropenia, and as a mitogenic support to adoptive cell transfer or dendritic cell (DC)-based anticancer vaccines] nor studies involving FDA-approved immunostimulatory cytokines (i.e., IFN-α2a, IFN-α2b and IL-2) employed as “on-label” interventions (see above) will be taken into further consideration here. Along similar lines, we will not discuss further therapeutic strategies that involve the use of potentially cytotoxic cytokines, in particular tumor necrosis factor α (TNFα), to directly trigger the demise of malignant cells38-41 or to damage the tumor vasculature.42,43

Literature Update

During the past 13 mo, results from multiple preclinical and clinical studies have corroborated the notion that cytokines play a critical role not only in the development and progression of a wide array of neoplasms, but also in their capability to elicit therapeutic immunity in response to treatment.

One large branch of clinical research has focused on the potential prognostic/predictive value of the circulating or intratumoral levels of distinct cytokines. Thus, the intra-platelet levels of vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF) and platelet factor 4 (PF4) have been proposed as independent predictors of colorectal carcinoma (CRC),44 while the bone marrow of pediatric neuroblastoma patients has been shown to exhibit a significant downregulation of CXCL12, as compared with its normal counterpart, as well as to express an IFN-related gene signature.45 Elevated baseline amounts of circulating CXCL10 have been demonstrated to predict disease relapse and poor overall survival in diffuse large B-cell lymphoma patients.46 High serum levels of CCL17 have been associated with established clinical risk factors and poor response to treatment in subjects affected by Hodgkin’s lymphoma.47 Specific cytokine and angiogenic factor (CAF) profiles have been linked to the efficacy and toxicity of the multitargeted tyrosine kinase inhibitor (TKI) pazopanib in advanced soft-tissue sarcoma patients,48 to disease type and general conditions at diagnosis in pediatric non-Hodgkin’s lymphoma (NHL) patients,49 to the propensity of individuals bearing renal cell carcinoma (RCC) to respond to sorafenib, another relatively unselective TKI,50,51 as well as to the responsiveness of acute myeloid leukemia (AML) patients to the sequential administration of 5-azacytidine (a DNA demethylating agent)52,53 and lenalidomide (a synthetic chemotherapeutic with multiple mechanisms of action).54 High baseline levels of circulating IL-2 and IL-8, but not of hepatocyte growth factor (HGF), IFNγ and osteopontin, have been suggested to constitute independent prognostic factors for head and neck cancer (HNC) patients.55-57 Conversely, high concentrations of TNFα in the serum of HNC patients before treatment have been shown to predict an unfavorable prognosis.57 Elevated amounts of VEGF and angiopoietin 2 in the serum have turned out to constitute independent predictors of survival (but not of sorafenib sensitivity) among patients with advanced hepatocellular carcinoma (HCC).58 Reduced levels of the IL-7 receptor as well as high amounts of the IL-12 receptor β2 on the surface of malignant cells have been associated with improved disease outcome among Stage I lung adenocarcinoma patients.59 Finally, a single nucleotide polymorphism in the promoter of IL10 (influencing IL-10 expression) has been suggested to predict survival and disease relapse among non-small cell lung carcinoma (NSCLC) patients subjected to tumor resection.60 Taken together, these observations are representative of an abundant recent literature (which we cannot exhaustively discuss here owing to space restrictions) demonstrating the critical influence that a large panel of cytokines exerts on the clinical course of cancer.

Alongside, consistent efforts have been devoted to the elucidation of the actual immunostimulatory potential of cytokines in cancer patients, resulting in an equally abundant literature. Some of these clinical reports refer to “on-label” settings, and the underlying studies were intended either to assess the safety and activity profile of FDA-approved cytokines combined with hitherto experimental chemotherapeutic regimens or to generate long-term follow-up data. Thus, Garcia and colleagues have combined GM-CSF with lenalidomide for the treatment of castration-resistant prostate cancer or with IL-2 and IFN-α for the therapy of metastatic RCC, in both cases observing a low incidence of acceptable side effects and modest antitumor activity.61,62 Tarhini et al. have tested the safety and efficacy of IFN-α2b combined with tremelimumab, an investigational monoclonal antibody targeting cytotoxic T lymphocyte-associated antigen 4 (CTLA4), in patients with Stage IV melanoma.63,64 In this context, IFN-α2b plus tremelimumab were associated with low toxicity and promising antitumor effects, which were paralleled by the recovery of effector T-cell (Teff) functions, by a robust downregulation of myeloid-derived suppressor cells (MDSCs), and (less so) by the inhibition of FOXP3+ regulatory T cells (Tregs).63,64 IFN-α has also been tested in combination with atrasentan (a selective endothelin A receptor antagonist) or metronomic chemotherapy in metastatic RCC patients,65,66 and with imatinib (a TKI specific for BCR-ABL, KIT and the PDGF receptor) in CML patients,67 in all settings being associated with acceptable adverse effects and (at least some extent of) antitumor efficacy. Along similar lines, it has been shown that IL-2 can be safely combined with stereotactic body radiation therapy for the treatment of subjects affected by metastatic melanoma or RCC.68 Finally, the long-term results of the randomized Phase III trials EORTC 18952 and 18991, comparing the administration of pegylated IFN-α2b with observation in resected Stage III melanoma patients,69 have been reported.70,71 In this setting (mean follow-up: 7.6 y), adjuvant pegylated IFN-α2b was found to have a positive impact on relapse-free survival that (1) was more modest than that observed earlier (at a mean follow-up of 3.8 y)69,71 and (2) was especially pronounced among patients exhibiting ulcerated lesions and low disease burden.70,71

A consistent number of clinical studies published in the last 13 mo have investigated the use of immunostimulatory cytokines as “off-label” antineoplastic interventions. Thus, the safety and immunostimulatory potential of IFN-α2b, combined with either the intraarterial administration of 5-fluorouracil (a nucleoside analog) or a chemoradiotherapeutic regimen involving 5-fluorouracil and cisplatin have been assessed in cohorts of advanced HCC and resected pancreatic carcinoma patients, respectively.72,73 The pegylated form of IFN-α2b has been evaluated as a standalone therapeutic intervention in children affected by pontine glioma,74 or in combination with imatinib in gastrointestinal stromal tumor (GIST) patients.75 IFN-β has been tested for its antineoplastic potential as a maintenance monotherapy for Stage II and III melanoma patients,76 and as part of a multimodal treatment involving 5-fluorouracil, cisplatin, folinic acid and radiotherapy for young HNC patients.77 In all these settings, IFN-α2b and IFN-β were well tolerated and—in some instances—significantly improved the efficacy of conventional therapeutic regimens. Petrella and colleagues have tested the safety, immunological outcome and antineoplastic activity of IL-21 administered as a standalone therapeutic interventions to metastatic melanoma patients,78 while Steele et al. have investigated whether this cytokine can be safely administered in combination with cetuximab, a monoclonal antibody specific for the epidermal growth factor receptor (EGFR), to metastatic CRC patients.79 In both these scenarios, IL-21 was not associated with severe side effects and induced immunological as well as clinical responses, at least in a fraction of patients. In a Phase I clinical trial, Selectikine, i.e., an immunocytokine consisting in an IL-2 molecule fused to a DNA-specific antibody (hence targeting necrotic tumor cells), turned out to be well tolerated by patients with advanced solid tumors, prompting the initiation of a Phase II study.80 Along similar lines, the systemic administration of h14.18-IL-2 (an immunocytokine comprising IL-2 and a ganglioside GD2-specific monoclonal antibody) to melanoma patients has been associated with an acceptable toxicity, yet also appeared to elicit clinical responses in a limited fraction of individuals.81 Of note, Pautier and colleagues have reported that IL-2 significantly increases the area under the curve and the maximum concentration of imatinib and its main metabolite CGP74588,82 suggesting that the antineoplastic and immunostimulatory effects of imatinib83-85 may be improved by IL-2 via both immunological and pharmacokinetic mechanisms. However, patients receiving IL-2 plus imatinib exhibited mild to moderate adverse effects such as fever, chills, fatigue, nausea and the elevation of hepatic enzymes. Thus, imatinib-associated side effects might also be exacerbated by the co-administration of IL-2.82 Finally, a few studies have evaluated immunostimulatory cytokines as adjuvants for peptide- and DC-based anticancer vaccines, or as a measure to boost the antineoplastic activity of adoptively transferred T cells, often with encouraging results.86-91

Besides such an abundant clinical literature, a number of top quality fundamental reports – unraveling the molecular and cellular cascades whereby cytokines influence oncogenesis, tumor progression and response to therapy – has been published during the last 13 mo. Among major discoveries in the field, (1) TNFα and IFNγ, but neither of these TH1 cytokines alone, have been shown to promote the senescence of malignant cells in vitro and in vivo;92 (2) the tumor stroma has been reported to underpin the resistance of melanoma cells to BRAF inhibitor owing to its ability to secrete high levels of HGF;93,94 (3) HGF has been involved in an autocrine signaling pathway that drives leukemogenesis;95 (4) the deubiquitinating enzyme ubiquitin-specific peptidase 15 (USP15) turned out to stabilize transforming growth factor β (TGFβ) receptor 1, hence promoting the development of glioblastoma via a TGFβ-dependent mechanism;96 (5) defects in the intestinal barrier have been linked to colorectal carcinogenesis through a microbial product-driven signaling cascade involving IL-23, TH17 cells and IL-17;97 (6) 5-fluorouracil and gemcitabine have been shown to trigger the activation of the inflammasome98 in MDSCs, hence initiating an IL-1β-dependent, IL-17-mediated mechanism that eventually promotes chemoresistance;99 (7) IL-22-binding protein (IL-22BP) has been reported to mediate oncosuppressive functions in the intestine by intercepting available IL-22, in particular during the recovery phase of inflammation;100 (8) a subset of CD8+CTLA4+ IL-35-secreting Tregs has been suggested to play a central immunosuppressive role in prostate cancer patients treated with a prostate acid phosphatase (PAP)-specific vaccine, suggesting that IL-35-blocking interventions may exert potent immunostimulatory effects (at least in this setting);101 (9) the administration of IL-2 has been associated with the expansion of naïve CD4+CD25highFOXP3+ Tregs in CRC patients;102 (10) GM-CSF dosing regimens have been shown to influence several immunological parameters in prostate cancer patients, including the abundance and activity of various immune cell subsets as well as the circulating levels of cytokines such as VEGF and TNFα;103 (11) a variant of IL-2 that is able to convey robust proliferative signals in the absence of CD25 has been developed;104 (12) CCL21 gradients have been reported to constitute the major chemotactic drivers of DC migration to lymph nodes in vivo;105 and (13) the tridimensional structure of human CXCR1, one of the high-affinity receptors for IL-8, has been elucidated by nuclear magnetic resonance (NMR) spectroscopy.106

Taken together, these observations suggest that immunostimulatory cytokines continue to attract a consistent amount of interest, both from clinicians and scientists dealing with fundamental research.

Update on Clinical Trials

When this Trial Watch was being redacted (April 2013), official sources listed no less than 116 clinical trials launched after April 1, 2012, to investigate the safety and therapeutic potential of cytokines in cancer patients (source www.clinicaltrials.gov). Of these, 89 trials involved IL-2 or other interleukins (24 studies), GM-CSF (5 studies), G-CSF (50 studies) and IFNs (10 studies) as fully “on-label” interventions, and therefore will not be discussed further here. Of the remaining 27 clinical trials, 9 aimed at assessing the immunostimulatory potential of IL-2 (as an “off-label” medication) and of other—hitherto investigational—interleukins (i.e., IL-11, IL-15, IL-18 and IL-21); 12 were launched to assess whether GM-CSF might be safely and efficiently used to boost tumor-specific immune responses, most often as induced by anticancer vaccines; and 7 were initiated to test IFN-α2b and IFNβ, again in “off-label” oncological settings (Table 1).

Table 1. Recent clinical trials evaluating the immunostimulatory potential of cytokines for cancer therapy*.

Cytokine Indication(s) Status Phase Route Co-therapy Ref.
GM-CSF
Breast cancer
Recruiting
n.a.
s.c.
Combined with a
multipeptide vaccine
NCT01660529
Chemoradiotherapy-associated mucositis
Recruiting
II
Mouthwash solution
Combined with vitamin B12
NCT01806272
Melanoma
Recruiting
II/III
n.a.
Combined with CSF470 and BCG
NCT01729663
MM
Recruiting
n.a.
s.c.
Combined with
WT1 analog peptide
NCT01827137
NB
Not yet recruiting
II
i.v.
Combined with ch14.18,
irinotecan and temozolomide
NCT01767194
Recruiting
I
n.a.
Combined with hu3F8
NCT01757626
I/II
i.v.
Combined with ch14.18,
IL-2 and isotretinoin
NCT01592045
NSCLC
Not yet recruiting
III
s.c.
Combined with a
hTERT-derived peptide
NCT01579188
Recruiting
I/II
s.c.
Combined with a
hTERT-derived peptide
NCT01789099
Ovarian cancer
Recruiting
I
s.c.
Combined with a
FBP-derived peptide
NCT01580696
Prostate cancer
Recruiting
I/II
s.c.
Combined with a
hTERT-derived peptide
NCT01784913
Reproductive tract cancer
Active not recruiting
I
s.c.
Combined with decitabine, PLD
and a NY-ESO-1-derived peptide
NCT01673217
IFNα-2a
Lymphoma
Completed
III
s.c.
Combined with rituximab
NCT01609010
RCC
Recruiting
II
n.a.
Combined with bevacizumab, everolimus and a TKI
NCT01731158
IFNα-2b
High-grade gliomas
Recruiting
III
n.a.
Combined with
radiotherapy and temozolomide
NCT01765088
Ovarian cancer
Recruiting
I/II
s.c.
Combined with standard chemotherapy and tocilizumab
NCT01637532
n.a.
Combined with gemcitabine
and a p53-derived SLP
NCT01639885
Solid tumors
Recruiting
II
n.a.
Combined with 5-FU
NCT01658813
IFNβ
Merkel cell carcinoma
Recruiting
I/II
i.t.
Combined with autologous T-cell transplantation and IL-2
NCT01758458
IL-2
Gastresophageal cancer
Recruiting
I
n.a.
Combined with IMAB362
and zoledronic acid
NCT01671774
Lung metastases
Recruiting
I/II
Intranasal nebulization
As single agent
NCT01590069
NB
Recruiting
I
s.c.
Combined with hu3F8
NCT01662804
I/II
i.v.
Combined with ch14.18,
GM-CSF and isotretinoin
NCT01592045
s.c.
Combined with ch14.18/CHO
and isotretinoin
NCT01701479
IL-15
HNC
Melanoma
NSCLC
RCC
Not yet recruiting
I
s.c.
As single agent
NCT01727076
Solid and hematological tumors
Recruiting
I
i.v.
As single agent
NCT01572493
IL-18
NHL
Recruiting
I
n.a.
Combined with ofatumumab
NCT01768338
 
IL-21 Solid tumors Recruiting I i.v. Combined with anti-PD1 mAbs NCT01629758  

5-FU, 5-fluorouracil; BCG, bacillus Calmette-Guérin; FBP, folate-binding protein; GM-CSF, granulocyte macrophage colony-stimulating factor; HNC, head and neck cancer; hTERT, human telomerase reverse transcriptase; IFN, interferon; IL, interleukin; i.t., intra tumorem; i.v., intra venam; mAb, monoclonal antibody; MM, multiple myeloma; n.a. not available; NB, neuroblastoma; NHL, Non-Hodgkin's lymphoma; NSCLC, non-small cell lung carcinoma; PD1, programmed death 1; PLD, pegylated liposomal doxorubicin; RCC, renal cell carcinoma; s.c., sub cutem; SLP, synthetic long peptide; TKI, tyrosine kinase inhibitor; WT1, Wilms tumor 1. *Limited to trials dealing with investigational cytokines or FDA-approved cytokines employed as “off-label” medications, started after April 1, 2012.

In particular, IL-2 is being tested (1) as a single agent, upon intranasal nebulization, for the treatment of patients bearing pulmonary metastases of sarcoma (NCT01590069); (2) in combination with IMAB362 (a monoclonal antibody specific for the gastric differentiation protein claudin 18, splice variant 2)107 and zoledronic acid (a bisphosphonate), for the therapy of subjects with adenocarcinomas of the stomach, esophagus and gastresophagic junction (NCT01671774); and (3) in combination with monoclonal antibodies targeting ganglioside GD2 (i.e., ch14.18, ch14.18/CHO and hu3F8), isotretinoin (a retinoid with antineoplastic properties) and GM-CSF (in 1 study), upon intravenous or subcutaneous delivery, for the treatment of neuroblastoma patients (NCT01592045; NCT01662804; NCT01701479). The safety and immunostimulatory potential of IL-15, given s.c. or i.v. as a standalone therapeutic intervention, are being evaluated in a mixed cohort of HNC, NSCLC, RCC and melanoma patients (NCT01727076) as well as in subjects affected by advanced hematopoietic and solid neoplasms (NCT01572493). In addition, IL-18 is being tested in combination with ofatumumab (a monoclonal antibody specific for CD20)108 in lymphoma patients who have undergone autologous peripheral blood stem cell transplantation (NCT01768338), while the therapeutic activity of intravenous IL-21 plus BMS-936558 (a PD1-blocking monoclonal antibody)109-111 is being investigated in individuals with advanced or metastatic solid tumors (NCT01629758). All these clinical trials but one (NCT01727076) are currently recruiting participants.

With the exceptions of NCT01592045, NCT01757626, NCT01767194 and NCT01806272, investigating whether GM-CSF can be conveniently combined either with anti-GD2 monoclonal antibodies, alone or together with irinotecan (an inhibitor of topoisomerase 1) and temozolomide (an alkylating agent), or with vitamin B12 for the treatment of neuroblastoma patients or chemoradiation-associated mucositis, respectively, all clinical trials launched during the last 13 mo to test “off-label” applications of GM-CSF involve anticancer vaccines. In particular, GM-CSF is being tested for its ability to boost immune responses as elicited by (1) a Wilms tumor 1 (WT1)-targeting vaccine in patients with multiple myeloma (NCT01827137); (2) a multipeptide vaccine in individuals affected by metastatic breast carcinoma (NCT01660529); (3) a telomerase-specific vaccine in subjects with inoperable Stage III NSCLC (NCT01579188); (4) a mixture of four, lethally irradiated melanoma cell lines combined with the bacillus Calmette-Guérin112 in Stage IIB, IIC and III melanoma patients (NCT01729663); (5) a telomerase-derived synthetic peptide in NSCLC (NCT01789099) and prostate cancer patients (NCT01784913); (6) a folate-binding protein (FBP)-targeting vaccine in ovarian carcinoma patients (NCT01580696); and (7) a NY-ESO-1-derived peptide in subjects with recurrent ovarian epithelial cancer, fallopian tube cancer or peritoneal cancer (NCT01673217). These trials are recruiting participants, with the exception of NCT01579188, NCT01673217 and NCT01767194.

IFN-α2a in combination with bevacizumab (a monoclonal antibody targeting VEGF) is being tested as a first-line intervention in RCC patients who are allocated to receive either everolimus (an inhibitor of the mammalian target of rapamycin, mTOR)113 followed by one TKI among sunitinib, sorafenib and pazopanib, or vice versa (NCT01731158). In addition, a clinical trial evaluating subcutaneous IFN-α2a plus rituximab (a CD20-targeting monoclonal antibody)114 in subjects with follicular lymphoma or other CD20+ indolent lymphomas that had originally been registered at www.clinicaltrials.gov on May 29, 2012, (NCT01609010) has already been completed (though results are not yet available). IFN-α2b is being investigated as an immunostimulatory intervention (1) in patients with newly diagnosed high-grade gliomas (i.e., anaplastic oligoastrocytoma, anaplastic astrocytoma, glioblastoma), combined with temozolomide upon radiotherapy (NCT01765088); (2) in subjects bearing recurrent or chemoresistant ovarian cancer, either in combination with tocilizumab (a monoclonal antibody specific for the IL-6 receptor) and carboplatin-based chemotherapy115 (NCT01637532), or together with gemcitabine and a p53-derived synthetic long peptide (NCT01639885); and (3) in previously-treated patients bearing metastatic gastrointestinal cancer, NSCLC or RCC, following a 5-fluorouracil-based therapeutic regimen (NCT01658813). Finally, IFN-β is being investigated for its ability to boost the antineoplastic activity of autologous T cells genetically manipulated to target tumor cells in individuals affected by metastatic Merkel cell carcinoma, a virus-associated cancer116 (NCT01758458). With the single exception of NCT01609010, all these studies are currently recruiting participants.

Of note, 21 clinical trials started after January 1, 2008, (which were included in the latest Trial Watch dealing with immunostimulatory cytokines)7 have been terminated (NCT00819169, for sponsor decision; NCT00724061, due to low accrual), suspended (NCT00923351; NCT01334515) or completed (NCT00602706; NCT00607048; NCT00609076; NCT00619268; NCT00626405; NCT00785122; NCT00796757; NCT00806598; NCT00822770; NCT00836407; NCT00939510; NCT00952237; NCT00990054; NCT01074060; NCT01082887; NCT01107756; NCT01324063) during the last 13 mo (source www.clinicaltrials.gov). Among completed studies, NCT00607048 and NCT00939510 are listed as having results, which however are not yet publicly available.

Concluding Remarks

Cytokines constitute a highly heterogeneous group of extracellular messengers that—taken as a whole—exerts an incredibly large range of biological effects, de facto regulating virtually all cellular functions.1-4 In line with this notion, during the past four decades great efforts have been devoted not only to elucidating the molecular and cellular circuitries that are set in motion by specific cytokines (or cytokine cocktails), but also to understanding whether cytokines can be safely and efficiently used to treat a variety of human conditions, including cancer.10,117 Nowadays, three distinct cytokines (i.e., IFN-α2a, IFN-α2b and IL-2) are approved by the FDA or other international regulatory agencies for use as therapeutic immunostimulants against cancer.10,117 In addition, cytokines such as G-CSF ad GM-CSF are part of the clinical routine as they potently favor immunoreconstitution in transplanted patients or in subjects who underwent aggressive chemotherapy.8,118

Arguably, the major obstacle against the clinical use of cytokines stems from their elevated pleiotropism, implying that these proteins may be intrinsically prone to elicit adverse effects, in particular upon systemic administration. For instance, high-dose IL-2 has been associated with tumor regression in a fraction of melanoma and RCC patients,119,120 yet this regimen also elicits moderate to severe toxicities in some individuals.121 To circumvent this issue, future efforts will have to focus at narrowing the spectrum of activity of particularly promising cytokines, tipping the balance away from a potentially dangerous pleiotropism and toward a highly specific biological activity. Immunocytokines, i.e., cytokines specifically delivered to target cells by means of monoclonal antibodies, constitute a promising approach in this sense, and some of them, such as Selectikine, have already entered clinical trials.122,123 In addition, it will be crucial to identify means to expand the clinical benefits of immunostimulatory cytokines—which currently are limited to individuals with highly immunosensitive tumors such as melanoma and RCC—to an ever larger population of cancer patients. Strategies to be pursued in this direction include the development of combinatorial therapeutic regimens involving Toll-like receptor (TLR) agonists,29,30 immune checkpoint-blocking antibodies27,28,124,125 or immunogenic chemotherapeutics.33,34,126 Well designed clinical trials are required to formally identify which of these approaches, if any, will successfully increase the number of cancer patients obtaining a clinical benefit from immunostimulatory cytokines.

Acknowledgments

Authors are supported by the European Commission (ArtForce); Agence National de la Recherche (ANR); Ligue Nationale contre le Cancer; Fondation pour la Recherche Médicale (FRM); Institut National du Cancer (INCa); Association pour la Recherche sur le Cancer (ARC), LabEx Immuno-Oncologie; Fondation de France; Fondation Bettencourt-Schueller; AXA Chair for Longevity Research; Cancéropôle Ile-de-France, Paris Alliance of Cancer Research Institutes (PACRI) and Cancer Research for Personalized Medicine (CARPEM).

Glossary

Abbreviations:

AML

acute myeloid leukemia

CAF

cytokine and angiogenic factor

CML

chronic myelogenous leukemia

CRC

colorectal carcinoma

CTLA4

cytotoxic T lymphocyte-associated antigen 4

DC

dendritic cell

EGFR

epidermal growth factor receptor

FBP

folate-binding protein

G-CSF

granulocyte colony-stimulating factor

GIST

gastrointestinal stromal tumor

GM-CSF

granulocyte monocyte colony-stimulating factor

HCC

hepatocellular carcinoma

HGF

hepatocyte growth factor

HNC

head and neck cancer

IFN

interferon

IL

interleukin

MDSC

myeloid-derived suppressor cell

NMR

nuclear magnetic resonance

NSCLC

non-small cell lung carcinoma

NHL

non-Hodgkin’s lymphoma

PAP

prostate acid phosphatase

PDGF

platelet-derived growth factor

PF4

platelet factor 4

RCC

renal cell carcinoma

Teff

effector T cell

TGFβ

transforming growth factor β

TKI

tyrosine kinase inhibitor

TLR

Toll-like receptor

TNFα

tumor necrosis factor α

Treg

FOXP3+ regulatory T cell

USP15

ubiquitin-specific peptidase 15

VEGF

vascular endothelial growth factor

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Footnotes

These authors contributed equally to this work.

References

  • 1.Tato CM, Cua DJ. SnapShot: Cytokines I. Cell. 2008;132:324–e1, e1. doi: 10.1016/j.cell.2008.01.001. [DOI] [PubMed] [Google Scholar]
  • 2.Tato CM, Cua DJ. SnapShot: cytokines II. Cell. 2008;132:500. doi: 10.1016/j.cell.2008.01.002. [DOI] [PubMed] [Google Scholar]
  • 3.Tato CM, Cua DJ. SnapShot: cytokines III. Cell. 2008;132:900. doi: 10.1016/j.cell.2008.02.023. [DOI] [PubMed] [Google Scholar]
  • 4.Tato CM, Cua DJ. SnapShot: Cytokines IV. Cell. 2008;132:e1–2. doi: 10.1016/j.cell.2008.02.024. [DOI] [PubMed] [Google Scholar]
  • 5.Borish LC, Steinke JW. 2. Cytokines and chemokines. J Allergy Clin Immunol. 2003;111(Suppl):S460–75. doi: 10.1067/mai.2003.108. [DOI] [PubMed] [Google Scholar]
  • 6.Steinke JW, Borish L. 3. Cytokines and chemokines. J Allergy Clin Immunol. 2006;117(Suppl Mini-Primer):S441–5. doi: 10.1016/j.jaci.2005.07.001. [DOI] [PubMed] [Google Scholar]
  • 7.Vacchelli E, Galluzzi L, Eggermont A, Galon J, Tartour E, Zitvogel L, et al. Trial Watch: Immunostimulatory cytokines. Oncoimmunology. 2012;1:493–506. doi: 10.4161/onci.20459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Metcalf D. Hematopoietic cytokines. Blood. 2008;111:485–91. doi: 10.1182/blood-2007-03-079681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol. 2005;6:1191–7. doi: 10.1038/ni1276. [DOI] [PubMed] [Google Scholar]
  • 10.Dranoff G. Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer. 2004;4:11–22. doi: 10.1038/nrc1252. [DOI] [PubMed] [Google Scholar]
  • 11.Coussens LM, Zitvogel L, Palucka AK. Neutralizing tumor-promoting chronic inflammation: a magic bullet? Science. 2013;339:286–91. doi: 10.1126/science.1232227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–7. doi: 10.1038/nature01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov. 2012;11:215–33. doi: 10.1038/nrd3626. [DOI] [PubMed] [Google Scholar]
  • 14.Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2013;31:51–72. doi: 10.1146/annurev-immunol-032712-100008. [DOI] [PubMed] [Google Scholar]
  • 15.Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331:1565–70. doi: 10.1126/science.1203486. [DOI] [PubMed] [Google Scholar]
  • 16.Arellano M, Lonial S. Clinical uses of GM-CSF, a critical appraisal and update. Biologics. 2008;2:13–27. doi: 10.2147/BTT.S1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Khoury HJ, Loberiza FR, Jr., Ringdén O, Barrett AJ, Bolwell BJ, Cahn JY, et al. Impact of posttransplantation G-CSF on outcomes of allogeneic hematopoietic stem cell transplantation. Blood. 2006;107:1712–6. doi: 10.1182/blood-2005-07-2661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sebban C, Lefranc A, Perrier L, Moreau P, Espinouse D, Schmidt A, 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. doi: 10.1016/j.ejca.2011.12.016. [DOI] [PubMed] [Google Scholar]
  • 19.Naeim A, Henk HJ, Becker L, Chia V, Badre S, Li X, et al. 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. doi: 10.1186/1471-2407-13-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.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. doi: 10.1200/JCO.2011.36.8647. [DOI] [PubMed] [Google Scholar]
  • 21.Pabst T, Vellenga E, van Putten W, Schouten HC, Graux C, Vekemans MC, 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. doi: 10.1182/blood-2011-11-389841. [DOI] [PubMed] [Google Scholar]
  • 22.Hosing C. Hematopoietic stem cell mobilization with G-CSF. Methods Mol Biol. 2012;904:37–47. doi: 10.1007/978-1-61779-943-3_3. [DOI] [PubMed] [Google Scholar]
  • 23.Demirer T, Ayli M, Ozcan M, Gunel N, Haznedar R, Dagli M, 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. doi: 10.1046/j.1365-2141.2002.03264.x. [DOI] [PubMed] [Google Scholar]
  • 24.Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, et al. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology. 2012;1:1323–43. doi: 10.4161/onci.22009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial watch: Peptide vaccines in cancer therapy. Oncoimmunology. 2012;1:1557–76. doi: 10.4161/onci.22428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, et al. Trial watch: Monoclonal antibodies in cancer therapy. Oncoimmunology. 2013;2:e22789. doi: 10.4161/onci.22789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, et al. Trial watch: Monoclonal antibodies in cancer therapy. Oncoimmunology. 2013;2:e22789. doi: 10.4161/onci.22789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology. 2012;1:28–37. doi: 10.4161/onci.1.1.17938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012;1:894–907. doi: 10.4161/onci.20931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012;1:699–716. doi: 10.4161/onci.20696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Senovilla L, Vacchelli E, Garcia P, Eggermont A, Fridman WH, Galon J, et al. Trial Watch: DNA vaccines for cancer therapy. OncoImmunology. 2013;2:e23803. doi: 10.4161/onci.23803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, et al. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology. 2012;1:1111–34. doi: 10.4161/onci.21494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Vacchelli E, Senovilla L, Eggermont A, Fridman WH, Galon J, Zitvogel L, et al. Trial Watch: Chemotherapy with immunogenic cell death inducers. OncoImmunology. 2012;2:e23510. doi: 10.4161/onci.23510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology. 2012;1:179–88. doi: 10.4161/onci.1.2.19026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Menger L, Vacchelli E, Kepp O, Eggermont A, Tartour E, Zitvogel L, et al. Trial watch: Cardiac glycosides and cancer therapy. Oncoimmunology. 2013;2:e23082. doi: 10.4161/onci.23082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology. 2012;1:306–15. doi: 10.4161/onci.19549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Vacchelli E, Eggermont A, Fridman WH, Galon J, Tartour E, Zitvogel L, et al. Trial Watch: Adoptive cell transfer for anticancer immunotherapy. OncoImmunology. 2013;2 doi: 10.4161/onci.24238. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.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. doi: 10.1038/ncponc0426. [DOI] [PubMed] [Google Scholar]
  • 39.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. doi: 10.1245/s10434-011-2030-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.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. doi: 10.1002/bjs.7621. [DOI] [PubMed] [Google Scholar]
  • 41.Bonvalot S, de Baere T, Mendiboure J, Paci A, Farace F, Drouard-Troalen L, et al. Hyperthermic pelvic perfusion with tumor necrosis factor-α for locally advanced cancers: encouraging results of a phase II study. Ann Surg. 2012;255:281–6. doi: 10.1097/SLA.0b013e318242ebe7. [DOI] [PubMed] [Google Scholar]
  • 42.Lorusso D, Scambia G, Amadio G, di Legge A, Pietragalla A, De Vincenzo R, et al. Phase II study of NGR-hTNF in combination with doxorubicin in relapsed ovarian cancer patients. Br J Cancer. 2012;107:37–42. doi: 10.1038/bjc.2012.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Zucali PA, Simonelli M, De Vincenzo F, Lorenzi E, Perrino M, Bertossi M, et al. Phase I and pharmacodynamic study of high-dose NGR-hTNF in patients with refractory solid tumours. Br J Cancer. 2013;108:58–63. doi: 10.1038/bjc.2012.506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Peterson JE, Zurakowski D, Italiano JE, Jr., Michel LV, Connors S, Oenick M, et al. VEGF, PF4 and PDGF are elevated in platelets of colorectal cancer patients. Angiogenesis. 2012;15:265–73. doi: 10.1007/s10456-012-9259-z. [DOI] [PubMed] [Google Scholar]
  • 45.Scaruffi P, Morandi F, Gallo F, Stigliani S, Parodi S, Moretti S, et al. Bone marrow of neuroblastoma patients shows downregulation of CXCL12 expression and presence of IFN signature. Pediatr Blood Cancer. 2012;59:44–51. doi: 10.1002/pbc.23339. [DOI] [PubMed] [Google Scholar]
  • 46.Ansell SM, Maurer MJ, Ziesmer SC, Slager SL, Habermann TM, Link BK, et al. Elevated pretreatment serum levels of interferon-inducible protein-10 (CXCL10) predict disease relapse and prognosis in diffuse large B-cell lymphoma patients. Am J Hematol. 2012;87:865–9. doi: 10.1002/ajh.23259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sauer M, Plütschow A, Jachimowicz RD, Kleefisch D, Reiners KS, Ponader S, et al. Baseline serum TARC levels predict therapy outcome in patients with Hodgkin lymphoma. Am J Hematol. 2013;88:113–5. doi: 10.1002/ajh.23361. [DOI] [PubMed] [Google Scholar]
  • 48.Sleijfer S, Gorlia T, Lamers C, Burger H, Blay JY, Le Cesne A, et al. Cytokine and angiogenic factors associated with efficacy and toxicity of pazopanib in advanced soft-tissue sarcoma: an EORTC-STBSG study. Br J Cancer. 2012;107:639–45. doi: 10.1038/bjc.2012.328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Mellgren K, Hedegaard CJ, Schmiegelow K, Müller K. Plasma cytokine profiles at diagnosis in pediatric patients with non-hodgkin lymphoma. J Pediatr Hematol Oncol. 2012;34:271–5. doi: 10.1097/MPH.0b013e3182431e02. [DOI] [PubMed] [Google Scholar]
  • 50.Zurita AJ, Jonasch E, Wang X, Khajavi M, Yan S, Du DZ, et al. A cytokine and angiogenic factor (CAF) analysis in plasma for selection of sorafenib therapy in patients with metastatic renal cell carcinoma. Ann Oncol. 2012;23:46–52. doi: 10.1093/annonc/mdr047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tran HT, Liu Y, Zurita AJ, Lin Y, Baker-Neblett KL, Martin AM, et al. Prognostic or predictive plasma cytokines and angiogenic factors for patients treated with pazopanib for metastatic renal-cell cancer: a retrospective analysis of phase 2 and phase 3 trials. Lancet Oncol. 2012;13:827–37. doi: 10.1016/S1470-2045(12)70241-3. [DOI] [PubMed] [Google Scholar]
  • 52.Lainey E, Wolfromm A, Marie N, Enot D, Scoazec M, Bouteloup C, et al. Azacytidine and erlotinib exert synergistic effects against acute myeloid leukemia. Oncogene. 2012 doi: 10.1038/onc.2012.469. In press. [DOI] [PubMed] [Google Scholar]
  • 53.Thépot S, Lainey E, Cluzeau T, Sébert M, Leroy C, Adès L, et al. Hypomethylating agents reactivate FOXO3A in acute myeloid leukemia. Cell Cycle. 2011;10:2323–30. doi: 10.4161/cc.10.14.16399. [DOI] [PubMed] [Google Scholar]
  • 54.Pollyea DA, Kohrt HE, Gallegos L, Figueroa ME, Abdel-Wahab O, Zhang B, et al. Safety, efficacy and biological predictors of response to sequential azacitidine and lenalidomide for elderly patients with acute myeloid leukemia. Leukemia. 2012;26:893–901. doi: 10.1038/leu.2011.294. [DOI] [PubMed] [Google Scholar]
  • 55.Le QT, Fisher R, Oliner KS, Young RJ, Cao H, Kong C, et al. Prognostic and predictive significance of plasma HGF and IL-8 in a phase III trial of chemoradiation with or without tirapazamine in locoregionally advanced head and neck cancer. Clin Cancer Res. 2012;18:1798–807. doi: 10.1158/1078-0432.CCR-11-2094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lim AM, Rischin D, Fisher R, Cao H, Kwok K, Truong D, et al. Prognostic significance of plasma osteopontin in patients with locoregionally advanced head and neck squamous cell carcinoma treated on TROG 02.02 phase III trial. Clin Cancer Res. 2012;18:301–7. doi: 10.1158/1078-0432.CCR-11-2295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lu K, Feng X, Deng Q, Sheng L, Liu P, Xu S, et al. Prognostic role of serum cytokines in patients with nasopharyngeal carcinoma. Onkologie. 2012;35:494–8. doi: 10.1159/000341827. [DOI] [PubMed] [Google Scholar]
  • 58.Llovet JM, Peña CE, Lathia CD, Shan M, Meinhardt G, Bruix J, SHARP Investigators Study Group Plasma biomarkers as predictors of outcome in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 2012;18:2290–300. doi: 10.1158/1078-0432.CCR-11-2175. [DOI] [PubMed] [Google Scholar]
  • 59.Suzuki K, Kadota K, Sima CS, Nitadori J, Rusch VW, Travis WD, et al. Clinical impact of immune microenvironment in stage I lung adenocarcinoma: tumor interleukin-12 receptor β2 (IL-12Rβ2), IL-7R, and stromal FoxP3/CD3 ratio are independent predictors of recurrence. J Clin Oncol. 2013;31:490–8. doi: 10.1200/JCO.2012.45.2052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wang YC, Sung WW, Wu TC, Wang L, Chien WP, Cheng YW, et al. Interleukin-10 haplotype may predict survival and relapse in resected non-small cell lung cancer. PLoS One. 2012;7:e39525. doi: 10.1371/journal.pone.0039525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Garcia JA, Elson P, Tyler A, Triozzi P, Dreicer R. Sargramostim (GM-CSF) and lenalidomide in castration-resistant prostate cancer (CRPC): Results from a phase I-II clinical trial(☆☆☆) Urol Oncol. 2013 doi: 10.1016/j.urolonc.2012.12.004. In press. [DOI] [PubMed] [Google Scholar]
  • 62.Garcia JA, Mekhail T, Elson P, Wood L, Bukowski RM, Dreicer R, et al. Phase I/II trial of subcutaneous interleukin-2, granulocyte-macrophage colony-stimulating factor and interferon-α in patients with metastatic renal cell carcinoma. BJU Int. 2012;109:63–9. doi: 10.1111/j.1464-410X.2010.10011.x. [DOI] [PubMed] [Google Scholar]
  • 63.Tarhini AA, Butterfield LH, Shuai Y, Gooding WE, Kalinski P, Kirkwood JM. Differing patterns of circulating regulatory T cells and myeloid-derived suppressor cells in metastatic melanoma patients receiving anti-CTLA4 antibody and interferon-α or TLR-9 agonist and GM-CSF with peptide vaccination. J Immunother. 2012;35:702–10. doi: 10.1097/CJI.0b013e318272569b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Tarhini AA, Cherian J, Moschos SJ, Tawbi HA, Shuai Y, Gooding WE, et al. Safety and efficacy of combination immunotherapy with interferon alfa-2b and tremelimumab in patients with stage IV melanoma. J Clin Oncol. 2012;30:322–8. doi: 10.1200/JCO.2011.37.5394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Groenewegen G, Walraven M, Vermaat J, de Gast B, Witteveen E, Giles R, et al. Targeting the endothelin axis with atrasentan, in combination with IFN-alpha, in metastatic renal cell carcinoma. Br J Cancer. 2012;106:284–9. doi: 10.1038/bjc.2011.515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Walter B, Schrettenbrunner I, Vogelhuber M, Grassinger J, Bross K, Wilke J, et al. Pioglitazone, etoricoxib, interferon-α, and metronomic capecitabine for metastatic renal cell carcinoma: final results of a prospective phase II trial. Med Oncol. 2012;29:799–805. doi: 10.1007/s12032-011-9982-0. [DOI] [PubMed] [Google Scholar]
  • 67.Hardan I, Stanevsky A, Volchek Y, Tohami T, Amariglio N, Trakhtenbrot L, et al. Treatment with interferon alpha prior to discontinuation of imatinib in patients with chronic myeloid leukemia. Cytokine. 2012;57:290–3. doi: 10.1016/j.cyto.2011.11.018. [DOI] [PubMed] [Google Scholar]
  • 68.Seung SK, Curti BD, Crittenden M, Walker E, Coffey T, Siebert JC, et al. Phase 1 study of stereotactic body radiotherapy and interleukin-2--tumor and immunological responses. Sci Transl Med. 2012;4:37ra74. doi: 10.1126/scitranslmed.3003649. [DOI] [PubMed] [Google Scholar]
  • 69.Eggermont AM, Suciu S, Santinami M, Testori A, Kruit WH, Marsden J, et al. EORTC Melanoma Group Adjuvant therapy with pegylated interferon alfa-2b versus observation alone in resected stage III melanoma: final results of EORTC 18991, a randomised phase III trial. Lancet. 2008;372:117–26. doi: 10.1016/S0140-6736(08)61033-8. [DOI] [PubMed] [Google Scholar]
  • 70.Eggermont AM, Suciu S, Testori A, Kruit WH, Marsden J, Punt CJ, et al. Ulceration and stage are predictive of interferon efficacy in melanoma: results of the phase III adjuvant trials EORTC 18952 and EORTC 18991. Eur J Cancer. 2012;48:218–25. doi: 10.1016/j.ejca.2011.09.028. [DOI] [PubMed] [Google Scholar]
  • 71.Eggermont AM, Suciu S, Testori A, Santinami M, Kruit WH, Marsden J, 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. doi: 10.1200/JCO.2011.41.3799. [DOI] [PubMed] [Google Scholar]
  • 72.Kasai K, Ushio A, Kasai Y, Sawara K, Miyamoto Y, Oikawa K, et al. Therapeutic efficacy of combination therapy with intra-arterial 5-fluorouracil and systemic pegylated interferon α-2b for advanced hepatocellular carcinoma with portal venous invasion. Cancer. 2012;118:3302–10. doi: 10.1002/cncr.26648. [DOI] [PubMed] [Google Scholar]
  • 73.Schmidt J, Abel U, Debus J, Harig S, Hoffmann K, Herrmann T, et al. Open-label, multicenter, randomized phase III trial of adjuvant chemoradiation plus interferon Alfa-2b versus fluorouracil and folinic acid for patients with resected pancreatic adenocarcinoma. J Clin Oncol. 2012;30:4077–83. doi: 10.1200/JCO.2011.38.2960. [DOI] [PubMed] [Google Scholar]
  • 74.Warren K, Bent R, Wolters PL, Prager A, Hanson R, Packer R, et al. A phase 2 study of pegylated interferon α-2b (PEG-Intron(®)) in children with diffuse intrinsic pontine glioma. Cancer. 2012;118:3607–13. doi: 10.1002/cncr.26659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chen LL, Chen X, Choi H, Sang H, Chen LC, Zhang H, et al. Exploiting antitumor immunity to overcome relapse and improve remission duration. Cancer Immunol Immunother. 2012;61:1113–24. doi: 10.1007/s00262-011-1185-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Aoyagi S, Hata H, Homma E, Shimizu H. Sequential local injection of low-dose interferon-beta for maintenance therapy in stage II and III melanoma: a single-institution matched case-control study. Oncology. 2012;82:139–46. doi: 10.1159/000336490. [DOI] [PubMed] [Google Scholar]
  • 77.Buehrlen M, Zwaan CM, Granzen B, Lassay L, Deutz P, Vorwerk P, et al. Multimodal treatment, including interferon beta, of nasopharyngeal carcinoma in children and young adults: preliminary results from the prospective, multicenter study NPC-2003-GPOH/DCOG. Cancer. 2012;118:4892–900. doi: 10.1002/cncr.27395. [DOI] [PubMed] [Google Scholar]
  • 78.Petrella TM, Tozer R, Belanger K, Savage KJ, Wong R, Smylie M, et al. Interleukin-21 has activity in patients with metastatic melanoma: a phase II study. J Clin Oncol. 2012;30:3396–401. doi: 10.1200/JCO.2011.40.0655. [DOI] [PubMed] [Google Scholar]
  • 79.Steele N, Anthony A, Saunders M, Esmarck B, Ehrnrooth E, Kristjansen PE, et al. A phase 1 trial of recombinant human IL-21 in combination with cetuximab in patients with metastatic colorectal cancer. Br J Cancer. 2012;106:793–8. doi: 10.1038/bjc.2011.599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Gillessen S, Gnad-Vogt US, Gallerani E, Beck J, Sessa C, Omlin A, et al. A phase I dose-escalation study of the immunocytokine EMD 521873 (Selectikine) in patients with advanced solid tumours. Eur J Cancer. 2013;49:35–44. doi: 10.1016/j.ejca.2012.07.015. [DOI] [PubMed] [Google Scholar]
  • 81.Albertini MR, Hank JA, Gadbaw B, Kostlevy J, Haldeman J, Schalch H, et al. Phase II trial of hu14.18-IL2 for patients with metastatic melanoma. Cancer Immunol Immunother. 2012;61:2261–71. doi: 10.1007/s00262-012-1286-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Pautier P, Locher C, Robert C, Deroussent A, Flament C, Le Cesne A, et al. Phase I clinical trial combining imatinib mesylate and IL-2 in refractory cancer patients: IL-2 interferes with the pharmacokinetics of imatinib mesylate. Oncoimmunology. 2013;2:e23079. doi: 10.4161/onci.23079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Delahaye NF, Rusakiewicz S, Martins I, Ménard C, Roux S, Lyonnet L, et al. Alternatively spliced NKp30 isoforms affect the prognosis of gastrointestinal stromal tumors. Nat Med. 2011;17:700–7. doi: 10.1038/nm.2366. [DOI] [PubMed] [Google Scholar]
  • 84.Ménard C, Blay JY, Borg C, Michiels S, Ghiringhelli F, Robert C, et al. Natural killer cell IFN-gamma levels predict long-term survival with imatinib mesylate therapy in gastrointestinal stromal tumor-bearing patients. Cancer Res. 2009;69:3563–9. doi: 10.1158/0008-5472.CAN-08-3807. [DOI] [PubMed] [Google Scholar]
  • 85.Balachandran VP, Cavnar MJ, Zeng S, Bamboat ZM, Ocuin LM, Obaid H, et al. Imatinib potentiates antitumor T cell responses in gastrointestinal stromal tumor through the inhibition of Ido. Nat Med. 2011;17:1094–100. doi: 10.1038/nm.2438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ohno S, Okuyama R, Aruga A, Sugiyama H, Yamamoto M. Phase I trial of Wilms’ Tumor 1 (WT1) peptide vaccine with GM-CSF or CpG in patients with solid malignancy. Anticancer Res. 2012;32:2263–9. [PubMed] [Google Scholar]
  • 87.Rahma OE, Ashtar E, Czystowska M, Szajnik ME, Wieckowski E, Bernstein S, et al. A gynecologic oncology group phase II trial of two p53 peptide vaccine approaches: subcutaneous injection and intravenous pulsed dendritic cells in high recurrence risk ovarian cancer patients. Cancer Immunol Immunother. 2012;61:373–84. doi: 10.1007/s00262-011-1100-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kameshima H, Tsuruma T, Kutomi G, Shima H, Iwayama Y, Kimura Y, et al. Immunotherapeutic benefit of α-interferon (IFNα) in survivin2B-derived peptide vaccination for advanced pancreatic cancer patients. Cancer Sci. 2013;104:124–9. doi: 10.1111/cas.12046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Zeestraten EC, Speetjens FM, Welters MJ, Saadatmand S, Stynenbosch LF, Jongen R, et al. Addition of interferon-α to the p53-SLP® vaccine results in increased production of interferon-γ in vaccinated colorectal cancer patients: a phase I/II clinical trial. Int J Cancer. 2013;132:1581–91. doi: 10.1002/ijc.27819. [DOI] [PubMed] [Google Scholar]
  • 90.van den Eertwegh AJ, Versluis J, van den Berg HP, Santegoets SJ, van Moorselaar RJ, van der Sluis TM, et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol. 2012;13:509–17. doi: 10.1016/S1470-2045(12)70007-4. [DOI] [PubMed] [Google Scholar]
  • 91.Dobrzanski MJ, Rewers-Felkins KA, Samad KA, Quinlin IS, Phillips CA, Robinson W, et al. Immunotherapy with IL-10- and IFN-γ-producing CD4 effector cells modulate “Natural” and “Inducible” CD4 TReg cell subpopulation levels: observations in four cases of patients with ovarian cancer. Cancer Immunol Immunother. 2012;61:839–54. doi: 10.1007/s00262-011-1128-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Braumüller H, Wieder T, Brenner E, Aßmann S, Hahn M, Alkhaled M, et al. T-helper-1-cell cytokines drive cancer into senescence. Nature. 2013;494:361–5. doi: 10.1038/nature11824. [DOI] [PubMed] [Google Scholar]
  • 93.Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR, Du J, et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature. 2012;487:500–4. doi: 10.1038/nature11183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature. 2012;487:505–9. doi: 10.1038/nature11249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kentsis A, Reed C, Rice KL, Sanda T, Rodig SJ, Tholouli E, et al. Autocrine activation of the MET receptor tyrosine kinase in acute myeloid leukemia. Nat Med. 2012;18:1118–22. doi: 10.1038/nm.2819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Eichhorn PJ, Rodón L, Gonzàlez-Juncà A, Dirac A, Gili M, Martínez-Sáez E, et al. USP15 stabilizes TGF-β receptor I and promotes oncogenesis through the activation of TGF-β signaling in glioblastoma. Nat Med. 2012;18:429–35. doi: 10.1038/nm.2619. [DOI] [PubMed] [Google Scholar]
  • 97.Grivennikov SI, Wang K, Mucida D, Stewart CA, Schnabl B, Jauch D, et al. Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature. 2012;491:254–8. doi: 10.1038/nature11465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Zitvogel L, Kepp O, Galluzzi L, Kroemer G. Inflammasomes in carcinogenesis and anticancer immune responses. Nat Immunol. 2012;13:343–51. doi: 10.1038/ni.2224. [DOI] [PubMed] [Google Scholar]
  • 99.Bruchard M, Mignot G, Derangère V, Chalmin F, Chevriaux A, Végran F, et al. Chemotherapy-triggered cathepsin B release in myeloid-derived suppressor cells activates the Nlrp3 inflammasome and promotes tumor growth. Nat Med. 2013;19:57–64. doi: 10.1038/nm.2999. [DOI] [PubMed] [Google Scholar]
  • 100.Huber S, Gagliani N, Zenewicz LA, Huber FJ, Bosurgi L, Hu B, et al. IL-22BP is regulated by the inflammasome and modulates tumorigenesis in the intestine. Nature. 2012;491:259–63. doi: 10.1038/nature11535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Olson BM, Jankowska-Gan E, Becker JT, Vignali DA, Burlingham WJ, McNeel DG. Human prostate tumor antigen-specific CD8+ regulatory T cells are inhibited by CTLA-4 or IL-35 blockade. J Immunol. 2012;189:5590–601. doi: 10.4049/jimmunol.1201744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Beyer M, Schumak B, Weihrauch MR, Andres B, Giese T, Endl E, et al. In vivo expansion of naïve CD4+ CD25(high) FOXP3+ regulatory T cells in patients with colorectal carcinoma after IL-2 administration. PLoS One. 2012;7:e30422. doi: 10.1371/journal.pone.0030422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Triozzi PL, Achberger S, Aldrich W, Elson P, Garcia J, Dreicer R. Differential immunologic and microRNA effects of 2 dosing regimens of recombinant human granulocyte/macrophage colony stimulating factor. J Immunother. 2012;35:587–94. doi: 10.1097/CJI.0b013e31826b20b6. [DOI] [PubMed] [Google Scholar]
  • 104.Levin AM, Bates DL, Ring AM, Krieg C, Lin JT, Su L, et al. Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine’. Nature. 2012;484:529–33. doi: 10.1038/nature10975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Weber M, Hauschild R, Schwarz J, Moussion C, de Vries I, Legler DF, et al. Interstitial dendritic cell guidance by haptotactic chemokine gradients. Science. 2013;339:328–32. doi: 10.1126/science.1228456. [DOI] [PubMed] [Google Scholar]
  • 106.Park SH, Das BB, Casagrande F, Tian Y, Nothnagel HJ, Chu M, et al. Structure of the chemokine receptor CXCR1 in phospholipid bilayers. Nature. 2012;491:779–83. doi: 10.1038/nature11580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Sahin U, Koslowski M, Dhaene K, Usener D, Brandenburg G, Seitz G, et al. Claudin-18 splice variant 2 is a pan-cancer target suitable for therapeutic antibody development. Clin Cancer Res. 2008;14:7624–34. doi: 10.1158/1078-0432.CCR-08-1547. [DOI] [PubMed] [Google Scholar]
  • 108.Keating MJ, Dritselis A, Yasothan U, Kirkpatrick P. Ofatumumab. Nat Rev Drug Discov. 2010;9:101–2. doi: 10.1038/nrd3100. [DOI] [PubMed] [Google Scholar]
  • 109.Brahmer JR. Harnessing the immune system for the treatment of non-small-cell lung cancer. J Clin Oncol. 2013;31:1021–8. doi: 10.1200/JCO.2012.45.8703. [DOI] [PubMed] [Google Scholar]
  • 110.Kesselring R, Jauch D, Fichtner-Feigl S. Interleukin 21 impairs tumor immunosurveillance of colitis-associated colorectal cancer. Oncoimmunology. 2012;1:537–8. doi: 10.4161/onci.19407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Stolfi C, Pallone F, Macdonald TT, Monteleone G. Interleukin-21 in cancer immunotherapy: Friend or foe? Oncoimmunology. 2012;1:351–4. doi: 10.4161/onci.19122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Jinesh G G, Kamat AM. Redirecting neutrophils against bladder cancer cells by BCG and Smac mimetic combination. Oncoimmunology. 2012;1:1161–2. doi: 10.4161/onci.20928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Galluzzi L, Kepp O, Kroemer G. TP53 and MTOR crosstalk to regulate cellular senescence. Aging (Albany NY) 2010;2:535–7. doi: 10.18632/aging.100202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Cheson BD. Hematology: Rituximab for follicular lymphoma: maintaining an open mind. Nat Rev Clin Oncol. 2011;8:257–8. doi: 10.1038/nrclinonc.2011.35. [DOI] [PubMed] [Google Scholar]
  • 115.Scheinecker C, Smolen J, Yasothan U, Stoll J, Kirkpatrick P. Tocilizumab. Nat Rev Drug Discov. 2009;8:273–4. doi: 10.1038/nrd2863. [DOI] [PubMed] [Google Scholar]
  • 116.Sihto H, Joensuu H. Tumor-infiltrating lymphocytes and outcome in Merkel cell carcinoma, a virus-associated cancer. Oncoimmunology. 2012;1:1420–1. doi: 10.4161/onci.21120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Seruga B, Zhang H, Bernstein LJ, Tannock IF. Cytokines and their relationship to the symptoms and outcome of cancer. Nat Rev Cancer. 2008;8:887–99. doi: 10.1038/nrc2507. [DOI] [PubMed] [Google Scholar]
  • 118.Mughal TI. Current and future use of hematopoietic growth factors in cancer medicine. Hematol Oncol. 2004;22:121–34. doi: 10.1002/hon.736. [DOI] [PubMed] [Google Scholar]
  • 119.Clement JM, McDermott DF. The high-dose aldesleukin (IL-2) “select” trial: a trial designed to prospectively validate predictive models of response to high-dose IL-2 treatment in patients with metastatic renal cell carcinoma. Clin Genitourin Cancer. 2009;7:E7–9. doi: 10.3816/CGC.2009.n.014. [DOI] [PubMed] [Google Scholar]
  • 120.Halama N, Zoernig I, Jaeger D. Advanced malignant melanoma: immunologic and multimodal therapeutic strategies. J Oncol. 2010;2010:689893. doi: 10.1155/2010/689893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Keeping IL-2 toxicity at bay. Nat Biotechnol. 2000;18:1131. doi: 10.1038/81087. [DOI] [PubMed] [Google Scholar]
  • 122.Rossi EA, Chang CH, Cardillo TM, Goldenberg DM. Optimization of multivalent bispecific antibodies and immunocytokines with improved in vivo properties. Bioconjug Chem. 2013;24:63–71. doi: 10.1021/bc300488f. [DOI] [PubMed] [Google Scholar]
  • 123.Pasche N, Neri D. Immunocytokines: a novel class of potent armed antibodies. Drug Discov Today. 2012;17:583–90. doi: 10.1016/j.drudis.2012.01.007. [DOI] [PubMed] [Google Scholar]
  • 124.Zitvogel L, Kroemer G. Targeting PD-1/PD-L1 interactions for cancer immunotherapy. Oncoimmunology. 2012;1:1223–5. doi: 10.4161/onci.21335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Peng W, Lizée G, Hwu P. Blockade of the PD-1 pathway enhances the efficacy of adoptive cell therapy against cancer. Oncoimmunology. 2013;2:e22691. doi: 10.4161/onci.22691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Egilmez NK, Harden JL, Rowswell-Turner RB. Chemoimmunotherapy as long-term maintenance therapy for cancer. Oncoimmunology. 2012;1:563–5. doi: 10.4161/onci.19369. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Oncoimmunology are provided here courtesy of Taylor & Francis

RESOURCES