Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jan 14.
Published in final edited form as: Cancer J. 2010 Jul-Aug;16(4):392–398. doi: 10.1097/PPO.0b013e3181eacbc4

Novel Gamma-Chain Cytokines as Candidate Immune Modulators in Immune Therapies for Cancer

Natasha M Fewkes *, Crystal L Mackall
PMCID: PMC6959548  NIHMSID: NIHMS1066325  PMID: 20693852

Abstract

Cytokines that signal through the common-gamma chain are potent growth factors for T cells and natural killer cells. Interleukin (IL)-2, the γc prototype, can mediate antitumor effects as a single agent or in the context of multimodality regimens but is limited by side effects and a propensity for expansion of regulatory T cells. IL-7, IL-15, and IL-21 each possess properties that can be exploited in the context of immunotherapy for cancer. Each has been demonstrated to mediate potent vaccine adjuvant effects in tumor models, and each can enhance the effectiveness of adoptive immunotherapies. Although the overlap among the agents is significant, IL-7 is uniquely immunorestorative and preferentially augments reactivity of naive populations, IL-15 potently augments reactivity of CD8+ memory cells and natural killer cells, and IL-21 preferentially expands the inflammatory Th17 subset and may limit terminal differentiation of effector CD8+ cells. Clinical trials of IL-7 and IL-21 have already been completed and, so far, demonstrate safety and biologic activity of these agents. Clinical trials of IL-15 are expected soon. Ultimately, these agents are expected to be most effective in the context of multimodal immunotherapy regimens, and careful clinical trial design will be needed to efficiently identify the proper doses, regimens, and settings in which to exploit their biologic properties for therapeutic gain.

Keywords: gamma c cytokines, IL-7, IL-15, IL-21, cancer immunotherapy


Immune responses to tumor antigens can be induced in patients with cancer using a variety of vaccine strategies, including genetic vectors, peptides, and dendritic cells. Despite this, tumor vaccines alone are generally not potent enough to shrink established tumors, and although some trials of tumor vaccines have demonstrated clinical benefit adjuvantly, this is not a universal finding.1 New approaches have become available to modulate endogenous immune responses using genetically modified T cells to target a wide array of tumors, but the effectiveness of these therapies remains limited, in part, by poor persistence of the transferred cells in vivo. Immunomodulatory agents have also shown promise against some established tumors, but response rates of single agents remain low.2 Together, the clinical data gleaned from a variety of models suggest that future progress in tumor immunotherapy will require multimodal regimens that incorporate agents to augment the strength and duration of immune responses. This can be accomplished by enhancing the number, quality, diversity, or longevity of tumor immune effectors and/or by diminishing suppressive factors that mediate tumor immune escape.

Cytokines are among the most promising agents available for potentiating the weak immune responses induced by current tumor vaccines and immunomodulators, and for augmenting persistence and functionality of adoptively transferred cells. Cytokines that signal through the common cytokine gamma chain (γc) can enhance the number, quality, diversity, and longevity of T cells and natural killer (NK) cells. Interleukin (IL)-2, the γc prototype, mediates antitumor effects in some settings but has significant limitations, including augmentation of regulatory T cell number and function.3 Recent studies demonstrate that other γc cytokines, namely IL-7, IL-15, and IL-21, have properties that could be exploited to amplify antitumor T cell and NK immunity. In this review, we will briefly discuss the basic biology of IL-7, IL-15, and IL-21 and then review translational and early clinical studies illustrating their potential to amplify cell-based immunity toward cancer.

INTERLEUKIN-7

IL-7 Biology and Preclinical Studies

IL-7 is required for development and survival of T cells, is important but not required for human B-cell development, and plays no significant role in NK-cell development and survival. IL-7 is produced primarily by stromal and parenchymal cells4 with lesser amounts produced by dendritic cells; IL-7 is not produced by B cells or T cells. The IL-7 receptor is a heterodimer comprising the common γc and IL-7Rα. IL-7Rα also signals thymic stromal lym-phopoietin (TSLP). Unlike receptors for most γc cytokines, (absent on resting cells but upregulated on activation), the IL-7R is expressed on most resting lymphocytes, downregulated after T-cell activation, then reexpressed on effector cells destined for the memory cell pool5 and on central memory T cells. Because IL-7 is produced by nonimmune cells in the absence of immune activation, is measurable in sera from healthy individuals, and the IL-7R heterodimer is present on most T cells, IL-7 can be conceptualized as a homeostatic cytokine whose signals are continuously available and play an important role in maintaining T-cell homeostasis.

The corollary to the central role of IL-7 in T-cell homeostasis is that the availability of IL-7 is increased during lymphopenia.68 Increased IL-7 availability plays a central role in driving immune reconstitution through homeostatic peripheral expansion, which involves exaggerated responses to cognate antigen,9 increased antigen-independent cycling of memory populations, and proliferative responses to low affinity antigens comprising both self-antigens and crossreactive environmental antigens.1012 IL-7 is notable for its immunorestorative properties, its ability to augment both CD4 and CD8 expansion, its tendency to induce cycling of naive T cells with broad repertoire diversity, and its ability to amplify low affinity, subdominant immune responses such as those required for antitumor immunity. In contrast to the immune activating effects described earlier, IL-7 signaling on dendritic cells has also been recently implicated in a regulatory axis that controls CD4 homeostatic expansion during lymphopenia.13 Cell-specific effects of IL-7 are summarized and compared with other γc cytokines in Table 1.

TABLE 1.

Cell-Specific Effects of γc Cytokines

B Cells CD4 T Cells CD8 T Cells NK Cells
IL-2 ↑ Especially Tregs ↑↑ ↑↑
IL-7 ↑ Especially non-Tregs ↑↑
IL-15 +/− ↑↑↑ ↑↑↑
IL-21 ↑ Th17 ↑ Tfh ↑ Cytotoxicity ↑ Cytotoxicity

Several studies have demonstrated the capacity of IL-7 to enhance antitumor effects of adoptively transferred cells when used to culture effectors ex vivo14,15 or when tumors were genetically engineered to express IL-7 in vivo.1618 In several studies, IL-7 outperformed IL-2 for ex vivo expansion of antigen-specific T cells, in terms of both cell yield and functionality after adoptive transfer.19,20 When administered systemically in animal models, IL-7 is also a potent vaccine adjuvant,21,22 which outperforms IL-2 when the 2 agents are compared directly. In the study by Melchionda et al, both IL-7 and IL-15 augmented immunodominant CD8 responses to a similar degree; however, IL-7 had more potent effects on CD4 effectors and more potently augmented subdominant antigens than IL-15.23 Similar to IL-15, IL-7 therapy provided as a short course during vaccination provided long-term benefits for memory cell survival, which persisted long after the cytokines were discontinued. A more recent study confirmed the capacity of IL-7 to enhance long-term memory cell responses, noting that this occurred after lentiviral-based tumor vaccination but not after peptide-based tumor vaccines,24 presumably because of upregulation of IL-7Rα on effector cells after lentiviral but not peptide-based vaccination. Nanjappa et al25 noted that IL-7 enhancement of memory cell responses is dependent on timing of administration, with IL-7 therapy administered during the contraction phase more effective than when the cytokine was administered during the initial expansion phase of lymphocyte choriomeningitis virus (LCMV) infection.

Work by Pellegrini et al dissected the effects of IL-7 in a murine islet cell tumor model that expresses LCMV glycoprotein. In this model, LCMV infection yields limited antitumor effects, but coadministration of IL-7 with LCMV infection prolonged survival, decreased tumor burden and increased tumor T-cell infiltration. As expected based on the capacity of IL-7 to expand T cells, this was associated with substantial increases in the number of antigen-specific T cells in vivo. In addition, however, these authors identified immunomodulatory effects of IL-7 that had not previously been appreciated. Specifically, increased numbers of tumor infiltrating Th17 cells and NK cells and increased serum levels of IL-6, IL-12, IL-17, and IL-1α were found when IL-7 was administered after LCMV infection compared with LCMV infection without cytokine. Interestingly, IL-7 did not induce inflammatory mediators in the absence of viral infection. T cells from IL-7-treated mice in this model were less susceptible to Treg and TGFβ-mediated inhibition and show diminished Cbl-b expression as a result of Nedd4 upregulation. Given that Cbl-b is a negative modulator of TCR signaling, IL-7 modulation of Cbl-b may be the mechanism by which IL-7 enhances T-cell responses to weak antigens. In summary, in addition to confirming the known effects of IL-7 on T-cell expansion and persistence, this work identified several novel mechanisms through which IL-7 augmented antitumor effects. It also clearly illustrated that potent IL-7-mediated antitumor responses are exquisitely dependent on concomitant administration of an effective vaccine because IL-7 had no significant antitumor effect in the absence of LCMV infection in these studies.

IL-7 Clinical Trials

Several clinical studies of rhIL-7 have been completed and are currently underway, and therefore, emerging data are available regarding the clinical potential of this agent. Thus far, rhIL-7 has been well tolerated, with no evidence for capillary leak syndrome. Toxicities have been predominantly low-grade fever, myalgias, transient flu-like symptoms, and self-limited transaminitis.26 Biologic effects have been observed in doses ranging from 3 to 60μg/kg, the highest dose studied thus far. Essentially, all studies have demonstrated dose-dependent increases in T-cell number following rhIL-7, with substantial increases in both CD4 and CD8 cells.2730 Early B-cell progenitors are also transiently increased, usually within the bone marrow but occasionally seen in the peripheral blood,26 and lesser increases in NK-cell numbers have been observed. In addition to changes in peripheral blood lymphocytes, rhIL-7 increases the size of the secondary lymphoid organs, including spleen and lymph nodes, which can be visualized by computed tomography scanning or positron emission tomography imaging.27 Increases in peripheral T-cell numbers seems to result primarily from peripheral effects including enhanced cycling and diminished apoptosis, rather than increased thymopoiesis. Notable is preferential expansion of rhIL-7 of naive T cells, which can enhance the repertoire diversity independent of age and independent of thymopoiesis.27 In addition, rhIL-7 has minimal effects on expansion of Tregs because of low IL-7Ra on this subset,31,32 which results in relative decreases in the frequency of Tregs in rhIL-7-treated subjects.28 This is in contrast to rhIL-2, which preferentially expands Tregs.3,33 Thus far, mature clinical data are not yet available to evaluate its effectiveness as a vaccine adjuvant or as a supportive cytokine for adoptive immunotherapy, but these studies are anxiously awaited. Clinical trials with IL-7 are summarized and compared with other γc cytokines in Table 2.

TABLE 2.

Clinical Trials With γc Cytokines

Dose Toxicity Biological Activity Antitumor Activity
IL-2 Subcutaneous: 700,000 IU–5,000,000 IU/m2 (43 μg–306 μg/m2) ≈ (1–7.7μg/kg) Capillary leak, hepatic dysfunction, flu-like malaise, low blood pressure, fever, nausea, vomiting, diarrhea, chills, edema, weight gain, confusion, skin rashes, and changes in blood chemistry. Early lymphopenia due to trafficking followed by expansion of peripheral lymphoid cells with continued administration. Renal cell carcinoma, melanoma, and possible enhanced activity of adoptively transferred cells.
Treg proliferation.
Intravenous: 360,000–720,000 IU/kg (22–44 μg/kg) Increased serum levels of gamma interferon.
IL-7 3–60 μg/kg Fever, myalgias, ↑ liver function tests, injection site erythema. Lymphocytosis None
Increased T-cell cycling and broadening of circulating TCR repertoire diversity.
Increased size of spleen and lymph nodes.
Bcl-2 upregulation.
IL-15 Recently approved for phase I trials, data not yet available
IL-21 3–100 μg/kg Fever, myalgias, ↑ liver function tests, neutropenia, and thrombocytopenia. Increased soluble CD25 Renal cell carcinoma, melanoma
Increased frequencies of NK and CD8+ T cells
Increased cytotoxicity of NK cells and CD8+ T cells

INTERLEUKIN-15

IL-15 Biology and Preclinical Studies

IL-15 was identified as a non-IL-2 growth factor that signaled through IL-2Rβ and γc.3436 It has structural similarities to IL-2 but distinct biologic properties.3739 Although IL-2 is produced by activated T cells, IL-15 is produced by a myriad of cell types, both bone marrow derived and nonbone marrow derived.40,41 Unlike the other γc cytokines, which function as soluble ligands, IL-15 largely functions as a cell associated cytokine bound to IL-15Rα expressing cells, predominantly monocytes4245 and dendritic cells.46 Thus, interleukin-15 signaling occurs when a cell-associated IL-15/IL-15Ra complex binds the IL-2Rβγc heterodimer on activated and memory T cells or NK cells. IL-15 is required for NK-cell development and maintenance and maintenance of the CD8 memory pool.47,48 IL-15 is not required to generate memory CD8+ T cells, but their survival depends on this factor.4952 IL-7 can substitute for IL-15 in this regard in lymphopenic hosts where elevated IL-7 levels occur53 or when IL-7 is overexpressed in T-cell replete hosts49 but not under normal conditions. IL-15 is also necessary for homeostasis of NKT cells.54

IL-15 potently costimulates CD8+ T cells, and IL-15 administration expands the memory pool. Several studies have demonstrated that IL-15 therapy increases the size of the effector and memory pool generated in response to infection or immuniza-tion.23,5557 For instance, IL-15 enhances CD8+ T-cell immunity in mice infected with T. gondii,56 IL-15 transgenic mice generate higher numbers of effectors after infection with L. monocytogenes,57 and therapy with either IL-7 or IL-15 improves survival of M. tuberculosis infected mice.22 Thus, IL-15 enhances the immune response generated to intracellular pathogens, leading to increased eradication in the short term. Although augmentation of NK-cell expansion and survival may contribute to these effects, T cells contribute substantially as well. IL-15 therapy also enhances T-cell responses to DNA vaccines,55,58 and dendritic cell vaccines in mice,23,59 and influenza and tetanus vaccines in rhesus macaques.60 The transpresentation of IL-15 has led to the insight that therapies aimed at enhancing availability of this agent in vivo are more effective when IL-15 is provided access to IL-15Rα.6163

In several studies evaluating vaccine adjuvant and antitumor effects, IL-15 was directly compared with IL-2 and demonstrated to be superior.23,55,60 In 1 study, evaluation of the effect of IL-15 on immunodominant and subdominant antigens demonstrated that IL-15 more potently augmented responses to subdominant antigens, when compared with dominant antigens, similar to the effects observed when IL-7 is administered in the setting of vaccination. This is particularly pertinent for tumor antigens, which are generally weak antigens, and which may be better modeled by subdominant antigens. In addition to potent effects on the T-cell effector pool, 2 studies undertook careful analysis of the effects of IL-15 on the long-lived memory pool. In both, treatment with rhIL-15 at the time of immunization led to durable increases in the survival of the memory pool, an effect which remained long after the therapeutic levels of IL-15 had cleared.55 Interestingly, a recent study also implicated IL-15 in enhancing humoral immune responses to a dendritic cell vaccine in a CD4+ independent manner, which contributed to antitumor effects.64

The potent survival of IL-15 and differentiation signals for CD8 memory cells and NK cells render it an attractive candidate for use in the context of adoptive immunotherapy for cancer. Preclinical models have confirmed its activity in this regard. For instance, T-cell receptor transgenic cells specific for the gp100 melanoma antigen cells cultured in the presence of IL-15 ex vivo were more potent than cells cultured in the absence of cytokines and more potent than cells cultured with IL-2 in inducing regression of established melanomas in a murine melanoma model.65 Similarly, IL-15 was superior to IL-2 after adoptive transfer of antigen-specific T cells in murine plasmacytoma,66 and transduction of tumors with IL-15 or tumor-specific T cells with IL-1567 induces potent antitumor effects.65,67 Similarly, ex vivo culture of tumor reactive CD8+ T cells found in tumor draining lymph nodes with IL-15 led to differentiation toward a central memory phenotype, directly implicating IL-15 in this process.68 Thus, substantial preclinical data demonstrate that IL-15 is a potent adjuvant for adoptive T-cell therapy, both when used to culture cells ex vivo and when administered in vivo after transfer.

NK cells are less proliferative than T cells; however, recent work suggests that substantial numbers of NK cells can be generated for use in adoptive therapy using artificial antigen-based culture conditions.69 IL-15 substantially augments ex vivo NK expansion in these systems, and although NK cells expanded with IL-2 also show enhanced cytotoxic activity, preliminary studies suggest that IL-15-expanded NK cells may be more potent.70 Similarly, in vivo therapy with IL-15 seems to induce more substantial NK expansion than IL-2 therapy.71 Thus, the central role of IL-15 as a growth and survival factor for developing and mature NK cells has led to the prediction that it will be a potent adjuvant for NK-cell-based immunotherapy. However, IL-2, IL-15, and IL-21, all have substantial effects on NK cells, and optimal use of these factors for ex vivo expansion and in vivo survival of adoptively transferred NK cells could potentially involve 1 or all these agents, and no clearly superior agent or combination has emerged thus far.72,73

Clinical studies have not yet been conducted with rhIL-15, but studies in primates demonstrated that the administration of primate IL-15 at a dose of 10 μg/kg every 2 to 7 days increased responses to tetanus and influenza vaccine.60 In simian immunodeficiency virus-infected macaques receiving antiretroviral therapy, IL-15 (10–100 μg/kg) administered twice weekly for 4 weeks increased circulating NK-cell numbers and CD8+ and CD4+ effector memory populations.74 When rhIL-15 was administered to a macaque using a daily dose of 15 μg/kg/d, Berger et al75 reported reversible neutropenia related to bone marrow suppression in the first animal, 20% weight loss in a second animal and a nonspecific inflammatory dermatitis in a 3rd animal treated with a reduced daily dose of 5 μg/kg. Despite these clinical toxicities, there was clear evidence for biologic activity with increased numbers and cycling rates of peripheral blood CD4+ T cells, CD8+ T cells, γδ T cells (CD4CD8), and NK cells, with preferential expansion of memory T cells, when compared with naive subsets. Subsequent studies in a total of 7 animals used an intermittent regimen (2.5–10 μg/kg every 3 days), which was well tolerated and also associated with substantial expansion of CD4+ and CD8+ memory T cells but lesser effects on NK cells and γδ T cells. Thus, preclinical data in primates suggest that a safe dose and regimen of IL-15 can be defined wherein the effects of IL-15 on memory T cells and NK cells could be exploited clinically.

INTERLEUKIN-21

IL-21 Biology and Preclinical Studies

IL-21 is the newest member added to the γc cytokine family, discovered in 2000.76,77 IL-21 is structurally similar to IL-2 and has many similar properties. The receptor for IL-21 comprises a unique subunit (IL-21R) and the common γc.78,79 The IL-21R complex is expressed on most mature lymphocytes, including T, B and NK cells, but IL-21 production is restricted to activated CD4+ T cells, especially Th17 cells8082 and T follicular helper (Tfh) cells. Because IL-21 production is restricted, cytokine availability likely plays an important role in regulating the effects of IL-21. IL-21 signaling on CD4+ T cells, in combination with TGFβ, drives development of the Th17 subset and restricts induced Treg generation.80,81 The combination of IL-21 signaling on, and IL-21 production by, activated Th17 cells results in an autocrine loop, which plays an important role in autoimmune and inflammatory states such as rheumatoid arthritis, SLE, psoriasis,83 and GVHD.84 In CD8 T cells, IL-21 signaling costimulates for activation and enhances effector function. In NK cells, IL-21 induces maturation and enhances cytotoxic killing.85 In B cells, IL-21 signaling alone induces apoptosis, but in the presence of CD40 and anti-IgM, IL-21 can promote class switching and differentiation of memory B and plasma cells. Moreover, IL-21 signaling on CD4+ Tfh cells, which reside within the germinal center, provides cognate help for isotype switching and high-affinity antibody production.86 Thus, although IL-21 induces apoptosis of B cells, it plays a major role in the generation of effective humoral immunity through indirect effects within the germinal center. In contrast to the immune activating effects described earlier, IL-21 inhibits dendritic cell differentiation and activation.87,88

The capacity of IL-21 to direct differentiation of inflammatory CD4+ T cells while inhibiting Tregs and enhancing cytolytic function of CD8 and NK cells makes it an attractive cytokine for use in cancer immunotherapy. IL-21 treatment can inhibit growth of syngeneic tumors including Rena renal carcinoma and B16 melanoma, both with89 and without90,91 adoptive transfer of tumor-specific T cells. Similarly, antitumor effects have been observed in several models where murine tumors were genetically modified to secrete IL-21.9295 In these models, tumors modified to secrete IL-21 make potent vaccines that induce memory responses and a capacity to respond to parental cells lines not modified with IL-21.95,96 IL-21 also enhances the effectiveness of antibody-dependent cell-mediated cytotoxicity, potentially by upregulation of CD247 on NK cells.97100

The costimulatory properties of IL-21 have led to its use in expansion of antitumor effectors ex vivo. Combination treatment with IL-21 and IL-15 yielded more CD8+ and CD56+ cells, fewer Tregs, and enhanced antigen-specific cytotoxicity, when compared with treatment with IL-2.101,102 Similarly, recent work demonstrated that IL-21 enhanced the effectiveness of human cells bearing a chimeric antigen receptor targeting CD19 in a xenograft model, compared with IL-15 and IL-2.103 In 1 study, IL-21 outperformed both IL-2 and IL-15 as an adjuvant for adoptive transfer of tumor antigen-specific T cells.104

Based on evidence that IL-21 can enhance both CD8 and NK-cell function, the results described earlier are not surprising. However, more detailed work demonstrates that IL-21 also induces a distinct pathway of CD8 differentiation that differs fundamentally from IL-2 mediated differentiation. Although IL-2 enhances the generation of CD44+ granzyme+ CD8+ cells that express Eomeso-dermin, IL-21 generates CD8+ T cells with repressed IL-2Rα but enhanced l-selectin expression and enhanced antitumor effects.105 Thus, through as yet poorly understood mechanisms, IL-21 may prevent terminal differentiation of cytolytic effectors ex vivo, and in turn enhance subsequent functionality of IL-21 cultured cells on transfer in vivo. In addition, to its distinct pathway of CD8 differentiation, another study administered IL-21 in concert with IL-2 as an adjuvant to T-cell transfer and noted simultaneous enhancement of cellular and humoral responses by IL-21 administration. Importantly, IL-21 mediated enhancement of humoral immunity contributed to the antitumor effects in this model.106 Thus, in addition to the capacity of IL-21 to enhance CD8 and NK-cell function and augment Th17 cells, IL-21 mediated effects on Tfh cells, which provide help for antibody generation in the germinal center, and may also contribute to the antitumor effects of IL-21.

Clinical Trials With IL-21

Three clinical trials have been conducted with IL-21. The first phase I study was an open-labeled, 2-armed trial of recombinant human IL-21 in patients with metastatic melanoma. Twenty-nine patients were given an intravenous bolus injection at doses from 1 to 100 μg/kg using 2 treatment regimens: thrice weekly for 6 weeks (3/wk) or 3 cycles of daily dosing for 5 days followed by 9 days of rest (5 + 9). IL-21 was generally well tolerated with a maximally tolerated dose of 30 μg/kg. Toxicities included increased alanine and aspartate aminotransferases, neutropenia, fatigue, thrombocytopenia, and lightheadedness with fevers and rigors. One partial response was observed after 3 weeks of treatment and became a complete response 3 months later.107 Pharmacodynamic studies showed elevated serum levels of several cytokines, chemokines, acute-phase proteins, and cell adhesion proteins in a dose-dependent fashion.108

In another phase I trial, 43 patients with metastatic melanoma or renal cell carcinoma were given an intravenous bolus injection of IL-21 at dose levels from 3 to 100 μg/kg, administered for 2 5-day cycles (days 1–5 and 15–19). The most common adverse events included flu-like symptoms, pruritis, and rash. Twelve patients received 5 additional 2-cycle courses of treatment without cumulative toxicity, except 1 patient with reversible grade 4 hepatotoxicity. Of 24 patients treated for malignant melanoma, there was 1 complete response and 11 patients with stable disease. Of 19 patients treated for renal cell carcinoma, there were 4 partial responses and 13 patients with stable disease.109 In a phase II open-labeled, single-armed, 2-staged trial, IL-21 was administered at 30 μg/kg/d in 5-day cycles every 2nd week. The primary objective of this study was antitumor efficacy. Of 24 patients treated, there was 1 complete response and 1 partial response.110 Thus, IL-21 is well tolerated and has shown a reproducible rate of antitumor effects when administered as a single agent to patients with malignant melanoma and renal cell carcinoma. Future studies are awaited combining IL-21 with vaccines and adoptive cell therapies (summarized in Table 2).

GAMMA-C CYTOKINES AND LYMPHOID MALIGNANCIES

Given the role that γc cytokines play in lymphoid development, incorporation of these agents into regimens used to treat lymphoid malignancies could be problematic. Indeed, mice transgenic for IL-7 develop significant lymphoproliferation and mice transgenic for IL-15 develop large granular cell leukemia. Furthermore, most immature and mature lymphocytes that comprise lymphoid leukemias and lymphomas can be expected to proliferate in response to these agents, raising a note of caution for use in this context. IL-21, however, is an exception to this rule. Because of the unique ability of IL-21 to induce apoptosis in B cells, a logical application of this cytokine in cancer immunotherapy would be against B-cell malignancies. Accordingly, in a murine model, IL-21 can induce caspase-dependant apoptosis of diffuse large B-cell lymphoma cells, which exhibit widespread expression of the IL-21 receptor.111 In another study, IL-21 was shown to induce apoptosis in vitro in mantle cell lymphoma, a subtype of B-cell lymphoma, by a STAT1-dependent pathway,112 and IL-21 has also been reported to induce apoptosis of CLL, through upregulation of the BH3 family member BIM.98,113 In a model using CD19+ lymphoma and adoptively transferred CD19-specific T-cells that constitutively express IL-2, IL-7, IL-15, or IL-21, each cytokine enhanced eradiation of B-cell malignancy, but IL-7 and IL-21 were most efficacious in vivo.103 Importantly, however, T-cell lymphomas do not apoptose after IL-21 signaling, and in anaplastic lymphoma kinase-positive anaplastic large cell lymphoma, IL-21 contributed to JAK/STAT3 activation and cell growth.114 Similarly, growth of Hodgkin lymphoma cell lines, although involving a B-cell lineage, can be potentiated by IL-21.115,116 Thus, special consideration should be given to the effects of γc cytokines on the tumors themselves when considering incorporation of these agents as adjuvant to immune based therapies targeting these diseases.

SUMMARY

Promising results continue to be generated with the use of immunomodulators, tumor vaccines, and adoptive cell therapy for cancer, but the overall effectiveness of immunotherapies against established tumors remains limited. Preclinical work demonstrates that the γc cytokines, which are potent T-cell and NK-cell growth factors, can augment the potency and antitumor effects of a variety of immune based therapies. Although IL-2 is the γc prototype, there is now ample evidence that IL-7, IL-15, and IL-21 are more effective adjuvants for vaccine-based and adoptive immunotherapy than IL-2. Their properties overlap somewhat, and each has been demonstrated to enhance long-term responses to vaccines, long after the cytokine has been discontinued. Furthermore, all have been demonstrated to induce antitumor activity when tumor cells are genetically engineered to secrete the cytokine, and all have augmented adoptive therapies when used to culture cells ex vivo or when administered in vivo in conjunction with cell transfer. Beyond this, each has unique properties that should serve to guide their incorporation into multimodal regimens in the clinic. Thus far, their toxicity profiles seem acceptable although only limited clinical studies have been conducted with rhIL-7 and rhIL-21, and rhIL-15 has not yet been tested in humans. Careful clinical trials are needed to define the dose, schedule, and setting where their biologic effects can be maximally leveraged to enhance the effectiveness of immunotherapy.

REFERENCES

  • 1.Finke LH, Wentworth K, Blumenstein B, et al. Lessons from randomized phase III studies with active cancer immunotherapies-outcomes from the 2006 meeting of the Cancer Vaccine Consortium (CVC). Vaccine. 2007;25(suppl 2):B97–B109. [DOI] [PubMed] [Google Scholar]
  • 2.Sznol M Betting on immunotherapy for melanoma. Curr Oncol Rep. 2009;11:397–404. [DOI] [PubMed] [Google Scholar]
  • 3.Zhang H, Chua KS, Guimond M, et al. Lymphopenia and interleukin-2 therapy alter homeostasis of CD4+ CD25+ regulatory T cells. Nat Med. 2005;11:1238–1243. [DOI] [PubMed] [Google Scholar]
  • 4.Mazzucchelli R, Durum SK. Interleukin-7 receptor expression: intelligent design. Nat Rev Immunol. 2007;7:144–154. [DOI] [PubMed] [Google Scholar]
  • 5.Kaech SM, Tan JT, Wherry EJ, et al. Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells. Nat Immunol. 2003;4:1191–1198. [DOI] [PubMed] [Google Scholar]
  • 6.Fry TJ, Connick E, Falloon J, et al. A potential role for interleukin-7 in T-cell homeostasis. Blood. 2001;97:2983–2990. [DOI] [PubMed] [Google Scholar]
  • 7.Bolotin E, Annett G, Parkman R, et al. Serum levels of IL-7 in bone marrow transplant recipients: relationship to clinical characteristics and lymphocyte count. Bone Marrow Transplant. 1999;23:783–788. [DOI] [PubMed] [Google Scholar]
  • 8.Napolitano LA, Grant RM, Deeks SG, et al. Increased production of IL-7 accompanies HIV-1-mediated T-cell depletion: implications for T-cell homeostasis. Nat Med. 2001;7:73–79. [DOI] [PubMed] [Google Scholar]
  • 9.Mackall CL, Bare CV, Titus JA, et al. Thymic-independent T cell regeneration occurs via antigen driven expansion of peripheral T cells resulting in a repertoire that is limited in diversity and prone to skewing. J Immunol. 1996;156:4609–4616. [PubMed] [Google Scholar]
  • 10.Goldrath AW, Bevan MJ. Low-affinity ligands for the TCR drive proliferation of mature CD8+ T cells in lymphopenic hosts. Immunity. 1999;11:183–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ernst B, Lee DS, Chang JM, et al. The peptide ligands mediating positive selection in the thymus control T cell survival and homeostatic proliferation in the periphery. Immunity. 1999;11:173–181. [DOI] [PubMed] [Google Scholar]
  • 12.Viret C, Wong FS, Janeway CA Jr. Designing and maintaining the mature TCR repertoire: the continuum of self-peptide:self-MHC complex recognition. Immunity. 1999;10:559–568. [DOI] [PubMed] [Google Scholar]
  • 13.Guimond M, Veenstra RG, Grindler DJ, et al. Interleukin 7 signaling in dendritic cells regulates the homeostatic proliferation and niche size of CD4+ T cells. Nat Immunol. 2009;10:149–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Murphy WJ, Back TC, Conlon KC, et al. Antitumor effects of interleukin-7 and adoptive immunotherapy on human colon carcinoma xenografts. J Clin Invest. 1993;92:1918–1924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Jicha DL, Mule JJ, Rosenberg SA. Interleukin 7 generates antitumor cytotoxic T lymphocytes against murine sarcomas with efficacy in cellular adoptive immunotherapy. J Exp Med. 1991;174:1511–1515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Aoki T, Tashiro K, Miyatake S, et al. Expression of murine interleukin 7 in a murine glioma cell line results in reduced tumorigenicity in vivo. Proc Natl Acad Sci USA. 1992;89:3850–3854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Schroten-Loef C, de Ridder CM, Reneman S, et al. A prostate cancer vaccine comprising whole cells secreting IL-7, effective against subcutaneous challenge, requires local GM-CSF for intra-prostatic efficacy. Cancer Immunol Immunother. 2009;58:373–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Gunnarsson S, Bexell D, Svensson A, et al. Intratumoral IL-7 delivery by mesenchymal stromal cells potentiates IFNgamma-transduced tumor cell immunotherapy of experimental glioma. JNeuroimmunol. 2010;218:140–144. [DOI] [PubMed] [Google Scholar]
  • 19.Cha E, Graham L, Manjili MH, et al. IL-7 + IL-15 are superior to IL-2 for the ex vivo expansion of 4T1 mammary carcinoma-specific T cells with greater efficacy against tumors in vivo. Breast Cancer Res Treat. 2010;122:359–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Caserta S, Alessi P, Basso V, et al. IL-7 is superior to IL-2 for ex vivo expansion of tumour-specific CD4(+) T cells. Eur J Immunol. 2010;40:470–479. [DOI] [PubMed] [Google Scholar]
  • 21.Pellegrini M, Calzascia T, Elford AR, et al. Adjuvant IL-7 antagonizes multiple cellular and molecular inhibitory networks to enhance immunotherapies. Nat Med. 2009;15:528–536. [DOI] [PubMed] [Google Scholar]
  • 22.Maeurer MJ, Trinder P, Hommel G, et al. Interleukin-7 or interleukin-15 enhances survival of Mycobacterium tuberculosis-infected mice. Infect Immun. 2000;68:2962–2970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Melchionda F, Fry TJ, Milliron MJ, et al. Adjuvant IL-7 or IL-15 overcomes immunodominance and improves survival of the CD8+ memory cell pool. JClin Invest. 2005;115:1177–1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Colombetti S, Levy F, Chapatte L. IL-7 adjuvant treatment enhances long-term tumor-antigen-specific CD8+ T-cell responses after immunization with recombinant lentivector. Blood. 2009;113:6629–6637. [DOI] [PubMed] [Google Scholar]
  • 25.Nanjappa SG, Walent JH, Morre M, et al. Effects of IL-7 on memory CD8 T cell homeostasis are influenced by the timing of therapy in mice. J Clin Invest. 2008;118:1027–1039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sportes C, Babb RR, Krumlauf MC, et al. Phase I study of recombinant human interleukin-7 administration in subjects with refractory malignancy. Clin Cancer Res. 2010;16:727–735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Sportes C, Hakim FT, Memon SA, et al. Administration of rhIL-7 in humans increases in vivo TCR repertoire diversity by preferential expansion of naive T cell subsets. J Exp Med. 2008;205:1701–1714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Rosenberg SA, Sportes C, Ahmadzadeh M, et al. IL-7 administration to humans leads to expansion of CD8+ and CD4+ cells but a relative decrease of CD4+ T-regulatory cells. J Immunother. 2006;29:313–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sereti I, Dunham RM, Spritzler J, et al. IL-7 administration drives T cell-cycle entry and expansion in HIV-1 infection. Blood. 2009;113:6304–6314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Levy Y, Lacabaratz C, Weiss L, et al. Enhanced T cell recovery in HIV-1-infected adults through IL-7 treatment. J Clin Invest. 2009;119:997–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Seddiki N, Santner-Nanan B, Martinson J, et al. Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J Exp Med. 2006;203:1693–1700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Liu W, Putnam AL, Xu-Yu Z, et al. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med. 2006;203:1701–1711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ahmadzadeh M, Rosenberg SA. IL-2 administration increases CD4 + CD25(hi) Foxp3+ regulatory T cells in cancer patients. Blood. 2006;107:2409–2414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bamford RN, Grant AJ, Burton JD, et al. The interleukin (IL) 2 receptor beta chain is shared by IL-2 and a cytokine, provisionally designated IL-T, that stimulates T-cell proliferation and the induction of lymphokine-activated killer cells. Proc Natl Acad Sci USA. 1994;91:4940–4944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Burton JD, Bamford RN, Peters C, et al. A lymphokine, provisionally designated interleukin T and produced by a human adult T-cell leukemia line, stimulates T-cell proliferation and the induction of lymphokine-activated killer cells. Proc Natl Acad Sci USA. 1994;91:4935–4939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Grabstein KH, Eisenman J, Shanebeck K, et al. Cloning of a T cell growth factor that interacts with the beta chain of the interleukin-2 receptor. Science. 1994;264:965–968. [DOI] [PubMed] [Google Scholar]
  • 37.Marks-Konczalik J, Dubois S, Losi JM, et al. IL-2-induced activation-induced cell death is inhibited in IL-15 transgenic mice. Proc Natl Acad Sci USA. 2000;97:11445–11450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ku CC, Murakami M, Sakamoto A, et al. Control of homeostasis of CD8+ memory T cells by opposing cytokines. Science. 2000;288:675–678. [DOI] [PubMed] [Google Scholar]
  • 39.Waldmann TA, Dubois S, Tagaya Y. Contrasting roles of IL-2 and IL-15 in the life and death of lymphocytes: implications for immunotherapy. Immunity. 2001;14:105–110. [PubMed] [Google Scholar]
  • 40.Blauvelt A, Asada H, Klaus-Kovtun V, et al. Interleukin-15 mRNA is expressed by human keratinocytes Langerhans cells, and blood-derived dendritic cells and is downregulated by ultraviolet B radiation. J Invest Dermatol. 1996;106:1047–1052. [DOI] [PubMed] [Google Scholar]
  • 41.Waldmann TA, Tagaya Y. The multifaceted regulation of interleukin-15 expression and the role of this cytokine in NK cell differentiation and host response to intracellular pathogens. Annu Rev Immunol. 1999;17:19–49. [DOI] [PubMed] [Google Scholar]
  • 42.Dubois S, Mariner J, Waldmann TA, et al. IL-15Ralpha recycles and presents IL-15 In trans to neighboring cells. Immunity. 2002;17:537–547. [DOI] [PubMed] [Google Scholar]
  • 43.Lodolce JP, Burkett PR, Boone DL, et al. T cell-independent interleukin 15Ralpha signals are required for bystander proliferation. J Exp Med. 2001;194:1187–1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Koka R, Burkett PR, Chien M, et al. Interleukin (IL)-15R[alpha]-deficient natural killer cells survive in normal but not IL-15R[alpha]-deficient mice. J Exp Med. 2003;197:977–984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Burkett PR, Koka R, Chien M, et al. Coordinate expression and trans presentation of interleukin (IL)-15Ralpha and IL-15 supports natural killer cell and memory CD8+ T cell homeostasis. J Exp Med. 2004;200:825–834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Koka R, Burkett P, Chien M, et al. Cutting edge: murine dendritic cells require IL-15R alpha to prime NK cells. JImmunol. 2004;173:3594–3598. [DOI] [PubMed] [Google Scholar]
  • 47.Cooper MA, Bush JE, Fehniger TA, et al. In vivo evidence for a dependence on interleukin 15 for survival of natural killer cells. Blood. 2002;100:3633–3638. [DOI] [PubMed] [Google Scholar]
  • 48.Ranson T, Vosshenrich CA, Corcuff E, et al. IL-15 is an essential mediator of peripheral NK-cell homeostasis. Blood. 2003;101:4887–4893. [DOI] [PubMed] [Google Scholar]
  • 49.Kieper WC, Tan JT, Bondi-Boyd B, et al. Overexpression of interleukin (IL)-7 leads to IL-15-independent generation of memory phenotype CD8+ T cells. JExp Med. 2002;195:1533–1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Tan JT, Ernst B, Kieper WC, et al. Interleukin (IL)-15 and IL-7 jointly regulate homeostatic proliferation of memory phenotype CD8+ cells but are not required for memory phenotype CD4+ cells. J Exp Med. 2002;195:1523–1532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Judge AD, Zhang X, Fujii H, et al. Interleukin 15 controls both proliferation and survival of a subset of memory-phenotype CD8(+) T cells. J Exp Med. 2002;196:935–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wu TS, Lee JM, Lai YG, et al. Reduced expression of Bcl-2 in CD8+ T cells deficient in the IL-15 receptor alpha-chain. JImmunol. 2002;168:705–712. [DOI] [PubMed] [Google Scholar]
  • 53.Goldrath AW, Sivakumar PV, Glaccum M, et al. Cytokine requirements for acute and Basal homeostatic proliferation of naive and memory CD8+ T cells. JExp Med. 2002;195:1515–1522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Ranson T, Vosshenrich CA, Corcuff E, et al. IL-15 availability conditions homeostasis of peripheral natural killer T cells. Proc Natl Acad Sci USA. 2003;100:2663–2668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Oh S, Berzofsky JA, Burke DS, et al. Coadministration of HIV vaccine vectors with vaccinia viruses expressing IL-15 but not IL-2 induces long-lasting cellular immunity. Proc Natl Acad Sci USA. 2003;100:3392–3397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Khan IA, Casciotti L. IL-15 prolongs the duration of CD8 + T cell-mediated immunity in mice infected with a vaccine strain of Toxoplasma gondii. J Immunol. 1999;163:4503–4509. [PubMed] [Google Scholar]
  • 57.Yajima T, Nishimura H, Ishimitsu R, et al. Overexpression of IL-15 in vivo increases antigen-driven memory CD8+ T cells following a microbe exposure. J Immunol. 2002;168:1198–1203. [DOI] [PubMed] [Google Scholar]
  • 58.Xin KQ, Hamajima K, Sasaki S, et al. IL-15 expression plasmid enhances cell-mediated immunity induced by an HIV-1 DNA vaccine. Vaccine. 1999;17:858–866. [DOI] [PubMed] [Google Scholar]
  • 59.Rubinstein MP, Kadima AN, Salem ML, et al. Systemic administration of IL-15 augments the antigen-specific primary CD8+ T cell response following vaccination with peptide-pulsed dendritic cells. J Immunol. 2002;169:4928–4935. [DOI] [PubMed] [Google Scholar]
  • 60.Villinger F, Miller R, Mori K, et al. IL-15 is superior to IL-2 in the generation of long-lived antigen specific memory CD4 and CD8 T cells in rhesus macaques. Vaccine. 2004;22:3510–3521. [DOI] [PubMed] [Google Scholar]
  • 61.Dubois S, Patel HJ, Zhang M, et al. Preassociation of IL-15 with IL-15R alpha-IgG1-Fc enhances its activity on proliferation of NK and CD8+/CD44high T cells and its antitumor action. J Immunol. 2008;180:2099–2106. [DOI] [PubMed] [Google Scholar]
  • 62.Bergamaschi C, Rosati M, Jalah R, et al. Intracellular interaction of interleukin-15 with its receptor alpha during production leads to mutual stabilization and increased bioactivity. JBiol Chem. 2008;283:4189–4199. [DOI] [PubMed] [Google Scholar]
  • 63.Stoklasek TA, Schluns KS, LefTancois L. Combined IL-15/IL-15Ralpha immunotherapy maximizes IL-15 activity in vivo. J Immunol. 2006;177:6072–6080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Steel JC, Ramlogan CA, Yu P, et al. Interleukin-15 and its receptor augment dendritic cell vaccination against the neu oncogene through the induction of antibodies partially independent of CD4 help. Cancer Res. 70:1072–1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Klebanoff CA, Finkelstein SE, Surman DR, et al. IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc Natl Acad Sci USA. 2004;101:1969–1974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Roychowdhury S, May KF Jr, Tzou KS, et al. Failed adoptive immunotherapy with tumor-specific T cells: reversal with low-dose interleukin 15 but not low-dose interleukin 2. Cancer Res. 2004;64:8062–8067. [DOI] [PubMed] [Google Scholar]
  • 67.Brentjens RJ, Latouche JB, Santos E, et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat Med. 2003;9:279–286. [DOI] [PubMed] [Google Scholar]
  • 68.Anichini A, Scarito A, Molla A, et al. Differentiation of CD8+ T cells from tumor-invaded and tumor-free lymph nodes of melanoma patients: role of common gamma-chain cytokines. J Immunol. 2003;171:2134–2141. [DOI] [PubMed] [Google Scholar]
  • 69.Fujisaki H, Kakuda H, Imai C, et al. Replicative potential of human natural killer cells. Br J Haematol. 2009;145:606–613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Fujisaki H, Kakuda H, Shimasaki N, et al. Expansion of highly cytotoxic human natural killer cells for cancer cell therapy. Cancer Res. 2009;69:4010–4017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ozdemir O, Ravindranath Y, Savasan S. Mechanisms of superior anti-tumor cytotoxic response of interleukin 15-induced lymphokine-activated killer cells. JImmunother. 2005;28:44–52. [DOI] [PubMed] [Google Scholar]
  • 72.Brady J, Hayakawa Y, Smyth MJ, et al. IL-21 induces the functional maturation of murine NK cells. J Immunol. 2004;172:2048–2058. [DOI] [PubMed] [Google Scholar]
  • 73.Sivori S, Cantoni C, Parolini S, et al. IL-21 induces both rapid maturation of human CD34 + cell precursors towards NK cells and acquisition of surface killer Ig-like receptors. Eur J Immunol. 2003;33:3439–3447. [DOI] [PubMed] [Google Scholar]
  • 74.Mueller YM, Petrovas C, Bojczuk PM, et al. Interleukin-15 increases effector memory CD8 + t cells and NK Cells in simian immunodeficiency virus-infected macaques. J Virol. 2005;79:4877–4885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Berger C, Berger M, Hackman RC, et al. Safety and immunologic effects of IL-15 administration in nonhuman primates. Blood. 2009;114:2417–2426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Dumoutier L, Van Roost E, Colau D, et al. Human interleukin-10-related T cell-derived inducible factor: molecular cloning and functional characterization as an hepatocyte-stimulating factor. Proc Natl Acad Sci USA. 2000;97:10144–10149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ozaki K, Kikly K, Michalovich D, et al. Cloning of a type I cytokine receptor most related to the IL-2 receptor beta chain. Proc Natl Acad Sci USA. 2000;97:11439–11444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Habib T, Senadheera S, Weinberg K, et al. The common gamma chain (gamma c) is a required signaling component of the IL-21 receptor and supports IL-21-induced cell proliferation via JAK3. Biochemistry. 2002;41:8725–8731. [DOI] [PubMed] [Google Scholar]
  • 79.Asao H, Okuyama C, Kumaki S, et al. Cutting edge: the common gamma-chain is an indispensable subunit of the IL-21 receptor complex. JImmunol. 2001;167:1–5. [DOI] [PubMed] [Google Scholar]
  • 80.Korn T, Bettelli E, Gao W, et al. IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells. Nature. 2007;448:484–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Nurieva R, Yang XO, Martinez G, et al. Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature. 2007;448:480–483. [DOI] [PubMed] [Google Scholar]
  • 82.Wei L, Laurence A, Elias KM, et al. IL-21 is produced by Th17 cells and drives IL-17 production in a STAT3-dependent manner. J Biol Chem. 2007;282:34605–34610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Nurieva RI, Dong C. Keeping autoimmunity in check: how to control a Th17 cell controller. Immunity. 2008;29:841–843. [DOI] [PubMed] [Google Scholar]
  • 84.Bucher C, Koch L, Vogtenhuber C, et al. IL-21 blockade reduces graft-versus-host disease mortality by supporting inducible T regulatory cell generation. Blood. 2009;114:5375–5384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kasaian MT, Whitters MJ, Carter LL, et al. IL-21 limits NK cell responses and promotes antigen-specific T cell activation: a mediator of the transition from innate to adaptive immunity. Immunity. 2002;16:559–569. [DOI] [PubMed] [Google Scholar]
  • 86.Vogelzang A, McGuire HM, Yu D, et al. A fundamental role for interleukin-21 in the generation of T follicular helper cells. Immunity. 2008;29:127–137. [DOI] [PubMed] [Google Scholar]
  • 87.Strengell M, Lehtonen A, Matikainen S, et al. IL-21 enhances SOCS gene expression and inhibits LPS-induced cytokine production in human monocyte-derived dendritic cells. J Leukoc Biol. 2006;79:1279–1285. [DOI] [PubMed] [Google Scholar]
  • 88.Brandt K, Bulfone-Paus S, Foster DC, et al. Interleukin-21 inhibits dendritic cell activation and maturation. Blood. 2003;102:4090–4098. [DOI] [PubMed] [Google Scholar]
  • 89.Petersen CC, Diernaes JE, Skovbo A, et al. Interleukin-21 restrains tumor growth and induces a substantial increase in the number of circulating tumor-specific T cells in a murine model of malignant melanoma. Cytokine. 49:80–88. [DOI] [PubMed] [Google Scholar]
  • 90.Eriksen KW, Sondergaard H, Woetmann A, et al. The combination of IL-21 and IFN-alpha boosts STAT3 activation, cytotoxicity and experimental tumor therapy. Mol Immunol. 2009;46:812–820. [DOI] [PubMed] [Google Scholar]
  • 91.Sondergaard H, Frederiksen KS, Thygesen P, et al. Interleukin 21 therapy increases the density of tumor infiltrating CD8+ T cells and inhibits the growth of syngeneic tumors. Cancer Immunol Immunother. 2007;56:1417–1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Di Carlo E, Comes A, Orengo AM, et al. IL-21 induces tumor rejection by specific CTL and IFN-gamma-dependent CXC chemokines in syngeneic mice. J Immunol. 2004;172:1540–1547. [DOI] [PubMed] [Google Scholar]
  • 93.Ma HL, Whitters MJ, Konz RF, et al. IL-21 activates both innate and adaptive immunity to generate potent antitumor responses that require perforin but are independent of IFN-gamma. J Immunol. 2003;171:608–615. [DOI] [PubMed] [Google Scholar]
  • 94.Kim-Schulze S, Kim HS, Fan Q, et al. Local IL-21 promotes the therapeutic activity of effector T cells by decreasing regulatory T cells within the tumor microenvironment. Mol Ther. 2009;17:380–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Croce M, Meazza R, Orengo AM, et al. Immunotherapy of neuroblastoma by an Interleukin-21-secreting cell vaccine involves survivin as antigen. Cancer Immunol Immunother. 2008;57:1625–1634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Zhao F, Dou J, He XF, et al. Enhancing therapy of B16F10 melanoma efficacy through tumor vaccine expressing GPI-anchored IL-21 and secreting GM-CSF in mouse model. Vaccine. 2010;28:2846–2852. [DOI] [PubMed] [Google Scholar]
  • 97.Roda JM, Parihar R, Lehman A, et al. Interleukin-21 enhances NK cell activation in response to antibody-coated targets. JImmunol. 2006;177:120–129. [DOI] [PubMed] [Google Scholar]
  • 98.Gowda A, Roda J, Hussain SR, et al. IL-21 mediates apoptosis through up-regulation of the BH3 family member BIM and enhances both direct and antibody-dependent cellular cytotoxicity in primary chronic lymphocytic leukemia cells in vitro. Blood. 2008;111:4723–4730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Roda JM, Joshi T, Butchar JP, et al. The activation of natural killer cell effector functions by cetuximab-coated, epidermal growth factor receptor positive tumor cells is enhanced by cytokines. Clin Cancer Res. 2007;13:6419–6428. [DOI] [PubMed] [Google Scholar]
  • 100.Watanabe M, Kono K, Kawaguchi Y, et al. Interleukin-21 can efficiently restore impaired antibody-dependent cell-mediated cytotoxicity in patients with oesophageal squamous cell carcinoma. Br J Cancer. 2010;102:520–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Huarte E, Fisher J, Turk MJ, et al. Ex vivo expansion of tumor specific lymphocytes with IL-15 and IL-21 for adoptive immunotherapy in melanoma. Cancer Lett. 2009;285:80–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Pouw N, Treffers-Westerlaken E, Kraan J, et al. Combination of IL-21 and IL-15 enhances tumour-specific cytotoxicity and cytokine production of TCR-transduced primary T cells. Cancer Immunol Immunother. 2010;59:921–931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Markley JC, Sadelain M. Interleukin (IL)-7 and IL-21 are superior to IL-2 and IL-15 in promoting human T-cell-mediated rejection of systemic lymphoma in immunodeficient mice. Blood. 2010;115:3508–3519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Moroz A, Eppolito C, Li Q, et al. IL-21 enhances and sustains CD8+ T cell responses to achieve durable tumor immunity: comparative evaluation of IL-2, IL-15, and IL-21. J Immunol. 2004;173:900–909. [DOI] [PubMed] [Google Scholar]
  • 105.Hinrichs CS, Spolski R, Paulos CM, et al. IL-2 and IL-21 confer opposing differentiation programs to CD8+ T cells for adoptive immunotherapy. Blood. 2008;111:5326–5333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Iuchi T, Teitz-Tennenbaum S, Huang J, et al. Interleukin-21 augments the efficacy of T-cell therapy by eliciting concurrent cellular and humoral responses. Cancer Res. 2008;68:4431–4441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Davis ID, Skrumsager BK, Cebon J, et al. An open-label, two-arm, phase I trial of recombinant human interleukin-21 in patients with metastatic melanoma. Clin Cancer Res. 2007;13:3630–3636. [DOI] [PubMed] [Google Scholar]
  • 108.Dodds MG, Frederiksen KS, Skak K, et al. Immune activation in advanced cancer patients treated with recombinant IL-21: multianalyte profiling of serum proteins. Cancer Immunol Immunother. 2009;58:843–854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Thompson JA, Curti BD, Redman BG, et al. Phase I study of recombinant interleukin-21 in patients with metastatic melanoma and renal cell carcinoma. J Clin Oncol. 2008;26:2034–2039. [DOI] [PubMed] [Google Scholar]
  • 110.Davis ID, Brady B, Kefford RF, et al. Clinical and biological efficacy of recombinant human interleukin-21 in patients with stage IV malignant melanoma without prior treatment: a phase IIa trial. Clin Cancer Res. 2009;15:2123–2129. [DOI] [PubMed] [Google Scholar]
  • 111.Sarosiek KA, Malumbres R, Nechushtan H, et al. Novel IL-21 signaling pathway up-regulates c-Myc and induces apoptosis of diffuse large B-cell lymphomas. Blood. 115:570–580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Gelebart P, Zak Z, Anand M, et al. Interleukin-21 effectively induces apoptosis in mantle cell lymphoma through a STAT1-dependent mechanism. Leukemia. 2009;23:1836–1846. [DOI] [PubMed] [Google Scholar]
  • 113.de Totero D, Meazza R, Capaia M, et al. The opposite effects of IL-15 and IL-21 on CLL B cells correlate with differential activation of the JAK/STAT and ERK1/2 pathways. Blood. 2008;111:517–524. [DOI] [PubMed] [Google Scholar]
  • 114.Dien Bard J, Gelebart P, Anand M, et al. IL-21 contributes to JAK3/STAT3 activation and promotes cell growth in ALK-positive anaplastic large cell lymphoma. Am J Pathol. 2009;175:825–834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Scheeren FA, Diehl SA, Smit LA, et al. IL-21 is expressed in Hodgkin lymphoma and activates STAT5: evidence that activated STAT5 is required for Hodgkin lymphomagenesis. Blood. 2008;111:4706–4715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Lamprecht B, Kreher S, Anagnostopoulos I, et al. Aberrant expression of the Th2 cytokine IL-21 in Hodgkin lymphoma cells regulates STAT3 signaling and attracts Treg cells via regulation of MIP-3alpha. Blood. 2008;112:3339–3347. [DOI] [PubMed] [Google Scholar]

RESOURCES