Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Mar 1.
Published in final edited form as: Immunotherapy. 2012 May;4(5):511–527. doi: 10.2217/imt.12.38

Combination strategies to enhance antitumor ADCC

Holbrook E Kohrt 1,, Roch Houot 2,3,, Aurélien Marabelle 1, Hearn Jay Cho 4, Keren Osman 4, Matthew Goldstein 1, Ronald Levy 1, Joshua Brody 4,*
PMCID: PMC3386352  NIHMSID: NIHMS384451  PMID: 22642334

Abstract

The clinical efficacy of monoclonal antibodies as cancer therapeutics is largely dependent upon their ability to target the tumor and induce a functional antitumor immune response. This two-step process of ADCC utilizes the response of innate immune cells to provide antitumor cytotoxicity triggered by the interaction of the Fc portion of the antibody with the Fc receptor on the immune cell. Immunotherapeutics that target NK cells, γδ T cells, macrophages and dendritic cells can, by augmenting the function of the immune response, enhance the antitumor activity of the antibodies. Advantages of such combination strategies include: the application to multiple existing antibodies (even across multiple diseases), the feasibility (from a regulatory perspective) of combining with previously approved agents and the assurance (to physicians and trial participants) that one of the ingredients – the antitumor antibody – has proven efficacy on its own. Here we discuss current strategies, including biologic rationale and clinical results, which enhance ADCC in the following ways: strategies that increase total target–monoclonal antibody–effector binding, strategies that trigger effector cell ‘activating’ signals and strategies that block effector cell ‘inhibitory’ signals.

Keywords: γδ T cells, ADCC, cancer, cytokines, IMiD, immunocytokines, immunomodulators, interleukins, monoclonal antibodies, NK cells, passive immunotherapy

ADCC

Monoclonal antibodies (mAbs) can target tumor antigens on the surface of cancer cells and have a favorable toxicity profile in comparison with cytotoxic chemotherapy. Expression of tumor antigens is dynamic and inducible through agents such as Toll-like receptor (TLR) agonists, immunomodulatory drugs (IMiDs) and hypomethylating agents [1]. Following binding of the mAb to the tumor antigen, the Fc portion of the mAb interacts with the Fc receptor (FcR) on the surface of effector cells (i.e., NK cells, γδ T cells and macrophages), leading to antitumor cytotoxicity and/or phagocytosis of the tumor cell. FcR interactions can be stimulatory or inhibitory to the killer cell, depending on which FcR is triggered and on which cell. Stimulatory effects are mediated through FcγRI on macrophages, dendritic cells (DCs) and neutrophils, and FcγRIIIa on NK cells, DCs and macrophages. In murine models, the cytotoxicity resulting from FcR activation on a NK cell, γδ T cell and macrophage is responsible for antitumor activity [2]. The role of DCs should be noted: although not considered to be primary ADCC effector cells, they can respond to mAb-bound tumor cells via their own FcR-mediated activation and probably play a significant role in activating effector cells. Preclinical models have shown that, although not the effector cell, DCs are critical to the efficacy of mAb-mediated tumor elimination [3]. Equally, mAb-activated ADCC effector cells can induce DC activation [4] and the importance of this crosstalk is an increasing focus of study [5].

The antitumor effects of mAbs are caused by multiple mechanisms of action, including cell signaling agonism/antagonism, complement activation and ligand sequestration, although ADCC probably plays a predominant role in the efficacy of some mAbs. In a clinical series, a correlation between the affinity of the receptor FcγRIIIa (determined by inherited FcR polymorphisms) and the clinical response to mAb therapy, supporting the significance of the innate immune response [610]. Several strategies could potentially improve the innate response following FcR activation by a mAb (Figure 1):

  • Quantitatively increasing the density of the bound target, mAb or the effector cells;

  • Stimulation of the effector cell by targeting the NK cell, γδ T cell and/or macrophage with small molecules, cytokines or agonistic antibodies;

  • Blocking an inhibitory interaction between the NK cell or macrophage and the tumor cell.

Figure 1. Enhancing ADCC.

Figure 1

FcR: Fc receptor; HDACi: Histone deacetylase inhibitor; IMiD: Immunomodulator; KIR: Killer immunoglobulin-like receptor;

MΦ: Macrophage; mAb: Monoclonal antibody; pDC: Plasmacytoid dendritic cell; TLR: Toll-like receptor.

The ability of the combination approaches to enhance ADCC is largely determined by the capacity of the mAb to induce ADCC. Since the approval of the first mAb for the treatment of non-Hodgkin’s lymphoma, rituximab (RTX), in 1997, several mAbs have become standard of care for the treatment of both solid tumors and hematologic malignancies, including trastuzumab (TRAST), alemtuzumab, cetuximab, panitumumab and ofatumumab [11]. As noted above, clinical series among lymphoma patients treated with an anti-CD20 mAb (RTX) [6,7], HER2-expressing breast cancer receiving anti-HER2 mAb therapy (TRAST) [8] or colorectal cancer patients treated with an anti-EGFR mAb (cetuximab) [9,10] observed a correlation between clinical benefit and FcγRIIIa genotype, with patients who have higher-affinity polymorphisms demonstrating superior clinical outcomes. By contrast, the anti-EGFR mAb panitumumab does not induce ADCC, owing to a different Fc isotype that does not bind to the FcγRIIIa. Therefore, when considering enhancement of ADCC, such approaches are limited to combinations with mAbs that activate the FcR. Nonetheless, an advantage of this dual therapy strategy is that mAbs yet to be discovered against currently unknown tumor antigens may be combined with the therapeutics discussed herein.

Increasing target–mAb–effector binding

As the central element in the target–mAb–effector cell unit, the mAb seems to be a probable candidate for improvements, either in its antigen-binding or its Fc-binding domains. This approach has been heavily pursued with some degree of success [1215]. Antibody engineering to improve interaction between the target or FcR requires that each new antibody be individually developed and tested as a new entity.

Increasing the antigen target

Tumor cells with a lower density of antigen targets are less responsive to mAbs than higher antigen-expressing diseases [16]. Therefore, it seems logical to try to increase the expression of the target on tumor cells. Antigen expression can be upregulated by cytokines [17], ionizing radiation [18], natural metabolites [19] and hypomethylating agents such as decitabine [20]. In addition, the family of TLR9 agonists known as CpG oligodeoxynucleotides (CpG ODN) can induce CD20 expression on malignant B cells [2123]. Taken together with data showing the activating effect of CpG ODN on effector cells (discussed below), it seems reasonable that the combination of CpG ODN with mAb might have synergistic efficacy. Clinical series, however, have tested CpG ODN administered intravenously or subcutaneously and have observed little efficacy in Phase I and II studies [2426] in low-grade lymphoma. One possible limitation of these studies has been their application to diseases (primarily follicular and mantle cell lymphoma) known to already have high expression of the relevant antigen (CD20). It is plausible that increasing antigen expression on low antigen-expressing diseases such as chronic lymphocytic leukemia could have a greater increase in relative efficacy. To this end, monotherapy studies have recently been undertaken [27,301] and should lead to combination trials.

Effector cells: NK cells

Given the importance of the effector cell in ADCC, it is conceivable that by increasing their number, in the context of adequate tumor antigens and mAbs, that the sum antitumor effect could be increased. One approach to test this hypothesis has been to generate high numbers of effector cells ex vivo using either IL-2 or a combination of IL-2, IFN-γ and anti-CD3 mAb, known as lymphokine-activated killer (LAK) cells and cytokine-induced killer cells, respectively, which both contain NK cells as the majority cell type present. Although LAK and cytokine-induced killer cell monotherapy have yet to show significant efficacy, their combination with mAbs has only recently been tested. Murine models have shown robust synergy between a variety of mAbs and transferred effector cells such as LAK and cytokine-induced killer cells [2830]. The same approach was studied combining LAK cells with RTX in a cohort of patients with RTX-refractory lymphoma. In the study, adoptive transfer of LAK cells improved peripheral blood NK cell counts and ADCC, and even induced clinical responses in some patients [31]. A similar combination of LAK cells and RTX is being studied in an ongoing Phase II trial powered for clinical end points [302]. Perhaps the most ambitious – and potentially significant – ongoing test of this idea is a randomized trial comparing the 3-year event-free survival in patients with newly diagnosed diffuse large B cell lymphoma treated with RTX, cyclophosphamide, doxorubicin, vincristine and prednisone (R-CHOP) versus R-CHOP plus autologous NK cell therapy [303]. To date, 273 out of the planned 276 have been enrolled [Cho SG, Pers. Comm.] and clinical results (as well as data describing the NK cell product) are pending.

Effector cells: γδ T cells

The role of NK cells and macrophages in mediating ADCC has been well established; however, only recently have γδ T cells been found to play a role as ADCC effectors. Typically, this population is considered as a minor subset (<5% of circulating T cells), although they may infiltrate tumors of epithelial origin preferentially and constitute a large portion of the tumor-infiltrating lymphocytes in cancers such as breast carcinoma. The combination of HLA-unrestricted cytotoxicity against multiple tumor cell lines of various histologies, secretion of cytolytic granules and proinflammatory cytokines such as TNF-α, IL-17 and IFN-γ make γδ T cells potentially potent antitumor effectors [32,33]. Clinical evidence for γδ T-cell function includes presence postchemotherapy predicting tumor response, and persistence following bone marrow transplantation correlating with survival [34,35]. The selectivity of γδ T cells was seen following culture and expansion of tumor-infiltrating lymphocytes that were unable to lyse normal tissues, but remained cytotoxic against autologous tumor [33].

Of significant relevance to ADCC, a subset of γ9δ2 T cells upregulate CD16 (FcγRIII) upon activation [36,37]. CD16high γδ T cells constitutively express several NK cell receptors including NKG2A–CD94 and express high amounts of perforin, but low levels of IFN-γ [36]. They specifically respond to activation via CD16 and are capable of lysing tumors following exposure to mAbs including RTX [3840], TRAST [39,40], alemtuzumab [39], ofatumumab [38] and GA101 [38], and may facilitate NK cell function [41]. The synergy between γδ T cells and NK cells relies on an interaction between the CD137 ligand and CD137; activated γ9γ2 T cells enhance antitumor cytotoxicity of NK cells through CD137 engagement [42].

Clinically, γδ T cells are promising for translation as they are easily collected by leukapheresis and can be used as adoptive therapy in combination with mAbs [43,44]. The highly restricted T-cell receptor repertoire (over 90% limited to the γ9δ2 subset) can be stimulated directly by synthetic compounds including bromohydrin pyrophosphate or indirectly by bisphosphonates [45]. Phase I and II clinical trials using γδ T cells a as monotherapy in solid tumors [4651] and hematological malignancies [52] have been initiated with modest results to date [32]. Combination approaches with mAbs including RTX and TRAST are in development.

Targeting effector cell activation: small molecules

TLR agonists

In addition to its aforementioned induction of CD20, CpG ODN also indirectly augments innate immune function. TLRs are specialized to recognize pathogen-associated molecular patterns; they stimulate plasmacytoid DCs and B cells [53], and one of many plasmacytoid DC responses to stimulation by CpG ODNs is activation of local NK cells, thus improving spontaneous cytotoxicity and ADCC [54]. CpG ODN effects on NK cells appeared to be indirect and IFN-γ production by T cells (possibly in response to plasmacytoid DC activation) has been hypothesized as the intermediary of NK cell activation. Preclinical data in lymphoma and HER2-expressing breast cancer show that CpG ODN augment antitumor efficacy of RTX [55,56] and TRAST [5759]. Similar results have been seen with non-CpG ODN TLR9 agonists, referred to as immunomodulatory oligonucleotides, in augmenting both cetuximab [60] and TRAST [61]. Similar preclinical boosting of TRAST- and RTX-mediated ADCC and increases in NK cell cytotoxicity has been with agonists of TLR2 [62], TLR3 [63], TLR4 [64], TLR7 [65] and TLR8 [66]. While each of these TLRs have a distinct expression profile, many are broadly expressed on myeloid subsets including monocytes, macrophages and DCs. By contrast to other myeloid activators (such as GM-CSF, discussed below), there is evidence that TLR agonists may stimulate immune cell function without the concomitant activation of suppressive cell types such as myeloid-derived suppressor cells [67]. Even beyond direct TLR effects on ADCC effector cells, DC activation status is extremely sensitive to TLR agonist exposure and, as noted previously, these cells profoundly impact ADCC effector cell function.

Immunomodulatory drugs

IMiDs have shown clinical activity in multiple hematologic malignancies despite their primary mechanism of action being unclear. Among their biologic effects (particularly lenalidomide) there are demonstrable and pleiotropic effects on immune cells and signaling molecules. These include enhancement of in vitro NK cell- and monocyte-mediated ADCC on RTX-coated [68] as well as TRAST- and cetuximab-coated tumor cells [69]. In vivo studies in a human lymphoma severe combined immune deficiency mouse model demonstrated significant increases in NK cell recruitment to tumors mediated via microenvironment cytokine changes and augmented RTX-associated ADCC [70]. Studies suggest that IMiD activation of NK cells occurs indirectly; partly via IL-2 induction by T cells [71]. Clinically, a recent study noted significant increases in peripheral blood NK cells, NK cell cytotoxicity and serum IL-2, IL-15 and GM-CSF [72], the potential ADCC-promoting effects of which are discussed below.

There have been several studies combining lenalidomide with RTX [7375], as well as ongoing studies with cetuximab [304306]. Among the most remarkable of these is a recent report of RTX plus lenalidomide for previously untreated indolent lymphoma, which yielded a complete response (CR) in 66% and partial response (PR) in 24% of the 70 patients evaluated [76]. Although interstudy comparisons are inherently difficult to interpret, lenalidomide monotherapy for relapsed disease has yielded CR in 7% and PR in 16% [77], while RTX for previously untreated disease has yielded CR in 37% and PR in 36% [78]. Taken together, these data suggest a degree of synergy between the agents, although whether this is by ADCC enhancement is unclear.

Inducers of NKG2D ligands

NKG2D is an activating immunoreceptor of primary importance on NK cells (also expressed by CD8 T cells, γδ T cells and macrophages). Activating signals mediated through engagement of NKG2D by its ligands on target cells can bypass signals transmitted through inhibitory NK cell receptors, allowing NKG2D to function as a ‘master switch’ in determining the activation status of NK cells [79]. Expression of NKG2D ligands determines the sensitivity of lymphoma cell lines to RTX-mediated ADCC [80]. Forced expression of either murine [81] or human [82] NKG2D ligands on tumor cells sensitizes them to NK cell-mediated cytolysis. Various agents have been shown to upregulate expression of NKG2D ligands including IFN-α [83], all-trans retinoic acid [84], hydroxyurea [85], proteasome inhibitors [86] and various histone deacetylase (HDAC) inhibitors [87]. HDAC inhibitors augment cytotoxic activity of NK cells against tumor cells [88] and ADCC of RTX [89] and TRAST [90], although it is unclear whether this is due to upregulation of NKG2D ligands or target expression on tumor cells. Clinical studies combining HDAC inhibitors have been initiated, for example in chronic lymphocytic leukemia [91].

Targeting effector cell activation: cytokines

GM-CSF/G-CSF

GM-CSF is an important cytokine with profound effects on the differentiation and activation of myeloid cells and, as such, has been widely studied as an adjuvant with vaccines, although its utility in potentiating ADCC has also been seen in preclinical models. Both ADCC and phagocytosis are enhanced in vitro by GM-CSF among multiple leukocyte subsets, including monocytes and lymphocytes [92,93], as well as neutrophils and eosinophils [94], and these effects are significantly greater than that observed with the related cytokines G-CSF and M-CSF [93]. Similar results have been seen in vivo [95] and have been shown to be tumor-protective in the setting of mAb therapy. Potentially important for combination mAb therapy is the observation that GM-CSF induces CD20 upregulation on malignant B cells [96].

In patients, GM-CSF has been shown to increase cells expressing FcRI+ (CD64), FcRII+ (CD32) and FcRIII+ (CD16), and to cause transient increases in ADCC [97]. Despite this, other correlative clinical studies have suggested that high doses of GM-CSF may have a negative impact on ADCC [98]. However, a variety of studies combining GM-CSF with different mAbs have not yielded clear evidence of a benefit in any disease, with the possible exception being neuroblastoma (see Table 1 and the ‘IL-2’ section below). Mechanistically, although GM-CSF augments important effectors including monocytes and macrophages, it may simultaneously activate suppressive immune elements such as myeloidderived suppressor cells [99], which can inhibit NK cell function [100], and, thus, potentially negate the beneficial effects.

Table 1.

Clinical studies combining cytokine and monoclonal antibody therapy.

Cytokine mAb Patients (n) Responses (%) Ref.
IFN-α2b RTX 64
38
CR: 33; PR: 37
CR: 11; PR: 34
[182]
[183]
IFN-α2b RTX 36 (−) IFN-α vs 33 (+) IFN-α CR ↑: 20 vs 21 [184]
PEG–IFN-α2b RTX 9 CR: 11; PR: 22 [111]
GM-CSF RTX 39 CR: 45; PR: 25 [185]
GM-CSF RTX (+) chemotherapy 39
38
20
15
CR: 64; PR: 23
CR: 53
CR: 60; PR: 15
CR: 80; PR: 20
[186]
[187]
[188]
[189]
GM-CSF TRAST 17 CR: 0; PR: 0 [190]
GM-CSF 17–1A (anti-EpCAM) 20 CR: 10; PR: 0 [191]
GM-CSF ch17–1A (chimeric anti-EpCAM) 24 CR: 0; PR: 0 [192]
GM-CSF 3F8 (anti-GD2) 15 CR in BM: 80 [193]
GM-CSF + IFN-α2b 17–1A (anti-EpCAM) 27 CR: 0; PR: 4 [194]
G-CSF RTX 19 CR: 16; PR: 26 [195]
IL-2 RTX 54
34
12
CR: 2; PR: 7
CR: 18; PR: 21
CR: 8; PR: 0
[196]
[116]
[197]
IL-2 TRAST 10
45
13
PR: 10
CR: 4; PR: 4
CR: 0; PR: 0
[198]
[199]
[200]
IL-2 14.2a (anti-GD2) 33 CR: 3; PR: 3 [201]
IL-2 ch14.18 (anti-GD2) 15 CR: 7; PR: 7 [202]
IL-2 + GM-CSF ch14.18 (anti-GD2) (+) BMT/ISO 113 (−) mAb/cytokine vs
113 (+) mAb/cytokine
2-year EFS: 46 vs 66 [119]
IL-12 RTX 45
13
25 (−) IL-12 vs 30 (+) IL-12
CR: 4; PR: 4
CR: 0; PR: 0
CR: 20; PR: 32 vs CR: 13; PR: 23
[199]
[200]
[203]
IL-12 TRAST 13
15
25 (−) IL-12 vs 30 (+) IL-12
CR: 0; PR: 0
CR: 8
CR: 20; PR: 32 vs CR: 13; PR: 23
[200]
[204]
[203]
IL-12 TRAST (+) chemotherapy 21 CR: 5; PR: 19 [205]
IL-21 RTX 21 CR: 10; PR: 29 [138]

Denotes increase in CR rate after induction RTX with continued RTX (−) or (+) IFN-α.

Study limited to previously RTX-refractory patients.

(+): Received the experimental agent; (−): Did not receive the experimental agent; BM: Bone marrow; BMT: Autologous bone marrow transplant; CR: Complete response; EFS: Event-free survival; ISO: Isotretinoin; mAb: Monoclonal antibody; PEG: Polyethylene glycol; PR: Partial response; RTX: Rituximab; TRAST: Trastuzumab.

G-CSF induces the expression of FcRI on neutrophils, enhances neutrophil cytotoxic capacity and ADCC, both in vitro and in vivo, and greatly increases the number of peripheral blood neutrophils. Therefore, it is possible that G-CSF could enhance the antitumor effect of mAbs. A small study testing this approach with RTX for low-grade lymphoma yielded clinical responses apparently comparable to RTX monotherapy.

IFN-α

Type I interferons (IFN-α and IFN-β) are strong stimuli for NK cells, inducing proliferation [101], cytotoxicity via the STAT1 and STAT2 pathways and IFN-γ secretion via the STAT4 pathway [102]. Addition of IFN-α to mAb therapy augments antitumor ADCC in vitro [103105], which could be attributed to effects on NK cells [106] as well as macrophages [107]. In vivo IFN-α has demonstrated significant enhancement of ADCC and antitumor mAb efficacy, particularly when combined with IL-2 [108]. Notably, NK cell cytotoxicity and ADCC from oral cancer patients could be augmented by IFN-α, although peritumoral NK cells were significantly impaired compared with peripheral blood NK cells [106].

Such preclinical data prompted an assessment of the effects of IFN-α on ADCC in early trials in cancer patients and, although these revealed minor augmentations of ADCC and NK cell cytolytic activity at lower doses, at higher doses, the predominant immunomodulatory effect was suppression of ADCC [109]. As another example of the potential inconsistencies between preclinical and clinical data (despite in vitro evidence that IFN-α induces CD20 upregulation on malignant B cells [110]) clinical studies showed no demonstrable CD20 upregulation in lymphoma tumor aspirates [111] and the combination of IFN-α with RTX has demonstrated poor clinical efficacy (Table 1).

IL-2

IL-2 was the first cytokine approved for the treatment of cancer (renal cell cancer and melanoma), based solely upon immunomodulatory effects (versus IFN-α, which has direct cytostatic effects as well). After its discovery as a T-cell growth factor [112], it was observed to induce cyto-toxicity in NK and T cells [113]. IL-2 acts via two distinct receptor complexes: the high-affinity receptor formed by IL-2Rα-, β- and γ-chains, and an intermediate-affinity receptor formed by only the β- and γ-chains. Whereas the high-affinity receptor is expressed on activated T and NK cells, the intermediate receptor is constitutively expressed on CD3-CD56+CD16+ NK cells [114] and a minor subsets of T cells. Notably, a small subset of CD56highCD16 NK cells constitutively expresses the high-affinity receptor and may respond to lower IL-2 concentrations [115]. These subtleties have prompted a variety of dosing schema in combination strategies with mAbs for a variety of tumor types (Table 1). One early-phase clinical study demonstrated significant proof of principle by showing the correlation between IL-2-mediated NK cell expansion and clinical response [116]. Ultimately, despite promising preclinical data [117,118], no study has shown a clear signal of greater efficacy than mAb monotherapy. Similar to the mechanistic concerns noted for GM-CSF, IL-2 also expands both immune stimulatory (e.g., effector T cell) and suppressive (Treg) components thath may diminish the potential benefits.

However, one of the first mAbs to be tested in combination with IL-2, the antiganglioside chimeric mAb ch14.18, was recently shown to be part of a combination immunotherapy (mAb–GM-CSF–IL-2) that improved overall survival in patients with neuroblastoma [119]. Which of the three immunotherapeutic components (or which combination thereof) is needed for this effect is unclear, although the same regimen is currently being tested in a randomized trial with or without the mAb [307].

IL-12

IL-12 is a member of a small family of heterodimeric cytokines and an important immune regulatory cytokine that exerts potent antitumor activity in various preclinical contexts [14,15]. IL-12 (previously known as NK cell-stimulating factor) enhances the ability of NK cells to lyse target cells [120] and provides a link between innate and adaptive immunity [121], as seen in its augmentation of NK cell-mediated killing of HER2+ tumor cells combined with TRAST [122], shown in other studies to depend on NK cell IFN-γ production [123]. Signaling by IL-12p70 through the IL-12 signaling network leads to the polarization of naive CD4+ T cells into a Th1 phenotype [124], which indirectly enhances NK cell ADCC [125]. Several clinical studies have combined IL-12 with mAb (Table 1), demonstrating pharmacodynamic proof of principle, although no clear benefit over a mAb monotherapy. A recently initiated study may yield data on whether cetuximab and IL-12 can be combined to treat patients with advanced oropharyngeal cancer [308].

IL-15

Despite numerous functional similarities to IL-2 and other members of the common γ-chain cytokines, IL-15 has been shown to be the primary signal responsible for NK cell differentiation and development [126128]. Given the central role of NK cells in ADCC, there could be marked synergy between IL-15 and mAb therapy. This has been the case in a variety of preclinical models testing the combination of RTX [54,129] and cetuximab [130]. In addition, there are data demonstrating that some of the potential downsides of IL-2 such as amplification of Tregs might be avoided, or even counteracted, by IL-15 [131].

IL-21

IL-21 regulates the proliferation, maturation and function of multiple leukocyte subtypes including B cells, T cells, NK cells and DCs. IL-21 upregulates the expression of CD16, the FcγRIII subtype required for ADCC, costimulates the secretion of IFN-γ and upregulates the expression of granzyme and perforins, increasing the cytotoxic potential of NK cells [132,133]. Combinations of IL-21 with TRAST [134], cetuximab [135] and RTX [136] have demonstrated increases in in vitro NK cell-mediated ADCC, as well as a mAb-potentiating in vivo antitumor effect [137]. An early-phase study combining IL-21 with RTX demonstrated CRs and PRs (Table 1), even demonstrating efficacy in the setting of RTX-refractory disease [138].

Immunocytokines

A primary limitation of mAb therapy in combination with systemic cytokines is the dose-limiting toxicities of the latter; this may also be a cause of the disparity between lackluster clinical results and encouraging preclinical data. One potential solution is to target the cytokine to the tumor site by conjugating it to the mAb, the resulting fusion proteins frequently being referred to as immunocytokines (ICKs). Still, by localizing the cytokine to the tumor site, the aim has usually been to increase ADCC by activating effector (generally NK) cells. The majority of the constructs have thus utilized IL-2, given its NK cell-activating effects (discussed earlier).

Initial studies of an anti-GD2–IL-2 ICK have shown antitumor activity in patients with neuroblastoma [139] and have even gone on to show that this effect may be NK cell-mediated by virtue of its increased efficacy in patients with (autologous) killer immunoglobulin-like receptor (KIR)–KIR ligand mismatch [140]. The same molecule has also shown biological activity in the melanoma setting [141], although in this setting it appeared to induce tumor infiltration by T cells rather than NK cells. An additional clinical assessment is ongoing [309].

A similar approach targeting CD20 with an IL-2 ICK has shown efficacy in a severe combined immune deficiency mouse lymphoma model [142], and is being developed for combination with both RTX [310] as well as CD19-targeted chimeric antigen receptor-expressing cells [143]. Other IL-2 ICKs have been tested in the clinic in patients with prostate cancer. These have targeted the angiogenesis-associated alternatively-spliced extra domain B of fibronectin [144,145], and the tumor-associated A1 domain of tenascin-C [146] and EpCAM [147]. Meanwhile, ICKs utilizing other cytokines, such as IFN-α, are being developed preclinically [148], as well as conjugates of mAbs with other immune stimulants such as CpG ODN [149].

Targeting effector cell activation: agonistic antibodies

Innate immune effectors such as NK cells and macrophages increase expression of activation markers, such as CD69, following FcR triggering during ADCC. As the process is dependent on cell contact, the activation occurs preferentially at the site of the tumor, which is a potential therapeutic advantage of a combination strategy targeting an innate activation marker. Expression of the activation marker CD137, also known as 4-1BB, a member of the TNF receptor superfamily (TNFRSF), is increased following FcR triggering [150]. Unlike other activation markers, minimal expression of CD137 is observed on the surface of NK cells that have not been stimulated by antibody-coated tumor cells. Agonistic antibodies targeting CD137, in preclinical models, both in vitro and in vivo, augment NK cell function, including degranulation, secretion of IFN-γ and antitumor cytotoxicity [41,151,152]. The combination anti-CD137 antibody approach in addition to RTX and TRAST is supported by innate immune response-dependent murine models [42,151153]. The selectivity of this approach to activated NK cells is clinically promising and being rapidly translated in a Phase I trial evaluating the combination of anti-CD137 mAbs with RTX in patients with lymphoma [311]. Taking a slightly different approach, Woo et al. have constructed a recombinant human 4-1BB ligand fusion protein which naturally dimerizes and activates human T cells in vitro [154]. As this molecule enters Phase I trials, it will be interesting to see whether it has a different activity and/or adverse effects profile compared with the mAb.

Other activating molecules expressed by NK cells may become promising candidates for enhancing ADCC. OX40 (CD134) is another member of the TNFRSF, with some homology and chromosomal proximity to CD137. It is best known for its upregulation on activated T cells but has also been shown to be expressed (as has its ligand OX40L) on NK cells [155]. An early-phase trial of an anti-OX40 mAb was recently initiated for patients with prostate cancer [312]. Upon activation, NK cells upregulate ICOS (CD278), a member of the CD28 superfamily, which is then involved in induction of cytotoxicity and IFN-γ production, and ICOS-mediated stimulation allows activated NK cells to more efficiently lyse tumor cells [156]. Whether these observations will lead to an ADCC-enhancing role for ICOS stimulation remains to be seen.

Blocking effector cell inhibition: antagonistic antibodies

Expression of HLA molecules on normal tissue prevents NK cell-mediated cytotoxicity through interaction with the KIRs. In the setting of virus-infected or tumor-transformed cells that have lost HLA expression, they become susceptible to NK cell spontaneous cytotoxicity [157]. However, by contrast to spontaneous cytotoxicity, ADCC results from the interaction of the activating FcR on the NK cell with the Fc portion of an antibody bound to a target cell. Following stimulation via the FcR, the NK cell becomes activated and lyses target cells through release of cytotoxic granules. Both preclinical and clinical evidence supports this role of NK cell function. A competent Fc receptor on the NK cell is required for the antitumor activity of mAbs such as RTX and TRAST [2,158]. The degree of NK cell infiltration into the tumor site as well as ex vivo function correlate with the clinical response to mAb therapy in patients with colorectal and breast cancer [159,160]. Despite the promising upfront efficacy of mAbs, patients heavily pretreated develop resistance to nearly all mAbs; for example, the response rate to TRAST in the metastatic HER2-expressing breast cancer setting is less than 10%. Multiple mechanisms may contribute to mAb resistance, including increased expression of HLA class I molecules. This results in a masked tumor cell appearing as ‘self ’ and inhibition of NK cell cytotoxicity through KIR interactions [161]. KIR engagement also inhibits FcR signaling through inhibition of ZAP-70, Syk, phosphorylation of both the ζ- and γ-subunits and PLC-γ [162,163].

Killer immunoglobulin-like receptors

Given the inhibitory effects of HLA molecule interaction with KIRs, blocking antibodies may prevent resistance to mAb therapy and tumor escape. In vitro, anti-KIR mAbs that prevent HLA ligation to KIRs lead to increased degranulation, secretion of IFN-γ and spontaneous lysis of tumor cells [164]. When combined in vivo, treatment with RTX plus anti-KIR mAbs augmented NK cell degranulation and IFN-γ secretion [165]. This effect was specific to blockage of the KIR2DL receptor and has since been validated in combination with RTX plus a different anti-KIR2DL mAb, resulting in enhanced cytotoxicity against a RTX-coated lymphoma cell line [166]. The ability of KIR inhibition to enhance cytotoxicity in an autologous system was recently demonstrated using effector NK cells and EBV-transformed B cells as target cells from the same donor. Interestingly, although blockade of KIR2DL enhanced NK cell cytotoxicity, significantly less functional enhancement was observed by blocking KIR3DL. This may suggest that that KIR3DL provides a less potent inhibitory signal, and that patient-specific variance in the impact of KIR inhibition needs to be considered [167]. Early clinical studies have focused on anti-KIR mAb monotherapy in multiple myeloma, although preclinical data has already shown that this same approach can be enhanced by the myeloma-specific targeting anti-CD38 mAb daratumumab [168].

GITR

GITR is, like CD137 and ICOS, another member of the TNFRSF expressed in various cell types, but apparently with differential functions between different cell subsets (as well as between mice and humans). GITR is constitutively expressed on NK cells and is upregulated following activation. Its inhibitory role is evident in that high levels of expression of its ligand (GITRL), observed on multiple tumor types, impairs RTX-induced NK cell ADCC and IFN-γ production, both of which are restored by GITR-blocking antibodies [169,170]. An anti-GITR mAb in development (TRX518) was shown to block the interaction between GITR and its ligand, and to enhance the cytotoxicity of NK cells [171]. An early-phase monotherapy trial was initiated in 2010 [313].

CD47

Similar to KIR inhibition of NK cell function, SIRPα engagement on the surface of macrophages provides a negative signal preventing phagocytosis and macrophage-mediated cytotoxicity following mAb therapy [172]. Recent in vitro studies demonstrated the dependence of mAb efficacy on macrophage phagocytosis of human RTX-opsonized lymphoma cells [173]. Murine studies have shown that macrophages play as important a role as NK cells in the therapeutic effect of anti-CD20 mAbs [174]. Clinical series have also shown that polymorphisms of FcγRIIa (CD32), expressed predominantly on macrophages, can predict the outcome after mAb therapy, including RTX [7], TRAST [175] and cetuximab therapy [10]. SIRPα on the macrophage binds to CD47, a transmembrane protein expressed on tumor cells, preventing their phagocytosis [176]. Although multiple tumor types express CD47, expression on tumor cells normally sensitive to ADCC and phagocytosis results in decreased efficacy of the mAbs such as RTX [177]. Blockade of CD47–SIRPα interaction in vitro results in enhanced phagocytosis and increased antitumor efficacy of RTX in mouse xenotransplant models. In a clinical correlative study, ex vivo RTX-mediated ADCC was impaired in CD47hi versus CD47lo patients. Anti-CD47 blocking mAb restored its function [178]. Anti-CD47 mAbs are in preclinical development with a Phase I trial anticipated.

PD-1

PD-1 is a negative regulatory member of the CD28 superfamily expressed on the surface of activated T cells, B cells, NK cells and macrophages, similar to but more broadly regulatory than CTLA-4. Its two known ligands, PD-L1 and PD-L2, are both expressed on a variety of tumor cell lines. The PD-1–PD-L1 axis modulates the NK cell versus multiple myeloma effect, as seen by its blockade enhancing NK cell function against autologous primary myeloma cells, seemingly through effects on NK cell trafficking, immune complex formation with myeloma cells and cytotoxicity specifically toward PD-L1(+) tumor cells [179]. Two anti-PD-1 mAbs (BMS-936558 and CT-011) are currently in clinical trials, the latter in a combination study with RTX for patients with low-grade follicular lymphoma [314].

Conclusion

The recent approval of an anti-CTLA4 mAb has demonstrated that modulating the immune response can improve patient survival [180,181]. As the immune response is a major determinant of mAb efficacy, the opportunity now exists to combine mAb therapy with IMiDs to enhance their antitumor efficacy. Remarkable advances in the basic science of cellular immunology have increased our understanding of the effector mechanisms of mAb antitumor efficacy. Whereas the earliest iterations of such combinations, for example IL-2 and GM-CSF, may have augmented both effector and suppressive cells, newer approaches such as IL-15 and TLR agonists may more efficiently activate effector cells while minimizing the influence of suppressive cells. Despite these encouraging rationale and preliminary data, clinical evidence is still required to demonstrate whether combination therapies will increase the antitumor effects of mAb.

Still, this approach is unique in combining a tumor-targeting therapy, the mAb, with an immune-enhancing therapy. If successful, these therapies may be combined with multiple mAbs in routine practice, as well as novel mAbs yet to be developed. Various approaches including augmenting antigen expression, stimulating the innate response and blocking inhibitory signals are being explored to determine the optimal synergy with mAb therapies. Therapies targeting NK cells, γδ T cells, macrophages and DCs may ultimately be used in combination to further augment ADCC. Encouraging preclinical studies have led to a number of promising therapeutics, and the results of proof-of-concept clinical trials are eagerly awaited.

Future perspective

Over the next decade it is certain that some combinations of mAbs with IMiDs will become an important weapon in the arsenal of anticancer therapies, with RTX and lenalidomide being one clear example. Ongoing preclinical investigations of the potential synergy of such combinations, and the underlying mechanisms thereof, will allow for rational design of early-phase clinical trials with rigorous measurement of meaningful biomarkers (e.g., increase in NK cell subsets, phenotypic activation state or cytolytic function). Those combinations that demonstrate proof of principle will be brought forward to later-phase trials assessing clinical end points that can confirm true synergy; examples of this could include patients who are refractory to either mAb or the IMiD monotherapy. The greater the effort towards higher-quality biomarker assessments and studying such ‘high-bar’ clinical settings, the more easily combination therapies will be proven when they are brought, ultimately, to randomized, controlled trials.

Executive summary.

ADCC

  • There are several potentially important mechanisms of antibody-mediated tumor killing, including ADCC, cell signaling agonism/antagonism, complement activation and ligand sequestration.

  • ADCC appears to be a primary antitumor mechanism for the majority of clinically approved monoclonal antibodies (mAbs), including rituximab, trastuzumab and cetuximab.

  • ADCC depends upon the binding of mAbs to Fc receptors on effector cells, which include NK cells, γδ T cells, macrophages and dendritic cells.

Increasing the number of mAbs bound to effector cell receptors

  • Antigen target density can be increased by tumoral exposure to cytokines, cytotoxins, hypomethylating agents and Toll-like receptor agonists, such as CpG oligodeoxynucleotides.

  • Effector cells (e.g., NK or γδ T cells) can be increased either by ex vivo expansion with cytokines and activating mAbs or by in vivo expansion with cytokines or other pharmacologics including bisphosphonates.

Stimulation of the effector cell

  • Small molecules including Toll-like receptor agonists, immunomodulatory drugs and inducers of NKG2D ligands can induce activation and proliferation of effector cells, including NK cells.

  • Cytokines such as GM-CSF, G-CSF, IFN-α, IL-2, IL-12, IL-15 or IL-21 can induce activation of effector cells including macrophages, NK cells and dendritic cells.

  • Immunocytokines are a novel class of therapeutics that target cytokines such as IL-2 or IFN-α to the tumor-infiltrating leukocytes by conjugating them to an antitumor mAb, limiting cytokine-mediated effector cell stimulation to the tumor site.

  • Agonistic antibodies to surface molecules such as the costimulators CD137, OX40 and ICOS can be used to activate effector cells such as NK cells. Some of these costimulators are upregulated upon FcR ligation such that the agonistic mAb specifically activate those effector cells exposed to the tumor–mAb target.

Blocking effector cell inhibition

  • Antagonistic antibodies can be used to block effector cell interactions that would otherwise inhibit ADCC or phagocytosis, such as: ligation of killer immunoglobulin-like receptor, GITR or PD-1 on NK cells to their respective ligands, which can be expressed on tumor cells or tumor-infiltrating leukocytes, and ligation of SIRPα on macrophages to the ‘don’t eat me signal’ CD47 molecule on tumor cells.

Acknowledgments

J Brody is supported by NIH funding R00CA140728-03.

Footnotes

Financial & competing interests disclosure

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

Papers of special note have been highlighted as:

▪ of interest

▪▪ of considerable interest

  • 1.Houot R, Kohrt H, Goldstein MJ, Levy R. Immunomodulating antibodies and drugs for the treatment of hematological malignancies. Cancer Metastasis Rev. 2011;30(1):97–109. doi: 10.1007/s10555-011-9274-3. [DOI] [PubMed] [Google Scholar]
  • 2.Clynes RA, Towers TL, Presta LG, Ravetch JV. Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets. Nat. Med. 2000;6(4):443–446. doi: 10.1038/74704. [DOI] [PubMed] [Google Scholar]
  • 3.Haynes NM, Hawkins ED, Li M, et al. CD11c+ dendritic cells and B cells contribute to the tumoricidal activity of anti-DR5 antibody therapy in established tumors. J. Immunol. 2010;185(1):532–541. doi: 10.4049/jimmunol.0903624. [DOI] [PubMed] [Google Scholar]
  • 4.Marquez ME, Millet C, Stekman H, et al. CD16 cross-linking induces increased expression of CD56 and production of IL-12 in peripheral NK cells. Cell. Immunol. 2010;264(1):86–92. doi: 10.1016/j.cellimm.2010.05.002. [DOI] [PubMed] [Google Scholar]
  • 5.Walzer T, Dalod M, Vivier E, Zitvogel L. Natural killer cell-dendritic cell crosstalk in the initiation of immune responses. Expert Opin Biol. Ther. 2005;5(Suppl. 1):S49–S59. doi: 10.1517/14712598.5.1.s49. [DOI] [PubMed] [Google Scholar]
  • 6.Cartron G, Dacheux L, Salles G, et al. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcγRIIIa gene. Blood. 2002;99(3):754–758. doi: 10.1182/blood.v99.3.754. [DOI] [PubMed] [Google Scholar]
  • 7.Weng WK, Levy R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J. Clin. Oncol. 2003;21(21):3940–3947. doi: 10.1200/JCO.2003.05.013. [DOI] [PubMed] [Google Scholar]
  • 8.Musolino A, Naldi N, Bortesi B, et al. Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J. Clin. Oncol. 2008;26(11):1789–1796. doi: 10.1200/JCO.2007.14.8957. [DOI] [PubMed] [Google Scholar]
  • 9.Zhang W, Gordon M, Schultheis AM, et al. FCGR2A and FCGR3A polymorphisms associated with clinical outcome of epidermal growth factor receptor expressing metastatic colorectal cancer patients treated with single-agent cetuximab. J. Clin. Oncol. 2007;25(24):3712–3718. doi: 10.1200/JCO.2006.08.8021. [DOI] [PubMed] [Google Scholar]
  • 10.Bibeau F, Lopez-Crapez E, Di Fiore F, et al. Impact of Fc{γ}RIIa-Fc{γ}RIIIa polymorphisms and KRAS mutations on the clinical outcome of patients with metastatic colorectal cancer treated with cetuximab plus irinotecan. J. Clin. Oncol. 2009;27(7):1122–1129. doi: 10.1200/JCO.2008.18.0463. [DOI] [PubMed] [Google Scholar]
  • 11.Weiner LM, Surana R, Wang S. Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nat. Rev. Immunol. 2010;10(5):317–327. doi: 10.1038/nri2744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wierda WG, Kipps TJ, Mayer J, et al. Ofatumumab as single-agent CD20 immunotherapy in fludarabine-refractory chronic lymphocytic leukemia. J. Clin. Oncol. 2010;28(10):1749–1755. doi: 10.1200/JCO.2009.25.3187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sehn LH, Goy A, Offner FC, et al. Randomized Phase II trial comparing GA101 (obinutuzumab) with rituximab in patients with relapsed CD20+ indolent B-cell non-Hodgkin lymphoma: preliminary analysis of the GAUSS study. Presented at: 53rd ASH Annual Meeting and Exposition; San Diego CA, USA. 10–13 December 2011; (Abstract 269). [Google Scholar]
  • 14.Robak T, Robak E. New anti-CD20 monoclonal antibodies for the treatment of B-cell lymphoid malignancies. BioDrugs. 2011;25(1):13–25. doi: 10.2165/11539590-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 15.Van Meerten T, Hagenbeek A. CD20-targeted therapy: the next generation of antibodies. Semin. Hematol. 2010;47(2):199–210. doi: 10.1053/j.seminhematol.2010.01.007. [DOI] [PubMed] [Google Scholar]
  • 16.Prevodnik VK, Lavrencak J, Horvat M, Novakovic BJ. The predictive significance of CD20 expression in B-cell lymphomas. Diagn. Pathol. 2011;6:33. doi: 10.1186/1746-1596-6-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sivaraman S, Deshpande CG, Ranganathan R, et al. Tumor necrosis factor modulates CD 20 expression on cells from chronic lymphocytic leukemia: a new role for TNF α? Microsc. Res. Tech. 2000;50(3):251–257. doi: 10.1002/1097-0029(20000801)50:3<251::AID-JEMT9>3.0.CO;2-7. [DOI] [PubMed] [Google Scholar]
  • 18.Kunala S, Macklis RM. Ionizing radiation induces CD20 surface expression on human B cells. Int. J. Cancer. 2001;96(3):178–181. doi: 10.1002/ijc.1018. [DOI] [PubMed] [Google Scholar]
  • 19.Dahl M, Bouchelouche P, Kramer-Marek G, Capala J, Nordling J, Bouchelouche K. Sarcosine induces increase in HER2/neu expression in androgen-dependent prostate cancer cells. Mol. Biol. Rep. 2011;38(7):4237–4243. doi: 10.1007/s11033-010-0442-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Takenouchi M, Hirai S, Sakuragi N, Yagita H, Hamada H, Kato K. Epigenetic modulation enhances the therapeutic effect of anti-IL-13R(α)2 antibody in human mesothelioma xenografts. Clin. Cancer Res. 2011;17(9):2819–2829. doi: 10.1158/1078-0432.CCR-10-2986. [DOI] [PubMed] [Google Scholar]
  • 21.Mankai A, Buhe V, Hammadi M, et al. Improvement of rituximab efficiency in chronic lymphocytic leukemia by CpG-mediated upregulation of CD20 expression independently of PU.1. Ann. NY Acad. Sci. 2009;1173:721–728. doi: 10.1111/j.1749-6632.2009.04614.x. [DOI] [PubMed] [Google Scholar]
  • 22.Decker T, Schneller F, Sparwasser T, et al. Immunostimulatory CpG-oligonucleotides cause proliferation, cytokine production, and an immunogenic phenotype in chronic lymphocytic leukemia B cells. Blood. 2000;95(3):999–1006. [PubMed] [Google Scholar]
  • 23.Jahrsdorfer B, Hartmann G, Racila E, et al. CpG DNA increases primary malignant B cell expression of costimulatory molecules and target antigens. J. Leukoc. Biol. 2001;69(1):81–88. [PubMed] [Google Scholar]
  • 24.Friedberg JW, Kelly JL, Neuberg D, et al. Phase II study of a TLR-9 agonist (1018 ISS) with rituximab in patients with relapsed or refractory follicular lymphoma. Br. J. Haematol. 2009;146(3):282–291. doi: 10.1111/j.1365-2141.2009.07773.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Friedberg JW, Kim H, McCauley M, et al. Combination immunotherapy with a CpG oligonucleotide (1018 ISS) and rituximab in patients with non-Hodgkin lymphoma: increased interferon-α/β-inducible gene expression, without significant toxicity. Blood. 2005;105(2):489–495. doi: 10.1182/blood-2004-06-2156. [DOI] [PubMed] [Google Scholar]
  • 26.Leonard JP, Link BK, Emmanouilides C, et al. Phase I trial of Toll-like receptor 9 agonist PF-3512676 with and following rituximab in patients with recurrent indolent and aggressive non Hodgkin’s lymphoma. Clin. Cancer Res. 2007;13(20):6168–6174. doi: 10.1158/1078-0432.CCR-07-0815. [DOI] [PubMed] [Google Scholar]
  • 27.Zent CS, Smith BJ, Ballas ZK, et al. Phase I clinical trial of CpG oligonucleotide 7909 (PF-03512676) in patients with previously treated chronic lymphocytic leukemia. Leuk. Lymphoma. 2011;53(2):211–217. doi: 10.3109/10428194.2011.608451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Schultz KR, Klarnet JP, Peace DJ, et al. Monoclonal antibody therapy of murine lymphoma: enhanced efficacy by concurrent administration of interleukin 2 or lymphokine-activated killer cells. Cancer Res. 1990;50(17):5421–5425. [PubMed] [Google Scholar]
  • 29.Senba T, Kuroki M, Arakawa F, et al. Tumor growth suppression by a mouse/human chimeric anti-CEA antibody and lymphokine-activated killer cells in vitro and in SCID mouse xenograft model. Anticancer Res. 1998;18(1A):17–24. [PubMed] [Google Scholar]
  • 30.Watanabe M, Kubota T, Kitajima M, Hakomori S. Synergetic effect of interleukin-2 and cellular cytotoxicity against a novel tumor-associated carbohydrate antigen Le(a)/Le(a) (dimeric Le(a)) mediated by monoclonal antibody NCC-ST-421 in adoptive immunization using SCID mice. Cancer Immunol. Immunother. 1993;37(4):245–250. doi: 10.1007/BF01518518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Berdeja JG, Hess A, Lucas DM, et al. Systemic interleukin-2 and adoptive transfer of lymphokine-activated killer cells improves antibody-dependent cellular cytotoxicity in patients with relapsed B-cell lymphoma treated with rituximab. Clin. Cancer Res. 2007;13(8):2392–2399. doi: 10.1158/1078-0432.CCR-06-1860. [DOI] [PubMed] [Google Scholar]
  • 32.Gomes AQ, Martins DS, Silva-Santos B. Targeting γδ T lymphocytes for cancer immunotherapy: from novel mechanistic insight to clinical application. Cancer Res. 2010;70(24):10024–10027. doi: 10.1158/0008-5472.CAN-10-3236. [DOI] [PubMed] [Google Scholar]
  • 33.Kabelitz D, Wesch D, He W. Perspectives of γδ T cells in tumor immunology. Cancer Res. 2007;67(1):5–8. doi: 10.1158/0008-5472.CAN-06-3069. [DOI] [PubMed] [Google Scholar]
  • 34.Ma Y, Aymeric L, Locher C, et al. Contribution of IL-17-producing γ δ T cells to the efficacy of anticancer chemotherapy. J. Exp. Med. 2011;208(3):491–503. doi: 10.1084/jem.20100269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Godder KT, Henslee-Downey PJ, Mehta J, et al. Long term disease-free survival in acute leukemia patients recovering with increased γδ T cells after partially mismatched related donor bone marrow transplantation. Bone Marrow Transplant. 2007;39(12):751–757. doi: 10.1038/sj.bmt.1705650. [DOI] [PubMed] [Google Scholar]
  • 36.Angelini DF, Borsellino G, Poupot M, et al. FcγRIII discriminates between 2 subsets of Vγ9Vδ2 effector cells with different responses and activation pathways. Blood. 2004;104(6):1801–1807. doi: 10.1182/blood-2004-01-0331. [DOI] [PubMed] [Google Scholar]
  • 37.Lafont V, Liautard J, Liautard JP, Favero J. Production of TNF-α by human V γ 9V δ 2 T cells via engagement of Fc γ RIIIA, the low affinity type 3 receptor for the Fc portion of IgG, expressed upon TCR activation by nonpeptidic antigen. J. Immunol. 2001;166(12):7190–7199. doi: 10.4049/jimmunol.166.12.7190. [DOI] [PubMed] [Google Scholar]
  • 38.Braza MS, Klein B, Fiol G, Rossi JF. γδ T-cell killing of primary follicular lymphoma cells is dramatically potentiated by GA101, a type II glycoengineered anti-CD20 monoclonal antibody. Haematologica. 2011;96(3):400–407. doi: 10.3324/haematol.2010.029520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Gertner-Dardenne J, Bonnafous C, Bezombes C, et al. Bromohydrin pyrophosphate enhances antibody-dependent cell-mediated cytotoxicity induced by therapeutic antibodies. Blood. 2009;113(20):4875–4884. doi: 10.1182/blood-2008-08-172296. [DOI] [PubMed] [Google Scholar]
  • 40.Tokuyama H, Hagi T, Mattarollo SR, et al. V γ 9 V δ 2 T cell cytotoxicity against tumor cells is enhanced by monoclonal antibody drugs – rituximab and trastuzumab. Int. J. Cancer. 2008;122(11):2526–2534. doi: 10.1002/ijc.23365. [DOI] [PubMed] [Google Scholar]
  • 41.Maniar A, Zhang X, Lin W, et al. Human γδ T lymphocytes induce robust NK cell-mediated antitumor cytotoxicity through CD137 engagement. Blood. 2010;116(10):1726–1733. doi: 10.1182/blood-2009-07-234211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Kohrt HE, Houot R, Goldstein MJ, et al. CD137 stimulation enhances the antilymphoma activity of anti-CD20 antibodies. Blood. 2011;117(8):2423–2432. doi: 10.1182/blood-2010-08-301945. ▪ Excellent preclinical model of agonist antibody-mediated activation of NK cells amplifying ADCC responses.
  • 43.Lamb LS, Lopez RD. γδ T cells: a new frontier for immunotherapy? Biol. Blood Marrow Transplant. 2005;11(3):161–168. doi: 10.1016/j.bbmt.2004.11.015. [DOI] [PubMed] [Google Scholar]
  • 44.Salot S, Laplace C, Saiagh S, et al. Large scale expansion of γ 9 δ 2 T lymphocytes: Innacell γ δ cell therapy product. J. Immunol. Methods. 2007;326(1–2):63–75. doi: 10.1016/j.jim.2007.07.010. [DOI] [PubMed] [Google Scholar]
  • 45.Hayday AC. [γ][δ] cells: a right time and a right place for a conserved third way of protection. Annu. Rev. Immunol. 2000;18:975–1026. doi: 10.1146/annurev.immunol.18.1.975. [DOI] [PubMed] [Google Scholar]
  • 46.Bennouna J, Bompas E, Neidhardt EM, et al. Phase-I study of Innacell γδ, an autologous cell-therapy product highly enriched in γ9δ2 T lymphocytes, in combination with IL-2, in patients with metastatic renal cell carcinoma. Cancer Immunol. Immunother. 2008;57(11):1599–1609. doi: 10.1007/s00262-008-0491-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Dieli F, Vermijlen D, Fulfaro F, et al. Targeting human {γ}δ} T cells with zoledronate and interleukin-2 for immunotherapy of hormone-refractory prostate cancer. Cancer Res. 2007;67(15):7450–7457. doi: 10.1158/0008-5472.CAN-07-0199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kobayashi H, Tanaka Y, Yagi J, et al. Safety profile and anti-tumor effects of adoptive immunotherapy using γ-δ T cells against advanced renal cell carcinoma: a pilot study. Cancer Immunol. Immunother. 2007;56(4):469–476. doi: 10.1007/s00262-006-0199-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Meraviglia S, Eberl M, Vermijlen D, et al. in vivo manipulation of Vγ9Vδ2 T cells with zoledronate and low-dose interleukin-2 for immunotherapy of advanced breast cancer patients. Clin. Exp. Immunol. 2010;161(2):290–297. doi: 10.1111/j.1365-2249.2010.04167.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Nakajima J, Murakawa T, Fukami T, et al. A Phase I study of adoptive immunotherapy for recurrent non-small-cell lung cancer patients with autologous γδ T cells. Eur. J. Cardiothorac. Surg. 2010;37(5):1191–1197. doi: 10.1016/j.ejcts.2009.11.051. [DOI] [PubMed] [Google Scholar]
  • 51.Nicol AJ, Tokuyama H, Mattarollo SR, et al. Clinical evaluation of autologous γ δ T cell-based immunotherapy for metastatic solid tumours. Br. J. Cancer. 2011;105(6):778–786. doi: 10.1038/bjc.2011.293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wilhelm M, Kunzmann V, Eckstein S, et al. γδ T cells for immune therapy of patients with lymphoid malignancies. Blood. 2003;102(1):200–206. doi: 10.1182/blood-2002-12-3665. [DOI] [PubMed] [Google Scholar]
  • 53.Jahrsdorfer B, Weiner GJ. Immunostimulatory CpG oligodeoxynucleotides and antibody therapy of cancer. Semin. Oncol. 2003;30(4):476–482. doi: 10.1016/s0093-7754(03)00236-7. [DOI] [PubMed] [Google Scholar]
  • 54.Moga E, Alvarez E, Canto E, et al. NK cells stimulated with IL-15 or CpG ODN enhance rituximab-dependent cellular cytotoxicity against B-cell lymphoma. Exp. Hematol. 2008;36(1):69–77. doi: 10.1016/j.exphem.2007.08.012. [DOI] [PubMed] [Google Scholar]
  • 55.Betting DJ, Yamada RE, Kafi K, Said J, van Rooijen N, Timmerman JM. Intratumoral but not systemic delivery of CpG oligodeoxynucleotide augments the efficacy of anti-CD20 monoclonal antibody therapy against B cell lymphoma. J. Immunother. 2009;32(6):622–631. doi: 10.1097/CJI.0b013e3181ab23f1. [DOI] [PubMed] [Google Scholar]
  • 56.Wooldridge JE, Ballas Z, Krieg AM, Weiner GJ. Immunostimulatory oligodeoxynucleotides containing CpG motifs enhance the efficacy of monoclonal antibody therapy of lymphoma. Blood. 1997;89(8):2994–2998. [PubMed] [Google Scholar]
  • 57.Wang H, Rayburn ER, Wang W, Kandimalla ER, Agrawal S, Zhang R. Immunomodulatory oligonucleotides as novel therapy for breast cancer: pharmacokinetics in vitro and in vivo anticancer activity, and potentiation of antibody therapy. Mol. Cancer Ther. 2006;5(8):2106–2114. doi: 10.1158/1535-7163.MCT-06-0158. [DOI] [PubMed] [Google Scholar]
  • 58.Van Ojik HH, Bevaart L, Dahle CE, et al. CpG-A and B oligodeoxynucleotides enhance the efficacy of antibody therapy by activating different effector cell populations. Cancer Res. 2003;63(17):5595–5600. [PubMed] [Google Scholar]
  • 59.Roda JM, Parihar R, Carson WE., 3rd CpG-containing oligodeoxynucleotides act through TLR9 to enhance the NK cell cytokine response to antibody-coated tumor cells. J. Immunol. 2005;175(3):1619–1627. doi: 10.4049/jimmunol.175.3.1619. [DOI] [PubMed] [Google Scholar]
  • 60.Rosa R, Melisi D, Damiano V, et al. Toll-like receptor 9 agonist IMO cooperates with cetuximab in K-ras mutant colorectal and pancreatic cancers. Clin. Cancer Res. 2011;17(20):6531–6541. doi: 10.1158/1078-0432.CCR-10-3376. [DOI] [PubMed] [Google Scholar]
  • 61.Damiano V, Garofalo S, Rosa R, et al. A novel Toll-like receptor 9 agonist cooperates with trastuzumab in trastuzumab-resistant breast tumors through multiple mechanisms of action. Clin. Cancer Res. 2009;15(22):6921–6930. doi: 10.1158/1078-0432.CCR-09-1599. [DOI] [PubMed] [Google Scholar]
  • 62.Lu H, Yang Y, Gad E, et al. TLR2 agonist PSK activates human NK cells and enhances the antitumor effect of HER2-targeted monoclonal antibody therapy. Clin. Cancer Res. 2011;17(21):6742–6753. doi: 10.1158/1078-0432.CCR-11-1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Nicodemus CF, Wang L, Lucas J, Varghese B, Berek JS. Toll-like receptor-3 as a target to enhance bioactivity of cancer immunotherapy. Am. J. Obstet. Gynecol. 2010;202(6):608.e1–608.e8. doi: 10.1016/j.ajog.2009.12.001. [DOI] [PubMed] [Google Scholar]
  • 64.Wang S, Astsaturov IA, Bingham CA, et al. Effective antibody therapy induces host-protective antitumor immunity that is augmented by TLR4 agonist treatment. Cancer Immunol. Immunother. 2012;61(1):49–61. doi: 10.1007/s00262-011-1090-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Hamm S, Rath S, Michel S, Baumgartner R. Cancer immunotherapeutic potential of novel small molecule TLR7 and TLR8 agonists. J. Immunotoxicol. 2009;6(4):257–265. doi: 10.3109/15476910903286733. [DOI] [PubMed] [Google Scholar]
  • 66.Lu H, Dietsch GN, Matthews MA, et al. VTX-2337 is a novel TLR8 agonist that activates NK cells and augments ADCC. Clin. Cancer Res. 2012;18(2):499–509. doi: 10.1158/1078-0432.CCR-11-1625. [DOI] [PubMed] [Google Scholar]
  • 67.Zoglmeier C, Bauer H, Norenberg D, et al. CpG blocks immunosuppression by myeloid-derived suppressor cells in tumor-bearing mice. Clin. Cancer Res. 2011;17(7):1765–1775. doi: 10.1158/1078-0432.CCR-10-2672. [DOI] [PubMed] [Google Scholar]
  • 68.Wu L, Adams M, Carter T, et al. Lenalidomide enhances natural killer cell and monocyte-mediated antibody-dependent cellular cytotoxicity of rituximab-treated CD20+ tumor cells. Clin. Cancer Res. 2008;14(14):4650–4657. doi: 10.1158/1078-0432.CCR-07-4405. [DOI] [PubMed] [Google Scholar]
  • 69.Wu L, Parton A, Lu L, Adams M, Schafer P, Bartlett JB. Lenalidomide enhances antibody-dependent cellular cytotoxicity of solid tumor cells in vitro: influence of host immune and tumor markers. Cancer Immunol. Immunother. 2011;60(1):61–73. doi: 10.1007/s00262-010-0919-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Reddy N, Hernandez-Ilizaliturri FJ, Deeb G, et al. Immunomodulatory drugs stimulate natural killer-cell function, alter cytokine production by dendritic cells, and inhibit angiogenesis enhancing the anti-tumour activity of rituximab in vivo. Br. J. Haematol. 2008;140(1):36–45. doi: 10.1111/j.1365-2141.2007.06841.x. [DOI] [PubMed] [Google Scholar]
  • 71.Hayashi T, Hideshima T, Akiyama M, et al. Molecular mechanisms whereby immunomodulatory drugs activate natural killer cells: clinical application. Br. J. Haematol. 2005;128(2):192–203. doi: 10.1111/j.1365-2141.2004.05286.x. [DOI] [PubMed] [Google Scholar]
  • 72.Berg SL, Cairo MS, Russell H, et al. Safety, pharmacokinetics, and immunomodulatory effects of lenalidomide in children and adolescents with relapsed/refractory solid tumors or myelodysplastic syndrome: a children’s oncology group Phase I consortium report. J. Clin. Oncol. 2011;29(3):316–323. doi: 10.1200/JCO.2010.30.8387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zinzani PL, Pellegrini C, Gandolfi L, et al. Combination of lenalidomide and rituximab in elderly patients with relapsed or refractory diffuse large B-cell lymphoma: a Phase 2 trial. Clin. Lymphoma Myeloma Leuk. 2011;11(6):462–466. doi: 10.1016/j.clml.2011.02.001. [DOI] [PubMed] [Google Scholar]
  • 74.Nowakowski GS, Laplant B, Habermann TM, et al. Lenalidomide can be safely combined with R-CHOP (R2CHOP) in the initial chemotherapy for aggressive B-cell lymphomas: Phase I study. Leukemia. 2011;25(12):1877–1881. doi: 10.1038/leu.2011.165. [DOI] [PubMed] [Google Scholar]
  • 75.Brown JR, Abramson J, Hochberg E, et al. A Phase I study of lenalidomide in combination with fludarabine and rituximab in previously untreated CLL/SLL. Leukemia. 2010;24(11):1972–1975. doi: 10.1038/leu.2010.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Fowler N, Hagemeister F, Mclaughlin P, et al. High response rates with lenalidomide plus rituximab for untreated indolent B cell non-Hodgkins lymphoma. Ann. Oncol. 2011;22(Suppl. 4):iv128–iv129. ▪▪ Among the highest clinical response rates seen in indolent lymphoma with a nonchemotherapeutic regimen.
  • 77.Witzig TE, Wiernik PH, Moore T, et al. Lenalidomide oral monotherapy produces durable responses in relapsed or refractory indolent non-Hodgkin’s lymphoma. J. Clin. Oncol. 2009;27(32):5404–5409. doi: 10.1200/JCO.2008.21.1169. [DOI] [PubMed] [Google Scholar]
  • 78.Hainsworth JD, Litchy S, Burris HA, 3rd, et al. Rituximab as first-line and maintenance therapy for patients with indolent non-Hodgkin’s lymphoma. J. Clin. Oncol. 2002;20(20):4261–4267. doi: 10.1200/JCO.2002.08.674. [DOI] [PubMed] [Google Scholar]
  • 79.Lanier LL. Up on the tightrope: natural killer cell activation and inhibition. Nat. Immunol. 2008;9(5):495–502. doi: 10.1038/ni1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Inagaki A, Ishida T, Yano H, et al. Expression of the ULBP ligands for NKG2D by B-NHL cells plays an important role in determining their susceptibility to rituximab-induced ADCC. Int. J. Cancer. 2009;125(1):212–221. doi: 10.1002/ijc.24351. [DOI] [PubMed] [Google Scholar]
  • 81.Diefenbach A, Jensen ER, Jamieson AM, Raulet DH. Rae1 and H60 ligands of the NKG2D receptor stimulate tumour immunity. Nature. 2001;413(6852):165–171. doi: 10.1038/35093109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Sutherland CL, Rabinovich B, Chalupny NJ, Brawand P, Miller R, Cosman D. ULBPs, human ligands of the NKG2D receptor, stimulate tumor immunity with enhancement by IL-15. Blood. 2006;108(4):1313–1319. doi: 10.1182/blood-2005-11-011320. [DOI] [PubMed] [Google Scholar]
  • 83.Zhang C, Niu J, Zhang J, Wang Y, Zhou Z, Tian Z. Opposing effects of interferon-α and interferon-γ on the expression of major histocompatibility complex class I chain-related A in tumors. Cancer Sci. 2008;99(6):1279–1286. doi: 10.1111/j.1349-7006.2008.00791.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Poggi A, Catellani S, Garuti A, Pierri I, Gobbi M, Zocchi MR. Effective in vivo induction of NKG2D ligands in acute myeloid leukaemias by all-trans-retinoic acid or sodium valproate. Leukemia. 2009;23(4):641–648. doi: 10.1038/leu.2008.354. [DOI] [PubMed] [Google Scholar]
  • 85.Lu X, Ohata K, Kondo Y, Espinoza JL, Qi Z, Nakao S. Hydroxyurea upregulates NKG2D ligand expression in myeloid leukemia cells synergistically with valproic acid and potentially enhances susceptibility of leukemic cells to natural killer cell-mediated cytolysis. Cancer Sci. 2010;101(3):609–615. doi: 10.1111/j.1349-7006.2009.01439.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Vales-Gomez M, Chisholm SE, Cassady-Cain RL, Roda-Navarro P, Reyburn HT. Selective induction of expression of a ligand for the NKG2D receptor by proteasome inhibitors. Cancer Res. 2008;68(5):1546–1554. doi: 10.1158/0008-5472.CAN-07-2973. [DOI] [PubMed] [Google Scholar]
  • 87.Kato N, Tanaka J, Sugita J, et al. Regulation of the expression of MHC class I-related chain A, B (MICA, MICB) via chromatin remodeling and its impact on the susceptibility of leukemic cells to the cytotoxicity of NKG2D-expressing cells. Leukemia. 2007;21(10):2103–2108. doi: 10.1038/sj.leu.2404862. [DOI] [PubMed] [Google Scholar]
  • 88.Diermayr S, Himmelreich H, Durovic B, et al. NKG2D ligand expression in AML increases in response to HDAC inhibitor valproic acid and contributes to allorecognition by NK-cell lines with single KIR-HLA class I specificities. Blood. 2008;111(3):1428–1436. doi: 10.1182/blood-2007-07-101311. [DOI] [PubMed] [Google Scholar]
  • 89.Shimizu R, Kikuchi J, Wada T, Ozawa K, Kano Y, Furukawa Y. HDAC inhibitors augment cytotoxic activity of rituximab by upregulating CD20 expression on lymphoma cells. Leukemia. 2010;24(10):1760–1768. doi: 10.1038/leu.2010.157. [DOI] [PubMed] [Google Scholar]
  • 90.Huang X, Wang S, Lee CK, Yang X, Liu B. HDAC inhibitor SNDX-275 enhances efficacy of trastuzumab in erbB2-overexpressing breast cancer cells and exhibits potential to overcome trastuzumab resistance. Cancer Lett. 2011;307(1):72–79. doi: 10.1016/j.canlet.2011.03.019. [DOI] [PubMed] [Google Scholar]
  • 91.Blum KA, Advani A, Fernandez L, et al. Phase II study of the histone deacetylase inhibitor MGCD0103 in patients with previously treated chronic lymphocytic leukaemia. Br. J. Haematol. 2009;147(4):507–514. doi: 10.1111/j.1365-2141.2009.07881.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Masucci G, Wersall P, Ragnhammar P, Mellstedt H. Granulocyte-monocyte-colonystimulating factor augments the cytotoxic capacity of lymphocytes and monocytes in antibody-dependent cellular cytotoxicity. Cancer Immunol. Immunother. 1989;29(4):288–292. doi: 10.1007/BF00199217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ragnhammar P, Frodin JE, Trotta PP, Mellstedt H. Cytotoxicity of white blood cells activated by granulocyte-colony-stimulating factor, granulocyte/macrophage-colony-stimulating factor and macrophage-colony-stimulating factor against tumor cells in the presence of various monoclonal antibodies. Cancer Immunol. Immunother. 1994;39(4):254–262. doi: 10.1007/BF01525989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Burgess AW, Begley CG, Johnson GR, et al. Purification and properties of bacterially synthesized human granulocyte-macrophage colony stimulating factor. Blood. 1987;69(1):43–51. [PubMed] [Google Scholar]
  • 95.Charak BS, Agah R, Mazumder A. Granulocyte-macrophage colony-stimulating factor-induced antibody-dependent cellular cytotoxicity in bone marrow macrophages: application in bone marrow transplantation. Blood. 1993;81(12):3474–3479. [PubMed] [Google Scholar]
  • 96.Venugopal P, Sivaraman S, Huang XK, Nayini J, Gregory SA, Preisler HD. Effects of cytokines on CD20 antigen expression on tumor cells from patients with chronic lymphocytic leukemia. Leuk. Res. 2000;24(5):411–415. doi: 10.1016/s0145-2126(99)00206-4. [DOI] [PubMed] [Google Scholar]
  • 97.Ragnhammar P, Masucci G, Frodin JE, Hjelm AL, Mellstedt H. Cytotoxic functions of blood mononuclear cells in patients with colorectal carcinoma treated with mAb 17-1A and granulocyte/macrophage-colony-stimulating factor. Cancer Immunol. Immunother. 1992;35(3):158–164. doi: 10.1007/BF01756182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Liljefors M, Nilsson B, Mellstedt H, Frodin JE. Influence of varying doses of granulocyte-macrophage colony-stimulating factor on pharmacokinetics and antibody-dependent cellular cytotoxicity. Cancer Immunol. Immunother. 2008;57(3):379–388. doi: 10.1007/s00262-007-0377-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Morales JK, Kmieciak M, Knutson KL, Bear HD, Manjili MH. GM-CSF is one of the main breast tumor-derived soluble factors involved in the differentiation of CD11b−Gr1− bone marrow progenitor cells into myeloid-derived suppressor cells. Breast Cancer Res. Treat. 2010;123(1):39–49. doi: 10.1007/s10549-009-0622-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Li H, Han Y, Guo Q, Zhang M, Cao X. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-β 1. J. Immunol. 2009;182(1):240–249. doi: 10.4049/jimmunol.182.1.240. [DOI] [PubMed] [Google Scholar]
  • 101.Biron CA, Sonnenfeld G, Welsh RM. Interferon induces natural killer cell blastogenesis in vivo. J. Leukoc. Biol. 1984;35(1):31–37. doi: 10.1002/jlb.35.1.31. [DOI] [PubMed] [Google Scholar]
  • 102.Nguyen KB, Salazar-Mather TP, Dalod MY, et al. Coordinated and distinct roles for IFN-α, β, IL-12, and IL-15 regulation of NK cell responses to viral infection. J. Immunol. 2002;169(8):4279–4287. doi: 10.4049/jimmunol.169.8.4279. [DOI] [PubMed] [Google Scholar]
  • 103.Masucci MG, Szigeti R, Klein E, et al. Effect of interferon-α 1 from E. coli on some cell functions. Science. 1980;209(4463):1431–1435. doi: 10.1126/science.6158096. [DOI] [PubMed] [Google Scholar]
  • 104.Yano H, Ishida T, Inagaki A, et al. Defucosylated anti CC chemokine receptor 4 monoclonal antibody combined with immunomodulatory cytokines: a novel immunotherapy for aggressive/refractory mycosis fungoides and sezary syndrome. Clin. Cancer Res. 2007;13(21):6494–6500. doi: 10.1158/1078-0432.CCR-07-1324. [DOI] [PubMed] [Google Scholar]
  • 105.Bungard S, Flieger D, Schweitzer S, Sauerbruch T, Spengler U. The combination of interleukin-2 and interferon α effectively augments the antibody-dependent cellular cytotoxicity of monoclonal antibodies 17-1A and BR55-2 against the colorectal carcinoma cell line HT29. Cancer Immunol. Immunother. 1998;46(4):213–220. doi: 10.1007/s002620050480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Mukhopadhyaya R, Tatake RJ, Krishnan N, et al. Immunoreactivity of lymphocytes from draining lymph nodes, peripheral blood and tumor infiltrates from oral cancer patients. J. Clin. Lab. Immunol. 1989;30(1):21–25. [PubMed] [Google Scholar]
  • 107.Fehr HG, Fan S, Lotzerich H, Appell HJ. Cytokine stimulation of antibody-dependent cellular cytotoxicity (ADCC) enhances cytolytic but not binding capacity of peritoneal macrophages. Anticancer Res. 1990;10(3):821–825. [PubMed] [Google Scholar]
  • 108.Eisenthal A, Cameron RB, Rosenberg SA. Induction of antibody-dependent cellular cytotoxicity in vivo by IFN-α and its antitumor efficacy against established B16 melanoma liver metastases when combined with specific anti-B16 monoclonal antibody. J. Immunol. 1990;144(11):4463–4471. [PubMed] [Google Scholar]
  • 109.Ozer H, Gavigan M, O’Malley J, et al. Immunomodulation by recombinant interferon-α 2 in a Phase I trial in patients with lymphoproliferative malignancies. J. Biol. Response Mod. 1983;2(6):499–515. [PubMed] [Google Scholar]
  • 110.Sivaraman S, Venugopal P, Ranganathan R, et al. Effect of interferon-α on CD20 antigen expression of B-cell chronic lymphocytic leukemia. Cytokines Cell. Mol. Ther. 2000;6(2):81–87. doi: 10.1080/13684730050515804. [DOI] [PubMed] [Google Scholar]
  • 111.Portlock CS, O’Connor OA, Straus DJ, et al. Pegylated interferon plus rituximab in advanced stage, indolent lymphoma: is there CD20 antigen upregulation? Leuk. Lymphoma. 2006;47(7):1260–1264. doi: 10.1080/10428190600565354. [DOI] [PubMed] [Google Scholar]
  • 112.Morgan DA, Ruscetti FW, Gallo R. Selective in vitro growth of T lymphocytes from normal human bone marrows. Science. 1976;193(4257):1007–1008. doi: 10.1126/science.181845. [DOI] [PubMed] [Google Scholar]
  • 113.Smith KA. Interleukin-2: inception, impact, and implications. Science. 1988;240(4856):1169–1176. doi: 10.1126/science.3131876. [DOI] [PubMed] [Google Scholar]
  • 114.Tsudo M, Goldman CK, Bongiovanni KF, et al. The p75 peptide is the receptor for interleukin 2 expressed on large granular lymphocytes and is responsible for the interleukin 2 activation of these cells. Proc. Natl Acad. Sci. USA. 1987;84(15):5394–5398. doi: 10.1073/pnas.84.15.5394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Caligiuri MA, Zmuidzinas A, Manley TJ, Levine H, Smith KA, Ritz J. Functional consequences of interleukin 2 receptor expression on resting human lymphocytes. Identification of a novel natural killer cell subset with high affinity receptors. J. Exp. Med. 1990;171(5):1509–1526. doi: 10.1084/jem.171.5.1509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Gluck WL, Hurst D, Yuen A, et al. Phase I studies of interleukin (IL)-2 and rituximab in B-cell non-Hodgkin’s lymphoma: IL-2 mediated natural killer cell expansion correlations with clinical response. Clin. Cancer Res. 2004;10(7):2253–2264. doi: 10.1158/1078-0432.ccr-1087-3. [DOI] [PubMed] [Google Scholar]
  • 117. Hooijberg E, Sein JJ, Van Den Berk PC, et al. Eradication of large human B cell tumors in nude mice with unconjugated CD20 monoclonal antibodies and interleukin 2. Cancer Res. 1995;55(12):2627–2634. ▪ Only randomized Phase III study showing an overall survival benefit of immunotherapy for neuroblastoma.
  • 118.Sondel PM, Hank JA. Combination therapy with interleukin-2 and antitumor monoclonal antibodies. Cancer J. Sci. Am. 1997;3(Suppl. 1):S121–S127. [PubMed] [Google Scholar]
  • 119.Yu AL, Gilman AL, Ozkaynak MF, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N. Engl. J. Med. 2010;363(14):1324–1334. doi: 10.1056/NEJMoa0911123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Colombo MP, Trinchieri G. Interleukin-12 in anti-tumor immunity and immunotherapy. Cytokine Growth Factor Rev. 2002;13(2):155–168. doi: 10.1016/s1359-6101(01)00032-6. [DOI] [PubMed] [Google Scholar]
  • 121.Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat. Rev. 2003;3(2):133–146. doi: 10.1038/nri1001. [DOI] [PubMed] [Google Scholar]
  • 122.Parihar R, Dierksheide J, Hu Y, Carson WE. IL-12 enhances the natural killer cell cytokine response to Ab-coated tumor cells. J. Clin. Invest. 2002;110(7):983–992. doi: 10.1172/JCI15950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Jaime-Ramirez AC, Mundy-Bosse BL, Kondadasula S, et al. IL-12 enhances the antitumor actions of trastuzumab via NK cell IFN-γ production. J. Immunol. 2011;186(6):3401–3409. doi: 10.4049/jimmunol.1000328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Jacobson NG, Szabo SJ, Weber-Nordt RM, et al. Interleukin 12 signaling in T helper type 1 (Th1) cells involves tyrosine phosphorylation of signal transducer and activator of transcription (Stat)3 and Stat4. J. Exp. Med. 1995;181(5):1755–1762. doi: 10.1084/jem.181.5.1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Nimmerjahn F, Ravetch JV. Divergent immunoglobulin g subclass activity through selective Fc receptor binding. Science. 2005;310(5753):1510–1512. doi: 10.1126/science.1118948. [DOI] [PubMed] [Google Scholar]
  • 126.Farag SS, Caligiuri MA. Human natural killer cell development and biology. Blood Rev. 2006;20(3):123–137. doi: 10.1016/j.blre.2005.10.001. [DOI] [PubMed] [Google Scholar]
  • 127.Mrozek E, Anderson P, Caligiuri MA. Role of interleukin-15 in the development of human CD56+ natural killer cells from CD34+ hematopoietic progenitor cells. Blood. 1996;87(7):2632–2640. [PubMed] [Google Scholar]
  • 128.Fehniger TA, Caligiuri MA. Interleukin 15: biology and relevance to human disease. Blood. 2001;97(1):14–32. doi: 10.1182/blood.v97.1.14. [DOI] [PubMed] [Google Scholar]
  • 129.Moga E, Canto E, Vidal S, Juarez C, Sierra J, Briones J. Interleukin-15 enhances rituximab-dependent cytotoxicity against chronic lymphocytic leukemia cells and overcomes transforming growth factor β-mediated immunosuppression. Exp. Hematol. 2011;39(11):1064–1071. doi: 10.1016/j.exphem.2011.08.006. [DOI] [PubMed] [Google Scholar]
  • 130.Roberti MP, Barrio MM, Bravo AI, et al. IL-15 and IL-2 increase cetuximab-mediated cellular cytotoxicity against triple negative breast cancer cell lines expressing EGFR. Breast Cancer Res. Treat. 2011;130(2):465–475. doi: 10.1007/s10549-011-1360-2. [DOI] [PubMed] [Google Scholar]
  • 131.Ben Ahmed M, Belhadj Hmida N, Moes N, et al. IL-15 renders conventional lymphocytes resistant to suppressive functions of regulatory T cells through activation of the phosphatidylinositol 3-kinase pathway. J. Immunol. 2009;182(11):6763–6770. doi: 10.4049/jimmunol.0801792. [DOI] [PubMed] [Google Scholar]
  • 132.Frederiksen KS, Lundsgaard D, Freeman JA, et al. IL-21 induces in vivo immune activation of NK cells and CD8(+) T cells in patients with metastatic melanoma and renal cell carcinoma. Cancer Immunol. Immunother. 2008;57(10):1439–1449. doi: 10.1007/s00262-008-0479-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Davis ID, Skak K, Smyth MJ, Kristjansen PE, Miller DM, Sivakumar PV. Interleukin-21 signaling: functions in cancer and autoimmunity. Clin. Cancer Res. 2007;13(23):6926–6932. doi: 10.1158/1078-0432.CCR-07-1238. [DOI] [PubMed] [Google Scholar]
  • 134.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(3):520–529. doi: 10.1038/sj.bjc.6605502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.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(21):6419–6428. doi: 10.1158/1078-0432.CCR-07-0865. [DOI] [PubMed] [Google Scholar]
  • 136.Eskelund CW, Nederby L, Thysen AH, Skovbo A, Roug AS, Hokland ME. Interleukin-21 and rituximab enhance NK cell functionality in patients with B-cell chronic lymphocytic leukaemia. Leuk. Res. 2011;35(7):914–920. doi: 10.1016/j.leukres.2011.02.006. [DOI] [PubMed] [Google Scholar]
  • 137.Smyth MJ, Teng MW, Sharkey J, et al. Interleukin 21 enhances antibody-mediated tumor rejection. Cancer Res. 2008;68(8):3019–3025. doi: 10.1158/0008-5472.CAN-07-6019. [DOI] [PubMed] [Google Scholar]
  • 138.Timmerman JM, Byrd JC, Andorsky DJ, et al. Efficacy and safety of recombinant interleukin-21 (rIL-21) and rituximab in relapsed/refractory indolent lymphoma. Presented at: ASCO 2008 Annual Meeting; Chicago, IL, USA. 30 May–3 June 2008; (Abstract 8554) [Google Scholar]
  • 139.Shusterman S, London WB, Gillies SD, et al. Antitumor activity of hu14.18–IL2 in patients with relapsed/refractory neuroblastoma: a Children’s Oncology Group (COG) Phase II study. J. Clin. Oncol. 2010;28(33):4969–4975. doi: 10.1200/JCO.2009.27.8861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Delgado DC, Hank JA, Kolesar J, et al. Genotypes of NK cell KIR receptors, their ligands, and Fcγ receptors in the response of neuroblastoma patients to Hu14.18–IL2 immunotherapy. Cancer Res. 2010;70(23):9554–9561. doi: 10.1158/0008-5472.CAN-10-2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Ribas A, Kirkwood JM, Atkins MB, et al. Phase I/II open-label study of the biologic effects of the interleukin-2 immunocytokine EMD 273063 (hu14.18–IL2) in patients with metastatic malignant melanoma. J. Transl Med. 2009;7:68. doi: 10.1186/1479-5876-7-68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Gillies SD, Lan Y, Williams S, et al. An anti-CD20-IL-2 immunocytokine is highly efficacious in a SCID mouse model of established human B lymphoma. Blood. 2005;105(10):3972–3978. doi: 10.1182/blood-2004-09-3533. [DOI] [PubMed] [Google Scholar]
  • 143.Singh H, Serrano LM, Pfeiffer T, et al. Combining adoptive cellular and immunocytokine therapies to improve treatment of B-lineage malignancy. Cancer Res. 2007;67(6):2872–2880. doi: 10.1158/0008-5472.CAN-06-2283. [DOI] [PubMed] [Google Scholar]
  • 144.Johannsen M, Spitaleri G, Curigliano G, et al. The tumour-targeting human L19-IL2 immunocytokine: preclinical safety studies, Phase I clinical trial in patients with solid tumours and expansion into patients with advanced renal cell carcinoma. Eur. J. Cancer. 2010;46(16):2926–2935. doi: 10.1016/j.ejca.2010.07.033. [DOI] [PubMed] [Google Scholar]
  • 145.Eigentler TK, Weide B, De Braud F, et al. A dose-escalation and signal-generating study of the immunocytokine L19-IL2 in combination with dacarbazine for the therapy of patients with metastatic melanoma. Clin. Cancer Res. 2011;17(24):7732–7742. doi: 10.1158/1078-0432.CCR-11-1203. [DOI] [PubMed] [Google Scholar]
  • 146.De Braud FG, Catania C, Onofri A, et al. Combination of the immunocytokine F16–IL2 with doxorubicin or paclitaxel in patients with solid tumors: results from two Phase Ib trials. J. Clin. Oncol. 2011;28(Suppl. 15) e13017. [Google Scholar]
  • 147.Davis CB, Gillies SD. Immunocytokines: amplification of anti-cancer immunity. Cancer Immunol. Immunother. 2003;52(5):297–308. doi: 10.1007/s00262-002-0349-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Rossi EA, Goldenberg DM, Cardillo TM, Stein R, Chang CH. CD20-targeted tetrameric interferon-α, a novel and potent immunocytokine for the therapy of B-cell lymphomas. Blood. 2009;114(18):3864–3871. doi: 10.1182/blood-2009-06-228890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Sharma S, Dominguez AL, Manrique SZ, Cavallo F, Sakaguchi S, Lustgarten J. Systemic targeting of CpG-ODN to the tumor microenvironment with anti-neu-CpG hybrid molecule and T regulatory cell depletion induces memory responses in BALB-neuT tolerant mice. Cancer Res. 2008;68(18):7530–7540. doi: 10.1158/0008-5472.CAN-08-1635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Lin W, Voskens CJ, Zhang X, et al. Fc-dependent expression of CD137 on human NK cells: insights into ‘agonistic’ effects of anti-CD137 monoclonal antibodies. Blood. 2008;112(3):699–707. doi: 10.1182/blood-2007-11-122465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Melero I, Johnston JV, Shufford WW, Mittler RS, Chen L. NK1.1 cells express 4-1BB (CDw137) costimulatory molecule and are required for tumor immunity elicited by anti-4-1BB monoclonal antibodies. Cell. Immunol. 1998;190(2):167–172. doi: 10.1006/cimm.1998.1396. [DOI] [PubMed] [Google Scholar]
  • 152.Wilcox RA, Tamada K, Strome SE, Chen L. Signaling through NK cell-associated CD137 promotes both helper function for CD8+ cytolytic T cells and responsiveness to IL-2 but not cytolytic activity. J. Immunol. 2002;169(8):4230–4236. doi: 10.4049/jimmunol.169.8.4230. [DOI] [PubMed] [Google Scholar]
  • 153.Stagg J, Loi S, Divisekera U, et al. Anti-ErbB-2 mAb therapy requires type I and II interferons and synergizes with anti-PD-1 or anti-CD137 mAb therapy. Proc. Natl Acad. Sci. USA. 2011;108(17):7142–7147. doi: 10.1073/pnas.1016569108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Meseck M, Huang T, Ma G, Wang G, Chen SH, Woo SL. A functional recombinant human 4-1BB ligand for immune costimulatory therapy of cancer. J. Immunother. 2011;34(2):175–182. doi: 10.1097/CJI.0b013e318206dac1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Croft M. Control of immunity by the TNFR-related molecule OX40 (CD134) Annu. Rev. Immunol. 2010;28:57–78. doi: 10.1146/annurev-immunol-030409-101243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Ogasawara K, Yoshinaga SK, Lanier LL. Inducible costimulator costimulates cytotoxic activity and IFN-γ production in activated murine NK cells. J. Immunol. 2002;169(7):3676–3685. doi: 10.4049/jimmunol.169.7.3676. [DOI] [PubMed] [Google Scholar]
  • 157.Lanier LL, Corliss B, Phillips JH. Arousal and inhibition of human NK cells. Immunol. Rev. 1997;155:145–154. doi: 10.1111/j.1600-065x.1997.tb00947.x. [DOI] [PubMed] [Google Scholar]
  • 158.Funakoshi S, Longo DL, Murphy WJ. Differential in vitro and in vivo antitumor effects mediated by anti-CD40 and anti-CD20 monoclonal antibodies against human B-cell lymphomas. J. Immunother. Emphasis Tumor Immunol. 1996;19(2):93–101. doi: 10.1097/00002371-199603000-00002. [DOI] [PubMed] [Google Scholar]
  • 159.Gennari R, Menard S, Fagnoni F, et al. Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2. Clin. Cancer Res. 2004;10(17):5650–5655. doi: 10.1158/1078-0432.CCR-04-0225. [DOI] [PubMed] [Google Scholar]
  • 160.Marechal R, De Schutter J, Nagy N, et al. Putative contribution of CD56 positive cells in cetuximab treatment efficacy in first-line metastatic colorectal cancer patients. BMC Cancer. 2010;10:340. doi: 10.1186/1471-2407-10-340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Borgerding A, Hasenkamp J, Engelke M, et al. B-lymphoma cells escape rituximab-triggered elimination by NK cells through increased HLA class I expression. Exp. Hematol. 2010;38(3):213–221. doi: 10.1016/j.exphem.2009.12.007. [DOI] [PubMed] [Google Scholar]
  • 162.Binstadt BA, Brumbaugh KM, Dick CJ, et al. Sequential involvement of Lck and SHP-1 with MHC-recognizing receptors on NK cells inhibits FcR-initiated tyrosine kinase activation. Immunity. 1996;5(6):629–638. doi: 10.1016/s1074-7613(00)80276-9. [DOI] [PubMed] [Google Scholar]
  • 163.Leibson PJ. Signal transduction during natural killer cell activation: inside the mind of a killer. Immunity. 1997;6(6):655–661. doi: 10.1016/s1074-7613(00)80441-0. [DOI] [PubMed] [Google Scholar]
  • 164.Koh CY, Blazar BR, George T, et al. Augmentation of antitumor effects by NK cell inhibitory receptor blockade in vitro and in vivo. Blood. 2001;97(10):3132–3137. doi: 10.1182/blood.v97.10.3132. [DOI] [PubMed] [Google Scholar]
  • 165.Romagne F, Andre P, Spee P, et al. Preclinical characterization of 1-7F9, a novel human anti-KIR receptor therapeutic antibody that augments natural killer-mediated killing of tumor cells. Blood. 2009;114(13):2667–2677. doi: 10.1182/blood-2009-02-206532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Srivastava S. Enhancing natural killer (NK) cell mediated killing of non-Hodgkin’s lymphoma. Blood. 2009;114 Abstract 2706. [Google Scholar]
  • 167.Binyamin L, Alpaugh RK, Hughes TL, Lutz CT, Campbell KS, Weiner LM. Blocking NK cell inhibitory self-recognition promotes antibody-dependent cellular cytotoxicity in a model of anti-lymphoma therapy. J. Immunol. 2008;180(9):6392–6401. doi: 10.4049/jimmunol.180.9.6392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Nijhof IS, De Weers M, Andre P, et al. Synergistic action of the human inhibitory KIR antibody IPH2102, and the human CD38 antibody daratumumab to enhance the lysis of primary multiple myeloma (MM) cells in the bone marrow mononuclear cells (MNCs) from myeloma patients. Presented at: 53rd ASH Annual Meeting and Exposition; San Diego, CA, USA. 10–13 December 2011; (Abstract 1865) [Google Scholar]
  • 169.Buechele C, Baessler T, Wirths S, Schmohl JU, Schmiedel BJ, Salih HR. Glucocorticoid-induced TNFR-related protein (GITR) ligand modulates cytokine release and NK cell reactivity in chronic lymphocytic leukemia (CLL) Leukemia. 2011 doi: 10.1038/leu.2011.313. (Epub ahead of print) [DOI] [PubMed] [Google Scholar]
  • 170.Baltz KM, Krusch M, Bringmann A, et al. Cancer immunoediting by GITR (glucocorticoid-induced TNF-related protein) ligand in humans: NK cell/tumor cell interactions. FASEB J. 2007;21(10):2442–2454. doi: 10.1096/fj.06-7724com. [DOI] [PubMed] [Google Scholar]
  • 171.Rosenzweig M, Ponte J, Apostolou I, et al. Development of TRX518, an aglycosyl humanized monoclonal antibody (Mab) agonist of huGITR. Presented at: ASCO 2010 Annual Meeting; Chicago, IL, USA. 4–8 June 2010; (Abstract e13028) [Google Scholar]
  • 172.Brown EJ, Frazier WA. Integrin-associated protein (CD47) and its ligands. Trends Cell Biol. 2001;11(3):130–135. doi: 10.1016/s0962-8924(00)01906-1. [DOI] [PubMed] [Google Scholar]
  • 173.Manches O, Lui G, Chaperot L, et al. in vitro mechanisms of action of rituximab on primary non-Hodgkin lymphomas. Blood. 2003;101(3):949–954. doi: 10.1182/blood-2002-02-0469. [DOI] [PubMed] [Google Scholar]
  • 174.Uchida J, Hamaguchi Y, Oliver JA, et al. The innate mononuclear phagocyte network depletes B lymphocytes through Fc receptor-dependent mechanisms during anti-CD20 antibody immunotherapy. J. Exp. Med. 2004;199(12):1659–1669. doi: 10.1084/jem.20040119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Tamura K, Shimizu C, Hojo T, et al. Fc{γ} R2A and 3A polymorphisms predict clinical outcome of trastuzumab in both neoadjuvant and metastatic settings in patients with HER2-positive breast cancer. Ann. Oncol. 2011;22(6):1302–1307. doi: 10.1093/annonc/mdq585. [DOI] [PubMed] [Google Scholar]
  • 176.Jaiswal S, Chao MP, Majeti R, Weissman IL. Macrophages as mediators of tumor immunosurveillance. Trends Immunol. 2010;31(6):212–219. doi: 10.1016/j.it.2010.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Chao MP, Alizadeh AA, Tang C, et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell. 2010;142(5):699–713. doi: 10.1016/j.cell.2010.07.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Ysebaert L, Laprevotte E, Klein C, et al. ‘Don’t eat me’: CD47 and LFA-1 modulate CLL response to nurselike cells-mediated antibody-dependent phagocytosis induced by rituximab and obinutuzumab (GA101). Presented at: 53rd ASH Annual Meeting and Exposition; San Diego, CA, USA. 10–13 December 2011; (Abstract 1762). [Google Scholar]
  • 179.Benson DM, Bakan CE, Mishra A, et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood. 2010;116(13):2286–2294. doi: 10.1182/blood-2010-02-271874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Hodi FS, O’Day SJ, Mcdermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010;363(8):711–723. doi: 10.1056/NEJMoa1003466. ▪ One of two randomized Phase III studies showing an overall survival benefit of immunotherapy for metastatic melanoma and a paradigm shift proving the principle of regulatory blockade allowing for a clinically meaningful antitumor effect.
  • 181. Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 2011;364(26):2517–2526. doi: 10.1056/NEJMoa1104621. ▪▪ One of two randomized Phase III studies showing overall survival benefit of immunotherapy for metastatic melanoma and a paradigm shift proving the principle of regulatory blockade allowing for a clinically meaningful antitumor effect.
  • 182.Sacchi S, Federico M, Vitolo U, et al. Clinical activity and safety of combination immunotherapy with IFN-α 2a and rituximab in patients with relapsed low grade non-Hodgkin’s lymphoma. Haematologica. 2001;86(9):951–958. [PubMed] [Google Scholar]
  • 183.Davis TA, Maloney DG, Grillo-Lopez AJ, et al. Combination immunotherapy of relapsed or refractory low-grade or follicular non-Hodgkin’s lymphoma with rituximab and interferon-α-2a. Clin. Cancer Res. 2000;6(7):2644–2652. [PubMed] [Google Scholar]
  • 184.Kimby E, Jurlander J, Geisler C, et al. Long-term molecular remissions in patients with indolent lymphoma treated with rituximab as a single agent or in combination with interferon α-2a: a randomized Phase II study from the Nordic lymphoma group. Leuk. Lymphoma. 2008;49(1):102–112. doi: 10.1080/10428190701704647. [DOI] [PubMed] [Google Scholar]
  • 185.Cartron G, Zhao-Yang L, Baudard M, et al. Granulocyte-macrophage colony-stimulating factor potentiates rituximab in patients with relapsed follicular lymphoma: results of a Phase II study. J. Clin. Oncol. 2008;26(16):2725–2731. doi: 10.1200/JCO.2007.13.7729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Karmali R, Larson ML, Wooldridge JE, et al. Granulocyte-macrophage colony stimulating factor-induced immune priming of cyclophosphamide, doxorubicin, vincristine, and prednisone with rituximab chemoimmunotherapy in previously untreated patients with diffuse large B-cell lymphoma and mantle cell lymphoma. Leuk. Lymphoma. 2011;52(11):2097–2104. doi: 10.3109/10428194.2011.589549. [DOI] [PubMed] [Google Scholar]
  • 187.Chang JE, Seo S, Kim KM, et al. Rituximab and CHOP chemotherapy plus GM-CSF for previously untreated diffuse large B-cell lymphoma in the elderly: a Wisconsin oncology network study. Clin. Lymphoma Myeloma Leuk. 2010;10(5):379–384. doi: 10.3816/CLML.2010.n.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Olivieri A, Lucesole M, Capelli D, et al. A new schedule of CHOP/rituximab plus granulocyte-macrophage colony-stimulating factor is an effective rescue for patients with aggressive lymphoma failing autologous stem cell transplantation. Biol. Blood Marrow Transplant. 2005;11(8):627–636. doi: 10.1016/j.bbmt.2005.05.002. [DOI] [PubMed] [Google Scholar]
  • 189.Niitsu N, Hayama M, Okamoto M, et al. Phase I study of rituximab–CHOP regimen in combination with granulocyte colony-stimulating factor in patients with follicular lymphoma. Clin. Cancer Res. 2004;10(12 Pt 1):4077–4082. doi: 10.1158/1078-0432.CCR-03-0658. [DOI] [PubMed] [Google Scholar]
  • 190.Cheng YC, Valero V, Davis ML, et al. Addition of GM-CSF to trastuzumab stabilises disease in trastuzumab-resistant HER2+ metastatic breast cancer patients. Br. J. Cancer. 2010;103(9):1331–1334. doi: 10.1038/sj.bjc.6605918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Ragnhammar P, Fagerberg J, Frodin JE, et al. Effect of monoclonal antibody 17-1A and GM-CSF in patients with advanced colorectal carcinoma – long-lasting, complete remissions can be induced. Int. J. Cancer. 1993;53(5):751–758. doi: 10.1002/ijc.2910530508. [DOI] [PubMed] [Google Scholar]
  • 192.Liljefors M, Nilsson B, Fagerberg J, Ragnhammar P, Mellstedt H, Frodin JE. Clinical effects of a chimeric anti-EpCAM monoclonal antibody in combination with granulocyte-macrophage colony-stimulating factor in patients with metastatic colorectal carcinoma. Int. J. Oncol. 2005;26(6):1581–1589. [PubMed] [Google Scholar]
  • 193.Kushner BH, Kramer K, Cheung NK. Phase II trial of the anti-G(D2) monoclonal antibody 3F8 and granulocyte-macrophage colony-stimulating factor for neuroblastoma. J. Clin. Oncol. 2001;19(22):4189–4194. doi: 10.1200/JCO.2001.19.22.4189. [DOI] [PubMed] [Google Scholar]
  • 194.Liljefors M, Ragnhammar P, Nilsson B, Ullenhag G, Mellstedt H, Frodin JE. Anti-EpCAM monoclonal antibody (MAb17-1A) based treatment combined with α-interferon, 5-fluorouracil and granulocyte-macrophage colony-stimulating factor in patients with metastatic colorectal carcinoma. Int. J. Oncol. 2004;25(3):703–711. [PubMed] [Google Scholar]
  • 195.van der Kolk LE, Grillo-Lopez AJ, Baars JW, van Oers MH. Treatment of relapsed B-cell non-Hodgkin’s lymphoma with a combination of chimeric anti-CD20 monoclonal antibodies (rituximab) and G-CSF: final report on safety and efficacy. Leukemia. 2003;17(8):1658–1664. doi: 10.1038/sj.leu.2402995. [DOI] [PubMed] [Google Scholar]
  • 196.Khan KD, Emmanouilides C, Benson DM, et al. A Phase 2 study of rituximab in combination with recombinant interleukin-2 for rituximab-refractory indolent non-Hodgkin’s lymphoma. Clin. Cancer Res. 2006;12(23):7046–7053. doi: 10.1158/1078-0432.CCR-06-1571. [DOI] [PubMed] [Google Scholar]
  • 197.Eisenbeis CF, Grainger A, Fischer B, et al. Combination immunotherapy of B-cell non-Hodgkin’s lymphoma with rituximab and interleukin-2: a preclinical and Phase I study. Clin. Cancer Res. 2004;10(18 Pt 1):6101–6110. doi: 10.1158/1078-0432.CCR-04-0525. [DOI] [PubMed] [Google Scholar]
  • 198.Repka T, Chiorean EG, Gay J, et al. Trastuzumab and interleukin-2 in HER2-positive metastatic breast cancer: a pilot study. Clin. Cancer Res. 2003;9(7):2440–2446. [PubMed] [Google Scholar]
  • 199.Fleming GF, Meropol NJ, Rosner GL, et al. A Phase I trial of escalating doses of trastuzumab combined with daily subcutaneous interleukin 2: report of Cancer and Leukemia Group B 9661. Clin. Cancer Res. 2002;8(12):3718–3727. [PubMed] [Google Scholar]
  • 200.Mani A, Roda J, Young D, et al. A Phase II trial of trastuzumab in combination with low-dose interleukin-2 (IL-2) in patients (PTS) with metastatic breast cancer (MBC) who have previously failed trastuzumab. Breast Cancer Res. Treat. 2009;117(1):83–89. doi: 10.1007/s10549-008-0251-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Frost JD, Hank JA, Reaman GH, et al. A Phase I/Ib trial of murine monoclonal anti-GD2 antibody 14.G2a plus interleukin-2 in children with refractory neuroblastoma: a report of the children’s cancer group. Cancer. 1997;80(2):317–333. doi: 10.1002/(sici)1097-0142(19970715)80:2<317::aid-cncr21>3.0.co;2-w. [DOI] [PubMed] [Google Scholar]
  • 202.Albertini MR, Hank JA, Schiller JH, et al. Phase Ib trial of chimeric antidisialoganglioside antibody plus interleukin 2 for melanoma patients. Clin. Cancer Res. 1997;3(8):1277–1288. [PubMed] [Google Scholar]
  • 203.Ansell SM, Geyer SM, Maurer MJ, et al. Randomized Phase II study of interleukin-12 in combination with rituximab in previously treated non-Hodgkin’s lymphoma patients. Clin. Cancer Res. 2006;12(20 Pt 1):6056–6063. doi: 10.1158/1078-0432.CCR-06-1245. [DOI] [PubMed] [Google Scholar]
  • 204.Parihar R, Nadella P, Lewis A, et al. A Phase I study of interleukin 12 with trastuzumab in patients with human epidermal growth factor receptor-2-overexpressing malignancies: analysis of sustained interferon gamma production in a subset of patients. Clin. Cancer Res. 2004;10(15):5027–5037. doi: 10.1158/1078-0432.CCR-04-0265. [DOI] [PubMed] [Google Scholar]
  • 205.Bekaii-Saab TS, Roda JM, Guenterberg KD, et al. A Phase I trial of paclitaxel and trastuzumab in combination with interleukin-12 in patients with HER2/neu-expressing malignancies. Mol. Cancer Ther. 2009;8(11):2983–2991. doi: 10.1158/1535-7163.MCT-09-0820. [DOI] [PMC free article] [PubMed] [Google Scholar]

References

RESOURCES