Skip to main content
Journal of Interferon & Cytokine Research logoLink to Journal of Interferon & Cytokine Research
. 2018 Feb 1;38(2):45–68. doi: 10.1089/jir.2017.0101

IL-2 and Beyond in Cancer Immunotherapy

John M Wrangle 1, Alicia Patterson 2, C Bryce Johnson 2, Daniel J Neitzke 2, Shikhar Mehrotra 2, Chadrick E Denlinger 2, Chrystal M Paulos 3, Zihai Li 3, David J Cole 2, Mark P Rubinstein 2,,3,
PMCID: PMC5815463  PMID: 29443657

Abstract

The development of the T- and natural killer (NK) cell growth factor IL-2 has been a sentinel force ushering in the era of immunotherapy in cancer. With the advent of clinical grade recombinant IL-2 in the mid-1980s, oncologists could for the first time directly manipulate lymphocyte populations with systemic therapy. By itself, recombinant IL-2 can induce clinical responses in up to 15% of patients with metastatic cancer or renal cell carcinoma. When administered with adoptively transferred tumor-reactive lymphocytes, IL-2 promotes T cell engraftment and response rates of up to 50% in metastatic melanoma patients. Importantly, these IL-2-driven responses can yield complete and durable responses in a subset of patients. However, the use of IL-2 is limited by toxicity and concern of the expansion of T regulatory cells. To overcome these limitations and improve response rates, other T cell growth factors, including IL-15 and modified forms of IL-2, are in clinical development. Administering T cell growth factors in combination with other agents, such as immune checkpoint pathway inhibitors, may also improve efficacy. In this study, we review the development of T- and NK cell growth factors and highlight current combinatorial approaches based on these reagents.

Keywords: : IL-2, IL-15, adoptive cellular therapy, T cells

IL-2: From Supernatant to Clinical Grade Therapy

The identification of IL-2 as a therapeutic agent began unwittingly in the mid-1960s when it was discovered that supernatants of antigen- or mitogen- activated leukocyte cultures contained a factor able to stimulate lymphocyte division (Gordon and MacLean 1965; Kasakura and Lowenstein 1965). By 1969, the term “lymphokine” had been coined (Dumonde and others 1969), and much research was dedicated to understanding the soluble factor or factors responsible (Dumonde and others 1969; Pick and Turk 1972; Chen and Di Sabato 1976). In 1976, Gallo and colleagues demonstrated that conditioned media from human lymphocytes contained this factor and could be used to maintain T cell cultures for over 9 months without the need for repetitive antigenic stimulation (Morgan and others 1976). This technique was quickly adapted to the culture of tumor-reactive T cells with sustained cytotoxic potential (Gillis and Smith 1977). These early studies led to the development of methods for enriching, purifying, and measuring this soluble factor (Farrar and others 1978; Gillis and others 1978; Shaw and others 1978; Watson and others 1979; Gillis and Watson 1980; Mier and Gallo 1980; Robb and others 1981; Stadler and others 1982), which would be named IL-2. Collectively, these advances allowed scientists to study this lymphocyte growth factor in greater depth.

In mice, both the persistence and antitumor efficacy of lymphocytes were greatly augmented upon injection of purified IL-2 (Cheever and others 1982, 1984; Donohue and others 1984). Furthermore, simply culturing lymphocytes in vitro with IL-2 could lead to the acquisition of ability to preferentially lyse tumor cells over healthy cells (Lotze and others 1981; Grimm and others 1982; Rayner and others 1985b). The effector cells mediating this tumor cytotoxicity were called lymphokine activated killer (LAK) cells and showed antitumor efficacy in preclinical models (Mazumder and Rosenberg 1984). These successes led to the evaluation of purified IL-2 in cancer and HIV-infected patients (Bindon and others 1983; Lotze and others 1984; Rayner and others 1985a). Although there was some evidence of biological activity, including toxicities, there were no clinical responses in the small number of patients treated.

In what was a critical milestone, the sequencing of the human IL-2 gene was reported in 1983 (Taniguchi and others 1983) and the murine IL-2 gene shortly thereafter (Kashima and others 1985). The cloning of IL-2 allowed the production of large quantities of purified recombinant IL-2 using Escherichia coli (Devos and others 1983; Taniguchi and others 1983; Lotze and others 1984; Wang and others 1984). Rosenberg and colleagues demonstrated that administration of recombinant IL-2 to mice mediated potent antitumor activity with regression of established pulmonary metastases and subcutaneous tumors (Lafreniere and Rosenberg 1985). In an initial clinical study reported in 1985, 20 patients with a variety of malignancies were treated with recombinant IL-2. This treatment resulted in the expansion of lymphoid populations but no clinical responses (Lotze and others 1985). An alternate clinical approach was suggested by experiments in mice showing that combining adoptive transfer of LAK cells with recombinant IL-2 was much more effective against tumor than either agent alone (Mule and others 1984, 1985, 1986; Lafreniere and Rosenberg 1985). While LAK cells had been evaluated clinically (Lotze and others 1980), these cells had never been coadministered to patients with recombinant IL-2. In the first human experience of LAK cells and recombinant IL-2 in patients with advanced cancer, 11 of 25 patients experienced objective responses defined as at least a 50% reduction in tumor volume and this included patients with metastatic melanoma, renal cell carcinoma, colon cancer, and lung adenocarcinoma (Rosenberg and others 1985). Among the responders was a patient with metastatic melanoma who experienced a complete response and has been disease free for 29 years (Rosenberg 2014). The conclusion that adding LAK cells improved IL-2 therapy was however complicated by the fact that a higher dose of IL-2 was used, as well as differences in the patient population. Therefore, in a subsequent study, Rosenberg and colleagues evaluated whether higher doses of IL-2 alone could be effective. In a small study of 10 patients, higher doses of IL-2 mediated clinical responses, including in 3 of 6 treated patients with metastatic melanoma (Lotze and others 1986a). These studies demonstrated for the first time that IL-2 administered as a single agent mediated antitumor efficacy in human patients with metastatic cancer.

An important remaining question was whether adoptively transferring LAK cells in addition to IL-2 therapy could improve efficacy. Therefore, Rosenberg and colleagues compared the administration of high-dose IL-2 alone versus high-dose IL-2 and LAK cells in metastatic melanoma and renal cell carcinoma patients. In a clinical trial with 181 patients randomized to two groups, 16 of 91 patients (18%) with IL-2 alone had objective responses, while 24 of 90 patients (24%) with IL-2 and LAK cells had objective responses (Rosenberg and others 1993). There was not a statistically significant difference in overall survival between patients receiving IL-2 versus IL-2 and LAK cells. However, there was a trend toward improved overall survival in the subset of metastatic melanoma patients that received IL-2 and LAK cells versus IL-2 alone. These results did not justify the addition of LAK cells to IL-2 therapy, particularly as the LAK cells could not be given as an off-the-shelf reagent. Subsequently, 2 other significant trials evaluated the efficacy of IL-2 alone. In 1 trial published in 1995, of 255 patients with renal cell carcinoma, roughly 15% of patients achieved objective responses with about one third of these being complete responses (Fyfe and others 1995). Similar response rates were reported in 1999 in a trial of over 270 patients with metastatic melanoma (Atkins and others 1999). Compared with other therapies available at the time, responses obtained with recombinant IL-2 were remarkable in their durability, with some patients achieving complete responses ongoing after 10 years (Rosenberg 2014). Notably, IL-2 administration in patients is associated with increased frequencies and activation of lymphocytes within the tumor (Cohen and others 1987; Swisher and others 1991). The Food and Drug Administration (FDA) approved IL-2 for the treatment of renal cell carcinoma in 1992 and metastatic melanoma in 1998 (Rosenberg 2014).

Receptors in the IL-2 Cytokine Family

IL-2 acts on lymphocytes by binding to the multimeric IL-2 receptor (IL-2R) and thereby engaging several intracellular signaling pathways that modulate lymphocyte survival, proliferation, and function (Smith 1988; Theze and others 1996; Nelson and Willerford 1998; Fehniger and others 2002; Kovanen and Leonard 2004; Ma and others 2006; Waldmann 2006, 2014, 2015; Boyman and others 2007; Bodnar and others 2008; D'Cruz and others 2009; Overwijk and Schluns 2009; Rochman and others 2009; Boyman and Sprent 2012; Carrette and Surh 2012; Liao and others 2013; Rosenberg 2014; Sim and Radvanyi 2014; Pulliam and others 2016). The heterotrimeric IL-2R is composed of 3 subunits: IL-2Rα (CD25), IL-2Rβ (CD122), and IL-2Rγ (CD132). IL-2Rβ and IL-2Rγ are essential for intracellular signaling and can form a functional dimeric receptor in the absence of IL-2Rα. Notably, IL-2Rγ is used as part of the receptors for IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, cytokines which can all act on various lymphocyte populations likely partially dictated by differing receptor subunit expression. IL-2, IL-7, and IL-15 have received the most attention for their ability to act on T cells, and in the case of IL-2 and IL-15, also on natural killer (NK) and NK T cells. IL-2 and IL-15 are closely related in that they both signal through the IL-2Rβγ heterodimeric receptor and engage the JAK/STAT, Ras/MAPK, and PI3K/Akt signaling pathways (Nelson and Willerford 1998; Bodnar and others 2008; Boyman and Sprent 2012; Liao and others 2013; Mishra and others 2014; Waldmann 2014). While not necessary for signaling, IL-2 and IL-15 also bind private α chains (IL-2Rα and IL-15Rα), which are structurally related and may have arisen evolutionarily from gene duplication (Giri and others 1995; Tagaya and others 1996). These α chains were initially thought to allow for high affinity receptor binding; however, as will be described below, it is clear that their functional contribution to cytokine signaling is more complicated.

While IL-2 and IL-15 both signal through the shared IL-2Rβγ and mediate similar signaling and functional activity on purified lymphocytes (Willerford and others 1995; Murakami and others 2002; Ring and others 2012; Arneja and others 2014), these cytokines have dramatically different biological activities in vivo. This is most apparent in a comparison of knockout mice. IL-2 knockout mice die prematurely as a result of autoimmune disease (Sadlack and others 1993), while IL-15 mice are relatively healthy with reduced numbers of IL-15-dependent cells such as NK cells and CD8 memory T cells (Kennedy and others 2000). These and other differential properties of IL-2 and IL-15 are likely mediated by IL-2 or IL-15 availability and expression of IL-2Rα and IL-15Rα. In particular, T regulatory cells express high and constitutive levels of IL-2Rα, a molecule critical for their survival. Thus, in the absence of IL-2Rα, mature T regulatory cells are absent and mice die prematurely unless IL-2Rα-competent T regulatory cells are provided (Willerford and others 1995; Almeida and others 2002). A patient with a mutated IL-2Rα gene, who had extensive lymphocytic infiltration of tissues accompanied by atrophy and inflammation, suggests that a similar process exists in humans (Sharfe and others 1997).

How IL-2Rα mediates its unique responsiveness to IL-2 has been the subject of much work. IL-2Rα is not known to mediate intracellular signaling and was initially characterized by its ability to promote high affinity binding of IL-2 to the IL-2Rβγ (Smith 1988; Nelson and Willerford 1998; Kovanen and Leonard 2004; Ma and others 2006). Interestingly, IL-2Rα can also promote sustained IL-2 signaling after removal of cytokine, which may be mediated by the ability of IL-2Rα to facilitate cell surface reservoirs of IL-2 and the ability of IL-2Rα to rescue IL-2 from degradation (Bergmann and others 1992; Fallon and others 2000; Rao and others 2004, 2005; Su and others 2015). Thus, in addition to facilitating high affinity binding of IL-2 to its receptor, IL-2Rα allows for temporal responsiveness of IL-2 (Su and others 2015). As activated T cells transiently express high levels of IL-2Rα (Cantrell and Smith 1983; Leonard and others 1985; Gullberg and Smith 1986; Andersson and others 1994; Cousens and others 1995; Obar and others 2010), these IL-2Rα-dependent mechanisms may facilitate the ability of effector T cells to function after being deprived of an IL-2-rich environment or for effector T cells to preserve access to limited amounts of IL-2. In addition to cell-intrinsic IL-2Rα signaling, it has been reported that IL-2Rα may be involved in the trans-presentation of IL-2 to neighboring cells (Eicher and Waldmann 1998; Kronin and others 1998; Fukao and Koyasu 2000; Wuest and others 2011). Soluble IL-2Rα may also play an important role in regulating immune responses (Rubin and others 1985; Baran and others 1988; Gooding and others 1995; Cabrera and others 2010; Hannani and others 2015; Li and others 2016).

Like IL-2Rα, IL-15Rα is unique to its cognate cytokine. IL-15Rα was initially recognized by its ability to facilitate high affinity IL-15 binding (Lodolce and others 1998; Fehniger and Caligiuri 2001; Waldmann and others 2001). However, recent work suggests that the ability of IL-15Rα to present IL-15 either in membrane or soluble form may be more critical (Dubois and others 2002; Burkett and others 2003; Koka and others 2004; Kobayashi and others 2005; Schluns and others 2005; Stonier and others 2008; Castillo and others 2009; Bergamaschi and others 2012). It is unclear whether IL-15 actually exists naturally by itself; rather it may always be associated with either membrane-bound or soluble IL-15Rα (Bergamaschi and others 2012; Castillo and Schluns 2012; Cole and Rubinstein 2012). There are some other notable findings related to IL-15Rα that may be relevant for its ability to function. There is evidence that IL-15Rα might engage membrane-bound IL-15 and mediate reverse signaling (Neely and others 2004; Khawam and others 2009). Unlike IL-2Rα subunit, IL-15Rα may retain some ability to mediate intracellular signaling upon IL-15 engagement, although there is not a consensus on the relevant pathways (Stevens and others 1997; Pereno and others 1999, 2000; Ratthe and Girard 2004; Marra and others 2014). Finally, naturally occurring soluble IL-15Rα has been reported which may impact the activity of IL-15 (Mortier and others 2004; Badoual and others 2008; Bergamaschi and others 2012).

IL-2 Combined with Adoptive Cellular Therapy

While IL-2 has efficacy as a single agent, preclinical work suggesting that coadministration of LAK cells could yield improved responses led to the search for other tumor-reactive lymphocyte populations to combine with IL-2. Yron and others (1980) reported the isolation and expansion of T cells from the tumors of mice using conditioned media containing IL-2. These T cells could kill tumor cells, but did not kill healthy lymphocytes. With the availability of recombinant IL-2, these tumor-infiltrating lymphocytes (TILs) could be more efficiently grown in vitro. In 1986, Rosenberg and colleagues reported that upon adoptive transfer in mice, TILs were 50–100-fold more efficacious than LAK cells in mediating antitumor immunity (Rosenberg and others 1986; Muul and others 1987). Importantly, in these experiments, the activity of TILs was enhanced by the administration of IL-2. In tandem with their characterization in mice, it was found that TILs could be generated from many types of human tumors (Kurnick and others 1986; Topalian and others 1987; Rosenberg 1992). In 1987, Kurnick and colleagues reported the administration of TIL to human patients with the treatment of 7 patients with metastatic adenocarcinoma of the lung (Kradin and others 1987). While there were no objective responses of at least 50% total reduction in tumor volume, the authors noted that 5 of 7 patients had some reduction in tumor volume. During the next 2 years, Kurnick and colleagues published 2 more studies in human patients using TIL with the addition of recombinant IL-2 (Kradin and others 1988, 1989). The authors observed partial (objective) responses in 3 of 13 melanoma patients and 2 of 7 renal cell carcinoma patients (Kradin and others 1988, 1989). During this same time, Rosenberg and colleagues also reported clinical trials using TIL with the addition of IL-2 administration (Rosenberg and others 1988; Topalian and others 1988). Based on mouse studies showing that preconditioning with cyclophosphamide was important for TIL efficacy (Rosenberg and others 1986), Rosenberg and colleagues administered cyclophosphamide to patients before TIL infusion. With the combination of cyclophosphamide, TIL, and high-dose IL-2, Rosenberg and colleagues achieved objective responses in 11 of 20 metastatic melanoma patients (Rosenberg and others 1988). Notably, 2 objective responses were observed among 5 patients in whom IL-2 therapy alone had failed to yield a response. Although there were limited numbers of patients and varying treatment protocols, there was great excitement that TIL therapy administered with IL-2 appeared to allow response rates much higher than that observed with IL-2 therapy alone. Importantly, and indicative of TIL's ability to mediate direct antitumor immunity, Griffith and others (1989) reported that indium-111 labeled TIL preferentially localized to tumor versus normal skin.

Several other reports warrant mention in better understanding TIL therapy and the relationship to IL-2. First, to more accurately define the response rate in a larger cohort of patients, Rosenberg and colleagues reported results of 86 patients with metastatic melanoma treated with TIL and IL-2 from May 1987 to December 1992 (Rosenberg and others 1994). The objective response rate from TIL therapy with IL-2 administration for these patients was 34%, a response rate much higher than what might have been expected with IL-2 alone. As part of the study, 59 of 86 patients received a single dose of cyclophosphamide (25 mg/kg) before TIL infusion. The response rate in patients without cyclophosphamide (31%) was not statistically different than patients with cyclophosphamide (35%) (Rosenberg and others 1994).

While a single injection of cyclophosphamide did not appear to significantly improve response rates of TIL therapy with IL-2, extensive studies in animals did suggest that lymphodepletion (using total body irradiation or chemotherapy) before adoptive cellular therapy would greatly improve donor lymphocyte persistence and antitumor efficacy (Glynn and others 1969; Berendt and North 1980; North 1982; Greenberg and others 1985, 1988; Rosenberg and others 1986; Muranski and others 2006; Paulos and others 2007; Wrzesinski and others 2010). Multiple mechanisms have been reported to mediate these effects, including the reduction of tumor burden, the destruction of suppressive cells, increased antigen presenting cells, and the release of lipopolysaccharide (LPS) from host microflora (Hill and others 1997; Zhang and others 2002; Brown and others 2004; Klebanoff and others 2005; Gattinoni and others 2006; Paulos and others 2007; Salem and others 2009; Yao and others 2012). Perhaps most relevant is the destruction of host lymphocytes which consume T cell growth factors such as IL-2, IL-7, or IL-15 (Schluns and others 2000; Tan and others 2001, 2002; Goldrath and others 2002; Gattinoni and others 2005; Johnson and others 2015; Martin and others 2017). The absence of host lymphocytes may also improve the availability of recombinant IL-2 for donor T cells. Furthermore, as IL-7 and IL-15 are produced by radiation-resistant cells, lymphodepletion leads to dramatically elevated IL-7 and IL-15 in mice and humans (Bolotin and others 1999; Fry and others 2001; Napolitano and others 2001; Miller and others 2005; Dudley and others 2008; Guimond and others 2009; Bergamaschi and others 2012). A demonstration of clinical relevance of lymphodepletion was reported in 2002 in patients with metastatic melanoma treated with TIL therapy and IL-2. In this trial, a much harsher conditioning regimen was provided with 2 days of cyclophosphamide (60 mg/kg) and 5 days of fludarabine (25 mg/m2) (Dudley and others 2002). This enhanced nonmyeloablative lymphodepletion conditioning regimen given with TIL and IL-2 resulted in objective responses in 6 of 13 patients. Notably, in this study TIL persisted at a greater level than in previous studies (Dudley and others 2002). Although the enhanced lymphodepletion regimen was likely critical, it is relevant that the T cell culture conditions in this study were different than earlier TIL studies in that a new rapid expansion protocol with both irradiated feeder cells and soluble anti-CD3 mAb was used to expand the cells immediately before infusion (Dudley and others 2002).

The success of lymphodepletion with cyclophosphamide and fludarabine led to additional interest in whether greater lymphodepletion might further improve response rates. Studies in mice suggested that the addition of total body irradiation to standard lymphodepletion would improve outcomes (Muranski and others 2006; Paulos and others 2007; Wrzesinski and others 2010). This rationale provided the framework for evaluating whether the addition of total body irradiation to standard cyclophosphamide and fludarabine would lead to improved clinical response of TIL therapy with IL-2. In initial studies, Rosenberg and colleagues observed higher response rates with the addition of 2 Gy (n = 25) or 12 Gy (n = 25) total body irradiation compared with the standard lymphodepletion nonmyeloablative conditioning regimen (n = 43) (Dudley and others 2008; Rosenberg and others 2011). However, another study reported no benefit of the addition of 6 Gy (n = 25) total body irradiation versus standard cyclophosphamide and fludarabine (n = 33) (Dudley and others 2010). To evaluate the value of total body irradiation, Rosenberg and colleagues performed a randomized study with (n = 50) or without (n = 51) 12 Gy total body irradiation in addition to standard cyclophosphamide and fludarabine (Goff and others 2016). In both groups, 24% of patients experienced complete responses, and there were no significant differences in overall survival (Goff and others 2016). Contrary to expectations, enhanced lymphodepletion did not improve the response rate of TIL and IL-2. These results suggest that there may be a threshold of lymphodepletion that is adequate for optimal antitumor responses. It is unclear if or how providing IL-2 support after administration of TIL may alter the optimal level of lymphodepletion.

While the addition of total body irradiation is unnecessary for the efficacy of TIL therapy, the ability of an enhanced, nonmyeloablative lymphodepletion regimen to mediate effective responses when given with TIL and IL-2 has now been reported by multiple groups (Pilon-Thomas and others 2012; Radvanyi and others 2012; Besser and others 2013). However, due to the difficulties in conducting these studies, the changing patient population, and evolving protocol designs, the optimal level of lymphodepletion and subsequent IL-2 support for patients receiving TIL therapy are yet to be determined.

While TIL has received much attention, other endogenous tumor-reactive T cell populations have been isolated, expanded, and used for cancer therapy (Yee and others 2002; Wallen and others 2009; Verdegaal and others 2011, 2016; Chapuis and others 2012, 2013; Yee 2014). In a striking example demonstrating the benefit of IL-2, Yee and others (2002) treated metastatic melanoma patients with multiple infusions of MART-1- and GP100-reactive CD8+ T cell clones in which patients received no lymphodepleting chemotherapy. After the first infusion, patients received no IL-2 therapy, but after subsequent infusions, patients received increasing amounts of low-dose IL-2 given subcutaneously (0.25, 0.5, and 1.0 × 106 U/m2 twice daily for 14 days) (Yee and others 2002). The authors found that T cells persisted much better when IL-2 was administered after adoptive transfer; however, most donor T cells failed to persist beyond 21 days in the blood. There were no objective responses, although 8 of 10 patients had minor, mixed, or stable responses for up to 21 months. It is relevant in this study that the T cells expressed IL-2Rα, which may have facilitated their responsiveness to low-dose IL-2. In a follow-up study in 11 patients with metastatic melanoma, Chapuis and others (2012) added cyclophosphamide (4 g/m2) before adoptive transfer of tumor-reactive T cell clones. Three patients received high-dose IL-2; however, this cohort was discontinued due to toxicity. Eight patients received low-dose IL-2, which was “well tolerated and safe” (Chapuis and others 2012). In this study, 4 of 11 patients had T cell persistence beyond 42 days. Furthermore, 1 patient had a complete response, and 5 had stable disease. These results further suggest that the appropriate balance between lymphodepletion and IL-2 therapy can lead to optimal T cell persistence and clinical responses.

IL-2 Combined with Genetically Modified T Cells

A significant advance in adoptive cellular therapy (ACT) was the development of methods to genetically engineer tumor-reactivity using the transfer of T cell receptor (TCR) or chimeric antigen receptor (CAR) genes (Gross and others 1989; Cole and others 1995; Kaplan and others 2003; Restifo and others 2012; Kochenderfer and Rosenberg 2013; Barrett and others 2014; Jensen and Riddell 2014; Kenderian and others 2014; Stromnes and others 2014; Nelson and Paulos 2015; Debets and others 2016; Sharma and Kranz 2016; Spear and others 2016; Turtle and others 2016; Lim and June 2017; Wang and Wang 2017). These approaches offer the possibility of generating tumor-reactive cells in patients who do not already possess such cells or where they cannot be isolated. Thus, for example, in metastatic melanoma it is estimated that only ∼27% to 45% of patients are able to receive TIL therapy due to factors such as inability to isolate tumor, inability to expand TIL, or patient disease progression during TIL preparation (Prieto and others 2010; Rosenberg and others 2011). The therapeutic utility of TCR- and CAR-modified T cells given with or without IL-2 is supported by studies in mice (Kessels and others 2001; Brentjens and others 2003; Chamoto and others 2004; Xue and others 2005; Chinnasamy and others 2010; Kochenderfer and others 2010b).

The successful translation of these efforts in humans was first reported in 2006 by Rosenberg and colleagues who adoptively transferred TCR-modified T cells into patients with metastatic melanoma (Morgan and others 2006). In this study, patients received nonmyeloablative chemotherapy, T cells genetically modified with a MART-1-reactive TCR, and high-dose IL-2. There were objective responses in 2 of 15 patients. Notably, these 2 patients had failed previous IL-2 therapy and also had remarkable persistence of their donor lymphocytes (Morgan and others 2006). Since this first experience, there have been multiple clinical trials using TCR-modified T cells, including TCRs reactive against CEA, MART-1, GP100, NY-ESO-1, MAGE-A3, and MAGE-A4 (Johnson and others 2009; Parkhurst and others 2011; Robbins and others 2011, 2015; Morgan and others 2013; Chodon and others 2014; Kageyama and others 2015; Rapoport and others 2015). In most of these studies, high-dose IL-2 was provided immediately after adoptive T cell transfer. Two studies of NY-ESO-1-reactive TCR-modified T cells are notable. Steven Rosenberg and colleagues used NY-ESO-1-reactive TCR-modified T cells in combination with lymphodepletion and high-dose IL-2 to successfully induce objective responses in 22 of 38 patients with either metastatic melanoma or synovial cell carcinoma (Robbins and others 2011, 2015). June and colleagues also used NY-ESO-1-reactive TCR-modified T cells to treat multiple myeloma and observed clinical responses in 16 of 20 patients, although without providing IL-2 (Rapoport and others 2015). As there were many differences between these studies, including the type of cancer, the methodology by which the T cells were expanded, and method of lymphodepletion, the value of IL-2 in these protocols is not clear.

While TCR-modified T cells show great promise in the treatment of solid tumors, the use of CAR-modified T cells has shown spectacular success in patients with B cell neoplasms. These recent successes followed a number of early clinical trials using CAR-modified T cells that did not result in clinical benefit (McGuinness and others 1999; Kershaw and others 2006; Lamers and others 2006; June and others 2014). In 2010 however, initial success was demonstrated by Kochenderfer and others (2010a) who used T cells modified with CD19-reactive CAR in combination with IL-2 to induce a response in a patient with advanced follicular lymphoma. A follow-up study by the same group in 2012 showed objective responses in 6 of 8 patients with advanced, progressive B cell malignancies that had failed other therapies (Kochenderfer and others 2012). These patients all received nonmyeloablative chemotherapy with cyclophosphamide and fludarabine followed by CAR-modified T cells and recombinant IL-2. In 2011, June and colleagues reported another major advance using another CD19-reactive CAR (Kalos and others 2011). In this case, the intracellular signaling domain of the CAR was modified with a 4-1BB signaling domain, and a modified, shorter T cell culture was used. Three patients with advance chronic lymphoid leukemia were treated with nonmyeloablative chemotherapy and CAR-modified T cells, but with no IL-2. All 3 patients had clinical responses, including 2 complete responses (Kalos and others 2011). June and colleagues, as well as other groups, have now extensively reported results demonstrating incredible efficacy of CD19-reactive CAR T cells in patients with a variety of B cell malignancies (Brentjens and others 2013; Grupp and others 2013; Maude and others 2014; Garfall and others 2015; Kochenderfer and others 2015; Lee and others 2015; Brudno and others 2016; Kebriaei and others 2016; Locke and others 2017). It is notable that for the most part, these studies have not used exogenous IL-2. Again, as was the case with other T cell therapies, the potential benefit and importance of IL-2 therapy are not clear, as between the various studies, there are differences, including cell culture conditions, lymphodepletion, and the CAR signaling domains. In the case of CD19-reactive CAR T cells, it may also be relevant that a large population of CD19+ nontumor cells might provide stimulation in such a manner that cytokine therapy is not necessary. However, a study by Kochenderfer and others (2017) reported an association between clinical response and elevated endogenous IL-15 (but not IL-2) in the serum, suggesting that IL-2 or IL-15-based therapy could have value as part of CAR therapy.

Alternatives to IL-2 Therapy

While administration of IL-2 is associated with remarkable clinical responses in certain patients, dose limiting toxicities make the widespread administration of this therapy infeasible. Side effects from high-dose IL-2 can be severe and include fever, chills, hypotension, tachycardia, oliguria, nausea, vomiting, diarrhea, capillary leak syndrome, renal failure, and thrombocytopenia requiring the use of blood pressure monitoring, volume replacements, and blood pressure support if needed. These side effects, however, are manageable when therapy is administered at experienced centers (Lotze and others 1986b; Rosenstein and others 1986; Kammula and others 1998; Dutcher and others 2001; Schwartzentruber 2001).

In addition to toxicity, IL-2 administration for cancer therapy carries the concern for expansion of T regulatory cells. T regulatory cells are especially responsive to IL-2 because they constitutively express high levels of the IL-2Rα subunit, necessary for high affinity IL-2 binding (Nishikawa and Sakaguchi 2014; Shevach and Thornton 2014; Yuan and others 2014; Waldmann 2015). The importance of IL-2 and T regulatory cells is particularly evident by the phenotypes of IL-2, IL-2Rα, and IL-2Rβ knockout mice, which all develop fatal autoimmunity (Sadlack and others 1993, 1995; Suzuki and others 1995; Willerford and others 1995; Yu and others 2003). Thus, while recombinant IL-2 can potently augment immune responses, it paradoxically plays a critical role in expanding a cell population that can shut down immune responses. The clinical relevance of this is suggested in patients treated with recombinant IL-2, where increased frequencies of FOXP3+ T regulatory cells have been reported (Zhang and others 2005; Ahmadzadeh 2006; Yao and others 2012). The ability of IL-2 to induce regulatory cells has been exploited to treat autoimmune disease. Thus, low doses of IL-2 have been given with clinical success to treat patients with chronic graft-versus-host disease, hepatitis C virus-induced vasculitis, and systemic lupus erythematosus (Koreth and others 2011; Saadoun and others 2011; Matsuoka and others 2013; He and others 2016). In addition to inducing T regulatory-mediated suppressive pathways (Matsuoka and others 2013; He and others 2016), low-dose IL-2 has also been shown in patients to induce the expansion of NK cell populations that may have antitumor potential (Caligiuri and others 1993). Thus, the interplay of IL-2-induced expansion of effector cells and regulatory cells is quite complicated.

One method to improve the utilization of high-dose IL-2 therapy would be to identify biomarkers to predict patients likely to respond. However, while there has been work to identify such biomarkers (Royal and others 1996; Bui and others 2003; Leibovich and others 2003; Atkins and others 2005; Upton and others 2005; Dudek and others 2010; Foureau and others 2014; Sim and others 2014; Kostner and others 2015; McDermott and others 2015a; Saraceni and others 2015; Chow and others 2016; Diller and others 2016; Kuzman and others 2017), there are no biomarkers that have been widely adopted to predict patients likely to respond. Newer methods and approaches for biomarkers have shown great promise in checkpoint therapy (Gibney and others 2016; Topalian and others 2016; Maleki Vareki and others 2017; Nishino and others 2017), but these methods have had limited evaluation in the context of IL-2 therapy. Notably, McDermott and others (2015a) reported that evidence in renal cell carcinoma that elevated PD-L1 expression in pretreatment tumors was favorably associated with clinical response. While being able to select patients likely to respond would be of value, there has been much effort to identify novel cytokine therapeutics with improved efficacy and reduced toxicities.

IL-15

The discovery in 1994 of IL-15 (Burton and others 1994; Grabstein and others 1994), which like IL-2 signals through the IL-2Rβγ subunits, raised the possibility that this novel cytokine might provide a therapeutic alternative to IL-2. Studies in mice suggested that toxicity at therapeutic doses associated with IL-15 might be more favorable than IL-2 (Munger and others 1995; Katsanis and others 1996; Kobayashi and others 2000). Given the similarities and potential advantages relative to IL-2, IL-15 was evaluated preclinically and showed efficacy in multiple models, including induction of antitumor immunity (Munger and others 1995; Evans and others 1997; Cao and others 1998; Di Carlo and others 2000; Rubinstein and others 2002; Kishida and others 2003; Oh and others 2003; Klebanoff and others 2004; Lasek and others 2004; Roychowdhury and others 2004; Melchionda and others 2005; Ugen and others 2006; Basak and others 2008; Habibi and others 2009; Liu and others 2013). Notably, recombinant IL-15 enhances the efficacy of adoptively transferred tumor-reactive T cells given concomitantly with vaccination in mice bearing established subcutaneous melanoma tumors (Klebanoff and others 2004). Given a variety of preclinical results, enthusiasm for IL-15 led to it being ranked first by the 2007 NCI immunotherapy agent workshop as the experimental agent with highest potential for treating cancer (Cheever 2008). In 2015, Waldmann and colleagues reported that they could safely administer IL-15 to patients and induce biological activity on NK cells and CD8+ memory T cells (Conlon and others 2015). Although there were no objective responses, there was clearance of lung lesions in 2 patients with metastatic melanoma (Conlon and others 2015). Waldmann and colleagues have recently reported a second cohort of patients treated with recombinant IL-15 and provided a detailed analysis of the response of NK cells to treatment (Dubois and others 2017).

While IL-15 has shown great promise as an antitumor agent, in 2006 it was found that the biological activity of IL-15 could be further improved by preassociation with its soluble receptor, IL-15Rα (Mortier and others 2006; Rubinstein and others 2006; Stoklasek and others 2006; Bergamaschi and others 2008; Dubois and others 2008), thereby creating IL-15/IL-15Rα complexes. Interestingly, in vitro association of IL-15 with either a monomeric or dimeric sIL-15Rα (fused to an Fc) led to greatly improved activity (Rubinstein and others 2006). These results suggest that when bound to IL-15Rα, IL-15 might undergo a conformational change into a superagonist (Rubinstein and others 2006). Structural evidence in support of this was subsequently reported by Garcia and colleagues (Ring and others 2012). While the activity of these IL-15/sIL-15Rα complexes was notable in vitro, upon infusion, IL-15/sIL-15Rα complexes mediated more than 50-fold greater activity than free IL-15 (Rubinstein and others 2006; Stoklasek and others 2006). As sIL-15Ra-Fc only improved IL-15 activity about 7-fold in vitro (Rubinstein and others 2006), these data suggest that there are additional mechanisms leading to improved biological activity in vivo. Potential mechanisms accounting for the improved activity in vivo include the ability of sIL-15Ra-Fc to act as a carrier protein, to redirect the localization of IL-15, and to protect IL-15 from proteolytic degradation.

The increased biological activity of IL-15/sIL-15Rα-Fc complexes in vivo is apparent in multiple readouts. IL-15/sIL-15Rα complexes induce the potent expansion of cell types important to antitumor immunity (including CD8+ memory T cells, NK cells, and NK T cells) and mediate potent antitumor immunity in mouse models (Rubinstein and others 2006; Stoklasek and others 2006; Bergamaschi and others 2008; Dubois and others 2008; Epardaud and others 2008; Bessard and others 2009; Desbois and others 2016). Following infection in mice, IL-15/sIL-15Rα-Fc complexes were also able to effectively augment the number of responding antigen-specific CD8+ T cells demonstrating the potential of this reagent to have efficacy with vaccination approaches (Epardaud and others 2008). In the interest of clinical translation, several groups have generated IL-15/sIL-15Rα complexes with or without inclusion of an Fc (Bouchaud and others 2008; Rowley and others 2009; Han and others 2011; Stone and others 2012; Tosic and others 2014). Notably, Wong and colleagues generated IL-15/IL-15Rα complexes with an amino acid mutation that further improved biological activity (Han and others 2011). This molecule has shown potent efficacy in augmenting antitumor responses in murine tumor models, and also, in inducing the expansion of lymphocytes in cynomolgus monkeys (Xu and others 2013; Gomes-Giacoia and others 2014; Rhode and others 2016). This molecule, which has been designated ALT-803, is currently in clinical testing. In addition to ALT-803 manufactured by Altor Bioscience, other IL-15/IL-15Ra complexes are in development by Novartis (Admune) and Cytune Pharma (Desbois and others 2016; Thaysen-Andersen and others 2016).

IL-7

IL-7 has been reviewed in detail elsewhere (Hofmeister and others 1999; Fry and Mackall 2002; Jiang and others 2005; Ma and others 2006; Mazzucchelli and Durum 2007; Overwijk and Schluns 2009; Mackall and others 2011; Carrette and Surh 2012; Lin and others 2017). IL-7 has many similarities with IL-2 and IL-15, including utilization of the shared IL-2Rγ subunit. IL-7 and its second receptor subunit, IL-7Rα, were cloned in the late 1980s (Namen and others 1988; Goodwin and others 1989, 1990). While IL-7Rα does not interact with any other IL-2Rγ-chain cytokine members, in conjunction with the thymic stromal lymphopoietin receptor (TSLPR), IL-7Rα mediates signaling and biological activity of TSLP in a wide range of immune cells (Levin and others 1999; Pandey and others 2000; Park and others 2000; Roan and others 2012; Lo Kuan and Ziegler 2014). The IL-7 receptor, IL-7Rα/IL-2Rγ, is expressed on both developing and mature B cells and T cells. Mice deficient in IL-7, IL-7Rα, or IL-2Rγ have a severe combined immunodeficiency (SCID) phenotype consistent with the critical role in these cytokines and receptors in lymphocyte development and survival (Peschon and others 1994; Cao and others 1995; von Freeden-Jeffry and others 1995). The lymphopenia of the IL-7Rα KO mouse is slightly more severe than the IL-7 KO mouse, suggesting that TSLP may play a role in supporting lymphocyte development and survival (Pandey and others 2000). Studies using IL-7 transgenic mice and administration of recombinant IL-7 into mice show that exogenous IL-7 can greatly augment the numbers of both T cells and B cells, although both mature and immature B cells are expanded (Morrissey and others 1991; Samaridis and others 1991; Damia and others 1992; Komschlies and others 1994; Fisher and others 1995; Mertsching and others 1995; Valenzona and others 1996; Melchionda and others 2005; Nanjappa and others 2008). In preclinical models, IL-7 administration can promote immune responses against tumor and infectious disease (Nanjappa and others 2008, 2011; Andersson and others 2009; Cui and others 2009; Pellegrini and others 2011; Tang and others 2014; Ruan and others 2016). It is noteworthy that surface expression of IL-7Rα is high on naive and memory T cells, but reduced upon T cell activation, suggesting that effector T cells may have reduced ability to respond to IL-7 (Foxwell and others 1992; Schluns and others 2000; Goldrath and others 2002; Xue and others 2002; Kaech and others 2003; Klonowski and others 2006). However, depending on the method of T cell activation, there may be sufficient surface IL-7Rα on effector T cells for IL-7 responsiveness (Johnson and others 2015).

The first human trial for the evaluation of IL-7 was reported in 2006 when Rosenberg and others (2006) demonstrated that IL-7 administration to cancer patients could enhance CD8+ and CD4+ T cell counts, with reduced frequencies of T regulatory cells, but with no objective responses. A number of other clinical studies have also evaluated IL-7 in human patients, including in cancer and HIV infection (Sportes and others 2008, 2010; Levy and others 2009, 2012; Sereti and others 2009; Perales and others 2012; Alstadhaug and others 2014; Gasnault and others 2014; Tredan and others 2015; Sheikh and others 2016; Thiebaut and others 2016). It is notable that the pre-B cell expansion observed in murine models was not apparent in humans, suggesting a critical difference between mouse and humans (Sportes and others 2010). Furthermore, unlike IL-2 therapy, IL-7 therapy does not lead to the expansion of CD4+CD25+ T regulatory cells (Rosenberg and others 2006; Sereti and others 2009; Sportes and others 2010).

Novel IL-2 based therapies

The design of mutant IL-2 molecules provides another option for improving the biological activity of IL-2. Several groups have designed IL-2 mutants that have reduced binding to IL-2Rα and in some cases have enhanced binding to IL-2Rβγ (Heaton and others 1993; Levin and others 2012; Carmenate and others 2013). These molecules have shown improved antitumor efficacy and reduced toxicity in animal models. An alternative approach has been the generation of IL-2 mutants with enhanced ability to bind IL-2Rαβγ relative to IL-2Rβγ. Shanafelt and others (2000) designed an IL-2 mutant with reduced IL-2Rβγ binding with the goal of minimizing toxicity mediated by IL-2Rβγ+ NK cells. This molecule, designated BAY 50-4798, was tested as a single agent in cancer patients. While able to induce some clinical responses in cancer patients, there were not obvious advantages compared with conventional IL-2 with the treatment protocol (Margolin and others 2007). In a related approach, other groups have designed mutant IL-2 molecules with improved binding of IL-2 to IL-2Rα. Fallon and others (2000) reported an IL-2 analog that may undergo increased endosomal recycling due to an alteration in the pH sensitivity between IL-2 and IL-2Rα. In separate studies, Rao and others also generated IL-2 mutants with improved affinity for IL-2Rα (Rao and others 2003, 2005). Unlike wild-type IL-2, these mutants could persist on the cell surface and mediate durable cell signaling as has been reported for IL-15 (Dubois and others 2002). Whether these mutant IL-2 molecules have clinical efficacy, and under what circumstances, remains to be determined.

While there has been much effort to augment or tailor IL-2 signaling, there are also mutants of IL-2 designed to inhibit signaling (Liu and others 2009; Mitra and others 2015). For example, Liu and others (2009) designed an IL-2 antagonist by selecting for high affinity binding to IL-2Rα and loss of binding to IL-2Rβγ. These molecules may selectively block high affinity IL-2 signaling and, for example, could suppress T regulatory responses. While mutant IL-2 molecules have promise, it is also worth mention that Tsytsikov and others (1996) reported natural variants of IL-2, generated by alternative splicing, that may competitively inhibit full length IL-2.

In addition to modifying the specificity of IL-2, other approaches seek to improve the half-life and biological activity of IL-2. Pegylation of recombinant molecules can improve half-life and biological activity (Pasut and Veronese 2009; Milla and others 2012; Turecek and others 2016), and pegylated IL-2 (PEG-IL-2) molecules have been generated with improved biological activity, half-life, and antitumor activity versus nonpegylated IL-2 (Katre and others 1987; Knauf and others 1988; Zimmerman and others 1989; Katre 1990; Yang and others 1991; Charych and others 2016). Administration of PEG-IL-2 to patients with metastatic melanoma and renal cell carcinoma was associated with objective responses (Meyers and others 1991; Yang and others 1995). Another benefit of this approach is that the pegylation process may alter the IL-2 to redirect its target cell specificity. NKTR-214 is IL-2 that has been pegylated in a way to reduce or mask its ability to engage to IL-2Rα, thereby making it more like IL-15 (Charych and others 2016). NKTR-214, which is produced by Nektar, is in clinical testing. Other methods for improving the half-life of IL-2 include the generation of fusion proteins, including the linkage of IL-2 with the Fc region of an antibody or albumin (Zheng and others 1999; Yao and others 2004; Melder and others 2005).

IL-2 therapy using antibody/cytokine complexes

Another method to increase the biological activity of IL-2 is by preassociation with anti-IL-2 mAb, to generate IL-2/mAb complexes, before infusion. IL-2/mAb complexes were first reported in 1993 and showed greatly enhanced antitumor efficacy versus free IL-2 (Sato and others 1993; Courtney and others 1994). The enhancement of activity was associated with improved IL-2 persistence in vivo. There are a number of potential enhancing mechanisms that may be responsible for this improved activity, including improved half-life, protection from degradation, and altered localization. Interestingly, despite these early studies demonstrating that antibodies could enhance the biological activity of IL-2, the administration of anticytokine monoclonal antibodies to reduce IL-2 responses was routine. In fact, it was thought that administration of anti-IL-2 mAb would block IL-2 and thus reduce T regulatory cell activity resulting in improved proliferation of CD8+ memory-phenotype T cells. However, in 2006, Boyman and others (2006) demonstrated that instead of blocking IL-2, anti-IL-2 mAb promoted the biological activity of endogenous IL-2, thus directly stimulating CD8+ memory-phenotype T cells. Furthermore, injection of preformed IL-2/mAb complexes exhibited potent biological activity on CD8+ memory-phenotype T cells and NK cells.

Another important property of IL-2/mAb complexes is that target cell specificity can be altered compared with native IL-2 (Boyman and others 2006). Thus, depending on the choice of anti-IL-2 mAb, IL-2 can be redirected either in favor of IL-2Rαhi T cells or independent of IL-2Rα expression. The ability to redirect IL-2 target cell specificity may relate to the ability of anti-IL-2 mAb to block the IL-2 interaction with certain cytokine receptor subunits or may conformationally stabilize certain interactions (Spangler and others 2015).

IL-2/mAb complexes have now shown efficacy in augmenting immune responses in a wide range of preclinical models, including antitumor immunity (Jin and others 2008; Mostbock and others 2008; Wilson and others 2008; Molloy and others 2009; Tomala and others 2009; Webster and others 2009; Hamilton and others 2010; Krieg and others 2010; Liu and others 2010; Smith and others 2011; Lee and others 2012; Kim and others 2013, 2015). In addition to antibody cytokine complexes generated with IL-2, it is relevant that cytokine complexes have also been reported with IL-4, IL-7, and IL-15 with potent biological activity, including on lymphocytes (Finkelman and others 1993; Boyman and others 2008; Phelan and others 2008; Rubinstein and others 2008; Morris and others 2009; Finch and others 2011).

Combination Therapies with IL-2

While IL-2 has shown remarkable efficacy, most patients do not achieve a clinical response. If IL-2 or related cytokines are to benefit a broader group of cancer patients, combinational therapies will likely be necessary. One advantage of IL-2-type therapies is the ability to integrate into almost any other form of immune- or nonimmune therapy. Thus, in any situation where T cells or NK cells participate in mediating immune responses, IL-2 therapy could mediate improved responses. Two therapies in particular warrant discussion and may benefit from the addition of IL-2 therapy: antibody-based therapies targeting tumor cells and immune-checkpoint therapies.

Antibody therapies can target tumor associated antigens and initiate antibody dependent cell-mediated cytotoxicity or complement-mediated lysis. IL-2 or IL-15-based therapies can expand and activate Fc+ lymphocytes such as NK cells that may directly improve the efficacy of such antibody-mediated therapeutics. A number of preclinical studies show synergy of IL-2 or IL-15 with such antibody-based therapies, and furthermore, depletion of NK cells can abrogate this effect (Eisenbeis and others 2004; Abes and others 2010; Tzeng and others 2015; Zhu and others 2015; Rosario and others 2016). These efforts have led to a number of clinical studies combining IL-2 therapy with rituximab (anti-CD20 mAb) or herceptin (anti-Her2/neu mAb) (Eisenbeis and others 2004; Khan and others 2006; Mani and others 2009; Poire and others 2010). An ongoing clinical trial involves combining ALT-803 (IL-15/IL-15Ra complexes) with rituximab for the treatment of non-Hodgkin's lymphoma (NCT02384954). This combinatorial approach may also have promise in the treatment of infectious diseases where antibodies are currently being evaluated.

Immune checkpoint therapy has shown efficacy in a growing number of cancers, including metastatic melanoma and renal cell carcinoma (Hodi and others 2010; Topalian and others 2012; Hamid and others 2013; Wolchok and others 2013; McDermott and others 2015b; Motzer and others 2015; Schadendorf and others 2015; Sharma and Allison 2015), but likely act in mechanistically distinct ways from IL-2-mediated therapy, thereby suggesting value for combinatorial therapy. Most simply, immune checkpoint therapies may overcome negative regulatory pathways, while cytokine therapies may expand and activate newly available tumor-specific T cell populations. In support of the potential synergy of these approaches, preclinical studies using tumor models and chronic viral infection have shown that IL-2- and IL-15-based therapies synergize with both anti-PD-1 and anti-CTLA-4 mAbs (Yu and others 2010; John and others 2013; West and others 2013; Desbois and others 2016; Mathios and others 2016), and other preclinical mechanistic studies also support the use of these combinations (Shi and others 2016; Asano and others 2017). In humans, there has been only 1 published report of a clinical trial directly combining immune checkpoint therapy with either IL-2 or IL-15. Rosenberg and others described treating 36 patients with metastatic melanoma with anti-CTLA-4 mAb (ipilimumab) and high-dose IL-2. The authors reported 3 (8%) complete responses in the initial analysis and 6 (17%) complete responses in the longer follow-up (Maker and others 2005; Prieto and others 2012). Although only a small number of patients were enrolled in this study, these results provide clinical support for combining checkpoint therapy with common γ-chain agonist cytokines. Notably, a follow-up study of high-dose IL-2 and ipilimumab is ongoing (NCT02203604). There are several other relevant trials ongoing, including the combination of IL-15/IL-15Rα complexes (ALT-803, NCT02523469), pegylated IL-2 (NKTR-214, NCT02983045, and NCT03138889), and high- and low-dose IL-2 (NCT02964078, NCT02989714, and NCT03111901) with anti-PD-1 mAbs (nivolumab and pembrolizumab) and anti-PD-L1 mAb (atezolizumab). While not concomitant therapy, also of interest is a case report of patient with metastatic renal cell carcinoma who did not respond to anti-PD-1 mAb, but did achieve a near complete response to subsequent IL-2 therapy (Brayer and Fishman 2014).

Conclusion

Immunotherapy is now commonplace for multiple malignancies, and in many ways, the use of IL-2 pioneered the introduction of this therapeutic modality into oncologic practice. While the use of high-dose IL-2 has remained a therapy only available at specialized centers, this therapy has given proof of principle for an alternate and potentially curative paradigm. While much has been learned about the IL-2 and related cytokines since their initial discovery, this area of research remains intensely active with multiple pharmaceutical companies aggressively pursuing clinical investigation of novel and safer IL-2 related reagents. Future directions will likely include combinatorial strategies that take advantage of the broad activity mediated by IL-2 and related cytokine therapeutics.

Acknowledgments

The authors are grateful for support from the National Cancer Institute (P01CA154778) and the Cancer Research Institute.

Author Disclosure Statement

MPR is an inventor on a patent application related to IL-2/mAb complexes. The other authors declare no competing financial interests.

References

  1. Abes R, Gelize E, Fridman WH, Teillaud JL. 2010. Long-lasting antitumor protection by anti-CD20 antibody through cellular immune response. Blood 116(6):926–934 [DOI] [PubMed] [Google Scholar]
  2. Ahmadzadeh M, Rosenberg SA. 2006. IL-2 administration increases CD4+CD25hiFoxp3+ regulatory Tcells in cancer patients. Blood 107(6):2409–2414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Almeida AR, Legrand N, Papiernik M, Freitas AA. 2002. Homeostasis of peripheral CD4+ T cells: IL-2R alpha and IL-2 shape a population of regulatory cells that controls CD4+ T cell numbers. J Immunol 169(9):4850–4860 [DOI] [PubMed] [Google Scholar]
  4. Alstadhaug KB, Croughs T, Henriksen S, Leboeuf C, Sereti I, Hirsch HH, Rinaldo CH. 2014. Treatment of progressive multifocal leukoencephalopathy with interleukin 7. JAMA Neurol 71(8):1030–1035 [DOI] [PubMed] [Google Scholar]
  5. Andersson A, Yang SC, Huang M, Zhu L, Kar UK, Batra RK, Elashoff D, Strieter RM, Dubinett SM, Sharma S. 2009. IL-7 promotes CXCR3 ligand-dependent T cell antitumor reactivity in lung cancer. J Immunol 182(11):6951–6958 [DOI] [PubMed] [Google Scholar]
  6. Andersson EC, Christensen JP, Marker O, Thomsen AR. 1994. Changes in cell adhesion molecule expression on T cells associated with systemic virus infection. J Immunol 152(3):1237–1245 [PubMed] [Google Scholar]
  7. Arneja A, Johnson H, Gabrovsek L, Lauffenburger DA, White FM. 2014. Qualitatively different T cell phenotypic responses to IL-2 versus IL-15 are unified by identical dependences on receptor signal strength and duration. J Immunol 192(1):123–135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Asano T, Meguri Y, Yoshioka T, Kishi Y, Iwamoto M, Nakamura M, Sando Y, Yagita H, Koreth J, Kim HT, Alyea EP, Armand P, Cutler CS, Ho VT, Antin JH, Soiffer RJ, Maeda Y, Tanimoto M, Ritz J, Matsuoka KI. 2017. PD-1 modulates regulatory T-cell homeostasis during low-dose interleukin-2 therapy. Blood 129(15):2186–2197 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Atkins M, Regan M, McDermott D, Mier J, Stanbridge E, Youmans A, Febbo P, Upton M, Lechpammer M, Signoretti S. 2005. Carbonic anhydrase IX expression predicts outcome of interleukin 2 therapy for renal cancer. Clin Cancer Res 11(10):3714–3721 [DOI] [PubMed] [Google Scholar]
  10. Atkins MB, Lotze MT, Dutcher JP, Fisher RI, Weiss G, Margolin K, Abrams J, Sznol M, Parkinson D, Hawkins M, Paradise C, Kinkel L, Rosenberg SA. 1999. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 17:2105–2116 [DOI] [PubMed] [Google Scholar]
  11. Badoual C, Bouchaud G, Agueznay Nel H, Mortier E, Hans S, Gey A, Fernani F, Peyrard S, Puig PL, Bruneval P, Sastre X, Plet A, Garrigue-Antar L, Quintin-Colonna F, Fridman WH, Brasnu D, Jacques Y, Tartour E. 2008. The soluble alpha chain of interleukin-15 receptor: a proinflammatory molecule associated with tumor progression in head and neck cancer. Cancer Res 68(10):3907–3914 [DOI] [PubMed] [Google Scholar]
  12. Baran D, Korner M, Theze J. 1988. Characterization of the soluble murine IL-2R and estimation of its affinity for IL-2. J Immunol 141(2):539–546 [PubMed] [Google Scholar]
  13. Barrett DM, Singh N, Porter DL, Grupp SA, June CH. 2014. Chimeric antigen receptor therapy for cancer. Annu Rev Med 65:333–347 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Basak GW, Zapala L, Wysocki PJ, Mackiewicz A, Jakobisiak M, Lasek W. 2008. Interleukin 15 augments antitumor activity of cytokine gene-modified melanoma cell vaccines in a murine model. Oncol Rep 19(5):1173–1179 [PubMed] [Google Scholar]
  15. Berendt MJ, North RJ. 1980. T-cell-mediated suppression of anti-tumor immunity. An explanation for progressive growth of an immunogenic tumor. J Exp Med 151(1):69–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bergamaschi C, Bear J, Rosati M, Beach RK, Alicea C, Sowder R, Chertova E, Rosenberg SA, Felber BK, Pavlakis GN. 2012. Circulating IL-15 exists as heterodimeric complex with soluble IL-15Ralpha in human and mouse serum. Blood 120(1):e1–e8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bergamaschi C, Rosati M, Jalah R, Valentin A, Kulkarni V, Alicea C, Zhang GM, Patel V, Felber BK, Pavlakis GN. 2008. Intracellular interaction of interleukin-15 with its receptor alpha during production leads to mutual stabilization and increased bioactivity. J Biol Chem 283(7):4189–4199 [DOI] [PubMed] [Google Scholar]
  18. Bergmann CA, Gould DL, Kaplan DR. 1992. Cytometrically detected specific binding of interleukin 2 to cells. Cytokine 4(3):192–200 [DOI] [PubMed] [Google Scholar]
  19. Bessard A, Sole V, Bouchaud G, Quemener A, Jacques Y. 2009. High antitumor activity of RLI, an interleukin-15 (IL-15)-IL-15 receptor alpha fusion protein, in metastatic melanoma and colorectal cancer. Mol Cancer Ther 8(9):2736–2745 [DOI] [PubMed] [Google Scholar]
  20. Besser MJ, Shapira-Frommer R, Itzhaki O, Treves AJ, Zippel DB, Levy D, Kubi A, Shoshani N, Zikich D, Ohayon Y, Ohayon D, Shalmon B, Markel G, Yerushalmi R, Apter S, Ben-Nun A, Ben-Ami E, Shimoni A, Nagler A, Schachter J. 2013. Adoptive transfer of tumor-infiltrating lymphocytes in patients with metastatic melanoma: intent-to-treat analysis and efficacy after failure to prior immunotherapies. Clin Cancer Res 19(17):4792–4800 [DOI] [PubMed] [Google Scholar]
  21. Bindon C, Czerniecki M, Ruell P, Edwards A, McCarthy WH, Harris R, Hersey P. 1983. Clearance rates and systemic effects of intravenously administered interleukin 2 (IL-2) containing preparations in human subjects. Br J Cancer 47(1):123–133 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Bodnar A, Nizsaloczki E, Mocsar G, Szaloki N, Waldmann TA, Damjanovich S, Vamosi G. 2008. A biophysical approach to IL-2 and IL-15 receptor function: localization, conformation and interactions. Immunol Lett 116(2):117–125 [DOI] [PubMed] [Google Scholar]
  23. Bolotin E, Annett G, Parkman R, Weinberg K. 1999. Serum levels of IL-7 in bone marrow transplant recipients: relationship to clinical characteristics and lymphocyte count. Bone Marrow Transplant 23(8):783–788 [DOI] [PubMed] [Google Scholar]
  24. Bouchaud G, Garrigue-Antar L, Sole V, Quemener A, Boublik Y, Mortier E, Perdreau H, Jacques Y, Plet A. 2008. The exon-3-encoded domain of IL-15ralpha contributes to IL-15 high-affinity binding and is crucial for the IL-15 antagonistic effect of soluble IL-15Ralpha. J Mol Biol 382(1):1–12 [DOI] [PubMed] [Google Scholar]
  25. Boyman O, Kovar M, Rubinstein MP, Surh CD, Sprent J. 2006. Selective stimulation of T cell subsets with antibody-cytokine immune complexes. Science 311(5769):1924–1927 [DOI] [PubMed] [Google Scholar]
  26. Boyman O, Purton JF, Surh CD, Sprent J. 2007. Cytokines and T-cell homeostasis. Curr Opin Immunol 19(3):320–326 [DOI] [PubMed] [Google Scholar]
  27. Boyman O, Ramsey C, Kim DM, Sprent J, Surh CD. 2008. IL-7/anti-IL-7 mAb complexes restore T cell development and induce homeostatic T Cell expansion without lymphopenia. J Immunol 180(11):7265–7275 [DOI] [PubMed] [Google Scholar]
  28. Boyman O, Sprent J. 2012. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol 12(3):180–190 [DOI] [PubMed] [Google Scholar]
  29. Brayer J, Fishman M. 2014. Regression of metastatic clear cell kidney cancer with interleukin-2 treatment following nivolumab (anti-PD-1) treatment. J Immunother 37(3):187–191 [DOI] [PubMed] [Google Scholar]
  30. Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, Bartido S, Stefanski J, Taylor C, Olszewska M, Borquez-Ojeda O, Qu J, Wasielewska T, He Q, Bernal Y, Rijo IV, Hedvat C, Kobos R, Curran K, Steinherz P, Jurcic J, Rosenblat T, Maslak P, Frattini M, Sadelain M. 2013. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 5(177):177ra38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Brentjens RJ, Latouche JB, Santos E, Marti F, Gong MC, Lyddane C, King PD, Larson S, Weiss M, Riviere I, Sadelain M. 2003. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat Med 9(3):279–286 [DOI] [PubMed] [Google Scholar]
  32. Brown S, Konopa J, Zhou D, Thompson J. 2004. Expression of TNFalpha by CD3+ and F4/80+ cells following irradiation preconditioning and allogeneic spleen cell transplantation. Bone Marrow Transplant 33(4):359–365 [DOI] [PubMed] [Google Scholar]
  33. Brudno JN, Somerville RP, Shi V, Rose JJ, Halverson DC, Fowler DH, Gea-Banacloche JC, Pavletic SZ, Hickstein DD, Lu TL, Feldman SA, Iwamoto AT, Kurlander R, Maric I, Goy A, Hansen BG, Wilder JS, Blacklock-Schuver B, Hakim FT, Rosenberg SA, Gress RE, Kochenderfer JN. 2016. Allogeneic T cells that express an anti-CD19 chimeric antigen receptor induce remissions of B-cell malignancies that progress after allogeneic hematopoietic stem-cell transplantation without causing graft-versus-host disease. J Clin Oncol 34(10):1112–1121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Bui MH, Seligson D, Han KR, Pantuck AJ, Dorey FJ, Huang Y, Horvath S, Leibovich BC, Chopra S, Liao SY, Stanbridge E, Lerman MI, Palotie A, Figlin RA, Belldegrun AS. 2003. Carbonic anhydrase IX is an independent predictor of survival in advanced renal clear cell carcinoma: implications for prognosis and therapy. Clin Cancer Res 9(2):802–811 [PubMed] [Google Scholar]
  35. Burkett PR, Koka R, Chien M, Chai S, Chan F, Ma A, Boone DL. 2003. IL-15R alpha expression on CD8+ T cells is dispensable for T cell memory. Proc Natl Acad Sci U S A 100(8):4724–4729 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Burton JD, Bamford RN, Peters C, Grant AJ, Kurys G, Goldman CK, Brennan J, Roessler E, Waldmann TA. 1994. A lymphokine, provisionally designated interleukin T and produced by a human adult T-cell leukemia line, stimulates T-cell proliferation and the induction of lymphokine-activated killer cells. Proc Natl Acad Sci U S A 91(11):4935–4939 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Cabrera R, Ararat M, Eksioglu EA, Cao M, Xu Y, Wasserfall C, Atkinson MA, Liu C, Nelson DR. 2010. Influence of serum and soluble CD25 (sCD25) on regulatory and effector T-cell function in hepatocellular carcinoma. Scand J Immunol 72(4):293–301 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Caligiuri MA, Murray C, Robertson MJ, Wang E, Cochran K, Cameron C, Schow P, Ross ME, Klumpp TR, Soiffer RJ, et al. . 1993. Selective modulation of human natural killer cells in vivo after prolonged infusion of low dose recombinant interleukin 2. J Clin Invest 91(1):123–132 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Cantrell DA, Smith KA. 1983. Transient expression of interleukin 2 receptors. Consequences for T cell growth. J Exp Med 158(6):1895–1911 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Cao S, Troutt AB, Rustum YM. 1998. Interleukin 15 protects against toxicity and potentiates antitumor activity of 5-fluorouracil alone and in combination with leucovorin in rats bearing colorectal cancer. Cancer Res 58(8):1695–1699 [PubMed] [Google Scholar]
  41. Cao X, Shores EW, Hu-Li J, Anver MR, Kelsall BL, Russell SM, Drago J, Noguchi M, Grinberg A, Bloom ET, et al. . 1995. Defective lymphoid development in mice lacking expression of the common cytokine receptor gamma chain. Immunity 2(3):223–238 [DOI] [PubMed] [Google Scholar]
  42. Carmenate T, Pacios A, Enamorado M, Moreno E, Garcia-Martinez K, Fuente D, Leon K. 2013. Human IL-2 mutein with higher antitumor efficacy than wild type IL-2. J Immunol 190(12):6230–6238 [DOI] [PubMed] [Google Scholar]
  43. Carrette F, Surh CD. 2012. IL-7 signaling and CD127 receptor regulation in the control of T cell homeostasis. Semin Immunol 24(3):209–217 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Castillo EF, Schluns KS. 2012. Regulating the immune system via IL-15 transpresentation. Cytokine 59(3):479–490 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Castillo EF, Stonier SW, Frasca L, Schluns KS. 2009. Dendritic cells support the in vivo development and maintenance of NK cells via IL-15 trans-presentation. J Immunol 183(8):4948–4956 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Chamoto K, Tsuji T, Funamoto H, Kosaka A, Matsuzaki J, Sato T, Abe H, Fujio K, Yamamoto K, Kitamura T, Takeshima T, Togashi Y, Nishimura T. 2004. Potentiation of tumor eradication by adoptive immunotherapy with T-cell receptor gene-transduced T-helper type 1 cells. Cancer Res 64(1):386–390 [DOI] [PubMed] [Google Scholar]
  47. Chapuis AG, Ragnarsson GB, Nguyen HN, Chaney CN, Pufnock JS, Schmitt TM, Duerkopp N, Roberts IM, Pogosov GL, Ho WY, Ochsenreither S, Wolfl M, Bar M, Radich JP, Yee C, Greenberg PD. 2013. Transferred WT1-reactive CD8+ T cells can mediate antileukemic activity and persist in post-transplant patients. Sci Transl Med 5(174):174ra27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Chapuis AG, Thompson JA, Margolin KA, Rodmyre R, Lai IP, Dowdy K, Farrar EA, Bhatia S, Sabath DE, Cao J, Li Y, Yee C. 2012. Transferred melanoma-specific CD8+ T cells persist, mediate tumor regression, and acquire central memory phenotype. Proc Natl Acad Sci U S A 109(12):4592–4597 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Charych DH, Hoch U, Langowski JL, Lee SR, Addepalli MK, Kirk PB, Sheng D, Liu X, Sims PW, VanderVeen LA, Ali CF, Chang TK, Konakova M, Pena RL, Kanhere RS, Kirksey YM, Ji C, Wang Y, Huang J, Sweeney TD, Kantak SS, Doberstein SK. 2016. NKTR-214, an engineered cytokine with biased IL2 receptor binding, increased tumor exposure, and marked efficacy in mouse tumor models. Clin Cancer Res 22(3):680–690 [DOI] [PubMed] [Google Scholar]
  50. Cheever MA. 2008. Twelve immunotherapy drugs that could cure cancers. Immunol Rev 222:357–368 [DOI] [PubMed] [Google Scholar]
  51. Cheever MA, Greenberg PD, Fefer A, Gillis S. 1982. Augmentation of the anti-tumor therapeutic efficacy of long-term cultured T lymphocytes by in vivo administration of purified interleukin 2. J Exp Med 155(4):968–980 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Cheever MA, Greenberg PD, Irle C, Thompson JA, Urdal DL, Mochizuki DY, Henney CS, Gillis S. 1984. Interleukin 2 administered in vivo induces the growth of cultured T cells in vivo. J Immunol 132(5):2259–2265 [PubMed] [Google Scholar]
  53. Chen DM, Di Sabato G. 1976. Further studies on the thymocyte stimulating factor. Cell Immunol 22(2):211–224 [DOI] [PubMed] [Google Scholar]
  54. Chinnasamy D, Yu Z, Theoret MR, Zhao Y, Shrimali RK, Morgan RA, Feldman SA, Restifo NP, Rosenberg SA. 2010. Gene therapy using genetically modified lymphocytes targeting VEGFR-2 inhibits the growth of vascularized syngenis tumors in mice. J Clin Invest 120(11):3953–3968 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Chodon T, Comin-Anduix B, Chmielowski B, Koya RC, Wu Z, Auerbach M, Ng C, Avramis E, Seja E, Villanueva A, McCannel TA, Ishiyama A, Czernin J, Radu CG, Wang X, Gjertson DW, Cochran AJ, Cornetta K, Wong DJ, Kaplan-Lefko P, Hamid O, Samlowski W, Cohen PA, Daniels GA, Mukherji B, Yang L, Zack JA, Kohn DB, Heath JR, Glaspy JA, Witte ON, Baltimore D, Economou JS, Ribas A. 2014. Adoptive transfer of MART-1 T-cell receptor transgenic lymphocytes and dendritic cell vaccination in patients with metastatic melanoma. Clin Cancer Res 20(9):2457–2465 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Chow S, Galvis V, Pillai M, Leach R, Keene E, Spencer-Shaw A, Shablak A, Shanks J, Liptrot T, Thistlethwaite F, Hawkins RE. 2016. High-dose interleukin2—a 10-year single-site experience in the treatment of metastatic renal cell carcinoma: careful selection of patients gives an excellent outcome. J Immunother Cancer 4:67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Cohen PJ, Lotze MT, Roberts JR, Rosenberg SA, Jaffe ES. 1987. The immunopathology of sequential tumor biopsies in patients treated with interleukin-2. Correlation of response with T-cell infiltration and HLA-DR expression. Am J Pathol 129(2):208–216 [PMC free article] [PubMed] [Google Scholar]
  58. Cole DJ, Rubinstein MP. 2012. Soluble IL-15/IL-15Ralpha complexes in human serum. Blood 120(1):1–2 [DOI] [PubMed] [Google Scholar]
  59. Cole DJ, Weil DP, Shilyansky J, Custer M, Kawakami Y, Rosenberg SA, Nishimura MI. 1995. Characterization of the functional specificity of a cloned T-cell receptor heterodimer recognizing the MART-1 melanoma antigen. Cancer Res 55(4):748–752 [PubMed] [Google Scholar]
  60. Conlon KC, Lugli E, Welles HC, Rosenberg SA, Fojo AT, Morris JC, Fleisher TA, Dubois SP, Perera LP, Stewart DM, Goldman CK, Bryant BR, Decker JM, Chen J, Worthy TA, Figg WD, Sr., Peer CJ, Sneller MC, Lane HC, Yovandich JL, Creekmore SP, Roederer M, Waldmann TA. 2015. Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. J Clin Oncol 33(1):74–82 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Courtney LP, Phelps JL, Karavodin LM. 1994. An anti-IL-2 antibody increases serum half-life and improves anti-tumor efficacy of human recombinant interleukin-2. Immunopharmacology 28(3):223–232 [DOI] [PubMed] [Google Scholar]
  62. Cousens LP, Orange JS, Biron CA. 1995. Endogenous IL-2 contributes to T cell expansion and IFN-gamma production during lymphocytic choriomeningitis virus infection. J Immunol 155(12):5690–5699 [PubMed] [Google Scholar]
  63. Cui Y, Zhang H, Meadors J, Poon R, Guimond M, Mackall CL. 2009. Harnessing the physiology of lymphopenia to support adoptive immunotherapy in lymphoreplete hosts. Blood 114(18):3831–3840 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. D'Cruz LM, Rubinstein MP, Goldrath AW. 2009. Surviving the crash: transitioning from effector to memory CD8+ T cell. Semin Immunol 21(2):92–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Damia G, Komschlies KL, Faltynek CR, Ruscetti FW, Wiltrout RH. 1992. Administration of recombinant human interleukin-7 alters the frequency and number of myeloid progenitor cells in the bone marrow and spleen of mice. Blood 79(5):1121–1129 [PubMed] [Google Scholar]
  66. Debets R, Donnadieu E, Chouaib S, Coukos G. 2016. TCR-engineered T cells to treat tumors: seeing but not touching? Semin Immunol 28(1):10–21 [DOI] [PubMed] [Google Scholar]
  67. Desbois M, Le Vu P, Coutzac C, Marcheteau E, Beal C, Terme M, Gey A, Morisseau S, Teppaz G, Boselli L, Jacques Y, Bechard D, Tartour E, Cassard L, Chaput N. 2016. IL-15 trans-signaling with the superagonist RLI promotes effector/memory CD8+ T cell responses and enhances antitumor activity of PD-1 antagonists. J Immunol 197(1):168–178 [DOI] [PubMed] [Google Scholar]
  68. Devos R, Plaetinck G, Cheroutre H, Simons G, Degrave W, Tavernier J, Remaut E, Fiers W. 1983. Molecular cloning of human interleukin 2 cDNA and its expression in E. coli. Nucleic Acids Res 11(13):4307–4323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Di Carlo E, Comes A, Basso S, De Ambrosis A, Meazza R, Musiani P, Moelling K, Albini A, Ferrini S. 2000. The combined action of IL-15 and IL-12 gene transfer can induce tumor cell rejection without T and NK cell involvement. J Immunol 165(6):3111–3118 [DOI] [PubMed] [Google Scholar]
  70. Diller ML, Kudchadkar RR, Delman KA, Lawson DH, Ford ML. 2016. Complete response to high-dose IL-2 and enhanced IFNgamma+Th17: TREG ratio in a melanoma patient. Melanoma Res 26(5):535–539 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Donohue JH, Rosenstein M, Chang AE, Lotze MT, Robb RJ, Rosenberg SA. 1984. The systemic administration of purified interleukin 2 enhances the ability of sensitized murine lymphocytes to cure a disseminated syngeneic lymphoma. J Immunol 132(4):2123–2128 [PubMed] [Google Scholar]
  72. Dubois S, Conlon KC, Muller JR, Hsu-Albert J, Beltran N, Bryant BR, Waldmann TA. 2017. IL15 Infusion of cancer patients expands the subpopulation of cytotoxic CD56bright NK cells and increases NK-cell cytokine release capabilities. Cancer Immunol Res 5(10):929–938 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Dubois S, Mariner J, Waldmann TA, Tagaya Y. 2002. IL-15Ralpha recycles and presents IL-15 in trans to neighboring cells. Immunity 17(5):537–547 [DOI] [PubMed] [Google Scholar]
  74. Dubois S, Patel HJ, Zhang M, Waldmann TA, Muller JR. 2008. Preassociation of IL-15 with IL-15R alpha-IgG1-Fc enhances its activity on proliferation of NK and CD8+/CD44high T cells and its antitumor action. J Immunol 180(4):2099–2106 [DOI] [PubMed] [Google Scholar]
  75. Dudek AZ, Yee RT, Manivel JC, Isaksson R, Yee HO. 2010. Carbonic anhydrase IX expression is associated with improved outcome of high-dose interleukin-2 therapy for metastatic renal cell carcinoma. Anticancer Res 30(3):987–992 [PubMed] [Google Scholar]
  76. Dudley ME, Gross CA, Langhan MM, Garcia MR, Sherry RM, Yang JC, Phan GQ, Kammula US, Hughes MS, Citrin DE, Restifo NP, Wunderlich JR, Prieto PA, Hong JJ, Langan RC, Zlott DA, Morton KE, White DE, Laurencot CM, Rosenberg SA. 2010. CD8+ enriched “young” tumor infiltrating lymphocytes can mediate regression of metastatic melanoma. Clin Cancer Res 16(24):6122–6131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Dudley ME, Wunderlich JR, Robbins PF, Yang JC, Hwu P, Schwartzentruber DJ, Topalian SL, Sherry R, Restifo NP, Hubicki AM, Robinson MR, Raffeld M, Duray P, Seipp CA, Rogers-Freezer L, Morton KE, Mavroukakis SA, White DE, Rosenberg SA. 2002. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 298(5594):850–854 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Dudley ME, Yang JC, Sherry R, Hughes MS, Royal R, Kammula U, Robbins PF, Huang J, Citrin DE, Leitman SF, Wunderlich J, Restifo NP, Thomasian A, Downey SG, Smith FO, Klapper J, Morton K, Laurencot C, White DE, Rosenberg SA. 2008. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J Clin Oncol 26(32):5233–5239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Dumonde DC, Wolstencroft RA, Panayi GS, Matthew M, Morley J, Howson WT. 1969. “Lymphokines”: non-antibody mediators of cellular immunity generated by lymphocyte activation. Nature 224(5214):38–42 [DOI] [PubMed] [Google Scholar]
  80. Dutcher J, Atkins MB, Margolin K, Weiss G, Clark J, Sosman J, Logan T, Aronson F, Mier J, Cytokine Working G. 2001. Kidney cancer: the Cytokine Working Group experience (1986–2001): Part II. Management of IL-2 toxicity and studies with other cytokines. Med Oncol 18(3):209–219 [DOI] [PubMed] [Google Scholar]
  81. Eicher DM, Waldmann TA. 1998. IL-2R alpha on one cell can present IL-2 to IL-2R beta/gamma(c) on another cell to augment IL-2 signaling. J Immunol 161(10):5430–5437 [PubMed] [Google Scholar]
  82. Eisenbeis CF, Grainger A, Fischer B, Baiocchi RA, Carrodeguas L, Roychowdhury S, Chen L, Banks AL, Davis T, Young D, Kelbick N, Stephens J, Byrd JC, Grever MR, Caligiuri MA, Porcu P. 2004. Combination immunotherapy of B-cell non-Hodgkin's lymphoma with rituximab and interleukin-2: a preclinical and phase I study. Clin Cancer Res 10(18 Pt 1):6101–6110 [DOI] [PubMed] [Google Scholar]
  83. Epardaud M, Elpek KG, Rubinstein MP, Yonekura AR, Bellemare-Pelletier A, Bronson R, Hamerman JA, Goldrath AW, Turley SJ. 2008. Interleukin-15/interleukin-15R alpha complexes promote destruction of established tumors by reviving tumor-resident CD8+ T cells. Cancer Res 68(8):2972–2983 [DOI] [PubMed] [Google Scholar]
  84. Evans R, Fuller JA, Christianson G, Krupke DM, Troutt AB. 1997. IL-15 mediates anti-tumor effects after cyclophosphamide injection of tumor-bearing mice and enhances adoptive immunotherapy: the potential role of NK cell subpopulations. Cell Immunol 179(1):66–73 [DOI] [PubMed] [Google Scholar]
  85. Fallon EM, Liparoto SF, Lee KJ, Ciardelli TL, Lauffenburger DA. 2000. Increased endosomal sorting of ligand to recycling enhances potency of an interleukin-2 analog. J Biol Chem 275(10):6790–6797 [DOI] [PubMed] [Google Scholar]
  86. Farrar JJ, Simon PL, Koopman WJ, Fuller-Bonar J. 1978. Biochemical relationship of thymocyte mitogenic factor and factors enhancing humoral and cell-mediated immune responses. J Immunol 121(4):1353–1360 [PubMed] [Google Scholar]
  87. Fehniger TA, Caligiuri MA. 2001. Interleukin 15: biology and relevance to human disease. Blood 97(1):14–32 [DOI] [PubMed] [Google Scholar]
  88. Fehniger TA, Cooper MA, Caligiuri MA. 2002. Interleukin-2 and interleukin-15: immunotherapy for cancer. Cytokine Growth Factor Rev 13(2):169–183 [DOI] [PubMed] [Google Scholar]
  89. Finch DK, Midha A, Buchanan CL, Cochrane D, Craggs RI, Cruwys S, Grahames C, Kolbeck R, Lowe DC, Maltby J, Pattison DV, Vousden KA, Ward A, Sleeman MA, Mallinder PR. 2011. Identification of a potent anti-IL-15 antibody with opposing mechanisms of action in vitro and in vivo. Br J Pharmacol 162(2):480–490 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Finkelman FD, Madden KB, Morris SC, Holmes JM, Boiani N, Katona IM, Maliszewski CR. 1993. Anti-cytokine antibodies as carrier proteins. Prolongation of in vivo effects of exogenous cytokines by injection of cytokine-anti-cytokine antibody complexes. J Immunol 151(3):1235–1244 [PubMed] [Google Scholar]
  91. Fisher AG, Burdet C, Bunce C, Merkenschlager M, Ceredig R. 1995. Lymphoproliferative disorders in IL-7 transgenic mice: expansion of immature B cells which retain macrophage potential. Int Immunol 7(3):415–423 [DOI] [PubMed] [Google Scholar]
  92. Foureau DM, Amin A, White RL, Anderson W, Jones CP, Sarantou T, McKillop IH, Salo JC. 2014. Sequential immune monitoring in patients with melanoma and renal cell carcinoma treated with high-dose interleukin-2: immune patterns and correlation with outcome. Cancer Immunol Immunother 63(12):1329–1340 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Foxwell BM, Taylor-Fishwick DA, Simon JL, Page TH, Londei M. 1992. Activation induced changes in expression and structure of the IL-7 receptor on human T cells. Int Immunol 4(2):277–282 [DOI] [PubMed] [Google Scholar]
  94. Fry TJ, Connick E, Falloon J, Lederman MM, Liewehr DJ, Spritzler J, Steinberg SM, Wood LV, Yarchoan R, Zuckerman J, Landay A, Mackall CL. 2001. A potential role for interleukin-7 in T-cell homeostasis. Blood 97(10):2983–2990 [DOI] [PubMed] [Google Scholar]
  95. Fry TJ, Mackall CL. 2002. Interleukin-7: from bench to clinic. Blood 99(11):3892–3904 [DOI] [PubMed] [Google Scholar]
  96. Fukao T, Koyasu S. 2000. Expression of functional IL-2 receptors on mature splenic dendritic cells. Eur J Immunol 30(5):1453–1457 [DOI] [PubMed] [Google Scholar]
  97. Fyfe G, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie AC. 1995. Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J Clin Oncol 13(3):688–696 [DOI] [PubMed] [Google Scholar]
  98. Garfall AL, Maus MV, Hwang WT, Lacey SF, Mahnke YD, Melenhorst JJ, Zheng Z, Vogl DT, Cohen AD, Weiss BM, Dengel K, Kerr ND, Bagg A, Levine BL, June CH, Stadtmauer EA. 2015. Chimeric antigen receptor T cells against CD19 for multiple myeloma. N Engl J Med 373(11):1040–1047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Gasnault J, de Goer de Herve MG, Michot JM, Hendel-Chavez H, Seta V, Mazet AA, Croughs T, Stankoff B, Bourhis JH, Lambotte O, Delfraissy JF, Taoufik Y. 2014. Efficacy of recombinant human interleukin 7 in a patient with severe lymphopenia-related progressive multifocal leukoencephalopathy. Open Forum Infect Dis 1(2):ofu074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Gattinoni L, Finkelstein SE, Klebanoff CA, Antony PA, Palmer DC, Spiess PJ, Hwang LN, Yu Z, Wrzesinski C, Heimann DM, Surh CD, Rosenberg SA, Restifo NP. 2005. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med 202(7):907–912 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Gattinoni L, Powell DJ, Jr., Rosenberg SA, Restifo NP. 2006. Adoptive immunotherapy for cancer: building on success. Nat Rev Immunol 6(5):383–393 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Gibney GT, Weiner LM, Atkins MB. 2016. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol 17(12):e542–e551 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Gillis S, Ferm MM, Ou W, Smith KA. 1978. T cell growth factor: parameters of production and a quantitative microassay for activity. J Immunol 120(6):2027–2032 [PubMed] [Google Scholar]
  104. Gillis S, Smith KA. 1977. Long term culture of tumour-specific cytotoxic T cells. Nature 268(5616):154–156 [DOI] [PubMed] [Google Scholar]
  105. Gillis S, Watson J. 1980. Biochemical and biological characterization of lymphocyte regulatory molecules. V. Identification of an interleukin 2-producing human leukemia T cell line. J Exp Med 152(6):1709–1719 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Giri JG, Anderson DM, Kumaki S, Park LS, Grabstein KH, Cosman D. 1995. IL-15, a novel T cell growth factor that shares activities and receptor components with IL-2. J Leukoc Biol 57(5):763–766 [DOI] [PubMed] [Google Scholar]
  107. Glynn JP, Halpern BL, Fefer A. 1969. An immunochemotherapeutic system for the treatment of a transplanted Moloney virus-induced lymphoma in mice. Cancer Res 29(3):515–520 [PubMed] [Google Scholar]
  108. Goff SL, Dudley ME, Citrin DE, Somerville RP, Wunderlich JR, Danforth DN, Zlott DA, Yang JC, Sherry RM, Kammula US, Klebanoff CA, Hughes MS, Restifo NP, Langhan MM, Shelton TE, Lu L, Kwong ML, Ilyas S, Klemen ND, Payabyab EC, Morton KE, Toomey MA, Steinberg SM, White DE, Rosenberg SA. 2016. Randomized, prospective evaluation comparing intensity of lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for patients with metastatic melanoma. J Clin Oncol 34(20):2389–2397 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Goldrath AW, Sivakumar PV, Glaccum M, Kennedy MK, Bevan MJ, Benoist C, Mathis D, Butz EA. 2002. Cytokine requirements for acute and Basal homeostatic proliferation of naive and memory CD8+ T cells. J Exp Med 195(12):1515–1522 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Gomes-Giacoia E, Miyake M, Goodison S, Sriharan A, Zhang G, You L, Egan JO, Rhode PR, Parker AS, Chai KX, Wong HC, Rosser CJ. 2014. Intravesical ALT-803 and BCG treatment reduces tumor burden in a carcinogen induced bladder cancer rat model; a role for cytokine production and NK cell expansion. PLoS One 9(6):e96705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Gooding R, Riches P, Dadian G, Moore J, Gore M. 1995. Increased soluble interleukin-2 receptor concentration in plasma predicts a decreased cellular response to IL-2. Br J Cancer 72(2):452–455 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Goodwin RG, Friend D, Ziegler SF, Jerzy R, Falk BA, Gimpel S, Cosman D, Dower SK, March CJ, Namen AE, et al. . 1990. Cloning of the human and murine interleukin-7 receptors: demonstration of a soluble form and homology to a new receptor superfamily. Cell 60(6):941–951 [DOI] [PubMed] [Google Scholar]
  113. Goodwin RG, Lupton S, Schmierer A, Hjerrild KJ, Jerzy R, Clevenger W, Gillis S, Cosman D, Namen AE. 1989. Human interleukin 7: molecular cloning and growth factor activity on human and murine B-lineage cells. Proc Natl Acad Sci U S A 86(1):302–306 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Gordon J, MacLean LD. 1965. A lymphocyte-stimulating factor produced in vitro. Nature 208(5012):795–796 [DOI] [PubMed] [Google Scholar]
  115. Grabstein KH, Eisenman J, Shanebeck K, Rauch C, Srinivasan S, Fung V, Beers C, Richardson J, Schoenborn MA, Ahdieh M, et al. . 1994. Cloning of a T cell growth factor that interacts with the beta chain of the interleukin-2 receptor. Science 264(5161):965–968 [DOI] [PubMed] [Google Scholar]
  116. Greenberg PD, Kern DE, Cheever MA. 1985. Therapy of disseminated murine leukemia with cyclophosphamide and immune Lyt-1+,2- T cells. Tumor eradication does not require participation of cytotoxic T cells. J Exp Med 161(5):1122–1134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Greenberg PD, Klarnet JP, Kern DE, Cheever MA. 1988. Therapy of disseminated tumors by adoptive transfer of specifically immune T cells. Prog Exp Tumor Res 32:104–127 [DOI] [PubMed] [Google Scholar]
  118. Griffith KD, Read EJ, Carrasquillo JA, Carter CS, Yang JC, Fisher B, Aebersold P, Packard BS, Yu MY, Rosenberg SA. 1989. In vivo distribution of adoptively transferred indium-111-labeled tumor infiltrating lymphocytes and peripheral blood lymphocytes in patients with metastatic melanoma. J Natl Cancer Inst 81(22):1709–1717 [DOI] [PubMed] [Google Scholar]
  119. Grimm EA, Mazumder A, Zhang HZ, Rosenberg SA. 1982. Lymphokine-activated killer cell phenomenon. Lysis of natural killer-resistant fresh solid tumor cells by interleukin 2-activated autologous human peripheral blood lymphocytes. J Exp Med 155(6):1823–1841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Gross G, Gorochov G, Waks T, Eshhar Z. 1989. Generation of effector T cells expressing chimeric T cell receptor with antibody type-specificity. Transplant Proc 21(1 Pt 1):127–130 [PubMed] [Google Scholar]
  121. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, Teachey DT, Chew A, Hauck B, Wright JF, Milone MC, Levine BL, June CH. 2013. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 368(16):1509–1518 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Guimond M, Veenstra RG, Grindler DJ, Zhang H, Cui Y, Murphy RD, Kim SY, Na R, Hennighausen L, Kurtulus S, Erman B, Matzinger P, Merchant MS, Mackall CL. 2009. Interleukin 7 signaling in dendritic cells regulates the homeostatic proliferation and niche size of CD4+ T cells. Nat Immunol 10(2):149–157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Gullberg M, Smith KA. 1986. Regulation of T cell autocrine growth. T4+ cells become refractory to interleukin 2. J Exp Med 163(2):270–284 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Habibi M, Kmieciak M, Graham L, Morales JK, Bear HD, Manjili MH. 2009. Radiofrequency thermal ablation of breast tumors combined with intralesional administration of IL-7 and IL-15 augments anti-tumor immune responses and inhibits tumor development and metastasis. Breast Cancer Res Treat 114(3):423–431 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, Dronca R, Gangadhar TC, Patnaik A, Zarour H, Joshua AM, Gergich K, Elassaiss-Schaap J, Algazi A, Mateus C, Boasberg P, Tumeh PC, Chmielowski B, Ebbinghaus SW, Li XN, Kang SP, Ribas A. 2013. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 369(2):134–144 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Hamilton SE, Schenkel JM, Akue AD, Jameson SC. 2010. IL-2 complex treatment can protect naive mice from bacterial and viral infection. J Immunol 185(11):6584–6590 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Han KP, Zhu X, Liu B, Jeng E, Kong L, Yovandich JL, Vyas VV, Marcus WD, Chavaillaz PA, Romero CA, Rhode PR, Wong HC. 2011. IL-15:IL-15 receptor alpha superagonist complex: high-level co-expression in recombinant mammalian cells, purification and characterization. Cytokine 56(3):804–810 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Hannani D, Vetizou M, Enot D, Rusakiewicz S, Chaput N, Klatzmann D, Desbois M, Jacquelot N, Vimond N, Chouaib S, Mateus C, Allison JP, Ribas A, Wolchok JD, Yuan J, Wong P, Postow M, Mackiewicz A, Mackiewicz J, Schadendorff D, Jaeger D, Zornig I, Hassel J, Korman AJ, Bahjat K, Maio M, Calabro L, Teng MW, Smyth MJ, Eggermont A, Robert C, Kroemer G, Zitvogel L. 2015. Anticancer immunotherapy by CTLA-4 blockade: obligatory contribution of IL-2 receptors and negative prognostic impact of soluble CD25. Cell Res 25(2):208–224 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. He J, Zhang X, Wei Y, Sun X, Chen Y, Deng J, Jin Y, Gan Y, Hu X, Jia R, Xu C, Hou Z, Leong YA, Zhu L, Feng J, An Y, Jia Y, Li C, Liu X, Ye H, Ren L, Li R, Yao H, Li Y, Chen S, Zhang X, Su Y, Guo J, Shen N, Morand EF, Yu D, Li Z. 2016. Low-dose interleukin-2 treatment selectively modulates CD4+ T cell subsets in patients with systemic lupus erythematosus. Nat Med 22(9):991–993 [DOI] [PubMed] [Google Scholar]
  130. Heaton KM, Ju G, Grimm EA. 1993. Human interleukin 2 analogues that preferentially bind the intermediate-affinity interleukin 2 receptor lead to reduced secondary cytokine secretion: implications for the use of these interleukin 2 analogues in cancer immunotherapy. Cancer Res 53(11):2597–2602 [PubMed] [Google Scholar]
  131. Hill GR, Crawford JM, Cooke KR, Brinson YS, Pan L, Ferrara JL. 1997. Total body irradiation and acute graft-versus-host disease: the role of gastrointestinal damage and inflammatory cytokines. Blood 90(8):3204–3213 [PubMed] [Google Scholar]
  132. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. 2010. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363(8):711–723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Hofmeister R, Khaled AR, Benbernou N, Rajnavolgyi E, Muegge K, Durum SK. 1999. Interleukin-7: physiological roles and mechanisms of action. Cytokine Growth Factor Rev 10(1):41–60 [DOI] [PubMed] [Google Scholar]
  134. Jensen MC, Riddell SR. 2014. Design and implementation of adoptive therapy with chimeric antigen receptor-modified T cells. Immunol Rev 257(1):127–144 [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Jiang Q, Li WQ, Aiello FB, Mazzucchelli R, Asefa B, Khaled AR, Durum SK. 2005. Cell biology of IL-7, a key lymphotrophin. Cytokine Growth Factor Rev 16(4–5):513–533 [DOI] [PubMed] [Google Scholar]
  136. Jin GH, Hirano T, Murakami M. 2008. Combination treatment with IL-2 and anti-IL-2 mAbs reduces tumor metastasis via NK cell activation. Int Immunol 20(6):783–789 [DOI] [PubMed] [Google Scholar]
  137. John LB, Devaud C, Duong CP, Yong CS, Beavis PA, Haynes NM, Chow MT, Smyth MJ, Kershaw MH, Darcy PK. 2013. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin Cancer Res 19(20):5636–5646 [DOI] [PubMed] [Google Scholar]
  138. Johnson CB, Riesenberg BP, May BR, Gilreath SC, Li G, Staveley-O'Carroll KF, Garrett-Mayer E, Mehrotra S, Cole DJ, Rubinstein MP. 2015. Effector CD8+ T-cell engraftment and antitumor immunity in lymphodepleted hosts is IL7Ralpha dependent. Cancer Immunol Res 3(12):1364–1374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, Kammula US, Royal RE, Sherry RM, Wunderlich JR, Lee CC, Restifo NP, Schwarz SL, Cogdill AP, Bishop RJ, Kim H, Brewer CC, Rudy SF, VanWaes C, Davis JL, Mathur A, Ripley RT, Nathan DA, Laurencot CM, Rosenberg SA. 2009. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 114(3):535–546 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. June CH, Maus MV, Plesa G, Johnson LA, Zhao Y, Levine BL, Grupp SA, Porter DL. 2014. Engineered T cells for cancer therapy. Cancer Immunol Immunother 63(9):969–975 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Kaech SM, Tan JT, Wherry EJ, Konieczny BT, Surh CD, Ahmed R. 2003. Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells. Nat Immunol 4(12):1191–1198 [DOI] [PubMed] [Google Scholar]
  142. Kageyama S, Ikeda H, Miyahara Y, Imai N, Ishihara M, Saito K, Sugino S, Ueda S, Ishikawa T, Kokura S, Naota H, Ohishi K, Shiraishi T, Inoue N, Tanabe M, Kidokoro T, Yoshioka H, Tomura D, Nukaya I, Mineno J, Takesako K, Katayama N, Shiku H. 2015. Adoptive transfer of MAGE-A4 T-cell receptor gene-transduced lymphocytes in patients with recurrent esophageal cancer. Clin Cancer Res 21(10):2268–2277 [DOI] [PubMed] [Google Scholar]
  143. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH. 2011. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med 3(95):95ra73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Kammula US, White DE, Rosenberg SA. 1998. Trends in the safety of high dose bolus interleukin-2 administration in patients with metastatic cancer. Cancer 83(4):797–805 [PubMed] [Google Scholar]
  145. Kaplan BL, Yu DC, Clay TM, Nishimura MI. 2003. Redirecting T lymphocyte specificity using T cell receptor genes. Int Rev Immunol 22(3–4):229–253 [DOI] [PubMed] [Google Scholar]
  146. Kasakura S, Lowenstein L. 1965. A factor stimulating DNA synthesis derived from the medium of leukocyte cultures. Nature 208(5012):794–795 [DOI] [PubMed] [Google Scholar]
  147. Kashima N, Nishi-Takaoka C, Fujita T, Taki S, Yamada G, Hamuro J, Taniguchi T. 1985. Unique structure of murine interleukin-2 as deduced from cloned cDNAs. Nature 313(6001):402–404 [DOI] [PubMed] [Google Scholar]
  148. Katre NV. 1990. Immunogenicity of recombinant IL-2 modified by covalent attachment of polyethylene glycol. J Immunol 144(1):209–213 [PubMed] [Google Scholar]
  149. Katre NV, Knauf MJ, Laird WJ. 1987. Chemical modification of recombinant interleukin 2 by polyethylene glycol increases its potency in the murine Meth A sarcoma model. Proc Natl Acad Sci U S A 84(6):1487–1491 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Katsanis E, Xu Z, Panoskaltsis-Mortari A, Weisdorf DJ, Widmer MB, Blazar BR. 1996. IL-15 administration following syngeneic bone marrow transplantation prolongs survival of lymphoma bearing mice. Transplantation 62(6):872–875 [DOI] [PubMed] [Google Scholar]
  151. Kebriaei P, Singh H, Huls MH, Figliola MJ, Bassett R, Olivares S, Jena B, Dawson MJ, Kumaresan PR, Su S, Maiti S, Dai J, Moriarity B, Forget MA, Senyukov V, Orozco A, Liu T, McCarty J, Jackson RN, Moyes JS, Rondon G, Qazilbash M, Ciurea S, Alousi A, Nieto Y, Rezvani K, Marin D, Popat U, Hosing C, Shpall EJ, Kantarjian H, Keating M, Wierda W, Do KA, Largaespada DA, Lee DA, Hackett PB, Champlin RE, Cooper LJ. 2016. Phase I trials using Sleeping Beauty to generate CD19-specific CAR T cells. J Clin Invest 126(9):3363–3376 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Kenderian SS, Ruella M, Gill S, Kalos M. 2014. Chimeric antigen receptor T-cell therapy to target hematologic malignancies. Cancer Res 74(22):6383–6389 [DOI] [PubMed] [Google Scholar]
  153. Kennedy MK, Glaccum M, Brown SN, Butz EA, Viney JL, Embers M, Matsuki N, Charrier K, Sedger L, Willis CR, Brasel K, Morrissey PJ, Stocking K, Schuh JC, Joyce S, Peschon JJ. 2000. Reversible defects in natural killer and memory CD8 T cell lineages in interleukin 15-deficient mice. J Exp Med 191(5):771–780 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, White DE, Wunderlich JR, Canevari S, Rogers-Freezer L, Chen CC, Yang JC, Rosenberg SA, Hwu P. 2006. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res 12(20 Pt 1):6106–6115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Kessels HW, Wolkers MC, van den Boom MD, van der Valk MA, Schumacher TN. 2001. Immunotherapy through TCR gene transfer. Nat Immunol 2(10):957–961 [DOI] [PubMed] [Google Scholar]
  156. Khan KD, Emmanouilides C, Benson DM, Jr., Hurst D, Garcia P, Michelson G, Milan S, Ferketich AK, Piro L, Leonard JP, Porcu P, Eisenbeis CF, Banks AL, Chen L, Byrd JC, Caligiuri MA. 2006. A phase 2 study of rituximab in combination with recombinant interleukin-2 for rituximab-refractory indolent non-Hodgkin's lymphoma. Clin Cancer Res 12(23):7046–7053 [DOI] [PubMed] [Google Scholar]
  157. Khawam K, Giron-Michel J, Gu Y, Perier A, Giuliani M, Caignard A, Devocelle A, Ferrini S, Fabbi M, Charpentier B, Ludwig A, Chouaib S, Azzarone B, Eid P. 2009. Human renal cancer cells express a novel membrane-bound interleukin-15 that induces, in response to the soluble interleukin-15 receptor alpha chain, epithelial-to-mesenchymal transition. Cancer Res 69(4):1561–1569 [DOI] [PubMed] [Google Scholar]
  158. Kim MG, Koo TY, Yan JJ, Lee E, Han KH, Jeong JC, Ro H, Kim BS, Jo SK, Oh KH, Surh CD, Ahn C, Yang J. 2013. IL-2/anti-IL-2 complex attenuates renal ischemia-reperfusion injury through expansion of regulatory T cells. J Am Soc Nephrol 24(10):1529–1536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Kim MT, Richer MJ, Gross BP, Norian LA, Badovinac VP, Harty JT. 2015. Enhancing dendritic cell-based immunotherapy with IL-2/Monoclonal antibody complexes for control of established tumors. J Immunol 195(9):4537–4544 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Kishida T, Asada H, Itokawa Y, Cui FD, Shin-Ya M, Gojo S, Yasutomi K, Ueda Y, Yamagishi H, Imanishi J, Mazda O. 2003. Interleukin (IL)-21 and IL-15 genetic transfer synergistically augments therapeutic antitumor immunity and promotes regression of metastatic lymphoma. Mol Ther 8(4):552–558 [DOI] [PubMed] [Google Scholar]
  161. Klebanoff CA, Finkelstein SE, Surman DR, Lichtman MK, Gattinoni L, Theoret MR, Grewal N, Spiess PJ, Antony PA, Palmer DC, Tagaya Y, Rosenberg SA, Waldmann TA, Restifo NP. 2004. IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc Natl Acad Sci U S A 101(7):1969–1974 [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Klebanoff CA, Khong HT, Antony PA, Palmer DC, Restifo NP. 2005. Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol 26(2):111–117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Klonowski KD, Williams KJ, Marzo AL, Lefrancois L. 2006. Cutting edge: IL-7-independent regulation of IL-7 receptor alpha expression and memory CD8 T cell development. J Immunol 177(7):4247–4251 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Knauf MJ, Bell DP, Hirtzer P, Luo ZP, Young JD, Katre NV. 1988. Relationship of effective molecular size to systemic clearance in rats of recombinant interleukin-2 chemically modified with water-soluble polymers. J Biol Chem 263(29):15064–15070 [PubMed] [Google Scholar]
  165. Kobayashi H, Carrasquillo JA, Paik CH, Waldmann TA, Tagaya Y. 2000. Differences of biodistribution, pharmacokinetics, and tumor targeting between interleukins 2 and 15. Cancer Res 60(13):3577–3583 [PubMed] [Google Scholar]
  166. Kobayashi H, Dubois S, Sato N, Sabzevari H, Sakai Y, Waldmann TA, Tagaya Y. 2005. Role of trans-cellular IL-15 presentation in the activation of NK cell-mediated killing, which leads to enhanced tumor immunosurveillance. Blood 105(2):721–727 [DOI] [PubMed] [Google Scholar]
  167. Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, Stetler-Stevenson M, Phan GQ, Hughes MS, Sherry RM, Yang JC, Kammula US, Devillier L, Carpenter R, Nathan DA, Morgan RA, Laurencot C, Rosenberg SA. 2012. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 119(12):2709–2720 [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Kochenderfer JN, Dudley ME, Kassim SH, Somerville RP, Carpenter RO, Stetler-Stevenson M, Yang JC, Phan GQ, Hughes MS, Sherry RM, Raffeld M, Feldman S, Lu L, Li YF, Ngo LT, Goy A, Feldman T, Spaner DE, Wang ML, Chen CC, Kranick SM, Nath A, Nathan DA, Morton KE, Toomey MA, Rosenberg SA. 2015. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol 33(6):540–549 [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Kochenderfer JN, Rosenberg SA. 2013. Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors. Nat Rev Clin Oncol 10(5):267–276 [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Kochenderfer JN, Somerville RPT, Lu T, Shi V, Bot A, Rossi J, Xue A, Goff SL, Yang JC, Sherry RM, Klebanoff CA, Kammula US, Sherman M, Perez A, Yuan CM, Feldman T, Friedberg JW, Roschewski MJ, Feldman SA, McIntyre L, Toomey MA, Rosenberg SA. 2017. Lymphoma remissions caused by anti-CD19 chimeric antigen receptor T cells are associated with high serum interleukin-15 levels. J Clin Oncol 35(16):1803–1813 [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA, Maric I, Raffeld M, Nathan DA, Lanier BJ, Morgan RA, Rosenberg SA. 2010a. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 116(20):4099–4102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Kochenderfer JN, Yu Z, Frasheri D, Restifo NP, Rosenberg SA. 2010b. Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. Blood 116(19):3875–3886 [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Koka R, Burkett P, Chien M, Chai S, Boone DL, Ma A. 2004. Cutting edge: murine dendritic cells require IL-15R alpha to prime NK cells. J Immunol 173(6):3594–3598 [DOI] [PubMed] [Google Scholar]
  174. Komschlies KL, Gregorio TA, Gruys ME, Back TC, Faltynek CR, Wiltrout RH. 1994. Administration of recombinant human IL-7 to mice alters the composition of B-lineage cells and T cell subsets, enhances T cell function, and induces regression of established metastases. J Immunol 152(12):5776–5784 [PubMed] [Google Scholar]
  175. Koreth J, Matsuoka K, Kim HT, McDonough SM, Bindra B, Alyea EP, 3rd, Armand P, Cutler C, Ho VT, Treister NS, Bienfang DC, Prasad S, Tzachanis D, Joyce RM, Avigan DE, Antin JH, Ritz J, Soiffer RJ. 2011. Interleukin-2 and regulatory T cells in graft-versus-host disease. N Engl J Med 365(22):2055–2066 [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Kostner AH, Ellegaard MB, Christensen IJ, Bastholt L, Schmidt H. 2015. Fever and the use of paracetamol during IL-2-based immunotherapy in metastatic melanoma. Cancer Immunol Immunother 64(3):349–355 [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Kovanen PE, Leonard WJ. 2004. Cytokines and immunodeficiency diseases: critical roles of the gamma(c)-dependent cytokines interleukins 2, 4, 7, 9, 15, and 21, and their signaling pathways. Immunol Rev 202:67–83 [DOI] [PubMed] [Google Scholar]
  178. Kradin R, Dubinett S, Mullin J, Boyle L, Strauss HW, Bourgoin PM, Preffer FI, Kurnick J. 1988. Treatment of patients with advanced cancer using tumor-infiltrating lymphocytes and interleukin 2. Transplant Proc 20(2):336–338 [PubMed] [Google Scholar]
  179. Kradin RL, Boyle LA, Preffer FI, Callahan RJ, Barlai-Kovach M, Strauss HW, Dubinett S, Kurnick JT. 1987. Tumor-derived interleukin-2-dependent lymphocytes in adoptive immunotherapy of lung cancer. Cancer Immunol Immunother 24(1):76–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Kradin RL, Kurnick JT, Lazarus DS, Preffer FI, Dubinett SM, Pinto CE, Gifford J, Davidson E, Grove B, Callahan RJ, et al. . 1989. Tumour-infiltrating lymphocytes and interleukin-2 in treatment of advanced cancer. Lancet 1(8638):577–580 [DOI] [PubMed] [Google Scholar]
  181. Krieg C, Letourneau S, Pantaleo G, Boyman O. 2010. Improved IL-2 immunotherapy by selective stimulation of IL-2 receptors on lymphocytes and endothelial cells. Proc Natl Acad Sci U S A 107(26):11906–11911 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Kronin V, Vremec D, Shortman K. 1998. Does the IL-2 receptor alpha chain induced on dendritic cells have a biological function? Int Immunol 10(2):237–240 [DOI] [PubMed] [Google Scholar]
  183. Kurnick JT, Kradin RL, Blumberg R, Schneeberger EE, Boyle LA. 1986. Functional characterization of T lymphocytes propagated from human lung carcinomas. Clin Immunol Immunopathol 38(3):367–380 [DOI] [PubMed] [Google Scholar]
  184. Kuzman JA, Stenehjem DD, Merriman J, Agarwal AM, Patel SB, Hahn AW, Alex A, Albertson D, Gill DM, Agarwal N. 2017. Neutrophil-lymphocyte ratio as a predictive biomarker for response to high dose interleukin-2 in patients with renal cell carcinoma. BMC Urol 17(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Lafreniere R, Rosenberg SA. 1985. Successful immunotherapy of murine experimental hepatic metastases with lymphokine-activated killer cells and recombinant interleukin 2. Cancer Res 45(8):3735–3741 [PubMed] [Google Scholar]
  186. Lamers CH, Sleijfer S, Vulto AG, Kruit WH, Kliffen M, Debets R, Gratama JW, Stoter G, Oosterwijk E. 2006. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol 24(13):e20–e22 [DOI] [PubMed] [Google Scholar]
  187. Lasek W, Basak G, Switaj T, Jakubowska AB, Wysocki PJ, Mackiewicz A, Drela N, Jalili A, Kaminski R, Kozar K, Jakobisiak M. 2004. Complete tumour regressions induced by vaccination with IL-12 gene-transduced tumour cells in combination with IL-15 in a melanoma model in mice. Cancer Immunol Immunother 53(4):363–372 [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, Fry TJ, Orentas R, Sabatino M, Shah NN, Steinberg SM, Stroncek D, Tschernia N, Yuan C, Zhang H, Zhang L, Rosenberg SA, Wayne AS, Mackall CL. 2015. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 385(9967):517–528 [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Lee SY, Cho ML, Oh HJ, Ryu JG, Park MJ, Jhun JY, Park MK, Stone JC, Ju JH, Hwang SY, Park SH, Surh CD, Kim HY. 2012. Interleukin-2/anti-interleukin-2 monoclonal antibody immune complex suppresses collagen-induced arthritis in mice by fortifying interleukin-2/STAT5 signalling pathways. Immunology 137(4):305–316 [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Leibovich BC, Han KR, Bui MH, Pantuck AJ, Dorey FJ, Figlin RA, Belldegrun A. 2003. Scoring algorithm to predict survival after nephrectomy and immunotherapy in patients with metastatic renal cell carcinoma: a stratification tool for prospective clinical trials. Cancer 98(12):2566–2575 [DOI] [PubMed] [Google Scholar]
  191. Leonard WJ, Kronke M, Peffer NJ, Depper JM, Greene WC. 1985. Interleukin 2 receptor gene expression in normal human T lymphocytes. Proc Natl Acad Sci U S A 82(18):6281–6285 [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Levin AM, Bates DL, Ring AM, Krieg C, Lin JT, Su L, Moraga I, Raeber ME, Bowman GR, Novick P, Pande VS, Fathman CG, Boyman O, Garcia KC. 2012. Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine’. Nature 484(7395):529–533 [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Levin SD, Koelling RM, Friend SL, Isaksen DE, Ziegler SF, Perlmutter RM, Farr AG. 1999. Thymic stromal lymphopoietin: a cytokine that promotes the development of IgM+ B cells in vitro and signals via a novel mechanism. J Immunol 162(2):677–683 [PubMed] [Google Scholar]
  194. Levy Y, Lacabaratz C, Weiss L, Viard JP, Goujard C, Lelievre JD, Boue F, Molina JM, Rouzioux C, Avettand-Fenoel V, Croughs T, Beq S, Thiebaut R, Chene G, Morre M, Delfraissy JF. 2009. Enhanced T cell recovery in HIV-1-infected adults through IL-7 treatment. J Clin Invest 119(4):997–1007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Levy Y, Sereti I, Tambussi G, Routy JP, Lelievre JD, Delfraissy JF, Molina JM, Fischl M, Goujard C, Rodriguez B, Rouzioux C, Avettand-Fenoel V, Croughs T, Beq S, Morre M, Poulin JF, Sekaly RP, Thiebaut R, Lederman MM. 2012. Effects of recombinant human interleukin 7 on T-cell recovery and thymic output in HIV-infected patients receiving antiretroviral therapy: results of a phase I/IIa randomized, placebo-controlled, multicenter study. Clin Infect Dis 55(2):291–300 [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Li J, Lu E, Yi T, Cyster JG. 2016. EBI2 augments Tfh cell fate by promoting interaction with IL-2-quenching dendritic cells. Nature 533(7601):110–114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Liao W, Lin JX, Leonard WJ. 2013. Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. Immunity 38(1):13–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Lim WA, June CH. 2017. The principles of engineering immune cells to treat cancer. Cell 168(4):724–740 [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Lin J, Zhu Z, Xiao H, Wakefield MR, Ding VA, Bai Q, Fang Y. 2017. The role of IL-7 in Immunity and Cancer. Anticancer Res 37(3):963–967 [DOI] [PubMed] [Google Scholar]
  200. Liu DV, Maier LM, Hafler DA, Wittrup KD. 2009. Engineered interleukin-2 antagonists for the inhibition of regulatory T cells. J Immunother 32(9):887–894 [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Liu R, Zhou Q, La Cava A, Campagnolo DI, Van Kaer L, Shi FD. 2010. Expansion of regulatory T cells via IL-2/anti-IL-2 mAb complexes suppresses experimental myasthenia. Eur J Immunol 40(6):1577–1589 [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Liu RB, Engels B, Schreiber K, Ciszewski C, Schietinger A, Schreiber H, Jabri B. 2013. IL-15 in tumor microenvironment causes rejection of large established tumors by T cells in a noncognate T cell receptor-dependent manner. Proc Natl Acad Sci U S A 110(20):8158–8163 [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Lo Kuan E, Ziegler SF. 2014. Thymic stromal lymphopoietin and cancer. J Immunol 193(9):4283–4288 [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Locke FL, Neelapu SS, Bartlett NL, Siddiqi T, Chavez JC, Hosing CM, Ghobadi A, Budde LE, Bot A, Rossi JM, Jiang Y, Xue AX, Elias M, Aycock J, Wiezorek J, Go WY. 2017. Phase 1 results of ZUMA-1: a multicenter study of KTE-C19 anti-CD19 CAR T cell therapy in refractory aggressive lymphoma. Mol Ther 25(1):285–295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Lodolce JP, Boone DL, Chai S, Swain RE, Dassopoulos T, Trettin S, Ma A. 1998. IL-15 receptor maintains lymphoid homeostasis by supporting lymphocyte homing and proliferation. Immunity 9(5):669–676 [DOI] [PubMed] [Google Scholar]
  206. Lotze MT, Chang AE, Seipp CA, Simpson C, Vetto JT, Rosenberg SA. 1986a. High-dose recombinant interleukin 2 in the treatment of patients with disseminated cancer. Responses, treatment-related morbidity, and histologic findings. JAMA 256(22):3117–3124 [PubMed] [Google Scholar]
  207. Lotze MT, Grimm EA, Mazumder A, Strausser JL, Rosenberg SA. 1981. Lysis of fresh and cultured autologous tumor by human lymphocytes cultured in T-cell growth factor. Cancer Res 41(11 Pt 1):4420–4425 [PubMed] [Google Scholar]
  208. Lotze MT, Line BR, Mathisen DJ, Rosenberg SA. 1980. The in vivo distribution of autologous human and murine lymphoid cells grown in T cell growth factor (TCGF): implications for the adoptive immunotherapy of tumors. J Immunol 125(4):1487–1493 [PubMed] [Google Scholar]
  209. Lotze MT, Matory YL, Ettinghausen SE, Rayner AA, Sharrow SO, Seipp CA, Custer MC, Rosenberg SA. 1985. In vivo administration of purified human interleukin 2. II. Half life, immunologic effects, and expansion of peripheral lymphoid cells in vivo with recombinant IL 2. J Immunol 135(4):2865–2875 [PubMed] [Google Scholar]
  210. Lotze MT, Matory YL, Rayner AA, Ettinghausen SE, Vetto JT, Seipp CA, Rosenberg SA. 1986b. Clinical effects and toxicity of interleukin-2 in patients with cancer. Cancer 58(12):2764–2772 [DOI] [PubMed] [Google Scholar]
  211. Lotze MT, Robb RJ, Sharrow SO, Frana LW, Rosenberg SA. 1984. Systemic administration of interleukin-2 in humans. J Biol Response Mod 3(5):475–482 [PubMed] [Google Scholar]
  212. Ma A, Koka R, Burkett P. 2006. Diverse functions of IL-2, IL-15, and IL-7 in lymphoid homeostasis. Annu Rev Immunol 24:657–679 [DOI] [PubMed] [Google Scholar]
  213. Mackall CL, Fry TJ, Gress RE. 2011. Harnessing the biology of IL-7 for therapeutic application. Nat Rev Immunol 11(5):330–342 [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Maker AV, Phan GQ, Attia P, Yang JC, Sherry RM, Topalian SL, Kammula US, Royal RE, Haworth LR, Levy C, Kleiner D, Mavroukakis SA, Yellin M, Rosenberg SA. 2005. Tumor regression and autoimmunity in patients treated with cytotoxic T lymphocyte-associated antigen 4 blockade and interleukin 2: a phase I/II study. Ann Surg Oncol 12(12):1005–1016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Maleki Vareki S, Garrigos C, Duran I. 2017. Biomarkers of response to PD-1/PD-L1 inhibition. Crit Rev Oncol Hematol 116:116–124 [DOI] [PubMed] [Google Scholar]
  216. Mani A, Roda J, Young D, Caligiuri MA, Fleming GF, Kaufman P, Brufsky A, Ottman S, Carson WE, 3rd, Shapiro CL. 2009. 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 117(1):83–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Margolin K, Atkins MB, Dutcher JP, Ernstoff MS, Smith JW, 2nd, Clark JI, Baar J, Sosman J, Weber J, Lathia C, Brunetti J, Cihon F, Schwartz B. 2007. Phase I trial of BAY 50–4798, an interleukin-2-specific agonist in advanced melanoma and renal cancer. Clin Cancer Res 13(11):3312–3319 [DOI] [PubMed] [Google Scholar]
  218. Marra P, Mathew S, Grigoriadis A, Wu Y, Kyle-Cezar F, Watkins J, Rashid M, De Rinaldis E, Hessey S, Gazinska P, Hayday A, Tutt A. 2014. IL15RA drives antagonistic mechanisms of cancer development and immune control in lymphocyte-enriched triple-negative breast cancers. Cancer Res 74(17):4908–4921 [DOI] [PubMed] [Google Scholar]
  219. Martin CE, Spasova DS, Frimpong-Boateng K, Kim HO, Lee M, Kim KS, Surh CD. 2017. Interleukin-7 availability is maintained by a hematopoietic cytokine sink comprising innate lymphoid cells and T cells. Immunity 47(1):171–182 e4 [DOI] [PubMed] [Google Scholar]
  220. Mathios D, Park CK, Marcus WD, Alter S, Rhode PR, Jeng EK, Wong HC, Pardoll DM, Lim M. 2016. Therapeutic administration of IL-15 superagonist complex ALT-803 leads to long-term survival and durable antitumor immune response in a murine glioblastoma model. Int J Cancer 138(1):187–194 [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Matsuoka K, Koreth J, Kim HT, Bascug G, McDonough S, Kawano Y, Murase K, Cutler C, Ho VT, Alyea EP, Armand P, Blazar BR, Antin JH, Soiffer RJ, Ritz J. 2013. Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease. Sci Transl Med 5(179):179ra43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey SF, Mahnke YD, Melenhorst JJ, Rheingold SR, Shen A, Teachey DT, Levine BL, June CH, Porter DL, Grupp SA. 2014. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 371(16):1507–1517 [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Mazumder A, Rosenberg SA. 1984. Successful immunotherapy of natural killer-resistant established pulmonary melanoma metastases by the intravenous adoptive transfer of syngeneic lymphocytes activated in vitro by interleukin 2. J Exp Med 159(2):495–507 [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Mazzucchelli R, Durum SK. 2007. Interleukin-7 receptor expression: intelligent design. Nat Rev Immunol 7(2):144–154 [DOI] [PubMed] [Google Scholar]
  225. McDermott DF, Cheng SC, Signoretti S, Margolin KA, Clark JI, Sosman JA, Dutcher JP, Logan TF, Curti BD, Ernstoff MS, Appleman L, Wong MK, Khushalani NI, Oleksowicz L, Vaishampayan UN, Mier JW, Panka DJ, Bhatt RS, Bailey AS, Leibovich BC, Kwon ED, Kabbinavar FF, Belldegrun AS, Figlin RA, Pantuck AJ, Regan MM, Atkins MB. 2015a. The high-dose aldesleukin “select” trial: a trial to prospectively validate predictive models of response to treatment in patients with metastatic renal cell carcinoma. Clin Cancer Res 21(3):561–568 [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. McDermott DF, Drake CG, Sznol M, Choueiri TK, Powderly JD, Smith DC, Brahmer JR, Carvajal RD, Hammers HJ, Puzanov I, Hodi FS, Kluger HM, Topalian SL, Pardoll DM, Wigginton JM, Kollia GD, Gupta A, McDonald D, Sankar V, Sosman JA, Atkins MB. 2015b. Survival, durable response, and long-term safety in patients with previously treated advanced renal cell carcinoma receiving nivolumab. J Clin Oncol 33(18):2013–2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. McGuinness RP, Ge Y, Patel SD, Kashmiri SV, Lee HS, Hand PH, Schlom J, Finer MH, McArthur JG. 1999. Anti-tumor activity of human T cells expressing the CC49-zeta chimeric immune receptor. Hum Gene Ther 10(2):165–173 [DOI] [PubMed] [Google Scholar]
  228. Melchionda F, Fry TJ, Milliron MJ, McKirdy MA, Tagaya Y, Mackall CL. 2005. Adjuvant IL-7 or IL-15 overcomes immunodominance and improves survival of the CD8+ memory cell pool. J Clin Invest 115(5):1177–1187 [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Melder RJ, Osborn BL, Riccobene T, Kanakaraj P, Wei P, Chen G, Stolow D, Halpern WG, Migone TS, Wang Q, Grzegorzewski KJ, Gallant G. 2005. Pharmacokinetics and in vitro and in vivo anti-tumor response of an interleukin-2-human serum albumin fusion protein in mice. Cancer Immunol Immunother 54(6):535–547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Mertsching E, Burdet C, Ceredig R. 1995. IL-7 transgenic mice: analysis of the role of IL-7 in the differentiation of thymocytes in vivo and in vitro. Int Immunol 7(3):401–414 [DOI] [PubMed] [Google Scholar]
  231. Meyers FJ, Paradise C, Scudder SA, Goodman G, Konrad M. 1991. A phase I study including pharmacokinetics of polyethylene glycol conjugated interleukin-2. Clin Pharmacol Ther 49(3):307–313 [DOI] [PubMed] [Google Scholar]
  232. Mier JW, Gallo RC. 1980. Purification and some characteristics of human T-cell growth factor from phytohemagglutinin-stimulated lymphocyte-conditioned media. Proc Natl Acad Sci U S A 77(10):6134–6138 [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Milla P, Dosio F, Cattel L. 2012. PEGylation of proteins and liposomes: a powerful and flexible strategy to improve the drug delivery. Curr Drug Metab 13(1):105–119 [DOI] [PubMed] [Google Scholar]
  234. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK, McKenna D, Le C, Defor TE, Burns LJ, Orchard PJ, Blazar BR, Wagner JE, Slungaard A, Weisdorf DJ, Okazaki IJ, McGlave PB. 2005. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 105(8):3051–3057 [DOI] [PubMed] [Google Scholar]
  235. Mishra A, Sullivan L, Caligiuri MA. 2014. Molecular pathways: interleukin-15 signaling in health and in cancer. Clin Cancer Res 20(8):2044–2050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Mitra S, Ring AM, Amarnath S, Spangler JB, Li P, Ju W, Fischer S, Oh J, Spolski R, Weiskopf K, Kohrt H, Foley JE, Rajagopalan S, Long EO, Fowler DH, Waldmann TA, Garcia KC, Leonard WJ. 2015. Interleukin-2 activity can be fine tuned with engineered receptor signaling clamps. Immunity 42(5):826–838 [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Molloy MJ, Zhang W, Usherwood EJ. 2009. Cutting edge: IL-2 immune complexes as a therapy for persistent virus infection. J Immunol 182(8):4512–4515 [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Morgan DA, Ruscetti FW, Gallo R. 1976. Selective in vitro growth of T lymphocytes from normal human bone marrows. Science 193(4257):1007–1008 [DOI] [PubMed] [Google Scholar]
  239. Morgan RA, Chinnasamy N, Abate-Daga D, Gros A, Robbins PF, Zheng Z, Dudley ME, Feldman SA, Yang JC, Sherry RM, Phan GQ, Hughes MS, Kammula US, Miller AD, Hessman CJ, Stewart AA, Restifo NP, Quezado MM, Alimchandani M, Rosenberg AZ, Nath A, Wang T, Bielekova B, Wuest SC, Akula N, McMahon FJ, Wilde S, Mosetter B, Schendel DJ, Laurencot CM, Rosenberg SA. 2013. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother 36(2):133–151 [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, Royal RE, Topalian SL, Kammula US, Restifo NP, Zheng Z, Nahvi A, de Vries CR, Rogers-Freezer LJ, Mavroukakis SA, Rosenberg SA. 2006. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 314(5796):126–129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Morris SC, Heidorn SM, Herbert DR, Perkins C, Hildeman DA, Khodoun MV, Finkelman FD. 2009. Endogenously produced IL-4 nonredundantly stimulates CD8+ T cell proliferation. J Immunol 182(3):1429–1438 [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Morrissey PJ, Conlon P, Charrier K, Braddy S, Alpert A, Williams D, Namen AE, Mochizuki D. 1991. Administration of IL-7 to normal mice stimulates B-lymphopoiesis and peripheral lymphadenopathy. J Immunol 147(2):561–568 [PubMed] [Google Scholar]
  243. Mortier E, Bernard J, Plet A, Jacques Y. 2004. Natural, proteolytic release of a soluble form of human IL-15 receptor alpha-chain that behaves as a specific, high affinity IL-15 antagonist. J Immunol 173(3):1681–1688 [DOI] [PubMed] [Google Scholar]
  244. Mortier E, Quemener A, Vusio P, Lorenzen I, Boublik Y, Grotzinger J, Plet A, Jacques Y. 2006. Soluble interleukin-15 receptor alpha (IL-15R alpha)-sushi as a selective and potent agonist of IL-15 action through IL-15R beta/gamma. Hyperagonist IL-15 × IL-15R alpha fusion proteins. J Biol Chem 281(3):1612–1619 [DOI] [PubMed] [Google Scholar]
  245. Mostbock S, Lutsiak ME, Milenic DE, Baidoo K, Schlom J, Sabzevari H. 2008. IL-2/anti-IL-2 antibody complex enhances vaccine-mediated antigen-specific CD8+ T cell responses and increases the ratio of effector/memory CD8+ T cells to regulatory T cells. J Immunol 180(7):5118–5129 [DOI] [PubMed] [Google Scholar]
  246. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, Castellano D, Choueiri TK, Gurney H, Donskov F, Bono P, Wagstaff J, Gauler TC, Ueda T, Tomita Y, Schutz FA, Kollmannsberger C, Larkin J, Ravaud A, Simon JS, Xu LA, Waxman IM, Sharma P, CheckMate I. 2015. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med 373(19):1803–1813 [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Mule JJ, Ettinghausen SE, Spiess PJ, Shu S, Rosenberg SA. 1986. Antitumor efficacy of lymphokine-activated killer cells and recombinant interleukin-2 in vivo: survival benefit and mechanisms of tumor escape in mice undergoing immunotherapy. Cancer Res 46(2):676–683 [PubMed] [Google Scholar]
  248. Mule JJ, Shu S, Rosenberg SA. 1985. The anti-tumor efficacy of lymphokine-activated killer cells and recombinant interleukin 2 in vivo. J Immunol 135(1):646–652 [PubMed] [Google Scholar]
  249. Mule JJ, Shu S, Schwarz SL, Rosenberg SA. 1984. Adoptive immunotherapy of established pulmonary metastases with LAK cells and recombinant interleukin-2. Science 225(4669):1487–1489 [DOI] [PubMed] [Google Scholar]
  250. Munger W, DeJoy SQ, Jeyaseelan R, Sr., Torley LW, Grabstein KH, Eisenmann J, Paxton R, Cox T, Wick MM, Kerwar SS. 1995. Studies evaluating the antitumor activity and toxicity of interleukin-15, a new T cell growth factor: comparison with interleukin-2. Cell Immunol 165(2):289–293 [DOI] [PubMed] [Google Scholar]
  251. Murakami M, Sakamoto A, Bender J, Kappler J, Marrack P. 2002. CD25+CD4+ T cells contribute to the control of memory CD8+ T cells. Proc Natl Acad Sci U S A 99(13):8832–8837 [DOI] [PMC free article] [PubMed] [Google Scholar]
  252. Muranski P, Boni A, Wrzesinski C, Citrin DE, Rosenberg SA, Childs R, Restifo NP. 2006. Increased intensity lymphodepletion and adoptive immunotherapy—how far can we go? Nat Clin Pract Oncol 3(12):668–681 [DOI] [PMC free article] [PubMed] [Google Scholar]
  253. Muul LM, Spiess PJ, Director EP, Rosenberg SA. 1987. Identification of specific cytolytic immune responses against autologous tumor in humans bearing malignant melanoma. J Immunol 138(3):989–995 [PubMed] [Google Scholar]
  254. Namen AE, Lupton S, Hjerrild K, Wignall J, Mochizuki DY, Schmierer A, Mosley B, March CJ, Urdal D, Gillis S. 1988. Stimulation of B-cell progenitors by cloned murine interleukin-7. Nature 333(6173):571–573 [DOI] [PubMed] [Google Scholar]
  255. Nanjappa SG, Kim EH, Suresh M. 2011. Immunotherapeutic effects of IL-7 during a chronic viral infection in mice. Blood 117(19):5123–5132 [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Nanjappa SG, Walent JH, Morre M, Suresh M. 2008. Effects of IL-7 on memory CD8 T cell homeostasis are influenced by the timing of therapy in mice. J Clin Invest 118(3):1027–1039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Napolitano LA, Grant RM, Deeks SG, Schmidt D, De Rosa SC, Herzenberg LA, Herndier BG, Andersson J, McCune JM. 2001. Increased production of IL-7 accompanies HIV-1-mediated T-cell depletion: implications for T-cell homeostasis. Nat Med 7(1):73–79 [DOI] [PubMed] [Google Scholar]
  258. Neely GG, Epelman S, Ma LL, Colarusso P, Howlett CJ, Amankwah EK, McIntyre AC, Robbins SM, Mody CH. 2004. Monocyte surface-bound IL-15 can function as an activating receptor and participate in reverse signaling. J Immunol 172(7):4225–4234 [DOI] [PubMed] [Google Scholar]
  259. Nelson BH, Willerford DM. 1998. Biology of the interleukin-2 receptor. Adv Immunol 70:1–81 [DOI] [PubMed] [Google Scholar]
  260. Nelson MH, Paulos CM. 2015. Novel immunotherapies for hematologic malignancies. Immunol Rev 263(1):90–105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Nishikawa H, Sakaguchi S. 2014. Regulatory T cells in cancer immunotherapy. Curr Opin Immunol 27:1–7 [DOI] [PubMed] [Google Scholar]
  262. Nishino M, Ramaiya NH, Hatabu H, Hodi FS. 2017. Monitoring immune-checkpoint blockade: response evaluation and biomarker development. Nat Rev Clin Oncol 14(11):655–668 [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. North RJ. 1982. Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on elimination of tumor-induced suppressor T cells. J Exp Med 155(4):1063–1074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Obar JJ, Molloy MJ, Jellison ER, Stoklasek TA, Zhang W, Usherwood EJ, Lefrancois L. 2010. CD4+ T cell regulation of CD25 expression controls development of short-lived effector CD8+ T cells in primary and secondary responses. Proc Natl Acad Sci U S A 107(1):193–198 [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Oh S, Berzofsky JA, Burke DS, Waldmann TA, Perera LP. 2003. Coadministration of HIV vaccine vectors with vaccinia viruses expressing IL-15 but not IL-2 induces long-lasting cellular immunity. Proc Natl Acad Sci U S A 100(6):3392–3397 [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Overwijk WW, Schluns KS. 2009. Functions of gammaC cytokines in immune homeostasis: current and potential clinical applications. Clin Immunol 132(2):153–165 [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Pandey A, Ozaki K, Baumann H, Levin SD, Puel A, Farr AG, Ziegler SF, Leonard WJ, Lodish HF. 2000. Cloning of a receptor subunit required for signaling by thymic stromal lymphopoietin. Nat Immunol 1(1):59–64 [DOI] [PubMed] [Google Scholar]
  268. Park LS, Martin U, Garka K, Gliniak B, Di Santo JP, Muller W, Largaespada DA, Copeland NG, Jenkins NA, Farr AG, Ziegler SF, Morrissey PJ, Paxton R, Sims JE. 2000. Cloning of the murine thymic stromal lymphopoietin (TSLP) receptor: formation of a functional heteromeric complex requires interleukin 7 receptor. J Exp Med 192(5):659–670 [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan DN, Feldman SA, Davis JL, Morgan RA, Merino MJ, Sherry RM, Hughes MS, Kammula US, Phan GQ, Lim RM, Wank SA, Restifo NP, Robbins PF, Laurencot CM, Rosenberg SA. 2011. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther 19(3):620–626 [DOI] [PMC free article] [PubMed] [Google Scholar]
  270. Pasut G, Veronese FM. 2009. PEGylation for improving the effectiveness of therapeutic biomolecules. Drugs Today (Barc) 45(9):687–695 [PubMed] [Google Scholar]
  271. Paulos CM, Wrzesinski C, Kaiser A, Hinrichs CS, Chieppa M, Cassard L, Palmer DC, Boni A, Muranski P, Yu Z, Gattinoni L, Antony PA, Rosenberg SA, Restifo NP. 2007. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J Clin Invest 117(8):2197–2204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  272. Pellegrini M, Calzascia T, Toe JG, Preston SP, Lin AE, Elford AR, Shahinian A, Lang PA, Lang KS, Morre M, Assouline B, Lahl K, Sparwasser T, Tedder TF, Paik JH, DePinho RA, Basta S, Ohashi PS, Mak TW. 2011. IL-7 engages multiple mechanisms to overcome chronic viral infection and limit organ pathology. Cell 144(4):601–613 [DOI] [PubMed] [Google Scholar]
  273. Perales MA, Goldberg JD, Yuan J, Koehne G, Lechner L, Papadopoulos EB, Young JW, Jakubowski AA, Zaidi B, Gallardo H, Liu C, Rasalan T, Wolchok JD, Croughs T, Morre M, Devlin SM, van den Brink MR. 2012. Recombinant human interleukin-7 (CYT107) promotes T-cell recovery after allogeneic stem cell transplantation. Blood 120(24):4882–4891 [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. Pereno R, Gaggero A, Scudeletti M, Lanza L, Meazza R, Mishal Z, Jasmin C, Indiveri F, Ferrini S, Azzarone B. 1999. IL-15/IL-15R alpha intracellular trafficking in human cells and protection from apoptosis. Ann N Y Acad Sci 876:236–245 [DOI] [PubMed] [Google Scholar]
  275. Pereno R, Giron-Michel J, Gaggero A, Cazes E, Meazza R, Monetti M, Monaco E, Mishal Z, Jasmin C, Indiveri F, Ferrini S, Azzarone B. 2000. IL-15/IL-15Ralpha intracellular trafficking in human melanoma cells and signal transduction through the IL-15Ralpha. Oncogene 19(45):5153–5162 [DOI] [PubMed] [Google Scholar]
  276. Peschon JJ, Morrissey PJ, Grabstein KH, Ramsdell FJ, Maraskovsky E, Gliniak BC, Park LS, Ziegler SF, Williams DE, Ware CB, Meyer JD, Davison BL. 1994. Early lymphocyte expansion is severely impaired in interleukin 7 receptor-deficient mice. J Exp Med 180(5):1955–1960 [DOI] [PMC free article] [PubMed] [Google Scholar]
  277. Phelan JD, Orekov T, Finkelman FD. 2008. Cutting edge: mechanism of enhancement of in vivo cytokine effects by anti-cytokine monoclonal antibodies. J Immunol 180(1):44–48 [DOI] [PubMed] [Google Scholar]
  278. Pick E, Turk JL. 1972. The biological activities of soluble lymphocyte products. Clin Exp Immunol 10(1):1–23 [PMC free article] [PubMed] [Google Scholar]
  279. Pilon-Thomas S, Kuhn L, Ellwanger S, Janssen W, Royster E, Marzban S, Kudchadkar R, Zager J, Gibney G, Sondak VK, Weber J, Mule JJ, Sarnaik AA. 2012. Efficacy of adoptive cell transfer of tumor-infiltrating lymphocytes after lymphopenia induction for metastatic melanoma. J Immunother 35(8):615–620 [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Poire X, Kline J, Grinblatt D, Zimmerman T, Conner K, Muhs C, Gajewski T, Van Besien K, Smith SM. 2010. Phase II study of immunomodulation with granulocyte-macrophage colony-stimulating factor, interleukin-2, and rituximab following autologous stem cell transplant in patients with relapsed or refractory lymphomas. Leuk Lymphoma 51(7):1241–1250 [DOI] [PubMed] [Google Scholar]
  281. Prieto PA, Durflinger KH, Wunderlich JR, Rosenberg SA, Dudley ME. 2010. Enrichment of CD8+ cells from melanoma tumor-infiltrating lymphocyte cultures reveals tumor reactivity for use in adoptive cell therapy. J Immunother 33(5):547–556 [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Prieto PA, Yang JC, Sherry RM, Hughes MS, Kammula US, White DE, Levy CL, Rosenberg SA, Phan GQ. 2012. CTLA-4 blockade with ipilimumab: long-term follow-up of 177 patients with metastatic melanoma. Clin Cancer Res 18(7):2039–2047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  283. Pulliam SR, Uzhachenko RV, Adunyah SE, Shanker A. 2016. Common gamma chain cytokines in combinatorial immune strategies against cancer. Immunol Lett 169:61–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
  284. Radvanyi LG, Bernatchez C, Zhang M, Fox PS, Miller P, Chacon J, Wu R, Lizee G, Mahoney S, Alvarado G, Glass M, Johnson VE, McMannis JD, Shpall E, Prieto V, Papadopoulos N, Kim K, Homsi J, Bedikian A, Hwu WJ, Patel S, Ross MI, Lee JE, Gershenwald JE, Lucci A, Royal R, Cormier JN, Davies MA, Mansaray R, Fulbright OJ, Toth C, Ramachandran R, Wardell S, Gonzalez A, Hwu P. 2012. Specific lymphocyte subsets predict response to adoptive cell therapy using expanded autologous tumor-infiltrating lymphocytes in metastatic melanoma patients. Clin Cancer Res 18(24):6758–6770 [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Rao BM, Driver I, Lauffenburger DA, Wittrup KD. 2004. Interleukin 2 (IL-2) variants engineered for increased IL-2 receptor alpha-subunit affinity exhibit increased potency arising from a cell surface ligand reservoir effect. Mol Pharmacol 66(4):864–869 [DOI] [PubMed] [Google Scholar]
  286. Rao BM, Driver I, Lauffenburger DA, Wittrup KD. 2005. High-affinity CD25-binding IL-2 mutants potently stimulate persistent T cell growth. Biochemistry 44(31):10696–10701 [DOI] [PubMed] [Google Scholar]
  287. Rao BM, Girvin AT, Ciardelli T, Lauffenburger DA, Wittrup KD. 2003. Interleukin-2 mutants with enhanced alpha-receptor subunit binding affinity. Protein Eng 16(12):1081–1087 [DOI] [PubMed] [Google Scholar]
  288. Rapoport AP, Stadtmauer EA, Binder-Scholl GK, Goloubeva O, Vogl DT, Lacey SF, Badros AZ, Garfall A, Weiss B, Finklestein J, Kulikovskaya I, Sinha SK, Kronsberg S, Gupta M, Bond S, Melchiori L, Brewer JE, Bennett AD, Gerry AB, Pumphrey NJ, Williams D, Tayton-Martin HK, Ribeiro L, Holdich T, Yanovich S, Hardy N, Yared J, Kerr N, Philip S, Westphal S, Siegel DL, Levine BL, Jakobsen BK, Kalos M, June CH. 2015. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med 21(8):914–921 [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Ratthe C, Girard D. 2004. Interleukin-15 enhances human neutrophil phagocytosis by a Syk-dependent mechanism: importance of the IL-15Ralpha chain. J Leukoc Biol 76(1):162–168 [DOI] [PubMed] [Google Scholar]
  290. Rayner AA, Grimm EA, Lotze MT, Chu EW, Rosenberg SA. 1985a. Lymphokine-activated killer (LAK) cells. Analysis of factors relevant to the immunotherapy of human cancer. Cancer 55(6):1327–1333 [DOI] [PubMed] [Google Scholar]
  291. Rayner AA, Grimm EA, Lotze MT, Wilson DJ, Rosenberg SA. 1985b. Lymphokine-activated killer (LAK) cell phenomenon. IV. Lysis by LAK cell clones of fresh human tumor cells from autologous and multiple allogeneic tumors. J Natl Cancer Inst 75(1):67–75 [PubMed] [Google Scholar]
  292. Restifo NP, Dudley ME, Rosenberg SA. 2012. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol 12(4):269–281 [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Rhode PR, Egan JO, Xu W, Hong H, Webb GM, Chen X, Liu B, Zhu X, Wen J, You L, Kong L, Edwards AC, Han K, Shi S, Alter S, Sacha JB, Jeng EK, Cai W, Wong HC. 2016. Comparison of the superagonist complex, ALT-803, to IL15 as cancer immunotherapeutics in animal models. Cancer Immunol Res 4(1):49–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  294. Ring AM, Lin JX, Feng D, Mitra S, Rickert M, Bowman GR, Pande VS, Li P, Moraga I, Spolski R, Ozkan E, Leonard WJ, Garcia KC. 2012. Mechanistic and structural insight into the functional dichotomy between IL-2 and IL-15. Nat Immunol 13(12):1187–1195 [DOI] [PMC free article] [PubMed] [Google Scholar]
  295. Roan F, Bell BD, Stoklasek TA, Kitajima M, Han H, Ziegler SF. 2012. The multiple facets of thymic stromal lymphopoietin (TSLP) during allergic inflammation and beyond. J Leukoc Biol 91(6):877–886 [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Robb RJ, Munck A, Smith KA. 1981. T cell growth factor receptors. Quantitation, specificity, and biological relevance. J Exp Med 154(5):1455–1474 [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Robbins PF, Kassim SH, Tran TL, Crystal JS, Morgan RA, Feldman SA, Yang JC, Dudley ME, Wunderlich JR, Sherry RM, Kammula US, Hughes MS, Restifo NP, Raffeld M, Lee CC, Li YF, El-Gamil M, Rosenberg SA. 2015. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res 21(5):1019–1027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  298. Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, Wunderlich JR, Nahvi AV, Helman LJ, Mackall CL, Kammula US, Hughes MS, Restifo NP, Raffeld M, Lee CC, Levy CL, Li YF, El-Gamil M, Schwarz SL, Laurencot C, Rosenberg SA. 2011. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol 29(7):917–924 [DOI] [PMC free article] [PubMed] [Google Scholar]
  299. Rochman Y, Spolski R, Leonard WJ. 2009. New insights into the regulation of T cells by gamma(c) family cytokines. Nat Rev Immunol 9(7):480–490 [DOI] [PMC free article] [PubMed] [Google Scholar]
  300. Rosario M, Liu B, Kong L, Collins LI, Schneider SE, Chen X, Han K, Jeng EK, Rhode PR, Leong JW, Schappe T, Jewell BA, Keppel CR, Shah K, Hess B, Romee R, Piwnica-Worms DR, Cashen AF, Bartlett NL, Wong HC, Fehniger TA. 2016. The IL-15-based ALT-803 complex enhances FcgammaRIIIa-triggered NK cell responses and in vivo clearance of B cell lymphomas. Clin Cancer Res 22(3):596–608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  301. Rosenberg SA. 1992. Karnofsky memorial lecture. The immunotherapy and gene therapy of cancer. J Clin Oncol 10(2):180–199 [DOI] [PubMed] [Google Scholar]
  302. Rosenberg SA. 2014. IL-2: the first effective immunotherapy for human cancer. J Immunol 192(12):5451–5458 [DOI] [PMC free article] [PubMed] [Google Scholar]
  303. Rosenberg SA, Lotze MT, Muul LM, Leitman S, Chang AE, Ettinghausen SE, Matory YL, Skibber JM, Shiloni E, Vetto JT, et al. . 1985. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N Engl J Med 313(23):1485–1492 [DOI] [PubMed] [Google Scholar]
  304. Rosenberg SA, Lotze MT, Yang JC, Topalian SL, Chang AE, Schwartzentruber DJ, Aebersold P, Leitman S, Linehan WM, Seipp CA, et al. . 1993. Prospective randomized trial of high-dose interleukin-2 alone or in conjunction with lymphokine-activated killer cells for the treatment of patients with advanced cancer. J Natl Cancer Inst 85(8):622–632 [DOI] [PubMed] [Google Scholar]
  305. Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, Simon P, Lotze MT, Yang JC, Seipp CA, et al. . 1988. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med 319(25):1676–1680 [DOI] [PubMed] [Google Scholar]
  306. Rosenberg SA, Spiess P, Lafreniere R. 1986. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science 233(4770):1318–1321 [DOI] [PubMed] [Google Scholar]
  307. Rosenberg SA, Sportes C, Ahmadzadeh M, Fry TJ, Ngo LT, Schwarz SL, Stetler-Stevenson M, Morton KE, Mavroukakis SA, Morre M, Buffet R, Mackall CL, Gress RE. 2006. IL-7 administration to humans leads to expansion of CD8+ and CD4+ cells but a relative decrease of CD4+ T-regulatory cells. J Immunother 29(3):313–319 [DOI] [PMC free article] [PubMed] [Google Scholar]
  308. Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, Citrin DE, Restifo NP, Robbins PF, Wunderlich JR, Morton KE, Laurencot CM, Steinberg SM, White DE, Dudley ME. 2011. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 17(13):4550–4557 [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Rosenberg SA, Yannelli JR, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, Seipp CA, Einhorn JH, White DE. 1994. Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. J Natl Cancer Inst 86(15):1159–1166 [DOI] [PubMed] [Google Scholar]
  310. Rosenstein M, Ettinghausen SE, Rosenberg SA. 1986. Extravasation of intravascular fluid mediated by the systemic administration of recombinant interleukin 2. J Immunol 137(5):1735–1742 [PubMed] [Google Scholar]
  311. Rowley J, Monie A, Hung CF, Wu TC. 2009. Expression of IL-15RA or an IL-15/IL-15RA fusion on CD8+ T cells modifies adoptively transferred T-cell function in cis. Eur J Immunol 39(2):491–506 [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. Royal RE, Steinberg SM, Krouse RS, Heywood G, White DE, Hwu P, Marincola FM, Parkinson DR, Schwartzentruber DJ, Topalian SL, Yang JC, Rosenberg SA. 1996. Correlates of response to IL-2 therapy in patients treated for metastatic renal cancer and melanoma. Cancer J Sci Am 2(2):91–98 [PubMed] [Google Scholar]
  313. Roychowdhury S, May KF, Jr., Tzou KS, Lin T, Bhatt D, Freud AG, Guimond M, Ferketich AK, Liu Y, Caligiuri MA. 2004. Failed adoptive immunotherapy with tumor-specific T cells: reversal with low-dose interleukin 15 but not low-dose interleukin 2. Cancer Res 64(21):8062–8067 [DOI] [PubMed] [Google Scholar]
  314. Ruan S, Samuelson DR, Assouline B, Morre M, Shellito JE. 2016. Treatment with interleukin-7 restores host defense against pneumocystis in CD4+ T-lymphocyte-depleted mice. Infect Immun 84(1):108–119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. Rubin LA, Kurman CC, Fritz ME, Biddison WE, Boutin B, Yarchoan R, Nelson DL. 1985. Soluble interleukin 2 receptors are released from activated human lymphoid cells in vitro. J Immunol 135(5):3172–3177 [PubMed] [Google Scholar]
  316. Rubinstein MP, Kadima AN, Salem ML, Nguyen CL, Gillanders WE, Cole DJ. 2002. Systemic administration of IL-15 augments the antigen-specific primary CD8+ T cell response following vaccination with peptide-pulsed dendritic cells. J Immunol 169(9):4928–4935 [DOI] [PubMed] [Google Scholar]
  317. Rubinstein MP, Kovar M, Purton JF, Cho JH, Boyman O, Surh CD, Sprent J. 2006. Converting IL-15 to a superagonist by binding to soluble IL-15R{alpha}. Proc Natl Acad Sci U S A 103(24):9166–9171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  318. Rubinstein MP, Lind NA, Purton JF, Filippou P, Best JA, McGhee PA, Surh CD, Goldrath AW. 2008. IL-7 and IL-15 differentially regulate CD8+ T-cell subsets during contraction of the immune response. Blood 112(9):3704–3712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  319. Saadoun D, Rosenzwajg M, Joly F, Six A, Carrat F, Thibault V, Sene D, Cacoub P, Klatzmann D. 2011. Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med 365(22):2067–2077 [DOI] [PubMed] [Google Scholar]
  320. Sadlack B, Lohler J, Schorle H, Klebb G, Haber H, Sickel E, Noelle RJ, Horak I. 1995. Generalized autoimmune disease in interleukin-2-deficient mice is triggered by an uncontrolled activation and proliferation of CD4+ T cells. Eur J Immunol 25(11):3053–3059 [DOI] [PubMed] [Google Scholar]
  321. Sadlack B, Merz H, Schorle H, Schimpl A, Feller AC, Horak I. 1993. Ulcerative colitis-like disease in mice with a disrupted interleukin-2 gene. Cell 75(2):253–261 [DOI] [PubMed] [Google Scholar]
  322. Salem ML, Diaz-Montero CM, Al-Khami AA, El-Naggar SA, Naga O, Montero AJ, Khafagy A, Cole DJ. 2009. Recovery from cyclophosphamide-induced lymphopenia results in expansion of immature dendritic cells which can mediate enhanced prime-boost vaccination antitumor responses in vivo when stimulated with the TLR3 agonist poly(I:C). J Immunol 182(4):2030–2040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  323. Samaridis J, Casorati G, Traunecker A, Iglesias A, Gutierrez JC, Muller U, Palacios R. 1991. Development of lymphocytes in interleukin 7-transgenic mice. Eur J Immunol 21(2):453–460 [DOI] [PubMed] [Google Scholar]
  324. Saraceni C, Agostino N, Weiss MJ, Harris K, Nair S. 2015. Clinical characteristics and treatment-related biomarkers associated with response to high-dose interleukin-2 in metastatic melanoma and renal cell carcinoma: retrospective analysis of an academic community hospital's experience. Springerplus 4:118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Sato J, Hamaguchi N, Doken K, Gotoh K, Ootsu K, Iwasa S, Ogawa Y, Toguchi H. 1993. Enhancement of anti-tumor activity of recombinant interleukin-2 (rIL-2) by immunocomplexing with a monoclonal antibody against rIL-2. Biotherapy 6(3):225–231 [DOI] [PubMed] [Google Scholar]
  326. Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K, Hamid O, Patt D, Chen TT, Berman DM, Wolchok JD. 2015. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J Clin Oncol 33(17):1889–1894 [DOI] [PMC free article] [PubMed] [Google Scholar]
  327. Schluns KS, Kieper WC, Jameson SC, Lefrancois L. 2000. Interleukin-7 mediates the homeostasis of naive and memory CD8 T cells in vivo. Nat Immunol 1(5):426–432 [DOI] [PubMed] [Google Scholar]
  328. Schluns KS, Stoklasek T, Lefrancois L. 2005. The roles of interleukin-15 receptor alpha: trans-presentation, receptor component, or both? Int J Biochem Cell Biol 37(8):1567–1571 [DOI] [PubMed] [Google Scholar]
  329. Schwartzentruber DJ. 2001. Guidelines for the safe administration of high-dose interleukin-2. J Immunother 24(4):287–293 [DOI] [PubMed] [Google Scholar]
  330. Sereti I, Dunham RM, Spritzler J, Aga E, Proschan MA, Medvik K, Battaglia CA, Landay AL, Pahwa S, Fischl MA, Asmuth DM, Tenorio AR, Altman JD, Fox L, Moir S, Malaspina A, Morre M, Buffet R, Silvestri G, Lederman MM, Team AS. 2009. IL-7 administration drives T cell-cycle entry and expansion in HIV-1 infection. Blood 113(25):6304–6314 [DOI] [PMC free article] [PubMed] [Google Scholar]
  331. Shanafelt AB, Lin Y, Shanafelt MC, Forte CP, Dubois-Stringfellow N, Carter C, Gibbons JA, Cheng SL, Delaria KA, Fleischer R, Greve JM, Gundel R, Harris K, Kelly R, Koh B, Li Y, Lantz L, Mak P, Neyer L, Plym MJ, Roczniak S, Serban D, Thrift J, Tsuchiyama L, Wetzel M, Wong M, Zolotorev A. 2000. A T-cell-selective interleukin 2 mutein exhibits potent antitumor activity and is well tolerated in vivo. Nat Biotechnol 18(11):1197–1202 [DOI] [PubMed] [Google Scholar]
  332. Sharfe N, Dadi HK, Shahar M, Roifman CM. 1997. Human immune disorder arising from mutation of the alpha chain of the interleukin-2 receptor. Proc Natl Acad Sci U S A 94(7):3168–3171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  333. Sharma P, Allison JP. 2015. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161(2):205–214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  334. Sharma P, Kranz DM. 2016. Recent advances in T-cell engineering for use in immunotherapy. F1000Res 5: pii: [DOI] [PMC free article] [PubMed] [Google Scholar]
  335. Shaw J, Monticone V, Paetkau V. 1978. Partial purification and molecular characterization of a lymphokine (costimulator) required for the mitogenic response of mouse thymocytes in vitro. J Immunol 120(6):1967–1973 [PubMed] [Google Scholar]
  336. Sheikh V, Porter BO, DerSimonian R, Kovacs SB, Thompson WL, Perez-Diez A, Freeman AF, Roby G, Mican J, Pau A, Rupert A, Adelsberger J, Higgins J, Bourgeois JS, Jr., Jensen SM, Morcock DR, Burbelo PD, Osnos L, Maric I, Natarajan V, Croughs T, Yao MD, Estes JD, Sereti I. 2016. Administration of interleukin-7 increases CD4 T cells in idiopathic CD4 lymphocytopenia. Blood 127(8):977–988 [DOI] [PMC free article] [PubMed] [Google Scholar]
  337. Shevach EM, Thornton AM. 2014. tTregs, pTregs, and iTregs: similarities and differences. Immunol Rev 259(1):88–102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  338. Shi LZ, Fu T, Guan B, Chen J, Blando JM, Allison JP, Xiong L, Subudhi SK, Gao J, Sharma P. 2016. Interdependent IL-7 and IFN-gamma signalling in T-cell controls tumour eradication by combined alpha-CTLA-4+alpha-PD-1 therapy. Nat Commun 7:12335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  339. Sim GC, Martin-Orozco N, Jin L, Yang Y, Wu S, Washington E, Sanders D, Lacey C, Wang Y, Vence L, Hwu P, Radvanyi L. 2014. IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients. J Clin Invest 124(1):99–110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  340. Sim GC, Radvanyi L. 2014. The IL-2 cytokine family in cancer immunotherapy. Cytokine Growth Factor Rev 25(4):377–390 [DOI] [PubMed] [Google Scholar]
  341. Smith C, Martinez M, Peet J, Khanna R. 2011. Differential outcome of IL-2/anti-IL-2 complex therapy on effector and memory CD8+ T cells following vaccination with an adenoviral vector encoding EBV epitopes. J Immunol 186(10):5784–5790 [DOI] [PubMed] [Google Scholar]
  342. Smith KA. 1988. Interleukin-2: inception, impact, and implications. Science 240(4856):1169–1176 [DOI] [PubMed] [Google Scholar]
  343. Spangler JB, Tomala J, Luca VC, Jude KM, Dong S, Ring AM, Votavova P, Pepper M, Kovar M, Garcia KC. 2015. Antibodies to interleukin-2 elicit selective T cell subset potentiation through distinct conformational mechanisms. Immunity 42(5):815–825 [DOI] [PMC free article] [PubMed] [Google Scholar]
  344. Spear TT, Nagato K, Nishimura MI. 2016. Strategies to genetically engineer T cells for cancer immunotherapy. Cancer Immunol Immunother 65(6):631–649 [DOI] [PMC free article] [PubMed] [Google Scholar]
  345. Sportes C, Babb RR, Krumlauf MC, Hakim FT, Steinberg SM, Chow CK, Brown MR, Fleisher TA, Noel P, Maric I, Stetler-Stevenson M, Engel J, Buffet R, Morre M, Amato RJ, Pecora A, Mackall CL, Gress RE. 2010. Phase I study of recombinant human interleukin-7 administration in subjects with refractory malignancy. Clin Cancer Res 16(2):727–735 [DOI] [PMC free article] [PubMed] [Google Scholar]
  346. Sportes C, Hakim FT, Memon SA, Zhang H, Chua KS, Brown MR, Fleisher TA, Krumlauf MC, Babb RR, Chow CK, Fry TJ, Engels J, Buffet R, Morre M, Amato RJ, Venzon DJ, Korngold R, Pecora A, Gress RE, Mackall CL. 2008. Administration of rhIL-7 in humans increases in vivo TCR repertoire diversity by preferential expansion of naive T cell subsets. J Exp Med 205(7):1701–1714 [DOI] [PMC free article] [PubMed] [Google Scholar]
  347. Stadler BM, Berenstein EH, Siraganian RP, Oppenheim JJ. 1982. Monoclonal antibody against human interleukin 2 (IL 2). Purification of IL 2 for the production of monoclonal antibodies. J Immunol 128(4):1620–1624 [PubMed] [Google Scholar]
  348. Stevens AC, Matthews J, Andres P, Baffis V, Zheng XX, Chae DW, Smith J, Strom TB, Maslinski W. 1997. Interleukin-15 signals T84 colonic epithelial cells in the absence of the interleukin-2 receptor beta-chain. Am J Physiol 272(5 Pt 1):G1201–G1208 [DOI] [PubMed] [Google Scholar]
  349. Stoklasek TA, Schluns KS, Lefrancois L. 2006. Combined IL-15/IL-15Ralpha immunotherapy maximizes IL-15 activity in vivo. J Immunol 177(9):6072–6080 [DOI] [PMC free article] [PubMed] [Google Scholar]
  350. Stone JD, Chervin AS, Schreiber H, Kranz DM. 2012. Design and characterization of a protein superagonist of IL-15 fused with IL-15Ralpha and a high-affinity T cell receptor. Biotechnol Prog 28(6):1588–1597 [DOI] [PMC free article] [PubMed] [Google Scholar]
  351. Stonier SW, Ma LJ, Castillo EF, Schluns KS. 2008. Dendritic cells drive memory CD8 T-cell homeostasis via IL-15 transpresentation. Blood 112(12):4546–4554 [DOI] [PMC free article] [PubMed] [Google Scholar]
  352. Stromnes IM, Schmitt TM, Chapuis AG, Hingorani SR, Greenberg PD. 2014. Re-adapting T cells for cancer therapy: from mouse models to clinical trials. Immunol Rev 257(1):145–164 [DOI] [PMC free article] [PubMed] [Google Scholar]
  353. Su EW, Moore CJ, Suriano S, Johnson CB, Songalia N, Patterson A, Neitzke DJ, Andrijauskaite K, Garrett-Mayer E, Mehrotra S, Paulos CM, Doedens AL, Goldrath AW, Li Z, Cole DJ, Rubinstein MP. 2015. IL-2Ralpha mediates temporal regulation of IL-2 signaling and enhances immunotherapy. Sci Transl Med 7(311):311ra170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  354. Suzuki H, Kundig TM, Furlonger C, Wakeham A, Timms E, Matsuyama T, Schmits R, Simard JJ, Ohashi PS, Griesser H, et al. . 1995. Deregulated T cell activation and autoimmunity in mice lacking interleukin-2 receptor beta. Science 268(5216):1472–1476 [DOI] [PubMed] [Google Scholar]
  355. Swisher SG, Anderson TM, Wen DR, Stene MA, Cochran AJ, Golub SH, Holmes EC. 1991. Immunopathological features of human pulmonary tumors following low-dose interleukin-2. Cancer Immunol Immunother 33(5):327–332 [DOI] [PMC free article] [PubMed] [Google Scholar]
  356. Tagaya Y, Bamford RN, DeFilippis AP, Waldmann TA. 1996. IL-15: a pleiotropic cytokine with diverse receptor/signaling pathways whose expression is controlled at multiple levels. Immunity 4(4):329–336 [DOI] [PubMed] [Google Scholar]
  357. Tan JT, Dudl E, LeRoy E, Murray R, Sprent J, Weinberg KI, Surh CD. 2001. IL-7 is critical for homeostatic proliferation and survival of naive T cells. Proc Natl Acad Sci U S A 98(15):8732–8737 [DOI] [PMC free article] [PubMed] [Google Scholar]
  358. Tan JT, Ernst B, Kieper WC, LeRoy E, Sprent J, Surh CD. 2002. Interleukin (IL)-15 and IL-7 jointly regulate homeostatic proliferation of memory phenotype CD8+ cells but are not required for memory phenotype CD4+ cells. J Exp Med 195(12):1523–1532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  359. Tang JC, Shen GB, Wang SM, Wan YS, Wei YQ. 2014. IL-7 inhibits tumor growth by promoting T cell-mediated antitumor immunity in Meth A model. Immunol Lett 158(1–2):159–166 [DOI] [PubMed] [Google Scholar]
  360. Taniguchi T, Matsui H, Fujita T, Takaoka C, Kashima N, Yoshimoto R, Hamuro J. 1983. Structure and expression of a cloned cDNA for human interleukin-2. Nature 302(5906):305–310 [DOI] [PubMed] [Google Scholar]
  361. Thaysen-Andersen M, Chertova E, Bergamaschi C, Moh ES, Chertov O, Roser J, Sowder R, Bear J, Lifson J, Packer NH, Felber BK, Pavlakis GN. 2016. Recombinant human heterodimeric IL-15 complex displays extensive and reproducible N- and O-linked glycosylation. Glycoconj J 33(3):417–433 [DOI] [PMC free article] [PubMed] [Google Scholar]
  362. Theze J, Alzari PM, Bertoglio J. 1996. Interleukin 2 and its receptors: recent advances and new immunological functions. Immunol Today 17(10):481–486 [DOI] [PubMed] [Google Scholar]
  363. Thiebaut R, Jarne A, Routy JP, Sereti I, Fischl M, Ive P, Speck RF, D'Offizi G, Casari S, Commenges D, Foulkes S, Natarajan V, Croughs T, Delfraissy JF, Tambussi G, Levy Y, Lederman MM. 2016. Repeated cycles of recombinant human interleukin 7 in HIV-infected patients with low CD4 T-cell reconstitution on antiretroviral therapy: results of 2 phase II multicenter studies. Clin Infect Dis 62(9):1178–1185 [DOI] [PMC free article] [PubMed] [Google Scholar]
  364. Tomala J, Chmelova H, Mrkvan T, Rihova B, Kovar M. 2009. In vivo expansion of activated naive CD8+ T cells and NK cells driven by complexes of IL-2 and anti-IL-2 monoclonal antibody as novel approach of cancer immunotherapy. J Immunol 183(8):4904–4912 [DOI] [PubMed] [Google Scholar]
  365. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen L, Sharfman WH, Anders RA, Taube JM, McMiller TL, Xu H, Korman AJ, Jure-Kunkel M, Agrawal S, McDonald D, Kollia GD, Gupta A, Wigginton JM, Sznol M. 2012. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366(26):2443–2454 [DOI] [PMC free article] [PubMed] [Google Scholar]
  366. Topalian SL, Muul LM, Solomon D, Rosenberg SA. 1987. Expansion of human tumor infiltrating lymphocytes for use in immunotherapy trials. J Immunol Methods 102(1):127–141 [DOI] [PubMed] [Google Scholar]
  367. Topalian SL, Solomon D, Avis FP, Chang AE, Freerksen DL, Linehan WM, Lotze MT, Robertson CN, Seipp CA, Simon P, et al. . 1988. Immunotherapy of patients with advanced cancer using tumor-infiltrating lymphocytes and recombinant interleukin-2: a pilot study. J Clin Oncol 6(5):839–853 [DOI] [PubMed] [Google Scholar]
  368. Topalian SL, Taube JM, Anders RA, Pardoll DM. 2016. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 16(5):275–287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  369. Tosic V, Thomas DL, Kranz DM, Liu J, McFadden G, Shisler JL, MacNeill AL, Roy EJ. 2014. Myxoma virus expressing a fusion protein of interleukin-15 (IL15) and IL15 receptor alpha has enhanced antitumor activity. PLoS One 9(10):e109801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  370. Tredan O, Menetrier-Caux C, Ray-Coquard I, Garin G, Cropet C, Verronese E, Bachelot T, Rebattu P, Heudel PE, Cassier P, Chabaud S, Croughs T, Dupont P, Cadore AC, Clapisson G, Delgado A, Bardin-dit-Courageot C, Rigal C, N'Kodia A, Gilles-Afchain L, Morre M, Perol D, Blay JY, Caux C. 2015. ELYPSE-7: a randomized placebo-controlled phase IIa trial with CYT107 exploring the restoration of CD4+ lymphocyte count in lymphopenic metastatic breast cancer patients. Ann Oncol 26(7):1353–1362 [DOI] [PubMed] [Google Scholar]
  371. Tsytsikov VN, Yurovsky VV, Atamas SP, Alms WJ, White B. 1996. Identification and characterization of two alternative splice variants of human interleukin-2. J Biol Chem 271(38):23055–23060 [DOI] [PubMed] [Google Scholar]
  372. Turecek PL, Bossard MJ, Schoetens F, Ivens IA. 2016. PEGylation of Biopharmaceuticals: a review of chemistry and nonclinical safety information of approved drugs. J Pharm Sci 105(2):460–475 [DOI] [PubMed] [Google Scholar]
  373. Turtle CJ, Riddell SR, Maloney DG. 2016. CD19-targeted chimeric antigen receptor-modified T cell immunotherapy for B cell malignancies. Clin Pharmacol Ther 100(3):252–258 [DOI] [PubMed] [Google Scholar]
  374. Tzeng A, Kwan BH, Opel CF, Navaratna T, Wittrup KD. 2015. Antigen specificity can be irrelevant to immunocytokine efficacy and biodistribution. Proc Natl Acad Sci U S A 112(11):3320–3325 [DOI] [PMC free article] [PubMed] [Google Scholar]
  375. Ugen KE, Kutzler MA, Marrero B, Westover J, Coppola D, Weiner DB, Heller R. 2006. Regression of subcutaneous B16 melanoma tumors after intratumoral delivery of an IL-15-expressing plasmid followed by in vivo electroporation. Cancer Gene Ther 13(10):969–974 [DOI] [PMC free article] [PubMed] [Google Scholar]
  376. Upton MP, Parker RA, Youmans A, McDermott DF, Atkins MB. 2005. Histologic predictors of renal cell carcinoma response to interleukin-2-based therapy. J Immunother 28(5):488–495 [DOI] [PubMed] [Google Scholar]
  377. Valenzona HO, Pointer R, Ceredig R, Osmond DG. 1996. Prelymphomatous B cell hyperplasia in the bone marrow of interleukin-7 transgenic mice: precursor B cell dynamics, microenvironmental organization and osteolysis. Exp Hematol 24(13):1521–1529 [PubMed] [Google Scholar]
  378. Verdegaal EM, de Miranda NF, Visser M, Harryvan T, van Buuren MM, Andersen RS, Hadrup SR, van der Minne CE, Schotte R, Spits H, Haanen JB, Kapiteijn EH, Schumacher TN, van der Burg SH. 2016. Neoantigen landscape dynamics during human melanoma-T cell interactions. Nature 536(7614):91–95 [DOI] [PubMed] [Google Scholar]
  379. Verdegaal EM, Visser M, Ramwadhdoebe TH, van der Minne CE, van Steijn JA, Kapiteijn E, Haanen JB, van der Burg SH, Nortier JW, Osanto S. 2011. Successful treatment of metastatic melanoma by adoptive transfer of blood-derived polyclonal tumor-specific CD4+ and CD8+ T cells in combination with low-dose interferon-alpha. Cancer Immunol Immunother 60(7):953–963 [DOI] [PMC free article] [PubMed] [Google Scholar]
  380. von Freeden-Jeffry U, Vieira P, Lucian LA, McNeil T, Burdach SE, Murray R. 1995. Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies IL-7 as a nonredundant cytokine. J Exp Med 181(4):1519–1526 [DOI] [PMC free article] [PubMed] [Google Scholar]
  381. Waldmann TA. 2006. The biology of interleukin-2 and interleukin-15: implications for cancer therapy and vaccine design. Nat Rev Immunol 6(8):595–601 [DOI] [PubMed] [Google Scholar]
  382. Waldmann TA. 2014. Interleukin-15 in the treatment of cancer. Expert Rev Clin Immunol 10(12):1689–1701 [DOI] [PMC free article] [PubMed] [Google Scholar]
  383. Waldmann TA. 2015. The shared and contrasting roles of IL2 and IL15 in the life and death of normal and neoplastic lymphocytes: implications for cancer therapy. Cancer Immunol Res 3(3):219–227 [DOI] [PMC free article] [PubMed] [Google Scholar]
  384. Waldmann TA, Dubois S, Tagaya Y. 2001. Contrasting roles of IL-2 and IL-15 in the life and death of lymphocytes: implications for immunotherapy. Immunity 14(2):105–110 [PubMed] [Google Scholar]
  385. Wallen H, Thompson JA, Reilly JZ, Rodmyre RM, Cao J, Yee C. 2009. Fludarabine modulates immune response and extends in vivo survival of adoptively transferred CD8 T cells in patients with metastatic melanoma. PLoS One 4(3):e4749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  386. Wang A, Lu SD, Mark DF. 1984. Site-specific mutagenesis of the human interleukin-2 gene: structure-function analysis of the cysteine residues. Science 224(4656):1431–1433 [DOI] [PubMed] [Google Scholar]
  387. Wang RF, Wang HY. 2017. Immune targets and neoantigens for cancer immunotherapy and precision medicine. Cell Res 27(1):11–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  388. Watson J, Gillis S, Marbrook J, Mochizuki D, Smith KA. 1979. Biochemical and biological characterization of lymphocyte regulatory molecules. I. Purification of a class of murine lymphokines. J Exp Med 150(4):849–861 [DOI] [PMC free article] [PubMed] [Google Scholar]
  389. Webster KE, Walters S, Kohler RE, Mrkvan T, Boyman O, Surh CD, Grey ST, Sprent J. 2009. In vivo expansion of T reg cells with IL-2-mAb complexes: induction of resistance to EAE and long-term acceptance of islet allografts without immunosuppression. J Exp Med 206(4):751–760 [DOI] [PMC free article] [PubMed] [Google Scholar]
  390. West EE, Jin HT, Rasheed AU, Penaloza-Macmaster P, Ha SJ, Tan WG, Youngblood B, Freeman GJ, Smith KA, Ahmed R. 2013. PD-L1 blockade synergizes with IL-2 therapy in reinvigorating exhausted T cells. J Clin Invest 123(6):2604–2615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  391. Willerford DM, Chen J, Ferry JA, Davidson L, Ma A, Alt FW. 1995. Interleukin-2 receptor alpha chain regulates the size and content of the peripheral lymphoid compartment. Immunity 3(4):521–530 [DOI] [PubMed] [Google Scholar]
  392. Wilson MS, Pesce JT, Ramalingam TR, Thompson RW, Cheever A, Wynn TA. 2008. Suppression of murine allergic airway disease by IL-2:anti-IL-2 monoclonal antibody-induced regulatory T cells. J Immunol 181(10):6942–6954 [DOI] [PMC free article] [PubMed] [Google Scholar]
  393. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K, Burke MM, Caldwell A, Kronenberg SA, Agunwamba BU, Zhang X, Lowy I, Inzunza HD, Feely W, Horak CE, Hong Q, Korman AJ, Wigginton JM, Gupta A, Sznol M. 2013. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 369(2):122–133 [DOI] [PMC free article] [PubMed] [Google Scholar]
  394. Wrzesinski C, Paulos CM, Kaiser A, Muranski P, Palmer DC, Gattinoni L, Yu Z, Rosenberg SA, Restifo NP. 2010. Increased intensity lymphodepletion enhances tumor treatment efficacy of adoptively transferred tumor-specific T cells. J Immunother 33(1):1–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  395. Wuest SC, Edwan JH, Martin JF, Han S, Perry JS, Cartagena CM, Matsuura E, Maric D, Waldmann TA, Bielekova B. 2011. A role for interleukin-2 trans-presentation in dendritic cell-mediated T cell activation in humans, as revealed by daclizumab therapy. Nat Med 17(5):604–609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  396. Xu W JM, Liu B, Zhu X, Johnson CB, Edwards AC, Kong L, Jeng EK, Han K, Marcus WD, Rubinstein MP, Rhode PR, Wong HC. 2013. Efficacy and mechanism-of-action of a novel superagonist interleukin-15: interleukin-15 receptor aSu/Fc fusion complex in syngeneic murine models of multiple myeloma. Cancer Res 73(10):3075–3086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  397. Xue HH, Kovanen PE, Pise-Masison CA, Berg M, Radovich MF, Brady JN, Leonard WJ. 2002. IL-2 negatively regulates IL-7 receptor alpha chain expression in activated T lymphocytes. Proc Natl Acad Sci U S A 99(21):13759–13764 [DOI] [PMC free article] [PubMed] [Google Scholar]
  398. Xue SA, Gao L, Hart D, Gillmore R, Qasim W, Thrasher A, Apperley J, Engels B, Uckert W, Morris E, Stauss H. 2005. Elimination of human leukemia cells in NOD/SCID mice by WT1-TCR gene-transduced human T cells. Blood 106(9):3062–3067 [DOI] [PubMed] [Google Scholar]
  399. Yang JC, Schwarz SL, Perry-Lalley DM, Rosenberg SA. 1991. Murine studies using polyethylene glycol-modified recombinant human interleukin 2 (PEG-IL-2): antitumor effects of PEG-IL2 alone and in combination with adoptive cellular transfer. Lymphokine Cytokine Res 10(6):475–480 [PubMed] [Google Scholar]
  400. Yang JC, Topalian SL, Schwartzentruber DJ, Parkinson DR, Marincola FM, Weber JS, Seipp CA, White DE, Rosenberg SA. 1995. The use of polyethylene glycol-modified interleukin-2 (PEG-IL-2) in the treatment of patients with metastatic renal cell carcinoma and melanoma. A phase I study and a randomized prospective study comparing IL-2 alone versus IL-2 combined with PEG-IL-2. Cancer 76(4):687–694 [DOI] [PubMed] [Google Scholar]
  401. Yao X, Ahmadzadeh M, Lu YC, Liewehr DJ, Dudley ME, Liu F, Schrump DS, Steinberg SM, Rosenberg SA, Robbins PF. 2012. Levels of peripheral CD4(+)FoxP3(+) regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer. Blood 119(24):5688–5696 [DOI] [PMC free article] [PubMed] [Google Scholar]
  402. Yao Z, Dai W, Perry J, Brechbiel MW, Sung C. 2004. Effect of albumin fusion on the biodistribution of interleukin-2. Cancer Immunol Immunother 53(5):404–410 [DOI] [PMC free article] [PubMed] [Google Scholar]
  403. Yee C. 2014. The use of endogenous T cells for adoptive transfer. Immunol Rev 257(1):250–263 [DOI] [PubMed] [Google Scholar]
  404. Yee C, Thompson JA, Byrd D, Riddell SR, Roche P, Celis E, Greenberg PD. 2002. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci U S A 99(25):16168–16173 [DOI] [PMC free article] [PubMed] [Google Scholar]
  405. Yron I, Wood TA, Jr., Spiess PJ, Rosenberg SA. 1980. In vitro growth of murine T cells. V. The isolation and growth of lymphoid cells infiltrating syngeneic solid tumors. J Immunol 125(1):238–245 [PubMed] [Google Scholar]
  406. Yu A, Zhou J, Marten N, Bergmann CC, Mammolenti M, Levy RB, Malek TR. 2003. Efficient induction of primary and secondary T cell-dependent immune responses in vivo in the absence of functional IL-2 and IL-15 receptors. J Immunol 170(1):236–242 [DOI] [PubMed] [Google Scholar]
  407. Yu P, Steel JC, Zhang M, Morris JC, Waldmann TA. 2010. Simultaneous blockade of multiple immune system inhibitory checkpoints enhances antitumor activity mediated by interleukin-15 in a murine metastatic colon carcinoma model. Clin Cancer Res 16(24):6019–6028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  408. Yuan X, Cheng G, Malek TR. 2014. The importance of regulatory T-cell heterogeneity in maintaining self-tolerance. Immunol Rev 259(1):103–114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  409. Zhang H, Chua KS, Guimond M, Kapoor V, Brown MV, Fleisher TA, Long LM, Bernstein D, Hill BJ, Douek DC, Berzofsky JA, Carter CS, Read EJ, Helman LJ, Mackall CL. 2005. Lymphopenia and interleukin-2 therapy alter homeostasis of CD4+CD25+ regulatory T cells. Nat Med 11(11):1238–1243 [DOI] [PubMed] [Google Scholar]
  410. Zhang Y, Louboutin JP, Zhu J, Rivera AJ, Emerson SG. 2002. Preterminal host dendritic cells in irradiated mice prime CD8+ T cell-mediated acute graft-versus-host disease. J Clin Invest 109(10):1335–1344 [DOI] [PMC free article] [PubMed] [Google Scholar]
  411. Zheng XX, Steele AW, Hancock WW, Kawamoto K, Li XC, Nickerson PW, Li Y, Tian Y, Strom TB. 1999. IL-2 receptor-targeted cytolytic IL-2/Fc fusion protein treatment blocks diabetogenic autoimmunity in nonobese diabetic mice. J Immunol 163(7):4041–4048 [PubMed] [Google Scholar]
  412. Zhu EF, Gai SA, Opel CF, Kwan BH, Surana R, Mihm MC, Kauke MJ, Moynihan KD, Angelini A, Williams RT, Stephan MT, Kim JS, Yaffe MB, Irvine DJ, Weiner LM, Dranoff G, Wittrup KD. 2015. Synergistic innate and adaptive immune response to combination immunotherapy with anti-tumor antigen antibodies and extended serum half-life IL-2. Cancer Cell 27(4):489–501 [DOI] [PMC free article] [PubMed] [Google Scholar]
  413. Zimmerman RJ, Aukerman SL, Katre NV, Winkelhake JL, Young JD. 1989. Schedule dependency of the antitumor activity and toxicity of polyethylene glycol-modified interleukin 2 in murine tumor models. Cancer Res 49(23):6521–6528 [PubMed] [Google Scholar]

Articles from Journal of Interferon & Cytokine Research are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES