Skip to main content
Antibody Therapeutics logoLink to Antibody Therapeutics
. 2021 Jun 25;4(2):123–133. doi: 10.1093/abt/tbab014

Next-generation cytokines for cancer immunotherapy

Diyuan Xue 1,2, Eric Hsu 3, Yang-Xin Fu 3,, Hua Peng 1,
PMCID: PMC8271143  PMID: 34263141

Abstract

Most studies focus on the first and second signals of T cell activation. However, the roles of cytokines in immunotherapy are not fully understood, and cytokines have not been widely used in patient care. Clinical application of cytokines is limited due to their short half-life in vivo, severe toxicity at therapeutic doses, and overall lack of efficacy. Several modifications have been engineered to extend their half-life and increase tumor targeting, including polyethylene glycol conjugation, fusion to tumor-targeting antibodies, and alteration of cytokine/cell receptor-binding affinity. These modifications demonstrate an improvement in either increased antitumor efficacy or reduced toxicity. However, these cytokine engineering strategies may still be improved further, as each strategy poses advantages and disadvantages in the delicate balance of targeting tumor cells, tumor-infiltrating lymphocytes, and peripheral immune cells. This review focuses on selected cytokines, including interferon-α, interleukin (IL)-2, IL-15, IL-21, and IL-12, in both preclinical studies and clinical applications. We review next-generation designs of these cytokines that improve half-life, tumor targeting, and antitumor efficacy. We also present our perspectives on the development of new strategies to potentiate cytokine-based immunotherapy.

Keywords: cytokines, immunotherapy, protein engineering, tumor targeting


STATEMENT OF SIGNIFICANCE

Clinical application of cytokines is limited due to their short half-life in vivo and severe toxicity at therapeutic doses. Here, we review several modifications of commonly used cytokines that have been engineered to extend their half-life and increase tumor targeting, we summarize their potential issues and newer designs of engineered cytokines.

INTRODUCTION

Cytokines are potent immune-modulating protein molecules that are used in treating cancer [1, 2]. Cytokines stimulate the function, survival, and proliferation of natural killer (NK) and T cells that mediate immune responses against tumors. The discovery of potent antitumor activity of cytokine therapy in animal models has prompted the evaluation of the potential application of some immune molecules for clinical cancer therapy. Such cytokines include interferon (IFN)-α, interleukin (IL)-2, IL-15, IL-21, and IL-12. IL-2 was the first Food and Drug Administration (FDA)-approved cytokine for treating metastatic renal cell cancer and advanced melanoma [3, 4]. IFN-α has been approved for the treatment of several human cancers [5–8]. However, in most patients, systemic administration of cytokines has limited efficacy in clinical trials due to their short half-life and severe adverse effects before reaching therapeutic doses [6, 9–12]. Novel strategies to improve cytokine antitumor effects as monotherapy or combination therapy for both preclinical and clinical applications will be discussed in this review.

Type I IFN-α

Type I IFNs, including IFN-α, IFN-β, IFN-ε, IFN-κ, and IFN-ν, are a family of monomeric cytokines with multiple functions [13]. IFN-α regulates the expression of various genes that modulate tumor cell growth, proliferation, apoptosis, and immune checkpoint-mediated immune suppression [14–18]. Several studies also have shown that the type I IFNs play critical roles in tumor control by promoting dendritic cell (DC) cross-priming to (re-) activate T cells [19–21]. IFN-α was FDA approved to treat hematological malignancies and melanoma at high doses [5–8].

Following the clinical success of IFN-α in cancer treatment, multiple strategies have been tested to address the limitations of IFN-α and further improve its clinical efficacy and safety (Table 1). To minimize filtration of IFN-α through the kidney prior to reaching a therapeutic dose, IFN-α requires a longer half-life. One IFN-α variant addresses this issue by conjugating polyethylene glycol (PEG) to IFN-α. PEGylation can cover the domain of IFN-α, which binds to its receptor to minimize peripheral IFN-α activity and uptake into off-target, nontumor tissues. PEGylated IFN-α has a comparable spectrum of biological activity to IFN-α, but with an approximately 10-fold longer plasma half-life, thus allowing for less frequent administration and patient burden. These significant benefits resulting from PEGylation of IFN-α have resulted in its approval as an adjuvant treatment of melanoma [22]. However, the type I IFN receptor is widely distributed on all nucleated cells including those in nontumor tissue, which suggests that PEGylated IFN-α can still induce toxic side effects [23]. Adjuvant therapy with PEGylated IFN-α2b has been associated with severe host toxicity, including fatigue (97 patients, 16%), hepatotoxicity (66, 11%), and depression (39, 6%). In all, 37% of patients discontinued adjuvant therapy because of these adverse toxicities [24]. These studies suggest that lack of tumor-targeted release of PEGylated IFN-α may ultimately limit positive clinical results.

Table 1.

Engineered IFN-α variants

Drug name Reference Drug features Drug benefits Clinical phase
Peginterferon alfa-2b Eggermont et al. [24] PEG conjugated to IFN-α at IFN/IFNR-binding interface Increases half-life and reduces peripheral toxicity with equivalent antitumor efficacy to IFN-α III
Anti-CD20/IFN-α Xuan et al. [30]; Liao et al. [31] Anti-CD20 conjugated to mouse IFN-α Kills lymphomas that express CD20 through adaptive immunity or direct IFN-α-mediated killing Preclinical
TAK-573 Collins et al. [33] Anti-CD38 conjugated to attenuated IFN-α Binds to CD38+ cells with high affinity; increases circulating IFN-associated cytokine levels in human patients I/II
IFN-α/anti-PDL1 Liang et al. [37] Anti-PDL1 conjugated to IFN-α Induces IFN-mediated upregulation of PDL1, which is then targeted to create feedforward antitumor immunity Preclinical

Various strategies have been developed to increase IFN-α delivery to tumor sites to improve efficacy with reduced toxicity. Fusing therapeutic cytokines to tumor-targeting antibodies or antibody fragments can promote the localization of such treatments within the tumor, thus minimizing cytokine therapy induce toxicity. IFNs have been conjugated to monoclonal antibodies (mAbs) that recognize and bind to tumor-associated proteins such as epidermal growth factor receptor (EGFR), Human Epidermal Growth Factor Receptor 2 (HER2) Erb-B2 Receptor Tyrosine Kinase 2 (ERBB2), cluster of differentiation (CD)20, CD38, CD138, and Vascular Endothelial Growth Factor (VEGF) receptor. These cytokine/antibody fusions may induce direct antiproliferative effects from both the conjugated IFNs and the mAbs [25–29]. A preclinical study discussing the treatment of a B cell lymphoma xenograft model that overexpresses human-CD20 (38C13-hCD20) reported that type I IFN linked to an anti-CD20 antibody increased the antitumor effects of type I IFN by directly killing off IFN-α-sensitive 38C13-hCD20 tumor cells [30]. Because many lymphomas are resistant to direct anti-CD20- and/or IFN-mediated apoptosis, other studies aimed to determine whether targeting tumors with IFN-α could mobilize the adaptive immune system for tumor control. Using a syngeneic anti-CD20- and IFN-α-resistant A20 B cell lymphoma tumor model, one study demonstrated that treating this lymphoma with a conjugated anti-CD20/IFN-α fusion protein abolished B cell lymphoma resistance to anti-CD20 while limiting IFN-associated systemic toxicity in the host. Mechanistically, the anti-CD20/IFN-α fusion employs tumor cells as the dominant antigen presenting cell (APC) for the reactivation of Cytotoxic T lymphocytes (CTLs), which ultimately induces potent antitumor efficacy [31].

Alternatively, conjugation of IFNs muteins with reduced receptor-binding affinity with high-affinity tumor-targeting antibodies can further induce preferential localization of such fusion proteins to the tumor and thus limit their consumption in circulation. Despite its fusion to a tumor-targeting modality, tumor-targeting antibody/wild-type IFN-α fusions can still induce toxicity through ubiquitously expressed IFN-α receptors (IFNAR) on nontumor cells. Furthermore, supposed tumor-associated antigens are not absolutely specific to tumors and can be expressed on nontumor cells, which take up and thus hijack the conjugated immunocytokines. An attenuated form of IFN-α was reported to reduce cell toxicity in the forms of the MLepR-targeting nanobody/IFN [32] and the anti-CD38/IFN-α fusions [27]. The anti-CD38/attenuated IFN-α fusion protein displays a 10 000-fold greater binding-specificity to CD38-positive (tumor) vs. CD38-negative (normal) cells than native IFN-α. In contrast, the corresponding wild-type IFN-α fusion protein showed only a 40-fold greater binding affinity. Therefore, the attenuating mutation in the IFN-α portion of the immunocytokine decreases IFN-α antigen specificity by approximately 250-fold. Treatment of established human multiple myeloma tumor (NCI-H929 xenograft model) bearing mice with CD38-targeting attenuated IFN-α leads to the complete elimination of such tumors. Patients may thus potentially be more safely treated with higher doses of the anti-CD38/attenuated IFN-α than native IFN-α or a “more conventional” IFN-α immunocytokine (anti-CD38/wild-type IFN-α). TAK-573 is a fusion protein of a humanized anti-CD38 IgG4 mAb with two attenuated IFN alpha-2b molecules. Ongoing phase I/II TAK-573-1501 clinical studies in patients with relapsed/refractory multiple myeloma (NCT03215030) indicate that TAK-573-treated patients exhibit an increased type I IFN gene signature and circulating IFN-associated cytokine levels. This preliminary biomarker data indicate that TAK-573 is a pharmacologically active molecule that mediates its effect through IFNAR pathway modulation [33].

IFNs are also the cytokines that most potently induce Programmed death ligand 1 (PD-L1) expression, which subsequently dampens Tc ell responses against the tumor via a negative feedback effect [34–36]. Activation of IFN signaling in the tumor microenvironment (TME) can therefore synergize with PD-L1 blockade therapy against advanced tumors by inducing more robust T cell activation with the absence of PD-L1 inhibition of T cells. Furthermore, the anti-PD-L1 antibody can be conjugated to immunomodulatory molecules to deliver them specifically into tumor tissues with minimal toxicity. Finally, IFN-α-armed anti-PD-L1 creates multiple feedforward responses that increase targeting effects to enhance responses to IFN-α treatment, thereby maximizing antitumor effects [37]. Nevertheless, this immunocytokine, when administered systematically, is still at risk for being taken up by peripheral IFN receptor-expressing cells, and its application might be improved by utilizing a mutant Fc that does not have antibody-dependent cellular cytotoxicity capability or incorporating an IFN activity blocking mask to even further minimize toxicity.

Common γ chain cytokines: IL-2, IL-15, and IL-21

The common γ chain cytokine family exerts numerous functions on T lymphocyte survival, function, and proliferation. This family consists of six members, including IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Most of these cytokines function through activating janus kinase (JAK)1/JAK3 and the downstream signaling of signal transducer and activator of transcription (STAT)1, STAT3, STAT5, MAPK, and PI3K/also known as Protein Kinase B (AKT).

IL-2

IL-2 was identified as a 15.5 kDa glycoprotein in the 1970s and is mainly produced by activated CD4+ T cells [38, 39]. As a potent inducer of cytotoxic T cells and NK cells, IL-2 was one of the first FDA-approved immunocytokines for metastatic melanoma and renal cell carcinoma [3, 4]. Clinical IL-2 immunotherapy has not been widely applied due to its short in vivo half-life, severe toxicity at therapeutic doses, and induction of immunosuppressive responses through regulatory T cell (Treg) expansion [10–12]. Many strategies for addressing these limitations have been implemented, such as fusing the Fc domains of immunoglobulins or PEG molecules to increase half-life, modifying IL-2 function by introducing targeted mutations, fusing IL-2 with antibodies that target the cytokine to the TME, masking IL-2 against Treg binding, and synthesizing tumor-associated protease-activated IL-2 prodrugs (Table 2).

Table 2.

Engineered IL-2 variants

Drug name Reference Drug features Drug benefits Clinical phase
NKTR-214 Charych et al. [45] PEG conjugated to IL-2 at IL-2/IL-2Rα-binding interface Increases half-life, reduces peripheral toxicity, increases affinity towards CD8 T cells II/III
IL-2 mutein (CD25) Carmenate et al. [51] Includes mutations that decrease IL-2 affinity for IL-2Rα Reduces IL-2-mediated toxicity and inhibits metastases Preclinical
IL-2 superkine Levin et al. [52] Includes mutations that increase IL-2 affinity for IL-2Rβ Reduces IL-2-mediated toxicity, more potently inhibits primary tumor growth Preclinical
NHS-IL2LT (Selectikine) Gillessen et al. [58] Antinecrotic tumor core conjugated to IL-2 mutein Exhibits very low toxicity in clinical trials I/II
CEA-IL2v Klein et al. [59] IL-2 mutein with decreased affinity for IL-2Rα, conjugated to anti-CEA Decreased mortality in preclinical tumor models, but was abandoned due to lack of efficacy in clinical trials II
DI-Leu16-IL2 Lansigan et al. [60] Anti-CD20 conjugated to IL-2 Induces several complete responses in lymphoma patients with few side effects I/II
SumIL2/Anti-EGFR Sun et al. [61] Anti-EGFR conjugated to CD8 preferential IL-2 mutein Inhibits tumor growth, can eliminate metastases when combined with surgery Preclinical
ALKS 4230 Lopes et al. [62] IL-2Rα conjugated to IL-2 Reduces peripheral toxicity and inhibits melanoma metastases preclinically Preclinical
IL-2/mAb complexes Krieg et al. [63] Antibody conjugated to IL-2 at IL-2/IL-2Rα-binding interface Unable to bind endogenous IL-2Rα, inhibits tumor growth with reduced toxicity Preclinical
Protease-activated IL-2 prodrug Hsu et al. [67] CD8 preferential IL-2 mutein masked with tumor protease removable IL-2Rβ Reduces peripheral toxicity, inhibits tumor growth, can eliminate metastases when combined with surgery Preclinical
Neo-2/15 Silva et al. [85] Computationally designed IL-2 variant with increased affinity for IL-2Rβ Mimics natural signaling of IL-2 with reduced peripheral toxicity I

The IL-2 receptor is a heterotrimeric complex formed by three subunits: the IL-2Rα, IL-2Rβ, and IL-2Rγ chains, also known as CD25, CD122, and CD132, respectively. IL-2 exerts stimulatory and regulatory functions by binding to IL-2 receptor subunits. Naïve CD8+ T cells and CD4+/CD8+ memory T cells express a medium-affinity dimeric receptor IL-2Rβ/IL-2Rγ and lack the IL-2Rα chain. When the IL-2Rα chain is also present in the receptor complex, IL-2 is bound with high affinity. IL-2Rα is highly expressed on Treg cells, thus increasing the affinity of IL-2 for Tregs over cytotoxic lymphocytes at lower doses. Therefore, high doses of IL-2 are immunostimulatory, but low doses are immunosuppressive [40–42].

Several second-generation IL-2 variants have been engineered to reduce affinity to IL-2Rα [43, 44] or increase IL-2Rβ affinity. IL-2Rα is highly expressed on pulmonary endothelial cells, a large percentage of Foxp3-negative CD4 T cells in humans, Treg cells, and some innate lymphoid cells. One strategy is by conjugating PEG to IL-2. NKTR-214 is a prodrug of human IL-2 that is PEGylated on the IL-2Rα-binding site, in which six PEG residues are slowly released to make an active and stable form of human IL-2. As the PEG molecules conjugated to IL-2 blocks IL-2 interaction with the IL-2Rα-binding motif, the binding of PEG-IL-2 to Treg cells is inhibited, whereas the binding of IL-2 to receptors on CD8+ T cells is not affected. PEG modulation also increases the half-life of IL-2 [45–47]. NKTR-214 significantly increased the ratio of CD8+ T cell to Treg cells in murine melanoma models and led to improved antitumor efficacy compared with recombinant IL-2 (Aldesleukin). However, NKTR-214 has limited efficacy as a monotherapy in solid tumors. On the other hand, the increased levels of the PD-1 protein on tumor-infiltrating T cells and PD-L1 on cancer cells in patients suggested that combination with immune checkpoint inhibitors might boost the antitumor efficacy of IL-2 variants. Clinically, NKTR-214 combined with nivolumab induced increased systemic and intratumor CD8+ T cell responses with no expansion of Tregs in the tumor and achieved encouraging overall response rates compared with nivolumab alone, according to immunohistochemistry data. Unfortunately, it has been reported that about seven in 10 humans have raised circulating anti-PEG antibodies. This high prevalence of anti-PEG antibodies may potentially limit the efficacy of NKTR-214 [48]. Lack of tumor-targeted release of PEGylated cytokines could result in toxicity before reaching therapeutic doses [49]. Moreover, most PEGylated proteins are prepared through nonsite-specific PEGylation, which may affect the consistency of manufacture and clinical administration of PEGylated IL-2 [45, 50].

Another approach to shifting IL-2 affinity towards CD8+ T cells is to synthesize IL-2 mutants with different binding properties for the IL-2 subunits. IL-2 exhibits a higher affinity towards Tregs over effector T cells due to the expression of IL-2Rα on Tregs. Therefore, the two major mutagenesis strategies to overcome IL-2-mediated immunosuppression involve decreasing IL-2 affinity for IL-2Rα or increasing IL-2 affinity for IL-2Rβ. One recent IL-2Rα affinity altering mutant has been observed to reduce IL-2-mediated toxicity while reducing metastatic disease. This mutant IL-2 was designed to mutate wild-type IL-2 on residues R38, F42, Y45, and E62. These residues, which all interface with IL-2Rα, were substituted with alanine. Although further in-depth in vivo primary tumor studies would provide a more comprehensive assessment of the efficacy of these muteins, this study still demonstrates that reducing IL-2 affinity for IL-2Rα can contribute to increased antitumor efficacy [51]. Other muteins, such as the IL-2 “superkine,” include mutants that significantly increase IL-2 affinity for IL-2Rβ [52]. The evolved mutations in the IL-2 superkine elicit potent phosphorylation of STAT5 and vigorous proliferation of T cells irrespective of IL-2Rα expression. Compared with IL-2, the IL-2 superkine induced superior expansion of cytotoxic T cell and NK, leading to improved antitumor responses in vivo. The IL-2 superkine also proportionally reduced the expansion of Treg cells. However, increasing binding affinity for IL-2Rβ still activates peripheral NK/T cells, which may lead to increased risk of toxicity at therapeutic doses [53].

To enhance IL-2 delivery to tumor sites, antibody-based tumor-targeted delivery of IL-2 has been attempted by many groups. IL-2 was fused to antibodies against different tumor antigens, such as EpCAM, CD20, ch14.18 (the human-mouse chimeric variant of 14.18) recognizing ganglioside D2 (GD2) and F8, L19, and F16 recognizing tumor angiogenesis markers. These IL-2/antibody fusions significantly improve antitumor effects with low toxicity than either antibody or cytokine monotherapy in both syngeneic mouse models and a human melanoma xenograft model. Therapeutic effects were mediated by both NK cells and CD8+ T cells, and reduced IL-2 retention in peripheral blood led to lower toxicity [54–56]. However, despite their improved tumor-targeting ability, these IL-2/antibody fusions can still lack potent antitumor efficacy [57]. As these fusion proteins still contain native IL-2, they may still exhibit increased binding and activation of Tregs over effector T cells. Multiple groups have thus combined the IL-2/IL-2R mutation and tumor-targeting antibody fusion strategies. Merck KGaA developed NHS-IL2LT (with LT standing for low toxicity), which contains an IL-2 (D20T) variant fused with an antibody (NHS76) that targets the necrotic core of tumors. NHS-IL2LT showed the lowest toxicity in clinical trials among all IL-2 variants to date [58]. Roche Pharma developed CEA-IL2v, which fuses an IL-2 mutein with an antibody that targets carcinoembryonic antigen (CEA). The mutein includes IL-2Rα affinity reducing mutations F42A, Y45A, and L72G. CEA-IL2v also induces significantly decreased mortality in a mouse CEA overexpressing colon cancer model compared with a native IL-2/CEA antibody fusion [59]. However, CEA-IL2v was abandoned due to lack of efficacy as monotherapy and in combination with check-point inhibitors (CPI) in clinical trials. In contrast, DI-Leu16-IL2 (anti-CD20 fused to wild-type IL2), when given subcutaneously, has resulted in several complete responses at doses with little or no side effects in a phase I dose-escalation study [60]. Another IL-2/antibody fusion, termed SumIL2, was invented by incorporating both F42A (an IL-2Rα affinity reducing mutation) and the IL-2 superkine mutations to reduce Treg binding and increase binding CD8+ T cells at the meanwhile [61]. SumIL2 is fused to an EGFR-targeting antibody. This fusion protein induced much more potent antitumor efficacy against murine cancers that overexpressed EGFR than SumIL2 fused to a nonexpressed tumor-associated antigen in preclinical models. This SumIL2/anti-EGFR fusion also preferentially binds to CD8+ T cells instead of Treg cells, resulting in improved tumor control over fusion proteins containing only F42A or IL-2 superkine [61]. However, EGFR is widely expressed in human tissues. Systemic delivery of the SumIL2/anti-EGFR fusion protein may still result in some off-target effects despite its high EGFR tumor-targeting capability.

Because IL-2 can complex with cell surface IL-2Rα and subsequently bind with high affinity to IL-2Rβγ, some groups aim to mask the amino acids on IL-2 that would normally first bind to IL-2Rα. One “natural” mask complex links IL-2 to IL-2Rα via a glycine/serine amino acid linker. In addition to masking IL-2 from endogenous cell surface IL-2Rα, soluble IL2Rα would also stabilize the structure of IL-2 for binding to IL-2Rβ. This approach was used to create the IL-2/IL-2Rα complex denoted ALKS 4230. ALKS 4230 has been observed to reduce peripheral toxicity and inhibit B16 melanoma metastases more effectively than wild-type IL-2 in mice [62]. Another example of a masked IL-2 involves complexing IL-2 to an IL-2 mAb. The investigators specifically used an IL-2 antibody that strongly binds to the IL2Rα-binding region of IL-2, thus creating an IL-2/mAb complex that is unable to bind to endogenous IL2Rα. Similar to the IL-2 superkine, this IL2Rβ preferential IL-2/mAb complex could more potently expand CD8 T cells and inhibit B16 primary tumor growth compared with wild-type IL-2 [63, 64]. In addition to the natural complex, this same group also rearranged the α-helices of IL-2 to graft IL-2 into the mAb. This grafted IL-2/mAb complex even more effectively induces antitumor immunity, expanding more CD8 T cells and fewer Tregs to more potently eliminate 4T1 and B16 metastases [65].

Like the IL-2/mask complexes, a protease-activated IL-2 is also a fusion protein that includes a mask. However, IL-2 is linked to the mask by a protease cleavable linker. The choice of the protease cleavage substrate is sensitive and specific for a protease that is preferentially overexpressed in the tumor. One group developed two protease-activated IL-2 fusion proteins that incorporate IL-2, IL-2Rα, and a cleavable linker specific to either prostate-specific antigen or matrix metalloproteinase (MMP). This fusion protein was able to inhibit peritoneal tumor cell growth in a mouse model [66]. Another group synthesized a protease-activated IL-2 fusion protein that incorporates multiple IL-2 enhancing strategies, including using the SumIL2 mutation to reduce Treg binding, fusion to Fc to increase half-life, and MMP cleavage activation in the tumor to reduce toxicity. This fusion protein reduced pulmonary and liver toxicity than the equivalent “cleaved” SumIL2 without compromising tumor-infiltrating lymphocyte (TIL) expansion and antitumor efficacy [67]. In summary, next-generation IL-2 variants can reduce toxicity while increasing TILs, which allows for more effective control of both primary and metastatic tumors.

IL-15

IL-15 is mainly produced by activated myeloid cells, such as monocytes, macrophages, and DCs [68]. When bound to the transmembrane IL-15Rα, IL-15 is transpresented to NK cells and T cells-expressing IL-2/IL-15Rβ and the common γ chain receptor. Notably, IL-15 is critical for NK cell development and the homeostasis of memory CD8+ T cells [69, 70]. IL-15 also affects other types of immune cells, such as innate lymphoid cells [71], further highlighting its role in potentiating the immune response. Preclinical observations strongly support the antitumor activity of IL-15 mediated by NK cells and T lymphocytes [72, 73]. More importantly, unlike IL-2, IL-15 does not stimulate Tregs since IL-15 does not bind to the IL-2Rα chain, which is required for the formation of the high-affinity receptor complex (IL-2Rαβγ) on Tregs to stimulate immunosuppressive signals [74].

A major challenge to clinical application of IL-15 is its short half-life and insufficient effectiveness in vivo [75, 76]. Preclinical studies indicate that the IL-15/IL-15Rα dimer, rather than the IL-15 monomer, is more bioactive when transpresented to NK and CD8+ memory T cells [77–79]. Fusion protein RLI, which consists of IL-15 linked to the cytokine-binding (sushi) domain of IL-15Rα, displayed super-agonistic activity towards the IL-15Rβ/γ complex and exerted potent antitumor properties in vivo [80]. ALT-803, another variant of IL-15 that encompasses human IL-15 covalently linked to the sushi domain of human IL-15Rα fused to an IgG1 Fc domain, has been tested in clinical trials. In the dose-escalation phase I clinical study with this recombinant protein, 33 patients with hematological cancer received ALT-803. In all, 19% of patients met the criteria for clinical benefit [81]. However, ALT-803 did not induce significantly better patient outcomes than DI-Leu16-IL2 [60]. Another clinical trial involves combining ALT-803 therapy with anti-PD-1 therapy in 21 patients with metastatic Non-small-cell lung carcinoma (NSCLC). In all, 29% of patients achieved an objective response with a tolerable safety profile [82]. However, the response rate has no advantage compared with the historical response rates of immune checkpoint blockade (ICB) alone [83]. All patients showed increased circulating NK and CD8+ T cells in the above clinical trials, indicating that IL-15-associated toxicity may exist at certain high doses [84].

In order to reduce IL-15/IL-15Rα dimer toxicity, several novel IL-15 variants have been developed and tested in preclinical studies. For example, Neo-2/15 is a computationally designed de novo protein mimic of IL-2 and IL-15 that binds to human and mouse IL-2Rβγ chains but not to IL-2Rα or IL-15Rα. Neo-2/15 can mimic the natural signaling function but does not carry the adverse effects induced by IL-2Rα or IL-15Rα binding. Neo-2/15 has been demonstrated to exhibit potent therapeutic activity with reduced toxicity in murine tumors [85]. Other clinical studies involving IL-15 proteins combined with other immunotherapies are ongoing, such as recombinant IL-15 with both anti-CTLA-4 and anti-PD-1 therapy, with a CD40 agonist, and with mAbs including anti-CD52 and anti-CD20 antibodies [71, 86].

IL-21

IL-21 is a 4α-helix bundle cytokine produced by follicular helper T (Tfh) cells, T helper 17 (Th17) cells, and natural killer T (NKT) cells. IL-21 signals via heterodimerization with the IL-21 receptor (IL-21R) [87, 88] and the common cytokine receptor γ-chain. Functional IL-21R is broadly expressed in hematopoietic cells, including T and B lymphocytes, NK cells, and myeloid cells [89]. IL-21 exerts pleiotropic functions by supporting the differentiation of CD4+ Th17 [90–92] and Tfh cells [93], facilitating the maturation and enhancing the cytotoxicity of CD8+ T cells and NK cells, and promoting the differentiation of memory CD8+ T cells [88, 94–96]. In addition, IL-21, in contrast to IL-2, blunts Treg expansion by suppressing Foxp3 expression and favors the enrichment of antigen-stimulated CD8+ T cells [97].

Clinical phase I/II studies have shown that nontumor targeting free IL-21 induces objective responses or disease stabilization in a fraction of metastatic melanoma and renal cancer patients [98–101]. However, systemic IL-21 therapy fails to achieve adequate concentrations in the TME to activate TILs due to its short half-life and peripheral consumption, which necessitates a high dose that induces significant toxicity. Local intratumoral administration of recombinant IL-21 is difficult to administer for most patients. Low patient responses to IL-21 limit its further administration in the clinic [102]. To address this limitation, immunocytokine fusion proteins have been generated. By conjugating IL-21 to tumor or T cell-targeting antibodies, IL-21 can be delivered directly to markers, such as PD-1 on T cells, which are more highly expressed in the tumor. Such conjugations can counterbalance the high affinity of a cytokine to ubiquitously expressed cytokine receptor on immune cells in nontumor tissues and therefore decrease toxicity [103]. An anti-PD-1/IL-21 mutant fusion protein, which incorporates a highly attenuated IL-21 mutein (R9E: R76A) fused to a PD-1 antibody, was designed to restrict cytokine activity to the targeted PD-1+ cells and exhibits an improved in vivo half-life [101]. This anti-PD-1/IL-21 fusion exhibits antitumor efficacy in a humanized mouse model that is refractory to anti-PD-1 monotherapy. Alternatively, it has also been shown that anti-CD20 and anti-EGFR fusions to IL-21 prolong the half-life of IL-21 and enhance its antitumor efficacy in non-Hodgkin lymphoma and colon adenocarcinoma models [104, 105]. Compared with free IL-21, systemic administration of anti-EGFR/IL-21 improves safety by enabling targeted delivery to EGFR positive tumor tissue. This anti-EGFR/IL-21 fusion protein can also synergistically combine with ICB therapy. ICB can “release the brakes” of host immune responses, but may have a limited effect on the rapid expansion of functional TILs. Providing T cell growth factors, for example, tumor-targeting IL-21, may more directly expand TILs. Mechanistically, anti-EGFR/IL-21 can increase antigen-specific CD8+ T function and proliferation, especially the PD-1intTim-3 population. In preclinical models, combining anti-EGFR/IL-21 therapy with ICB therapy achieved significantly increased antitumor effects and decreased mortality compared with any single treatment.

IL-12

IL-12 is a heterodimer composed of two independent subunits linked together by disulfide bonds: α (IL-12p35) and β (IL-12p40). IL-12 is mainly produced by activated APCs such as DCs, macrophages, monocytes, and B cells. IL-12 binds to the IL-12 receptor (IL-12R), which is expressed as a high-affinity heterodimer of IL-12Rβ1 and IL-12Rβ2 subunits, on T cells and NK cells. The β1 subunit is constitutively expressed on immune cells, whereas expression of the β2 subunit is upregulated in T cells and NK cells upon activation [106]. In preclinical studies, systemic administration of recombinant mouse IL-12 elicited potent antitumor effects in various mouse models. IL-12 stimulates the effector functions of activated T cells and NK cells via induction of cytotoxic enzymes and cytokines, such as IFN-gamma (IFN-γ), which are required for potent antitumor immunity [107, 108].

However, systemic delivery of IL-12 in the clinic at therapeutic doses has been limited due to its short half-life in vivo, severe side effects, and lack of tumor targeting [109, 110]. In clinical studies, antitumor effects of IL-12 have been evaluated in numerous malignancies such as cutaneous T cell lymphoma, acquired immunodeficiency syndrome (AIDS)-related Kaposi sarcoma, and non-Hodgkin’s lymphoma. However, frequent systemic administrations of IL-12 have been associated with severe host toxicity through the undesirable release of proinflammatory cytokines from peripheral T and NK cells. Therefore, various strategies have been implemented to improve efficacy with reduced toxicity (Table 3). In current clinical trials, intratumoral delivery of IL-12 via plasmid or virus vectors has been observed to generate systemic adaptive immune responses, activate or reactivate tumor-infiltrating CD8+ T cells, and control both primary tumors and metastases [111, 112].

Table 3.

Engineered IL-12 variants

Drug name Reference Drug features Drug benefits Clinical phase
IL12-GEMy Kaczanowska et al. [119] Genetically engineered myeloid cells that express IL-12 Reverses immune suppression in the premetastatic niche, reduces primary and metastatic tumor burden through T and NK cell activation Preclinical
CAR-T-expressing IL-12 Zhang et al. [122] Tumor-infiltrating lymphocytes engineered to secrete IL-12 Induces objective responses in metastatic melanoma patients, but induces toxicity I/II
CBD-IL-12 Mansurov et al. [123] Collagen-binding domain conjugated to IL-12 Localizes to exposed collagen on tumor vasculature, induces sustained levels of IFN-γ in TME, reduces toxicity Preclinical
NHS-IL-12 Xu et al. [126] DNA/DNA–histone complex antibody conjugated to IL-12 Combines with anti-PDL1 to potently expand and activate T cells in tumor tissue, expands TCR diversity of tumor-infiltrating T cells in human patients I/II
IL-12 mutein partial agonists Glassman et al. [128] Includes mutations that decrease IL-12 affinity for IL-12Rβ1 Preferentially supports CD8 T cell function over NK cells, producing potent antitumor immunity with reduced toxicity Preclinical

Many preclinical studies have used IL-12-transduced cells for local cancer therapy. DCs [113–115] and macrophages [116–118] have been engineered to express IL-12. Transduction of these APCs increases their ability to induce robust antitumor immune responses. In a recent study, genetically engineered myeloid cells (GEMys)-expressing IL-12 (IL12-GEMy) successfully reversed immunosuppression in the premetastatic niche and reduced metastatic and primary tumor burden through T and NK cell activation [119]. Adoptive T cell therapy has potent efficacy against hematologic malignancies, but has yet to achieve clinical benefit against solid tumors. Engineering Chimeric antigen receptor T (CAR-T) cells to express IL-12 using an Nuclear factor of activated T-cells (NFAT)-responsive promoter is under investigation to improve antitumor efficacy against solid tumors [120, 121]. Etxeberria et al. [122] also engineered tumor-specific CD8+ T cells that transiently express IL-12. Preclinically, when injected intratumorally but not intravenously, these engineered T cells induced to complete rejections not only of the injected lesion but also of distant concomitant tumors [123]. Further improvement of this strategy is necessary to achieve longer-lasting tumor control and systemic administration to avoid the difficulties involved with local intratumoral injection.

In addition, tumor-targeting antibody fragment/IL-12/Fc fusions have been synthesized to increase IL-12 half-life and delivery to tumor sites. Indeed, tumor-targeting antibody/IL-12 fusion proteins selectively accumulate in tumors following intravenous administration in multiple murine tumor models [124, 125]. Mechanistically, the fusion protein alone can stimulate effective CD8+ T cell-mediated antitumor immune responses, activate NK cells, and induce IFN-γ production and STAT4 phosphorylation [124, 125]. More importantly, reducing IL-12 retention in peripheral tissues and blood leads to less adverse weight loss. Finally, the fusion protein controls both primary and metastatic tumor growth more effectively than either antibody or cytokine monotherapy.

A variety of cytokines, including IL-12 immunocytokines, are also administrated in combination with ICB therapy. Because simply removing suppression by ICB may not restore sufficient immunity against tumors, additional T cell cytokines may be necessarily to expand effector TILs and sustain antitumor immunity. For example, the administration of NHS-muIL-12, which incorporates an antibody (NHS76) recognizing DNA/DNA–histone complexes fused with two molecules of murine IL-12, with avelumab (an anti-PDL1 antibody) expanded CD8+ T cells and enhanced T cell activation in tumor tissue more than either agent alone [126]. Furthermore, these combined treatments increased proliferation of cytotoxic NK and CD8+ T cells, T-bet expression, plasma cytokine levels, and messenger RNA levels of innate and adaptive immune genes [127]. Preliminary clinical trials report increased NK cell frequencies in the peripheral blood and broadened T-cell receptor (TCR) diversity of tumor-infiltrating T cells in patients receiving NHS-IL-12. In addition, five out of 59 patients experienced stable disease conditions, and a dose-tolerated safety profile was also reported [128].

Nevertheless, such immunocytokine fusions retain complete cytokine activity in circulation, allowing them to interact with circulating lymphocytes and induce cytokine-mediated toxicities. In another recent study, a set of IL-12 mutations were screened out to preferentially activate CD8+ T cells but not NK cells [129]. Based on critical differences in IL-12Rβ1 expression between activated CD8+ T cells and NK cells, Glassman et al. [129] introduced a series of alanine mutations in murine p40 to reduce the affinity to IL-12Rβ1. These IL-12 partial agonists preferentially support T cell function and preserve IFN-γ induction by CD8+ T cells while impairing cytokine production from NK cells in vitro. Finally, the IL-12 partial agonists produced potent antitumor immunity with reduced toxicity relative to IL-12. Further investigation of the potential immunogenicity of a nonendogenous mutation may assist in the evaluation of the therapeutic potential of this IL-12 variant. Taken together, systemic administration of IL-12 is still challenging in the clinic because of many unwanted adverse effects.

Next-generation pro-cytokines in cancer immunotherapy

All of the modified cytokines mentioned above are innovatively designed to reduce toxicity and improve the antitumor efficacy of systemically delivered cytokine drugs. Many biotechnology and pharmaceutical companies have conducted multiple clinical trials for cancer therapies with these modified cytokines [49, 58, 60, 130]. Balancing maximum cytokine activity and minimum peripheral toxicity is crucial for clinical cytokine immunotherapy. Tumor-targeted therapy has been explored to solve this problem, but the potential expression of the targeted antigens in normal tissue, even at low levels, often results in off-target effects. In addition, immunogenicity is a general issue for all muteins and conjugated nonendogenous proteins. Promising next-generation cytokine prodrugs (“pro-cytokines” [67]) may overcome the above obstacles in multiple mouse tumor models. Ideal pro-cytokines mask cytokine activity with blocking polypeptides, such as natural cytokine receptor-binding domains, to achieve minimal toxicity. Using natural cytokine receptors minimizes immunogenicity to the host without compromising tight control over the reduction in peripheral cytokine activity. More extended receptor domains potentially block cytokines more effectively but might reduce efficacy, whereas shorter domains may enhance efficacy but increase toxicity. Moreover, different receptor domains can be linked to cytokine subunits with a specific peptide sequence selectively cleavable by proteases that are overexpressed in the TME. This strategy masks the toxic activity of the cytokine until it enters tumor tissue, thus providing effective “passive” tumor targeting. Although tumors may enter a dormant phase, metastasis is always accompanied by the production of various tumor-specific proteases, which may indicate increased activation of protease activated cytokines in more advanced cancers [131]. In addition, different types of tumors may contain distinguished proteases, which can be evaluated for use in pro-cytokine engineering and design. Therefore, personalized pro-cytokines may potentially significantly improve patient outcomes. This concept is similar to the technology of antibody-directed enzyme prodrug therapy (ADEPT). However, in ADEPT designs, either extraordinarily high or specific expression of tumor antigens is required. In contrast, pro-cytokines target membrane-type proteases, for example, MMP 14, a membrane-bound MMP expressed on tumor cells and inflammatory cells, is greatly enriched inside the TME of almost all tumors [132, 133]. The membrane-bound property of such proteases also decreases the potential leakage of pro-cytokines out of tumor tissue after entering the TME. These tumor-specific membrane-bound MMPs therefore increase antitumor efficacy and tumor targeting of pro-cytokines. Moreover, the pro-cytokine concept is highly flexible and easily applicable to different cytokines and fusion proteins. Altogether, multiple approaches to reducing toxicity without compromising antitumor efficacy generate a diverse portfolio of therapeutic modalities, holding considerable promise for translation into effective antitumor immunotherapy.

DATA AVAILABILITY STATEMENT

All data included in this review are available upon request by contact with the corresponding author.

CONFLICT OF INTEREST STATEMENT

The authors declared that they have no conflicts of interest to this work.

FUNDING

None declared

References

  • 1.Chen, DS, Mellman, I. Elements of cancer immunity and the cancer–immune set point. Nature 2017; 541: 321–30. [DOI] [PubMed] [Google Scholar]
  • 2.Waldmann, TA. Cytokines in cancer immunotherapy. Cold Spring Harb Perspect Biol 2018; 10: a028472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Rosenberg, SA, Yang, JC, White, DEet al. . Durability of complete responses in patients with metastatic cancer treated with high-dose interleukin-2: identification of the antigens mediating response. Ann Surg 1998; 228: 307–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Rosenberg, SA. IL-2: the first effective immunotherapy for human cancer. J Immunol 2014; 192: 5451–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Golomb, HM, Jacobs, A, Fefer, Aet al. . Alpha-2 interferon therapy of hairy-cell leukemia: a multicenter study of 64 patients. J Clin Oncol 1986; 4: 900–5. [DOI] [PubMed] [Google Scholar]
  • 6.Solal-Celigny, P, Lepage, E, Brousse, Net al. . Recombinant interferon alfa-2b combined with a regimen containing doxorubicin in patients with advanced follicular lymphoma. N Engl J Med 1993; 329: 1608–14. [DOI] [PubMed] [Google Scholar]
  • 7.Kirkwood, JM, Strawderman, MH, Ernstoff, MSet al. . Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: the Eastern Cooperative Oncology Group Trial EST 1684. J Clin Oncol 1996; 14: 7–17. [DOI] [PubMed] [Google Scholar]
  • 8.Groopman, JE, Gottlieb, MS, Goodman, Jet al. . Recombinant alpha-2 interferon therapy for Kaposi’s sarcoma associated with the acquired immunodeficiency syndrome. Ann Intern Med 1984; 100: 671–6. [DOI] [PubMed] [Google Scholar]
  • 9.Baldo, P, Rupolo, M, Compagnoni, Aet al. . Interferon-alpha for maintenance of follicular lymphoma. Cochrane Database Syst Rev 2010; 1: CD004629. [DOI] [PubMed] [Google Scholar]
  • 10.Skrombolas, D, Frelinger, JG. Challenges and developing solutions for increasing the benefits of IL-2 treatment in tumor therapy. Expert Rev Clin Immunol 2014; 10: 207–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Panelli, MC, White, R, Foster, Met al. . Forecasting the cytokine storm following systemic interleukin (IL)-2 administration. J Transl Med 2004; 2: 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chavez, ARV, Buchser, W, Basse, PHet al. . Pharmacologic administration of interleukin-2. Ann N Y Acad Sci 2009; 1182: 14–27. [DOI] [PubMed] [Google Scholar]
  • 13.Pestka, S, Krause, CD, Walter, MR. Interferons, interferon-like cytokines, and their receptors. Immunol Rev 2004; 202: 8–32. [DOI] [PubMed] [Google Scholar]
  • 14.Brassard, DL, Grace, MJ, Bordens, RW. Interferon-α as an immunotherapeutic protein. J Leukoc Biol 2002; 71: 565–81. [PubMed] [Google Scholar]
  • 15.Balkwill, F, Watling, D, Taylor-Papadimitriou, J. Inhibition by lymphoblastoid interferon of growth of cells derived from the human breast. Int J Cancer 1978; 22: 258–65. [DOI] [PubMed] [Google Scholar]
  • 16.Hobeika, AC, Subramaniam, PS, Johnson, HM. IFNα induces the expression of the cyclin-dependent kinase inhibitor p21 in human prostate cancer cells. Oncogene 1997; 14: 1165–70. [DOI] [PubMed] [Google Scholar]
  • 17.Xiao, W, Klement, JD, Lu, Cet al. . IFNAR1 controls autocrine type I IFN regulation of PD-L1 expression in myeloid-derived suppressor cells. J Immunol 2018; 201: 264–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Chawla-Sarkar, M, Lindner, DJ, Liu, YFet al. . Apoptosis and interferons: role of interferon-stimulated genes as mediators of apoptosis. Apoptosis 2003; 8: 237–49. [DOI] [PubMed] [Google Scholar]
  • 19.Deng, L, Liang, H, Xu, Met al. . STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 2014; 41: 843–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ren, Z, Guo, J, Liao, Jet al. . CTLA-4 limits anti-CD20–mediated tumor regression. Clin Cancer Res 2017; 23: 193–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Sistigu, A, Yamazaki, T, Vacchelli, Eet al. . Cancer cell–autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat Med 2014; 20: 1301–9. [DOI] [PubMed] [Google Scholar]
  • 22.Herndon, TM, Demko, SG, Jiang, Xet al. . U.S. Food and Drug Administration approval: peginterferon‐alfa‐2b for the adjuvant treatment of patients with melanoma. Oncologist 2012; 17: 1323–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Dunn, GP, Koebel, CM, Schreiber, RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol 2006; 6: 836–48. [DOI] [PubMed] [Google Scholar]
  • 24.Eggermont, AM, Suciu, S, Santinami, Met al. . Adjuvant therapy with pegylated interferon alfa-2b versus observation alone in resected stage III melanoma: final results of EORTC 18991, a randomised phase III trial. Lancet 2008; 372: 117–26. [DOI] [PubMed] [Google Scholar]
  • 25.Li, Z, Zhu, Y, Li, Cet al. . Anti-VEGFR2-interferon-α2 regulates the tumor microenvironment and exhibits potent anti-tumor efficacy against colorectal cancer. Oncoimmunology 2017; 6: e1290038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Pelham, JM, Gray, JD, Flannery, GRet al. . Interferon-alpha conjugation to human osteogenic sarcoma monoclonal antibody 791T/36. Cancer Immunol Immunother 1983; 15: 210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Pogue, SL, Taura, T, Bi, Met al. . Targeting attenuated interferon-α to myeloma cells with a CD38 antibody induces potent tumor regression with reduced off-target activity. PLoS One 2016; 11: e0162472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Huang, TH, Chintalacharuvu, KR, Morrison, SL. Targeting IFN-alpha to B cell lymphoma by a tumor-specific antibody elicits potent antitumor activities. J Immunol 2007; 179: 6881–8. [DOI] [PubMed] [Google Scholar]
  • 29.Yang, X, Zhang, X, Fu, MLet al. . Targeting the tumor microenvironment with interferon-β bridges innate and adaptive immune responses. Cancer Cell 2014; 25: 37–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Xuan, C, Steward, KK, Timmerman, JMet al. . Targeted delivery of interferon-alpha via fusion to anti-CD20 results in potent antitumor activity against B-cell lymphoma. Blood 2010; 115: 2864–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Liao, J, Luan, Y, Ren, Zet al. . Converting lymphoma cells into potent antigen-presenting cells for interferon-induced tumor regression. Cancer Immunol Res 2017; 5: 560–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Garcin, G, Paul, F, Staufenbiel, Met al. . High efficiency cell-specific targeting of cytokine activity. Nat Commun 2014; 5: 3016. [DOI] [PubMed] [Google Scholar]
  • 33.Collins, S, Joshi, A, Shen, Let al. . 357 TAK-573, an anti-CD38–attenuated interferon alpha (IFNα) fusion protein (Attenukine™), has demonstrated IFNα receptor (IFNAR) pathway modulation in patients with relapsed/refractory multiple myeloma. J Immunother Cancer 2020; 8: A382–2. [Google Scholar]
  • 34.Blank, C, Brown, I, Peterson, ACet al. . PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res 2004; 64: 1140–5. [DOI] [PubMed] [Google Scholar]
  • 35.Mühlbauer, M, Fleck, M, Schütz, Cet al. . PD-L1 is induced in hepatocytes by viral infection and by interferon-α and -γ and mediates T cell apoptosis. J Hepatol 2006; 45: 520–8. [DOI] [PubMed] [Google Scholar]
  • 36.Bald, T, Landsberg, J, Lopez-Ramos, Det al. . Immune cell–poor melanomas benefit from PD-1 blockade after targeted type I IFN activation. Cancer Discov 2014; 4: 674–87. [DOI] [PubMed] [Google Scholar]
  • 37.Liang, Y, Tang, H, Guo, Jet al. . Targeting IFNα to tumor by anti-PD-L1 creates feedforward antitumor responses to overcome checkpoint blockade resistance. Nat Commun 2018; 9: 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Leonard, WJ. Cytokines and immunodeficiency diseases. Nat Rev Immunol 2001; 1: 200–8. [DOI] [PubMed] [Google Scholar]
  • 39.Liao, W, Lin, J-X, Leonard, WJ. Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. Immunity 2013; 38: 13–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Maloy, KJ, Powrie, F. Fueling regulation: IL-2 keeps CD4+ Treg cells fit. Nat Immunol 2005; 6: 1071. [DOI] [PubMed] [Google Scholar]
  • 41.Fontenot, JD, Rasmussen, JP, Gavin, MAet al. . A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nat Immunol 2005; 6: 1142–51. [DOI] [PubMed] [Google Scholar]
  • 42.Boyman, O, Sprent, J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol 2012; 12: 180–90. [DOI] [PubMed] [Google Scholar]
  • 43.Heaton, KM, Ju, G, Grimm, EA. 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 1993; 53: 2597–602. [PubMed] [Google Scholar]
  • 44.Mott, HR, Baines, BS, Hall, RMet al. . The solution structure of the F42A mutant of human interleukin 2. J Mol Biol 1995; 247: 979–94. [DOI] [PubMed] [Google Scholar]
  • 45.Charych, DH, Hoch, U, Langowski, JLet al. . NKTR-214, an engineered cytokine with biased IL2 receptor binding, increased tumor exposure, and marked efficacy in mouse tumor models. Clin Cancer Res 2016; 22: 680–90. [DOI] [PubMed] [Google Scholar]
  • 46.Charych, D, Khalili, S, Dixit, Vet al. . Modeling the receptor pharmacology, pharmacokinetics, and pharmacodynamics of NKTR-214, a kinetically-controlled interleukin-2 (IL2) receptor agonist for cancer immunotherapy. PLoS One 2017; 12: e0179431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Garber, K. Cytokine resurrection: engineered IL-2 ramps up immuno-oncology responses [J]. Nature biotechnology 2018; 36: 378–380. [DOI] [PubMed] [Google Scholar]
  • 48.Chen, BM, Su, YC, Chang, CJet al. . Measurement of pre-existing IgG and IgM antibodies against polyethylene glycol in healthy individuals. Anal Chem 2016; 88: 10661. [DOI] [PubMed] [Google Scholar]
  • 49.Bentebibel, SE, Hurwitz, ME, Bernatchez, Cet al. . A first-in-human study and biomarker analysis of NKTR-214, a novel IL2Rβγ-biased cytokine, in patients with advanced or metastatic solid tumors. Cancer Discov 2019; 9: 711–21. [DOI] [PubMed] [Google Scholar]
  • 50.Jonathan, D, Mark, D. Site-specific PEGylation of therapeutic proteins. Int J Mol Sci 2015; 16: 25831–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Carmenate, T, Pacios, A, Enamorado, Met al. . Human IL-2 mutein with higher antitumor efficacy than wild type IL-2. J Immunol 2013; 190: 6230–8. [DOI] [PubMed] [Google Scholar]
  • 52.Levin, AM, Bates, DL, Ring, AMet al. . Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine’. Nature 2012; 484: 529–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Yan, L, Strick-Marchand, H, Lim, AIet al. . Regulatory T cells control toxicity in a humanized model of IL-2 therapy. Nat Commun 2017; 8: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Gutbrodt, KL, Schliemann, C, Giovannoni, Let al. . Antibody-based delivery of interleukin-2 to neovasculature has potent activity against acute myeloid leukemia. Sci Transl Med 2013; 5: 201ra118. [DOI] [PubMed] [Google Scholar]
  • 55.Becker, JC, Varki, N, Gillies, SDet al. . An antibody-interleukin 2 fusion protein overcomes tumor heterogeneity by induction of a cellular immune response. Proc Natl Acad Sci U S A 1996; 93: 7826–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Yang, RK, Kalogriopoulos, NA, Rakhmilevich, ALet al. . Intratumoral treatment of smaller mouse neuroblastoma tumors with a recombinant protein consisting of IL-2 linked to the Hu14.18 antibody increases intratumoral CD8+ T and NK cells and improves survival. Cancer Immunol Immunother 2013; 62: 1303–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Weiss, T, Puca, E, Silginer, Met al. . Immunocytokines are a promising immunotherapeutic approach against glioblastoma. Sci Transl Med 2020; 12: eabb2311. [DOI] [PubMed] [Google Scholar]
  • 58.Gillessen, S, Gnad-Vogt, US, Gallerani, Eet al. . A phase I dose-escalation study of the immunocytokine EMD 521873 (Selectikine) in patients with advanced solid tumours. Eur J Cancer 2013; 49: 35–44. [DOI] [PubMed] [Google Scholar]
  • 59.Klein, C, Waldhauer, I, Nicolini, VGet al. . Cergutuzumab amunaleukin (CEA-IL2v), a CEA-targeted IL-2 variant-based immunocytokine for combination cancer immunotherapy: overcoming limitations of aldesleukin and conventional IL-2-based immunocytokines. Onco Targets Ther 2017; 6: e1277306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lansigan, F, Nakamura, R, Quick, Det al. . Phase I/II study of an anti-CD20-interleukin-2 immunocytokine DI-Leu16-IL2 in patients with relapsed B-cell lymphoma (NHL). J Clin Oncol 2016; 34: e19046. [Google Scholar]
  • 61.Sun, Z, Ren, Z, Yang, Ket al. . A next-generation tumor-targeting IL-2 preferentially promotes tumor-infiltrating CD8+ T-cell response and effective tumor control. Nat Commun 2019; 10: 3874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lopes, JE, Fisher, JL, Flick, HLet al. . ALKS 4230: a novel engineered IL-2 fusion protein with an improved cellular selectivity profile for cancer immunotherapy. J Immunother Cancer 2020; 8: e000673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Krieg, C, Letourneau, S, Pantaleo, Get al. . Improved IL-2 immunotherapy by selective stimulation of IL-2 receptors on lymphocytes and endothelial cells. Proc Natl Acad Sci U S A 2010; 107: 11906–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Boyman, O, Kovar, M, Rubinstein, MPet al. . Selective stimulation of T cell subsets with antibody-cytokine immune complexes. Science 2006; 311: 1924–7. [DOI] [PubMed] [Google Scholar]
  • 65.Sahin, D, Arenas-Ramirez, N, Rath, Met al. . An IL-2-grafted antibody immunotherapy with potent efficacy against metastatic cancer. Nat Commun 2020; 11: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Puskas, J, Skrombolas, D, Sedlacek, Aet al. . Development of an attenuated interleukin-2 fusion protein that can be activated by tumour-expressed proteases. Immunology 2011; 133: 206–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hsu, EJ, Cao, X, Moon, Bet al. . A cytokine receptor-masked IL2 prodrug selectively activates tumor-infiltrating lymphocytes for potent antitumor therapy. Nat Commun 2021; 12: 2768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Bamford, R, Battiata, A, Waldmann, T. IL-15: the role of translational regulation in their expression. J Leukoc Biol 1996; 59: 476–80. [DOI] [PubMed] [Google Scholar]
  • 69.Kennedy, MK, Glaccum, M, Brown, SNet al. . Reversible defects in natural killer and memory CD8 T cell lineages in interleukin 15–deficient mice. J Exp Med 2000; 191: 771–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lodolce, JP, Boone, DL, Chai, Set al. . IL-15 receptor maintains lymphoid homeostasis by supporting lymphocyte homing and proliferation. Immunity 1998; 9: 669–76. [DOI] [PubMed] [Google Scholar]
  • 71.Waldmann, TA, Miljkovic, MD, Conlon, KC. Interleukin-15 (dys) regulation of lymphoid homeostasis: implications for therapy of autoimmunity and cancer. J Exp Med 2020; 1: 217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Evans, R, Fuller, JA, Christianson, Get al. . 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 1997; 179: 66–73. [DOI] [PubMed] [Google Scholar]
  • 73.Klebanoff, CA, Finkelstein, SE, Surman, DRet al. . IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc Natl Acad Sci 2004; 101: 1969–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Marshall, D, Sinclair, C, Tung, Set al. . Differential requirement for IL-2 and IL-15 during bifurcated development of thymic regulatory T cells. J Immunol 2014; 193: 5525–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Conlon, KC, Lugli, E, Welles, HCet al. . 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 2015; 33: 74–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Miller, JS, Morishima, C, McNeel, DGet al. . A first-in-human phase I study of subcutaneous outpatient recombinant human IL15 (rhIL15) in adults with advanced solid tumors. Clin Cancer Res 2018; 24: 1525–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Dubois, S, Patel, HJ, Zhang, Met al. . Preassociation of IL-15 with IL-15Rα-IgG1-Fc enhances its activity on proliferation of NK and CD8+/CD44highT cells and its antitumor action. J Immunol 2008; 180: 2099–106. [DOI] [PubMed] [Google Scholar]
  • 78.Kobayashi, H, Dubois, S, Sato, Net al. . Role of trans-cellular IL-15 presentation in the activation of NK cell–mediated killing, which leads to enhanced tumor immunosurveillance. Blood 2005; 105: 721–7. [DOI] [PubMed] [Google Scholar]
  • 79.Castillo, EF, Schluns, KS. Regulating the immune system via IL-15 transpresentation. Cytokine 2012; 59: 479–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Mortier, E, Quéméner, A, Vusio, Pet al. . Soluble interleukin-15 receptor α (IL-15Rα)-sushi as a selective and potent agonist of IL-15 action through IL-15Rβ/γ. J Biol Chem 2006; 281: 1612–9. [DOI] [PubMed] [Google Scholar]
  • 81.Romee, R, Cooley, S, Berrien-Elliott, MMet al. . First-in-human phase 1 clinical study of the IL-15 superagonist complex ALT-803 to treat relapse after transplantation. Blood 2018; 131: 2515–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Wrangle, JM, Velcheti, V, Patel, MRet al. . ALT-803, an IL-15 superagonist, in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1b trial. Lancet Oncol 2018; 19: 694–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Onoi, K, Chihara, Y, Uchino, Jet al. . Immune checkpoint inhibitors for lung cancer treatment: a review. J Clin Med 2020; 9: 1362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Guo, Y, Luan, L, Rabacal, Wet al. . IL-15 superagonist–mediated immunotoxicity: role of NK cells and IFN-γ. J Immunol 2015; 195: jimmunol.1500300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Silva, D-A, Yu, S, Ulge, UYet al. . De novo design of potent and selective mimics of IL-2 and IL-15. Nature 2019; 565: 186–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Waldmann, TA, Dubois, S, Miljkovic, MDet al. . IL-15 in the combination immunotherapy of cancer. Front Immunol 2020; 11: 868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ozaki, K, Kikly, K, Michalovich, Det al. . Cloning of a type I cytokine receptor most related to the IL-2 receptor beta chain. Proc Natl Acad Sci 2000; 97: 11439–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Parrish-Novak, J, Dillon, SR, Nelson, Aet al. . Interleukin 21 and its receptor are involved in NK cell expansion and regulation of lymphocyte function. Nature 2000; 408: 57–63. [DOI] [PubMed] [Google Scholar]
  • 89.Spolski, R, Leonard, WJ. Interleukin-21: basic biology and implications for cancer and autoimmunity. Annu Rev Immunol 2008; 26: 57–79. [DOI] [PubMed] [Google Scholar]
  • 90.Nurieva, R, Yang, XO, Martinez, Get al. . Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature 2007; 448: 480–3. [DOI] [PubMed] [Google Scholar]
  • 91.Zhou, L, Ivanov, II, Spolski, Ret al. . IL-6 programs TH-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol 2007; 8: 967–74. [DOI] [PubMed] [Google Scholar]
  • 92.Korn, T, Bettelli, E, Gao, Wet al. . IL-21 initiates an alternative pathway to induce proinflammatory TH 17 cells. Nature 2007; 448: 484–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Eto, D, Lao, C, DiToro, Det al. . IL-21 and IL-6 are critical for different aspects of B cell immunity and redundantly induce optimal follicular helper CD4 T cell (Tfh) differentiation. PLoS One 2011; 6: e17739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Tian, Y, Cox, MA, Kahan, SMet al. . A context-dependent role for IL-21 in modulating the differentiation, distribution, and abundance of effector and memory CD8 T cell subsets. J Immunol 2016; 196: 2153–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Allard, EL, Hardy, MP, Leignadier, Jet al. . Overexpression of IL-21 promotes massive CD8+ memory T cell accumulation. Eur J Immunol 2007; 37: 3069–77. [DOI] [PubMed] [Google Scholar]
  • 96.Zeng, R, Spolski, R, Finkelstein, SEet al. . Synergy of IL-21 and IL-15 in regulating CD8+ T cell expansion and function. J Exp Med 2005; 201: 139–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Li, Y, Yee, C. IL-21–mediated Foxp3 suppression leads to enhanced generation of antigen-specific CD8+ cytotoxic T lymphocytes. Blood 2008; 111: 229–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Davis, ID, Skrumsager, BK, Cebon, Jet al. . An open-label, two-arm, phase I trial of recombinant human interleukin-21 in patients with metastatic melanoma. Clin Cancer Res 2007; 13: 3630–6. [DOI] [PubMed] [Google Scholar]
  • 99.Steele, N, Anthony, A, Saunders, Met al. . A phase 1 trial of recombinant human IL-21 in combination with cetuximab in patients with metastatic colorectal cancer. Br J Cancer 2012; 106: 793–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Timmerman, JM, Byrd, JC, Andorsky, DJet al. . A phase I dose-finding trial of recombinant interleukin-21 and rituximab in relapsed and refractory low grade B-cell lymphoproliferative disorders. Clin Cancer Res 2012; 18: 5752–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Schmidt, H, Brown, J, Mouritzen, Uet al. . Safety and clinical effect of subcutaneous human interleukin-21 in patients with metastatic melanoma or renal cell carcinoma: a phase I trial. Clin Cancer Res 2010; 16: 5312–9. [DOI] [PubMed] [Google Scholar]
  • 102.Petrella, TM, Mihalcioiu, CLD, McWhirter, Eet al. . Final efficacy results of NCIC CTG IND.202: a randomized phase II study of recombinant interleukin-21 (rIL21) in patients with recurrent or metastatic melanoma (MM). J Clin Oncol 2013; 31: 9032–2. [Google Scholar]
  • 103.Tzeng, A, Kwan, BH, Opel, CFet al. . Antigen specificity can be irrelevant to immunocytokine efficacy and biodistribution. Proc Natl Acad Sci 2015; 112: 3320–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Bhatt, S, Parvin, S, Zhang, Yet al. . Anti-CD20-interleukin-21 fusokine targets malignant B cells via direct apoptosis and NK-cell–dependent cytotoxicity. Blood 2017; 129: 2246–56. [DOI] [PubMed] [Google Scholar]
  • 105.Deng, S, Sun, Z, Qiao, Jet al. . Targeting tumors with IL-21 reshapes the tumor microenvironment by proliferating PD-1intTim-3–CD8+ T cells. JCI Insight 2020; 5: 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Szabo, SJ, Dighe, AS, Gubler, Uet al. . Regulation of the interleukin (IL)-12R β2 subunit expression in developing T helper 1 (Th1) and Th2 cells. J Exp Med 1997; 185: 817–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Vignali, DA, Kuchroo, VK. IL-12 family cytokines: immunological playmakers. Nat Immunol 2012; 13: 722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Tugues, S, Burkhard, SH, Ohs, Iet al. . New insights into IL-12-mediated tumor suppression. Cell Death Differ 2015; 22: 237–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Cohen, J. IL-12 deaths: explanation and a puzzle. Science 1995; 270: 908–8. [DOI] [PubMed] [Google Scholar]
  • 110.Leonard, JP, Sherman, ML, Fisher, GLet al. . Effects of single-dose interleukin-12 exposure on interleukin-12–associated toxicity and interferon-γ production. Blood 1997; 90: 2541–8. [PubMed] [Google Scholar]
  • 111.Anandaroop, M, Wright, J, Shirley, Set al. . Characterization of abscopal effects of intratumoral electroporation-mediated IL-12 gene therapy. Gene Ther 2018; 26: 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Sangro, B, Melero, I, Qian, Cet al. . Gene therapy of cancer based on interleukin 12. Curr Gene Ther 2005; 5: 573–81. [DOI] [PubMed] [Google Scholar]
  • 113.Yoshida, M, Jo, JI, Tabata, Y. Augmented anti-tumor effect of dendritic cells genetically engineered by interleukin-12 plasmid DNA. J Biomater Sci Polym Ed 2010; 21: 659–75. [DOI] [PubMed] [Google Scholar]
  • 114.Tatsumi, T, Takehara, T, Yamaguchi, Set al. . Injection of IL-12 gene-transduced dendritic cells into mouse liver tumor lesions activates both innate and acquired immunity. Gene Ther 2007; 14: 863–71. [DOI] [PubMed] [Google Scholar]
  • 115.Tatsumi, T, Huang, J, Gooding, WEet al. . Intratumoral delivery of dendritic cells engineered to secrete both interleukin (IL)-12 and IL-18 effectively treats local and distant disease in association with broadly reactive Tc1-type immunity. Cancer Res 2003; 63: 6378. [PubMed] [Google Scholar]
  • 116.Satoh, T, Saika, T, Ebara, Set al. . Macrophages transduced with an adenoviral vector expressing interleukin 12 suppress tumor growth and metastasis in a preclinical metastatic prostate cancer model. Cancer Res 2003; 63: 7853–60. [PubMed] [Google Scholar]
  • 117.Moyes, KW, Lieberman, NAP, Kreuser, SAet al. . Genetically engineered macrophages: a potential platform for cancer immunotherapy. Hum Gene Ther 2017; 28: 200–15. [DOI] [PubMed] [Google Scholar]
  • 118.Brempelis, KJ, Cowan, CM, Kreuser, SAet al. . Genetically engineered macrophages persist in solid tumors and locally deliver therapeutic proteins to activate immune responses. J Immunother Cancer 2020; 8: e001356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Kaczanowska, S, Beury, DW, Gopalan, Vet al. . Genetically engineered myeloid cells rebalance the core immune suppression program in metastasis. Cell 2021; 184: 2033–52.e21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Zhang, L, Kerkar, SP, Yu, Zet al. . Improving adoptive T cell therapy by targeting and controlling IL-12 expression to the tumor environment. Mol Ther 2011; 19: 751–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kerkar, SP, Muranski, P, Kaiser, Aet al. . Tumor-specific CD8+ T cells expressing interleukin-12 eradicate established cancers in lymphodepleted hosts. Cancer Res 2010; 70: 6725–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Etxeberria I, Bolaños E, Quetglas JI, et al. Intratumor adoptive transfer of IL-12 mRNA transiently engineered antitumor CD8+ T cells. Cancer Cell 2019; 36: 613–629. [DOI] [PubMed] [Google Scholar]
  • 123.Zhang, L, Morgan, RA, Beane, JDet al. . Tumor-infiltrating lymphocytes genetically engineered with an inducible gene encoding interleukin-12 for the immunotherapy of metastatic melanoma. Clin Cancer Res 2015; 21: 2278–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Mansurov, A, Ishihara, J, Hosseinchi, Pet al. . Collagen-binding IL-12 enhances tumour inflammation and drives the complete remission of established immunologically cold mouse tumours. Nat Biomed Eng 2020; 4: 531–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Venetz, D, Koovely, D, Weder, Bet al. . Targeted reconstitution of cytokine activity upon antigen binding using split cytokine antibody fusion proteins. J Biol Chem 2016; 291: 18139–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Jonathan, KF, Vandeveer, AJ, Schlom, Jet al. . Enhanced antitumor effects by combining an IL-12/anti-DNA fusion protein with avelumab, an anti-PD-L1 antibody. Oncotarget 2017; 8: 20558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Xu, C, Zhang, Y, Rolfe, PAet al. . Combination therapy with NHS-muIL12 and avelumab (anti-PD-L1) enhances antitumor efficacy in preclinical cancer models. Clin Cancer Res 2017; 23: 5869–80. [DOI] [PubMed] [Google Scholar]
  • 128.Strauss, J, Heery, CR, Kim, JWet al. . First-in-human phase I trial of a tumor-targeted cytokine (NHS-IL12) in subjects with metastatic solid tumors. Clin Cancer Res 2019; 25: 99–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Glassman, CR, Mathiharan, YK, Jude, KMet al. . Structural basis for IL-12 and IL-23 receptor sharing reveals a gateway for shaping actions on T versus NK cells. Cell 2021; 184: 983–99.e24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Margolin, K, Atkins, MB, Dutcher, JPet al. . Phase I trial of BAY 50-4798, an interleukin-2–specific agonist in advanced melanoma and renal cancer. Clin Cancer Res 2007; 13: 3312–9. [DOI] [PubMed] [Google Scholar]
  • 131.Kessenbrock, K, Plaks, V, Werb, Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell 2010; 141: 52–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.McGowan, P, Duffy, M. Matrix metalloproteinase expression and outcome in patients with breast cancer: analysis of a published database. Ann Oncol 2008; 19: 1566–72. [DOI] [PubMed] [Google Scholar]
  • 133.Turunen, SP, Tatti-Bugaeva, O, Lehti, K. Membrane-type matrix metalloproteases as diverse effectors of cancer progression. Biochim Biophys Acta Mol Cell Res 2017; 1864: 1974–88. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

All data included in this review are available upon request by contact with the corresponding author.


Articles from Antibody Therapeutics are provided here courtesy of Oxford University Press

RESOURCES