The clinical development of cytokines as cancer therapeutics has been limited due to significant toxicities generally observed with systemic administration. This narrow therapeutic window, together with relatively modest efficacy, has made natural cytokines unattractive drug candidates. Immunocytokines represent a class of next-generation cytokines designed to overcome the challenges associated with traditional cytokines. These agents seek to improve the therapeutic index of cytokines by using antibodies as vehicles for the targeted delivery of immunomodulatory agents within the local tumor microenvironment (TME). Various molecular formats and cytokine payloads have been studied. In this review, we provide an overview of the rationale, preclinical support, and current clinical development strategies for immunocytokines.

Cytokines are potent small molecules involved in cell–cell interactions and in mediating host immune responses. They play important functions in anticancer immunity through modulation of various cellular responses, including the regulation of immune proliferation, differentiation, and effector functions, thus making them longstanding targets for cancer therapy (1, 2). However, deploying natural cytokines clinically has been challenging. Despite being the first form of cancer immunotherapy to receive regulatory approval - IFNα in 1986 and IL2 in 1992 - there has been little subsequent progress in clinical development of cytokine-based therapeutics (3, 4). This may be attributable to several factors. The pleiotropic activity of cytokines affects multiple immune compartments, some without beneficial antitumor response. Furthermore, the precise mechanisms underlying tumor regressions with cytokine administration are generally unknown. Thus, biomarkers are not available to guide dose or scheduling. Finally, cytokines have traditionally been administered at extremely high levels in early studies. Such dosing may bypass or obscure normal physiologic antitumor mechanisms and lead to unwanted toxicity. To address these limitations, various modifications have been made to: (i) increase tumor targeting, (ii) enhance binding selectivity, and (iii) improve the pharmacokinetic profile of these agents (5).

In recent years, improved understanding of cytokine immunobiology and enhancements in protein engineering have sparked renewed interest in this arena (2, 6). “Next generation cytokines” have since been developed, including immunocytokines. Immunocytokines are antibody-based immunoconjugates, consisting of monoclonal antibodies or antibody fragments targeting a tumor antigen or component of the tumor microenvironment (TME) fused with cytokine payloads (Fig. 1; ref. 7). These agents seek to improve the therapeutic index of cytokines by using antibodies as vehicles for the targeted delivery of immunomodulatory agents within the local TME Fig. 2; ref. 8).

Figure 1.

Schematic representation of immunocytokines currently in clinical trials (based on antibodies in IgG format) comprised of antibody, linker, and cytokine payload. Other antibody formats include monovalent, F(ab′), divalent, F(ab′)2, scFv, diabodies, and triabodies. Antibody targets for immunocytokines include tumor associated antigens, components of the ECM, tumor DNA, and T cells. Tumor antigen targets including EpCAM, GD2, HER2, EGFR, CD20, CEA, and FAP. ECM targets include tenascin-C and fibronectin extradomain-A and -B, T-cell targets include PD-1 and PD-L1, and DNA targets include DNA/histone complex. Numerous cytokine payloads, including IL2, IL12, TNFα, IL15, and IL21, are being studied. (Adapted from an image created with BioRender.com.)

Figure 1.

Schematic representation of immunocytokines currently in clinical trials (based on antibodies in IgG format) comprised of antibody, linker, and cytokine payload. Other antibody formats include monovalent, F(ab′), divalent, F(ab′)2, scFv, diabodies, and triabodies. Antibody targets for immunocytokines include tumor associated antigens, components of the ECM, tumor DNA, and T cells. Tumor antigen targets including EpCAM, GD2, HER2, EGFR, CD20, CEA, and FAP. ECM targets include tenascin-C and fibronectin extradomain-A and -B, T-cell targets include PD-1 and PD-L1, and DNA targets include DNA/histone complex. Numerous cytokine payloads, including IL2, IL12, TNFα, IL15, and IL21, are being studied. (Adapted from an image created with BioRender.com.)

Close modal
Figure 2.

Targets of immunocytokines within the TME. Various approaches have been explored, including efforts to target (i) tumor cell-surface antigens, (ii) ECM targets, (iii) DNA damage targets, and (iv) T-cell targets. (Adapted from an image created with BioRender.com.)

Figure 2.

Targets of immunocytokines within the TME. Various approaches have been explored, including efforts to target (i) tumor cell-surface antigens, (ii) ECM targets, (iii) DNA damage targets, and (iv) T-cell targets. (Adapted from an image created with BioRender.com.)

Close modal

In this review, we provide an overview of the development of immunocytokines as novel cancer immunotherapies. We outline key factors in immunocytokine design, from tumor targeting components to molecular formats and cytokine payloads, emphasizing the clinical translation of such developments. Finally, we review challenges in the clinical development of these agents and summarize important future directions in the field.

One key component of an immunocytokine is the target antigen (9). Common targets have been tumor-associated antigens expressed on the cell surface of tumor cells. One early-target was epithelial cell adhesion molecule (EpCAM), a transmembrane glycoprotein overexpressed in various epithelial cancers (e.g., colon or prostate) and linked to tumor growth and metastases (10). Another target, disialoganglioside 2 (GD2), is a glycosphingolipid highly expressed on neuroectoderm-derived tumors (e.g., melanoma and neuroblastoma), with limited expression in normal tissues (11). Immunocytokines have also been developed to target HER2, CD20, and carcinoembryonic antigen (CEA), among others (Table 1). One limitation in targeting tumor-associated antigens (TAA) is that target expression can be transient and/or heterogeneous. Furthermore, only select patients may express the target of interest, potentially limiting populations most likely to benefit.

Table 1.

Select immunocytokines in clinical trials.

Cytokine
NamepayloadAntigenIndicationTreatmentNCTStudy status
L19-IL2 IL2 EDB Melanoma Combination with dacarbazine NCT02076646, Phase I/II Not recruiting 
(Darleukin)    Monotherapy NCT01253096, Phase II Completed 
    Combination with dacarbazine NCT01055522, Phase II Terminated 
    Monotherapy NCT02938299, Phase III Recruiting 
   Oligometastatic Combination with radiotherapy NCT02086721, Phase I Completed 
   Solid tumor Monotherapy NCT01058538, Phase I Completed 
   B-cell lymphoma Combination with rituximab NCT02957019, Phase I/II Not recruiting 
   Pancreatic Combination with gemcitabine NCT01198522, Phase I Terminated 
   NSCLC Combination with radiotherapy NCT03705403, Phase II Recruiting 
F16-IL2 IL2 Tenascin C AML Combination with cytarabine NCT02957032, Phase I Not recruiting 
(Teleukin)    Combination with BI 836858 NCT03207191, Phase I Completed 
   Solid tumors Combination with doxorubicin NCT01131364, Phase I/II Terminated 
    Combination with paclitaxel NCT01134250, Phase I/II Completed 
   Merkel cell Combination with paclitaxel NCT02054884, Phase II Terminated 
DI-Leu16-IL2 IL2 CD20 B-cell NHL Monotherapy, following rituximab blood NCT00720135, Phase I Completed 
    B-cell depletion NCT02151903. Phase I/II Terminated 
     NCT01874288, Phase I/II Terminated 
Hu14.18-IL2 IL2 GD2 Neuroblastoma Monotherapy NCT00003750, Phase I Completed 
   Melanoma Combination with ICI, radiotherapy NCT03958383, Phase I/II Suspended 
    Monotherapy NCT00590824, Phase II Completed 
    Monotherapy NCT00109863, Phase II Completed 
   Neuroblastoma and osteosarcoma Combination with NK cells NCT03209869. Phase I Withdrawn 
   Neuroblastoma Combination with sargramostim and isotretinoin NCT01334515, Phase II Completed 
    Monotherapy NCT00082758, Phase II Completed 
    Combination with chemo, ICI, NK cells NCT01576692, Phase I Completed 
EMD27306 (Tucotuzumab celmoleukin) IL2 EpCAM EpCAM-positive ovarian, prostate CRC or NSCLC Combination with cyclophosphamide NCT00132522, Phase I Completed 
huKS-IL2, EpCAM G733-2   Refractory kidney bladder, lung Monotherapy NCT00016237, Phase I Completed 
RO6874281 (RG7461; Simlukafusp alfa) IL2v FAP Breast, head, and neck solid tumors Combination with trastuzumab or cetuximab NCT02627274, Phase I   Completed 
   RCC Combination with ICI ± bevazicumab NCT03063762, Phase I Completed 
   Head and neck, esophageal and cervical cancers Combination with atezolizumab NCT03386721, Phase II Completed 
   Melanoma Combination with pembrolizumab NCT03875079, Phase I Completed 
   Pancreatic Multiple ICI combinations NCT03193190, Phase I/II Recruiting 
RO6895882 (Cergutuzumab amunaleukin) IL2v CEA Solid CEA positive tumors Monotherapy NCT02004106, Phase I Completed 
   Solid tumors Combination with atezolizumab NCT02350673, Phase I Completed 
EMD521873 (Selectikine) IL2v DNA/histone complex NSCLC Combination with radiotherapy NCT00879866, Phase I Completed 
   Solid tumors and B-cell NHL Monotherapy or in combination with cyclophosphamide NCT01032681, Phase I Completed 
   Melanoma Combination with radiotherapy NCT01973608, Phase I Terminated 
RO7284755 IL2v PD-1 Solid tumors Monotherapy or combination with atezolizumab NCT04303858, Phase I Recruiting 
NHS-IL-12 (M7824) IL12 DNA/histone complex Hormone-positive breast cancers Combination with radiation, avelumab NCT04756505, Phase I Withdrawn 
   Solid tumors Monotherapy NCT01417546, Phase I Completed 
   Prostate Combination with docetaxel NCT04633252, Phase I/II Recruiting 
   Genitourinary malignancies Combination with radiotherapy NCT04235777, Phase I Recruiting 
   Advanced Kaposi sarcoma Monotherapy and in combination M7824 NCT04303117, Phase I/II Recruiting 
   HPV tumors Combination with bintrafusp alfa and entinostat NCT04708470, Phase I/II Recruiting 
    Combination with vaccine NCT04287868, Phase I/II Not recruiting 
   MSS small bowel and CRC Combination with vaccine and superagonist NCT04491955, Phase II Not recruiting 
   Pancreas Combination with radiation NCT04327986, Phase I/II Terminated 
   Solid tumors Combination with avelumab NCT02994953, Phase I Terminated 
   Prostate cancer Combination with docetaxel NCT04633252, Phase I/II Recruiting 
BC1-IL12 (AS1409) IL12 EDB RCC, melanoma Monotherapy NCT00625768, Phase I Completed 
L19-TNF Onfekafusp alfa; Fibromun) TNF EDB Melanoma Combination with dacarbazine NCT02076646, Phase I/II Not recruiting 
    Combination with dacarbazine NCT01055522, Phase II Terminated 
    Combination with L19-IL2 NCT02938299, Phase III Recruiting 
   Oligometastatic solid tumor Combination with radiation therapy NCT02086721, Phase I Completed 
   IDH WT glioma Monotherapy NCT03779230, Phase I/II Not recruiting 
   Glioblastoma Combination with lomustine NCT04573192, Phase I/II Recruiting 
Cytokine
NamepayloadAntigenIndicationTreatmentNCTStudy status
L19-IL2 IL2 EDB Melanoma Combination with dacarbazine NCT02076646, Phase I/II Not recruiting 
(Darleukin)    Monotherapy NCT01253096, Phase II Completed 
    Combination with dacarbazine NCT01055522, Phase II Terminated 
    Monotherapy NCT02938299, Phase III Recruiting 
   Oligometastatic Combination with radiotherapy NCT02086721, Phase I Completed 
   Solid tumor Monotherapy NCT01058538, Phase I Completed 
   B-cell lymphoma Combination with rituximab NCT02957019, Phase I/II Not recruiting 
   Pancreatic Combination with gemcitabine NCT01198522, Phase I Terminated 
   NSCLC Combination with radiotherapy NCT03705403, Phase II Recruiting 
F16-IL2 IL2 Tenascin C AML Combination with cytarabine NCT02957032, Phase I Not recruiting 
(Teleukin)    Combination with BI 836858 NCT03207191, Phase I Completed 
   Solid tumors Combination with doxorubicin NCT01131364, Phase I/II Terminated 
    Combination with paclitaxel NCT01134250, Phase I/II Completed 
   Merkel cell Combination with paclitaxel NCT02054884, Phase II Terminated 
DI-Leu16-IL2 IL2 CD20 B-cell NHL Monotherapy, following rituximab blood NCT00720135, Phase I Completed 
    B-cell depletion NCT02151903. Phase I/II Terminated 
     NCT01874288, Phase I/II Terminated 
Hu14.18-IL2 IL2 GD2 Neuroblastoma Monotherapy NCT00003750, Phase I Completed 
   Melanoma Combination with ICI, radiotherapy NCT03958383, Phase I/II Suspended 
    Monotherapy NCT00590824, Phase II Completed 
    Monotherapy NCT00109863, Phase II Completed 
   Neuroblastoma and osteosarcoma Combination with NK cells NCT03209869. Phase I Withdrawn 
   Neuroblastoma Combination with sargramostim and isotretinoin NCT01334515, Phase II Completed 
    Monotherapy NCT00082758, Phase II Completed 
    Combination with chemo, ICI, NK cells NCT01576692, Phase I Completed 
EMD27306 (Tucotuzumab celmoleukin) IL2 EpCAM EpCAM-positive ovarian, prostate CRC or NSCLC Combination with cyclophosphamide NCT00132522, Phase I Completed 
huKS-IL2, EpCAM G733-2   Refractory kidney bladder, lung Monotherapy NCT00016237, Phase I Completed 
RO6874281 (RG7461; Simlukafusp alfa) IL2v FAP Breast, head, and neck solid tumors Combination with trastuzumab or cetuximab NCT02627274, Phase I   Completed 
   RCC Combination with ICI ± bevazicumab NCT03063762, Phase I Completed 
   Head and neck, esophageal and cervical cancers Combination with atezolizumab NCT03386721, Phase II Completed 
   Melanoma Combination with pembrolizumab NCT03875079, Phase I Completed 
   Pancreatic Multiple ICI combinations NCT03193190, Phase I/II Recruiting 
RO6895882 (Cergutuzumab amunaleukin) IL2v CEA Solid CEA positive tumors Monotherapy NCT02004106, Phase I Completed 
   Solid tumors Combination with atezolizumab NCT02350673, Phase I Completed 
EMD521873 (Selectikine) IL2v DNA/histone complex NSCLC Combination with radiotherapy NCT00879866, Phase I Completed 
   Solid tumors and B-cell NHL Monotherapy or in combination with cyclophosphamide NCT01032681, Phase I Completed 
   Melanoma Combination with radiotherapy NCT01973608, Phase I Terminated 
RO7284755 IL2v PD-1 Solid tumors Monotherapy or combination with atezolizumab NCT04303858, Phase I Recruiting 
NHS-IL-12 (M7824) IL12 DNA/histone complex Hormone-positive breast cancers Combination with radiation, avelumab NCT04756505, Phase I Withdrawn 
   Solid tumors Monotherapy NCT01417546, Phase I Completed 
   Prostate Combination with docetaxel NCT04633252, Phase I/II Recruiting 
   Genitourinary malignancies Combination with radiotherapy NCT04235777, Phase I Recruiting 
   Advanced Kaposi sarcoma Monotherapy and in combination M7824 NCT04303117, Phase I/II Recruiting 
   HPV tumors Combination with bintrafusp alfa and entinostat NCT04708470, Phase I/II Recruiting 
    Combination with vaccine NCT04287868, Phase I/II Not recruiting 
   MSS small bowel and CRC Combination with vaccine and superagonist NCT04491955, Phase II Not recruiting 
   Pancreas Combination with radiation NCT04327986, Phase I/II Terminated 
   Solid tumors Combination with avelumab NCT02994953, Phase I Terminated 
   Prostate cancer Combination with docetaxel NCT04633252, Phase I/II Recruiting 
BC1-IL12 (AS1409) IL12 EDB RCC, melanoma Monotherapy NCT00625768, Phase I Completed 
L19-TNF Onfekafusp alfa; Fibromun) TNF EDB Melanoma Combination with dacarbazine NCT02076646, Phase I/II Not recruiting 
    Combination with dacarbazine NCT01055522, Phase II Terminated 
    Combination with L19-IL2 NCT02938299, Phase III Recruiting 
   Oligometastatic solid tumor Combination with radiation therapy NCT02086721, Phase I Completed 
   IDH WT glioma Monotherapy NCT03779230, Phase I/II Not recruiting 
   Glioblastoma Combination with lomustine NCT04573192, Phase I/II Recruiting 

Various immunocytokines have also been directed against components of the extracellular matrix (ECM) of tumors (12). These include vascular antigens that are upregulated in cancer angiogenesis, including tenascin-C and extra domain A (ED-A) and B (ED-B) of fibronectin (13). Beyond targeting angiogenesis, immunocytokines have also been directed against: (i) components of collagen within tumor stroma, (ii) antigens exposed within necrotic tissues (e.g., DNA/histone complexes), and (iii) antigens expressed on T cells within the TME (e.g., programmed cell death 1; PD-1; refs. 14–17).

Antibody formats

The molecular composition of immunocytokines may also have important implications for antitumor activity, affecting drug clearance rates and ability to penetrate and be retained within tumors (18).

Intact IgG

Broadly, there are two major delivery platforms for immunocytokines: intact IgG or antibody fragment formats. Early generations of immunocytokines commonly featured IgG formats (7). This format has several advantages. Intact IgG has a long serum half-life, mediated by large size, which reduces renal clearance. Intact IgGs can also bind to the neonatal Fc receptor (FcRn), which prevents degradation by the lysosomal compartment, allowing them to be retained in circulation (19, 20). Full size IgG antibodies have two heavy chains and two light chains, both with variable regions. This dual variable region structure leads to higher avidity for a given target, resulting in higher rates of retention at the target (21). Nonetheless, the large size of the IgG format can also pose important challenges. For example, it may impede extravasation from vasculature and therefore penetration into tumor. Furthermore, the long-half life and tissue retention may lead to significant off-target toxicity (22).

Antibody fragment formats

To overcome limitations of IgG-based immunocytokines, various antibody fragment formats have been developed, including monovalent, F(ab′), divalent, F(ab′)2, and single-chain variable fragments (scFv; ref. 23). A principal advantage of these formats is their smaller size, which may promote tumor penetration, while retaining antigen specificity of the full-length antibody (8). Nonetheless, smaller size and lack of an Fc region also serve to decrease serum half-life – although this may decrease off-target toxicity (8). Importantly, depending on the antibody format, the avidity for the target may also be decreased. To overcome this, other antibody fragment formats (e.g., dia- and triabodies) have been developed (24). These contain more antigen-binding moieties and thereby have increased avidity while retaining other advantages of antibody fragment formats (21).

To date, various cytokine payloads have been incorporated into immunocytokines. The most extensively studied payloads include IL2, IL12, and tumor necrosis factor (TNF). Below, we summarize preliminary data behind each cytokine payload, emphasizing the existing clinical data generated thus far.

IL2

IL2 is a 15-kDa proinflammatory cytokine produced by activated T cells (25). IL2 mediates its protean immunologic effects through binding to the IL2 receptor (IL-2R), which is comprised of α, β, and γ subunits. IL2Rα (CD25) confers high-affinity binding to IL2 and is essential for the expansion of FOXP3+ T regulatory cells (Tregs), while IL2Rβ (CD122), and IL2Rγ (CD132) subunits, expressed on natural killer (NK) cells and resting CD4+ and CD8+ T cells, mediate signal transduction (13, 26). At low-doses, IL2 preferentially binds to its high-affinity CD25 receptors on Treg cells leading to immune evasion (27, 28). At high-doses, IL2 therapy induced durable responses in a small subset of patients with renal cell carcinoma (RCC) and melanoma (4, 29). However, significant safety concerns, including potentially life-threatening organ dysfunction, capillary leak syndrome, and coma, have limited its use (30). Various strategies to improve upon initial IL2 formulations have since been explored, such as PEGylation to increase half-life, introducing targeted mutations to modify function (e.g., CD25-biased, CD25/CD122-biased formulations), masking IL2 against Treg binding, and developing IL2-based immunocytokines to improve antitumor activity and reduce toxicity (31, 32).

IL2 is the most extensively studied immunocytokine payload to date (Table 1), with various agents entering phase I–II clinical trials. Broadly, these agents can be categorized according to IL2 formulation (unbiased vs. biased) and targeting approach (cell surface antigens vs. ECM). Unbiased IL2 formulations are those that are not selective for the intermediate-affinity dimeric or high-affinity trimeric IL2R whereas biased IL2 agents selectively target either CD25, CD25/CD122, or CD122 alone (25).

IL2 targeting tumor antigens

Many clinical trials have been performed with IL2 immunocytokines targeting tumor antigens. For example, Hu14.18-IL2 is a humanized mAb covalently linked to two molecules of IL2 that targets GD2. In preclinical murine models of neuroblastoma, Hu14.18-IL2 showed antitumor activity, and early-phase clinical studies demonstrated it was relatively well tolerated with evidence of immune activation (33). However, clinical activity has been disappointing. In one phase II study of Hu14.18-IL2 that enrolled 14 patients with melanoma, the objective response rate (ORR) was only 7.1% (34). Likewise, in a phase II study that enrolled 13 neuroblastoma patients with measurable disease, no objective responses were observed (35).

IL2-based immunocytokines have also been explored within hematologic malignancies. For example, DI-Leu16-IL2, a humanized anti-CD20 antibody fused to IL2, has been explored in patients with CD20-positive non–Hodgkin's lymphoma (NHL). A phase 1 trial of DI-Leu16-IL2 following peripheral B-cell depletion with low-dose rituximab in patients with relapsed/refractory disease showed durable tumor regression or stabilization in 14 of 16 (87.5%) evaluable patients (36). Among five patients with objective responses, the median duration of response was 13 months, with one ongoing complete response observed at 11 months. This drug was generally well-tolerated and demonstrated higher permissible doses with s.c. administration than could be achieved with i.v. therapy (37).

Cergutuzumab amunaleukin (CEA–IL2v) and simlukafusp α (fibroblast activation protein; FAP-IL2v) are IL2 variant (IL2v)-based immunocytokines that target carcinoembryonic antigen (CEA) and fibroblast activation protein α (FAPα) tumor antigens, respectively (38, 39). These are CD122-biased IL2 agents fused to the C-terminus of their respective antibodies together with novel Fc mutations that eliminate FcγR and C1q-binding. Preclinically, these variants lack preferential induction of Tregs due to abolished CD25-binding but retain capacity to activate and expand NK and CD8+ effector T-cells through IL2Rβγ in the periphery and the TME (13). A phase Ib trial of CEA-IL2v in combination with atezolizumab has completed enrolment (NCT02350673), but results have not been reported to date.

IL2 targeting ECM components

In addition to tumor antigens, IL2 immunocytokines have also been developed against components of the ECM. For example, L19-IL2 is a fully human recombinant monoclonal antibody comprised of IL2 fused to the L19 diabody, where L19 targets an alternatively spliced isoform of fibronectin (ED-B domain) expressed in the setting of angiogenesis (14). In preclinical models, IL19-IL2 selectively accumulates in tumor vasculature and demonstrated antitumor activity with concomitant increases in T lymphocytes, NK cells, and macrophages, along with a corresponding increase in IFNγ (14). L19-IL2 has been studied in multiple different tumor types, both as monotherapy and in combination with other agents, including systemic therapy and radiotherapy (40–42). Among metastatic solid tumors patients enrolled in a phase I/II study of L19-IL2, a best response of stable disease was observed in 17 of 33 patients (51%; ref. 40). Among the metastatic RCC subgroup, 15 of 18 (83%) patients experienced disease stabilization, but no objective responses were observed. Given the modest efficacy of L19-IL2 monotherapy, this agent has also been explored in combination with dacarbazine in metastatic melanoma (43). In an initial single-arm phase II study, L19-IL2 showed an ORR of 28%, with one complete response. However, the subsequent randomized phase II portion of this trial was disappointing, with no significant difference in progression-free survival (PFS) between L19-IL2 and dacarbazine alone (44). Nonetheless, L19-IL2 (darleukin) is still being explored in ongoing clinical trials, outlined below.

IL2: cis-targeted cytokines

Another approach to developing immunocytokines is to target immune cells directly. The IL2v antibody fusion protein RO7284755 targets programmed cell death 1 (PD-1), which is overexpressed on the surface of activated effector T cells. Preclinical data show high-affinity PD-1 binding allows IL2v to trigger a proliferation cascade to generate a distinct population of tumor antigen–specific CD8+ T cells (17). By docking in cis to PD-1, it also overcomes the need for CD25-binding (45, 46). PD-1-IL2v (RO7284755) is currently being studied both as monotherapy and in combination with the anti–programmed death ligand 1 (PD-L1) antibody atezolizumab for the treatment of immune checkpoint inhibitor (ICI)-responsive advanced cancers (NCT04303858).

IL12

IL12 is a pleiotropic proinflammatory cytokine primarily produced by antigen presenting cells (APC). Comprised of two subunits, p40 and p35, IL12 stimulates the effector function of activated T cells and NK cells and production of IFNγ (47). In various murine models, treatment with recombinant IL12 leads to enhanced innate and adaptive antitumor immunity and dramatic antitumor activity (48). However, in clinical trials, systemic administration of recombinant IL12 has produced disappointing results, marked by low therapeutic efficacy and significant systemic toxicity, including fatal inflammatory syndrome (8, 49). IL12-based immunocytokines have been developed as one approach to overcome this. NHS IL12 consists of two IL12 heterodimers fused to the human IgG1 NHS76 antibody that targets areas of tumor necrosis by binding exposed histone complexes in DNA (50). A phase I study of s.c. administration of NHS IL-12 in patients with metastatic or locally advanced epithelial or mesenchymal tumors demonstrated that this agent was well tolerated and increased T- cell receptor (TCR) diversity and tumor-infiltrating lymphocyte density, indicative of an antitumor immune response (51). Interestingly, reductions in prostate-specific antigen (PSA) levels were seen in five of eight evaluable patients with prostate cancer, but no objective responses were observed. Nonetheless, given evidence of immune activation with monotherapy, NHS IL12 is being explored in combination with other agents.

Preclinical data suggests that IL12 may be more effective as part of combination therapy (52, 53). Currently, NHS IL2 is being studied in combination with docetaxel in patients with metastatic prostate cancer (NCT04633252) and in combination with the anti–PD-L1 inhibitor, avelumab (NCT04756505). Other strategies in the development of IL12-based immunocytokines have focused on modifications in target antibodies (e.g., IL12–7NP2, which targets FAP), delivery systems (e.g., intratumor delivery), or novel combination strategies (e.g., histone deacetylase inhibitors; refs. 54, 55).

TNF

TNFα is a multifunctional cytokine that plays key roles in vascular permeability, inflammation, and apoptosis (2). TNFα acts on endothelial cells to increase vascular permeability and mediates hemorrhagic tumor necrosis (56). Systemic administration of TNFα has been limited due to significant adverse events including severe hypotension and organ dysfunction (57). Furthermore, studies of TNFα have shown minimal to no tumor response (57–59). TNFα has since been studied as a payload in immunocytokine development. L19-TNF is an immunocytokine comprised of the single-chain human monoclonal L19 antibody fused to TNFα (50). In a phase 1 trial of L19-TNF plus doxorubicin in patients with advanced cancers, primarily soft tissue sarcomas (STS), the combination was overall well tolerated and demonstrated preliminary evidence of antitumor activity (60). In an expansion cohort of 15 patients with STS, there was 1 complete response, 1 partial response and 7 patients with disease stabilization (60). A randomized phase II trial of L19-TNF plus dacarbazine versus dacarbazine alone in advanced STS is ongoing (NCT04733183). L19-TNF is also being studied in other tumor types including glioblastoma and melanoma (NCT03779230; refs. 61, 62).

Despite a decade of progress in immunocytokine development, no agent has yet gained regulatory approval, leaving important challenges for the field. Here, we outline potential strategies to improve upon existing immunocytokines, highlighting the need for: (i) improved antibody targets, (ii) enhanced antibody designs, (iii) improved drug delivery techniques, (iv) novel combination strategies, and (v) modifications in payloads (Fig. 3).

Figure 3.

Strategies for improving upon the effectiveness of immunocytokines, including enhancement in antibodies, improved delivery techniques, modifications in payloads, improved antibody targets, and novel combination approaches. (Adapted from an image created with BioRender.com.)

Figure 3.

Strategies for improving upon the effectiveness of immunocytokines, including enhancement in antibodies, improved delivery techniques, modifications in payloads, improved antibody targets, and novel combination approaches. (Adapted from an image created with BioRender.com.)

Close modal

Improved antibody targets

Lack of highly specific targets has been a fundamental limitation of immunocytokines. With recent progress in transcriptomic and proteomic profiling technologies, efforts are ongoing to elucidate new tumor targets (18). Efforts in antibody target discovery may also benefit from the recent explosion in antibody–drug conjugate (ADC) development, to facilitate identification of new cell surface, tumor-specific targets (63).

Enhancements in antibodies

Selectively engineering modifications in the antibody component of immunocytokines may be another strategy to improve efficacy. For example, point mutations in the Fc tail have been suggested to enhance effector function (21) by increasing antibody-dependent cellular phagocytosis and cytotoxicity (64). Other antibody enhancement strategies have focused on modifying pharmacokinetics by manipulating posttranslational glycosylation (65, 66). Lastly, cross-linking cytokines to bispecific (e.g., BITEs or BiKEs) and tri-specific (TriKEs) immune engagers have also been developed, aiming to increase immune activation, recruit effector cell populations, and increase specificity for the tumor by targeting multiple antigens (2, 21, 67).

Improved delivery techniques

Novel delivery techniques have also been explored to improve potency and reduce off-target toxicity as immunocytokines with unblocked and nonattenuated cytokines are likely to be associated with cytokine-related toxicities. One innovative strategy is the use of protease-cleavable epitope masking. In this approach, an epitope-masking molecule is linked to the antibody, which is cleavable by matrix metalloproteases specifically expressed at tumor sites. This essentially renders immune-active antibody moieties inactive until the tumor site is reached, thereby reducing systemic toxicity (68). One recent such example is WTX-124, which links IL2 to an inactivation domain that is cleaved by dysregulated proteases in the TME (69). In preclinical murine models, WTX-124 retained antitumor activity while minimizing IL2 effects in nontumor tissues.

“Activity-on-target cytokines” (AcTakines) are yet another emerging immunocytokine class with a novel delivery method. AcTakines replace the wild-type (WT) cytokine payload with a mutant form that has reduced affinity for its cognate receptor, thereby rendering it inactive in circulation. Upon cell-specific–binding through a targeting moiety, the cytokine regains full activity (70). This strategy was first studied to improve the therapeutic index of IFNα but has since been applied to many other cytokines including phase I trials with IL15 and IL21 (17).

Modifications in payloads

Immunocytokines have been developed that carry multiple payloads, termed dual-cytokine immunocytokines. These are still in the preclinical stage but combinations such as IL2/TNF have shown promise (71, 72). In addition, coformulations of multiple immunocytokines have been explored. For example, Daromun is an intralesional therapy consisting of two immunocytokines (L19IL2 and L19TNF). In a phase II trial of Daromun that enrolled 20 efficacy evaluable patients with unresectable stage IIIC and IVM1a melanoma, the ORR of injected lesions was 55%. Interestingly, treatment responses were observed in 7 of 13 lesions that were uninjected, suggestive of a bystander effect (73). A phase III trial of intralesional Daromun as neoadjuvant therapy in patients with high-risk, resectable IIIB/C melanoma is underway (NCT02938299 and NCT03567889; ref. 74).

Combinations

To augment the activity of immunocytokines, there is growing interest in developing combination strategies with chemotherapy, radiation, and immunotherapy partners. Given the success of ICIs across multiple tumor types, these agents represent appealing combination partners. Combinations of immunocytokines plus ICIs have shown encouraging results in preclinical models (52, 75). For example, L19-TNF in combination with the PD-1 inhibitor nivolumab eradicated tumors in sarcoma models (76). Recently, there have also been efforts to combine a PD-L1 inhibitor with the cytokine IL15 in a single novel immunocytokine, LH01. Interestingly, Shi and colleagues reported that LH01 demonstrated superior antitumor activity than the combination of anti–PD-L1 and IL15 (16). Clinically, ICIs are being investigated in combination with multiple other immunocytokines or engineered cytokines (Table 1).

Immunocytokines have the potential to improve the therapeutic window of cytokine-based therapeutics. Over the last decade, immunocytokine development has centered around antibody engineering, identification of antibody targets, and selection of optimal payloads. Although these studies established proof of principle in preclinical studies, clinical development has lagged. Moving forward, further refinements in drug delivery, payloads, and antibody constructs may help facilitate successful translation into the clinic. In parallel, efforts to identify robust combination strategies and ideal clinical opportunities to deploy such approaches will be important to build upon the advances in immunocytokine development.

A. Pabani reports grants and personal fees from Bristol-Myers Squibb; personal fees from AstraZeneca, Pfizer, Merck, Roche, and Canadian Agency for Drugs and Technologies in Health; and grants from Alberta Cancer Foundation outside the submitted work. J.F. Gainor reports personal fees and other support from Bristol-Myers Squibb, Merck, AstraZeneca, Moderna, and Blueprint; personal fees from Loxo/Lilly, Gilead, Pfizer, EMD Serono, iTeos, Amgen, Mirati, Nuvalent, Karyopharm, Beigene, Silverback Therapeutics, GlydeBio, Takeda, Curie Therapeutics, and Merus Pharmaceuticals; grants, personal fees, and other support from Genentech/Roche and Novartis; and other support from Jounce and Alexo outside the submitted work. In addition, J.F. Gainor's spouse is an employee with equity in Ironwood Pharmaceuticals.

1.
Chen
DS
,
Mellman
I
.
Elements of cancer immunity and the cancer-immune set point
.
Nature
2017
;
541
:
321
30
.
2.
Zheng
X
,
Wu
Y
,
Bi
J
,
Huang
Y
,
Cheng
Y
,
Li
Y
, et al
.
The use of supercytokines, immunocytokines, engager cytokines, and other synthetic cytokines in immunotherapy
.
Cell Mol Immunol
2022
;
19
:
192
209
.
3.
Golomb
HM
,
Jacobs
A
,
Fefer
A
,
Ozer
H
,
Thompson
J
,
Portlock
C
, et al
.
Alpha-2 interferon therapy of hairy-cell leukemia: a multicenter study of 64 patients
.
J Clin Oncol
1986
;
4
:
900
5
.
4.
Fyfe
G
,
Fisher
RI
,
Rosenberg
SA
,
Sznol
M
,
Parkinson
DR
,
Louie
AC
.
Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy
.
J Clin Oncol
1995
;
13
:
688
96
.
5.
Berraondo
P
,
Sanmamed
MF
,
Ochoa
MC
,
Etxeberria
I
,
Aznar
MA
,
Perez-Gracia
JL
, et al
.
Cytokines in clinical cancer immunotherapy
.
Br J Cancer
2019
;
120
:
6
15
.
6.
Di Trani
CA
,
Cirella
A
,
Arrizabalaga
L
,
Fernandez-Sendin
M
,
Bella
A
,
Aranda
F
, et al
.
Overcoming the limitations of cytokines to improve cancer therapy
.
Int Rev Cell Mol Biol
2022
;
369
:
107
41
.
7.
Neri
D
.
Antibody-cytokine fusions: versatile products for the modulation of anticancer immunity
.
Cancer Immunol Res
2019
;
7
:
348
54
.
8.
Jin
S
,
Sun
Y
,
Liang
X
,
Gu
X
,
Ning
J
,
Xu
Y
, et al
.
Emerging new therapeutic antibody derivatives for cancer treatment
.
Signal Transduct Target Ther
2022
;
7
:
39
.
9.
Neri
D
,
Sondel
PM
.
Immunocytokines for cancer treatment: past, present and future
.
Curr Opin Immunol
2016
;
40
:
96
102
.
10.
Went
P
,
Vasei
M
,
Bubendorf
L
,
Terracciano
L
,
Tornillo
L
,
Riede
U
, et al
.
Frequent high-level expression of the immunotherapeutic target Ep-CAM in colon, stomach, prostate and lung cancers
.
Br J Cancer
2006
;
94
:
128
35
.
11.
Ahmed
M
,
Cheung
NK
.
Engineering anti-GD2 monoclonal antibodies for cancer immunotherapy
.
FEBS Lett
2014
;
588
:
288
97
.
12.
Runbeck
E
,
Crescioli
S
,
Karagiannis
SN
,
Papa
S
.
Utilizing immunocytokines for cancer therapy
.
Antibodies (Basel)
2021
;
10
:
10
.
13.
Mortara
L
,
Balza
E
,
Bruno
A
,
Poggi
A
,
Orecchia
P
,
Carnemolla
B
.
Anti-cancer therapies employing IL-2 cytokine tumor targeting: contribution of innate, adaptive and immunosuppressive cells in the anti-tumor efficacy
.
Front Immunol
2018
;
9
:
2905
.
14.
Carnemolla
B
,
Borsi
L
,
Balza
E
,
Castellani
P
,
Meazza
R
,
Berndt
A
, et al
.
Enhancement of the antitumor properties of interleukin-2 by its targeted delivery to the tumor blood vessel extracellular matrix
.
Blood
2002
;
99
:
1659
65
.
15.
Ishihara
J
,
Ishihara
A
,
Sasaki
K
,
Lee
SS
,
Williford
JM
,
Yasui
M
, et al
.
Targeted antibody and cytokine cancer immunotherapies through collagen affinity
.
Sci Transl Med
2019
;
11
:
eaau3259
.
16.
Shi
W
,
Lv
L
,
Liu
N
,
Wang
H
,
Wang
Y
,
Zhu
W
, et al
.
A novel anti-PD-L1/IL-15 immunocytokine overcomes resistance to PD-L1 blockade and elicits potent antitumor immunity
.
Mol Ther
2023
;
31
:
66
77
.
17.
Holder
PG
,
Lim
SA
,
Huang
CS
,
Sharma
P
,
Dagdas
YS
,
Bulutoglu
B
, et al
.
Engineering interferons and interleukins for cancer immunotherapy
.
Adv Drug Deliv Rev
2022
;
182
:
114112
.
18.
Gutbrodt
KL
,
Neri
D
.
Immunocytokines
.
Antibodies (Basel)
2012
;
1
:
70
87
.
19.
Thurber
GM
,
Schmidt
MM
,
Wittrup
KD
.
Factors determining antibody distribution in tumors
.
Trends Pharmacol Sci
2008
;
29
:
57
61
.
20.
Chames
P
,
Van Regenmortel
M
,
Weiss
E
,
Baty
D
.
Therapeutic antibodies: successes, limitations and hopes for the future
.
Br J Pharmacol
2009
;
157
:
220
33
.
21.
Gout
DY
,
Groen
LS
,
van Egmond
M
.
The present and future of immunocytokines for cancer treatment
.
Cell Mol Life Sci
2022
;
79
:
509
.
22.
Gillies
SD
,
Lo
KM
,
Burger
C
,
Lan
Y
,
Dahl
T
,
Wong
WK
.
Improved circulating half-life and efficacy of an antibody-interleukin 2 immunocytokine based on reduced intracellular proteolysis
.
Clin Cancer Res
2002
;
8
:
210
6
.
23.
Kholodenko
RV
,
Kalinovsky
DV
,
Doronin
II
,
Ponomarev
ED
,
Kholodenko
IV
.
Antibody fragments as potential biopharmaceuticals for cancer therapy: success and limitations
.
Curr Med Chem
2019
;
26
:
396
426
.
24.
Adams
GP
,
Tai
MS
,
McCartney
JE
,
Marks
JD
,
Stafford
WF
III
,
Houston
LL
, et al
.
Avidity-mediated enhancement of in vivo tumor targeting by single-chain Fv dimers
.
Clin Cancer Res
2006
;
12
:
1599
605
.
25.
Raeber
ME
,
Sahin
D
,
Boyman
O
.
Interleukin-2-based therapies in cancer
.
Sci Transl Med
2022
;
14
:
eabo5409
.
26.
Boyman
O
,
Sprent
J
.
The role of interleukin-2 during homeostasis and activation of the immune system
.
Nat Rev Immunol
2012
;
12
:
180
90
.
27.
Xue
D
,
Hsu
E
,
Fu
YX
,
Peng
H
.
Next-generation cytokines for cancer immunotherapy
.
Antib Ther
2021
;
4
:
123
33
.
28.
Briukhovetska
D
,
Dorr
J
,
Endres
S
,
Libby
P
,
Dinarello
CA
,
Kobold
S
.
Interleukins in cancer: from biology to therapy
.
Nat Rev Cancer
2021
;
21
:
481
99
.
29.
Rosenberg
SA
,
Yang
JC
,
Topalian
SL
,
Schwartzentruber
DJ
,
Weber
JS
,
Parkinson
DR
, et al
.
Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2
.
JAMA
1994
;
271
:
907
13
.
30.
Rosenberg
SA
.
IL-2: the first effective immunotherapy for human cancer
.
J Immunol
2014
;
192
:
5451
8
.
31.
Silva
DA
,
Yu
S
,
Ulge
UY
,
Spangler
JB
,
Jude
KM
,
Labao-Almeida
C
, et al
.
De novo design of potent and selective mimics of IL-2 and IL-15
.
Nature
2019
;
565
:
186
91
.
32.
Sahin
D
,
Arenas-Ramirez
N
,
Rath
M
,
Karakus
U
,
Humbelin
M
,
van Gogh
M
, et al
.
An IL-2-grafted antibody immunotherapy with potent efficacy against metastatic cancer
.
Nat Commun
2020
;
11
:
6440
.
33.
Osenga
KL
,
Hank
JA
,
Albertini
MR
,
Gan
J
,
Sternberg
AG
,
Eickhoff
J
, et al
.
A phase I clinical trial of the hu14.18-IL2 (EMD 273063) as a treatment for children with refractory or recurrent neuroblastoma and melanoma: a study of the Children's Oncology Group
.
Clin Cancer Res
2006
;
12
:
1750
9
.
34.
Albertini
MR
,
Hank
JA
,
Gadbaw
B
,
Kostlevy
J
,
Haldeman
J
,
Schalch
H
, et al
.
Phase II trial of hu14.18-IL2 for patients with metastatic melanoma
.
Cancer Immunol Immunother
2012
;
61
:
2261
71
.
35.
Shusterman
S
,
London
WB
,
Gillies
SD
,
Hank
JA
,
Voss
SD
,
Seeger
RC
, et al
.
Antitumor activity of hu14.18-IL2 in patients with relapsed/refractory neuroblastoma: a children's oncology group (COG) phase II study
.
J Clin Oncol
2010
;
28
:
4969
75
.
36.
Lansigan
F
,
Nakamura
R
,
Quick
DP
,
Vlock
D
,
Raubitschek
A
,
Gillies
S
, et 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
34:15s
,
2016
(
suppl; abstr e19046
).
37.
Lansigan
F
,
Nakamura
R
,
Quick
DP
,
Vlock
D
,
Raubitschek
A
,
Gillies
SD
, et al
.
DI-Leu16-IL2, an anti-CD20-interleukin-2 immunocytokine, is safe and active in patients with relapsed and refractory B-cell lymphoma: a report of maximum tolerated dose, optimal biologic dose, and recommended phase 2 dose
.
Blood
2016
:
128
:
620
.
38.
Klein
C
,
Waldhauer
I
,
Nicolini
VG
,
Freimoser-Grundschober
A
,
Nayak
T
,
Vugts
DJ
, et 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
.
Oncoimmunology
2017
;
6
:
e1277306
.
39.
Xin
L
,
Gao
J
,
Zheng
Z
,
Chen
Y
,
Lv
S
,
Zhao
Z
, et al
.
Fibroblast activation protein-alpha as a target in the bench-to-bedside diagnosis and treatment of tumors: a narrative review
.
Front Oncol
2021
;
11
:
648187
.
40.
Johannsen
M
,
Spitaleri
G
,
Curigliano
G
,
Roigas
J
,
Weikert
S
,
Kempkensteffen
C
, et al
.
The tumour-targeting human L19-IL2 immunocytokine: preclinical safety studies, phase I clinical trial in patients with solid tumours and expansion into patients with advanced renal cell carcinoma
.
Eur J Cancer
2010
;
46
:
2926
35
.
41.
Weide
B
,
Eigentler
TK
,
Pflugfelder
A
,
Zelba
H
,
Martens
A
,
Pawelec
G
, et al
.
Intralesional treatment of stage III metastatic melanoma patients with L19-IL2 results in sustained clinical and systemic immunologic responses
.
Cancer Immunol Res
2014
;
2
:
668
78
.
42.
Rekers
NH
,
Olivo Pimentel
V
,
Yaromina
A
,
Lieuwes
NG
,
Biemans
R
,
Zegers
CML
, et al
.
The immunocytokine L19-IL2: an interplay between radiotherapy and long-lasting systemic anti-tumour immune responses
.
Oncoimmunology
2018
;
7
:
e1414119
.
43.
Eigentler
TK
,
Weide
B
,
de Braud
F
,
Spitaleri
G
,
Romanini
A
,
Pflugfelder
A
, et al
.
A dose-escalation and signal-generating study of the immunocytokine L19-IL2 in combination with dacarbazine for the therapy of patients with metastatic melanoma
.
Clin Cancer Res
2011
;
17
:
7732
42
.
44.
Weide
B
,
Eigentler
T
,
Catania
C
,
Ascierto
PA
,
Cascinu
S
,
Becker
JC
, et al
.
A phase II study of the L19IL2 immunocytokine in combination with dacarbazine in advanced metastatic melanoma patients
.
Cancer Immunol Immunother
2019
;
68
:
1547
59
.
45.
Codarri Deak
L
,
Nicolini
V
,
Hashimoto
M
,
Karagianni
M
,
Schwalie
PC
,
Lauener
L
, et al
.
PD-1-cis IL-2R agonism yields better effectors from stem-like CD8(+) T cells
.
Nature
2022
;
610
:
161
72
.
46.
Hashimoto
M
,
Araki
K
,
Cardenas
MA
,
Li
P
,
Jadhav
RR
,
Kissick
HT
, et al
.
PD-1 combination therapy with IL-2 modifies CD8(+) T cell exhaustion program
.
Nature
2022
;
610
:
173
81
.
47.
D'Andrea
A
,
Rengaraju
M
,
Valiante
NM
,
Chehimi
J
,
Kubin
M
,
Aste
M
, et al
.
Production of natural killer cell stimulatory factor (interleukin 12) by peripheral blood mononuclear cells
.
J Exp Med
1992
;
176
:
1387
98
.
48.
Colombo
MP
,
Trinchieri
G
.
Interleukin-12 in anti-tumor immunity and immunotherapy
.
Cytokine Growth Factor Rev
2002
;
13
:
155
68
.
49.
Motzer
RJ
,
Rakhit
A
,
Schwartz
LH
,
Olencki
T
,
Malone
TM
,
Sandstrom
K
, et al
.
Phase I trial of subcutaneous recombinant human interleukin-12 in patients with advanced renal cell carcinoma
.
Clin Cancer Res
1998
;
4
:
1183
91
.
50.
Fallon
J
,
Tighe
R
,
Kradjian
G
,
Guzman
W
,
Bernhardt
A
,
Neuteboom
B
, et al
.
The immunocytokine NHS-IL12 as a potential cancer therapeutic
.
Oncotarget
2014
;
5
:
1869
84
.
51.
Strauss
J
,
Heery
CR
,
Kim
JW
,
Jochems
C
,
Donahue
RN
,
Montgomery
AS
, et 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
.
52.
Xu
C
,
Zhang
Y
,
Rolfe
PA
,
Hernandez
VM
,
Guzman
W
,
Kradjian
G
, et 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
.
53.
Lee
P
,
Wang
F
,
Kuniyoshi
J
,
Rubio
V
,
Stuges
T
,
Groshen
S
, et al
.
Effects of interleukin-12 on the immune response to a multipeptide vaccine for resected metastatic melanoma
.
J Clin Oncol
2001
;
19
:
3836
47
.
54.
Hicks
KC
,
Chariou
PL
,
Ozawa
Y
,
Minnar
CM
,
Knudson
KM
,
Meyer
TJ
, et al
.
Tumour-targeted interleukin-12 and entinostat combination therapy improves cancer survival by reprogramming the tumour immune cell landscape
.
Nat Commun
2021
;
12
:
5151
.
55.
Nadal
L
,
Peissert
F
,
Elsayed
A
,
Weiss
T
,
Look
T
,
Weller
M
, et al
.
Generation and in vivo validation of an IL-12 fusion protein based on a novel anti-human FAP monoclonal antibody
.
J Immunother Cancer
2022
;
10
:
e005282
.
56.
Borsi
L
,
Balza
E
,
Carnemolla
B
,
Sassi
F
,
Castellani
P
,
Berndt
A
, et al
.
Selective targeted delivery of TNFalpha to tumor blood vessels
.
Blood
2003
;
102
:
4384
92
.
57.
Roberts
NJ
,
Zhou
S
,
Diaz
LA
Jr
,
Holdhoff
M
.
Systemic use of tumor necrosis factor alpha as an anticancer agent
.
Oncotarget
2011
;
2
:
739
51
.
58.
Budd
GT
,
Green
S
,
Baker
LH
,
Hersh
EP
,
Weick
JK
,
Osborne
CK
.
A southwest oncology group Phase II trial of recombinant tumor necrosis factor in metastatic breast cancer
.
Cancer
1991
;
68
:
1694
5
.
59.
Brown
TD
,
Goodman
P
,
Fleming
T
,
Macdonald
JS
,
Hersh
EM
,
Braun
TJ
.
A Phase II trial of recombinant tumor necrosis factor in patients with adenocarcinoma of the pancreas: a southwest oncology group study
.
J Immunother (1991)
1991
;
10
:
376
8
.
60.
Schliemann
C
,
Hemmerle
T
,
Berdel
AF
,
Angenendt
L
,
Kerkhoff
A
,
Hering
JP
, et al
.
Dose escalation and expansion phase I studies with the tumour-targeting antibody-tumour necrosis factor fusion protein L19TNF plus doxorubicin in patients with advanced tumours, including sarcomas
.
Eur J Cancer
2021
;
150
:
143
54
.
61.
Weiss
T
,
Puca
E
,
Silginer
M
,
Hemmerle
T
,
Pazahr
S
,
Bink
A
, et al
.
Immunocytokines are a promising immunotherapeutic approach against glioblastoma
.
Sci Transl Med
2020
;
12
:
eabb2311
.
62.
Papadia
F
,
Basso
V
,
Patuzzo
R
,
Maurichi
A
,
Di Florio
A
,
Zardi
L
, et al
.
Isolated limb perfusion with the tumor-targeting human monoclonal antibody-cytokine fusion protein L19-TNF plus melphalan and mild hyperthermia in patients with locally advanced extremity melanoma
.
J Surg Oncol
2013
;
107
:
173
9
.
63.
Criscitiello
C
,
Morganti
S
,
Curigliano
G
.
Antibody-drug conjugates in solid tumors: a look into novel targets
.
J Hematol Oncol
2021
;
14
:
20
.
64.
Lazar
GA
,
Dang
W
,
Karki
S
,
Vafa
O
,
Peng
JS
,
Hyun
L
, et al
.
Engineered antibody Fc variants with enhanced effector function
.
Proc Natl Acad Sci U S A.
2006
;
103
:
4005
10
.
65.
Dekkers
G
,
Treffers
L
,
Plomp
R
,
Bentlage
AEH
,
de Boer
M
,
Koeleman
CAM
, et al
.
Decoding the human immunoglobulin G-glycan repertoire reveals a spectrum of fc-receptor- and complement-mediated-effector activities
.
Front Immunol
2017
;
8
:
877
.
66.
Umana
P
,
Jean-Mairet
J
,
Moudry
R
,
Amstutz
H
,
Bailey
JE
.
Engineered glycoforms of an antineuroblastoma IgG1 with optimized antibody-dependent cellular cytotoxic activity
.
Nat Biotechnol
1999
;
17
:
176
80
.
67.
Vallera
DA
,
Felices
M
,
McElmurry
R
,
McCullar
V
,
Zhou
X
,
Schmohl
JU
, et al
.
IL15 trispecific killer engagers (TriKE) make natural killer cells specific to CD33+ targets while also inducing persistence, in vivo expansion, and enhanced function
.
Clin Cancer Res
2016
;
22
:
3440
50
.
68.
Erster
O
,
Thomas
JM
,
Hamzah
J
,
Jabaiah
AM
,
Getz
JA
,
Schoep
TD
, et al
.
Site-specific targeting of antibody activity in vivo mediated by disease-associated proteases
.
J Control Release
2012
;
161
:
804
12
.
69.
Nirschl
CJ
,
Brodkin
HR
,
Hicklin
DJ
,
Ismail
N
,
Morris
K
,
Seidel-Dugan
C
, et al
.
Discovery of a conditionally activated IL-2 that promotes antitumor immunity and induces tumor regression
.
Cancer Immunol Res
2022
;
10
:
581
96
.
70.
Garcin
G
,
Paul
F
,
Staufenbiel
M
,
Bordat
Y
,
Van der Heyden
J
,
Wilmes
S
, et al
.
High efficiency cell-specific targeting of cytokine activity
.
Nat Commun
2014
;
5
:
3016
.
71.
De Luca
R
,
Soltermann
A
,
Pretto
F
,
Pemberton-Ross
C
,
Pellegrini
G
,
Wulhfard
S
, et al
.
Potency-matched dual cytokine-antibody fusion proteins for cancer therapy
.
Mol Cancer Ther
2017
;
16
:
2442
51
.
72.
De Luca
R
,
Gouyou
B
,
Ongaro
T
,
Villa
A
,
Ziffels
B
,
Sannino
A
, et al
.
A novel fully-human potency-matched dual cytokine-antibody fusion protein targets carbonic anhydrase IX in renal cell carcinomas
.
Front Oncol
2019
;
9
:
1228
.
73.
Danielli
R
,
Patuzzo
R
,
Di Giacomo
AM
,
Gallino
G
,
Maurichi
A
,
Di Florio
A
, et al
.
Intralesional administration of L19-IL2/L19-TNF in stage III or stage IVM1a melanoma patients: results of a phase II study
.
Cancer Immunol Immunother
2015
;
64
:
999
1009
.
74.
Miura
JT
,
Zager
JS
.
Neo-DREAM study investigating Daromun for the treatment of clinical stage IIIB/C melanoma
.
Future Oncol
2019
;
15
:
3665
74
.
75.
Fallon
JK
,
Vandeveer
AJ
,
Schlom
J
,
Greiner
JW
.
Enhanced antitumor effects by combining an IL-12/anti-DNA fusion protein with avelumab, an anti-PD-L1 antibody
.
Oncotarget
2017
;
8
:
20558
71
.
76.
Corbellari
R
,
Nadal
L
,
Villa
A
,
Neri
D
,
De Luca
R
.
The immunocytokine L19-TNF eradicates sarcomas in combination with chemotherapy agents or with immune check-point inhibitors
.
Anticancer Drugs
2020
;
31
:
799
805
.