Review article
Antibody–cytokine fusion proteins for the therapy of cancer

https://doi.org/10.1016/S0022-1759(00)00345-8Get rights and content

Abstract

Advances in genetic engineering and expression systems have led to rapid progress in the development of antibodies fused to other proteins. These ‘antibody fusion proteins’ have novel properties and include antibodies with specificity for tumor associated antigens fused to cytokines such as interleukin-2 (IL2), granulocyte/macrophage colony-stimulating factor (GM-CSF), and interleukin-12 (IL12). The goal of this approach to cancer therapy is to concentrate the cytokine in the tumor microenvironment and in so doing directly enhance the tumoricidal effect of the antibody and/or enhance the host immune response (T-cell, B-cell or NK) against the tumor. In the past decade, multiple antibody–cytokine fusion proteins have been developed with different specificities targeting a broad variety of tumors. These novel molecules retain both antibody and cytokine associated functions. In addition, in animals bearing tumors, antibody–cytokine fusion proteins are able to target the tumor and to elicit a significant anti-tumor response that in some cases results in a complete elimination of the tumor. These results suggest that antibody–cytokine fusion proteins have potential for use in the treatment of human cancer. In the present review, we describe strategies for construction of antibody–cytokine fusion proteins and discuss the properties of several antibody–cytokine fusion proteins with IgG genetically fused to the cytokines IL2, GM-CSF or IL12.

Introduction

The management of residual disease is a central problem in the treatment of cancer. Despite improvements in treatment protocols, relapse remains a critical and generally fatal problem in high risk cancer patients. Chemotherapeutic strategies are necessarily limited by various toxicities and of limited efficacy. Therefore, additional modalities are needed to achieve disease containment or elimination. Systemic treatment with cytokines such as IL2, IL12 and GM-CSF can render some non-immunogenic tumors immunogenic, activating a protective immune response (Rosenberg et al., 1998, Ruef and Coleman, 1990, Tsung et al., 1997). However, when cytokines are given systemically there are frequently problems with severe toxic side effects that make it impossible to achieve an effective dose at the site of the tumor (Cohen, 1995, Maas et al., 1993, Ruef and Coleman, 1990, Siegel and Puri, 1991).

More effective treatment with cytokines could be achieved if methods were developed to provide effective concentrations in the tumor while limiting generalized toxicity. Direct injection into the site of the tumor has been one approach to this problem (Cortesina et al., 1988, Forni et al., 1987, Maas et al., 1989, Maas et al., 1991, Pizza et al., 1984, Rutten et al., 1989). However, this requires that the tumor be localized and accessible and direct injection into micrometastases is not possible. Another approach has been cytokine gene therapy whereby tumor cells are removed from the patients, transduced or transfected with the cytokine of interest, and reintroduced into the patient with the expectation that a systemic immune response will be elicited against the tumors (Dranoff and Mulligan, 1995, Hurford et al., 1995, Maass et al., 1995, Schmidt et al., 1995, Soiffer et al., 1998, Su et al., 1994, Zatloukal et al., 1995). Although the results suggest that this immunization strategy has potential application in the treatment of minimal residual disease, the ex vivo genetic modification and reintroduction of cells into patients is limited by its patient specific nature. Additionally, it is technically difficult, time consuming and expensive to expand primary autologous human tumor cells to the numbers required for vaccination (Hrouda et al., 1999, Simons et al., 1997, Simons et al., 1999, Soiffer et al., 1998). While in vivo gene delivery using viral vectors has been considered, the in vivo low transfer efficiency of viral vectors and their immunogenicity and safety limit their use (Hrouda et al., 1999). In addition, surface glycoproteins of many viral vectors bind to receptors prevalent on a variety of cells, such non-specific interaction directly decreases the transfection efficiency in vivo (Smith and Wu, 1999). Moreover, a significant fraction of the human population carries preexisting antibodies to viral vectors and such unfavorable immune responses decrease the half-life of vectors (Piedra et al., 1998). Thus the challenge is to develop an alternative approach for achieving effective local concentrations of cytokines.

Tumor specific Abs genetically fused to cytokines provide an alternative approach for concentrating in the region of the tumors quantities of cytokine sufficient to elicit a significant anti-tumor activity without accompanying systemic toxicity. In fact, in the past decade, we and others have developed several Ab-cytokine fusion proteins specific for different TAAs. In preclinical trials using murine model such Ab-cytokine fusion proteins have been shown to be very effective anti-cancer agents suggesting that they may be useful in the treatment of human cancer. As the number and diversity of Ab-cytokine fusion proteins has dramatically increased in the last years the present review cannot include all of them. Instead we will focus this review on a subset of Ab-cytokine fusion proteins consisting of IgG genetically fused to the cytokines IL2, GM-CSF or IL12.

Section snippets

Ab-(IL2) fusion proteins

Originally known as ‘T cell growth factor’, IL2 is a cytokine produced by T helper cells which stimulates T cells to proliferate and become cytotoxic (Grimm et al., 1982, Hank et al., 1990, Lotze et al., 1981, Yron et al., 1980) and NK cells to respond with increased cytotoxicity toward tumor cells (Grimm et al., 1982). These properties suggest that Ab-IL2 fusion proteins targeting cancer cells may be effective for cancer treatment. Indeed, among the Ab-cytokine fusion proteins, Ab-IL2 fusion

Ab-(GM-CSF) fusion proteins

GM-CSF is a cytokine associated with the growth and differentiation of hematopoietic cells. It is also a potent immunostimulator with pleiotropic effects, including the augmentation of Ag presentation in a variety of cells (Blanchard and Djeu, 1991, Fischer et al., 1988, Heufler et al., 1988, Morrissey et al., 1987, Smith et al., 1990, Steis et al., 1990), increased expression of MHC class II on monocytes and adhesion molecules on granulocytes and monocytes (Arnaout et al., 1986, Grabstein et

Ab–(IL12) fusion proteins

IL12, a cytokine normally released by professional Ag-presenting cells, promotes cell-mediated immunity (Trinchieri, 1995) by inducing naive CD4+T cells to differentiate into Th1 cells (Gracie and Bradley, 1996, Hsieh et al., 1993). In addition, IL12 has the ability to enhance the cytotoxicity of NK and CD8+ T cells (Farrar and Schreiber, 1993, Gately et al., 1994). Moreover, the IFN-γ produced by IL12 stimulated T and NK cells can retard tumor growth by inhibiting tumor angiogenesis (Voest et

Conclusions

In the last decade many studies have shown that the genetic fusion of anti-cancer Ab with cytokines results in novel proteins which retain both Ab and cytokine functions and show superior anti-cancer activity compared with equivalent amount of free Ab and cytokines or non tumor specific Ab cytokine fusion proteins. These findings support the hypothesis that Ab–cytokine fusion proteins can specifically target the cytokine to the tumor microenvironment and in so doing stimulate the immune

Acknowledgements

Our work was supported in part by grants CA-16858, CA-68465, AI-39187, AI-29470 from the National Institutes of Health, ACS-IM-77313 from the American Cancer Society, 3CB-0245 from the University of California Breast Cancer Research Program, Susan G. Komen Breast Cancer Foundation Grant 9855, Department of Defense Breast Cancer Research Program Grant BC980134, Tumor Immunology Training Grant 5-T32-CA09120-24 from NCI (NIH) and Cancer Center Core grant CA-16042 (UCLA).

References (82)

  • J.C. Becker et al.

    Long-lived and transferable tumor immunity in mice after targeted interleukin-2 therapy

    J. Clin. Invest.

    (1996)
  • D.K. Blanchard et al.

    Differential modulation of surface antigens on human macrophages by IFN-gamma and GM-CSF: effect on susceptibility to LAK lysis

    J. Leukoc. Biol.

    (1991)
  • P.M. Challita-Eid et al.

    A B7.1–antibody fusion protein retains antibody specificity and ability to activate via the T cell costimulatory pathway

    J. Immunol.

    (1998)
  • J. Cohen

    Clinical trials — IL-12 deaths — Explanation and a puzzle

    Science

    (1995)
  • G. Cortesina et al.

    Treatment of recurrent squamous cell carcinoma of the head and neck with low doses of interleukin-2 injected perilymphatically

    Cancer

    (1988)
  • J.L. Dangl et al.

    Segmental flexibility and complement fixation of genetically engineered chimeric human, rabbit and mouse antibodies

    EMBO J.

    (1988)
  • N.K. Damle et al.

    IL-2-activated human killer lymphocytes but not their secreted products mediate increase in albumin flux across cultured endothelial monolayers. Implications for vascular leak syndrome

    J. Immunol.

    (1989)
  • J.H. Donohue et al.

    The fate of interleukin-2 after in vivo administration

    J. Immunol.

    (1983)
  • A. Dowlati et al.

    Soluble and cell-associated transferrin receptor in lung cancer

    Br. J. Cancer

    (1997)
  • T. Dreier et al.

    Recombinant immunocytokines targeting the mouse transferrin receptor: Construction and biological activities

    Bioconjug. Chem.

    (1998)
  • M.A. Farrar et al.

    The molecular cell biology of interferon-gamma and its receptor

    Annu. Rev. Immunol.

    (1993)
  • H.G. Fischer et al.

    Granulocyte-macrophage colony-stimulating factor activates macrophages derived from bone marrow cultures to synthesis of MHC class II molecules and to augmented antigen presentation function

    J. Immunol.

    (1988)
  • G. Forni et al.

    Local administration of interleukin-2 activates lymphocytes from tumor bearing mice to recruit host immunoreactivity and inhibit tumor growth

    Prog. Clin. Biol. Res.

    (1987)
  • M.K. Gately et al.

    Administration of recombinant IL-12 to normal mice enhances cytolytic lymphocyte activity and induces production of IFN-gamma in vivo

    Int. Immunol.

    (1994)
  • S.D. Gillies et al.

    Antibody–IL-12 fusion proteins are effective in SCID mouse models of prostate and colon carcinoma metastases

    J. Immunol.

    (1998)
  • K.H. Grabstein et al.

    Induction of macrophage tumoricidal activity by granulocyte-macrophage colony-stimulating factor

    Science

    (1986)
  • J.A. Gracie et al.

    Interleukin-12 induces interferon-gamma-dependent switching of IgG alloantibody subclass

    Eur. J. Immunol.

    (1996)
  • E.A. Grimm et al.

    Lymphokine-activated killer cell phenomenon. Lysis of natural killer-resistant fresh solid tumor cells by interleukin 2-activated autologous human peripheral blood lymphocytes

    J. Exp. Med.

    (1982)
  • U. Gubler et al.

    Coexpression of two distinct genes is required to generate secreted bioactive cytotoxic lymphocyte maturation factor

    Proc. Natl. Acad. Sci. USA

    (1991)
  • J.A. Hank et al.

    Augmentation of antibody dependent cell mediated cytotoxicity following in vivo therapy with recombinant interleukin 2

    Cancer Res.

    (1990)
  • E.T. Harvill et al.

    In vivo properties of an IgG3–IL-2 fusion protein. A general strategy for immune potentiation

    J. Immunol.

    (1996)
  • C. Heufler et al.

    Granulocyte/macrophage colony-stimulating factor and interleukin 1 mediate the maturation of murine epidermal Langerhans cells into potent immunostimulatory dendritic cells

    J. Exp. Med.

    (1988)
  • J.L. Hornick et al.

    Pretreatment with a monoclonal antibody/interleukin-2 fusion protein directed against DNA enhances the delivery of therapeutic molecules to solid tumors

    Clin. Cancer. Res.

    (1999)
  • D. Hrouda et al.

    Gene therapy for prostate cancer

    Semin. Oncol.

    (1999)
  • C.S. Hsieh et al.

    Development of Th1 Cd4+ T-cells through IL-12 produced by listeria-induced macrophages

    Science

    (1993)
  • P. Hu et al.

    A chimeric Lym-1/interleukin 2 fusion protein for increasing tumor vascular permeability and enhancing antibody uptake

    Cancer Res.

    (1996)
  • R.K.J. Hurford et al.

    Gene therapy of metastatic cancer by in vivo retroviral gene targeting

    Nat. Genet.

    (1995)
  • N.V. Katre et al.

    Chemical modification of recombinant interleukin 2 by polyethylene glycol increases its potency in the murine Meth A sarcoma model

    Proc. Natl. Acad. Sci. USA

    (1987)
  • T.S. Kim et al.

    An ovalbumin–IL-12 fusion protein is more effective than ovalbumin plusfree recombinant IL-12 in inducing a T helper cell type 1-dominated immune response and inhibiting antigen-specific IgE production

    J. Immunol.

    (1997)
  • G.J. Lieschke et al.

    Bioactive murine and human interleukin-12 fusion proteins which retain antitumor activity in vivo

    Nat. Biotechnol.

    (1997)
  • M.T. Lotze et al.

    Lysis of fresh and cultured autologous tumor by human lymphocytes cultured in T-cell growth factor

    Cancer Res.

    (1981)
  • Cited by (77)

    • Interferon-γ inhibits retinal neovascularization in a mouse model of ischemic retinopathy

      2021, Cytokine
      Citation Excerpt :

      The antibody is linked to a cytokine to provide target specificity of cytokines. As a result, if IFNG is conjugated with EC-specific antibody, this cytokine could be delivered to ECs selectively to regulate angiogenic responses [22–24]. Recently, our group reported that the TSPAN12 antagonist antibody can potently inhibit both cellular angiogenesis and pathological blood vessel formation in the OIR model.

    • Targeted Delivery of TNF Potentiates the Antibody-Dependent Cell-Mediated Cytotoxicity of an Anti-Melanoma Immunoglobulin

      2019, Journal of Investigative Dermatology
      Citation Excerpt :

      Figure 5). Antibody-cytokine fusions, capable of selective localization in the tumor mass, can substantially increase the local density of NK cells and other leukocytes (Borsi et al., 2003; Carnemolla et al., 2002; De Luca et al., 2017; Halin et al., 2002; Lode et al., 1998; Moschetta et al., 2012; Penichet and Morrison, 2001). The antibody-based delivery of TNF to components of the tumor extracellular matrix can mediate a hemorrhagic necrosis of the tumor mass (van Horssen et al., 2006), followed by activation of the immune system against the (few) residual tumor cells (De Luca et al., 2017; Probst et al., 2017).

    • Antibody-cytokine fusion proteins for improving efficacy and safety of cancer therapy

      2017, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      These “antibody-cytokine fusion proteins” merge the specific targeting capability of antibodies with the immunomodulatory functions of cytokines [12–14]. The primary objective of this approach is to concentrate cytokines in the tumor site, enhance the tumoricidal effect of the antibody molecule, and increase the host immune reaction against tumor, while severe toxic effects caused by high amounts of systemic cytokine are prevented [15–18]. Now, most of antibody fusion proteins developed for tumor immunotherapy are antibody–cytokine fusion proteins, which consist of an antibody able to target a tumor-associated antigen and a cytokine able to raise the immune reaction (Table 1).

    • Construction, expression and characterization of a fusion protein HBscFv-IFNγ in Komagatella (Pichia) pastoris X33

      2017, Enzyme and Microbial Technology
      Citation Excerpt :

      The scFv antibody fragment is a small recombinant protein in which the variable light (VL) and heavy (VH) chain domains of an antibody molecule are connected by a short peptide linker, usually the (Gly4Ser)3 linker [4]. The small fragment, whose molecular weight is only one-sixth of the whole antibody, is considered promising for medical applications since it has several advantages including superior tissue penetration, absence of side reactions involving the constant domains, and facile engineering for fusion proteins, such as scFv-coupled toxins, enzymes and cytokines for prodrug activation or the creation of multivalent or bispecific proteins [5,6]. Recently, possible clinical utility of scFv has been reported [7–10].

    View all citing articles on Scopus
    View full text