Survey
Tumor necrosis factor, cancer and anticancer therapy

https://doi.org/10.1016/j.cytogfr.2004.11.001Get rights and content

Abstract

Tumor necrosis factor (TNF) is being utilized as an antineoplastic agent for the treatment of patients with locally advanced solid tumors. However, its role in cancer therapy is debated. Although a large body of evidence supports TNF's antineoplastic activity, the cascade of molecular events underlying TNF-mediated tumor regression observed in vivo is still incompletely elucidated. Intriguingly, some pre-clinical findings suggest that TNF may promote cancer development and progression, which has led to propose anti-TNF therapy as a novel approach to malignancies. In the present work, we summarize the molecular biology of TNF with particular regard to its tumor-related properties, and review the experimental and clinical evidence currently available describing the complex and sometime conflicting relationship between this cytokine, cancer and antitumor therapy. Recent insights that might pave the way to further exploitation of the antineoplastic potential of TNF are also discussed.

Introduction

Several cytokines have been tested as antineoplastic agents, both alone and in combination with conventional chemotherapeutic agents or antitumor vaccines [1], [2]. From the historical viewpoint, tumor necrosis factor (TNF, formerly referred to as TNF-α) was the first cytokine to be employed for cancer biotherapy. TNF is a pleiotropic protein [3] that was first isolated from the serum of mice treated with bacterial endotoxin, and was shown to replicate the ability of endotoxin to induce haemorrhagic necrosis of methylcholanthrene-induced sarcomas [4]. This discovery ended a long search for the active component of “Coley's toxin”, which consisted of a crude bacterial filtrate developed by Willam Coley, a New York surgeon and pioneer of cancer biotherapy at the turn of the 20th century [5]. Coley's toxin-induced high fever and tumor necrosis in some patients affected with sarcoma, carcinoma and lymphoma. The phenomenon is now attributed, at least in part, to the effect of macrophage-derived TNF released upon activation of these leukocytes by the endotoxin contained within Coley's mixture.

In the 1980s, TNF was also characterized as cachectin [6] for its role in the wasting syndrome, and as T-lymphocyte differentiation factor [7]. Cloning of TNF gene in 1984 [8] led to the era of clinical experimentation. As systemic TNF administration was associated with lack of objective tumor response and serious side-effects, researchers investigated the delivery of TNF through the locoregional route, which culminated in a license from the European Medicine Evaluation Agency (EMEA) for the TNF-based treatment of limb-threatening soft tissue sarcomas [9].

Section snippets

TNF and its receptors

The TNF gene is located on chromosome 6p21.3 and is mainly expressed by activated macrophages, NK-cells and T-lymphocytes, even though other cell types (e.g. fibroblasts, astrocytes, Kupffer cells, smooth-muscle cells, keratinocytes, and tumor cells) have been shown to express it [10].

In its soluble form, TNF acts as a homotrimer with a subunit molecular mass of 17 kDa (157 amino acids). TNF is first synthesized as a 26 kDa (233 amino acids) membrane-bound pro-peptide (pro-TNF) and is released

Direct cytotoxic effects

The in vitro antiproliferative/cytotoxic property of TNF was among the first activities attributed to the cytokine [2]. TNF-R1 is expressed on virtually any cell type and has been demonstrated to exert cytostatic/cytotoxic effects on some animal and human tumor cell lines. Intriguingly, the genomic derangement proper of cancer development can itself sensitize malignant cells to TNF-driven cell death by leading to the overexpression of cathepsin-B [30], which might partly explain the selective

TNF tumor promoting effects

While physiologically TNF plays a major role in growth regulation, cell differentiation, response to viral, bacterial, fungal, and parasitic infections, its inappropriate overexpression has been implicated in the pathogenesis of a wide spectrum of human disorders, such as autoimmunity (e.g. multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease), allergy, septic shock, allograft rejection and insulin resistance [10]. Despite the above-discussed evidence of its antitumor activity,

TNF circulating levels and gene polymorphism

Cancer anorexia-cachexia syndrome (CACS) is a combination of anorexia, tissue wasting, weight loss and poor performance status. Although CACS signs/symptoms also depend upon macrophage-derived IL-1 and tumor-derived IL-6, TNF is considered a major mediator of this syndrome [107]. A consistent correlation has been reported between TNF plasma levels and occurrence of cachexia in patients with different tumor types. Following the promising results obtained in animals [108], the use of TNF

TNF-based anticancer therapy

The dose-limiting side effect of TNF systemic administration is represented by dose-dependent hypotension that can progress to a shock-like syndrome due to a depression of the cardiac function combined with a capillary leak syndrome resembling that of septic shock. Unfortunately, at the maximum tolerated dose (150–300 μg/m2), TNF systemic administration is not associated with significant antitumor activity, as demonstrated by phases I–II trials [2]. This is the reason why in the clinical setting

TNF muteins

Splitting the molecular domains of TNF responsible for the cytokine antitumor properties from those causing systemic toxicity has been the objective of studies on TNF analogues [142]. Moreover, muteins selectively binding to a specific TNF-R might show reduced tumor promoting effects, as supported by the observation that TNF-induced cytotoxicity towards some malignant cells lines is mediated by interaction with TNF-R1, whereas TNF-induced proliferation of these cells is mediated by the

Conclusions

The interest in TNF as an antineoplastic agent is witnessed by the continuous flow of scientific production on this subject [203], [204], [205]. The role played by TNF in cancer development/progression, even if less understood than that in the pathogenesis of autoimmune/inflammatory disorders, might appear to shadow the anticancer potential of this cytokine. Although further elucidation of TNF biology and the conduction of novel clinical trials are warranted to explain some apparent

Acknowledgements

We apologize to those authors whose work was not cited due to space limitations.

References (222)

  • A.M. Eggermont et al.

    Current uses of isolated limb perfusion in the clinic and a model system for new strategies

    Lancet Oncol

    (2003)
  • A. Bierhaus et al.

    Mechanism of the tumor necrosis factor alpha-mediated induction of endothelial tissue factor

    J Biol Chem

    (1995)
  • E. Napoleone et al.

    Monocytes upregulate endothelial cell expression of tissue factor: a role for cell–cell contact and cross-talk

    Blood

    (1997)
  • J.C. Lewis et al.

    Tissue factor expression during coculture of endothelial cells and monocytes

    Exp Mol Pathol

    (1995)
  • B. Stoelcker et al.

    Tumor necrosis factor induces tumor necrosis via tumor necrosis factor receptor type 1-expressing endothelial cells of the tumor vasculature

    Am J Pathol

    (2000)
  • P.C. Brooks et al.

    Integrin alpha v beta 3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels

    Cell

    (1994)
  • T.W. Bauer et al.

    A human melanoma xenograft in a nude rat responds to isolated limb perfusion with TNF plus melphalan

    Surgery

    (2003)
  • F. Balkwill

    Tumor necrosis factor or tumor promoting factor?

    Cytokine Growth Factor Rev

    (2002)
  • A. Ferrajoli et al.

    The clinical significance of tumor necrosis factor-alpha plasma level in patients having chronic lymphocytic leukemia

    Blood

    (2002)
  • R.Y. Liu et al.

    Activation of p38 mitogen-activated protein kinase is required for tumor necrosis factor-alpha supported proliferation of leukemia and lymphoma cell lines

    J Biol Chem

    (2000)
  • S.T. Malik et al.

    Cells secreting tumour necrosis factor show enhanced metastasis in nude mice

    Eur J Cancer

    (1990)
  • S.A. Rosenberg

    Principles and practice of the biologic therapy of cancer

    (2000)
  • E.A. Carswell et al.

    An endotoxin-induced serum factor that causes necrosis of tumors

    Proc Natl Acad Sci USA

    (1975)
  • W. Coley

    Further observations upon the treatment of malignant tumors with the toxins of erysipelas and Bacillus prodigiosus with a report of 160 cases

    Bull Johns Hopkins Hosp

    (1896)
  • B. Beutler et al.

    Identity of tumour necrosis factor and the macrophage-secreted factor cachectin

    Nature

    (1985)
  • K. Takeda et al.

    Identity of differentiation inducing factor and tumour necrosis factor

    Nature

    (1986)
  • T. Shirai et al.

    Cloning and expression in Escherichia coli of the gene for human tumour necrosis factor

    Nature

    (1985)
  • J. Oppenheim et al.

    Cytokine reference

    (2000)
  • M.H. Bemelmans et al.

    Tumor necrosis factor: function, release and clearance

    Crit Rev Immunol

    (1996)
  • B. Aggarwal

    Signaling pathways of the TNF superfamily: a double edged sword

    Nat Rev Immunol

    (2003)
  • G. Chen et al.

    TNF-R1 signaling: a beautiful pathway

    Science

    (2002)
  • S. Ghosh et al.

    Missing pieces in the NF-kappaB puzzle

    Cell

    (2002)
  • X. Li et al.

    TNF-RII and c-IAP1 mediate ubiquitination and degradation of TRAF2

    Nature

    (2002)
  • M. Leist et al.

    Four deaths and a funeral: from caspases to alternative mechanisms

    Nat Rev Mol Cell Biol

    (2001)
  • L. Foghsgaard et al.

    Cathepsin B acts as a dominant execution protease in tumor cell apoptosis induced by tumor necrosis factor

    J Cell Biol

    (2001)
  • J.W. Kim et al.

    Activation of death-inducing signaling complex (DISC) by pro-apoptotic C-terminal fragment of RIP

    Oncogene

    (2000)
  • M. Irmler et al.

    Inhibition of death receptor signals by cellular FLIP

    Nature

    (1997)
  • M. Jaattela et al.

    Bcl-x and Bcl-2 inhibit TNF and Fas-induced apoptosis and activation of phospholipase A2 in breast carcinoma cells

    Oncogene

    (1995)
  • S. Fulda et al.

    Sensitization for death receptor- or drug-induced apoptosis by re-expression of caspase-8 through demethylation or gene transfer

    Oncogene

    (2001)
  • B.B. Aggarwal

    Tumour necrosis factors receptor associated signalling molecules and their role in activation of apoptosis JNK and NF-kappaB

    Ann Rheum Dis

    (2000)
  • C. De Nadai et al.

    Nitric oxide inhibits tumor necrosis factor-alpha-induced apoptosis by reducing the generation of ceramide

    Proc Natl Acad Sci USA

    (2000)
  • N. Fehrenbacher et al.

    Sensitization to the lysosomal cell death pathway upon immortalization and transformation

    Cancer Res

    (2004)
  • M.A. Palladino et al.

    Characterization of the antitumor activities of human tumor necrosis factor-alpha and the comparison with other cytokines: induction of tumor-specific immunity

    J Immunol

    (1987)
  • K. Shimomura et al.

    Recombinant human tumor necrosis factor-alpha: thrombus formation is a cause of anti-tumor activity

    Int J Cancer

    (1988)
  • P.A. Van de Wiel et al.

    Macroscopic and microscopic early effects of tumour necrosis factor on murine Meth A sarcoma, and relation to curative activity

    J Pathol

    (1989)
  • M. Furrer et al.

    Lack of antitumour activity of human recombinant tumour necrosis factor-alpha, alone or in combination with melphalan in a nude mouse human melanoma xenograft system

    Melanoma Res

    (1997)
  • F.R. Balkwill et al.

    Human tumor xenografts treated with recombinant human tumor necrosis factor alone or in combination with interferons

    Cancer Res

    (1986)
  • L.F. Fajardo et al.

    Dual role of tumor necrosis factor-alpha in angiogenesis

    Am J Pathol

    (1992)
  • N. Watanabe et al.

    Toxic effect of tumor necrosis factor on tumor vasculature in mice

    Cancer Res

    (1988)
  • P.L. Naredi et al.

    The effects of tumour necrosis factor alpha on the vascular bed and blood flow in an experimental rat hepatoma

    Int J Cancer

    (1993)
  • Cited by (0)

    View full text