当前位置: 首页 > 期刊 > 《肿瘤学家》 > 2006年第4期 > 正文
编号:11025968
TNF- in Cancer Treatment: Molecular Insights, Antitumor Effects, and Clinical Utility
http://www.100md.com 《肿瘤学家》
     LEARNING OBJECTIVES

    After completing this course, the reader will be able to:

    Discuss the role of TNF-a in cancer survival and apoptosis.

    Describe the mechanism of chemotherapy potentiation by TNF-a.

    Explain the selective targeting of tumor vasculature by TNF-a.

    Discuss TNFR-1 and TNFR-2 signaling.

     ABSTRACT

    Tumor necrosis factor alpha (TNF-), isolated 30 years ago, is a multifunctional cytokine playing a key role in apoptosis and cell survival as well as in inflammation and immunity. Although named for its antitumor properties, TNF has been implicated in a wide spectrum of other diseases. The current use of TNF in cancer is in the regional treatment of locally advanced soft tissue sarcomas and metastatic melanomas and other irresectable tumors of any histology to avoid amputation of the limb. It has been demonstrated in the isolated limb perfusion setting that TNF- acts synergistically with cytostatic drugs. The interaction of TNF- with TNF receptor 1 and receptor 2 (TNFR-1, TNFR-2) activates several signal transduction pathways, leading to the diverse functions of TNF-. The signaling molecules of TNFR-1 have been elucidated quite well, but regulation of the signaling remains unclear. Besides these molecular insights, laboratory experiments in the past decade have shed light upon TNF- action during tumor treatment. Besides extravasation of erythrocytes and lymphocytes, leading to hemorrhagic necrosis, TNF- targets the tumor-associated vasculature (TAV) by inducing hyperpermeability and destruction of the vascular lining. This results in an immediate effect of selective accumulation of cytostatic drugs inside the tumor and a late effect of destruction of the tumor vasculature. In this review, covering TNF- from the molecule to the clinic, we provide an overview of the use of TNF- in cancer starting with molecular insights into TNFR-1 signaling and cellular mechanisms of the antitumor activities of TNF- and ending with clinical response. In addition, possible factors modulating TNF- actions are discussed.

    INTRODUCTION

    Tumor necrosis factor alpha (TNF-) is a multifunctional cytokine involved in apoptosis, cell survival, inflammation, and immunity acting via two receptors [1, 2]. Currently it is used in cancer treatment in the isolated limb perfusion (ILP) setting for soft tissue sarcoma (STS), irresectable tumors of various histological types, and melanoma intransit metastases confined to the limb [3]. TNF- was isolated in 1975 from the serum of mice treated with bacterial endotoxin as the active component of "Coley’s toxin" and was shown to induce hemorrhagic necrosis of mice tumors [4, 5]. It was almost a century ago that William Coley, a surgeon from New York, observed high fever and tumor necrosis in some cancer patients treated with his bacterial filtrate ("Coley’s mixed toxins") [6]. A decade after its isolation, TNF- was also characterized as "cachectin" and as T-lymphocyte differentiation factor [7, 8]. In 1984, the human TNF- gene was cloned [9, 10], and a range of clinical experiments were set up, leading to a license from the European Agency for the Evaluation of Medicinal Products (EMEA) for the treatment of limb-threatening STS in an isolated perfusion setting [11].

    TNF- AND TNF RECEPTOR 1 SIGNALING

    TNF- is a 17-kDa protein consisting of 157 amino acids that is a homotrimer in solution. In humans, the gene is mapped to chromosome 6 [12]. Its bioactivity is mainly regulated by soluble TNF-–binding receptors. TNF- is mainly produced by activated macrophages, T lymphocytes, and natural killer (NK) cells. Lower expression is known for a variety of other cells, including fibroblasts, smooth muscle cells, and tumor cells. In cells, TNF- is synthesized as pro-TNF (26 kDa), which is membrane-bound and is released upon cleavage of its pro domain by TNF-converting enzyme (TACE) [13].

    As mentioned above, TNF- acts via two distinct receptors [14]. Although the affinity for TNF receptor 2 (TNFR-2) is five times higher than that for TNFR-1 [15], the latter initiates the majority of the biological activities of TNF-. TNFR-1 (p60) is expressed on all cell types, while TNFR-2 (p80) expression is mainly confined to immune cells [16]. The major difference between the two receptors is the death domain (DD) of TNFR-1 that is absent in TNFR-2. For this reason, TNFR-1 is an important member of the death receptor family that shares the capability of inducing apoptotic cell death [17]. Besides this apoptotic signaling, TNFR-1 is widely studied because it is a dual role receptor: next to induction of apoptosis, it also has the ability to transduce cell survival signals. Although signaling pathways are well defined nowadays, the life-death signaling regulation is still poorly understood [18, 19]. The TNFR-1 signaling pathways are depicted in Figure 1. Upon binding of the homotrimer TNF-, TNFR-1 trimerizes, and silencer of death domain (SODD) protein is released [20]. TNFR-associated death domain (TRADD) binds to the DD of TNFR-1 and recruits the adaptor proteins receptor interacting protein (RIP), TNFR-associated factor 2 (TRAF-2), and Fas-associated death domain (FADD) [21]. In turn, these adaptor proteins recruit key molecules that are responsible for further intra-cellular signaling. When TNFR-1 signals apoptosis, FADD binds pro-caspase-8, which is subsequently activated. This activation initiates a protease cascade leading to apoptosis, also involving the mitochondria and with caspases as key regulators [22]. The ultimate event in this apoptotic signaling is the activation of endonucleases, like EndoG, resulting in DNA fragmentation. Alternatively, when TNFR-1 signals survival, TRAF-2 is recruited to the complex, which inhibits apoptosis via cytoplasmic inhibitor of apoptosis protein (cIAP). The binding of TRAF-2 initiates a pathway of phosphorylation steps resulting in the activation of cFos/cJun transcription factors via mitogen-activated protein kinase (MAPK) and cJun N-terminal kinase (JNK) [23]. The major signaling event of TRAF-2 and RIP is the widely studied activation of nuclear factor kappa B (NF-B) transcription factor via NF-B–inducing kinase (NIK) and the inhibitor of B kinase (IKK) complex [24]. Both the NF-B and cFos/cJun transcription factors induce transcription of antiapoptotic, proliferative, immunomodulatory, and inflammatory genes. NF-B is the major survival factor in preventing TNF-–induced apoptosis, and inhibition of this transcription factor may improve the efficacy of apoptosis-inducing cancer therapies [25]. NF-B activation in many human malignancies is aberrant or constitutive, and its role in the regulation of the apoptosis–proliferation balance in tumor cells indicates its role in oncogenesis [26, 27]. For further details on the dual signaling of TNFR-1, see Figure 1.

    IMPLICATIONS FOR CELLULAR MECHANISMS UNDERLYING TNF- EFFECTS DURING SOLID TUMOR TREATMENT

    It is widely known that TNF- induces hemorrhagic necrosis in a certain set of tumor types. To investigate the underlying mechanisms of TNF- action during ILP of solid tumors in humans, we set up perfusion models in rats and reported that hemorrhagic necrosis was much greater in tumors treated with TNF- and chemotherapeutic drugs [28]. In addition, we showed a synergistic antitumor effect of the combination treatment with TNF- and chemotherapeutic drugs [29]. In contrast, TNF- alone induced only a mild central necrosis, and there was no objective tumor response observed. The same rat models also revealed that the addition of TNF- improved the accumulation of chemotherapeutic drugs selectively in the tumor up to three- to sixfold. The augmented uptake of melphalan added to the molecular properties of this small molecule (distribution by gradient instead of convection) resulted in intratumoral concentrations close to the 50% inhibitory concentration (IC50) in STS cells in vitro [30, 31]. These levels result in tumor cell kill in the ILP setting, and melphalan can distribute within the well-perfused parts of the tumor even though the intratumoral pressure is high. This selective uptake of melphalan by the tumor was also observed when other vasoactive drugs were used in the ILP setting (see below). It is important to note that the cell lines we used were not sensitive to TNF- in vitro, which is in accordance with other reports describing a lack of effect of TNF- and no synergism with cytotoxic drugs in cell lines [32, 33]. Next to these ILP data, studies in mice and rats showed that a systemic low dose of TNF- augments the antitumor activity of pegylated liposomal doxorubicin [34, 35]. These observations are comprehensible clues that mechanisms underlying the TNF- effect during solid tumor treatment cannot be caused by a direct cytotoxic or cytostatic effect of TNF- toward the tumor cells. It was suggested that, rather than tumor cells themselves, cells of the tumor stroma may be responsible for the observed antitumor effect of TNF- in patients. This hypothesis was confirmed by data from mice experiments revealing that TNF- had a cytotoxic effect on tumor vasculature [36].

    ANGIOGENESIS AND TUMOR-ASSOCIATED VASCULATURE

    Angiogenesis, the formation of new blood vessels from pre-existing ones, has become a major field of research, mainly in cancer [37]. Angiogenesis is essential for a tumor to provide the tumor cells with oxygen and essential nutrients for growth and to metastasize hematogenically [38]. A growing tumor activates surrounding vessels by secreting angiogenic factors, thereby changing the dormant tumor phenotype toward an angiogenic one, the so-called "angiogenic switch" [39]. Activated endothelial cells have to migrate toward the tumor along a newly formed matrix, the components of which are synthesized by themselves, tumor cells, and other cells such as macrophages and fibroblasts [40]. Figure 2 shows schematically the process of tumor angiogenesis, which can be divided into four different stages. A small, dormant tumor (stage 1) can, depending on the nature of the tumor and its microenvironment, make the angiogenic switch to ensure exponential growth. The tumor secretes growth factors to activate endothelial cells of surrounding vessels (stage 2). Upon activation, these endothelial cells start to migrate and proliferate toward the tumor. Only one endothelial cell starts an angiogenic sprout and develops into an endothelial tip cell migrating along the extracellular matrix (ECM) and guiding the following so-called stalk endothelial cells (stage 3) [41]. Finally, the growing tumor is connected to the vasculature (stage 4). In addition to growth and proliferation, the tumor can metastasize. Malignant tumor cells, by invasion of the vessels, ECM degradation, attachment, and homing to target sites can form distal metastases [42]. The process of tumor angiogenesis results in a tumor-associated vasculature (TAV) that is rather chaotic, both in structure and function. In comparison with normal vessels, tumor vessels have a noncontinuous endothelium, an enlarged basal membrane, and an aberrant pericyte coverage[43]. Frequently in tumors, the vascular hierarchy of arterioles, capillaries, and venules is absent, resulting in loosely associated pericytes [44]. From animal experiments, it is known that pericytes are present in small tumors and more abundant in large tumors [45]. The contribution of pericytes to (anti)-angiogenic therapies is currently an attractive focus of research. On one hand, these characteristics impair tumor blood flow, delivery of oxygen, and therapeutics to the tumor cells and vessel functionality, but on the other hand, these differences may be used as a target. The solid tumors treated by ILP with TNF- have a massive vascular structure consisting of vessels with a phenotype specific to tumor vessels, although detailed study needs to clarify the exact contribution of the TAV to the observed antitumor responses.

     ACTIVITY OF TNF- IN SOLID TUMORS: HYPOTHETICAL MECHANISM

    The vascular differences mentioned above are depicted in Figure 3A. These differences are responsible for a more leaky vasculature in the tumor, with average intraendothelial gaps of 400nm, depending on the tumor type[46]. Blood cells such as lymphocytes and monocytes easily adhere and extravasate into the tumor. We speculate that the endothelial cells of the tumor vessels, compared with normal vessels, have an upregulation of TNFR-1 on their membranes, which may be dependent on TNFR-1–upregulating factors produced by vessel-surrounding cells like tumor cells and macrophages. This upregulation, along with the specific architecture of the endothelial lining, defines the tumor vessels as a specific target for TNF- treatment (Fig. 3B). When TNF- is administered via ILP to treat solid tumors, it binds soluble receptors, and because of the high dosage, TNFR-1 receptors on tumor endothelial cells become occupied. Healthy endothelium, in contrast, also binds TNF-; however, because of a lower number of membrane-bound TNFR-1 receptors (most TNFR-1 is stored in the golgi apparatus [47]), there is no toxicity. We propose that this TNF- to TNFR-1 binding results in hyperpermeability of the tumor vessels, and erythrocytes and other blood cells extravasate. The strong extravasation of erythrocytes results in massive hemorrhagic necrosis of the tumor. As a result of the direct cytotoxicity of high-dose TNF- to endothelial cells, some of these cells undergo apoptosis, and this process strongly enhances the induced hyperpermeability (Fig. 3B). Several studies have shown that a lower dose of TNF- results in comparable responses [48, 49], suggesting that a lower dose still may induce these antivascular effects. The healthy vessels, however, stay intact; no apoptosis and no extravasation occurs. The observed synergistic activity of TNF- and chemotherapeutic drugs is a consequence of this double-induced hyperpermeability. This hyperpermeability throughout the tumor facilitates the augmented accumulation and distribution of the drug in the tumor, resulting in better exposure of the tumor cells to the cytostatic agent [30]. This double-induced hyperpermeability, along with the dual targeting—the TAV (by TNF-) and the tumor cells (by the chemotherapy drug)—is one explanation for the observed synergistic response of tumors to TNF- and chemotherapy that results in high response rates in patients.

     CLINICAL EFFICACY OF TNF-–BASED ISOLATED LIMB PERFUSION

    The use of TNF- in the ILP setting was pioneered by Lienard et al. [50]. In 19 melanoma patients and a few STS cases, impressive and very rapid responses were observed. This observation was followed by multicenter trials in patients with locally advanced STS and melanoma. In Table 1, we present an overview of the multicenter trials in Europe that led to the approval of TNF- by the EMEA in 1998 for its application in ILP for the treatment of high-grade (2–3) STS. In these multicenter trials, an overall response rate of 76% and a median limb salvage rate of 82% were observed. Moreover, this table lists the largest single-center studies in STS that confirm the results of the multicenter experience [49, 51–67]. We observe strikingly consistent major response rates, with a median of 76% (range, 58%–91%), and with a median limb salvage rate of 84% (range, 58%–97%). TNF-–based ILP now is performed in 35 cancer centers in Europe with national referral patterns for limb salvage. ILP with melphalan alone for melanoma in-transit metastases is reported in the literature to result in about a 50% complete response (CR) rate and an 80% overall response rate [68]. The introduction of TNF- in this setting was reported to increase CR rates to 70%–90% and overall response rates to 95%–100%. These results are summarized in Table 2 [50, 69–78]. Early on, however, it was observed that ILP with TNF- plus melphalan (TM-ILP) was especially effective against bulky tumors such as STSs, in which ILP with melphalan alone [79] or doxorubicin alone [79, 80] fails. It should also be noted, of course, that both drugs have no activity against melanoma in the systemic setting and that melphalan has no activity against STS in the systemic setting. TNF-–based ILP with melphalan or doxorubicin results in similar tumor response rates, but because of less locoregional toxicity, melphalan is preferred over doxorubicin in the ILP setting [65–67]. In our own series of 50 ILPs in patients with bulky melanoma in-transit metastases, the CR rate was still 58% [73], identical to the CR rate that was seen in an interim analysis of a randomized trial by Fraker et al. [74], in which TNF- based ILP was shown to be of significant benefit in patients with a high tumor load, increasing the CR rate from 19% for M-ILP to 58% for TM-ILP. Apart from bulky melanoma, a further indication for TNF-–based ILP is response failure to a prior ILP because excellent response rates have been reported in this situation [76, 77]. Similarly, high response rates have been reported for TNF-–based ILP for nonmelanoma locally advanced skin cancers [78]. Because TNF- acts primarily on the tumor vasculature, these observations make sense, and the propensity to respond to a TNF- based ILP is assumed to depend more on tumor vasculature than on the histologic type of the tumor.

    Response of STS to TNF-–based ILP is shown in Figure 4A. Magnetic resonance imaging of a patient with high-grade (6–7) leiomyosarcoma in the upper leg shows clear dark tumor masses with high gadolinium uptake before ILP. Five weeks after ILP, all tumor masses are gadolinium-negative. Along the distal femur, only small tumor remnants are visible, but at the proximal femur, a large but gadolinium-negative tumor mass without signs of regression is visible. All lesions were resected and found to be 100% necrotic. Thus the response was classified as a histopathologic CR.

    Targeting by TNF- of the tumor vasculature is revealed in patients by angiographies before and after ILP. The TAV is selectively destroyed by TNF-–based ILP; the TAV is gone while normal vessels of the limbs are still intact after ILP (Fig. 4B). TNF- targets the vasculature of tumors with completely different histologies, but as the TAV is well developed in all these tumors, combination therapy in the ILP setting is very effective for the specific tumors treated. Synergistic and high response rates are achieved in sarcomas consisting of a broad range of subtypes, as well as in melanomas. At the histopathological level, massive hemorrhagic necrosis is observed inside melanomas treated with ILP [81], an effect likely caused by TNF-–induced coagulation and extravasation of erythrocytes [82]. In accordance with the angiographies of STS, the vascular lining of melanoma tumor vessels is destroyed (Fig. 4C). Staining for endothelial cells reveals that, upon treatment in skin vessels, the endothelium is intact and continuous while this lining is heavily disrupted and the cells detach from the underlying basement membrane in melanoma-associated vessels. These antivascular TNF- effects are achieved by the high concentration reached during ILP. At these high levels, TNF- activity is antivascular and antiangiogenic, while at lower concentration TNF- is known to promote angiogenesis [83]. In addition to direct TAV-mediated effects, TNF- reduces blood flow in tumors in a dose-dependent fashion [84]. This set of antivascular TNF- effects was recently confirmed by experiments revealing that tumor response to TNF- correlates with the degree of tumor vascularity [85]. Along with this dual role in angiogenesis, TNF- is also known for its dual role in cancer treatment, anti-TNF- therapy is also used for several types of cancer. The anti-neoplastic and tumor-promoting effects of TNF- are discussed in a recent review [86].

    APPROACHES TO MODULATE TNF- ACTION IN CANCER TREATMENT

    High response rates in the ILP setting do not avoid the need to search for factors that modulate the TNF- effect in solid tumors. In addition to the possible application of TNF- in other settings(e.g., systemic treatment)and for other tumors types, nonresponding patients in the ILP setting may also benefit from TNF- sensitizers. Some of these approaches are mentioned below. An obvious target is inhibition of the NF-B survival pathway. Inactivation of NF-B is known to sensitize several tumors to TNF- [87]. NF-B can be blocked in several ways: overexpression of its inhibitor IB and selective NF-B inhibitors have been shown to increase TNF-–induced apoptosis of tumor cells [88, 89]. One such inhibitor, bortezomib, has entered the clinical arena as a combination therapy with chemotoxic drugs for prostate cancer and myeloma [90].

    Nitric oxide (NO) is involved in survival of TNF-–treated cells through NF-B–induced expression of inducible NO synthase (iNOS) [91, 92]. We have previously shown that inhibition of NOS by the addition of L-NAME (NG-nitro-L-arginine methyl ester) during TNF-–based ILP resulted in an increased tumor response in rats bearing STSs [93]. These observations were confirmed by a recent study showing that NOS inhibition in endothelial cells reduces their sensitivity to TNF- in vitro, leading to the hypothesis that tumor vessels exhibit a higher level of NOS, which might explain their higher TNF- sensitivity [94]. These studies justify further evaluation of NOS inhibition in tumors of patients treated by ILP to stimulate the anti-TAV activities of TNF-.

    Apoptosis induced by TNF- is also associated with the generation of reactive oxygen species (ROS). It has been shown that the key survival factor NF-B induces ROS-neutralizing enzymes like superoxide dismutase [95]. Induction of ROS production or an inhibition of the NF-B pathway by cyclooxygenase-2 (COX-2) inhibitors is reported to be successful in sensitizing tumor cells to TNF-–induced apoptosis [96].

    Procedures with liposomal encapsulation of TNF- to elongate its circulation time and to achieve effective TNF- concentrations in the tumor have been shown to be effective in systemic treatment in combination with chemotherapeutic drugs in rats [97].

    Another experimental approach is the development of TNF- analogues like TNF- mutants that selectively bind to TNFR-1 [98] and mutants affecting the pharmacokinetics of TNF- to TNFR-1 interactions at the tumor vascular level [99]. In animal models, these approaches have shown promising results versus wild-type TNF-, but this research is still too experimental, and no TNF- analogues have reached clinical studies yet.

    A more physiological and therefore potent way involves the role of other cytokines that serve as TNF- sensitizers. Interferon gamma (IFN-) is one of the widely studied cytokines, and although no beneficial effects and some toxicity were observed in the ILP setting for melanomas and sarcomas, for other cancers, IFN- might be very suitable because of its reported actions on TNFR-1 and caspase-8, thereby regulating TNF-–induced apoptosis [100]. Endothelial monocyte-activating polypeptide II (EMAP-II) is a cytokine produced by tumor cells, so local production of this cytokine is suggested to facilitating TNF- antitumor activity. EMAP-II can sensitize reportedly TNF-–resistant tumors to TNF-, which was shown by the upregulation of EMAP-II resulting in an increased TNF- effect [101]. Underlying mechanisms for this effect remain unclear, although it is postulated that an upregulation of TNFR-1 on endothelial cells might be one explanation [102]. Besides TNF-–modulating effects, EMAP-II exhibits other antitumor effects including antiangiogenic and immunosuppressive activities [103, 104]. Combining these cytokines with TNF- and investigating the expression patterns and local protein production might be very promising in modulating TNF- activities in cancer treatment.

    Besides modulating TNF- actions, other TAV-manipulating agents are being tested in ILP as well. Recently, we showed that both histamine (via its own vascular activity) and interleukin 2 (via upregulation of TNF- production) have strong synergistic antitumor effects when combined with melphalan. Importantly, with both these agents, an augmented accumulation of melphalan in the tumor specifically was observed, confirming TAV as an effective target in solid tumor treatment [105, 106].

    CONCLUSIONS

    The use of high-dose TNF- locally administered in combination with melphalan for patients with metastatic intransit melanoma and STS confined to the limb is a well-established treatment modality nowadays. Furthermore, the modulation of tumor pathophysiology by low-dose TNF- indicates that, in combination with liposomal drugs, systemic therapy should be investigated in the clinic. Besides a greater understanding of the molecular events of the TNFR-1 signaling that takes place during tumor treatment, these studies likely will expand the use of TNF- for other cancer types and for nonresponding ILP patients, as well. The multifunctional properties of TNF- may well result in a more varied application of this cytokine.

    DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST

    Dr. Eggermont has acted as a consultant for Boehringer-Ingelheim, GmbH.

    REFERENCES

    Bazzoni F, Beutler B. The tumor necrosis factor ligand and receptor families. N Engl J Med 1996;334:1717–1725.

    Locksley RM, Killeen N, Lenardo MJ. The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 2001;104:487–501.

    Eggermont AM, de Wilt JH, ten Hagen TL. Current uses of isolated limb perfusion in the clinic and a model system for new strategies. Lancet Oncol 2003;4:429–437.

    Carswell EA, Old LJ, Kassel RL et al. An endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci U S A 1975;72: 3666–3670.

    Old LJ. Tumor necrosis factor (TNF). Science 1985;230:630–632.

    Nauts HC, Fowler GA, Bogatko FH. A review of the influence of bacterial infection and of bacterial products (Coley’s toxins) on malignant tumors in man; a critical analysis of 30 inoperable cases treated by Coley’s mixed toxins, in which diagnosis was confirmed by microscopic examination selected for special study. Acta Med Scand Suppl 1953;276:1–103.

    Beutler B, Greenwald D, Hulmes JD et al. Identity of tumour necrosis factor and the macrophage-secreted factor cachectin. Nature 1985;316: 552–554.

    Takeda K, Iwamoto S, Sugimoto H et al. Identity of differentiation inducing factor and tumour necrosis factor. Nature 1986;323:338–340.

    Shirai T, Yamaguchi H, Ito H et al. Cloning and expression in Escherichia coli of the gene for human tumour necrosis factor. Nature 1985;313: 803–806.

    Marmenout A, Fransen L, Tavernier J et al. Molecular cloning and expression of human tumor necrosis factor and comparison with mouse tumor necrosis factor. Eur J Biochem 1985;152:515–522.

    Lejeune FJ, Ruegg C, Lienard D. Clinical applications of TNF-alpha in cancer. Curr Opin Immunol 1998;10:573–580.

    Muller U, Jongeneel CV, Nedospasov SA et al. Tumour necrosis factor and lymphotoxin genes map close to H-2D in the mouse major histocompatibility complex. Nature 1987;325:265–267.

    Bemelmans MH, van Tits LJ, Buurman WA. Tumor necrosis factor: function, release and clearance. Crit Rev Immunol 1996;16:1–11.

    Vandenabeele P, Declercq W, Beyaert R et al. Two tumour necrosis factor receptors: structure and function. Trends Cell Biol 1995;5:392–399.

    Tartaglia LA, Goeddel DV. Two TNF receptors. Immunol Today 1992;13:151–153.

    Aggarwal BB. Signalling pathways of the TNF superfamily: a double-edged sword. Nat Rev Immunol 2003;3:745–756.

    Ashkenazi A, Dixit VM. Death receptors: signaling and modulation. Science 1998;281:1305–1308.

    Baker SJ, Reddy EP. Modulation of life and death by the TNF receptor superfamily. Oncogene 1998;17:3261–3270.

    Muppidi JR, Tschopp J, Siegel RM. Life and death decisions: secondary complexes and lipid rafts in TNF receptor family signal transduction. Immunity 2004;21:461–465.

    Takada H, Chen NJ, Mirtsos C et al. Role of SODD in regulation of tumor necrosis factor responses. Mol Cell Biol 2003;23:4026–4033.

    Rath PC, Aggarwal BB. TNF-induced signaling in apoptosis. J Clin Immunol 1999;19:350–364.

    Degterev A, Boyce M, Yuan J. A decade of caspases. Oncogene 2003;22:8543–8567.

    Natoli G, Costanzo A, Ianni A et al. Activation of SAPK/JNK by TNF receptor 1 through a noncytotoxic TRAF2-dependent pathway. Cell Signal 1997;275:200–203.

    Devin A, Cook A, Lin Y et al. The distinct roles of TRAF2 and RIP in IKK activation by TNF-R1: TRAF2 recruits IKK to TNF-R1 while RIP mediates IKK activation. Immunity 2000;12:419–429.

    Beg AA, Baltimore D. An essential role for NF-kappaB in preventing TNF-alpha-induced cell death. Science 1996;274:782–784.

    Dolcet X, Llobet D, Pallares J et al. NF-kB in development and progression of human cancer. Virchows Arch 2005;446:475–482.

    Lin A, Karin M. NF-kappaB in cancer: a marked target. Semin Cancer Biol 2003;13:107–114.

    Manusama ER, Stavast J, Durante NM et al. Isolated limb perfusion with TNF alpha and melphalan in a rat osteosarcoma model: a new anti-tumour approach. Eur J Surg Oncol 1996;22:152–157.

    Manusama ER, Nooijen PT, Stavast J et al. Synergistic antitumour effect of recombinant human tumour necrosis factor alpha with melphalan in isolated limb perfusion in the rat. Br J Surg 1996;83:551–555.

    van der Veen AH, de Wilt JH, Eggermont AM et al. TNF-alpha augments intratumoural concentrations of doxorubicinin TNF-alpha-based isolated limb perfusion in rat sarcoma models and enhances anti-tumour effects. Br J Cancer 2000;82:973–980.

    de Wilt JH, ten Hagen TL, de Boeck G et al. Tumour necrosis factor alpha increases melphalan concentration in tumour tissue after isolated limb perfusion. Br J Cancer 2000;82:1000–1003.

    Ruggiero V, Latham K, Baglioni C. Cytostatic and cytotoxic activity of tumor necrosis factor on human cancer cells. J Immunol 1987;138: 2711–2717.

    Watanabe N, Niitsu Y, Yamauchi N et al. Synergistic cytotoxicity of recombinant human TNF and various anti-cancer drugs. Immunopharmacol Immunotoxicol 1988;10:117–127.

    Brouckaert P, Takahashi N, van Tiel ST et al. Tumor necrosis factor-alpha augmented tumor response in B16BL6 melanoma-bearing mice treated with stealth liposomal doxorubicin (Doxil) correlates with altered Doxil pharmacokinetics. Int J Cancer 2004;109:442–448.

    ten Hagen TL, van der Veen AH, Nooijen PT et al. Low-dose tumor necrosis factor-alpha augments antitumor activity of stealth liposomal doxorubicin (DOXIL) in soft tissue sarcoma-bearing rats. Int J Cancer 2000;87:829–837.

    Watanabe N, Niitsu Y, Umeno H et al. Toxic effect of tumor necrosis factor on tumor vasculature in mice. Cancer Res 1988;48:2179–2183.

    Carmeliet P. Angiogenesis in health and disease. Nat Med 2003;9: 653–60.

    Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med 1971;285:1182–1186.

    Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nat Rev Cancer 2003;3:401–410.

    Bussolino F, Mantovani A, Persico G. Molecular mechanisms of blood vessel formation. Trends Biochem Sci 1997;22:251–256.

    Gerhardt H, Golding M, Fruttiger M et al. VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. J Cell Biol 2003;161: 1163–1177.

    Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer 2002;2:563–572.

    Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature 2000;407:249–257.

    Baluk P, Hashizume H, McDonald DM. Cellular abnormalities of blood vessels as targets in cancer. Curr Opin Genet Dev 2005;15:102–111.

    Morikawa S, Baluk P, Kaidoh T et al. Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors. Am J Pathol 2002;160: 985–1000.

    Hashizume H, Baluk P, Morikawa S et al. Openings between defective endothelial cells explain tumor vessel leakiness. Am J Pathol 2000;156:1363–1380.

    Bradley JR, Thiru S, Pober JS. Disparate localization of 55-kD and 75-kD tumor necrosis factor receptors in human endothelial cells. Am J Pathol 1995;146:27–32.

    Hill S, Fawcett WJ, Sheldon J et al. Low-dose tumour necrosis factor alpha and melphalan in hyperthermic isolated limb perfusion. Br J Surg 1993;80:995–997.

    Bonvalot S, Laplanche A, Lejeune F et al. Limb salvage with isolated perfusion for soft tissue sarcoma: could less TNF-alpha be better? Ann Oncol 2005;16:1061–1068.

    Lienard D, Ewalenko P, Delmotte JJ et al. High-dose recombinant tumor necrosis factor alpha in combination with interferon gamma and melphalan in isolation perfusion of the limbs for melanoma and sarcoma. J Clin Oncol 1992;10:52–60.

    Eggermont AMM, Lienard D, Schraffordt Koops H et al. Treatment of irresectable soft tissue sarcomas of the limbs by isolation perfusion with high dose TNF-alpha in combination with interferon-gamma and melphalan. In: Fiers W, Buurman WA, eds. Tumor Necrosis Factor: Molecular and Cellular Biology and Clinical Relevance. Basel: Karger, 1993: 239–243.

    Eggermont AM, Schraffordt Koops H, Lienard D et al. Isolated limb perfusion with high-dose tumor necrosis factor-alpha in combination with interferon-gamma and melphalan for nonresectable extremity soft tissue sarcomas: a multicenter trial. J Clin Oncol 1996;14:2653–2665.

    Eggermont AM, Schraffordt Koops H, Klausner JM et al. Isolated limb perfusion with tumor necrosis factor and melphalan for limb salvage in 186 patients with locally advanced soft tissue extremity sarcomas. The cumulative multicenter European experience. Ann Surg 1996;224: 756–764.

    Eggermont AM, Schraffordt Koops H, Klaase JM et al. Limb salvage by isolated limb perfusion with tumor necrosis factor alpha and melphalan for locally advanced extremity soft tissue sarcomas: results of 270 perfusions in 246 patients. Proc Am Soc Clin Oncol 1999;11:497.

    Gutman M, Inbar M, Lev-Shlush D et al. High dose tumor necrosis factor-alpha and melphalan administered via isolated limb perfusion for advanced limb soft tissue sarcoma results in a >90% response rate and limb preservation. Cancer 1997;79:1129–1137.

    Olieman AF, Pras E, van Ginkel RJ et al. Feasibility and efficacy of external beam radiotherapy after hyperthermic isolated limb perfusion with TNF-alpha and melphalan for limb-saving treatment in locally advanced extremity soft-tissue sarcoma. Int J Radiat Oncol Biol Phys 1998;40: 807–814.

    Rossi CR, Foletto M, Di Filippo F et al. Soft tissue limb sarcomas: Italian clinical trials with hyperthermic antiblastic perfusion. Cancer 1999;86:1742–1749.

    Lejeune FJ, Pujol N, Lienard D et al. Limb salvage by neoadjuvant isolated perfusion with TNFalpha and melphalan for non-resectable soft tissue sarcoma of the extremities. Eur J Surg Oncol 2000;26:669–678.

    Hohenberger P, Kettelhack C, Hermann A et al. Functional outcome after preoperative isolated limb perfusion with RhTNFalpha/melphalan for high-grade extremity sarcoma. Eur J Cancer 2001;37:S34–S35.

    Noorda EM, Vrouenraets BC, Nieweg OE et al. Isolated limb perfusion with TNFalpha and melphalan for irresectable soft tissue sarcoma of the extremities. Ann Surg Oncol 2003;10:S36.

    van Etten B, van Geel AN, de Wilt JH et al. Fifty tumor necrosis factor-based isolated limb perfusions for limb salvage in patients older than 75 years with limb-threatening soft tissue sarcomas and other extremity tumors. Ann Surg Oncol 2003;10:32–37.

    Grünhagen DJ, Brunstein F, Graveland WJ et al. Isolated limb perfusion with tumor necrosis factor and melphalan prevents amputation in patients with multiple sarcomas in arm or leg. Ann Surg Oncol 2005;12:473–479.

    Lans TE, Grünhagen DJ, de Wilt JH et al. Isolated limb perfusions with tumor necrosis factor and melphalan for locally recurrent soft tissue sarcoma in previously irradiated limbs. Ann Surg Oncol 2005;12:406–411.

    Grünhagen DJ, de Wilt JH, Graveland WJ et al. The palliative value of tumor necrosis factor alpha-based isolated limb perfusion in patients with metastatic sarcoma and melanoma. Cancer 2006;106:156–162.

    Rossi CR, Mocellin S, Pilati P et al. Hyperthermic isolated perfusion with low-dose tumor necrosis factor alpha and doxorubicin for the treatment of limb-threatening soft tissue sarcomas. Ann Surg Oncol 2005;12: 398–405.

    Grünhagen DJ, Brunstein F, Verhoef C et al. Outcome and prognostic factor analysis of 217 consecutive isolated limb perfusions with TNFalpha and melphalan for limb-threatening soft tissue sarcoma. Cancer March 15, 2006 [Epub ahead of print].

    Grünhagen DJ, de Wilt JH, van Geel AN et al. TNF dose reduction in isolated limb perfusion. Eur J Surg Oncol 2005;31:1011–1019.

    Eggermont AM. Treatment of melanoma in-transit metastases confined to the limb. Cancer Surv 1996;26:335–349.

    Lejeune FJ, Lienard D, Leyvraz S et al. Regional therapy of melanoma. Eur J Cancer 1993;29A:606–612.

    Eggermont AM, Lienard D, Schraffordt Koops H et al. High dose TNFalpha in isolated perfusion of the limb: highly effective treatment for melanoma in transit metastases or unresectable sarcoma. Reg Cancer Treat 1995;7:32–36.

    Fraker DL, Alexander HR, Andrich M et al. Treatment of patients with melanoma of the extremity using hyperthermic isolated limb perfusion with melphalan, tumor necrosis factor, and interferon gamma: results of a tumor necrosis factor dose-escalation study. J Clin Oncol 1996;14:479–489.

    Lienard D, Eggermont AM, Koops HS et al. Isolated limb perfusion with tumour necrosis factor-alpha and melphalan with or without interferon-gamma for the treatment of in-transit melanoma metastases: a multicentre randomized phase II study. Melanoma Res 1999;9:491–502.

    Grünhagen DJ, Brunstein F, Graveland WJ et al. One hundred consecutive isolated limb perfusions with TNF-alpha and melphalan in melanoma patients with multiple in-transit metastases. Ann Surg 2004;240:939–947; discussion 947–948.

    Fraker DL, Alexander HR, Ross R. A phase III trial of isolated limb perfusion for extremity melanoma comparing melphalan alone versus melphalan plus TNFalpha plus IFNgamma. Ann Surg Oncol 2002;9:S8.

    Rossi CR, Foletto M, Mocellin S et al. Hyperthermic isolated limb perfusion with low-dose tumor necrosis factor-alpha and melphalan for bulky in-transit melanoma metastases. Ann Surg Oncol 2004;11:173–177.

    Bartlett DL, Ma G, Alexander HR et al. Isolated limb reperfusion with tumor necrosis factor and melphalan in patients with extremity melanoma after failure of isolated limb perfusion with chemotherapeutics. Cancer 1997;80:2084–2090.

    Grünhagen DJ, van Etten B, Brunstein F et al. Efficacy of repeat isolated limb perfusions with tumor necrosis factor alpha and melphalan for multiple in-transit metastases in patients with prior isolated limb perfusion failure. Ann Surg Oncol 2005;12:609–615.

    Olieman AF, Lienard D, Eggermont AM et al. Hyperthermic isolated limb perfusion with tumor necrosis factor alpha, interferon gamma, and melphalan for locally advanced nonmelanoma skin tumors of the extremities: a multicenter study. Arch Surg 1999;134:303–307.

    Klaase JM, Kroon BB, Benckhuijsen C et al. Results of regional isolation perfusion with cytostatics in patients with soft tissue tumors of the extremities. Cancer 1989;64:616–621.

    Feig BW, Ross MI, Cornieer J et al. A prospective evaluation of isolated limb perfusion with doxorubicin in patients with unresectable extremity sarcomas. Ann Surg Oncol 2004;11:S80.

    Nooijen PT, Manusama ER, Eggermont AM et al. Synergistic effects of TNF-alpha and melphalan in an isolated limb perfusion model of rat sarcoma: a histopathological, immunohistochemical and electron microscopical study. Br J Cancer 1996;74:1908–1915.

    Shimomura K, Manda T, Mukumoto S et al. Recombinant human tumor necrosis factor-alpha: thrombus formation is a cause of anti-tumor activity. Int J Cancer 1988;41:243–247.

    Fajardo LF, Kwan HH, Kowalski J et al. Dual role of tumor necrosis factor-alpha in angiogenesis. Am J Pathol 1992;140:539–544.

    Naredi PL, Lindner PG, Holmberg SB 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;54:645–649.

    van Etten B, de Vries MR, van Ijken MG et al. Degree of tumour vascularity correlates with drug accumulation and tumour response upon TNF-alpha-based isolated hepatic perfusion. Br J Cancer 2003;88:314–319.

    Mocellin S, Rossi CR, Pilati P et al. Tumor necrosis factor, cancer and anticancer therapy. Cytokine Growth Factor Rev 2005;16:35–53.

    Orlowski RZ, Baldwin AS Jr. NF-kappaB as a therapeutic target in cancer. Trends Mol Med 2002;8:385–389.

    Wang CY, Cusack JC Jr, Liu R et al. Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-kappaB. Nat Med 1999;5:412–417.

    Lee KY, Chang W, Qiu D et al. PG490 (triptolide) cooperates with tumor necrosis factor-alpha to induce apoptosis in tumor cells. J Biol Chem 1999;274:13451–13455.

    Richardson PG, Barlogie B, Berenson J et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med 2003;348:2609–2617.

    Ganster RW, Taylor BS, Shao L et al. Complex regulation of human inducible nitric oxide synthase gene transcription by Stat 1 and NF-kappa B. Proc Natl Acad Sci U S A 2001;98:8638–8643.

    Binder C, Schulz M, Hiddemann W et al. Induction of inducible nitric oxide synthase is an essential part of tumor necrosis factor-alpha-induced apoptosis in MCF-7 and other epithelial tumor cells. Lab Invest 1999;79:1703–1712.

    de Wilt JH, Manusama ER, van Etten B et al. Nitric oxide synthase inhibition results in synergistic anti-tumour activity with melphalan and tumour necrosis factor alpha-based isolated limb perfusions. Br J Cancer 2000;83:1176–1182.

    Mocellin S, Provenzano M, Rossi CR et al. Induction of endothelial nitric oxide synthase expression by melanoma sensitizes endothelial cells to tumor necrosis factor-driven cytotoxicity. Clin Cancer Res 2004;10:6879–6886.

    Delhalle S, Deregowski V, Benoit V et al. NF-kappaB-dependent MnSOD expression protects adenocarcinoma cells from TNF-alpha-induced apoptosis. Oncogene 2002;21:3917–3924.

    Totzke G, Schulze-Osthoff K, Janicke RU. Cyclooxygenase-2 (COX-2) inhibitors sensitize tumor cells specifically to death receptor-induced apoptosis independently of COX-2 inhibition. Oncogene 2003;22: 8021–8030.

    ten Hagen TL, Seynhaeve AL, van Tiel ST et al. Pegylated liposomal tumor necrosis factor-alpha results in reduced toxicity and synergistic antitumor activity after systemic administration in combination with liposomal doxorubicin (Doxil) in soft tissue sarcoma-bearing rats. Int J Cancer 2002;97:115–120.

    Ameloot P, Brouckaert P. Production and characterization of receptor-specific TNF muteins. Methods Mol Med 2004;98:33–46.

    Curnis F, Sacchi A, Corti A. Improving chemotherapeutic drug penetration in tumors by vascular targeting and barrier alteration. J Clin Invest 2002;110:475–482.

    Fulda S, Debatin KM. IFNgamma sensitizes for apoptosis by upregulating caspase-8 expression through the Stat1 pathway. Oncogene 2002;21:2295–2308.

    Gnant MF, Berger AC, Huang J et al. Sensitization of tumor necrosis factor alpha-resistant human melanoma by tumor-specific in vivo transfer of the gene encoding endothelial monocyte-activating polypeptide II using recombinant vaccinia virus. Cancer Res 1999;59:4668–4674.

    Berger AC, Alexander HR, Wu PC et al. Tumour necrosis factor receptor I (p55) is upregulated on endothelial cells by exposure to the tumour-derived cytokine endothelial monocyte-activating polypeptide II (EMAP-II). Cytokine 2000;12:992–1000.

    Schwarz MA, Kandel J, Brett J et al. Endothelial-monocyte activating polypeptide II, a novel antitumor cytokine that suppresses primary and metastatic tumor growth and induces apoptosis in growing endothelial cells. J Exp Med 1999;190:341–54.

    Murray JC, Symonds P, Ward W et al. Colorectal cancer cells induce lymphocyte apoptosis by an endothelial monocyte-activating polypeptide-II-dependent mechanism. J Immunol 2004;172:274–281.

    Brunstein F, Hoving S, Seynhaeve AL et al. Synergistic antitumor activity of histamine plus melphalan in isolated limb perfusion: preclinical studies. J Natl Cancer Inst 2004;96:1603–1610.

    Hoving S, Brunstein F, aan de Wiel-Ambagtsheer G et al. Synergistic anti-tumor response of interleukin 2 with melphalan in isolated limb perfusion in soft tissue sarcoma-bearing rats. Cancer Res 2005;65:4300–4308.(Remco van Horssen, Timo L)