throbber
Downloaded from
`
`http://theoncologist.alphamedpress.org/
`
` by guest on April 17, 2018
`
`This material is protected by U.S. Copyright law.
`Unauthorized reproduction is prohibited.
`For reprints contact: Reprints@AlphaMedPress.com
`
`Sarcomas
`
`TNF-α in Cancer Treatment:
`Molecular Insights, Antitumor Effects, and Clinical Utility
`
`Remco van Horssen, Timo L. M. ten Hagen, Alexander M. M. Eggermont
`
`Department of Surgical Oncology, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam, The Netherlands
`
`Key Words. Cancer • TNF-α • TNFR-1 • Tumor vasculature • Isolated limb perfusion
`
`Learning Objectives
`After completing this course, the reader will be able to:
` 1. Discuss the role of TNF-a in cancer survival and apoptosis.
` 2. Describe the mechanism of chemotherapy potentiation by TNF-a.
` 3. Explain the selective targeting of tumor vasculature by TNF-a.
` 4. Discuss TNFR-1 and TNFR-2 signaling.
`
`CMECME
`
`Access and take the CME test online and receive 1 AMA PRA category 1 credit at CME.TheOncologist.com
`
`Abstract
`Tumor necrosis factor alpha (TNF-α), 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 prop-
`erties, 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 sarco-
`mas 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-α acts synergistically with cytostatic
`drugs. The interaction of TNF-α with TNF receptor 1
`and receptor 2 (TNFR-1, TNFR-2) activates several
`signal transduction pathways, leading to the diverse
`functions of TNF-α. 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-α action during tumor treatment.
`Besides extravasation of erythrocytes and lymphocytes,
`leading to hemorrhagic necrosis, TNF-α targets the
`tumor-associated vasculature (TAV) by inducing hyper-
`permeability 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-α from the molecule to the clinic, we pro-
`vide an overview of the use of TNF-α in cancer starting
`with molecular insights into TNFR-1 signaling and cel-
`lular mechanisms of the antitumor activities of TNF-α
`and ending with clinical response. In addition, possible
`factors modulating TNF-α actions are discussed. The
`Oncologist 2006;11:397–408
`
`Introduction
`cytokine involved in apoptosis, cell survival, inflammation,
`Tumor necrosis factor alpha (TNF-α) is a multifunctional
`and immunity acting via two receptors [1, 2]. Currently it
`Correspondence: Alexander M. M. Eggermont, M.D., Ph.D., Erasmus MC–Daniel den Hoed Cancer Center, Department of Surgical
`Oncology, 301 Groene Hilledijk, 3075 EA Rotterdam, The Netherlands. Telephone: 31-0-10-439-1911; Fax: 31-0-10-439-1011; e-mail:
`a.m.m.eggermont@erasmusmc.nl Received August 26, 2005; accepted for publication February 10, 2006. ©AlphaMed Press 1083-
`7159/2006/$20.00/0
`The Oncologist 2006;11:397–408 www.TheOncologist.com
`
`IPR2018-00685
`Celgene Ex. 2028, Page 1
`
`

`

`Downloaded from
`
`http://theoncologist.alphamedpress.org/
`
` by guest on April 17, 2018
`
`398
`
`TNF-α in Cancer Treatment
`
`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 in-
`transit metastases confined to the limb [3]. TNF-α was iso-
`lated 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 iso-
`lation, TNF-α was also characterized as “cachectin” and
`as T-lymphocyte differentiation factor [7, 8]. In 1984, the
`human TNF-α gene was cloned [9, 10], and a range of clini-
`cal 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-α and TNF Receptor 1 Signaling
`TNF-α 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-α–binding receptors. TNF-α
`is mainly produced by activated macrophages, T lympho-
`cytes, 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-α 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-α acts via two distinct recep-
`tors [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-α.
`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 recep-
`tor 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-α, 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 signal-
`ing 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 inhib-
`its apoptosis via cytoplasmic inhibitor of apoptosis protein
`(cIAP). The binding of TRAF-2 initiates a pathway of phos-
`phorylation 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-κB) transcription
`factor via NF-κB–inducing kinase (NIK) and the inhibitor
`of κB kinase (IKK) complex [24]. Both the NF-κB and cFos/
`cJun transcription factors induce transcription of antiapop-
`totic, proliferative, immunomodulatory, and inflammatory
`genes. NF-κB is the major survival factor in preventing TNF-
`α–induced apoptosis, and inhibition of this transcription fac-
`tor may improve the efficacy of apoptosis-inducing cancer
`therapies [25]. NF-κB activation in many human malignan-
`cies 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-α Effects During Solid
`Tumor Treatment
`It is widely known that TNF-α induces hemorrhagic necro-
`sis in a certain set of tumor types. To investigate the underly-
`ing mechanisms of TNF-α 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-α and chemotherapeutic drugs [28]. In
`addition, we showed a synergistic antitumor effect of the
`combination treatment with TNF-α and chemotherapeutic
`drugs [29]. In contrast, TNF-α alone induced only a mild
`central necrosis, and there was no objective tumor response
`observed. The same rat models also revealed that the addi-
`tion of TNF-α improved the accumulation of chemothera-
`peutic 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 concentra-
`TheOncologist®
`
`IPR2018-00685
`Celgene Ex. 2028, Page 2
`
`

`

`van Horssen, ten Hagen, Eggermont
`
`
`
`
`
`
`
`399
`
`Downloaded from
`
`http://theoncologist.alphamedpress.org/
`
` by guest on April 17, 2018
`
`Figure 1. Tumor necrosis factor receptor 1 (TNFR-1) signaling pathway. Tumor necrosis factor alpha (TNF-α) activates both
`survival and proliferation pathways along with apoptotic pathways via TNFR-1. The separate pathways are well defined, while the
`survival-death balance regulation remains unclear. Abbreviations: APAF-1, apoptosis protein activating factor 1; Bcl-2, B-cell
`lymphoma 2; Bid, Bak, Bax, and Bcl-XL, mitochondrial proteins of the Bcl-2 family; CAD, caspase-activated DNAse; Caspase-
`3/8/9, cysteine aspartase (apoptotic protease) 3/8/9; Cdc37, co-chaperon of HSP90; cIAP, cytoplasmic inhibitor of apoptosis; cFos/
`cJun, transcription factors; DD, death domain; EndoG, mitochondrial DNAse; FADD, Fas-associated DD; HSP90, heat shock
`protein 90; I-CAD, inhibitor of CAD; IκB, inhibitor of NF- κB; IKKα/β, IκB kinase; JNK, cJun n-terminal kinase; MEKK1, mito-
`gen-activated protein kinase/extracellular signal–related kinase kinase 1; MKK3/7, MAPK kinase 3/7; NEMO, NF-κB essential
`modulator; NF-κB, nuclear factor kappa B transcription factor; NIK, NF-κB inducing kinase; p38MAPK, p38 mitogen-activated
`protein kinase; RIP, receptor interacting protein; SODD, silencer of DD; sTNFR-1, soluble TNFR-1; TNF-α, tumor necrosis factor
`alpha; TNFR-1, TNF receptor 1; TRADD, TNF receptor-associated DD; TRAF-2, TNF receptor-associated factor-2.
`
`tions 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 intratu-
`moral 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-α
`in vitro, which is in accordance with other reports describing
`a lack of effect of TNF-α 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-α aug-
`ments the antitumor activity of pegylated liposomal doxoru-
`bicin [34, 35]. These observations are comprehensible clues
`
`that mechanisms underlying the TNF-α effect during solid
`tumor treatment cannot be caused by a direct cytotoxic or
`cytostatic effect of TNF-α toward the tumor cells. It was sug-
`gested that, rather than tumor cells themselves, cells of the
`tumor stroma may be responsible for the observed antitumor
`effect of TNF-α in patients. This hypothesis was confirmed
`by data from mice experiments revealing that TNF-α 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 pro-
`
`www.TheOncologist.com
`
`IPR2018-00685
`Celgene Ex. 2028, Page 3
`
`

`

`Downloaded from
`
`http://theoncologist.alphamedpress.org/
`
` by guest on April 17, 2018
`
`400
`
`TNF-α in Cancer Treatment
`
`vide 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 guid-
`ing the following so-called stalk endothelial cells (stage 3)
`[41]. Finally, the growing tumor is connected to the vascu-
`lature (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 vascula-
`ture (TAV) that is rather chaotic, both in structure and func-
`tion. 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 tar-
`get. The solid tumors treated by ILP with TNF-α have a mas-
`sive vascular structure consisting of vessels with a pheno-
`type specific to tumor vessels, although detailed study needs
`to clarify the exact contribution of the TAV to the observed
`antitumor responses.
`
`Activity of TNF-α 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 400 nm, depending on the tumor type [46]. Blood cells such
`as lymphocytes and monocytes easily adhere and extrava-
`sate 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
`
`Figure 2. The sequential steps during tumor angiogenesis. The dormant tumor in stage 1 starts to secrete angiogenic growth factors
`(GF) after its “angiogenic switch”, which is accomplished by an imbalance in pro- and antiangiogenic factors. These GFs activate
`endothelial cells of surrounding vessels, and these cells start to migrate (stage 2) and proliferate toward the tumor. An endothelial tip
`cell (TC) is guiding this sprouting process (stage 3). In stage 4, the novel sprout has formed a lumen and the tumor is connected to the
`vasculature, thereby ensuring its growth and enabling it to metastasize hematogenically. Abbreviation: ECM, extracellular matrix.
`
`TheOncologist®
`
`IPR2018-00685
`Celgene Ex. 2028, Page 4
`
`

`

`van Horssen, ten Hagen, Eggermont
`
`
`
`
`
`
`
`401
`
`Downloaded from
`
`http://theoncologist.alphamedpress.org/
`
` by guest on April 17, 2018
`
`Figure 3. Differences between healthy and tumor endothelium and a proposed mechanism of the tumor necrosis factor alpha
`(TNF-α) effect. (A): Schematic representation of a vessel with healthy endothelial lining (upper) and tumor endothelial lining
`(lower). Healthy endothelium is a continuous lining of endothelial cells, covered with pericytes and with a thin basement mem-
`brane. The permeability is low, and extravasation is tightly organized. In contrast, tumor endothelium does not consist of a con-
`tinuous lining of endothelial cells; it lacks pericyte coverage, and the basement membrane is thickened. This phenotype results
`in greater permeability of the tumor vessel (small arrows). We hypothesize that tumor endothelium exhibits a higher TNFR-1
`expression level because of TNFR-1–upregulating factors produced by vessel-surrounding cells. (B): Upon TNF-α treatment
`with isolated limb perfusion, the healthy endothelium stays intact because it is TNF-α insensitive by its lack of TNFR-1 expres-
`sion on the membrane. Tumor endothelium binds TNF-α, which affects the endothelial phenotype and induces apoptosis in some
`endothelial cells. These two processes result in an enormous induction of vessel permeability (big arrows). As a result, the chemo-
`therapy drug is well distributed throughout the tumor, and a strong extravasation of erythrocytes results in massive hemorrhagic
`tumor necrosis. The selectively targeted tumor vessels are no longer functional and regress.
`
`endothelial lining, defines the tumor vessels as a specific tar-
`get for TNF-α treatment (Fig. 3B). When TNF-α is adminis-
`tered 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-α; however, because of a lower num-
`ber 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-α to TNFR-1 binding results in hyper-
`permeability of the tumor vessels, and erythrocytes and other
`blood cells extravasate. The strong extravasation of erythro-
`cytes results in massive hemorrhagic necrosis of the tumor.
`As a result of the direct cytotoxicity of high-dose TNF-α to
`endothelial cells, some of these cells undergo apoptosis, and
`this process strongly enhances the induced hyperpermeabil-
`
`ity (Fig. 3B). Several studies have shown that a lower dose of
`TNF-α 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-α and chemotherapeutic drugs is a consequence of this
`double-induced hyperpermeability. This hyperpermeability
`throughout the tumor facilitates the augmented accumula-
`tion and distribution of the drug in the tumor, resulting in bet-
`ter exposure of the tumor cells to the cytostatic agent [30].
`This double-induced hyperpermeability, along with the dual
`targeting—the TAV (by TNF-α) and the tumor cells (by the
`chemotherapy drug)—is one explanation for the observed
`synergistic response of tumors to TNF-α and chemotherapy
`that results in high response rates in patients.
`
`www.TheOncologist.com
`
`IPR2018-00685
`Celgene Ex. 2028, Page 5
`
`

`

`Downloaded from
`
`http://theoncologist.alphamedpress.org/
`
` by guest on April 17, 2018
`
`402
`
`TNF-α in Cancer Treatment
`
`Clinical Efficacy of TNF-α–Based Isolated
`Limb Perfusion
`The use of TNF-α in the ILP setting was pioneered by Lien-
`ard 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-α 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
`
`Table 1. Principle studies on tumor necrosis factor (TNF)–based isolated limb perfusion (ILP) for irresectable soft tissue sarcomas
`ILP Studies
`Pts
`CR
`PR
`NC/PD
`Limb Salvage
`(n)
`Drugs
`(%)
`(%)
`(%)
`(%)
`Pivotal Multicenter Studies
`TNF + IFN + M
`TNF + IFN + M
`
`20
`59
`
`TNF ± IFN + M
`
`TNF ± IFN + M
`
`195
`
`270
`196
`
`55 a
`18 a
`36 b
`18 a
`29 b
`28 c
`17 c
`
`40 a
`64 a
`51 b
`57 a
`53 b
`48 c
`48 b
`
`5 a
`18 a
`13 b
`25 a
`18 b
`24 c
`35 b
`
`Reference
`
`90
`84
`
`Eggermont et al. [51]
`Eggermont et al. [52]
`
`82
`
`76
`71 d
`
`Eggermont et al. [53]
`
`Eggermont et al. [54]
`
`Single-center studies (>20 pts)
`TNF ± IFN + M
`TNF ± IFN + M
`TNF + Dox
`TNF ± IFN + M
`TNF ± IFN + M
`TNF ± IFN + M
`TNF ± IFN + M j
`TNF ± IFN + M i
`TNF ± IFN + M k
`TNF ± IFN + M g
`TNF + Dox g
`
`TNF ± IFN + M
`
`TNF + M g
`
`35
`34
`20
`22
`55
`49
`29
`64
`29
`37
`21
`
`217
`
`100
`
`85
`85
`85
`77
`84
`58
`76
`82
`65
`97
`71
`
`87
`
`87
`
`82
`
`Gutman et al. [55]
`Olieman et al. [56]
`Rossi et al. [57]
`Lejeune et al. [58]
`Hohenberger et al. [59]
`Noorda et al. [60]
`van Etten et al. [61]
`Grünhagen et al. [62]
`Lans et al. [63]
`Grünhagen et al. [64]
`Rossi et al. [65]
`
`Grünhagen et al. [66]
`
`Bonvalot et al. [49]
`
`Grünhagen et al. [67]
`
`37 b
`9 b
`54 b
`35 b
`6 b
`59 b
`26 e
`10 e
`64 e
`18 b
`18 b
`64 b
`-
`-
`-
`8 b
`37 b
`55 b
`18 b
`38 b
`38 b
`13 b
`45 b
`42 b
`30 b
`50 b
`20 b
`16 b
`68 b
`16 b
`38 a
`57 a
`5 a
`55 b
`10 b
`35 b
`31 a
`51 a
`18 a
`26 b
`25 b
`49 b
`34 f
`17 f
`49 f
`35 f
`43 f
`22 f
`26 b
`50 b
`24 b
`240
`TNF ± IFN + M g
`a Objective clinical response rate by World Health Organization criteria.
`b CR, clinical CR or 100% necrosis; PR, clinical PR or >50%–90% necrosis.
`c CR only recognized by the European Medicines Agency when histopathology showed 100% necrosis.
`d Independent committee recognized 196 patients as pure amputation candidates.
`e No clinical response data; CR >90%; PR, radiological and/or hisopathological >50% necrosis.
`f CR/PR, loss of vasculature on ultrasound MRI; lower panel CR, >90% necrosis on histopathology.
`g Low-dose TNF of 1 mg or various doses of TNF-α (0.5–4 mg); Grünhagen et al. (1–4 mg), soft tissue sarcomas patients.
`h Patients with metastatic disease.
`i Patients with multiple tumors in extremity.
`j Patients >75 years old.
`k Patients with recurrent sarcomas in 60–70 Gray irradiated fields.
`Abbreviations: CR, complete remission; IFN, interferon gamma; M, melphalan; NC, no change; PD, progressive disease; PR,
`partial remission; Pts, patients.
`
`TheOncologist®
`
`IPR2018-00685
`Celgene Ex. 2028, Page 6
`
`

`

`Downloaded from
`
`http://theoncologist.alphamedpress.org/
`
` by guest on April 17, 2018
`
`van Horssen, ten Hagen, Eggermont
`
`
`
`
`
`
`
`403
`
`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-α–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-α 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-α plus melphalan (TM-
`ILP) was especially effective against bulky tumors such as
`STSs, in which ILP with melphalan alone [79] or doxoru-
`bicin 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-α–based ILP with mel-
`phalan 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], identi-
`cal to the CR rate that was seen in an interim analysis of
`a randomized trial by Fraker et al. [74], in which TNF-α–
`
`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-α–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-α–based ILP for nonmel-
`anoma locally advanced skin cancers [78]. Because TNF-α
`acts primarily on the tumor vasculature, these observations
`make sense, and the propensity to respond to a TNF-α–
`based ILP is assumed to depend more on tumor vasculature
`than on the histologic type of the tumor.
`Response of STS to TNF-α–based ILP is shown in Fig-
`ure 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 gadolin-
`ium-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 regres-
`sion is visible. All lesions were resected and found to be
`100% necrotic. Thus the response was classified as a histo-
`pathologic CR.
`Targeting by TNF-α of the tumor vasculature is
`revealed in patients by angiographies before and after ILP.
`The TAV is selectively destroyed by TNF-α–based ILP;
`the TAV is gone while normal vessels of the limbs are still
`intact after ILP (Fig. 4B). TNF-α targets the vasculature
`
`Table 2. Tumor necrosis factor (TNF)–based isolated limb perfusion (ILP) in melanoma and nonmelanoma skin cancer patients.
`TTLP
`Median (Mo) Reference
`8+
`Lienard et al. [50]
`18+
`Lejeune et al. [69]
`26
`Eggermont et al. [70]
`ns
`Fraker et al. [71]
`14
`Lienard et al. [72]
`14
`Lienard et al. [72]
`16+
`Grünhagen et al. [73]
`ns
`Fraker et al. [74]
`ns
`Rossi et al. [75]
`8
`Grünhagen et al. [73]
`6
`Bartlett et al. [76]
`14
`Grünhagen et al. [77]
`20+
`Olieman et al. [78]
`
`CR (%)
`89
`90
`88
`76
`78
`69
`69
`59
`70
`58
`65
`75
`60
`
`PR (%)
`11
`10
`12
`16
`22
`22
`26
`16
`25
`34
`29
`25
`27
`
`Overall RR
`100 a
`100 a
`100 a
`92 a
`100 a
`91
`95
`75
`95
`83
`94
`100
`87
`
`Nonmelanoma skin cancers
`a TNF + IFN
`Abbreviations: CR, complete remission; Overall RR, overall (CR + PR) response rate; PR, partial remission rate; Pts, patients;
`TTLP, time to local progression; ns, not specified.
`
`ILP
`All melanoma patients
`
`Bulky melanoma only
`
`Repeat ILP for melanoma
`
`Pts (n)
`19
`44
`58
`26
`32
`32
`100
`39
`20
`50
`17
`26
`15
`
`www.TheOncologist.com
`
`IPR2018-00685
`Celgene Ex. 2028, Page 7
`
`

`

`404
`
`TNF-α in Cancer Treatment
`
`Downloaded from
`
`http://theoncologist.alphamedpress.org/
`
` by guest on April 17, 2018
`
`vascular TNF-α effects are achieved by the high concen-
`tration reached during ILP. At these high levels, TNF-α
`activity is antivascular and antiangiogenic, while at lower
`concentration TNF-α is known to promote angiogenesis
`[83]. In addition to direct TAV-mediated effects, TNF-α
`reduces blood flow in tumors in a dose-dependent fashion
`[84]. This set of antivascular TNF-α effects was recently
`confirmed by experiments revealing that tumor response
`to TNF-α correlates with the degree of tumor vascularity
`[85]. Along with this dual role in angiogenesis, TNF-α is
`also known for its dual role in cancer treatment, anti-TNF-
`α therapy is also used for several types of cancer. The anti-
`neoplastic and tumor-promoting effects of TNF-α are dis-
`cussed in a recent review [86].
`
`Approaches to Modulate TNF-α Action in
`Cancer Treatment
`High response rates in the ILP setting do not avoid the need
`to search for factors that modulate the TNF-α effect in solid
`tumors. In addition to the possible application of TNF-α in
`other settings (e.g., systemic treatment) and for other tumors
`types, nonresponding patients in the ILP setting may also
`benefit from TNF-α sensitizers. Some of these approaches
`are mentioned below. An obvious target is inhibition of the
`NF-κB survival pathway. Inactivation of NF-κB is known
`to sensitize several tumors to TNF-α [87]. NF-κB can be
`blocked in several ways: overexpression of its inhibitor IκB
`and selective NF-κB inhibitors have been shown to increase
`TNF-α–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-α–
`treated cells through NF-κB–induced expression of induc-
`ible 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-α–based
`ILP resulted in an increased tumor response in rats bear-
`ing STSs [93]. These observations were confirmed by a
`recent study showing that NOS inhibition in endothelial
`cells reduces their sensitivity to TNF-α in vitro, leading to
`the hypothesis that tumor vessels exhibit a higher level of
`NOS, which might explain their higher TNF-α sensitivity
`[94]. These studies justify further evaluation of NOS inhi-
`bition in tumors of patients treated by ILP to stimulate the
`anti-TAV activities of TNF-α.
`Apoptosis induced by TNF-α is also associated with
`the generation of reactive oxygen species (ROS). It has
`been shown that the key survival factor NF-κB induces
`ROS-neutralizing enzymes like superoxide dismutase
`[95]. Induction of ROS production or an inhibition of the
`TheOncologist®
`
`Figure 4. Antitumor and antivascular effects of tumor necro-
`sis factor alpha (TNF-α) upon isolated limb perfusion (ILP)
`treatment of sarcoma and melanoma patients. (A): Magnetic
`resonance imaging of a patient with a high-grade leiomyosar-
`coma in the upper leg showing the tumor mass before treatment
`on the left. Five weeks after ILP with TNF-α and melphalan,
`there is no gadolinium uptake in the tumor remnants (right).
`Tumor remnants were resected, and all were necrotic. (B):
`Angiographies of two patients with rapidly growing sarcomas
`in the leg before (left) and after (right) ILP with TNF-α and
`melphalan. Angiographies clearly show the well-developed
`tumor vasculature before ILP, which is selectively destroyed
`after treatment while the normal vessels are still present and
`intact. Both patients were classified as complete responders.
`(C): Endothelial lining of tumor vessels is destroyed. Endo-
`thelial staining (CD31) of vessels in normal skin (left) and
`melanoma (right) in biopsies of a melanoma patient taken after
`ILP. Vessels in the normal skin show a continuous endothelial
`lining (arrow), while in the melanoma-associated vessel, this
`lining is disrupted and the endothelial cells detach from the
`basement membrane (arrowheads). The elastic fibers (stars,
`stained aspecific) in the thickened basement membrane are
`also visible in the melanoma vessel. Scale bar, 50 μm.
`
`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 sub-
`types, 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-α–induced coagulation and extravasation of
`erythrocytes [82]. In accordance with the angiographies
`of STS, the vascular lining of m

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket