throbber
Tumor necrosis factor-a in the pathogenesis and treatment
`of cancer
`G Mark Anderson , Marian T Nakada and Mark DeWitte
`
`The critical pathogenic role of tumor necrosis factor (TNF)a
`in inflammatory disorders such as rheumatoid arthritis and
`inflammatory bowel disease is well established. The role
`played by TNFa in both the treatment and pathogenesis of
`cancer remains less understood. Recent advances help to
`create a framework for understanding seemingly paradoxical
`effects of TNFa as both an anti-tumour agent and a mediator
`of tumour growth. High pharmacological doses of TNFa
`combined with chemotherapy can regress otherwise
`intractable tumours, and efforts continue to optimize delivery
`to avoid severe toxicities. Mounting evidence demonstrates
`that pathophysiological concentrations of endogenous TNFa
`act to promote tumour genesis and growth. The cellular and
`molecular pathways mediating these phenomena are starting
`to be clarified. Current data support the continued
`development of both TNFa and anti-TNFa therapy for
`clinical treatment of cancers in distinct settings.
`
`Addresses
`Centocor, 200 Great Valley Parkway, Malvern, PA 19355, USA
` e-mail: MAnders4@cntus.jnj.com
`
`Current Opinion in Pharmacology 2004, 4:314–320
`
`This review comes from a themed issue on
`Cancer
`Edited by Aristides Eliopoulos and Lawrence Young
`
`Available online 17th June 2004
`
`1471-4892/$ – see front matter
`ß 2004 Elsevier Ltd. All rights reserved.
`
`DOI 10.1016/j.coph.2004.04.004
`
`Abbreviations
`CLL
`chronic lymphocytic leukemia
`GVHD graft versus host disease
`IL
`interleukin
`MDS
`myelodysplastic syndrome
`MMP
`matrix metalloproteinase
`NF-jB nuclear factor-kB
`TNF
`tumor necrosis factor
`TNFR
`TNF receptor
`
`has previously been reviewed [1,2,3]. TNFa mediates
`tumour regression, and recombinant TNFa is approved in
`Europe to be administered locoregionally at supra-
`physiological levels as a therapy for sarcoma [4]. The
`concept that the major effects of endogenous TNFa in
`cancer are opposite to the effects observed with high dose
`TNFa therapy has gained momentum recently [5].
`Instead of causing tumour regression, cancer-derived
`TNFa can mediate tumour progression by causing the
`proliferation, invasion and metastasis of tumour cells (see
`also Update). The paradox that this cytokine is both a
`‘tumour necrosis factor’ and a ‘tumour-promoting factor’
`is lucidly explored by Balkwill [6]. This paradox can be
`explained partly by the differences in levels of TNFa in
`distinct settings. When TNFa is administered therapeu-
`tically in extremely high doses, it acts as a vasculotoxic
`tumour-regressing agent. However, when TNFa is pro-
`duced by tumours and tumour-associated macrophages or
`stromal cells at physiological levels, it promotes tumour
`growth and additional macrophage recruitment, stimulat-
`ing the elaboration of angiogenic and growth factors from
`infiltrating cells.
`
`TNFa-mediated cellular signaling
`The signaling pathways that determine whether a cell
`will respond to endogenous TNF by proliferating or by
`undergoing apoptosis continue to be elucidated. Micheau
`and Tschopp [7] reported a new model with evidence
`for the production of two distinct intracellular complexes
`after TNFa binds to TNFR1 in HT1080 fibrosarcoma
`cells. The first complex promotes cell survival by induc-
`tion of nuclear factor-kB (NF-kB). The second complex,
`formed substantially later after receptor ligation, is cyto-
`plasmic and spatially removed from the initial receptor
`complex. The second complex is pro-apoptotic but can be
`inhibited by anti-apoptotic gene products previously
`induced via the NF-kB pathway. This system represents
`a switch that can determine the fate of a given cell
`receiving a TNFa signal. That fate clearly will vary
`depending upon the presence or absence of key signaling
`factors resulting from cell-specific gene expression or the
`mutational status of the cell.
`
`Introduction
`Tumor necrosis factor (TNF)a is a potent pleiotropic
`proinflammatory cytokine produced by macrophages,
`neutrophils, fibroblasts, keratinocytes, NK cells, T and
`B cells, and tumour cells. TNFa mediates host responses
`in acute and chronic inflammatory conditions, and is a
`mediator of protection from infection and malignancy.
`The biology of the TNF/TNF receptor (TNFR) system
`
`TNFa as a therapy for cancer
`Systemically administered TNF-a was evaluated in the
`clinic in the 1980s as a therapy for solid tumours, and was
`found to have severe toxicities, most notably hypotension
`and organ failure [8]. Studies revealed that the maximally
`tolerated dose of TNFa was significantly lower than that
`required to cause anti-tumour effects. The delivery of
`locally high levels of TNFa, without systemic exposure,
`
`Current Opinion in Pharmacology 2004, 4:314–320
`
`www.sciencedirect.com
`
`IPR2018-00685
`Celgene Ex. 2029, Page 1
`
`

`

`TNF-a in the pathogenesis and treatment of cancer Anderson, Nakada and DeWitte 315
`
`is now accomplished by isolated limb perfusion in which
`the circulation of the limb is connected surgically to a
`bypass circuit into which drugs are administered [9].
`Isolated limb perfusion with TNFa in combination with
`chemotherapy is approved in Europe for the treatment of
`locally advanced unresectable soft-tissue sarcomas (for a
`summary of the clinical studies of isolated limb perfusion
`with TNFa, see [9]). Positive clinical data have been
`reported with this TNFa therapy for in-transit melanoma
`and drug-resistant bony sarcomas [9]. Although effective,
`this procedure is highly specialized and not widely used.
`The mechanism of action is believed to involve direct
`toxic effects on the tumour, without affecting normal
`vasculature. TNFa causes vessel regression, as well as
`hemorrhagic necrosis, because it induces endothelial cell
`apoptosis,
`resulting in a procoagulant endothelium.
`TNFa enhances vascular permeability, facilitating the
`uptake and accumulation of chemotherapeutic drugs
`such as melphalan and doxorubicin [9], as well as anti-
`bodies [10]. TNFa alone is not effective when adminis-
`tered via isolated limb perfusion,
`suggesting that
`chemotherapy accumulation is a major mechanism of
`action. The specific effect of TNFa on angiogenic,
`but not normal, vasculature might be attributable to
`the deactivation of the angiogenesis-associated integrin
`avb3. TNFa in combination with interferon-g leads to
`endothelial apoptosis [11].
`
`Many approaches are being used to target therapeutic
`TNFa directly to tumours, without causing systemic
`toxicity. These include gene therapy (TNFerade) [12]
`and TNFa conjugated to targeting peptides or single
`chain antibody fragments (see also Update) [13–15].
`
`Role of TNFa in the pathogenesis of cancer
`In contrast to the use of pharmacologic doses of TNFa as
`an anti-cancer therapeutic, mounting evidence suggests
`that TNFa acts to promote tumour growth and progres-
`sion at physiologically relevant concentrations [16]. Some
`of the most detailed work regarding the role of TNFa as a
`tumour promoter was published in a series of papers from
`the laboratory of Fran Balkwill [16,17,18]. The authors
`used the induction of skin tumours in mice as a model
`system to understand the growth-promoting actions of
`TNFa and its
`signaling pathway.
`In this
`system,
`9,10-dimethyl-1,2-benzanthracene (DMBA) is used as a
`topical DNA-damaging tumour inducer; 12-O-tetrade-
`canoylphorbol-13-acetate (TPA) is then applied as a
`tumour promoter. TNFa was first implicated in tumour
`growth in this model by the observation that TNFa
`knockout mice are highly resistant to the generation of
`skin tumours by this method. The presence of functional
`TNFa has no effect on DNA mutation rates or tumour
`initiation, but
`instead profoundly influences tumour
`promotion. TPA was shown to induce TNFa in skin
`keratinocytes of wild-type mice. The production of
`gene products induced by the activating protein-1 tran-
`
`such as granulocyte-macrophage
`scription pathway,
`factor, matrix metalloproteinase
`colony-stimulating
`(MMP)-3 and MMP-9, is important for the tumour-pro-
`moting effect. Anti-tumour effects were also observed
`following pharmacologic intervention with a neutralizing
`anti-mouse TNFa antibody. The potential role of TNFa
`in the growth of human skin cancers, initiated largely by
`UV radiation, is an important area for future investigation.
`
`Clinical observations supporting a role for
`TNFa in tumor growth
`TNFa plasma levels in cancer patients
`Several reports have associated detection of abnormally
`high levels of TNFa protein in the blood of cancer
`patients with a wide range of tumour types [19], including
`pancreatic [20], kidney [21], breast [22], asbestosis
`induced lung [23] and prostate cancer [24]. Within groups
`of patients with the same tumour type, higher levels of
`TNFa have been correlated with advanced tumour
`stage, greater paraneoplastic complications and shorter
`survival time. However, circulating TNFa is not always
`detectable in cancer patients and can vary within indi-
`vidual patients over time and course of disease [25].
`Regulation of TNFRs is critical to tumour cell respon-
`siveness to TNFa, and tumour tissue levels of TNFa
`might be more relevant than blood levels in explaining
`pro-tumourigenic associations.
`
`Endometriosis and ovarian cancer develop along a con-
`tinuum of malignant
`transformation and promotion.
`Serum TNFa has been associated significantly with
`endometriomas and malignant, cystic and ovarian cancers,
`but not with benign tumours [26]. Inflammatory condi-
`tions associated with endometriosis and ovarian cancer
`include exposure to exogenous irritants and ovulation,
`accompanied by cell proliferation, oxidative stress, vas-
`cular permeability, and overproduction of prostaglandins,
`leukotrienes, TNFa, interleukin (IL)-6 and IL-1 [27].
`mRNA for these cytokines is found in epithelial ovarian
`tumours and in related ascites. TNFa is prevalent in
`peritoneal fluid around endometrial foci. Attracted plate-
`lets and macrophages secrete vascular endothelial growth
`factor, MMP-9 and transforming growth factor-b, and
`promote infiltration of ectopic endometrium and/or inva-
`sion and metastasis. The invasive tissue of endometriosis
`is surrounded by an ineffective immune response, just as
`ovarian tumours are surrounded by inflammatory cells
`impotent against the neoplasia. Chronic inflammation
`might
`invoke a switch from Th1-dominant
`to a
`Th2-dominant microenvironment linked to mutagenesis.
`Progression can be modified by immunosuppression, sug-
`gesting a therapeutic role for anti-inflammatory agents.
`
`in the
`Esophageal metaplasia (Barrett’s oesophagus),
`setting of chronic inflammation can progress to adeno-
`carcinoma (Barrett’s adenocarcinoma). Tselepsis et al.
`[28] showed that epithelial TNFa expression was
`
`www.sciencedirect.com
`
`Current Opinion in Pharmacology 2004, 4:314–320
`
`IPR2018-00685
`Celgene Ex. 2029, Page 2
`
`

`

`316 Cancer
`
`absent in normal gastric and esophageal squamous muco-
`sae, but increased in sections from metaplasia through
`dysplasia to carcinoma. Elevated TNFa immunoreactiv-
`ity in non-dysplastic Barrett’s oesophagus is localized to
`mucosal regions of prominent lymphoid infiltrate where
`metaplastic stem cells are located. Within individual
`glands, the most dysplastic cells express TNFa, and
`TNFR1 levels are increased during disease progression,
`potentially amplifying the TNFa signal. This study
`demonstrated a novel signaling pathway for oncogene
`activation by an inflammatory cytokine.
`
`In chronic lymphocytic leukemia (CLL), neoplastic lym-
`phocytes release TNFa spontaneously in vitro, and leu-
`kaemic lymphocytes are more proliferative and viable
`when exposed to TNFa. TNFa in CLL patients corre-
`lated with extent of disease, serum b2-microglobulin, low
`hemoglobin and low platelets, as well as with karyotype.
`TNFa levels were predictive of survival, independent of
`staging, b2-microglobulin, hemoglobin, and white blood
`cell and platelet counts [29].
`
`Anti-TNFa therapy in cancer
`Tsimberidou and Giles [30] review potential clinical
`indications for agents that block or inactivate TNFa
`(Figure 1), specifically the soluble TNFa receptor fusion
`protein etanercept and the anti-TNFa antibody inflix-
`imab, in multiple myeloma, myelodysplastic syndrome
`
`Figure 1
`
`Multiple anti-TNF agents are currently marketed or in clinical
`development. Several anti-TNF biologics are illustrated above,
`including antibodies (infliximab and adalimumab), the modified
`antibody fragment CDP870, and the soluble receptor etanercept.
`The various domains present in the structures are illustrated for
`each agent. Reproduced with permission from [63].
`
`(MDS), acute myelogenous leukemia and myelofibrosis
`[30]. The limited efficacy of monotherapy points to
`the need to optimize dose and schedule, to combine
`anti-TNFa agents with active biological or cytotoxic
`agents, and to understand individual patient proteomic
`patterns of disease and gene polymorphisms that could
`predict response.
`
`Investigative and clinical uses of TNFa blockade in graft
`versus host disease (GVHD) are founded on the estab-
`lished role of upregulated TNFa as a consequence of
`conditioning regimens and as an effector molecule in
`multi-organ reactivity to allogeneic graft cells [31,32].
`Infliximab has shown activity in refractory acute and
`chronic GVHD [33]. With added immunosuppression
`and risk of serious infections, vigilance is required when
`using anti-TNF agents in patients with GVHD [34]. No
`studies yet have elucidated the kinetics, optimal dose and
`schedule of anti-TNF agents in GVHD [35].
`
`Anti-TNFa agents can inhibit the excessive apoptosis
`in haematopoietic cells, which is suspected as the cause
`of cytopenias in MDS. The hyperproliferative marrow of
`MDS, with excessive apoptosis and overexpression of
`TNFa, compels intervention to modulate the dysregu-
`lated cytokine milieu in haematopoietic tissues. Thera-
`peutic
`activity
`of
`infliximab suggests
`that
`the
`microenvironment of the marrow is more directly affected
`than are the dysplastic haematopoietic cells [36,37]. A
`logical next step would be to use a specific anti-TNF
`agent to modulate cell–cell signaling and stromal inter-
`actions in the marrow, combined with a cytotoxic agent
`known to be active against the myelodysplastic clone.
`Etanercept was tested in a Phase I clinical study in
`combination with IL-2 to determine if it could modulate
`the biological effects, and reduce toxicity, of high-dose
`IL-2 administration. TNFa bioactivity was inhibited, and
`the polymorphonuclear leukocyte chemotactic defect
`normally seen with IL-2 was not observed. Increases in
`levels of C-reactive protein, IL-6, IL-8 and IL-1 receptor
`antagonist were partially suppressed relative to historical
`controls [38].
`
`A pilot study of etanercept in patients with refractory
`multiple myeloma reported tolerability but no objective
`responses, and some evidence of acceleration of disease
`soon after starting therapy [39]. TNFa was increased
`during treatment compared with pretreatment values;
`this is probable evidence of accumulation of the bound
`cytokine. More research on mRNA expression in the
`myeloma and stromal cells might determine if the bioac-
`tivity of increased TNFa was attenuated fully. Infliximab
`and etanercept differ in function: etanercept binds and
`blocks both TNFa and lymphotoxin (TNFb), whereas
`infliximab binds only to TNFa. Infliximab, but not eta-
`nercept, binds to soluble TNFa bound to cell-surface
`receptors. Soluble TNFa forms stable complexes with
`
`Current Opinion in Pharmacology 2004, 4:314–320
`
`www.sciencedirect.com
`
`IPR2018-00685
`Celgene Ex. 2029, Page 3
`
`

`

`TNF-a in the pathogenesis and treatment of cancer Anderson, Nakada and DeWitte 317
`
`infliximab; TNFa dissociates more rapidly from etaner-
`cept, releasing bioactive TNFa [40]. Given these differ-
`ences and the expectation that blocking TNFa should
`show anti-myeloma activity [41], it could be worthwhile
`to investigate infliximab in this disease.
`
`Potential risks of anti-TNF-a therapy
`Because of the adaptive protective purpose of inflamma-
`tion, pharmacological inhibition of this proinflammatory
`cytokine could have adverse effects in the host, unre-
`lated to the target disease of the anti-TNFa therapy. The
`risk of reactivation of latent tuberculosis is addres-
`sed in the prescribing information for
`infliximab
`(REMICADE1; Centocor Inc, Malvern, PA). Pre-
`emptive systemic antifungal therapy is recommended
`for patients receiving infliximab for treatment of GVHD
`[34]. Smith and Skelton [42] reported cases of squamous
`cell carcinoma that became evident and grew rapidly
`during an initial period of etanercept therapy for rheu-
`matoid arthritis. The tumours might have been present,
`but occult before disruption of immunological control.
`Etanercept could disable innate anti-tumour surveil-
`lance by blockade of both lymphotoxin a and the cyto-
`toxic effects of TNFa. Additional proposed mechanisms
`include the inhibition of the Th1 cytokine pattern and
`impairment of cytotoxic T cells. All cases were in chron-
`ically UV-damaged actinic skin predisposed to tumour-
`igenesis by long-term, low-level production of TNFa.
`No new squamous cell carcinomas developed in patients
`who continued treatment
`for more than one year,
`suggesting prolonged anti-TNFa therapy could be pre-
`ventive of cutaneous malignancies.
`
`Pharmacovigilance data on etanercept, infliximab and
`adalimumab were reviewed by the FDA in 2003, with
`a focus on lymphoproliferative disease in patients treated
`with these anti-TNFa agents,
`relative to the rate
`expected in populations with immune-mediated diseases
`[43]. The potential role of TNFa-blocking therapy in the
`development of malignancies is not known. A prospective
`study of 18 572 patients with rheumatoid arthritis treated
`with anti-TNFa therapy plus methotrexate reported
`an increased standard incidence ratio compared with
`patients not receiving methotrexate or biologics, but
`confidence intervals overlapped for all treatments [44].
`Patients with highly active disease and/or chronic expo-
`sure to immunosuppressant therapies could have several-
`fold higher risk for development of lymphoma, thus
`caution should be exercised when considering anti-TNFa
`agents in patients either with a history of malignancy or
`who develop malignancy during treatment.
`
`The FDA also reported on the risk of histoplasmosis [45],
`lymphoma [46] and/or listeriosis [47]. The Mayo clinic
`reviewed the safety of infliximab in 500 patients with
`Crohn’s disease treated with infliximab [48]. The biolo-
`gical basis of concern warrants heightened vigilance and
`
`consideration of the benefit to risk ratio when prescribing
`anti-TNF therapies.
`
`Anti-TNFa therapy in cancer supportive care
`In addition to direct anti-tumour effects, anti-TNFa
`therapies are being tested for supportive care indications.
`These include cancer-related cachexia, fatigue, depres-
`sion, amelioration of chemotherapy-induced toxicities
`and metastatic bone pain [49,50]. TNFa has long been
`associated with cachexia, and trials are underway testing
`anti-TNFa agents in this condition [51,52]. Ramesh and
`Reeves [53] recently demonstrated a role for TNFa in a
`mouse model of cisplatin-mediated renal injury. Treat-
`ment of animals with TNFa synthesis inhibitors or anti-
`TNFa antibodies prevented kidney damage in the
`model.
`
`TNFa appears to play a complex role in side effects of
`radiotherapy, and anti-TNFa treatments could be useful
`in their management [54,55]. TNFa is also a known
`activator of osteoclasts [56] and mediator of neuropathic
`pain [57]. An intriguing pair of clinical cases, in which
`etanercept was used to treat refractory metastatic bone
`pain, suggest that anti-TNFa agents may ultimately play
`a role in controlling cancer pain [58]. The potential value
`of anti-TNFa agents in these debilitating conditions
`presents broad opportunities to improve cancer care.
`
`Conclusions
`It is likely that there is no ‘right’ or ‘wrong’ answer to the
`question of whether TNFa is good or bad for the
`tumour. Research should continue to optimize the use
`of pharmacological TNFa to treat cancer, as well as the
`use of neutralizing therapies to inhibit the effects of
`pathophysiologically derived TNFa. As we increase our
`understanding of the complex biology of TNFa in
`differing contexts within oncology and hematology,
`these opposing approaches should prove more effective
`against cancer.
`
`Update
`Recent results further support the anti-tumor effective-
`ness of TNFa when delivered by gene therapy
`approaches. Hecht et al. [59] reported preliminary evi-
`dence for dose-dependent improvement in progression-
`free survival in a phase I/II trial of TNFerade in combi-
`nation with radiation and 5-flourouracil
`in pancreatic
`cancer patients. Zarovni et al. [60] demonstrated substan-
`tially improved anti-tumor efficacy in animal models with
`tumor vessel-targeted peptide–TNFa fusion proteins.
`The molecules were delivered by intramuscular injection
`of cDNA expression vectors.
`
`Additional evidence demonstrating the pro-metastatic
`effects of TNFa has been published recently. Mochizuki
`et al. [61] reported that TNFa promoted metastasis of
`xenografted human gastric cancer cells to the abdominal
`
`www.sciencedirect.com
`
`Current Opinion in Pharmacology 2004, 4:314–320
`
`IPR2018-00685
`Celgene Ex. 2029, Page 4
`
`

`

`318 Cancer
`
`wall in a mouse model. Hagemann et al. [62] showed that
`macrophage-induced invasion by human breast cancer
`cells was mediated by MMPs and was dependent upon
`TNFR activity.
`
`Acknowledgements
`The authors would like to thank Patti Sassoli for her able and patient
`assistance in creating this review.
`
`References and recommended reading
`Papers of particular interest, published within the annual period of
`review, have been highlighted as:
` of special interest
` of outstanding interest
`Palladino MA, Bahjat FR, Theodorakis EA, Moldawer LL:
`1.
`Anti-TNF-alpha therapies: the next generation.
`Nat Rev Drug Discov 2003, 2:736-746.
`A comprehensive review of the biology of TNFa, the role of TNFa in human
`disease and animal models, and the use and comparison of TNFa
`antagonists approved and in clinical development.
`
`2. Younes A, Kadin ME: Emerging applications of the tumor
`
`necrosis factor family of ligands and receptors in cancer
`therapy. J Clin Oncol 2003, 21:3526-3534.
`This comprehensive, thoughtful and provocative review focuses on
`clinical applications of four TNF family member-pairs for cancer treat-
`ment: CD30/CD30 ligand, CD40/CD40 ligand, receptor activator of
`nuclear factor-kB (RANK)/RANK ligand, and TNF-related apoptosis-indu-
`cing ligand (TRAIL)/Apo-2L receptors. Structures, roles and pathways are
`clearly and succinctly illustrated. The 252 references (a tour de force)
`provide the best foundation currently available on which to build under-
`standing of the biology of these molecules relevant to the growth and,
`potentially, the interventional eradication of cancer cells.
`
`Younes A, Aggarwall BB: Clinical implications of the tumor
`necrosis factor family in benign and malignant hematologic
`disorders. Cancer 2003, 98:458-467.
`
`3.
`
`4.
`
`11. Ruegg C, Yilmaz A, Bieler G, Bamat J, Chaubert P, Lejeune FJ:
`Evidence for the involvement of endothelial cell integrin
`alphaVbeta3 in the disruption of the tumor vasculature induced
`by TNF and IFN-gamma. Nat Med 1998, 4:408-414.
`
`12. Senzer N, Mani S, Rosemurgy A, Nemunaitis J, Cunningham C,
`Guha C, Bayol N, Gillen M, Chu K, Rasmussen C et al.:
`TNFerade biologic, an adenovector with a radiation-inducible
`promoter, carrying the human tumor necrosis factor alpha
`gene: a phase I study in patients with solid tumors.
`J Clin Oncol 2004, 22:592-601.
`
`13. 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.
`
`14. Curnis F, Gasparri A, Sacchi A, Longhi R, Corti A: Coupling tumor
`necrosis factor-alpha with alphaV integrin ligands improves its
`antineoplastic activity. Cancer Res 2004, 64:565-571.
`
`15. Borsi L, Balza E, Carnemolla B, Sassi F, Castellani P, Berndt A,
`Kosmehl H, Biro A, Siri A, Orecchia P et al.: Selective targeted
`delivery of TNFalpha to tumor blood vessels. Blood 2003,
`102:4384-4392.
`
`16. Szlosarek PW, Balkwill FR: Tumour necrosis factor alpha:
`a potential target for the therapy of solid tumours.
`Lancet Oncol 2003, 4:565-573.
`
`17.
`
`Scott KA, Moore RJ, Arnott CH, East N, Thompson RG, Scallon BJ,
`Shealy DJ, Balkwill FR: An anti-tumor necrosis factor-alpha
`antibody inhibits the development of experimental skin tumors.
`Mol Cancer Ther 2003, 2:445-451.
`This paper establishes the anti-tumour effects of an anti-TNF antibody
`in a well-defined model system.
`
`18. Arnott CH, Scott KA, Moore RJ, Robinson SC, Thompson RG,
`Balkwill FR: Expression of both TNF-alpha receptor subtypes is
`essential for optimal skin tumour development. Oncogene 2003.
`
`19. Mantovani G, Maccio A, Mura L, Massa E, Mudu MC, Mulas C,
`Lusso MR, Madeddu C, Dessi A: Serum levels of leptin and
`proinflammatory cytokines in patients with advanced-stage
`cancer at different sites. J Mol Med 2000, 78:554-561.
`
`Eggermont AM, ten Hagen TL: Tumor necrosis factor-based
`isolated limb perfusion for soft tissue sarcoma and melanoma:
`ten years of successful antivascular therapy. Curr Oncol Rep
`2003, 5:79-80.
`
`20. Karayiannakis AJ, Syrigos KN, Polychronidis A, Pitiakoudis M,
`Bounovas A, Simopoulos K: Serum levels of tumor necrosis
`factor-alpha and nutritional status in pancreatic cancer
`patients. Anticancer Res 2001, 21:1355-1358.
`
`5. Dranoff G: Cytokines in cancer pathogenesis and cancer
`
`therapy. Nat Rev Cancer 2004, 4:11-22.
`An excellent review of the conflicting role of cytokines in inhibiting tumour
`development and progression, as well as in promoting tumour growth and
`mediating invasion and metastasis. The cell sources and roles for endo-
`genous cytokines in tumour progression, as well a listing of cytokines as
`cancer therapy, are presented in table form.
`
`6. Balkwill F: Tumor necrosis factor or tumor promoting factor?
`Cytokine Growth Factor Rev 2002, 13:135-141.
`An excellent and very thorough review of evidence for both the pro- and
`anti-tumour effects of TNFa.
`7. Micheau O, Tschopp J: Induction of TNF receptor I-mediated
`
`apoptosis via two sequential signaling complexes.
`Cell 2003, 114:181-190.
`Provides a new model
`for the cellular and molecular mechanisms
`determining the fate of cells following TNFa binding.
`
`8.
`
`Lejeune FJ, Ruegg C, Lienard D: Clinical applications of
`TNF-alpha in cancer. Curr Opin Immunol 1998, 10:573-580.
`
`Eggermont AM, de Wilt JH, ten Hagen TL: Current uses of isolated
`9.
`limb perfusion in the clinic and a model system for new
`strategies. Lancet Oncol 2003, 4:429-437.
`A review of the use of drugs, including TNFa, with delivery via isolated
`limb perfusion. Includes a summary of response rates for the treatment of
`in-transit melanoma metastases and soft-tissue sarcomas, and reviews
`preclinical research being performed to advance the clinical applications
`of this technology.
`
`10. Folli S, Pelegrin A, Chalandon Y, Yao X, Buchegger F, Lienard D,
`Lejeune F, Mach JP: Tumor-necrosis factor can enhance
`radio-antibody uptake in human colon carcinoma xenografts
`by increasing vascular permeability. Int J Cancer 1993,
`53:829-836.
`
`21. Yoshida N, Ikemoto S, Narita K, Sugimura K, Wada S, Yasumoto R,
`Kishimoto T, Nakatani T: Interleukin-6, tumour necrosis factor
`alpha and interleukin-1beta in patients with renal cell
`carcinoma. Br J Cancer 2002, 86:1396-1400.
`
`22. Leek RD, Landers R, Fox SB, Ng F, Harris AL, Lewis CE:
`Association of tumour necrosis factor alpha and its receptors
`with thymidine phosphorylase expression in invasive breast
`carcinoma. Br J Cancer 1998, 77:2246-2251.
`
`23. Partanen R, Koskinen H, Hemminki K: Tumour necrosis
`factor-alpha (TNF-alpha) in patients who have asbestosis
`and develop cancer. Occup Environ Med 1995, 52:316-319.
`
`24. Pfitzenmaier J, Vessella R, Higano CS, Noteboom JL, Wallace D Jr,
`Corey E: Elevation of cytokine levels in cachectic patients with
`prostate carcinoma. Cancer 2003, 97:1211-1216.
`
`25. Bossola M, Muscaritoli M, Bellantone R, Pacelli F, Cascino A,
`Sgadari A, Battaglia F, Piccioni E, Scambia G, Doglietto GB et al.:
`Serum tumour necrosis factor-alpha levels in cancer patients
`are discontinuous and correlate with weight loss.
`Eur J Clin Invest 2000, 30:1107-1112.
`
`26. Darai E, Detchev R, Hugol D, Quang NT: Serum and cyst fluid
`levels of interleukin (IL) -6, IL-8 and tumour necrosis factor-
`alpha in women with endometriomas and benign and malignant
`cystic ovarian tumours. Hum Reprod 2003, 18:1681-1685.
`
`27. Ness RB: Endometriosis and ovarian cancer: thoughts on
`
`shared pathophysiology. Am J Obstet Gynecol 2003,
`189:280-294.
`In this excellent and comprehensive review, the author develops and
`explores a unifying concept of local hormonal and inflammatory condi-
`tions existent within a causal pathway of malignant transformation.
`The broad range of evidence is from the cellular to the epidemiologic,
`
`Current Opinion in Pharmacology 2004, 4:314–320
`
`www.sciencedirect.com
`
`IPR2018-00685
`Celgene Ex. 2029, Page 5
`
`

`

`TNF-a in the pathogenesis and treatment of cancer Anderson, Nakada and DeWitte 319
`
`connecting inflammation with endometriosis and ovarian cancer; the
`bibliography is extensive.
`
`28.
`
`Tselepis C, Perry I, Dawson C, Hardy R, Darnton SJ, McConkey C,
`Stuart RC, Wright N, Harrison R, Jankowski JA: Tumour necrosis
`factor-alpha in Barrett’s oesophagus: a potential novel
`mechanism of action. Oncogene 2002, 21:6071-6081.
`The authors interpret the immunohistochemistry and assess expression
`of TNFa and TNFR1 in Barrett’s metaplasia, dysplasia and adenocarci-
`noma, linking the cytokine to histopathological progression of disease.
`Moreover, they elegantly show elevated transcription of the oncogenec-
`myc, mediated by b-catenin, following stimulation by TNFa, and present
`evidence for an early role of TNFa along the progression continuum. The
`discussion is thorough, cogent and hypothesis-provoking.
`
`29.
`
`Ferrajoli A, Keating MJ, Manshouri T, Giles FJ, Dey A, Estrov Z,
`Koller CA, Kurzrock R, Thomas DA, Faderl S et al.: The clinical
`significance of tumor necrosis factor-alpha plasma level in
`patients having chronic lymphocytic leukemia. Blood 2002,
`100:1215-1219.
`This report provides an example of a potentially therapeutic application of
`biomedical research to clinical practice, based on correlations with extent
`of disease and determination of prognostic factors. From a well-explained
`rationale and with carefully established associations, the authors suggest
`that TNFa is involved with progression of CLL, with variability related
`to complexity of the TNFa protein and receptor system in vivo. The
`discussion raises several
`important questions and suggests specific
`directions for additional studies of cytokine regulation in hematologic
`malignancy.
`
`30. Tsimberidou AM, Giles FJ: TNF-alpha targeted therapeutic
`approaches in patients with hematologic malignancies.
`Expert Rev Anticancer Ther 2002, 2:277-286.
`
`31. Korngold R, Marini JC, de Baca ME, Murphy GF, Giles-Komar J:
`Role of tumor necrosis factor-alpha in graft-versus-host
`disease and graft-versus-leukemia responses. Biol Blood
`Marrow Transplant 2003, 9:292-303.
`
`32.
`
`Jacobsohn DA, Hallick J, Anders V, McMillan S, Morris L,
`Vogelsang GB: Infliximab for steroid-refractory acute
`GVHD: a case series. Am J Hematol 2003, 74:119-124.
`The authors present a balanced assessment of the therapeutic use of an
`anti- TNFa monoclonal antibody in the excruciatingly difficult context of
`refractory GVHD. This case series report clearly demonstrates the need to
`understand the risk:benefit ratio in each individual patient when pharma-
`cologically blocking TNFa. The unmet medical need is identified by these
`authors who participate in this speciality of, perhaps, the most pragmatic
`and courageous of clinical immunologists.
`
`33. Couriel DR, Saliba R, Hicks K, Cohen A, Ippoliti C, Neumann J,
`Giralt S, De Lima MJ, Champlin R: ASH 44th annual meeting,
`6-10 December 2002, Philadelphia, PA [Abstract 3343].
`
`34. Marty FM, Lee SJ, Fahey MM, Alyea EP, Soiffer RJ, Antin JH,
`Baden LR: Infliximab use in patients with severe graft-versus-
`host disease and other emerging risk factors of non-Candida
`invasive fungal infections in allogeneic hematopoietic stem
`cell transplant recipients: a cohort study. Blood 2003,
`102:2768-2776.
`
`35. Campos A, Vaz CP, Costa N, Mendes J, Roncon S, Barbosa I,
`Carvalhais A, Campilho F, Pimentel P: ASH 45th Annual Meeting,
`6-9 December 2003, San Diego, CA [Abstract 5528].
`
`36. Stasi R, Amadori S: Infliximab chimaeric anti-tumour necrosis
`factor alpha monoclonal antibody treatment for patients
`with myelodysplastic syndromes. Br J Haematol 2002,
`116:334-337.
`
`37. Raza A, Lisak LA, Tahir S, Billmeier JM, Willmann HS, Bellak DJ,
`Alvi MI, Gezer S, Venugopal P: ASH 44th Annual Meeting, 6-1

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