Tumor necrosis factor in lung cancer: Complex roles in biology and resistance to treatment

Ke Gong, Gao Guo, Nicole Beckley, Yue Zhang, Xiaoyao Yang, Mishu Sharma, Amyn A Habib, Ke Gong, Gao Guo, Nicole Beckley, Yue Zhang, Xiaoyao Yang, Mishu Sharma, Amyn A Habib

Abstract

Tumor necrosis factor (TNF) and its receptors are widely expressed in non-small cell lung cancer (NSCLC). TNF has an established role in inflammation and also plays a key role in inflammation-induced cancer. TNF can induce cell death in cancer cells and has been used as a treatment in certain types of cancer. However, TNF is likely to play an oncogenic role in multiple types of cancer, including NSCLC. TNF is a key activator of the transcription factor NF-κB. NF-κB, in turn, is a key effector of TNF in inflammation-induced cancer. Data from The Cancer Genome Atlas database suggest that TNF could be a biomarker in NSCLC and indicate a complex role for TNF and its receptors in NSCLC. Recent studies have reported that TNF is rapidly upregulated in NSCLC in response to targeted treatment with epidermal growth factor receptor (EGFR) inhibition, and this upregulation leads to NF-κB activation. The TNF upregulation and consequent NF-κB activation play a key role in mediating both primary and secondary resistance to EGFR inhibition in NSCLC, and a combined inhibition of EGFR and TNF can overcome therapeutic resistance in experimental models. TNF may mediate the toxic side effects of immunotherapy and may also modulate resistance to immune checkpoint inhibitors. Drugs inhibiting TNF are widely used for the treatment of various inflammatory and rheumatologic diseases and could be quite useful in combination with targeted therapy of NSCLC and other cancers.

Trial registration: ClinicalTrials.gov NCT00410059.

Keywords: EGFR inhibition; Immunotherapy; NSCLC; TCGA; TNF; Therapeutic resistance.

Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1
Figure 1
TNF affects TCGA-NSCLC patient survival. (A−C) The Cancer Genome Atlas Lung Squamous Cell Carcinoma (TCGA-LUSC) patients with RNAseq and survival data were divided into high-50% and low-50% groups by TNF, TNFR1, and TNFR2 mRNA levels and the effect on overall survival was examined. Kaplan–Meier (KM) survival curves were drawn and compared by log-rank test and Gehan's test. (D−F) 500 primary TCGA-Lung Adenocarcinoma (TCGA-LUAD) patients’ RNAseq and survival data was analyzed. (G−I). Among the 500 primary TCGA-LUAD patients, there are 41 patients harboring EGFR-activating mutation (with L858R or exon 19 deletion, but without T790M mutation). Roles of TNF/TNFR1/TNFR2 on these patients’ survival data was examined. P <0.05 of log-rank was considered statistically significant. Analysis was performed by GraphPad 9. TCGA, The Cancer Genome Atlas; TNF, tumor necrosis factor; TNFR1, TNF receptor 1; TNFR2, TNF receptor 2.
Figure 2
Figure 2
Higher TNF superfamily ligands and receptors determine worse prognosis of NSCLC patients treated with EGFR inhibition. (A) A summary table of survival analysis on TNF superfamily ligands and receptors in the BATTLE trial data (ClinicalTrials.gov NCT00410059 for erlotinib). The higher HR (Hazard Ratio) >1 means the gene would likely promote tumor growth and shorten the progression-free survival (PFS). BATTLE data were provided by the BATTLE group at MD Anderson Cancer Center . (B−D) KM survival curves were drawn on 3 TNF superfamily ligands (TNFSFs) with top 5 HR, the other 2 ligands TNF and LTB was reported in our previous study . (E) The first principle component (PC1) score of current known TNFSFs and TNF superfamily receptors (TNFRSFs) as listed in the Figure 2A for each patient in NCT00410059 was calculated. Patients with higher or lower PC1 scores were compared by KM survival analysis. (F) All gene survival analysis was performed on NCT00410059. All genes were ranked by HR to distinguish oncogenes and tumor suppressors. The enrichment plot by Gene Set Enrichment Analysis (GSEA) shows TNFSF & TNFRSF signature (genes in the Figure 2A) was significantly enriched in oncogenes, genes with higher HR that bring in shorter PFS under erlotinib treatment. (G) KM survival curves of IκBα. P <0.05 of log-rank was considered statistically significant. Analysis was performed by GraphPad 9, and GSEA: www.gsea-msigdb.org. BATTLE, Biomarker-integrated Approaches of Targeted Therapy for Lung Cancer Elimination; NSCLC, non-small cell lung cancer; TNF, tumor necrosis factor; TNFSFs, TNF superfamily ligands.
Figure 3
Figure 3
Schematic diagram of TNF signaling and the effect of EGFR and TNF inhibition in NSCLC. Representative TNF superfamily ligands and receptors predicting worse prognosis in the BATTLE trial with statistically significance were labelled in red. BATTLE, Biomarker-integrated Approaches of Targeted Therapy for Lung Cancer Elimination; NSCLC, non-small cell lung cancer; TNF, tumor necrosis factor. (Color version of figure is available online.)

References

    1. Wajant H., Pfizenmaier K., Scheurich P. Tumor necrosis factor signaling. Cell Death Differ. 2003;10:45–65.
    1. Kalliolias G.D., Ivashkiv L.B. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat Rev Rheumatol. 2016;12:49–62.
    1. Jarosz-Griffiths H.H., Holbrook J., Lara-Reyna S., McDermott M.F. TNF receptor signalling in autoinflammatory diseases. Int Immunol. 2019;31:639–648.
    1. Sedger L.M., McDermott M.F. TNF and TNF-receptors: From mediators of cell death and inflammation to therapeutic giants - past, present and future. Cytokine Growth Factor Rev. 2014;25:453–472.
    1. Cruceriu D., Baldasici O., Balacescu O., Berindan-Neagoe I. The dual role of tumor necrosis factor-alpha (TNF-alpha) in breast cancer: molecular insights and therapeutic approaches. Cell Oncol (Dordr) 2020;43:1–18.
    1. Lis K., Kuzawinska O., Balkowiec-Iskra E. Tumor necrosis factor inhibitors - state of knowledge. Arch Med Sci. 2014;10:1175–1185.
    1. Ma X., Xu S. TNF inhibitor therapy for rheumatoid arthritis. Biomed Rep. 2013;1:177–184.
    1. Carswell E.A., Old L.J., Kassel R.L., Green S., Fiore N., Williamson B. An endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci U S A. 1975;72:3666–3670.
    1. Balkwill F. Tumour necrosis factor and cancer. Nat Rev Cancer. 2009;9:361–371.
    1. Montfort A., Colacios C., Levade T., Andrieu-Abadie N., Meyer N., Segui B. The TNF paradox in cancer progression and immunotherapy. Front Immunol. 2019;10:1818.
    1. Waters J.P., Pober J.S., Bradley J.R. Tumour necrosis factor and cancer. J Pathology. 2013;230:241–248.
    1. Aggarwal B.B. Signalling pathways of the TNF superfamily: a double-edged sword. Nat Rev Immunol. 2003;3:745–756.
    1. Bertazza L., Mocellin S. Tumor necrosis factor (TNF) biology and cell death. Front Biosci. 2008;13:2736–2743.
    1. Cordero J.B., Macagno J.P., Stefanatos R.K., Strathdee K.E., Cagan R.L., Vidal M. Oncogenic Ras diverts a host TNF tumor suppressor activity into tumor promoter. Dev Cell. 2010;18:999–1011.
    1. Guo G., Gong K., Ali S., Ali N., Shallwani S., Hatanpaa K.J., Pan E., Mickey B., Burma S., Wang D.H. A TNF-JNK-Axl-ERK signaling axis mediates primary resistance to EGFR inhibition in glioblastoma. Nat Neurosci. 2017;20:1074–1084.
    1. Guo G., Gong K., Puliyappadamba V.T., Panchani N., Pan E., Mukherjee B., Damanwalla Z., Bharia S., Hatanpaa K.J., Gerber D.E. Efficacy of EGFR plus TNF inhibition in a preclinical model of temozolomide-resistant glioblastoma. Neuro Oncol. 2019;21:1529–1539.
    1. Gong K., Guo G., Gerber D.E., Gao B., Peyton M., Huang C., Minna J.D., Hatanpaa K.J., Kernstine K., Cai L. TNF-driven adaptive response mediates resistance to EGFR inhibition in lung cancer. J Clin Invest. 2018;128:2500–2518.
    1. Gong K., Guo G., Panchani N., Bender M.E., Gerber D.E., Minna J.D., Fattah F., Gao B., Peyton M., Kernstine K. EGFR inhibition triggers an adaptive response by co-opting antiviral signaling pathways in lung cancer. Nat Cancer. 2020;1:394–409.
    1. Ma L., She C., Shi Q., Yin Q., Ji X., Wang Y., Fan Y., Kong X., Li P., Sun Z. TNFalpha inhibitor C87 sensitizes EGFRvIII transfected glioblastoma cells to gefitinib by a concurrent blockade of TNFalpha signaling. Cancer Biol Med. 2019;16:606–617.
    1. Luo Z., Wang B., Liu H., Shi L. TNF inhibitor pomalidomide sensitizes glioblastoma cells to EGFR inhibition. Ann Clin Lab Sci. 2020;50:474–480.
    1. Perez-Ruiz E., Minute L., Otano I., Alvarez M., Ochoa M.C., Belsue V., de Andrea C., Rodriguez-Ruiz M.E., Perez-Gracia J.L., Marquez-Rodas I. Prophylactic TNF blockade uncouples efficacy and toxicity in dual CTLA-4 and PD-1 immunotherapy. Nature. 2019;569:428–432.
    1. Bertrand F., Montfort A., Marcheteau E., Imbert C., Gilhodes J., Filleron T., Rochaix P., Andrieu-Abadie N., Levade T., Meyer N. TNFalpha blockade overcomes resistance to anti-PD-1 in experimental melanoma. Nat Commun. 2017;8:2256.
    1. Brenner D., Blaser H., Mak T.W. Regulation of tumour necrosis factor signalling: live or let die. Nat Rev Immunol. 2015;15:362–374.
    1. Faustman D., Davis M. TNF receptor 2 pathway: drug target for autoimmune diseases. Nat Rev Drug Discov. 2010;9:482–493.
    1. Tran T.A., Kallakury B.V., Ambros R.A., Ross J.S. Prognostic significance of tumor necrosis factors and their receptors in nonsmall cell lung carcinoma. Cancer. 1998;83:276–282.
    1. Zhang Y.W., Chen Q.Q., Cao J., Xu L.Q., Tang X., Wang J., Zhang J., Dong L.X. Expression of tumor necrosis factor receptor 2 in human non-small cell lung cancer and its role as a potential prognostic biomarker. Thorac Cancer. 2019;10:437–444.
    1. Balkwill F. TNF-alpha in promotion and progression of cancer. Cancer Metastasis Rev. 2006;25:409–416.
    1. Sethi G., Sung B., Aggarwal B.B. TNF: a master switch for inflammation to cancer. Front Biosci. 2008;13:5094–5107.
    1. Hazra A., Gogtay N. Biostatistics series module 9: survival analysis. Indian J Dermatol. 2017;62:251–257.
    1. Midha A., Dearden S., McCormack R. EGFR mutation incidence in non-small-cell lung cancer of adenocarcinoma histology: a systematic review and global map by ethnicity (mutMapII) Am J Cancer Res. 2015;5:2892–2911.
    1. Gazdar A.F. Activating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene. 2009;28(Suppl 1):S24–S31.
    1. Chen F., Chen N., Yu Y., Cui J. Efficacy and safety of epidermal growth factor receptor (egfr) inhibitors plus antiangiogenic agents as first-line treatments for patients with advanced EGFR-mutated non-small cell lung cancer: a meta-analysis. Front Oncol. 2020;10:904.
    1. Hess L.M., Brnabic A., Mason O., Lee P., Barker S. Relationship between progression-free survival and overall survival in randomized clinical trials of targeted and biologic agents in oncology. J Cancer. 2019;10:3717–3727.
    1. Kim E.S., Herbst R.S., Wistuba I.I., Lee J.J., Blumenschein G.R., Jr., Tsao A., Stewart D.J., Hicks M.E., Erasmus J., Jr., Gupta S. The BATTLE trial: personalizing therapy for lung cancer. Cancer Discov. 2011;1:44–53.
    1. Nikolaev A., McLaughlin T., O'Leary D.D., Tessier-Lavigne M. APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature. 2009;457:981–989.
    1. Shen Y.J., Huang S.G. Improve survival prediction using principal components of gene expression data. Genomics Proteomics Bioinformatics. 2006;4:110–119.
    1. Chen D.T., Davis-Yadley A.H., Huang P.Y., Husain K., Centeno B.A., Permuth-Wey J., Pimiento J.M., Malafa M. Prognostic fifteen-gene signature for early stage pancreatic ductal adenocarcinoma. PLoS One. 2015;10
    1. Subramanian A., Tamayo P., Mootha V.K., Mukherjee S., Ebert B.L., Gillette M.A., Paulovich A., Pomeroy S.L., Golub T.R., Lander E.S. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–15550.
    1. Taniguchi K., Karin M. NF-kappaB, inflammation, immunity and cancer: coming of age. Nat Rev Immunol. 2018;18:309–324.
    1. Karin M., Cao Y., Greten F.R., Li Z.W. NF-kappaB in cancer: from innocent bystander to major culprit. Nat Rev Cancer. 2002;2:301–310.
    1. Hayden M.S., Ghosh S. Regulation of NF-kappaB by TNF family cytokines. Seminars in immunology. 2014;26:253–266.
    1. Shi J.H., Sun S.C. Tumor necrosis factor receptor-associated factor regulation of nuclear factor kappaB and mitogen-activated protein kinase pathways. Front Immunol. 2018;9:1849.
    1. Zhang Q., Lenardo M.J., Baltimore D. 30 years of NF-kappaB: a blossoming of relevance to human pathobiology. Cell. 2017;168:37–57.
    1. Napetschnig J., Wu H. Molecular basis of NF-kappaB signaling. Annu Rev Biophys. 2013;42:443–468.
    1. Collart M.A., Baeuerle P., Vassalli P. Regulation of tumor necrosis factor alpha transcription in macrophages: involvement of four kappa B-like motifs and of constitutive and inducible forms of NF-kappa B. Mol Cell Biol. 1990;10:1498–1506.
    1. Bivona T.G., Hieronymus H., Parker J., Chang K., Taron M., Rosell R., Moonsamy P., Dahlman K., Miller V.A., Costa C. FAS and NF-kappaB signalling modulate dependence of lung cancers on mutant. EGFR Nature. 2011;471:523–526.
    1. Blakely C.M., Pazarentzos E., Olivas V., Asthana S., Yan J.J., Tan I., Hrustanovic G., Chan E., Lin L., Neel D.S. NF-kappaB-activating complex engaged in response to EGFR oncogene inhibition drives tumor cell survival and residual disease in lung cancer. Cell Rep. 2015;11:98–110.
    1. Hatanpaa K.J., Burma S.., Zhao D.., Habib A..A. Epidermal growth factor receptor (EGFR) in glioma: signal transduction, neuropathology, imaging and radioresistance. Neoplasia. 2010;12:675–684.
    1. An Z., Aksoy O., Zheng T., Fan Q.W., Weiss W.A. Epidermal growth factor receptor and EGFRvIII in glioblastoma: signaling pathways and targeted therapies. Oncogene. 2018;37:1561–1575.
    1. Thorne A.H., Zanca C., Furnari F. Epidermal growth factor receptor targeting and challenges in glioblastoma. Neuro Oncol. 2016;18:914–918.
    1. Warta R., Herold-Mende C. Helping EGFR inhibition to block cancer. Nat Neurosci. 2017;20:1035–1037.
    1. Schueler J., Tschuch C., Klingner K., Bug D., Peille A.L., de Koning L., Oswald E., Klett H., Sommergruber W. Induction of acquired resistance towards egfr inhibitor gefitinib in a patient-derived xenograft model of non-small cell lung cancer and subsequent molecular characterization. Cells. 2019;8:740.
    1. Pan Y.H., Lin C.Y., Lu C.H., Li L., Wang Y.B., Chen H.Y., He Y. Metformin synergistically enhances the antitumor activity of the third-generation EGFR-TKI CO-1686 in lung cancer cells through suppressing NF-kappaB signaling. Clin Respir J. 2018;12:2642–2652.
    1. Yoo D.G., Kim C.S., Lee S.K., Kim H.S., Cho E.J., Park M.S., Lee S.D., Park J.B., Jeon B.H. Redox factor-1 inhibits cyclooxygenase-2 expression via inhibiting of p38 MAPK in the A549 cells Korean. J Physiol Pharmacol. 2010;14:139–144.
    1. Chen C.C., Sun Y.T., Chen J.J., Chiu K.T. TNF-alpha-induced cyclooxygenase-2 expression in human lung epithelial cells: involvement of the phospholipase C-gamma 2, protein kinase C-alpha, tyrosine kinase, NF-kappa B-inducing kinase, and I-kappa B kinase 1/2 pathway. J Immunol. 2000;165:2719–2728.
    1. Xiao J., Wang F., Lu H., Xu S., Zou L., Tian Q., Fu Y., Lin X., Liu L., Yuan P. Targeting the COX2/MET/TOPK signaling axis induces apoptosis in gefitinib-resistant NSCLC cells. Cell Death Dis. 2019;10:777.
    1. Kim Y.M., Park S.Y., Pyo H. Cyclooxygenase-2 (COX-2) negatively regulates expression of epidermal growth factor receptor and causes resistance to gefitinib in COX-2-overexpressing cancer cells. Mol Cancer Res. 2009;7:1367–1377.
    1. Lu Y., Shi C., Qiu S., Fan Z. Identification and validation of COX-2 as a co-target for overcoming cetuximab resistance in colorectal cancer cells. Oncotarget. 2016;7:64766–64777.
    1. Gadgeel S.M., Ruckdeschel J.C., Heath E.I., Heilbrun L.K., Venkatramanamoorthy R., Wozniak A. Phase II study of gefitinib, an epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), and celecoxib, a cyclooxygenase-2 (COX-2) inhibitor, in patients with platinum refractory non-small cell lung cancer (NSCLC) J Thorac Oncol. 2007;2:299–305.
    1. Reckamp K.L., Koczywas M., Cristea M.C., Dowell J.E., Wang H.J., Gardner B.K., Milne G.L., Figlin R.A., Fishbein M.C., Elashoff R.M. Randomized phase 2 trial of erlotinib in combination with high-dose celecoxib or placebo in patients with advanced non-small cell lung cancer. Cancer. 2015;121:3298–3306.
    1. Terzuoli E., Costanza F., Ciccone V., Ziche M., Morbidelli L., Donnini S. mPGES-1 as a new target to overcome acquired resistance to gefitinib in non-small cell lung cancer cell lines. Prostaglandins Other Lipid Mediat. 2019;143
    1. Hwang H.S., Park Y.Y., Shin S.J., Go H., Park J.M., Yoon S.Y., Lee J.L., Cho Y.M. Involvement of the TNF-alpha pathway in TKI resistance and suggestion of TNFR1 as a predictive biomarker for TKI responsiveness in clear cell renal cell carcinoma. J Korean Med Sci. 2020;35:e31.
    1. Yoshida R., Barbie D.A. EGFR blockade activates interferon. Nat Cancer. 2020;1:376–378.
    1. Doroshow D.B., Herbst R.S. Treatment of advanced non-small cell lung cancer in 2018. JAMA Oncol. 2018;4:569–570.
    1. Thungappa S., Ferri J., Caglevic C., Passiglia F., Raez L., Rolfo C. Immune checkpoint inhibitors in lung cancer: the holy grail has not yet been found. ESMO Open. 2017;2
    1. Lee C.K., Man J., Lord S., Links M., Gebski V., Mok T., Yang J.C. Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer-a meta-analysis. J Thorac Oncol. 2017;12:403–407.
    1. Gainor J.F. Square peg, round hole? Programmed death-1 inhibitors in epidermal growth factor receptor-mutant non-small cell lung cancer. J Thorac Dis. 2018;10:25–29.
    1. Oronsky B., Ma P., Reid T.R., Cabrales P., Lybeck M., Oronsky A., Oronsky N., Carter C.A. Navigating the "No Man's Land" of TKI-failed EGFR-mutated non-small cell lung cancer (NSCLC): a review. Neoplasia. 2018;20:92–98.
    1. Kennedy L.B., Salama A.K.S. A review of cancer immunotherapy toxicity CA. Cancer J Clin. 2020;70:86–104.
    1. Marin-Acevedo J.A., Chirila R.M., Dronca R.S. Immune checkpoint inhibitor toxicities. Mayo Clin Proc. 2019;94:1321–1329.
    1. Thompson J.A., Schneider B.J., Brahmer J., Andrews S., Armand P., Bhatia S., Budde L.E., Costa L., Davies M., Dunnington D. Management of immunotherapy-related toxicities, version 1.2019. J Natl Compr Canc Netw. 2019;17:255–289.
    1. Weber J.S., Postow M.A. TNFalpha blockade in checkpoint inhibition: the good, the bad, or the ugly? Clin Cancer Res. 2020;26:2085–2086.
    1. Arriola E., Wheater M., Karydis I., Thomas G., Ottensmeier C. Infliximab for IPILIMUMAB-related colitis-letter. Clin Cancer Res. 2015;21:5642–5643.
    1. Badran Y.R., Cohen J.V., Brastianos P.K., Parikh A.R., Hong T.S., Dougan M. Concurrent therapy with immune checkpoint inhibitors and TNFalpha blockade in patients with gastrointestinal immune-related adverse events. J Immunother Cancer. 2019;7:226.
    1. Johnson D.H., Zobniw C.M., Trinh V.A., Ma J., Bassett R.L., Jr., Abdel-Wahab N., Anderson J., Davis J.E., Joseph J., Uemura M. Infliximab associated with faster symptom resolution compared with corticosteroids alone for the management of immune-related enterocolitis. J Immunother Cancer. 2018;6:103.
    1. Horwood P.F., Soli K.W., Maure T., Naito Y.I., Morita A., Natsuhara K., Tadokoro K., Baba J., Odani S., Tomitsuka E. A high burden of asymptomatic gastrointestinal infections in traditional communities in Papua New Guinea. Am J Trop Med Hyg. 2017;97:1872–1875.
    1. Zhang C., Zhang G., Sun N., Zhang Z., Zhang Z., Luo Y., Che Y., Xue Q., He J. Comprehensive molecular analyses of a TNF family-based signature with regard to prognosis, immune features, and biomarkers for immunotherapy in lung adenocarcinoma. EBioMedicine. 2020;59
    1. Vredevoogd D.W., Kuilman T., Ligtenberg M.A., Boshuizen J., Stecker K.E., de Bruijn B., Krijgsman O., Huang X., Kenski J.C.N., Lacroix R. Augmenting immunotherapy impact by lowering tumor TNF cytotoxicity. Threshold Cell. 2020;180:404–405.
    1. Verheijden R.J., May A.M., Blank C.U., Aarts M.J.B., van den Berkmortel F., van den Eertwegh A.J.M., de Groot J.W.B., Boers-Sonderen M.J., van der Hoeven J.J.M., Hospers G.A. Association of Anti-TNF with decreased survival in steroid refractory ipilimumab and anti-PD1-treated patients in the dutch melanoma treatment registry. Clin Cancer Res. 2020;26:2268–2274.

Source: PubMed

3
Abonnieren