Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 3979 word(s) 3979 2021-02-25 07:50:51 |
2 update layout and reference -2147 word(s) 1832 2021-02-26 04:47:59 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Tubin, S. Biology of NSCLC. Encyclopedia. Available online: https://encyclopedia.pub/entry/7593 (accessed on 02 July 2024).
Tubin S. Biology of NSCLC. Encyclopedia. Available at: https://encyclopedia.pub/entry/7593. Accessed July 02, 2024.
Tubin, Slavisa. "Biology of NSCLC" Encyclopedia, https://encyclopedia.pub/entry/7593 (accessed July 02, 2024).
Tubin, S. (2021, February 25). Biology of NSCLC. In Encyclopedia. https://encyclopedia.pub/entry/7593
Tubin, Slavisa. "Biology of NSCLC." Encyclopedia. Web. 25 February, 2021.
Biology of NSCLC
Edit

The overall prognosis and survival of non-small cell lung cancer (NSCLC) patients remain poor. The immune system plays an integral role in driving tumor control, tumor progression, and overall survival of NSCLC patients. While the tumor cells possess many ways to escape the immune system, conventional radiotherapy (RT) approaches, which are directly cytotoxic to tumors, can further add additional immune suppression to the tumor microenvironment by destroying many of the lymphocytes that circulate within the irradiated tumor environment.

non-small cell lung cancer immunosuppression immunostimulation immunotherapy radiother-apy

1. Introduction

The World Health Organization (WHO) estimated 1.76 million deaths caused by non-small cell lung cancer (NSCLC) in 2018. Lung cancer represents the leading cause of deaths worldwide with NSCLC representing approximately 85% of all lung malignancies [1]. Despite substantial improvements in therapy, 5-year overall survival (OS) for NSCLC does not exceed 25% [2][3]. Prognosis and survival of patients affected by NSCLC are correlated to disease stage, with OS being prognostically favorable by earlier diagnosis and treatment [4][5]. Nevertheless, a significant percent of patients are diagnosed in advanced stage with little chance to be cured.

Chronic inflammation plays a key role in tumorigenesis of NSCLC [6]. Inflammatory factors like cigarette smoking are associated with chronic bronchitis and emphysema, which lead to the development of lung cancer [7]. The process of cellular malignant transformation consists of many steps over long time periods, ranging from pre-carcinogenic chronic inflammation, progressing, if not treated, towards the development of invasive carcinoma and systemic disease [8][9][10]. Some chronically inflamed pre-carcinogenic environments will never result in malignant cell transformation, while others exposed to the same carcinogen will undergo tumorigenesis. This uncertainty of initiating tumorigenesis events highlights the malignant “modulating” role of genetic predisposition in cancer [11] and underscores that inflammation may assist in this process. The profile and status of the inflammatory-changed environment following the chronic carcinogen(s) exposure is exceedingly variable, and its potential for malignant transformation is related to polymorphic immune response genes affected by a diversity of anti-oxidant and DNA repair associated genes [11]. The immune system, in its cell-mediated and humoral form, is deeply involved in generation of an inflammatory environment and is considered to be the first step of tumorigenesis. Chronic exposure of normal cells to carcinogen(s) leads to initiation of immune cell activation with subsequent upregulation of the pro-inflammatory cytokines like Interleukin-1 alpha (IL-1α) and IL-1-β, and production of Cyclooxygenase (COX)-1 and COX-2 immune-regulatory enzymes in epithelial and mesenchymal. This activation of the inflammatory pathways is associated with the development of malignant disease [12][13][14][15]. Additionally, the induction of COX-2 favors increased angiogenesis in an inflammatory environment and helps pave the way for hyper-vascularization needed for tumor development and progression [16][17][18].

In addition to assisting tumorigenesis, the immune system also plays a fundamental role in the defense against cancer by surveillance and identification of foreign or “non-self” from self and assisting with elimination of DNA-damaged cancer cells from the body. However, tumor cells can escape by suppressing the immune system. Furthermore, therapies, especially if they cause immune-suppression, may further aid the escape of the tumor cells from the immune system. Most patients with NSCLC will eventually require radiotherapy (RT) alone (especially those that are medically inoperable) or in combination with systemic therapy (especially those with advanced stage disease). Regardless, there are many reports that show that conventional RT induces an immunosuppression, and thereby can negatively affect the overall survival [19][20][21][22]. Thus, the current immunogenic balance, determined by the inhibitory effects caused by tumor cells themselves and radiation therapy can lead to poor clinical outcomes. There is newer emerging evidence suggesting that immunosuppression is an “elastic process” that can be transformed into an immune-stimulating environment by “correcting” many manipulable components of this triangle radiation, tumor, and immune cells by using combination approaches that change the way RT, chemotherapy, and immunotherapy are used. Significant efforts are being deployed in the development of novel treatment strategies and protocols aimed to convert radiation- and tumor-induced immunosuppression into a predominant immunostimulation by means of immunotherapy, unconventional RT and the combination of these modalities in order to improve the outcomes of NSCLC patients.

2. Tumor-Related Immune Suppression in NSCLC

The interplay between the tumor and immune cells is the subject of an extended and ongoing research. Table 1 represents latest evidence on tumor-related immunosuppressive effects [23][24][25][26][27][28][29][30][31][32][33][34][35][36][37][38][39][40][41][42][43][44][45][46][47][48][49][50][51][52][53][54][55][56]. Tumor cells escape immune surveillance by downregulation of HLA and co-stimulatory molecules, or by production of immunosuppressive factors and upregulation of immune cell apoptosis inducing molecules [23][24][25][26]. As a consequence, the immune system will “ignore and tolerate” tumor cell proliferation and progression. The presence of tumor-infiltrating lymphocytes (TIL) in cancer cell nests is an independent prognostic factor of survival in various types of cancers including NSCLC [57], and if used properly, could be used to help convert immune cells to fight against cancer. The inefficiency of the immune response against tumor is inversely proportional to tumor growth, being weaker in larger tumors and stronger in smaller tumors [58]. There is a gap in the current knowledge and understanding of the mechanisms behind the immune response against NSCLC, and additional attention to these mechanisms are needed if we are to improve the outcome of patients in the future.

Table 1. Tumor-related immunosuppressive effects.

Tumor-Related Effect

Consequences

Production of immunosuppressive factors and upregulation of immune cell apoptosis inducing molecules [23][24].

Escape from immune surveillance.

Downregulation of HLA and co-stimulatory molecules [25][26].

Escape from immune surveillance.

Tumor-related secretion of soluble molecules VEGF, PGE2, TGF-β, IL-10 [39][40][41].

Immune editing, escape from immune control.

Release of anti-inflammatory mediators such as IL-10 and TGF-β [39][40][41].

Inhibition of dendritic cells and T-cells.

Tumor-associated antigens overexpression in NSCLC [42][43].

Immune system tolerance and less responsiveness to immune checkpoint blockade.

Tumor specific (neo) antigens present on MHC molecules, often downregulated in NSCLC [44][45].

Tumor cells evasion of immune destruction.

Lung cancer cells overexpress the immunosuppressive protein, PD-L1 [46][47].

Inhibitory effects on signaling pathways involved in T-cell activation and cytokine secretion.

Tumor cells mediate a checkpoint/“brake” on T-cell activation and thus anti-tumor immunity, by expressing CTLA-4, a B7 ligand and an inhibitory homolog of CD28 [48][49].

Tumor cells evasion of immune destruction.

PD-1’s arrangement with self-ligand PD-L1, found on lung tumor cells dampens the apoptotic pathway [49][50]

Induction of anergy and T-cell depletion.

Tumor cells expand a local immunosuppressive microenvironment, induce dysfunctional T-cell signaling, and upregulate inhibitory immune checkpoints [51].

Evasion of host immune-mediated surveillance and destruction.

Tumor cells express ligands for PD-1 interacting in that way with surface molecules on CD8+ T-cells; influence the microenvironment via orchestration by cytokines [52].

Apoptosis of CD8+ T-cells; immune tolerance.

Tumor cells do not express many neoantigens, and some of those even if expressed might be low immunogenic eliciting only a mild reaction with low affinity antibodies [53].

Cytotoxic lymphocytes unable to recognize tumor cells, inhibited combined cytotoxic reaction together with T-cells.

Release of soluble amino acids tryptophan and arginine within the tumor microenvironment [54][55].

Inhibition of T-cells and NK function, tumor immune tolerance.

Tumor cells express ectonucleotidases CD73 and CD38 which create adenosine from ATP via ADP-AMP [56].

Induction of immunotolerance in cytotoxic lymphocytes.

Abbreviations: Non-small cell lung cancer (NSCLC), Major Histocompatibility Complex (MHC), Cytotoxic T-Lymphocyte-associated Protein-4 (CTLA-4), Programmed Death-Ligand 1 (PD-L1), Vascular Endothelial Growth Factor (VEGF), Prostaglandin E2 (PGE2), Tumor Growth Factor beta (TGF-β), Interleukin-10 (IL-10), Adenosine triphosphate (ATP), Adenosine diphosphate (ADP), Adenosine monophosphate (AMP).

The currently available data comes from peripheral blood or surgically removed NSCLC tumor tissue, with the latter being limited to less than a third of operable NSCLC patients. The vast majority of the patients have an unresectable disease or are inoperable and undergo radio/chemotherapy, and therefore NSCLC tumor tissue in these advanced stage patients is typically unavailable for detailed immunological analysis. However, based on the limited histological and immunological analysis obtained from the available tissue, the main components of a tumor-directed immune response are represented by a complex interaction between various immune cells operating a composite cytokine network that is supported by the surrounding mesenchymal, epithelial and endothelial cells. The immune cells are a large, highly cooperative family consisting of tumor infiltrating lymphocytes (TILs), the tumor associated macrophages (TAMs), the tumor associated neutrophils (TANs), tissue eosinophilia, and T-cell lymphocytes [59][60][61][62][63][64]. The lung anti-tumor immune response is imitated by activation of the pulmonary antigen presenting cells (APCs), represented by macrophages and dendritic cells [65]. This is a fundamental step towards the beginning of an effective anti-tumor immune response. Following tumor antigen(s) recognition and distinguishing “self” from “non-self”, the APCs migrate to the regional lymph nodes and activate the effector immune cells that aid in destruction of tumor cells. These effector immune cells, also known as cytotoxic lymphocytes, include the CD4+ lymphocytes, natural killers, natural killer T-cells, CD8+ lymphocytes and B lymphocytes [66][67][68]. The activation of these cells are enhanced by the secretion of inflammatory cytokines such as IL-12 and Interferon gamma (IFN-γ). These cytokines are released by the activated macrophages, growing tumor cells, and stromal cells surrounding the tumor. Additionally, membrane-receptor induction of programmed death by the cytotoxic lymphocytes also aids cytokine release and apoptosis of tumor cells [69], as the final coordinate anti-tumor response.

For the purpose of effective antigen presentation, a critical role is played by interaction between co-stimulating molecules on antigen-presenting cells and corresponding receptors on cytotoxic lymphocyte [70]. One mechanisms of immune suppression that tumor cells use, are to block this antigen/cytotoxic lymphocyte interaction and prevent the cytotoxic lymphocytes from getting activated against the tumor (Table 1). Several additional tumor-related factors and mechanisms that result in immune suppression have also been described. One of those involved is alterations in signal transduction molecules on effector T-cells leading to the lack of tumor antigen recognition and missing anti-tumor immune response [71]. In this case, increased tumor-related anti-inflammatory humoral factors like IL-10 or Tumor Growth Factor-beta (TGF-β) induce the loss of signal transducer CD3-ε chain (CD3-ε) in TIL. With that, the signaling pathway for T-cell activation is inhibited, the immune response cannot be initiated and results in immunosuppression.

Alteration of CD3-ε, which is involved in tumor-induced T-cell apoptosis, leads to tumor induced caspase-dependent apoptosis in high proportion of tumor infiltrating T-lymphocytes [72][73]. Further, tumor escapes immune control through the process of immune editing, having as the target the loco-regional tumor microenvironment (TME). Several different tumor-related soluble molecules are involved in this form of immunosuppression: Vascular Endothelial Growth Factor (VEGF), Prostaglandin E2, TGF-β, IL-10, soluble phosphatidylserine, MICA Fas and FasL100 [27][28][29][30][31][32][33][34]. Their immunosuppressive effects include inactivation of dendritic cells and T-cells, inhibition of Fas-mediated and NKG2D-mediated killing of immune cells, and release of anti-inflammatory mediators such as IL-10 and TGF-β that inhibit dendritic cells and T-cells [35][36][37][38]. All those effects promote metastatic spread and progression in NSCLC patients [74][75].

Finally, the stromal cells from the TME also exhibit an important immunosuppressive role through modulation and binding of tumor antigens. By binding tumor antigens, these cells compete with the antigen-presenting cells so that many tumor antigens will be down-regulated, resulting in immunosuppression and tumor progression [76][77][78]. By increasing interstitial fluid pressure in the tumor, stromal cells will make significant quantity of tumor antigens to be unavailable and therefore, ignored by T-cells [79]. Besides the tumor and the TME causing immune-suppression, therapies such as convention RT and chemotherapy can also lead to additional immune suppression.

References

  1. Liu, X.; Wu, S.; Yang, Y.; Zhao, M.; Zhu, G.; Hou, Z. The prognostic landscape of tumor-infiltrating immune cell and immunomodula-tors in lung cancer. Pharmacother. 2017, 95, 55–61.
  2. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA Cancer, J. Clin. 2019, 69, 7–34, doi:10.3322/caac.21551.
  3. Abedi, S.; Janbabaei, G.; Afshari, M.; Moosazadeh, M.; Alashti, M.R.; Hedayatizadeh-Omran, A.; Alizadeh-Navaei, R.; Abedini, E. Estimating the Survival of Patients With Lung Cancer: What Is the Best Statistical Model? Prev. Med. Public Health 2019, 52, 140–144, doi:10.3961/jpmph.17.090.
  4. Doria-Rose, V.P.; Marcus, P.M.; Miller, A.B.; Bergstralh, E.J.; Mandel, J.S.; Tockman, M.S.; Prorok, P.C.; Doria-Rose, V. Does the source of death information affect cancer screening efficacy results? A study of the use of mortality review versus death certificates in four randomized trials. Trials 2010, 7, 69–77, doi:10.1177/1740774509356461.
  5. Collins, L.G.; Haines, C.; Perkel, R.; Enck R.E. Lung cancer: Diagnosis and management. Fam. Physician 2007, 75, 56–63.
  6. O’Byrne, K.J.; Dalgleish, A.G. Chronic immune activation and inflammation as the cause of malignancy. J. Cancer 2001, 85, 473–483, doi:10.1054/bjoc.2001.1943.
  7. Mayne, S.T.; Buenconsejo, J.; Janerich, D.T. Previous lung disease and risk oflung cancer among men and women nonsmokers. J. Epidemiol. 1999, 149, 13–20.
  8. Raza, A. Consilience across evolving dysplasias affecting myeloid, cervical,esophageal, gastric and liver cells: Common themes and emerging patterns. Res. 2000, 24, 63–72.
  9. Vogelstein, B.; Fearon, E.R.; Hamilton, S.R.; Kern, S.E.; Preisinger, A.C.; Leppert, M.; Nakamura, Y.; White, R.; Smits, A.M.; Bos, J.L. Genetic alterations duringcolorectal-tumor development. Engl. J. Med. 1988, 319, 525–532.
  10. Lengauer, C.; Kinzler, K.W.; Vogelstein, B. Genetic instabilities in humancancers. Nature 1998, 396, 643–649.
  11. Spitz, M.R.; Wei, Q.; Li, G.; Wu, X. Genetic susceptibility to tobaccocarcinogenesis. Cancer Invest. 1999, 17, 645–659.
  12. Taketo, M.M. Cyclooxygenase-2 Inhibitors in Tumorigenesis (Part II). Natl. Cancer Inst. 1998, 90, 1609–1620, doi:10.1093/jnci/90.21.1609.
  13. Vane, J.R.; Bakhle, Y.S.; Botting, R.M. Cyclooxygenases 1 and 2. Rev. Pharmacol. Toxicol. 1998, 38, 97–120.
  14. Della Bella, S.; Molteni, M.; Compasso, S.; Zulian, C.; Vanoli, M.; Scorza, R. Differential effects of cy-clo-oxygenase pathway metabolites on cytokine production by T lymphocytes. Prostaglandins Leukot. Essent. Fatty Acids 1997, 56, 177–184.
  15. Subbaramaiah, K.; Zakim, D.; Weksler, B.B.; Dannenberg, A.J. Inhibition ofcyclooxygenase: A novel ap-proach to cancer prevention. Soc. Exp. Biol. Med. 1997, 216, 201–210.
  16. Gjertsen, M.K.; Bjorheim, J.; Saeterdal, I.; Myklebust, J.; Gaudernack, G. Cytotoxic CD4+ and CD8+ T lym-phocytes, generated by mutant p21-ras(12Val) peptide vaccination of a patient, recognize 12Val-dependent nestedepitopes present within the vaccine peptide and kill autologous tumour cellscarrying this mutation. J. Cancer 1997, 72, 784–790.
  17. Tsujii, M.; Kawano, S.; Tsuji, S.; Sawaoka, H.; Hori, M.; Dubois, R.N. Cyclooxygenase Regulates Angiogenesis Induced by Colon Cancer Cells. Cell 1998, 93, 705–716, doi:10.1016/s0092-8674(00)81433-6.
  18. Takahashi, Y.; Kawahara, F.; Noguchi, M.; Miwa, K.; Sato, H.; Seiki, M.; Inoue, H.; Tanabe, T.; Yoshimoto, T. Ac-tivation of matrix metalloproteinase-2 in human breast cancer cells overexpressing cyclooxygenase-1 or -2. FEBS Lett. 1999, 460, 145–148.
  19. Wirsdörfer, F.; Jendrossek, V. The Role of Lymphocytes in Radiotherapy-Induced Adverse Late Effects in the Lung. Immunol. 2016, 7, doi:10.3389/fimmu.2016.00591.
  20. Grossman, S.A.; Ellsworth, S.; Campian, J.; Wild, A.T.; Herman, J.M.; Laheru, D.; Brock, M.; Balmanoukian, A.; Ye, X. Survival in Patients With Severe Lymphopenia Following Treatment With Radiation and Chemotherapy for Newly Diagnosed Solid Tumors. Natl. Compr. Cancer Netw. 2015, 13, 1225–1231, doi:10.6004/jnccn.2015.0151.
  21. Campian, J.L.; Ye, X.; Brock, M.; Grossman S.A. Treatment-related Lymphopenia in patients with stage III non-small-cell lung Cancer. Cancer Investig. 2013, 31, 183–138.
  22. Tang, C.; Liao, Z.; Gomez, D.; Levy L.; Zhuang Y.; Gebremichael R.A.; Hong D.S.; Komaki R.; Welsh J.W. Lymphopenia association with gross tumor volume and lung V5 and its effects on non-small cell lung Cancer patientoutcomes. J. Radiat. Oncol. Biol. Phys. 2014, 89, 1084–1091.
  23. Wang, Y.; Hays, E.; Rama, M.; Bonavida, B. Cell-mediated immune resistance in cancer. Cancer Drug Resist. 2019, 3, 232–251, doi:10.20517/cdr.2019.98.
  24. Gadiyar, V.; Lahey, K.C.; Calianese, D.; DeVoe, C.; Mehta, D.; Bono, K.; Desind, S.; Davra, V.; Birge, R.B. Cell Death in the Tumor Microenvironment: Implications for Cancer Immunotherapy. Cells 2020, 9, 2207, doi:10.3390/cells9102207.
  25. Couture, A.; Garnier, A.; Docagne, F.; Boyer, O.; Vivien, D.; Le-Mauff, B.; Latouche, J.-B.; Toutirais, O. HLA-Class II Artificial Antigen Presenting Cells in CD4+ T Cell-Based Immunotherapy. Immunol. 2019, 10, doi:10.3389/fimmu.2019.01081.
  26. Sabbatino, F.; Liguori, L.; Polcaro, G.; Salvato, I.; Caramori, G.; Salzano, F.A.; Casolaro, V.; Stellato, C.; Col, J.D.; Pepe, S. Role of Human Leukocyte Antigen System as A Predictive Biomarker for Checkpoint-Based Immunotherapy in Cancer Patients. J. Mol. Sci. 2020, 21, 7295, doi:10.3390/ijms21197295.
  27. Gabrilovich, D.; Ishida, T.; Oyama, T.; Ran, S.; Kravtsov, V.; Nadaf, S.; Carbone, D.P. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood 1998, 92, 4150–4166.
  28. Urosevic, M.; Dummer, R. HLA-G and IL-10 expression in human cancer—different stories with the same mes-sage. Cancer Biol. 2003, 13, 337–342.
  29. Beck, C.; Schreiber, H.; Rowley, D. Role of TGF-beta in immune evasion of cancer. Res. Tech. 2001, 52, 387–395.
  30. CHe, X.; Stuart, J.M. Prostaglandin E2 selectively inhibits human CD4+ T cells secreting low amounts of both IL-2 and IL-4. Immunol. 1999, 163, 6173–6179.
  31. Erdogan, B.; Uzaslan, E.; Budak, F.; Karadağ M.; Ediger D.; Oral B.; Göral G.; Ege E.; Gözü O. The evaluation of soluble Fas and soluble Fas ligand levels of bronchoalveolar lavage fluid in lung cancer patients. Toraks 2005, 53, 127–131.
  32. Kim, R.; Emi, M.; Tanabe, K.; Uchida, Y.; Toge, T. The role of Fas ligand and transforming growth factor beta in tumor progression: Molecular mechanisms of immune privilege via Fas-mediated apoptosis and potential targets for cancer therapy. Cancer 2004, 100, 2281–2291.
  33. Holdenrieder, S.; Stieber, P.; Peterfi, A.; Nagel, D.; Steinle, A.; Salih, H.R. Soluble MICB in malignant diseases: Analysis of diagnostic significance and correlation with soluble MICA. Cancer Immunol. Immunother. 2006, 55, 1584–1589, doi:10.1007/s00262-006-0167-1.
  34. Kim, R.; Emi, M.; Tanabe, K. Cancer cell immune escape and tumor progression by exploitation of an-ti-inflammatory and pro-inflammatory responses. Cancer Biol. Ther. 2005, 4, 924–933.
  35. Wang, T.; Niu, G.; Kortylewski, M.; Burdelya L.; Shain K.; Zhang S.; Bhattacharya R.; Gabrilovich D.; Heller R.; Coppola D.; et al. Regulation of the innate and adaptive immune responses by Stat-3 signal-ing in tumor cells. Med. 2004, 10, 48–54.
  36. Kortylewski, M.; Kujawski, M.; Wang, T.; Wei S.; Zhang S.; Pilon-Thomas S.; Niu G.; Kay H.; Mulé J.; Kerr W.G.; et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multi-component antitumor immunity. Med. 2005, 11, 1314–1121.
  37. Webb, S.D.; Sherratt, J.A.; Fish, R.G. Cells behaving badly: A theoretical model for the Fas/FasL system in tumour immunology. Biosci. 2002, 179, 113–129, doi:10.1016/s0025-5564(02)00120-7.
  38. Doubrovina, E.S.; Doubrovin, M.M.; Vider, E.; Sisson, R.B.; O’Reilly, R.J.; Dupont, B.; Vyas, Y.M. Evasion from NK Cell Immunity by MHC Class I Chain-Related Molecules Expressing Colon Adenocarcinoma. Immunol. 2003, 171, 6891–6899, doi:10.4049/jimmunol.171.12.6891.
  39. Shrihari, T.G. Dual role of inflammatory mediators in cancer. Ecancermedicalscience 2017, 11, 721, doi:10.3332/ecancer.2017.721.
  40. Finetti, F.; Travelli, C.; Ercoli, J.; Colombo, G.; Buoso, E.; Trabalzini, L. Prostaglandin E2 and Cancer: Insight into Tumor Progression and Immunity. Biology 2020, 9, 434, doi:10.3390/biology9120434.
  41. Thepmalee, C.; Panya, A.; Junking, M.; Chieochansin T.; Yenchitsomanus PT. Inhibition of IL-10 and TGF-β receptors on dendritic cells enhances activation of effector T-cells to kill cholangiocarcinoma cells. Vaccin. Immunother. 2018, 14, 1423–1431, doi:10.1080/21645515.2018.1431598.42.
  42. Yasumoto, K.; Hanagiri, T.; Takenoyama, M. Lung cancer-associated tumor antigens and the present status of immunotherapy against non-small-cell lung cancer. Genes Thorac Cardiovasc. Surg. 2009, 57, 449–457, doi:10.1007/s11748-008-0433-6.
  43. Smith, C.C.; Selitsky, S.R.; Chai, S.; Armistead, P.M.; Vincent, B.G.; Serody, J.S. Alternative tumour-specific antigens. Rev. Cancer 2019, 19, 465–478, doi:10.1038/s41568-019-0162-4.
  44. Cornel, A.M.; Mimpen, I.L.; Nierkens, S. MHC Class I Downregulation in Cancer: Underlying Mechanisms and Potential Targets for Cancer Immunotherapy. Cancers 2020, 12, 1760, doi:10.3390/cancers12071760.
  45. Berghmans, T.; Dingemans, A.-M.; Hendriks, L.E.; Cadranel, J. Immunotherapy for nonsmall cell lung cancer: A new therapeutic algorithm. Respir. J. 2020, 55, 1901907, doi:10.1183/13993003.01907-2019.
  46. Azuma, K.; Ota, K.; Kawahara, A.; Hattori, S.; Iwama, E.; Harada, T.; Matsumoto, K.; Takayama, K.; Takamori, S.; Kage, M.; et al. Association of PD-L1 overexpression with activating EGFR mutations in surgically resected nonsmall-cell lung cancer. Oncol. 2014, 25, 1935–1940, doi:10.1093/annonc/mdu242.
  47. Meyers, D.; Bryan, P.; Banerji, S.; Morris, D.G. Targeting the PD-1/PD-L1 axis for the treatment of non-small-cell lung cancer. Oncol. 2018, 25, e324–e334, doi:10.3747/co.25.3976.
  48. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674, doi:10.1016/j.cell.2011.02.013.
  49. Sznol, M.; Chen, L. Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced human can-cer. Cancer Res. 2013, 19, 1021–1034.
  50. Patsoukis, N.; Wang, Q.; Strauss, L.; Boussiotis, V.A. Revisiting the PD-1 pathway. Adv. 2020, 6, eabd2712, doi:10.1126/sciadv.abd2712.
  51. Anderson, K.G.; Stromnes, I.M.; Greenberg, P.D. Obstacles Posed by the Tumor Microenvironment to T cell Activity: A Case for Synergistic Therapies. Cancer Cell 2017, 31, 311–325, doi:10.1016/j.ccell.2017.02.008.
  52. Lanitis, E.; Dangaj, D.; Irving, M.; Coukos, G. Mechanisms regulating T-cell infiltration and activity in solid tumors. Oncol. 2017, 28, xii18–xii32, doi:10.1093/annonc/mdx238.
  53. Yarchoan, M.; Johnson, B.A.; Lutz, E.R.; Laheru, D.A.; Jaffee, M.Y. Targeting neoantigens to augment antitumour immunity. Rev. Cancer 2017, 17, 209–222, doi:10.1038/nrc.2016.154.
  54. Lieu, E.L.; Nguyen, T.; Rhyne, S.; Kim, J. Amino acids in cancer. Mol. Med. 2020, 52, 15–30, doi:10.1038/s12276-020-0375-3.
  55. Timosenko, E.; Hadjinicolaou, A.V.; Cerundolo, V. Modulation of cancer-specific immune responses by amino acid degrading enzymes. Immunotherapy 2017, 9, 83–97.
  56. Allard, B.; Longhi, M.S.; Robson, S.C.; Stagg, J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Rev. 2017, 276, 121–144, doi:10.1111/imr.12528.
  57. Villegas, F.R.; Coca, S.; Villarrubia, V.G.; Jiménez, R.; Chillón, M.J.; Jareño, J.; Zuil, M.; Callol, L. Prognostic significance of tumor infiltrating natural killer cells subset CD57 in patients with squamous cell lung cancer. Lung Cancer 2002, 35, 23–28, doi:10.1016/s0169-5002(01)00292-6.
  58. Tartour, E.; Zitvogel, L. Lung cancer: Potential targets for immunotherapy. Lancet Respir. Med. 2013, 1, 551–563, doi:10.1016/s2213-2600(13)70159-0.
  59. Yoshino, I.; Yano, T.; Murata, M.; Miyamoto, M.; Ishida, T.; Sugimachi, K.; Kimura, G.; Nomoto, K. Phenotypes of lymphocytes infiltrating non-small cell lung cancer tissues and its variation with histological types of cancer. Lung Cancer 1993, 10, 13–19, doi:10.1016/0169-5002(93)90305-h.
  60. Al-Shibli, K.I.; Donnem, T.; Al-Saad, S.; Persson, M.; Bremnes, R.M.; Busund, L.-T. Prognostic Effect of Epithelial and Stromal Lymphocyte Infiltration in Non–Small Cell Lung Cancer. Cancer Res. 2008, 14, 5220–5227, doi:10.1158/1078-0432.ccr-08-0133.
  61. Allavena, P.; Mantovani, A. Immunology in the clinic review series; focus on cancer: Tumour-associated macro-phages: Undisputed stars of the inflammatory tumour microenvironment. Exp. Immunol. 2012, 167, 195–205.
  62. Través, P.G.; Luque, A.; Hortelano, S.; Macrophages, Inflammation, and Tumor Suppressors: ARF, a New Player in the Game. Inflamm. 2012, 2012, 1–11, doi:10.1155/2012/568783.
  63. Fridlender, Z.G.; Albelda, S.M. Tumor-associated neutrophils: Friend or foe? Carcinogenesis 2012, 33, 949–955, doi:10.1093/carcin/bgs123.
  64. Gatault, S.; Legrand, F.; Delbeke, M.; Loiseau, S.; Capron, M. Involvement of eosinophils in the anti-tumor response. Cancer Immunol. Immunother. 2012, 61, 1527–1534, doi:10.1007/s00262-012-1288-3.
  65. Vermaelen, K.; Pauwels, R. Pulmonary dendritic cells. J. Respir Crit. Care Med. 2005, 172, 530–551.
  66. Aerts, J.; Hegmans, J. Tumor-Specific Cytotoxic T Cells Are Crucial for Efficacy of Immunomodulatory Antibodies in Patients with Lung Cancer. Cancer Res. 2013, 73, 2381–2388, doi:10.1158/0008-5472.can-12-3932.
  67. Salagianni, M.; Baxevanis, C.N.; Papamichail, M.; Perez S.A. New insights into the role of NK cells in cancer immuno-therapy. Oncoimmunology 2012, 1, 205-207.
  68. Motohashi, S.; Okamoto, Y.; Yoshino, I.; Nakayama T. Anti-tumor immune responses induced by iNKT cell-based immuno-therapy for lung cancer and head and neck cancer. Clin. Immunol. 2011, 140, 167–176.
  69. Martínez-Lostao, L.; Anel, A.; Pardo, J. How Do Cytotoxic Lymphocytes Kill Cancer Cells? Cancer Res. 2015, 21, 5047–5056, doi:10.1158/1078-0432.ccr-15-0685.
  70. Domagala-Kulawik J, Osinska I, Hoser G. Mechanisms of immune response regulation in lung cancer. Transl Lung Cancer Res. 2014 Feb;3(1):15-22. doi: 10.3978/j.issn.2218-6751.2013.11.03. PMID: 25806277; PMCID: PMC4367608.
  71. von Bernstorff, W.; Voss, M.; Freichel, S.; Schmid A.; Vogel I.; Jöhnk C.; Henne-Bruns D.; Kremer B.; Kalthoff H. Systemic and local immunosuppression in pancreatic cancer patients. Cancer Res. 2001, 7, 925s–932s.
  72. Gastman, B.R.; E.; Johnson, D.; Whiteside, T.L.; Rabinowich, H. Caspase-mediated degradation of T-cell receptor zeta-chain. Cancer Res. 1999, 59, 1422–1427.
  73. Gastman, B.R.; Johnson, D.E.; Whiteside, T.L.; Rabinowich, H. Tumor-induced apoptosis of T lymphocytes: Elucidation of intracellular apoptotic events. Blood 2000, 95, 2015–2023, doi:10.1182/blood.v95.6.2015.
  74. Suzuki, M.; Iizasa, T.; Ko, E.; Baba, M.; Saitoh, Y.; Shibuya, K.; Sekine, Y.; Yoshida, S.; Hiroshima, K.; Fujisawa, T. Serum endostatin correlates with progression and prognosis of non-small cell lung cancer. Lung Cancer 2002, 35, 29–34, doi:10.1016/s0169-5002(01)00285-9.
  75. Kim, R.; Emi, M.; Tanabe, K.; Arihiro, K. Tumor-Driven Evolution of Immunosuppressive Networks during Malignant Progression. Cancer Res. 2006, 66, 5527–5536, doi:10.1158/0008-5472.can-05-4128.
  76. Juprelle-Soret, M.; Coninck, S.W.-D.; Wattiaux, R. Subcellular localization of transglutaminase. Effect of collagen. J. 1988, 250, 421–427, doi:10.1042/bj2500421.
  77. Savinov, A.Y.; Wong, F.S.; Stonebraker, A.C.; Chervonsky, A.V. Presentation of antigen by endothelial cells and chem-oattraction are required for homing of insulin-specific CD8+ T cells. Exp. Med. 2003, 97, 643–656.
  78. Spiotto, M.T.; Yu, P.; Rowley, D.A.; Nishimura, M.I.; Meredith, S.C.; Gajewski, T.F.; Fu, Y.-X.; Schreiber, H. Increasing Tumor Antigen Expression Overcomes “Ignorance” to Solid Tumors via Crosspresentation by Bone Marrow-Derived Stromal Cells. Immunity 2002, 17, 737–747, doi:10.1016/s1074-7613(02)00480-6.
  79. Pietras, K.; Ostman, A.; Sjöquist, M.; Buchdunger, E.; Reed, R.K.; Heldin, C.H.; Rubin, K. Inhibition of platelet-derived growth factor receptors reduces interstitial hypertension and increases transcapillary transport in tumors. Cancer Res. 2001, 61, 2929–2934.
More
Information
Subjects: Cell Biology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 356
Revisions: 2 times (View History)
Update Date: 26 Feb 2021
1000/1000
Video Production Service