Skip to main content
Erschienen in: Clinical and Translational Oncology 8/2018

08.01.2018 | Review Article

Perspective on immune oncology with liquid biopsy, peripheral blood mononuclear cells, and microbiome with non-invasive biomarkers in cancer patients

verfasst von: A. Mitsuhashi, Y. Okuma

Erschienen in: Clinical and Translational Oncology | Ausgabe 8/2018

Einloggen, um Zugang zu erhalten

Abstract

Antibodies against immune checkpoint inhibitors such as anti-programmed cell death protein 1 (PD-1) and anti-programmed death ligand 1 (PD-L1) play a key role in the treatment of advanced lung cancer. To examine the clinical benefits of these agents, preclinical and clinical studies have been conducted to identify definitive biomarkers associated with cancer status. Analysis of the blood and feces of tumor patients has attracted attention in recent studies attempting to identify non-invasive biomarkers such as cytokines, soluble PD-L1, peripheral blood mononuclear cells, and gut microbiota. These factors are believed to interact with each other to produce synergistic effects and contribute to the formation of the tumor immune microenvironment through the seven steps of the cancer immunity cycle. The immunogram was first introduced as a novel indicator to define the immunity status of cancer patients. In this review, we discuss the progress in the identification of predictive biomarkers as well as future prospects for anti-PD-1/PD-L1 therapy.
Literatur
1.
Zurück zum Zitat Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol. 2007;19:813–24.CrossRefPubMed Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol. 2007;19:813–24.CrossRefPubMed
3.
Zurück zum Zitat Kloor M. Lymphocyte infiltration and prognosis in colorectal cancer. Lancet Oncol. 2009;10:840–1.CrossRefPubMed Kloor M. Lymphocyte infiltration and prognosis in colorectal cancer. Lancet Oncol. 2009;10:840–1.CrossRefPubMed
4.
Zurück zum Zitat Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8:239–45.CrossRefPubMed Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8:239–45.CrossRefPubMed
5.
Zurück zum Zitat Guan J, Lim KS, Mekhail T, Chang CC. Programmed death ligand-1 (PD-L1) expression in the programmed death receptor-1 (PD-1)/PD-L1 blockade: a key player against various cancers. Arch Pathol Lab Med. 2017;141:851–61.CrossRefPubMed Guan J, Lim KS, Mekhail T, Chang CC. Programmed death ligand-1 (PD-L1) expression in the programmed death receptor-1 (PD-1)/PD-L1 blockade: a key player against various cancers. Arch Pathol Lab Med. 2017;141:851–61.CrossRefPubMed
6.
Zurück zum Zitat Mu CY, Huang JA, Chen Y, Chen C, Zhang XG. High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor-infiltrating dendritic cells maturation. Med Oncol. 2011;28:682–8.CrossRefPubMed Mu CY, Huang JA, Chen Y, Chen C, Zhang XG. High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor-infiltrating dendritic cells maturation. Med Oncol. 2011;28:682–8.CrossRefPubMed
7.
Zurück zum Zitat Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14:847–56.CrossRefPubMed Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14:847–56.CrossRefPubMed
8.
Zurück zum Zitat Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti–PD–1 antibody in cancer. N Engl J Med. 2012;366:2443–54.CrossRefPubMedPubMedCentral Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti–PD–1 antibody in cancer. N Engl J Med. 2012;366:2443–54.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, et al. Phase I study of single–agent anti-programmed death–1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28:3167–75.CrossRefPubMedPubMedCentral Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, et al. Phase I study of single–agent anti-programmed death–1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28:3167–75.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369:134–44.CrossRefPubMedPubMedCentral Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369:134–44.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Callahan MK, Postow MA, Wolchok JD. Targeting T cell co-receptors for cancer therapy. Immunity. 2016;44:1069–78.CrossRefPubMed Callahan MK, Postow MA, Wolchok JD. Targeting T cell co-receptors for cancer therapy. Immunity. 2016;44:1069–78.CrossRefPubMed
12.
Zurück zum Zitat Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. Pembrolizumab versus chemotherapy for pd–l1–positive non-small-cell lung cancer. N Engl J Med. 2016;375:1823–33.CrossRefPubMed Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. Pembrolizumab versus chemotherapy for pd–l1–positive non-small-cell lung cancer. N Engl J Med. 2016;375:1823–33.CrossRefPubMed
13.
Zurück zum Zitat Rimm DL, Han G, Taube JM, Yi ES, Bridge JA, Flieder DB, et al. A prospective, multi-institutional, pathologist-based assessment of 4 immunohistochemistry assays for pd-l1 expression in non-small cell lung cancer. JAMA Oncol. 2017;3:1051–8.CrossRefPubMedPubMedCentral Rimm DL, Han G, Taube JM, Yi ES, Bridge JA, Flieder DB, et al. A prospective, multi-institutional, pathologist-based assessment of 4 immunohistochemistry assays for pd-l1 expression in non-small cell lung cancer. JAMA Oncol. 2017;3:1051–8.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–8.CrossRefPubMedPubMedCentral Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–8.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Chen PL, Roh W, Reuben A, Cooper ZA, Spencer CN, Prieto PA, et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 2016;6:827–37.CrossRefPubMedPubMedCentral Chen PL, Roh W, Reuben A, Cooper ZA, Spencer CN, Prieto PA, et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 2016;6:827–37.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 2017;376:2415–26.CrossRefPubMed Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 2017;376:2415–26.CrossRefPubMed
18.
Zurück zum Zitat Yamazaki N, Kiyohara Y, Uhara H, Iizuka H, Uehara J, Otsuka F, et al. Cytokine biomarkers to predict antitumor responses to nivolumab suggested in a phase 2 study for advanced melanoma. Cancer Sci. 2017;108:1022–31.CrossRefPubMedPubMedCentral Yamazaki N, Kiyohara Y, Uhara H, Iizuka H, Uehara J, Otsuka F, et al. Cytokine biomarkers to predict antitumor responses to nivolumab suggested in a phase 2 study for advanced melanoma. Cancer Sci. 2017;108:1022–31.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Sanmamed MF, Perez-Gracia JL, Schalper KA, Fusco JP, Gonzalez A, Rodriguez-Ruiz ME, et al. Changes in serum interleukin-8 (IL–8) levels reflect and predict response to anti-PD-1 treatment in melanoma and non-small cell lung cancer patients. Ann Oncol. 2017;28:1988–95.CrossRefPubMedPubMedCentral Sanmamed MF, Perez-Gracia JL, Schalper KA, Fusco JP, Gonzalez A, Rodriguez-Ruiz ME, et al. Changes in serum interleukin-8 (IL–8) levels reflect and predict response to anti-PD-1 treatment in melanoma and non-small cell lung cancer patients. Ann Oncol. 2017;28:1988–95.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Akiyama Y, Nonomura C, Kondou R, Miyata H, Ashizawa T, Maeda C, et al. Immunological effects of the anti-programmed death-1 antibody on human peripheral blood mononuclear cells. Int J Oncol. 2016;49:1099–107.CrossRefPubMed Akiyama Y, Nonomura C, Kondou R, Miyata H, Ashizawa T, Maeda C, et al. Immunological effects of the anti-programmed death-1 antibody on human peripheral blood mononuclear cells. Int J Oncol. 2016;49:1099–107.CrossRefPubMed
21.
Zurück zum Zitat Akyüz N, Brandt A, Stein A, Schliffke S, Mährle T, Quidde J, et al. T–cell diversification reflects antigen selection in the blood of patients on immune checkpoint inhibition and may be exploited as liquid biopsy biomarker. Int J Cancer. 2017;140:2535–44.CrossRefPubMed Akyüz N, Brandt A, Stein A, Schliffke S, Mährle T, Quidde J, et al. T–cell diversification reflects antigen selection in the blood of patients on immune checkpoint inhibition and may be exploited as liquid biopsy biomarker. Int J Cancer. 2017;140:2535–44.CrossRefPubMed
22.
Zurück zum Zitat Botticelli A, Zizzari I, Mazzuca F, Ascierto PA, Putignani L, Marchetti L, et al. Cross-talk between microbiota and immune fitness to steer and control response to anti PD-1/PDL-1 treatment. Oncotarget. 2017;8:8890–9.PubMed Botticelli A, Zizzari I, Mazzuca F, Ascierto PA, Putignani L, Marchetti L, et al. Cross-talk between microbiota and immune fitness to steer and control response to anti PD-1/PDL-1 treatment. Oncotarget. 2017;8:8890–9.PubMed
23.
Zurück zum Zitat Xu LH, Chi XY, Li FY, Jia QT, Zha QB, He XH. Preparation and identification of human soluble sPD-L1 and its antibodies. Sheng Wu Gong Cheng Xue Bao. 2007;23:106–11.CrossRefPubMed Xu LH, Chi XY, Li FY, Jia QT, Zha QB, He XH. Preparation and identification of human soluble sPD-L1 and its antibodies. Sheng Wu Gong Cheng Xue Bao. 2007;23:106–11.CrossRefPubMed
24.
Zurück zum Zitat Okuma Y, Hosomi Y, Nakahara Y, Watanabe K, Sagawa Y, Homma S. High plasma levels of soluble programmed cell death ligand 1 are prognostic for reduced survival in advanced lung cancer. Lung Cancer. 2017;104:1–6.CrossRefPubMed Okuma Y, Hosomi Y, Nakahara Y, Watanabe K, Sagawa Y, Homma S. High plasma levels of soluble programmed cell death ligand 1 are prognostic for reduced survival in advanced lung cancer. Lung Cancer. 2017;104:1–6.CrossRefPubMed
25.
Zurück zum Zitat Rossille D, Gressier M, Damotte D, Maucort-Boulch D, Pangault C, Semana G, et al. High level of soluble programmed cell death ligand 1 in blood impacts overall survival in aggressive diffuse large B-Cell lymphoma: results from a French multicenter clinical trial. Leukemia. 2014;28:2367–75.CrossRefPubMed Rossille D, Gressier M, Damotte D, Maucort-Boulch D, Pangault C, Semana G, et al. High level of soluble programmed cell death ligand 1 in blood impacts overall survival in aggressive diffuse large B-Cell lymphoma: results from a French multicenter clinical trial. Leukemia. 2014;28:2367–75.CrossRefPubMed
26.
Zurück zum Zitat Finkelmeier F, Canli Ö, Tal A, Pleli T, Trojan J, Schmidt M, et al. High levels of the soluble programmed death–ligand (sPD–L1) identify hepatocellular carcinoma patients with a poor prognosis. Eur J Cancer. 2016;59:152–9.CrossRefPubMed Finkelmeier F, Canli Ö, Tal A, Pleli T, Trojan J, Schmidt M, et al. High levels of the soluble programmed death–ligand (sPD–L1) identify hepatocellular carcinoma patients with a poor prognosis. Eur J Cancer. 2016;59:152–9.CrossRefPubMed
27.
Zurück zum Zitat Takahashi N, Iwasa S, Sasaki Y, Shoji H, Honma Y, Takashima A, et al. Serum levels of soluble programmed cell death ligand 1 as a prognostic factor on the first–line treatment of metastatic or recurrent gastric cancer. J Cancer Res Clin Oncol. 2016;142:1727–38.CrossRefPubMed Takahashi N, Iwasa S, Sasaki Y, Shoji H, Honma Y, Takashima A, et al. Serum levels of soluble programmed cell death ligand 1 as a prognostic factor on the first–line treatment of metastatic or recurrent gastric cancer. J Cancer Res Clin Oncol. 2016;142:1727–38.CrossRefPubMed
28.
Zurück zum Zitat Zhou J, Mahoney KM, Giobbie-Hurder A, Zhao F, Lee S, Liao X, et al. Soluble PD–L1 as a biomarker in malignant melanoma treated with checkpoint blockade. Cancer Immunol Res. 2017;5:480–92.CrossRefPubMedPubMedCentral Zhou J, Mahoney KM, Giobbie-Hurder A, Zhao F, Lee S, Liao X, et al. Soluble PD–L1 as a biomarker in malignant melanoma treated with checkpoint blockade. Cancer Immunol Res. 2017;5:480–92.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Ruf M, Moch H, Schraml P. PD-L1 expression is regulated by hypoxia inducible factor in clear cell renal cell carcinoma. Int J Cancer. 2016;139:396–403.CrossRefPubMed Ruf M, Moch H, Schraml P. PD-L1 expression is regulated by hypoxia inducible factor in clear cell renal cell carcinoma. Int J Cancer. 2016;139:396–403.CrossRefPubMed
30.
Zurück zum Zitat Frigola X, Inman BA, Krco CJ, Liu X, Harrington SM, Bulur PA, et al. Soluble B7-H1: differences in production between dendritic cells and T cells. Immunol Lett. 2012;142(1–2):78–82.CrossRefPubMed Frigola X, Inman BA, Krco CJ, Liu X, Harrington SM, Bulur PA, et al. Soluble B7-H1: differences in production between dendritic cells and T cells. Immunol Lett. 2012;142(1–2):78–82.CrossRefPubMed
31.
Zurück zum Zitat Davies LC, Heldring N, Kadri N, Le Blanc K. Mesenchymal stromal cell secretion of programmed death–1 ligands regulates t cell mediated immunosuppression. Stem Cells. 2017;35:766–76.CrossRefPubMed Davies LC, Heldring N, Kadri N, Le Blanc K. Mesenchymal stromal cell secretion of programmed death–1 ligands regulates t cell mediated immunosuppression. Stem Cells. 2017;35:766–76.CrossRefPubMed
32.
Zurück zum Zitat Dezutter-Dambuyant C, Durand I, Alberti L, Bendriss-Vermare N, Valladeau-Guilemond J, Duc A, et al. A novel regulation of PD-1 ligands on mesenchymal stromal cells through MMP-mediated proteolytic cleavage. Oncoimmunology. 2015;5:e1091146.CrossRefPubMedPubMedCentral Dezutter-Dambuyant C, Durand I, Alberti L, Bendriss-Vermare N, Valladeau-Guilemond J, Duc A, et al. A novel regulation of PD-1 ligands on mesenchymal stromal cells through MMP-mediated proteolytic cleavage. Oncoimmunology. 2015;5:e1091146.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Weide B, Martens A, Hassel JC, Berking C, Postow MA, Bisschop K, et al. Baseline biomarkers for outcome of melanoma patients treated with pembrolizumab. Clin Cancer Res. 2016;22:5487–96.CrossRefPubMedPubMedCentral Weide B, Martens A, Hassel JC, Berking C, Postow MA, Bisschop K, et al. Baseline biomarkers for outcome of melanoma patients treated with pembrolizumab. Clin Cancer Res. 2016;22:5487–96.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Nakamura Y, Kitano S, Takahashi A, Tsutsumida A, Namikawa K, Tanese K, et al. Nivolumab for advanced melanoma: pretreatment prognostic factors and early outcome markers during therapy. Oncotarget. 2016;7:77404–15.PubMedPubMedCentral Nakamura Y, Kitano S, Takahashi A, Tsutsumida A, Namikawa K, Tanese K, et al. Nivolumab for advanced melanoma: pretreatment prognostic factors and early outcome markers during therapy. Oncotarget. 2016;7:77404–15.PubMedPubMedCentral
35.
Zurück zum Zitat McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351:1463–9.CrossRefPubMedPubMedCentral McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351:1463–9.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Gros A, Parkhurst MR, Tran E, Pasetto A, Robbins PF, Ilyas S, et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat Med. 2016;22:433–8.CrossRefPubMed Gros A, Parkhurst MR, Tran E, Pasetto A, Robbins PF, Ilyas S, et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat Med. 2016;22:433–8.CrossRefPubMed
37.
Zurück zum Zitat Kamphorst AO, Pillai RN, Yang S, Nasti TH, Akondy RS, Wieland A, et al. Proliferation of PD-1 + CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A. 2017;114:4993–8.CrossRefPubMedPubMedCentral Kamphorst AO, Pillai RN, Yang S, Nasti TH, Akondy RS, Wieland A, et al. Proliferation of PD-1 + CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A. 2017;114:4993–8.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science. 2017;355:1428–33.CrossRefPubMed Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science. 2017;355:1428–33.CrossRefPubMed
39.
Zurück zum Zitat Kamphorst AO, Wieland A, Nasti T, Yang S, Zhang R, Barber DL, et al. Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science. 2017;355:1423–7.CrossRefPubMedPubMedCentral Kamphorst AO, Wieland A, Nasti T, Yang S, Zhang R, Barber DL, et al. Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science. 2017;355:1423–7.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Dronca RS, Liu X, Harrington SM, Chen L, Cao S, Kottschade LA, et al. T cell Bim levels reflect responses to anti-PD-1 cancer therapy. JCI Insight. 2016;1:e86014.CrossRefPubMedPubMedCentral Dronca RS, Liu X, Harrington SM, Chen L, Cao S, Kottschade LA, et al. T cell Bim levels reflect responses to anti-PD-1 cancer therapy. JCI Insight. 2016;1:e86014.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Dulos J, Carven GJ, van Boxtel SJ, Evers S, Driessen-Engels LJ, Hobo W, et al. PD-1 blockade augments Th1 and Th17 and suppresses Th2 responses in peripheral blood from patients with prostate and advanced melanoma cancer. J Immunother. 2012;35:169–78.CrossRefPubMed Dulos J, Carven GJ, van Boxtel SJ, Evers S, Driessen-Engels LJ, Hobo W, et al. PD-1 blockade augments Th1 and Th17 and suppresses Th2 responses in peripheral blood from patients with prostate and advanced melanoma cancer. J Immunother. 2012;35:169–78.CrossRefPubMed
42.
Zurück zum Zitat Nonomura Y, Otsuka A, Nakashima C, Seidel JA, Kitoh A, Dainichi T, et al. Peripheral blood Th9 cells are a possible pharmacodynamic biomarker of nivolumab treatment efficacy in metastatic melanoma patients. Oncoimmunology. 2016;5:e1248327.CrossRefPubMedPubMedCentral Nonomura Y, Otsuka A, Nakashima C, Seidel JA, Kitoh A, Dainichi T, et al. Peripheral blood Th9 cells are a possible pharmacodynamic biomarker of nivolumab treatment efficacy in metastatic melanoma patients. Oncoimmunology. 2016;5:e1248327.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Weber JS, Ramakrishnan R, Laino A, Berglund AE, Woods D. Association of changes in T regulatory cells (Treg) during nivolumab treatment with melanoma outcome. J Clin Oncol. 2017;35:(suppl; abstr 3031). Weber JS, Ramakrishnan R, Laino A, Berglund AE, Woods D. Association of changes in T regulatory cells (Treg) during nivolumab treatment with melanoma outcome. J Clin Oncol. 2017;35:(suppl; abstr 3031).
44.
Zurück zum Zitat Kagamu H, Yamaguchi O, Shiono A, Mouri A, Miyauchi S, Utsugi H et al. CD4+ T cells in PBMC to predict the outcome of anti–PD–1 therapy. J Clin Oncol. 2017;35:(suppl; abstr 11525). Kagamu H, Yamaguchi O, Shiono A, Mouri A, Miyauchi S, Utsugi H et al. CD4+ T cells in PBMC to predict the outcome of anti–PD–1 therapy. J Clin Oncol. 2017;35:(suppl; abstr 11525).
45.
Zurück zum Zitat Mahnke YD, Brodie TM, Sallusto F, Roederer M, Lugli E. The who’s who of T cell differentiation: human memory T cell subsets. Eur J Immunol. 2013;43:2797–809.CrossRefPubMed Mahnke YD, Brodie TM, Sallusto F, Roederer M, Lugli E. The who’s who of T cell differentiation: human memory T cell subsets. Eur J Immunol. 2013;43:2797–809.CrossRefPubMed
46.
Zurück zum Zitat Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun. 2016;7:12150.CrossRefPubMedPubMedCentral Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun. 2016;7:12150.CrossRefPubMedPubMedCentral
47.
48.
Zurück zum Zitat Weber J, Gibney G, Kudchadkar R, Yu B, Cheng P, Martinez AJ, et al. Phase I/II study of metastatic melanoma patients treated with nivolumab who had progressed after ipilimumab. Cancer Immunol Res. 2016;4:345–53.CrossRefPubMedPubMedCentral Weber J, Gibney G, Kudchadkar R, Yu B, Cheng P, Martinez AJ, et al. Phase I/II study of metastatic melanoma patients treated with nivolumab who had progressed after ipilimumab. Cancer Immunol Res. 2016;4:345–53.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Youn JI, Gabrilovich DI. The biology of myeloid-derived suppressor cells: the blessing and the curse of morphological and functional heterogeneity. Eur J Immunol. 2010;40:2969–75.CrossRefPubMedPubMedCentral Youn JI, Gabrilovich DI. The biology of myeloid-derived suppressor cells: the blessing and the curse of morphological and functional heterogeneity. Eur J Immunol. 2010;40:2969–75.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Naing A, Papadopoulos KP, Infante JR, Wong DJ, Autio KA, Ott PA, et al. Clinical activity and safety of pegylated human IL–10 (AM0010) in combination with anti–PD1. J Clin Oncol. 2016;34:3018.CrossRef Naing A, Papadopoulos KP, Infante JR, Wong DJ, Autio KA, Ott PA, et al. Clinical activity and safety of pegylated human IL–10 (AM0010) in combination with anti–PD1. J Clin Oncol. 2016;34:3018.CrossRef
51.
Zurück zum Zitat Wu X, Giobbie-Hurder A, Liao X, Connelly C, Connolly EM, Li J, et al. Angiopoietin–2 as a biomarker and target for immune checkpoint therapy. Cancer Immunol Res. 2017;5:17–28.CrossRefPubMed Wu X, Giobbie-Hurder A, Liao X, Connelly C, Connolly EM, Li J, et al. Angiopoietin–2 as a biomarker and target for immune checkpoint therapy. Cancer Immunol Res. 2017;5:17–28.CrossRefPubMed
52.
Zurück zum Zitat Ott PA, Hodi FS, Buchbinder EI. Inhibition of immune checkpoints and vascular endothelial growth factor as combination therapy for metastatic melanoma: an overview of rationale, preclinical evidence, and initial clinical data. Clin Ther. 2015;37:764–82.CrossRef Ott PA, Hodi FS, Buchbinder EI. Inhibition of immune checkpoints and vascular endothelial growth factor as combination therapy for metastatic melanoma: an overview of rationale, preclinical evidence, and initial clinical data. Clin Ther. 2015;37:764–82.CrossRef
53.
Zurück zum Zitat Courau T, Nehar-Belaid D, Florez L, Levacher B, Vazquez T, Brimaud F, et al. TGF–β and VEGF cooperatively control the immunotolerant tumor environment and the efficacy of cancer immunotherapies. JCI Insight. 2016;1:e85974.CrossRefPubMedPubMedCentral Courau T, Nehar-Belaid D, Florez L, Levacher B, Vazquez T, Brimaud F, et al. TGF–β and VEGF cooperatively control the immunotolerant tumor environment and the efficacy of cancer immunotherapies. JCI Insight. 2016;1:e85974.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Yuan J, Zhou J, Dong Z, Tandon S, Kuk D, Panageas KS, et al. Pretreatment serum VEGF is associated with clinical response and overall survival in advanced melanoma patients treated with ipilimumab. Cancer Immunol Res. 2014;2:127–32.CrossRefPubMed Yuan J, Zhou J, Dong Z, Tandon S, Kuk D, Panageas KS, et al. Pretreatment serum VEGF is associated with clinical response and overall survival in advanced melanoma patients treated with ipilimumab. Cancer Immunol Res. 2014;2:127–32.CrossRefPubMed
55.
Zurück zum Zitat Hodi FS, Lawrence D, Lezcano C, Wu X, Zhou J, Sasada T, et al. Bevacizumab plus ipilimumab in patients with metastatic melanoma. Cancer Immunol Res. 2014;2:632–42.CrossRefPubMedPubMedCentral Hodi FS, Lawrence D, Lezcano C, Wu X, Zhou J, Sasada T, et al. Bevacizumab plus ipilimumab in patients with metastatic melanoma. Cancer Immunol Res. 2014;2:632–42.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Wallin JJ, Bendell JC, Funke R, Sznol M, Korski K, Jones S, et al. Atezolizumab in combination with bevacizumab enhances antigen–specific T–cell migration in metastatic renal cell carcinoma. Nat Commun. 2016;7:12624.CrossRefPubMedPubMedCentral Wallin JJ, Bendell JC, Funke R, Sznol M, Korski K, Jones S, et al. Atezolizumab in combination with bevacizumab enhances antigen–specific T–cell migration in metastatic renal cell carcinoma. Nat Commun. 2016;7:12624.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early-and late-stage human malignancies. Sci Transl Med. 2014;6:224ra24.CrossRefPubMedPubMedCentral Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early-and late-stage human malignancies. Sci Transl Med. 2014;6:224ra24.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Newman AM, Bratman SV, To J, Wynne JF, Eclov NC, Modlin LA, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014;20:548–54.CrossRefPubMedPubMedCentral Newman AM, Bratman SV, To J, Wynne JF, Eclov NC, Modlin LA, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014;20:548–54.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Lipson EJ, Velculescu VE, Pritchard TS, Sausen M, Pardoll DM, Topalian SL, et al. Circulating tumor DNA analysis as a real-time method for monitoring tumor burden in melanoma patients undergoing treatment with immune checkpoint blockade. J Immunother Cancer. 2014;2:42.CrossRefPubMedPubMedCentral Lipson EJ, Velculescu VE, Pritchard TS, Sausen M, Pardoll DM, Topalian SL, et al. Circulating tumor DNA analysis as a real-time method for monitoring tumor burden in melanoma patients undergoing treatment with immune checkpoint blockade. J Immunother Cancer. 2014;2:42.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Cabel L, Riva F, Servois V, Livartowski A, Daniel C, Rampanou A, et al. Circulating tumor DNA changes for early monitoring of anti-PD1 immunotherapy: a proof-of-concept study. Ann Oncol. 2017;28:1996–2001.CrossRefPubMed Cabel L, Riva F, Servois V, Livartowski A, Daniel C, Rampanou A, et al. Circulating tumor DNA changes for early monitoring of anti-PD1 immunotherapy: a proof-of-concept study. Ann Oncol. 2017;28:1996–2001.CrossRefPubMed
61.
Zurück zum Zitat Guibert N, Mazieres J, Delaunay M, Casanova A, Farella M, Keller L, et al. Monitoring of KRAS-mutated ctDNA to discriminate pseudo-progression from true progression during anti-PD-1 treatment of lung adenocarcinoma. Oncotarget. 2017;8:38056–60.CrossRefPubMedPubMedCentral Guibert N, Mazieres J, Delaunay M, Casanova A, Farella M, Keller L, et al. Monitoring of KRAS-mutated ctDNA to discriminate pseudo-progression from true progression during anti-PD-1 treatment of lung adenocarcinoma. Oncotarget. 2017;8:38056–60.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Lee CK, Man J, Lord S, Links M, Gebski V, Mok T, et al. Checkpoint inhibitors in metastatic EGFR–mutated non-small cell lung cancer–a meta–analysis. J Thorac Oncol. 2017;12:403–7.CrossRefPubMed Lee CK, Man J, Lord S, Links M, Gebski V, Mok T, et al. Checkpoint inhibitors in metastatic EGFR–mutated non-small cell lung cancer–a meta–analysis. J Thorac Oncol. 2017;12:403–7.CrossRefPubMed
63.
Zurück zum Zitat Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, et al. Mutations associated with acquired resistance to PD–1 blockade in melanoma. N Engl J Med. 2016;375:819–29.CrossRefPubMedPubMedCentral Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, et al. Mutations associated with acquired resistance to PD–1 blockade in melanoma. N Engl J Med. 2016;375:819–29.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Shin DS, Zaretsky JM, Escuin-Ordinas H, Garcia-Diaz A, Hu-Lieskovan S, Kalbasi A, et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov. 2017;7:188–201.CrossRefPubMed Shin DS, Zaretsky JM, Escuin-Ordinas H, Garcia-Diaz A, Hu-Lieskovan S, Kalbasi A, et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov. 2017;7:188–201.CrossRefPubMed
65.
Zurück zum Zitat Nomizo T, Ozasa H, Tsuji T, Funazo T, Yasuda Y, Yoshida H, et al. Clinical impact of single nucleotide polymorphism in PD–l1 on response to nivolumab for advanced non-small-cell lung cancer patients. Sci Rep. 2017;7:45124.CrossRefPubMedPubMedCentral Nomizo T, Ozasa H, Tsuji T, Funazo T, Yasuda Y, Yoshida H, et al. Clinical impact of single nucleotide polymorphism in PD–l1 on response to nivolumab for advanced non-small-cell lung cancer patients. Sci Rep. 2017;7:45124.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Aiello MM, Vigneri PG, Bruzzi P, Verderame F, Paratore S, Restuccia N, et al. Excision repair cross complementation group 1 (ERCC-1) gene polymorphisms and response to nivolumab in advanced non-small cell lung cancer (NSCLC). J Clin Oncol. 2017;35:3032.CrossRef Aiello MM, Vigneri PG, Bruzzi P, Verderame F, Paratore S, Restuccia N, et al. Excision repair cross complementation group 1 (ERCC-1) gene polymorphisms and response to nivolumab in advanced non-small cell lung cancer (NSCLC). J Clin Oncol. 2017;35:3032.CrossRef
67.
Zurück zum Zitat Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342:967–70.CrossRefPubMed Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342:967–70.CrossRefPubMed
68.
Zurück zum Zitat Zitvogel L, Ayyoub M, Routy B, Kroemer G. Microbiome and anticancer immunosurveillance. Cell. 2016;165:276–87.CrossRefPubMed Zitvogel L, Ayyoub M, Routy B, Kroemer G. Microbiome and anticancer immunosurveillance. Cell. 2016;165:276–87.CrossRefPubMed
69.
Zurück zum Zitat Pushalkar S, Ji X, Li Y, Estilo C, Yegnanarayana R, Singh B, et al. Comparison of oral microbiota in tumor and non-tumor tissues of patients with oral squamous cell carcinoma. BMC Microbiol. 2012;12:144.CrossRefPubMedPubMedCentral Pushalkar S, Ji X, Li Y, Estilo C, Yegnanarayana R, Singh B, et al. Comparison of oral microbiota in tumor and non-tumor tissues of patients with oral squamous cell carcinoma. BMC Microbiol. 2012;12:144.CrossRefPubMedPubMedCentral
70.
Zurück zum Zitat Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillere R, Hannani D, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342:971–6.CrossRefPubMedPubMedCentral Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillere R, Hannani D, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342:971–6.CrossRefPubMedPubMedCentral
71.
Zurück zum Zitat Rabe H, Nordström I, Andersson K, Lundell AC, Rudin A. Staphylococcus aureus convert neonatal conventional CD4(+) T cells into FOXP3(+) CD25(+) CD127(low) T cells via the PD-1/PD-L1 axis. Immunology. 2014;141:467–81.CrossRefPubMedPubMedCentral Rabe H, Nordström I, Andersson K, Lundell AC, Rudin A. Staphylococcus aureus convert neonatal conventional CD4(+) T cells into FOXP3(+) CD25(+) CD127(low) T cells via the PD-1/PD-L1 axis. Immunology. 2014;141:467–81.CrossRefPubMedPubMedCentral
72.
Zurück zum Zitat Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079–84.CrossRefPubMedPubMedCentral Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079–84.CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350:1084–9.CrossRefPubMedPubMedCentral Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350:1084–9.CrossRefPubMedPubMedCentral
76.
Zurück zum Zitat Kaderbhai C, Richard C, Fumet JD, Aarnink A, Foucher P, Coudert B, et al. Antibiotic use does not appear to influence response to nivolumab. Anticancer Res. 2017;37:3195–200.PubMed Kaderbhai C, Richard C, Fumet JD, Aarnink A, Foucher P, Coudert B, et al. Antibiotic use does not appear to influence response to nivolumab. Anticancer Res. 2017;37:3195–200.PubMed
77.
Zurück zum Zitat Derosa L, Routy B, Enot D, Baciarello G, Massard C, Loriot Y, et al. Impact of antibiotics on outcome in patients with metastatic renal cell carcinoma treated with immune checkpoint inhibitors. J Clin Oncol. 2017;35:462.CrossRef Derosa L, Routy B, Enot D, Baciarello G, Massard C, Loriot Y, et al. Impact of antibiotics on outcome in patients with metastatic renal cell carcinoma treated with immune checkpoint inhibitors. J Clin Oncol. 2017;35:462.CrossRef
78.
Zurück zum Zitat Chen DS, Mellman I. Oncology meets immunology: the cancer–immunity cycle. Immunity. 2013;39:1–10.CrossRefPubMed Chen DS, Mellman I. Oncology meets immunology: the cancer–immunity cycle. Immunity. 2013;39:1–10.CrossRefPubMed
79.
Zurück zum Zitat Blank CU, Haanen JB, Ribas A, Schumacher TN. The “cancer immunogram”. Science. 2016;352:658–60.CrossRefPubMed Blank CU, Haanen JB, Ribas A, Schumacher TN. The “cancer immunogram”. Science. 2016;352:658–60.CrossRefPubMed
80.
Zurück zum Zitat Karasaki T, Nagayama K, Kuwano H, Nitadori JI, Sato M, Anraku M, et al. An immunogram for the cancer–immunity cycle: towards personalized immunotherapy of lung cancer. J Thorac Oncol. 2017;12:791–803.CrossRefPubMed Karasaki T, Nagayama K, Kuwano H, Nitadori JI, Sato M, Anraku M, et al. An immunogram for the cancer–immunity cycle: towards personalized immunotherapy of lung cancer. J Thorac Oncol. 2017;12:791–803.CrossRefPubMed
Metadaten
Titel
Perspective on immune oncology with liquid biopsy, peripheral blood mononuclear cells, and microbiome with non-invasive biomarkers in cancer patients
verfasst von
A. Mitsuhashi
Y. Okuma
Publikationsdatum
08.01.2018
Verlag
Springer International Publishing
Erschienen in
Clinical and Translational Oncology / Ausgabe 8/2018
Print ISSN: 1699-048X
Elektronische ISSN: 1699-3055
DOI
https://doi.org/10.1007/s12094-017-1827-7

Weitere Artikel der Ausgabe 8/2018

Clinical and Translational Oncology 8/2018 Zur Ausgabe

Hodgkin Lymphom: BrECADD-Regime übertrifft die Erwartungen

05.06.2024 ASCO 2024 Kongressbericht

Das Kombinationsregime BrECADD mit Brentuximab vedotin ermöglichte in der Studie HD21 beim fortgeschrittenen klassischen Hodgkin-Lymphom eine unerwartet hohe progressionsfreie Überlebensrate von 94,3% nach vier Jahren. Gleichzeitig war das Regime besser tolerabel als der bisherige Standard eBEACOPP.

Antikörper-Drug-Konjugat verdoppelt PFS bei Multiplem Myelom

05.06.2024 ASCO 2024 Nachrichten

Zwei Phase-3-Studien deuten auf erhebliche Vorteile des Antikörper-Wirkstoff-Konjugats Belantamab-Mafodotin bei vorbehandelten Personen mit Multiplem Myelom: Im Vergleich mit einer Standard-Tripeltherapie wurde das progressionsfreie Überleben teilweise mehr als verdoppelt.

Neuer TKI gegen CML: Höhere Wirksamkeit, seltener Nebenwirkungen

05.06.2024 Chronische myeloische Leukämie Nachrichten

Der Tyrosinkinasehemmer (TKI) Asciminib ist älteren Vertretern dieser Gruppe bei CML offenbar überlegen: Personen mit frisch diagnostizierter CML entwickelten damit in einer Phase-3-Studie häufiger eine gut molekulare Response, aber seltener ernste Nebenwirkungen.

Brustkrebs-Prävention wird neu gedacht

04.06.2024 ASCO 2024 Kongressbericht

Zurzeit untersuchen Forschende verschiedene neue Ansätze zur Prävention von Brustkrebs bei Personen mit hohem Risiko. Darunter Denosumab, die prophylaktische Bestrahlung der Brust – und Impfungen.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.