Skip to main content
Top
Published in: BMC Cancer 1/2023

Open Access 01-12-2023 | Colorectal Cancer | Research

Screening of prognosis-related Immune cells and prognostic predictors in Colorectal Cancer Patients

Authors: Shuangshuang Deng, Qiping Zhu, Hongyan Chen, Tianyu Xiao, Yinshen Zhu, Jinli Gao, Qing Li, Yong Gao

Published in: BMC Cancer | Issue 1/2023

Login to get access

Abstract

Objective

To accurately screen potential immune cells that can predict the survival of colorectal cancer (CRC) patients and identify related prognostic predictors.

Methods

The sample data of CRC patients were downloaded from the GEO database as a training set to establish a prognosis-scoring model and screen prognosis-related immune cells. The sample data of CRC patients from the TCGA database were used as the validation set. Simultaneously, cancer tissue samples from 116 patients with CRC diagnosed pathologically in Shanghai Dongfang Hospital were collected to analyze the relationship of prognosis-related immune cells with patients’ survival, and clinical and pathological parameters, and to screen prognostic predictors.

Results

Prognosis-related immune cells screened from GEO and TCGA databases mainly included Follicular Helper T cells (Tfh), Monocytes and M2 Macrophages. In the training set, the 2,000- and 4,000-day survival rates were 48.3% and 10.7% in the low-risk group (N = 234), and 42.1% and 7.5% in the high-risk group (N = 214), respectively. In the validation set, the 2,000- and 4,000-day survival rates were 34.8% and 8.6% in the low-risk group (N = 187), and 28.9% and 6.1% in the high-risk group (N = 246), respectively. The prognosis of patients in the high-risk group was worse than that in the low-risk group (P < 0.05). Furthermore, the screened primary prognostic predictors were CD163 and CD4 + CXCR5. CD163 protein expression was distributed in Monocytes and M2 Macrophages. The 1,000- and 2,000-day survival rates were 56.1% and 7.0% in the CD163 low-expression group, and 40.7% and 1.7% in the high-expression group (N = 214), respectively, showing a worse prognosis in the high-expression group than that in the low-expression group. Meanwhile, the immune marker CD4 + CXCR5 could identify Tfh. The 1,000- and 2,000-day survival rates were 63.9% and 5.6% in the CD4 + CXCR5 high-expression group, and 33.3% and 2.8% in the low-expression group (N = 214), respectively, with a better prognosis in the high-expression group than that in the low-expression group.

Conclusion

Prognostic-related immune cells of CRC mainly include Tfh cells, Monocytes and M2 Macrophages. Monocytes and M2 Macrophages correlate negatively, while Tfh cells correlate positively with the prognosis of CRC patients. Immune markers CD163 and CD4 + CXCR5 can be considered as the prognostic predictors of CRC with clinical value of the application.
Literature
1.
go back to reference Keum, NaNa. Giovannucci Edward,Global burden of colorectal cancer: emerging trends, risk factors and prevention strategies[J]. Nat Rev Gastroenterol Hepatol. 2019;16:713–32.PubMedCrossRef Keum, NaNa. Giovannucci Edward,Global burden of colorectal cancer: emerging trends, risk factors and prevention strategies[J]. Nat Rev Gastroenterol Hepatol. 2019;16:713–32.PubMedCrossRef
2.
go back to reference Feng R-M. Current cancer situation in China: good or bad news from the 2018 Global Cancer Statistics?[J]. Cancer Commun (Lond). 2019;39:22. Zong Yi-Nan,Cao Su-Mei. Feng R-M. Current cancer situation in China: good or bad news from the 2018 Global Cancer Statistics?[J]. Cancer Commun (Lond). 2019;39:22. Zong Yi-Nan,Cao Su-Mei.
3.
go back to reference Siegel Rebecca L, Miller Kimberly D, Fuchs Hannah E, et al. Cancer Stat 2021[J] CA Cancer J Clin. 2021;71:7–33.CrossRef Siegel Rebecca L, Miller Kimberly D, Fuchs Hannah E, et al. Cancer Stat 2021[J] CA Cancer J Clin. 2021;71:7–33.CrossRef
4.
go back to reference Miller Kimberly DNogueira, Leticia B et al. Cancer treatment and survivorship statistics, 2019[J].CA Cancer J Clin, 2019, 69: 363–385. Miller Kimberly DNogueira, Leticia B et al. Cancer treatment and survivorship statistics, 2019[J].CA Cancer J Clin, 2019, 69: 363–385.
5.
go back to reference Brenner Hermann,Kloor Matthias,Pox Christian Peter,Colorectal cancer[J].Lancet, 2014, 383: 1490–1502. Brenner Hermann,Kloor Matthias,Pox Christian Peter,Colorectal cancer[J].Lancet, 2014, 383: 1490–1502.
6.
go back to reference Kalyan Aparna,Kircher Sheetal,Shah Hiral. Updates on immunotherapy for colorectal cancer[J]. J Gastrointest Oncol. 2018;9:160–9.PubMedCrossRef Kalyan Aparna,Kircher Sheetal,Shah Hiral. Updates on immunotherapy for colorectal cancer[J]. J Gastrointest Oncol. 2018;9:160–9.PubMedCrossRef
7.
go back to reference Linnekamp Janneke F, Wang Xin,Medema Jan Paul. Colorectal cancer heterogeneity and targeted therapy: a case for molecular disease subtypes[J]. Cancer Res. 2015;75:245–9.PubMedCrossRef Linnekamp Janneke F, Wang Xin,Medema Jan Paul. Colorectal cancer heterogeneity and targeted therapy: a case for molecular disease subtypes[J]. Cancer Res. 2015;75:245–9.PubMedCrossRef
8.
go back to reference Tosolini Marie,Kirilovsky Amos,Mlecnik Bernhard. Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, th2, treg, th17) in patients with colorectal cancer[J]. Cancer Res. 2011;71:1263–71.PubMedCrossRef Tosolini Marie,Kirilovsky Amos,Mlecnik Bernhard. Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, th2, treg, th17) in patients with colorectal cancer[J]. Cancer Res. 2011;71:1263–71.PubMedCrossRef
9.
go back to reference Ioannides CG, Whiteside TL. T cell recognition of human tumors: implications for molecular immunotherapy of cancer. Clin Immunol Immunopathol. 1993;66(2):91–106.PubMedCrossRef Ioannides CG, Whiteside TL. T cell recognition of human tumors: implications for molecular immunotherapy of cancer. Clin Immunol Immunopathol. 1993;66(2):91–106.PubMedCrossRef
10.
go back to reference T GS: Innate and adaptive immune cells in Tumor microenvironment.The Gulf journal of oncology2021, 1(35):77–81. T GS: Innate and adaptive immune cells in Tumor microenvironment.The Gulf journal of oncology2021, 1(35):77–81.
11.
go back to reference Xie Yajuan,Hu Cheng,Feng Yi. Osteoimmunomodulatory effects of biomaterial modification strategies on macrophage polarization and bone regeneration[J]. Regen Biomater. 2020;7:233–45.CrossRef Xie Yajuan,Hu Cheng,Feng Yi. Osteoimmunomodulatory effects of biomaterial modification strategies on macrophage polarization and bone regeneration[J]. Regen Biomater. 2020;7:233–45.CrossRef
12.
go back to reference Franklin RA, Liao W, Sarkar A, Kim MV, Bivona MR, Liu K, Pamer EG, Li MO. The cellular and molecular origin of tumor-associated macrophages. Sci (New York NY). 2014;344(6186):921–5.CrossRef Franklin RA, Liao W, Sarkar A, Kim MV, Bivona MR, Liu K, Pamer EG, Li MO. The cellular and molecular origin of tumor-associated macrophages. Sci (New York NY). 2014;344(6186):921–5.CrossRef
13.
go back to reference Shi X, Shiao SL. The role of macrophage phenotype in regulating the response to radiation therapy. Translational research: the journal of laboratory and clinical medicine. 2018;191:64–80.PubMedCrossRef Shi X, Shiao SL. The role of macrophage phenotype in regulating the response to radiation therapy. Translational research: the journal of laboratory and clinical medicine. 2018;191:64–80.PubMedCrossRef
14.
go back to reference Fu X-L. Interleukin 6 induces M2 macrophage differentiation by STAT3 activation that correlates with gastric cancer progression[J]. Cancer Immunol Immunother. 2017;66:1597–608. Duan Wei,Su. Fu X-L. Interleukin 6 induces M2 macrophage differentiation by STAT3 activation that correlates with gastric cancer progression[J]. Cancer Immunol Immunother. 2017;66:1597–608. Duan Wei,Su.
15.
go back to reference Crumley Andrew BC, Going James J, Hilmy Mustafa, et al. Interrelationships between tumor proliferative activity, leucocyte and macrophage infiltration, systemic inflammatory response, and survival in patients selected for potentially curative resection for gastroesophageal cancer[J]. Ann Surg Oncol. 2011;18:2604–12.PubMedCrossRef Crumley Andrew BC, Going James J, Hilmy Mustafa, et al. Interrelationships between tumor proliferative activity, leucocyte and macrophage infiltration, systemic inflammatory response, and survival in patients selected for potentially curative resection for gastroesophageal cancer[J]. Ann Surg Oncol. 2011;18:2604–12.PubMedCrossRef
16.
go back to reference Turcotte Simon CMichaelJ,Katz, Steven C et al. Tumor-Associated Macrophage Infiltration in Colorectal Cancer Liver Metastases is Associated With Better Outcome[J].Ann Surg Oncol, 2017, 24: 1835–1842. Turcotte Simon CMichaelJ,Katz, Steven C et al. Tumor-Associated Macrophage Infiltration in Colorectal Cancer Liver Metastases is Associated With Better Outcome[J].Ann Surg Oncol, 2017, 24: 1835–1842.
17.
go back to reference Fu X-L. Interleukin 6 induces M2 macrophage differentiation by STAT3 activation that correlates with gastric cancer progression[J]. Cancer Immunol Immunother. 2017;66:1597–608. Duan Wei,Su Chong-Yu. Fu X-L. Interleukin 6 induces M2 macrophage differentiation by STAT3 activation that correlates with gastric cancer progression[J]. Cancer Immunol Immunother. 2017;66:1597–608. Duan Wei,Su Chong-Yu.
18.
go back to reference Kurahara Hiroshi,Shinchi Hiroyuki,Mataki Yuko. Significance of M2-polarized tumor-associated macrophage in pancreatic cancer[J]. J Surg Res. 2011;167:e211–219.PubMedCrossRef Kurahara Hiroshi,Shinchi Hiroyuki,Mataki Yuko. Significance of M2-polarized tumor-associated macrophage in pancreatic cancer[J]. J Surg Res. 2011;167:e211–219.PubMedCrossRef
19.
go back to reference Liu Hang,Wang Jingxuan,Zhang Minghui. Jagged1 promotes aromatase inhibitor resistance by modulating tumor-associated macrophage differentiation in breast cancer patients[J]. Breast Cancer Res Treat. 2017;166:95–107.CrossRef Liu Hang,Wang Jingxuan,Zhang Minghui. Jagged1 promotes aromatase inhibitor resistance by modulating tumor-associated macrophage differentiation in breast cancer patients[J]. Breast Cancer Res Treat. 2017;166:95–107.CrossRef
20.
go back to reference WEI C, YANG C, WANG S, et al. Crosstalk between cancer cells and tumor associated macrophages is required for mesenchymal circulating tumor cell-mediated colorectal cancer metastasis[J]. Mol Cancer. 2019;18(1):64.PubMedPubMedCentralCrossRef WEI C, YANG C, WANG S, et al. Crosstalk between cancer cells and tumor associated macrophages is required for mesenchymal circulating tumor cell-mediated colorectal cancer metastasis[J]. Mol Cancer. 2019;18(1):64.PubMedPubMedCentralCrossRef
21.
go back to reference Afik Ran,Zigmond Ehud,Vugman Milena. Tumor macrophages are pivotal constructors of tumor collagenous matrix[J]. J Exp Med. 2016;213:2315–31.PubMedCrossRef Afik Ran,Zigmond Ehud,Vugman Milena. Tumor macrophages are pivotal constructors of tumor collagenous matrix[J]. J Exp Med. 2016;213:2315–31.PubMedCrossRef
22.
go back to reference Wen, et al. MK2 contributes to tumor progression by promoting M2 macrophage polarization and tumor angiogenesis[J]. Proc Natl Acad Sci U S A. 2018;115:E4236–44. Sriram Ganapathy,Kong Yi. Wen, et al. MK2 contributes to tumor progression by promoting M2 macrophage polarization and tumor angiogenesis[J]. Proc Natl Acad Sci U S A. 2018;115:E4236–44. Sriram Ganapathy,Kong Yi.
23.
go back to reference Zhong Xiaoming,Chen Bin,Yang Zhiwen,The Role of Tumor-Associated Macrophages in Colorectal Carcinoma Progression[J].Cell Physiol Biochem, 2018, 45:356–365. Zhong Xiaoming,Chen Bin,Yang Zhiwen,The Role of Tumor-Associated Macrophages in Colorectal Carcinoma Progression[J].Cell Physiol Biochem, 2018, 45:356–365.
24.
go back to reference Tomita T, Sakurai Y,Ishibashi S, et al. Imbalance of Clara cell-mediated homeostatic inflammation is involved in lung metastasis[J]. Oncogene. 2011;30:3429–39.PubMedCrossRef Tomita T, Sakurai Y,Ishibashi S, et al. Imbalance of Clara cell-mediated homeostatic inflammation is involved in lung metastasis[J]. Oncogene. 2011;30:3429–39.PubMedCrossRef
25.
go back to reference Smyth MJ, Swann J, Hayakawa Y. Innate tumor immune surveillance. Adv Exp Med Biol. 2007;590:103–11.PubMedCrossRef Smyth MJ, Swann J, Hayakawa Y. Innate tumor immune surveillance. Adv Exp Med Biol. 2007;590:103–11.PubMedCrossRef
26.
27.
go back to reference Zhuang Y, Peng LS, Zhao YL, Shi Y, Mao XH, Chen W, Pang KC, Liu XF, Liu T, Zhang JY, et al. CD8(+) T cells that produce interleukin-17 regulate myeloid-derived suppressor cells and are associated with survival time of patients with gastric cancer. Gastroenterology. 2012;143(4):951–962e958.PubMedCrossRef Zhuang Y, Peng LS, Zhao YL, Shi Y, Mao XH, Chen W, Pang KC, Liu XF, Liu T, Zhang JY, et al. CD8(+) T cells that produce interleukin-17 regulate myeloid-derived suppressor cells and are associated with survival time of patients with gastric cancer. Gastroenterology. 2012;143(4):951–962e958.PubMedCrossRef
28.
go back to reference Johnston Robert J, Poholek Amanda C,DiToro, Daniel, et al. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation[J]. Science. 2009;325:1006–10.PubMedPubMedCentralCrossRef Johnston Robert J, Poholek Amanda C,DiToro, Daniel, et al. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation[J]. Science. 2009;325:1006–10.PubMedPubMedCentralCrossRef
29.
go back to reference Barnett Goenka Radhika G, Silver Jonathan S, et al. Cutting edge: dendritic cell-restricted antigen presentation initiates the follicular helper T cell program but cannot complete ultimate effector differentiation[J]. J Immunol. 2011;187:1091–5.PubMedCrossRef Barnett Goenka Radhika G, Silver Jonathan S, et al. Cutting edge: dendritic cell-restricted antigen presentation initiates the follicular helper T cell program but cannot complete ultimate effector differentiation[J]. J Immunol. 2011;187:1091–5.PubMedCrossRef
30.
go back to reference Choi Youn Soo,Eto Danelle,Yang, Jessica A, et al. Cutting edge: STAT1 is required for IL-6-mediated Bcl6 induction for early follicular helper cell differentiation[J]. J Immunol. 2013;190:3049–53.PubMedCrossRef Choi Youn Soo,Eto Danelle,Yang, Jessica A, et al. Cutting edge: STAT1 is required for IL-6-mediated Bcl6 induction for early follicular helper cell differentiation[J]. J Immunol. 2013;190:3049–53.PubMedCrossRef
31.
32.
go back to reference Victora GD, Dominguez-Sola D, Homles AB, et al. Identification of human germinal center light and dark zone cells and their relation-ship to human B-cell lymphomas[J]. Blood. 2012;120(11):2240–8.PubMedPubMedCentralCrossRef Victora GD, Dominguez-Sola D, Homles AB, et al. Identification of human germinal center light and dark zone cells and their relation-ship to human B-cell lymphomas[J]. Blood. 2012;120(11):2240–8.PubMedPubMedCentralCrossRef
33.
go back to reference Good-Jacobson KL, Szumilas CG, Chen L, et al. PD-1 regulates germinal center B cell survival and the formation and affinity of long-lived plasma cells[J]. Nat Immunol. 2010;11(6):535–42.PubMedPubMedCentralCrossRef Good-Jacobson KL, Szumilas CG, Chen L, et al. PD-1 regulates germinal center B cell survival and the formation and affinity of long-lived plasma cells[J]. Nat Immunol. 2010;11(6):535–42.PubMedPubMedCentralCrossRef
34.
go back to reference Yu MZhiyuanXieY et al. [Peripheral blood CD4+;CXCR5+; follicular helper T cells are related to hyperglobulinemia of patients with chronic hepatitis B][J].Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi, 2013, 29: 515-8, 521. Yu MZhiyuanXieY et al. [Peripheral blood CD4+;CXCR5+; follicular helper T cells are related to hyperglobulinemia of patients with chronic hepatitis B][J].Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi, 2013, 29: 515-8, 521.
35.
go back to reference Akiba Hisaya,Takeda Kazuyoshi,Kojima Yuko. The role of ICOS in the CXCR5 + follicular B helper T cell maintenance in vivo[J]. J Immunol. 2005;175:2340–8.PubMedCrossRef Akiba Hisaya,Takeda Kazuyoshi,Kojima Yuko. The role of ICOS in the CXCR5 + follicular B helper T cell maintenance in vivo[J]. J Immunol. 2005;175:2340–8.PubMedCrossRef
36.
go back to reference Pallikkuth Suresh,Parmigiani Anita,Silva, Sandra Y, et al. Impaired peripheral blood T-follicular helper cell function in HIV-infected nonresponders to the 2009 H1N1/09 vaccine[J]. Blood. 2012;120:985–93.CrossRef Pallikkuth Suresh,Parmigiani Anita,Silva, Sandra Y, et al. Impaired peripheral blood T-follicular helper cell function in HIV-infected nonresponders to the 2009 H1N1/09 vaccine[J]. Blood. 2012;120:985–93.CrossRef
37.
go back to reference Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133(5):775–87.PubMedCrossRef Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133(5):775–87.PubMedCrossRef
38.
go back to reference Michie CA, McLean A, Alcock C, Beverley PC. Lifespan of human lymphocyte subsets defined by CD45 isoforms. Nature. 1992;360(6401):264–5.PubMedCrossRef Michie CA, McLean A, Alcock C, Beverley PC. Lifespan of human lymphocyte subsets defined by CD45 isoforms. Nature. 1992;360(6401):264–5.PubMedCrossRef
39.
go back to reference Erler JT, Weaver VM. Three-dimensional context regulation of metastasis. Clin Exp Metastasis. 2009;26(1):35–49.PubMedCrossRef Erler JT, Weaver VM. Three-dimensional context regulation of metastasis. Clin Exp Metastasis. 2009;26(1):35–49.PubMedCrossRef
40.
go back to reference Luen SJ, Savas P, Fox SB, Salgado R, Loi S. Tumour-infiltrating lymphocytes and the emerging role of immunotherapy in breast cancer. Pathology. 2017;49(2):141–55.PubMedCrossRef Luen SJ, Savas P, Fox SB, Salgado R, Loi S. Tumour-infiltrating lymphocytes and the emerging role of immunotherapy in breast cancer. Pathology. 2017;49(2):141–55.PubMedCrossRef
41.
go back to reference Ingold Heppner B, Untch M, Denkert C, Pfitzner BM, Lederer B, Schmitt W, Eidtmann H, Fasching PA, Tesch H, Solbach C, et al. Tumor-infiltrating lymphocytes: a predictive and prognostic biomarker in Neoadjuvant-Treated HER2-Positive breast Cancer. Clin cancer research: official J Am Association Cancer Res. 2016;22(23):5747–54.CrossRef Ingold Heppner B, Untch M, Denkert C, Pfitzner BM, Lederer B, Schmitt W, Eidtmann H, Fasching PA, Tesch H, Solbach C, et al. Tumor-infiltrating lymphocytes: a predictive and prognostic biomarker in Neoadjuvant-Treated HER2-Positive breast Cancer. Clin cancer research: official J Am Association Cancer Res. 2016;22(23):5747–54.CrossRef
42.
go back to reference Althobiti M, Aleskandarany MA, Joseph C, Toss M, Mongan N, Diez-Rodriguez M, Nolan CC, Ashankyty I, Ellis IO, Green AR, et al. Heterogeneity of tumour-infiltrating lymphocytes in breast cancer and its prognostic significance. Histopathology. 2018;73(6):887–96.PubMedCrossRef Althobiti M, Aleskandarany MA, Joseph C, Toss M, Mongan N, Diez-Rodriguez M, Nolan CC, Ashankyty I, Ellis IO, Green AR, et al. Heterogeneity of tumour-infiltrating lymphocytes in breast cancer and its prognostic significance. Histopathology. 2018;73(6):887–96.PubMedCrossRef
43.
go back to reference Jiang D, Liu Y, Wang H, Wang H, Song Q, Sujie A, Huang J, Xu Y, Zeng H, Tan L, et al. Tumour infiltrating lymphocytes correlate with improved survival in patients with esophageal squamous cell carcinoma. Sci Rep. 2017;7:44823.PubMedPubMedCentralCrossRef Jiang D, Liu Y, Wang H, Wang H, Song Q, Sujie A, Huang J, Xu Y, Zeng H, Tan L, et al. Tumour infiltrating lymphocytes correlate with improved survival in patients with esophageal squamous cell carcinoma. Sci Rep. 2017;7:44823.PubMedPubMedCentralCrossRef
44.
go back to reference Schumacher K, Haensch W, Röefzaad C, Schlag PM. Prognostic significance of activated CD8(+) T cell infiltrations within esophageal carcinomas. Cancer Res. 2001;61(10):3932–6.PubMed Schumacher K, Haensch W, Röefzaad C, Schlag PM. Prognostic significance of activated CD8(+) T cell infiltrations within esophageal carcinomas. Cancer Res. 2001;61(10):3932–6.PubMed
45.
go back to reference Lu L, Weng C, Mao H, Fang X, Liu X, Wu Y, Cao X, Li B, Chen X, Gan Q, et al. IL-17A promotes migration and tumor killing capability of B cells in esophageal squamous cell carcinoma. Oncotarget. 2016;7(16):21853–64.PubMedPubMedCentralCrossRef Lu L, Weng C, Mao H, Fang X, Liu X, Wu Y, Cao X, Li B, Chen X, Gan Q, et al. IL-17A promotes migration and tumor killing capability of B cells in esophageal squamous cell carcinoma. Oncotarget. 2016;7(16):21853–64.PubMedPubMedCentralCrossRef
46.
go back to reference Yoshioka T, Miyamoto M, Cho Y, Ishikawa K, Tsuchikawa T, Kadoya M, Li L, Mishra R, Ichinokawa K, Shoji Y, et al. Infiltrating regulatory T cell numbers is not a factor to predict patient’s survival in oesophageal squamous cell carcinoma. Br J Cancer. 2008;98(7):1258–63.PubMedPubMedCentralCrossRef Yoshioka T, Miyamoto M, Cho Y, Ishikawa K, Tsuchikawa T, Kadoya M, Li L, Mishra R, Ichinokawa K, Shoji Y, et al. Infiltrating regulatory T cell numbers is not a factor to predict patient’s survival in oesophageal squamous cell carcinoma. Br J Cancer. 2008;98(7):1258–63.PubMedPubMedCentralCrossRef
47.
go back to reference Mori M, Ohtani H, Naito Y, Sagawa M, Sato M, Fujimura S, Nagura H. Infiltration of CD8 + T cells in non-small cell lung cancer is associated with dedifferentiation of cancer cells, but not with prognosis. Tohoku J Exp Med. 2000;191(2):113–8.PubMedCrossRef Mori M, Ohtani H, Naito Y, Sagawa M, Sato M, Fujimura S, Nagura H. Infiltration of CD8 + T cells in non-small cell lung cancer is associated with dedifferentiation of cancer cells, but not with prognosis. Tohoku J Exp Med. 2000;191(2):113–8.PubMedCrossRef
48.
go back to reference Shaban M, Khurram SA, Fraz MM, Alsubaie N, Masood I, Mushtaq S, Hassan M, Loya A, Rajpoot NM. A Novel Digital score for abundance of Tumour infiltrating lymphocytes predicts Disease Free Survival in oral squamous cell carcinoma. Sci Rep. 2019;9(1):13341.PubMedPubMedCentralCrossRef Shaban M, Khurram SA, Fraz MM, Alsubaie N, Masood I, Mushtaq S, Hassan M, Loya A, Rajpoot NM. A Novel Digital score for abundance of Tumour infiltrating lymphocytes predicts Disease Free Survival in oral squamous cell carcinoma. Sci Rep. 2019;9(1):13341.PubMedPubMedCentralCrossRef
49.
go back to reference Zeng Y, Zeng Y, Yin H, Chen F, Wang Q, Yu X, Zhou Y. Exploration of the immune cell infiltration-related gene signature in the prognosis of melanoma. Aging. 2021;13(3):3459–82.PubMedPubMedCentralCrossRef Zeng Y, Zeng Y, Yin H, Chen F, Wang Q, Yu X, Zhou Y. Exploration of the immune cell infiltration-related gene signature in the prognosis of melanoma. Aging. 2021;13(3):3459–82.PubMedPubMedCentralCrossRef
50.
go back to reference Lu G, Chen L, Wu S, Feng Y, Lin T. Comprehensive Analysis of Tumor-Infiltrating Immune Cells and Relevant Therapeutic Strategy in Esophageal Cancer. 2020, 2020:8974793. Lu G, Chen L, Wu S, Feng Y, Lin T. Comprehensive Analysis of Tumor-Infiltrating Immune Cells and Relevant Therapeutic Strategy in Esophageal Cancer. 2020, 2020:8974793.
51.
go back to reference Vigano L, Soldani C, Franceschini B, Cimino M, Lleo A, Donadon M, Roncalli M, Aghemo A, Di Tommaso L, Torzilli G. Tumor-infiltrating lymphocytes and macrophages in Intrahepatic Cholangiocellular Carcinoma. Impact on Prognosis after Complete Surgery. 2019;23(11):2216–24. Vigano L, Soldani C, Franceschini B, Cimino M, Lleo A, Donadon M, Roncalli M, Aghemo A, Di Tommaso L, Torzilli G. Tumor-infiltrating lymphocytes and macrophages in Intrahepatic Cholangiocellular Carcinoma. Impact on Prognosis after Complete Surgery. 2019;23(11):2216–24.
52.
go back to reference Sui S, An X, Xu C, Li Z, Hua Y, Huang G, Sui S, Long Q, Sui Y, Xiong Y, et al. An immune cell infiltration-based immune score model predicts prognosis and chemotherapy effects in breast cancer. Theranostics. 2020;10(26):11938–49.PubMedPubMedCentralCrossRef Sui S, An X, Xu C, Li Z, Hua Y, Huang G, Sui S, Long Q, Sui Y, Xiong Y, et al. An immune cell infiltration-based immune score model predicts prognosis and chemotherapy effects in breast cancer. Theranostics. 2020;10(26):11938–49.PubMedPubMedCentralCrossRef
Metadata
Title
Screening of prognosis-related Immune cells and prognostic predictors in Colorectal Cancer Patients
Authors
Shuangshuang Deng
Qiping Zhu
Hongyan Chen
Tianyu Xiao
Yinshen Zhu
Jinli Gao
Qing Li
Yong Gao
Publication date
01-12-2023
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2023
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-10667-y

Other articles of this Issue 1/2023

BMC Cancer 1/2023 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine