C reactive protein impairs adaptive immunity in immune cells of patients with melanoma

Tatsuya Yoshida, Junya Ichikawa, Iulia Giuroiu, Andressa S Laino, Yuhan Hao, Michelle Krogsgaard, Melinda Vassallo, David M Woods, F Stephen Hodi, Jeffrey Weber, Tatsuya Yoshida, Junya Ichikawa, Iulia Giuroiu, Andressa S Laino, Yuhan Hao, Michelle Krogsgaard, Melinda Vassallo, David M Woods, F Stephen Hodi, Jeffrey Weber

Abstract

Background: High C reactive protein (CRP) levels have been reported to be associated with a poor clinical outcome in a number of malignancies and with programmed cell death protein 1 immune checkpoint blockade in patients with advanced cancer. Little is known about the direct effects of CRP on adaptive immunity in cancer. Therefore, we investigated how CRP impacted the function of T cells and dendritic cells (DCs) from patients with melanoma.

Methods: The effects of CRP on proliferation, function, gene expression and phenotype of patient T cells and DCs, and expansion of MART-1 antigen-specific T cells were analyzed by multicolor flow cytometry and RNA-seq. Additionally, serum CRP levels at baseline from patients with metastatic melanoma treated on the Checkmate-064 clinical trial were assessed by a Luminex assay.

Results: In vitro, CRP inhibited proliferation, activation-associated phenotypes and the effector function of activated CD4+ and CD8+ T cells from patients with melanoma. CRP-treated T cells expressed high levels of interleukin-1β, which is known to enhance CRP production from the liver. CRP also suppressed formation of the immune synapse and inhibited early events in T-cell receptor engagement. In addition, CRP downregulated the expression of costimulatory molecules on mature DCs and suppressed expansion of MART-1-specific CD8+ T cells in a dose-dependent manner by impacting on both T cells and antigen-presenting cells. High-serum CRP levels at baseline were significantly associated with a shorter survival in both nivolumab-treated and ipilimumab-treated patients.

Conclusions: These findings suggest that high levels of CRP induce an immunosuppressive milieu in melanoma and support the blockade of CRP as a therapeutic strategy to enhance immune checkpoint therapies in cancer.

Trial registration number: NCT01783938 and NCT02983006.

Keywords: immunology; medicine; oncology.

Conflict of interest statement

Competing interests: JW: honoraria and travel from BMS, Merck, GSK, Genentech, AstraZeneca, Pfizer, CytoMx, EMD Serono, Incyte. Stock in Biond, Altor, Protean, CytoMx, Celldex, Sellas. Research funding from NextCure. All other clinical research funding to my institution, not me. Named on a patent for a PD-1 biomarker by Biodesix not used in this work. Named on a CTLA-4 biomarker patent by Moffitt Cancer Center not used in this work. Named on a patent for the use of 41-BB antibody for tumor infiltrating lymphocyte growth by Moffitt Cancer Center not used in this work.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
C reactive protein (CRP) affects the proliferation on activated T cells. CD3+ T cells were isolated by negative magnetic separation from baseline patient peripheral blood mononuclear cells (PBMC), and activated with CD3/CD28 antibody for 72 hours in the presence of different CRP concentration. (A) Proliferation analysis of CellTrace Violet-labeled CD3+ T cells stimulated with anti-CD3 and CD28 monoclonal antibodies (mAbs) for 72 hours. The cell ratio and CRP+ population of CD8+ cells in each generation (G1 to over G4) were detected. *P

Figure 2

C reactive protein (CRP) affects…

Figure 2

C reactive protein (CRP) affects the function of activated T cells. Expression of…

Figure 2
C reactive protein (CRP) affects the function of activated T cells. Expression of checkpoint, stimulatory and cytolytic proteins by flow cytometry on activated CD8 (A) and CD4 (B) T cells treated with CRP at 10, 20 and 40 µg/mL as a heat map. Representative data from three patients. (C) CD3+ T cells isolated from baseline peripheral blood mononuclear cells (PBMC) samples of patients with melanoma prior to any treatment were cultured for 72 hours with or without CRP at doses of 10, 40 and 100 µg/mL, and subsequently stimulated for 4 hours with phorbol 12-myristate 13-acetate (PMA)/ionomycin for analysis of intracellular cytokine production (interferon (IFN)-γ, tumor necrosis factor (TNF)-α and interleukin (IL)-2) and granzyme B. Representative data from five patients. Error bars are +SEM for triplicate samples assessed. **P

Figure 3

C reactive protein (CRP) inhibits…

Figure 3

C reactive protein (CRP) inhibits T-cell receptor (TCR) signaling pathway in T cells.…

Figure 3
C reactive protein (CRP) inhibits T-cell receptor (TCR) signaling pathway in T cells. (A) Expression of phosphorylated LCK (pY505), ZAP70(pY319)/Syk(pY352), LAT(pY717), ERK(pT202/pY204) and AKT(pS473) on CD3+ T cells pretreated with CRP at 10, 40 and 100 µg/mL for 72 hours and concomitantly stimulated with H2O2 are from one representative of five similar patients. *P<0.05, **p<0.01, ***p<0.001 relative to cells with no treatment. (B) Representative data from three patients of calcium influx on T cells. CD3+ T cells treated with CRP for 24 hours at 10–100 μg/mL were activated with phorbol 12-myristate 13-acetate (PMA) and ionomycin. (C and D) Anti-CD3/CD28 monoclonal antibodies (mAbs) Dynabeads recruit lipid rafts of CD3+ cells to form immunological synapses. CRP (40 µg/mL) treated CD3+ T cells stained with anti-CRP antibody (green) were significantly less likely to recruit lipid rafts to form the immunological synapse at the bead-T cell interface denoted by Phallodin staining (red) that control treated CD3+ T cells. Representative tiled image and high-magnification images from one of three patients.

Figure 4

C reactive protein (CRP) inhibits…

Figure 4

C reactive protein (CRP) inhibits function, costimulatory and checkpoint proteins on dendritic cells…

Figure 4
C reactive protein (CRP) inhibits function, costimulatory and checkpoint proteins on dendritic cells (DCs). Frequency of mature (A) and immature (B) DCs treated with CRP at 10 and 40 μg/mL for 48 hours expressed costimulatory markers (CD40, CD80, CD83, CD86, PD-L1, HLA-ABC, HLA-DR and beta-2 microglobulin). Representative data are from one of five patients. Error bars are +SEM for triplicate samples assessed. **P

Figure 5

C reactive protein (CRP) inhibits…

Figure 5

C reactive protein (CRP) inhibits the expansion of antigen-specific T cells. (A) MART-1-specific…

Figure 5
C reactive protein (CRP) inhibits the expansion of antigen-specific T cells. (A) MART-1-specific CD8 T-cell expansion. CD8+ T cells isolated from HLA-A*0201 patients with melanoma were stimulated by γ-irradiated, MART-126-35 peptide-pulsed peripheral blood mononuclear cells (PBMCs) (antigen-presenting cells (APCs)) for 7 days in presence of different CRP concentration (0, 10, 40 µg/mL). (B and C) Ki67 expression and proliferation by CFSE-labeled Melan-A-specific CD8+ T cells and MART-1 tetramer-positive T cells stimulated by γ-irradiated MART-126-35 peptide pulsed APC. (D) Scheme for assessing whether CRP-impaired generation of Melan-A-positive CD8+ T cells was due to its effect on T cells and/or on mature dendritic cells (mDCs). (E) Purified CD8+ T cells and mDCs separately before co-culture were treated with CRP for 48 hours, and co-cultured with MART-1 peptide-pulsed mDCs. Results shown are representative of three patients assessed over three independent experiments. (F) Volcano plot for the differential expression of total genes. The y-axis corresponds to negative log10 transformed adjusted p value, and the x-axis displays the log2 fold change gene expression value. The red and blue dots represent the significantly differential expressed transcripts (adjusted p value < 0.05); the gray dots represent the transcripts whose expression levels did not reach statistical significance (adjusted p value > 0.05). (G) Expression heat map of genes associated with cytokine-cytokine receptor interaction pathway, and osteoclast differentiation pathway. Error bars are +SEM for triplicate samples assessed. **P<0.05, **p<0.01, ***p<0.001 relative to cells with no CRP treatment.

Figure 6

Serum C reactive protein (CRP)…

Figure 6

Serum C reactive protein (CRP) levels in treatment-naïve patients receiving nivolumab (cohort A)…

Figure 6
Serum C reactive protein (CRP) levels in treatment-naïve patients receiving nivolumab (cohort A) or ipilimumab (cohort B) from the Checkmate-064 trial. (A) Consolidated Standards of Reporting Trials diagram for the study. Among 140 patients enrolled in the Checkmate-064 trial, 95 serum samples before administration of study drug were obtained for CRP analysis (29 from cohort A, 66 from cohort B). (B) Kaplan-Meier plot of the relation of overall survival (OS) to CRP levels before treatment. Cut-off point was at the median (15.48 µg/mL). The red curve represents survival for patients below the median, and the blue curve shows survival for those patients above the median. The survival probabilities were estimated using the Kaplan-Meier method, where differences in the variables were calculated using the log-rank test.
Similar articles
Cited by
References
    1. Du Clos TW. Function of C-reactive protein. Ann Med 2000;32:274–8. 10.3109/07853890009011772 - DOI - PubMed
    1. Gabay C, Kushner I. Acute-Phase proteins and other systemic responses to inflammation. N Engl J Med 1999;340:448–54. 10.1056/NEJM199902113400607 - DOI - PubMed
    1. Yamashita H, Shimada K, Seki E, et al. . Concentrations of interleukins, interferon, and C-reactive protein in stable and unstable angina pectoris. Am J Cardiol 2003;91:133–6. 10.1016/S0002-9149(02)03097-7 - DOI - PubMed
    1. Yang J, Liu Z, Liu H, et al. . C-Reactive protein promotes bone destruction in human myeloma through the CD32-p38 MAPK-Twist axis. Sci Signal 2017;10 10.1126/scisignal.aan6282. [Epub ahead of print: 12 Dec 2017]. - DOI - PMC - PubMed
    1. Agnoli C, Grioni S, Pala V, et al. . Biomarkers of inflammation and breast cancer risk: a case-control study nested in the EPIC-Varese cohort. Sci Rep 2017;7:12708 10.1038/s41598-017-12703-x - DOI - PMC - PubMed
Show all 44 references
Publication types
MeSH terms
Associated data
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 2
Figure 2
C reactive protein (CRP) affects the function of activated T cells. Expression of checkpoint, stimulatory and cytolytic proteins by flow cytometry on activated CD8 (A) and CD4 (B) T cells treated with CRP at 10, 20 and 40 µg/mL as a heat map. Representative data from three patients. (C) CD3+ T cells isolated from baseline peripheral blood mononuclear cells (PBMC) samples of patients with melanoma prior to any treatment were cultured for 72 hours with or without CRP at doses of 10, 40 and 100 µg/mL, and subsequently stimulated for 4 hours with phorbol 12-myristate 13-acetate (PMA)/ionomycin for analysis of intracellular cytokine production (interferon (IFN)-γ, tumor necrosis factor (TNF)-α and interleukin (IL)-2) and granzyme B. Representative data from five patients. Error bars are +SEM for triplicate samples assessed. **P

Figure 3

C reactive protein (CRP) inhibits…

Figure 3

C reactive protein (CRP) inhibits T-cell receptor (TCR) signaling pathway in T cells.…

Figure 3
C reactive protein (CRP) inhibits T-cell receptor (TCR) signaling pathway in T cells. (A) Expression of phosphorylated LCK (pY505), ZAP70(pY319)/Syk(pY352), LAT(pY717), ERK(pT202/pY204) and AKT(pS473) on CD3+ T cells pretreated with CRP at 10, 40 and 100 µg/mL for 72 hours and concomitantly stimulated with H2O2 are from one representative of five similar patients. *P<0.05, **p<0.01, ***p<0.001 relative to cells with no treatment. (B) Representative data from three patients of calcium influx on T cells. CD3+ T cells treated with CRP for 24 hours at 10–100 μg/mL were activated with phorbol 12-myristate 13-acetate (PMA) and ionomycin. (C and D) Anti-CD3/CD28 monoclonal antibodies (mAbs) Dynabeads recruit lipid rafts of CD3+ cells to form immunological synapses. CRP (40 µg/mL) treated CD3+ T cells stained with anti-CRP antibody (green) were significantly less likely to recruit lipid rafts to form the immunological synapse at the bead-T cell interface denoted by Phallodin staining (red) that control treated CD3+ T cells. Representative tiled image and high-magnification images from one of three patients.

Figure 4

C reactive protein (CRP) inhibits…

Figure 4

C reactive protein (CRP) inhibits function, costimulatory and checkpoint proteins on dendritic cells…

Figure 4
C reactive protein (CRP) inhibits function, costimulatory and checkpoint proteins on dendritic cells (DCs). Frequency of mature (A) and immature (B) DCs treated with CRP at 10 and 40 μg/mL for 48 hours expressed costimulatory markers (CD40, CD80, CD83, CD86, PD-L1, HLA-ABC, HLA-DR and beta-2 microglobulin). Representative data are from one of five patients. Error bars are +SEM for triplicate samples assessed. **P

Figure 5

C reactive protein (CRP) inhibits…

Figure 5

C reactive protein (CRP) inhibits the expansion of antigen-specific T cells. (A) MART-1-specific…

Figure 5
C reactive protein (CRP) inhibits the expansion of antigen-specific T cells. (A) MART-1-specific CD8 T-cell expansion. CD8+ T cells isolated from HLA-A*0201 patients with melanoma were stimulated by γ-irradiated, MART-126-35 peptide-pulsed peripheral blood mononuclear cells (PBMCs) (antigen-presenting cells (APCs)) for 7 days in presence of different CRP concentration (0, 10, 40 µg/mL). (B and C) Ki67 expression and proliferation by CFSE-labeled Melan-A-specific CD8+ T cells and MART-1 tetramer-positive T cells stimulated by γ-irradiated MART-126-35 peptide pulsed APC. (D) Scheme for assessing whether CRP-impaired generation of Melan-A-positive CD8+ T cells was due to its effect on T cells and/or on mature dendritic cells (mDCs). (E) Purified CD8+ T cells and mDCs separately before co-culture were treated with CRP for 48 hours, and co-cultured with MART-1 peptide-pulsed mDCs. Results shown are representative of three patients assessed over three independent experiments. (F) Volcano plot for the differential expression of total genes. The y-axis corresponds to negative log10 transformed adjusted p value, and the x-axis displays the log2 fold change gene expression value. The red and blue dots represent the significantly differential expressed transcripts (adjusted p value < 0.05); the gray dots represent the transcripts whose expression levels did not reach statistical significance (adjusted p value > 0.05). (G) Expression heat map of genes associated with cytokine-cytokine receptor interaction pathway, and osteoclast differentiation pathway. Error bars are +SEM for triplicate samples assessed. **P<0.05, **p<0.01, ***p<0.001 relative to cells with no CRP treatment.

Figure 6

Serum C reactive protein (CRP)…

Figure 6

Serum C reactive protein (CRP) levels in treatment-naïve patients receiving nivolumab (cohort A)…

Figure 6
Serum C reactive protein (CRP) levels in treatment-naïve patients receiving nivolumab (cohort A) or ipilimumab (cohort B) from the Checkmate-064 trial. (A) Consolidated Standards of Reporting Trials diagram for the study. Among 140 patients enrolled in the Checkmate-064 trial, 95 serum samples before administration of study drug were obtained for CRP analysis (29 from cohort A, 66 from cohort B). (B) Kaplan-Meier plot of the relation of overall survival (OS) to CRP levels before treatment. Cut-off point was at the median (15.48 µg/mL). The red curve represents survival for patients below the median, and the blue curve shows survival for those patients above the median. The survival probabilities were estimated using the Kaplan-Meier method, where differences in the variables were calculated using the log-rank test.
Similar articles
Cited by
References
    1. Du Clos TW. Function of C-reactive protein. Ann Med 2000;32:274–8. 10.3109/07853890009011772 - DOI - PubMed
    1. Gabay C, Kushner I. Acute-Phase proteins and other systemic responses to inflammation. N Engl J Med 1999;340:448–54. 10.1056/NEJM199902113400607 - DOI - PubMed
    1. Yamashita H, Shimada K, Seki E, et al. . Concentrations of interleukins, interferon, and C-reactive protein in stable and unstable angina pectoris. Am J Cardiol 2003;91:133–6. 10.1016/S0002-9149(02)03097-7 - DOI - PubMed
    1. Yang J, Liu Z, Liu H, et al. . C-Reactive protein promotes bone destruction in human myeloma through the CD32-p38 MAPK-Twist axis. Sci Signal 2017;10 10.1126/scisignal.aan6282. [Epub ahead of print: 12 Dec 2017]. - DOI - PMC - PubMed
    1. Agnoli C, Grioni S, Pala V, et al. . Biomarkers of inflammation and breast cancer risk: a case-control study nested in the EPIC-Varese cohort. Sci Rep 2017;7:12708 10.1038/s41598-017-12703-x - DOI - PMC - PubMed
Show all 44 references
Publication types
MeSH terms
Associated data
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 3
Figure 3
C reactive protein (CRP) inhibits T-cell receptor (TCR) signaling pathway in T cells. (A) Expression of phosphorylated LCK (pY505), ZAP70(pY319)/Syk(pY352), LAT(pY717), ERK(pT202/pY204) and AKT(pS473) on CD3+ T cells pretreated with CRP at 10, 40 and 100 µg/mL for 72 hours and concomitantly stimulated with H2O2 are from one representative of five similar patients. *P<0.05, **p<0.01, ***p<0.001 relative to cells with no treatment. (B) Representative data from three patients of calcium influx on T cells. CD3+ T cells treated with CRP for 24 hours at 10–100 μg/mL were activated with phorbol 12-myristate 13-acetate (PMA) and ionomycin. (C and D) Anti-CD3/CD28 monoclonal antibodies (mAbs) Dynabeads recruit lipid rafts of CD3+ cells to form immunological synapses. CRP (40 µg/mL) treated CD3+ T cells stained with anti-CRP antibody (green) were significantly less likely to recruit lipid rafts to form the immunological synapse at the bead-T cell interface denoted by Phallodin staining (red) that control treated CD3+ T cells. Representative tiled image and high-magnification images from one of three patients.
Figure 4
Figure 4
C reactive protein (CRP) inhibits function, costimulatory and checkpoint proteins on dendritic cells (DCs). Frequency of mature (A) and immature (B) DCs treated with CRP at 10 and 40 μg/mL for 48 hours expressed costimulatory markers (CD40, CD80, CD83, CD86, PD-L1, HLA-ABC, HLA-DR and beta-2 microglobulin). Representative data are from one of five patients. Error bars are +SEM for triplicate samples assessed. **P

Figure 5

C reactive protein (CRP) inhibits…

Figure 5

C reactive protein (CRP) inhibits the expansion of antigen-specific T cells. (A) MART-1-specific…

Figure 5
C reactive protein (CRP) inhibits the expansion of antigen-specific T cells. (A) MART-1-specific CD8 T-cell expansion. CD8+ T cells isolated from HLA-A*0201 patients with melanoma were stimulated by γ-irradiated, MART-126-35 peptide-pulsed peripheral blood mononuclear cells (PBMCs) (antigen-presenting cells (APCs)) for 7 days in presence of different CRP concentration (0, 10, 40 µg/mL). (B and C) Ki67 expression and proliferation by CFSE-labeled Melan-A-specific CD8+ T cells and MART-1 tetramer-positive T cells stimulated by γ-irradiated MART-126-35 peptide pulsed APC. (D) Scheme for assessing whether CRP-impaired generation of Melan-A-positive CD8+ T cells was due to its effect on T cells and/or on mature dendritic cells (mDCs). (E) Purified CD8+ T cells and mDCs separately before co-culture were treated with CRP for 48 hours, and co-cultured with MART-1 peptide-pulsed mDCs. Results shown are representative of three patients assessed over three independent experiments. (F) Volcano plot for the differential expression of total genes. The y-axis corresponds to negative log10 transformed adjusted p value, and the x-axis displays the log2 fold change gene expression value. The red and blue dots represent the significantly differential expressed transcripts (adjusted p value < 0.05); the gray dots represent the transcripts whose expression levels did not reach statistical significance (adjusted p value > 0.05). (G) Expression heat map of genes associated with cytokine-cytokine receptor interaction pathway, and osteoclast differentiation pathway. Error bars are +SEM for triplicate samples assessed. **P<0.05, **p<0.01, ***p<0.001 relative to cells with no CRP treatment.

Figure 6

Serum C reactive protein (CRP)…

Figure 6

Serum C reactive protein (CRP) levels in treatment-naïve patients receiving nivolumab (cohort A)…

Figure 6
Serum C reactive protein (CRP) levels in treatment-naïve patients receiving nivolumab (cohort A) or ipilimumab (cohort B) from the Checkmate-064 trial. (A) Consolidated Standards of Reporting Trials diagram for the study. Among 140 patients enrolled in the Checkmate-064 trial, 95 serum samples before administration of study drug were obtained for CRP analysis (29 from cohort A, 66 from cohort B). (B) Kaplan-Meier plot of the relation of overall survival (OS) to CRP levels before treatment. Cut-off point was at the median (15.48 µg/mL). The red curve represents survival for patients below the median, and the blue curve shows survival for those patients above the median. The survival probabilities were estimated using the Kaplan-Meier method, where differences in the variables were calculated using the log-rank test.
Figure 5
Figure 5
C reactive protein (CRP) inhibits the expansion of antigen-specific T cells. (A) MART-1-specific CD8 T-cell expansion. CD8+ T cells isolated from HLA-A*0201 patients with melanoma were stimulated by γ-irradiated, MART-126-35 peptide-pulsed peripheral blood mononuclear cells (PBMCs) (antigen-presenting cells (APCs)) for 7 days in presence of different CRP concentration (0, 10, 40 µg/mL). (B and C) Ki67 expression and proliferation by CFSE-labeled Melan-A-specific CD8+ T cells and MART-1 tetramer-positive T cells stimulated by γ-irradiated MART-126-35 peptide pulsed APC. (D) Scheme for assessing whether CRP-impaired generation of Melan-A-positive CD8+ T cells was due to its effect on T cells and/or on mature dendritic cells (mDCs). (E) Purified CD8+ T cells and mDCs separately before co-culture were treated with CRP for 48 hours, and co-cultured with MART-1 peptide-pulsed mDCs. Results shown are representative of three patients assessed over three independent experiments. (F) Volcano plot for the differential expression of total genes. The y-axis corresponds to negative log10 transformed adjusted p value, and the x-axis displays the log2 fold change gene expression value. The red and blue dots represent the significantly differential expressed transcripts (adjusted p value < 0.05); the gray dots represent the transcripts whose expression levels did not reach statistical significance (adjusted p value > 0.05). (G) Expression heat map of genes associated with cytokine-cytokine receptor interaction pathway, and osteoclast differentiation pathway. Error bars are +SEM for triplicate samples assessed. **P<0.05, **p<0.01, ***p<0.001 relative to cells with no CRP treatment.
Figure 6
Figure 6
Serum C reactive protein (CRP) levels in treatment-naïve patients receiving nivolumab (cohort A) or ipilimumab (cohort B) from the Checkmate-064 trial. (A) Consolidated Standards of Reporting Trials diagram for the study. Among 140 patients enrolled in the Checkmate-064 trial, 95 serum samples before administration of study drug were obtained for CRP analysis (29 from cohort A, 66 from cohort B). (B) Kaplan-Meier plot of the relation of overall survival (OS) to CRP levels before treatment. Cut-off point was at the median (15.48 µg/mL). The red curve represents survival for patients below the median, and the blue curve shows survival for those patients above the median. The survival probabilities were estimated using the Kaplan-Meier method, where differences in the variables were calculated using the log-rank test.

References

    1. Du Clos TW. Function of C-reactive protein. Ann Med 2000;32:274–8. 10.3109/07853890009011772
    1. Gabay C, Kushner I. Acute-Phase proteins and other systemic responses to inflammation. N Engl J Med 1999;340:448–54. 10.1056/NEJM199902113400607
    1. Yamashita H, Shimada K, Seki E, et al. . Concentrations of interleukins, interferon, and C-reactive protein in stable and unstable angina pectoris. Am J Cardiol 2003;91:133–6. 10.1016/S0002-9149(02)03097-7
    1. Yang J, Liu Z, Liu H, et al. . C-Reactive protein promotes bone destruction in human myeloma through the CD32-p38 MAPK-Twist axis. Sci Signal 2017;10 10.1126/scisignal.aan6282. [Epub ahead of print: 12 Dec 2017].
    1. Agnoli C, Grioni S, Pala V, et al. . Biomarkers of inflammation and breast cancer risk: a case-control study nested in the EPIC-Varese cohort. Sci Rep 2017;7:12708 10.1038/s41598-017-12703-x
    1. Yasuda Y, Saito K, Yuasa T, et al. . Early response of C-reactive protein as a predictor of survival in patients with metastatic renal cell carcinoma treated with tyrosine kinase inhibitors. Int J Clin Oncol 2017;22:1081–6. 10.1007/s10147-017-1166-2
    1. Hang J, Xue P, Yang H, et al. . Pretreatment C-reactive protein to albumin ratio for predicting overall survival in advanced pancreatic cancer patients. Sci Rep 2017;7:2993 10.1038/s41598-017-03153-6
    1. Pastorino U, Morelli D, Leuzzi G, et al. . Baseline and postoperative C-reactive protein levels predict mortality in operable lung cancer. Eur J Cancer 2017;79:90–7. 10.1016/j.ejca.2017.03.020
    1. Akamine T, Takada K, Toyokawa G, et al. . Association of preoperative serum CRP with PD-L1 expression in 508 patients with non-small cell lung cancer: a comprehensive analysis of systemic inflammatory markers. Surg Oncol 2018;27:88–94. 10.1016/j.suronc.2018.01.002
    1. Zhang L, Liu S-H, Wright TT, et al. . C-Reactive protein directly suppresses Th1 cell differentiation and alleviates experimental autoimmune encephalomyelitis. J Immunol 2015;194:5243–52. 10.4049/jimmunol.1402909
    1. Zhang R, Becnel L, Li M, et al. . C-Reactive protein impairs human CD14+ monocyte-derived dendritic cell differentiation, maturation and function. Eur J Immunol 2006;36:2993–3006. 10.1002/eji.200635207
    1. Tarhini AA, Cherian J, Moschos SJ, et al. . Safety and efficacy of combination immunotherapy with interferon alfa-2b and tremelimumab in patients with stage IV melanoma. J Clin Oncol 2012;30:322–8. 10.1200/JCO.2011.37.5394
    1. Oya Y, Yoshida T, Kuroda H, et al. . Predictive clinical parameters for the response of nivolumab in pretreated advanced non-small-cell lung cancer. Oncotarget 2017;8:103117–28. 10.18632/oncotarget.21602
    1. Weber JS, Sznol M, Sullivan RJ, et al. . A serum protein signature associated with outcome after anti-PD-1 therapy in metastatic melanoma. Cancer Immunol Res 2018;6:79–86. 10.1158/2326-6066.CIR-17-0412
    1. Weber JS, Gibney G, Sullivan RJ, et al. . Sequential administration of nivolumab and ipilimumab with a planned switch in patients with advanced melanoma (CheckMate 064): an open-label, randomised, phase 2 trial. Lancet Oncol 2016;17:943–55. 10.1016/S1470-2045(16)30126-7
    1. Maeda Y, Nishikawa H, Sugiyama D, et al. . Detection of self-reactive CD8⁺ T cells with an anergic phenotype in healthy individuals. Science 2014;346:1536–40. 10.1126/science.aaa1292
    1. Lin Y, Gallardo HF, Ku GY, et al. . Optimization and validation of a robust human T-cell culture method for monitoring phenotypic and polyfunctional antigen-specific CD4 and CD8 T-cell responses. Cytotherapy 2009;11:912–22. 10.3109/14653240903136987
    1. Dobin A, Davis CA, Schlesinger F, et al. . Star: ultrafast universal RNA-seq aligner. Bioinformatics 2013;29:15–21. 10.1093/bioinformatics/bts635
    1. Liao Y, Smyth GK, Shi W. featureCounts: an efficient General purpose program for assigning sequence reads to genomic features. Bioinformatics 2014;30:923–30. 10.1093/bioinformatics/btt656
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-Seq data with DESeq2. Genome Biol 2014;15:550 10.1186/s13059-014-0550-8
    1. Viola A, Schroeder S, Sakakibara Y, et al. . T lymphocyte costimulation mediated by reorganization of membrane microdomains. Science 1999;283:680–2. 10.1126/science.283.5402.680
    1. Nicolaou SA, Neumeier L, Peng Y, et al. . The Ca(2+)-activated K(+) channel KCa3.1 compartmentalizes in the immunological synapse of human T lymphocytes. Am J Physiol Cell Physiol 2007;292:C1431–9. 10.1152/ajpcell.00376.2006
    1. Lu J, Marnell LL, Marjon KD, et al. . Structural recognition and functional activation of FcgammaR by innate pentraxins. Nature 2008;456:989–92. 10.1038/nature07468
    1. Griffith CE, Zhang W, Wange RL. ZAP-70-dependent and -independent activation of ERK in Jurkat T cells. Differences in signaling induced by H2O2 and CD3 cross-linking. J Biol Chem 1998;273:10771–6. 10.1074/jbc.273.17.10771
    1. Schieven GL, Mittler RS, Nadler SG, et al. . Zap-70 tyrosine kinase, CD45, and T cell receptor involvement in UV- and H2O2-induced T cell signal transduction. J Biol Chem 1994;269:20718–26.
    1. Chen EY, Tan CM, Kou Y, et al. . Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 2013;14:128 10.1186/1471-2105-14-128
    1. Kuleshov MV, Jones MR, Rouillard AD, et al. . Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res 2016;44:W90–7. 10.1093/nar/gkw377
    1. McFadyen JD, Kiefer J, Braig D, et al. . Dissociation of C-reactive protein localizes and amplifies inflammation: evidence for a direct biological role of C-reactive protein and its conformational changes. Front Immunol 2018;9:1351 10.3389/fimmu.2018.01351
    1. Litvack ML, Palaniyar N. Review: soluble innate immune pattern-recognition proteins for clearing dying cells and cellular components: implications on exacerbating or resolving inflammation. Innate Immun 2010;16:191–200. 10.1177/1753425910369271
    1. Agrawal A, Cha-Molstad H, Samols D, et al. . Overexpressed nuclear factor-kappaB can participate in endogenous C-reactive protein induction, and enhances the effects of C/EBPbeta and signal transducer and activator of transcription-3. Immunology 2003;108:539–47. 10.1046/j.1365-2567.2003.01608.x
    1. Jimenez RV, Wright TT, Jones NR, et al. . C-Reactive protein impairs dendritic cell development, maturation, and function: implications for peripheral tolerance. Front Immunol 2018;9:372 10.3389/fimmu.2018.00372
    1. Kuehnemuth B, Piseddu I, Wiedemann GM, et al. . Ccl1 is a major regulatory T cell attracting factor in human breast cancer. BMC Cancer 2018;18:1278 10.1186/s12885-018-5117-8
    1. D'Amico L, Roato I. Cross-Talk between T cells and osteoclasts in bone resorption. Bonekey Rep 2012;1:82 10.1038/bonekey.2012.82
    1. Yunis I, Whitehead AS. The mouse C-reactive protein gene maps to distal chromosome 1 and, like its human counterpart, is closely linked to the serum amyloid P component gene. Immunogenetics 1990;32:361–3. 10.1007/BF00211651
    1. Thomas-Rudolph D, Du Clos TW, Snapper CM, et al. . C-Reactive protein enhances immunity to Streptococcus pneumoniae by targeting uptake to Fc gamma R on dendritic cells. J Immunol 2007;178:7283–91. 10.4049/jimmunol.178.11.7283
    1. Fang S, Wang Y, Sui D, et al. . C-Reactive protein as a marker of melanoma progression. J Clin Oncol 2015;33:1389–96. 10.1200/JCO.2014.58.0209
    1. Findeisen P, Zapatka M, Peccerella T, et al. . Serum amyloid A as a prognostic marker in melanoma identified by proteomic profiling. J Clin Oncol 2009;27:2199–208. 10.1200/JCO.2008.18.0554
    1. Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 2016;127:3321–30. 10.1182/blood-2016-04-703751
    1. Teachey DT, Lacey SF, Shaw PA, et al. . Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov 2016;6:664–79. 10.1158/-16-0040
    1. Rossi J-F, Lu Z-Y, Jourdan M, et al. . Interleukin-6 as a therapeutic target. Clin Cancer Res 2015;21:1248–57. 10.1158/1078-0432.CCR-14-2291
    1. Norelli M, Camisa B, Barbiera G, et al. . Monocyte-Derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med 2018;24:739–48. 10.1038/s41591-018-0036-4
    1. Uemura M, Trinh VA, Haymaker C, et al. . Selective inhibition of autoimmune exacerbation while preserving the anti-tumor clinical benefit using IL-6 blockade in a patient with advanced melanoma and Crohn's disease: a case report. J Hematol Oncol 2016;9:81 10.1186/s13045-016-0309-7
    1. Ridker PM, Everett BM, Thuren T, et al. . Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med 2017;377:1119–31. 10.1056/NEJMoa1707914
    1. Ridker PM, MacFadyen JG, Thuren T, et al. . Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet 2017;390:1833–42. 10.1016/S0140-6736(17)32247-X

Source: PubMed

3
Tilaa