Cancer-driven changes link T cell frequency to muscle strength in people with cancer: a pilot study

Aditi Narsale, Rosa Moya, Jasmin Ma, Lindsey J Anderson, Daniel Wu, Jose M Garcia, Joanna D Davies, Aditi Narsale, Rosa Moya, Jasmin Ma, Lindsey J Anderson, Daniel Wu, Jose M Garcia, Joanna D Davies

Abstract

Background: Tumour growth can promote the loss of muscle mass and function. This is particularly disturbing because overall survival is significantly reduced in people with weaker and smaller skeletal muscle. The risk of cancer is also greater in people who are immune deficient. Muscle wasting in mice with cancer can be inhibited by infusion of CD4+ precursor T cells that restore balanced ratios of naïve, memory, and regulatory T cells. These data are consistent with the hypothesis that stronger anti-cancer T cell immunity leads to improved muscle mass and function. As a first step to testing this hypothesis, we determined whether levels of circulating T cell subsets correlate with levels of muscle strength in people with cancer.

Methods: The frequency of circulating CD4+ and CD8+ naïve, memory, and regulatory T cell subsets was quantified in 11 men with gastrointestinal cancer (aged 59.3 ± 10.1 years) and nine men without cancer (aged 60 ± 13 years), using flow cytometry. T cell marker expression was determined using real-time PCR and western blot analyses in whole blood and peripheral blood mononuclear cells. Handgrip strength, one-repetition maximum chest press, and knee extension tests were used to determine muscle strength. Performance was determined using a stair climb test. Body composition was determined using dual-energy X-ray absorptiometry scan. The Karnofsky and ECOG scales were used to assess functional impairment. Correlations between frequencies of cell subsets with strength, performance, and body composition were determined using regression analyses.

Results: Our data show significant correlations between (i) higher frequencies of CD8+ naïve (P = 0.02) and effector memory (P = 0.003) T cells and lower frequencies of CD8+ central memory T cells (P = 0.002) with stronger handgrip strength, (ii) lower frequency of regulatory cells with greater lean mass index (P = 0.04), (iii) lower frequency of CD8+ T cells that express CD95 with greater stair climb power (P = 0.003), (iv) higher frequency of T cells that co-express CD197 and CD45RA and greater one-repetition maximum knee extension strength (P = 0.008), and (iv) higher expression of CD4 in whole blood with greater functional impairment (P = 0.004) in people with cancer.

Conclusions: We have identified significant correlations between levels of T cell populations and muscle strength, performance, and body composition in people with cancer. These data justify a follow-up study with a larger cohort to test the validity of the findings.

Keywords: Cancer; Correlations; Flow cytometry; Muscle; T cells.

Conflict of interest statement

None declared.

© 2019 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of the Society on Sarcopenia, Cachexia and Wasting Disorders.

Figures

Figure 1
Figure 1
Handgrip strength (HGS) and one‐repetition maximum chest press strength correlate with the frequency of circulating recent thymic emigrants (RTEs), central memory (CM), and effector memory (EM) in subjects with, but not without, cancer. Peripheral blood mononuclear cells were isolated from fresh whole blood and labelled for naïve and memory cell subsets as described in Materials and methods section and Table2. Panel A shows the correlation between the frequency of naïve CD8+ T cells that express CD31 but not Ki67 (CD8+ RTE, n = 7), CD8+ T cells that are CM (CD8+ CM, n = 9), and CD8+ memory cells that are EM (CD8+ EM, n = 9) and either HGS or chest press strength in the cancer group. Each symbol represents a patient. Spearman correlation (r) and statistical significance (P) are shown on each panel in red for the cancer group and in black for the non‐cancer group. (B) The frequency of each cell subset in people with cancer is compared with non‐cancer‐matched controls using the Student's t‐test. Data shown are mean ± SEM. There is no statistically significant difference between groups.
Figure 2
Figure 2
T cells and non‐T cells, identified by their co‐expression of CD197 and CD45RA, correlate with muscle strength and performance. The frequency of CD197+ CD3+ (A) and CD197+ CD3− (B) cells that express CD45RA. Correlations with handgrip strength (HGS, chest press strength, knee extension strength, and stair climb power (SCP) were calculated. Each symbol represents a participant. Spearman correlation (r) and statistical significance (P) are shown on each panel for the cancer group.
Figure 3
Figure 3
CD197 expression is significantly reduced in people with cancer. CD197 mRNA expression in whole blood from people with (n = 11) and without (n = 9) cancer was quantified using real‐time RT‐PCR (A). CD197 and CD45RA protein expression was determined on a single western blot (B and C). Panel B shows the combined data from all samples in both groups. Panel C shows all samples from each group on the same gel. Groups are compared using the Student's t‐test. Data shown are mean ± SEM. The P value is shown on panel A.
Figure 4
Figure 4
The frequency of CD95+ T cells and non‐T cells correlates with lower body muscle strength in people with cancer. Correlations between relative frequency of CD8+ CD95+ T cells and stair climb power (SCP) (A) and knee extension strength (B). Correlations between the relative frequency of CD3− peripheral blood mononuclear cell (PBMC) in people with cancer that express CD95 and either SCP (C) or knee extension strength (D). Spearman correlation (r) and statistical significance (P) are shown on panels A–D. Each symbol represents a single participant.
Figure 5
Figure 5
The level of CD95 gene expression in whole blood correlates with muscle strength. CD95 mRNA in whole blood (A) and CD95 and caspase 3 protein expression in peripheral blood mononuclear cell (B and C) was quantified using real‐time RT‐PCR and western blot from people with (n = 11) and without (n = 9) cancer. Panel B shows the combined data from all samples in both groups. Panel C shows all samples from each group on the same gel. Groups were compared using the Student's t‐test. Data shown are mean ± SEM. The P value is shown on panel A. Spearman correlations (r) in the group with cancer are shown between the levels of CD95 mRNA expression in whole blood, quantified by real‐time RT‐PCR, and either knee extension strength (D) or stair climb power (SCP) (E).
Figure 6
Figure 6
Treg cell frequency correlates with a low lean mass index (LMI) in people with cancer. Correlations between the relative frequency of Treg (A) with LMI in people with cancer. The LMI shown is the LMI in kg/m2 × 1000. Spearman correlation (r) and statistical significance (P) are shown. Panel B, Treg cell frequency, is compared between groups using the Student's t‐test. Data shown are mean ± SEM.
Figure 7
Figure 7
Interleukin (IL) 2‐expressing CD8+ T cells and body composition. Correlations between the relative frequency IL‐2 expressing CD8+ T cells and either lean mass index (LMI) (A) or body mass index (BMI) (B) in people with cancer (n = 8). The LMI shown is the LMI in kg/m2 × 1000. Spearman correlation (r) and statistical significance (P) are shown on panels A and B. Each symbol represents a single participant. Panel C, IL‐2+ CD8+ cell subset frequency, is compared between groups with (n = 8) and without (n = 8) cancer using the Student's t‐test. Data shown are mean ± SEM.
Figure 8
Figure 8
Immune correlations with the Karnofsky Performance Scale Index and ECOG‐PS. The level of CD4 expression in whole blood from people with (n = 11) and without (n = 9) cancer was determined by real‐time RT‐PCR. In the same subjects, performance was determined using both the Karnofsky index and the ECOG‐PS. Panels A and B compare the Karnofsky index (A) and ECOG‐PS (B) in people with and without cancer. Correlations between CD4 levels and either the Karnofsky index (C) or the ECOG‐PS (D) are shown for people with cancer. In panels A and B, groups are compared using the Student's t‐test. Data shown are mean ± SEM. The P value is shown on each panel. Spearman correlation (r) and statistical significance (P) are shown on panels C and D for the cancer group. Data are not significant for the non‐cancer group.

References

    1. Lepone LM, Donahue RN, Grenga I, Metenou S, Richards J, Heery CR, et al. Analysis of 123 peripheral human immune cell subsets: defining differences with age and between healthy donors and cancer patients not detected in analysis of standard immune cell types. J Circ Biomarkers 2016;5:5–21.
    1. Wang T, Zhang Z, Xing H, Wang L, Zhang G, Yu N, et al. Elevated Th22 cells and related cytokines in patients with epithelial ovarian cancer. Medicine 2017;96:43–48.
    1. Czystowska M, Gooding W, Szczepanski MJ, Lopez‐Abaitero A, Ferris RL, Johnson JT, et al. The immune signature of CD8+ CCR7+ T cells in the peripheral circulation associates with disease recurrence in patients with HNSCC. Clin Cancer Res 2013;19:889–899.
    1. Kotsakis A, Koinis F, Katsarou A, Gioulbasani M, Aggouraki D, Kentepozidis N, et al. Prognostic value of circulating regulatory T cell subsets in untreated non‐small cell lung cancer patients. Sci Rep 2016;6:39247.
    1. Schafer ZT, Brugge JS. IL‐6 involvement in epithelial cancers. J Clin Invest 2007;117:3660–3663.
    1. Lucey DR, Clerici M, Shearer GM. Type 1 and type 2 cytokine dysregulation in human infectious, neoplastic, and inflammatory diseases. Clin Microbiol Rev 1996;9:532–562.
    1. Utech AE, Tadros EM, Hayes TG, Garcia JM. Predicting survival in cancer patients: the role of cachexia and hormonal, nutritional and inflammatory markers. J Cachexia Sarcopenia Muscle 2012;3:245–251.
    1. van Vledder MG, Levolger S, Ayez N, Verhoef C, Tran TC, Ijzermans JN. Body composition and outcome in patients undergoing resection of colorectal liver metastases. Br J Surg 2012;99:550–557.
    1. Caan BJ, Cespedes Feliciano EM, Prado CM, Alexeeff S, Kroenke CH, Bradshaw P, et al. Association of muscle and adiposity measured by computed tomography with survival in patients with nonmetastatic breast cancer. JAMA Oncol 2018;4:798–804.
    1. Versteeg KS, Blauwhoff‐Buskermolen S, Buffart LM, de van der Schueren MAE, Langius JAE, Verheul HMW, et al. Higher muscle strength is associated with prolonged survival in older patients with advanced cancer. Oncologist 2018;23:580–585.
    1. Chen CH, Ho‐Chang HYZ, Hung TT. Hand‐grip strength is a simple and effective outcome predictor in esophageal cancer following esophagectomy with reconstruction: a prospective study. J Cardiothoracic Surg 2011;6:98.
    1. Sato S, Nagai E, Taki Y, Watanabe M, Watanabe Y, Nakano K, et al. Hand grip strength as a predictor of postoperative complications in esophageal cancer patients undergoing esophagectomy. Esophagus 2018;15:10–18.
    1. Aversa Z, Costelli P, Muscaritoli M. Cancer‐induced muscle wasting: latest findings in prevention and treatment. Ther Adv Med Oncol 2017;9:369–382.
    1. Porporato PE. Understanding cachexia as a cancer metabolism syndrome. Oncogene 2016;5:e200.
    1. Duchowny K. Do nationally representative cutpoints for clinical muscle weakness predict mortality? Results from nine years of follow‐up in the health and retirement study. J Gerontol A Biol Sci Med Sci 2018;10.1093/gerona/gly169. [Epub ahead of print].
    1. Yorke AM, Curtis AB, Shoemaker M, Vangsnes E. Grip strength values stratified by age, gender, and chronic disease status in adults aged 50 years and older. J Geriatr Phys Ther 2015;38:115–121.
    1. Herrero F, San Juan A, Fleck S, Balmer J, Pérez M, Cañete S, et al. A. combined aerobic and resistance training in breast cancer survivors: a randomized, controlled pilot trial. Int J Sports Med 2006;27:573–580.
    1. Winters‐Stone KM, Dobek J, Bennett JA, Nail LM, Leo MC, Schwartz A. The effect of resistance training on muscle strength and physical function in older, postmenopausal breast cancer survivors: a randomized controlled trial. J Cancer Surviv 2012;6:189–199.
    1. De Backer IC, Van Breda E, Vreugdenhil A, Nijziel MR, Kester AD, Schep G. High‐intensity strength training improves quality of life in cancer survivors. Acta Oncol 2007;46:1143–1151.
    1. Brunelli A, Monteverde M, Al Refai M, Fianchini A. Stair climbing test as a predictor of cardiopulmonary complications after pulmonary lobectomy in the elderly. Ann Thorac Surg 2004;77:266–270.
    1. Reddy S, Contreras CM, Singletary B, Bradford TM, Waldrop MG, Mims AH, et al. Timed stair climbing is the single strongest predictor of perioperative complications in patients undergoing abdominal surgery. J Am Coll Surg 2016;222:559–566.
    1. Bennell K, Dobson F, Hinman R. Measures of physical performance assessments: Self‐Paced Walk Test (SPWT), Stair Climb Test (SCT), Six‐Minute Walk Test (6MWT), Chair Stand Test (CST), Timed Up & amp; Go (TUG), Sock Test, Lift and Carry Test (LCT), and Car Task. Arthritis Care Res (Hoboken) 2011;63:S350–S370.
    1. Garcia JM, Boccia RV, Graham CD, Yan Y, Duus EM, Allen S, et al. Anamorelin for patients with cancer cachexia: an integrated analysis of two phase 2, randomized, placebo‐controlled, double‐blind trials. Lancet Oncol 2015;16:108–116.
    1. Temel JS, Abernethy AP, Currow DC, Friend J, Duus EM, Yan Y, et al. Anamorelin in patients with non‐small‐cell lung cancer and cachexia (ROMANA 1 and ROMANA 2): results from two randomised, double‐blind, phase 3 trials. Lancet Oncol 2016;17:519–531.
    1. Kinsey E, Ajazi E, Wang X, Johnston MAM, Crawford J. Predictors of physical and functional loss in advanced stage lung cancer patients receiving platinum chemotherapy. J Thorac Oncol 2018;S1556–S0864, (18)30676–2.
    1. Crawford J, Prado CM, Johnston MA, Gralla RJ, Taylor RP, Hancock ML, et al. Study design and rationale for the phase 3 clinical development program of enobosarm, a selective androgen receptor modulator, for the prevention and treatment of muscle wasting in cancer patients (POWER trials). Curr Oncol Rep 2016;18:37.
    1. Dobs AS, Boccia RV, Croot CC, Gabrail NY, Dalton JT, Hancock ML, et al. Effects of enobosarm on muscle wasting and physical function in patients with cancer: a double‐blind, randomized controlled phase 2 trial. Lancet Oncol 2013;14:335–345.
    1. Zhao C, Davies JD. A peripheral CD4+ T cell precursor for naïve, memory and regulatory T cells. J Exp Med 2010;207:2883–2894.
    1. Zhao C, Marrero I, Narsale A, Moya R, Davies JD. CD4+ CD44v.low cells are unique peripheral precursors that are distinct from recent thymic emigrants and stem cell‐like memory cells. Cell Immunol 2015;296:106–114.
    1. Wang Z, Zhao C, Moya R, Davies JD. A novel role for CD4+ T cells in the control of cachexia. J Immunol 2008;181:4676–4684.
    1. Anderson LJ, Liu H, Garcia JM. Sex differences in muscle wasting. Adv Exp Med Biol 2017;1043:153–197.
    1. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol 2004;22:745–763.
    1. Rosenblum MD, Way SS, Abbas AK. Regulatory T cell memory. Nat Rev Immunol 2015;16:90–101.
    1. Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 2009;30:899–911.
    1. Liu W, Putnam AL, Xu‐Yu Z, Szot GL, Lee MR, Zhu S, et al. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4 + T reg cells. J Exp Med 2006;203:1701–1711.
    1. Seddiki N, Santner‐Nanan B, Martinson J, Zaunders J, Sasson S, Landay A, et al. Expression of interleukin (IL)‐2 and IL‐7 receptors discriminates between human regulatory and activated T cells. J Exp Med 2006;203:1693–1700.
    1. Gattinoni L, Zhong XS, Palmer DC, Yi Y, Hinrichs CS, Yu Z, et al. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat Med 2009;15:808–813.
    1. Gattinoni L, Lugli E, Ji Y, Pos Z, Paulos CM, Quigley MF, et al. A human memory T cell subset with stem cell‐like properties. Nat Med 2011;17:1290–1297.
    1. Junge S, Kloeckener‐Gruissem B, Zufferey R, Keisker A, Salgo B, Fauchere JC, et al. Correlation between recent thymic emigrants and CD31+ (PECAM‐1) CD4+ T cells in normal individuals during aging and in lymphopenic children. Eur J Immunol 2007;37:3270–3280.
    1. Kohler S, Thiel A. Life after the thymus: CD31+ and CD31− human naïve CD4+ T‐cell subsets. Blood 2009;113:769–774.
    1. Moya R, Robertson HK, Payne D, Narsale A, Koziol J, Davies JD, et al. A pilot study showing associations between frequency of CD4(+) memory cell subsets at diagnosis and duration of partial remission in type 1 diabetes. Clin Immunol 2016;166‐167:72–80.
    1. Narsale A, Moya R, Robertson HK, Davies JD, Type 1Diabetes TrialNet Study Group . Data on correlations between T cell subset frequencies and length of partial remission in type 1 diabetes. Data Brief 2016;8:1348–1351.
    1. Zhang DH, Cohn L, Ray P, Bottomly K, Ray A. Transcription factor GATA‐3 is differentially expressed in murine Th1 and Th2 cells and controls Th2‐specific expression of the interleukin‐5 gene. J Biol Chem 1997;272:21597–21603.
    1. Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher LH. A novel transcription factor, T‐bet, directs Th1 lineage commitment. Cell 2000;100:655–659.
    1. Weber DR. A comparison of fat and lean body mass index to BMI for the identification of metabolic syndrome in children and adolescents. J Clin Endocrinol Metab 2014;99:3208–3216.
    1. Lerner L, Hayes TG, Tao N, Krieger B, Feng B, Wu Z, et al. Plasma growth differentiation factor 15 is associated with weight loss and mortality in cancer patients. J Cachexia Sarcopenia Muscle 2015;6:317–324.
    1. Hollen PJ, Gralla RJ, Kris MG, Cox C, Belani CP, Grunberg SM, et al. Measurement of quality of life in patients with lung cancer in multicenter trials of new therapies. Cancer 1994;73:2087–2098.
    1. ECOG Performance Status. 2009. dataset/ecog.html
    1. Burney BO, Hayes TG, Smiechowska J, Cardwell G, Papusha V, Bhargava P, et al. Low testosterone levels and increased inflammatory markers in patients with cancer and relationship with cachexia. J Clin Endocrinol Metab 2012;97:E700–E709.
    1. Garcia JM, Friend J, Allen S. Therapeutic potential of anamorelin, a novel, oral ghrelin mimetic, in patients with cancer‐related cachexia: a multicenter, randomized, double‐blind, crossover, pilot study. Support Care Cancer 2013;21:129–137.
    1. Gruber I, Landenberger N, Staebler A, Hahn M, Wallwiener D, Fehm T. Relationship between circulating tumor cells and peripheral T‐cells in patients with primary breast cancer. Anticancer Res 2013;33:2233–2238.
    1. Malek TR. The biology of interleukin‐2. Annu Rev Immunol 2008;26:453–479.
    1. Zhang N, Bevan MJ. CD8(+) T cells: foot soldiers of the immune system. Immunity 2011;35:161–168.
    1. Kaech SM, Cui W. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat Rev Immunol 2012;12:749–761.
    1. Durgeau A, Virk Y, Corgnac S, Mami‐Chouaib F. Recent advances in targeting CD8 T‐cell immunity for more effective cancer immunotherapy. Front Immunol 2018;9:14–27.
    1. Celis‐Morales CA, Welsh P, Lyall DM, Steel L, Petermann F, Anderson J, et al. Associations of grip strength with cardiovascular, respiratory, and cancer outcomes and all acause mortality: prospective cohort study of half a million UK Biobank participants. Br Med J 2018;k1651:361.
    1. Kohler S, Wagner U, Pierer M, Kimmig S, Oppmann B, Möwes B, et al. Post‐thymic in vivo proliferation of naïve CD4+ T cells constrains the TCR repertoire in healthy human adults. Eur J Immunol 2005;35:1987–1994.
    1. Forster R, Schubel A, Breitfeld D, Kremmer E, Renner‐Müller I, Wolf E, et al. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell 1999;99:23–33.
    1. Nolz JC, Starbeck‐Miller GR, Harty JT. Naïve, effector and memory CD8 T‐cell trafficking: parallels and distinctions. Immunotherapy 2011;3:1223–1233.
    1. Narsale A, Moya R, Davies JD. Human CD4+ CD25+ CD127hi cells and the Th1/Th2 phenotype. Clin Immunol 2018;188:103–112.
    1. Cederholm T, Wretlind B, Hellstrom K, Andersson B, Engstrom L, Brismar K, et al. Enhanced generation of interleukins 1 beta and 6 may contribute to the cachexia of chronic disease. Am J Clin Nutr 1997;65:876–882.
    1. Yasumoto K, Mukaida N, Harada A, Kuno K, Akiyama M, Nakashima E, et al. Molecular analysis of the cytokine network involved in cachexia in colon 26 adenocarcinoma‐bearing mice. Cancer Res 1995;55:921–927.
    1. Mantovani G, Maccio A, Lai P, Massa E, Ghiani M, Santona MC. Cytokine involvement in cancer anorexia/cachexia: role of megestrol acetate and medroxyprogesterone acetate on cytokine down regulation and improvement of clinical symptoms. Crit Rev Oncol 1998;9:99–106.
    1. Strassmann G, Fong M, Kenney JS, Jacob CO. Evidence for the involvement of interleukin 6 in experimental cancer cachexia. J Clin Invest 1992 1992;89:1681–1684.
    1. Sturlan S, Beinhauer BG, Oberhuber G, Huang L, Aasen AO, Rogy MA. In vivo gene transfer of murine interleukin‐4 inhibits colon‐26‐mediated cancer cachexia in mice. Anticancer Res 2002;22:2547–2554.
    1. Fujiki F, Mukaida N, Hirose K, Ishida H, Harada A, Ohno S, et al. Prevention of adenocarcinoma colon 26‐induced cachexia by interleukin 10 gene transfer. Cancer Res 1997;57:94–99.
    1. Fӧrster R, Davalos‐Misslitz AC, Rot A. CCR7 and its ligands: balancing immunity and tolerance. Nat Rev Immunol 2008;8:362–371.
    1. Penna G, Sozzani S, Adorini L. Cutting edge: selective usage of chemokine receptors by plasmacytoid dendritic cells. J Immunol 2001;167:1862–1866.
    1. Umemoto E, Otani K, Ikeno T, Verjan Garcia N, Hayasaka H, Bai Z, et al. Constitutive plasmacytoid dendritic cell migration to the splenic white pulp is cooperatively regulated by CCR7‐ and CXCR4‐mediated signaling. J Immunol 2012;189:191–199.
    1. Yoshida R, Imai T, Hieshima K, Kusuda J, Baba M, Kitaura M, et al. Molecular cloning of a novel human CC chemokine EBI1‐ligand chemokine that is a specific functional ligand for EBI1, CCR7. J Biol Chem 1997;272:13803–13809.
    1. Yoshida R, Nagira M, Kitaura M, Imagawa N, Imai T, Yoshie O. Secondary lymphoid‐tissue chemokine is a functional ligand for the CC chemokine receptor CCR7. J Biol Chem 1998;273:7118–7122.
    1. Wang J, Seethala PR, Zang Q, Gooding W, van Waes C, Hasegawa H, et al. Autocrine and paracrine chemokine receptor 7 activation in head and neck cancer: implications for therapy. J Natl Cancer Inst 2008;100:502–512.
    1. Forster R, Davalos‐Misslitz AC, Rot A. CCR7 and its ligands: balancing immunity ad tolerance. Nat Rev Immunol 2008;8:362–371.
    1. Zhang B, Sun T, Xue L, Han X, Zhang B, Lu N, et al. Functional polymorphisms in FAS and FASL contribute to increased apoptosis of tumor infiltrating lymphocytes and risk of breast cancer. Carcinogenesis 2007;28:1067–1073.
    1. Strand S, Hofmann WJ, Hug H, Müller M, Otto G, Strand D, et al. Lymphocyte apoptosis induced by CD95 (APO‐1/FAS) ligand expressing tumor cells—a mechanism of immune evasion? Nat Med 1996;2:1361–1366.
    1. Paulsen M, Janssen O. Pro‐ and anti‐apoptotic CD95 signaling in T cells. Cell Commun Signal 2011;9:7–14.
    1. Yurchenko M, Shlapatska LM, Sidorenko SP. The multilevel regulation of CD95 signaling outcome. Exp Oncol 2012;34:153–159.
    1. von Haehling S, Morley JE, Coats AJS, Anker SD. Ethical guidelines for publishing in the Journal of Cachexia, Sarcopenia and Muscle: update 2017. J Cachexia Sarcopenia Muscle 2017;8:1081–1083.

Source: PubMed

3
Suscribir