Combination immunotherapy using G-CSF and oncolytic virotherapy reduces tumor growth in osteosarcoma

Alvaro Morales-Molina, Stefano Gambera, Angela Leo, Javier García-Castro, Alvaro Morales-Molina, Stefano Gambera, Angela Leo, Javier García-Castro

Abstract

Background: Osteosarcoma is the most common malignant solid tumor that affects bones, however, survival rates of patients with relapsed osteosarcoma have not improved in the last 30 years. Oncolytic virotherapy, which uses viruses designed to selectively replicate in cancer cells, has emerged as a promising treatment for solid tumors. Our group uses mesenchymal stem cells (MSCs) to transport oncolytic adenoviruses (OAds) to the tumor site, a therapeutic strategy called Celyvir. This treatment has been already applied in human patients, canine patients and different mouse models. In parallel, previous results have probed that administration of granulocyte-colony stimulating factor (G-CSF) increased immune infiltration in tumors. We then hypothesized that the mobilization of immune cells by G-CSF may increase the antitumor efficacy of Celyvir treatment by increasing the immune infiltration into the tumors.

Methods: In this study, we use a murine version of Celyvir consisting in murine MSCs carrying the murine OAd dlE102-here called OAd-MSCs-in an immunocompetent model of osteosarcoma. We tested the antitumoral efficacy of the combination of OAd-MSCs plus G-CSF.

Results: Our results show that treatment with OAd-MSCs or the union of OAd-MSCs with G-CSF (Combination) significantly reduced tumor growth of osteosarcoma in vivo. Moreover, treated tumors presented higher tumor infiltration of immune cells-especially tumor-infiltrating lymphocytes-and reduced T cell exhaustion, which seems to be enhanced in tumors treated with the Combination. The comparison of our results to those obtained from a cohort of pediatric osteosarcoma patients showed that the virotherapy induces immunological changes similar to those observed in patients with good prognosis.

Conclusions: The results open the possibility of using cellular virotherapy for the treatment of bone cancers. Indeed, its combination with G-CSF may be considered for the improvement of the therapy.

Trial registration: ClinicalTrials.gov NCT01844661.

Keywords: immunotherapy; lymphocytes; oncolytic virotherapy; programmed cell death 1 receptor; t-lymphocytes; tumor-infiltrating.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
In vitro oncolytic effect of oncolytic adenovirus dlE102 in murine osteosarcoma cell lines. (A) For flow cytometry viability assay, cells were infected at different MOI (1–50) and cytotoxicity was measured 7 days later using LIVE/DEAD viability kit, showing red fluorescence for dead cells and green fluorescence for live cells. (B) Percentages refers to dead cells and statistical differences are compared with control group. (C) For luciferase viability assay, K5 cells and p53−/−Rb−/− MSCs were transduced with a lentiviral vector containing a firefly luciferase cassette and infected at MOI 1 and 10. Luminescence of alive cells was quantified 4 days later and expressed as infected/control ratio. (D) Bright field pictures show the cytopathic effect of K5 cells and p53−/−Rb−/− MSCs 4 days after infection with dlE102 at MOI 10. The experiment was repeated twice (N=2). Unpaired t-test. *p<0.05, **p<0.01, ***p<0.001. MOI, multiplicity of infection; MSCs, mesenchymal stem cells.
Figure 2
Figure 2
In vivo antitumor efficacy of OAd-MSCs and Combination in osteosarcoma. (A) Graph represents the experimental design and antitumor effect of OAd-MSCs and Combination in BALB/c mice bearing subcutaneous K5 tumors. Lines represent the mean+SEM of tumor volume in groups treated with PBS (black, n=11), MSCs (gray, n=4), G-CSF (dark gray, n=9), OAd-MSCs (blue, n=9) or Combination (red, n=7). (B) Representative pictures and tumor volumes at end point. Mann-Whitney U test. (C) Follow-up of tumor volume in mice treated with MSCs, G-CSF, OAd-MSCs or Combination (individual values) and PBS group (mean+SEM, black line). (D, E) Tumor weight and tumor growth at end point. Mann-Whitney U test. (F) Kaplan-Meier survival curve of treated mice. Log-rank (Mantel-Cox) test. *p

Figure 3

Tumor-infiltrating immune cells in osteosarcoma.…

Figure 3

Tumor-infiltrating immune cells in osteosarcoma. (A) Density of tumor-infiltrating leukocytes, NK cells, and…

Figure 3
Tumor-infiltrating immune cells in osteosarcoma. (A) Density of tumor-infiltrating leukocytes, NK cells, and adaptive immune cells at end point expressed as percentage per gram of tumor. CD4+/CD8+ ratio is also represented. (B, C) Percentage of tumor-infiltrating lymphocytes, CD4+ and CD8+ T cells expressing PD-1 or CD137. (D) Density of tumor-infiltrating innate immune cells. (E) Ratio of pro-inflammatory/anti-inflammatory status of macrophages (M1/M2) and neutrophils (N1/N2). Mann-Whitney U test (n=3–9). *p<0.05. G-CSF, granulocyte-colony stimulating factor; LMR, lymphocyte-monocyte ratio; MSCs, mesenchymal stem cells; NLR, neutrophil-lymphocyte ratio; NK, natural killer; OAd-MSCs, MSCs carrying the murine oncolytic adenovirus dlE102; PBS, phosphate-buffered saline; PD-1, programmed cell death protein 1

Figure 4

Leukocyte infiltration and tumor microenvironment…

Figure 4

Leukocyte infiltration and tumor microenvironment in osteosarcoma after treatment with OAd-MSCs and Combination.…

Figure 4
Leukocyte infiltration and tumor microenvironment in osteosarcoma after treatment with OAd-MSCs and Combination. (A–C) Representative images of immunohistochemistry of CD45+ cells in osteosarcoma tumors treated with PBS (A), OAd-MSCs (B) or Combination (C). Upper panel shows images of the periphery, while lower panel shows images of the core of the tumors. (D) Infiltration of CD45+ cells in tumors quantified in 40× high-power fields. (E) Protein expression of CD45 and TBP in tumors treated with PBS, OAd-MSCs or Combination. (F) Heat-map showing the pro-inflammatory cytokine array performed in tumors treated with PBS, OAd-MSCs or Combination. White represents the lowest expression while dark blue represents the highest expression. (G) Quantification by integrated density of pro-inflammatory cytokines differentially expressed in tumors treated with PBS, OAd-MSCs or Combination. Unpaired t-test. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001. OAd-MSCs, mesenchymal stem cells carrying the murine oncolytic adenovirus dlE102; PBS, phosphate-buffered saline; TBP, TATA-box binding protein.

Figure 5

Histopathological features in osteosarcoma after…

Figure 5

Histopathological features in osteosarcoma after treatment with OAd-MSCs or Combination. (A) Representative X-ray…

Figure 5
Histopathological features in osteosarcoma after treatment with OAd-MSCs or Combination. (A) Representative X-ray imaging of osteosarcoma tumors treated with PBS, G-CSF, OAd-MSCs or Combination. Mineralized area is marked in red, while cellular area is marked in yellow. (B) Percentage of cellular tumor measured in X-ray images. (C) Representative images of Masson’s-Trichrome staining. (D) Percentage of cellular tumor measured from Masson’s stained slide images. Unpaired t-test. *p

Figure 6

Comparison of preclinical results in…

Figure 6

Comparison of preclinical results in the murine osteosarcoma model with clinical osteosarcoma patients.…

Figure 6
Comparison of preclinical results in the murine osteosarcoma model with clinical osteosarcoma patients. (A) Graph represents the experimental flow followed for the genomic analysis of osteosarcoma patients. (B) Hazard ratio of protective (1, red) regarding immune cell populations present in osteosarcoma tumors. (C) Kaplan-Meier survival curve of osteosarcoma patients regarding high (purple) or low (yellow) levels of different immune biomarkers. Log-rank (Mantel-Cox) test. (D) Violin plots represent differences in NLR, CD4+/CD8+ ratio, T cells and CD8+ T cells in patients according to their current clinical status (dead in orange; live in green). (E) Violin plots represent differences in NLR, CD4+/CD8+ ratio, T cells and CD8+ T cells in mice treated with PBS, OAd-MSCs or Combination. Mann-Whitney U test. *p<0.05. NLR, neutrophil-lymphocyte ratio; OAd-MSCs, mesenchymal stem cells carrying the murine oncolytic adenovirus dlE102; PBS, phosphate-buffered saline.
Similar articles
Cited by
References
    1. Valery PC, Laversanne M, Bray F. Bone cancer incidence by morphological subtype: a global assessment. Cancer Causes Control 2015;26:1127–39. 10.1007/s10552-015-0607-3 - DOI - PubMed
    1. ESMO/European Sarcoma Network Working Group . Bone sarcomas: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2014;25 Suppl 3:iii113–23. 10.1093/annonc/mdu256 - DOI - PubMed
    1. Grünewald TG, Alonso M, Avnet S, et al. . Sarcoma treatment in the era of molecular medicine. EMBO Mol Med 2020;12:e11131. 10.15252/emmm.201911131 - DOI - PMC - PubMed
    1. Savage SA, Mirabello L. Using epidemiology and genomics to understand osteosarcoma etiology. Sarcoma 2011;2011:1–13. 10.1155/2011/548151 - DOI - PMC - PubMed
    1. Duong LM, Richardson LC. Descriptive epidemiology of malignant primary osteosarcoma using population-based registries, United States, 1999-2008. J Registry Manag 2013;40:59–64. - PMC - PubMed
Show all 58 references
Publication types
MeSH terms
Substances
Associated data
Related information
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 3
Figure 3
Tumor-infiltrating immune cells in osteosarcoma. (A) Density of tumor-infiltrating leukocytes, NK cells, and adaptive immune cells at end point expressed as percentage per gram of tumor. CD4+/CD8+ ratio is also represented. (B, C) Percentage of tumor-infiltrating lymphocytes, CD4+ and CD8+ T cells expressing PD-1 or CD137. (D) Density of tumor-infiltrating innate immune cells. (E) Ratio of pro-inflammatory/anti-inflammatory status of macrophages (M1/M2) and neutrophils (N1/N2). Mann-Whitney U test (n=3–9). *p<0.05. G-CSF, granulocyte-colony stimulating factor; LMR, lymphocyte-monocyte ratio; MSCs, mesenchymal stem cells; NLR, neutrophil-lymphocyte ratio; NK, natural killer; OAd-MSCs, MSCs carrying the murine oncolytic adenovirus dlE102; PBS, phosphate-buffered saline; PD-1, programmed cell death protein 1
Figure 4
Figure 4
Leukocyte infiltration and tumor microenvironment in osteosarcoma after treatment with OAd-MSCs and Combination. (A–C) Representative images of immunohistochemistry of CD45+ cells in osteosarcoma tumors treated with PBS (A), OAd-MSCs (B) or Combination (C). Upper panel shows images of the periphery, while lower panel shows images of the core of the tumors. (D) Infiltration of CD45+ cells in tumors quantified in 40× high-power fields. (E) Protein expression of CD45 and TBP in tumors treated with PBS, OAd-MSCs or Combination. (F) Heat-map showing the pro-inflammatory cytokine array performed in tumors treated with PBS, OAd-MSCs or Combination. White represents the lowest expression while dark blue represents the highest expression. (G) Quantification by integrated density of pro-inflammatory cytokines differentially expressed in tumors treated with PBS, OAd-MSCs or Combination. Unpaired t-test. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001. OAd-MSCs, mesenchymal stem cells carrying the murine oncolytic adenovirus dlE102; PBS, phosphate-buffered saline; TBP, TATA-box binding protein.
Figure 5
Figure 5
Histopathological features in osteosarcoma after treatment with OAd-MSCs or Combination. (A) Representative X-ray imaging of osteosarcoma tumors treated with PBS, G-CSF, OAd-MSCs or Combination. Mineralized area is marked in red, while cellular area is marked in yellow. (B) Percentage of cellular tumor measured in X-ray images. (C) Representative images of Masson’s-Trichrome staining. (D) Percentage of cellular tumor measured from Masson’s stained slide images. Unpaired t-test. *p

Figure 6

Comparison of preclinical results in…

Figure 6

Comparison of preclinical results in the murine osteosarcoma model with clinical osteosarcoma patients.…

Figure 6
Comparison of preclinical results in the murine osteosarcoma model with clinical osteosarcoma patients. (A) Graph represents the experimental flow followed for the genomic analysis of osteosarcoma patients. (B) Hazard ratio of protective (1, red) regarding immune cell populations present in osteosarcoma tumors. (C) Kaplan-Meier survival curve of osteosarcoma patients regarding high (purple) or low (yellow) levels of different immune biomarkers. Log-rank (Mantel-Cox) test. (D) Violin plots represent differences in NLR, CD4+/CD8+ ratio, T cells and CD8+ T cells in patients according to their current clinical status (dead in orange; live in green). (E) Violin plots represent differences in NLR, CD4+/CD8+ ratio, T cells and CD8+ T cells in mice treated with PBS, OAd-MSCs or Combination. Mann-Whitney U test. *p<0.05. NLR, neutrophil-lymphocyte ratio; OAd-MSCs, mesenchymal stem cells carrying the murine oncolytic adenovirus dlE102; PBS, phosphate-buffered saline.
Figure 6
Figure 6
Comparison of preclinical results in the murine osteosarcoma model with clinical osteosarcoma patients. (A) Graph represents the experimental flow followed for the genomic analysis of osteosarcoma patients. (B) Hazard ratio of protective (1, red) regarding immune cell populations present in osteosarcoma tumors. (C) Kaplan-Meier survival curve of osteosarcoma patients regarding high (purple) or low (yellow) levels of different immune biomarkers. Log-rank (Mantel-Cox) test. (D) Violin plots represent differences in NLR, CD4+/CD8+ ratio, T cells and CD8+ T cells in patients according to their current clinical status (dead in orange; live in green). (E) Violin plots represent differences in NLR, CD4+/CD8+ ratio, T cells and CD8+ T cells in mice treated with PBS, OAd-MSCs or Combination. Mann-Whitney U test. *p<0.05. NLR, neutrophil-lymphocyte ratio; OAd-MSCs, mesenchymal stem cells carrying the murine oncolytic adenovirus dlE102; PBS, phosphate-buffered saline.

References

    1. Valery PC, Laversanne M, Bray F. Bone cancer incidence by morphological subtype: a global assessment. Cancer Causes Control 2015;26:1127–39. 10.1007/s10552-015-0607-3
    1. ESMO/European Sarcoma Network Working Group . Bone sarcomas: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2014;25 Suppl 3:iii113–23. 10.1093/annonc/mdu256
    1. Grünewald TG, Alonso M, Avnet S, et al. . Sarcoma treatment in the era of molecular medicine. EMBO Mol Med 2020;12:e11131. 10.15252/emmm.201911131
    1. Savage SA, Mirabello L. Using epidemiology and genomics to understand osteosarcoma etiology. Sarcoma 2011;2011:1–13. 10.1155/2011/548151
    1. Duong LM, Richardson LC. Descriptive epidemiology of malignant primary osteosarcoma using population-based registries, United States, 1999-2008. J Registry Manag 2013;40:59–64.
    1. Kansara M, Teng MW, Smyth MJ, et al. . Translational biology of osteosarcoma. Nat Rev Cancer 2014;14:722–35. 10.1038/nrc3838
    1. Garcia-Moure M, Martinez-Vélez N, Patiño-García A, et al. . Oncolytic adenoviruses as a therapeutic approach for osteosarcoma: a new hope. J Bone Oncol 2017;9:41–7. 10.1016/j.jbo.2016.12.001
    1. Alonso MM, Cascallo M, Gomez-Manzano C, et al. . ICOVIR-5 shows E2F1 addiction and potent antiglioma effect in vivo. Cancer Res 2007;67:8255–63. 10.1158/0008-5472.CAN-06-4675
    1. Cascallo M, Alonso MM, Rojas JJ, et al. . Systemic toxicity-efficacy profile of ICOVIR-5, a potent and selective oncolytic adenovirus based on the pRB pathway. Mol Ther 2007;15:1607–15. 10.1038/sj.mt.6300239
    1. Kidd S, Spaeth E, Dembinski JL, et al. . Direct evidence of mesenchymal stem cell tropism for tumor and wounding microenvironments using in vivo bioluminescent imaging. Stem Cells 2009;27:2614–23. 10.1002/stem.187
    1. Ramírez M, García-Castro J, Melen GJ, et al. . Patient-Derived mesenchymal stem cells as delivery vehicles for oncolytic virotherapy: novel state-of-the-art technology. Oncolytic Virother 2015;4:149–55. 10.2147/OV.S66010
    1. Hadryś A, Sochanik A, McFadden G, et al. . Mesenchymal stem cells as carriers for systemic delivery of oncolytic viruses. Eur J Pharmacol 2020;874:172991. 10.1016/j.ejphar.2020.172991
    1. Mahasa KJ, de Pillis L, Ouifki R, et al. . Mesenchymal stem cells used as carrier cells of oncolytic adenovirus results in enhanced oncolytic virotherapy. Sci Rep 2020;10:425. 10.1038/s41598-019-57240-x
    1. García-Castro J, Alemany R, Cascalló M, et al. . Treatment of metastatic neuroblastoma with systemic oncolytic virotherapy delivered by autologous mesenchymal stem cells: an exploratory study. Cancer Gene Ther 2010;17:476–83. 10.1038/cgt.2010.4
    1. Melen GJ, Franco-Luzón L, Ruano D, et al. . Influence of carrier cells on the clinical outcome of children with neuroblastoma treated with high dose of oncolytic adenovirus delivered in mesenchymal stem cells. Cancer Lett 2016;371:161–70. 10.1016/j.canlet.2015.11.036
    1. Ramirez M, Ruano D, Moreno L, et al. . First-in-child trial of celyvir (autologous mesenchymal stem cells carrying the oncolytic virus ICOVIR-5) in patients with relapsed and refractory pediatric solid tumors. Journal of Clinical Oncology 2018;36:10543–43. 10.1200/JCO.2018.36.15_suppl.10543
    1. Ruano D, López-Martín JA, Moreno L, et al. . First-in-human, first-in-child trial of autologous MscS carrying the oncolytic virus Icovir-5 in patients with advanced tumors. Mol Ther 2020;28:1033–42. 10.1016/j.ymthe.2020.01.019
    1. Laborda E, Puig-Saus C, Rodriguez-García A, et al. . A pRb-responsive, RGD-modified, and hyaluronidase-armed canine oncolytic adenovirus for application in veterinary oncology. Mol Ther 2014;22:986–98. 10.1038/mt.2014.7
    1. Cejalvo T, Perisé-Barrios AJ, Del Portillo I, et al. . Remission of spontaneous canine tumors after systemic cellular Viroimmunotherapy. Cancer Res 2018;78:4891–901. 10.1158/0008-5472.CAN-17-3754
    1. Martinez-Velez N, Xipell E, Jauregui P, et al. . The oncolytic adenovirus Δ24-RGD in combination with cisplatin exerts a potent anti-osteosarcoma activity. J Bone Miner Res 2014;29:2287–96. 10.1002/jbmr.2253
    1. Geiss C, Kis Z, Leuchs B, et al. . Preclinical testing of an oncolytic parvovirus: standard Protoparvovirus H-1PV efficiently induces osteosarcoma cell lysis in vitro. Viruses 2017;9. 10.3390/v9100301. [Epub ahead of print: 17 10 2017].
    1. Jeong S-N, Yoo SY. Novel oncolytic virus armed with cancer suicide gene and normal vasculogenic gene for improved anti-tumor activity. Cancers 2020;12. 10.3390/cancers12051070. [Epub ahead of print: 25 04 2020].
    1. Rincón E, Cejalvo T, Kanojia D, et al. . Mesenchymal stem cell carriers enhance antitumor efficacy of oncolytic adenoviruses in an immunocompetent mouse model. Oncotarget 2017;8:45415–31. 10.18632/oncotarget.17557
    1. Morales-Molina Álvaro, Gambera S, Cejalvo T, et al. . Antitumor virotherapy using syngeneic or allogeneic mesenchymal stem cell carriers induces systemic immune response and intratumoral leukocyte infiltration in mice. Cancer Immunol Immunother 2018;67:1589–602. 10.1007/s00262-018-2220-2
    1. Morales-Molina A, Rodríguez-Milla Miguel Ángel, Gimenez-Sanchez A, et al. . Cellular Virotherapy Increases Tumor-Infiltrating Lymphocytes (TIL) and Decreases their PD-1+ Subsets in Mouse Immunocompetent Models. Cancers 2020;12. 10.3390/cancers12071920. [Epub ahead of print: 16 07 2020].
    1. Franco-Luzón L, González-Murillo África, Alcántara-Sánchez C, et al. . Systemic oncolytic adenovirus delivered in mesenchymal carrier cells modulate tumor infiltrating immune cells and tumor microenvironment in mice with neuroblastoma. Oncotarget 2020;11:347–61. 10.18632/oncotarget.27401
    1. Robinson M, Li B, Ge Y, et al. . Novel immunocompetent murine tumor model for evaluation of conditionally replication-competent (oncolytic) murine adenoviral vectors. J Virol 2009;83:3450–62. 10.1128/JVI.02561-08
    1. Chen X, Bahrami A, Pappo A, et al. . Recurrent somatic structural variations contribute to tumorigenesis in pediatric osteosarcoma. Cell Rep 2014;7:104–12. 10.1016/j.celrep.2014.03.003
    1. Lyman GH, Yau L, Nakov R, et al. . Overall survival and risk of second malignancies with cancer chemotherapy and G-CSF support. Ann Oncol 2018;29:1903–10. 10.1093/annonc/mdy311
    1. Lewis IJ, Nooij MA, Whelan J, et al. . Improvement in histologic response but not survival in osteosarcoma patients treated with intensified chemotherapy: a randomized phase III trial of the European osteosarcoma intergroup. J Natl Cancer Inst 2007;99:112–28. 10.1093/jnci/djk015
    1. Lyman GH, Dale DC, Culakova E, et al. . The impact of the granulocyte colony-stimulating factor on chemotherapy dose intensity and cancer survival: a systematic review and meta-analysis of randomized controlled trials. Ann Oncol 2013;24:2475–84. 10.1093/annonc/mdt226
    1. Espinoza ML. Movilización de linfocitos infiltrantes de tumor a sangre periférica en un modelo murino de neuroblastoma. Análisis inmunofenotípico, clonal Y funcional. Madrid: Universidad Autónoma de Madrid, 2020.
    1. Wang Z, Li B, Ren Y, et al. . T-Cell-based immunotherapy for osteosarcoma: challenges and opportunities. Front Immunol 2016;7:353. 10.3389/fimmu.2016.00353
    1. Li J, Byrne KT, Yan F, et al. . Tumor cell-intrinsic factors underlie heterogeneity of immune cell infiltration and response to immunotherapy. Immunity 2018;49:178–93. 10.1016/j.immuni.2018.06.006
    1. Zhang J, Endres S, Kobold S. Enhancing tumor T cell infiltration to enable cancer immunotherapy. Immunotherapy 2019;11:201–13. 10.2217/imt-2018-0111
    1. Schmidt J, Strauss GP, Schön A, et al. . Establishment and characterization of osteogenic cell lines from a spontaneous murine osteosarcoma. Differentiation 1988;39:151–60. 10.1111/j.1432-0436.1988.tb00090.x
    1. Rubio R, Abarrategi A, Garcia-Castro J, et al. . Bone environment is essential for osteosarcoma development from transformed mesenchymal stem cells. Stem Cells 2014;32:1136–48. 10.1002/stem.1647
    1. Gambera S, Abarrategi A, González-Camacho F, et al. . Clonal dynamics in osteosarcoma defined by RGB marking. Nat Commun 2018;9:3994. 10.1038/s41467-018-06401-z
    1. Smith K, Ying B, Ball AO, et al. . Interaction of mouse adenovirus type 1 early region 1A protein with cellular proteins pRb and p107. Virology 1996;224:184–97. 10.1006/viro.1996.0520
    1. Miao Y-R, Zhang Q, Lei Q, et al. . ImmuCellAI: a unique method for comprehensive T-cell subsets abundance prediction and its application in cancer immunotherapy. Adv Sci 2020;7:1902880. 10.1002/advs.201902880
    1. Rupji M, Zhang X, Kowalski J. CASAS: cancer survival analysis suite, a web based application. F1000Res 2017;6:919. 10.12688/f1000research.11830.1
    1. Li X, Wang P, Li H, et al. . The efficacy of oncolytic adenovirus is mediated by T-cell responses against virus and tumor in Syrian hamster model. Clin Cancer Res 2017;23:239–49. 10.1158/1078-0432.CCR-16-0477
    1. Liu B, Huang Y, Sun Y, et al. . Prognostic value of inflammation-based scores in patients with osteosarcoma. Sci Rep 2016;6:39862. 10.1038/srep39862
    1. Heymann M-F, Lézot F, Heymann D. The contribution of immune infiltrates and the local microenvironment in the pathogenesis of osteosarcoma. Cell Immunol 2019;343:103711. 10.1016/j.cellimm.2017.10.011
    1. Galon J, Pagès F, Marincola FM, et al. . Cancer classification using the immunoscore: a worldwide Task force. J Transl Med 2012;10:205. 10.1186/1479-5876-10-205
    1. Wu C-C, Beird HC, Andrew Livingston J, et al. . Immuno-genomic landscape of osteosarcoma. Nat Commun 2020;11:1008. 10.1038/s41467-020-14646-w
    1. Gomez-Brouchet A, Illac C, Gilhodes J, et al. . CD163-positive tumor-associated macrophages and CD8-positive cytotoxic lymphocytes are powerful diagnostic markers for the therapeutic stratification of osteosarcoma patients: an immunohistochemical analysis of the biopsies fromthe French OS2006 phase 3 trial. Oncoimmunology 2017;6:e1331193. 10.1080/2162402X.2017.1331193
    1. Granier C, Dariane C, Combe P, et al. . Tim-3 expression on Tumor-infiltrating PD-1+CD8+ T cells correlates with poor clinical outcome in renal cell carcinoma. Cancer Res 2017;77:1075–82. 10.1158/0008-5472.CAN-16-0274
    1. Kollmann D, Schweiger T, Schwarz S, et al. . PD1-positive tumor-infiltrating lymphocytes are associated with poor clinical outcome after pulmonary metastasectomy for colorectal cancer. Oncoimmunology 2017;6:e1331194. 10.1080/2162402X.2017.1331194
    1. Paget C, Duret H, Ngiow SF, et al. . Studying the role of the immune system on the antitumor activity of a hedgehog inhibitor against murine osteosarcoma. Oncoimmunology 2012;1:1313–22. 10.4161/onci.21680
    1. Melero I, Shuford WW, Newby SA, et al. . Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nat Med 1997;3:682–5. 10.1038/nm0697-682
    1. Feist M, Zhu Z, Dai E, et al. . Oncolytic virus promotes tumor-reactive infiltrating lymphocytes for adoptive cell therapy. Cancer Gene Ther 2020. 10.1038/s41417-020-0189-4. [Epub ahead of print: 07 Jul 2020].
    1. Savola S, Klami A, Myllykangas S, et al. . High expression of complement component 5 (C5) at tumor site associates with superior survival in Ewing's sarcoma family of tumour patients. ISRN Oncol 2011;2011:1–10. 10.5402/2011/168712
    1. Corre I, Verrecchia F, Crenn V, et al. . The osteosarcoma microenvironment: a complex but targetable ecosystem. Cells 2020;9:976. 10.3390/cells9040976
    1. Takeshima T, Pop LM, Laine A, et al. . Key role for neutrophils in radiation-induced antitumor immune responses: potentiation with G-CSF. Proc Natl Acad Sci U S A 2016;113:11300–5. 10.1073/pnas.1613187113
    1. Du R, Hu P, Liu Q, et al. . Granulocyte colony-stimulating factor treatment during radiotherapy is associated with survival benefit in patients with lung cancer. Technol Cancer Res Treat 2018;17:153303381881607. 10.1177/1533033818816076
    1. Charrier S, Blundell M, Cédrone G, et al. . Wiskott-Aldrich syndrome protein-deficient hematopoietic cells can be efficiently mobilized by granulocyte colony-stimulating factor. Haematologica 2013;98:1300–8. 10.3324/haematol.2012.077040
    1. Winkler IG, Bendall LJ, Forristal CE, et al. . B-Lymphopoiesis is stopped by mobilizing doses of G-CSF and is rescued by overexpression of the anti-apoptotic protein BCL2. Haematologica 2013;98:325–33. 10.3324/haematol.2012.069260

Source: PubMed

3
Abonner