Radiotherapy planning parameters correlate with changes in the peripheral immune status of patients undergoing curative radiotherapy for localized prostate cancer

Elgin Hoffmann, Frank Paulsen, Philipp Schaedle, Daniel Zips, Cihan Gani, Hans-Georg Rammensee, Cécile Gouttefangeas, Franziska Eckert, Elgin Hoffmann, Frank Paulsen, Philipp Schaedle, Daniel Zips, Cihan Gani, Hans-Georg Rammensee, Cécile Gouttefangeas, Franziska Eckert

Abstract

Purpose: The influence of radiotherapy on patient immune cell subsets has been established by several groups. Following a previously published analysis of immune changes during and after curative radiotherapy for prostate cancer, this analysis focused on describing correlations of changes of immune cell subsets with radiation treatment parameters.

Patients and methods: For 13 patients treated in a prospective trial with radiotherapy to the prostate region (primary analysis) and five patients treated with radiotherapy to prostate and pelvic nodal regions (exploratory analysis), already published immune monitoring data were correlated with clinical data as well as radiation planning parameters such as clinical target volume (CTV) and volumes receiving 20 Gy (V20) for newly contoured volumes of pelvic blood vessels and bone marrow.

Results: Most significant changes among immune cell subsets were observed at the end of radiotherapy. In contrast, correlations of age and CD8+ subsets (effector and memory cells) were observed early during and 3 months after radiotherapy. Ratios of T cells and T cell proliferation compared to baseline correlated with CTV. Early changes in regulatory T cells (Treg cells) and CD8+ effector T cells correlated with V20 of blood vessels and bone volumes.

Conclusions: Patient age as well as radiotherapy planning parameters correlated with immune changes during radiotherapy. Larger irradiated volumes seem to correlate with early suppression of anti-cancer immunity. For immune cell analysis during normofractionated radiotherapy and correlations with treatment planning parameters, different time points should be looked at in future projects.

Trial registration number: NCT01376674, 20.06.2011.

Keywords: DVH; IMRT; Immune status; Localized; Prostate cancer; T cells.

Conflict of interest statement

E. Hoffmann: Research and educational grants from Elekta, Philips, Siemens, and Sennewald. F. Paulsen: Research and educational grants from Elekta, Philips, Siemens, and Sennewald. P. Schaedle: No conflict of interest. D. Zips: Research and educational grants by Elekta, Philips, Siemens, Sennewald, Kaikuu, and TheraPanacea C. Gani: Research and educational grants, sponsoring for symposia by Elekta, Philips, Siemens, Sennewald, Kaikuu, and TheraPanacea. H.-G. Rammensee: No conflict of interest. C. Gouttefangeas: No conflict of interest. F. Eckert: Research and educational grants from Elekta, Philips, Siemens, and Sennewald. Speaker’s honoraria by Sennewald.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
To compare immune cell changes with radiation planning parameters, the ratio of immune cell subsets and proliferative fractions within each of these cell subsets is displayed for time point B (4 weeks into radiotherapy), time point C (end of radiotherapy) and time point D (3 months after end of radiotherapy) compared to baseline levels (time point A). Significant changes have been observed for proliferation of most cell subsets at time point B, most subsets at time point C, and only a few long-lasting effects at time point D. The red line indicates a ratio of 1 and thus no change compared to baseline frequencies. * indicates significant changes with one sample t-test compared to a ratio of 1
Fig. 2
Fig. 2
Correlation of patient age at time of radiotherapy and CD8+ effector cells as well as CD8+ memory cells is shown for all time points in the subgroup of patients treated with radiotherapy to the prostate. Patient age correlated positively with CD8+ effector cells and negatively with CD8+ memory cells at early and late time points (B/A and D/A, respectively)
Fig. 3
Fig. 3
Volumes contoured in addition to standard radiotherapy target volume (turquoise) and organs at risk such as bladder (yellow) and rectum (brown) are pelvic bone marrow (PBM union) comprising iliac bone marrow (red), lumbosacral spine (yellow) and lower pelvic bone marrow (light blue) as well as arteries (orange) and veins (dark blue) combined as vessels are shown
Fig. 4
Fig. 4
Dose-volume histograms of PBM union and vessels (upper panels) are shown for two patients comparing radiotherapy to the prostate only and radiotherapy including pelvic node irradiation. Correlations of CTV and V20 of vessels and PBM are shown for all patients. The clinical target volume (CTV) used for radiation planning and dose parameters for PBM and vessels showed no to moderate correlation to each other. Thus, the volume of CTV is not an adequate surrogate parameter for the dose to PBM and vessels (V20). V20 for PBM and V20 for vessels showed a moderate correlation
Fig. 5
Fig. 5
Changes of T cells and T cell proliferation are shown for time points B/A, C/A and D/A in the primary analysis of patients receiving radiotherapy to prostate only. T cells negatively correlated with CTV volume when comparing time points B and D, but not C, to A. T cell proliferation was positively correlated with CTV volume for ratio D/A, while having the lowest total ratio at this time point
Fig. 6
Fig. 6
Ratios of Treg cells as well as CD8+ effector cells are shown in correlation with V20 (blood vessels) and V20 (PBM union) for time points B/A, C/A and D/A. Treg cells correlated positively with V20 (blood vessels) as well as V20 (PBM) at time point B/A in the primary analysis. At the same time, CD8+ effector cell ratios showed a negative correlation with V20 (blood vessels) as well as V20 (PBM). No such correlation was found with the ratios at time points C/A or D/A

References

    1. Viani GA, Stefano EJ, Afonso SL. Higher-than-conventional radiation doses in localized prostate cancer treatment: a meta-analysis of randomized, controlled trials. Int J Radiat Oncol Biol Phys. 2009;74:1405–1418. doi: 10.1016/j.ijrobp.2008.10.091.
    1. Podder TK, Fredman ET, Ellis RJ. Advances in radiotherapy for prostate cancer treatment. Adv Exp Med Biol. 2018;1096:31–47. doi: 10.1007/978-3-319-99286-0_2.
    1. Mohler JL, Antonarakis ES, Armstrong AJ, et al. Prostate cancer, version 2.2019, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2019;17:479–505. doi: 10.6004/jnccn.2019.0023.
    1. Yang F, Markovic SN, Molina JR, et al. Association of sex, age, and eastern cooperative oncology group performance status with survival benefit of cancer immunotherapy in randomized clinical trials: a systematic review and meta-analysis. JAMA Netw Open. 2020;3:e2012534. doi: 10.1001/jamanetworkopen.2020.12534.
    1. Kim TJ, Koo KC. Current status and future perspectives of checkpoint inhibitor immunotherapy for prostate cancer: a comprehensive review. Int J Mol Sci. 2020 doi: 10.3390/ijms21155484.
    1. Comiskey MC, Dallos MC, Drake CG. Immunotherapy in prostate cancer: teaching an old dog new tricks. Curr Oncol Rep. 2018;20:75. doi: 10.1007/s11912-018-0712-z.
    1. Feyerabend S, Stevanovic S, Gouttefangeas C, et al. Novel multi-peptide vaccination in Hla-A2+ hormone sensitive patients with biochemical relapse of prostate cancer. Prostate. 2009;69:917–927. doi: 10.1002/pros.20941.
    1. Grassberger C, Ellsworth SG, Wilks MQ, Keane FK, Loeffler JS. Assessing the interactions between radiotherapy and antitumour immunity. Nat Rev Clin Oncol. 2019;16:729–745. doi: 10.1038/s41571-019-0238-9.
    1. Reynders K, De Ruysscher D. Radiotherapy and immunotherapy: improving cancer treatment through synergy. Prog Tumor Res. 2015;42:67–78. doi: 10.1159/000437185.
    1. Wang Y, Liu ZG, Yuan H, Deng W, Li J, Huang Y, Kim BYS, Story MD, Jiang W. The Reciprocity between radiotherapy and cancer immunotherapy. Clin Cancer Res. 2019;25:1709–1717. doi: 10.1158/1078-0432.Ccr-18-2581.
    1. Deutsch E, Chargari C, Galluzzi L, Kroemer G. Optimising efficacy and reducing toxicity of anticancer radioimmunotherapy. Lancet Oncol. 2019;20:e452–e463. doi: 10.1016/s1470-2045(19)30171-8.
    1. Dovedi SJ, Lipowska-Bhalla G, Beers SA, Cheadle EJ, Mu L, Glennie MJ, Illidge TM, Honeychurch J. Antitumor efficacy of radiation plus immunotherapy depends upon dendritic cell activation of effector CD8+ T Cells. Cancer Immunol Res. 2016;4:621–630. doi: 10.1158/2326-6066.Cir-15-0253.
    1. Harris TJ, Hipkiss EL, Borzillary S, et al. Radiotherapy augments the immune response to prostate cancer in a time-dependent manner. Prostate. 2008;68:1319–1329. doi: 10.1002/pros.20794.
    1. Vanpouille-Box C, Alard A, Aryankalayil MJ, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. 2017;8:15618. doi: 10.1038/ncomms15618.
    1. Eckert F, Gaipl US, Niedermann G, Hettich M, Schilbach K, Huber SM, Zips D. Beyond checkpoint inhibition - Immunotherapeutical strategies in combination with radiation. Clin Transl Radiat Oncol. 2017;2:29–35. doi: 10.1016/j.ctro.2016.12.006.
    1. Eckert F, Zwirner K, Boeke S, Thorwarth D, Zips D, Huber SM. Rationale for combining radiotherapy and immune checkpoint inhibition for patients with hypoxic tumors. Front Immunol. 2019;10:407. doi: 10.3389/fimmu.2019.00407.
    1. Donaubauer AJ, Rühle PF, Becker I, Fietkau R, Gaipl US, Frey B. One-tube multicolor flow cytometry assay (OTMA) for comprehensive immunophenotyping of peripheral blood. Methods Mol Biol. 2019;1904:189–212. doi: 10.1007/978-1-4939-8958-4_8.
    1. Neo SY, O'Reilly A, Pico de Coaña Y. Immune monitoring of cancer patients by multi-color flow cytometry. Methods Mol Biol. 2019;1913:49–65. doi: 10.1007/978-1-4939-8979-9_4.
    1. van Meir H, Nout RA, Welters MJ, et al. Impact of (chemo)radiotherapy on immune cell composition and function in cervical cancer patients. Oncoimmunology. 2016;6:e1267095. doi: 10.1080/2162402X.2016.1267095.
    1. Riemann D, Cwikowski M, Turzer S, Giese T, Grallert M, Schütte W, Seliger B. Blood immune cell biomarkers in lung cancer. Clin Exp Immunol. 2019;195:179–189. doi: 10.1111/cei.13219.
    1. Eckert F, Schaedle P, Zips D, Schmid-Horch B, Rammensee HG, Gani C, Gouttefangeas C. Impact of curative radiotherapy on the immune status of patients with localized prostate cancer. Oncoimmunology. 2018;7:e1496881. doi: 10.1080/2162402x.2018.1496881.
    1. Yan K, Ramirez E, Xie XJ, Gu X, Xi Y, Albuquerque K. Predicting severe hematologic toxicity from extended-field chemoradiation of para-aortic nodal metastases from cervical cancer. Pract Radiat Oncol. 2018;8:13–19. doi: 10.1016/j.prro.2017.07.001.
    1. Bazan JG, Luxton G, Mok EC, Koong AC, Chang DT. Normal tissue complication probability modeling of acute hematologic toxicity in patients treated with intensity-modulated radiation therapy for squamous cell carcinoma of the anal canal. Int J Radiat Oncol Biol Phys. 2012;84:700–706. doi: 10.1016/j.ijrobp.2011.12.072.
    1. Cozzarini C, Noris Chiorda B, Sini C, Fiorino C, Briganti A, Montorsi F, Di Muzio N. Hematologic toxicity in patients treated with postprostatectomy whole-pelvis irradiation with different intensity modulated radiation therapy techniques is not negligible and is prolonged: preliminary results of a longitudinal, observational study. Int J Radiat Oncol Biol Phys. 2016;95:690–695. doi: 10.1016/j.ijrobp.2016.01.022.
    1. Sini C, Fiorino C, Perna L, et al. Dose-volume effects for pelvic bone marrow in predicting hematological toxicity in prostate cancer radiotherapy with pelvic node irradiation. Radiother Oncol. 2016;118:79–84. doi: 10.1016/j.radonc.2015.11.020.
    1. Farhood B, Najafi M, Mortezaee K. CD8(+) cytotoxic T lymphocytes in cancer immunotherapy: a review. J Cell Physiol. 2019;234:8509–8521. doi: 10.1002/jcp.27782.
    1. Reading JL, Gálvez-Cancino F, Swanton C, Lladser A, Peggs KS, Quezada SA. The function and dysfunction of memory CD8(+) T cells in tumor immunity. Immunol Rev. 2018;283:194–212. doi: 10.1111/imr.12657.
    1. Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017;27:109–118. doi: 10.1038/cr.2016.151.
    1. Baum C, Alber M, Birkner M, Nusslin F. Robust treatment planning for intensity modulated radiotherapy of prostate cancer based on coverage probabilities. Radiother Oncol. 2006;78:27–35. doi: 10.1016/j.radonc.2005.09.005.
    1. Eckert F, Alloussi S, Paulsen F, et al. Prospective evaluation of a hydrogel spacer for rectal separation in dose-escalated intensity-modulated radiotherapy for clinically localized prostate cancer. BMC Cancer. 2013;13:27. doi: 10.1186/1471-2407-13-27.
    1. Li N, Liu X, Zhai F, Liu B, Cao X, Li S, Zhang M, Liu M. Association between dose-volume parameters and acute bone marrow suppression in rectal cancer patients treated with concurrent chemoradiotherapy. Oncotarget. 2017;8:92904–92913. doi: 10.18632/oncotarget.21646.
    1. Nakamura N, Kusunoki Y, Akiyama M. Radiosensitivity of CD4 or CD8 positive human T-lymphocytes by an in vitro colony formation assay. Radiat Res. 1990;123:224–227. doi: 10.2307/3577549.
    1. Kalina JL, Neilson DS, Comber AP, Rauw JM, Alexander AS, Vergidis J, Lum JJ. Immune modulation by androgen deprivation and radiation therapy: implications for prostate cancer immunotherapy. Cancers. 2017 doi: 10.3390/cancers9020013.
    1. Fülöp T, Larbi A, Pawelec G. Human T cell aging and the impact of persistent viral infections. Front Immunol. 2013;4:271. doi: 10.3389/fimmu.2013.00271.
    1. Schreiber K, Arina A, Engels B, et al. Spleen cells from young but not old immunized mice eradicate large established cancers. Clin Cancer Res. 2012;18:2526–2533. doi: 10.1158/1078-0432.Ccr-12-0127.
    1. Sekido K, Tomihara K, Tachinami H, Heshiki W, Sakurai K, Moniruzzaman R, Imaue S, Fujiwara K, Noguchi M. Alterations in composition of immune cells and impairment of anti-tumor immune response in aged oral cancer-bearing mice. Oral Oncol. 2019;99:104462. doi: 10.1016/j.oraloncology.2019.104462.
    1. Kugel CH, 3rd, Douglass SM, Webster MR, et al. Age Correlates with Response to Anti-PD1, Reflecting Age-Related Differences in Intratumoral Effector and Regulatory T-Cell Populations. Clin Cancer Res. 2018;24:5347–5356. doi: 10.1158/1078-0432.Ccr-18-1116.
    1. Wang S, Nie D, Qu L, Shao Y, Lian J, Wang Q, Shen D. CT Male pelvic organ segmentation via hybrid loss network with incomplete annotation. IEEE Trans Med Imaging. 2020;39:2151–2162. doi: 10.1109/tmi.2020.2966389.
    1. Jin JY, Mereniuk T, Yalamanchali A, Wang W, Machtay M, Spring Kong FM, Ellsworth S. A framework for modeling radiation induced lymphopenia in radiotherapy. Radiother Oncol. 2020;144:105–113. doi: 10.1016/j.radonc.2019.11.014.
    1. Wang X, Wang P, Zhao Z, Mao Q, Yu J, Li M. A review of radiation-induced lymphopenia in patients with esophageal cancer: an immunological perspective for radiotherapy. Ther Adv Med Oncol. 2020;12:1758835920926822. doi: 10.1177/1758835920926822.
    1. Lambin P, Lieverse RIY, Eckert F, Marcus D, Oberije C, van der Wiel AMA, Guha C, Dubois LJ, Deasy JO. Lymphocyte-sparing radiotherapy: the rationale for protecting lymphocyte-rich organs when combining radiotherapy with immunotherapy. Semin Radiat Oncol. 2020;30:187–193. doi: 10.1016/j.semradonc.2019.12.003.
    1. Sage EK, Schmid TE, Geinitz H, Gehrmann M, Sedelmayr M, Duma MN, Combs SE, Multhoff G. Effects of definitive and salvage radiotherapy on the distribution of lymphocyte subpopulations in prostate cancer patients. Strahlenther Onkol. 2017;193:648–655. doi: 10.1007/s00066-017-1144-7.
    1. Verma A, Mathur R, Farooque A, Kaul V, Gupta S, Dwarakanath BS. T-Regulatory Cells In Tumor Progression And Therapy. Cancer Manag Res. 2019;11:10731–10747. doi: 10.2147/cmar.S228887.
    1. Vacchelli E, Semeraro M, Enot DP, et al. Negative prognostic impact of regulatory T cell infiltration in surgically resected esophageal cancer post-radiochemotherapy. Oncotarget. 2015;6:20840–20850. doi: 10.18632/oncotarget.4428.
    1. Demaria S, Formenti SC. Role of T lymphocytes in tumor response to radiotherapy. Front Oncol. 2012;2:95. doi: 10.3389/fonc.2012.00095.
    1. Evans JD, Morris LK, Zhang H, et al. Prospective immunophenotyping of CD8(+) t cells and associated clinical outcomes of patients with oligometastatic prostate cancer treated with metastasis-directed SBRT. Int J Radiat Oncol Biol Phys. 2019;103:229–240. doi: 10.1016/j.ijrobp.2018.09.001.
    1. Gupta A, Probst HC, Vuong V, et al. Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation. Journal of immunology. 2012;189:558–66. doi: 10.4049/jimmunol.1200563.
    1. Grabenbauer GG, Lahmer G, Distel L, Niedobitek G. Tumor-infiltrating cytotoxic T cells but not regulatory T cells predict outcome in anal squamous cell carcinoma. Clin Cancer Res. 2006;12:3355–3360. doi: 10.1158/1078-0432.Ccr-05-2434.
    1. Friedrich T, Henthorn N, Durante M. Modeling radioimmune response-current status and perspectives. Front Oncol. 2021;11:647272. doi: 10.3389/fonc.2021.647272.
    1. Franco P, Ragona R, Arcadipane F, Mistrangelo M, Cassoni P, Rondi N, Morino M, Racca P, Ricardi U. Lumbar-sacral bone marrow dose modeling for acute hematological toxicity in anal cancer patients treated with concurrent chemo-radiation. Med Oncol. 2016;33:137. doi: 10.1007/s12032-016-0852-7.
    1. Franco P, Ragona R, Arcadipane F, Mistrangelo M, Cassoni P, Rondi N, Morino M, Racca P, Ricardi U. Dosimetric predictors of acute hematologic toxicity during concurrent intensity-modulated radiotherapy and chemotherapy for anal cancer. Clin Transl Oncol. 2017;19:67–75. doi: 10.1007/s12094-016-1504-2.
    1. Jianyang W, Yuan T, Yuan T, et al. A prospective phase II study of magnetic resonance imaging guided hematopoietical bone marrow-sparing intensity-modulated radiotherapy with concurrent chemotherapy for rectal cancer. Radiol Med. 2016;121:308–314. doi: 10.1007/s11547-015-0605-2.
    1. Kumar T, Schernberg A, Busato F, Laurans M, Fumagalli I, Dumas I, Deutsch E, Haie-Meder C, Chargari C. Correlation between pelvic bone marrow radiation dose and acute hematological toxicity in cervical cancer patients treated with concurrent chemoradiation. Cancer Manag Res. 2019;11:6285–6297. doi: 10.2147/cmar.S195989.
    1. Mell LK, Schomas DA, Salama JK, et al. Association between bone marrow dosimetric parameters and acute hematologic toxicity in anal cancer patients treated with concurrent chemotherapy and intensity-modulated radiotherapy. Int J Radiat Oncol Biol Phys. 2008;70:1431–1437. doi: 10.1016/j.ijrobp.2007.08.074.
    1. Rose B, Mitra D, Hong TS, et al. Irradiation of anatomically defined pelvic subsites and acute hematologic toxicity in anal cancer patients undergoing chemoradiation. Pract Radiat Oncol. 2017;7:e291–e297. doi: 10.1016/j.prro.2017.03.008.
    1. Mell LK, Tiryaki H, Ahn KH, Mundt AJ, Roeske JC, Aydogan B. Dosimetric comparison of bone marrow-sparing intensity-modulated radiotherapy versus conventional techniques for treatment of cervical cancer. Int J Radiat Oncol Biol Phys. 2008;71:1504–1510. doi: 10.1016/j.ijrobp.2008.04.046.
    1. Platta CS, Bayliss A, McHaffie D, Tome WA, Straub MR, Bradley KA. A dosimetric analysis of tomotherapy based intensity modulated radiation therapy with and without bone marrow sparing in gynecologic malignancies. Technol Cancer Res Treat. 2013;12:19–29. doi: 10.7785/tcrt.2012.500300.

Source: PubMed

3
購読する