A hypofractionated radiation regimen avoids the lymphopenia associated with neoadjuvant chemoradiation therapy of borderline resectable and locally advanced pancreatic adenocarcinoma

Todd Crocenzi, Benjamin Cottam, Pippa Newell, Ronald F Wolf, Paul D Hansen, Chet Hammill, Matthew C Solhjem, Yue-Yun To, Amy Greathouse, Garth Tormoen, Zeljka Jutric, Kristina Young, Keith S Bahjat, Michael J Gough, Marka R Crittenden, Todd Crocenzi, Benjamin Cottam, Pippa Newell, Ronald F Wolf, Paul D Hansen, Chet Hammill, Matthew C Solhjem, Yue-Yun To, Amy Greathouse, Garth Tormoen, Zeljka Jutric, Kristina Young, Keith S Bahjat, Michael J Gough, Marka R Crittenden

Abstract

Background: Preclinical studies have shown synergy between radiation therapy and immunotherapy. However, in almost all preclinical models, radiation is delivered in single doses or short courses of high doses (hypofractionated radiation). By contrast in most clinical settings, radiation is delivered as standard small daily fractions of 1.8-2 Gy to achieve total doses of 50-54 Gy (fractionated radiation). We do not yet know the optimal dose and scheduling of radiation for combination with chemotherapy and immunotherapy.

Methods: To address this, we analyzed the effect of neoadjuvant standard fractionated and hypofractionated chemoradiation on immune cells in patients with locally advanced and borderline resectable pancreatic adenocarcinoma.

Results: We found that standard fractionated chemoradiation resulted in a significant and extended loss of lymphocytes that was not explained by a lack of homeostatic cytokines or response to cytokines. By contrast, treatment with hypofractionated radiation therapy avoided the loss of lymphocytes associated with conventional fractionation.

Conclusion: Hypofractionated neoadjuvant chemoradiation is associated with reduced systemic loss of T cells.

Trial registration: ClinicalTrials.gov NCT01342224, April 21, 2011; NCT01903083, July 2, 2013.

Keywords: Chemotherapy; Fractionation; Gemcitabine; Homeostatic repopulation; IL-15; IL-7; Immunotherapy; Lymphocytes; Lymphodepletion; Radiation.

Figures

Fig. 1
Fig. 1
Effect of conventional neoadjuvant chemoradiation on immune cells in patient blood. a Absolute numbers of i) CD3+, ii) CD8+, iii) CD4+ and iv) CD4+CD25+ T cells as well as v) SSCintCD14+ monocytes and vi) SSChiCD15+ granulocytes by flow cytometry of fresh peripheral blood over the course of the study. Individual patients are gray, the mean is black. Dotted lines show sampling times and shows periods of neoadjuvant and adjuvant chemotherapy (red rectangles), neoadjuvant chemoradiation (blue bar) according to the trial schema provided in Additional file 1: Figure S1. b Analysis of the proportions of i) CD4 and ii) CD8 subpopulations in PBMC collected pretreatment (open circles) and post-treatment (closed circles). Key: NS – not significant. * p < 0.05
Fig. 2
Fig. 2
Role of homeostatic repopulation through cytokines and cytokine responses. a Patient serum collected prior to treatment (pre) and at the end of treatment (post) were tested for cytokine levels by multiplex assay. Individual patients are gray, the mean is black. b-d PBMC collected prior to treatment and at the end of treatment were treated with a range of cytokines for and analyzed for i) pSTAT1, ii) pSTAT3 or iii) pSTAT5 expression by intracellular flow cytometry. Surface staining of these mixed populations identified pSTAT activation in (b) CD4+ T cells, (c) CD8+ T cells or (d) CD14+ monocytes. Individual patient’s pre and post treatment values are connected. Graphs show change in pSTAT MFI over control (vehicle alone) stimulated cells. Colors highlight STATs that respond to a particular stimulation in each cell type. Key: NS – not significant. * p < 0.05, ** p < 0.01
Fig. 3
Fig. 3
Effect of hypofraction on immune cells in patient blood. a Absolute numbers of i) CD3+, ii) CD8+, iii) CD4+ and iv) SSCintCD14+ monocytes by flow cytometry of fresh peripheral blood in patients on a conventionally fractionated regimen (Fract, open symbols) versus a hypofractionated regimen (Hypo, closed symbols). The samples immediately pre-RT and immediately post-RT are highlighted on the first graph. b comparison of absolute numbers of i) CD3+, ii) CD8+, iii) CD4+ and iv) SSCintCD14+ monocytes by flow cytometry of fresh peripheral blood immediately following completion of radiation therapy in patients on a conventionally fractionated regimen (Fract, open symbols) versus a hypofractionated regimen (Hypo, closed symbols). c Analysis of the proportions of i) CD4 and ii) CD8 subpopulations in PBMC collected pretreatment (open circles) and post-treatment with a hypofractionated regimen (closed circles)
Fig. 4
Fig. 4
Effect of treatment regimen on early homeostatic cytokines. Patient serum collected prior to treatment (pre), immediately following completion of radiation therapy (post) and at the end of treatment (end) from patients on i) a conventionally fractionated regimen (Fract) versus ii) a hypofractionated regimen (Hypo) were tested for (a) IL-7 and (b) IL-15 cytokine levels by ultrasensitive multiplex assay. Key: NS – not significant. * p < 0.05, ** p < 0.01, *** p < 0.001
Fig. 5
Fig. 5
Link between planning target volume (PTV), spleen dose and T cell count. a) i) Graph showing the PTV for patients receiving a conventionally fractionated regimen (Fract) versus a hypofractionated regimen (Hypo). A subset of patients receiving a conventionally fractionated regimen exhibited a PTV greater than 400 (square symbols). ii) Post-chemoRT blood T cell counts are shown for patients with PTV greater than or less than 400 receiving a fractionated regimen (Fract) versus a hypofractionated regimen (Hypo). b) Relationship between mean spleen dose (Gy) and post-chemoRT blood T cell counts for patients receiving a hypofractionated regimen. Each symbol represents one patient. Key: NS – not significant. ** p < 0.01

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):5–29. doi: 10.3322/caac.21254.
    1. Sutton JM, Abbott DE. Neoadjuvant therapy for pancreas cancer: past lessons and future therapies. World J Gastroenterol. 2014;20(42):15564–79. doi: 10.3748/wjg.v20.i42.15564.
    1. Callery M, Chang K, Fishman E, Talamonti M, Traverso W, Linehan D. Pretreatment assessment of resectable and borderline resectable pancreatic cancer: expert consensus statement. Ann Surg Oncol. 2009;16(7):1727–33. doi: 10.1245/s10434-009-0408-6.
    1. Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–25. doi: 10.1056/NEJMoa1011923.
    1. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):1691–703. doi: 10.1056/NEJMoa1304369.
    1. Seung SK, Curti BD, Crittenden M, Walker E, Coffey T, Siebert JC, et al. Phase 1 study of stereotactic body radiotherapy and interleukin-2--tumor and immunological responses. Sci Transl Med. 2012;4(137):137ra74. doi: 10.1126/scitranslmed.3003649.
    1. Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med. 2012;366(10):925–31. doi: 10.1056/NEJMoa1112824.
    1. Crittenden MR, Thanarajasingam U, Vile RG, Gough MJ. Intratumoral immunotherapy: using the tumour against itself. Immunology. 2005;114(1):11–22. doi: 10.1111/j.1365-2567.2004.02001.x.
    1. Marabelle A, Kohrt H, Caux C, Levy R. Intratumoral immunization: a new paradigm for cancer therapy. Clin Cancer Res. 2014;20(7):1747–56. doi: 10.1158/1078-0432.CCR-13-2116.
    1. Stewart CC, Perez CA. Effect of irradiation on immune responses. Radiology. 1976;118(1):201–10. doi: 10.1148/118.1.201.
    1. Rosen EM, Fan S, Rockwell S, Goldberg ID. The molecular and cellular basis of radiosensitivity: implications for understanding how normal tissues and tumors respond to therapeutic radiation. Cancer Invest. 1999;17(1):56–72. doi: 10.1080/07357909909011718.
    1. Chang BK, Timmerman RD. Stereotactic body radiation therapy: a comprehensive review. Am J Clin Oncol. 2007;30(6):637–44. doi: 10.1097/COC.0b013e3180ca7cb1.
    1. Liblau RS, Tisch R, Shokat K, Yang X, Dumont N, Goodnow CC, et al. Intravenous injection of soluble antigen induces thymic and peripheral T-cells apoptosis. Proc Natl Acad Sci U S A. 1996;93(7):3031–6. doi: 10.1073/pnas.93.7.3031.
    1. Griffith TS, Ferguson TA. Cell death in the maintenance and abrogation of tolerance: the five Ws of dying cells. Immunity. 2011;35(4):456–66. doi: 10.1016/j.immuni.2011.08.011.
    1. Wild AT, Ye X, Ellsworth SG, Smith JA, Narang AK, Garg T, et al. The association between chemoradiation-related lymphopenia and clinical outcomes in patients with locally advanced pancreatic adenocarcinoma. Am J Clin Oncol. 2013
    1. Savage AM, Pritchard JA, Deeley TJ, Davies BH. Immunological state of patients with carcinoma of the bronchus before and after radiotherapy. Thorax. 1980;35(7):500–5. doi: 10.1136/thx.35.7.500.
    1. Gough MJ, Crittenden MR, Young KH. Comparing equals when evaluating immunotherapy with different doses and fractions of radiation therapy. Immunotherapy. 2015:1–3. doi:10.2217/IMT.15.58.
    1. Mahadevan A, Jain S, Goldstein M, Miksad R, Pleskow D, Sawhney M, et al. Stereotactic body radiotherapy and gemcitabine for locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys. 2010;78(3):735–42. doi: 10.1016/j.ijrobp.2009.08.046.
    1. Maecker HT, McCoy JP, Nussenblatt R. Standardizing immunophenotyping for the human immunology project. Nat Rev Immunol. 2012;12(3):191–200.
    1. Clutter MR, Krutzik PO, Nolan GP. Phospho-specific flow cytometry in drug discovery. Drug Discov Today Technol. 2005;2(3):295–302. doi: 10.1016/j.ddtec.2005.08.010.
    1. Krutzik PO, Irish JM, Nolan GP, Perez OD. Analysis of protein phosphorylation and cellular signaling events by flow cytometry: techniques and clinical applications. Clin Immunol. 2004;110(3):206–21. doi: 10.1016/j.clim.2003.11.009.
    1. Callery MP, Chang KJ, Fishman EK, Talamonti MS, William Traverso L, Linehan DC. Pretreatment assessment of resectable and borderline resectable pancreatic cancer: expert consensus statement. Ann Surg Oncol. 2009;16(7):1727–33. doi: 10.1245/s10434-009-0408-6.
    1. Gattinoni L, Finkelstein SE, Klebanoff CA, Antony PA, Palmer DC, Spiess PJ, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med. 2005;202(7):907–12. doi: 10.1084/jem.20050732.
    1. Dummer W, Niethammer AG, Baccala R, Lawson BR, Wagner N, Reisfeld RA, et al. T cell homeostatic proliferation elicits effective antitumor autoimmunity. J Clin Invest. 2002;110(2):185–92. doi: 10.1172/JCI0215175.
    1. Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, Restifo NP, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol. 2005;23(10):2346–57. doi: 10.1200/JCO.2005.00.240.
    1. Prlic M, Blazar BR, Khoruts A, Zell T, Jameson SC. Homeostatic expansion occurs independently of costimulatory signals. J Immunol. 2001;167(10):5664–8. doi: 10.4049/jimmunol.167.10.5664.
    1. Goldrath AW, Bogatzki LY, Bevan MJ. Naive T cells transiently acquire a memory-like phenotype during homeostasis-driven proliferation. J Exp Med. 2000;192(4):557–64. doi: 10.1084/jem.192.4.557.
    1. Fuks Z, Strober S, Bobrove AM, Sasazuki T, McMichael A, Kaplan HS. Long term effects of radiation of T and B lymphocytes in peripheral blood of patients with Hodgkin’s disease. J Clin Invest. 1976;58(4):803–14. doi: 10.1172/JCI108532.
    1. Grossman SA, Ellsworth S, Campian J, Wild AT, Herman JM, Laheru D, et al. Survival in patients with severe lymphopenia following treatment with radiation and chemotherapy for newly diagnosed solid tumors. J Natl Compr Cancer Netw. 2015;13(10):1225–31.
    1. Yovino S, Kleinberg L, Grossman SA, Narayanan M, Ford E. The etiology of treatment-related lymphopenia in patients with malignant gliomas: modeling radiation dose to circulating lymphocytes explains clinical observations and suggests methods of modifying the impact of radiation on immune cells. Cancer Invest. 2013;31(2):140–4. doi: 10.3109/07357907.2012.762780.
    1. Kachikwu EL, Iwamoto KS, Liao YP, DeMarco JJ, Agazaryan N, Economou JS, et al. Radiation enhances regulatory T cell representation. Int J Radiat Oncol Biol Phys. 2011;81(4):1128–35. doi: 10.1016/j.ijrobp.2010.09.034.
    1. Crittenden MR, Savage T, Cottam B, Bahjat KS, Redmond WL, Bambina S, et al. The peripheral myeloid expansion driven by murine cancer progression is reversed by radiation therapy of the tumor. PLoS One. 2013;8(7):e69527. doi: 10.1371/journal.pone.0069527.
    1. Schuler PJ, Harasymczuk M, Schilling B, Saze Z, Strauss L, Lang S, et al. Effects of adjuvant chemoradiotherapy on the frequency and function of regulatory T cells in patients with head and neck cancer. Clin Cancer Res. 2013;19(23):6585–96. doi: 10.1158/1078-0432.CCR-13-0900.
    1. Ellsworth S, Balmanoukian A, Kos F, Nirschl CJ, Nirschl TR, Grossman SA, et al. Sustained CD4 T cell-driven lymphopenia without a compensatory IL-7/IL-15 response among high-grade glioma patients treated with radiation and temozolomide. Oncoimmunology. 2014;3(1):e27357. doi: 10.4161/onci.27357.
    1. Bos PD, Plitas G, Rudra D, Lee SY, Rudensky AY. Transient regulatory T cell ablation deters oncogene-driven breast cancer and enhances radiotherapy. J Exp Med. 2013;210(11):2435–66. doi: 10.1084/jem.20130762.
    1. Simpson TR, Li F, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F, et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med. 2013;210(9):1695–710. doi: 10.1084/jem.20130579.
    1. Selby MJ, Engelhardt JJ, Quigley M, Henning KA, Chen T, Srinivasan M, et al. Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res. 2013;1(1):32–42. doi: 10.1158/2326-6066.CIR-13-0013.
    1. Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP, et al. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res. 2005;11(2 Pt 1):728–34. doi:11/2/728.
    1. Sharabi AB, Nirschl CJ, Kochel CM, Nirschl TR, Francica BJ, Velarde E, et al. Stereotactic radiation therapy augments antigen-specific PD-1 mediated anti-tumor immune responses via cross-presentation of tumor antigen. Cancer Immunol Res. 2014
    1. Herman JM, Chang DT, Goodman KA, Dholakia AS, Raman SP, Hacker-Prietz A, et al. Phase 2 multi-institutional trial evaluating gemcitabine and stereotactic body radiotherapy for patients with locally advanced unresectable pancreatic adenocarcinoma. Cancer. 2015;121(7):1128–37. doi: 10.1002/cncr.29161.
    1. Chuong MD, Springett GM, Freilich JM, Park CK, Weber JM, Mellon EA, et al. Stereotactic body radiation therapy for locally advanced and borderline resectable pancreatic cancer is effective and well tolerated. Int J Radiat Oncol Biol Phys. 2013;86(3):516–22. doi: 10.1016/j.ijrobp.2013.02.022.
    1. Chen G, Emens LA. Chemoimmunotherapy: reengineering tumor immunity. Cancer Immunol Immunother. 2013;62(2):203–16. doi: 10.1007/s00262-012-1388-0.
    1. Lynch TJ, Bondarenko I, Luft A, Serwatowski P, Barlesi F, Chacko R, et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II study. J Clin Oncol. 2012;30(17):2046–54. doi: 10.1200/JCO.2011.38.4032.
    1. Reck M, Bondarenko I, Luft A, Serwatowski P, Barlesi F, Chacko R, et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase 2 trial. Ann Oncol. 2013;24(1):75–83. doi: 10.1093/annonc/mds213.
    1. Antonia SJ, Mirza N, Fricke I, Chiappori A, Thompson P, Williams N, et al. Combination of p53 cancer vaccine with chemotherapy in patients with extensive stage small cell lung cancer. Clin Cancer Res. 2006;12(3 Pt 1):878–87. doi: 10.1158/1078-0432.CCR-05-2013.
    1. Winograd R, Byrne KT, Evans RA, Odorizzi PM, Meyer AR, Bajor DL, et al. Induction of T-cell immunity overcomes complete resistance to PD-1 and CTLA-4 blockade and improves survival in pancreatic carcinoma. Cancer Immunol Res. 2015;3(4):399–411. doi: 10.1158/2326-6066.CIR-14-0215.
    1. Young KH, Baird JR, Savage T, Cottam B, Friedman D, Bambina S, et al. Optimizing timing of immunotherapy improves control of tumors by hypofractionated radiation therapy. PLoS One. 2016;11(6):e0157164. doi: 10.1371/journal.pone.0157164.
    1. Gough MJ, Crittenden MR, Sarff M, Pang P, Seung SK, Vetto JT, et al. Adjuvant therapy with agonistic antibodies to CD134 (OX40) increases local control after surgical or radiation therapy of cancer in mice. J Immunother. 2010;33(8):798–809. doi: 10.1097/CJI.0b013e3181ee7095.
    1. Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013;73(3):1128–41. doi: 10.1158/0008-5472.CAN-12-2731.
    1. Campian JL, Sarai G, Ye X, Marur S, Grossman SA. Association between severe treatment-related lymphopenia and progression-free survival in patients with newly diagnosed squamous cell head and neck cancer. Head Neck. 2014;36(12):1747–53. doi: 10.1002/hed.23535.
    1. Ku GY, Yuan J, Page DB, Schroeder SE, Panageas KS, Carvajal RD, et al. Single-institution experience with ipilimumab in advanced melanoma patients in the compassionate use setting: lymphocyte count after 2 doses correlates with survival. Cancer. 2010;116(7):1767–75. doi: 10.1002/cncr.24951.

Source: PubMed

3
Prenumerera