PD-1 Inhibitor Combined With Radiotherapy and GM-CSF (PRaG) in Patients With Metastatic Solid Tumors: An Open-Label Phase II Study

Yuehong Kong, Xiangrong Zhao, Meiling Xu, Jie Pan, Yifu Ma, Li Zou, Qiliang Peng, Junjun Zhang, Cunjin Su, Zhi Xu, Wei Zhou, Yong Peng, Jiabao Yang, Chengliang Zhou, Yujia Li, Qiuchen Guo, Guangqiang Chen, Hongya Wu, Pengfei Xing, Liyuan Zhang, Yuehong Kong, Xiangrong Zhao, Meiling Xu, Jie Pan, Yifu Ma, Li Zou, Qiliang Peng, Junjun Zhang, Cunjin Su, Zhi Xu, Wei Zhou, Yong Peng, Jiabao Yang, Chengliang Zhou, Yujia Li, Qiuchen Guo, Guangqiang Chen, Hongya Wu, Pengfei Xing, Liyuan Zhang

Abstract

Patients with metastatic cancer refractory to standard systemic therapies have a poor prognosis and few therapeutic options. Radiotherapy can shape the tumor microenvironment (TME) by inducing immunogenic cell death and promoting tumor recognition by natural killer cells and T lymphocytes. Granulocyte macrophage-colony stimulating factor (GM-CSF) was known to promote dendric cell maturation and function, and might also induce the macrophage polarization with anti-tumor capabilities. A phase II trial (ChiCTR1900026175) was conducted to assess the clinical efficacy and safety of radiotherapy, PD-1 inhibitor and GM-CSF (PRaG regimen). This trial was registered at http://www.chictr.org.cn/index.aspx. A PRaG cycle consisted of 3 fractions of 5 or 8 Gy delivered for one metastatic lesion from day 1, followed by 200 μg subcutaneous injection of GM-CSF once daily for 2 weeks, and intravenous infusion of PD-1 inhibitor once within one week after completion of radiotherapy. The PRaG regimen was repeated every 21 days for at least two cycles. Once the PRaG therapy was completed, the patient continued PD-1 inhibitor monotherapy until confirmed disease progression or unacceptable toxicity. The primary endpoint was objective response rate (ORR). A total of 54 patients were enrolled with a median follow-up time of 16.4 months. The ORR was 16.7%, and the disease control rate was 46.3% in intent-to-treat patients. Median progression-free survival was 4.0 months (95% confidence interval [CI], 3.3 to 4.8), and median overall survival was 10.5 months (95% CI, 8.7 to 12.2). Grade 3 treatment-related adverse events occurred in five patients (10.0%) and grade 4 in one patient (2.0%). Therefore, the PRaG regimen was well tolerated with acceptable toxicity and may represent a promising salvage treatment for patients with chemotherapy-refractory solid tumors. It is likely that PRaG acts via heating upthe TME with radiotherapy and GM-CSF, which was further boosted by PD-1 inhibitors.

Keywords: PD-1 inhibitor; chemotherapy refractory; granulocyte macrophage-colony stimulating factor; radiotherapy; tumor microenvironment.

Conflict of interest statement

Author ZX was employed by ICON Plc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Kong, Zhao, Xu, Pan, Ma, Zou, Peng, Zhang, Su, Xu, Zhou, Peng, Yang, Zhou, Li, Guo, Chen, Wu, Xing and Zhang.

Figures

Figure 1
Figure 1
Treatment schedule of the study.
Figure 2
Figure 2
Consort diagram.
Figure 3
Figure 3
Waterfall plots of maximum percent change in nonirradiated RECIST target lesions.
Figure 4
Figure 4
Kaplan–Meier curves of progression-free survival and overall survival.
Figure 5
Figure 5
Lymphocyte subset percentage changes after treatment from baseline between the three groups (CR+PR, SD, PD). The red boxplot represents percentage changes after one cycle of treatment from baseline. The green boxplot represents percentage changes after two treatment cycles from baseline. The blue boxplot represents percentage changes after three cycles of treatment from baseline. The differences in the proportion of changes after the first cycle of treatment, after the second cycle of treatment, and after the third treatment cycle was compared separately between the three groups (CR+PR, SD, PD). The one-way ANOVA was used for the homogeneity of consistent variance, and the rank-sum test was used for the homogeneity of inconsistent variance. None of the other lymphocyte subset percentage changes showed statistical differences (p > 0.05). (A) CD3+T cells percentage changes from baseline. (B) CD3+CD4+T cells percentage changes from baseline. (C) CD3+CD8+T cells percentage changes from baseline. (D) CD16+CD56+T cells percentage changes from baseline.

References

    1. Ribas A, Wolchok JD. Cancer Immunotherapy Using Checkpoint Blockade. Science (2018) 359(6382):1350–55. doi: 10.1126/science.aar4060
    1. Navani V, Graves MC, Bowden NA, van der Westhuizen A. Immune Checkpoint Blockade in Solid Organ Tumours: Choice, Dose and Predictors of Response. Br J Clin Pharmacol (2020) 86(9):1736–52. doi: 10.1111/bcp.14352
    1. Brooks ED, Schoenhals JE, Tang C, Micevic G, Gomez DR, Chang JY, et al. . Stereotactic Ablative Radiation Therapy Combined With Immunotherapy for Solid Tumors. Cancer J (2016) 22(4):257–66. doi: 10.1097/ppo.0000000000000210
    1. Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, et al. . Irradiation and Anti-PD-L1 Treatment Synergistically Promote Antitumor Immunity in Mice. J Clin Invest (2014) 124(2):687–95. doi: 10.1172/jci67313
    1. Luke JJ, Lemons JM, Karrison TG, Pitroda SP, Melotek JM, Zha Y, et al. . Safety and Clinical Activity of Pembrolizumab and Multisite Stereotactic Body Radiotherapy in Patients With Advanced Solid Tumors. J Clin Oncol (2018) 36(16):1611–18. doi: 10.1200/JCO.2017.76.2229
    1. Coleman CN, Eke I, Makinde AY, Chopra S, Demaria S, Formenti SC, et al. . Radiation-Induced Adaptive Response: New Potential for Cancer Treatment. Clin Cancer Res (2020) 26(22):5781–90. doi: 10.1158/1078-0432.Ccr-20-0572
    1. Demaria S, Coleman CN, Formenti SC. Radiotherapy: Changing the Game in Immunotherapy. Trends Cancer (2016) 2(6):286–94. doi: 10.1016/j.trecan.2016.05.002
    1. Kong Y, Ma Y, Zhao X, Pan J, Xu Z, Zhang L. Optimizing the Treatment Schedule of Radiotherapy Combined With Anti-PD-1/PD-L1 Immunotherapy in Metastatic Cancers. Front Oncol (2021) 11:638873. doi: 10.3389/fonc.2021.638873
    1. Timmerman RD, Herman J, Cho LC. Emergence of Stereotactic Body Radiation Therapy and its Impact on Current and Future Clinical Practice. J Clin Oncol (2014) 32(26):2847–54. doi: 10.1200/jco.2014.55.4675
    1. Brooks ED, Chang JY. Time to Abandon Single-Site Irradiation for Inducing Abscopal Effects. Nat Rev Clin Oncol (2019) 16(2):123–35. doi: 10.1038/s41571-018-0119-7
    1. Galluzzi L, Vitale I, Warren S, Adjemian S, Agostinis P, Martinez AB, et al. . Consensus Guidelines for the Definition, Detection and Interpretation of Immunogenic Cell Death. J Immunother Cancer (2020) 8(1):e000337. doi: 10.1136/jitc-2019-000337
    1. Baumann M, Krause M, Overgaard J, Debus J, Bentzen SM, Daartz J, et al. . Radiation Oncology in the Era of Precision Medicine. Nat Rev Cancer (2016) 16(4):234–49. doi: 10.1038/nrc.2016.18
    1. Bernstein MB, Krishnan S, Hodge JW, Chang JY. Immunotherapy and Stereotactic Ablative Radiotherapy (ISABR): A Curative Approach? Nat Rev Clin Oncol (2016) 13(8):516–24. doi: 10.1038/nrclinonc.2016.30
    1. Breen WG, Leventakos K, Dong H, Merrell KW. Radiation and Immunotherapy: Emerging Mechanisms of Synergy. J Thorac Dis (2020) 12(11):7011–23. doi: 10.21037/jtd-2019-cptn-07
    1. Sato H, Niimi A, Yasuhara T, Permata TBM, Hagiwara Y, Isono M, et al. . DNA Double-Strand Break Repair Pathway Regulates PD-L1 Expression in Cancer Cells. Nat Commun (2017) 8(1):1751. doi: 10.1038/s41467-017-01883-9
    1. Theelen W, Chen D, Verma V, Hobbs BP, Peulen HMU, Aerts J, et al. . Pembrolizumab With or Without Radiotherapy for Metastatic non-Small-Cell Lung Cancer: A Pooled Analysis of Two Randomised Trials. Lancet Respir Med (2021) 9(5):467–75. doi: 10.1016/s2213-2600(20)30391-x
    1. Theelen W, Peulen HMU, Lalezari F, van der Noort V, de Vries JF, Aerts J, et al. . Effect of Pembrolizumab After Stereotactic Body Radiotherapy vs Pembrolizumab Alone on Tumor Response in Patients With Advanced Non-Small Cell Lung Cancer: Results of the PEMBRO-RT Phase 2 Randomized Clinical Trial. JAMA Oncol (2019) 5(9):1276–82. doi: 10.1001/jamaoncol.2019.1478
    1. Welsh J, Menon H, Chen D, Verma V, Tang C, Altan M, et al. . Pembrolizumab With or Without Radiation Therapy for Metastatic non-Small Cell Lung Cancer: A Randomized Phase I/II Trial. J Immunother Cancer (2020) 8(2):e001001. doi: 10.1136/jitc-2020-001001
    1. Seo I, Lee HW, Byun SJ, Park JY, Min H, Lee SH, et al. . Neoadjuvant Chemoradiation Alters Biomarkers of Anticancer Immunotherapy Responses in Locally Advanced Rectal Cancer. J Immunother Cancer (2021) 9(3):e001610. doi: 10.1136/jitc-2020-001610
    1. Parikh AR, Szabolcs A, Allen JN, Clark JW, Wo JY, Raabe M, et al. . Radiation Therapy Enhances Immunotherapy Response in Microsatellite Stable Colorectal and Pancreatic Adenocarcinoma in a Phase II Trial. Nat Cancer (2021) 2(11):1124–35. doi: 10.1038/s43018-021-00269-7
    1. Arina A, Gutiontov SI, Weichselbaum RR. Radiotherapy and Immunotherapy for Cancer: From "Systemic" to "Multisite". Clin Cancer Res (2020) 26(12):2777–82. doi: 10.1158/1078-0432.Ccr-19-2034
    1. Golden EB, Chhabra A, Chachoua A, Adams S, Donach M, Fenton-Kerimian M, et al. . Local Radiotherapy and Granulocyte-Macrophage Colony-Stimulating Factor to Generate Abscopal Responses in Patients With Metastatic Solid Tumours: A Proof-of-Principle Trial. Lancet Oncol (2015) 16(7):795–803. doi: 10.1016/S1470-2045(15)00054-6
    1. Zhao X, Kong Y, Zhang L. Anti-PD-1 Immunotherapy Combined With Stereotactic Body Radiation Therapy and GM-CSF as Salvage Therapy in a PD-L1-Negative Patient With Refractory Metastatic Esophageal Squamous Cell Carcinoma: A Case Report and Literature Review. Front Oncol (2020) 10:1625. doi: 10.3389/fonc.2020.01625
    1. Golden EB, Marciscano AE, Formenti SC. Radiation and In Situ Tumor Vaccination. Int J Radiat Oncol Biol Phys (2020) 108(4):891–8. doi: 10.1016/j.ijrobp.2020.08.023
    1. Lazarus HM, Ragsdale CE, Gale RP, Lyman GH. Sargramostim (Rhu GM-CSF) as Cancer Therapy (Systematic Review) and An Immunomodulator. A Drug Before Its Time? Front Immunol (2021) 12:706186(3196). doi: 10.3389/fimmu.2021.706186
    1. Lotfi N, Zhang GX, Esmaeil N, Rostami A. Evaluation of the Effect of GM-CSF Blocking on the Phenotype and Function of Human Monocytes. Sci Rep (2020) 10(1):1567. doi: 10.1038/s41598-020-58131-2
    1. Mehta HM, Malandra M, Corey SJ. G-CSF and GM-CSF in Neutropenia. J Immunol (2015) 195(4):1341–9. doi: 10.4049/jimmunol.1500861
    1. Mandula JK, Rodriguez PC. Tumor-Directed Dysregulation of Erythroid Progenitors Drives Immunosuppressive Myeloid Cells. Cancer Cell (2022) 40(6):597–9. doi: 10.1016/j.ccell.2022.04.017
    1. Venkatesulu BP, Mallick S, Lin SH, Krishnan S. A Systematic Review of the Influence of Radiation-Induced Lymphopenia on Survival Outcomes in Solid Tumors. Crit Rev Oncol Hematol (2018) 123:42–51. doi: 10.1016/j.critrevonc.2018.01.003
    1. Chen D, Patel RR, Verma V, Ramapriyan R, Barsoumian HB, Cortez MA, et al. . Interaction Between Lymphopenia, Radiotherapy Technique, Dosimetry, and Survival Outcomes in Lung Cancer Patients Receiving Combined Immunotherapy and Radiotherapy. Radiother Oncol (2020) 150:114–20. doi: 10.1016/j.radonc.2020.05.051
    1. Koukourakis MI, Giatromanolaki A. Tumor Draining Lymph Nodes, Immune Response, and Radiotherapy: Towards a Revisal of Therapeutic Principles. Biochim Biophys Acta Rev Cancer (2022) 1877(3):188704. doi: 10.1016/j.bbcan.2022.188704
    1. Zhou X, Yao Z, Bai H, Duan J, Wang Z, Wang X, et al. . Treatment-Related Adverse Events of PD-1 and PD-L1 Inhibitor-Based Combination Therapies in Clinical Trials: A Systematic Review and Meta-Analysis. Lancet Oncol (2021) 22(9):1265–74. doi: 10.1016/s1470-2045(21)00333-8
    1. Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. . Durvalumab After Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N Engl J Med (2017) 377(20):1919–29. doi: 10.1056/NEJMoa1709937
    1. Shaverdian N, Lisberg AE, Bornazyan K, Veruttipong D, Goldman JW, Formenti SC, et al. . Previous Radiotherapy and the Clinical Activity and Toxicity of Pembrolizumab in the Treatment of non-Small-Cell Lung Cancer: A Secondary Analysis of the KEYNOTE-001 Phase 1 Trial. Lancet Oncol (2017) 18(7):895–903. doi: 10.1016/s1470-2045(17)30380-7
    1. Bartlett EK, Flynn JR, Panageas KS, Ferraro RA, Sta Cruz JM, Postow MA, et al. . High Neutrophil-to-Lymphocyte Ratio (NLR) is Associated With Treatment Failure and Death in Patients Who Have Melanoma Treated With PD-1 Inhibitor Monotherapy. Cancer (2020) 126(1):76–85. doi: 10.1002/cncr.32506
    1. Capone M, Giannarelli D, Mallardo D, Madonna G, Festino L, Grimaldi AM, et al. . Baseline Neutrophil-to-Lymphocyte Ratio (NLR) and Derived NLR Could Predict Overall Survival in Patients With Advanced Melanoma Treated With Nivolumab. J Immunother Cancer (2018) 6(1):74. doi: 10.1186/s40425-018-0383-1
    1. Diem S, Schmid S, Krapf M, Flatz L, Born D, Jochum W, et al. . Neutrophil-To-Lymphocyte Ratio (NLR) and Platelet-To-Lymphocyte Ratio (PLR) as Prognostic Markers in Patients With Non-Small Cell Lung Cancer (NSCLC) Treated With Nivolumab. Lung Cancer (2017) 111:176–81. doi: 10.1016/j.lungcan.2017.07.024
    1. Keegan A, Ricciuti B, Garden P, Cohen L, Nishihara R, Adeni A, et al. . Plasma IL-6 Changes Correlate to PD-1 Inhibitor Responses in NSCLC. J Immunother Cancer (2020) 8(2):e000678. doi: 10.1136/jitc-2020-000678
    1. Weber R, Groth C, Lasser S, Arkhypov I, Petrova V, Altevogt P, et al. . IL-6 as a Major Regulator of MDSC Activity and Possible Target for Cancer Immunotherapy. Cell Immunol (2021) 359:104254. doi: 10.1016/j.cellimm.2020.104254
    1. Gjoerup O, Brown CA, Ross JS, Huang RSP, Schrock A, Creeden J, et al. . Identification and Utilization of Biomarkers to Predict Response to Immune Checkpoint Inhibitors. AAPS J (2020) 22(6):132. doi: 10.1208/s12248-020-00514-4

Source: PubMed

3
S'abonner