Radiation-induced lymphopenia may negatively affect outcomes in patients receiving PD-1 directed immunotherapy
Letter to the Editor

Radiation-induced lymphopenia may negatively affect outcomes in patients receiving PD-1 directed immunotherapy

Luke R. G. Pike1,2, Jonathan D. Schoenfeld2

1Harvard Radiation Oncology Program, Massachusetts General Hospital, Boston, MA, USA; 2Dana-Farber/Brigham and Women’s Cancer Center, Boston, MA, USA

Correspondence to: Luke R. G. Pike, MD, DPhil. Harvard Radiation Oncology Program, 55 Fruit Street, Massachusetts General Hospital, Boston, MA. 02114, USA. Email: lrpike@partners.org.

Response to: Chen VE, Greenberger BA, Taylor JM, et al. Key immune system markers in immunotherapy: absolute lymphocyte count, neutrophil to lymphocyte ratio, and alternative immune system metrics. Ther Radiol Oncol 2019;3:33.


Received: 06 October 2019; Accepted: 30 October 2019; Published: 11 December 2019.

doi: 10.21037/tro.2019.10.01


We thank the editors for the invitation to contribute to Therapeutic Radiology and Oncology and Chen et al. for their excellent piece “Key immune system markers in immunotherapy: absolute lymphocyte count, neutrophil to lymphocyte ratio, and alternative immune system metrics” in which they discuss our recently published work, “The Impact of Radiation Therapy on Lymphocyte Count and Survival in Metastatic Cancer Patients Receiving PD-1 Immune Checkpoint Inhibitors” (1).

Indeed, it is becoming increasingly clear that the combination of CTLA-4- or PD-(L)1-directed immune checkpoint inhibitors (ICI) and radiation therapy (RT) is largely safe (2,3), and in the setting of the otherwise excellent systemic disease control that can be attained with these drugs in the setting of diseases such as NSCLC, renal cell carcinoma, and metastatic melanoma, definitive and/or ablative irradiation of gross disease may confer an overall survival benefit (4-6). Likewise, in preclinical models and limited clinical case series, it has been shown that the combination of RT and ICIs has the potential to yield so-called “abscopal responses” through neoantigen release, the diversification of the T cell receptor (TCR) repertoire, and both local and systemic effects on cytokine release (7-12). To date, clinical abscopal responses are uncommon, but we eagerly await the results of ongoing combination studies for evidence of increased activity as well as deeper and more informative translational correlate studies that might identify means by which to modulate these responses.

However, we and others have recently shown that RT also has the potential for harm in the setting of treatment with PD1-directed ICI (1,13). In our recently published study, we analyzed a cohort of 110 patients with metastatic NSCLC, melanoma, or renal cell carcinoma, who had received either of the approved PD-1 directed ICIs, nivolumab or pembrolizumab, as well as palliative RT as part of standard of care. We found that receipt of prolonged courses of extracranial RT was associated with the development of severe lymphopenia (<500 cells/µL), which often persisted for many months, and elevated neutrophil to lymphocyte ratio (NTL), and that this lymphopenic state at the time of initiation PD-1 directed ICI was associated with poor overall survival (1). As Chen et al. correctly point out, this work suggests that radiation-induced lymphopenia may impact oncologic outcomes in patients receiving PD-1 ICI. Limitations of this work include retrospective design, heterogeneous patient population, and lack of data on other endpoints such as progression free survival, cancer-specific survival or competing risks. In addition, we point out that the association of NTL with survival was weaker than the association of severe lymphopenia with survival, and that the former association was lost when a multivariable analysis was performed including other factors known to be associated with survival, such as performance status and albumin. We speculate that this difference may be due to the multitude of confounding causes for elevated neutrophil count, such as receipt of corticosteroids, infection, and systemic inflammation.

Mechanistically, RT-induced lymphopenia could affect the efficacy of PD-1 directed ICI in several ways. Firstly, direct killing of CD8+ T effector cells, which are thought to be the primary mediators of tumor cell eradication could occur in the irradiated tumor microenvironment as well as in abutting normal tissues, lymph nodes, and circulating blood. Recent modeling studies have determined that the volume of irradiated blood pool could play an important role, and suggest that the total volume receiving low dose to be an important determinant of lymphopenia (14,15). Thus, RT to a tumor, although effectively eradicating cancer cells and releasing antigen for further T cell diversification, could also plausibly preferentially kill exhausted tumor-infiltrating lymphocytes that would be expected to exact cell mediated cytoxicity through disinhibition with a PD-(L)1 inhibitor. However, it should be noted that preclinical data to this point have suggested contradictory effects with potential activation of tumor resident T cells (16). Secondly, recent data suggests that T regulatory cells may be resistant to RT and could in fact be drawn to the site of irradiation via unclear mechanisms (17-19). Likewise, immunosuppressive myeloid-derived cells also appear to be relatively radioresistant, and thus could further push the balance towards immunosuppression (20,21). Thus, the culmination of these effect may result in a systemic deficit of cytotoxic T cells and an abundance of tumor specific T regulatory cells and myeloid-derived immunosuppressive cells, which could in sum abrogate any beneficial effects of RT on IO, or worse.

Ultimately, these results, though intriguing, are hypothesis-generating and warrant deeper evaluation through prospective study. We suggest that trials which include both radiation and ICI consider the inclusion of rigorous correlate studies to evaluate the potential positive and negative effects of radiotherapy on the lymphoid compartment and the resultant efficacy of ICIs.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned and reviewed by the Section Editor Dr. Hsin-Hua Nien (Attending physician, Department of Radiation Oncology, Cathay General Hospital, Taiwan).

Conflicts of Interest: Luke R. G. Pike receives consulting fees from Myst Therapeutics, Clarus Ventures/Blackstone, and Third Rock Ventures. Jonathan D. Schoenfeld receives research support paid to the institution from Merck, BMS, Regeneron and consulting/scientific advisory board work with Debiopharm, BMS, Nanobiotix, Tilos, AstraZeneca, LEK, Catenion, ACI Clinical, as well as expert witness fees.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Pike LRG, Bang A, Mahal BA, et al. The Impact of Radiation Therapy on Lymphocyte Count and Survival in Metastatic Cancer Patients Receiving PD-1 Immune Checkpoint Inhibitors. Int J Radiat Oncol Biol Phys 2019;103:142-51. [Crossref] [PubMed]
  2. Hwang WL, Pike LRG, Royce TJ, et al. Safety of combining radiotherapy with immune-checkpoint inhibition. Nat Rev Clin Oncol 2018;15:477-94. [Crossref] [PubMed]
  3. Bang A, Wilhite TJ, Pike LRG, et al. Multicenter Evaluation of the Tolerability of Combined Treatment With PD-1 and CTLA-4 Immune Checkpoint Inhibitors and Palliative Radiation Therapy. Int J Radiat Oncol Biol Phys 2017;98:344-51. [Crossref] [PubMed]
  4. Pike LRG, Bang A, Ott P, et al. Radiation and PD-1 inhibition: Favorable outcomes after brain-directed radiation. Radiother Oncol 2017;124:98-103. [Crossref] [PubMed]
  5. Gomez DR, Tang C, Zhang J, et al. Local consolidative therapy vs. Maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer: Long-term results of a multi-institutional, phase II, randomized study. J Clin Oncol 2019;37:1558-65. [Crossref] [PubMed]
  6. Antonia SJ, Villegas A, Daniel D, et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N Engl J Med 2017;377:1919-29. [Crossref] [PubMed]
  7. Dewan MZ, Galloway AE, Kawashima N, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin. Cancer Res 2009;15:5379-88. [Crossref] [PubMed]
  8. Demaria S, Formenti SC. Role of T lymphocytes in tumor response to radiotherapy. Front Oncol 2012;2:95. [Crossref] [PubMed]
  9. Vanpouille-Box C, Alard A, Aryankalayil MJ, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun 2017;8:15618. [Crossref] [PubMed]
  10. Bang A, Schoenfeld JD. Immunotherapy and radiotherapy for metastatic cancers. Ann Palliat Med 2019;8:312-25. [Crossref] [PubMed]
  11. Twyman-Saint Victor C, Rech AJ, Maity A, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015;520:373-7. [Crossref] [PubMed]
  12. Postow MA, Callahan MK, Barker CA, et al. Immunologic Correlates of the Abscopal Effect in a Patient with Melanoma. N Engl J Med 2012;366:925-31. [Crossref] [PubMed]
  13. Diehl A, Yarchoan M, Hopkins A, et al. Relationships between lymphocyte counts and treatment-related toxicities and clinical responses in patients with solid tumors treated with PD-1 checkpoint inhibitors. Oncotarget 2017;8:114268-80. [Crossref] [PubMed]
  14. Tang C, Liao Z, Gomez D, et al. Lymphopenia Association With Gross Tumor Volume and Lung V5 and Its Effects on Non-Small Cell Lung Cancer Patient Outcomes. Int J Radiat Oncol Biol Phys 2014;89:1084-91. [Crossref] [PubMed]
  15. Yovino S, Kleinberg L, Grossman SA, et al. 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;31:140-4. [Crossref] [PubMed]
  16. Arina A, Beckett M, Fernandez C, et al. Tumor-reprogrammed resident T cells resist radiation to control tumors. Nat Commun 2019;10:3959. [Crossref] [PubMed]
  17. Kachikwu EL, Iwamoto KS, Liao YP, et al. Radiation enhances regulatory T cell representation. Int J Radiat Oncol Biol Phys 2011;81:1128-35. [Crossref] [PubMed]
  18. Muroyama Y, Nirschl TR, Kochel CM, et al. Stereotactic radiotherapy increases functionally suppressive regulatory T cells in the tumor microenvironment. Cancer Immunol Res 2017;5:992-1004. [Crossref] [PubMed]
  19. Oweida AJ, Darragh L, Phan A, et al. STAT3 Modulation of Regulatory T Cells in Response to Radiation Therapy in Head and Neck Cancer. J Natl Cancer Inst 2019; [Crossref] [PubMed]
  20. Ostrand-Rosenberg S, Horn LA, Ciavattone NG. Radiotherapy Both Promotes and Inhibits Myeloid-Derived Suppressor Cell Function: Novel Strategies for Preventing the Tumor-Protective Effects of Radiotherapy. Front Oncol 2019;9:215. [Crossref] [PubMed]
  21. Sampath S, Won H, Massarelli E, et al. Combined modality radiation therapy promotes tolerogenic myeloid cell populations and STAT3-related gene expression in head and neck cancer patients. Oncotarget 2018;9:11279-90. [Crossref] [PubMed]
doi: 10.21037/tro.2019.10.01
Cite this article as: Pike LRG, Schoenfeld JD. Radiation-induced lymphopenia may negatively affect outcomes in patients receiving PD-1 directed immunotherapy. Ther Radiol Oncol 2019;3:37.

Download Citation