Increased abscopal responses after radiation therapy for oligoprogressive patients receiving immune checkpoint inhibitors?
Editorial Commentary

Increased abscopal responses after radiation therapy for oligoprogressive patients receiving immune checkpoint inhibitors?

Jérôme Doyen1,2, Antonin Levy3,4

1Department of Radiation Oncology, Centre Antoine-Lacassagne, Nice, France; 2Université Côte d’Azur, Nice, France; 3Department of Radiation Oncology, International Center for Thoracic Cancers (CICT), Université Paris-Saclay, Gustave Roussy, Villejuif, France; 4INSERM U1030, Molecular Radiotherapy and Therapeutic Innovations, Université Paris-Saclay, Villejuif, France

Correspondence to: Jérôme Doyen. Centre Antoine-Lacassagne, Nice, France. Email: jerome.doyen@nice.unicancer.fr.

Comment on: Chicas-Sett R, Zafra J, Rodriguez-Abreu D, et al. Combination of SABR With Anti-PD-1 in Oligoprogressive Non-Small Cell Lung Cancer and Melanoma: Results of a Prospective Multicenter Observational Study. Int J Radiat Oncol Biol Phys 2022;114:655-65.


Keywords: Radiation therapy; cancer; immune checkpoint inhibitors; abscopal


Submitted Jan 09, 2023. Accepted for publication Mar 01, 2023. Published online Mar 15, 2023.

doi: 10.21037/tcr-23-37


The definition of oligometastatic disease has been recently refined by national societies (1). With advances in systemic treatments, drug exposition time tends to be longer, and a subsequent proportion of patients develop limited acquired resistance or “oligoprogression” while receiving systemic treatments. In this setting, local ablative treatments with stereotactic ablative radiation therapy (SABR) could allow maintaining responses by inducing tumor subclones elimination without modifying systemic treatments. Moreover, some systemic treatments such as immune-checkpoint inhibitors (ICI) and SABR could work together with possible stimulation of radiation immune-stimulatory effects, counteraction of radiation immune-suppressive effects and possible immune-resistance tackling (2). SABR could reinvigorate anti-tumoral immunity by (I) the presentation of new tumoral antigen to the immune system/expansion of neoantigen-reactive CD8 T cells [described in patients by Postow et al. (3) and Formenti et al. (4)], (II) the increase of immunogenic cell death, (III) the increase of the viral mimicry and (IV) the reversal of vascular barrier as reviewed by Charpentier et al. (5). This could ultimately lead to increased radiated out-of-field “abscopal” responses.

The observational study of Chicas-Sett et al. (6) is one of the first to prospectively report the efficacy of SABR in advanced melanoma or non-small cell lung cancer (NSCLC) patients showing oligoprogression while on ICI with anti-programmed cell death protein 1 (anti-PD1) (nivolumab and pembrolizumab). SABR was administered concurrently at a dose of 35 Gy (5 fractions) or 24 Gy (3 fractions). Interestingly, brain lesions, partial tumor irradiation (if >65 cc, but % not described afterwards) and patients with oncogenic driver mutations (although low proportion: 8%) could be enrolled. It should also be highlighted that further course of SABR after new oligoprogression was allowed if the patient maintained clinical benefit, and if so, earlier disease progression was not counted. Endpoints included objective response rate (ORR), overall (OS) and progression-free survival (PFS) and an abscopal response was defined as a ≥30% reduction in 1 to 2 predefined non-irradiated lesions at 2 months.

In total, 50 previously treated (median of 2 prior lines of systemic therapy) patients with 76 irradiated lesions were analyzed. Positron emission tomography was performed in 64% of patients [n=32; brain magnetic resonance imaging (MRI) not reported]. The majority (64%) of patients presented more than 5 metastases (polymetastatic disease) but with five or fewer progressive tumor sites while on ICI (median of 6 cycles of ICI; range, 2–43) (6). Halve patients were considered “primary refractory” to ICI. Main irradiated lesions were nodes, lung and bones (83%) and one third of patients (38%) had more than one course of SABR.

Outcomes reported by Chicas-Sett et al. are promising. At a median follow-up of 32.8 months, 42% ORR (30% complete response rate, 12% partial responses) and 10% stabilizations while maintaining anti-PD1 were observed (6). The median OS and PFS, calculated from SABR delivery, were respectively 37.4 and 14.2 months (2-year PFS: 36%). Some non-irradiated tumor sites that presented no further tumor response before SABR also decreased in size or were stabilized after SABR. This abscopal effect was observed in 26/40 (65%) evaluable patients, with improved outcome in this subgroup of patients as compared to other patients (median PFS of 21.2 vs. 3 months, respectively; P<0.0001).

Although it may not be directly compared, Results of Chicas-Sett et al. exceeded outcomes observed with single agent ICI in pre-treated patients or retrospective reports in oligoprogressive patients (7,8). Only one early report of a randomized trial (CURB) is available in this setting (9). Tsai et al. evaluated in advanced NSCLC or breast cancer with five or fewer oligoprogressive lesions standard of care with systemic therapy (almost 50% patients with ICI) +/− SBRT to oligoprogressive sites. PFS was increased in the 59 patients with NSCLC after SBRT (44 vs. 9 weeks; P=0.001) but no difference was observed in the 47 patients with breast cancer (P=0.48).

Beside randomized trial, the strategy of adding a local treatment in oligoprogressive patients is also the only, scientific way to prove that the abscopal effect exist. The definition of the abscopal response is however not standardized. Many non-randomized report-initiated ICI and SABR concurrently with uncertainties regarding additional abscopal responses. Formenti et al., Saiag et al., and Sumodhee et al. observed a 18%, 31.5% and 9.5% abscopal response rates, respectively, and a 10%, 0% and 9.5% stable disease rates, respectively (4,10,11).

The high abscopal response rate observed by Chicas-Sett et al. could be partially biased since authors included few patients (median: 6 cycles) that only received 2 ICI cycles (6). For such patients, progression could have been pseudoprogression and not true progression, reflecting delayed response on ICI. The pseudoprogression rate among solid tumors is around 10–15% [reviewed by Wang et al. (12)], which is consequently not a frequent event. The use of stereotactic technique as compared to conventional radiotherapy or limited volume/partial irradiation may have also played a role since the limited dose delivered to healthy organs could decrease radiation-induced lymphopenia (RIL) (13). Correlation between survival and RIL was demonstrated in many cancers (14). Techniques such as lymph-node sparing radiotherapy, proton therapy or the use of fewer (15) fractions (less than 5) could potentially limit RIL since lymphocytes are highly radiosensitive [0.95 Gy causing 37% of cell death (16)]. The best dose per fraction is anyway unknown. According to Vanpouille-Box et al. the amount of cytosolic DNA which reflects the immunogenicity of tumor cells, depends on the dose per fraction (17). Interestingly, most studies that reported abscopal response used dose per fraction less than 10 Gy: 9.5 Gy for Postow et al. (3), 6–9.5 Gy for Formenti et al. (4), 7–8 Gy for Chicas-Sett et al. (6), 6 Gy for Saiag et al. (10) and Sumodhee et al. (11). Of note, in these studies abscopal response was not only observed after SABR but also after conventional 3-dimensional conformal radiotherapy.

Only few randomized trials assessed the addition of SABR in patient’s receiving ICI. A pooled analysis of two phase 2 randomized studies already demonstrated survival benefit in advanced NSCLC if pembrolizumab was combined with radiotherapy, concurrently with pembrolizumab (18). At the opposite, two randomized phase II trial showed no additional effects of SABR. In 62 patients with advanced head and neck squamous cell carcinoma (HNSCC) the addition of 3×9 Gy was assessed on 1 target between the first and second cycle of nivolumab: no survival benefit was observed (19). Another randomized phase II trial assessed nivolumab-ipilimumab +/− SABR (3×8 Gy to one lesion) in 50 patients (50% ICI naïve only) advanced Merkel cell carcinomas (20). No difference of ORR was observed between the two arms. Although the dose per fraction and number of fractions seemed appropriate, the limited number of radiated targets could have limited he efficacy of SABR given it did not consider the tumor heterogeneity (21,22). Some primary tumors such as HNSCC carcinomas could not be a good model for this association yet suggested in earlier stages (23). Finally, the best ICI-SABR sequence is unknown. Preclinical data suggest indeed that irradiation after the beginning of ICI reduces lymphocyte infiltration and the magnitude of the abscopal effect (24).

Several phase III trials are ongoing (25), testing the association of radiotherapy with ICI (NCT03391869, NCT03774732, NCT04929041, NCT04402788, NCT04944914, NCT04747054) with some focusing on oligoprogression (CURB, STOP: NCT02756793). Prospectively clinical and surrogate predictive biomarkers are eagerly needed for selecting oligoprogressive patients that could best benefit from additional local treatments.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, Translational Cancer Research. The article did not undergo external peer review.

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-37/coif). The authors have no conflicts of interest to declare.

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. Guckenberger M, Lievens Y, Bouma AB, et al. Characterisation and classification of oligometastatic disease: a European Society for Radiotherapy and Oncology and European Organisation for Research and Treatment of Cancer consensus recommendation. Lancet Oncol 2020;21:e18-28. [Crossref] [PubMed]
  2. Mondini M, Levy A, Meziani L, et al. Radiotherapy-immunotherapy combinations - perspectives and challenges. Mol Oncol 2020;14:1529-37. [Crossref] [PubMed]
  3. 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]
  4. Formenti SC, Rudqvist NP, Golden E, et al. Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat Med 2018;24:1845-51. [Crossref] [PubMed]
  5. Charpentier M, Spada S, Van Nest SJ, et al. Radiation therapy-induced remodeling of the tumor immune microenvironment. Semin Cancer Biol 2022;86:737-47. [Crossref] [PubMed]
  6. Chicas-Sett R, Zafra J, Rodriguez-Abreu D, et al. Combination of SABR With Anti-PD-1 in Oligoprogressive Non-Small Cell Lung Cancer and Melanoma: Results of a Prospective Multicenter Observational Study. Int J Radiat Oncol Biol Phys 2022;114:655-65. [Crossref] [PubMed]
  7. Schoenfeld AJ, Rizvi HA, Memon D, et al. Systemic and Oligo-Acquired Resistance to PD-(L)1 Blockade in Lung Cancer. Clin Cancer Res 2022;28:3797-803. [Crossref] [PubMed]
  8. Mahmood U, Huynh MA, Killoran JH, et al. Retrospective Review of Outcomes After Radiation Therapy for Oligoprogressive Disease on Immune Checkpoint Blockade. Int J Radiat Oncol Biol Phys 2022;114:666-75. [Crossref] [PubMed]
  9. Tsai CJ, Yang JT, Guttmann DM, et al. Consolidative use of radiotherapy to block (CURB) oligoprogression–interim analysis of the first randomized study of stereotactic body radiotherapy in patients with oligoprogressive metastatic cancers of the lung and breast. International Journal of Radiation Oncology*Biology*Physics 2021;111:1325-6.
  10. Saiag P, Molinier R, Roger A, et al. Efficacy of Large Use of Combined Hypofractionated Radiotherapy in a Cohort of Anti-PD-1 Monotherapy-Treated Melanoma Patients. Cancers (Basel) 2022;14:4069. [Crossref] [PubMed]
  11. Sumodhee S, Guo L, Bouhlel L, et al. Impact of radiotherapy schedule on survival of patients treated with immune-checkpoint inhibitors for advanced melanoma and non-small cell lung cancer. Cancer Radiother 2022;26:1045-53. [Crossref] [PubMed]
  12. Wang Q, Gao J, Wu X. Pseudoprogression and hyperprogression after checkpoint blockade. Int Immunopharmacol 2018;58:125-35. [Crossref] [PubMed]
  13. Chen D, Patel RR, Verma V, 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. [Crossref] [PubMed]
  14. Venkatesulu BP, Mallick S, Lin SH, et al. A systematic review of the influence of radiation-induced lymphopenia on survival outcomes in solid tumors. Crit Rev Oncol Hematol 2018;123:42-51. [Crossref] [PubMed]
  15. Deutsch E, Cengel KA, Galluzzi L, et al. Could Protons Promote Tumor Control by Avoiding Lymphopenia? J Thorac Oncol 2021;16:e39-41. [Crossref] [PubMed]
  16. Dainiak N. Hematologic consequences of exposure to ionizing radiation. Exp Hematol 2002;30:513-28. [Crossref] [PubMed]
  17. Vanpouille-Box C, Alard A, Aryankalayil MJ, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun 2017;8:15618. [Crossref] [PubMed]
  18. Theelen WSME, Chen D, Verma V, 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;9:467-75. [Crossref] [PubMed]
  19. McBride S, Sherman E, Tsai CJ, et al. Randomized Phase II Trial of Nivolumab With Stereotactic Body Radiotherapy Versus Nivolumab Alone in Metastatic Head and Neck Squamous Cell Carcinoma. J Clin Oncol 2021;39:30-7. [Crossref] [PubMed]
  20. Kim S, Wuthrick E, Blakaj D, et al. Combined nivolumab and ipilimumab with or without stereotactic body radiation therapy for advanced Merkel cell carcinoma: a randomised, open label, phase 2 trial. Lancet 2022;400:1008-19. [Crossref] [PubMed]
  21. Brooks ED, Chang JY. Time to abandon single-site irradiation for inducing abscopal effects. Nat Rev Clin Oncol 2019;16:123-35. [Crossref] [PubMed]
  22. Doyen J, Picard A, Naghavi AO, et al. Clinical Outcomes of Metastatic Melanoma Treated With Checkpoint Inhibitors and Multisite Radiotherapy. JAMA Dermatol 2017;153:1056-9. [Crossref] [PubMed]
  23. Lee NY, Ferris RL, Psyrri A, et al. Avelumab plus standard-of-care chemoradiotherapy versus chemoradiotherapy alone in patients with locally advanced squamous cell carcinoma of the head and neck: a randomised, double-blind, placebo-controlled, multicentre, phase 3 trial. Lancet Oncol 2021;22:450-62. [Crossref] [PubMed]
  24. Wei J, Montalvo-Ortiz W, Yu L, et al. Sequence of αPD-1 relative to local tumor irradiation determines the induction of abscopal antitumor immune responses. Sci Immunol 2021;6:eabg0117. [Crossref] [PubMed]
  25. Doyen J, Besse B, Texier M, et al. PD-1 iNhibitor and chemotherapy with concurrent IRradiation at VAried tumor sites in advanced Non-small cell lung cAncer: the Prospective Randomized Phase 3 NIRVANA-Lung Trial. Clin Lung Cancer 2022;23:e252-6. [Crossref] [PubMed]
Cite this article as: Doyen J, Levy A. Increased abscopal responses after radiation therapy for oligoprogressive patients receiving immune checkpoint inhibitors? Transl Cancer Res 2023;12(4):713-716. doi: 10.21037/tcr-23-37

Download Citation