Quantitative spatial profiling of PD-1/PD-L1 interaction in patients with cancer
Editorial Commentary

Quantitative spatial profiling of PD-1/PD-L1 interaction in patients with cancer

Taiki Hakozaki1,2^

1Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan; 2Graduate School of Advanced Science and Engineering, Faculty of Science and Engineering, Waseda University, Tokyo, Japan

^ORCID: 0000-0002-9980-4417.

Correspondence to: Taiki Hakozaki, MD. Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo, Tokyo 113-0021, Japan. Email: t-hakozaki@akane.waseda.jp.

Comment on: Gavrielatou N, Liu Y, Vathiotis I, et al. Association of PD-1/PD-L1 Co-location with Immunotherapy Outcomes in Non-Small Cell Lung Cancer. Clin Cancer Res 2022;28:360-7.


Keywords: Immune checkpoint inhibitor; non-small cell lung cancer (NSCLC); biomarker; programmed death ligand-1 (PD-L1); prognosis


Submitted Jan 29, 2023. Accepted for publication Mar 06, 2023. Published online Mar 17, 2023.

doi: 10.21037/tlcr-23-56


Immunotherapy (ITx) has emerged in less than a decade as one of the most important treatment modalities for patients with cancer. Immune checkpoint inhibitors, represented by anti-programmed death receptor-1 (PD-1)/programmed death ligand-1 (PD-L1) antibodies, are now widely used as a standard of care across primary organs and histologic subtypes (1-3). Although the prognosis of the overall population with advanced cancer has been improved by anti-PD-1/PD-L1 antibody-based regimens, their tailored prescriptions have not yet been achieved. In other words, ITx still lacks satisfactory biomarkers to predict its efficacy, unlike molecularly targeted agents for which companion biomarkers with high predictive accuracy (i.e., the presence of driver alterations) are available. Although tissue PD-L1 expression assessed by immunohistochemistry is an approved method for selecting patients for anti-PD-1/PD-L1 antibody-based regimens, better outcomes are not necessarily guaranteed by PD-L1 positivity.

PD-L1 expression, calculated as a tumor proportion score (TPS), is a biomarker available for routine use in the selection of a treatment regimen for an individual patient with advanced non-small cell lung cancer (NSCLC) (4). PD-L1 positivity is crucial in selecting anti-PD-1/PD-L1 antibody monotherapy or other regimens in the first-line setting. However, even in patients with high PD-L1 expression, objective responses to anti-PD-1/PD-L1 antibody monotherapy are achieved in up to 40–50% of the population (5). Additionally, TPS has further limited utility as a biomarker for combination regimens with cytotoxic agents or anti-cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) antibodies used independently of TPS in advanced NSCLC. Such a limitation of the approach based solely on tissue PD-L1 expression would still drive researchers to diligently search for novel biomarkers with a more predictive ability (6).

Gavrielatou et al. investigated the association between “PD-1/PD-L1 colocation score” and the efficacy of ITx in 154 patients with advanced NSCLC (7). Patients with a high PD-1/PD-L1 colocation score in the overall cohort had a significant benefit from ITx compared to those with a low score in an objective response, progression-free survival (PFS), and overall survival (OS). The predictive ability of the PD-1/PD-L1 colocation score was marked in the subgroup analysis stratified by line of treatment in the cohort receiving ITx as a second or subsequent line of treatment (n=85). At the same time, no similar dissociation was observed in the first-line setting (n=69). It was also shown that the colocation score did not correlate with the tissue PD-L1 expression levels. Thus, a key message from the analysis was the potential utility of the PD-1/PD-L1 colocation score as a novel biomarker distinct from conventional PD-L1 expression assessment, particularly for those receiving ITx in the second or subsequent line of treatment.

“PD-1/PD-L1 colocation score” may be an unfamiliar term. However, the basic idea behind it is simple and in line with the mechanism of action of anti-PD-1/PD-L1 antibodies. In short, the PD-1/PD-L1 colocation score reflects the spatial proximity between PD-1 and PD-L1 in formalin-fixed paraffin-embedded tumor samples, which is quantified by the algorithm using multiplexed quantitative immunofluorescence (QIF). The present method assesses receptor/ligand interactions, in contrast to conventional PD-L1 assays focusing solely on the ligand. Thus, it attempts to reflect the interplay between tumor and immune cells within the tumor microenvironment in predicting ITx results. In addition to the report by Gavrielatou et al., the association between PD-1/PD-L1 colocation and ITx outcomes in patients with advanced cancer treated with anti-PD-1 antibodies has been addressed by several previous studies (Table 1).

Table 1

Studies on PD-1/PD-L1 colocation and anti-PD-1 antibody efficacy among patients with cancer

Authors Subjects ITx regimens Methods Key results
Gavrielatou et al. [2022] (7) NSCLC (n=124) aPD-1/PD-L1 monotherapy, aPD-1/PD-L1 + aCTLA-4, aPD-1/PD-L1 + chemo Multiplexed QIF A high PD-1/PD-L1 colocation score was associated with better response, PFS, and OS
Giraldo et al. [2018] (8) Meckel cell carcinoma (n=16) aPD-1 monotherapy Multiplexed QIF The density of PD-1+ cells adjacent to a PD-L1+ cell was higher in the responder than non-responder
Johnson et al. [2018] (9) Melanoma (n=166) aPD-1 monotherapy Multiplexed QIF High PD-1/PD-L1 colocation score ± IDO-1/HLA-DR co-expression were associated with better response, PFS, and OS
Sánchez-Magraner et al. [2020] (10) NSCLC (n=40) aPD-1 monotherapy FRET Patients with the highest 40% of FRET efficiency had a better OS than those with the lowest 60%
Sánchez-Magraner et al. [2020] (10) Melanoma (n=176) aPD-1 monotherapy FRET Patients with the highest 20% of FRET efficiency had a better OS than those with the lowest 80%

PD-1, programmed death receptor-1; PD-L1, programmed death ligand-1; ITx, immunotherapy; NSCLC, non-small cell lung cancer; aPD-1/PD-L1, anti-programmed death receptor-1/programmed death ligand-1 antibody; aCTLA-4, anti-cytotoxic T-lymphocyte-associated protein-4 antibody; QIF, quantitative immunofluorescence; PFS, progression-free survival; OS, overall survival; aPD-1, anti-programmed death receptor-1 antibody; IDO-1, indoleamine 2;3-dioxygenase-1; HLA-DR, human leukocyte antigen-D related; FRET, fluorescence resonance energy transfer.

Giraldo et al. showed that the density of PD-1+ cells adjacent to a PD-L1+ cell was higher in responder than non-responder in a cohort of Meckel cell carcinoma (n=16) (8). Johnson et al. showed that a high PD-1/PD-L1 colocation score was associated with better response, PFS, and OS on ITx in the larger melanoma cohort (n=166) using a similar approach using multiplexed QIF (9). As another approach more focused on PD-1/PD-L1 functional interaction over their simple spatial proximity, Sánchez-Magraner et al. applied the fluorescence resonance energy transfer (FRET) method in quantifying PD-1/PD-L1 colocation (10). There, higher FRET efficiency was associated with better OS in the cohort of NSCLC (n=40) and melanoma (n=176). Gavrielatou et al. added data with a specific value. It provided additional supporting data on this issue by examining the largest samples from patients with NSCLC under diverse clinical scenarios.

Despite accumulating positive signals on the quantitative spatial profiling of PD-1/PD-L1 interaction, several points need to be solved before clinical application as a predictive biomarker for ITx. First, it is not yet clear in which clinical setting (e.g., ITx regimen, treatment regimen) the novel parameter can be used to guide clinical decisions. Gavrielatou et al. found that the PD-1/PD-L1 colocation score may be most useful in the second or later-line setting. On the other hand, the score failed to stratify survival for those who received ITx in the first-line setting. On this point, we must be careful of the cohort characteristics in that one-third of patients received ITx as a combination regimen with cytotoxic agents (n=33) or other (n=18), as shown in the Tab. S1. Today, most patients with advanced NSCLC receive ITx primarily in the first-line setting. In contrast, the same populations received ITx as an anti-PD-1/PD-L1 monotherapy in the second or later-line setting. Combination regimens would be more likely used in the first-line setting or in patients with low or negative PD-L1 expression under such circumstances. In other words, the difference in the predictive ability of the colocation score may also be due to differences in the ITx regimens and not just the treatment lines.

Second, the available data have not yet demonstrated the superiority of the colocation score as a predictive biomarker for ITx over conventional PD-L1 expression levels. Although conventional PD-L1 assessment failed to stratify ITx outcomes in their cohort, it would still be considered a practical biomarker, at least for anti-PD-1/PD-L1 monotherapy in advanced NSCLC. Head-to-head comparisons are necessary to establish the colocation score as a new standard for this population. As an alternative, the combination of the conventional PD-L1-based approach and the quantitative spatial profiling of PD-1/PD-L1 interaction may be worth exploring. Future studies may also need to investigate this novel parameter as a biomarker for combination regimens that are standard options for broader populations, regardless of PD-L1 expression, to address unmet clinical needs. A standardized protocol for the quantitative spatial profiling of PD-1/PD-L1 interaction would also be needed. Further validation by a unified approach in the larger clinical scenario-based cohorts and optimization of the cutoff value for the PD-1/PD-L1 colocation score are still warranted based on the results obtained in small cohorts using different assays (multiplexed QIF, FRET, etc.). Future research should consider these points in light of the updated standard of care, not only in NSCLC but also in other cancers for which ITx regimens are available.


Acknowledgments

Funding: None.


Footnote

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

Conflicts of Interest: The author has completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-56/coif). TH received personal fees from Chugai Pharmaceutical, Ono Pharmaceutical, and Eisai outside the submitted work. The author has no other conflicts of interest to declare.

Ethical Statement: The author is accountable for all aspects of the work to ensure 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. Ettinger DS, Wood DE, Aisner DL, et al. NCCN Guidelines Insights: Non-Small Cell Lung Cancer, Version 2.2021. J Natl Compr Canc Netw 2021;19:254-66. [Crossref] [PubMed]
  2. Benson AB, Venook AP, Al-Hawary MM, et al. Colon Cancer, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021;19:329-59. [Crossref] [PubMed]
  3. Swetter SM, Thompson JA, Albertini MR, et al. NCCN Guidelines® Insights: Melanoma: Cutaneous, Version 2.2021. J Natl Compr Canc Netw 2021;19:364-76. [Crossref] [PubMed]
  4. Yu H, Boyle TA, Zhou C, et al. PD-L1 Expression in Lung Cancer. J Thorac Oncol 2016;11:964-75. [Crossref] [PubMed]
  5. Reck M, Remon J, Hellmann MD. First-Line Immunotherapy for Non-Small-Cell Lung Cancer. J Clin Oncol 2022;40:586-97. [Crossref] [PubMed]
  6. Topalian SL, Taube JM, Anders RA, et al. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 2016;16:275-87. [Crossref] [PubMed]
  7. Gavrielatou N, Liu Y, Vathiotis I, et al. Association of PD-1/PD-L1 Co-location with Immunotherapy Outcomes in Non-Small Cell Lung Cancer. Clin Cancer Res 2022;28:360-7. [Crossref] [PubMed]
  8. Giraldo NA, Nguyen P, Engle EL, et al. Multidimensional, quantitative assessment of PD-1/PD-L1 expression in patients with Merkel cell carcinoma and association with response to pembrolizumab. J Immunother Cancer 2018;6:99. [Crossref] [PubMed]
  9. Johnson DB, Bordeaux J, Kim JY, et al. Quantitative Spatial Profiling of PD-1/PD-L1 Interaction and HLA-DR/IDO-1 Predicts Improved Outcomes of Anti-PD-1 Therapies in Metastatic Melanoma. Clin Cancer Res 2018;24:5250-60. [Crossref] [PubMed]
  10. Sánchez-Magraner L, Miles J, Baker CL, et al. High PD-1/PD-L1 Checkpoint Interaction Infers Tumor Selection and Therapeutic Sensitivity to Anti-PD-1/PD-L1 Treatment. Cancer Res 2020;80:4244-57. [Crossref] [PubMed]
Cite this article as: Hakozaki T. Quantitative spatial profiling of PD-1/PD-L1 interaction in patients with cancer. Transl Lung Cancer Res 2023;12(6):1346-1349. doi: 10.21037/tlcr-23-56

Download Citation