Unravelling the puzzle of immunotherapeutic efficacy in lung cancer
Editorial Commentary

Unravelling the puzzle of immunotherapeutic efficacy in lung cancer

Petros Christopoulos1,2 ORCID logo

1Department of Medical Oncology, Thoraxklinik and National Center for Tumor Diseases at Heidelberg University Hospital, Heidelberg, Germany; 2Translational Lung Research Center at Heidelberg University Hospital, the German Center for Lung Research (DZL), Heidelberg, Germany

Correspondence to: Petros Christopoulos, MD, PhD. Department of Medical Oncology, Thoraxklinik and National Center for Tumor Diseases at Heidelberg University Hospital, Röntgenstr. 1, Heidelberg 691269, Germany; Translational Lung Research Center at Heidelberg University Hospital, the German Center for Lung Research (DZL), Heidelberg, Germany. Email: petros.christopoulos@med.uni-heidelberg.de.

Comment on: Lo Russo G, Prelaj A, Dolezal J, et al. PEOPLE (NTC03447678), a phase II trial to test pembrolizumab as first-line treatment in patients with advanced NSCLC with PD-L1


Keywords: Non-small cell lung cancer (NSCLC); immunotherapy; biomarkers; flow cytometry; gene expression


Submitted Mar 06, 2024. Accepted for publication Apr 17, 2024. Published online May 17, 2024.

doi: 10.21037/tlcr-24-221


Higher response rates and longer survival have established programmed death cell (ligand)-1 [PD-(L)-1] inhibitors in addition to chemotherapy as the new first-line standard for non-small cell lung cancers (NSCLCs) without treatable genetic alterations (1). Nonetheless, combined chemoimmunotherapy has a relatively high rate of grade ≥3 adverse events at >60%, which makes PD-(L)1 inhibitor monotherapy a more attractive option for older and frail patients, as recently demonstrated by the IPSOS phase 3 trial in patients unfit for platinum and several retrospective analyses (2,3). The respective approval in Europe is, however, currently restricted to tumors with a high PD-L1 tumor proportion score (TPS) ≥50%, which show the greatest immunotherapeutic (IO) sensitivity (1).

The identification of further biomarkers to expand the scope and improve patient selection for IO has been extremely difficult so far. Since tumor profiling with next-generation sequencing (NGS) is already being performed at initial diagnosis (4), most previous efforts have focussed on the potential correlation of additional genetic parameters with IO benefit. However, despite promising early results in pancancer studies, the tumor mutational burden (TMB) did not show a strong enough association with clinical enpoints in phase 3 trials for formal regulatory approval (5), which is partly due to the technical challenges of panel-based TMB estimation (6). More recently, other genetic markers, like STK11, KEAP1, KRAS/TP53 and METΔex14/TP53 co-mutations were also linked to IO benefit in retrospective studies, but they concern relatively small patient groups without formal prospective validation (7-9).

An alternative strategy with increasing momentum is the exploration of purely immunologic, instead of genetic, biomarkers, which could theoretically capture the immunopathophysiology of each individual patient better and provide insights for the development of tailored IO strategies. With a recent study in the Journal for Immunotherapy of Cancer, Lo Russo et al. nicely demonstrate the methodological principles, power, and prospects, but also limitations of this approach (10). One first challenge arises due to the complexity and plasticity of the immune system, which necessitate multiparametric “omics” studies for adequate representation. Another issue is the continuous nature of most immunologic variables, in contrast to the binary presence or absence of genetic alterations, which complicates statistical analyses and requires the introduction of cut-offs with unclear biologic significance. Finally, the relatively low efficacy of monotherapy with PD-(L)1 inhibitors in NSCLC with PD-L1 TPS <50%, as exemplified by the progression-free survival (PFS) of only 2.9 months and the short overall survival (OS) of 12.1 months observed by Russo et al., is an additional hurdle, because the distinction between favorable and unfavorable patient courses becomes more difficult within such a narrow range.

Despite these difficulties, Lo Russo et al. could identify a plethora of correlates for IO efficacy in univariate testing and furthermore construct a composite model of independent predictors (10). These were 4 blood immune cells populations, i.e., the abundance of peripheral blood natural killer cells/CD56dimCD16+ [hazard ratio (HR) 0.56, P=0.006] at baseline, as well as the abundance of non-classical CD14dimCD16+ monocytes (HR 0.52, P=0.004), eosinophils (CD15+CD16) (HR 0.62, P=0.03) and lymphocytes (HR 0.32, P=0.001) after first radiologic evaluation, along with the tissue expression levels of 5 genes at baseline, i.e., CD244 (HR 0.74, P=0.05), PTPRC (HR 0.55, P=0.098), KLRB1 (HR 0.76, P=0.05), IRF9 (HR 3.03, P=0.08) and COMP (HR =1.22, P=0.06). While the performance of the model is currently being validated within the INT-led I3LUNG Horizon Europe project (https://cordis.europa.eu/project/id/101057695), several other points are also worth noting here. First, 2 out of the 4 significant immune cell populations have a myeloid origin and 3 out of 4 refer to innate cells, which underlines the relative importance of natural besides adaptive immunity for the efficacy of PD-(L)1 inhibitors. Similar observations were made by a recent study of miRNA in the blood on NSCLC patients, which identified a signature of 5 myeloid-derived species as reliable predictor of IO benefit outperforming tissue-based PD-L1 staining (11). Moreover, the association of gene expression results with the patient outcome is of direct practical relevance, as targeted RNA profiling with the Nanostring platform has been found suitable for integration with the routine molecular diagnostics of NSCLC patients, whose tumor RNA is anyway isolated from tissue biopsies at initial diagnosis for the sake of RNA NGS (4). One remaining point of interest here is the relationship of IO outcomes with the tissue abundance of immune cell populations, which can be calculated both in relative and absolute terms from Nanostring results and have demonstrated strong predictive potential in previous studies (12), but were not analyzed by Lo Russo et al. (10). As for the intestinal microbiome, the lack of independent representation in the final IO predictive model derived by the broad analysis of Lo Russo et al. is not surprising, since the presence of favorable bacterial taxa in stool is known to correlate strongly with immunologic parameters of the host (13).

Overall, the results by Lo Russo et al. add to the growing body of evidence supporting multiple immunologic parameters as potentially useful non-invasive IO biomarkers in the future. Other examples so far range from simple and readily available laboratory values, like the neutrophil-to-lymphocyte ratio (NLR) and advanced lung cancer inflammation index (ALI) (14), to more technically demanding parameters, like the proximity of tumor infiltrating CD8+ to PD-L1+ cancer cells (15), various serum cytokines (16), the T-cell receptor repertoire (17), miRNA signatures (18) and the course of ctDNA levels in longitudinal measurements (19). Considering the rapidly accumulating data, there is an urgent need for head-to-head comparison of promising assays to prioritize further development of novel technologies with the highest likelihood of successful clinical application and identify special patient groups that would likely require alternative approaches. Furthermore, we will need to systematically consider additional unmet biomarker needs, such as the development of predictors for immune-related adverse events (20), criteria for therapy de-escalation (21), IO biomarkers in other thoracic tumors inherently resistant to PD-(L)1 inhibitors, like EGFR+/ALK+ NSCLC (22) and thymomas (23), as well as tools to guide application of emerging IO, including next-generation antibodies (24) and TCR-T cells (25). Similar biomarker-centered phase 2 studies to deepen biologic understanding of the disease and improve patient management are ongoing also for tyrosine kinase inhibitor (TKI)-treated NSCLC, for example the ABP trial for ALK+ tumors (NCT04318938), whose analysis can be inspired by the premium systematic work and follow the avenues of further research nicely demonstrated by Lo Russo et al. (10).


Acknowledgments

Funding: This work was funded by the German Center for Lung Research (DZL).


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, Translational Lung Cancer Research. The article has undergone external peer review.

Peer Review File: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-221/prf

Conflicts of Interest: The author has completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-221/coif). The author declares research funding from AstraZeneca, Amgen, Boehringer Ingelheim, Merck, Novartis, Roche, and Takeda; speaker’s honoraria from AstraZeneca, Gilead, Janssen, Novartis, Roche, Pfizer, Thermo Fisher, and Takeda; support for attending meetings from AstraZeneca, Eli Lilly, Daiichi Sankyo, Janssen, Gilead, Novartis, Pfizer, and Takeda, and personal fees for participating to advisory boards from AstraZeneca, Boehringer Ingelheim, Chugai, Pfizer, Novartis, MSD, Takeda and Roche, all 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 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. Planchard D, Popat S, Kerr K, et al. Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2018;29:iv192-iv237. Erratum in: Ann Oncol 2019;30:863-70. [Crossref] [PubMed]
  2. Lee SM, Schulz C, Prabhash K, et al. First-line atezolizumab monotherapy versus single-agent chemotherapy in patients with non-small-cell lung cancer ineligible for treatment with a platinum-containing regimen (IPSOS): a phase 3, global, multicentre, open-label, randomised controlled study. Lancet 2023;402:451-63. [Crossref] [PubMed]
  3. Blasi M, Kuon J, Shah R, et al. Pembrolizumab Alone or With Chemotherapy for 70+ Year-Old Lung Cancer Patients: A Retrospective Study. Clin Lung Cancer 2023;24:e282-90. [Crossref] [PubMed]
  4. Volckmar AL, Leichsenring J, Kirchner M, et al. Combined targeted DNA and RNA sequencing of advanced NSCLC in routine molecular diagnostics: Analysis of the first 3,000 Heidelberg cases. Int J Cancer 2019;145:649-61. [Crossref] [PubMed]
  5. Gadgeel S, Rodríguez-Abreu D, Speranza G, et al. Updated Analysis From KEYNOTE-189: Pembrolizumab or Placebo Plus Pemetrexed and Platinum for Previously Untreated Metastatic Nonsquamous Non-Small-Cell Lung Cancer. J Clin Oncol 2020;38:1505-17. [Crossref] [PubMed]
  6. Budczies J, Kazdal D, Allgäuer M, et al. Quantifying potential confounders of panel-based tumor mutational burden (TMB) measurement. Lung Cancer 2020;142:114-9. [Crossref] [PubMed]
  7. Papillon-Cavanagh S, Doshi P, Dobrin R, et al. STK11 and KEAP1 mutations as prognostic biomarkers in an observational real-world lung adenocarcinoma cohort. ESMO Open 2020;5:e000706. [Crossref] [PubMed]
  8. Bischoff P, Reck M, Overbeck T, et al. Outcome of First-Line Treatment With Pembrolizumab According to KRAS/TP53 Mutational Status for Nonsquamous Programmed Death-Ligand 1-High (≥50%) NSCLC in the German National Network Genomic Medicine Lung Cancer. J Thorac Oncol 2024;19:803-17. [Crossref] [PubMed]
  9. Blasi M, Kuon J, Lüders H, et al. First-line immunotherapy for lung cancer with MET exon 14 skipping and the relevance of TP53 mutations. Eur J Cancer 2024;199:113556. [Crossref] [PubMed]
  10. Lo Russo G, Prelaj A, Dolezal J, et al. PEOPLE (NTC03447678), a phase II trial to test pembrolizumab as first-line treatment in patients with advanced NSCLC with PD-L1 <50%: a multiomics analysis. J Immunother Cancer 2023;11:e006833. Erratum in: J Immunother Cancer 2024;12:e006833corr1. [Crossref] [PubMed]
  11. Rajakumar T, Horos R, Jehn J, et al. A blood-based miRNA signature with prognostic value for overall survival in advanced stage non-small cell lung cancer treated with immunotherapy. NPJ Precis Oncol 2022;6:19. [Crossref] [PubMed]
  12. Budczies J, Kirchner M, Kluck K, et al. A gene expression signature associated with B cells predicts benefit from immune checkpoint blockade in lung adenocarcinoma. Oncoimmunology 2021;10:1860586. [Crossref] [PubMed]
  13. Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018;359:97-103. [Crossref] [PubMed]
  14. Mountzios G, Samantas E, Senghas K, et al. Association of the advanced lung cancer inflammation index (ALI) with immune checkpoint inhibitor efficacy in patients with advanced non-small-cell lung cancer. ESMO Open 2021;6:100254. [Crossref] [PubMed]
  15. Ghiringhelli F, Bibeau F, Greillier L, et al. Immunoscore immune checkpoint using spatial quantitative analysis of CD8 and PD-L1 markers is predictive of the efficacy of anti- PD1/PD-L1 immunotherapy in non-small cell lung cancer. EBioMedicine 2023;92:104633. [Crossref] [PubMed]
  16. Schindler H, Lusky F, Daniello L, et al. Serum cytokines predict efficacy and toxicity, but are not useful for disease monitoring in lung cancer treated with PD-(L)1 inhibitors. Front Oncol 2022;12:1010660. [Crossref] [PubMed]
  17. Christopoulos P, Schneider MA, Bozorgmehr F, et al. Large cell neuroendocrine lung carcinoma induces peripheral T-cell repertoire alterations with predictive and prognostic significance. Lung Cancer 2018;119:48-55. [Crossref] [PubMed]
  18. Rajakumar T, Horos R, Kittner P, et al. Brief Report: A Blood-Based MicroRNA Complementary Diagnostic Predicts Immunotherapy Efficacy in Advanced-Sta ge NSCLC With High Programmed Death-Ligand 1 Express ion. JTO Clin Res Rep 2022;3:100369. [Crossref] [PubMed]
  19. Bratman SV, Yang SYC, Iafolla MAJ, et al. Personalized circulating tumor DNA analysis as a predictive biomarker in solid tumor patients treated with pembrolizumab. Nat Cancer 2020;1:873-81. [Crossref] [PubMed]
  20. Daniello L, Elshiaty M, Bozorgmehr F, et al. Therapeutic and Prognostic Implications of Immune-Related Adverse Events in Advanced Non-Small-Cell Lung Cancer. Front Oncol 2021;11:703893. [Crossref] [PubMed]
  21. Hirsch I, Goldstein DA, Tannock IF, et al. Optimizing the dose and schedule of immune checkpoint inhibitors in cancer to allow global access. Nat Med 2022;28:2236-7. [Crossref] [PubMed]
  22. Budczies J, Kirchner M, Kluck K, et al. Deciphering the immunosuppressive tumor microenvironment in ALK- and EGFR-positive lung adenocarcinoma. Cancer Immunol Immunother 2022;71:251-65. [Crossref] [PubMed]
  23. Christopoulos P, Dopfer EP, Malkovsky M, et al. A novel thymoma-associated immunodeficiency with increased naive T cells and reduced CD247 expression. J Immunol 2015;194:3045-53. [Crossref] [PubMed]
  24. Elshiaty M, Schindler H, Christopoulos P. Principles and Current Clinical Landscape of Multispecific Antibodies against Cancer. Int J Mol Sci 2021;22:5632. [Crossref] [PubMed]
  25. Gaissmaier L, Elshiaty M, Christopoulos P. Breaking Bottlenecks for the TCR Therapy of Cancer. Cells 2020;9:2095. [Crossref] [PubMed]
Cite this article as: Christopoulos P. Unravelling the puzzle of immunotherapeutic efficacy in lung cancer. Transl Lung Cancer Res 2024;13(5):1173-1176. doi: 10.21037/tlcr-24-221

Download Citation