New insights in predictive determinants of the tumor immune microenvironment for immune checkpoint inhibition: a never ending story?
Editorial Commentary

New insights in predictive determinants of the tumor immune microenvironment for immune checkpoint inhibition: a never ending story?

Markus Eckstein1, Shilpa Gupta2

1Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bavaria, Germany;2Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA

Correspondence to: Shilpa Gupta, MD. Associate Professor of Medicine, Lerner College of Medicine, Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Ave., Desk CA60, Cleveland, OH 44195, USA. Email: guptas5@ccf.org; Markus Eckstein, MD. Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Krankenhausstraße 8-10, 91054 Erlangen, Germany. Email: markus.eckstein@uk-erlangen.de.

Provenance: This is an invited article commissioned by the Guest Section Editor Tao Shi (Clinical Cancer Institute of Nanjing University, Nanjing, China).

Comment on: Ott PA, Bang YJ, Piha-Paul SA, et al. T-Cell-Inflamed Gene-Expression Profile, Programmed Death Ligand 1 Expression, and Tumor Mutational Burden Predict Efficacy in Patients Treated With Pembrolizumab Across 20 Cancers: KEYNOTE-028. J Clin Oncol 2019;37:318-27.


Submitted May 31, 2019. Accepted for publication Jun 05, 2019.

doi: 10.21037/atm.2019.06.12


Introduction

Checkpoint inhibitors such as Pembrolizumab (Keytruda®, MSD), Atezolizumab (Tecentriq®, Roche), Durvalumab (Imfinzi®, AstraZeneca) or Nivolumab (Opdivo®, BMS) have significantly improved curative and palliative treatment of solid malignancies (1-4). However, clinical responses vary largely across different tumor entities. Since the currently used standard predictive tool—PD-L1 assessment by immunohistochemistry—shows largely varying predictive efficacy in different tumor types, e.g., in urothelial cancer (5-7), there is a heavy need for additional complementary biomarkers which can improve the current immunotherapy selection and prediction of therapy success.


Recent results of the KEYNOTE-028

Recently published results from the KEYNOTE-028 study which investigated the clinical efficacy of Pembrolizumab in patients with PD-L1 positive advanced solid malignancies revealed new insights in immune-oncological determinants for immunotherapy responsiveness (8). This basket trial consists of patients suffering from multiple solid malignancies beyond the big entities where immune checkpoint inhibition is already standard of care in many indications. The investigators applied a variety of different molecular analysis in particular analysis of a previously published T-cell-inflammation related gene expression signature consisting of 18 genes (9,10), PD-L1 assessment using the 22c3 pharmDx assay for combined positive scoring (CPS) (11), and TMB analysis using a common NGS platform (8). The investigators found that patients out of this basket trial had improved objective response rates in case of elevated expression values as detected by the 18-gene gene expression signature, high PD-L1 expression and high TMB. Since these parameters showed only weak inter-marker correlations each single biomarker was able to exclusively identify patients who benefitted from anti-PD-1 targeted treatment (8). These findings build optimism that complementary biomarkers beyond PD-L1 IHC assessment could identify additional patients profiting from immune checkpoint inhibition. If a sequential testing of PD-L1, the inflammatory milieu (e.g., by gene expression signatures) and TMB might be an option for the clinical practice has to be investigated in upcoming studies. However, these results once more indicate that one biomarker alone will not be sufficient for a proper selection of patients who would benefit from immunotherapy, especially not in the setting where immune checkpoint inhibition is no current standard of care e.g., in sarcomas, tumors of the salivary glands, neuroendocrine tumors or other rare entities which were part of the KEYNOTE-028 (8).


Current state of IO biomarker testing

The immunotherapeutic field in oncology is currently one of the most dynamic research areas. Drugs targeting PD-1, its ligand PD-L1 or CTLA-4 revolutionized the field of anti-cancer treatment of several solid malignancies such as melanoma, NSCLC or urothelial carcinoma (2,4,12). In the particular case of urothelial carcinoma, after a long void of over 40 years, checkpoint inhibitors were the first effective therapy option in platinum-refractory metastatic urothelial carcinoma patients (13,14).

Despite the undeniable therapeutic advances through checkpoint inhibition there are several critical aspects which need to be discussed. At the moment, PD-L1 testing is current standard of predictive testing in several indications in NSCLC, head and neck squamous cell carcinoma or urothelial carcinoma (15-17). One important example for the current PD-L1 “misery” is the predictive role of PD-L1 in urothelial cancer: Recently, the FDA and EMA restricted the use of Keytruda (Pembrolizumab) and Tecentriq (Atezolizumab) in urothelial carcinoma based on still unpublished interim data (15), although PD-L1 testing was previously not indicated in other approved indications due to low correlation of PD-L1 status with progression free and overall survival benefits of immune checkpoint inhibition (2,6,13,14). Reasons for this “consistent inconsistency” could be numerous, but important issues lay in the nature of immunohistochemical PD-L1 assessment: although recent studies suggested that all approved companion diagnostic assay could be used more or less interchangeably for immune cell scoring and for tumor cell scoring with the exception of the SP142 assay, which detects significantly lower amounts of tumor cells, the inter-algorithm variability remains (7,18). Our group could show that the now prescribed algorithms for Atezolizumab (5%-IC-score) and Pembrolizumab (CPS10) identify different patient populations (7): of 125 patients which were positive for at least one of the two scoring systems only 41.6% were diagnosed as eligible for both drugs, while the other 58.4% were only eligible for one of the both drugs. Assuming that different PD-1/PD-L1 targeting drugs show comparable efficacy, this inter-algorithm variability could explain at least a part of the inconsistent predictive value of PD-L1 IHC scoring with different algorithms. Further issues with PD-L1 testing lay in the lack of trained pathologists which are aware of all relevant PD-L1 algorithms for different indications as well as reimbursement and supply issues in several countries. Since all approved companion diagnostic assays are quite expensive, many health insurances—especially in Europe—are not reimbursing those tests. Amongst this, the platform dependency of Dako (Dako 22c3, Dako 28-8; Dako autostainer dependent) and Ventana assays (Ventana SP142, Ventana SP263; Ventana autostainer dependent) led to the widespread use of lab developed tests most based on freely available antibody clones such as the E1L3N (CellSignaling), 28-8 (Dako) or 22c3 (Dako) which might lack sufficient validation (19,20). Furthermore, inter-observer variability could also lead to significant issues in proper patient selection which could be prevented by widespread and systematic training of pathologists for important scoring algorithms for different indications (21). Large-scale harmonization trials led to substantial improvements of PD-L1 assessment in NSCLC (22), and are therefore heavily needed for other entities such as urothelial carcinoma or head and neck squamous cell carcinomas.

Among the “PD-L1 testing misery”, there are further promising, more objective biomarkers for predicting improved outcomes under immune checkpoint inhibition such as the deficiency of DNA mismatch repair genes which causes the truncation of the subsequent DNA repair proteins (23,24). The most prominent genetic alteration linked with these important mechanisms to maintain the genomic integrity of normal cells, are the four central DNA mismatch repair proteins MSH2, MSH6, MLH1 and PMS2. Deficiency of one of these four genes is causing classical microsatellite instability (MSI)—either sporadic or as germline variant (Lynch-syndrome) (25). In the pre-NGS era, MSI was considered to occur more or less exclusively in patients with Lynch-syndrome which most often develop colorectal malignancies, but recent large-scale tumor genetic analysis showed MSI to be more frequent as sporadic event in multiple cancer types than previously thought (26). Due to the impaired DNA repair, MSI-H tumors are considered to achieve numerous non-synonymous somatic mutations which lead to an increased neoantigen burden (26). Recent studies clearly demonstrated that patients suffering from MSI-H malignancies—irrespective of primary localization and sporadic or hereditary occurrence—are benefiting from checkpoint inhibitors (27-31). Approval of pembrolizumab for MSI-H malignancies represented the first agnostic histology approval and set the stage for biomarker-based drug approvals. Patients with different solid tumors achieving high non-synonymous tumor mutational burden due to other genetic alterations causing genetic instability have also been shown to benefit from anti-PD-1/PD-L1 targeted immunotherapy. At the moment this has been proven for e.g., NSCLC, melanoma and also urothelial carcinoma (32-36). As highlighted once more by the Keynote-028 other immunologic determinants such as T-cell inflammation related gene expression correlates with improved response to checkpoint inhibition in several solid tumors (9).

Beside those promising biomarker strategies other concepts are currently investigated to further improve immunotherapy of solid tumors: TGF-β is a cytokine connected with several pro-tumorigenic effects such as promoting immunosuppression, angiogenesis, fibroblast activation and metastasis by actively excluding immune cells from the tumor mass due to fibroblast induced expansion of the extracellular matrix (34). Anti-TGF-β targeted antibodies were previously shown to potentiate the anti-tumoral activity of Atezolizumab in a murine model by reprogramming fibroblasts and increasing the amounts of CD8+ T-cells in the tumor mass (34). The benefit of additional TGF-beta blockade is currently investigated in several ongoing clinical trials.


Conclusions

Taken together, the current era of immunotherapy in solid tumors is an exciting and rapidly changing field. Although the therapeutic effects are undeniable it will be absolutely necessary to develop new diagnostic strategies beyond “simple” PD-L1 IHC assessment to identify patients who are truly benefitting from immune checkpoint inhibition while patients who won’t benefit should rather undergo other targeted therapies if possible. Since biomarkers like PD-L1, immune cell infiltration, TMB or MSI-status are not always correlated a sequential or parallel panel testing of these markers could potentially improve a proper patient selection for immunotherapy.


Acknowledgments

None.


Footnote

Conflicts of Interest: S Gupta is an advisor for Merck, Pfizer, BMS. Has received honorarium from Exelixis and Janssen; M Eckstein is advisor for AstraZeneca and Janssen-Cilag; received speaker’s honoraria from AstraZeneca, Roche, Astellas and Janssen-Cilag.


References

  1. Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015;348:56-61. [Crossref] [PubMed]
  2. Bellmunt J, Powles T, Vogelzang NJ. A review on the evolution of PD-1/PD-L1 immunotherapy for bladder cancer: The future is now. Cancer Treat Rev 2017;54:58-67. [Crossref] [PubMed]
  3. Park YJ, Kuen DS, Chung Y. Future prospects of immune checkpoint blockade in cancer: from response prediction to overcoming resistance. Exp Mol Med 2018;50:109. [Crossref] [PubMed]
  4. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science 2018;359:1350-5. [Crossref] [PubMed]
  5. Rosenberg JE, Hoffman-Censits J, Powles T, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 2016;387:1909-20. [Crossref] [PubMed]
  6. Balar AV, Galsky MD, Rosenberg JE, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet 2017;389:67-76. [Crossref] [PubMed]
  7. Eckstein M, Erben P, Kriegmair MC, et al. Performance of the Food and Drug Administration/EMA-approved programmed cell death ligand-1 assays in urothelial carcinoma with emphasis on therapy stratification for first-line use of atezolizumab and pembrolizumab. Eur J Cancer 2019;106:234-43. [Crossref] [PubMed]
  8. Ott PA, Bang YJ, Piha-Paul SA, et al. T-Cell-Inflamed Gene-Expression Profile, Programmed Death Ligand 1 Expression, and Tumor Mutational Burden Predict Efficacy in Patients Treated With Pembrolizumab Across 20 Cancers: KEYNOTE-028. J Clin Oncol 2019;37:318-27. [Crossref] [PubMed]
  9. Ayers M, Lunceford J, Nebozhyn M, et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 2017;127:2930-40. [Crossref] [PubMed]
  10. Danaher P, Warren S, Lu R, et al. Pan-cancer adaptive immune resistance as defined by the Tumor Inflammation Signature (TIS): results from The Cancer Genome Atlas (TCGA). J Immunother Cancer 2018;6:63. [Crossref] [PubMed]
  11. Kulangara K, Zhang N, Corigliano E, et al. Clinical Utility of the Combined Positive Score for Programmed Death Ligand-1 Expression and the Approval of Pembrolizumab for Treatment of Gastric Cancer. Arch Pathol Lab Med 2019;143:330-7. [Crossref] [PubMed]
  12. Mehra R, Seiwert TY, Gupta S, et al. Efficacy and safety of pembrolizumab in recurrent/metastatic head and neck squamous cell carcinoma: pooled analyses after long-term follow-up in KEYNOTE-012. Br J Cancer 2018;119:153-9. [Crossref] [PubMed]
  13. Bellmunt J, de Wit R, Vaughn DJ, et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med 2017;376:1015-26. [Crossref] [PubMed]
  14. Powles T, Duran I, van der Heijden MS, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2018;391:748-57. [Crossref] [PubMed]
  15. Gourd E. EMA restricts use of anti-PD-1 drugs for bladder cancer. Lancet Oncol 2018;19:e341. [Crossref] [PubMed]
  16. Pabani A, Butts CA. Current landscape of immunotherapy for the treatment of metastatic non-small-cell lung cancer. Curr Oncol 2018;25:S94-102. [Crossref] [PubMed]
  17. Yang B, Liu T, Qu Y, et al. Progresses and Perspectives of Anti-PD-1/PD-L1 Antibody Therapy in Head and Neck Cancers. Front Oncol 2018;8:563. [Crossref] [PubMed]
  18. Rijnders M, van der Veldt AAM, Zuiverloon TCM, et al. PD-L1 Antibody Comparison in Urothelial Carcinoma. Eur Urol 2019;75:538-40. [Crossref] [PubMed]
  19. Velcheti V, Patwardhan PD, Liu FX, et al. Real-world PD-L1 testing and distribution of PD-L1 tumor expression by immunohistochemistry assay type among patients with metastatic non-small cell lung cancer in the United States. PLoS One 2018;13:e0206370. [Crossref] [PubMed]
  20. Scheel AH, Baenfer G, Baretton G, et al. Interlaboratory concordance of PD-L1 immunohistochemistry for non-small-cell lung cancer. Histopathology 2018;72:449-59. [Crossref] [PubMed]
  21. Scheel AH, Dietel M, Heukamp LC, et al. Harmonized PD-L1 immunohistochemistry for pulmonary squamous-cell and adenocarcinomas. Mod Pathol 2016;29:1165-72. [Crossref] [PubMed]
  22. Hirsch FR, McElhinny A, Stanforth D, et al. PD-L1 Immunohistochemistry Assays for Lung Cancer: Results from Phase 1 of the Blueprint PD-L1 IHC Assay Comparison Project. J Thorac Oncol 2017;12:208-22. [Crossref] [PubMed]
  23. Jiricny J. The multifaceted mismatch-repair system. Nat Rev Mol Cell Biol 2006;7:335-46. [Crossref] [PubMed]
  24. Li GM. Mechanisms and functions of DNA mismatch repair. Cell Res 2008;18:85-98. [Crossref] [PubMed]
  25. Umar A, Boland CR, Terdiman JP, et al. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst 2004;96:261-8. [Crossref] [PubMed]
  26. Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med 2017;9:34. [Crossref] [PubMed]
  27. Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409-13. [Crossref] [PubMed]
  28. Le DT, Uram JN, Wang H, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med 2015;372:2509-20. [Crossref] [PubMed]
  29. Overman MJ, McDermott R, Leach JL, et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol 2017;18:1182-91. [Crossref] [PubMed]
  30. Yarchoan M, Hopkins A, Jaffee EM. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N Engl J Med 2017;377:2500-1. [Crossref] [PubMed]
  31. Overman MJ, Lonardi S, Wong KYM, et al. Durable Clinical Benefit With Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer. J Clin Oncol 2018;36:773-9. [Crossref] [PubMed]
  32. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124-8. [Crossref] [PubMed]
  33. Cristescu R, Mogg R, Ayers M, et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018;362. [Crossref] [PubMed]
  34. Mariathasan S, Turley SJ, Nickles D, et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018;554:544-8. [Crossref] [PubMed]
  35. Chan TA, Wolchok JD, Snyder A. Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. N Engl J Med 2015;373:1984. [Crossref] [PubMed]
  36. Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014;371:2189-99. [Crossref] [PubMed]
Cite this article as: Eckstein M, Gupta S. New insights in predictive determinants of the tumor immune microenvironment for immune checkpoint inhibition: a never ending story? Ann Transl Med 2019;7(Suppl 3):S135. doi: 10.21037/atm.2019.06.12

Download Citation