Immunotherapy, checkpoint inhibitor advance to frontline non-small cell lung cancer
Editorial

Immunotherapy, checkpoint inhibitor advance to frontline non-small cell lung cancer

John Nemunaitis, Neil Senzer

Mary Crowley Cancer Research Center, Dallas, TX, USA

Correspondence to: John Nemunaitis, M.D. 12222 Merit Drive, Suite 1500, Dallas, Texas 75251, USA. Email: jnemunaitis@marycrowley.org.

Comment on: Reck M, Rodríguez-Abreu D, Robinson AG, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2016;375:1823-1833.


Submitted Dec 22, 2016. Accepted for publication Feb 06, 2017.

doi: 10.21037/tcr.2017.02.22


For many years, immune-based therapies have been explored in a selected array of cancers traditionally considered “immunogenic”, namely melanoma, prostate, renal cell, non-Hodgkin’s lymphoma, bladder cancer, and renal cell carcinoma. Until recently, lung cancer has not been considered part of this group. Indeed, the designation “immunogenic” has essentially been ex post facto descriptive. However, our increased understanding of dynamic molecular immunology, including the multifarious immune editing process, and an expanded array of technologic tools now allow for a mechanistic classification with regard to immunogenicity. So yes, in non-small cell lung cancer (NSCLC) there can be impaired peptide transport and MHC-peptide affinity, a lack of effective MHC-peptide:TCR binding, intratumoral cytokine and microenvironmental cytokine and cellular [both stromal and tumor infiltrating lymphocytes (TIL)] immune suppressors and, in counter-response to an endogenous or exogenous elicited adaptive immune response, the development of adaptive immune resistance. Determining the tumor/host specific mechanism(s) rendering NSCLC “non-immunogenic” in an individual patient allows the opportunity for mechanism-specific therapeutic intervention (1). Prior to the clinical use of the FDA approved checkpoint inhibitors (nivolumab and pembrolizumab), vaccine success in NSCLC was limited at best [i.e., belagenpumatucel-L (2), L-BLP 25 (3), GVAX (4), EP2101 (5) and MAGE-3 vaccines (6)].

Having elucidated, in part, the endogenous and adaptive immune resistance role of immune checkpoint inhibitors along with the availability of a variety of PD-L1 assays (as yet to be standardized), some remarkable immune mediated responses in NSCLC have been described. In previous phase II/III studies of Pembrolizumab 2 vs. 10 mg/kg vs. docetaxel 75 mg/m2 every 3 weeks in patients with 2nd/3rd line advanced NSCLC, those with greater than 50% membranous expression of PD-L1 (at both dose levels) achieved a statistically significant progression free survival (PFS) and overall survival (OS) compared to those receiving docetaxel (7,8). In the current publication, Reck et al. (9) present the results of a phase III study of front-line therapy in 305 NSCLC patient comparing Pembrolizumab 200 mg every 3 weeks (n=154) to standard of care (pathology specific) chemotherapy (n=151) which not only confirms a significant improvement in PFS (10.3 vs. 6 months) but also a significant survival advantage (P=0.005). The OS results are all the more impressive insofar as 43.7% of the chemotherapy comparator group crossed over to Pembrolizumab treatment after disease progression. It would be of interest to have a breakdown of the crossover and non-crossover groups. The limited number of non-smokers (10) precludes any conclusion in this group with a lower mutational burden (11). These results strongly support the mechanism of action of the human IgG4κ monoclonal antibody against PD-1 as well as the “immunogenicity” of NSCLC. But despite these results, the objective response rate to Pembrolizumab was only 44.8% and the achievement of a “cure” is rare. Furthermore, although a higher rate of response to PD-L1/PD-1 inhibitors is seen in PD-L1 expressing tumors, responses are not infrequently seen in PD-L1 negative patients (12). In part, this may be due to both dynamic temporal changes in PD-L1 expression as well as intrapatient expression discordance (13). Thus, the need and opportunity to pursue more effective biomarkers related to mechanisms of primary and secondary resistance with thought towards use of rational combinatorial therapeutic regimens is an increasingly appropriate focus of clinical management.

A recent meta-analysis (14) (using random effects modeling) of patients with advanced NSCLC comprising 6,756 patients enrolled in 18 randomized controlled trials reported a clinical advantage for “tumor vaccines” and “cellular immunotherapies” compared to protocol-specific best supportive care, placebo, or matched chemotherapy. Immunotherapy was associated with an OS advantage of 5.43 months (P=0.005) and a PFS difference of 3.24 months (P=0.005). Excluded from analysis were studies of immune checkpoint blockade therapy, autologous tumor vaccines and biologic response modifiers. The significant benefits derived from first generation immunotherapeutics as suggested by this meta-analysis combined with current insight into immune molecular mechanisms support the exploration of “combination” immunotherapy field as envisioned almost 10 years ago (15).

So are there other biomarkers for targeted immunotherapy in addition to the non-exclusionary predictive PD-L1?

Recent preclinical testing (16) in immune competent models reveals a correlation between high nonsynonymous tumor mutation burden (TMB) and both response and survival following immunotherapy with PD-L1/PD-1 axis checkpoint inhibitor therapy. Rizvi et al. (10) showed that a higher clinical benefit rate (PR/CR or SD >6 mo) to Pembrolizumab correlates with TMB in NSCLC patients. PFS was also improved in high vs. low TMB patients (14.5 vs. 3.7 mo, P=0.01). Interestingly, the analysis of mutational patterns in patients with high TMB revealed a response correlation with mutations involving DNA repair genes (i.e., POLD1, POLE, MSH2). Rizvi et al. addressed the underlying mechanism by hypothesizing (as others have) that recognition of tumor specific neoantigens, not subject to central processing, formed as a consequence of somatic mutations (predominantly missense), is important for the activity of anti PD-1 therapy. They then characterized the neoantigen tumor landscape on these same patients and found a direct correlation with TMB (P<0.0001). Cancers (regardless of histology type) with a mean mutational load of >10 somatic mutations per Mb of coding DNA have a higher probability of processing and presenting neoantigens recognizable by T cells (17). For example, Rosenberg showed that the TH1 cell clone responsible for clinical response in a patient with metastatic cholangiocarcinoma treated on an adoptive T cell protocol was a single unique mutation in ERBB2 (18). Schreiber, using genomic and bioinformatic approaches, identified autologous tumor specific mutation antigens responsible for anti-PD-1 mediated rejection of an aggressive sarcoma in a mouse model (19) and Verdegaal (20) ascribed the significant tumor responses to adoptive autologous cell transfer in two advanced stage melanoma patients to unique tumor neoantigens. However, insofar as these neoantigens elicit antitumor immune responses, they also have the potential to induce off-setting counter responses including CTLA4, PD-1, and PD-L1, i.e., adaptive immune resistance. This would explain the benefit shown to be derived from checkpoint inhibitors. It stands to reason that the presence of both cytotoxic T-cells (CTLs) (as a subset of TIL) and an operative and dominant PD-1/PD-L1 checkpoint in the tumor and/or TIL would provide the optimal scenario for effective PD-1/PD-L1 axis inhibition. Given that CTL PD-1/PD-L1 expression can be induced following TCR activation and tumor PD-L1 expression induced following IFNγ and STAT3 stimulation (21), it would account for the finding that quantitative TIL and PD-L1 expression appear to be conjoint predictors of response to PD-1/PD-L1 pathway inhibition.

For the above reasons, a rational combination strategy would be to pair a PD-1/PD-L1 inhibitor (e.g., avelumab) with a treatment strategy that attracts TIL and enhances tumor (neo)antigen processing and presentation particularly in the presence of a low TMB (22). There is evidence of enhanced activated T-cell infiltration into tumor in response to adaptive immunotherapy. A recent study demonstrated PD-L1 IHC positivity in 12.5% (3 of 25) of resected specimens from unvaccinated patients with pancreatic cancer (23). Two weeks following autologous GVAX vaccine, specimen membranous PD-L1 expression was increased to 25% (10 of 40), and was found in vaccine induced intratumoral tertiary nodules in >80% of patients. In the same report, cyclophosphamide + GVAX treatment of Panc02 xenografts in C57B16 mice elicited an 11% cure rate which was increased to 30% with the addition of monoclonal antibody targeting PD-L1. Further, the combination of cyclophosphamide/GVAX + monoclonal antibody targeting PD-1 significantly increased the percentage of IFNγ-producing T cells within TILs as well as greater CD8+ T cell IFNγ secretion compared to either cyclophosphamide/GVAX or anti-PD-1 alone.

Vigil (24) is an autologous whole tumor cell vaccine, which incorporates a non-viral plasmid vector to simultaneously drive GMCSF production (via rhGMCSF transgene) and TGFβ1 and β2 knockdown (via bifunctional shRNAfurin). It provides patient-specific, tumor-specific antigenic matrix (including neoantigens and cancer-testis antigens, when present) capable of activating CD8+ T-cell antigen-specific effector function and T-cell effector memory acquisition. It is one of the combinatorial therapeutic pathways in development that would obviate the necessity of identifying tumor specific neoantigens that appears to be required for optimizing peptide-based vaccines. In addition, by incorporating GMCSF and furin mediated TGFβ1/β2 knockdown, Vigil drives antigen-presenting cell (APC) recruitment, tumor-associated/specific antigen uptake, processing, maturation, and (cross-)presentation. Results from the phase I and II trials (24,25) demonstrated safety, confirmed transgene product expression, and effectively activated T-cells (IFNγ-ELISPOT conversion) against autologous tumor cells that correlated with survival and time to relapse in a range of tumor types (24,25). Vigil is currently being evaluated in combination with avelumab in a phase I trial.

Reck and colleagues have expanded the role of immunotherapeutics by demonstrating the effectiveness of single agent pembrolizumab as first-line therapy in NSCLC. Immunotherapy can now be added to surgery, radiation, chemotherapy and targeted therapy as standards of care in this second most commonly diagnosed malignancy with 224,390 new cases and 158,080 deaths expected this year.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned and reviewed by the Section Editor Lei Wen (Department of Clinical Oncology Xijing Hospital, the Fourth Military Medical University, Xi’an, China).

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/tcr.2017.02.22). 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. Blankenstein T, Coulie PG, Gilboa E, et al. The determinants of tumour immunogenicity. Nat Rev Cancer 2012;12:307-13. [Crossref] [PubMed]
  2. Giaccone G, Bazhenova LA, Nemunaitis J, et al. A phase III study of belagenpumatucel-L, an allogeneic tumour cell vaccine, as maintenance therapy for non-small cell lung cancer. Eur J Cancer 2015;51:2321-9. [Crossref] [PubMed]
  3. Palmer M, Parker J, Modi S, et al. Phase I study of the BLP25 (MUC1 peptide) liposomal vaccine for active specific immunotherapy in stage IIIB/IV non-small-cell lung cancer. Clin Lung Cancer 2001;3:49-57; discussion 58. [Crossref] [PubMed]
  4. Nemunaitis J, Jahan T, Ross H, et al. Phase 1/2 trial of autologous tumor mixed with an allogeneic GVAX vaccine in advanced-stage non-small-cell lung cancer. Cancer Gene Ther 2006;13:555-62. [Crossref] [PubMed]
  5. Nemunaitis J, Nemunaitis J. A review of vaccine clinical trials for non-small cell lung cancer. Expert Opin Biol Ther 2007;7:89-102. [Crossref] [PubMed]
  6. Van den Eynde BJ, van der Bruggen P. T cell defined tumor antigens. Curr Opin Immunol 1997;9:684-93. [Crossref] [PubMed]
  7. Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016;387:1540-50. [Crossref] [PubMed]
  8. Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015;372:2018-28. [Crossref] [PubMed]
  9. Reck M, Rodríguez-Abreu D, Robinson AG, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2016;375:1823-33. [Crossref] [PubMed]
  10. 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]
  11. Govindan R, Ding L, Griffith M, et al. Genomic landscape of non-small cell lung cancer in smokers and never-smokers. Cell 2012;150:1121-34. [Crossref] [PubMed]
  12. Carbognin L, Pilotto S, Milella M, et al. Differential Activity of Nivolumab, Pembrolizumab and MPDL3280A according to the Tumor Expression of Programmed Death-Ligand-1 (PD-L1): Sensitivity Analysis of Trials in Melanoma, Lung and Genitourinary Cancers. PLoS One 2015;10:e0130142 [Crossref] [PubMed]
  13. Pinato DJ, Shiner RJ, White SD, et al. Intra-tumoral heterogeneity in the expression of programmed-death (PD) ligands in isogeneic primary and metastatic lung cancer: Implications for immunotherapy. Oncoimmunology 2016;5:e1213934 [Crossref] [PubMed]
  14. Dammeijer F, Lievense LA, Veerman GD, et al. Efficacy of Tumor Vaccines and Cellular Immunotherapies in Non-Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis. J Clin Oncol 2016;34:3204-12. [Crossref] [PubMed]
  15. Nemunaitis JJ. Are vaccines making a comeback in non-small-cell lung cancer? J Clin Oncol 2008;26:1402-3. [Crossref] [PubMed]
  16. Brown SD, Warren RL, Gibb EA, et al. Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival. Genome Res 2014;24:743-50. [Crossref] [PubMed]
  17. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015;348:69-74. [Crossref] [PubMed]
  18. Tran E, Turcotte S, Gros A, et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014;344:641-5. [Crossref] [PubMed]
  19. Gubin MM, Zhang X, Schuster H, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 2014;515:577-81. [Crossref] [PubMed]
  20. Verdegaal EM, de Miranda NF, Visser M, et al. Neoantigen landscape dynamics during human melanoma-T cell interactions. Nature 2016;536:91-5. [Crossref] [PubMed]
  21. Yao S, Chen L. Adaptive resistance: a tumor strategy to evade immune attack. Eur J Immunol 2013;43:576-9. [Crossref] [PubMed]
  22. Bobisse S, Foukas PG, Coukos G, et al. Neoantigen-based cancer immunotherapy. Ann Transl Med 2016;4:262. [Crossref] [PubMed]
  23. Soares KC, Rucki AA, Wu AA, et al. PD-1/PD-L1 blockade together with vaccine therapy facilitates effector T-cell infiltration into pancreatic tumors. J Immunother 2015;38:1-11. [Crossref] [PubMed]
  24. Senzer N, Barve M, Kuhn J, et al. Phase I trial of "bi-shRNAi(furin)/GMCSF DNA/autologous tumor cell" vaccine (FANG) in advanced cancer. Mol Ther 2012;20:679-86. [Crossref] [PubMed]
  25. Oh J, Barve M, Matthews CM, et al. Phase II study of Vigil® DNA engineered immunotherapy as maintenance in advanced stage ovarian cancer. Gynecol Oncol 2016;143:504-510. [Crossref] [PubMed]
Cite this article as: Nemunaitis J, Senzer N. Immunotherapy, checkpoint inhibitor advance to frontline non-small cell lung cancer. Transl Cancer Res 2017;6(Suppl 1):S141-S144. doi: 10.21037/tcr.2017.02.22

Download Citation