Changing paradigms of non-small cell lung cancer treatment
Editorial

Changing paradigms of non-small cell lung cancer treatment

Francesco Guerrera1,2, Fabrizio Tabbò3, Enrico Ruffini1,2, Pietro Bertoglio4

1Department of Thoracic Surgery, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, Torino, Italy;2Department of Surgical Sciences, 3Department of Oncology, University of Torino, Torino, Italy;4Division of Thoracic Surgery, Sacro Cuore-Don Calabria Research Hospital and Cancer Care Centre Negrar-Verona, Verona, Italy

Correspondence to: Francesco Guerrera, MD. Department of Thoracic Surgery, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, Corso Dogliotti, 14 10126 Torino, Italy. Email: fra.guerrera@gmail.com.

Provenance: This is an invited Editorial commissioned by the Executive Editor-in-Chief Jianxing He (Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China).

Comment on: Bott MJ, Cools-Lartigue J, Tan KS, et al. Safety and Feasibility of Lung Resection After Immunotherapy for Metastatic or Unresectable Tumors. Ann Thorac Surg 2018;106:178-83.


Submitted Oct 23, 2018. Accepted for publication Oct 31, 2018.

doi: 10.21037/jtd.2018.11.04


Nowadays, the use of immune checkpoint inhibitors (ICI) in locally advanced and metastatic non-small cell lung cancer (NSCLC) is quickly gaining a general consensus due to interesting results in terms of tumor response and overall survival (1,2). Nevertheless, the role of ICI in an adjuvant or neo-adjuvant setting is still under investigation and restricted to clinical trials (3). Undoubtedly, the use of the immunotherapy in combination with radical local treatments might open new perspective for all medical experts (i.e., medical oncologists, surgeons, pathologists, radiation oncologists), who will have to face a radical change in the standard treatment of NSCLC.

Bott and colleagues (4) reported outcomes of 19 patients who underwent surgical resection for residual intrathoracic disease after ICI treatment for unresectable or metastatic lung cancer (mainly NSCLC and metastatic melanoma), between 2012 and 2016. Patients were treated, without an induction intent, with different immune checkpoint blockade agents: anti-PD-1 agents (nivolumab and pembrolizumab), anti-CTLA-4 agents (ipilimumab) or anti-PD-L1 agents (durvalumab and atezolizumab). Of note, all the patients present a resolution of the extra-thoracic disease after the ICI regimens. Authors reported a 32% rate of complete pathological response and 95% (all cases apart one) of radical resection. Concerning surgical technical aspects and post-operative complications, they observed one case of conversion from mini-invasive approach to thoracotomy, and one case of grade-4 pneumonitis with no post-operative mortality. Overall survival and disease-free survival were 77% and 42% respectively. Authors conclude that surgery after immunotherapy is feasible and safe, with good post-operative results and acceptable long-term outcomes.

To date, only few studies reported results of the use of ICI as pre-operative treatment for resectable NSCLC (5,6) but several trials are currently ongoing, and results will be available in the next years clarifying the possible benefits of this approach (7,8). Even though, preliminary reports seem to be favourable to this approach, as in the advanced disease, nevertheless the best setting for immunotherapy in combination with surgery is yet to be found. Indeed, ICI could be used either alone or in combination with radiotherapy or chemotherapy agents, and in adjuvant or neoadjuvant setting. These different approaches reflect diverse rationales and should be tailored on each specific patient. For instance, pre-operative immunotherapy could be used in order to achieve a tumour volume reduction, and consequently a higher rate of lung-sparring and/or of radical resection, while post-operative administration could be used in unexpected locally advanced disease (9). Therefore, the recognition of the correct time frame of medical treatment, the correct dose, the diverse chemotherapy combinations, and the possible combination with radiotherapy administration represents future challenges in this field. On the other hands, post-operative complications rate and intra-operative tissue changes (e.g., inflammation, fibrosis) determined by ICI need to be elucidated, in order to define the best choice amongst different surgical resections achievable (e.g., lobar, sub-lobar, extended) and diverse surgical approaches available (classic vs. mini-invasive).

In this context, the results presented by Bott and colleagues are reassuring, with an impact on surgical procedures (e.g., conversion to thoracotomy, operative time) and on post-operative complications largely comparable with classical induction agents and a low rate of positive margins.

Surely, one of the greatest potential innovations of immunotherapy is to enormously enlarge the cohort of resectable cases. On one hand, it may assure a better control of unforeseen micro metastatic sites in early stage NSCLC, which would undergo to upfront surgical resection (10); on the other hand, it might open new perspectives and indications for surgery also in patients with a systemic disease at the moment of diagnosis (4). Pre-clinical studies have triggered these captivating questions: analyzing the role of immunotherapy for breast cancer in mice, Liu and coworkers (11) found a significant survival advantage when immunotherapy was administered in a neoadjuvant setting compared to adjuvant setting, also when compared with chemotherapy; this advantage was still present at metastatic sites regardless dimensions of metastasis. Authors propose that the additional survival advantage of immunotherapy as neoadjuvant treatment could lie in the activation of T cell antitumor immunity, which is not possible, or at least much less effective, with chemotherapeutic agents. The exact mechanism explaining why immunotherapy in a neoadjuvant setting showed an advantage in terms of survival and tumor control is still not clear (12); we can speculate that it might be due to a vaccines-like mechanism spurring the circulation of tumor antigens from dead tumor cells, that allow a prime and expansion of tumor specific T-cells and might also enhance their affinity for tumor cells (11). The perspective of a potential larger cohort of patients that could benefit from a surgical resection after an induction treatment using immune checkpoint blockade could reveal some important and challenging questions. Regardless of the use of minimally invasive techniques, surgery causes a temporary postoperative immunological unbalance [the so-called postoperative systemic inflammatory response syndrome (SIRS)] (13), which may vary considerably between different patients based on genetic susceptibility. Indeed, the development of SIRS is strictly related to postoperative complications, morbidity and mortality (14). Several risk factors for development of this syndrome have been analyzed and consequent therapeutically solutions have been proposed with disappointing results. The importance of an immunological disequilibrium seems to be even more dramatic in patients treated with immune checkpoint blockade, in whom this might results in a loss of immunological control of the cancer, causing even its growth and spread.

Remarkably, the authors reported that 32% of patients had not residual tumor found at the pathological evaluation of surgical specimens. Similarly, in a study analyzing early stage NSCLC patients treated with neoadjuvant nivolumab, some cases showed size increment of tumor lesion despite a major pathological response was identified in the specimen (5). As matter of fact, radiological re-evaluation after neoadjuvant treatment is usually based on dimensional criteria well-known as the RECIST guidelines (15), which are mainly based on unidimensional parameters; beside RECIST, WHO bi-dimensional criteria might also be use. Nonetheless, radiological and pathological re-evaluation are not always consistent, since a good radiological response might hide persistency of viable tumor cell (16). This inconsistency between radiological and pathological re-evaluation might be explained by the immune-cell infiltration of the tumor that is triggered by the therapy itself and cause some microenvironment change in the surrounding stromal tissue potentially misinterpreted by the imaging. Recently, developments in radiomics disclose new parameters that might be used and interpreted to define more precisely neoadjuvant response (17). In the light of these evidences, it might be important to rethink evaluation criteria of tumor response to therapies. Circulating tumor cells (18) or circulating biomarker will play a role of paramount importance, giving the real-time feedback of tumor status and possible treatment efficacy; these parameters could be therefore interpreted with radiological and clinical data in order to give a final report and a consistent base for surgical indication.

In conclusion, the immunotherapy has been changing our habitual every-day clinical decision process in the treatment of NSCLC. Surgery and medical oncology will have to redefine their roles and possibly a larger amount of patient will benefit from immunotherapy, chemotherapy and surgery in different settings according to clinic-pathological features of cancer patients. Medical oncologists and surgeons will be called to collaborate and find the best way to integrate new therapies for new patients in new settings.


Acknowledgements

None.


Footnote

Conflicts of Interest: The authors have no conflicts of interest to declare.


References

  1. Raju S, Joseph R, Sehgal S. Review of checkpoint immunotherapy for the management of non-small cell lung cancer. Immunotargets Ther 2018;7:63-75. [Crossref] [PubMed]
  2. Filosso PL, Guerrera F, Lausi PO, et al. Locally advanced non-small cell lung cancer treatment: another step forward. J Thorac Dis 2017;9:4908-11. [Crossref] [PubMed]
  3. Ghysen K, Vansteenkiste J. Immunotherapy in patients with early stage resectable nonsmall cell lung cancer. Curr Opin Oncol 2018. [Epub ahead of print]. [Crossref] [PubMed]
  4. Bott MJ, Cools-Lartigue J, Tan KS, et al. Safety and Feasibility of Lung Resection After Immunotherapy for Metastatic or Unresectable Tumors. Ann Thorac Surg 2018;106:178-83. [Crossref] [PubMed]
  5. Forde PM, Chaft JE, Pardoll DM. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N Engl J Med 2018;379:e14. [Crossref] [PubMed]
  6. Shu CA, Grigg C, Chiuzan C, et al. Neoadjuvant atezolizumab + chemotherapy in resectable non-small cell lung cancer (NSCLC). J Clin Oncol 2018;36:8532. [Crossref]
  7. Yeh J, Marrone KA, Forde PM. Neoadjuvant and consolidation immuno-oncology therapy in stage III non-small cell lung cancer. J Thorac Dis 2018;10:S451-9. [Crossref] [PubMed]
  8. Ledford H. Cancer treatment: The killer within. Nature 2014;508:24-6. [Crossref] [PubMed]
  9. Guerrera F, Renaud S, Tabbò F, et al. How to design a randomized clinical trial: tips and tricks for conduct a successful study in thoracic disease domain. J Thorac Dis 2017;9:2692-6. [Crossref] [PubMed]
  10. Bertoglio P, Renaud S, Guerrera F. Unless I see, I will not believe. J Thorac Dis 2017;9:2835-8. [Crossref] [PubMed]
  11. Liu J, Blake SJ, Yong MC, et al. Improved Efficacy of Neoadjuvant Compared to Adjuvant Immunotherapy to Eradicate Metastatic Disease. Cancer Discov 2016;6:1382-99. [Crossref] [PubMed]
  12. Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568-71. [Crossref] [PubMed]
  13. Okamura A, Watanabe M, Fukudome I, et al. Relationship Between Visceral Obesity and Postoperative Inflammatory Response Following Minimally Invasive Esophagectomy. World J Surg 2018. [Epub ahead of print]. [Crossref] [PubMed]
  14. Dieleman JM, Peelen LM, Coulson TG, et al. Age and other perioperative risk factors for postoperative systemic inflammatory response syndrome after cardiac surgery. Br J Anaesth 2017;119:637-44. [Crossref] [PubMed]
  15. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228-47. [Crossref] [PubMed]
  16. William WN Jr, Pataer A, Kalhor N, et al. Computed tomography RECIST assessment of histopathologic response and prediction of survival in patients with resectable non-small-cell lung cancer after neoadjuvant chemotherapy. J Thorac Oncol 2013;8:222-8. [Crossref] [PubMed]
  17. Coroller TP, Agrawal V, Huynh E, et al. Radiomic-Based Pathological Response Prediction from Primary Tumors and Lymph Nodes in NSCLC. J Thorac Oncol 2017;12:467-76. [Crossref] [PubMed]
  18. Sun W, Li G, Wan J, et al. Circulating tumor cells: A promising marker of predicting tumor response in rectal cancer patients receiving neoadjuvant chemo-radiation therapy. Oncotarget 2016;7:69507-17. [Crossref] [PubMed]
Cite this article as: Guerrera F, Tabbò F, Ruffini E, Bertoglio P. Changing paradigms of non-small cell lung cancer treatment. J Thorac Dis 2018;10(Suppl 33):S4170-S4172. doi: 10.21037/jtd.2018.11.04

Download Citation