Vol 16, No 6 (2020)
Case report
Published online: 2020-12-07

open access

Page views 623
Article views/downloads 582
Get Citation

Connect on Social Media

Connect on Social Media

The search for causes of resistance to pembrolizumab in lung adenocarcinoma with PD-L1 expression — focus on intestinal microbiome

Anna Grenda1, Ewelina Iwan2, Paweł Krawczyk1, Izabela Chmielewska1, Bożena Jarosz3, Katarzyna Reszka1, Tomasz Kucharczyk1, Kamila Wojas-Krawczyk1, Michał Gil1, Magdalena Słomiany-Szwarc2, Arkadiusz Bomba2, Dariusz Wasyl2, Janusz Milanowski1
Oncol Clin Pract 2020;16(6):364-368.

Abstract

Anti-PD-1 or PD-L1 immunotherapy in some patients with non-small cell lung cancer (NSCLC) may not be effective, despite the high percentage of cancer cells with PD-L1 expression (≥ 50%). TMB (tumor mutation burden), smoking status and low intestinal microbiome diversity may be associated with lack of efficacy of immune checkpoints inhibitors treatment in NSCLC patients. The case presented here concerns a non-smoking female patient with lung adenocarcinoma, in whom, despite the high percentage of PD-L1 positive tumor cells (50%), pembrolizumab therapy was ineffective. Next generation sequencing (NGS) was performed using the FOCUS panel allowing the analysis of 52 genes whose damage is associated with various types of solid tumors, including lung cancer. Benign genetic changes clinically irrelevant for patients with non-small cell lung cancer have been observed. In the meantime, profiling of the patient’s intestinal microbiome was performed, due to the fact that the composition of the intestinal microbiome may be a decisive factor in the lack of response to immunotherapy in patients with high PD-L1 expression and no driver mutations. Low diversity of bacteria in the intestines, with a noticeable dysbiosis (dysbacteriosis), was observed. The presence of bacteria Akkermansia, Enterococcaceae, Bifidobacteriaceae or Coriobacteriaceae, especially the presence of Akkermansia mucinifila seems to be a favourable factor of the possibility of obtaining response to immunotherapy and prolongation of progression-free survival (PFS). In the intestinal microbiome of the presented case, no bacteria from the Verrucomicrobia phylum, to which A. mucinifila belongs, were found. In addition, only 0.011% of Enterococcaceae were found. Studies on the intestinal microbiome in cancer patients receiving immunotherapy appear to be necessary to correctly understand the effect of microbiome composition on the effectiveness of this treatment method.

Article available in PDF format

View PDF Download PDF file

References

  1. Rinninella E, Raoul P, Cintoni M, et al. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms. 2019; 7(1).
  2. Kasai C, Sugimoto K, Moritani I, et al. Comparison of human gut microbiota in control subjects and patients with colorectal carcinoma in adenoma: Terminal restriction fragment length polymorphism and next-generation sequencing analyses. Oncol Rep. 2016; 35(1): 325–333.
  3. Dinan TG, Cryan JF. The microbiome-gut-brain axis in health and disease. Gastroenterol Clin North Am. 2017; 46(1): 77–89.
  4. Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature. 2012; 486(7402): 207–214.
  5. Vetizou M, Trinchieri G. Anti-PD1 in the wonder-gut-land. Cell Res. 2018; 28(3): 263–264.
  6. Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018; 359(6371): 97–103.
  7. Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018; 359(6371): 91–97.
  8. Derosa L, Hellmann MD, Spaziano M, et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Ann Oncol. 2018; 29(6): 1437–1444.
  9. Davis A, Chae Y, Agte S, et al. Association of tumor mutational burden with smoking and mutation status in non-small cell lung cancer (NSCLC). J Clin Oncol. 2017; 35(7_suppl): 24.
  10. Nagahashi M, Sato S, Yuza K, et al. Common driver mutations and smoking history affect tumor mutation burden in lung adenocarcinoma. J Surg Res. 2018; 230: 181–185.
  11. Norum J, Nieder C. Tobacco smoking and cessation and PD-L1 inhibitors in non-small cell lung cancer (NSCLC): a review of the literature. ESMO Open. 2018; 3(6): e000406.
  12. Garon EB, Rizvi NA, Hui R, et al. KEYNOTE-001 Investigators. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015; 372(21): 2018–2028.
  13. Reck M, Rodríguez-Abreu D, Robinson AG, et al. KEYNOTE-024 Investigators. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med. 2016; 375(19): 1823–1833.