Carcinoma gástrico com estroma linfoide: potencial biomarcador para imunoterapia “contra” o cancro do estômago?

envie a um amigo share this

Carcinoma gástrico com estroma linfoide: potencial biomarcador para imunoterapia “contra” o cancro do estômago?

Terça, 09.10.2018

O carcinoma gástrico é uma doença heterogénea do ponto de vista morfológico e molecular. Entre as variantes morfológicas, o carcinoma gástrico com estroma linfoide caracteriza-se pela abundância do infiltrado inflamatório rico em linfócitos no microambiente tumoral.

Através da investigação histopatológica, da análise digital e bioinformática (da expressão de proteínas e genes associados à resposta imunológica), os autores identificaram as características do carcinoma gástrico com estroma linfoide que são potencialmente relevantes para a aplicação de terapêuticas dirigidas contra as moléculas do checkpoint imunológico PD-1/PD-L1 (“imunoterapias”): associação com infecção por virus de Epstein-Barr e instabilidade de microsatellites, que definem dois subtipos moleculares de carcinoma gástrico com melhor resposta ao Pembrolizumab (anticorpo contra PD-1); imuno-expressão frequente (em 92% dos casos) de PD-L1, o biomarcador utilizado para seleccionar os doentes para imunoterapia; abundante infiltrado inflamatório intra- e peritumoral; enriquecimento de vias moleculares associadas à resposta T citotóxica e ativação de checkpoints imunológicos. Os resultados permitiram atribuir à infecção por virus de Epstein-Barr um papel determinante na constituição e promoção de um microambiente tumoral ativo do ponto de vista imunológico, com potencial aplicação terapêutica.

 

Autores e afiliações:

Irene Gullo1,2,3,4, Patrícia Oliveira3,4, Maria Athelogou5, Gilza Gonçalves2,3,4,6, Marta L Pinto4,7,8, Joana Carvalho3,4, Ana Valente3,4, Hugo Pinheiro3,4,9, Sara Andrade3,4,10, Gabriela M. Almeida3,4, Ralf Huss5, Kakoli Das11, Patrick Tan11,12,13, José C. Machado2,3,4, Carla Oliveira2,3,4, Fátima Carneiro1,2,3,4

 

1Department of Pathology, Centro Hospitalar de São João, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal

2Department of Pathology, Faculty of Medicine of the University of Porto (FMUP), Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal

3Institute of Molecular Pathology and Immunology at the University of Porto (Ipatimup), Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal

4Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal

5Definiens AG, Bernhard-Wicki Str 5, 80636 Munich, Germany

6Department of Biomedical Sciences and Medicine, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal

7INEB-Institute of Biomedical Engineering, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal

8ICBAS-Institute of Biomedical Sciences Abel Salazar, University of Porto, Rua Jorge de Viterbo Ferreira 228, 4050-343 Porto, Portugal

9Hospital Senhora da Oliveira, Rua dos Cutileiros 114, 4835-044 Guimarães, Portugal

10 Department of Biomedicine, Faculty of Medicine of the University of Porto (FMUP), Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal

11 Cancer and Stem Cell Biology Program, Duke-NUS Medical School, 8 College Road, 169857, Singapore

12 Genome Institute of Singapore, Biopolis, 60 Biopolis St, 138672, Singapore

13 Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Dr, 117599, Singapore

 

 

 

Abstract:

Background: Gastric cancer with lymphoid stroma (GCLS) is characterized by prominent stromal infiltration of T-lymphocytes. The aim of this study was to investigate GCLS biology through analysis of clinicopathological features, EBV infection, microsatellite instability (MSI), immune gene-expression profiling and PD-L1 status in neoplastic cells and tumor immune microenvironment.

Methods: Twenty-four GCLSs were analyzed by RNA in situ hybridization for EBV (EBER), PCR/fragment analysis for MSI, immunohistochemistry (PD-L1, cytokeratin, CD3, CD8), co-immunofluorescence (CK/PD-L1, CD68/PD-L1), NanoString gene-expression assay for immune related genes and PD-L1 copy number alterations. CD3+ and CD8+ T-cell densities were calculated by digital analysis. Fifty-four non-GCLSs were used as control group.

Results: GCLSs displayed distinctive clinicopathological features, such as lower pTNM stage (p = 0.02) and better overall survival (p = 0.01). EBV+ or MSI-high phenotype was found in 66.7 and 16.7% cases, respectively. GCLSs harbored a cytotoxic T-cell-inflamed profile, particularly at the invasive front of tumors (p < 0.01) and in EBV+ cases (p = 0.01). EBV+ GCLSs, when compared to EBV− GCLSs, showed higher mRNA expression of genes related to Th1/cytotoxic and immunosuppressive biomarkers. PD-L1 protein expression, observed in neoplastic and immune stromal cells (33.3 and 91.7%, respectively), and PD-L1 amplification (18.8%) were restricted to EBV+/MSI-high tumors and correlated with high values of PD-L1 mRNA expression.

Conclusions: This study shows that GCLS has a distinctive clinico-pathological and molecular profile. Furthermore, through an in-depth study of tumor immune microenvironment—by digital analysis and mRNA expression profiling—it highlights the role of EBV infection in promoting an inflamed tumor microenvironment, with putative therapeutic implications.

 

Revista: Gastric Cancer, 2018

 

Link: https://link.springer.com/article/10.1007/s10120-018-0836-8