Few studies were conducted investigating the immunological profiles in gastrointestinal stromal tumors (GIST). Adaptive and innate immune cells are present in the tumor microenvironment, indicating GIST as inflamed tumors. In addition, murine models suggested a potential interaction between immune components and imatinib. In this retrospective study, the GIST immunological profile was investigated through in silico analysis and immunohistochemistry (IHC), exploring the basis for immunotherapy approaches. Gene expression profiles (GEP) from 31 KIT/PDGFRA-mutant GIST were analyzed to evaluate the tumor microenvironment and immunotherapy predictive signatures such as the expanded IFN-γ-induced immune signature (EIIS) and the T-cell-inflamed signature (TIS). GEP and IHC supported the presence of immune infiltrate in GIST, with dominance of CD4+ and CD8+ T cells and M2 macrophages showing a remarkable similarity with melanoma microenvironment. The EIIS genes were expressed in most of GIST samples and positively correlated with PD-L1 abundance (p < .0001). Co-expression was also found between PD-L1 and CD8A (p < .0001) or CD8B (p = .0003). Moreover, the median TIS score for GIST was between the 65th and 70th percentile of the Cancer Genome Atlas dataset, in the same range of tumors responding to anti-PD-1/PD-L1. Analysis of the Gene Expression Omnibus database GIST samples pre- and post-treatment confirmed that imatinib downregulates PD-L1 and IRF1 expression through the inhibition of KIT and PDGFRA, thus contributing to counteract the suppressed adaptive immune response against GIST. The presence of a rich immune infiltrate in GIST along with the presence of TIS and EIIS suggests that GIST may benefit from immunotherapy along with tyrosine kinase inhibitors.

Immune microenvironment profiling of gastrointestinal stromal tumors (GIST) shows gene expression patterns associated to immune checkpoint inhibitors response / Pantaleo M.A.; Tarantino G.; Agostinelli C.; Urbini M.; Nannini M.; Saponara M.; Castelli C.; Stacchiotti S.; Fumagalli E.; Gatto L.; Santini D.; De Leo A.; Marafioti T.; Akarca A.; Sabattini E.; pession A.; Ardizzoni A.; Indio V.; Astolfi A.. - In: ONCOIMMUNOLOGY. - ISSN 2162-4011. - ELETTRONICO. - 8:9(2019), pp. e1617588.e1617588-e1617588.e1617588-10. [10.1080/2162402X.2019.1617588]

Immune microenvironment profiling of gastrointestinal stromal tumors (GIST) shows gene expression patterns associated to immune checkpoint inhibitors response

Pantaleo M. A.;Tarantino G.;Agostinelli C.;Urbini M.;Nannini M.;Saponara M.;Gatto L.;De Leo A.;pession A.;Ardizzoni A.;Indio V.;Astolfi A.
2019

Abstract

Few studies were conducted investigating the immunological profiles in gastrointestinal stromal tumors (GIST). Adaptive and innate immune cells are present in the tumor microenvironment, indicating GIST as inflamed tumors. In addition, murine models suggested a potential interaction between immune components and imatinib. In this retrospective study, the GIST immunological profile was investigated through in silico analysis and immunohistochemistry (IHC), exploring the basis for immunotherapy approaches. Gene expression profiles (GEP) from 31 KIT/PDGFRA-mutant GIST were analyzed to evaluate the tumor microenvironment and immunotherapy predictive signatures such as the expanded IFN-γ-induced immune signature (EIIS) and the T-cell-inflamed signature (TIS). GEP and IHC supported the presence of immune infiltrate in GIST, with dominance of CD4+ and CD8+ T cells and M2 macrophages showing a remarkable similarity with melanoma microenvironment. The EIIS genes were expressed in most of GIST samples and positively correlated with PD-L1 abundance (p < .0001). Co-expression was also found between PD-L1 and CD8A (p < .0001) or CD8B (p = .0003). Moreover, the median TIS score for GIST was between the 65th and 70th percentile of the Cancer Genome Atlas dataset, in the same range of tumors responding to anti-PD-1/PD-L1. Analysis of the Gene Expression Omnibus database GIST samples pre- and post-treatment confirmed that imatinib downregulates PD-L1 and IRF1 expression through the inhibition of KIT and PDGFRA, thus contributing to counteract the suppressed adaptive immune response against GIST. The presence of a rich immune infiltrate in GIST along with the presence of TIS and EIIS suggests that GIST may benefit from immunotherapy along with tyrosine kinase inhibitors.
2019
Immune microenvironment profiling of gastrointestinal stromal tumors (GIST) shows gene expression patterns associated to immune checkpoint inhibitors response / Pantaleo M.A.; Tarantino G.; Agostinelli C.; Urbini M.; Nannini M.; Saponara M.; Castelli C.; Stacchiotti S.; Fumagalli E.; Gatto L.; Santini D.; De Leo A.; Marafioti T.; Akarca A.; Sabattini E.; pession A.; Ardizzoni A.; Indio V.; Astolfi A.. - In: ONCOIMMUNOLOGY. - ISSN 2162-4011. - ELETTRONICO. - 8:9(2019), pp. e1617588.e1617588-e1617588.e1617588-10. [10.1080/2162402X.2019.1617588]
Pantaleo M.A.; Tarantino G.; Agostinelli C.; Urbini M.; Nannini M.; Saponara M.; Castelli C.; Stacchiotti S.; Fumagalli E.; Gatto L.; Santini D.; De Leo A.; Marafioti T.; Akarca A.; Sabattini E.; pession A.; Ardizzoni A.; Indio V.; Astolfi A.
File in questo prodotto:
Eventuali allegati, non sono esposti

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11585/693171
 Attenzione

Attenzione! I dati visualizzati non sono stati sottoposti a validazione da parte dell'ateneo

Citazioni
  • ???jsp.display-item.citation.pmc??? ND
  • Scopus 37
  • ???jsp.display-item.citation.isi??? 36
social impact