KRAS and ERBB-family genetic alterations affect response to PD-1 inhibitors in metastatic nonsquamous NSCLC

Background: Programmed cell death 1 (PD-1) and PD-ligand 1 (PD-L1) inhibitors represent novel therapeutic options for advanced non-small cell lung cancer (NSCLC). However, approximately 50% of patients do not benefit from therapy and experience rapid disease progression. PD-L1 expression is the only...

Full description

Bibliographic Details
Main Authors: Marika Cinausero, Noemi Laprovitera, Giovanna De Maglio, Lorenzo Gerratana, Mattia Riefolo, Marianna Macerelli, Michelangelo Fiorentino, Elisa Porcellini, Vanessa Buoro, Francesco Gelsomino, Anna Squadrilli, Gianpiero Fasola, Massimo Negrini, Marcello Tiseo, Manuela Ferracin, Andrea Ardizzoni
Format: Article
Language:English
Published: SAGE Publishing 2019-11-01
Series:Therapeutic Advances in Medical Oncology
Online Access:https://doi.org/10.1177/1758835919885540
_version_ 1819290565163876352
author Marika Cinausero
Noemi Laprovitera
Giovanna De Maglio
Lorenzo Gerratana
Mattia Riefolo
Marianna Macerelli
Michelangelo Fiorentino
Elisa Porcellini
Vanessa Buoro
Francesco Gelsomino
Anna Squadrilli
Gianpiero Fasola
Massimo Negrini
Marcello Tiseo
Manuela Ferracin
Andrea Ardizzoni
author_facet Marika Cinausero
Noemi Laprovitera
Giovanna De Maglio
Lorenzo Gerratana
Mattia Riefolo
Marianna Macerelli
Michelangelo Fiorentino
Elisa Porcellini
Vanessa Buoro
Francesco Gelsomino
Anna Squadrilli
Gianpiero Fasola
Massimo Negrini
Marcello Tiseo
Manuela Ferracin
Andrea Ardizzoni
author_sort Marika Cinausero
collection DOAJ
description Background: Programmed cell death 1 (PD-1) and PD-ligand 1 (PD-L1) inhibitors represent novel therapeutic options for advanced non-small cell lung cancer (NSCLC). However, approximately 50% of patients do not benefit from therapy and experience rapid disease progression. PD-L1 expression is the only approved biomarker of benefit to anti-PD-1/PD-L1 therapy. However, its weakness has been evidenced in many studies. More recently, tumor mutational burden (TMB) has proved to be a suitable biomarker, but its calculation is difficult to obtain for all patients. Methods: We tested specific NSCLC genetic alterations as potential immunotherapy biomarkers. Tumor DNA was obtained from advanced NSCLC patients treated with anti-PD-1 monoclonal antibody nivolumab ( n =  44) or pembrolizumab ( n =  3). The mutational status of 22 genes was assessed by targeted next-generation sequencing and the association with survival was tested in uni- and multivariate models. The association between gene mutations and clinical benefit was also investigated. Results: The most frequently mutated genes were TP53 (49%), KRAS (43%), ERBB2 (13%), SMAD4 (13%), DDR2 (13%), STK11 (9%), ERBB4 (6%), EGFR (6%), BRAF (6%), and MET (6%). We confirmed that KRAS mut patients have a better response to PD-1 inhibitors, showing a longer progression-free survival (PFS) and overall survival (OS) than KRAS wt patients. In addition, we observed that patients with ERBB -family mutations, including EGFR, ERBB2 , and ERBB4 all failed to respond to PD-1 antibodies, independently of KRAS status. Conclusions: This study suggests that the analysis of KRAS and ERBB -family gene mutational status is valuable when assessing the clinical practice for the selection of NSCLC patients to treat with PD-1 inhibitors.
first_indexed 2024-12-24T03:24:46Z
format Article
id doaj.art-bbe74f39c6134ab4a55c01b9a32b30e3
institution Directory Open Access Journal
issn 1758-8359
language English
last_indexed 2024-12-24T03:24:46Z
publishDate 2019-11-01
publisher SAGE Publishing
record_format Article
series Therapeutic Advances in Medical Oncology
spelling doaj.art-bbe74f39c6134ab4a55c01b9a32b30e32022-12-21T17:17:23ZengSAGE PublishingTherapeutic Advances in Medical Oncology1758-83592019-11-011110.1177/1758835919885540KRAS and ERBB-family genetic alterations affect response to PD-1 inhibitors in metastatic nonsquamous NSCLCMarika CinauseroNoemi LaproviteraGiovanna De MaglioLorenzo GerratanaMattia RiefoloMarianna MacerelliMichelangelo FiorentinoElisa PorcelliniVanessa BuoroFrancesco GelsominoAnna SquadrilliGianpiero FasolaMassimo NegriniMarcello TiseoManuela FerracinAndrea ArdizzoniBackground: Programmed cell death 1 (PD-1) and PD-ligand 1 (PD-L1) inhibitors represent novel therapeutic options for advanced non-small cell lung cancer (NSCLC). However, approximately 50% of patients do not benefit from therapy and experience rapid disease progression. PD-L1 expression is the only approved biomarker of benefit to anti-PD-1/PD-L1 therapy. However, its weakness has been evidenced in many studies. More recently, tumor mutational burden (TMB) has proved to be a suitable biomarker, but its calculation is difficult to obtain for all patients. Methods: We tested specific NSCLC genetic alterations as potential immunotherapy biomarkers. Tumor DNA was obtained from advanced NSCLC patients treated with anti-PD-1 monoclonal antibody nivolumab ( n =  44) or pembrolizumab ( n =  3). The mutational status of 22 genes was assessed by targeted next-generation sequencing and the association with survival was tested in uni- and multivariate models. The association between gene mutations and clinical benefit was also investigated. Results: The most frequently mutated genes were TP53 (49%), KRAS (43%), ERBB2 (13%), SMAD4 (13%), DDR2 (13%), STK11 (9%), ERBB4 (6%), EGFR (6%), BRAF (6%), and MET (6%). We confirmed that KRAS mut patients have a better response to PD-1 inhibitors, showing a longer progression-free survival (PFS) and overall survival (OS) than KRAS wt patients. In addition, we observed that patients with ERBB -family mutations, including EGFR, ERBB2 , and ERBB4 all failed to respond to PD-1 antibodies, independently of KRAS status. Conclusions: This study suggests that the analysis of KRAS and ERBB -family gene mutational status is valuable when assessing the clinical practice for the selection of NSCLC patients to treat with PD-1 inhibitors.https://doi.org/10.1177/1758835919885540
spellingShingle Marika Cinausero
Noemi Laprovitera
Giovanna De Maglio
Lorenzo Gerratana
Mattia Riefolo
Marianna Macerelli
Michelangelo Fiorentino
Elisa Porcellini
Vanessa Buoro
Francesco Gelsomino
Anna Squadrilli
Gianpiero Fasola
Massimo Negrini
Marcello Tiseo
Manuela Ferracin
Andrea Ardizzoni
KRAS and ERBB-family genetic alterations affect response to PD-1 inhibitors in metastatic nonsquamous NSCLC
Therapeutic Advances in Medical Oncology
title KRAS and ERBB-family genetic alterations affect response to PD-1 inhibitors in metastatic nonsquamous NSCLC
title_full KRAS and ERBB-family genetic alterations affect response to PD-1 inhibitors in metastatic nonsquamous NSCLC
title_fullStr KRAS and ERBB-family genetic alterations affect response to PD-1 inhibitors in metastatic nonsquamous NSCLC
title_full_unstemmed KRAS and ERBB-family genetic alterations affect response to PD-1 inhibitors in metastatic nonsquamous NSCLC
title_short KRAS and ERBB-family genetic alterations affect response to PD-1 inhibitors in metastatic nonsquamous NSCLC
title_sort kras and erbb family genetic alterations affect response to pd 1 inhibitors in metastatic nonsquamous nsclc
url https://doi.org/10.1177/1758835919885540
work_keys_str_mv AT marikacinausero krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT noemilaprovitera krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT giovannademaglio krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT lorenzogerratana krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT mattiariefolo krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT mariannamacerelli krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT michelangelofiorentino krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT elisaporcellini krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT vanessabuoro krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT francescogelsomino krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT annasquadrilli krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT gianpierofasola krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT massimonegrini krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT marcellotiseo krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT manuelaferracin krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc
AT andreaardizzoni krasanderbbfamilygeneticalterationsaffectresponsetopd1inhibitorsinmetastaticnonsquamousnsclc