Cargando…

Mismatch repair deficiency, next-generation sequencing-based microsatellite instability, and tumor mutational burden as predictive biomarkers for immune checkpoint inhibitor effectiveness in frontline treatment of advanced stage endometrial cancer

OBJECTIVE: Molecular profiling is developing to inform treatment in endometrial cancer. Using real world evidence, we sought to evaluate frontline immune checkpoint inhibitor vs chemotherapy effectiveness in advanced endometrial cancer, stratified by Tumor Mutational Burden (TMB) ≥10 mut/MB and micr...

Descripción completa

Detalles Bibliográficos
Autores principales: Hill, Breana L, Graf, Ryon P, Shah, Kunal, Danziger, Natalie, Lin, Douglas I, Quintanilha, Julia, Li, Gerald, Haberberger, James, Ross, Jeffrey S, Santin, Alessandro D, Slomovitz, Brian, Elvin, Julia A, Eskander, Ramez N
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BMJ Publishing Group 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10086481/
https://www.ncbi.nlm.nih.gov/pubmed/36750267
http://dx.doi.org/10.1136/ijgc-2022-004026
_version_ 1785022163117408256
author Hill, Breana L
Graf, Ryon P
Shah, Kunal
Danziger, Natalie
Lin, Douglas I
Quintanilha, Julia
Li, Gerald
Haberberger, James
Ross, Jeffrey S
Santin, Alessandro D
Slomovitz, Brian
Elvin, Julia A
Eskander, Ramez N
author_facet Hill, Breana L
Graf, Ryon P
Shah, Kunal
Danziger, Natalie
Lin, Douglas I
Quintanilha, Julia
Li, Gerald
Haberberger, James
Ross, Jeffrey S
Santin, Alessandro D
Slomovitz, Brian
Elvin, Julia A
Eskander, Ramez N
author_sort Hill, Breana L
collection PubMed
description OBJECTIVE: Molecular profiling is developing to inform treatment in endometrial cancer. Using real world evidence, we sought to evaluate frontline immune checkpoint inhibitor vs chemotherapy effectiveness in advanced endometrial cancer, stratified by Tumor Mutational Burden (TMB) ≥10 mut/MB and microsatellite instability (MSI). METHODS: Patients with advanced endometrial cancer in the US-based de-identified Flatiron Health-Foundation Medicine Clinico-Genomic Database were included. Data originated from patients treated between January 2011- March 2022 at 280 US clinics. Next-generation sequencing assays were performed via FoundationOne or FoundationOneCDx. Longitudinal clinical data were derived from electronic health records. Immune checkpoint inhibitor treatment included pembrolizumab, dostarlimab, and nivolumab monotherapies. Time to next treatment, time to treatment discontinuation, and overall survival were assessed with the log-rank test and Cox proportional hazard models with adjusted hazard ratios (aHR) for known prognostic factors. We used the Likelihood ratio test to compare biomarker performance. RESULTS: A total of 343 patients received chemotherapy and 28 received immune checkpoint inhibitor monotherapy as frontline treatment. Patients who received monotherapy were more likely to be stage III at diagnosis (immune checkpoint inhibitor: 54.6% vs chemotherapy: 15.0%; p<0.001) and more likely to test MSI-high via next-generation sequencing (immune checkpoint inhibitor: 53.6% vs chemotherapy: 19.2%; p<0.001). In MSI-high cancers, single-agent immune checkpoint inhibitor had a more favorable time to next treatment (aHR: 0.18, p=0.001) and overall survival (aHR 0.29, p=0.045). Additional analyses on 70 unique tumor specimens revealed mismatch repair deficiency (dMMR) via immunohistochemistry and MSI-high via next-generation sequencing concordance (91%), with nominal improvement of MSI over dMMR to predict time to treatment discontinuation (p=0.030), time to next treatment (p=0.032), and overall survival (p=0.22). MSI status was concordant with tumor mutational burden ≥10 in 94.3% of cases. CONCLUSION: Immune checkpoint inhibitors may have improved efficacy over chemotherapy in frontline treatment for advanced endometrial cancer defined by MSI-high using next-generation sequencing as a nominally better predictor of outcomes than dMMR with immunohistochemistry. This provides the biologic rationale of active phase III trials.
format Online
Article
Text
id pubmed-10086481
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BMJ Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-100864812023-04-12 Mismatch repair deficiency, next-generation sequencing-based microsatellite instability, and tumor mutational burden as predictive biomarkers for immune checkpoint inhibitor effectiveness in frontline treatment of advanced stage endometrial cancer Hill, Breana L Graf, Ryon P Shah, Kunal Danziger, Natalie Lin, Douglas I Quintanilha, Julia Li, Gerald Haberberger, James Ross, Jeffrey S Santin, Alessandro D Slomovitz, Brian Elvin, Julia A Eskander, Ramez N Int J Gynecol Cancer Original Research OBJECTIVE: Molecular profiling is developing to inform treatment in endometrial cancer. Using real world evidence, we sought to evaluate frontline immune checkpoint inhibitor vs chemotherapy effectiveness in advanced endometrial cancer, stratified by Tumor Mutational Burden (TMB) ≥10 mut/MB and microsatellite instability (MSI). METHODS: Patients with advanced endometrial cancer in the US-based de-identified Flatiron Health-Foundation Medicine Clinico-Genomic Database were included. Data originated from patients treated between January 2011- March 2022 at 280 US clinics. Next-generation sequencing assays were performed via FoundationOne or FoundationOneCDx. Longitudinal clinical data were derived from electronic health records. Immune checkpoint inhibitor treatment included pembrolizumab, dostarlimab, and nivolumab monotherapies. Time to next treatment, time to treatment discontinuation, and overall survival were assessed with the log-rank test and Cox proportional hazard models with adjusted hazard ratios (aHR) for known prognostic factors. We used the Likelihood ratio test to compare biomarker performance. RESULTS: A total of 343 patients received chemotherapy and 28 received immune checkpoint inhibitor monotherapy as frontline treatment. Patients who received monotherapy were more likely to be stage III at diagnosis (immune checkpoint inhibitor: 54.6% vs chemotherapy: 15.0%; p<0.001) and more likely to test MSI-high via next-generation sequencing (immune checkpoint inhibitor: 53.6% vs chemotherapy: 19.2%; p<0.001). In MSI-high cancers, single-agent immune checkpoint inhibitor had a more favorable time to next treatment (aHR: 0.18, p=0.001) and overall survival (aHR 0.29, p=0.045). Additional analyses on 70 unique tumor specimens revealed mismatch repair deficiency (dMMR) via immunohistochemistry and MSI-high via next-generation sequencing concordance (91%), with nominal improvement of MSI over dMMR to predict time to treatment discontinuation (p=0.030), time to next treatment (p=0.032), and overall survival (p=0.22). MSI status was concordant with tumor mutational burden ≥10 in 94.3% of cases. CONCLUSION: Immune checkpoint inhibitors may have improved efficacy over chemotherapy in frontline treatment for advanced endometrial cancer defined by MSI-high using next-generation sequencing as a nominally better predictor of outcomes than dMMR with immunohistochemistry. This provides the biologic rationale of active phase III trials. BMJ Publishing Group 2023-04 2023-02-07 /pmc/articles/PMC10086481/ /pubmed/36750267 http://dx.doi.org/10.1136/ijgc-2022-004026 Text en © IGCS and ESGO 2023. Re-use permitted under CC BY-NC. No commercial re-use. Published by BMJ. https://creativecommons.org/licenses/by-nc/4.0/This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, an indication of whether changes were made, and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/ (https://creativecommons.org/licenses/by-nc/4.0/) .
spellingShingle Original Research
Hill, Breana L
Graf, Ryon P
Shah, Kunal
Danziger, Natalie
Lin, Douglas I
Quintanilha, Julia
Li, Gerald
Haberberger, James
Ross, Jeffrey S
Santin, Alessandro D
Slomovitz, Brian
Elvin, Julia A
Eskander, Ramez N
Mismatch repair deficiency, next-generation sequencing-based microsatellite instability, and tumor mutational burden as predictive biomarkers for immune checkpoint inhibitor effectiveness in frontline treatment of advanced stage endometrial cancer
title Mismatch repair deficiency, next-generation sequencing-based microsatellite instability, and tumor mutational burden as predictive biomarkers for immune checkpoint inhibitor effectiveness in frontline treatment of advanced stage endometrial cancer
title_full Mismatch repair deficiency, next-generation sequencing-based microsatellite instability, and tumor mutational burden as predictive biomarkers for immune checkpoint inhibitor effectiveness in frontline treatment of advanced stage endometrial cancer
title_fullStr Mismatch repair deficiency, next-generation sequencing-based microsatellite instability, and tumor mutational burden as predictive biomarkers for immune checkpoint inhibitor effectiveness in frontline treatment of advanced stage endometrial cancer
title_full_unstemmed Mismatch repair deficiency, next-generation sequencing-based microsatellite instability, and tumor mutational burden as predictive biomarkers for immune checkpoint inhibitor effectiveness in frontline treatment of advanced stage endometrial cancer
title_short Mismatch repair deficiency, next-generation sequencing-based microsatellite instability, and tumor mutational burden as predictive biomarkers for immune checkpoint inhibitor effectiveness in frontline treatment of advanced stage endometrial cancer
title_sort mismatch repair deficiency, next-generation sequencing-based microsatellite instability, and tumor mutational burden as predictive biomarkers for immune checkpoint inhibitor effectiveness in frontline treatment of advanced stage endometrial cancer
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10086481/
https://www.ncbi.nlm.nih.gov/pubmed/36750267
http://dx.doi.org/10.1136/ijgc-2022-004026
work_keys_str_mv AT hillbreanal mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer
AT grafryonp mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer
AT shahkunal mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer
AT danzigernatalie mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer
AT lindouglasi mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer
AT quintanilhajulia mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer
AT ligerald mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer
AT haberbergerjames mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer
AT rossjeffreys mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer
AT santinalessandrod mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer
AT slomovitzbrian mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer
AT elvinjuliaa mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer
AT eskanderramezn mismatchrepairdeficiencynextgenerationsequencingbasedmicrosatelliteinstabilityandtumormutationalburdenaspredictivebiomarkersforimmunecheckpointinhibitoreffectivenessinfrontlinetreatmentofadvancedstageendometrialcancer