Early decrease of blood myeloid-derived suppressor cells during checkpoint inhibition is a favorable biomarker in metastatic melanoma.
T-lymphocytes
biomarkers, tumor
immunotherapy
melanoma
myeloid-derived suppressor cells
Journal
Journal for immunotherapy of cancer
ISSN: 2051-1426
Titre abrégé: J Immunother Cancer
Pays: England
ID NLM: 101620585
Informations de publication
Date de publication:
06 2023
06 2023
Historique:
accepted:
28
04
2023
medline:
9
6
2023
pubmed:
8
6
2023
entrez:
7
6
2023
Statut:
ppublish
Résumé
The need for reliable clinical biomarkers to predict which patients with melanoma will benefit from immune checkpoint blockade (ICB) remains unmet. Several different parameters have been considered in the past, including routine differential blood counts, T cell subset distribution patterns and quantification of peripheral myeloid-derived suppressor cells (MDSC), but none has yet achieved sufficient accuracy for clinical utility. Here, we investigated potential cellular biomarkers from clinical routine blood counts as well as several myeloid and T cell subsets, using flow cytometry, in two independent cohorts of a total of 141 patients with stage IV M1c melanoma before and during ICB. Elevated baseline frequencies of monocytic MDSCs (M-MDSC) in the blood were confirmed to predict shorter overall survival (OS) (HR 2.086, p=0.030) and progression-free survival (HR 2.425, p=0.001) in the whole patient cohort. However, we identified a subgroup of patients with highly elevated baseline M-MDSC frequencies that fell below a defined cut-off during therapy and found that these patients had a longer OS that was similar to that of patients with low baseline M-MDSC frequencies. Importantly, patients with high M-MDSC frequencies exhibited a skewed baseline distribution of certain other immune cells but these did not influence patient survival, illustrating the paramount utility of MDSC assessment. We confirmed that in general, highly elevated frequencies of peripheral M-MDSC are associated with poorer outcomes of ICB in metastatic melanoma. However, one reason for an imperfect correlation between high baseline MDSCs and outcome for individual patients may be the subgroup of patients identified here, with rapidly decreasing M-MDSCs on therapy, in whom the negative effect of high M-MDSC frequencies was lost. These findings might contribute to developing more reliable predictors of late-stage melanoma response to ICB at the individual patient level. A multifactorial model seeking such markers yielded only MDSC behavior and serum lactate dehydrogenase as predictors of treatment outcome.
Sections du résumé
BACKGROUND
The need for reliable clinical biomarkers to predict which patients with melanoma will benefit from immune checkpoint blockade (ICB) remains unmet. Several different parameters have been considered in the past, including routine differential blood counts, T cell subset distribution patterns and quantification of peripheral myeloid-derived suppressor cells (MDSC), but none has yet achieved sufficient accuracy for clinical utility.
METHODS
Here, we investigated potential cellular biomarkers from clinical routine blood counts as well as several myeloid and T cell subsets, using flow cytometry, in two independent cohorts of a total of 141 patients with stage IV M1c melanoma before and during ICB.
RESULTS
Elevated baseline frequencies of monocytic MDSCs (M-MDSC) in the blood were confirmed to predict shorter overall survival (OS) (HR 2.086, p=0.030) and progression-free survival (HR 2.425, p=0.001) in the whole patient cohort. However, we identified a subgroup of patients with highly elevated baseline M-MDSC frequencies that fell below a defined cut-off during therapy and found that these patients had a longer OS that was similar to that of patients with low baseline M-MDSC frequencies. Importantly, patients with high M-MDSC frequencies exhibited a skewed baseline distribution of certain other immune cells but these did not influence patient survival, illustrating the paramount utility of MDSC assessment.
CONCLUSION
We confirmed that in general, highly elevated frequencies of peripheral M-MDSC are associated with poorer outcomes of ICB in metastatic melanoma. However, one reason for an imperfect correlation between high baseline MDSCs and outcome for individual patients may be the subgroup of patients identified here, with rapidly decreasing M-MDSCs on therapy, in whom the negative effect of high M-MDSC frequencies was lost. These findings might contribute to developing more reliable predictors of late-stage melanoma response to ICB at the individual patient level. A multifactorial model seeking such markers yielded only MDSC behavior and serum lactate dehydrogenase as predictors of treatment outcome.
Identifiants
pubmed: 37286306
pii: jitc-2023-006802
doi: 10.1136/jitc-2023-006802
pmc: PMC10254874
pii:
doi:
Substances chimiques
Biomarkers
0
Types de publication
Journal Article
Research Support, Non-U.S. Gov't
Langues
eng
Sous-ensembles de citation
IM
Informations de copyright
© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.
Déclaration de conflit d'intérêts
Competing interests: TA reports institutional grants from SkylineDx, institutional grants and personal fees from Novartis, institutional grants from NeraCare, personal fees from BMS, institutional grants from Sanofi, personal fees from CeCaVa, personal fees from Pierre Fabre, outside the submitted work. NW reports an advisory role for Pierre Fabre and Sanofi, consultant's honoraria from Novartis, and has received travel support from AbbVie and Amgen outside the submitted work. FM reports receiving commercial research grants from Novartis and Roche; and has received travel support and/or speaker’s fees and/or advisor’s honoraria by Novartis, Roche, Bristol-Myers Squibb, Merck Sharp & Dohme and Pierre Fabre. PT has received travel support and/or speaker’s fees and/or advisor’s honoraria by Almirall, Biofrontera, Bristol-Myers Squibb, Curevac, Kyowa Kirin, Merck, Merck Sharp & Dohme, Novartis, Pierre Fabre, Roche, Sanofi and 4SC. CG reports receiving commercial research grants from Bristol-Myers Squibb, Novartis and Roche; and is a consultant/advisory board member for Amgen, Bristol-Myers Squibb, Merck Sharp & Dohme, Novartis and Roche. GP has received speaker’s honoraria from Novartis, Roche, Pfizer, GlaxoSmithKline and Astellas. TE has received travel support and/or speaker’s fees and/or advisor’s honoraria by Sanofi, Novartis, Bristol-Myers Squibb, Merck Sharp & Dohme, Almiral Hermal and Pierre Fabre. BW reports receiving commercial research grants from, is a consultant/advisory board member for and reports receiving travel reimbursement from Bristol-Myers Squibb and Merck Sharp & Dohme. KW-H received commercial research grants from CatalYm GmbH and travel support from Society for Immunotherapy of Cancer. No potential conflicts of interest were disclosed by the other authors.
Références
Oncotarget. 2017 Mar 28;8(13):21539-21553
pubmed: 28423487
Clin Cancer Res. 2017 May 1;23(9):2346-2355
pubmed: 27799249
Clin Cancer Res. 2014 Mar 15;20(6):1601-9
pubmed: 24323899
J Clin Oncol. 2013 Nov 20;31(33):4252-9
pubmed: 24127443
Cancer Immunol Res. 2016 Apr;4(4):345-53
pubmed: 26873574
J Clin Oncol. 1998 Mar;16(3):1103-11
pubmed: 9508197
Nature. 2014 Nov 27;515(7528):568-71
pubmed: 25428505
Cancer Discov. 2017 Mar;7(3):264-276
pubmed: 28031159
Oncoimmunology. 2020 Aug 28;9(1):1786888
pubmed: 32939320
Cancer Immunol Immunother. 2014 Sep;63(9):977-83
pubmed: 24966003
Eur J Cancer. 2021 Oct 30;158:191-207
pubmed: 34757258
Eur J Cancer. 2009 Jan;45(2):228-47
pubmed: 19097774
Proc Natl Acad Sci U S A. 2014 Mar 18;111(11):4221-6
pubmed: 24591638
J Immunother Cancer. 2018 Mar 6;6(1):18
pubmed: 29510697
Sci Immunol. 2020 Jan 3;5(43):
pubmed: 31901074
J Immunother Cancer. 2018 Jul 16;6(1):74
pubmed: 30012216
Front Immunol. 2020 Jul 30;11:1680
pubmed: 32849585
Oncol Lett. 2022 Jun 16;24(2):265
pubmed: 35765279
J Transl Med. 2020 Mar 11;18(1):121
pubmed: 32160899
J Immunother Cancer. 2020 Sep;8(2):
pubmed: 32929051
Clin Cancer Res. 2016 Nov 15;22(22):5487-5496
pubmed: 27185375
Br J Cancer. 2018 Aug;119(3):339-346
pubmed: 29950611
Transl Oncol. 2020 Dec;13(12):100865
pubmed: 32920330
Front Immunol. 2019 May 22;10:1147
pubmed: 31191529
Cancer Cell. 2019 Feb 11;35(2):238-255.e6
pubmed: 30753825
PLoS One. 2014 Feb 03;9(2):e87705
pubmed: 24498358
Front Immunol. 2022 Jul 07;13:906352
pubmed: 35874702
Nat Rev Immunol. 2021 Aug;21(8):485-498
pubmed: 33526920
Nat Med. 2018 Feb;24(2):144-153
pubmed: 29309059
Cancer Immunol Immunother. 2009 Jan;58(1):49-59
pubmed: 18446337
Cancer Immunol Res. 2014 Aug;2(8):812-21
pubmed: 24844912
Nat Immunol. 2019 Sep;20(9):1231-1243
pubmed: 31358999
Medicine (Baltimore). 2022 Aug 12;101(32):e29536
pubmed: 35960066
Exp Dermatol. 2014 Mar;23(3):202-4
pubmed: 24495013
N Engl J Med. 2017 Oct 5;377(14):1345-1356
pubmed: 28889792
Br J Cancer. 2019 Jan;120(1):16-25
pubmed: 30413826
Cancers (Basel). 2021 Dec 18;13(24):
pubmed: 34944986
Clin Cancer Res. 2004 Nov 1;10(21):7252-9
pubmed: 15534099
Nature. 2017 May 4;545(7652):60-65
pubmed: 28397821
Clin Cancer Res. 2016 Jun 15;22(12):2908-18
pubmed: 26787752
Vaccines (Basel). 2016 Nov 03;4(4):
pubmed: 27827871
J Immunother Cancer. 2021 Dec;9(12):
pubmed: 34933966
J Immunother Cancer. 2021 Feb;9(2):
pubmed: 33589521
J Clin Oncol. 2009 Dec 20;27(36):6199-206
pubmed: 19917835
Clin Cancer Res. 2016 Oct 1;22(19):4848-4858
pubmed: 27169993
Science. 2018 Mar 23;359(6382):1350-1355
pubmed: 29567705
Proc Natl Acad Sci U S A. 2019 Nov 5;116(45):22699-22709
pubmed: 31636208
Immunol Res. 2014 Aug;59(1-3):56-65
pubmed: 24825777
Lancet Oncol. 2018 Nov;19(11):1480-1492
pubmed: 30361170