Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 Jul;31(7):2385-2396.
doi: 10.1038/s41591-025-03699-3. Epub 2025 Jun 5.

Inherited mitochondrial genetics as a predictor of immune checkpoint inhibition efficacy in melanoma

Affiliations

Inherited mitochondrial genetics as a predictor of immune checkpoint inhibition efficacy in melanoma

Kelsey R Monson et al. Nat Med. 2025 Jul.

Abstract

Response to immune checkpoint inhibitors (ICIs) in metastatic melanoma (MM) varies among patients, and current baseline biomarkers predicting treatment outcomes are limited. As mitochondrial (MT) metabolism has emerged as an important regulator of host immune function, we explored the association of host MT genetics (MT haplogroups) with ICI efficacy in 1,225 ICI-treated patients with MM from the clinical trial CheckMate-067 and the International Germline Immuno-Oncology Melanoma Consortium. We discovered and validated significant associations of MT haplogroup T (HG-T) with resistance to anti-programmed cell death protein-1-based ICI (both single-agent and combination) and have shown that HG-T is independent from established tumor predictors. We also found that patients belonging to HG-T exhibit a unique nivolumab-resistant baseline peripheral CD8+ T cell repertoire compared to other MT haplogroups, providing, to our knowledge, the first link between MT inheritance, host immunity and ICI resistance. The study proposes a host blood-based biomarker with stand-alone clinical value predicting ICI efficacy and points to an ICI-resistance mechanism associated with MT metabolism, with clinical relevance in immuno-oncology.

PubMed Disclaimer

Conflict of interest statement

Competing interests: J.E.H., J.X., V.C., S.D., A.K.-J., D.S., E.K., A.B., C.S., M.I., W.O., R.M.L., M.C., G.M., Y.S., K.C., W.R., M.S.E. and I.O. report no competing interests. K.R.M., R.F., L.M. and T.K. are inventors on a patent application filed by NYU covering the findings described in this manuscript. Y.L., H.T., S.D. and D.T. are employed by Bristol Myers Squibb. A.C.P. reports consultancy roles with BMS, Merck, Replimune and Regeneron and participation in a speaker’s bureau for BMS. J.B.A.G.H. reports advisory roles for AZ, Achilles Therapeutics, BioNTech, CureVac, Immunocore, Iovance Biotherapeutics, Instil Bio, MSD, Molecular Partners, Neogene Therapeutics, Novartis, Roche, Sanofi, T-Knife and Third Rock Ventures; grant support from Amgen, Asher Bio, BioNTech, BMS, Novartis and Sastra Cell Therapy; and stock options in Neogene Therapeutics and Sastra Cell Therapy. T.F.G. reports commercial research grants from Roche-Genentech, Bristol Myers Squibb, Merck, Incyte, Seagen, Celldex, Evelo, Bayer, Aduro, Pyxis; is a consultant/advisory board member for Roche-Genentech, Merck, AbbVie, Bayer, Jounce, Aduro, Fog Pharma, Adaptimmune, FivePrime, Pyxis and Allogene and holds ownership interest (including patents) in Jounce, Pyxis, Aduro, Evelo and Bristol Myers Squibb. J.M. reports stock and other ownership in Pfizer and honoraria from Medscape and HMP; consulting fees from Merck, Sanofi/Regeneron, Bristol Myers Squibb, Seagen and Novartis; research support from GV20 Therapeutics, Astra Zeneca, Amgem, Macrogenics, Incyte, Bristol Meyers Squibb, Merck, Novartis, Regeneron, Kinnate Biopharma; and travel awards from Merck and Sharpe Dohme. M.K. serves on the scientific advisory boards of Genentech and Merck and Co. and received research support from Merck Sharp & Dohme, a subsidiary of Merck and Co., Genentech, Biogen, Novartis and the Mark Foundation. F.S.H. reports personal fees and other from Incyte; grants from Bristol Myers Squibb; grants and personal fees from Novartis; personal fees from Merck, Aduro, Amgen, 7 Hills Pharma, Apricity, Bicara, Bioentre, Catalym, Checkpoint Therapeutics, Compass Therapeutics, Genentech, Gossamer, Immunocore, Iovance, Merck, Pieris Pharmaceutical, Surface, Trillium, AstraZeneca, Corner Therapeutics and Zumutor. K.T.F. reports leadership positions at Strata Oncology, Kinnate Biopharma, Scorpion Therapeutics, Clovis Oncology; stock and other ownership interests in Clovis Oncology, Loxo, X4 Pharma, Strata Oncology, PIC Therapeutics, Apricity Health, Oncoceutics, FOGPharma, Tvardi Therapeutics, Checkmate Pharmaceuticals, Kinnate Biopharma, Scorpion Therapeutics, ALX Oncology, xCures, Monopteros Therapeutics, Vibliome Therapeutics, Transcode Therapeutics, Soley Therapeutics, Nextech Invest, Alterome Therapeutics and PreDICTA; consulting or advisory roles for Novartis, Lilly, Oncoceutics, Tvardi Therapeutics, Takeda, Debiopharm Group, OmRx Oncology, Quanta Therapeutics, Immagene and Karkinos Healthcare. I.P. owns stock in Ideaya and has consulted for Nouscom, Iovance, Nektar and Regeneron. O.R. reports that he is an employee and holds stocks at AstraZeneca. M.A.P. reports consulting fees from BMS, Merck, Novartis, Eisai, Pfizer, Chugai, Erasca, Nektar and Lyvgen and institutional support from RGenix, Infinity, BMS, Merck, Novartis, BioAtla and Genentech. R.J.S. has received grants or contracts from Merck (to institution); consulting fees from BMS, Novartis, Merck, Pfizer, Replimune and Marengo; participated on a data safety monitoring board or advisory board of Yale University and Duke University. J.J.L. reports grants or contracts from AbbVie, Astellas, AstraZeneca, Bristol Myers Squibb, Corvus, Day One, EMD Serono, F-star, Genmab, Ikena, Immatics, Incyte, Kadmon, KAHR, MacroGenics, Merck, Moderna, Nektar, Next Cure, Numab, Palleon, Pfizer, Replimune, Rubius, Servier, Scholar Rock, Synlogic, Takeda, Trishula, Tizona and Xencor; consulting fees from 7 Hills Pharma, Bright Peak, Exo, F-star, Inzen, RefleXion, Xilio, Actym, Alphamab Oncology, Arch Oncology, Duke Street Bio, Kanaph, Mavu, NeoTx, Onc.AI, OncoNano, Pyxis, Saros, Stipe, Tempest, AbbVie, Alnylam, Atomwise, Bayer, Bristol Myers Squibb, Castle, Checkmate, Codiak, Crown, Cugene, Curadev, Day One, Eisai, EMD Serono, Endeavor, Flame, G1 Therapeutics, Genentech, Gilead, Glenmark, HotSpot, Kadmon, KSQ, Janssen, Ikena, Inzen, Immatics, Immunocore, Incyte, Instil, IO Biotech, MacroGenics, Merck, Mersana, Nektar, Novartis, Partner, Pfizer, Pioneering Medicines, PsiOxus, Regeneron, Ribon, Roivant, Servier, Stingthera, Synlogic and Synthekine; participation on a data safety monitoring board or advisory board with AbbVie, Immutep and Evaxion; leadership or fiduciary role with the Society for Immunotherapy of Cancer; stock or stock options with Actym, Alphamab Oncology, Arch Oncology, Duke Street Bio, Kanaph, Mavu, NeoTx, Onc.AI, OncoNano, Pyxis, Saros, STipe and Tempest. P.A.A. has/had a consultant/advisory role for Bristol Myers Squibb, Roche-Genentech, Merck Sharp & Dohme, Novartis, Merck Serono, Pierre-Fabre, AstraZeneca, Sun Pharma, Sanofi, Sandoz, Immunocore, Italfarmaco, Nektar, Boehringer-Ingelheim, Eisai, Regeneron, Daiichi Sankyo, Pfizer, Oncosec, Nouscom, Lunaphore, Seagen, iTeos, Medicenna, Bio-Al Health, ValoTX, Replimmune, Bayer and Erasca. P.A.A. also received research funding from Bristol Myers Squibb, Roche-Genentech, Pfizer and Sanofi and travel support by Pfizer, Bio-Al Health and Replimmune.

Figures

Fig. 1
Fig. 1. NIVO treatment efficacy by European haplogroup.
Dark bars represent participants with NIVO CB (CR, PR or dSD). Light bars represent participants with NIVO NCB (pSD or PoD). a, Phylogenetic tree depicting MT-HG lineages and CM-067 NIVO discovery cohort treatment efficacy (n = 115 patients) with CB/NCB frequencies (counts) for the major European haplogroups (H, U/K, T, J, V, I, X, W). Inset shows CB/NCB proportions within each major European haplogroup (H, J, T, U/K). b, CM-067 and IO-GEM NIVO treatment efficacy frequencies by European haplogroup (H, U, T, J, K and Other, including W, R, V, X, I). Left to right (from top left), CM-067 NIVO discovery cohort (n = 115), CM-067 NIVO validation cohort (n = 82), CM-067 pooled (n = 197) and IO-GEM validation cohort (n = 174). c, Forest plot from logistic regression analysis showing the ORs, log odds, CIs and two-tailed P values when comparing NIVO treatment outcomes for HG-T versus other MT-HGs. Black boxes represent the point estimate for the main effect size (log odds) in discovery and validation cohorts and the lines extending from them represent the 95% CIs. Diamonds represent the log odds point estimate for pooled analyses estimating the overall magnitude of effect. Point estimates to the left of the dashed line indicate increased odds of NIVO CB, while point estimates to the right indicate increased odds of no NIVO CB. CIs that do not cross the dashed line indicate a statistically significant point estimate (P < 0.05). Source data
Fig. 2
Fig. 2. ICI treatment efficacy in IPI, NIVO or IPI-NIVO cohorts comparing HG-T and other (H, U, J, K, W, R, V, X and I) MT-HGs.
a, Meta-analysis proportions of CB and NCB comparing HG-T (n = 113) and other MT-HGs (n = 1,087) for CM-067 and IO-GEM IPI cohort (CTLA-4); CM-067 and IO-GEM NIVO cohort (PD-1); CM-067 and IO-GEM COMBO cohort. bd, Forest plots from logistic regression analysis illustrating the improvement in ICI outcomes across treatments for HG-T and other MT-HGs for the patient population in a, showing the ORs, log odds, CIs and two-tailed P values. When a treatment is shown to be a significant improvement over another, it is not presented again in subsequent comparisons. Comparison groups are as follows: other MT-HGs treated with IPI (n = 431), NIVO (n = 307) or COMBO (n = 349); HG-T treated with IPI (n = 46), NIVO (n = 39) or COMBO (n = 28). b, Logistic regression comparing ICI treatment outcomes to outcomes for other MT-HGs treated with IPI (n = 431); point estimates to the left of the dashed line indicate an improvement in outcomes over those of other MT-HGs treated with IPI. For example, for other MT-HGs, treatment outcomes in NIVO were a significant improvement over treatment outcomes in IPI (row 1), while there was no statistical difference in treatment outcomes for patients belonging to HG-T who were treated with NIVO compared to other MT-HGs treated with IPI (row 4). c, Logistic regression comparing ICI treatment outcomes to outcomes for other MT-HGs treated with NIVO (n = 307). d, Logistic regression comparing ICI treatment outcomes to outcomes for other MT-HGs treated with COMBO (n = 349). Black boxes represent the point estimate for the main effect size (log odds) and the lines extending from them represent the 95% CIs. Point estimates to the left of the dashed line indicate increased odds of ICI CB compared to the reference (patients belonging to other European haplogroups who were treated with IPI, NIVO or COMBO), while point estimates to the right indicate increased odds of no ICI CB compared to the reference. CIs that do not cross the dashed line indicate a statistically significant point estimate (P < 0.05). CB (CR, PR, dSD); NCB (pSD, PoD). Source data
Fig. 3
Fig. 3. Tumor markers and haplogroups in CM-067.
a, Correlation of tumor markers with clinical variables including log-transformed TMB, tumor PD-L1 status, composite ‘immune’ score (CD8+ T cell tumor infiltration > median, tumor IFNγ expression score > 0, or both), haplogroup (HG-T versus others) and sex (n = 255). b, Distribution of tumor markers stratified by haplogroup and ICI response for IFNγ score (n = 166), log-transformed TMB (n = 376), percentage tumor infiltration by CD8+ T cells (n = 182) and PD-L1 status (n = 476). Box plots depict lower and upper hinges (25th to 75th percentiles), median value (central line), whiskers extending from the hinges (largest and smallest values no more than 1.5 times the interquartile range) and outlying points outside this range (solid points beyond the whiskers). P values derived from Kruskal–Wallis tests. Source data
Fig. 4
Fig. 4. scRNA-seq analysis for pretreatment circulating CD8+ T cells from CM-067 NIVO patients (n = 21) and CM-915 patients (n = 31).
a, UMAP dimensionality reduction plot (CM-067, n = 21 patients). b, Bubble plot showing per-cluster cell-type classification based on T cell marker gene expression (CM-067, n = 21 patients). c, Box plots showing single-cell CD8+ T cell-type proportions stratified by NIVO treatment response (CB, NCB) and haplogroup (HGT; other haplogroups, HGO; CM-067, n = 21 patients). P value from one-tailed Wilcoxon rank-sum test for the following comparisons: cluster 1, HGT versus CB, P = 0.038; cluster 2, HGT versus CB, P = 0.021, HGT versus HGO, P = 0.045; cluster 10, HGO versus HGT, P = 0.011, HGO versus CB, P = 0.011. d, Scatterplot showing per-cluster mean expression of T cell exhaustion markers grouped by cell type. The connecting lines are used for the visualization of the changes in the gene expression of the selected markers per cluster. The clusters are ordered from least differentiated to exhausted phenotypes, based on the levels of differentiation markers as derived from b. e, Box plots showing single-cell CD8+ T cell-type proportions of baseline (postsurgical resection, pre-ICI) patient samples from CM-915 stratified by haplogroup (T haplogroup versus other haplogroups; CM-915, n = 31 patients). P value from one-tailed Wilcoxon rank-sum test, cluster 1, HG-T versus other MT-HGs, P = 0.020. Box plots depict lower and upper hinges (25th to 75th percentiles), median value (central line), whiskers extending from the hinges (largest and smallest values no more than 1.5 times the interquartile range) and outlying points outside this range (solid points beyond the whiskers). Source data
Fig. 5
Fig. 5. Baseline differences in gene expression by haplogroup comparing bulk RNA-seq from peripheral blood CD8+ T cells in CM-067.
a, Top five significantly enriched pathways from GO analysis on top DEGs from CM-067 NIVO NCB cohort (n = 62 patients) comparing HG-T (n = 9) versus other MT-HGs (n = 53). −log10-transformed P values derived from two-tailed Fisher’s exact test. b,c, Top five significantly enriched pathways from GO analysis (Fig. 5b) and COMPARTMENTS database (Fig. 5c) on top DEGs from CM-067 NIVO and COMBO cohorts (n = 212 patients) comparing HG-T (n = 17) versus other MT-HGs (n = 195). −log10-transformed P values derived from two-tailed Fisher’s exact test. d, Volcano plot from differential expression analysis from NIVO and COMBO CM-067 analysis (n = 212) showing upregulation of DEGs in the SOD pathway in HG-T. Genes with negative log2 fold change (log2FC) are downregulated in HG-T; genes with positive log2FC are upregulated in HG-T. Red points are the significant DEGs in both NIVO NCB and NIVO and COMBO analyses that are present in the SOD pathway. Yellow and blue points are DEGs that are significantly upregulated and downregulated, respectively, in HG-T in both NIVO NCB and NIVO and COMBO analyses. Light gray points are significant DEGs in the NIVO and COMBO analysis that did not overlap with DEGs in the NIVO NCB comparison. Dark gray points are genes that were not significantly differentially expressed and did not overlap between the NIVO NCB comparison and NIVO and COMBO comparison. −log10-transformed P values derived from negative binomial Wald test in DESeq2. Source data
Fig. 6
Fig. 6. PFS probability by haplogroup in CM-067.
a, PFS (in years) comparing HG-T (n = 39) and other MT-HGs (n = 339) in CM-067 NIVO and COMBO cohorts (n = 378). P value from two-tailed Kaplan–Meier (log-rank) test. b, PFS (in years) stratified by treatment, comparing HG-T treated by NIVO (n = 24) and COMBO (n = 15) and other MT-HGs treated by NIVO (n = 173) and COMBO (n = 166). P value from two-tailed Kaplan–Meier (log-rank) test. Source data
Extended Data Fig. 1
Extended Data Fig. 1. Percentage of subjects in each European haplogroup.
Haplogroup distributions comparing subjects in BMS-067 Nivo Discovery Cohort (left; n=115) and subjects with European ancestry from the 1000 Genomes Project (right; n=414). Most frequent European haplogroups (H, U, T, J, and K) are plotted individually, with less common haplogroups (W, R, V, X, I) grouped together as “Other”.
Extended Data Fig. 2
Extended Data Fig. 2. Response to ICI by haplogroup in CM-067 and IO-GEM cohorts.
Proportion of CB and NCB ICI treatment responses in NIVO, COMBO (IPI-NIVO), and IPI cohorts comparing HG-T and other (H, U, J, K, W, R, V, X, and I) MT-HGs in: (a) CM-067 NIVO cohort (n=197), two-tailed Chi-square p-value = 5.75E-03; (b) IO-GEM NIVO cohort (n=174), two-tailed Chi-square p-value = 0.04; (c) CM-067 COMBO cohort (n=181), two-tailed Chi-square p-value = 3.55E-03; (d) IO-GEM COMBO cohort (n=196), two-tailed Chi-square p-value = 0.12; (e) CM-067 IPI cohort (n=98), two-tailed Chi-square p-value = 0.39; (f) IO-GEM IPI cohort (n=379), two-tailed Chi-square p-value = 0.03; Total CheckMate-067, n=476; Total IO-GEM, n=749.
Extended Data Fig. 3
Extended Data Fig. 3. Association between HG-T and tumor markers in CM-067.
a. Results of correlation analysis comparing HG-T to demographic and tumor characteristics (n=255): age, sex, log-transformed tumor mutation burden (TMB), tumor PD-L1 status, and tumor “immune” score (a composite variable for either positive IFN-γ gene expression, increased CD8+ tumor infiltration, or both). b-d. Distribution of tumor markers by haplogroup: b. IFN-γ score (n=166); c. log-transformed tumor mutation burden (TMB) (n=376); d. % Tumor Infiltration by CD8+ T cells (n=182). Boxplots depict lower and upper hinges (25th to 75th percentiles), median value (central line), whiskers extending from the hinges (largest and smallest values no more than 1.5 times the interquartile range), and outlying points outside this range (solid points beyond the whiskers). P-values derived from two-tailed Kruskal-Wallis tests.
Extended Data Fig. 4
Extended Data Fig. 4. Cluster annotation and cell-type determination from CM-067 and CM-915 scRNA-seq.
a. Top 8 significantly differentially expressed genes per cluster from single-cell (sc)RNA-seq of peripheral blood CD8+ T cells isolated from NIVO-treated CM-067 patients (n=21). b. Per-cluster cell-type classification based on differential expression of marker genes from scRNA-seq of peripheral blood CD8+ T cells isolated from NIVO-treated CM-067 patients (n=21). c. Per-cluster gene expression of T cell exhaustion markers from scRNA-seq of peripheral blood CD8+ T cells isolated from NIVO-treated CM-067 patients (n=21). d. Per-cluster gene expression of T cell exhaustion markers in CD8+ T cells from scRNA-seq of pre-treatment PBMCs from CM-915 patients (n=31). As this was an independent analysis, clusters are labeled by the cluster identified in CM-915 (top cluster number) and, based on their expression profiles, the corresponding cluster first identified in CM-067 (bottom cluster number).
Extended Data Fig. 5
Extended Data Fig. 5. Differentially expressed genes from CM-067 bulk RNA-seq analysis of circulating CD8+ T cells comparing HG-T and other MT-HGs.
Heatmaps showing upregulated (red) and downregulated (green) genes for HG-T (pink) or other MT-HGs (blue) among a. NIVO NCB patients (n=62) and b. cohort of NIVO and COMBO CM-067 patients with bulk RNA-sequencing regardless of outcome (n=212).
Extended Data Fig. 6
Extended Data Fig. 6. Progression-free and overall survival stratified by MT-HG in CM-067 NIVO cohort (n=197) and COMBO cohort (n=181), and IMCG prognostic cohort.
a-b. Progression-free survival (PFS) stratified by MT-HG in CM-067 a. NIVO and b. COMBO cohorts. c-d. Overall survival (OS) stratified by MT-HG in CM-067 c. NIVO and d. COMBO cohorts. e-f. OS stratified by MT-HG (HG-T vs Other MT-HGs) and response in CM-067 e. NIVO and f. COMBO cohorts. Light bands represent 95% confidence intervals. g-h. OS by MT-HG in IMCG prognostic cohort for g. early-stage (Stage I-II) disease (n=915) and h. late-stage (Stage III) disease (n=109). All p-values from two-tailed Kaplan-Meier (log-rank) test.
Extended Data Fig. 7
Extended Data Fig. 7. Summary of circulating pre-treatment CD8+ T cell phenotypes defined using single-cell (sc)RNA-seq on patients from BMS-067 NIVO (n=21).
1. Clinical Benefit (CB) “response” phenotype: predominantly T cells able to be reinvigorated by anti-PD-1. 2. No Clinical Benefit in haplogroups other than HG-T (NCB-O): predominantly dysfunctional or terminally exhausted effector cells unable to be reinvigorated by anti-PD-1. 3. No Clinical Benefit in HG-T (NCB-T): few cytotoxic or exhausted effector cells, anti-PD-1 reinvigoration is insufficient. Created with BioRender.com.
Extended Data Fig. 8
Extended Data Fig. 8. Study population flow diagram.
Flow diagram illustrating the patient population, sample collection, multi-omic material extraction, sequencing/genotyping, and haplogroup determination for the analytic cohorts. Created with BioRender.com.
Extended Data Fig. 9
Extended Data Fig. 9. Genomics data analysis workflow.
Workflow for computationally determining mitochondrial haplogroups.

References

    1. Larkin, J. et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med.373, 23–34 (2015). - PMC - PubMed
    1. Hodi, F. S. et al. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol.19, 1480–1492 (2018). - PubMed
    1. Tawbi, H. A. et al. Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma. N. Engl. J. Med.386, 24–34 (2022). - PMC - PubMed
    1. Larkin, J. et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med.381, 1535–1546 (2019). - PubMed
    1. Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med.371, 2189–2199 (2014). - PMC - PubMed