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 15;6(7):102210.
doi: 10.1016/j.xcrm.2025.102210. Epub 2025 Jun 27.

Overcoming resistance to immunotherapy by targeting CD38 in human tumor explants

Affiliations

Overcoming resistance to immunotherapy by targeting CD38 in human tumor explants

Or-Yam Revach et al. Cell Rep Med. .

Abstract

CD38, an ecto-enzyme involved in NAD+ catabolism, is highly expressed in exhausted CD8+ T cells and has emerged as an attractive target to improve response to immune checkpoint blockade (ICB) by blunting T cell exhaustion. However, the precise role(s) and regulation of CD38 in exhausted T cells and the efficacy of CD38-directed therapeutic strategies in human cancer remain incompletely defined. Here, we show that CD38+CD8+ T cells are induced by chronic TCR activation and type I interferon stimulation and confirm their association with ICB resistance in human melanoma. Disrupting CD38 restores cellular NAD+ pools and improves T cell bioenergetics and effector functions. Targeting CD38 restores ICB sensitivity in a cohort of patient-derived organotypic tumor spheroids from explanted melanoma specimens. These results support further preclinical and clinical evaluation of CD38-directed therapies in melanoma and underscore the importance of NAD+ as a vital metabolite to enhance those therapies.

Keywords: 3D microfluidic culture; CD38; NAD(+); PD-1; T cell exhaustion; cytokines; ex vivo; immunotherapy; organotypic tumor spheroids.

PubMed Disclaimer

Conflict of interest statement

Declaration of interests R.W.J. is a member of the advisory board for and has a financial interest in Xsphera Biosciences Inc., a company focused on using ex vivo profiling technology to deliver functional, precision immuno-oncology solutions for patients, providers, and drug-development companies. R.W.J. has received honoraria from Incyte (invited speaker), G1 Therapeutics (advisory board), and Bioxcel Therapeutics (invited speaker). R.W.J. has an ownership interest in US patents US20200399573A9 and US20210363595A1. R.W.J.’s interests were reviewed and are managed by Massachusetts General Hospital and Mass General Brigham in accordance with their conflict-of-interest policies. A.M. has served a consultant/advisory role for Third Rock Ventures, Asher Biotherapeutics, Abata Therapeutics, ManaT Bio, Flare Therapeutics, venBio Partners, BioNTech, Rheos Medicines, and Checkmate Pharmaceuticals, is currently a part-time Entrepreneur in Residence at Third Rock Ventures, is an equity holder in ManaT Bio, Asher Biotherapeutics, and Abata Therapeutics, and has received research funding support from Bristol-Myers Squibb. A.M.’s interests were reviewed and are managed by Massachusetts General Hospital and Mass General Brigham in accordance with their conflict-of-interest policies. J.M.S., L-C.C., N.S., M.M., N.N., and R.S. are current employees with Teiko.bio and own stock. E.E. and S.A.E. were employed with Teiko.bio in the past 2 years and own stock. R.S. and M.H.S. are Teiko.bio’s co-founders, and R.S. serves on the company board. M.H.S. serves as an advisor for Teiko.bio and owns stock. M.E.W. worked as a contractor for Teiko.bio during this project. M.H.S. is a co-founder and owns stock in Prox Biosciences, has received a speaking honorarium from Standard BioTools, Arsenal, and Kumquat Bio, has been a paid consultant for Teiko.bio, Prox Biosciences, Five Prime, Ono, January, Earli, Astellas, and Indaptus, and has received research funding from Roche/Genentech, Pfizer, Valitor, and Bristol Myers Squibb. X.W. and C.R.F. report a patent on the B7-H3 CAR T cells (US10519214B2). K.T.F. serves on the Board of Directors of Clovis Oncology, Strata Oncology, Kinnate, and Scorpion Therapeutics and on Scientific Advisory Boards of PIC Therapeutics, Apricity, C-Reveal, Tvardi, ALX Oncology, xCures, Monopteros, Vibliome, Karkinos, Soley Therapeutics, Alterome, Immagene, and intrECate, and is a consultant to Nextech, Takeda, Novartis, Transcode Therapeutics, and Roche/Genentech. R.T.M. consults for Bristol Myers Squibb. G.M.B. has sponsored research agreements with Olink Proteomics, Teiko.bio, InterVenn Biosciences, and Palleon Pharmaceuticals. She has served on advisory boards for Iovance, Merck, Nektar Therapeutics, Novartis, and Ankyra Therapeutics. She consults for Merck, InterVenn Biosciences, Iovance, and Ankyra Therapeutics. She holds equity in Ankyra Therapeutics. M.S.-F. has received funding from Calico Life Sciences, Bristol-Myers Squibb, and Istari Oncology and has served as a consultant for Galvanize Therapeutics. N.H. holds equity in BioNTech, is an advisor for Related Sciences/Danger Bio, Repertoire Immune Medicines, and CytoReason, and receives research funding from Calico Life Sciences and Bristol-Myers Squibb. D.L. serves on the scientific advisory board for Oncovalent Therapeutics and has received honoraria from Genentech.

Figures

None
Graphical abstract
Figure 1
Figure 1
CD38+CD8+ TILs predict ICB resistance (A–H) scRNA-seq of CD45+ immune cells from melanoma patients. (A) CD8+ T cell clusters (n = 6,350). (B–D) Expression of (B) CD38, (C) PD-1 (PDCD1), and (D) TCF7. (E) CD8+ T cell clusters by ICB response, (F) Proportion of CD38 expressing CD8+ T cells in ICB responders (ICB-R) and non-responders (ICB-NR). Two-sided unpaired t test. Means (bars) and individual values (open circles) are shown. (G and H) Receiver-operating characteristic (ROC) curves, demonstrating (G) the predictive power of CD38+CD8+ TILs in melanoma tumors and (H) the specific performance of cluster 6 exhausted CD8+ T cells for ICB resistance. FPR, false-positive rate; TPR, true-positive rate. (I–L) scRNA-seq analysis of CD8 T cells from melanoma validation cohort. (I) Uniform manifold approximation and projection (UMAP) of CD8 T cells (n = 20,210). (J) Proportion of CD38 expressing CD8+ T cells. Two-sided unpaired t test. Means (bars) and individual values (open circles) are shown. (K) CD38 expression and (L) TCF7 expression. (M) Proportion of CD38 expressing CD8+ T cells from NSCLC (MPR, major pathological response; ICB-R, n = 23, non-MPR; ICB-NR, n = 34, two-sided unpaired t test). Means (bars) and individual values (open circles) are shown. (N) ROC curve demonstrating the predictive power of CD38+CD8+ T cells for lack of ICB treatment benefit in NSCLC. See also Figures S1 and S2; Table S1.
Figure 2
Figure 2
Intratumoral CD38+CD8+ T cells accumulate during tumor progression (A) scRNA-seq of T/NK cells from B16 tumors (Tum), tumor-draining lymph nodes (dLN), and normal lymph nodes (nLN). (B and C) Dotplots indicating (B) Cd38 expression and (C) Cd38 and Tcf7 expression at days 7, 10, and 16. (D–G) CyTOF analysis of CD38+CD8+ T cells in peripheral blood from melanoma patients (D) by response to ICB; before (E) and after (F) ICB treatment. (G) CD8+CD38+IgG4+ by response to ICB. Two-sided unpaired t test. Means (bars) and individual values (open circles) are shown. (H–M) scRNA-seq of T/NK tumor-infiltrating leukocytes from control/IgG (n = 3) and αPD-1 (n = 4) B16-ova tumors. (H) UMAP of T/NK cell clusters by condition; (I and J) Cd38 expression and (K) proportion of Cd38 expressing terminal effector CD8+ TILs per condition. Two-sided unpaired t test. Median (line) and individual values (open circles) are shown. (L and M) UMAP and track plots showing Cd38 gene expression. Immune population statistics can be found in Figure S3D. ∗p < 0.05. See also Figure S3 and Table S2.
Figure 3
Figure 3
CD38hiCD8+ T cells are dysfunctional (A) Expression of exhaustion and effector/memory-related genes in CD8+ TILs from human melanoma tumors. (B) Co-expression deviation proportion plot demonstrating co-expression of exhaustion-related genes and TCF7 from melanoma validation cohort. (C and D) differentially expressed genes based on CD38 expression in (C) CD8+ TILs from human melanoma and (D) CD3+ TILs from B16-ova murine melanoma. (E and F) CD38hi and CD38lo B7-H3.CAR-T cells (E) surface staining of PD-1+CD39+TIM-3+ (n = 3; two-sided paired t test) and (F) TCF7 expression (n = 3; two-sided unpaired t test). Means (bars) and individual values (open circles) are shown. (G) Scheme depicting acute and chronic TCR stimulation. (H–J) (H) Acute and chronic B7-H3.CAR-T proliferation assay (n = 3; two-way ANOVA with Sidak correction for multiple comparisons). Means ± SEM (shaded area) are shown. Staining of (I) CD38+CD39+ and (J) PD-1+TIM-3+ (n = 3, two-sided paired t test). Means (bars) and individual values (open circles) are shown. (K) Cytotoxicity assay toward 10164 patient-derived melanoma cell line. A representative experiment out of three is presented; two more are in Figures S4E and S4F. (n = 3 biological replicates; three independent experiments; two-way ANOVA with Sidak correction for multiple comparisons). Means ± SEM (shaded area) are shown. (L–N) Analysis of chronically stimulated control sgRNA and CD38 sgRNA B7-H3.CAR-T cells. (L) Proliferation assay (n = 3 biological replicates; three independent experiments; two-way ANOVA with Sidak correction for multiple comparisons). Means ± SEM (shaded area) are shown. (M) TCF7 intracellular staining (n = 4; two-sided paired t test). Means (bars) and individual values (open circles) are shown. (N) Cytotoxicity assay against 10164 melanoma cells (n = 3 biological replicates; three independent experiments; two-way ANOVA with Sidak correction for multiple comparisons). Means ± SEM (shaded area) are shown. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗∗p < 0.0001. See also Figure S4; Tables S3 and S4.
Figure 4
Figure 4
CD38+ T cells exhibit altered bioenergetics (A) Correlation analysis between the GSEA of CD38+/−CD8+ T cells in human melanoma and CD38+/−CD3+ T cells in B16-ova murine melanoma. (B–E) Flow cytometry of (B and D) mitochondrial mass and (C and E) MMP in indicated groups (n = 3; two-sided paired t test). Means (bars) and individual values (open circles) are shown. (F) Oxygen consumption rate (OCR) under basal condition and in response to inhibitors (n = 5, two biological replicates, two-way ANOVA with Sidak correction for multiple comparisons). Data are presented as mean ± SEM. (G and H) Relative levels of (G) NAD+ (nicotinamide adenine dinucleotide) and (H) NADP+ (nicotinamide adenine dinucleotide phosphate); log2 fold change (L2FC) from control is shown (n = 6 biological replicates; two independent experiments; two-sided unpaired t test). Means (bars) and individual values (open circles) are shown. (I) scheme of NAD+ metabolism and L2FC of indicated analytes. (J) OCR as in (F) of B7-H3.CAR-T cells ± CD38i (n = 5, two biological replicates, two-way ANOVA with Sidak correction for multiple comparisons). Data are presented as mean ± SEM. (K and L) staining of (K) TIM-3+PD-1+ and (L) CD39+TIM-3+ in B7-H3.CAR-T ± CD38i (n ≥ 4; two-sided paired t test). Means (bars) and individual values (open circles) are shown. (M) TCF7 expression by RT-qPCR in B7-H3.CAR-T cells in indicated groups (n = 4; two-sided unpaired t test). Means (bars) and individual values (open circles) are shown. (N and O) Relative levels of (N) NAM (nicotinamide) and (O) ADPR (adenosine diphosphate ribose) in B7-H3.CAR-T cells ± CD38i (n = 6 biological replicates; two independent experiments; two-sided unpaired t test). Means (bars) and individual values (open circles) are shown. ∗p < 0.05, ∗∗p < 0.01. See also Figures S5–S7; Tables S5, S6, and S8.
Figure 5
Figure 5
Type I interferon induces CD38 expression in T cells (A and B) Expression of type I IFN-stimulated genes in CD8+ TILs from human melanoma (A) by cluster and (B) by response to ICB. (C–E) Staining of CD38 in human CD8+ TILs (n = 3; in C, two-sided paired t test; in E, two-way ANOVA with Sidak correction for multiple comparisons). Means (bars) and individual values (open circles) are shown. (F) Analysis of relative NAD(H) in control or IFN-β-treated CD8+ TILs (n = 3; two-sided unpaired t test). Means (bars) and individual values (open circles) are shown. (G and H) staining of TIM-3 in CD8+ TILs in indicated groups (n > 4; two-sided paired t test). (I and J) Flow-cytometry analysis of (I) MMP and (J) mitochondrial mass of CD8+ TILs ± IFN-β (n = 3; two-sided unpaired t test). Means (bars) and individual values (open circles) are shown. ∗p < 0.05; ns, not significant. See also Figure S8.
Figure 6
Figure 6
CD38 blockade overcomes ICB resistance (A) Scheme of PDOTS preparation. (B) Viability assessment of melanoma PDOTS (n = 27) following treatment with anti-PD-1 (pembrolizumab), anti-CD38 (daratumumab), or the combination. Individual values (open circles) indicate the mean for each PDOTS specimen; one-way ANOVA with Greenhouse-Geisser correction for multiple comparisons. (C) Waterfall plot of melanoma PDOTS (n = 27). Response is defined as a 30% reduction from control (lower dashed lines); growth is defined as a 20% increase from control (upper dashed lines). Viability percentages of controls are in Table S10. (D and E) PDOTS viability assessment with indicated treatments (n = 3 biological replicates per PDOTS specimen, one-way ANOVA with Tukey correction for multiple comparisons). Means (bars) and individual values (open circles) are shown. (F) Representative images of PDOTS in (E). HO, Hoechst (blue); PI, propidium iodide (dead cells [red]). Scale bars, 100 μm. (G) scRNA-seq analysis of CD8+ T cells (n = 39,621) from tumors in (B) and (C) (n = 21), showing CD38 expression. (H) CD38 expression in indicated clusters (n = 21, one-way ANOVA with Tukey correction for multiple comparisons). (I) GSEA of CD38+ PD-1 blockade responding tumors in Prolif.CD8 and CXCL13+ T cell clusters (n = 12). (J) Module score of IFNG and GZMA in Prolif.CD8 and CXCL13+ T cells discriminate PDOTS responsive (R) and non-responsive (NR) to dual PD-1/CD38 blockade (R, n = 12; NR, n = 8; individual graphs are in Figures S11H and S11I). (K and L) Viability assessment of (K) B16-ova MDOTS (n = 6 biological replicates, two independent experiments) and (L) CT26-GFP MDOTS treated with indicated treatments (n = 12 biological replicates, four independent experiments, one-way ANOVA with Tukey correction for multiple comparisons). Means (bars) and individual values (open circles) are shown. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001; ns, not significant. See also Figures S9–S12; Tables S10, S11, S12, and S13.
Figure 7
Figure 7
Disrupting CD38 restores NAD+ and overcomes ICB resistance (A and B) Viability assessment of (A) B16-ova MDOTS (n = 6 biological replicates, two independent experiments) and (B) CT26-GFP MDOTS (n = 12 biological replicates, four independent experiments). One-way ANOVA with Tukey correction for multiple comparisons. Means (bars) and individual values (open circles) are shown. (C) Representative images of MDOTS in (B). HO, Hoechst (blue); PI, propidium iodide (dead cells [red]); tumor, GFP-tumor cells. (D) Viability assessment of CT26-GFP MDOTS with indicated treatments (n = 6 biological replicates, two independent experiments, one-way ANOVA with Tukey correction for multiple comparisons). Means (bars) and individual values (open circles) are shown. (E) Viability assessment of melanoma PDOTS with indicated treatments. (n = 18, six independent specimens; mixed-effects one-way ANOVA with Tukey correction for multiple comparisons). (F) Viability assessment of melanoma PDOTS treated with indicated treatments (n = 3 biological replicates; one-way ANOVA with Tukey correction for multiple comparisons). Means (bars) and individual values (open circles) are shown. (G) Scheme demonstrating the effect of targeting CD38 in T cells by increasing NAD+ and TCF7 expression along with restoring mitochondrial function and overcoming resistance to ICB. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001; ns, not significant. See also Figure S12 and Table S14.

Similar articles

Cited by

References

    1. Jenkins R.W., Barbie D.A., Flaherty K.T. Mechanisms of resistance to immune checkpoint inhibitors. Br. J. Cancer. 2018;118:9–16. - PMC - PubMed
    1. Sharma P., Hu-Lieskovan S., Wargo J.A., Ribas A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell (Cambridge, MA, U. S.) 2017;168:707–723. - PMC - PubMed
    1. VanderWalde A., Bellasea S.L., Kendra K.L., Khushalani N.I., Campbell K.M., Scumpia P.O., Kuklinski L.F., Collichio F., Sosman J.A., Ikeguchi A., et al. Ipilimumab with or without nivolumab in PD-1 or PD-L1 blockade refractory metastatic melanoma: a randomized phase 2 trial. Nat. Med. 2023;29:2278–2285. - PMC - PubMed
    1. Chesney J., Lewis K.D., Kluger H., Hamid O., Whitman E., Thomas S., Wermke M., Cusnir M., Domingo-Musibay E., Phan G.Q., et al. Efficacy and safety of lifileucel, a one-time autologous tumor-infiltrating lymphocyte (TIL) cell therapy, in patients with advanced melanoma after progression on immune checkpoint inhibitors and targeted therapies: pooled analysis of consecutive cohorts of the C-144-01 study. J. Immunother. Cancer. 2022;10 doi: 10.1136/jitc-2022-005755. - DOI - PMC - PubMed
    1. Oliveira G., Egloff A.M., Afeyan A.B., Wolff J.O., Zeng Z., Chernock R.D., Zhou L., Messier C., Lizotte P., Pfaff K.L., et al. Preexisting tumor-resident T cells with cytotoxic potential associate with response to neoadjuvant anti-PD-1 in head and neck cancer. Sci. Immunol. 2023;8 - PMC - PubMed

MeSH terms