Graphical abstract graphic file with name fx1.jpg [125]Open in a new tab Highlights * • CD38^hiCD8^+ T cells are associated with ICB resistance in melanoma * • CD38 expression is induced by chronic TCR stimulation and type I IFN signaling * • Blocking CD38 restores NAD^+ pools and mitochondrial function * • Dual PD-1/CD38 blockade overcomes ICB resistance in patient-derived 3D tumor models __________________________________________________________________ Revach et al. show that CD38 is elevated in dysfunctional T cells in melanoma, driving an impaired metabolic state, and that disrupting CD38 improves T cell function and restores immune checkpoint blockade (ICB) sensitivity in ICB-resistant patient-derived melanoma organotypic tumor spheroids, supporting further clinical evaluation of CD38-directed therapy in melanoma. Introduction Cancer immunotherapy with immune checkpoint blockade (ICB) has revolutionized cancer treatment, providing durable responses and cures for a subset of patients with advanced metastatic disease.[126]^1 Despite the success of ICB in melanoma and other cancers, resistance remains a central challenge and is ultimately fatal.[127]^2 Dual PD-1/CTLA-4 blockade demonstrated a modest objective response rate (28%) in the second-line setting and is associated with significant toxicities.[128]^3 An improved response rate (31.4%) was observed in ICB-resistant melanoma patients treated with adoptive T cell therapy (lifileucel),[129]^4 but a durable benefit was observed in a minority of patients, underscoring the need for rational therapeutic strategies to overcome ICB resistance. Tumor-infiltrating CD8^+ T lymphocytes (TILs) are key determinants of anti-tumor immunity and successful response to ICB.[130]^5^,[131]^6^,[132]^7 Single-cell sequencing has revealed that CD8^+ T cells defined by expression of stem-like transcription factors (e.g., TCF7) are enriched in melanoma patients sensitive to ICB, whereas exhausted CD8^+ T cells, defined by high expression of co-inhibitory receptors, are enriched in ICB-resistant melanoma patients.[133]^8 Exhausted T cells develop during chronic antigen stimulation, are characterized by loss of proliferative potential and diminished effector function, and may acquire immunosuppressive properties.[134]^7^,[135]^8^,[136]^9^,[137]^10 Therapeutic strategies directed at promoting stem-like T cell expansion, enhancing effector functions, and preventing (or reversing) T cell exhaustion would represent rational approaches to overcome ICB resistance. CD38, an ecto-enzyme involved in NAD^+ catabolism, is upregulated during T cell activation and is associated with T cell exhaustion in cancer and chronic viral infection.[138]^11^,[139]^12 CD38^hiCD8^+ T cells are induced following unsuccessful response to ICB contributing to ICB resistance, while depletion of CD38^hiCD8^+ cells improved anti-tumor immunity in murine tumor models.[140]^13 Yet the precise role of the CD38-NAD^+ axis in governing the balance of tumor-infiltrating CD8^+ T cell states is unknown. Importantly, the potential impact of targeting CD38 to overcome T cell dysfunction and ICB resistance in human melanoma tumor models has not been tested. Here, we show that the upregulation of CD38 in CD8^+ T cells is strongly associated with T cell exhaustion and ICB resistance in melanoma. CD38^+CD8^+ T cells accumulate specifically in the tumor microenvironment (TME) and exhibit features of T cell dysfunction, with low TCF7 expression, decreased proliferative capacity, and altered mitochondrial bioenergetics. Pharmacological or genetic disruption of CD38 limits the development of these functional and metabolic defects, resulting in increased TCF7 expression, improved T cell function, and restored ICB sensitivity. Importantly, we demonstrate the therapeutic potential of CD38 blockade to overcome resistance to ICB using a cohort of patient-derived organotypic tumor spheroids (PDOTS),[141]^14^,[142]^15 which retain autologous tumor-infiltrating immune and stromal cells and are amenable to ex vivo 3D microfluidic culture and drug sensitivity testing. Results CD38^+CD8^+ TILs predict ICB resistance We first examined the expression of CD38 in CD8^+ TILs using published single-cell RNA sequencing (scRNA-seq) data from patients with advanced melanoma treated with ICB ([143]Figures 1A, [144]S1A, and S1C).[145]^8 We observed increased CD38 expression in exhausted CD8^+ TILs (clusters 1–3), which closely mirrored the expression of PD-1 (PDCD1) ([146]Figures 1A–1C). CD8^+ T cell states associated with early T cell activation or effector memory function (clusters 4–6) were marked with low CD38 expression and high expression of the transcription factor TCF7 ([147]Figures 1B and 1D). As TCF7-expressing CD8^+ T cells are associated with ICB response,[148]^8 we sought to determine whether CD38-expressing CD8^+ T cells were associated with ICB resistance in human cancer. Consistent with prior observations,[149]^8^,[150]^13 we observed a higher proportion of CD38 expressing CD8^+ TILs in ICB-resistant (ICB-NR) melanoma tumors ([151]Figures 1E and 1F). CD8-specific expression of CD38 also demonstrated high predictive power for ICB resistance in melanoma patients (area under the curve [AUC] 0.87, p = 1.53 × 10^−5) ([152]Figure 1G), and higher proportions of CD38-expressing CD8^+ T cells were observed in both baseline (pre-treatment) and post-treatment tumors from ICB-resistant melanoma patients, though more robustly in the post-treatment setting ([153]Figures S1D and S1E). Furthermore, CD8^+ T cell-specific CD38 expression was associated with resistance to both single-agent PD-1 blockade and dual PD-1/CTLA-4 blockade ([154]Figures S1F and S1G). Figure 1. [155]Figure 1 [156]Open in a new tab CD38^+CD8^+ TILs predict ICB resistance (A–H) scRNA-seq of CD45^+ immune cells from melanoma patients.[157]^8 (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.[158]^16 (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[159]^17 (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 [160]Figures S1 and [161]S2; [162]Table S1. Further subcluster analysis demonstrated that exhausted CD8^+ T cells (cluster 6 in [163]Figure S1A) showed the highest predictive ability for ICB resistance (AUC 0.83, p = 5.3 × 10^−4) compared to the exhausted/cell-cycle cluster (cluster 11 in [164]Figure S1A, AUC 0.64, p = 0.068) and exhausted/heat-shock cluster (cluster 9 in [165]Figure S1A, AUC 0.62, p = 0.300) ([166]Figures 1H, [167]S1H, and S1I). CD38 expression in other T cell and immune clusters also showed lower predictive power for ICB resistance and were mostly not significant ([168]Figures S1J–S1Q). While tumor-specific expression of CD38 has been observed in non-small cell lung cancer (NSCLC) and other cancers and is associated with ICB resistance,[169]^18 CD38 expression in human melanoma tumor cells was rarely observed compared to CD45^+ immune cells ([170]Figures S1R and [171]S2A–S2D).[172]^19^,[173]^20 To validate these findings, we examined an independent cohort of ICB-treated melanoma patient tumors and evaluated tumor-infiltrating CD8^+ T cells using scRNA-seq ([174]Figures 1I and [175]S2E; [176]Table S1).[177]^16 Enrichment of CD38-expressing CD8^+ T cells was again observed in ICB-NR compared to ICB-R patients ([178]Figure 1J), with CD38 expression enriched in exhausted T cell clusters and anti-correlated with TCF7 expression ([179]Figures 1K and 1L). To determine whether the association between CD38-expressing CD8^+ T cells and ICB resistance could be observed in other cancers, we examined the expression of CD38 in CD8^+ T cells from patients with NSCLC.[180]^17 Consistent with our observations in melanoma, CD38 expression in CD8^+ T cells was associated with a lack of ICB treatment benefit in NSCLC and was predictive for lack of response (AUC 0.75, p = 0.002) ([181]Figures 1M and 1N). Taken together, these analyses indicate that CD38-expressing tumor-infiltrating CD8^+ T cells are strongly associated with ICB resistance in multiple cancer types. Intratumoral CD38^+CD8^+ T cells accumulate during tumor progression To determine the temporal dynamics of CD8^+ T cell-specific CD38 upregulation and examine their tumor specificity, we examined time-resolved scRNA-seq data from B16 murine melanoma tumors with matched tumor draining lymph nodes (dLNs) and normal lymph nodes (nLNs).[182]^21 While Cd38 expression was observed in other immune populations in tumors, dLNs, and nLNs ([183]Figure S3A), Cd38-expressing CD8^+ T cells were observed primarily within tumors ([184]Figures 2A and 2B). Further, Cd38 expression was largely absent from nLNs and dLNs but was detectable in tumors by day 7 with elevated expression at days 10 and 16 with increasing tumor burden ([185]Figure 2C). In contrast, Tcf7 expression was observed largely in CD8^+ T cells in nLNs and dLNs, with decreased expression in CD8^+ TILs ([186]Figure 2C). Further, while modest residual Tcf7 expression was observed at early time points (day 7), Tcf7 was absent by day 10 and remained undetectable at day 16 as tumor burden increased ([187]Figure 2C). Therefore, while CD38^+CD8^+ T cells can be detected in the tumor periphery, Cd38 upregulation and the parallel loss of Tcf7 expression in CD8^+ T cells occurs specifically in the TME during tumor progression. Figure 2. [188]Figure 2 [189]Open in a new tab Intratumoral CD38^+CD8^+ T cells accumulate during tumor progression (A) scRNA-seq[190]^21 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.[191]^14 (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 [192]Figure S3D. ∗p < 0.05. See also [193]Figure S3 and [194]Table S2. Higher circulating levels of CD38^+CD4^+ T cells have been associated with diminished ICB sensitivity in melanoma.[195]^9 Multiplexed cytometry by time of flight (CyTOF) analysis of peripheral blood from melanoma patients (before and after initiating ICB treatment) revealed higher levels of CD38^+CD8^+ T cells in ICB-NR patients ([196]Figure 2D and [197]Table S2). In contrast to tumors, pre-treatment circulating levels of CD38^+CD8^+ T cells were not significantly different between responders and non-responders ([198]Figure 2E), whereas higher levels of CD38^+CD8^+ T cells were observed in post-treatment ICB-NR samples ([199]Figure 2F). Increased IgG4^+ detection was also observed on peripheral blood CD38^+CD8^+ T cells in ICB-resistant patients, identifying cells bound by clinically administered IgG[4] isotype anti-PD-1 antibodies ([200]Figures 2G and [201]S3B).[202]^22 In agreement with these observations, circulating plasma levels of CD38 were increased in melanoma patients 6 weeks after initiating ICB treatment but remained elevated at 6 months only in non-responder patients ([203]Figure S3C). ICB treatment increases CD38^+CD8^+ T cells Ineffective ICB treatment promotes the induction of dysfunctional CD8^+ T cells that contribute to ICB resistance.[204]^13 To evaluate the accumulation of Cd38 expressing TILs following ICB treatment, we examined scRNA-seq data of tumor-infiltrating CD45^+ immune cells from B16-ova tumors treated with ICB.[205]^14 Anti-PD-1 treatment resulted in the expansion of early-effector and terminal exhausted/effector CD8^+ T cells with a reduction of progenitor CD8^+ T cells ([206]Figures 2H–2J and [207]S3D). Yet Cd38 expression was significantly increased only in terminal exhausted/effector CD8^+ T cells following PD-1 blockade ([208]Figures 2K–2M, [209]S3E, and S3F). Thus, while Cd38 is expressed in various T/natural killer (NK) cell populations (and multiple immune cell types), its upregulation following ICB treatment is observed primarily in exhausted CD8^+ T cells. CD38^hiCD8^+ T cells are dysfunctional We next sought to examine the association between CD38 and T cell exhaustion. Increased expression of multiple co-inhibitory receptors (PDCD1, HAVCR2, ENTPD1, CTLA4, and LAG3) was observed together with CD38 in exhausted CD8^+ T cells (clusters 1–3, see [210]Figure 1A) from ICB-treated melanoma patients,[211]^8 whereas CD38 expression was comparatively low in effector-memory CD8^+ T cells (clusters 4–6, see [212]Figure 1A) expressing TCF7 ([213]Figure 3A). Analysis of co-expression deviation proportions (CDPs) in our validation melanoma scRNA-seq dataset[214]^16 (see [215]Figures 1I–1L) also demonstrated co-expression of CD38 with multiple co-inhibitory receptors and an inverse correlation with TCF7 expression ([216]Figure 3B). Examination of differentially expressed genes in CD38^+ and CD38^− tumor-infiltrating CD8^+ T cells from melanoma patients treated with ICB[217]^8 revealed increased expression of co-inhibitory receptors (e.g., PDCD1 and HAVCR2) in CD38^+ CD8 T cells and increased expression in T cell stem/memory markers (e.g., TCF7, IL7R, and CCR7) in CD38^−CD8^+ T cells ([218]Figure 3C and [219]Table S3). Further, analysis of TILs from B16-ova tumors[220]^14 revealed similar upregulation of co-inhibitory receptors and exhaustion-related molecules in CD38^+ TILs, whereas increased expression of Tcf7, Il7r, and Ccr7 was observed in CD38^− TILs ([221]Figure 3D and [222]Table S4). Figure 3. [223]Figure 3 [224]Open in a new tab CD38^hiCD8^+ T cells are dysfunctional (A) Expression of exhaustion and effector/memory-related genes in CD8^+ TILs from human melanoma tumors.[225]^8 (B) Co-expression deviation proportion plot demonstrating co-expression of exhaustion-related genes and TCF7 from melanoma validation cohort.[226]^16 (C and D) differentially expressed genes based on CD38 expression in (C) CD8^+ TILs from human melanoma[227]^8 and (D) CD3^+ TILs from B16-ova murine melanoma.[228]^14 (E and F) CD38^hi and CD38^lo 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 [229]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 [230]Figure S4; [231]Tables S3 and [232]S4. Chimeric antigen receptor T (CAR-T) cells are increasingly utilized to study T cell biology, providing a renewable source of antigen-specific effector T cells.[233]^23^,[234]^24 To further study the mechanism of CD38 function in human T cells, we examined human B7-H3-directed CAR-T cells (B7-H3.CAR-T).[235]^25^,[236]^26 Following isolation of CD38^hi and CD38^lo B7-H3.CAR-T cells, CD38^hi CAR-T cells exhibited increased expression of exhaustion-related surface markers (PD-1, TIM-3, and CD39) and decreased expression of TCF7 and FOXO1 compared to CD38^lo B7-H3.CAR-T cells ([237]Figures 3E, 3F, and [238]S4A–S4C). To model the development of T cell dysfunction, we performed repetitive in vitro T cell receptor (TCR) stimulation (every 2–4 days) with αCD3/αCD28 beads for 10–14 days (“chronic” stimulation) compared to a one-time activation with αCD3/αCD28, followed by interleukin-2 (IL-2) expansion (“acute” stimulation) ([239]Figure 3G). Chronic TCR stimulation of CAR-T cells resulted in reduced proliferative capacity ([240]Figure 3H), increased levels of CD38^+CD39^+ and PD-1^+TIM-3^+ cells ([241]Figures 3I and 3J), and reduced ability to lyse target cancer cells compared to standard (acute) stimulation ([242]Figures 3K and [243]S4D–S4F), recapitulating key features of T cell exhaustion. Similar observations were obtained with patient-derived CD8^+ TILs and matched primary tumor cells ([244]Figures S4G and S4H). To determine whether CD38 is necessary for the development of T cell dysfunction, we deleted CD38 in B7-H3.CAR-T cells using CRISPR-Cas9 gene editing ([245]Figure S4I) followed by chronic TCR stimulation. CD38 single-guide RNA (sgRNA) B7-H3.CAR-T cells displayed a clear proliferative advantage over control sgRNA CAR-T cells ([246]Figure 3L) accompanied by an increased proportion of TCF7^+ CAR-T cells ([247]Figure 3M), indicating a higher T cell stemness potential.[248]^8 Further, CD38 sgRNA CAR-T cells exhibited increased cytotoxic capacity toward patient-derived melanoma cell lines compared to control sgRNA CAR-T cells ([249]Figure 3N), indicating that CD38 deletion prevents T cell dysfunction in a human T cell exhaustion model. CD38^+ T cells exhibit altered bioenergetics To identify key cellular processes associated with upregulation of CD38 in T cells, we performed gene set enrichment analysis (GSEA) using CD8^+ TILs from ICB-treated melanoma patients[250]^8 and CD3^+ TILs from B16-ova tumors.[251]^14 Top scoring gene sets included type I/II interferon signaling (IFN-α/IFN-γ response) as well as oxidative phosphorylation (OXPHOS), which correlated strongly between mouse and human data ([252]Figures 4A and [253]S5A–S5D; [254]Tables S5 and [255]S6). Given these findings and previous reports linking CD38 to altered mitochondrial fitness,[256]^12^,[257]^27 we examined the mitochondrial mass and mitochondrial membrane potential (MMP) of CD38^lo and CD38^hi T cells. Increased mitochondrial mass and MMP were observed in CD38^hi B7-H3.CAR-T cells and human melanoma CD8^+ TILs ([258]Figures 4B, 4C, and [259]S5E–S5G), consistent with previous reports.[260]^12^,[261]^28 Importantly, CD38 deletion resulted in reduced mitochondrial mass and lower MMP compared to control sgRNA following chronic TCR stimulation ([262]Figures 4D and 4E), indicating enhanced T cell mitochondrial fitness.[263]^12^,[264]^28^,[265]^29 In line with these observations, CD38 sgRNA CAR-T cells exhibited increased extracellular acidification (ECAR) ([266]Figure S5H), indicating enhanced glycolysis[267]^30 and increased mitochondrial spare respiratory capacity following the addition of the mitochondrial uncoupling agent, FCCP (carbonyl cyanide-p-trifluoromethoxyphenylhydrazone), suggesting improved T cell fitness under conditions of increased metabolic stress ([268]Figure 4F).[269]^31 Figure 4. [270]Figure 4 [271]Open in a new tab CD38^+ T cells exhibit altered bioenergetics (A) Correlation analysis between the GSEA of CD38^+/−CD8^+ T cells in human melanoma[272]^8 and CD38^+/−CD3^+ T cells in B16-ova murine melanoma.[273]^14 (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); log[2] 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 [274]Figures S5–S7; [275]Tables S5, [276]S6, and [277]S8. Disrupting CD38 restores NAD^+ in exhausted T cells As NAD^+ boosting has been shown to restore mitochondrial fitness and improve T cell function,[278]^29 we next sought to determine whether disrupting CD38 could improve mitochondrial metabolic fitness by restoring cellular NAD^+ levels. Genetic deletion or pharmacological inhibition of CD38 with 78c (CD38i)[279]^32^,[280]^33 in B7-H3.CAR-T cells resulted in increased NAD(H) levels that were further enhanced with nicotinamide riboside (NR) supplementation to boost cellular NAD^+ levels ([281]Figures S6A and S6B).[282]^29 To define the broader metabolic consequences of targeting CD38 in T cells, we performed intracellular metabolite profiling using liquid chromatography-mass spectrometry (LC-MS) of acute (CD38^hi) and chronically TCR-stimulated (CD38^low) B7-H3.CAR-T cells (see [283]Figure 3I). Chronic TCR stimulation was associated with changes in a number of metabolites, including intermediate-chain acylcarnitines, glycerol-3-phosphate shuttle metabolic intermediates (glycerol-3-phosphate and dihydroxyacetone phosphate), malate-aspartate shuttle intermediates (malate and aspartate), and NADPH ([284]Figures S6C and S6D; [285]Table S7). Of note, NADPH can be generated from NAD^+ as part of the pentose phosphate pathway (PPP) via phosphorylation by NAD^+ kinase to NADP^+ and subsequent reduction of NADP^+ to NADPH by glucose-6-phosphate dehydrogenase (G6PD), an enzyme also enriched in exhausted CD8^+ T cells in melanoma.[286]^34 Further metabolomic analysis of CD38i-treated, chronically TCR-stimulated B7-H3.CAR-T cells demonstrated increased levels of both NAD^+ and NADP^+ ([287]Figures 4G–4I and [288]Table S8), showing restoration of cellular NAD^+ pools in exhausted T cells upon CD38 inhibition. CD38i also increased the mitochondrial spare respiratory capacity of chronically TCR-stimulated B7-H3.CAR-T ([289]Figure 4J), similar to CD38-knockout T cells. Restoration of cellular NAD^+ pools was accompanied by decreased proportions of PD-1^+TIM-3^+ and CD39^+TIM-3^+ B7-H3.CAR-T cells and increased TCF7 expression ([290]Figures 4K–4M), whereas NAD^+ depletion using the nicotinamide phosphoribosyltransferase inhibitor FK866[291]^35 (NAMPTi) reduced NAD^+ levels and TCF7 expression ([292]Figures S6E and S6F), increased CD8^+PD-1^+TIM-3^+ and CD8^+CD38^+ T cells ([293]Figures S6G and S6H), and impaired proliferation and cytotoxicity ([294]Figures S6I and S6J). Further, NAD^+ boosting during chronic stimulation by NR supplementation (mimicking the effect of CD38i)[295]^36 ([296]Figure S6B) resulted in increased T cell proliferation and cytotoxicity ([297]Figures S6K and S6L), similar to the effect of CD38 inhibition (see [298]Figures 3L and 3N), with no significant changes in CD38 expression ([299]Figure S6M). Compared to CD38 inhibition or deletion, the effect of NR supplementation on TCF7 expression was modest ([300]Figure S6N), suggesting that TCF7 regulation might also be governed by other CD38-regulated metabolites. In addition to NAD^+ catabolism, CD38 promotes the production of cyclic ADP-ribose (cADPR), which was recently shown to regulate the expression of TCF7 via the ADPR/Ca^2+/RyR2/AKT pathway.[301]^36^,[302]^37 CD38 inhibition in chronically stimulated T cells resulted in decreased levels of the NAD^+ precursor nicotinamide (NAM)[303]^38 and ADPR[304]^39 ([305]Figures 4N and 4O; [306]Table S8). Further, we observed elevations in cytoplasmic Ca^2+ levels following chronic stimulation (CD38^hi) ([307]Figure S6O) that was reduced upon CD38 inhibition ([308]Figure S6P). These findings implicate CD38 in the regulation of TCF7 expression via modulation of both NAD^+ and cADPR levels. ADPR can be further metabolized to the immunosuppressive metabolite adenosine[309]^40 by the sequential action of CD203a and CD73 ([310]Figure 4I).[311]^41 While adenosine was not reliably detected in our analysis, related metabolites (e.g., AMP, ATP, ADP, S-adenosyl-L-methionine [SAM], and deoxyadenosine) were detectable and not significantly altered upon CD38i treatment ([312]Figures S7A and S7E). It is, therefore, possible that other ectoenzymes, such as CD73, CD203a, and CD39 expressed on chronically stimulated CAR-T cells ([313]Figures S7F and S7G), compensate for the inhibition of CD38 and contribute to the maintenance of these metabolites.[314]^41 Further, modulation of adenosine levels might depend on T cell extrinsic processes involving multiple cell types in the TME.[315]^9^,[316]^10^,[317]^41 LC-MS evaluation of conditioned media of control and CD38i-treated CAR-T cells failed to detect extracellular NAD(P)^+, and no significant changes in extracellular levels of ADPR, NAM, SAM, or deoxyadenosine were observed ([318]Figures S7H–S7K and [319]Table S9), consistent with prior reports that CD38 primarily governs intracellular NAD^+ levels.[320]^42 Using surface and intracellular immunofluorescence staining of CD38 and PD-1 in acute or chronically stimulated T cells, we showed that CD38 can be localized both in the cell membrane and in the cytoplasm of T cells ([321]Figures S7L–S7N), as previously shown.[322]^43^,[323]^44 CD38 cytoplasmic aggregates were more prevalent in chronically stimulated T cells and specifically in PD-1-positive T cells ([324]Figures S7L–S7N). Type I IFN induces CD38 expression in T cells GSEA also revealed a strong association between CD38 expression and type I/II IFN signaling ([325]Figures 4A, [326]S5A, and S5B; [327]Tables S5 and [328]S6). While important for anti-tumor immunity, recent reports have suggested a role for type I IFN signaling in the development of T cell dysfunction,[329]^45^,[330]^46^,[331]^47 and exposure of healthy CD8^+ T cells to type I IFN promoted mitochondrial and T cell dysfunction via upregulation of CD38 and enhanced NAD^+ consumption.[332]^48 Analysis of scRNA-seq of human melanoma CD8^+ TILs showed co-expression of CD38 and multiple type I IFN-stimulated genes (ISGs)[333]^49 in clusters 1–3 (exhausted T cells, see [334]Figure 1A) ([335]Figure 5A) with higher expression of numerous ISGs in CD8^+ T cells from ICB-NR compared to ICB-R patients ([336]Figure 5B). Type I IFN has well-established anti-tumor properties and can be secreted by tumor cells, macrophages, and dendritic cells in the TME.[337]^45^,[338]^50 Analysis of the expression of type I IFN signaling machinery (IFNAR1/2, STAT1/2, and IRF9)[339]^51 in CD45^+ cell clusters in human melanoma showed high expression of these genes in macrophages and dendritic cells (clusters 3 and 4, see [340]Figure S1A) and T cells (clusters 6, 7, and 11, see [341]Figure S1A) ([342]Figure S8A). More importantly, this signature was associated with resistance to ICB ([343]Figure S8B). Figure 5. [344]Figure 5 [345]Open in a new tab 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.[346]^8 (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 [347]Figure S8. To test the hypothesis that exposure of CD8^+ TILs to type I IFN in the TME can reinforce the expression of CD38 and drive T cell dysfunction, CD8^+ TILs expanded from melanoma patients were treated with IFN-β (type I IFN)[348]^50 with and without TCR activation. CD38 expression was highly induced following IFN-β treatment alone and was increased further when combined with TCR activation ([349]Figures 5C, 5D, and [350]S8C). Interestingly, low-dose IFN-β was sufficient to induce significant elevation of CD38-positive cells ([351]Figure S8D). Further, the effect was specific to type I IFN, as IFN-γ (type II) did not induce CD38 upregulation ([352]Figure S8E). Ruxolitinib, a JAK1/2 inhibitor,[353]^52 prevented upregulation of CD38 expression observed following stimulation of CD8^+ TILs with IFN-β ([354]Figure 5E), confirming a key role of IFN sensing in the T cell-specific upregulation of CD38. Consistent with the observations in TILs, IFN-β induced CD38 expression in a JAK1/2-sensitive manner in B7-H3.CAR-T cells, which was not observed following IFN-γ treatment ([355]Figures S8F and S8G). In both CD8^+ TILs and CAR-T cells, IFN-β-driven CD38 upregulation resulted in decreased cellular NAD^+ ([356]Figures 5F and [357]S8H) and was accompanied by increased surface expression of PD-1 and TIM-3 ([358]Figures 5G and [359]S8I), while CD38i treatment blunted IFN-β-driven upregulation of TIM-3, indicating a less exhausted T cell phenotype ([360]Figures 5H and [361]S8J). CD38 expression itself was not affected by CD38i treatment ([362]Figure S8K). Along with the loss of cellular NAD^+, we observed increased MMP and mitochondrial mass following IFN-β treatment with chronic TCR activation compared to chronic stimulation alone ([363]Figures 5I, 5J, [364]S8L, and S8M). These mitochondrial changes required longer IFN-β stimulation, together with TCR activation, to occur (over 6 days) and were not observed after short-term IFN-β stimulation (3 days) ([365]Figures S8N and S8O), as was shown in T cells from patients with systemic lupus erythematosus.[366]^48 These results indicate that both chronic TCR stimulation and type I IFN signaling can drive CD38 expression, resulting in increased NAD^+ consumption and an aberrant mitochondrial phenotype. CD38 blockade overcomes ICB resistance Given that upregulation of CD38 in CD8^+ T cells is associated with T cell dysfunction and ICB resistance and that disrupting CD38 is able to restore T cell effector function, we next examined CD38 blockade as a strategy to overcome ICB resistance. CD38 blockade was previously shown to enhance in vivo response to PD-1 blockade in murine tumor models.[367]^13^,[368]^37 Yet clinical trials of dual PD-(L)1 blockade plus CD38 blockade have failed to demonstrate meaningful clinical responses in NSCLC, prostate cancer, and multiple myeloma,[369]^53^,[370]^54^,[371]^55 for which CD38 targeting was largely directed at the cancer cells themselves. Importantly, the effect of dual PD-1/CD38 blockade in ICB-resistant melanoma, where CD38 expression in CD8^+ T cells correlates with lack of response to ICB ([372]Figures 1F and 1J), has not been tested. To examine the therapeutic potential of CD38 blockade in enhancing tumor immunity and overcoming resistance to ICB using a clinically relevant model of human melanoma, we performed ex vivo profiling of PDOTS.[373]^14^,[374]^15^,[375]^56 PDOTS, unlike traditional organoid models, are short-term 3D cultures of multicellular spheroids derived from freshly explanted patient tumors and retain tumor-infiltrating immune and stromal cells, including CD8^+ TILs.[376]^15 PDOTS derived from patients with advanced melanoma (n = 27) were treated with anti-PD-1 (pembrolizumab) ± anti-CD38 (daratumumab), compared to untreated controls ([377]Figure 6A and [378]Table S10). Following 5–7 days of treatment, PDOTS viability was analyzed using a microscopy-based viability assay.[379]^15^,[380]^57 We observed a significant reduction in PDOTS viability following a dual PD-1/CD38 blockade, which demonstrated superior anti-tumor activity compared to anti-PD-1 or anti-CD38 alone ([381]Figures 6B, 6C, and [382]S9; [383]Table S10). Examination of patient-specific responses to PD-1 ± CD38 blockade revealed an 18% response rate (5/27) to anti-CD38 treatment and a 55% response rate to dual PD-1/CD38 blockade (15/27), compared to a 7.4% response rate to single-agent PD-1 blockade (2/27) ([384]Figure 6C). Exceptional ex vivo response to dual PD-1/CD38 blockade was observed in several PDOTS from patients with clinical ICB resistance (60% of responders, 9 of 15), including PDOTS 10101 and 10213 derived from patients with ICB-resistant cutaneous and mucosal melanoma, respectively ([385]Figures 6D–6F and [386]S10A). Importantly, 53% of the PDOTS (8/15) that responded to dual PD-1/CD38 blockade did not respond ex vivo to PD-1 or CD38 blockade alone (e.g., PDOTS 10095, 10101, and 10190) ([387]Figures 6C and 6E). Dual PD-1/CD38 blockade was also active in ICB-resistant Merkel cell carcinoma[388]^58 and showed significant tumor killing with the combination ([389]Figure S10B). For select PDOTS specimens, we were able to show that dual PD-1/CD38 blockade was more effective than dual PD-1/CTLA-4 in reducing tumor growth ([390]Figure S10C). Multispectral flow-cytometry analysis (prior to PDOTS culture) and immunofluorescence staining of PDOTS “on-chip” in the 3D microfluidic culture confirmed the expression of CD38 and PD-1 on tumor-infiltrating immune cells ([391]Figures S10D and S10E). Post hoc multivariate analysis of multispectral flow-cytometry data, mutational status, and selected clinical features failed to identify any statistically significant association with PDOTS response ([392]Figures S11A–S11C). Figure 6. [393]Figure 6 [394]Open in a new tab 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 [395]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 [396]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 [397]Figures S9–S12; [398]Tables S10, [399]S11, [400]S12, and [401]S13. To identify CD8^+ T cell-intrinsic features associated with response or resistance to dual PD-1/CD38 blockade, scRNA-seq analysis was performed on CD8^+ T cells from matched tumor digests for selected PDOTS sensitive (n = 12) and resistant (n = 9) to dual PD-1/CD38 blockade ([402]Table S10). No differences were observed in the proportions of the four identified CD8^+ T cell clusters or in the expression of CD38 and TCF7 between PDOTS responsive or resistant to dual PD-1/CD38 blockade ([403]Figures 6G and [404]S11D–S11F; [405]Table S11). The proliferating T cell cluster (Prolif.CD8^+) and the CXCL13^+ T cell cluster exhibited the highest expression of CD38 ([406]Figures 6G and 6H), the latter of which has previously been shown to be enriched for tumor-reactive T cells.[407]^59^,[408]^60 Gene sets associated with effector T cell function,[409]^61 including TCR signaling, antigen presentation, T cell co-stimulation, and mitosis, were enriched in Prolif.CD8 and CXCL13^+ T cell clusters from tumor explants responsive to dual PD-1/CD38 blockade ([410]Figure 6I and [411]Table S12). Further, genes associated with cytotoxic potential (GZMA, IFNG),[412]^61 were up-regulated in Prolif.CD8 and CXCL13^+ T cells from tumor explants responsive to dual PD-1/CD38 blockade ([413]Figure S11G and [414]Table S13). Accordingly, the module score of these two genes in Prolif.CD8 and CXCL13^+ T cells could accurately discriminate responding from non-responding samples with an accuracy score of 85% (17 samples out of 20) ([415]Figures 6J, [416]S11H, and S11I; [417]Table S13). Interestingly, CD8^+ T cells from matched tumor digests from PDOTS resistant to dual PD-1/CD38 blockade exhibited higher expression of genes associated with Ca^2+ signaling and related pathways (RYR2, PLCG2, and AKT2)[418]^37^,[419]^62 and amphiregulin (AREG), which has been shown to promote the immunosuppressive effects of regulatory T cells[420]^63 ([421]Figure S11G and [422]Table S13). These results suggest that while specific CD8^+ T cell states and genes (e.g., CD38 and TCF7) do not clearly associate with sensitivity to dual PD-1/CD38 blockade, relative enrichment of CD38-expressing, tumor-reactive CD8^+ T cells with high expression of effector molecules is associated with response. To confirm the efficacy of dual PD-1/CD38 blockade in ICB-resistant murine tumor models, we performed ex vivo profiling of B16-ova and CT26-GFP murine-derived organotypic tumor spheroids (MDOTS).[423]^15 To model a more immunosuppressive microenvironment with reduced ICB sensitivity,[424]^64 tumors were harvested for MDOTS profiling on days 10–14 when the proportion of CD38^+CD8^+ T cells exceeded 50% ([425]Figures 2C and [426]S12A). MDOTS profiling revealed a modest response to single-agent PD-1 or CD38 blockade but showed significantly improved tumor control with combined PD-1/CD38 blockade in both models ([427]Figures 6K and 6L), supporting the effectiveness of CD38 blockade in overcoming ICB resistance. Lastly, using B16-ova MDOTS ± anti-CD8b treatment, we confirmed that CD8^+ T cell activity was required for the combinatorial effect of dual PD-1/CD38 blockade ([428]Figure S12B). Taken together, these findings demonstrate that CD38 blockade can overcome ICB resistance in human and murine tumor models. Restoring NAD^+ to overcome ICB resistance CD38 blockade with anti-human CD38 (e.g., daratumumab) or anti-mouse CD38 antibodies can partially inhibit CD38 enzymatic activity but also results in enzyme internalization.[429]^65 To determine whether pharmacological inhibition of CD38 enzymatic activity and restoration of cellular NAD^+ pools is sufficient to enhance sensitivity to PD-1 blockade, we performed MDOTS profiling of explanted B16-ova and CT26-GFP tumors treated with or without CD38i. CD38i exhibited modest single-agent activity but significantly increased the response to PD-1 blockade in both B16-ova and CT26-GFP MDOTS ([430]Figures 7A–7C). Consistent with this observation, NR supplementation[431]^36 enhanced the response to PD-1 blockade to a similar extent as disrupting CD38 in CT26-GFP MDOTS, while NR by itself had a modest effect on MDOTS viability more similar to CD38 blockade alone ([432]Figures 7D and [433]S12C). Further, NAD^+ depletion using FK866[434]^35 (NAMPTi) blunted the effect of dual PD-1/CD38 blockade ([435]Figure S12D), confirming a key role for the CD38-NAD^+ axis in mediating response to ICB. Further evaluation of CD38i ± PD-1 blockade in an additional cohort of melanoma PDOTS showed that similar to daratumumab treatment, inhibition of CD38 enzymatic activity is sufficient to sensitize ICB-resistant melanoma PDOTS to PD-1 blockade ([436]Figure 7E and [437]Table S14), an effect that could be further enhanced with NR supplementation ([438]Figure 7F and [439]Table S14). Taken together, these findings demonstrate the therapeutic potential of targeting CD38 in CD8^+ T cells in melanoma to overcome ICB resistance and the role of chronic TCR and type I IFN signaling in its upregulation while underscoring the importance of NAD^+ as a vital metabolite for maintaining and restoring T cell function ([440]Figure 7G). Figure 7. [441]Figure 7 [442]Open in a new tab 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 [443]Figure S12 and [444]Table S14. Discussion Here, we show that CD38^+CD8^+ T cells accumulate during tumor progression and, following ineffective ICB treatment, exhibit features of T cell exhaustion and correlate with ICB resistance. Disrupting CD38 restores cellular NAD^+ pools and TCF7 expression, thereby improving T cell function and sensitizing tumors to ICB, underscoring the importance of NAD^+ as a vital metabolite necessary for sustaining T cell-mediated tumor immunity. CD38-mediated ADPR generation was recently shown to contribute to T cell dysfunction via AKT-mediated repression of TCF7 expression.[445]^37 Interestingly, NAD^+-dependent signaling was also shown to regulate TCF7 via the NAD^+/SIRT1/FOXO1 pathway.[446]^66 Following CD38i treatment of chronically stimulated T cells, we observed increased NAD^+ levels with a concomitant reduction in ADPR. Thus, upregulation of CD38 can regulate T cell function by simultaneously depleting a driver of TCF7 expression (NAD^+) while promoting ADPR formation that represses TCF7 expression via increased intracellular Ca^2+ levels, RyR2 calcium channel activation, and downstream AKT activation.[447]^37 Interestingly, increased expression of genes associated with Ca^2+ signaling (e.g., PLCG2, RYR2, and AKT2) was observed in CD8^+ T cells from PDOTS matched tumor digests resistant to dual PD-1/CD38 blockade. Importantly, we showed that CD38 can be localized in small cytoplasmic aggregates that are enriched in chronically stimulated T cells, particularly those expressing PD-1. Those aggregates suggest an increased intracellular activity of CD38 in exhausted PD-1^+ T cells that can both decrease intracellular NAD^+ and enhance intracellular Ca^+2 signaling. Further studies are required to elucidate the intersecting metabolic pathways governed by CD38 and the effect of its cellular locations on T cell function. Chronic TCR stimulation and type I IFN[448]^50 promote upregulation of CD38, leading to mitochondrial dysregulation and T cell dysfunction. The role of type I IFN in CD38 upregulation in CD8^+ TILs and human CAR-T cells can provide another link between type I IFN signaling and T cell exhaustion and dysfunction[449]^48 as well as a better understanding of the dual role of type I IFN in cancer, providing tumor-suppressive effects as well as immune suppression and tumor progression in different contexts.[450]^47 Interestingly, recent clinical trials in patients with Hodgkin lymphoma and NSCLC have shown that JAK inhibitors can restore ICB sensitivity with an associated increased proportion of effector-memory T cells with a concomitant reduction in exhausted T cells.[451]^52^,[452]^67 Resistance to ICB in melanoma and other cancers remains a significant clinical challenge.[453]^3 The observed efficacy of dual PD-1/CD38 blockade, particularly in PDOTS from ICB refractory melanoma patients, supports further clinical development of CD38-directed therapeutic strategies to overcome ICB resistance. As CD38 monoclonal antibodies are approved by the Food and Drug Administration for other indications,[454]^33 there is a clear path to rapid translation of our observations to evaluate dual PD-1/CD38 blockade in patients with ICB-refractory advanced melanoma. Based on recent findings[455]^68 and our observations using B7-H3.CAR-T cells, targeting CD38/NAD^+ signaling may also be an appealing approach to prevent the development of CAR-T cell dysfunction. Thus, disrupting CD38 represents an emerging approach to enhance response to ICB and cellular cancer immunotherapy. Limitations of the study In this study, we tested the efficacy of dual PD-1/CD38 blockade using ex vivo profiling of melanoma PDOTS, many of which were derived from patients with clinical ICB resistance. Despite the conceptual advantages of using an established ex vivo immunocomponent human model, PDOTS are short-term (5–7 days) cultures that incompletely recapitulate every aspect of the tumor-immunity cycle, such as recruitment of peripheral immune cells from the lymph nodes and blood. As such, the biological consequences of PD-1 ± CD38 blockade in PDOTS reflect the impact of targeting these molecules within the model TME and within the time frame of the assay. A second limitation of the study is that we were not able to dissect the specific contributions of type II (extracellular-facing catalytic domain) and type III (intracellular-facing catalytic domain) CD38 using commercially available reagents. As disrupting CD38 exclusively influenced intracellular NAD^+ levels, this suggests that the catalytic domain of CD38 in exhausted T cells is in a type III orientation. Future studies using metabolic labeling, select disrupting of downstream metabolic pathways, and novel chemical probes are expected to provide new insights into the subcellular compartmentalization and downstream metabolic consequences of disrupting CD38 in exhausted T cells. Resource availability Lead contact Any information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Russell W. Jenkins (rjenkins@mgh.harvard.edu). Materials availability Human melanoma cell lines will be made available on request, but we may require a payment and/or a completed materials transfer agreement especially if there is potential for commercial application. There are restrictions to the availability of human melanoma cell lines because of the lack of an external centralized repository for their distribution and our need to maintain the stock. We are glad to share cell lines with reasonable compensation by the requestor for its processing and shipping. Data and code availability * • This paper analyzed publicly available scRNA-seq available at GEO: [456]GSE120575 , [457]GSE173351, [458]GSE225713, and [459]GSE217160. New scRNA-seq data have been deposited at GEO: [460]GSE296471 and [461]GSE296926 and are publicly available as of the date of publication. Metabolomics data have been deposited at NMDR: ST003779 and ST003830 and are publicly available as of the date of publication. * • All original code has been deposited at Zenodo at DOIs 15108338, 15120392, 15164848, 15107287, 15115199, and 15381446 and are publicly available as of the date of publication. * • Any additional information required to reanalyze the data reported in this paper is available from the [462]lead contact upon request. Acknowledgments