Weekly Digests
‹ Back to July

Meet the newest T cell checkpoint: CD38

July 23, 2025

Immune checkpoint blockade (ICB) can unleash immune responses against a variety of cancers, but its efficacy is often limited by T cell exhaustion. In addition to well established checkpoints like PD-1 and CTLA-4, T cell exhaustion has also been linked to CD38 and NAD+. In recent work published in Cell Reports Medicine, Revach et al. explored this axis and the potential of targeting it to limit T cell exhaustion and improve ICB.

To begin, Revach et al. used publicly available scRNAseq datasets from patients with advanced melanoma treated with ICB, and found that CD38 expression was increased in exhausted CD8+ TILs and decreased in active or effector memory T cells, which expressed higher levels of TCF7. Further, a higher proportion of CD8+ TILs expressing CD38 were identified in patients that were resistant to ICB (ICB-NR; both at baseline and to a greater extent post-treatment) than in those responsive to ICB (ICB-R). Further, high CD38 expression could be used to predict resistance to both anti-PD-1 and anti-PD-1 plus anti-CTLA-4. Similar results were noted in an independent cohort and in a cohort of patients with NSCLC.

Next, the researchers generated time-resolved scRNAseq data from murine B16 melanoma, tumor draining lymph nodes (dLNs), and normal lymph nodes (nLNs), and found that Cd38 expression was evident in CD8+ TILs by day 7 and increased over time with increasing tumor burden. Meanwhile, Tcf7 expression was observed largely in CD8+ T cells in dLNs and nLNs, and was lost in CD8+ TILs. In B16-OVA tumors treated with anti-PD-1, Cd38 expression was increased only in terminal exhausted/effector CD8+ TILs. Looking at peripheral blood from patients with melanoma before and after ICB treatment, the researchers identified higher levels of CD38+CD8+ T cells in ICB-NR patients mainly after treatment. CD38 was also increased in the plasma in both ICB-R and ICB-NR patients at 6 weeks, but remained elevated long-term only in ICB-NR patients.

In data from patients with melanoma, the researchers found that CD38 was generally expressed alongside multiple co-inhibitory receptors, and similar patterns were seen in TILs from B16-OVA. Similarly, comparing CD38hi and CD38lo B7-H3-directed CAR T cells, the researchers found that CD38-high cells had increased expression of exhaustion-related surface markers and decreased expression of TCF7.

To model the development of dysfunction in CAR T cells, the researchers showed that chronic TCR stimulation increased CD38 expression, reduced proliferation, increased exhaustion marker expression, and reduced lysis of target cancer cells compared to acute stimulation. Similar results were observed in patient-derived CD8+ TILs cultured with matched primary tumor cells. Using CRISPR-Cas9, Revach et al. showed that cells deficient in CD38 had a proliferative advantage following chronic TCR stimulation, with an increased proportion expressing TCF7 and showing cytotoxicity towards patient-derived melanoma cell lines.

Next, the researchers investigated how CD38 relates to exhaustion by using CD8+ TILs from ICB-treated melanoma patients and CD3+ TILs from B16-OVA tumors. In these cells, genes related to type I/II IFN signaling and OXPHOS were enriched, suggesting differences in mitochondrial fitness. Upon further investigation, the researchers found evidence that mitochondrial fitness was reduced in CD38hi versus CD38lo B7-H3 CAR T cells and CD8+ TILs from human melanoma, and that CD38 deletion improved mitochondrial fitness. As NAD+ boosting has also been shown to restore mitochondrial fitness, the researchers demonstrated that chronically TCR-stimulated B7-H3 CAR T cells demonstrated decreased levels of NAD+, and that deletion or inhibition of CD38 restored NAD+ pools, reduced expression of exhaustion markers, and increased expression of TCF7. Further, NAD+ depletion induced a more exhausted state, while NAD+ boosting increased T cell functionality without affecting CD38 expression. However, the effect of NAD+ on TCF7 expression was limited, so the researchers investigated other pathways, and found that CD38 also promoted expression of cADPR, which regulated expression of TCF7 via the ADPR/Ca2+/RyR2/AKT pathway.

To better understand the mechanism underlying the increase in CD38 expression in exhausted TILs, the researchers evaluated the association between CD38 and type I/II IFN signaling. Analysis of scRNAseq data of CD8+ TILs in human melanoma revealed co-expression of CD38 and several type I IFN-stimulated genes (ISGs) in exhausted T cells, with higher expression of ISGs in T cells from ICB-NR patients. Further, high expression of IFN signaling machinery in macrophages and DCs was associated with resistance to ICB, leading the researchers to hypothesize that IFNs in the TME could induce the expression of CD38 and drive T cell dysfunction. Testing this, the researchers found that CD8+ TILs expanded from melanoma patients upregulated CD38 expression when treated with IFN-β (type I IFN) – but not IFNγ (type II IFN) – and this upregulation was increased further when IFN-β was combined with TCR activation. Treatment with IFN-β also induced features of exhaustion similar to those in CD38hiCD8+ T cells. A JAK1/2 inhibitor prevented this upregulation of CD38, confirming the role of IFN sensing in T cell-specific upregulation of CD38.

To investigate the effects of CD38 on resistance to anti-PD-1, the researchers utilized melanoma PDOTS – short-term 3D cultures of multicellular spheroids derived from freshly explanted patient tumors, retaining tumor-infiltrating immune and stromal cells, including CD8+ TILs. Treatment of PDOTS with anti-PD-1 plus anti-CD38 was more effective than either monotherapy, with strong ex vivo responses (monitored by tumor cell viability) to the combination even in samples derived from patients that were clinically resistant to ICB. Proliferating CD8+ T cell and CXCL13+ T cell clusters showed the highest expression CD38, and in PDOTs that were responsive to dual-blockade, cells in these clusters showed increased expression of genes associated with effector functions and cytotoxic potential. Module scores of these gene sets could predict responding from non-responding PDOTS with 85% accuracy. Similar results were observed in MDOTS derived from murine B16-OVA or CT26-GFP, which were also shown to require CD8+ T cells for the antitumor efficacy of dual blockade. Finally, the researchers investigated whether CD38 inhibition (CD38i) or restoring NAD+ could overcome CD38-mediated exhaustion and found that indeed, in MDOTS, both strategies enhanced responses to anti-PD-1. Further, NAD+ depletion blunted the effects of anti-PD-1 plus anti-CD38, underscoring its role.

Overall, these results show that exposure to chronic TCR stimulation and increased IFN-I in tumors drives increased expression of CD38 in CD8+ T cells, which in turn, reduces mitochondrial fitness and depletes NAD+, driving exhaustion and contributing to resistance to ICB in melanoma and other cancers. Targeting CD38 or supplementing NAD+ could potentially overcome these effects and sensitize tumors to ICB.

Write-up and image by Lauren Hitchings

Meet the researcher

This week, first author Or-Yam Revach and lead author Russell W. Jenkins answered our questions.

First author Or-Yam Revach

What was the most surprising finding of this study for you?
CD38, an ecto-enzyme involved in NAD⁺ catabolism, is highly expressed in exhausted CD8⁺ T cells and has emerged as a promising target to enhance immune checkpoint blockade (ICB) by blunting T cell exhaustion. However, its efficacy in human cancer remained unclear. Using ex vivo profiling of a cohort of patient-derived organotypic tumor spheroids (PDOTS), we were amazed to see over 50% of tumors respond to CD38/PD1 blockade combination, compared to only 7.4% to PD-1 alone. Even more striking was that half of these responders were ICB resistant tumors clinically, suggesting that targeting CD38 could both enhance responses and overcome resistance.

What is the outlook?
In our study, we repurposed daratumumab, an FDA-approved anti-CD38 antibody for multiple myeloma, to restore ICB sensitivity by targeting T cell exhaustion in melanoma. The striking response observed in PDOTS to combined PD-1/CD38 blockade supports rapid clinical translation using existing antibodies. While prior trials of CD38/PD-(L)1 blockade in some solid tumors where CD38 targeting was directed at the cancer cells showed limited efficacy, they didn’t reveal major safety concerns. Thus, there is a clear path to rapid translation of the CD8-specific targeting of CD38 in combination with ICB in refractory melanoma.

What was the coolest thing you’ve learned (about) recently outside of work?
I’m a mother of two and a scientist. During my postdoctoral training, my kids reached the age where they became curious about my work and even asked why I haven’t solved the cancer problem yet! Their innocent, thoughtful questions have been surprisingly inspiring. They push me to see problems from fresh perspectives and often spark ideas or solutions I might not have considered otherwise.

References:

Revach OY, Cicerchia AM, Shorer O, Palin CA, Petrova B, Anderson S, Liu B, Park J, Chen L, Mehta A, Wright SJ, McNamee N, Tal-Mason A, Cattaneo G, Tiwari P, Xie H, Sweere JM, Cheng LC, Sigal N, Enrico E, Miljkovic M, Evans SA, Nguyen N, Whidden ME, Srinivasan R, Spitzer MH, Sun Y, Sharova T, Lawless AR, Michaud WA, Rasmussen MQ, Fang J, Brook JR, Chen F, Wang X, Ferrone CR, Lawrence DP, Sullivan RJ, Liu D, Sachdeva UM, Sen DR, Flaherty KT, Manguso RT, Bod L, Kellis M, Boland GM, Yizhak K, Yang J, Kanarek N, Sade-Feldman M, Hacohen N, Jenkins RW. Overcoming resistance to immunotherapy by targeting CD38 in human tumor explants. Cell Rep Med. 2025 Jul 15.

In the Spotlight...

A bispecific antibody-drug conjugate targeting EGFR and HER3 in metastatic esophageal squamous cell carcinoma: a phase 1b trial

In a phase 1b trial, 82 patients with esophageal squamous cell carcinoma were treated with an anti-EGFR/HER3 bispecific antibody linked to a topoisomerase I inhibitor, at 2.0 or 2.5 mg/kg q3wks. Treatment response and drug Cmax were dose-related; overall response and disease control rates were 39.6/79.2% at 2.5mg/kg and 15/50% at 2 mg/kg. Although all patients experienced AEs, more strongly so at the higher dose, treatment was generally safe, and the 2.5mg/kg dose was selected for a Phase 2 trial. Pretreatment EGFR/HER3 expression did not correlate with treatment response (however most patients were positive).

Contributed by Alex Najibi

CD4+ T cells license Kupffer cells to reverse CD8+ T cell dysfunction induced by hepatocellular priming

Venzin and Beccaria et al. demonstrate that pre-activated CD4+ T cells actively license local hepatic APCs and restore antiviral CD8+ T cell function. HBV-specific CD4+ T cell receptor transgenic mice show that effector CD4+ T cells reprogram Kupffer cells (KCs), but not DCs, to rescue dysfunctional CD8+ T cells in a tolerogenic hepatic environment. CD4+ T cells activated KCs via CD40L-CD40 interactions prompting IL-12 and IL-27 production. IL-12 amplified the helper CD4+ T cells and IL-27 reinvigorated dysfunctional CD8+ T cells. Exogenous IL-27 reinstated HBV-specific CD8+ T cell effector function in T cells from chronically infected patients.

Contributed by Shishir Pant

Multimodal delineation of a layer of effector function among exhausted CD8 T cells in tumors

Ray et al. discovered that Cd69 mRNA levels could be used to differentiate between potent and dysfunctional activation states among intratumoral CD69+CD8+ T cells. CD8 T cells with high Cd69 mRNA and CD69 protein showed increased levels of AP-1 transcription factors, produced potent effector molecules, displayed reduced features of exhaustion, showed enhanced antigen engagement, and superior tumor cell killing in vivo compared to Cd69RNAlowCD69+ cells. Simultaneous Cd69 mRNA and protein expression analysis identified Cd69RNAhighCD69+CD8+ T cells associated with antitumor effector function across human cancers.

Contributed by Shishir Pant

IGM-7354, an immunocytokine with IL-15 fused to an anti-PD-L1 IgM, induces NK and CD8+ T cell-mediated cytotoxicity of PD-L1 positive tumor cells

Desbois et al. generated IGM-7354, an IgM comprising ten PD-L1 binding sites which blocked PD-L1/PD-1 binding and a single IL15/IL15Rα complex which activated IL15Rβγ+ human NK and CD8+ T cells and rescued TEX cells in vitro. IGM-7354 delivered i.t. expanded human CD8+ T cells and reduced tumor growth in a humanized mouse breast cancer model, and boosted daratumumab (anti-CD38)-mediated ADCC in a multiple myeloma xenograft model and anti-CD19 CAR-T cell efficacy in a systemic lymphoma model even after tumor rechallenge. In monkeys, IgM-7354 increased NK and CD8+ T, particularly TEM cell proliferation, in blood, LN and BM.

Contributed by Paula Hochman

Enhanced antitumor immunity of VNP20009-CCL2-CXCL9 via the cGAS/STING axis in osteosarcoma lung metastasis

Focused on improving bacteria-mediated cancer immunotherapy responses in osteosarcoma (OS) with liver metastases, Liu, Liu, and Yang et al. engineered a novel Salmonella strain to express CCL2 and CCL9 (VNP-C-C). VNP-C-C accumulated within tumors and induced immunogenic cell death via the cGAS/STING pathway (leading to IFNα secretion), even in the absence of T cells. In immunocompetent mice, CCL2 and CCL9 further amplified anti-tumor responses by recruiting DCs, polarized M1-like macrophages and T cells into the TME, resulting in enhanced efficacy and survival in an immunocompetent OS lung metastasis model.

Contributed by Katherine Turner

CTG-initiated cryptic peptide translation up and downstream of a canonical ATG start codon is enhanced by TLR stimulation and induces tumor regression in mice

Song et al used stop codon-separated tandem constructs encoding a control peptide via canonical ATG and SIINFEKL via noncanonical CTG to examine noncanonical translation. TLR stimulation selectively enhanced translation of CTG-SIINFEKL (and genome wide noncanonical-ORF) in BMDCs, and stronger OTI T cell priming was observed when CTG peptide was upstream of a canonical ORF. OTI T cells efficiently killed cancer cells expressing very low levels of CTG-SIINFEKL, where MHCI expression was the limiting factor. In vivo, tumors expressing the construct could be controlled by OTI T cells, but efficacy varied by context.

Contributed by Morgan Janes

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.