CD38 is a multifunctional type II transmembrane glycoprotein that serves as an ectoenzyme, catalyzing the synthesis and hydrolysis of cyclic ADP-ribose (cADPR) from NAD⁺ and nicotinic acid adenine dinucleotide phosphate (NAADP) from NADP⁺, which act as second messengers to mobilize intracellular calcium stores.[1][2] Expressed predominantly on hematopoietic cells such as plasma cells, activated T and B lymphocytes, and natural killer cells, CD38 functions as a receptor in cell adhesion and signal transduction, particularly through interactions with CD31, and is recognized as a marker of immune cell activation.[3][2]Structurally, CD38 features a short N-terminal cytoplasmic tail, a single transmembrane helix, and an extensive C-terminal extracellular domain rich in cysteine residues that form stabilizing disulfide bonds, enabling the protein to dimerize.[1] Its enzymatic active site, centered around a conserved glutamate residue (Glu226), supports bidirectional catalysis: at neutral or alkaline pH, it generates cADPR to trigger calcium release from the endoplasmic reticulum, while at acidic pH, it produces NAADP to mobilize calcium from lysosomes, influencing diverse physiological processes including insulin secretion from pancreatic β-cells, neutrophilchemotaxis, and oocyteactivation during fertilization.[1][2]In pathological contexts, CD38 is overexpressed in various hematologic malignancies, notably multiple myeloma where it serves as both a prognostic biomarker and a therapeutic target; as of 2025, anti-CD38 monoclonal antibodies are integrated into standard quadruplet regimens for newly diagnosed multiple myeloma, in addition to their established use in relapsed/refractory cases.[4] Monoclonal antibodies like daratumumab bind CD38 to induce tumor cell death via antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, and phagocytosis, achieving significant response rates in relapsed/refractory cases.[2][5][6] Beyond cancer, CD38 modulates inflammation by regulating cytokine production (e.g., IL-6, TNF-α) and immune cell recruitment in conditions such as rheumatoid arthritis and inflammatory bowel disease, while CD38 deficiency accelerates autoimmunity in mouse models of type 1 diabetes but suppresses disease progression in some models of systemic lupus erythematosus.[3] High expression is also noted in non-hematopoietic tissues like the pancreas, brain, and placenta, linking CD38 to broader roles in metabolism and neurodegeneration.[2]
Molecular Structure and Genetics
Gene Organization and Protein Structure
The CD38 gene is located on the short arm of human chromosome 4 at position 4p15.32, spanning approximately 71 kb of genomic DNA and consisting of 8 exons that encode a 300-amino acid precursor protein.[7] The gene structure includes a CpG island in the 5'-flanking promoter region, which spans about 900 bp and incorporates exon 1, facilitating transcriptional initiation.[7] The CD38 gene is highly polymorphic, with notable single nucleotide polymorphisms (SNPs) such as rs6449182 located in intron 1. This C>G variant affects gene expression levels, with the G allele associated with reduced promoter activity and increased susceptibility to B-cell chronic lymphocytic leukemia and multiple myeloma.[8][9]The mature CD38 protein is a type II transmembrane glycoprotein with a molecular weight of approximately 45 kDa due to N-linked glycosylation at four sites in the extracellular domain.[10] It features a short N-terminal cytoplasmic tail of 21 amino acids (residues 1-21), a single hydrophobic transmembrane domain of 21 amino acids (residues 22-42), and a large C-terminal extracellular domain of 256 amino acids (residues 43-300), which positions the enzymatic and receptor functions on the cell surface.[10][7] This topology enables CD38 to act as a bifunctional ectoenzyme and signaling receptor, with the extracellular domain oriented outward to interact with extracellular substrates and ligands.[3]Key structural motifs in the extracellular domain include the active site pocket, formed by conserved residues such as Glu-226, Trp-125, Trp-189, Asp-155, Ser-193, Glu-146, Arg-127, and others, which facilitate NAD+ hydrolysis to produce cyclic ADP-ribose (cADPR) and ADP-ribose (ADPR).[11] Additionally, the extracellular region contains binding sites that mediate interactions with non-substrate ligands, including hyaluronic acid in the extracellular matrix and CD31 (PECAM-1) on adjacent cells, supporting cell adhesion and migration functions.[12][13]CD38 exhibits strong evolutionary conservation across mammalian species, with high sequence identity in the catalytic domain, reflecting its essential physiological roles. In non-mammalian organisms, CD38 homologs show structural and functional homology to soluble ADP-ribosyl cyclases, such as the enzyme from Aplysia californica, which shares a similar dinucleotide-binding fold but lacks the transmembrane topology.[14][15]The three-dimensional structure of the human CD38 extracellular domain has been elucidated through X-ray crystallography, with the first high-resolution model (PDB entry 1YH3) determined at 1.9 Å resolution, revealing a compact α/β fold dominated by a central β-sheet flanked by α-helices.[16][17] This structure highlights the dinucleotide-binding fold, characterized by a deep pocket that accommodates NAD+ substrates and supports both cyclase and hydrolase activities, with conserved motifs aligning closely to those in related enzymes like CD157 and Aplysia ADP-ribosyl cyclase. Subsequent structures, such as those complexed with inhibitors or analogs (e.g., PDB 3DZK), further confirm the flexibility of the active site loop, which undergoes conformational changes during catalysis.[18]
Expression Regulation
The expression of the CD38 gene is primarily regulated at the transcriptional level through interactions between its promoter region and various transcription factors activated by cytokines and other signaling molecules. The human CD38 promoter contains binding sites for specificity protein 1 (Sp1), retinoic acid response elements (RARE), and interferon regulatory factor 1 (IRF-1), which facilitate induction by all-trans retinoic acid (ATRA) and interferons. For instance, ATRA stimulates CD38 transcription via RARE located in the first intron, leading to increased mRNA levels in cells such as airway smooth muscle and leukemic B cells. Similarly, interferons (types I and II, including IFN-γ and IFN-α) rapidly upregulate CD38 expression in B cells through IRF-1 binding to the promoter, enhancing ectoenzymatic activity. Although direct evidence for NF-κB and STAT1/3 binding sites in the CD38 promoter is limited, cytokine-induced pathways involving these factors contribute to inducible expression in activated immune cells.Epigenetic modifications play a key role in fine-tuning CD38 expression, particularly in immune cells, by altering chromatin accessibility and promoter activity. The CD38 promoter includes a CpG island approximately 900 bp long encompassing exon 1, where DNA methylation patterns can silence gene expression in quiescent states. Histone acetylation, mediated by histone acetyltransferases like p300 recruited by PTIP (PAX-interacting protein 1), promotes CD38 activation by relaxing chromatin structure at cis-regulatory elements in hematopoietic cells. In murine memory B cells, histone modifications control hallmark genes like CD38, maintaining low expression in resting states but allowing rapid induction upon stimulation.Post-transcriptional regulation of CD38 occurs via microRNAs (miRNAs) that target its mRNA, modulating protein levels during cellular differentiation. For example, miR-26a directly suppresses CD38 translation in multiple myeloma cells, acting as a tumor suppressor by reducing surface expression and enhancing anti-tumor responses. In plasma celldifferentiation, networks of miRNAs, including those identified in human B-cell stimulation models, regulate CD38 alongside other differentiation markers, preventing excessive expression that could impair maturation.CD38 exhibits developmental and inducible expression patterns tightly linked to immune cell activation and lineage commitment. During B-cell differentiation, CD38 is upregulated from early precursors (pre-pro-B to immature stages) in bone marrow, serving as a marker of progression. In T cells, activation induces CD38 surface expression, correlating with glycolytic shifts and effector functions in both human and murine models. These patterns are inducible by inflammatory cytokines, reflecting adaptive responses in immune homeostasis.Species-specific differences in CD38 promoter elements contribute to variations in expression profiles across mammals. The human promoter's CpG-rich structure and RARE sites differ from murine counterparts, leading to distinct regulation; for instance, human CD38 shows broader inducibility by ATRA in hematopoietic cells compared to mice, where expression is more restricted to certain lineages. Phylogenetic analyses of primate CD38 sequences highlight epitope variations that may influence promoter accessibility, underscoring evolutionary adaptations in immune regulation.
Biological Functions
Enzymatic Activities
CD38 exhibits multifaceted enzymatic activities as a multifunctional ectoenzyme primarily involved in NAD⁺ metabolism. Its core function is NAD⁺ glycohydrolase activity, which hydrolyzes extracellular NAD⁺ into nicotinamide (NAM) and ADP-ribose (ADPR), with a minor fraction cyclized to cyclic ADP-ribose (cADPR).[19] This hydrolysis predominates, accounting for over 90% of CD38's catalytic output, and occurs with a Michaelis constant (Kₘ) for NAD⁺ in the range of 15–56 μM.[20] Additionally, CD38 displays ADP-ribosyl cyclase activity, converting NAD⁺ to cADPR, a potent second messenger that mobilizes intracellular calcium stores via ryanodine receptors.[21] The cyclase activity shares a similar Kₘ for NAD⁺ (approximately 15–56 μM) and exhibits optimal efficiency at neutral pH (6–8).[20]CD38 also catalyzes base-exchange reactions, particularly the exchange of the nicotinamide moiety of NADP⁺ with nicotinic acid to produce nicotinic acid-adenine dinucleotide phosphate (NAADP), another calcium-mobilizing second messenger.[21] This reaction is favored under acidic conditions (pH 4–5).[20] Kinetic parameters vary slightly across activities, with the glycohydrolase showing higher turnover rates than the cyclase, reflecting the enzyme's preference for hydrolysis.[22]These activities differ in localization: the predominant type II transmembrane orientation positions the catalytic domain extracellularly, enabling hydrolysis of NAD⁺ in the cellular microenvironment and contributing to local NAD⁺ depletion.[20] In contrast, type III isoforms localize intracellularly (e.g., in the nucleus or mitochondria), where they may modulate NAD⁺ levels within organelles.[20] The catalytic mechanisms are underpinned by the enzyme's crystal structure, resolved at 1.9 Å resolution, which reveals a bilobal active site pocket facilitating substrate binding and catalysis.[23] Key residues include the catalytic glutamate (Glu²²⁶), which nucleophilically attacks the NAD⁺ anomeric carbon to form a covalent intermediate, and tryptophan residues (Trp¹²⁵, Trp¹⁸⁹) that position the substrate via hydrophobic interactions; glutamate (Glu¹⁴⁶) and aspartate (Asp¹⁵⁵) residues modulate pH-dependent partitioning between hydrolysis and cyclization pathways.[23]
Receptor and Signaling Roles
CD38 functions as a cell surface receptor that mediates heterotypic adhesion between leukocytes and endothelial cells primarily through its interaction with CD31 (also known as PECAM-1), a member of the immunoglobulin superfamily expressed on vascular endothelium. This CD38-CD31 binding promotes transendothelial migration of leukocytes, facilitating their extravasation into tissues during inflammatory responses and supporting immune cell homing to lymphoid organs. Additionally, CD38 binds hyaluronic acid, a glycosaminoglycan component of the extracellular matrix, which further regulates leukocyte adhesion and motility by anchoring cells within the stromal environment.Beyond adhesion, CD38 transduces signals upon ligand engagement or antibody crosslinking, often through lateral associations with tetraspanins such as CD9 and CD81, which organize signaling complexes in lipid rafts. These interactions lead to tyrosine phosphorylation of associated proteins, including linker for activation of T cells (LAT) and CD3 chain components, initiating downstream cascades.[24] A key outcome is calcium mobilization, involving influx from extracellular sources and release from intracellular stores, which is essential for cytoskeletal rearrangements and cellular activation.[3]In B cells, CD38 acts as a costimulatory receptor that enhances antigen receptor signaling by associating with the CD19/CD81 complex, promoting proliferation and differentiation upon ligation.[25] Similarly, in T cells, CD38 costimulates activation through its dependence on the T cell receptor (TCR)/CD3 complex, amplifying responses to antigenic stimulation and supporting cytokine production.[26] These roles underscore CD38's contribution to adaptive immune responses by integrating adhesion and signaling to fine-tune lymphocyte function.Downstream of receptor engagement, CD38 triggers intracellular signaling cascades, including activation of phospholipase C (PLC)-γ, which generates inositol 1,4,5-trisphosphate (IP3) to mediate calcium release from the endoplasmic reticulum.[27] This calcium signaling intersects with the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway via Raf-1 activation, promoting gene transcription and cellular proliferation.[24] Enzymatic products like cyclic ADP-ribose (cADPR), generated by CD38, further contribute to calcium release through ryanodine receptors.[3]The bifunctional nature of CD38 links its receptor functions to enzymatic amplification, where ligand-induced clustering enhances ADP-ribosyl cyclase activity, thereby potentiating intracellular signaling loops.[28]
Expression Patterns
Tissue and Cellular Distribution
CD38 exhibits prominent expression on hematopoietic cells, serving as a key marker for various immune cell types. In humans, it is highly expressed on plasma cells, with nearly 100% of these cells displaying surface CD38 at high levels (approximately 10^5 molecules per cell), as well as on activated T and B lymphocytes, natural killer (NK) cells, monocytes, macrophages, dendritic cells, and neutrophils, particularly within bone marrow and lymphoid tissues.[3][29][30] This pattern underscores CD38's role as an activation-associated antigen in the immune system.In non-hematopoietic tissues, CD38 shows low to moderate expression. Notable sites include pancreatic islet cells, where it is detected on beta cells; neuronal cells such as astrocytes and oligodendrocytes in the brain; vascular endothelium and smooth muscle cells; as well as prostatic epithelial cells, retinal cells, kidney, and gut tissues.[3][31][32] Protein expression is often cytoplasmic and membranous, with enhanced levels in lymphoid tissues compared to other organs, as summarized in human tissue atlases.[33]Developmentally, CD38 is absent or low on early lymphocyte precursors but is induced during maturation and activation processes. For instance, it appears on human B-cell precursors and germinal center B cells, persisting at high levels on plasma cells, while mature peripheral B cells show reduced surface expression unless reactivated. Similarly, in T cells, CD38 marks thymocyte precursors and double-positive CD4+CD8+ cells, with upregulation on mature T cells following activation.[3]Expression patterns vary across species, with broader distribution in rodents compared to humans. In mice, CD38 is retained on mature B cells throughout differentiation and on resting T cells, whereas human mature B cells lose surface expression post-maturation, and resting T cells lack it until activation; murine plasma cells show lower levels than their human counterparts.[3][34]Detection of CD38 expression typically employs flow cytometry for surface analysis on live cells or immunohistochemistry for tissue sections, utilizing monoclonal antibodies such as HB-7, which specifically binds the extracellular domain.[3][35]
Pathological Upregulation
CD38 is pathologically upregulated in various hematological malignancies, particularly multiple myeloma (MM), where it is highly and uniformly expressed on nearly all malignant plasma cells, often exceeding 90% positivity in primary samples and cell lines.[36][37] This overexpression contrasts with lower baseline levels in normal lymphoid and myeloid cells and is influenced by inflammatory cytokines such as IL-6, IFN-γ, TNF-α, and CXCL-16 secreted within the tumor microenvironment, which induce CD38 transcription via pathways like STAT3 activation.[38] In MM, such upregulation supports tumor survival and immune evasion, though certain genetic alterations like 1q gain/amplification can lead to paradoxically reduced CD38 levels through enhanced IL-6 receptor signaling and JAK-STAT3-mediated suppression.[39]In solid tumors, CD38 expression is elevated in cervical cancer, where it correlates with disease progression and is linked to activation of the PI3K-AKT pathway, promoting cellenergymetabolism and proliferation.[40][41] In ovarian cancer, high CD38 levels enhance immune infiltration and are associated with favorable prognosis.[42][43]Pathological upregulation of CD38 also occurs in inflammatory conditions, including rheumatoid arthritis (RA), where it is significantly increased in synovial tissues and fibroblasts compared to healthy controls, contributing to chronic inflammation through enhanced T cell activation and cytokine production.[44][3] In neuroinflammatory models of Alzheimer's disease, amyloid-β promotes CD38 expression in senescent microglia, exacerbating neuroinflammation and neuronal damage via NAD+ depletion and calcium dysregulation.[45][46]Conversely, CD38 expression is downregulated in certain immunodeficiencies, such as subsets of common variable immunodeficiency (CVID), where diminished CD38 on B cells impairs plasma cell differentiation and antibody production, leading to hypogammaglobulinemia.[47] This contrasts with its upregulation in chronic T cell exhaustion states, though low CD38 on HIV-specific CD8+ T cells in elite controllers suggests a protective role against viral persistence in some immunodeficiency contexts.[48]Within the tumor microenvironment, CD38 is upregulated on stromal fibroblasts and tumor-associated macrophages, fostering an immunosuppressive niche that promotes tumor invasion, angiogenesis, and immune evasion through adenosine production and altered cytokine signaling.[49][50] In cancer-associated fibroblasts, CD38 enhances pro-tumoral activity by enabling secretion of growth factors, while on tumor-associated macrophages, it modulates polarization toward an M2-like immunosuppressive phenotype.[51][52] Recent studies as of 2024 also highlight upregulated CD38 on plasma cells and immune infiltrates in kidney diseases such as lupus nephritis and antibody-mediated rejection.[53]
Physiological and Pathological Roles
Immune Modulation
CD38 serves as a costimulatory molecule on T cells, enhancing their proliferation and cytokine production through ligation with CD31 on antigen-presenting cells or endothelial cells. This interaction triggers calcium mobilization via cyclic ADP-ribose (cADPR) production, amplifying T-cell receptor signaling and leading to increased secretion of pro-inflammatory cytokines such as IFN-γ and IL-12. In experimental models, blockade of CD38-CD31 engagement reduces antigen-induced T-cell activation and cytokine release, underscoring its role in promoting effector T-cell responses during immune activation.[54][55]In B-cell development, CD38 expression is dynamically regulated, serving as a key marker of maturation stages. It is highly expressed on immature pro-B and pre-B cells, where ligation induces apoptosis to maintain B-cell homeostasis, but diminishes on naive mature B cells. Upon activation in germinal centers, CD38 is reinduced on centroblasts and centrocytes, facilitating somatic hypermutation and isotype switching through calcium-dependent signaling. During terminal differentiation, sustained high CD38 expression (CD38hi) identifies plasmablasts and plasma cells, correlating with their antibody-secreting capacity; for instance, in vitro differentiation of switched-memory B cells yields CD38hiCD138+ plasma cells that produce elevated IgG and IgA levels.[56][57]CD38 modulates neutrophil function by regulating chemotaxis and degranulation through its enzymatic generation of cADPR and ADP-ribose, which mobilize intracellular calcium stores. In CD38-deficient mice, neutrophils exhibit defective migration toward chemoattractants like fMLP, resulting in delayed recruitment to infection sites such as the lungs during bacterial challenges. This calcium signaling also supports actin polymerization and granule release, enhancing degranulation and antimicrobial responses; human neutrophils with upregulated CD38 show improved chemotactic accuracy and effector functions in inflammatory contexts.[58][54]In the tumor microenvironment, CD38 expression on regulatory T cells (Tregs) drives metabolic shifts that enhance their stability and suppressive capacity, thereby inhibiting anti-tumor immunity. CD38-mediated NAD+ depletion redirects pyruvate metabolism toward gluconeogenesis, elevating phosphoenolpyruvate while limiting α-ketoglutarate accumulation in the TCA cycle; this maintains hypomethylation at the Foxp3 locus, preserving Treg identity and function. Inhibition of CD38 in tumor-bearing models reduces intratumoral Treg frequencies, improves CD8+ T effector-to-Treg ratios, and slows tumor progression, highlighting its role in immune evasion.[59]Elevated CD38 expression on immune cells contributes to hyperactivation in autoimmune diseases such as systemic lupus erythematosus (SLE) and multiple sclerosis (MS). In SLE, increased CD38 on T cells, B cells, and monocytes correlates with disease activity, promoting calcium flux that exacerbates cytokine production and autoantibody generation; CD38 deficiency in murine models attenuates SLE pathology by reducing type I IFN and autoantibodies. Similarly, in MS and its experimental model (EAE), upregulated CD38 on T cells and glia drives neuroinflammation and demyelination through NAD+ dysregulation and enhanced T-cell priming, with CD38 knockout mice showing milder disease severity and impaired autoreactive responses.[54][60][61]
Metabolic Regulation and Aging
CD38 plays a central role in metabolic regulation by acting as a major NAD+ glycohydrolase, hydrolyzing NAD+ into nicotinamide and other products, which depletes cellular NAD+ pools and impairs sirtuin activity. Sirtuins, NAD+-dependent deacetylases, are essential for maintaining mitochondrial biogenesis, oxidative metabolism, and energy homeostasis; reduced NAD+ availability due to CD38 activity leads to diminished sirtuin function, resulting in mitochondrial dysfunction characterized by decreased respiratory capacity and increased reactive oxygen species production.[62][63] Additionally, CD38 catalyzes the synthesis of cyclic ADP-ribose (cADPR) from NAD+, which acts as a second messenger to mobilize intracellular calcium stores, thereby influencing calcium-dependent signaling pathways that regulate metabolic gene expression, including those involved in glucose uptake and lipid oxidation.[62][63]With advancing age, CD38 expression is upregulated in various tissues, notably the brain and heart, contributing to progressive NAD+ depletion. In aged mice, this upregulation correlates with substantial NAD+ reductions, such as approximately 50% drops in brain and cardiac tissues compared to younger counterparts, exacerbating metabolic decline and cellular senescence.[64][65] This age-related CD38 increase disrupts NAD+ homeostasis, linking it to several degenerative processes.CD38-mediated NAD+ exhaustion has been implicated in age-related diseases through metabolic dysregulation. In neurodegeneration, such as Alzheimer's disease, elevated CD38 promotes neuroinflammation and amyloid-beta accumulation by impairing NAD+-dependent neuroprotection, while in cardiovascular aging, it contributes to endothelial dysfunction and vascular stiffness via mitochondrial impairment in cardiac tissues.[66][65] In diabetes, CD38 upregulation in pancreatic beta cells leads to exhaustion and reduced insulin secretion due to depleted NAD+ and altered calcium signaling, accelerating beta-cell failure.[64][63]Recent studies from 2025 demonstrate that pharmacological inhibition of CD38 effectively restores NAD+ levels in aging models, mitigating metabolic deficits and improving physiological outcomes. For instance, CD38 inhibitors have been shown to enhance cognitive function in tauopathy mouse models by reducing neuroinflammation and preserving neuronal NAD+ homeostasis, while also bolstering cardiac performance through improved mitochondrial function and exercise capacity in aged rodents.[62][63][66] These findings underscore CD38's potential as a target for countering age-associated metabolic decline.
Clinical Relevance
Disease Associations
CD38 has been implicated as a prognostic marker in hematological malignancies, particularly multiple myeloma (MM), where elevated levels of circulating CD38-positive clonal plasma cells at diagnosis correlate with poorer treatment response and overall survival.[67] In extramedullary MM, lower CD38 expression on tumor cells is associated with worse overall survival compared to higher expression, highlighting CD38's role in disease aggressiveness.[68] These associations underscore CD38's utility in risk stratification for MM patients, independent of therapeutic targeting.In solid tumors, CD38 overexpression contributes to disease progression and metastasis across several cancer types. In epithelial ovarian cancer, cancer-cell-derived CD38 promotes tumor growth and metastatic spread both in vitro and in vivo by modulating the tumor microenvironment.[69] Similarly, in prostate cancer, CD38 expression in tumor epithelium is linked to poorer overall survival and increased risk of recurrence, with epigenetic regulation via methylation influencing its levels.[70][71] In breast cancer, CD38 expression on tumor cells is necessary for metastatic potential, as demonstrated in experimental models where its knockdown reduces primary tumor growth and dissemination.[72]CD38 upregulation in pancreatic islets is associated with type 2 diabetes mellitus, where it contributes to impaired insulin secretion through NAD+ depletion and disrupted calcium signaling. A missense mutation in the CD38 gene has been identified as a factor in insulin secretion defects in non-insulin-dependent diabetes mellitus, linking genetic variants to beta-cell dysfunction.[73] Elevated CD38 activity in diabetic islets exacerbates beta-cell stress, reducing glucose-stimulated insulin release and promoting disease progression.[74]In neurological disorders, CD38 plays a role in HIV-associated neurocognitive decline through its regulation of astrocyte calcium signaling and neuroinflammation, with increased expression correlating to dementia severity in HIV-1 infection.[75] For Alzheimer's disease, CD38 deficiency attenuates amyloid-beta pathology and cognitive deficits in mouse models, primarily via reduced microglial activation and pro-inflammatory responses in the brain.[76][77] CD38's dual influence on microglial function—promoting activation while also inducing cell death—links it to exacerbated neurodegeneration in these conditions.[78]Regarding cardiovascular diseases, CD38 contributes to atherosclerosis by driving cholesterol-induced macrophagesenescence via NAD+ depletion, which promotes foam cell accumulation and plaque instability.[79] Recent studies highlight CD38's involvement in the heart-brain axis, where its overexpression links cardiac dysfunction to neurological outcomes, including in ischemic conditions as of 2025.[80]In infectious diseases, particularly HIV, CD38 facilitates viral persistence and chronic inflammation by sustaining T-cell exhaustion and immune activation during infection.[81] Elevated CD38 expression on CD8+ T cells correlates with disease progression and higher levels of pro-inflammatory markers, contributing to ongoing inflammation even in treated chronic viral infections.[82][83]
Diagnostic Biomarkers
CD38 serves as a valuable diagnostic biomarker in various hematological malignancies and inflammatory conditions due to its overexpression on malignant cells and involvement in immune activation. In multiple myeloma, flow cytometry is widely employed to detect CD38-positive plasma cells for assessing minimal residual disease (MRD), where CD38 is combined with markers like CD138 and CD45 to achieve high sensitivity in bone marrow samples, enabling detection limits as low as 10^{-5} to 10^{-6}.[84] This approach has been standardized in consensus guidelines, correlating MRD negativity with improved progression-free survival in patients post-therapy.[84] Innovations such as camelid-derived anti-CD38 nanobodies enhance detection specificity, particularly in cases where standard antibodies face epitope masking after prior treatments.[85]Immunohistochemistry (IHC) scoring of CD38 expression in tumor biopsies provides prognostic insights and predicts therapeutic responses, particularly in solid tumors like hepatocellular carcinoma (HCC). High CD38 positivity in the tumor microenvironment, quantified via IHC, is associated with better outcomes in patients receiving anti-PD-1/PD-L1 immunotherapy, with median progression-free survival exceeding 10 months in responders compared to under 5 months in non-responders.[86] This scoring method evaluates both tumor cells and infiltrating immune cells, offering a non-invasive means to stratify patients for immune checkpoint blockade.[86]Soluble CD38 (sCD38) in serum acts as a non-invasive biomarker for monitoring inflammation and cancer progression, especially in multiple myeloma, where elevated plasma levels correlate with tumor burden and disease activity.[87] Assays like nanobody-based detection quantify sCD38, reflecting ectodomain shedding from malignant plasma cells, and levels above 50 ng/mL indicate active proliferation and poorer prognosis.[87] In inflammatory contexts, such as chronic immune activation, sCD38 contributes to systemic signaling, though its utility is more established in oncological monitoring than broad inflammation.[3]High CD38 expression on leukemic cells holds significant prognostic value in leukemias, particularly chronic lymphocytic leukemia (CLL), where surface CD38 positivity exceeding 30% identifies patients with aggressive disease and shorter overall survival, independent of other risk factors like IGHV mutation status.[88] In acute myeloid leukemia (AML), CD38 levels enhance cytogenetic risk stratification, with overexpression linked to adverse outcomes and reduced event-free survival rates below 40% at five years.[89] Flow cytometric quantification of CD38 thus aids in risk-adapted therapy decisions.[90]As of 2025, emerging positron emission tomography (PET) imaging with CD38-targeted radiotracers shows promise for theranostics in solid tumors, extending beyond hematological applications. Agents like ^{68}Ga-labeled peptides specifically bind CD38-overexpressing cells in lung cancer models, enabling non-invasive visualization of tumor lesions with high tumor-to-background ratios greater than 5:1.[91] Recent developments, including ^{89}Zr-DFO-isatuximab conjugates, demonstrate feasibility in preclinical solid tumor xenografts, supporting diagnostic and therapeutic monitoring.[92] These tracers facilitate personalized theranostics by assessing CD38 heterogeneity prior to targeted interventions.[38]
Therapeutic Applications
Monoclonal Antibodies
Daratumumab is a human IgG1κ monoclonal antibody approved by the U.S. Food and Drug Administration in 2015 for the treatment of multiple myeloma in patients who have received at least three prior lines of therapy.[93][94] It binds to CD38 on myeloma cells, triggering multiple antitumor mechanisms, including antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and antibody-dependent cellular phagocytosis (ADCP), as well as direct apoptosis through immune-mediated crosslinking.[95] These effects are particularly effective against CD38-expressing myeloma cells, even in the presence of bone marrow stromal cells that may otherwise shield tumor cells from immune attack.[95]Isatuximab, another anti-CD38 monoclonal antibody, differs in its enhanced capacity for direct tumor cell killing. It induces apoptosis in myeloma cells independently of Fc-dependent immune mechanisms, primarily through CD38 receptor crosslinking that activates intracellular signaling pathways, including caspase activation and lysosomal permeabilization.[96][97] Like daratumumab, isatuximab also engages ADCC and CDC, but its unique epitope on CD38 allows for more potent direct induction of programmed cell death in CD38-positive malignant cells.[98]In clinical trials, combinations of these antibodies with immunomodulatory drugs (IMiDs) such as lenalidomide have demonstrated high efficacy in relapsed multiple myeloma. For instance, the phase 3 POLLUX trial showed that daratumumab plus lenalidomide and dexamethasone achieved an overall response rate of 93% in patients with relapsed or refractory disease, compared to 75% with lenalidomide and dexamethasone alone, with deepened responses including complete response rates of 13% versus 6%.[99][100] Similarly, isatuximab combined with pomalidomide and dexamethasone in the ICARIA-MM trial yielded a 60% overall response rate versus 35% for pomalidomide and dexamethasone, highlighting the synergistic enhancement of IMiD activity through CD38 targeting.[101]Resistance to CD38-targeting monoclonal antibodies can emerge through mechanisms such as CD38 antigen loss on tumor cells or trogocytosis, where immune effector cells strip CD38 from the myeloma cell surface during ADCC, reducing target availability.[102][103] These processes contribute to diminished antibody efficacy over time, particularly in heavily pretreated patients, and underscore the need for strategies to monitor and overcome antigen modulation.[104]As of 2025, next-generation developments include bispecific antibodies such as CD38xCD3 constructs that redirect T cells to CD38-expressing tumors, expanding beyond traditional Fc-mediated effects to enhance T-cell cytotoxicity. These agents are under investigation primarily for hematological malignancies but show promise for T-cell redirection in CD38-positive solid tumors, with ongoing preclinical and early-phase trials exploring their broader applicability.[105][106]
Small Molecule Inhibitors
Small molecule inhibitors of CD38 primarily target its enzymatic activities, including NAD+ glycohydrolase and ADP-ribosyl cyclase functions, to prevent NAD+ depletion and cyclic ADP-ribose (cADPR) production. These compounds offer a pharmacological approach to modulate CD38-mediated signaling without affecting its role as a cell surface receptor, distinguishing them from antibody-based therapies. By binding to the active site or adjacent regions, such inhibitors can selectively block catalysis while exhibiting favorable pharmacokinetics for systemic administration.[107]Natural modulators of CD38 include flavonoids like quercetin and apigenin, which act as partial inhibitors by occupying a flavone-binding pocket near the enzyme's active site. Apigenin, in particular, inhibits CD38 NADase activity at micromolar concentrations, leading to elevated intracellular NAD+ levels and reduced protein acetylation in cellular models. Similarly, quercetin competes for the same binding site, promoting NAD+ accumulation and demonstrating partial inhibition of both hydrolase and cyclase activities in vitro. These plant-derived compounds highlight the potential for dietary interventions to fine-tune CD38 function, though their low potency limits standalone therapeutic use.[108][109]Among synthetic small molecules, 78c (a thiazoloquinazolinone derivative nicknamed "appetite for destruction" in early studies) stands out as a potent, reversible inhibitor of CD38, with IC50 values of 1.9 nM for mouse CD38 and 7.3 nM for human CD38 across hydrolase and cyclase activities. Other synthetic inhibitors, such as 8-amino-N1-inosine 5'-monophosphate (8-NH2-N1-IMP) derivatives, target the active site to block cADPR hydrolysis, achieving sub-micromolar inhibition and showing selectivity over related enzymes. These compounds are designed based on structural insights into CD38's NAD+-bindingpocket, incorporating nucleoside mimics or heterocyclic scaffolds to enhance potency and membrane permeability. Lucifer yellow derivatives have also been explored as fluorescent probes that bind the active site, informing the development of non-fluorescent inhibitors with similar structural features.[110][111]The therapeutic rationale for CD38 small molecule inhibitors centers on restoring NAD+ homeostasis, which declines with age and in pathological states. In aging models, 78c administration reverses tissue NAD+ decline, improves mitochondrial function, and ameliorates metabolic dysfunction in aged mice, extending median lifespan by approximately 10%. In cancer contexts, CD38 inhibition suppresses tumor growth; for instance, pharmacological blockade or genetic knockout reduces clonogenic growth of lung cancer cells and attenuates tumor progression in mouse xenografts by limiting NAD+-dependent metabolic reprogramming in tumor cells. These effects underscore CD38's role in supporting immunosuppressive microenvironments and fueling glycolysis in malignancies.[112][113][114]As of 2025, small molecule CD38 inhibitors remain primarily in preclinical development for neurodegeneration, with studies demonstrating neuroprotection through NAD+ elevation and reduced neuroinflammation in Alzheimer's disease models. Compounds like 78c exhibit favorable safety profiles in rodent toxicity assays, showing minimal off-target effects on related hydrolases and no significant cardiovascular or hepatic liabilities at therapeutic doses. Emerging candidates, such as VRG201 (a small-molecule CD38 inhibitor developed by Verge Genomics for metabolic diseases), are advancing toward clinical evaluation.[115][116]