TNFRSF8 (Tumor Necrosis Factor Receptor Superfamily Member 8), also known as CD30 or Ki-1 antigen, is a transmembrane glycoprotein encoded by the TNFRSF8 gene located on chromosome 1p36.22 . It belongs to the TNF receptor superfamily and plays critical roles in immune regulation, apoptosis, and cellular signaling . Primarily expressed on activated T and B cells, TNFRSF8 is a biomarker and therapeutic target in lymphoid malignancies, including Hodgkin’s lymphoma (HL), systemic anaplastic large cell lymphoma (sALCL), and mycosis fungoides .
Gene ID: 943 (NCBI Gene)
HGNC ID: 11923
UniProt ID: P28908
TNFRSF8 is a 120 kDa type I transmembrane glycoprotein composed of:
TNFRSF8 is predominantly expressed in subsets of immune cells, with membranous and cytoplasmic localization observed in:
Tissue-Specific Expression (Human Protein Atlas ):
High Expression | Moderate/Low Expression |
---|---|
Lymph nodes, spleen, thymus | Lung, liver, adrenal gland |
Pro-apoptotic signaling: Limits autoreactive CD8+ T-cell proliferation .
Immune modulation: Promotes Th2-biased responses and B-cell antibody production .
BV, an anti-CD30 antibody-drug conjugate, is FDA-approved for:
Study Cohort | Patients (n) | Objective Response Rate (ORR) | Median Progression-Free Survival (PFS) |
---|---|---|---|
HL (post-transplant) | 102 | 75% (34% CR) | 6.7 months |
Systemic ALCL | 58 | 86% (57% CR) | 12.6 months |
CTCL (vs. standard therapy) | 128 | ORR4: 56% vs. 13% | 16.7 vs. 3.5 months |
Early trials of anti-CD30 monoclonal antibodies (e.g., SGN-30, 5F11) showed limited efficacy, with ORR <20% in HL .
SGN-30: Induced tumor regression in ALCL xenografts via receptor cross-linking .
5F11 (Iratumumab): Enhanced antibody-dependent cytotoxicity in vitro but limited clinical benefit .
Human CD30/TNFRSF8 is a 120 kDa type I transmembrane glycoprotein belonging to the TNF receptor superfamily. The mature protein consists of a 361 amino acid extracellular domain (ECD) with six cysteine-rich repeats, a 28 amino acid transmembrane segment, and a 188 amino acid cytoplasmic domain. There is also an alternatively spliced isoform that includes only the C-terminal 132 amino acids of the cytoplasmic domain. This structure differs significantly from mouse and rat CD30, which contain only three cysteine-rich repeats and lack 90 amino acids of the ECD, with sequence identity of only 53% and 49% in common regions, respectively .
CD30 signaling involves the recruitment of TNF receptor-associated factors (TRAFs), particularly TRAF2 and TRAF5, to its cytoplasmic domain upon ligation with CD30L/TNFSF8. This interaction initiates signal transduction that leads to the activation of NF-κB and other downstream pathways. In T cells, CD30 signaling costimulates antigen-induced Th0 and Th2 proliferation and cytokine secretion, typically favoring a Th2-biased immune response. Even without antigenic stimulation, CD30 can induce T cell expression of IL-13. In B cells, CD30 ligation promotes cellular proliferation, antibody production, and expression of chemokines including CXCR4, CCL3, and CCL5 .
For detecting CD30 expression, flow cytometry remains the gold standard when working with cellular suspensions. Research-grade antibodies conjugated to various fluorophores (including Alexa Fluor 405, 488, and 750) are available for multicolor flow cytometry applications. For optimal results, use fresh samples when possible and include appropriate isotype controls. For tissue sections, immunohistochemistry using anti-CD30 antibodies such as clone 81316 provides excellent sensitivity and specificity. For quantitative analysis of CD30 protein levels, ELISA-based methods can be employed using validated antibody pairs. When developing novel detection methods, validation against established antibodies (such as the research-grade Brentuximab biosimilar) is strongly recommended to ensure specificity .
When designing experiments to study CD30 signaling, consider both ligand-dependent and antibody-mediated receptor activation approaches. For physiologically relevant activation, recombinant human CD30L or cells expressing CD30L should be used. Alternatively, agonistic anti-CD30 antibodies can provide more controlled stimulation. Include appropriate time course experiments (typical signaling events occur within minutes to hours) and analyze multiple downstream pathways including NF-κB activation, MAP kinase phosphorylation, and cytokine production. For comprehensive analysis, complement biochemical assays with transcriptomic approaches to identify the full spectrum of CD30-regulated genes. Controls should include CD30-negative cells and blocking antibodies to confirm specificity of observed effects .
When translating CD30 research between species, researchers must account for significant structural differences between human and rodent CD30. Mouse CD30 lacks 90 amino acids in the extracellular domain and contains only three cysteine-rich repeats (compared to six in humans), with sequence identity of only 53%. These differences may impact ligand binding properties and downstream signaling outcomes. When using animal models, characterize the expression pattern of CD30 in the specific model and validate antibody cross-reactivity. For more translational relevance, consider humanized mouse models expressing human CD30, or use recombinant human CD30 Fc chimera proteins for in vivo studies. Always include appropriate controls that account for species-specific differences in CD30 biology .
Isolation of CD30-positive primary cells requires careful planning due to the activation-dependent expression of this receptor. For T cells, initial activation with anti-CD3/CD28 or PHA/IL-2 for 48-72 hours will upregulate CD30 expression. Subsequently, positive selection using anti-CD30 antibodies conjugated to magnetic beads or fluorescence-activated cell sorting (FACS) can isolate the CD30+ population. Culture these cells in RPMI-1640 supplemented with 10% FBS and appropriate cytokines (IL-2 at 100 U/mL maintains viability). For B cells, stimulation with CD40L and IL-4 effectively induces CD30 expression. Monitor purity by flow cytometry and CD30 expression levels, as receptor expression may decrease without continued activation signals. For long-term studies, consider periodic restimulation protocols to maintain CD30 expression .
Studying CD30-CD30L interactions requires multiple complementary approaches. For binding kinetics, surface plasmon resonance (SPR) using recombinant CD30 extracellular domain and CD30L provides quantitative affinity measurements. Cell-based assays utilizing CD30L-expressing cells (activated T cells, monocytes, or transfected cell lines) co-cultured with CD30+ cells can demonstrate functional interactions. Reporter systems where CD30 signaling activates luciferase or fluorescent protein expression provide convenient readouts. For visualizing interactions in situ, proximity ligation assays or FRET-based approaches can be employed. When designing blocking experiments, use validated antibodies against both CD30 and CD30L, and include recombinant soluble CD30 as a competitive inhibitor of CD30L binding .
For lymphoma research, multi-parameter analysis is essential. Begin with immunophenotyping panels that include CD30 alongside other lymphoma markers (CD15, CD20, PAX5, ALK) to accurately classify CD30+ lymphomas. For functional studies in lymphoma cell lines (such as L428, KM-H2 for Hodgkin lymphoma; SU-DHL-1, Karpas 299 for ALCL), both genetic approaches (CRISPR/Cas9, shRNA) and pharmacological interventions (CD30-targeted antibody-drug conjugates) provide complementary insights. Patient-derived xenograft models maintain the heterogeneity of primary lymphomas and allow for therapeutic testing. When evaluating potential CD30-targeted therapies, assess both direct cytotoxicity and immune-mediated effects, as CD30 antibodies can function through multiple mechanisms. Correlate CD30 expression levels with treatment response to identify predictive biomarkers .
CD30 expression heterogeneity presents a significant analytical challenge. When encountering variable expression, first verify technical consistency in staining protocols and antibody performance using appropriate positive controls (such as stimulated T cells or CD30+ cell lines). For clinical samples, standardize reporting using quantitative metrics (mean fluorescence intensity for flow cytometry; H-score or percentage positive cells for IHC) rather than qualitative assessments. Consider that CD30 expression follows a continuum rather than binary positive/negative categorization. In lymphoma samples, CD30 expression may vary between different regions of the same tumor, necessitating multiple sampling. Temporal variations in CD30 expression may occur during disease progression or treatment response, requiring longitudinal analysis when possible .
Contradictory findings in CD30 signaling literature often stem from differences in experimental systems. When encountering discrepancies, catalog the key variables: cell type (primary cells vs. cell lines), activation state prior to CD30 stimulation, stimulation method (recombinant ligand vs. agonistic antibodies), timing, and downstream readouts. Cell context is particularly important—CD30 signaling outcomes differ substantially between T cells, B cells, and non-lymphoid cells. For mechanistic resolution, employ genetic approaches (knockout/knockin of specific signaling components) to identify the precise pathways contributing to phenotypic outcomes. Techniques like proximity labeling can identify cell-type-specific CD30 interactors that may explain differential signaling. Finally, consider that CD30 may function in signaling complexes with other TNFR family members, creating context-dependent outcomes .
Precision medicine approaches targeting CD30 extend beyond conventional antibody-drug conjugates to include several innovative platforms. Bispecific antibodies linking CD30 with CD3 or other immune effector molecules can enhance immune-mediated killing of CD30+ cells. For mechanistic studies, degrader technologies (PROTACs) targeting CD30 can provide insights into receptor dependency distinct from signaling blockade. CAR-T cells directed against CD30 represent an emerging cellular therapy approach, particularly for relapsed/refractory CD30+ lymphomas. When designing precision targeting studies, consider implementing dynamic monitoring of CD30 expression, as receptor levels may change during treatment. Combination strategies that address potential resistance mechanisms (such as CD30 downregulation or shedding) should be evaluated concurrently. For these advanced platforms, detailed characterization of binding properties, mechanism of action, and off-target effects is essential .
Single-cell technologies offer unprecedented insights into CD30 biology by revealing heterogeneity within seemingly homogeneous populations. Single-cell RNA sequencing of CD30+ cells can identify distinct transcriptional states and novel marker combinations that define functional subsets. For studying rare CD30+ populations, consider index sorting approaches that preserve single-cell protein expression data linked to transcriptional profiles. Single-cell CyTOF/mass cytometry with metal-conjugated anti-CD30 antibodies enables high-dimensional phenotyping in complex samples. For spatial context, imaging mass cytometry or multiplexed immunofluorescence can map CD30+ cells within tissue microenvironments and their interactions with other cell types. Computational tools for trajectory analysis can reveal dynamics of CD30 expression during cellular differentiation or activation processes .
Emerging research suggests CD30 plays more complex roles in immune regulation than previously recognized. Key research directions include investigating CD30's contribution to T cell exhaustion in chronic diseases, as sustained CD30 signaling may lead to altered functional states distinct from classical activation. The role of CD30 in specialized immune compartments such as tertiary lymphoid structures and tumor microenvironments represents another promising area. The bidirectional crosstalk between CD30+ lymphocytes and other tissue-resident cells (including stromal cells and innate lymphoid cells) warrants deeper investigation. For these studies, spatial transcriptomics combined with in situ protein detection provides valuable insights into local signaling networks. Metabolism-focused approaches examining how CD30 signaling reprograms cellular energetics could reveal new therapeutic vulnerabilities. Finally, examining CD30's role in maintaining immunological memory provides important insights for vaccine development and chronic infection management .
The CD30 ligand receptor, also known as TNFRSF8, is a type I transmembrane glycoprotein that belongs to the tumor necrosis factor (TNF) receptor superfamily. It is commonly referred to as CD30 and is recognized for its role in various immune responses and its association with certain types of lymphomas .
The CD30 receptor is a 120 kDa protein that consists of a 361 amino acid extracellular domain (ECD) with six cysteine-rich repeats, a 28 amino acid transmembrane segment, and a 188 amino acid cytoplasmic domain . The receptor is primarily expressed on activated T cells, B cells, and certain lymphoma cells, including Hodgkin’s and Reed-Sternberg cells .
The CD30 ligand (CD30L), also known as TNFSF8, is a type-II membrane-associated glycoprotein. It is expressed on activated T cells, monocyte-macrophages, granulocytes, eosinophils, and some Burkitt-like lymphoma cell lines . The interaction between CD30 and its ligand plays a crucial role in the regulation of immune responses, including cell proliferation, activation, differentiation, and apoptosis .
The CD30-CD30L interaction has been implicated in various biological processes. It is known to mediate pleiotropic effects, including:
The CD30 receptor and its ligand are of particular interest in the context of hematologic malignancies. CD30 is a marker for Hodgkin lymphoma and anaplastic large cell lymphoma, making it a valuable target for diagnostic and therapeutic purposes . The recombinant human CD30 ligand has been utilized in research to study its effects on various cell lines and to develop potential treatments for these malignancies .
Recombinant human CD30 ligand is produced using various expression systems, such as HEK293 cells. It is typically purified to high levels of purity and is used in research to investigate the CD30-CD30L interaction and its biological effects . The recombinant protein is often used in studies related to immune response modulation and the development of targeted therapies for CD30-positive lymphomas .