Fact-checked by Grok 2 weeks ago

CD30

CD30 is a transmembrane glycoprotein and member of the tumor necrosis factor receptor (TNFR) superfamily, also known as TNFRSF8, that functions as a cell surface receptor primarily expressed on activated T and B lymphocytes as well as certain malignant cells. It was first identified in 1982 as a marker on Reed-Sternberg cells in Hodgkin lymphoma using a monoclonal antibody derived from a Hodgkin lymphoma cell line. Structurally, CD30 is a type I protein consisting of approximately 595 amino acids with a molecular weight of 105–120 kDa, featuring an extracellular domain with six cysteine-rich repeats, a single transmembrane domain, and an intracellular domain that binds TRAF proteins to activate signaling pathways such as NF-κB and MAPK/ERK. In healthy individuals, CD30 expression is tightly regulated and limited to subsets of activated immune cells, including CD4+ memory T cells and Th2-type cytokine-producing lymphocytes, where it plays a key role in modulating T- and B-cell interactions, memory T-cell maintenance, and germinal center responses essential for humoral immunity. Its ligand, CD30L (TNFSF8), is expressed on activated monocytes, B cells, and T cells, facilitating bidirectional signaling that influences cell proliferation, apoptosis, and cytotoxicity while regulating lymphocyte trafficking and effector functions. Dysregulated CD30 expression is associated with various diseases, particularly hematopoietic malignancies such as classical Hodgkin lymphoma (cHL), where it is highly expressed on nearly all Reed-Sternberg cells, and anaplastic large cell lymphoma (ALCL), present in over 90% of cases; it also appears in subsets of peripheral T-cell lymphomas, cutaneous T-cell lymphomas, and some non-lymphoid tumors like embryonal carcinoma. In these contexts, CD30 signaling promotes tumor cell survival through anti-apoptotic mechanisms and immune evasion, contributing to lymphomagenesis. Due to its restricted normal expression and prominent role in malignancies, CD30 serves as a valuable diagnostic marker in for identifying CD30-positive lymphomas and guiding therapeutic decisions. Therapeutically, CD30 has emerged as a prime target for antibody-based interventions, most notably , an antibody-drug conjugate approved by the FDA in 2011 for relapsed or refractory cHL and systemic ALCL, which delivers a cytotoxic payload to CD30-expressing cells and has demonstrated objective response rates of 75% in cHL and complete remissions in up to 34% of patients in clinical trials. As of 2025, ongoing research continues to explore additional modalities, including bispecific antibodies like AFM13 (acimtamig) for redirection, which has shown high response rates (up to 90%) in Phase II trials combined with NK cells for relapsed/refractory lymphomas, and chimeric antigen receptor () T-cell therapies demonstrating safety and efficacy in early-phase studies for CD30-positive lymphomas; emerging antibody-drug conjugates such as TUB-010 are also entering clinical development, while addressing challenges such as antigen downregulation and neuropathy-related toxicities.

Molecular characteristics

Gene

The TNFRSF8 gene, which encodes the CD30 protein (also known as member 8), is located on the short arm of human at cytogenetic band 1p36.22, with genomic coordinates spanning from 12,063,303 to 12,144,207 (GRCh38.p14 assembly). The gene covers approximately 81 kb and consists of 16 exons, providing the foundational structure for transcription into mRNA variants. Alternative splicing of TNFRSF8 pre-mRNA generates multiple transcript variants, including the canonical full-length isoform (NM_001243.5, encoding a 595-amino-acid precursor) and a shorter isoform (NM_001281430.3, encoding a 419-amino-acid protein with a truncated cytoplasmic domain). These splice variants result in protein isoforms that differ primarily in their intracellular regions, potentially modulating downstream signaling, though the soluble CD30 (sCD30) detected in is primarily produced through proteolytic shedding of the membrane-bound form by enzymes such as ADAM17/TACE rather than direct splicing. Ensembl annotations identify five transcripts in total, underscoring the 's capacity for isoform diversity. TNFRSF8 exhibits strong evolutionary conservation across mammals, with orthologs reported in 459 species, including mice (Tnfrsf8 on ), rats, and non-human , highlighting its preserved role in immune cell activation and regulation. Gene expression is tightly controlled by promoter elements responsive to transcription factors; for instance, the oncoprotein binds directly to the TNFRSF8 promoter to drive CD30 upregulation in lymphoid malignancies, while JunB relieves transcriptional repression to enhance expression in activated immune cells. Certain polymorphisms in TNFRSF8 influence expression levels and protein processing; a notable example is the low-frequency missense variant rs2230624 (p.Cys273Tyr), which disrupts a bond, leading to reduced surface CD30 expression, lower precursor-to-mature protein ratios, and decreased shedding of sCD30 in cell lines and primary cells. This variant associates with diminished risk (odds ratio 0.82) and lower counts, likely through impaired CD30-mediated T-cell regulation. Other single nucleotide variants may affect splicing efficiency, though specific impacts on TNFRSF8 isoform ratios remain under investigation.

Protein structure

CD30 is a type I transmembrane and a member of the receptor (TNFR) superfamily. The full-length human isoform comprises 595 amino acids, including a 19-amino-acid N-terminal that is cleaved to yield the mature protein. This mature form features a 361-amino-acid extracellular domain, a 28-amino-acid , and a 187-amino-acid cytoplasmic domain. The extracellular domain is characterized by six cysteine-rich domains (CRDs), which form an elongated structure typical of the TNFR superfamily and mediate interactions. These CRDs are stabilized by conserved intramolecular bonds that maintain the pseudorepeat motifs essential for receptor folding and function. CD30 undergoes extensive post-translational N-linked at multiple sites within the extracellular domain, contributing to its and increasing the apparent molecular weight to approximately 120 , compared to the unglycosylated core of about 64 . High-resolution structural data, such as crystal or cryo-EM structures, are not available for CD30; however, models based on related TNFR family members depict the CRDs as a linear array facilitating trimerization upon binding. In addition to the membrane-bound form, a soluble isoform (sCD30) arises primarily through proteolytic shedding by metalloproteinases, releasing the majority of the extracellular domain while omitting the transmembrane and cytoplasmic regions. This ~88 kDa soluble variant retains the six CRDs for but lacks anchoring, allowing it to circulate and potentially modulate signaling by competing with the full-length receptor.

Expression patterns

In normal cells

CD30 is primarily expressed on activated T and B lymphocytes, but not on resting lymphocytes, with expression transiently upregulated during immune in response to antigenic or mitogens. This activation-dependent pattern is observed in a small subset of + and + T cells, particularly those producing Th2-type cytokines, as well as in stimulated B immunoblasts located at the edges of germinal centers and in extrafollicular regions of lymphoid tissues. Low-level expression also occurs in subsets of macrophages, natural killer cells, and , often following or culture conditions that mimic immune responses. During fetal development, CD30 exhibits ontogenic expression starting as early as the 8th week of in various tissues from all three germ layers, including non-hematopoietic systems such as the , , and musculoskeletal system. Expression in the hematolymphoid system, including developing lymphoid organs such as the and nascent lymph nodes, appears from the 10th week onwards, suggesting a role in immunoregulation during T-cell maturation in the thymic medulla and other lymphoid structures. While restricted to cells of hematopoietic origin in adults, CD30 is expressed in various non-hematopoietic fetal tissues early in . The surface density of CD30 on these normal cells is regulated by various cytokines and co-stimulatory signals, including upregulation by IL-4 following TCR engagement and co-stimulation. IL-2 can enhance CD30 expression in certain activated T-cell contexts, such as regulatory T cells, while broader environments influence its modulation during immune responses.

In pathological conditions

CD30 exhibits aberrant overexpression in several hematological malignancies, most notably in the Reed-Sternberg cells of classical , where it serves as a key diagnostic marker and contributes to tumor cell survival and proliferation. In anaplastic large cell lymphoma (ALCL), a subtype of peripheral , CD30 is uniformly and strongly expressed on the surface of nearly all tumor cells, defining the disease's hallmark morphology and supporting its classification as a CD30-positive lymphoproliferative disorder. This contrasts with its limited, activation-induced expression in normal lymphoid cells. CD30 is also prominently expressed in embryonal carcinomas, particularly those of testicular origin, where it aids in distinguishing these aggressive tumors from other subtypes like . In virus-associated lymphomas, such as Epstein-Barr virus (EBV)-positive , CD30 expression is significantly elevated compared to EBV-negative counterparts, with positivity rates of approximately 43% in EBV+ cases versus 16% in EBV- cases (as of 2022) and correlating with viral oncogenesis. Subsets of T-cell non-Hodgkin lymphomas beyond ALCL, including certain peripheral T-cell lymphomas, show variable but notable CD30 positivity, particularly in activated or anaplastic variants. Elevated levels of soluble CD30 (sCD30), a cleaved extracellular form released from activated or malignant cells, are detectable in the of patients with autoimmune diseases, reflecting dysregulated immune activation. In systemic lupus erythematosus, sCD30 concentrations are markedly increased and associate with Th2 bias and disease flares. Similarly, in , sCD30 is higher than in healthy controls and correlates with titers and active . In lymphomas, CD30 expression and sCD30 serum levels often correlate with advanced stages and poorer prognosis; for instance, in , elevated sCD30 at predicts higher clinical , bulky , and reduced event-free . Relapsed or refractory cases of ALCL and other CD30-positive lymphomas frequently show intensified expression, underscoring its role in progression and potential for monitoring.

Biological function

Signaling pathways

Upon binding of CD30 ligand (CD30L), the CD30 receptor, a member of the receptor (TNFR) superfamily, undergoes trimerization, which is essential for initiating intracellular signaling cascades. This trimerization facilitates the recruitment of adaptor proteins, particularly TNFR-associated factors (TRAFs), to the cytoplasmic tail of CD30. TRAF2 and TRAF5 are the primary adaptors involved, binding to specific TRAF-interaction motifs in the CD30 intracellular tail and forming a signaling complex that activates downstream pathways. These TRAFs mediate the activation of the pathway by recruiting the (IKK) complex, leading to phosphorylation and proteasomal degradation of IκB inhibitors, thereby allowing translocation to the nucleus and transcription of target genes. Additionally, TRAF2 contributes to the activation of (MAPK) pathways, including ERK and JNK, which promote activity. The signaling through exerts an anti-apoptotic effect by upregulating survival genes such as and , while MAPK/ERK signaling supports , particularly in activated lymphocytes and certain tumor cells. This dual role enhances cell survival and expansion in contexts where CD30 is expressed. Negative regulation of CD30 signaling occurs through signal-induced depletion of TRAF2, where ligand stimulation triggers rapid TRAF2 via calpain-mediated cleavage, limiting sustained activation and preventing excessive survival signaling. Furthermore, CD30 receptor following ligand binding leads to its lysosomal degradation, often involving ubiquitination of associated proteins to attenuate prolonged signaling.

Physiological roles

CD30 contributes to the regulation of T-cell proliferation and survival after activation by delivering costimulatory signals that enhance cell persistence and prevent exhaustion in activated lymphocytes. This process involves transient activation to support survival signals in post-activation phases. Additionally, CD30 limits the expansion of autoreactive + T cells, thereby promoting and restricting potentially harmful self-reactive responses. In B-cell biology, CD30 facilitates differentiation and production during immune responses to infections by accelerating entry and enhancing formation. It supports isotype switching and the expansion of IgG1-switched B cells, leading to improved high-affinity titers in secondary responses. These functions are evident in studies of CD30-deficient models, which show impaired maintenance and reduced . CD30 contributes to lymphoid organ development through interactions involving CD30L expression on lymphoid inducer (LTi) cells. Furthermore, it participates in responses to pathogens such as viruses and , where its absence leads to diminished + T-cell expansion and production against certain infections like Mycobacterium avium, though it appears dispensable for others like .

Molecular interactions

Ligand

CD30 ligand (CD30L), also known as TNFSF8 or CD153, is a type II transmembrane belonging to the (TNF) superfamily. It consists of a short N-terminal cytoplasmic domain, a transmembrane region, and a C-terminal extracellular domain with homology to other TNF family ligands. CD30L is expressed on the surface of activated T cells, stimulated monocyte-macrophages, granulocytes, , and certain Burkitt-like cell lines, with expression tightly regulated by cellular activation states. In unstimulated cells such as resting T cells or monocytes, CD30L is undetectable, but activation induces its transcription and surface presentation. As the primary for the CD30 receptor (TNFRSF8), CD30L binds with high , typically in the range of 1–5 (Kd ≈ 1.1 reported for soluble CD30-Fc to CD30L-expressing cells). Like other TNF superfamily members, CD30L forms homotrimers via its TNF homology domain, enabling it to engage and cluster multiple CD30 monomers on opposing cells, which is essential for efficient . This trimeric stoichiometry promotes receptor oligomerization without requiring additional adaptor proteins for initial binding. A soluble form of CD30L (sCD30L) arises from proteolytic ectodomain shedding of the membrane-bound protein, primarily catalyzed by metalloproteases. This process releases bioactive sCD30L into the extracellular milieu, where it can modulate signaling at distant sites. Shedding and overall CD30L expression are upregulated by inflammatory stimuli, including cytokines like (IFN-γ), which enhance transcription in monocytes and T cells during immune activation. CD30L demonstrates strong evolutionary conservation within the lineage, with functional homologs present in mammals, birds (e.g., TNFSF8), and certain basal vertebrates like the (a lobe-finned fish), underscoring its fundamental role in adaptive immunity. The TNF homology domain shows particularly high sequence similarity across species (e.g., >70% identity between and murine CD30L), while species-specific variations occur in regulatory regions, potentially influencing expression patterns. This conservation highlights CD30L's emergence early in gnathostome evolution as a key costimulatory molecule.

Protein partners

CD30 primarily interacts with members of the tumor necrosis factor receptor-associated factor (TRAF) family through specific motifs in its cytoplasmic domain, facilitating . The cytoplasmic tail of CD30 contains two distinct TRAF-binding regions: the proximal region binds TRAF1, TRAF2, and TRAF5, while the distal region interacts with TRAF3. These interactions, particularly with TRAF2 and TRAF5, are crucial for activating downstream pathways such as , which promote cell survival and proliferation upon binding. In addition to pro-survival signaling, CD30 associates with components of the death-inducing signaling complex (DISC), including Fas-associated death domain () and , to mediate pro-apoptotic effects under specific conditions, such as in anaplastic large cell lymphoma cells. FADD recruitment to CD30 enables activation, leading to when survival pathways are inhibited, for instance, by p38 MAPK blockade. CD30 exhibits potential cross-talk with other tumor necrosis factor receptor (TNFR) superfamily members, including CD40 and . CD30 expression can be induced by CD40 signaling, and both receptors share overlapping TRAF-dependent pathways that regulate activation and proliferation. Similarly, CD30-mediated may involve autocrine Fas/FasL interactions, amplifying apoptotic signals in certain cellular contexts. Following ligand-induced oligomerization, CD30 triggers post-interaction modifications of its partners, notably the ubiquitination and subsequent proteasomal degradation of TRAF2, TRAF3, and TRAF5, which attenuates prolonged survival signaling and may promote . This TRAF degradation serves as a mechanism to balance CD30's dual roles in fate decisions.

Clinical relevance

Biomarker applications

CD30 serves as a key in the and of various lymphoid malignancies, particularly those with aberrant expression in neoplastic cells. (IHC) is widely employed on tissue biopsies to detect CD30 expression, revealing characteristic membranous and/or cytoplasmic staining patterns in CD30-positive lymphomas such as classical (cHL) and anaplastic large cell (ALCL). This method aids in identifying CD30+ tumor cells, with strong positivity often correlating with diagnostic confirmation in over 90% of cHL cases and most systemic ALCL subtypes. Serum levels of soluble CD30 (sCD30), a shed ectodomain of the receptor, provide a non-invasive for assessing activity and in CD30-expressing malignancies. Elevated sCD30 concentrations, typically exceeding 100 U/mL, are associated with active , response, and poor outcomes in patients with and ALCL, enabling serial monitoring without repeated biopsies. For instance, pre-treatment sCD30 levels above this threshold predict inferior event-free survival in advanced-stage cHL. Flow cytometry offers a quantitative approach to evaluate CD30 surface expression on peripheral blood or bone marrow cells, useful in detecting minimal residual disease or aberrant expression in autoimmune and lymphoproliferative disorders like lymphomatoid papulosis or certain T-cell lymphomas. This technique, utilizing monoclonal antibodies such as Ber-H2, allows for precise gating and measurement of CD30+ populations, facilitating early detection in peripheral samples. In differential diagnosis, CD30 expression is often integrated with other markers, such as CD15, to distinguish cHL from non-Hodgkin lymphomas; CD30+ CD15- patterns are hallmark for Reed-Sternberg cells in cHL, enhancing specificity when combined with CD20 or CD45 staining.

Role in disease

CD30 plays a significant oncogenic role in certain lymphomas, particularly through ligand-independent signaling that constitutively activates the pathway, thereby promoting tumor survival and proliferation. In classical (cHL), overexpressed CD30 on Hodgkin-Reed-Sternberg cells drives persistent activation, which sustains anti-apoptotic gene expression and enhances proliferation independent of ligand binding. Similarly, in anaplastic large lymphoma (ALCL), high CD30 expression contributes to by inducing constitutive signaling via degradation of TRAF3, although the NPM-ALK in ALK-positive ALCL can modulate this pathway, leading to complex effects on survival. This activation fosters an inflammatory microenvironment that supports tumor growth in both cHL and ALCL. In autoimmune diseases, CD30 is associated with dysregulated T-cell responses, where CD30-positive T cells accumulate at sites of inflammation, and elevated levels of soluble CD30 (sCD30) correlate with disease activity and inflammatory processes. For instance, in , high serum sCD30 levels reflect an attempt by CD30+ T cells to downregulate excessive inflammation through regulatory mechanisms, though this often fails to control chronic . Similarly, increased sCD30 is observed in , , and thyroid autoimmunity such as and , linking CD30 to Th2-biased immune dysregulation that exacerbates tissue damage. CD30 expression facilitates viral persistence in infections like those caused by Epstein-Barr virus (EBV) and human T-lymphotropic virus type 1 (HTLV-1), where it supports infected cell survival and immune evasion. In EBV-associated , such as EBV-positive , CD30 is frequently co-expressed with EBV markers like , protecting infected cells from and aiding latent viral maintenance through NF-κB-mediated survival signals. In HTLV-1 infection leading to adult T-cell leukemia/ (ATLL), CD30 is upregulated in up to 36% of cases, particularly in lymphomatous subtypes, where it drives constitutive NF-κB activation in infected T cells, promoting clonal expansion and viral persistence. High CD30 expression serves as a prognostic indicator in various lymphomas, often associating with more aggressive disease subtypes while showing variable survival implications. In peripheral T-cell lymphomas and extranodal /T-cell lymphomas, CD30 positivity identifies subtypes with distinct molecular profiles and potential for aggressive behavior, though it may confer better outcomes in certain contexts like regional-stage extranodal /T-cell lymphoma. In , CD30 expression marks a subgroup linked to EBV association and aggressive features, but it paradoxically correlates with favorable prognosis in some cohorts due to enhanced responsiveness to immune modulation. Overall, elevated CD30 highlights high-risk, aggressive amenable to targeted evaluation.

Therapeutic targeting

Antibody-drug conjugates

Antibody-drug conjugates (ADCs) targeting CD30 represent a targeted therapeutic approach for CD30-expressing malignancies, such as and anaplastic large cell lymphoma (ALCL), where the component binds specifically to the on tumor cells. (Adcetris), the first and most established CD30-directed , consists of a chimeric IgG1 (cAC10) covalently linked to the microtubule-disrupting agent (MMAE) via a protease-cleavable valine-citrulline linker. Upon binding to CD30, the conjugate internalizes through ; in the , the linker is cleaved by , releasing MMAE, which binds to , inhibits microtubule polymerization, induces G2/M phase arrest, and triggers selectively in CD30-positive cells. The U.S. (FDA) approved in August 2011 for relapsed or refractory after failure of autologous transplantation or at least two prior multi-agent regimens, and for systemic ALCL after failure of at least one prior , based on pivotal 2 trials demonstrating objective response rates (ORR) of 75% (complete response [CR] 34%) in and 86% (CR 57%) in ALCL. The standard dosing regimen is 1.8 mg/kg administered as an intravenous infusion over 30 minutes every 3 weeks, up to a maximum of 16 cycles or until disease progression or unacceptable toxicity. In these trials, common adverse reactions (≥20%) included peripheral sensory (often grade 2 or higher, leading to discontinuation in 10%), , , , pyrexia, rash, nausea, diarrhea, and abdominal pain, with peripheral attributed to MMAE's inhibition affecting neuronal cells. Subsequent FDA approvals have expanded brentuximab vedotin's indications, including in 2015 as consolidation therapy post-autologous transplant in high-risk , in 2018 for previously untreated stage IV classical in combination with , , and (A+AVD; 5-year modified 82% vs. 77% with ), and in 2018 for frontline peripheral (PTCL) with , , and (CHP; ORR 83%, 68%). By 2022, approval extended to pediatric patients aged ≥2 years with previously untreated high-risk classical in combination with , reducing the risk of event-free survival by 59% compared to standard regimens. In frontline settings, the combination has shown improved efficacy with manageable toxicity, though remains a key dose-limiting factor, often mitigated by schedule adjustments or discontinuation. Emerging CD30-targeted ADCs aim to enhance and reduce off-target toxicity through optimized , linkers, or antibodies. For instance, TUB-010, a novel ADC using a tubulysin derivative with a cleavable linker, demonstrated potent antitumor activity in preclinical models of CD30-positive lymphomas, with improved stability and bystander killing compared to . Next-generation linkers, such as enzyme-cleavable or pH-sensitive variants, are under investigation to further minimize premature release and neuropathy incidence in ongoing clinical trials for relapsed CD30-expressing lymphomas.

Cellular therapies

CD30-targeted chimeric receptor () T-cell therapies represent a key advancement in adoptive cellular for CD30-expressing malignancies, particularly relapsed or refractory lymphomas. These second-generation CAR constructs typically incorporate a (scFv) derived from the CD30-specific cAC10, linked to CD3ζ signaling and costimulatory domains such as or 4-1BB to promote robust T-cell expansion and effector function. Clinical development has focused on autologous CAR-T cells, with phase 1/2 trials demonstrating substantial antitumor activity while highlighting the need for optimized lymphodepletion regimens to enhance efficacy. In pivotal phase 1/2 studies, CD30 CAR-T cells have achieved complete response () rates of 40-70% in heavily pretreated patients with CD30-positive lymphomas. The RELY-30 trial (NCT02917083), evaluating autologous CD30 CAR-T cells in relapsed CD30+ Hodgkin lymphoma, reported an overall response rate (ORR) of 72% and a rate of 59% among 32 patients receiving fludarabine-based lymphodepletion, with durable responses observed in a subset at median follow-up exceeding 500 days. Similarly, the CHARIOT trial (NCT04268706), a multicenter phase 2 study of autologous CD30 CAR-T (TT11) in relapsed/refractory classical , yielded an ORR of 73.3% and a rate of 60% in evaluable patients, with low rates of severe . These outcomes position CD30 CAR-T as a viable option post-brentuximab vedotin , though relapse due to loss or T-cell exhaustion remains a concern. Advancements from 2023 to 2025 have aimed to overcome limitations in specificity and access. Bispecific CD20/CD30 CAR-T cells, designed for dual targeting to mitigate escape in B-cell lymphomas with heterogeneous expression, showed feasibility and efficacy in a 2025 case report of bulky transformed , achieving rapid tumor reduction without dose-limiting toxicities. In 2025, the HSP-CAR30 therapy, featuring a high proportion of less-differentiated T cells for improved persistence, achieved 100% ORR (50% CR) in phase I (n=10) and over 55% CR in phase II (n=32) for refractory CD30+ lymphoma, with mild adverse effects and long-term remissions in 60% of complete responders at 34 months median follow-up. For , where skin-homing is critical, CD30 CAR-T cells co-expressing the CCR4 (to enhance migration toward CCL17/CCL22 gradients) demonstrated safety and preliminary activity in a phase 1 trial, with data from the 2024 American Society of Hematology () annual meeting indicating T-cell expansion and responses in CD30+ patients. Key challenges include managing (CRS), which occurs in most patients but is predominantly grade 1-2 and responsive to or steroids, and ensuring CAR-T persistence beyond 3-6 months to prevent relapse. A 2025 analysis reported that most infections following CD30 CAR-T infusion were mild (grade 1-2), with lower severity compared to concurrent CAR-T recipients, supporting the favorable profile. Trials like NCT05634785 are extending applications to non-Hodgkin lymphomas and CD30+ nonseminomatous tumors, evaluating CD30 CAR-T as a bridge to consolidation or standalone therapy. Autologous manufacturing, while personalized, faces scalability hurdles due to vein-to-vein times of 3-4 weeks and high costs per patient; allogeneic CD30 CAR-T variants, such as those engineered on EBV-specific T cells, promise off-the-shelf access and batch production but require gene editing (e.g., via ) to reduce alloreactivity and improve yield.

References

  1. [1]
    Understanding CD30 biology and therapeutic targeting: a historical ...
    Sep 8, 2017 · CD30 is a member of the tumor necrosis factor receptor superfamily. It is characteristically expressed in certain hematopoietic malignancies, ...Role Of Cd30 In Health And... · Cd30 As A Tumor Target · Immunoconjugates
  2. [2]
    CD30 Expression and Its Functions during the Disease Progression ...
    May 13, 2023 · CD30, a member of TNFRSF, is a type I single transmembrane protein consisting of 595 amino acids, whose molecular weight is 105–120 kDa [5].2.2. Cd30 Signal... · 3. Cd30 Gene Induction · 3.1. Cd30 Promoter And...
  3. [3]
    The Polyvalent Role of CD30 for Cancer Diagnosis and Treatment
    CD30 (TNFRSF8) is a protein receptor located on the surface of cells and belongs to the tumor necrosis factor receptor superfamily. It can be found on in ...2. Cd30 In Disease · 2.3. 1. Alk-Positive (alk+)... · 3.1. Brentuximab Vedotin
  4. [4]
    The Opposite Functions of CD30 Ligand Isoforms - PMC - NIH
    Mar 21, 2024 · TNFSF8/CD30 ligand is a TNF superfamily member expressed on several major immune cell types, including activated monocytes, B, and T cells.
  5. [5]
    Deciphering CD30 ligand biology and its role in humoral immunity
    CD30 ligand (CD30L) and CD30 are interacting cell-surface glycoproteins that are members of the tumour necrosis factor (TNF) and tumour necrosis factor ...Expression Of Cd30l And Cd30 · Studies In Cd30l And Cd30... · Cd30/cd30l Interactions...
  6. [6]
    Mechanism of action and therapeutic targeting of CD30 molecule in ...
    Dec 22, 2023 · This review will elaborate on the mechanism of CD30-mediated tumor promotion, explore the molecular factors that regulate CD30 expression and function,3. Cd30-Mediated Tumor... · 3.1. Cd30-Mediated Signaling... · 5.1. 3. B Cell Lymphomas
  7. [7]
    Design and Assessment of Novel Anti-CD30 Chimeric Antigen ...
    CD30 is a cell surface protein expressed on Hodgkin's lymphoma, some T cell lymphomas, and some B cell lymphomas. CD30 has a restricted expression pattern in ...Cd30 Has A Favorable... · 5f11-28z Car T Cells... · Loss Of Cd30 T Cells From...
  8. [8]
    TNFRSF8 TNF receptor superfamily member 8 [ (human)] - NCBI
    Aug 19, 2025 · The protein encoded by this gene is a member of the TNF-receptor superfamily. This receptor is expressed by activated, but not by resting, T and B cells.
  9. [9]
    Soluble cytokine receptor levels in aqueous humour of patients with ...
    Dec 5, 2019 · A soluble form of CD30 is released by CD30+ cells in vitro and in vivo by proteolytic cleavage.
  10. [10]
    Gene: TNFRSF8 (ENSG00000120949) - Summary - Homo_sapiens
    Chromosome 1: 12,063,303-12,144,207 forward strand. ... This gene has 5 transcripts (splice variants), 459 orthologues and 21 paralogues. Transcripts. Show ...
  11. [11]
    The oncogenic transcription factor IRF4 is regulated by a novel ... - NIH
    In addition, IRF4 binds the TNFRSF8 promoter, resulting in increased expression of CD30. CD30 subsequently activates NF-κB, further increasing IRF4 expression ...
  12. [12]
    TNFRSF8 Gene
    Soluble CD30 levels in serum have been employed as an indicator of T‐cell activation in conditions ranging from transplant rejection and HIV persistence to ...
  13. [13]
    Eighty-eight variants highlight the role of T cell regulation and airway ...
    Jan 20, 2020 · We show evidence that two low frequency variants in TNFRSF8 and TGFBR1 associate with decreased asthma risk through loss of function and gain of ...
  14. [14]
    Molecular cloning and expression of a new member of the nerve ...
    1992 Feb 7;68(3):421-7. doi: 10.1016/0092-8674(92)90180-k. Authors. H Dürkop ... CD30 were cloned from expression libraries of the human HUT-102 cell line ...
  15. [15]
    TNFRSF8 Gene - GeneCards | TNR8 Protein | TNR8 Antibody
    The protein encoded by this gene is a member of the TNF-receptor superfamily. This receptor is expressed by activated, but not by resting, T and B cells.
  16. [16]
    Recombinant Human CD30/TNFRSF8 Fc Chimera Protein, CF
    Mature human CD30 consists of a 361 amino acid (aa) extracellular domain (ECD) with six cysteine-rich repeats, a 28 aa transmembrane segment, and a 188 aa ...<|control11|><|separator|>
  17. [17]
    Structural principles of tumor necrosis factor superfamily signaling
    Jan 2, 2018 · The ectodomains of TNFRSFs are made up of several cysteine-rich domains (CRDs) that form elongated structures. Receptors can be classified ...
  18. [18]
    Recombinant Human CD30/TNFRSF8 Protein, CF 6126-CD-100
    Rating 5.0 (1) CD30, also known as Ki-1 antigen and TNFRSF8, is a 120 kDa type I transmembrane glycoprotein belonging to the TNF receptor superfamily.Missing: sCD30 | Show results with:sCD30
  19. [19]
    Understanding CD30 biology and therapeutic targeting - Nature
    Sep 8, 2017 · CD30 is a member of the tumor necrosis factor receptor superfamily. It is characteristically expressed in certain hematopoietic malignancies ...
  20. [20]
    Human macrophages can express the Hodgkin's cell ... - PubMed
    Previous reports on the expression of the Hodgkin's cell-associated antigen Ki-1, CD30, on normal cells have been limited to activated lymphocytes. This study ...
  21. [21]
    Increased expression and activation of CD30 induce apoptosis in ...
    Aug 1, 2004 · CD30 surface expression was up-regulated in eosinophils incubated for 24 h as compared with freshly isolated eosinophils, and both CD30 ...
  22. [22]
    The expression of CD30 and its clinico-pathologic significance in ...
    CD30 expression is minimal in healthy adults but is most prominent in activated B cells, T cells, and NK cells, although it accounts for less than 1% of ...
  23. [23]
    Ber-H2 (CD30) immunohistochemical staining of human fetal tissues
    Our results demonstrated that CD30 is expressed early in human fetal development (8th to 10th week of gestation) in several fetal tissues derived from all ...Missing: lymphoid | Show results with:lymphoid
  24. [24]
    Ber-H2 (CD30) Immunohistochemical Staining of Human Fetal Tissues
    CD30 appears to have an important immunoregulatory role in normal T cell development. Within the thymus, CD30L is highly expressed on medullary thymic ...
  25. [25]
    Expression of the costimulatory receptor CD30 is regulated by both ...
    Expression of the costimulatory receptor CD30 is regulated by both CD28 and cytokines. J Immunol. 1998 Mar 1;160(5):2180-7. Authors. M C Gilfillan , P J ...
  26. [26]
    PD-1 receptor deficiency enhances CD30+ Treg cell function in ...
    Jun 2, 2025 · In the presence of IL-2, PD-1 inhibition led to significant increases in both protein and mRNA CD30 expression compared with the PD-L1 Fc ...
  27. [27]
    New molecular targets in Hodgkin and Reed-Sternberg cells
    CD30 signaling is an important pathway for survival and proliferation of HRS cells. CD30 forms a 120 kDa type I transmembrane glycoprotein, with both ...
  28. [28]
    CD30 + anaplastic large cell lymphoma: a review of its ...
    Defining features consist of a proliferation of predominantly large lymphoid cells with strong expression of the cytokine receptor CD30 and a characteristic ...Abstract · Structure and pathogenic role... · Clinical features and subforms
  29. [29]
    Treatment of CD30‐Expressing Germ Cell Tumors and Sex Cord ...
    Dec 8, 2017 · Expression of CD30 has been observed in testicular cancers, and patients with CD30‐expressing embryonal carcinomas have worse progression ...
  30. [30]
    CD30 protects EBV-positive diffuse large B-cell lymphoma cells ...
    Feb 28, 2024 · Increasing evidence has proved that CD30 expression is significantly higher in EBV+ DLBCL patients compared to EBV-negative (EBV‒) DLBCL and ...
  31. [31]
    Soluble CD30 in early rheumatoid arthritis as a predictor of good ...
    Circulating levels of soluble CD30, a marker of cells producing Th2-type cytokines, are increased in patients with systemic lupus erythematosus and correlate ...
  32. [32]
    Soluble CD30 Levels in Patients with Rheumatoid Arthritis | Archives ...
    The serum levels of sCD30 were significantly increased in RA patients (25.1±18.3 IU/ml) compared to healthy controls (17.2±8.1 IU/ml) (p=0.004). The mean serum ...
  33. [33]
    Serum levels of soluble CD30 are elevated in the majority of ...
    Serum levels of soluble CD30 are elevated in the majority of untreated patients with Hodgkin's disease and correlate with clinical features and prognosis.
  34. [34]
    Clinical implications of serum levels of soluble CD30 in 70 adult ...
    In particular, soluble CD30 (sCD30) levels were elevated in most patients with Hodgkin's disease (HD) and anaplastic large-cell lymphoma (ALCL). Patients and ...
  35. [35]
    CD30: receptor, marker, target | PLMI - Dove Medical Press
    Jun 10, 2016 · Expression of the costimulatory receptor CD30 is regulated by both CD28 and cytokines. J Immunol. 1998;160(5):2180–2187. 7. Stein H, Gerdes ...
  36. [36]
    CD30-dependent degradation of TRAF2: implications for negative ...
    Here we show that one of the effects of CD30 signal transduction is to render cells sensitive to apoptosis induced by the type 1 tumor necrosis factor receptor ...
  37. [37]
    CD30: from basic research to cancer therapy - PMC - PubMed Central
    The picture emerging is that one of the primary functions of CD30 is the control of memory cells providing costimulation and trafficking information or inducing ...Cd30: From Basic Research To... · Murine Cd30 And Cd30-L · Cd30 In Colitis In Animal...
  38. [38]
  39. [39]
    Regulation of CD30 Antigen Expression and Its Potential ... - NIH
    CD30 is a specific marker of a subset of peripheral T cell NHLs known as anaplastic large cell lymphomas (ALCL).
  40. [40]
    CD30 influences germinal center B-cell dynamics and the ... - NIH
    Oct 17, 2024 · Subsequent studies demonstrated that CD30 signaling plays a role in the maintenance of GCs and recall of immune responses.
  41. [41]
  42. [42]
    CD30 Is Dispensable for T-Cell Responses to Influenza Virus and ...
    CD30 is a tumor necrosis factor receptor (TNFR) family member whose expression is associated with Hodgkin's disease, anaplastic large cell lymphomas, and other ...
  43. [43]
    Expression and regulation of CD30 ligand and CD30 in human ...
    These data provide strong evidence for the involvement in growth regulation of recombinant and natural CD30L through its interaction with the CD30 receptor.Missing: transmembrane | Show results with:transmembrane
  44. [44]
    The Opposite Functions of CD30 Ligand Isoforms - MDPI
    TNFSF8/CD30 ligand is a TNF superfamily member expressed on several major immune cell types, including activated monocytes, B, and T cells.
  45. [45]
    CD30L is involved in the regulation of the inflammatory response ...
    CD30L could promote the inflammatory response by inducing the homing and differentiation of monocytes via the chemokine ligand 2 (CCL2)/CCR2 axis and NF-κB ...
  46. [46]
    Vertebrate TNF Superfamily: Evolution and Functional Insights - MDPI
    This study characterizes the evolution of the tumor necrosis factor superfamily (TNFSF) across vertebrate lineages, both cyclostomes and gnathostomes.
  47. [47]
    Tumor Necrosis Factor Superfamily: Ancestral Functions and ...
    Jul 6, 2020 · The evolution of the tumor necrosis factor superfamily (TNFSF) in early vertebrates is inferred by comparing the TNFSF genes found in humans and nine fishes.
  48. [48]
    CD30 induction of human immunodeficiency virus gene transcription ...
    To confirm the interaction of CD30 with TRAF1 and TRAF2, we expressed the intracellular regions of CD30, Fas, and CD40 as GST fusion bacterial proteins, ...Missing: review | Show results with:review
  49. [49]
    Inhibition of p38 mitogen-activated protein kinase unmasks a CD30 ...
    Signaling via TNFR2 was shown to induce c-IAP1–dependent ubiquitination and subsequent proteosomal degradation of the cellular TNFR-associated factor-2 pool ...
  50. [50]
    Inhibition of p38 mitogen-activated protein kinase unmasks a CD30 ...
    Importantly, we show that expression of the caspase-8 inhibitor c-FLIP(S) is strongly induced by the CD30 ligand, and that this is dependent on the activation ...
  51. [51]
    CD30 Is a CD40-Inducible Molecule that Negatively Regulates ...
    Thus, in normal B cells, CD40L-induced CD30 expression is negatively regulated by BCR coengagement and exposure to IL-6 and/or IL-12. CD30 Engagement Hampers ...
  52. [52]
    Induction of cell death by tumour necrosis factor (TNF) receptor 2 ...
    Here we demonstrate in different cellular systems that cytotoxic effects induced by TNF‐R2, CD40 and CD30 are mediated by endogenous production of TNF.<|control11|><|separator|>
  53. [53]
    CD30-dependent degradation of TRAF2: implications for negative ...
    These data suggest a model in which CD30 limits its own ability to transduce cell survival signals through signal-coupled depletion of TRAF2.
  54. [54]
    A novel model of alternative NF-κB pathway activation in Anaplastic ...
    Nov 12, 2020 · Endogenous expression of CD30 induces constitutive NF-κB activation through binding and degrading of TRAF3. In ALK+ ALCL, the CD30 pathway is ...
  55. [55]
    The NPM-ALK oncoprotein abrogates CD30 signaling and ...
    Here we show that NPM-ALK impedes CD30 signaling and NF-κB activation, dependent on both ALK kinase activity and the N-terminal NPM domain.
  56. [56]
    In rheumatoid arthritis soluble CD30 ligand is present at high levels ...
    In RA high sCD30 reflects the attempt by CD30+T cells to downmodulate inflammation. · Evaluation of sCD30 serum levels in RA may predict a good response to ...Missing: elevated | Show results with:elevated
  57. [57]
    Soluble CD30, the Immune Response, and Acute Rejection in ...
    Feb 27, 2020 · Moreover, CD30+ T cells are available at inflammatory sites of several autoimmune diseases, such as atopic dermatitis, rheumatoid arthritis ...
  58. [58]
    Elevated levels of soluble CD26 and CD30 in multiple sclerosis
    Oct 26, 2015 · The present results suggest that the elevated levels of sCD30 are associated with the regulatory immune responses predisposing to clinically ...
  59. [59]
    Increased Serum Concentration of Soluble CD30 in Patients with ...
    In this study, we demonstrated that increased levels of sCD30 are present in patients with Graves' disease and Hashimoto's thyroiditis, especially in the ...
  60. [60]
    CD30 associates with EBER-EBV but not HCV-NS3 in T-cell non ...
    Jun 3, 2025 · CD30, a transmembrane glycoprotein, is known to be uniformly expressed in Hodgkin lymphoma (HL) and anaplastic large cell lymphoma (ALCL) (3).
  61. [61]
    Clinicopathological features and survival in EBV-positive diffuse ...
    Aug 24, 2021 · We found a positive expression of CD30 in almost all EBV+ DLBCL-NOS cases (98%), in contrast with the literature, where CD30 positivity ...
  62. [62]
    Differential CD30 expression in adult T-cell leukemia-lymphoma ...
    Our preliminary data show that 36% of ATLLs are CD30+ and therefore may be amenable to anti-CD30 therapy.
  63. [63]
    Elevated expression of CD30 in adult T-cell leukemia cell lines
    May 6, 2005 · Elevated CD30 expression is considered one of the causes of constitutive NF-κB activation in ATL cells, and may be involved in ATL development.
  64. [64]
    Molecular heterogeneity of CD30+ peripheral T-cell lymphoma with ...
    Molecular heterogeneity of CD30 + peripheral T-cell lymphoma with prognostic significance and therapeutic implications: a retrospective multi-centre study
  65. [65]
    CD30 expression and survival in extranodal NK/T-cell lymphoma
    Mar 27, 2018 · This study indicates that the CD30 expression is significantly associated with better prognosis in ENKTL, especially for patients with regional lymphoma ...Cd30 Expression And Survival... · Results · Study Characteristics<|control11|><|separator|>
  66. [66]
    CD30 expression defines a novel subgroup of diffuse large B-cell ...
    Here we report that CD30 expression is a favorable prognostic factor in a cohort of 903 de novo DLBCL patients. CD30 was expressed in ∼14% of DLBCL patients.
  67. [67]
    Clinical Implications of CD30 Expression in Aggressive B-Cell ...
    We evaluated CD30 expression in 116 cases of aggressive B-cell lymphomas diagnosed at Penn State Milton S. Hershey Medical Center between 2000 and 2012.
  68. [68]
    [PDF] Brentuximab vedotin. - accessdata.fda.gov
    ADCETRIS (brentuximab vedotin) is a CD30-directed antibody-drug conjugate (ADC) consisting of three components: 1) the chimeric IgG1 antibody cAC10, specific ...
  69. [69]
    Brentuximab Vedotin - an overview | ScienceDirect Topics
    Brentuximab vedotin (BV) is an antibody–drug conjugate (ADC) that selectively delivers a toxic microtubule-disrupting agent into CD30-expressing cells, thereby ...
  70. [70]
    Brentuximab Vedotin (SGN-35) for Relapsed CD30-Positive ...
    Nov 4, 2010 · Brentuximab vedotin induced durable objective responses and resulted in tumor regression for most patients with relapsed or refractory CD30-positive lymphomas.Missing: Emerging | Show results with:Emerging<|control11|><|separator|>
  71. [71]
    [PDF] ADCETRIS® (brentuximab vedotin) - accessdata.fda.gov
    In case of overdosage, the patient should be closely monitored for adverse reactions, particularly neutropenia, and supportive treatment should be administered.
  72. [72]
    Brentuximab vedotin - PMC - NIH
    The most common adverse events, predominantly of grade 1 and 2, were fatigue (36%), pyrexia (33%), diarrhea (22%), nausea (22%), neutropenia (22%), and ...
  73. [73]
    FDA Expands Approval of Brentuximab for Hodgkin Lymphoma - NCI
    Apr 17, 2018 · The Food and Drug Administration (FDA) has expanded the approved uses for the drug brentuximab vedotin (Adcetris) in people with Hodgkin lymphoma.
  74. [74]
    Brentuximab Vedotin with Chemotherapy for Stage III or IV ...
    Dec 10, 2017 · Brentuximab vedotin plus AVD (A+AVD) had an acceptable side-effect profile and resulted in complete response in 24 of 25 patients (96%).
  75. [75]
    Adcetris plus Chemotherapy Now FDA Approved for Pediatric ...
    On November 10, 2022, the FDA approved brentuximab vedotin (Adcetris; Seagen) in combination with a chemotherapy regimen of doxorubicin, vincristine, ...Missing: expanded | Show results with:expanded
  76. [76]
    Brentuximab vedotin: clinical updates and practical guidance - PMC
    Brentuximab vedotin (BV), a potent antibody-drug conjugate, targets the CD30 antigen. Owing to the remarkable efficacy shown in CD30-positive lymphomas ...Missing: side | Show results with:side
  77. [77]
    TUB-010, a novel anti-CD30 antibody-drug conjugate based on Tub ...
    Jan 19, 2025 · Researchers have developed TUB-010, a new type of antibody-drug conjugate (ADC) that targets CD30, a protein present in various lymphomas.Missing: Emerging | Show results with:Emerging
  78. [78]
    Antibody-Drug Conjugates Targeting CD30 in T-Cell Lymphomas
    Feb 2, 2025 · The antibody-drug conjugate brentuximab vedotin (BV), targeting CD30-positive cells, has been approved for the treatment of relapsed or ...
  79. [79]
    The third-generation anti-CD30 CAR T-cells specifically homing to ...
    Jun 21, 2022 · The ORR in 32 patients who received fludarabine-based lymphodepletion was 72% including a CR rate of 59%. However, 1-year progression-free ...
  80. [80]
    Anti-CD30 CAR-T Cell Therapy in Relapsed and Refractory Hodgkin ...
    Chimeric antigen receptor (CAR) T-cell therapy of B-cell malignancies has proved to be effective. We show how the same approach of CAR T cells specific for ...Missing: domain 1BB<|control11|><|separator|>
  81. [81]
    Anti-CD30 CAR-T Cell Therapy for Lymphoma
    Nov 10, 2020 · The overall response rate in the 32 patients with active disease who received fludarabine-based lymphodepletion was 72%, including 19 ...
  82. [82]
    Anti-CD30 CAR-T Therapy for Hodgkin Lymphoma: Review
    Jan 14, 2025 · Ramos et al., the objective response rate (ORR) for the 32 patients with HL was 72%, with 19 patients (59%) achieving a complete response (CR), ...
  83. [83]
    Case Report: Bispecific CD20/CD30-targeted chimeric antigen ...
    May 7, 2025 · We report that a patient with bulky transformed follicular lymphoma was successfully treated with CD20/CD30-directed CAR-T cells.
  84. [84]
    Transient responses and significant toxicities of anti-CD30 CAR T ...
    Feb 13, 2024 · ... CD30 CAR T cells for CD30+ lymphomas: results of a phase 1 trial ... The overall response rate was 43%; 1 patient achieved a complete remission.
  85. [85]
    Study Details | NCT05634785 | CD30 CAR for CD30+ NSGCT
    This is a phase 2 research study that enrolls adult subjects with Nonseminomatous Germ Cell Tumors (NSGCT). The purpose of this study is to create a ...Missing: Hodgkin | Show results with:Hodgkin
  86. [86]
    Adoptive T-cell therapy for Hodgkin lymphoma - ScienceDirect.com
    Oct 26, 2021 · The results of clinical trials with EBV-specific CTLs and anti-CD30 CAR T cells to target tumor-associated antigens in patients with extensively ...
  87. [87]
    Scalable Manufacturing of CAR T Cells for Cancer Immunotherapy
    Autologous scaling out versus allogeneic scaling up. A ... CAR T cells is a strategy being implemented in autologous and allogeneic product manufacturing.