Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 426
1.
Cells ; 13(10)2024 May 17.
Article En | MEDLINE | ID: mdl-38786084

Relevant advances have been made in the management of relapsed/refractory (r/r) Hodgkin Lymphomas (HL) with the use of the anti-CD30 antibody-drug conjugate (ADC) brentuximab-vedotin (Bre-Ved). Unfortunately, most patients eventually progress despite the excellent response rates and tolerability. In this report, we describe an ADC composed of the aminobisphosphonate zoledronic acid (ZA) conjugated to Bre-Ved by binding the free amino groups of this antibody with the phosphoric group of ZA. Liquid chromatography-mass spectrometry, inductively coupled plasma-mass spectrometry, and matrix-assisted laser desorption ionization-mass spectrometry analyses confirmed the covalent linkage between the antibody and ZA. The novel ADC has been tested for its reactivity with the HL/CD30+ lymphoblastoid cell lines (KMH2, L428, L540, HS445, and RPMI6666), showing a better titration than native Bre-Ved. Once the HL-cells are entered, the ADC co-localizes with the lysosomal LAMP1 in the intracellular vesicles. Also, this ADC exerted a stronger anti-proliferative and pro-apoptotic (about one log fold) effect on HL-cell proliferation compared to the native antibody Bre-Ved. Eventually, Bre-Ved-ZA ADC, in contrast with the native antibody, can trigger the proliferation and activation of cytolytic activity of effector-memory Vδ2 T-lymphocytes against HL-cell lines. These findings may support the potential use of this ADC in the management of r/r HL.


Brentuximab Vedotin , Immunoconjugates , Ki-1 Antigen , Zoledronic Acid , Humans , Zoledronic Acid/pharmacology , Zoledronic Acid/therapeutic use , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Immunoconjugates/chemistry , Brentuximab Vedotin/pharmacology , Brentuximab Vedotin/therapeutic use , Ki-1 Antigen/metabolism , Ki-1 Antigen/immunology , Cell Line, Tumor , Hodgkin Disease/drug therapy , Hodgkin Disease/pathology , Hodgkin Disease/immunology , Apoptosis/drug effects , Cell Proliferation/drug effects
3.
J Exp Med ; 220(8)2023 08 07.
Article En | MEDLINE | ID: mdl-37097292

Control of Mycobacterium tuberculosis (Mtb) infection requires generation of T cells that migrate to granulomas, complex immune structures surrounding sites of bacterial replication. Here we compared the gene expression profiles of T cells in pulmonary granulomas, bronchoalveolar lavage, and blood of Mtb-infected rhesus macaques to identify granuloma-enriched T cell genes. TNFRSF8/CD30 was among the top genes upregulated in both CD4 and CD8 T cells from granulomas. In mice, CD30 expression on CD4 T cells is required for survival of Mtb infection, and there is no major role for CD30 in protection by other cell types. Transcriptomic comparison of WT and CD30-/- CD4 T cells from the lungs of Mtb-infected mixed bone marrow chimeric mice showed that CD30 directly promotes CD4 T cell differentiation and the expression of multiple effector molecules. These results demonstrate that the CD30 co-stimulatory axis is highly upregulated on granuloma T cells and is critical for protective T cell responses against Mtb infection.


Mycobacterium tuberculosis , Tuberculosis , Animals , Mice , CD4-Positive T-Lymphocytes , Cell Differentiation , Granuloma/metabolism , Macaca mulatta , Tuberculosis/microbiology , Ki-1 Antigen/immunology
4.
Dermatol Online J ; 29(1)2023 Feb 15.
Article En | MEDLINE | ID: mdl-37040909

Methotrexate (MTX) is a first-line systemic medication used to treat rheumatoid arthritis because of its immunomodulatory effects. However, MTX has also been linked to the development of lymphoproliferative disorders (LPD) in patients with rheumatoid arthritis. We describe a patient with long-standing rheumatoid arthritis treated with MTX who developed cutaneous Epstein-Barr virus (EBV)-positive B cell lymphoproliferative disease resembling grade III lymphomatoid granulomatosis localized to the right leg. The lymphomatoid process resolved with withdrawal of the MTX. The pathogenesis of iatrogenic lymphoproliferative disorder was most likely triggered by the rheumatoid inflammation and the immunosuppressing effects of MTX, which led to EBV reactivation. We recommend a trial of MTX discontinuation prior to considering chemotherapy in patients with rheumatoid arthritis treated with MTX who develop EBV-positive B cell lymphoproliferative disease resembling a high grade B-cell lymphoma.


Arthritis, Rheumatoid , Epstein-Barr Virus Infections , Lymphomatoid Granulomatosis , Lymphoproliferative Disorders , Humans , Arthritis, Rheumatoid/drug therapy , Epstein-Barr Virus Infections/chemically induced , Herpesvirus 4, Human , Lymphomatoid Granulomatosis/chemically induced , Lymphomatoid Granulomatosis/drug therapy , Methotrexate/therapeutic use , Ki-1 Antigen/immunology , Lymphoproliferative Disorders/complications , Lymphoproliferative Disorders/immunology , B-Lymphocytes/immunology
5.
J Clin Invest ; 132(2)2022 01 18.
Article En | MEDLINE | ID: mdl-34813503

Tertiary lymphoid tissues (TLTs) facilitate local T and B cell interactions in chronically inflamed organs. However, the cells and molecular pathways that govern TLT formation are poorly defined. Here, we identified TNF superfamily CD153/CD30 signaling between 2 unique age-dependent lymphocyte subpopulations, CD153+PD-1+CD4+ senescence-associated T (SAT) cells and CD30+T-bet+ age-associated B cells (ABCs), as a driver for TLT expansion. SAT cells, which produced ABC-inducing factors IL-21 and IFN-γ, and ABCs progressively accumulated within TLTs in aged kidneys after injury. Notably, in kidney injury models, CD153 or CD30 deficiency impaired functional SAT cell induction, which resulted in reduced ABC numbers and attenuated TLT formation with improved inflammation, fibrosis, and renal function. Attenuated TLT formation after transplantation of CD153-deficient bone marrow further supported the importance of CD153 in immune cells. Clonal analysis revealed that SAT cells and ABCs in the kidneys arose from both local differentiation and recruitment from the spleen. In the synovium of aged rheumatoid arthritis patients, T peripheral helper/T follicular helper cells and ABCs also expressed CD153 and CD30, respectively. Together, our data reveal a previously unappreciated function of CD153/CD30 signaling in TLT formation and propose targeting the CD153/CD30 signaling pathway as a therapeutic target for slowing kidney disease progression.


Acute Kidney Injury/immunology , Aging/immunology , CD30 Ligand/immunology , Ki-1 Antigen/immunology , Lymphoid Tissue/immunology , Signal Transduction/immunology , Acute Kidney Injury/genetics , Aging/genetics , Animals , CD30 Ligand/genetics , CD4-Positive T-Lymphocytes/immunology , Ki-1 Antigen/genetics , Male , Mice , Mice, Knockout , Signal Transduction/genetics
6.
Clin Cancer Res ; 27(13): 3744-3756, 2021 07 01.
Article En | MEDLINE | ID: mdl-33986022

PURPOSE: Natural killer (NK)-cell recognition and function against NK-resistant cancers remain substantial barriers to the broad application of NK-cell immunotherapy. Potential solutions include bispecific engagers that target NK-cell activity via an NK-activating receptor when simultaneously targeting a tumor-specific antigen, as well as enhancing functionality using IL12/15/18 cytokine pre-activation. EXPERIMENTAL DESIGN: We assessed single-cell NK-cell responses stimulated by the tetravalent bispecific antibody AFM13 that binds CD30 on leukemia/lymphoma targets and CD16A on various types of NK cells using mass cytometry and cytotoxicity assays. The combination of AFM13 and IL12/15/18 pre-activation of blood and cord blood-derived NK cells was investigated in vitro and in vivo. RESULTS: We found heterogeneity within AFM13-directed conventional blood NK cell (cNK) responses, as well as consistent AFM13-directed polyfunctional activation of mature NK cells across donors. NK-cell source also impacted the AFM13 response, with cNK cells from healthy donors exhibiting superior responses to those from patients with Hodgkin lymphoma. IL12/15/18-induced memory-like NK cells from peripheral blood exhibited enhanced killing of CD30+ lymphoma targets directed by AFM13, compared with cNK cells. Cord-blood NK cells preactivated with IL12/15/18 and ex vivo expanded with K562-based feeders also exhibited enhanced killing with AFM13 stimulation via upregulation of signaling pathways related to NK-cell effector function. AFM13-NK complex cells exhibited enhanced responses to CD30+ lymphomas in vitro and in vivo. CONCLUSIONS: We identify AFM13 as a promising combination with cytokine-activated adult blood or cord-blood NK cells to treat CD30+ hematologic malignancies, warranting clinical trials with these novel combinations.


Antibodies, Bispecific , Immunotherapy , Killer Cells, Natural , Leukemia , Lymphoma , Humans , Antibodies, Bispecific/therapeutic use , Blood/drug effects , Blood/immunology , Cells, Cultured , Combined Modality Therapy , Cytokines/pharmacology , Fetal Blood/drug effects , Fetal Blood/immunology , Immunotherapy/methods , Ki-1 Antigen/immunology , Killer Cells, Natural/immunology , Leukemia/therapy , Lymphoma/therapy , Receptors, IgG/immunology
7.
Bioconjug Chem ; 32(3): 595-606, 2021 03 17.
Article En | MEDLINE | ID: mdl-33630573

To overcome stability and heterogeneity issues of antibody-drug conjugates (ADCs) produced with existing bioconjugation technologies incorporating a maleimide motif, we developed McSAF Inside, a new technology based on a trifunctionalized di(bromomethyl)pyridine scaffold. Our solution allows the conjugation of a linker-payload to previously reduced interchain cysteines of a native antibody, resulting in disulfide rebridging. This leads to highly stable and homogeneous ADCs with control over the drug-to-antibody ratio (DAR) and the linker-payload position. Using our technology, we synthesized an ADC, MF-BTX-MMAE, built from anti-CD30 antibody cAC10 (brentuximab), and compared it to Adcetris, the first line treatment against CD30-positive lymphoma, in a CD30-positive lymphoma model. MF-BTX-MMAE displayed improved DAR homogeneity, with a solid batch-to-batch reproducibility, as well as enhanced stability in thermal stress conditions or in the presence of a free thiol-containing protein, such as human serum albumin (HSA). MF-BTX-MMAE showed antigen-binding, in vitro cytotoxicity, in vivo efficacy, and tolerability similar to Adcetris. Therefore, in accordance with current regulatory expectations for the development of new ADCs, McSAF Inside technology gives access to relevant ADCs with improved characteristics and stability.


Immunoconjugates/metabolism , Ki-1 Antigen/immunology , Lymphoma/immunology , Animals , Disease Models, Animal , Mice , Proof of Concept Study
8.
Cancer Immunol Immunother ; 70(8): 2353-2365, 2021 Aug.
Article En | MEDLINE | ID: mdl-33527196

Regulatory T cells (Tregs) are often enriched in tumors, where their immunosuppressive function has a key role in tumor persistence and progression. In colorectal cancer (CRC), however, Tregs are frequently associated with an improved clinical outcome. Tumor-infiltrating Tregs have been shown to exhibit a distinct signature comprising the co-stimulatory molecules (OX40, 4-1BB), cytokine receptors (IL1R2, IL21R, CCR8, CD30), and co-inhibitory molecules (PD-L1, TIGIT). Here, we showed by flow cytometry that circulating CD45RO+ Tregs from patients with CRC (n = 25) have elevated CD30 and OX40 expression compared to healthy subjects (n = 14). We identified co-expression of CD30 and OX40 on circulating CD45RO+ Tregs using single-cell images captured by the DEPArray™ system. The frequency of CD30+OX40+CD45RO+ Tregs was significantly higher in CRC patients than in healthy subjects (P < 0.001). Importantly, receiver operating characteristic analysis confirmed that this CD30+OX40+ Treg subset could strongly discriminate between CRC patients and healthy subjects with the highest accuracy of 92.3%, an AUC of 0.92, a sensitivity of 88%, a specificity of 100%, a positive predictive value of 100%, a negative predictive value of 82.35%, and a trade-off value of 3.44%, compared to other Treg subsets. Consistently, multiplex-IHC/IF of tumor-infiltrating Tregs revealed a significant association between high densities of CD30+OX40+ Tregs and improved overall survival; no such association was found for other subsets. These data suggest a potential role for CD30+OX40+ Tregs as a diagnostic or prognostic biomarker in CRC.


Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Ki-1 Antigen/immunology , Receptors, OX40/immunology , T-Lymphocytes, Regulatory/immunology , Biomarkers, Tumor/immunology , Cells, Cultured , Humans , Leukocyte Common Antigens/immunology , Prospective Studies , Receptors, Cytokine/immunology , Retrospective Studies
10.
Hematol Oncol Stem Cell Ther ; 14(2): 95-103, 2021 Jun.
Article En | MEDLINE | ID: mdl-32603659

Hodgkin lymphoma (HL) is a highly responsive disease with nearly 70% of patients experiencing cure after front-line chemotherapy. Patients who experience disease relapse receive salvage chemotherapy followed by consolidation with autologous hematopoietic cell transplantation (auto-HCT). Nearly 50% of patients relapse after an auto-HCT and constitute a subgroup with poor prognosis. Novel treatments such as immune checkpoint inhibitors and an anti-CD30 monoclonal antibody are currently approved for patients relapsing after auto-HCT; however, the duration of remission with these therapies remains limited. Allogeneic HCT is currently the only potentially curative treatment modality for patients relapsing after a prior auto-HCT. Early clinical trials with chimeric antigen receptor T-cell therapy targeting CD30 are underway for patients with relapsed/refractory HL and are already demonstrating safety and promising efficacy.


Hematopoietic Stem Cell Transplantation , Hodgkin Disease/therapy , Neoplasm Recurrence, Local/therapy , Animals , Antineoplastic Agents, Immunological/therapeutic use , Disease Management , Hodgkin Disease/diagnosis , Hodgkin Disease/immunology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive , Ki-1 Antigen/antagonists & inhibitors , Ki-1 Antigen/immunology , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/immunology , Prognosis , Transplantation, Autologous
11.
JAMA Dermatol ; 157(3): 317-321, 2021 03 01.
Article En | MEDLINE | ID: mdl-33377934

Importance: Treatment options for Sézary syndrome (SS) are limited and associated with low response rates. Brentuximab vedotin is a CD30-directed antibody-drug conjugate approved for refractory CD30-positive cutaneous T-cell lymphoma. However, limited data exist on its efficacy in SS, including in the pivotal phase 3 ALCANZA (A Phase 3 Trial of Brentuximab Vedotin (SGN-35) Versus Physician's Choice [Methotrexate or Bexarotene] in Participants With CD30-Positive Cutaneous T-Cell Lymphoma) trial. Objective: To assess the preliminary efficacy and tolerability of brentuximab vedotin for SS. Design, Setting, and Participants: From January 1, 2017, to July 31, 2020, a total of 13 patients with SS received brentuximab vedotin and were analyzed as part of a retrospective case series. Median follow-up was 10.4 months (range, 1.4-34.6 months). All patients were 18 years or older with a diagnosis of SS and with B2 blood involvement at the time brentuximab vedotin therapy was initiated. This single-center study was conducted at a major academic referral center. Interventions: Intravenous brentuximab vedotin administration approximately every 3 weeks. Main Outcomes and Measures: The primary end point was the global response rate. Outcomes were assessed in the skin and lymph nodes per the 2011 European Organization for Research and Treatment of Cancer-International Society of Cutaneous Lymphoma response criteria and in the blood per the 2018 Prospective Cutaneous Lymphoma International Prognostic Index revised blood response criteria. Results: The study included 13 patients (8 [62%] male; mean [SD] age, 68.2 [8.6] years). Of these 13 patients, 5 (38%) achieved a global response after a median of 6 cycles, including 1 complete response. Response rates by disease compartment were 38% in the skin, 63% in the blood, and 50% in the lymph nodes. Three of 11 patients (27%) with pruritus reported improvement. Skin CD30 positivity (>10%) was detected in 9 patients but was not associated with responses. Among responders, the median time to response was 6 weeks (range, 6-9 weeks), and the median duration of response was 5.5 months (range, 2.5-28.9 months). The median time to next treatment was 3.2 months (range, 1.5-36.7 months). Peripheral neuropathy occurred in 4 patients but resolved in 2 patients. Grade 2 adverse events were neuropathy (n = 2), constipation (n = 1), and hand-foot syndrome (n = 1). Conclusions and Relevance: In this case series, brentuximab vedotin use was associated with some efficacy in SS across multiple disease compartments and in the setting of refractory disease or low CD30 skin expression. Brentuximab vedotin may offer a manageable treatment schedule and low incidence of significant toxic effects.


Antineoplastic Agents, Immunological/administration & dosage , Brentuximab Vedotin/administration & dosage , Sezary Syndrome/drug therapy , Skin Neoplasms/drug therapy , Aged , Antineoplastic Agents, Immunological/adverse effects , Brentuximab Vedotin/adverse effects , Female , Humans , Ki-1 Antigen/immunology , Male , Middle Aged , Retrospective Studies , Sezary Syndrome/pathology , Skin Neoplasms/pathology , Time Factors , Treatment Outcome
12.
J Neurovirol ; 26(6): 863-869, 2020 12.
Article En | MEDLINE | ID: mdl-33025348

It is estimated that about 10-20 million peoples are infected with human T-cell leukemia virus type 1 (HTLV-1) around the world and suffered from HTLV-related diseases. The present study was aimed to evaluate the cellular immunity, T-cell activation, humoral immunity, and inflammatory response hallmarks which affect HTLV-1-associated disease progression. A total of 78 participants were included in the study, comprising 39 HTLV-1 asymptomatic careers (ACs) and 39 healthy controls. The HTLV-proviral load (PVL) was determined via real-time PCR technique, and anti-HTLV antibody, sIL2R, sCD30, Neoptrin, hs-CRP, IgE, anti-VCA, anti-EBNA, and anti-EA were assessed by ELISA method. Mean PVL in ACs was 352.7 ± 418.7 copies/104 PBMCs. A significant higher level of sIL-2R was observed in ACs (P < 0.0001). Anti-VCA antibody titer in ACs and healthy controls was 80.72 ± 105.95 and 156.05 ± 130.71, respectively (P = 0.007). Intriguingly, suppression in ACs immune response was not observed. Resultantly, HTLV-1 infection has no effect on the humoral immune response in ACs but greater T-cell activation and function cellular responses were detected. Finally, more studies on various immune markers in adult T-cell leukemia/lymphoma (ATLL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) patients are greatly needed to illuminate the association of ACs' immune status with the development of the related diseases.


CD4-Positive T-Lymphocytes/immunology , HTLV-I Infections/immunology , Human T-lymphotropic virus 1/immunology , Immunity, Cellular , Immunity, Innate , Adult , Antibodies, Viral/blood , Asymptomatic Diseases , C-Reactive Protein/immunology , C-Reactive Protein/metabolism , CD4-Positive T-Lymphocytes/virology , Case-Control Studies , Female , HTLV-I Infections/blood , HTLV-I Infections/diagnosis , HTLV-I Infections/virology , Humans , Immunoglobulin E/blood , Interleukin-2 Receptor alpha Subunit/blood , Interleukin-2 Receptor alpha Subunit/immunology , Iran , Ki-1 Antigen/blood , Ki-1 Antigen/immunology , Lymphocyte Activation , Male , Middle Aged , Neopterin/blood , Neopterin/immunology , Viral Load
15.
J Clin Oncol ; 38(32): 3794-3804, 2020 11 10.
Article En | MEDLINE | ID: mdl-32701411

PURPOSE: 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 CD30 (CD30.CAR-Ts) can be used to treat Hodgkin lymphoma (HL). METHODS: We conducted 2 parallel phase I/II studies (ClinicalTrials.gov identifiers: NCT02690545 and NCT02917083) at 2 independent centers involving patients with relapsed or refractory HL and administered CD30.CAR-Ts after lymphodepletion with either bendamustine alone, bendamustine and fludarabine, or cyclophosphamide and fludarabine. The primary end point was safety. RESULTS: Forty-one patients received CD30.CAR-Ts. Treated patients had a median of 7 prior lines of therapy (range, 2-23), including brentuximab vedotin, checkpoint inhibitors, and autologous or allogeneic stem cell transplantation. The most common toxicities were grade 3 or higher hematologic adverse events. Cytokine release syndrome was observed in 10 patients, all of which were grade 1. No neurologic toxicity was observed. The overall response rate in the 32 patients with active disease who received fludarabine-based lymphodepletion was 72%, including 19 patients (59%) with complete response. With a median follow-up of 533 days, the 1-year progression-free survival and overall survival for all evaluable patients were 36% (95% CI, 21% to 51%) and 94% (95% CI, 79% to 99%), respectively. CAR-T cell expansion in vivo was cell dose dependent. CONCLUSION: Heavily pretreated patients with relapsed or refractory HL who received fludarabine-based lymphodepletion followed by CD30.CAR-Ts had a high rate of durable responses with an excellent safety profile, highlighting the feasibility of extending CAR-T cell therapies beyond canonical B-cell malignancies.


Hodgkin Disease/therapy , Immunotherapy, Adoptive/methods , Ki-1 Antigen/immunology , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bendamustine Hydrochloride/administration & dosage , Bendamustine Hydrochloride/therapeutic use , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Epitopes , Female , Hodgkin Disease/drug therapy , Hodgkin Disease/immunology , Humans , Immunotherapy, Adoptive/adverse effects , Ki-1 Antigen/antagonists & inhibitors , Lymphocyte Depletion , Male , Middle Aged , Stem Cell Transplantation/methods , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives , Young Adult
16.
Blood ; 136(21): 2401-2409, 2020 11 19.
Article En | MEDLINE | ID: mdl-32730586

In relapsed/refractory Hodgkin lymphoma (R/R HL), immunotherapies such as the anti-programmed death-1 inhibitor pembrolizumab have demonstrated efficacy as monotherapy and are playing an increasingly prominent role in treatment. The CD30/CD16A-bispecific antibody AFM13 is an innate immune cell engager, a first-in-class, tetravalent antibody, designed to create a bridge between CD30 on HL cells and the CD16A receptor on natural killer cells and macrophages, to induce tumor cell killing. Early studies of AFM13 have demonstrated signs of efficacy as monotherapy for patients with R/R HL and the combination of AFM13 with pembrolizumab represents a rational new treatment modality. Here, we describe a phase 1b, dose-escalation study to assess the safety and preliminary efficacy of AFM13 in combination with pembrolizumab in patients with R/R HL. The primary objective was estimating the maximum tolerated dose; the secondary objectives were to assess safety, tolerability, antitumor efficacy, pharmacokinetics, and pharmacodynamics. In this heavily pretreated patient population, treatment with the combination of AFM13 and pembrolizumab was generally well tolerated, with similar safety profiles compared to the known profiles of each agent alone. The combination of AFM13 with pembrolizumab demonstrated an objective response rate of 88% at the highest treatment dose, with an 83% overall response rate for the overall population. Pharmacokinetic assessment of AFM13 in the combination setting revealed a half-life of up to 20.6 hours. This proof-of-concept study holds promise as a novel immunotherapy combination worthy of further investigation. This phase 1b study was registered at www.clinicaltrials.gov as NCT02665650.


Antigens, Neoplasm/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hodgkin Disease/drug therapy , Immunotherapy , Ki-1 Antigen/antagonists & inhibitors , Receptors, IgG/antagonists & inhibitors , Adolescent , Adult , Aged , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/pharmacokinetics , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Combined Modality Therapy , Dose-Response Relationship, Immunologic , Female , Half-Life , Hematopoietic Stem Cell Transplantation , Hodgkin Disease/therapy , Humans , Immunity, Innate/drug effects , Ki-1 Antigen/immunology , Male , Maximum Tolerated Dose , Middle Aged , Proof of Concept Study , Receptors, IgG/immunology , Recurrence , Transplantation, Autologous , Young Adult
17.
Curr Hematol Malig Rep ; 15(4): 333-342, 2020 08.
Article En | MEDLINE | ID: mdl-32435988

PURPOSE OF REVIEW: Primary cutaneous CD30+ T-cell lymphoproliferative disorders (CD30+ LPDs) are the second most common cutaneous lymphomas after mycosis fungoides and Sezary syndrome. They include primary cutaneous anaplastic large cell lymphoma (pcALCL), lymphomatoid papulosis (LyP), and borderline lesions. The purpose of this literature review is to consolidate the available evidence on the primary cutaneous CD30+ LPD in order to define the tools for correct diagnosis and appropriate treatment. RECENT FINDINGS: The current body of knowledge regarding the clinical features, histopathologic changes, recently described genetic alterations, and therapeutic options will be covered in this comprehensive review. Primary cutaneous CD30+ LPD represent rare cutaneous lymphomas that have significant histologic overlap within the defined group as well as with other neoplastic and reactive entities. The importance of differentiating these entities is crucial, as each one has a different clinical course and prognosis.


Ki-1 Antigen/immunology , Lymphoma, Primary Cutaneous Anaplastic Large Cell , Lymphomatoid Papulosis , Lymphoproliferative Disorders , Skin Neoplasms , Diagnosis, Differential , Humans , Lymphoma, Primary Cutaneous Anaplastic Large Cell/genetics , Lymphoma, Primary Cutaneous Anaplastic Large Cell/immunology , Lymphoma, Primary Cutaneous Anaplastic Large Cell/therapy , Lymphomatoid Papulosis/genetics , Lymphomatoid Papulosis/immunology , Lymphomatoid Papulosis/therapy , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/therapy , Predictive Value of Tests , Prognosis , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/therapy
18.
Int Immunopharmacol ; 84: 106531, 2020 Jul.
Article En | MEDLINE | ID: mdl-32353687

Inflammatory bowel disease is one of the major causes of colitis-associated colon cancer (CAC). Therefore, it is necessary to explore new therapies to prevent colon cancer (CRC) in view of the relationship between chronic inflammation and tumor development. Previous studies on the correlation between CD30L/CD30 and cancer were mostly limited to lymphoid or homogenous tumors, while there have been only a few reports on the role of CD30L/CD30 signal transduction in the pathogenesis of CAC. In this study, we established an AOM/DSS-induced CAC model with CD30LKO mice to explore the effect of CD30L/CD30 signal transduction on the formation of the intestinal tumor immune microenvironment (TIME) during the development of intestinal tumors. Our results revealed that CD30L deficiency promoted the accumulation of myeloid derived suppressor cells (MDSCs), increased the expression of PD-L1 on MDSCs and tumor associated macrophages (TAMs), and enhanced the secretion of various inflammatory and immunosuppressive factors in the intestinal mucosa of CAC mice. Furthermore, CD30L gene deletion could selectively promote the upregulation of PD-1 expression on CD4+ and CD8+ T cells and inhibit their activation, differentiation and secretion of effector cytokines, which led to an attenuation of antitumor immune responses mediated by TEM (CD44+CD62L-) cells. Thus, our data suggest that CD30L/CD30 signaling might be a potential candidate target for immunological therapy in CAC.


CD30 Ligand/immunology , Colitis-Associated Neoplasms/immunology , Colitis/immunology , Ki-1 Antigen/immunology , Animals , Azoxymethane , CD30 Ligand/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinogens , Colitis/chemically induced , Colitis/complications , Colitis-Associated Neoplasms/etiology , Dextran Sulfate , Female , Intestines/immunology , Ki-1 Antigen/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
19.
Front Immunol ; 11: 398, 2020.
Article En | MEDLINE | ID: mdl-32296414

Cancer cells generally recruit and influence non-malignant immune cells to support the tumor growth. Classical Hodgkin lymphoma (cHL) is a good example because the affected lymphoid tissue contains only a few malignant Hodgkin and Reed-Sternberg (H-RS) cells, which are supported by a massive infiltrate of lymphocytes, fibroblasts, and innate immune cells. The transmembrane receptor CD30, which is selectively expressed on the H-RS cells, plays an important role, not only in cell stimulation and intercellular communication but also in tumor diagnosis and targeted tumor therapy. Different protein processing pathways influence its functionality. Depending on the conditions, the receptor is internalized or released. The release of CD30 occurs either as an intact molecule, embedded in the membrane of extracellular vesicles (EVs), or as a cleaved soluble ectodomain (sCD30). CD30 cleavage is predominantly catalyzed by ADAM10. The enzyme is catalytically active in cells as well as in EVs and gradually releases sCD30. Because the circulation contains no CD30+ donor cells, this mechanism explains that the cleaved ectodomain represents the predominant form of CD30 in the plasma of cHL patients. CD30 processing might influence the impact of CD30 antibody-drug conjugates, such as Brentuximab Vedotin (BV). Whereas, ADAM10-degraded CD30 impedes the BV efficacy, tumor-derived EVs load bystander cells with CD30 and generate new targets among supporter cells. This crossfire effect might contribute to the enormous clinical impact of BV, whereas the ADAM10-dependent cleavage to the mild systemic off-target effects of the treatment with BV.


ADAM10 Protein/immunology , Cell Communication/immunology , Hodgkin Disease/immunology , Ki-1 Antigen/immunology , Extracellular Vesicles/immunology , Humans
20.
Leukemia ; 34(12): 3242-3255, 2020 12.
Article En | MEDLINE | ID: mdl-32203142

While cancer stem cells are well established in certain hematologic and solid malignancies, their existence in T cell lymphoma is unclear and the origin of disease is not fully understood. To examine the existence of lymphoma stem cells, we utilized a mouse model of anaplastic large cell lymphoma. Established NPM-ALK+ lymphomas contained heterogeneous cell populations ranging from mature T cells to undifferentiated hematopoietic stem cells. Interestingly, CD4-/CD8- double negative (DN) lymphoma cells aberrantly expressed the T cell receptor α/ß chain. Serial transplantation of sorted CD4/CD8 and DN lymphoma subpopulations identified lymphoma stem cells within the DN3/DN4 T cell population, whereas all other subpopulations failed to establish serial lymphomas. Moreover, transplanted lymphoma DN3/DN4 T cells were able to differentiate and gave rise to mature lymphoma T cells. Gene expression analyses unmasked stem-cell-like transcriptional regulation of the identified lymphoma stem cell population. Furthermore, these lymphoma stem cells are characterized by low CD30 expression levels, which might contribute to limited long-term therapeutic success in patients treated with anti-CD30-targeted therapies. In summary, our results highlight the existence of a lymphoma stem cell population in a NPM-ALK-driven CD30+ mouse model, thereby giving the opportunity to test innovative treatment strategies developed to eradicate the origin of disease.


Lymphoma, Large-Cell, Anaplastic/immunology , Lymphoma, T-Cell/immunology , Stem Cells/immunology , Anaplastic Lymphoma Kinase/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Disease Models, Animal , Female , Gene Expression/immunology , Ki-1 Antigen/immunology , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Translocation, Genetic/immunology
...