Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.039
Filter
1.
Zhonghua Bing Li Xue Za Zhi ; 53(6): 535-540, 2024 Jun 08.
Article in Chinese | MEDLINE | ID: mdl-38825896

ABSTRACT

The 5th edition of the World Health Organization (WHO) classification of haematolymphoid tumours used the hierarchical system to classify T-cell and NK-cell lymphoid proliferations and lymphomas (T/NK-LPD/LYM) based on research advances and clinicopathological characteristics of the diseases. In this edition of classification, tumour-like lesions were included, some tumors were added/deleted, the names or terms of certain diseases were refined, and the diagnostic criteria or subtypes of some diseases were revised. This group of diseases was reintegrated from non-clonal hyperplasia to highly aggressive lymphoma, which would further reflect the nature of T/NK-LPD/LYM and benefit to clinical application.


Subject(s)
Killer Cells, Natural , Lymphoma , T-Lymphocytes , World Health Organization , Humans , Killer Cells, Natural/pathology , Killer Cells, Natural/immunology , T-Lymphocytes/pathology , T-Lymphocytes/immunology , Lymphoma/pathology , Lymphoma/classification , Lymphoma/immunology , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell/classification , Lymphoma, T-Cell/immunology , Lymphoproliferative Disorders/pathology , Lymphoproliferative Disorders/classification , Lymphoproliferative Disorders/immunology
3.
Science ; 383(6686): eadh4059, 2024 03.
Article in English | MEDLINE | ID: mdl-38422122

ABSTRACT

We describe humans with rare biallelic loss-of-function PTCRA variants impairing pre-α T cell receptor (pre-TCRα) expression. Low circulating naive αß T cell counts at birth persisted over time, with normal memory αß and high γδ T cell counts. Their TCRα repertoire was biased, which suggests that noncanonical thymic differentiation pathways can rescue αß T cell development. Only a minority of these individuals were sick, with infection, lymphoproliferation, and/or autoimmunity. We also report that 1 in 4000 individuals from the Middle East and South Asia are homozygous for a common hypomorphic PTCRA variant. They had normal circulating naive αß T cell counts but high γδ T cell counts. Although residual pre-TCRα expression drove the differentiation of more αß T cells, autoimmune conditions were more frequent in these patients compared with the general population.


Subject(s)
Autoimmunity , Intraepithelial Lymphocytes , Membrane Glycoproteins , Receptors, Antigen, T-Cell, alpha-beta , Humans , Autoimmunity/genetics , Cell Differentiation , Homozygote , Intraepithelial Lymphocytes/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Membrane Glycoproteins/genetics , Loss of Function Mutation , Lymphocyte Count , Alleles , Infections/immunology , Lymphoproliferative Disorders/immunology , Pedigree , Male , Female , Middle Aged , Aged , Aged, 80 and over
4.
Dermatol Online J ; 29(1)2023 Feb 15.
Article in English | MEDLINE | ID: mdl-37040909

ABSTRACT

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.


Subject(s)
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 Allergy Clin Immunol ; 149(2): 758-766, 2022 02.
Article in English | MEDLINE | ID: mdl-34329649

ABSTRACT

BACKGROUND: Pediatric nonmalignant lymphoproliferative disorders (PLPDs) are clinically and genetically heterogeneous. Long-standing immune dysregulation and lymphoproliferation in children may be life-threatening, and a paucity of data exists to guide evaluation and treatment of children with PLPD. OBJECTIVE: The primary objective of this study was to ascertain the spectrum of genomic immunologic defects in PLPD. Secondary objectives included characterization of clinical outcomes and associations between genetic diagnoses and those outcomes. METHODS: PLPD was defined by persistent lymphadenopathy, lymph organ involvement, or lymphocytic infiltration for more than 3 months, with or without chronic or significant Epstein-Barr virus (EBV) infection. Fifty-one subjects from 47 different families with PLPD were analyzed using whole exome sequencing. RESULTS: Whole exome sequencing identified likely genetic errors of immunity in 51% to 62% of families (53% to 65% of affected children). Presence of a genetic etiology was associated with younger age and hemophagocytic lymphohistiocytosis. Ten-year survival for the cohort was 72.4%, and patients with viable genetic diagnoses had a higher survival rate (82%) compared to children without a genetic explanation (48%, P = .03). Survival outcomes for individuals with EBV-associated disease and no genetic explanation were particularly worse than outcomes for subjects with EBV-associated disease and a genetic explanation (17% vs 90%; P = .002). Ascertainment of a molecular diagnosis provided targetable treatment options for up to 18 individuals and led to active management changes for 12 patients. CONCLUSIONS: PLPD defines children at high risk for mortality, and whole exome sequencing informs clinical risks and therapeutic opportunities for this diagnosis.


Subject(s)
Lymphoproliferative Disorders/genetics , Adolescent , Autoimmunity , Child , Child, Preschool , Female , Genetic Association Studies , Genetic Testing , Herpesvirus 4, Human/isolation & purification , Humans , Immunity/genetics , Infant , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/mortality , Male , Exome Sequencing , Young Adult
6.
Br J Haematol ; 196(3): 548-558, 2022 02.
Article in English | MEDLINE | ID: mdl-34649298

ABSTRACT

Patients affected by lymphoid malignancies (LM) are frequently immune-compromised, suffering increased mortality from COVID-19. This prospective study evaluated serological and T-cell responses after complete mRNA vaccination in 263 patients affected by chronic lymphocytic leukaemia, B- and T-cell lymphomas and multiple myeloma. Results were compared with those of 167 healthy subjects matched for age and sex. Overall, patient seroconversion rate was 64·6%: serological response was lower in those receiving anti-cancer treatments in the 12 months before vaccination: 55% vs 81·9% (P < 0·001). Anti-CD20 antibody plus chemotherapy treatment was associated with the lowest seroconversion rate: 17·6% vs. 71·2% (P < 0·001). In the multivariate analysis conducted in the subgroup of patients on active treatment, independent predictors for seroconversion were: anti-CD20 treatment (P < 0·001), aggressive B-cell lymphoma diagnosis (P = 0·002), and immunoglobulin M levels <40 mg/dl (P = 0·030). The T-cell response was evaluated in 99 patients and detected in 85 of them (86%). Of note, 74% of seronegative patients had a T-cell response, but both cellular and humoral responses were absent in 13·1% of cases. Our findings raise some concerns about the protection that patients with LM, particularly those receiving anti-CD20 antibodies, may gain from vaccination. These patients should strictly maintain all the protective measures.


Subject(s)
2019-nCoV Vaccine mRNA-1273/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , BNT162 Vaccine/administration & dosage , COVID-19 , Hematologic Neoplasms , Immunity, Cellular/drug effects , Lymphoproliferative Disorders , SARS-CoV-2/immunology , T-Lymphocytes/immunology , 2019-nCoV Vaccine mRNA-1273/immunology , Aged , Antibodies, Viral/immunology , BNT162 Vaccine/immunology , COVID-19/immunology , COVID-19/prevention & control , Female , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/immunology , Humans , Immunoglobulin M/immunology , Lymphoproliferative Disorders/drug therapy , Lymphoproliferative Disorders/immunology , Male , Middle Aged , Prospective Studies , Seroconversion
7.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34768899

ABSTRACT

Over the last few years, treatment principles have been changed towards more targeted therapy for many B-cell lymphoma subtypes and in chronic lymphocytic leukemia (CLL). Immunotherapeutic modalities, namely monoclonal antibodies (mAbs), bispecific antibodies (bsAbs), antibody-drug conjugates (ADCs), and chimeric antigen receptor T (CAR-T) cell therapy, commonly use B-cell-associated antigens (CD19, CD20, CD22, and CD79b) as one of their targets. T-cell engagers (TCEs), a subclass of bsAbs, work on a similar mechanism as CAR-T cell therapy without the need of previous T-cell manipulation. Currently, several anti-CD20xCD3 TCEs have demonstrated promising efficacy across different lymphoma subtypes with slightly better outcomes in the indolent subset. Anti-CD19xCD3 TCEs are being developed as well but only blinatumomab has been evaluated in clinical trials yet. The results are not so impressive as those with anti-CD19 CAR-T cell therapy. Antibody-drug conjugates targeting different B-cell antigens (CD30, CD79b, CD19) seem to be effective in combination with mAbs, standard chemoimmunotherapy, or immune checkpoint inhibitors. Further investigation will show whether immunotherapy alone or in combinatory regimens has potential to replace chemotherapeutic agents from the first line treatment.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunoconjugates/therapeutic use , Immunotherapy/methods , Lymphoma, B-Cell/drug therapy , Lymphoproliferative Disorders/drug therapy , Animals , Humans , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology
9.
Cancer Med ; 10(21): 7629-7640, 2021 11.
Article in English | MEDLINE | ID: mdl-34558211

ABSTRACT

BACKGROUND: Lymphoid neoplasms treatment has recently been renewed to increase antitumor efficacy and conventional chemotherapies toxicities. Limited data have been published about the infection risk associated with these new drugs, therefore this study analyzes the infectious complications in patients with lymphoproliferative diseases (LPD) treated with monoclonal antibodies (obinutuzumab, ofatumumab, brentuximab, nivolumab, or pembrolizumab), BTK inhibitors (ibrutinib and acalabrutinib), PI3K inhibitors (idelalisib) and BCL2 inhibitors (venetoclax). METHODS: Multicenter retrospective study of 458 LPD patients treated with targeted therapies in real-life setting, in 18 Spanish institutions, from the time of their commercial availability to August 2020. RESULTS: Severe infections incidence was 23% during 17-month median follow-up; cumulative incidence was higher in the first 3-6 months of targeted drug treatment and then decreased. The most frequent etiology was bacterial (54%). Nine (6%) Invasive fungal infections (IFI) were observed, in its majority in chronic lymphocytic leukemia (CLL) patients treated predominantly with ibrutinib. Significant risk factors for severe infection were: severe lymphopenia (p = 0.009, OR 4.7, range 1.3-1.7), combined targeted treatment vs single agent treatment (p = 0.014 OR 2.2 range 1.1-4.2) and previous rituximab (p = 0.03 OR 1.8, range 1.05-3.3). Infection-related mortality was 6%. In 22% of patients with severe infections, definitive discontinuation of the targeted drug was observed. CONCLUSION: A high proportion of patients presented severe infections during follow-up, with non-negligible attributable mortality, but infection incidence is not superior to the one observed during the chemotherapy era. In selected cases with specific risk factors for infection, antimicrobial prophylaxis should be considered.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Immunocompromised Host , Infections/etiology , Lymphoproliferative Disorders/drug therapy , Lymphoproliferative Disorders/immunology , Adenine/adverse effects , Adenine/analogs & derivatives , Adolescent , Adult , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Benzamides/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Female , Humans , Lymphopenia/complications , Lymphoproliferative Disorders/complications , Male , Middle Aged , Piperidines/adverse effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Purines/adverse effects , Pyrazines/adverse effects , Quinazolinones/adverse effects , Retrospective Studies , Risk Factors , Sulfonamides/adverse effects , Young Adult
10.
Immunology ; 164(4): 722-736, 2021 12.
Article in English | MEDLINE | ID: mdl-34534359

ABSTRACT

Bruton's tyrosine kinase (BTK) is a TEC kinase with a multifaceted role in B-cell biology and function, highlighted by its position as a critical component of the B-cell receptor signalling pathway. Due to its role as a therapeutic target in several haematological malignancies including chronic lymphocytic leukaemia, BTK has been gaining tremendous momentum in recent years. Within the immune system, BTK plays a part in numerous pathways and cells beyond B cells (i.e. T cells, macrophages). Not surprisingly, BTK has been elucidated to be a driving factor not only in lymphoproliferative disorders but also in autoimmune diseases and response to infection. To extort this role, BTK inhibitors such as ibrutinib have been developed to target BTK in other diseases. However, due to rising levels of resistance, the urgency to develop new inhibitors with alternative modes of targeting BTK is high. To meet this demand, an expanding list of BTK inhibitors is currently being trialled. In this review, we synopsize recent discoveries regarding BTK and its role within different immune cells and pathways. Additionally, we discuss the broad significance and relevance of BTK for various diseases ranging from haematology and rheumatology to the COVID-19 pandemic. Overall, BTK signalling and its targetable nature have emerged as immensely important for a wide range of clinical applications. The development of novel, more specific and less toxic BTK inhibitors could be revolutionary for a significant number of diseases with yet unmet treatment needs.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , B-Lymphocytes/enzymology , Immune System/enzymology , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/enzymology , Autoimmune Diseases/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , COVID-19/enzymology , COVID-19/immunology , Humans , Immune System/drug effects , Immune System/immunology , Lymphoproliferative Disorders/drug therapy , Lymphoproliferative Disorders/enzymology , Lymphoproliferative Disorders/immunology , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Receptors, Antigen, B-Cell/metabolism , Receptors, Chemokine/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , COVID-19 Drug Treatment
11.
ACS Appl Mater Interfaces ; 13(36): 42370-42381, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34486371

ABSTRACT

Natural killer (NK) cells serve as key innate effectors and their activity has been considered a prognostic biomarker in diverse human diseases. Currently, NK cell functional assays have several problems primarily related to adequate preparation, labeling, or treatment of target cells, which are cumbersome and often hamper consistent sensitivity for NK cells. Here, bispecific antibodies (BsAb's) targeting NKG2D and 2B4 receptors, whose combination mounts selective cytotoxicity and IFN-γ production of NK cells, are developed as acellular, consistent, and easy-to-use strategies for assessing NK cell functions. These NK cell activator BsAb's (NKABs) are constructed in symmetric dual bivalent formats with different interdomain spacings [immunoglobulin G (IgG)-single-chain variable fragment (scFv) and dual-variable domain (DVD)-Ig] and kappa constant (Cκ)-scFv format linking two scFv's with a Cκ domain. These NKABs are specific and superior to a combination of monospecific antibodies for NK cell activation. NKAB elicits both direct cytotoxicity and IFN-γ production via integration of NKG2D and 2B4 signals. Moreover, stimulation with NKAB IgG-scFv and Cκ-scFv reveals defective NK cell functions in X-linked lymphoproliferative disease involving 2B4 dysfunction in NK cells and multiple myeloma in peripheral blood mononuclear cells and whole blood, respectively. Hence, this work provides a proof of concept that NKAB facilitates the reliable and comprehensive measurement of NK cell function in clinical settings for diagnostic and prognostic purposes.


Subject(s)
Antibodies, Bispecific/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Signaling Lymphocytic Activation Molecule Family/immunology , HEK293 Cells , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Lymphoproliferative Disorders/diagnosis , Lymphoproliferative Disorders/immunology , Multiple Myeloma/diagnosis , Multiple Myeloma/immunology , Proof of Concept Study , Single-Chain Antibodies/immunology
13.
Pediatr Transplant ; 25(8): e14111, 2021 12.
Article in English | MEDLINE | ID: mdl-34405495

ABSTRACT

BACKGROUND: CNI-free immunosuppression with conversion to mTORi-based immunosuppression has been demonstrated to reduce CNI-toxicity and to exhibit anti-proliferative properties. However, the experience of CNI-free immunosuppression in paediatric heart transplantation is limited. METHODS: A retrospective analysis was conducted of 129 paediatric heart transplants performed between 1997 and 2015. Fifteen patients with clinically indicated conversion from CNI-based to CNI-free immunosuppression were identified. Survival data, rejection episodes, renal function, post-transplantation lymphoproliferative disorder and CAV, including examination with OCT were analysed. RESULTS: Immunosuppression conversion was successful in all patients. Fourteen of 15 patients (93%) are currently living with good graft function. Median post-transplant survival was 15 years (range, 5-23 years), and median follow-up since conversion was 6 years (range, 1-11 years). Mild (grade 1R) ACR was present in three patients after discontinuation of CNIs. The recovery of renal function with a significant increase in eGFR was observed at 1 and 3 years after conversion. No patient had angiographic signs of macroscopic CAV according to the current ISHLT classification; however, OCT showed the signs of angiographically silent CAV in all patients. CAV did not progress in any patient, implying CAV was stabilised by mTORi-based CNI-free immunosuppression. CONCLUSIONS: CNI-free immunosuppression based on mTORis is a safe and appropriate strategy for maintenance therapy in selected paediatric patients, significantly improves renal function and stabilises CAV. OCT revealed early development of angiographically silent CAV.


Subject(s)
Graft Rejection/diagnostic imaging , Graft Rejection/immunology , Heart Transplantation , Immunosuppression Therapy/methods , Immunosuppressive Agents/therapeutic use , Adolescent , Calcineurin Inhibitors , Child , Child, Preschool , Everolimus/therapeutic use , Female , Glomerular Filtration Rate , Graft Survival , Humans , Lymphoproliferative Disorders/immunology , Male , Retrospective Studies , Sirolimus/therapeutic use , Tomography, Optical Coherence , Young Adult
14.
Am J Clin Pathol ; 156(5): 871-885, 2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34406351

ABSTRACT

OBJECTIVES: Follicular hyperplasias (FHs) with light chain-restricted (LCR) plasmacytoid/plasma cells (PCs) within germinal centers (GCs) based on immunohistochemistry (IHC)/in situ hybridization (ISH) can potentially lead to diagnostic error. This study aims to better characterize such cases, including their clinical implications. METHODS: LC expression by IHC/ISH was quantitatively assessed in GCs of 17 FHs with LCRGCs. BCL2, CD10, BCL6, BCL2, immunoglobulin (Ig) heavy chains, IgG4, and Epstein-Barr encoding region stains were performed. In total, 8 cases had polymerase chain reaction (PCR)-based clonality studies. RESULTS: All cases showed FH, including 4 with progressively transformed GCs (PTGCs); 0.8% to 52% (median, 21%) of the GCs were LCR; 13 of 17 had both κ- and λ-LCRGCs, and 4 of 17 had only κ-LCRGCs; 7 of 16 had prominent intrafollicular IgG4-positive cells. One case demonstrated BCL2-positive cells in focal LCRGCs but lacked BCL2 rearrangement. B-cell monoclonality was demonstrated in 3 of 8 cases (only after microdissection). Seven patients had autoimmune disorders, and 1 had had a transplant. Three patients had a history of lymphoma, 1 developed lymphoma, and 1 developed lymphomatoid granulomatosis subsequently. CONCLUSIONS: FHs with LCRGC by IHC/ISH are typically not associated with the development of lymphoma, even though they can express BCL2 and show monoclonality by PCR. They may be associated with increased intrafollicular IgG4-positive cells, PTGC, and autoimmunity.


Subject(s)
Germinal Center/immunology , Germinal Center/pathology , Immunoglobulin Light Chains/immunology , Plasma Cells/immunology , Adult , Aged , Aged, 80 and over , Diagnosis, Differential , Female , Humans , Hyperplasia/immunology , Hyperplasia/pathology , Lymphoma/diagnosis , Lymphoma/immunology , Lymphoma/pathology , Lymphoproliferative Disorders/diagnosis , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology , Male , Middle Aged , Plasma Cells/pathology , Retrospective Studies
15.
Front Immunol ; 12: 677027, 2021.
Article in English | MEDLINE | ID: mdl-34168649

ABSTRACT

Epstein-Barr virus (EBV) is a human herpesvirus that is common among the global population, causing an enormous disease burden. EBV can directly cause infectious mononucleosis and is also associated with various malignancies and autoimmune diseases. In order to prevent primary infection and subsequent chronic disease, efforts have been made to develop a prophylactic vaccine against EBV in recent years, but there is still no vaccine in clinical use. The outbreak of the COVID-19 pandemic and the global cooperation in vaccine development against SARS-CoV-2 provide insights for next-generation antiviral vaccine design and opportunities for developing an effective prophylactic EBV vaccine. With improvements in antigen selection, vaccine platforms, formulation and evaluation systems, novel vaccines against EBV are expected to elicit dual protection against infection of both B lymphocytes and epithelial cells. This would provide sustainable immunity against EBV-associated malignancies, finally enabling the control of worldwide EBV infection and management of EBV-associated diseases.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/physiology , Lymphoproliferative Disorders/immunology , SARS-CoV-2/physiology , Viral Vaccines/immunology , Animals , Epstein-Barr Virus Infections/prevention & control , Humans , Lymphoproliferative Disorders/prevention & control , Pre-Exposure Prophylaxis
16.
Transplant Proc ; 53(5): 1519-1527, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34134932

ABSTRACT

Posttransplant lymphoproliferative disorder (PTLD) occurs in 1% to 3% of adult renal transplant recipients (RTRs). PTLD has a heterogeneous presentation and is often associated with Epstein-Barr virus (EBV) and immunosuppression. We present a descriptive case series of 16 RTRs who demonstrate a variety of PTLD manifestations. Fifty-six percent received rabbit antithymocyte globulin induction, and 37.5% received basiliximab. Maintenance immunosuppression included glucocorticoids, tacrolimus, and mycophenolate mofetil. Median time from transplantation to PTLD diagnosis was 96.5 months. PTLD involved a single site in 44% of RTRs and multiple sites in 56%. PTLD was localized to the gastrointestinal tract in 9 RTRs, in lymph nodes in 9, central nervous system in 4, bone marrow in 3, skin in 3, lungs in 2, perinephric space in 2, mediastinum in 1, and native kidney in 1. PTLD was EBV positive in 8 RTRs, monomorphic/monoclonal in 14, and of B-cell lineage in 13. Three RTRs had T-cell PTLD. Immunosuppressive agents, except glucocorticoids, were discontinued at diagnosis. Treatment was chemotherapy either alone (in 14 RTRs) or in combination with radiation. Complete remission was achieved in 62.5% of RTRs. Renal dysfunction developed in 62.5% of RTRs, and 4 received dialysis. The overall mortality rate was 62.5%, with median time of death 6.5 months after diagnosis. PTLD that was EBV negative and had T-cell involvement presented with aggressive disease and a higher mortality. Clinicians should be aware of the various PTLD manifestations. Early diagnosis and a multidisciplinary approach to treatment is crucial for improved patient outcomes.


Subject(s)
Immunosuppression Therapy/adverse effects , Kidney Transplantation/adverse effects , Lymphoma/immunology , Lymphoproliferative Disorders/immunology , Postoperative Complications/immunology , Adult , Epstein-Barr Virus Infections/immunology , Female , Herpesvirus 4, Human/immunology , Humans , Immunosuppressive Agents/adverse effects , Male , Mycophenolic Acid/adverse effects , Renal Dialysis , Tacrolimus/adverse effects , Treatment Outcome
17.
Pharm Biol ; 59(1): 741-747, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34155950

ABSTRACT

CONTEXT: Simiao Qingwen Baidu decoction (SQBD), a traditional Chinese medicine prescription, can ameliorate Epstein-Barr virus (EBV) induced disease. However, its mechanism still remains unknown. OBJECTIVE: To detect the mechanism of SQBD in EBV-induced B lymphoproliferative disease in vitro. MATERIALS AND METHODS: Sprague-Dawley (SD) rats (n = 20) were given SQBD (10 mL/kg) by gavage once a day for 7 d. SQBD-containing serum was obtained from abdominal aortic blood of rats, and diluted with medium to obtain 5%, 10% or 20%-medicated serum. SD rats (n = 10) were given normal saline, and normal serum was collected as a control. EBV-transformed B cells (CGM1) were cultured in medium containing 5%, 10% or 20%-medicated serum. CGM1 cells were treated with normal serum as a control. Cell viability and apoptosis were examined. The expression and activity of proteins were assessed. RESULTS: We found that IC50 (83 ± 26.07%, 24 h; 69.88 ± 4.69%, 48 h) of 10% medicated serum was higher than that of 5% (25.47 ± 6.98%, 24 h; 21.62 ± 7.30%, 48 h) and 20%-medicated serum (51 ± 7.25%, 24 h; 56.03 ± 2.56%, 48 h). Moreover, SQBD promoted apoptosis of CGM1 cells by regulating EBV latency proteins expression. SQBD inhibited EBV-induced lytic viral replication. CONCLUSIONS: Our data confirmed that SQBD inhibits EBV-induced B lymphoproliferative disease and lytic viral replication. This work provides a theoretical basis for the mechanism of SQBD in EBV-induced B lymphoproliferative disease, and SQBD may be an effectively therapeutic drug for EBV-induced B lymphoproliferative disease.


Subject(s)
B-Lymphocytes/drug effects , Drugs, Chinese Herbal/therapeutic use , Herpesvirus 4, Human/drug effects , Lymphoproliferative Disorders/drug therapy , Virus Replication/drug effects , Animals , B-Lymphocytes/physiology , Drugs, Chinese Herbal/pharmacology , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/physiology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/metabolism , Male , Rats , Rats, Sprague-Dawley , Virus Replication/physiology
18.
Clin Exp Immunol ; 205(3): 288-305, 2021 09.
Article in English | MEDLINE | ID: mdl-34008169

ABSTRACT

Lymphadenopathies can be part of the clinical spectrum of several primary immunodeficiencies, including diseases with immune dysregulation and autoinflammatory disorders, as the clinical expression of benign polyclonal lymphoproliferation, granulomatous disease or lymphoid malignancy. Lymphadenopathy poses a significant diagnostic dilemma when it represents the first sign of a disorder of the immune system, leading to a consequently delayed diagnosis. Additionally, the finding of lymphadenopathy in a patient with diagnosed immunodeficiency raises the question of the differential diagnosis between benign lymphoproliferation and malignancies. Lymphadenopathies are evidenced in 15-20% of the patients with common variable immunodeficiency, while in other antibody deficiencies the prevalence is lower. They are also evidenced in different combined immunodeficiency disorders, including Omenn syndrome, which presents in the first months of life. Interestingly, in the activated phosphoinositide 3-kinase delta syndrome, autoimmune lymphoproliferative syndrome, Epstein-Barr virus (EBV)-related lymphoproliferative disorders and regulatory T cell disorders, lymphadenopathy is one of the leading signs of the entire clinical picture. Among autoinflammatory diseases, the highest prevalence of lymphadenopathies is observed in patients with periodic fever, aphthous stomatitis, pharyngitis, and cervical adenitis (PFAPA) and hyper-immunoglobulin (Ig)D syndrome. The mechanisms underlying lymphoproliferation in the different disorders of the immune system are multiple and not completely elucidated. The advances in genetic techniques provide the opportunity of identifying new monogenic disorders, allowing genotype-phenotype correlations to be made and to provide adequate follow-up and treatment in the single diseases. In this work, we provide an overview of the most relevant immune disorders associated with lymphadenopathy, focusing on their diagnostic and prognostic implications.


Subject(s)
Common Variable Immunodeficiency/immunology , Immunologic Deficiency Syndromes/immunology , Lymphadenopathy/immunology , Lymphoproliferative Disorders/immunology , Common Variable Immunodeficiency/diagnosis , Common Variable Immunodeficiency/genetics , Common Variable Immunodeficiency/pathology , Epstein-Barr Virus Infections/immunology , Genetic Predisposition to Disease/genetics , Herpesvirus 4, Human/immunology , Humans , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/pathology , Infant , Infant, Newborn , Lymphadenopathy/diagnosis , Lymphadenopathy/pathology , Lymphoproliferative Disorders/diagnosis , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/pathology
20.
Am J Dermatopathol ; 43(12): e234-e236, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-33899771

ABSTRACT

ABSTRACT: Post-transplant lymphoproliferative disorder (PTLD) is a term used to describe a range of lymphoproliferative disorders that occur after solid organ transplant. Although the clinical presentation is variable, primary cutaneous PTLD typically presents as isolated nodules that appear as dermal-based proliferations. We present a case of a 70-year-old woman with a history of a kidney transplant who presented with a 2-month history of an asymptomatic, erythematous plaque on the right shin, clinically suspected to be squamous cell carcinoma in situ. Histomorphology demonstrated a dermal proliferation of atypical plasma cells with dense chromatin, variable nucleoli, and irregular nuclear borders. The atypical plasma cells were positive for Epstein-Barr virus by in situ hybridization and markedly kappa-restricted by RNAscope in situ hybridization. A diagnosis of cutaneous monomorphic PTLD, plasma cell neoplasm variant, was rendered, a rare diagnosis in the skin. Treatment for PTLD typically involves reduction of immunosuppression, although our patient progressed and developed new lesions despite this intervention. In this study, we present an atypical presentation of cutaneous PTLD, plasma cell neoplasm variant, presenting as squamous cell carcinoma in situ.


Subject(s)
Immunocompromised Host , Kidney Transplantation/adverse effects , Lymphoproliferative Disorders/diagnosis , Postoperative Complications/immunology , Skin Diseases/diagnosis , Aged , Carcinoma, Squamous Cell/diagnosis , Diagnosis, Differential , Female , Humans , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology , Postoperative Complications/diagnosis , Postoperative Complications/pathology , Skin Diseases/immunology , Skin Diseases/pathology , Skin Neoplasms/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...