Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
2.
biorxiv; 2023.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2023.03.14.532609

ABSTRACT

Several studies have identified the presence of functionally depleted CD8+ T cells in COVID-19 patients, and particularly abnormally reduced CD8+ T cells in severe/critical patients, which may be a major cause of disease progression and poor prognosis. In this study, a proliferating-depleted CD8+ T cell phenotype was observed in severe/critical COVID-19 patients through scRNA-seq and scTCR-seq analysis. These CD8+ T cells were subsequently found to be characterized by cell cycle arrest and downregulation of mitochondrial biogenesis and respiratory chain complex genes. Cellchat analysis revealed that the Galectin signaling pathways between infected lung epithelial cells and CD8+ T cells play the key role in inducing CD8+ T cell reduction and dysfunction in severe/critical COVID-19. We used SARS-COV-2 ORF3a to transfect A549 epithelial cells, and co-cultured with CD8+ T cells. The ex vivo experiments confirmed that galectin-3 inhibited the transcription of mitochondrial respiratory chain complex III/IV genes in CD8+ T cells by suppressing the nuclear translocation of nuclear respiratory factor 1 (NRF1). In addition, the regulatory effect of galectin-3 was correlated with the activation of ERK signaling and/or the inhibition of Akt signaling. Galectin-3 inhibitor, TD-139, promoted nuclear translocation of NRF1, and enhanced mitochondrial respiratory chain complex III/IV gene expression and mitochondrial biogenesis, then restore the expansion ability of CD8+ T cells. Our study improved the understanding the immunopathogenesis and provided new target for the prevention and treatment of severe/critical COVID-19.


Subject(s)
Lymphoma, T-Cell , COVID-19
3.
medrxiv; 2023.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2023.01.09.23284366

ABSTRACT

Background Dengue which is endemic in India and has been occurring for decades apparently witnessed a rise in disease burden in 2021 in specific regions of the nation. We aim to explore less studied risk factors of Dengue occurrence and severity in the post-COVID-19 and post-COVID-19 vaccination era. Methods This was an exploratory analysis involving participants from two prior observational studies conducted during the period of Feb 2021-April 2022 in a tertiary hospital in North India. Healthcare workers constituted the majority of study participants. Individuals were stratified into five groups based on COVID-19 infection and timing of vaccination: CovidNoVaccine (CNV), VaccineNoCOVID (VNC), CovidAfterVaccine (CAV), VaccineAfterCOVID (VAC) and NoVaccineNoCovid (NVNC) groups. The occurrence of lab-confirmed Dengue and severe forms of Dengue were the main outcomes of interest. We tried to predict determinants of Dengue occurrence and severity with a particular focus on COVID-19 history and vaccination status. Results A total of 1520 vaccinated individuals and 181 unvaccinated individuals were included. Of these 1701 participants, symptomatic Dengue occurred in 133 (7.8%) and was of 'severe' category in 42 (31.6%). Individuals with a history of COVID-19 in 2020 had 2 times higher odds of developing symptomatic Dengue. The VAC group had 3.6, 2- and 1.9 times higher odds of developing Dengue than the NVNC, VNC, and CAV groups. The severity of dengue was not affected by COVID-19 or COVID-19 vaccination. Conclusions COVID-19 may enhance the risk of developing symptomatic dengue. Future research dealing with long COVID should explore the propensity of COVID-19 victims towards symptomatic forms of other viral illnesses. Individuals receiving the COVID-19 vaccine after recovering from COVID-19 particularly seem to be at greater risk of symptomatic dengue and need long-term watchfulness. Possible mechanisms, such as antibody-mediated enhancement or T-cell dysfunction, should be investigated in COVID-19-recovered and vaccinated individuals. Further large-scale, multicentric, robust studies with a better enrolment of unvaccinated people will help understand the interplay of factors involved in COVID-19 and Dengue.


Subject(s)
COVID-19 , Lymphoma, T-Cell
4.
PLoS One ; 17(12): e0266584, 2022.
Article in English | MEDLINE | ID: covidwho-2140382

ABSTRACT

Patients with hematologic malignancies have poor outcomes from COVID infection and are less likely to mount an antibody response after COVID infection. This is a retrospective study of adult lymphoma patients who received the COVID vaccine between 12/1/2020 and 11/30/2021. The primary endpoint was a positive anti-COVID spike protein antibody level following the primary COVID vaccination series. The primary vaccination series was defined as 2 doses of the COVID mRNA vaccines or 1 dose of the COVID adenovirus vaccine. Subgroups were compared using Fisher's exact test, and unadjusted and adjusted logistic regression models were used for univariate and multivariate analyses. A total of 243 patients were included in this study; 72 patients (30%) with indolent lymphomas; 56 patients (23%) with Burkitt's, diffuse large B-cell lymphoma (DLBCL), and primary mediastinal B-cell lymphoma (PMBL) combined; 55 patients (22%) with chronic lymphocytic leukemia or small lymphocytic lymphoma (CLL/SLL); 44 patients (18%) with Hodgkin and T-cell lymphomas (HL/TCL) combined; 12 patients (5%) with mantle cell lymphoma; and 4 patients (2%) with other lymphoma types. One-hundred fifty-eight patients (65%) developed anti-COVID spike protein antibodies after completing the primary COVID vaccination series. Thirty-eight of 46 (83%) patients who received an additional primary shot and had resultant levels produced anti-COVID spike protein antibodies. When compared to other lymphoma types, patients with CLL/SLL had a numerically lower seroconversion rate of 51% following the primary vaccination series whereas patients with HL/TCL appeared to have a robust antibody response with a seropositivity rate of 77% (p = 0.04). Lymphoma patients are capable of mounting a humoral response to the COVID vaccines. Further studies are required to confirm our findings, including whether T-cell immunity would be of clinical relevance in this patient population.


Subject(s)
COVID-19 Vaccines , COVID-19 , Leukemia, Lymphocytic, Chronic, B-Cell , Lymphoma, Large B-Cell, Diffuse , Lymphoma, T-Cell , Adult , Humans , Antibodies , Antibody Formation , COVID-19/prevention & control , COVID-19 Vaccines/immunology , Retrospective Studies , Spike Glycoprotein, Coronavirus/immunology
5.
Int J Mol Sci ; 23(16)2022 Aug 19.
Article in English | MEDLINE | ID: covidwho-1997646

ABSTRACT

Follicular dendritic cell (FDC) proliferation in angioimmunoblastic T-cell lymphoma (AITL) is still not well defined, challenging the accurate differential diagnosis between the AITL with expanded follicular dendritic cell meshwork and the combined AITL and follicular dendritic cell sarcoma (FDCS). Herein, we reported the case of a 58-year-old male with coexisting SARS-CoV-2 infection and AITL with an exuberant CD30-positive FDC proliferation, in which genetic analysis identified mutations of genes commonly involved in AITL but not in FDC sarcoma (i.e., RHOA, TET2, DNMT3A, and IDH2), thus supporting the reactive nature of the CD30-positive FDC expansion.


Subject(s)
COVID-19 , Dendritic Cell Sarcoma, Follicular , Immunoblastic Lymphadenopathy , Lymphoma, T-Cell , Cell Proliferation , Dendritic Cell Sarcoma, Follicular/diagnosis , Dendritic Cell Sarcoma, Follicular/genetics , Dendritic Cell Sarcoma, Follicular/pathology , Dendritic Cells, Follicular/pathology , Humans , Immunoblastic Lymphadenopathy/diagnosis , Immunoblastic Lymphadenopathy/genetics , Immunoblastic Lymphadenopathy/pathology , Ki-1 Antigen/genetics , Lymphoma, T-Cell/diagnosis , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/pathology , Male , Middle Aged , SARS-CoV-2
6.
biorxiv; 2022.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2022.06.24.497299

ABSTRACT

Palatine tonsils are secondary lymphoid organs representing the first line of immunological defense against inhaled or ingested pathogens. Here, we present a comprehensive census of cell types forming the human tonsil by applying single-cell transcriptome, epigenome, proteome and adaptive immune repertoire sequencing as well as spatial transcriptomics, resulting in an atlas of >357,000 cells. We provide a glossary of 121 annotated cell types and states, and disentangle gene regulatory mechanisms that drive cells through specialized lineage trajectories. Exemplarily, we stratify multiple tonsil-resident myeloid slancyte subtypes, establish a distant BCL6 superenhancer as locally active in both follicle-associated T and B cells, and describe SIX5 as a potentially novel transcriptional regulator of plasma cell maturation. Further, our atlas is a reference map to understand alterations observed in disease. Here, we discover immune-phenotype plasticity in tumoral cells and microenvironment shifts of mantle cell lymphomas (MCL). To facilitate such reference-based analysis, we develop HCATonsilData and SLOcatoR, a computational framework that provides programmatic and modular access to our dataset; and allows the straightforward annotation of future single-cell profiles from secondary lymphoid organs.


Subject(s)
Lymphoma, T-Cell , Neoplasms , Lymphoma, Mantle-Cell
7.
Dermatol Online J ; 27(12)2021 Dec 15.
Article in English | MEDLINE | ID: covidwho-1789588

ABSTRACT

Primary cutaneous diffuse large B-cell lymphoma, leg type is a rare entity accounting for 4% of all primary cutaneous lymphomas whose clinical presentation encompasses a range of possibilities. COVID-19 has caused a delay in diagnosis of malignant neoplasms and consequently, this has resulted in poorer prognoses. A 62-year-old woman presented with two smooth-surfaced, mobile, well-circumscribed, oval, skin-colored nodules approximately one-cm in diameter with nonerythematous borders on the lower third of the left leg. Two months later, eleven nodules measuring between one and 1.5cm with erythematous halo, slight scaling, central erosion, and crusting had appeared. Histological study showed moderate pericapillary lymphocytic infiltration in the papillary and reticular dermis and prominent diffuse proliferation of medium to large cells in the subcutis. These exhibited irregular vesicular nuclei, a conspicuous solitary nucleolus of two to three small nucleoli, and three mitoses per high power field. Adipocytes were consistently encircled by neoplastic lymphocytes. Primary cutaneous diffuse large B-cell lymphoma, leg type is a high-grade lymphoma that can manifest as a diagnostic challenge and requires adequate immunohistochemistry and in situ hybridization studies for proper diagnosis, treatment, and prognosis.


Subject(s)
COVID-19 , Lymphoma, Large B-Cell, Diffuse , Panniculitis , Skin Neoplasms , COVID-19/diagnosis , Female , Humans , Leg/pathology , Lymphoma, Large B-Cell, Diffuse/diagnosis , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, T-Cell , Middle Aged , Panniculitis/diagnosis , Panniculitis/pathology , Skin Neoplasms/diagnosis , Skin Neoplasms/pathology
8.
biorxiv; 2022.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2022.04.07.487556

ABSTRACT

Pathogenic infections cause thymic atrophy, perturb thymic-T cell development and alter immunological response. Previous studies reported dysregulated T cell function and lymphopenia in coronavirus disease-19 (COVID-19) patients. However, immune-pathological changes, in the thymus, post severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection have not been elucidated. Here, we report SARS-CoV-2 infects thymocytes, depletes CD4+CD8+ (double positive DP) T cell population associated with an increased apoptosis of thymocytes, which leads to severe thymic atrophy in K18-hACE2-Tg mice. CD44+CD25- T cells were found to be enriched in infected thymus, indicating an early arrest in the T cell developmental pathway. Further, Interferon gamma was crucial for thymic atrophy, as anti-IFN{gamma}; antibody neutralization rescued the loss of thymic involution. Therapeutic use of remdesivir (prototype anti-viral drug) was also able to rescue thymic atrophy. While Omicron variant of SARS-CoV2 caused marginal thymic atrophy, delta variant of SARS-CoV-2 exhibited most profound thymic atrophy characterized by severely depleted DP T cells. Recently characterized broadly SARS-CoV-2 neutralizing monoclonal antibody P4A2 was able to rescue thymic atrophy and restore thymic developmental pathway of T cells. Together, we provide the first report of SARS-CoV-2 associated thymic atrophy resulting from impaired T cell developmental pathway and also explains dysregulated T cell function in COVID-19.


Subject(s)
Lymphoma, T-Cell , Coronavirus Infections , Severe Acute Respiratory Syndrome , Chronobiology Disorders , COVID-19 , Atrophy , Lymphopenia
9.
medrxiv; 2022.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2022.03.24.22272883

ABSTRACT

Patients with hematologic malignancies have poor outcomes from COVID infection and are less likely to mount an antibody response after COVID infection. There is limited data on the efficacy of the COVID vaccines in lymphoma patients, and to suggest the optimal timing of vaccination to elicit immunity in patients receiving immunochemotherapy. This is a retrospective study of adult lymphoma patients who received the COVID vaccine between 12/1/2020 and 11/30/2021. The primary endpoint was a positive anti-COVID spike protein antibody titer following the primary COVID vaccination series. The primary series was defined as 2 doses of the COVID mRNA vaccines or 1 dose of the COVID adenovirus vaccine. Subgroups were compared using Fisher’s exact test, and unadjusted and adjusted logistic regression models were used for univariate (UVA) and multivariate (MVA) analyses. A total of 243 patients were included in this study; 72 patients (30%) with indolent lymphomas; 56 patients (23%) with Burkitt’s, diffuse large B-cell lymphoma (DLBCL), and primary mediastinal B-cell lymphoma (PMBL) combined; 55 patients (22%) with chronic lymphocytic leukemia or small lymphocytic lymphoma (CLL/SLL); and 44 patients (18%) with Hodgkin and T-cell lymphomas (HL/TCL) combined. One-hundred fifty-eight patients (65%) developed anti-COVID spike protein antibodies after completing the primary COVID vaccination series. Thirty-eight of 46 (83%) patients who received an additional primary shot and had resultant levels produced anti-COVID spike protein antibodies. When compared to other lymphoma types, patients with CLL/SLL had a numerically lower seroconversion rate of 51% following the primary series whereas patients with HL/TCL appeared to have a robust antibody response with a seropositivity rate of 77% (p=0.04). Lymphoma patients are capable of mounting a humoral response to the COVID mRNA vaccines. Further studies are required to confirm our findings, including whether T-cell immunity would be of clinical relevance in this patient population.


Subject(s)
Lymphoma, T-Cell , Lymphoma, B-Cell , Mastocytosis, Systemic , Lymphoma , Leukemia, Lymphocytic, Chronic, B-Cell , Hematologic Neoplasms , Hodgkin Disease , Adenoviridae Infections
10.
biorxiv; 2022.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2022.03.18.484954

ABSTRACT

To provide a unique global view of the relative potential for evasion of CD8+ and CD4+ T cells by SARS-CoV-2 lineages as they evolve over time, we performed a comprehensive analysis of predicted HLA-I and HLA-II binding peptides in spike (S) and nucleocapsid (N) protein sequences of all available SARS-CoV-2 genomes as provided by NIH NCBI at a bi-monthly interval between March and December of 2021. A data supplement of all B.1.1.529 (Omicron) genomes from GISAID in early December was also used to capture the rapidly spreading variant. A key finding is that throughout continued viral evolution and increasing rates of mutations occurring at T-cell epitope hotspots, protein instances with worst case binding loss did not become the most frequent for any Variant of Concern (VOC) or Variant of Interest (VOI) lineage; suggesting T-cell evasion is not likely to be a dominant evolutionary pressure on SARS-CoV-2. We also determined that throughout the course of the pandemic in 2021, there remained a relatively steady ratio of viral variants that exhibit conservation of epitopes in the N protein, despite significant potential for epitope loss in S relative to other lineages. We further localized conserved regions in N with high epitope yield potential, and illustrated HLA-I binding heterogeneity across the S protein consistent with empirical observations. Although Omicron’s high volume of mutations caused it to exhibit more epitope loss potential than most frequently observed versions of proteins in almost all other VOCs, epitope candidates across its most frequent N proteins were still largely conserved. This analysis adds to the body of evidence suggesting that N may have merit as an additional antigen to elicit immune responses to vaccination with increased potential to provide sustained protection against COVID-19 disease in the face of emerging variants.


Subject(s)
Lymphoma, T-Cell , COVID-19
11.
researchsquare; 2021.
Preprint in English | PREPRINT-RESEARCHSQUARE | ID: ppzbmed-10.21203.rs.3.rs-1083825.v1

ABSTRACT

The mechanisms explaining progression to severe COVID-19 remain poorly understood. It has been proposed that immune system dysregulation/over-stimulation may be implicated, but it is not clear how such processes would lead to respiratory failure. We performed comprehensive multiparameter immune monitoring in a tightly controlled cohort of 128 COVID-19 patients, and used the ratio of oxygen saturation to fraction of inspired oxygen (SpO2 / FiO2) as a physiologic measure of disease severity. Machine learning algorithms integrating 139 parameters identified IL-6 and CCL2 as two factors predictive of severe disease, consistent with the therapeutic benefit observed with anti-IL6-R antibody treatment. However, transcripts encoding these cytokines were not detected among circulating immune cells. Rather, in situ analysis of lung specimens using RNAscope and immunofluorescent staining revealed that elevated IL-6 and CCL2 were dominantly produced by infected lung type II pneumocytes. Severe disease was not associated with higher viral load, deficient antibody responses, or dysfunctional T cell responses. These results refine our understanding of severe COVID-19 pathophysiology, indicating that aberrant cytokine production by infected lung epithelial cells is a major driver of immunopathology. We propose that these factors cause local immune regulation towards the benefit of the virus.


Subject(s)
Lymphoma, T-Cell , Lung Diseases , Immune System Diseases , COVID-19 , Respiratory Insufficiency
12.
ssrn; 2021.
Preprint in English | PREPRINT-SSRN | ID: ppzbmed-10.2139.ssrn.3962440

ABSTRACT

By largely unknown mechanisms, dysregulated gene-specific translation directly contributes to chronic inflammation-associated diseases such as sepsis and ARDS. Here, we report that G9a, a histone methyltransferase and well-regarded transcriptional repressor, non-canonically or non-epigenetically activates translation of select antimicrobial genes to promote proliferation of cytokine producing macrophages and to impair T cell function; all hallmarks of endotoxin-tolerance related complications including sepsis, ARDS and COVID19. Mechanistically, G9a interacts with translation regulators including METTL3, an N6-methyladenosine or m6A RNA methyltransferase, and methylates it to cooperatively upregulate the translation of certain m6A-modified mRNAs that encode immune checkpoint and anti-inflammatory proteins. Further, translatome proteomic analysis of ET macrophages progressively treated by a G9a inhibitor identified proteins showing G9a-dependent translation that unite the networks associated with hyperinflammation and T cell dysfunction. Overall, we identified a previously unrecognized function of G9a in gene-specific translation that can be leveraged to treat ET-related chronic inflammatory diseases.


Subject(s)
COVID-19 , Lymphoma, T-Cell
13.
ssrn; 2021.
Preprint in English | PREPRINT-SSRN | ID: ppzbmed-10.2139.ssrn.3945001

ABSTRACT

Since nucleoside-modified mRNA vaccines strongly activate T follicular helper cells, it is important to explore the possible impact of approved SARS-CoV-2 mRNA vaccines on neoplasms affecting this cell type. Herein, we report and discuss unexpected rapid progression of lymphomatous lesions after administration of a BNT162b2 mRNA vaccine booster in a man recently diagnosed with AITL.


Subject(s)
Lymphoma, T-Cell , Thyroiditis, Autoimmune , Neoplasms
14.
researchsquare; 2021.
Preprint in English | PREPRINT-RESEARCHSQUARE | ID: ppzbmed-10.21203.rs.3.rs-820910.v1

ABSTRACT

T-cell immunity is central for the control of viral infections. CoVac-1 is a peptide-based vaccine candidate, composed of SARS-CoV-2 T-cell epitopes derived from various viral proteins, combined with the toll-like receptor 1/2 agonist XS15 emulsified in Montanide TM ISA51 VG, aiming to induce superior SARS-CoV-2 T-cell immunity to combat COVID-19. We conducted a Phase I open-label trial, including 36 participants aged 18 to 80 years, who received one single subcutaneous CoVAC-1 vaccination. The primary endpoint was safety analyzed until day 56. Immunogenicity in terms of CoVac-1-induced T-cell response was analyzed as main secondary endpoint until day 28. No serious adverse events and no grade 4 adverse events were observed. Expected local granuloma formation was observed in all study subjects, while systemic reactogenicity was absent or mild. SARS-CoV-2-specific T-cell responses targeting multiple vaccine peptides were induced in all study participants, mediated by multifunctional T-helper 1 CD4 + and CD8 + T cells. CoVac-1-induced interferon-γ T-cell responses by far surpassed those detected in COVID-19 convalescents and were unaffected by current SARS-CoV-2 variants of concern (VOC). Together, CoVac-1 showed a favorable safety profile and induced broad, potent, and VOC-independent T-cell responses, supporting the presently ongoing evaluation in a Phase II trial for patients with B-cell/antibody deficiency. Funded by the Ministry of Science, Research and the Arts Baden-Württemberg, Germany; ClinicalTrials.gov number, NCT04546841.


Subject(s)
COVID-19 , Granuloma , Immunologic Deficiency Syndromes , Lymphoma, T-Cell
15.
Blood ; 138(9): 811-814, 2021 09 02.
Article in English | MEDLINE | ID: covidwho-1288619
16.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.04.02.438262

ABSTRACT

Severe COVID-19 has been associated with T cell lymphopenia 1,2, but no causal effect of T cell deficiency on disease severity has been established. To investigate the specific role of T cells in recovery from SARS-CoV-2 infections we studied rhesus macaques that were depleted of either CD4+, CD8+ or both T cell subsets prior to infection. Peak virus loads were similar in all groups, but the resolution of virus in the T cell-depleted animals was slightly delayed compared to controls. The T cell-depleted groups developed virus-neutralizing antibody responses and also class-switched to IgG. When re-infected six weeks later, the T cell-depleted animals showed anamnestic immune responses characterized by rapid induction of high-titer virus-neutralizing antibodies, faster control of virus loads and reduced clinical signs. These results indicate that while T cells play a role in the recovery of rhesus macaques from acute SARS-CoV-2 infections, their depletion does not induce severe disease, and T cells do not account for the natural resistance of rhesus macaques to severe COVID-19. Neither primed CD4+ or CD8+ T cells appeared critical for immunoglobulin class switching, the development of immunological memory or protection from a second infection.


Subject(s)
Lymphoma, T-Cell , Severe Acute Respiratory Syndrome , Immune System Diseases , COVID-19 , Lymphopenia
17.
Eur J Haematol ; 106(5): 654-661, 2021 May.
Article in English | MEDLINE | ID: covidwho-1057986

ABSTRACT

INTRODUCTION: Hemophagocytic lymphohistiocytosis (HLH) is a cytokine storm syndrome associated with mortality rates of up to 88%. Standard therapy with high-dose glucocorticoids and etoposide used in adults is extrapolated from pediatric trials, with significant toxicity in older patients and those with poor performance status. The JAK1/2 inhibitor ruxolitinib has recently gained attention as a treatment option for HLH due to its broad cytokine-modulating abilities and safety profile. Herein we report our center's experience using ruxolitinib in the treatment of adult-onset secondary HLH. CASE SERIES: We report four patients with profound secondary HLH provoked by diverse triggers, including invasive pulmonary aspergillosis on background systemic lupus erythematosus, disseminated tuberculosis, and T-cell lymphoma treated with ruxolitinib as monotherapy or combination therapy in upfront and salvage settings. RESULTS: All four patients had rapid, sustained improvement in clinical status, inflammatory markers, and hematological cell counts followed by durable remission. Three patients developed manageable infectious complications postruxolitinib. CONCLUSIONS: This series demonstrates the effective use of JAK inhibition with ruxolitinib to control pathological immune activation in critically ill patients with secondary HLH and otherwise limited therapeutic options. JAK inhibition is also an area of urgent investigation for the treatment of cytokine storm associated with COVID-19.


Subject(s)
Janus Kinases/antagonists & inhibitors , Lymphohistiocytosis, Hemophagocytic/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Adult , COVID-19/complications , Combined Modality Therapy , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/etiology , Cytokines/blood , Female , Humans , Invasive Pulmonary Aspergillosis/complications , Lupus Erythematosus, Systemic/complications , Lymphohistiocytosis, Hemophagocytic/etiology , Lymphoma, T-Cell/complications , Male , Middle Aged , Nitriles , Pyrimidines , SARS-CoV-2 , Salvage Therapy , Tuberculosis/complications , Young Adult
18.
preprints.org; 2021.
Preprint in English | PREPRINT-PREPRINTS.ORG | ID: ppzbmed-10.20944.preprints202101.0249.v1

ABSTRACT

Background: The t (2; 5) chromosomal rearrangement and resulting nucleophosmin (NPM1) -ALK fusion was first observed in 1994 in anaplastic large cell lymphoma (ALCL), a T-cell lymphoma responsive to cyclophosphamide, abriblastine, vincristine and prednisone in approximately 80% of cases; refractory cases usually respond favorably to brentuximab-vedotin. These treatments are regarded as a bridge to allogeneic hematopoietic stem cell transplantation (allo-SCT). Nowadays, transplant procedures and monitoring of chemotherapy patients proceed very slowly because the SARS-CoV-2 pandemic has heavily clogged the hospitals in all countries. Results: A 40-year-old Caucasian woman was first seen at our clinical center in June 2020. She had ALCL ALK +, a history of failure to two previous therapeutic lines and was in complete remission after 12 courses of Brentuximab, still pending allo-SCT after two failed donor selection. Facing of a new therapeutic failure, we requested the Italian drug regulatory agency, and obtained the authorization, to administer 250 mg twice a day of Crizotinib, a drug incomprehensibly not registered for ALCL ALK +. Conclusions: The response to Crizotinib was optimal, since no adverse event occurred, and CT-PET persisted negative; this drug has proved to be a valid bridge to allo-SCT


Subject(s)
Lymphoma, T-Cell , Lymphoma, Large-Cell, Anaplastic , Lymphoma , COVID-19
20.
ssrn; 2020.
Preprint in English | PREPRINT-SSRN | ID: ppzbmed-10.2139.ssrn.3682253

ABSTRACT

T cells play an essential role in COVID-19, but characteristics of beneficial clones remain unclear. By analysis of clonal trajectories, we identified recovery-associated expanding T cell clones many of which carrying public T cell receptors (TCRs). Mining the immune repertoires of unrelated COVID-19 cases for these sequences revealed T cells with exact TCR complementarity-determining region 3 identity that were more abundant in recovering than in fatal cases. These TCRs were also found in subjects not previously exposed to the virus, with lower representation in repertoires from risk groups like aged individuals or cancer patients. Together, our data indicate that a significant part of the recovery-associated T cell response in COVID-19 may be mediated by public TCRs that are present in repertoires of unexposed individuals. The lower representation of these clones in repertoires of risk groups or failure to expand such clones may contribute to more unfavorable clinical COVID-19 courses.Funding: This project was partially funded by the CRC 841 of the German Research Foundation (toMB) as well as by the Martin-Luther-University Halle (Saale).Conflict of Interest: The authors declare no competing interests.Ethical Approval: Blood collection of COVID-19 patients was performed under institutional review board approvals number 2020-039 and 11/17. Blood collection of pre-pandemic samples was performed under approval of ethics committee of the medical association Hamburg (project number PV4767) and the ethics committee of the medical faculty of MartinLuther-University Halle-Wittenberg (project number 2014-75). Written informed consent was received from all participants. The study has been performed in accordance with the declaration of Helsinki of 1975.


Subject(s)
Lymphoma, T-Cell , COVID-19 , Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL