Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
Front Immunol ; 15: 1326757, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38390330

RESUMEN

Despite significant advances in our knowledge regarding the genetics and molecular biology of gliomas over the past two decades and hundreds of clinical trials, no effective therapeutic approach has been identified for adult patients with newly diagnosed glioblastoma, and overall survival remains dismal. Great hopes are now placed on combination immunotherapy. In clinical trials, immunotherapeutics are generally tested after standard therapy (radiation, temozolomide, and steroid dexamethasone) or concurrently with temozolomide and/or steroids. Only a minor subset of patients with progressive/recurrent glioblastoma have benefited from immunotherapies. In this review, we comprehensively discuss standard therapy-related systemic immunosuppression and lymphopenia, their prognostic significance, and the implications for immunotherapy/oncolytic virotherapy. The effectiveness of immunotherapy and oncolytic virotherapy (viro-immunotherapy) critically depends on the activity of the host immune cells. The absolute counts, ratios, and functional states of different circulating and tumor-infiltrating immune cell subsets determine the net immune fitness of patients with cancer and may have various effects on tumor progression, therapeutic response, and survival outcomes. Although different immunosuppressive mechanisms operate in patients with glioblastoma/gliomas at presentation, the immunological competence of patients may be significantly compromised by standard therapy, exacerbating tumor-related systemic immunosuppression. Standard therapy affects diverse immune cell subsets, including dendritic, CD4+, CD8+, natural killer (NK), NKT, macrophage, neutrophil, and myeloid-derived suppressor cell (MDSC). Systemic immunosuppression and lymphopenia limit the immune system's ability to target glioblastoma. Changes in the standard therapy are required to increase the success of immunotherapies. Steroid use, high neutrophil-to-lymphocyte ratio (NLR), and low post-treatment total lymphocyte count (TLC) are significant prognostic factors for shorter survival in patients with glioblastoma in retrospective studies; however, these clinically relevant variables are rarely reported and correlated with response and survival in immunotherapy studies (e.g., immune checkpoint inhibitors, vaccines, and oncolytic viruses). Our analysis should help in the development of a more rational clinical trial design and decision-making regarding the treatment to potentially improve the efficacy of immunotherapy or oncolytic virotherapy.


Asunto(s)
Glioblastoma , Glioma , Linfopenia , Viroterapia Oncolítica , Adulto , Humanos , Viroterapia Oncolítica/efectos adversos , Glioblastoma/patología , Pronóstico , Temozolomida/uso terapéutico , Estudios Retrospectivos , Inmunoterapia/efectos adversos , Terapia de Inmunosupresión , Glioma/terapia , Esteroides/uso terapéutico , Linfopenia/terapia
2.
Cytotherapy ; 26(1): 25-35, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37897472

RESUMEN

BACKGROUND AIMS: There are currently no effective anti-viral treatments for coronavirus disease 2019 (COVID-19)-hospitalized patients with hypoxemia. Lymphopenia is a biomarker of disease severity usually present in patients who are hospitalized. Approaches to increasing lymphocytes exerting an anti-viral effect must be considered to treat these patients. Following our phase 1 study, we performed a phase 2 randomized multicenter clinical trial in which we evaluated the efficacy of the infusion of allogeneic off-the-shelf CD45RA- memory T cells containing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cells from convalescent donors plus the standard of care (SoC) versus just the SoC treatment. METHODS: Eighty-four patients were enrolled in three Spanish centers. The patients were randomized into the infusion of 1 × 106/kg CD45RA- memory T cells or the SoC. We selected four unvaccinated donors based on the expression of interferon gamma SARS-CoV-2-specific response within the CD45RA- memory T cells and the most frequent human leukocyte antigen typing in the Spanish population. RESULTS: We analyzed data from 81 patients. The primary outcome for recovery, defined as the proportion of participants in each group with normalization of fever, oxygen saturation sustained for at least 24 hours and lymphopenia recovery through day 14 or at discharge, was met for the experimental arm. We also observed faster lymphocyte recovery in the experimental group. We did not observe any treatment-related adverse events. CONCLUSIONS: Adoptive cell therapy with off-the-shelf CD45RA- memory T cells containing SAR-CoV-2-specific T cells is safe, effective and accelerates lymphocyte recovery of patients with COVID-19 pneumonia and/or lymphopenia. TRIAL REGISTRATION: NCT04578210.


Asunto(s)
COVID-19 , Linfopenia , Humanos , SARS-CoV-2 , COVID-19/terapia , Células T de Memoria , Resultado del Tratamiento , Linfopenia/terapia , Antivirales
3.
J Exp Clin Cancer Res ; 42(1): 50, 2023 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-36814272

RESUMEN

Leucocyte subpopulations in both lymphoid and myeloid lineages have a significant impact on antitumor immune response. While radiation-induced lymphopenia is being studied extensively, radiation effects on lymphoid and myeloid subtypes have been relatively less addressed. Interactions between leucocyte subpopulations, their specific radiation sensitivity and the specific kinetics of each subpopulation can be modeled based on both experimental data and knowledge of physiological leucocyte depletion, production, proliferation, maturation and homeostasis. Modeling approaches of the leucocyte kinetics that may be used to unravel mechanisms underlying radiation induced-leucopenia and prediction of changes in cell counts and compositions after irradiation are presented in this review. The approaches described open up new possibilities for determining the influence of irradiation parameters both on a single-time point of acute effects and the subsequent recovery of leukocyte subpopulations. Utilization of these approaches to model kinetic data in post-radiotherapy states may be a useful tool for further development of new treatment strategies or for the combination of radiotherapy and immunotherapy.


Asunto(s)
Leucocitos , Linfopenia , Humanos , Cinética , Leucocitos/fisiología , Linfopenia/terapia
4.
Bratisl Lek Listy ; 124(1): 70-73, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36519611

RESUMEN

The hematological toxicity associated with radiotherapy is represented by neutropenia, anemia, thrombocytopenia, being associated with the increased risk of infection with opportunists, with fatigue and intolerance to effort, but also with the increased risk of bleeding. In the context of the preclinical and clinical results that mention the synergistic effect of the immunotherapy-radiotherapy association, radiation-induced lymphopenia (RIL) becomes an immunosuppression factor, a factor that would tip the fragile antitumor immunopotentiation-immunosuppression balance in favor of the immunosuppressive effect. Both the number of lymphocytes and the neutrophil/lymphocyte ratio (NLR) are prognostic and predictive biomarkers, providing information on the immune status of the host and on a possible response of the tumor to immunotherapy. Modern radiation techniques can increase the risk of lymphopenia by irradiating large volumes of tissue with low doses of radiation. In this context, a redefinition of the dose-volume constraints and the definition of bone marrow, lymphoid organs and lymph nodes not involved in tumors as organs at risk (OARs) is strictly necessary in the case of using irradiation through intensity-modulated radiation therapy (IMRT) techniques or volumetric modulated arc therapy (VMAT) for solid tumors that benefit from immune checkpoint inhibitor (ICI) therapy (Ref. 22). Text in PDF www.elis.sk Keywords: lymphopenia, immunotherapy, radiotherapy, toxicity, spleen, nodes irradiation.


Asunto(s)
Anemia , Linfopenia , Trombocitopenia , Humanos , Planificación de la Radioterapia Asistida por Computador/efectos adversos , Planificación de la Radioterapia Asistida por Computador/métodos , Linfopenia/etiología , Linfopenia/terapia , Anemia/complicaciones , Inmunoterapia , Trombocitopenia/complicaciones , Dosificación Radioterapéutica
5.
Ann Hematol ; 101(10): 2337-2345, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35836007

RESUMEN

Patients with hematological malignancies (HMs) are at a higher risk of developing severe form and protracted course of COVID-19 disease. We investigated whether the combination of viral replication inhibition with remdesivir and administration of anti-SARS-CoV-2 immunoglobulins with convalescent plasma (CP) therapy might be sufficient to treat B-cell-depleted patients with COVID-19. We enrolled 20 consecutive patients with various HMs with profound B-cell lymphopenia and COVID-19 pneumonia between December 2020 and May 2021. All patients demonstrated undetectable baseline anti-SARS-CoV-2 immunoglobulin levels before CP. Each patient received at least a complete course of remdesivir and at least one unit of CP. Previous anti-CD20 therapy resulted in a more prolonged SARS-CoV-2 PCR positivity compared to other causes of B-cell lymphopenia (p = 0.004). Timing of CP therapy showed a significant impact on the clinical outcome. Simultaneous use of remdesivir and CP reduced time period for oxygen weaning after diagnosis (p = 0.017), length of hospital stay (p = 0.007), and PCR positivity (p = 0.012) compared to patients who received remdesivir and CP consecutively. In addition, time from the diagnosis to CP therapy affected the length of oxygen dependency (p < 0.001) and hospital stay (p < 0.0001). In those cases where there were at least 10 days from the diagnosis to plasma administration, oxygen dependency was prolonged vs. patients with shorter interval (p = 0.006). In conclusion, the combination of inhibition of viral replication with passive immunization was proved to be efficient and safe. Our results suggest the clear benefit of early, combined administration of remdesivir and CP to avoid protracted COVID-19 disease among patients with HMs and B-cell lymphopenia.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Neoplasias Hematológicas , Linfopenia , Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , COVID-19/terapia , Neoplasias Hematológicas/complicaciones , Neoplasias Hematológicas/terapia , Humanos , Inmunización Pasiva/métodos , Linfopenia/etiología , Linfopenia/terapia , Oxígeno , SARS-CoV-2 , Sueroterapia para COVID-19
6.
Trials ; 22(1): 674, 2021 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-34600562

RESUMEN

BACKGROUND: Moderate/severe cases of COVID-19 present a dysregulated immune system with T cell lymphopenia and a hyper-inflammatory state. This is a study protocol of an open-label, multi-center, double-arm, randomized, dose-finding phase I/II clinical trial to evaluate the safety, tolerability, alloreactivity, and efficacy of the administration of allogeneic memory T cells and natural killer (NK) cells in COVID-19 patients with lymphopenia and/or pneumonia. The aim of the study is to determine the safety and the efficacy of the recommended phase 2 dose (RP2D) of this treatment for patients with moderate/severe COVID-19. METHODS: In the phase I trial, 18 patients with COVID-19-related pneumonia and/or lymphopenia with no oxygen requirement or with an oxygen need of ≤ 2.5 liters per minute (lpm) in nasal cannula will be assigned to two arms, based on the biology of the donor and the patient. Treatment of arm A consists of the administration of escalating doses of memory T cells, plus standard of care (SoC). Treatment of arm B consists of the administration of escalating doses of NK cells, plus SoC. In the phase II trial, a total of 182 patients with COVID-19-related pneumonia and/or lymphopenia requiring or not oxygen supplementation but without mechanical ventilation will be allocated to arm A or B, considering HLA typing. Within each arm, they will be randomized in a 1:1 ratio. In arm A, patients will receive SoC or RP2D for memory T cells plus the SoC. In arm B, patients will receive SoC or RP2D for NK cells plus the SoC. DISCUSSION: We hypothesized that SARS-CoV-2-specific memory T-lymphocytes obtained from convalescent donors recovered from COVID-19 can be used as a passive cell immunotherapy to treat pneumonia and lymphopenia in moderate/severe patients. The lymphopenia induced by COVID-19 constitutes a therapeutic window that may facilitate donor engraftment and viral protection until recovery. TRIAL REGISTRATION: ClinicalTrials.gov NCT04578210 . First Posted : October 8, 2020.


Asunto(s)
COVID-19 , Linfopenia , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Humanos , Memoria Inmunológica , Células Asesinas Naturales , Linfopenia/diagnóstico , Linfopenia/terapia , Estudios Multicéntricos como Asunto , Ensayos Clínicos Controlados Aleatorios como Asunto , SARS-CoV-2 , Linfocitos T , Resultado del Tratamiento
7.
Front Immunol ; 12: 759558, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34650571

RESUMEN

Adoptive cell therapy (ACT) using chimeric antigen receptor (CAR) T cells holds impressive clinical outcomes especially in patients who are refractory to other kinds of therapy. However, many challenges hinder its clinical applications. For example, patients who undergo chemotherapy usually have an insufficient number of autologous T cells due to lymphopenia. Long-term ex vivo expansion can result in T cell exhaustion, which reduces the effector function. There is also a batch-to-batch variation during the manufacturing process, making it difficult to standardize and validate the cell products. In addition, the process is labor-intensive and costly. Generation of universal off-the-shelf CAR T cells, which can be broadly given to any patient, prepared in advance and ready to use, would be ideal and more cost-effective. Human induced pluripotent stem cells (iPSCs) provide a renewable source of cells that can be genetically engineered and differentiated into immune cells with enhanced anti-tumor cytotoxicity. This review describes basic knowledge of T cell biology, applications in ACT, the use of iPSCs as a new source of T cells and current differentiation strategies used to generate T cells as well as recent advances in genome engineering to produce next-generation off-the-shelf T cells with improved effector functions. We also discuss challenges in the field and future perspectives toward the final universal off-the-shelf immunotherapeutic products.


Asunto(s)
Inmunoterapia Adoptiva , Células Madre Pluripotentes Inducidas/inmunología , Linfopenia/terapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Humanos , Células Madre Pluripotentes Inducidas/citología , Linfopenia/inmunología , Linfocitos T/citología
11.
J Immunol ; 206(12): 2828-2838, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34108260

RESUMEN

Differentially and functionally distinct T cell subsets are involved in the development of complications after allogeneic hematopoietic stem cell transplantation (HSCT), but little is known about factors regulating their recovery after HSCT. In this study, we investigated associations between immune-regulating cytokines, T cell differentiation, and clinical outcomes. We included 80 children undergoing allogeneic HSCT for acute leukemia using bone marrow or peripheral blood stem cells grafted from a matched sibling or unrelated donor. Cytokines (IL-7, IL-15, IL-18, SCF, IL-6, IL-2, and TNF-α) and active anti-thymocyte globulin (ATG) levels were longitudinally measured along with extended T cell phenotyping. The cytokine profiles showed a temporary rise in IL-7 and IL-15 during lymphopenia, which was strongly dependent on exposure to active ATG. High levels of IL-7 and IL-15 from graft infusion to day +30 were predictive of slower T cell recovery during the first 2 mo post-HSCT; however, because of a major expansion of memory T cell stages, only naive T cells remained decreased after 3 mo (p < 0.05). No differential effect was seen on polarization of CD4+ T cells into Th1, Th2, or Th17 cells or regulatory T cells. Low levels of IL-7 and IL-15 at day +14 were associated with acute graft-versus-host disease grades II-IV in ATG-treated patients (p = 0.0004 and p = 0.0002, respectively). Children with IL-7 levels comparable to healthy controls at day +14 post-HSCT were less likely to develop EBV reactivation posttransplant. These findings suggest that quantification of IL-7 and IL-15 may be useful as biomarkers in assessing the overall T cell depletion and suggest a potential for predicting complications after HSCT.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/efectos adversos , Interleucina-15/análisis , Interleucina-7/análisis , Leucemia Mieloide Aguda/terapia , Linfopenia/terapia , Células T de Memoria/inmunología , Adolescente , Adulto , Niño , Preescolar , Humanos , Lactante , Interleucina-15/inmunología , Interleucina-7/inmunología , Leucemia Mieloide Aguda/inmunología , Depleción Linfocítica , Linfopenia/inmunología , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
12.
Acta Haematol ; 144(6): 698-705, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34062545

RESUMEN

Idiopathic CD4+ lymphocytopenia (ICL) is the depletion of CD4+ lymphocytes to <300 cells/mm3 without human immunodeficiency virus infection or other causes of lymphocytopenia. ICL causes fatal infections; its etiology remains unclear and it lacks consensus regarding therapeutic options. We report the first patient with ICL who had a successful clinical course following a cord blood transplant (CBT). A 45-year-old woman was diagnosed with ICL and underwent partial hepatectomy for an abscess caused by the Mycobacterium avium complex. No specific gene alterations were detected through next generation sequencing-based evaluation. Following a reduced-intensity conditioning (RIC) regimen consisting of fludarabine, busulfan, and 4 Gy total body irradiation, a single-unit CBT was performed. Neutrophils were engrafted on day +14. CD4+ lymphocyte counts increased to over 300 cells/mm3 on day +436. After 75 months, she was alive without any sequelae. CBT with an RIC regimen could be a curable treatment option for ICL.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Trasplante de Células Madre de Sangre del Cordón Umbilical , Linfopenia/terapia , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Femenino , Hepatectomía , Humanos , Absceso Hepático/etiología , Absceso Hepático/cirugía , Recuento de Linfocitos , Linfopenia/diagnóstico , Linfopenia/inmunología , Persona de Mediana Edad , Complejo Mycobacterium avium/patogenicidad , Neutrófilos/trasplante , Tomografía Computarizada por Rayos X , Irradiación Corporal Total
14.
Electromagn Biol Med ; 40(1): 11-25, 2021 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-33073612

RESUMEN

Among haematological parameters of patients seriously ill with the coronavirus infectious disease 2019 (COVID-19), leucocytosis, lymphocytopenia, and the abnormal release of circulating cytokines, termed cytokine storm syndrome (CSS, also known as cytokine release syndrome or CRS), were found associated with disease severity. In particular, according to the serum cytokine profiling, pro-inflammatory interleukin 6 (IL-6) and anti-inflammatory interleukin 10 (IL-10) were observed to be considerably higher in patients experiencing respiratory distress, septic shock and/or multi-organ failure, namely "critical cases" requiring intensive care unit (ICU) admission, very often resulting in death. Interestingly, the production of these cytokines from human lymphocytes was found to be modulated by exposure of 24 h to a 554.2-553.8 mT inhomogeneous static magnetic field (SMF), which elicits IL-10 and suppresses IL-6. Thus, herein, with the aim of restoring lymphocyte count and physiological serum levels of IL-6 and IL-10, the infusion of human leukocyte antigen (HLA)-matched and SMF-exposed allogenic lymphocytes is proposed for the first time as an easy and affordable treatment option for COVID-19 patients. Even if the count of lymphocytes in COVID-19 patients is very low, SMF exposure may be a valuable tool for reprogramming autologous lymphocytes towards physiological conditions. Furthermore, the same procedure could be extended to include the whole autologous or allogenic white blood cells (WBCs). Time-varying/pulsed magnetic fields exerting comparable cell effects could also be employed.


Asunto(s)
COVID-19/complicaciones , Síndrome de Liberación de Citoquinas/terapia , Antígenos HLA/inmunología , Linfocitos/citología , Linfopenia/terapia , Campos Magnéticos , Síndrome de Liberación de Citoquinas/complicaciones , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/patología , Humanos , Inmunoterapia , Interleucina-6/química , Interleucina-6/metabolismo , Linfocitos/inmunología , Linfopenia/complicaciones , Linfopenia/inmunología , Linfopenia/patología , Modelos Moleculares , Conformación Proteica , Transducción de Señal/inmunología
15.
Nat Rev Immunol ; 21(5): 277-291, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33097917

RESUMEN

Following periods of haematopoietic cell stress, such as after chemotherapy, radiotherapy, infection and transplantation, patient outcomes are linked to the degree of immune reconstitution, specifically of T cells. Delayed or defective recovery of the T cell pool has significant clinical consequences, including prolonged immunosuppression, poor vaccine responses and increased risks of infections and malignancies. Thus, strategies that restore thymic function and enhance T cell reconstitution can provide considerable benefit to individuals whose immune system has been decimated in various settings. In this Review, we focus on the causes and consequences of impaired adaptive immunity and discuss therapeutic strategies that can recover immune function, with a particular emphasis on approaches that can promote a diverse repertoire of T cells through de novo T cell formation.


Asunto(s)
Regeneración/inmunología , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Linfocitos B/inmunología , Linfocitos B/fisiología , Microambiente Celular/inmunología , Microambiente Celular/fisiología , Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunosenescencia , Inmunoterapia , Linfopenia/inmunología , Linfopenia/terapia , Modelos Inmunológicos , Regeneración/fisiología , Estrés Fisiológico/inmunología , Estrés Fisiológico/fisiología , Linfocitos T/fisiología
17.
Acta Haematol ; 144(1): 10-23, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32721958

RESUMEN

Coronavirus disease 2019 (COVID-19) is affecting millions of patients worldwide. It is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which belongs to the family Coronaviridae, with 80% genomic similarities to SARS-CoV. Lymphopenia was commonly seen in infected patients and has a correlation to disease severity. Thrombocytopenia, coagulation abnormalities, and disseminated intravascular coagulation were observed in COVID-19 patients, especially those with critical illness and non-survivors. This pandemic has caused disruption in communities and hospital services, as well as straining blood product supply, affecting chemotherapy treatment and haematopoietic stem cell transplantation schedule. In this article, we review the haematological manifestations of the disease and its implication on the management of patients with haematological disorders.


Asunto(s)
Coagulación Intravascular Diseminada , Trasplante de Células Madre Hematopoyéticas , Linfopenia , Pandemias , SARS-CoV-2/metabolismo , Trombocitopenia , COVID-19/sangre , COVID-19/mortalidad , COVID-19/terapia , Coagulación Intravascular Diseminada/sangre , Coagulación Intravascular Diseminada/mortalidad , Coagulación Intravascular Diseminada/terapia , Coagulación Intravascular Diseminada/virología , Humanos , Linfopenia/sangre , Linfopenia/mortalidad , Linfopenia/terapia , Linfopenia/virología , Trombocitopenia/sangre , Trombocitopenia/mortalidad , Trombocitopenia/terapia , Trombocitopenia/virología
18.
J Med Virol ; 93(2): 794-802, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32672840

RESUMEN

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA was found in the intestines and feces, but its clinical significance is not completely clear. We aim to characterize the longitudinal test results of SARS-CoV-2 RNA in anal swabs and to explore the association with disease severity. METHODS: We included laboratory-confirmed coronavirus disease 2019 (COVID-19) patients, who were hospitalized in Guangzhou Eighth People's Hospital and excluded those who had not received anal swabs for SARS-COV-2 RNA testing. Epidemiological, clinical, and laboratory data were obtained. Throat swabs and anal swabs were collected periodically for SARS-COV-2 RNA detection. RESULTS: Two hundred and seventeen eligible patients (median aged 50 years, 50.2% were females) were analyzed. 21.2% (46/217) of the patients were detected with SARS-CoV-2 RNA in anal swabs. The duration of viral RNA was longer, but the viral load was lower in anal swabs than throat swabs in the early stage of the disease. During a median follow-up of 20 days, 30 (13.8%) patients were admitted to the intensive care unit (ICU) for high-flow nasal cannula or higher-level oxygen support measures to correct hypoxemia. Detectable viral RNA in anal swabs (adjusted hazard ratio [aHR], 2.50; 95% confidence interval [CI], 1.20-5.24), increased C-reactive protein (aHR, 3.14; 95% CI, 1.35-7.32) and lymphocytopenia (aHR, 3.12; 95% CI, 1.46-6.67) were independently associated with ICU admission. The cumulative incidence of ICU admission was higher among patients with detectable viral RNA in anal swabs (26.3% vs 10.7%, P = .006). CONCLUSION: Detectable SARS-CoV-2 RNA in the digestive tract was a potential warning indicator of severe disease.


Asunto(s)
Canal Anal/virología , COVID-19/diagnóstico , Linfopenia/diagnóstico , ARN Viral/genética , SARS-CoV-2/genética , Adulto , Antivirales/uso terapéutico , Proteína C-Reactiva/metabolismo , COVID-19/patología , COVID-19/terapia , COVID-19/virología , Prueba de COVID-19 , Cloroquina/uso terapéutico , Femenino , Hospitalización/estadística & datos numéricos , Humanos , Indoles/uso terapéutico , Unidades de Cuidados Intensivos/estadística & datos numéricos , Linfopenia/patología , Linfopenia/terapia , Linfopenia/virología , Masculino , Persona de Mediana Edad , Oseltamivir/uso terapéutico , Faringe/virología , Estudios Retrospectivos , SARS-CoV-2/patogenicidad , Índice de Severidad de la Enfermedad , Carga Viral/efectos de los fármacos
19.
Hum Antibodies ; 29(1): 49-54, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32986663

RESUMEN

Coronavirus disease 2019 (COVID-19) has caused a global pandemic in early 2020. This infectious disorder has a heterogeneous course ranging from asymptomatic disorder to a critical situation needing intensive cares. In the current study, we present a report of affected patients admitted in a single hospital in Iran. Eighty-two hospitalized patients with COVID-19 were assessed. Demographic, clinical, and paraclinical parameters were gathered and statistically analyzed. The median age (IQR) of the patients was 57.32 (45.75, 70) years. At primary evaluation, fever was present in 45.12% of the affected individuals. The most common clinical symptoms were dyspnea (81.71%) and cough (65.85%). Totally, 12 (14.63%) and 14 (17.07%) of patients had low and high WBC counts, respectively. Lymphopenia was detected in 36 (43.9%) of patients, while 6 (7.32%) of patients had lymphocytosis. High levels of Il-6 were detected in 4 (4.88%) of patients. CRP levels were elevated in 69 (84.1%) of patients. The median (IQR) of hospitalization was 7 (5, 9) days. Totally, 26 patients (31%) were hospitalized in ICU. All patients were discharged with good health conditions except for one patient who died. The current study shows the heterogeneous clinical manifestations and paraclinical parameters of COVID-19 patients.


Asunto(s)
COVID-19/fisiopatología , Tos/fisiopatología , Disnea/fisiopatología , Fiebre/fisiopatología , Linfocitosis/fisiopatología , Linfopenia/fisiopatología , Anciano , Proteína C-Reactiva/metabolismo , COVID-19/mortalidad , COVID-19/terapia , COVID-19/virología , Tos/mortalidad , Tos/terapia , Tos/virología , Diabetes Mellitus/mortalidad , Diabetes Mellitus/fisiopatología , Diabetes Mellitus/terapia , Diabetes Mellitus/virología , Disnea/mortalidad , Disnea/terapia , Disnea/virología , Femenino , Fiebre/mortalidad , Fiebre/terapia , Fiebre/virología , Hospitales , Humanos , Hipertensión/mortalidad , Hipertensión/fisiopatología , Hipertensión/terapia , Hipertensión/virología , Irán , Recuento de Leucocitos , Linfocitosis/mortalidad , Linfocitosis/terapia , Linfocitosis/virología , Linfopenia/mortalidad , Linfopenia/terapia , Linfopenia/virología , Masculino , Persona de Mediana Edad , Obesidad/mortalidad , Obesidad/fisiopatología , Obesidad/terapia , Obesidad/virología , Oxígeno/uso terapéutico , Respiración Artificial/métodos , Estudios Retrospectivos , SARS-CoV-2/patogenicidad , Índice de Severidad de la Enfermedad , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA