Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Annals of the Rheumatic Diseases ; 82(Suppl 1):1498, 2023.
Article in English | ProQuest Central | ID: covidwho-20235066

ABSTRACT

BackgroundFatigue is a difficult subject for both physicians and patients. It is barely addressed during consultations and can therefore burden patient-physician-relations. To improve communication regarding fatigue, we developed a checklist that includes suggestions for evaluating possible causes for fatigue. In this analysis, we describe our study population and report first results 3 and 6 months after using the checklist.ObjectivesThe aims of our study are to validate the use of our newly developed fatigue checklist and to demonstrate that addressing fatigue in daily clinical practice and offering possible interventions can improve fatigue.MethodsWe recruited n=110 SLE patients with fatigue from our university hospital-based lupus reference centre in Duesseldorf. Fatigue was measured using the FSS (Fatigue Severity Scale). Our checklist included signs of depression and anxiety using the PHQ-4 (Patient Health Questionnaire), BMI (body mass index), physical activity, anemia, hypothyroidism and vitamin D deficiency. For each applicable cause, we listed possible interventions for free selection by the treating physician, such as replacement therapy (vitamin D, vitamin B12, iron, folic acid, erythropoietin), physical activity programs and psychosomatic consultations that were discussed with the patients. We re-evaluated our patients after 3 (T1) and 6 months (T2).ResultsBaseline characteristics of patients are summarized in Table 1.Table 1.BMI=body mass index, TSH=thyroidea stimulating hormone, PHQ4=patient health questionnaire (cut-off >3 points), HAQ=health assessment questionnaire, IMET= Index for measuring restrictions on social participation (higher scores point towards more restrictions on social participation), FSS=fatigue severity scale (≥4 points equal severe fatigue)N = 110n (%)Mean (SD)Age (years)49.0 (12.34)Female sex99.0 (90.0)BMI (kg/m2)25.9 (5.55)Disease duration (years)19.1 (10.05)TSH (µIU/ml)1.5 (1.05)25-OH-Vitamin D (ng/ml)39.5 (15.35)Haemoglobin (g/dl)13.0 (1.64)Sports activities>4h/week6.0 (5.5)2-4h/week18.0 (16.4)1-2h/week16.0 (14.5)<1h/week28.0 (25.5)No sport42.0 (38.2)Depression (PHQ4 score)2.3 (1.63)Anxiety (PHQ4 score)2.0 (1.71)Functional status (HAQ score)0.8 (0.49)Participation (IMET score)2.8 (2.31)Fatigue (FSS score)5.3 (1.35)After 3 and 6 months, we re-evaluated 83 patients and saw a significant reduction in fatigue measured by the FSS score (T1: mean difference estimate 0.367 and p-value <0.001;T2: mean difference estimate 0.305;p-value <0.005).Figure 1.Comparing FSS-Scores from T0, T1 and T2[Figure omitted. See PDF]ConclusionThe preliminary analysis of our study shows for the first time that incorporation of a checklist procedure into the management of patients with fatigue may improve short-term outcome after 3 and 6 months of observation. The improvement of symptoms documented in our study occurred even though the suggested exercise program and psychosomatic counseling sessions were not available for use during the current observation period because of the COVID-19 pandemic. At present, the mechanisms behind the observed effect remain unclear. Our ongoing analysis will clarify whether an additional effect on fatigue will occur after all suggested interventions resulting from the use of the checklist have been executed. Finally, it will demonstrate whether the incorporation of our checklist into routine clinical practice is capable to reduce fatigue over a prolonged time period.REFERENCES:NIL.Acknowledgements:NIL.Disclosure of InterestsNone Declared.

2.
Biomolecules ; 13(5)2023 05 11.
Article in English | MEDLINE | ID: covidwho-20239134

ABSTRACT

It is estimated that, at minimum, 500 million individuals suffer from cellular metabolic dysfunction, such as diabetes mellitus (DM), throughout the world. Even more concerning is the knowledge that metabolic disease is intimately tied to neurodegenerative disorders, affecting both the central and peripheral nervous systems as well as leading to dementia, the seventh leading cause of death. New and innovative therapeutic strategies that address cellular metabolism, apoptosis, autophagy, and pyroptosis, the mechanistic target of rapamycin (mTOR), AMP activated protein kinase (AMPK), growth factor signaling with erythropoietin (EPO), and risk factors such as the apolipoprotein E (APOE-ε4) gene and coronavirus disease 2019 (COVID-19) can offer valuable insights for the clinical care and treatment of neurodegenerative disorders impacted by cellular metabolic disease. Critical insight into and modulation of these complex pathways are required since mTOR signaling pathways, such as AMPK activation, can improve memory retention in Alzheimer's disease (AD) and DM, promote healthy aging, facilitate clearance of ß-amyloid (Aß) and tau in the brain, and control inflammation, but also may lead to cognitive loss and long-COVID syndrome through mechanisms that can include oxidative stress, mitochondrial dysfunction, cytokine release, and APOE-ε4 if pathways such as autophagy and other mechanisms of programmed cell death are left unchecked.


Subject(s)
Alzheimer Disease , COVID-19 , Diabetes Mellitus , Metabolic Diseases , Neurodegenerative Diseases , Humans , AMP-Activated Protein Kinases/metabolism , Post-Acute COVID-19 Syndrome , TOR Serine-Threonine Kinases/metabolism , Alzheimer Disease/metabolism , Neurodegenerative Diseases/metabolism , Brain/metabolism
3.
Organ Transplantation ; 12(2):169-176, 2021.
Article in Chinese | EMBASE | ID: covidwho-2327450

ABSTRACT

Renal transplantation is the optimal approach to improve the quality of life and restore normal life for patients with end-stage renal diseases. With the development of medical techniques and immunosuppressants, the short-term survival of renal graft has been significantly prolonged, whereas the long-term survival remains to be urgently solved. Renal ischemia-reperfusion injury (IRI), acute rejection, chronic renal allograft dysfunction, renal fibrosis and other factors are still the major problems affecting the survival of renal graft. Relevant researches have always been hot spots in the field of renal transplantation. Meantime, 2020 is an extraordinary year. The novel coronavirus pneumonia (COVID-19) pandemic severely affects the development of all walks of life. Researches related to renal transplantation have also sprung up. In this article, the frontier hotspots of clinical and basic studies related to renal transplantation and the COVID-19 related researches in the field of renal transplantation in China were reviewed, aiming to provide novel therapeutic ideas and strategies.Copyright © 2021 Journal of Zhongshan University. All Rights Reserved.

4.
Allergy: European Journal of Allergy and Clinical Immunology ; 78(Supplement 111):706, 2023.
Article in English | EMBASE | ID: covidwho-2294116

ABSTRACT

Case report We present the case of a 63-year- old man with two consecutive admissions, due to COVID19 infection and subsequent bacterial superinfection. Three days after the second admission (04/28), and 43 days from the beginning of the infection an assessment by dermatology and allergology is then requested. The patient had generalized erythematous maculopapular rash in the trunk, back, groin and limbs. On the left side and back, pustular lesions not focused on follicles were also added, with a fever of 37.7degreeC. There were no oral and genital lesions. No psoriasis. The drugs used during the present and previous admissions were reviewed. Previous admission (04/04-22/ 20): Linezolid, ciprofloxacin, meropenem 04/13-22, piperacillin/tazobactam, hydroxychloroquine, azithromycin, ceftriaxone. Upon discharge amoxicillin/acid clavulanic. Present admission (04/25) Cutaneous reaction 04/28. 04/25: meropenem, paracetamol, enoxaparin, insulin, omeprazole, venlafaxine. 04/26: Darbepoetin, furosemide, mycophenolate in single dose. 04/27: Linezolid, macrogol, Clopidogrel, Magnesium, Calcitriol. Medical records: DM type 2, liver transplantation due to HCV cirrhosis, HCV recurrence, uninodular hepatocarcinoma, advanced CKD, secondary hyperparathyroidism, multiple neurological antecedents. We performed a detailed study. We hypothesized with a pharmagological/ drug reaction with several drugs possibly involved and our main suspicion was an allergic reaction to beta-lactams. Biopsy: Subcorneal pustules, basal spongiosis and presence in the superficial dermis of edema and an inflammatory infiltrate with abundant neutrophils. No fungi. Findings compatible with clinical diagnosis of generalized acute exanthematic pustulosis (PEGA). Immunohistochemical study Covid19. (Jimenez Diaz Foundation) Finely granular positivity in endothelium and more coarse in sweaty epithelium. Neutrophilic superficial inflammatory component with presumably spure staining. ACe-2 (positive external control) is not detected. The patient presents a EuroSCAR score of 9, sum of the clinic and the pathological anatomy, and therefore defined diagnosis. Clinical diagnosis: PEGA secondary to meropenem. Conclusion(s): We present the case of a PEGA by meropenem, not very often described in the literature. We highlight the importance of differential diagnosis with viral infections. Skin tests, especially epicutaneous tests, are key to the diagnosis. (Figure Presented).

5.
Hum Cell ; 36(3): 908-922, 2023 May.
Article in English | MEDLINE | ID: covidwho-2275550

ABSTRACT

Severe acute respiratory coronavirus 2 (SARS-CoV-2) infection in the young and healthy usually results in an asymptomatic or mild viral syndrome, possibly through an erythropoietin (EPO)-dependent, protective evolutionary landscape. In the old and in the presence of co-morbidities, however, a potentially lethal coronavirus disease 2019 (COVID-19) cytokine storm, through unrestrained renin-angiotensin aldosterone system (RAAS) hyperactivity, has been described. Multifunctional microRNA-155 (miR-155) elevation in malaria, dengue virus (DENV), the thalassemias, and SARS-CoV-1/2, plays critical antiviral and cardiovascular roles through its targeted translational repression of over 140 genes. In the present review, we propose a plausible miR-155-dependent mechanism whereby the translational repression of AGRT1, Arginase-2 and Ets-1, reshapes RAAS towards Angiotensin II (Ang II) type 2 (AT2R)-mediated balanced, tolerable, and SARS-CoV-2-protective cardiovascular phenotypes. In addition, it enhances EPO secretion and endothelial nitric oxide synthase activation and substrate availability, and negates proinflammatory Ang II effects. Disrupted miR-155 repression of AT1R + 1166C-allele, significantly associated with adverse cardiovascular and COVID-19 outcomes, manifests its decisive role in RAAS modulation. BACH1 and SOCS1 repression creates an anti-inflammatory and cytoprotective milieu, robustly inducing antiviral interferons. MiR-155 dysregulation in the elderly, and in comorbidities, allows unimpeded RAAS hyperactivity to progress towards a particularly aggressive COVID-19 course. Elevated miR-155 in thalassemia plausibly engenders a favorable cardiovascular profile and protection against malaria, DENV, and SARS-CoV-2. MiR-155 modulating pharmaceutical approaches could offer novel therapeutic options in COVID-19.


Subject(s)
COVID-19 , MicroRNAs , Humans , SARS-CoV-2 , COVID-19/genetics , Peptidyl-Dipeptidase A , Angiotensin II , Antiviral Agents , MicroRNAs/genetics
6.
Turkish Journal of Pediatric Disease ; 15(5):434-436, 2021.
Article in English | EMBASE | ID: covidwho-2231627

ABSTRACT

We describe a case of a patient with Chronic Kidney Disease who developed polycythemia due to Erythropoiesis Stimulating Agents overuse during COVID-19 isolation. A 12-year-old male had not been able to attend routine controls since had been in isolation for 4 months after the COVID-19 outbreak. He had continued to take Erythropoiesis-Stimulating Agents during that period at the starting dose of 150 U/kg/week. He had been on peritoneal dialysis in the last year because of end-stage renal failure. Laboratory investigation revealed a hemoglobin (Hb) level of 20.8 g/dl, hematocrit level of 66%, creatinine level of 6.5 mgr/dl. He underwent daily phlebotomy sessions (10cc/kg/session). During this period aspirin was also started (5mg/kg). After 5 sessions his Hb level decreased to 14 gr/dl and hematocrit to 40%. Pediatric nephrologist should be aware that there is a potential risk of polycythemia with Erythropoiesis Stimulating Agents when Hb level is not appropriately followed on a routine basis. Copyright © 2021 Ankara Pediatric Hematology Oncology Training and Research Hospital. All rights reserved.

7.
Pan Afr Med J ; 43: 129, 2022.
Article in English | MEDLINE | ID: covidwho-2164238

ABSTRACT

The COVID-19 pandemic is arguably one of the greatest public health crises since the 1918 influenza pandemic. Although several vaccines have been approved and rolled out, effective antiviral treatment options are very limited. Here, we present a case of severe COVID-19 that failed to respond to the standard interventions and continued to deteriorate. On day 22 of his illness, after informed consent, the patient was administered 4000IU of erythropoietin (EPO) subcutaneously, in the hope of improving his O2 saturation. Positive response was observed in the patient within 24 hours. This prompted us to continued EPO treatment for a total of 42 days until full recovery and discharge. Our findings warrant further studies to ascertain the use of EPO in severe cases COVID-19.


Subject(s)
Antiviral Agents , COVID-19 , Erythropoietin , Humans , Antiviral Agents/therapeutic use , COVID-19/epidemiology , Erythropoietin/therapeutic use , SARS-CoV-2
8.
Journal of Clinical and Basic Research ; 6(1):11-27, 2022.
Article in English | CAB Abstracts | ID: covidwho-2057219

ABSTRACT

Coronavirus disease 2019 (COVID-19) is a severe acute respiratory disease with a high prevalence. According to the research and statistical data, in January 2021, there have been 92,262,621 confirmed cases of COVID-19 and more than two million deaths. Infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the main cause of this disease. In addition to the respiratory system, the disease affects the gastrointestinal tract, central-peripheral nervous system, circulatory system, and kidneys. Therefore, any therapeutic action to reduce COVID-19-related symptoms and complications is essential. In this study, we conducted a systematic review of the published literature and preprints on the efficacy of erythropoietin (EPO) and recombinant human EPO as a safe stimulant and tissue protector in the treatment of COVID-19. We also briefly described the structure of coronavirus, its pathogenesis, and the structure of EPO and recombinant human EPO. All relevant articles published in the Science Direct, PubMed, and Google Scholar databases were searched. According to the results, EPO is a cytoprotective cytokine induced by hypoxia. The pleiotropic effects of EPO are associated with its erythrocyte-forming, anti-apoptotic, anti-inflammatory activities. It also exerts protective effects on the heart, lungs, kidneys, arteries, and central and peripheral nervous systems. It has been demonstrated that EPO can increase hemoglobin levels, thereby increasing oxygen delivery to the tissues. Therefore, recombinant human EPO therapy can be used for counteracting the adverse effects of COVID-19 including hypoxic myocarditis, acute renal failure, pulmonary edema, and brain-spinal cord ischemic injury. Overall, the use of EPO and recombinant human EPO therapy increases blood coagulation, tumor growth, thromboembolism, and purification of red blood cells, which must be accompanied by anticoagulants such as heparin.

9.
Sports Health ; : 19417381221120639, 2022 Sep 25.
Article in English | MEDLINE | ID: covidwho-2053793

ABSTRACT

BACKGROUND: Recent studies have indicated that people who live at altitude have a lower incidence of coronavirus disease (COVID-19) and lesser severity in infection cases. HYPOTHESIS: Hypoxia exposure could lead to health benefits, and it could be used in the recovery process as an additional stimulus to physical training to improve cardiorespiratory fitness (CRF). STUDY DESIGN: Randomized controlled clinical trial. LEVEL OF EVIDENCE: Level 2. METHODS: The 43 participants, aged 30 to 69 years, were divided into control group (CG, n = 18) and 2 training groups: normoxia (NG, n = 9) and hypoxia (HG, n = 16). Before and after the intervention were evaluated the lactate threshold 2 (L2), peak oxygen uptake (VO2peak), and a blood sample was collected at rest to evaluate hematological adaptation. Both groups performed an 8-week moderate-intensity physical training on a bike. The HG were trained under normobaric hypoxic conditions (fractional inspired oxygen [FiO2] = 13.5%). RESULTS: The 8-week intervention promoted a similar improvement in CRF of people recovered from COVID-19 in the HG (L2 = 34.6%; VO2peak = 16.3%; VO2peak intensity = 24.6%) and NG (L2 = 42.6%; VO2peak = 16.7%; VO2peak intensity = 36.9%). Only the HG presented differences in hematological variables (erythropoietin = 191.7%; reticulocytes = -32.4%; off-score = 28.2%) in comparison with the baseline. CONCLUSION: The results of the present study provide evidence that moderate-intensity training in normoxia or hypoxia promoted similar benefits in CRF of people recovered from COVID-19. Furthermore, the hypoxia offered an additional stimulus to training promoting erythropoietin increase and hematological stimulation. CLINICAL RELEVANCE: The present exercise protocol can be used for the rehabilitation of people recovered from COVID-19, with persistent low CRF. In addition, this is the first study demonstrating that physical training combined with hypoxia, as well as improving CRF, promotes greater hematological stimulation in people recovered from COVID-19.

10.
Journal of Kermanshah University of Medical Sciences ; 26(2), 2022.
Article in English | EMBASE | ID: covidwho-1969721

ABSTRACT

Background: Erythropoietin plays a significant role in the growth of red blood cells, hemoglobin levels, and tissue oxygenation in critically ill patients, as well as anti-inflammatory and neuroprotective effects. Objectives: This study aimed to evaluate the effect of recombinant erythropoietin on improving COVID-19 patients. Methods: This study was conducted on 20 COVID-19 participants with hemoglobin of ≥ 9. The inclusion criteria was at least one severe COVID-19 symptom/sign in this interventional study. The primary outcome was a combination of hospital stay length and paraclinical evaluation (LDH and hemoglobin level). The outcomes and side effects were evaluated on day 0 (before the intervention) and five (post-intervention). Results: The mean hemoglobin level was 10 ± 1.1 gr/dL in the intervention group and 8 ± 0.7 gr/dL in the control group post-treatment, indicating a significant difference between the groups (P = 0.04). The mean hospital stay length (6 ± 2 days) in the intervention group was significantly less than the control group (9 ± 4 days) (P = 0.001). At the end of the treatment, the mean LDH was significantly lower in the intervention group (503 ± 264 µ/L) than in the control group (725 ± 320 µ/L;P = 0.017). Conclusions: According to the results, this study provides the first solid evidence for the positive effects of recombinant erythropoietin on COVID-19.

11.
Nephrology Dialysis Transplantation ; 37(SUPPL 3):i641-i643, 2022.
Article in English | EMBASE | ID: covidwho-1915773

ABSTRACT

BACKGROUND AND AIMS: Patients with end-stage kidney disease (ESKD) face higher risk for severe outcomes from COVID-19 infection. Moreover, it is not well known to which extent potentially modifiable risk factors contribute to mortality risk. In this study, we investigated the incidence and risk factors for 30-day case-fatality of COVID-19 in haemodialysis patients treated in the European Fresenius Medical Care (FMC) Nephrocare network. METHOD: In this historical cohort study, we included unvaccinated adult dialysis patients with a first documented SARS-CoV-2 infection between 1 February 2020 and 31 March 2021 (study period) registered in the European Clinical Database (EuCliD ® ). The first SARS-CoV-2 suspicion date for all documented infections was considered the index date for the analysis. Patients were followed for up to 30 days. Follow-up time was defined from the index date until the date of death, end of follow-up period or lost to follow-up, whichever occurred first. We ascertained patients' characteristics in the 6-month period prior to index date. We used logistic regression and XGBoost to assess risk factors for 30-day mortality. RESULTS: We included 9211 patients meeting the inclusion criteria for the study (Table 1). Age was 65.4 ± 13.7 years, dialysis vintage was 4.2 ± 3.7 years. In the follow up period, 1912 patients died within 30 days (20.8%, 95% confidence interval: 19.9%- 21.6%). Correlates of COVID-19 related mortality are summarized in Table 2. Several potentially modifiable factors were associated with increased risk of death: patients on HD compared with online haemodiafiltration had shorter survival after presentation with COVID-19 as well as those who did not achieve the therapeutic targets for serum albumin, erythropoietin resistance index, protein catabolic rate, haemodynamic status, C-reactive protein, single-pool Kt/V, hydration status and serum sodium in the months before infection. The discrimination accuracy of prediction models developed with XGBoost was similar to that observed for main-effect logistic regression (AUC 0.69 and 0.71, respectively) suggesting that no major cross-interaction and non-linear effect could improve prediction accuracy. CONCLUSION: We observed high 30-day COVID-19 related mortality among unvaccinated dialysis patients. Older patients, men and those with greater.

12.
Italian Journal of Medicine ; 16(SUPPL 1), 2022.
Article in English | EMBASE | ID: covidwho-1912859

ABSTRACT

The proceedings contain 357 papers. The topics discussed include: evaluation of the nurse staffing level in the IRCCS Rizzoli through the use of ICA method;gender differences in patients hospitalized for COVID-19 disease;clinical study on the efficacy and safety of arginine administered orally in association with other active ingredients for the prevention and treatment of sarcopenia in patients with COVID-19-related pneumonia;regional efficacy and safety results of roxadustat compared with placebo or darbepoetin alfa in non-dialysis-dependent chronic kidney disease patients with anemia;C1s-targeted inhibition of classical complement pathway by sutimlimab in cold agglutinin disease: efficacy and safety results from the 26-week, randomized, placebo-controlled phase 3 CADENZA study (NCT03347422);octreotide LAR in patients with intestinal angiectasies and anticoagulation treatment;and implementation of 'Bedside modified-ISBAR' in nursing handover of COVID-patients in urgent-care medicine of Azienda Sanitaria Locale Biella (ASL BI): a phenomenological qualitative study.

13.
Bull Exp Biol Med ; 173(1): 46-50, 2022 May.
Article in English | MEDLINE | ID: covidwho-1872572

ABSTRACT

Morphological and functional characteristics of erythrocytes, hemoglobin, and erythropoietin level in the venous blood were evaluated by laser interference microscopy, Raman spectroscopy with a short-focus extreme aperture lens monochromator, and by ELISA, respectively, in 30 patients with verified moderate COVID-19 at the time of hospitalization and 30 healthy volunteers. The patients whose course of COVID-19 has worsened to critical by day 5 had already had lower (p<0.001) indicators at the time of hospitalization such as the area and thickness of erythrocytes, the hemoglobin distribution and packing density, hemoglobin conformation index (I1355/I1550)/(I1375/I1580) reflecting its oxygen affinity, and blood erythropoietin content. Our findings suggest that these characteristics of erythrocytes, hemoglobin, and erythropoietin can serve as potential predictors of COVID-19 aggravation in hospitalized patients.


Subject(s)
COVID-19 , Erythropoietin , Erythrocytes/chemistry , Hemoglobins/chemistry , Humans
14.
Breast ; 56:S58, 2021.
Article in English | EMBASE | ID: covidwho-1735076

ABSTRACT

Goals: ESA are recombinant human erythropoietin preparations which stimulate bone marrow to produce RBC. Apart from relieving anaemia symptoms, the application of ESA can prevent the necessity of the red blood cells transfusion in patients with CIA and thus avoid transfusion-related complications. During the Covid-19 pandemic, the absence of the necessity of hospitalization to transfuse blood products presents an additional value. The aim of the study is to assess the results of the application of darbepoetin alfa in the treatment of anaemia in pts undergoing neoadjuvant chemotherapy for EBC. Methods: A retrospective assessmentwas carried out of the results of darbepoetin alfa (Aranesp®) treatment of 98 pts receiving neoadjuvant chemotherapy for EBC in the National Institute of Oncology in Poland in whom therapy was initiated in the period from 02 January 2019 to 16 February 2020. The drug was given to symptomatic anaemia patients, with a haemoglobin (Hb) level of 8–11 g/dL. In addition, it was also applied in chemotherapy-undergoing patients, with asymptomatic anaemia, with a haemoglobin concentration of <8 g/dL. The treatment was continued until a stable Hb level was reached ensuring the security of further oncological treatment without the necessity of RBC transfusion or termination of chemotherapy. Results: The results of darbepoetin alfa treatment were assessed in a group of 98 subsequent pts, in the course of neadjuvant chemotherapy for EBC. The pts received the following chemotherapy regimens: AC/PCL (35%), AC/PCL + carboplatin (9%), TCH (33%), TCH-P (8%), other (15%). At the moment of the initiation of ESA administration, 18% pts were aged 65 or more (max 76) while 82% below 65 yrs. of age (min 28). In the majority (90) of pts the number of Aranesp® doses administered did not exceed 3, only 8 received 4 doses. The estimated effectiveness in the <65 vs ≥65 years of age groups was 84.8% vs 79.8%, respectively. The estimated effectiveness values in the following BMI-dependent groups, namely: underweight + normal weight (BMI: 16–24.99), overweight (BMI 25–29.99) and obesity (BMI 30–43.1), were 86.3%, 76.6% and 72.0%, respectively. Conclusion(s): Darbepoetin alfa proved effective in the treatment of anaemia in chemotherapy-treated pts for EBC. The response to the treatment in the assessed group of pts was 85.7% There were no statistically significant age and BMI-related differences in ESA effectiveness. No significant side effects of darbepoetin alfa therapy were observed. Conflict of Interest: No significant relationships.

15.
Journal of Investigative Medicine ; 70(2):461, 2022.
Article in English | EMBASE | ID: covidwho-1706882

ABSTRACT

Case Report The purpose of the study is to explore the possible diagnosis of Gaisbock in a patient with long-standing erythrocytosis and hypertension. Methods Used Case Study Summary of Results A 40-year-old Caucasian man with obesity was admitted with recurrent leg swelling and increasing oxygen requirements two weeks after hospitalization with COVID-19 pneumonia. Upon review of the patient's history, he was found to have untreated hypertension over several medical encounters and an erythrocytosis spanning ten years. Recent medical history included a diagnosis of deep vein thrombosis (DVT) in the same leg two and a half months prior and was treated with Xarelto. The patient reported a history of low testosterone for 12 years. However, he had not used any testosterone supplementation for the last nine months. He reported daytime fatigue, frequent bouts of nighttime awakenings, and frequent snoring. The patient never had a sleep study or used a CPAP. The patient used half a can of chewing tobacco daily for thirteen years, and he smoked one pack per day for ten years but quit 12 years ago. He worked strenuous jobs in the construction industry most of his life. On this admission, the patient's lab work was notable for hemoglobin of 18.7 gm/dL (13.7-17.5) and a normal erythropoietin level of 5.7 MIU/mL (2.6-18.5) without thrombocytosis or leukocytosis and a positive factor V Leiden mutation. His blood pressure was 132/91 mmHg. On review of previous records, the patient was found to have consistently elevated hemoglobin The patient had a stocky, ruddy appearance without hepatosplenomegaly. Conclusion Erythrocytosis can be categorized as primary, secondary, or relative. Patients with relative erythrocytosis have a decreased plasma volume with a relative increase in hemoglobin. Additionally, elevated hemoglobin levels have been associated with hypertension. Gaisbock's syndrome, first described in 1905, is characterized by hypertension and erythrocytosis without splenomegaly, leukocytosis, or thrombocytosis. It is associated with mild obesity, elevated blood pressure, and increased blood viscosity, which may explain why these patients often develop cardiovascular complications. Patients with relative erythrocytosis are at a higher risk for thromboembolic complications. In this case, Gaisbock's syndrome was suspected because the patient had had a stocky, plethoric appearance with persistently elevated hemoglobin and blood pressure with a normal erythropoietin level. Gaisbock's syndrome establishes a relationship between benign erythrocytosis, hypertension, and an increased risk for cardiovascular events. (Table Presented).

16.
Eur J Neurol ; 28(10): 3443-3447, 2021 Oct.
Article in English | MEDLINE | ID: covidwho-1607745

ABSTRACT

BACKGROUND AND PURPOSE: COVID-19 affects the brain in various ways, amongst which delirium is worrying. An assessment was made of whether a specific, long-lasting, COVID-19-related brain injury develops in acute respiratory distress syndrome patients after life-saving re-oxygenation. METHODS: Ten COVID+ patients (COVID+) with unusual delirium associated with neuroimaging suggestive of diffuse brain injury and seven controls with non-COVID encephalopathy were studied. The assessment took place when the intractable delirium started at weaning off ventilation support. Brain magnetic resonance imaging (MRI) was performed followed by standard cerebrospinal fluid (CSF) analyses and assessment of CSF erythropoietin concentrations (as a marker for the assessment of tissue repair), and of non-targeted CSF metabolomics using liquid chromatography high resolution mass spectrometry. RESULTS: Patients were similar as regards severity scores, but COVID+ were hospitalized longer (25 [11.75; 25] vs. 9 [4.5; 12.5] days, p = 0.03). On admission, but not at MRI and lumbar puncture performance, COVID+ were more hypoxic (p = 0.002). On MRI, there were leptomeningeal enhancement and diffuse white matter haemorrhages only in COVID+. In the latter, CSF erythropoietin concentration was lower (1.73 [1.6; 2.06] vs. 3.04 [2.9; 3.91] mIU/ml, p = 0.01), and CSF metabolomics indicated (a) increased compounds such as foodborne molecules (sesquiterpenes), molecules from industrialized beverages and micro-pollutants (diethanolamine); and (b) decreased molecules such as incomplete breakdown products of protein catabolism and foodborne molecules (glabridin). At 3-month discharge, fatigue, anxiety and depression as well as MRI lesions persisted in COVID+. CONCLUSIONS: Some COVID+ are at risk of a specific delirium. Imperfect brain repair after re-oxygenation and lifestyle factors might influence long-lasting brain injuries in a context of foodborne micro-pollutants.


Subject(s)
COVID-19 , Delirium , Environmental Pollutants , Brain/diagnostic imaging , Critical Care , Humans , SARS-CoV-2
17.
Blood ; 138:2195, 2021.
Article in English | EMBASE | ID: covidwho-1582436

ABSTRACT

Introduction: Aplastic anemia (AA) is a life-threatening disorder characterized by pancytopenia and a hypocellular bone marrow. Pure red cell aplasia (PRCA) is a similar disorder with primary reduction in the red blood cell population and virtual absence of erythroid precursors in the bone marrow. While the etiology of immune mediated marrow failure is multifactorial, preceding viral infections have been associated with the disease;these include parvovirus B19, cytomegalovirus, and Epstein-Barr virus. We present four cases of immune mediated marrow failure with either preceding or simultaneous SARS-CoV-2 infection. Methods: The medical records of patients treated for AA or PRCA at the University of Texas Southwestern Medical Center, Parkland Hospital, and the National Institutes of Health (NIH) were reviewed for SARS-CoV-2 infection. Four patients without prior hematological diseases were identified who had SARS-CoV-2 infection prior to or with simultaneous the diagnosis of AA or PRCA. Results: Patient #1 was a 22-year-old white female who was diagnosed with asymptomatic COVID-19 10 days prior to her pancytopenia and AA diagnosis was confirmed by bone marrow biopsy (5% cellularity;Table 1). Her extensive work-up including HIV, hepatitis panel, immunoglobulins, B12 and folate was negative, and she underwent HLA-matched family donor hematopoietic stem cell transplant. Patient #2 was a 69-year-old Asian female who presented to her primary care physician with symptoms of fatigue and was found to be pancytopenic. CBC from a few months prior was completely normal. Further work-up was positive for COVID-19 and negative for HIV, nutritional deficiency, or hemolysis. She did not have respiratory symptoms, was eventually diagnosed with pRBC and platelet transfusion-dependent severe AA (5-10% cellularity on bone marrow), and underwent treatment with cyclosporine, equine antithymocyte globulin, and eltrombopag. She has had a partial response to this therapy. Both patients had bone marrow specimens stained for SARS-CoV-2 by immunohistochemistry that were negative. Patient #3 was a 76-year-old white male who was diagnosed with COVID-19 4 months prior to presenting with a non-ST segment myocardial infarction and found to be profoundly anemic, requiring pRBC transfusion. He re-presented with chest pain one week later and was found to be anemic again, and required transfusion. A trial of darbepoetin alfa was unsuccessful. Extensive work-up for malignancy, infection, and autoimmune etiologies were negative. He was diagnosed with PRCA based on the bone marrow biopsy and initiated treatment with cyclosporine. Patient # 4 was diagnosed with severe AA (presenting as pancytopenia) and COVID-19 infection. He had fatigue for one month and fever, chills and sore throat one-week prior seeking medical care. Testing for hepatitis, HIV, EBV, and CMV was negative. He was treated on a clinical trial (NCT04304820) at NIH with cyclosporine and eltrombopag until SARS-CoV-2 PCR was negative then received equine anti-thymocyte globulin. He has achieved a complete hematologic response at 6 months and remains well at last follow-up. Conclusion: The four patients described had minimal respiratory COVID-19 symptoms, but they presented with cytopenia and were eventually diagnosed with bone marrow failure. It is possible that this is co-incidental due to the high prevalence of SARS-CoV-2. However, there is emerging evidence that COVID-19 pneumonia is a hyperinflammatory and immune dysregulated state improved by dexamethasone therapy. Other immune mediated hematologic conditions, such as autoimmune hemolytic anemia and immune thrombocytopenia, have been reported. The onset from infection to cytopenia appears rapid, although patients often presented with symptoms for many days prior to diagnosis and thus testing may have been delayed from the onset of infection. This case series does not provide a mechanistic link between SARS-CoV-2 infection and bone marrow failure, but it raises the possibility that SARS-CoV-2 may mediate an immunologic response that cont ibutes to marrow failure. Patients appear to respond well to standard immunosuppressive treatment. Further cases and studies are needed to determine if this is directly linked to SARS-CoV-2 and whether the natural history and response to standard therapy is different than idiopathic cases. [Formula presented] Disclosures: Young: Novartis: Research Funding.

18.
Blood ; 138:2180, 2021.
Article in English | EMBASE | ID: covidwho-1582290

ABSTRACT

SARS-CoV-2 infection and vaccination have raised concerns in paroxysmal nocturnal hemoglobinuria (PNH). In fact, PNH patients carry an increased infectious risk secondary to complement inhibition treatment or associated bone marrow failure (BMF), and may therefore benefit from preventive strategies such as vaccinations. On the contrary, vaccines can be numbered among inflammatory complement amplifiers (e.g., infections, traumas, surgery), potentially triggering a disease exacerbation. In PNH patients on complement inhibitors, this phenomenon has been defined pharmacodynamic breakthrough hemolysis (BTH). Based on isolated reports of BTH following SARS-CoV-2 vaccines, we conducted a survey among 5 Italian reference centers to evaluate complications and BTH occurrence in PNH patients who completed the SARS-CoV-2 vaccination schedule from January, 2 2021 until the time of writing. Adverse events, hematologic and hemolytic parameters were recorded within 7-10 days before and after each dose of vaccine. A total of 67 patients (females/males 43/24, median age 47.6 years, range 21-90.5) were eligible for the analysis. According to the International PNH Interest Group classification, 45 patients suffered from hemolytic PNH, 20 from PNH in the context of BMF syndromes (aplastic anemia or myelodysplastic syndrome), and 2 from subclinical PNH. Fifty-five subjects (82%) were on regular complement inhibition therapy, i.e., eculizumab (N=35), ravulizumab (N=13), subcutaneous anti-C5 (N=3), anti-factor B (N=2) and ravulizumab + anti-factor D combination (N=2). Vaccines (Comirnaty/Pfizer-BioNTech N=53, mRNA-1273/Moderna N=12, and ChAdOx1 nCov-19/AstraZeneca N=2) were complessively well-tolerated, with 3 non-hematologic adverse events after the first dose (2 fever and 1 exercise-induced tachycardia, grade 1 according to CTCAE v5.0) and 2 after the second one (fever, accompanied by vomit in one patient, grade 1). During the observation period, 3 BTH and 1 hemolytic exacerbation were recorded (5.9% of patients), as detailed in Table 1. The most severe episode occurred in a young woman (Patient 3) on subcutaneous ravulizumab who experienced a hemoglobin (Hb) drop >2 g/dL, marked clinical signs of intravascular hemolysis and lactate dehydrogenase (LDH) increase >1.5 x upper limit of normal (ULN) from baseline, which is considered a clinical BTH according to the criteria proposed by the Severe Aplastic Anemia Working Party of the European group for Bone Marrow Transplantation. The patient required hospitalization for additional treatment with recombinant erythropoietin and anti-thombotic/bacterial prophylaxis. The second more severe BTH was registered in a male patient (Patient 1) on oral anti-factor B who experienced a Hb drop >2 g/dL without an overt hemolytic flare, and required hospitalization for intravenous antibiotic therapy (concomitant urinary tract infection). The remaining two patients experienced a subclinical BTH (Patient 2) and a hemolytic flare (Patient 4, not on complement inhibition). On the whole, a median delta variation from usual values of Hb and LDH of -25% (range -26+3%) and +80% (+18+105%) were observed, respectively. Of note, 3 episodes occurred after the second dose of vaccine, generally within 24-48 hours. Anti-complement drugs were not modified/discontinued in any of the 3 patients on regular treatment. Patients not experiencing BTH (94.1%) showed stable hematologic parameters after the first dose (Hb/LDH median delta variations from baseline -1%/+1%, range -14+12%/-32+40%) and the second dose of vaccine (Hb/LDH median delta variations from baseline +1%/0%, range -18+47%/-76+41%). Of note, 4 patients with a previous SARS-CoV-2 infection completed the vaccination without any complication/PNH exacerbation. In conclusion, this survey shows that BTH/hemolytic flares following SARS-CoV-2 vaccines are observed in about 6% of PNH patients, may be clinically relevant but manageable, and should not discourage vaccination. BTH has been registered mostly few days after the second dose of vaccine, suggesting a “booster⠝ effect favoring a higher inflammatory response. Watchful clinical and laboratory monitoring is advised, in order to promptly recognize severe hemolytic flares in both treated and naïve patients. [Formula presented] Disclosures: Fattizzo: Novartis: Speakers Bureau;Kira: Speakers Bureau;Alexion: Speakers Bureau;Annexon: Consultancy;Momenta: Honoraria, Speakers Bureau;Apellis: Speakers Bureau;Amgen: Honoraria, Speakers Bureau. Bianchi: Agios pharmaceutics: Consultancy, Membership on an entity's Board of Directors or advisory committees. Sica: Jazz Pharma: Consultancy;Alexion: Consultancy. Barcellini: Novartis: Other: Invited speaker, Research Funding;Agios: Membership on an entity's Board of Directors or advisory committees;Alexion Pharmaceuticals, Inc: Membership on an entity's Board of Directors or advisory committees, Other: Invited speaker, Research Funding;Bioverativ: Membership on an entity's Board of Directors or advisory committees;Incyte: Membership on an entity's Board of Directors or advisory committees.

19.
Blood ; 138:4573, 2021.
Article in English | EMBASE | ID: covidwho-1582276

ABSTRACT

Introduction:Plasmablastic lymphoma (PBL) is a rare and aggressive lymphoma most commonly seen in the setting of chronic immunosuppression, such as HIV infection and organ transplantation, or in patients with pre-existing lymphoproliferative or autoimmune disorders. PBL commonly presents at extranodal sites and carries a poor prognosis with a global overall survival of 9-15 months after initial diagnosis. Despite poor prognosis for patients with PBL, therapeutic strategies to target this disease are limited, as CHOP-like regimens have failed to produce durable remission, and no standard of care has been established. The cell of origin for PBL is believed to be the plasmablast, as PBL cells possess immunoblastic morphology and contain an immunohistochemical profile positive for plasmablast markers, such as CD38, CD138, and MUM1/IRF4, and negative for B cell markers, such as CD20, CD19, and PAX5. The similarities between PBL cells and multiple myeloma (MM) cells, a plasma cell neoplasm, have led to investigations of the efficacy of MM therapeutics for the treatment of PBL. Daratumumab is a first-in-class monoclonal antibody directed against CD38 that has shown efficacy in treating relapsed/refractory and newly diagnosed MM. Here, we describe the treatment of four patients with advanced-stage PBL in the context of varying degrees of immunosuppression using combination treatment with daratumumab and EPOCH. Methods:Four consecutive patients were treated with daratumumab plus chemotherapy. Three of the four patients were treated in the frontline setting and received low-dose EPOCH (vincristine, doxorubicin, etoposide daily for 4 days;cyclophosphamide day 5) with 16mg/kg daratumumab dosed on days 1 and 8 for 6 cycles. Patient #4 showed partial CD20 positivity, prompting Rituximab co-administration (R-EPOCH). Patient #3 was treated in the relapsed setting with 16mg/kg daratumumab in combination with lenalidomide, dexamethasone, and doxorubicin. She was treated for 12 months. Responses were followed by PET/CT imaging. Results:The four consecutive patients (2 female, 2 male) ranged in age from 26-88 and all had advanced-stage PBL (Table 1). Three of the four patients had some degree of immunosuppression (Patient #1- post-transplantation lymphoproliferative disorder (PTLD), Patient #2- HIV/AIDS, Patient #3- Chron's disease, Patient #4- no history of immunosuppression). All patients had a Ki67 proliferation index over 70% and demonstrated extranodal involvement of disease (bone n=3, intestine n=2, liver n=2;kidney n=1;adrenal n=1). The three patients that received daratumumab in combination with EPOCH demonstrated a complete response at their first disease assessment by PET/CT scan after cycle 2 (Patient #1) or cycle 4 (Patients #2,4) (Fig1). Patient #3, who demonstrated a mixed response to previous therapy, achieved a complete response 5 months after starting treatment with daratumumab in combination with chemotherapy. As of July 2021, three of the four patients continued to have no evidence of disease for a median of 17 months (range 15-19 months). Patient #4 relapsed in July 2021, 3 months after demonstrating a complete response. Adverse events that required hospitalization were rarely noted following daratumumab treatment but included neutropenic fever (n=2, one event following treatment cycle 4 and the other event 2 months after completion of daratumumab administration), COVID-19 infection (n=1), and a PICC line-associated thrombus (n=1). Minor events were also noted and included self-limiting bradycardia (n=1), neuropathy (n=1), and rigor (n=1). Conclusions:We describe four patients with varying degrees of immunosuppression and HIV-status with aggressive-stage PBL that achieved complete response following treatment with daratumumab in combination with low-dose EPOCH or other chemotherapy. Three of four patients obtained durable responses. At the time of this writing, three additional HIV +patients have initiated treatment with daratumumab but have not yet reached their first disease assessment. Disease progression for these patients will be monitored and presented as part of this study. Our findings suggest a potential efficacy and warrant further investigation of using daratumumab for the treatment of HIV +and HIV - PBLs. [Formula presented] Disclosures: Amengual: Epizyme, Inc.: Speakers Bureau;Appia Pharmaceuticals: Research Funding;Daiichi Sankyo, Inc: Consultancy;Seagen: Consultancy. OffLabel Disclosure: our work uses daratumumab, a first-in-class monoclonal antibody against CD38 for the treatment of plasmablastic lymphoma

20.
World J Stem Cells ; 13(10): 1513-1529, 2021 Oct 26.
Article in English | MEDLINE | ID: covidwho-1524342

ABSTRACT

Erythropoietin (EPO) is the main mediator of erythropoiesis and an important tissue protective hormone that appears to mediate an ancestral neuroprotective innate immune response mechanism at an early age. When the young brain is threatened-prematurity, neonatal hyperbilirubinemia, malaria- EPO is hyper-secreted disproportionately to any concurrent anemic stimuli. Under eons of severe malarial selection pressure, neuroprotective EPO augmenting genetic determinants such as the various hemoglobinopathies, and the angiotensin converting enzyme (ACE) I/D polymorphism, have been positively selected. When malarial and other cerebral threats abate and the young child survives to adulthood, EPO subsides. Sustained high ACE and angiotensin II (Ang II) levels through the ACE D allele in adulthood may then become detrimental as witnessed by epidemiological studies. The ubiquitous renin angiotensin system (RAS) influences the α-klotho/fibroblast growth factor 23 (FGF23) circuitry, and both are interconnected with EPO. Here we propose that at a young age, EPO augmenting genetic determinants through ACE D allele elevated Ang II levels in some or HbE/beta thalassemia in others would increase EPO levels and shield against coronavirus disease 2019, akin to protection from malaria and dengue fever. Human evolution may use ACE2 as a "bait" for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) to gain cellular entry in order to trigger an ACE/ACE2 imbalance and stimulate EPO hypersecretion using tissue RAS, uncoupled from hemoglobin levels. In subjects without EPO augmenting genetic determinants at any age, ACE2 binding and internalization upon SARS-CoV-2 entry would trigger an ACE/ACE2 imbalance, and Ang II oversecretion leading to protective EPO stimulation. In children, low nasal ACE2 Levels would beneficially augment this imbalance, especially for those without protective genetic determinants. On the other hand, in predisposed adults with the ACE D allele, ACE/ACE2 imbalance, may lead to uncontrolled RAS overactivity and an Ang II induced proinflammatory state and immune dysregulation, with interleukin 6 (IL-6), plasminogen activator inhibitor, and FGF23 elevations. IL-6 induced EPO suppression, aggravated through co-morbidities such as hypertension, diabetes, obesity, and RAS pharmacological interventions may potentially lead to acute respiratory distress syndrome, cytokine storm and/or autoimmunity. HbE/beta thalassemia carriers would enjoy protection at any age as their EPO stimulation is uncoupled from the RAS system. The timely use of rhEPO, EPO analogs, acetylsalicylic acid, bioactive lipids, or FGF23 antagonists in genetically predisposed individuals may counteract those detrimental effects.

SELECTION OF CITATIONS
SEARCH DETAIL