Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (43)

Search Parameters:
Keywords = vaso-occlusive crisis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
9 pages, 523 KiB  
Article
The Direct Medical Costs of Sickle Cell Disease in Saudi Arabia: Insights from a Single Center Study
by Yazed AlRuthia
Healthcare 2025, 13(4), 420; https://doi.org/10.3390/healthcare13040420 - 15 Feb 2025
Abstract
Background: Sickle cell disease (SCD) is a rare autosomal recessive disorder that is common in countries with consanguineous marriages. It leads to various complications, including painful episodes, infections, delayed growth, stroke, and organ damage, which contribute to high healthcare utilization and costs. [...] Read more.
Background: Sickle cell disease (SCD) is a rare autosomal recessive disorder that is common in countries with consanguineous marriages. It leads to various complications, including painful episodes, infections, delayed growth, stroke, and organ damage, which contribute to high healthcare utilization and costs. In Saudi Arabia, the prevalence of SCD is notably high, largely due to the frequency of consanguineous marriages. However, there has not yet been a study estimating the direct medical costs of managing SCD based on real-world data. This study aims to assess these costs in Saudi Arabia. Methods: Data were collected from electronic medical records (EMRs) at a university-affiliated tertiary care center. A micro-costing approach was used to estimate the direct medical costs (e.g., laboratory tests, imaging, emergency department visits, hospitalizations, prescription medications, outpatient visits, etc.) retrospectively over a 12-month follow-up period. The baseline characteristics of the patients were presented using frequencies and percentages. The costs of different healthcare services were analyzed using means and the 95% confidence intervals. A generalized linear model (GLM) with a gamma distribution was utilized to examine the association between the overall costs and patient characteristics (e.g., age, gender, duration of illness, surgeries, blood transfusions, etc.), allowing for the estimation of the adjusted mean costs. Results: A total of 100 patients met the inclusion criteria and were included in the analysis. The mean age of the patients was 10.21 years (±6.87 years); 53% were male, and a substantial majority (96%) had the HbSS genotype. Sixty-one percent of the patients had undergone at least one red blood cell (RBC) exchange transfusion, while 21% had undergone surgical procedures, including tonsillectomy, splenectomy, and cholecystectomy. Additionally, 45% had experienced at least one vaso-occlusive crisis (VOC), and 59% had been hospitalized at least once in the past 12 months. Factors such as the frequency of laboratory tests and imaging studies, the length of hospital stay (LOS), the rate of emergency department (ED) visits, surgical procedures, the number of prescription medications, and the frequency of blood transfusions were all significant predictors of higher direct medical costs (p < 0.05). The estimated mean annual direct medical costs per patient were USD 26,626.45 (95% CI: USD 22,716.89–USD 30,536.00). After adjusting for various factors, including age, gender, duration of illness, frequency of lab and imaging tests, LOS, ED visits, surgical procedures, number of prescription medications, rates of VOCs, and RBC exchange transfusions, the adjusted mean annual direct medical cost per patient was calculated to be USD 14,604.72 (95% CI: USD 10,943.49–USD 19,525.96). Conclusions: The results of this study emphasize the substantial direct medical costs linked to sickle cell disease (SCD), which are greatly affected by the frequency of related complications. These insights should motivate policymakers and healthcare researchers to assess both the national direct and indirect costs associated with SCD, especially given the significant number of SCD patients in Saudi Arabia. Full article
Show Figures

Figure 1

Figure 1
<p>The percentages of different classes of direct medical costs for SCD management.</p>
Full article ">
10 pages, 2336 KiB  
Review
The Management of Postpartum Cardiorespiratory Failure in a Patient with COVID-19 and Sickle Cell Trait Requiring Extraorporeal Membrane Oxygenation Support and Airflight Transportation
by Alexandre Pelouze, Sylvain Massias, Diae El Manser, Adrien Koeltz, Patricia Shri Balram Christophe, Mohamed Soualhi and Marc Licker
J. Clin. Med. 2025, 14(1), 213; https://doi.org/10.3390/jcm14010213 - 2 Jan 2025
Viewed by 672
Abstract
Acute cardiovascular disorders are incriminated in up to 33% of maternal deaths, and the presence of sickle cell anemia (SCA) aggravates the risk of peripartum complications. Herein, we present a 24-year-old Caribbean woman with known SCA who developed a vaso-occlusive crisis at 36 [...] Read more.
Acute cardiovascular disorders are incriminated in up to 33% of maternal deaths, and the presence of sickle cell anemia (SCA) aggravates the risk of peripartum complications. Herein, we present a 24-year-old Caribbean woman with known SCA who developed a vaso-occlusive crisis at 36 weeks of gestation that required emergency Cesarean section. In the early postpartum period, she experienced fever with rapid onset of acute respiratory distress in the context of COVID-19 infection that required tracheal intubation and mechanical ventilatory support with broad-spectrum antibiotics and blood exchange transfusion. Shortly thereafter, transthoracic echocardiography documented severe biventricular dysfunction associated with raising levels of cardiac troponin and ECG signs of myocardial ischemia. Medical treatment with incremental dobutamine and noradrenaline infusion failed to improve cardiac output and blood gas exchange. After consultation with the regional cardiac center, a prompt decision was made to provide cardiac and respiratory support via implantation of femoral cannula and initiation of veno-arterial extracorporeal membrane oxygenation (ECMO, Cardiohelp®). Under stable ECMO, the patient was transferred by helicopter to a specialized cardiac center. There were no signs of ongoing hemolysis, and progressive recovery of the right and left ventricular function facilitated forward blood flow through the aortic valve. Three days after implantation, ECMO was weaned, and the cannula were removed. One day later, the patient’s chest X-rays showed partial resolution of lung edema. The patient was successfully extubated, and non-invasive ventilation with pulmonary rehabilitation was initiated to speed up her functional recovery. Full article
(This article belongs to the Special Issue Clinical Advances in Cardiac Anesthesia and Critical Care)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Chest X-rays showing persistent lung edema with bilateral infiltrates under mechanical ventilation and extracorporeal membrane oxygenation; (<b>B</b>) chest X-rays showing partial regression of lung edema and improved aeration on spontaneous ventilation after tracheal extubation.</p>
Full article ">Figure 2
<p>Timeline of key events (Cesarian section, respiratory failure and heart failure) and therapeutic interventions from hospital admission to weaning extracorporeal membrane oxygenation (ECMO) and mechanical ventilation followed by airflight transportation back to the referaal hospital.</p>
Full article ">
25 pages, 373 KiB  
Review
Emergencies in Hematology: Why, When and How I Treat?
by Andrea Duminuco, Vittorio Del Fabro, Paola De Luca, Dario Leotta, Miriana Carmela Limoli, Ermelinda Longo, Antonella Nardo, Gabriella Santuccio, Alessandro Petronaci, Gaia Stanzione, Francesco Di Raimondo and Giuseppe Alberto Palumbo
J. Clin. Med. 2024, 13(24), 7572; https://doi.org/10.3390/jcm13247572 - 12 Dec 2024
Cited by 1 | Viewed by 3517
Abstract
Hematological emergencies are critical medical conditions that require immediate attention due to their rapid progression and life-threatening nature. As various examples, hypercalcemia, often associated with cancers such as multiple myeloma, can lead to severe neurological and cardiac dysfunction. Hyperleukocytosis, common in acute myeloid [...] Read more.
Hematological emergencies are critical medical conditions that require immediate attention due to their rapid progression and life-threatening nature. As various examples, hypercalcemia, often associated with cancers such as multiple myeloma, can lead to severe neurological and cardiac dysfunction. Hyperleukocytosis, common in acute myeloid leukemias, increases the risk of leukostasis and multiorgan failure. Sickle cell crisis, a common complication in sickle cell disease, results from vaso-occlusion, leading to acute pain and tissue ischemia. Tumor lysis syndrome, reported in cases of rapid destruction of cancer cells, causes electrolyte imbalances and acute kidney injury. Acute transfusion reactions, fundamental in hematological conditions, can range from mild allergic responses to severe hemolysis and shock, requiring prompt management. Disseminated intravascular coagulation, involving excessive coagulation and bleeding, is commonly triggered by hematological malignancies, common in the first phases of acute promyelocytic leukemia. Recently, in the era of bispecific antibodies and chimeric antigen receptor T cells, cytokine release syndrome is a manifestation that must be recognized and promptly treated. Understanding the pathophysiology, recognizing the clinical manifestations, and ensuring adequate diagnostic strategies and management approaches for each condition are central to early intervention in improving patient outcomes and reducing mortality. Full article
(This article belongs to the Section Hematology)
17 pages, 2084 KiB  
Article
Newborn Screening for Sickle Cell Disease in Catalonia between 2015 and 2022—Epidemiology and Impact on Clinical Events
by José Manuel González de Aledo-Castillo, Ana Argudo-Ramírez, David Beneitez-Pastor, Anna Collado-Gimbert, Francisco Almazán Castro, Sílvia Roig-Bosch, Anna Andrés-Masó, Anna Ruiz-Llobet, Georgina Pedrals-Portabella, David Medina-Santamaria, Gemma Nadal-Rey, Marina Espigares-Salvia, Maria Teresa Coll-Sibina, Marcelina Algar-Serrano, Montserrat Torrent-Español, Pilar Leoz-Allegretti, Anabel Rodríguez-Pebé, Marta García-Bernal, Elisabet Solà-Segura, Amparo García-Gallego, Blanca Prats-Viedma, Rosa María López-Galera, Abraham J. Paredes-Fuentes, Sonia Pajares García, Giovanna Delgado-López, Adoración Blanco-Álvarez, Bárbara Tazón-Vega, Cristina Díaz de Heredia, María del Mar Mañú-Pereira, José Luis Marín-Soria, Judit García-Villoria, Pablo Velasco-Puyó and on behalf of the Sickle Cell Disease Newborn Screening Group of Cataloniaadd Show full author list remove Hide full author list
Int. J. Neonatal Screen. 2024, 10(4), 69; https://doi.org/10.3390/ijns10040069 - 3 Oct 2024
Viewed by 1930
Abstract
In 2015, Catalonia introduced sickle cell disease (SCD) screening in its newborn screening (NBS) program along with standard-of-care treatments like penicillin, hydroxyurea, and anti-pneumococcal vaccination. Few studies have assessed the clinical impact of introducing NBS programs on SCD patients. We analyzed the incidence [...] Read more.
In 2015, Catalonia introduced sickle cell disease (SCD) screening in its newborn screening (NBS) program along with standard-of-care treatments like penicillin, hydroxyurea, and anti-pneumococcal vaccination. Few studies have assessed the clinical impact of introducing NBS programs on SCD patients. We analyzed the incidence of SCD and related hemoglobinopathies in Catalonia and the change in clinical events occurring after introducing NBS. Screening 506,996 newborns from 2015 to 2022, we conducted a retrospective multicenter study including 100 screened (SG) and 95 unscreened (UG) SCD patients and analyzed SCD-related clinical events over the first six years of life. We diagnosed 160 cases of SCD, with an incidence of 1 in 3169 newborns. The SG had a significantly lower median age at diagnosis (0.1 y vs. 1.68 y, p < 0.0001), and initiated penicillin prophylaxis (0.12 y vs. 1.86 y, p < 0.0001) and hydroxyurea treatment earlier (1.42 y vs. 4.5 y, p < 0.0001). The SG experienced fewer median SCD-related clinical events (vaso-occlusive crisis, acute chest syndrome, infections of probable bacterial origin, acute anemia requiring transfusion, acute splenic sequestration, and pathological transcranial Doppler echography) per year of follow-up (0.19 vs. 0.77, p < 0.0001), a reduced number of annual emergency department visits (0.37 vs. 0.76, p < 0.0001), and fewer hospitalizations (0.33 vs. 0.72, p < 0.0001). SCD screening in Catalonia’s NBS program has effectively reduced morbidity and improved affected children’s quality of life. Full article
Show Figures

Figure 1

Figure 1
<p>Catalonian NBS process. DBS: dried blood spots; SCD: sickle cell disease; CRU: Clinical Reference Unit; HPLC: high-performance liquid chromatography.</p>
Full article ">Figure 2
<p>Clinical events by year of follow-up. SG: screened group; UG: unscreened group; Total: total clinical events; SCD-related: sickle cell disease-related clinical events; ER: visits to the emergency department; HOS: hospitalizations. The level of statistical significance is indicated by asterisks: ** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.00001.</p>
Full article ">Figure 3
<p>Mean number of SCD-related clinical events by year of age in the study cohorts. SG: screened group; UG: unscreened group. The level of statistical significance is indicated by asterisks: * for <span class="html-italic">p</span> &lt; 0.05, ** for <span class="html-italic">p</span> &lt; 0.01, and *** for <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Clinical events by genotype in the study cohorts. SG: screening group; UG: unscreened group; Total: total clinical events; SCD-related: sickle cell disease-related clinical events; ER: visits to the emergency department; HOS: hospitalizations. The level of statistical significance is indicated by asterisks: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01, ns (not significant). Genotypes: SS (HbSS); SC(HbSC); Sβ<sup>0</sup>(HBSβ<sup>0</sup>).</p>
Full article ">Figure 5
<p>Impact of hydroxyurea treatment on the events in both the UG and SG. UG-PreHU: unscreened group pre-hydroxyurea; UG-PostHU: unscreened group post-hydroxyurea; SG-PreHU: screened group pre-hydroxyurea; SG-PostHU: screened group post-hydorxyurea; Total: total clinical events; SCD-related: sickle cell disease-related clinical events; ER: visits to the emergency department; HOS: hospitalizations. The level of statistical significance is indicated by asterisks: **** <span class="html-italic">p</span> &lt; 0.0001, ns (not significant).</p>
Full article ">Figure 6
<p>Kaplan–Meier curves for event-free survival by specific events in the SG and UG cohorts. Each graph represents the six-year survival estimate since birth without the corresponding SCD-related event. SG: screened group; UG: unscreened group. (<b>a</b>) Six-year Kaplan–Meier estimate without vaso-occlusive crisis (VOC) was different in both groups (57.0% vs. 30.3%, <span class="html-italic">p</span> = 0.03). (<b>b</b>) Six-year Kaplan–Meier estimate without acute chest syndrome (ACS) was not different in both groups (73.5% vs. 54.3%, <span class="html-italic">p</span> = 0.06). (<b>c</b>) Six-year Kaplan–Meier estimate without infections of probable bacterial origin (BI) was not different in both groups (71.3% vs. 69.8% <span class="html-italic">p</span> = 1.0). (<b>d</b>) Six-year Kaplan–Meier estimate without infections of probable acute anemia requiring transfusion (TRF) was not different in both groups (64.5% vs. 69.7%, <span class="html-italic">p</span> = 0.9). (<b>e</b>) Six-year Kaplan–Meier estimate without acute splenic sequestration (ASSC) was not different in both groups (93.8% vs. 85.0%, <span class="html-italic">p</span> = 0.12).</p>
Full article ">
14 pages, 25497 KiB  
Article
GBT1118, a Voxelotor Analog, Ameliorates Hepatopathy in Sickle Cell Disease
by Elio Haroun, Seah H. Lim and Dibyendu Dutta
Medicina 2024, 60(10), 1581; https://doi.org/10.3390/medicina60101581 - 26 Sep 2024
Viewed by 1089
Abstract
Background and Objectives: In sickle cell disease (SCD), hepatopathy is a cumulative consequence of ischemia/reperfusion (I/R) injury from a vaso-occlusive crisis, tissue inflammation, and iron overload due to blood transfusion. Hepatopathy is a major contributing factor of shortened life span in SCD patients. [...] Read more.
Background and Objectives: In sickle cell disease (SCD), hepatopathy is a cumulative consequence of ischemia/reperfusion (I/R) injury from a vaso-occlusive crisis, tissue inflammation, and iron overload due to blood transfusion. Hepatopathy is a major contributing factor of shortened life span in SCD patients. We hypothesized that the voxelotor, a hemoglobin allosteric modifier, ameliorates sickle hepatopathy. Materials and Methods: Townes SCD mice and their controls were treated with either chow containing GBT1118, a voxelotor analog, or normal chow. We evaluated inflammation, fibrosis, apoptosis and ferroptosis in their livers using qPCR, ELISA, histology, and immunohistochemistry. Results: GBT1118 treatment resulted in reduced hemolysis, iron overload and inflammation in the liver of SCD mice. There were significant reductions in the liver enzyme levels and bile acids. Furthermore, GBT1118-treated mice exhibited reduced apoptosis, necrosis, and fibrosis. Increased ferroptosis as evident from elevated 4-hydroxynonenal (4-HNE) staining, malondialdehyde (MDA) levels, and expression of Ptgs2 and Slc7a11 mRNAs, were also significantly reduced after GBT1118 treatment. To explain the increased ferroptosis, we evaluated iron homeostasis markers in livers. SCD mice showed decreased expression of heme oxygenase-1, ferritin, hepcidin, and ferroportin mRNA levels. GBT1118 treatment significantly increased expressions of these genes. Conclusions: Our results suggest GBT1118 treatment in SCD confers the amelioration of sickle hepatopathy by reducing inflammation, fibrosis, apoptosis, iron overload and ferroptosis. Full article
(This article belongs to the Section Gastroenterology & Hepatology)
Show Figures

Figure 1

Figure 1
<p>Treatment schema. Non-SCD or SCD Townes were randomized and treated either with control chow or chow containing GBT1118 for 120 days. Mice were kept under strict water deprivation for 12 h for seven consecutive nights every three weeks starting from Week 3 to induce VOC.</p>
Full article ">Figure 2
<p>Hemoglobin (Hb) levels, red blood cell (RBC) counts, hematocrit (HCT) percentage, total bilirubin (TBIL) concentration and spleen weight and size at Day 120. N = 6, (***) <span class="html-italic">p</span> &lt; 0.001 for non-SCD compared to SCD and GBT1118. (##) <span class="html-italic">p</span> &lt; 0.01, (###) <span class="html-italic">p</span> &lt; 0.001 for GBT1118 compared to SCD using 2-way repeated measures ANOVA with Bonferroni post hoc multiple comparison test.</p>
Full article ">Figure 3
<p>Liver weights in non-SCD, SCD and GBT1118-treated SCD mice after 4 months of treatment, and levels of alkaline phosphatase (ALP), alanine transaminase (ALT), and bile acid at baseline, 2 months and 4 months. N = 6, (*) <span class="html-italic">p</span> &lt; 0.05, (**) <span class="html-italic">p</span> &lt; 0.01, (***) <span class="html-italic">p</span> &lt; 0.001 for non-SCD compared to SCD and GBT1118. (#) <span class="html-italic">p</span> &lt; 0.05, (###) <span class="html-italic">p</span> &lt; 0.001 for GBT1118 compared to SCD using 2-way repeated measures ANOVA with Bonferroni post hoc multiple comparison test.</p>
Full article ">Figure 4
<p>(<b>A</b>) Leukocyte infiltration, and (<b>B</b>) necrosis area in liver tissues, quantified by hematoxylin and eosin (H&amp;E) staining. Area within yellow-colored dashed line shows leukocyte infiltration, and area within white-colored dashed line shows necrosis. Scale bar 100 μm; (<b>C</b>) inflammatory cell marker infiltrated in the liver quantified by qPCR using mRNAs for lymphocyte antigen 6 family member G (<span class="html-italic">Ly6g</span>), monocyte chemoattractant protein 1 (<span class="html-italic">Mcp1</span>), and adhesion G protein-coupled receptor E1 (<span class="html-italic">Adgre1)</span> gene encoding F4/80 protein in non-SCD, SCD and GBT1118-treated SCD mice at 4 months post-treatment. qPCR mRNA values normalized to 18S rRNA and compared using 2-way repeated measures ANOVA with Bonferroni post hoc multiple comparison test. Necrosis area percentage was evaluated using Mann–Whitney test. N = 6, (***) <span class="html-italic">p</span> &lt; 0.001, for non-SCD compared to SCD and GBT1118. (###) <span class="html-italic">p</span> &lt; 0.001 for GBT1118 compared to SCD.</p>
Full article ">Figure 5
<p>Serum hemopexin concentration, and expression of mRNAs for genes regulating iron homeostasis in the liver, transferrin (<span class="html-italic">Trf</span>), transferrin receptor 1 (<span class="html-italic">Tfr1</span>), heme oxygenase 1 (<span class="html-italic">Hmox1</span>), ferritin L (<span class="html-italic">Ftl</span>) and ferritin H (<span class="html-italic">Fth</span>) hepcidin (<span class="html-italic">Hamp1</span>), ferroportin 1a (<span class="html-italic">Fpn1a</span>) and ferroportin 1b (<span class="html-italic">Fpn1b</span>). qPCR mRNA values normalized to 18S rRNA using 2-way repeated measures ANOVA with Bonferroni post hoc multiple comparison test. N = 6, (***) <span class="html-italic">p</span> &lt; 0.001 for non-SCD compared to SCD and GBT1118. (#) <span class="html-italic">p</span> &lt; 0.05, (###) <span class="html-italic">p</span> &lt; 0.001 for GBT1118 compared to SCD.</p>
Full article ">Figure 6
<p>Apoptosis quantified by <span class="html-italic">TUNEL</span> staining in liver tissues. Apoptotic nuclei (brown stain) detected using 3,3-diaminobenzidine (DAB). Tissues counterstained with methyl green. Scale bar, 100 μm; fibrosis quantified by Masson’s trichrome staining in liver tissues. Scale bar, 400 μm. N = 6, for non-SCD compared to SCD and GBT1118. (#) <span class="html-italic">p</span> &lt; 0.05, (###) <span class="html-italic">p</span> &lt; 0.001 for GBT1118 compared to SCD using Mann–Whitney test.</p>
Full article ">Figure 7
<p>Perls Prussian blue stain in liver sections. Scale bar, 400 µm, and quantification of iron deposit area percentage; 4-hydroxynonenal (4-HNE) in the liver. Scale bar: 100 μm. Quantification of 4-HNE stained cells, <span class="html-italic">Malondialdehyde</span> (MDA) concentration, and expression of ferroptotic markers <span class="html-italic">Ptgs2</span> and <span class="html-italic">Slc7a11</span> mRNAs in the liver. qPCR mRNA values normalized to 18S rRNA and compared using 2-way repeated measures ANOVA with Bonferroni post hoc multiple comparison test. Iron deposit area percentage was evaluated using Mann–Whitney test N = 6, (*) <span class="html-italic">p</span> &lt; 0.05 (**) <span class="html-italic">p</span> &lt; 0.01, (***) <span class="html-italic">p</span> &lt; 0.001 for non-SCD compared to SCD and GBT1118. (##) <span class="html-italic">p</span> &lt; 0.01, (###) <span class="html-italic">p</span> &lt; 0.001 for GBT1118 compared to SCD.</p>
Full article ">
12 pages, 2178 KiB  
Review
Inpatient Management of Pain Episodes in Children with Sickle Cell Disease: A Review
by Zhour Barnawi, Ronay Thomas, Radhika Peddinti and Nabil Abou Baker
Children 2024, 11(9), 1106; https://doi.org/10.3390/children11091106 - 10 Sep 2024
Cited by 1 | Viewed by 1701
Abstract
Sickle cell disease (SCD) is the most common hemoglobinopathy in the world. Sickle cell vaso-occlusive episodes (VOEs) are very painful acute events and the most common complication as well as reason for hospitalization. SCD pain is best evaluated holistically with a pain functional [...] Read more.
Sickle cell disease (SCD) is the most common hemoglobinopathy in the world. Sickle cell vaso-occlusive episodes (VOEs) are very painful acute events and the most common complication as well as reason for hospitalization. SCD pain is best evaluated holistically with a pain functional assessment to aid in focusing pain management on reducing pain in addition to improving function. Patients with SCD have long endured structural racism and negative implicit bias surrounding the management of pain. Thus, it is important to approach the management of inpatient pain systematically with the use of multi-modal medications and nonpharmacologic treatments. Furthermore, equitable pain management care can be better achieved with standardized pain plans for an entire system and individualized pain plans for patients who fall outside the scope of the standardized pain plans. In this article, we discuss the best practices to manage SCD VOEs during an inpatient hospitalization. Full article
(This article belongs to the Special Issue Sickle Cell Disease in Infancy and Childhood)
Show Figures

Figure 1

Figure 1
<p>This figure shows the pain’s pathway from the peripheral nerve to the brain. During moments of factors that trigger pain, mast cells activate and release neuropeptide substance P. There are increased cytokines such as tumor necrosis factor-alpha (TNFα), interleukin 6 (IL-6), and prostaglandin E1 (PGE1). In turn, this initiates pain signaling down the dorsal root ganglion and spinal cord to the brain. In central sensitization, there is increased reactive oxygen species, endoplasmic reticulum (ER) stress, glial activation, toll-like receptor 4 phosphorylation of p38MAPK.</p>
Full article ">Figure 2
<p>This flow chart shows the different types of pain related to SCD. There are four main types of acute pain: acute VOE pain without chronic pain between VOEs, acute VOE pain with chronic pain in between VOEs, acute on chronic pain exacerbation without VOE, and pain due to another diagnosis. It is important to evaluate the pain and ensure that a VOE pain mimicker such as infection is not the cause as management may change.</p>
Full article ">Figure 3
<p>This figure displays different interventions to manage pain related to SCD. In general, there are two large subgroups of interventions: pharmacological and non-pharmacological. It is important that both are used to manage acute pain for the most effective resolution to the pain.</p>
Full article ">Figure 4
<p>This diagram illustrates the general approach to pharmacological SCD-related pain management. At all levels of the pyramid, patients may utilize non-pharmacological methods to support pain management. At the bottom of the pyramid are inventions that should apply to many patients with SCD while the top of the pyramid is reserved for select patients that require complex management. In the first or bottom level of the pyramid, patients should manage general pain at home with oral medications such as acetaminophen, nonsteroidal anti-inflammatory drugs (NSAIDs), or opioids for severe pain unresponsive to the aforementioned medications. In the 2nd level, the emergency department (ED) may utilize short-acting intravenous (IV) or intranasal (IN) medication to manage the pain. In the 3rd level of the pyramid, patients are admitted to the hospital and should obtain multimodal analgesia for management including the use of a patient-controlled analgesia. In the 4th level during hospitalization, the pain may be refractory to conventional pain medication and need IV ketamine or lidocaine infusion. Of note, if this treatment is highly effective for a patient, then it may be offered with conventional management. On the 5th and final level, additional support from a pain specialist in addition to a multidisciplinary team will need to be considered to manage the pain.</p>
Full article ">Figure 5
<p>This is an example of an individualized pain plan for a patient with SCD. There should be 3 sections with a plan for home-related pain, emergency room, and inpatient pain plans. Medications should be calculated on weight-based dosing, not predetermined doses. Routes such as oral (PO), intravenous (IV), intranasal, and transdermal should be individually discussed with each patient and their caregivers to ensure understanding and agreement.</p>
Full article ">
10 pages, 538 KiB  
Systematic Review
Acute Pancreatitis in Individuals with Sickle Cell Disease: A Systematic Review
by Chinenye R. Dike, Adefunke DadeMatthews, Oluwagbemiga DadeMatthews, Maisam Abu-El-Haija, Jeffrey Lebensburger, Abigail Smith and Aamer Imdad
J. Clin. Med. 2024, 13(16), 4712; https://doi.org/10.3390/jcm13164712 - 11 Aug 2024
Viewed by 1571
Abstract
Background/Objectives: Sickle cell disease (SCD) impacts about 100,000 people in the US. SCD increases the risk of cholelithiasis and microvascular ischemia, which could increase the risk of acute pancreatitis (AP). Abdominal pain is a common presenting symptom of AP and sickle cell [...] Read more.
Background/Objectives: Sickle cell disease (SCD) impacts about 100,000 people in the US. SCD increases the risk of cholelithiasis and microvascular ischemia, which could increase the risk of acute pancreatitis (AP). Abdominal pain is a common presenting symptom of AP and sickle cell vaso-occlusive crisis. The purpose of our systematic review is to estimate the prevalence and determine the severity of AP in individuals with SCD compared to the general population. Methods: Multiple electronic databases were searched. We included studies that included children and adults (population) and addressed the association of SCD (exposure) with AP (outcome) compared to the same population without SCD (control). Two authors screened titles and abstracts independently, and data were abstracted in duplication from included studies. We registered this protocol in PROSPERO-CRD42023422397. Results: Out of 296 studies screened from multiple electronic databases, we identified 33 studies. These studies included 17 case reports, one case series, and 15 retrospective cohort studies, and 18 studies included children. Eight of the AP case reports were in patients with HbSS genotype, two with sickle beta thalassemia, and one with HbSoArab, and in six case reports, a genotype was not specified. Complications were reported in 11 cases—respiratory complication (in at least four cases), splenic complications (three cases), pancreatic pseudocyst (two cases) and death from AP (one case). Of the four AP cases in the case series, three had HbSS genotype, and two cases had complications and severe pancreatitis. AP prevalence in SCD was estimated to be 2% and 7% in two retrospective studies, but they lacked a comparison group. In retrospective studies that evaluated the etiology of AP in children, biliary disease caused mostly by SCD was present in approximately 12% and 34%, respectively. Conclusions: Data on the prevalence of AP in individuals with SCD are limited. Prospectively designed studies aiming to proactively evaluate AP in individuals with SCD who present with abdominal pain are needed to improve timely diagnosis of AP in SCD and outcomes. Full article
(This article belongs to the Section Gastroenterology & Hepatopancreatobiliary Medicine)
Show Figures

Figure 1

Figure 1
<p>Flow diagram of included studies.</p>
Full article ">
18 pages, 929 KiB  
Review
End Organ Affection in Sickle Cell Disease
by Tanvi Bathla, Saran Lotfollahzadeh, Matthew Quisel, Mansi Mehta, Marina Malikova and Vipul C. Chitalia
Cells 2024, 13(11), 934; https://doi.org/10.3390/cells13110934 - 29 May 2024
Viewed by 2009
Abstract
Sickle cell disease is an orphan disease affecting ethnic minorities and characterized by profound systemic manifestations. Although around 100,000 individuals with SCD are living in the US, the exact number of individuals is unknown, and it is considered an orphan disease. This single-gene [...] Read more.
Sickle cell disease is an orphan disease affecting ethnic minorities and characterized by profound systemic manifestations. Although around 100,000 individuals with SCD are living in the US, the exact number of individuals is unknown, and it is considered an orphan disease. This single-gene disorder leads to red blood cell sickling and the deoxygenation of hemoglobin, resulting in hemolysis. SCD is associated with acute complications such as vaso-occlusive crisis, infections, and chronic target organ complications such as pulmonary disease and renal failure. While genetic therapy holds promise to alter the fundamental disease process, the major challenge in the field remains the target end organ damage and ways to mitigate or reverse it. Here, we provide an overview of the clinical manifestations and pathogenesis with a focus on end-organ damage and current therapeutic options, including recent FDA-approved stem cell and gene editing therapies. Full article
Show Figures

Figure 1

Figure 1
<p>Clinical manifestations of SCD.</p>
Full article ">Figure 2
<p>Pathophysiology of SCN. Green arrow: Increased levels; Red arrow: Decreased levels; Black arrow: Leads to.</p>
Full article ">
16 pages, 1682 KiB  
Article
Unraveling the Complexity of Vaso-Occlusive Crises in Sickle Cell Disease: Insights from a Resource-Limited Setting
by Ali Kaponda, Kalunga Muya, Jules Panda, Kodondi Kule Koto and Bruno Bonnechère
J. Clin. Med. 2024, 13(9), 2528; https://doi.org/10.3390/jcm13092528 - 25 Apr 2024
Viewed by 1294
Abstract
Background/Objectives: This study investigated vaso-occlusive crises (VOCs) in sickle cell disease in Lubumbashi, Democratic Republic of Congo, aiming to understand the disease complexities amidst limited resources. With sickle cell hemoglobinopathies on the rise in sub-Saharan Africa, this nine-year study explored factors associated [...] Read more.
Background/Objectives: This study investigated vaso-occlusive crises (VOCs) in sickle cell disease in Lubumbashi, Democratic Republic of Congo, aiming to understand the disease complexities amidst limited resources. With sickle cell hemoglobinopathies on the rise in sub-Saharan Africa, this nine-year study explored factors associated with VOCs and hematological components. Methods: This study comprised 838 patients, analyzing VOCs and hematological changes over time. Demographic characteristics and blood composition changes were carefully categorized. A total of 2910 crises were observed and managed, with analyses conducted on severity, localization, and age groups using statistical methods. Results: The majority of crises were mild or moderate, primarily affecting osteoarticular regions. Statistical analysis revealed significant disparities in crisis intensity based on location and age. The association between blood samples and the number of comorbidities was investigated. Significant positive associations were found for all parameters, except monocytes, indicating a potential link between blood variables and complication burden. Survival analysis using Cox regression was performed to predict the probability of experiencing a second crisis. No significant effects of medication or localization were observed. However, intensity (p < 0.001), age (p < 0.001), and gender (p < 0.001) showed significant effects. Adjusted Hazard Ratios indicated increased risk with age and male gender and reduced risk with mild or severe crisis intensity compared to light. Conclusions: This research sheds light on the complexities of VOCs in resource-limited settings where sickle cell disease is prevalent. The intricate interplay between clinical, laboratory, and treatment factors is highlighted, offering insights for improved patient care. It aims to raise awareness of patient challenges and provide valuable information for targeted interventions to alleviate their burden. Full article
(This article belongs to the Section Hematology)
Show Figures

Figure 1

Figure 1
<p>Relationship between the age of the patients and the number of complications.</p>
Full article ">Figure 2
<p>Relationship between VOC severity and type of medication used, according to gender. The list of the medications’ abbreviations is presented in <a href="#jcm-13-02528-t003" class="html-table">Table 3</a>.</p>
Full article ">Figure 3
<p>Relationship between the number of complications and the number of medications.</p>
Full article ">Figure 4
<p>Survival analysis according to gender (<b>A</b>) and the severity of VOCs (<b>B</b>).</p>
Full article ">
11 pages, 1023 KiB  
Opinion
Enhancing the Management of Pediatric Sickle Cell Disease by Integrating Functional Evaluation to Mitigate the Burden of Vaso-Occlusive Crises
by Paul Muteb Boma, Alain Ali Kaponda, Jules Panda and Bruno Bonnechère
J. Vasc. Dis. 2024, 3(1), 77-87; https://doi.org/10.3390/jvd3010007 - 1 Mar 2024
Viewed by 1549
Abstract
Sickle cell disease (SCD) imposes a significant health burden, particularly in low- and middle-income countries where healthcare professionals and resources are scarce. This opinion paper delves into the management strategies employed for vaso-occlusive crises (VOCs) in pediatric patients with SCD, advocating for the [...] Read more.
Sickle cell disease (SCD) imposes a significant health burden, particularly in low- and middle-income countries where healthcare professionals and resources are scarce. This opinion paper delves into the management strategies employed for vaso-occlusive crises (VOCs) in pediatric patients with SCD, advocating for the adoption of a transformative strategy. We explore the integration of functional assessment approaches into existing procedures, highlighting the potential of technology-assisted rehabilitation, including wearable sensors and digital biomarkers, to enhance the effectiveness of managing and preventing VOCs. Rehabilomics, as a comprehensive framework, merges rehabilitation-related data with biomarkers, providing a basis for personalized therapeutic interventions. Despite the promising advantages of these approaches, persistent obstacles such as the limited availability of rehabilitation programs, especially in resource-limited settings, pose challenges. This paper underscores the importance of a collaborative strategy to effectively address the unique obstacles faced by patients with SCD. This collaborative approach involves improving accessibility to rehabilitation services, incorporating technology-supported therapy, and fostering focused research endeavors. The primary objective of this comprehensive approach is to enhance the overall care of SCD patients, with a specific focus on preventing VOCs, as well as providing tailored (neuro)rehabilitation services in resource-limited settings. By examining the current state of SCD management and proposing transformative strategies, this opinion paper seeks to inspire collective action and collaboration to improve outcomes for pediatric SCD patients globally. Full article
(This article belongs to the Section Peripheral Vascular Diseases)
Show Figures

Figure 1

Figure 1
<p>Most common clinical manifestations of SCD: in pink are those in the chronic phase; in orange are those in the acute phase [<a href="#B20-jvd-03-00007" class="html-bibr">20</a>].</p>
Full article ">Figure 2
<p>Multidimensional and functional data collection.</p>
Full article ">
23 pages, 1308 KiB  
Review
Sickle Cell Disease Update: New Treatments and Challenging Nutritional Interventions
by Victoria Bell, Theodoros Varzakas, Theodora Psaltopoulou and Tito Fernandes
Nutrients 2024, 16(2), 258; https://doi.org/10.3390/nu16020258 - 15 Jan 2024
Cited by 10 | Viewed by 8939
Abstract
Sickle cell disease (SCD), a distinctive and often overlooked illness in the 21st century, is a congenital blood disorder characterized by considerable phenotypic diversity. It comprises a group of disorders, with sickle cell anemia (SCA) being the most prevalent and serious genotype. Although [...] Read more.
Sickle cell disease (SCD), a distinctive and often overlooked illness in the 21st century, is a congenital blood disorder characterized by considerable phenotypic diversity. It comprises a group of disorders, with sickle cell anemia (SCA) being the most prevalent and serious genotype. Although there have been some systematic reviews of global data, worldwide statistics regarding SCD prevalence, morbidity, and mortality remain scarce. In developed countries with a lower number of sickle cell patients, cutting-edge technologies have led to the development of new treatments. However, in developing settings where sickle cell disease (SCD) is more prevalent, medical management, rather than a cure, still relies on the use of hydroxyurea, blood transfusions, and analgesics. This is a disease that affects red blood cells, consequently affecting most organs in diverse manners. We discuss its etiology and the advent of new technologies, but the aim of this study is to understand the various types of nutrition-related studies involving individuals suffering from SCD, particularly in Africa. The interplay of the environment, food, gut microbiota, along with their respective genomes collectively known as the gut microbiome, and host metabolism is responsible for mediating host metabolic phenotypes and modulating gut microbiota. In addition, it serves the purpose of providing essential nutrients. Moreover, it engages in direct interactions with host homeostasis and the immune system, as well as indirect interactions via metabolites. Nutrition interventions and nutritional care are mechanisms for addressing increased nutrient expenditures and are important aspects of supportive management for patients with SCD. Underprivileged areas in Sub-Saharan Africa should be accompanied by efforts to define and promote of the nutritional aspects of SCD. Their importance is key to maintaining well-being and quality of life, especially because new technologies and products remain limited, while the use of native medicinal plant resources is acknowledged. Full article
(This article belongs to the Special Issue Dietary Approaches and Prevention of Chronic Diseases)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The most common clinical manifestation of sickle cell disease, a vaso-occlusive crisis (VOC) occurring when blood flow is blocked by sickled red blood cells (crescent-shaped) to the point that tissues and organs become deprived of oxygen, causing pain.</p>
Full article ">Figure 2
<p>Some of the major complications associated with SCD development.</p>
Full article ">Figure 3
<p>Some of the tropical plants used in SCA in Sub-Saharan Africa.</p>
Full article ">
12 pages, 1598 KiB  
Article
Pain Control for Sickle Cell Crisis, a Novel Approach? A Retrospective Study
by Amélie Rollé, Elsa Vidal, Pierre Laguette, Yohann Garnier, Delphine Delta, Frédéric Martino, Patrick Portecop, Maryse Etienne-Julan, Pascale Piednoir, Audrey De Jong, Marc Romana and Emmanuelle Bernit
Medicina 2023, 59(12), 2196; https://doi.org/10.3390/medicina59122196 - 18 Dec 2023
Cited by 2 | Viewed by 2523
Abstract
Background and Objectives: Pain management poses a significant challenge for patients experiencing vaso-occlusive crisis (VOC) in sickle cell disease (SCD). While opioid therapy is highly effective, its efficacy can be impeded by undesirable side effects. Local regional anesthesia (LRA), involving the deposition [...] Read more.
Background and Objectives: Pain management poses a significant challenge for patients experiencing vaso-occlusive crisis (VOC) in sickle cell disease (SCD). While opioid therapy is highly effective, its efficacy can be impeded by undesirable side effects. Local regional anesthesia (LRA), involving the deposition of a perineural anesthetic, provides a nociceptive blockade, local vasodilation and reduces the inflammatory response. However, the effectiveness of this therapeutic approach for VOC in SCD patients has been rarely reported up to now. The objective of this study was to assess the effectiveness of a single-shot local regional anesthesia (LRA) in reducing pain and consequently enhancing the management of severe vaso-occlusive crisis (VOC) in adults with sickle cell disease (SCD) unresponsive to conventional analgesic therapy. Materials and Methods: We first collected consecutive episodes of VOC in critical care (ICU and emergency room) for six months in 2022 in a French University hospital with a large population of sickle cell patients in the West Indies population. We also performed a systematic review of the use of LRA in SCD. The primary outcome was defined using a numeric pain score (NPS) and/or percentage of change in opioid use. Results: We enrolled nine SCD adults (28 years old, 4 females) for ten episodes of VOC in whom LRA was used for pain management. Opioid reduction within the first 24 h post block was −75% (50 to 96%). Similarly, the NPS decreased from 9/10 pre-block to 0–1/10 post-block. Five studies, including one case series with three patients and four case reports, employed peripheral nerve blocks for regional anesthesia. In general, local regional anesthesia (LRA) exhibited a reduction in pain and symptoms, along with a decrease in opioid consumption post-procedure. Conclusions: LRA improves pain scores, reduces opioid consumption in SCD patients with refractory pain, and may mitigate opioid-related side effects while facilitating the transition to oral analgesics. Furthermore, LRA is a safe and effective procedure. Full article
(This article belongs to the Special Issue Latest Advances in Regional Anesthesia)
Show Figures

Figure 1

Figure 1
<p>Comparison of opioid consumption before and after LRA. Local regional anesthesia (LRA) was effective in treating sickle cell crises for the reductions in opioid consumption. Opioid reduction within the first 24 h post-block was −75% (50 to 96%, <span class="html-italic">p</span> = 0.016, *). The percentage change of morphine consumption 24 h before and 24 h after LRA (%change = [(pre-block use) − (post-block use)]/(pre-block use) × 100. Then, we performed a paired <span class="html-italic">t</span>-test (<span class="html-italic">p</span>-value).</p>
Full article ">Figure 2
<p>Comparison of numeric pain scale before and after LRA. Local regional anesthesia (LRA) was effective in treating sickle cell crises for the reduction in pain trajectory. Numeric pain scale (NPS) decreased from 9/10 pre-block to 0–1/10 post-block (less than 6 h later) (<span class="html-italic">p</span> &lt; 0.001, ****).</p>
Full article ">
28 pages, 3397 KiB  
Review
Thrombo-Inflammation in COVID-19 and Sickle Cell Disease: Two Faces of the Same Coin
by Kate Chander Chiang, Ajay Gupta, Prithu Sundd and Lakshmanan Krishnamurti
Biomedicines 2023, 11(2), 338; https://doi.org/10.3390/biomedicines11020338 - 25 Jan 2023
Cited by 5 | Viewed by 4493
Abstract
People with sickle cell disease (SCD) are at greater risk of severe illness and death from respiratory infections, including COVID-19, than people without SCD (Centers for Disease Control and Prevention, USA). Vaso-occlusive crises (VOC) in SCD and severe SARS-CoV-2 infection are both characterized [...] Read more.
People with sickle cell disease (SCD) are at greater risk of severe illness and death from respiratory infections, including COVID-19, than people without SCD (Centers for Disease Control and Prevention, USA). Vaso-occlusive crises (VOC) in SCD and severe SARS-CoV-2 infection are both characterized by thrombo-inflammation mediated by endothelial injury, complement activation, inflammatory lipid storm, platelet activation, platelet-leukocyte adhesion, and activation of the coagulation cascade. Notably, lipid mediators, including thromboxane A2, significantly increase in severe COVID-19 and SCD. In addition, the release of thromboxane A2 from endothelial cells and macrophages stimulates platelets to release microvesicles, which are harbingers of multicellular adhesion and thrombo-inflammation. Currently, there are limited therapeutic strategies targeting platelet-neutrophil activation and thrombo-inflammation in either SCD or COVID-19 during acute crisis. However, due to many similarities between the pathobiology of thrombo-inflammation in SCD and COVID-19, therapies targeting one disease may likely be effective in the other. Therefore, the preclinical and clinical research spurred by the COVID-19 pandemic, including clinical trials of anti-thrombotic agents, are potentially applicable to VOC. Here, we first outline the parallels between SCD and COVID-19; second, review the role of lipid mediators in the pathogenesis of these diseases; and lastly, examine the therapeutic targets and potential treatments for the two diseases. Full article
(This article belongs to the Special Issue Sickle Cell Disease: Recent Advances in Pathophysiology and Therapy)
Show Figures

Figure 1

Figure 1
<p>Putative mechanism of complement-mediated microvascular thrombosis and vaso-occlusive disease in SCD and COVID-19: SARS-CoV-2 infection and sickle cell disease induce complement activation and formation of membrane attack complex leading to necrosis and pyroptosis of endothelial cells, platelets, and monocytes and accumulation of IL-1α. IL-1α stimulates the IL-1 receptor expressed on endothelial cells leading to thromboxane synthesis. Thromboxane A<sub>2</sub> via the TP receptor activates platelets leading to platelet activation, platelet neutrophil partnership, neutrophil activation, and the release of neutrophil extracellular traps (NETs), thrombo-inflammation, oxidative stress, and subsequent end-organ damage and failure. The current review article is primarily focused on eicosanoid signaling in platelets; therefore, other receptors and pathways were excluded from <a href="#biomedicines-11-00338-f001" class="html-fig">Figure 1</a> for the reader’s convenience. COX, cyclooxygenase; IL, interleukin; NETs, neutrophil extracellular traps; TP, thromboxane prostanoid receptor; MAC, membrane attack complex; VTE, venous thromboembolism; TMA, thrombotic microangiopathy; DIC, disseminated intravascular thrombosis; ARDS, acute respiratory distress syndrome; AKI, acute kidney injury.</p>
Full article ">Figure 2
<p>Mechanisms of heme and thromboxane A<sub>2</sub>-mediated thrombo-inflammation in COVID-19 and sickle cell disease (SCD): Vaso-occlusion due to sickling or direct entry by SARS-CoV-2 virus leads to endothelial cell activation and damage, and hemolysis. COX-2 expression in endothelial cells promotes thromboxane A<sub>2</sub> synthesis. Thromboxane A<sub>2</sub> inhibits nitric oxide (NO) synthesis and promotes leukocyte adhesion and thrombo-inflammation. Free heme released from red blood cells is spontaneously oxidized to its ferric form, hemin. Hemin stimulates platelet CLEC2 signaling and thromboxane A<sub>2</sub>/TP receptor-dependent Syk phosphorylation leading to platelet activation, spreading, and degranulation. Platelets release exosomes and microvesicles, which stimulate the CLEC5A and TLR2 receptors on neutrophils. Subsequently, NLRP3 activation in neutrophils and monocytes promotes activation and assembly of gasdermin D, leading to the release of neutrophil extracellular traps and monocyte pyroptosis. NLRP3 inflammasome activation induces the release of proinflammatory cytokines, including IL-18 and IL-1β, thereby fueling thrombo-inflammation in COVID-19 and SCD. NO, nitric oxide; COX, cyclooxygenase; IL, interleukin; NETs, neutrophil extracellular traps; TP, thromboxane prostanoid receptor; CLEC, C-type lectin-like receptor; Syk, spleen tyrosine kinase; PLC, phospholipase C; PKC, protein kinase C; TLR, toll-like receptor; ADP, adenosine diphosphate; EGF, epidermal growth factor; PDGF, platelet-derived growth factor; TGF, transforming growth factor; NLRP3, NLR family pyrin domain containing 3.</p>
Full article ">
13 pages, 779 KiB  
Review
Mitochondria: Emerging Consequential in Sickle Cell Disease
by Mohammad S. Akhter, Hassan A. Hamali, Hina Rashid, Gasim Dobie, Aymen M. Madkhali, Abdullah A. Mobarki, Johannes Oldenburg and Arijit Biswas
J. Clin. Med. 2023, 12(3), 765; https://doi.org/10.3390/jcm12030765 - 18 Jan 2023
Cited by 4 | Viewed by 3044
Abstract
Advanced mitochondrial multi-omics indicate a multi-facet involvement of mitochondria in the physiology of the cell, changing the perception of mitochondria from being just the energy-generating organelles to organelles that highly influence cell structure, function, signaling, and cell fate. This sets mitochondrial dysfunction in [...] Read more.
Advanced mitochondrial multi-omics indicate a multi-facet involvement of mitochondria in the physiology of the cell, changing the perception of mitochondria from being just the energy-generating organelles to organelles that highly influence cell structure, function, signaling, and cell fate. This sets mitochondrial dysfunction in the centerstage of numerous acquired and genetic diseases. Sickle cell disease is also being increasingly associated with mitochondrial anomalies and the pathophysiology of sickle cell disease finds mitochondria at crucial intersections in the pathological cascade. Altered mitophagy, increased ROS, and mitochondrial DNA all contribute to the condition and its severity. Such mitochondrial aberrations lead to consequent mitochondrial retention in red blood cells in sickle cell diseases, increased oxidation in the cellular environment, inflammation, worsened vaso-occlusive crisis, etc. There are increasing studies indicating mitochondrial significance in sickle cell disease, consequently providing an opportunity to target it for improving the outcomes of treatment. Identification of the impaired mitochondrial attributes in sickle cell disease and their modulation by therapeutic interventions can impart a better management of the disease. This review aims to describe the mitochondria in the perspective of sicke cell disease so as to provide the reader an overview of the emerging mitochondrial stance in sickle cell disease. Full article
(This article belongs to the Special Issue Sickle Cell Disease: Current Understanding and Future Options)
Show Figures

Figure 1

Figure 1
<p>The mitochondrial dynamics and energetics are impaired in SCD, and are indicated by mitochondrial retention in RBCs, increased oxidative stress, and free circulating mtDNA.</p>
Full article ">Figure 2
<p>An all-inclusive approach to modulate bioenergetics, dynamics, and genome of mitochondria is of therapeutic importance in improving the treatment outcome in SCD.</p>
Full article ">
15 pages, 1599 KiB  
Article
Real-World Evidence on Disease Burden and Economic Impact of Sickle Cell Disease in Italy
by Lucia De Franceschi, Chiara Castiglioni, Claudia Condorelli, Diletta Valsecchi, Eleonora Premoli, Carina Fiocchi, Valentina Perrone, Luca Degli Esposti, Gian Luca Forni and on behalf of the GREATalyS Study Group
J. Clin. Med. 2023, 12(1), 117; https://doi.org/10.3390/jcm12010117 - 23 Dec 2022
Cited by 7 | Viewed by 2892
Abstract
A real-world analysis was conducted in Italy among sickle cell disease (SCD) patients to evaluate the epidemiology of SCD, describe patients’ characteristics and the therapeutic and economic burden. A retrospective analysis of administrative databases of various Italian entities was carried out. All patients [...] Read more.
A real-world analysis was conducted in Italy among sickle cell disease (SCD) patients to evaluate the epidemiology of SCD, describe patients’ characteristics and the therapeutic and economic burden. A retrospective analysis of administrative databases of various Italian entities was carried out. All patients with ≥1 hospitalization with SCD diagnosis were included from 01/2010-12/2017 (up to 12/2018 for epidemiologic analysis). The index date corresponded to the first SCD diagnosis. In 2018, SCD incidence rate was 0.93/100,000, the prevalence was estimated at 13.1/100,000. Overall, 1816 patients were included. During the 1st year of follow-up, 50.7% of patients had one all-cause hospitalization, 27.8% had 2, 10.4% had 3, and 11.1% had ≥4. Over follow-up, 6.1–7.2% of patients were treated with SCD-specific, 58.4–69.4% with SCD-related, 60.7–71.3% with SCD-complications-related drugs. Mean annual number per patient of overall treatments was 14.9 ± 13.9, hospitalizations 1.1 ± 1.1, and out-patient services 5.3 ± 7.6. The total mean direct cost per patient was EUR 7918/year (EUR 2201 drugs, EUR 3320 hospitalizations, and EUR 2397 out-patient services). The results from this real-world analysis showed a high disease burden for SCD patients with multiple hospitalizations during the follow-up. High healthcare resource utilization and costs were associated with patient’ management and were most likely underestimated since indirect costs and Emergency Room admissions were not included. Full article
Show Figures

Figure 1

Figure 1
<p>(Upper plots <b>A</b>–<b>D</b>) Epidemiology of SCD in the sample population (N = 15,300,00). (<b>A</b>) Incidence rate overall and by gender per year included in the study (2010–2018). (<b>B</b>) Prevalence by gender and age (&lt;18 years and ≥18 years) for year 2018. (<b>C</b>) Prevalence by age classes in adult SCD patients with and without VOC at diagnosis for year 2018. (<b>D</b>) Prevalence by geographic area and presence of crisis for year 2018. (Lower plots <b>E</b>–<b>G</b>) Projections to Italian population for year 2018. (<b>E</b>) Number of SCD patients estimated in Italy by age and gender. (<b>F</b>) Number of SCD patients estimated in Italy by age ranges. (<b>G</b>) Number of SCD patients estimated in Italy by presence of VOCs at diagnosis.</p>
Full article ">Figure 2
<p>Study flow-chart of patients in analysis. The inclusion period was from 1st January 2010 to 31st December 2017. Patients were identified by SCD diagnosis retrieved from the hospitalization database (ICD-9-CM codes 282.41, 282.42, 282.60, 282.61, 282.62, 282.63, 282.64, 282.68, and 282.69; code 282.50 related to the Sickle cell trait was not considered for inclusion).</p>
Full article ">Figure 3
<p>Hospitalization of patients with SCD. (<b>A</b>) More frequent primary diagnosis in SCD patients without crisis with secondary SCD diagnosis at inclusion (<span class="html-italic">n</span> = 1129). (<b>B</b>) Percentages of patients with all-cause hospitalizations during follow-up period. Hospitalizations comprised ordinary admissions and day hospitals; Emergency Room access not requiring hospitalization was not included. Hospitalization corresponding to the index date was counted in the number of hospitalizations. (<b>C</b>) Percentages of SCD patients with VOC episodes or complications during follow-up.</p>
Full article ">Figure 4
<p>Therapeutic pathways and healthcare resource costs. (<b>A</b>) Patients treated with at least one SCD-specific drug, SCD-related drug, and SCD complication-related drug during the follow-up period (1–4 years). (<b>B</b>) Patients with crisis treated with at least one SCD-specific drug, SCD-related drug, and SCD complication-related drug during the follow-up period (1–4 years). (<b>C</b>) Mean annual healthcare resource consumption and related costs considering all available follow-up. List of SCD treatments in analysis and the related ATC code is reported in <a href="#app1-jcm-12-00117" class="html-app">Supplementary Table S2</a>. Abbreviation: OSS, outpatient services.</p>
Full article ">
Back to TopTop