Melanin nanoparticles for safe and effective iron chelation therapy for Beta-thalassemia during pregnancy

2022 ◽  
Vol 226 (1) ◽  
pp. S54-S55
Author(s):  
Nahla Bakhamis ◽  
Helen Townley
Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5165-5165
Author(s):  
Christian Junghanss ◽  
Rudolf Schlag ◽  
Bernd Gaede ◽  
Matthias Moelle ◽  
Steffen Doerfel ◽  
...  

Abstract Abstract 5165 Background: Progressive anaemia is highly prevalent amongst many malignant diseases leading to RBC transfusion-dependency. Therefore transfusion-related iron overload (IOL) is common in these patients (pts) and can result in multiple organ failure. Iron chelation therapy prevents organ failure, reduces the risk of infections and can improve hematopoesis in some diseases. The once-daily oral iron chelator deferasirox has been shown to reduce iron overload in pts with various transfusion-dependent anaemias assessed by serum ferritin (SF). Despite extensive knowledge of iron chelation in MDS or beta-thalassemia pts, data in pts with other anaemias is limited. Here, we present data from a subgroup of transfusion-related IOL pts that were included two non-interventional studies (EXTEND, EXJANGE) performed in Germany and who suffered from diseases other than MDS or beta thalassemia. Methods: 130 pts with various malignant diseases such as myeloproliferative disorders (43 pts, including 31 pts particular specified as myelofibrosis), acute myeloid leukaemia (14 pts), sickle cell anaemia (6 pts), aplastic anaemia (11), congenital aplastic anaemia (5) or Non-Hodgkin's lymphoma (6 pts) were treated with deferasirox in the daily-routine setting of office-based physicians and included in either the EXTEND or EXJANGE study. Patient with MDS or beta-thalassemia were also included in the studies, but are excluded from this analysis. Analysis is based on 1-year pooled data of these two, multicenter, non-interventional observational studies. Transfusion-dependent pts with IOL with or without prior chelation were enrolled and received the iron chelator deferasirox. Prescription of deferasirox, just as inclusion and exclusion criteria was in accordance with the terms of Exjade marketing authorization in the EU. Efficacy and safety parameters, including serum ferritin and adverse events (AEs), were collected in 2-monthly intervals. Results: 98 pts had no prior chelation therapy (51 M, 45 F, 2 missing; mean age 63.3, range 3.2–91.9 yrs) and a median baseline SF of 2,968 (range 561–11, 423) ng/mL. 32 pts had prior received prior chelation therapy (mainly with desferal; 17 M, 15 F; mean age 50.1, range 3.5–80.9 yrs) and a median baseline SF of 2,635 (range 539–19, 540) ng/mL. The mean number of prior red blood cell transfusions was 55. The mean prescribed daily dose of deferasirox at the first visit was 16.3 mg/kg/d rising up to 18.1 mg/kg/d after 12 months. During treatment, median SF levels clearly decreased from first to final visit [-806 ng/mL; p<0.0001 (explorative analysis)] in the chelation-naïve and also in the pre-chelated population [-300 ng/ml; p = 0.1705 (explorative analysis)]. The median observation period and days on therapy was 349 and 343 days, respectively. At final visit 74 pts (56.9%) were still on deferasirox therapy. Reasons for discontinuation by the final visit included 19 AEs (35.2%). 45 pts (34.6%) experienced an investigator assessed drug-related AE. The most common drug-related AEs were diarrhea (n=17; 37.8%), nausea (n=11; 24.4%) and blood creatinine increased (n=6; 13.3%). As in previous clinical trials, serum creatinine clearances showed a minor decrease over the study period (median decrease until final visit: 4 ml/min). Conclusion: Our analysis confirmed that deferasirox is effective and well tolerated in chelation-naïve as well as in previously chelated pts with transfusion-related IOL and diseases other than MDS or beta thalassemia. As baseline serum ferritin values were >2,500 ng/mL even in pts with prior chelation therapy, adequate chelation treatment should be considered earlier at a serum ferritin >1,000 ng/mL in pts with transfusion-dependent IOL for adequate iron chelation therapy. Disclosures: Junghanss: Novartis: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Haus:Novartis Pharma: Employment. Junkes:Novartis: Employment. Leismann:Novartis: Employment.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 752-752 ◽  
Author(s):  
Antonio Piga ◽  
Silverio Perrotta ◽  
M.Rita Gamberini ◽  
Ersi Voskaridou ◽  
Angela Melpignano ◽  
...  

Background. Luspatercept is a fusion protein (modified activin receptor IIB-IgG Fc) being investigated for the treatment of anemias with ineffective erythropoiesis. In beta-thalassemia, imbalanced production of alpha and beta globin chains in erythroid precursors inhibits late-stage erythroid differentiation, leading to anemia, ineffective erythropoiesis (IE), and dysregulated iron homeostasis. Luspatercept promotes late-stage erythroid differentiation, corrects ineffective erythropoiesis, and reduces alpha globin aggregates, hemolysis, and disease complications including iron overload in a beta-thalassemia mouse model (Suragani R, Blood 2014). Aims. This is an ongoing, phase 2, multicenter, open-label, dose-finding study to evaluate luspatercept in adults with transfusion-dependent (TD) or non-transfusion dependent (NTD) beta-thalassemia. Efficacy outcomes include erythroid response including Hb increase in NTD patients and reduction in RBC transfusion burden in TD patients, and reduction in liver iron concentration (LIC) by MRI in both TD and NTD patients. Methods. Inclusion criteria included age ≥ 18 yr and either TD (defined as ≥4 RBC units transfused in the 8 weeks prior to first dose, confirmed over 6 months) or NTD (defined as <4 RBC units transfused in the 8 weeks prior to first dose) with baseline Hb <10.0 g/dL. Luspatercept was administered SC every 3 weeks for up to 5 doses (12 weeks) with an 8-week follow-up (20 weeks total). Six sequential cohorts (n=up to 6 each) were treated at escalating dose levels from 0.2 to 1.25 mg/kg and an expansion cohort (n=30) is ongoing with starting dose level 0.8 mg/kg and individual dose titration. Patients who completed the core study may have been eligible to enroll in an ongoing 24-month extension study. Results. Preliminary data (as of 10-Apr-2015) were available for 39 patients, 35 patients from dose escalation, and 4 patients from the expansion. Median age was 40 yr, ranging from 20-57 yr, and 82% had prior splenectomy. TD patients (n=14). Transfusion burden prior to treatment ranged from 4 to 12 units/12 weeks. Ten of 14 (71%) patients were treated for ≥12 weeks and were evaluable for the primary endpoint of ≥20% reduction in transfusion burden. All 10 of these patients responded with >40% reduction in transfusion burden over any 12-week period during the study. Reduction in transfusion burden correlated with reduction in LIC. Two of the 3 TD patients with baseline LIC ≥7 mg/g dw (all on iron chelation therapy) had a reduction in LIC at 16 weeks (-2.0 and -4.7 mg/g dw). NTD patients (n=25). Median baseline Hb was 8.4 g/dL (range 6.5-9.6 g/dL). Four of 8 (50%) patients in the 0.8-1.25 mg/kg dose groups achieved the primary endpoint of increase in Hb ≥1.5 g/dL sustained for ≥2 weeks, compared with 0 of 17 (0%) patients in the 0.2-0.6 mg/kg dose groups. Four of eight (50%) patients in the 0.8-1.25 mg/kg dose groups had a mean increase in Hb ≥1.0 g/dL sustained for ≥12 weeks, compared with 5 of 17 (29%) patients in the 0.2-0.6 mg/kg dose groups. Eight of 12 (67%) NTD patients with baseline LIC ≥5 mg/g dw (including 6 patients on iron chelation therapy) had a reduction of at least 1 mg/g dw in LIC at 16 weeks; reduction in LIC correlated with increase in hemoglobin. All 3 patients with chronic leg ulcers at baseline (2 NTD, 1 TD) had substantial, rapid healing, evident within 6 weeks after the first dose of luspatercept. Luspatercept was generally well tolerated, with no related serious adverse events reported to date. Adverse events were mostly mild-moderate; most frequent related adverse events (>10% patients) were bone pain, myalgia, headache and asthenia. Longer-term treatment data from the extension study for TD and NTD patients will be presented. Conclusions. Luspatercept treatment for up to 3 months was well-tolerated, increased Hb levels in NTD patients, and decreased transfusion requirement in TD patients with beta-thalassemia. Both TD and NTD patients also had decreases in LIC, and healing of leg ulcers occurred in 3 of 3 patients. These treatment effects represent a significant reduction in disease burden for patients with beta-thalassemia. Phase 3 studies of luspatercept in beta-thalassemia are planned. Disclosures Piga: Celgene Corporation: Honoraria; Acceleron Pharma: Research Funding. Zhang:Acceleron: Employment. Bellevue:Acceleron: Employment. Wilson:Acceleron: Employment. Laadem:Celgene Corporation: Employment. Sherman:Acceleron Pharma: Employment. Attie:Acceleron: Employment.


2021 ◽  
Vol 10 (18) ◽  
pp. 1365-1368
Author(s):  
Anamika Giri ◽  
Sameera Dronamraju ◽  
Sourya Acharya ◽  
Samarth Shukla ◽  
Sunil Kumar

Thalassemia, derived from Thalassa [Greek: Sea] refers to a cluster of hereditary haemoglobinopathies initially reported in areas of Sub-Saharan Africa, the Indian subcontinent, Southeast Asia, and the Mediterranean where malaria was (or is) endemic. Currently, it is the most frequently recognized haemoglobinopathy, and has been correlated with a plethora of immune system changes such as neutropenia, natural killer cell dysfunctions, increased activity of CD8 suppressor cells, along with disturbances in macrophage function, chemotaxis, phagocytosis, and interferons INFγ production.1 The reduction in the alpha or beta chain that leads to the production of haemoglobin is the key pathophysiology behind thalassemia. Therefore, weakened red blood cells can contribute to inadequate erythropoiesis and haemolytic anaemia. Thalassemia diseases are known as transfusion-dependent or non-dependent according to either their beta-globin chain genotype or their clinical path. The most serious type of thalassemia is beta-thalassemia major, and it is transfusiondependent, presenting in early infancy.2 A diagnosis of β-thalassemia major (BTM) means lifelong, regular transfusions, supplemented with adequate iron chelation therapy for the patient.3 This also means that unless adequate blood screening and hygiene procedures are in place, patients with BTM are at a risk of transfusion transmitted infections (TTI).4 Many countries have been prompted by the spread of Covid-19 infection to shutter routine outpatient coronary health care units before the pandemic is over, which also extends to people with thalassemia, who require regular follow-ups. While Covid-19 infection rates remain high, patients with thalassemia with planned cardiovascular examinations are advised to delay their medical appointments for a minimum of 3 months.1 In addition, according to the statement published by the Thalassemia International Foundation on the 13th of July, 2020, patients with hemoglobinopathies are at a greater risk in acquiring the Covid-19 infection, posing an additional challenge to the patients, families, and medical staff managing these disorders.5 Thalassaemic patients have reduced levels of protein S and protein C; elevated aggregation of platelets; and recruitment of monocytes, granulocytes, and endothelial cells. Patients of thalassemia also show elevated markers of platelet and coagulation activation, even in the absence of major thromboembolic events. An increase in D-Dimer levels is also seen in most Covid-19 patients who are hospitalised, attributable to the underlying inflammatory process.1 A variety of questions have been raised during the ongoing Covid-19 pandemic with respect to diagnostic and clinical approaches towards this particular population. The precise path of infection with Covid-19 in those patients has yet to be elucidated.2 The progression of Covid-19 infection in patients with hemoglobinopathy in general and thalassemia in particular is not fully known and is still an area of discussion and under review. We report a case of a 30-year-old male patient, a known case of beta thalassemia major on maintenance transfusion and iron chelation therapy who acquired Covid-19 infection.


2010 ◽  
Vol 123 (3) ◽  
pp. 148-152 ◽  
Author(s):  
Marina Economou ◽  
Nikoletta Printza ◽  
Aikaterini Teli ◽  
Vassiliki Tzimouli ◽  
Ioanna Tsatra ◽  
...  

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2469-2469
Author(s):  
Francesca Vinchi ◽  
Gregory M Vercellotti ◽  
John D. Belcher ◽  
Eitan Fibach ◽  
Hala Zreid ◽  
...  

Abstract Beta thalassemia is an inherited hemoglobinopathy due to reduced synthesis of Beta globin chains and, consequently, of hemoglobin A (a2b2). The clinical manifestations are mainly the result of chronic anemia and iron overload. The latter is due to increased iron absorption, induced by accelerated but ineffective erythropoiesis, and recurrent red blood cell transfusions. Alfa-chains and iron excess promote oxidative damage of red blood cell membrane, resulting in macrophage sequestration and extravascular hemolysis, and to a lower extent, in intravascular hemolysis, with consequent release of hemoglobin (Hb), heme and iron. Increasing evidence suggests that free heme exerts vasculotoxic, pro-inflammatory and procoagulant effects due to its ability to trigger endothelial and immune cells activation. In addition, a role for heme and iron has been postulated in the pathogenesis of other vascular diseases, including atherosclerosis. In mouse models of Beta thalassemia and sickle cell disease, circulating heme levels are elevated and correlate with the exhaustion of systemic scavengers for hemoglobin and heme, haptoglobin and hemopexin, respectively, as well as with severe endothelial dysfunction and inflammation. Hemopexin-based therapies significantly improve endothelial damage, vascular oxidative stress and inflammation in these mice (Vinchi et al., Circulation 2013, Blood 2016; Vercellotti GM. et al., Mol Med 2016). Whereas more data are reported on sickle patients in this regard, few data are available in patients with Beta thalassemia. In the present study, we examined serum samples from a cohort of 60 patients with Beta thalassemia major (age 11.5 ± 6.8, 44% males-56% females, Hb 7.69 ± 1.22 mg/dl, transfused every 3-4 weeks) and 7 patients with Beta thalassemia intermedia (age 14 ± 12 , 70% males-30% females, Hb 8.4 ± 0.74 mg/dl, transfused every 4-5 weeks). 10% of the patients received inconsistent iron chelation therapy. Serum from 10 healthy subjects (age 22.7±15.3, 50% males-50% females, Hb 13.12±1.15 mg/dl) served as control. Both groups of patients show high systemic heme and iron levels, which associate with a severe drop in serum haptoglobin, hemopexin and transferrin. Consistently, transferrin saturation (12.4±2 vs 79.6±24 %) and serum ferritin (55.14 ±0.23 vs 4919.2 ±2657.4 ng/ml) are elevated. Interestingly, these patients present with high systemic levels of the soluble adhesion molecules sVCAM-1 and sICAM-1, markers of enhanced endothelial activation. In addition, they show increased levels of serum malondialdehyde, a well-known marker of lipid peroxidation and oxidative stress, and high levels of circulating oxidized low density lipoproteins (oxLDL). All parameters significantly correlate with increased systemic heme and iron indices as well as decreased haptoglobin, hemopexin and transferrin levels. In conclusion, Beta thalassemia patients show a strong correlation between systemic heme and iron overload, depletion of the respective scavengers, and markers of oxidative stress and endothelial dysfunction, thus confirming studies in animal models. These results emphasize the involvement of serum hemoglobin, heme and iron in the pathophysiology of Beta thalassemia, including vascular dysfunction, and the key protective role of their carriers. These findings are relevant for disorders hallmarked by vasculopathy, such as sickle cell disease and Beta thalassemia, as well as cardiovascular diseases, such as atherosclerosis. Our data support the potential therapeutic benefit of the administration of hemoglobin/heme scavengers along with efficient iron chelation therapy to counteract heme- and iron-driven toxicity. (The last three authors equally contributed to the work) ****P<0.0001 Disclosures Vercellotti: CSL-Behring: Research Funding; Imara: Research Funding. Belcher:Cydan/Imara: Research Funding; CSL-Behring: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document