scholarly journals PREPARATION AND EVALUATION OF IRON OXIDE NANOPARTICLES FOR TREATMENT OF IRON DEFICIENCY ANEMIA

Author(s):  
Fahima Hashem ◽  
Mohamed Nasr ◽  
Yomna Ahmed

Objective: The objective of this research was to formulate and evaluate iron oxide nanoparticles for treatment of iron deficiency anemia (IDA).Methods: Iron oxide nanoparticles were prepared by co-precipitation method and stabilized by coating with folic acid or chitosan. The prepared nanoparticles were characterized in vitro for morphology, particle size, zeta potential, crystallinity and ultraviolet-visible (UV-Vis) absorption. In vivo studies were performed to evaluate the efficacy of the prepared nanoparticles in treating iron-deficient anemic rats compared to the commercial iron product.Results: In vitro results revealed that particle sizes were 65.95±5 nm, 220.2±12 nm and 295.3±19 nm for uncoated iron oxide nanoparticles, folic acid-coated iron oxide nanoparticles and chitosan coated iron oxide nanoparticles, respectively. UV-Vis absorption spectrum and x-ray diffraction (XRD) patterns confirmed that the prepared nanoparticles were iron oxide nanoparticles. In vivo results indicated that folic acid-coated iron oxide nanoparticles showed effective restorative action, returning haemoglobin (Hb) concentration to normal levels, where not only complete recovery of Hb within short time from the anemic state to the high normal level, but also improved Hb concentrations compared to the commercial iron product.Conclusion: The results obtained in this research work clearly indicated a promising potential of folic acid-coated iron oxide nanoparticles for the effective treatment of IDA.

2019 ◽  
Vol 9 (2) ◽  
pp. 166-172
Author(s):  
Ahmed A.G. El-Shahawy ◽  
Gamal Elghnam ◽  
Alsayed A.M. Alsherbini

Background:Gold and Iron Oxide nanoparticles NPs play as nanocarriers for a specific drug delivery and contrast agents. Intercellular uptake of these nanoparticles and targeting to individual cell and sub-cellular compartment is essential.Objective:The aim of the current study is to evaluate the intracellular uptake of these NPs to specific tumor cells in vitro conjugated with folic acid with a goal of enhancing the efficiency of specific targeting to tumor cells.Methods:We synthesized the nanoparticles by a chemical method and characterized by UV-Visible, FTIR, XRD, and TEM.Results & Conclusion:The results revealed the conjugation of Gold and Iron Oxide nanoparticles with folic acid increased the intercellular uptake with high percent compared to non- conjugated nanoparticles.


2010 ◽  
Vol 19 (4) ◽  
pp. 419-429 ◽  
Author(s):  
Po-Wah So ◽  
Tammy Kalber ◽  
David Hunt ◽  
Michael Farquharson ◽  
Alia Al-Ebraheem ◽  
...  

Determination of the dynamics of specific cell populations in vivo is essential for the development of cell-based therapies. For cell tracking by magnetic resonance imaging (MRI), cells need to internalize, or be surface labeled with a MRI contrast agent, such as superparamagnetic iron oxide nanoparticles (SPIOs): SPIOs give rise to signal loss by gradient-echo and T2-weighted MRI techniques. In this study, cancer cells were chemically tagged with biotin and then magnetically labeled with anti-biotin SPIOs. No significant detrimental effects on cell viability or death were observed following cell biotinylation. SPIO-labeled cells exhibited signal loss compared to non-SPIO-labeled cells by MRI in vitro. Consistent with the in vitro MRI data, signal attenuation was observed in vivo from SPIO-labeled cells injected into the muscle of the hind legs, or implanted subcutaneously into the flanks of mice, correlating with iron detection by histochemical and X-ray fluorescence (XRF) methods. To further validate this approach, human mesenchymal stem cells (hMSCs) were also employed. Chemical biotinylation and SPIO labeling of hMSCs were confirmed by fluorescence microscopy and flow cytometry. The procedure did not affect proliferation and multipotentiality, or lead to increased cell death. The SPIO-labeled hMSCs were shown to exhibit MRI signal reduction in vitro and was detectable in an in vivo model. In this study, we demonstrate a rapid, robust, and generic methodology that may be a useful and practical adjuvant to existing methods of cell labeling for in vivo monitoring by MRI. Further, we have shown the first application of XRF to provide iron maps to validate MRI data in SPIO-labeled cell tracking studies.


2019 ◽  
Vol 21 ◽  
pp. 102063 ◽  
Author(s):  
Vladimir Mulens-Arias ◽  
José Manuel Rojas ◽  
Laura Sanz-Ortega ◽  
Yadileiny Portilla ◽  
Sonia Pérez-Yagüe ◽  
...  

PLoS ONE ◽  
2015 ◽  
Vol 10 (2) ◽  
pp. e0115636 ◽  
Author(s):  
Christian NDong ◽  
Jennifer A. Tate ◽  
Warren C. Kett ◽  
Jaya Batra ◽  
Eugene Demidenko ◽  
...  

2012 ◽  
Vol 5 (1) ◽  
pp. 108-114 ◽  
Author(s):  
He Wei ◽  
Oliver T. Bruns ◽  
Ou Chen ◽  
Moungi G. Bawendi

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1998-1998
Author(s):  
Nancy D Doolittle ◽  
Edit Dosa ◽  
Edward A Neuwelt

Abstract Abstract 1998 Poster Board I-1020 Ferumoxytol (Feraheme™), an iron oxide nanoparticle, was approved as an iron replacement product for the treatment of iron deficiency anemia in adult patients with chronic kidney disease (CKD) (United States Food and Drug Administration, June 2009). Potential uses of ferumoxytol extend beyond the treatment of iron deficiency anemia and may have important implications for hematology-oncology. Following administration, iron oxide nanoparticles initially serve as blood pool agents which is an ideal characteristic for assessing blood volume and blood flow. Subsequently, because of their long plasma half-life and ability to target inflammatory cells in the central nervous system (CNS), these compounds serve as excellent anatomic imaging agents. We have given adult patients with malignant CNS tumors and other CNS inflammatory lesions, ferumoxytol (n = 68) and ferumoxytran-10 [Combidex™’ (n = 97) as magnetic resonance (MR) contrast agents, on Institutional Review Board approved protocols. We hypothesize that iron oxide nanoparticles provide additional MR imaging information which may be beneficial in the differential diagnosis and in monitoring therapy in patients with CNS lesions. This report focuses on our experience with 16 patients with central nervous system lymphoma (CNSL). The patients underwent brain MR with gadolinium followed in 10 ± 9 days (mean ± SD) by MR with ferumoxytran-10 (2.6 mg/kg, intravenous [IV’ over 30 min) (n = 11) or ferumoxytol (510 mg, IV bolus) (n = 5). Findings of interest include: 1) in 3 patients, MR with iron oxide nanoparticles showed additional areas of CNS enhancement when compared with MR with gadolinium; 2) in 2 patients, increased enhancement patterns with iron oxide nanoparticles were diagnostically useful in distinguishing lymphoma from non-lymphomatous inflammatory conditions; and 3) in 2 patients with histories of CKD and post-renal transplant lymphoproliferative disorder confined to the CNS, MR with ferumoxytol was used to monitor therapy, as this agent appears to avoid the risk of nephrogenic systemic fibrosis (NSF), which is associated with exposure to gadolinium-containing contrast agents. Of note, in 1 patient with thromboembolic complications during therapy, brain MR enhancement was seen approximately 5 weeks after ferumoxytol infusion. The enhancement was initially interpreted as a hemorrhagic brainstem event and led to insertion of a superior vena cava filter rather than anticoagulation. This is an example of MR alteration which generally resolves in 3 to 5 days following ferumoxytol, however in rare instances may persist for up to 3 months. In summary, in addition to their use as iron replacement agents in iron deficiency anemia, the use of iron oxide compounds as MR contrast agents compliment the use of gadolinium. These agents offer exciting potential in the differential diagnosis of CNS inflammatory lesions versus CNSL, in monitoring CNS therapy, and may provide dual benefit in patients with CKD by possibly eliminating the gadolinium-associated risk of NSF. Disclosures: Off Label Use: Feraheme is an iron replacement product indicated for the treatment of iron deficiency in adult patients with chronic kidney disease (CKD).


2015 ◽  
Vol 16 (10) ◽  
pp. 24417-24450 ◽  
Author(s):  
Ujwal Patil ◽  
Shiva Adireddy ◽  
Ashvin Jaiswal ◽  
Sree Mandava ◽  
Benjamin Lee ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document