Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
2.
Int J Mol Sci ; 25(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38673932

ABSTRACT

Platinum-containing chemotherapeutic drugs are efficacious in many forms of cancer but are dose-restricted by serious side effects, of which peripheral neuropathy induced by oxidative-nitrosative-stress-mediated chain reactions is most disturbing. Recently, hope has been raised regarding the catalytic antioxidants mangafodipir (MnDPDP) and calmangafodipir [Ca4Mn(DPDP)5; PledOx®], which by mimicking mitochondrial manganese superoxide dismutase (MnSOD) may be expected to overcome oxaliplatin-associated chemotherapy-induced peripheral neuropathy (CIPN). Unfortunately, two recent phase III studies (POLAR A and M trials) applying Ca4Mn(DPDP)5 in colorectal cancer (CRC) patients receiving multiple cycles of FOLFOX6 (5-FU + oxaliplatin) failed to demonstrate efficacy. Instead of an anticipated 50% reduction in the incidence of CIPN in patients co-treated with Ca4Mn(DPDP)5, a statistically significant increase of about 50% was seen. The current article deals with confusing differences between early and positive findings with MnDPDP in comparison to the recent findings with Ca4Mn(DPDP)5. The POLAR failure may also reveal important mechanisms behind oxaliplatin-associated CIPN itself. Thus, exacerbated neurotoxicity in patients receiving Ca4Mn(DPDP)5 may be explained by redox interactions between Pt2+ and Mn2+ and subtle oxidative-nitrosative chain reactions. In peripheral sensory nerves, Pt2+ presumably leads to oxidation of the Mn2+ from Ca4Mn(DPDP)5 as well as from Mn2+ in MnSOD and other endogenous sources. Thereafter, Mn3+ may be oxidized by peroxynitrite (ONOO-) into Mn4+, which drives site-specific nitration of tyrosine (Tyr) 34 in the MnSOD enzyme. Conformational changes of MnSOD then lead to the closure of the superoxide (O2•-) access channel. A similar metal-driven nitration of Tyr74 in cytochrome c will cause an irreversible disruption of electron transport. Altogether, these events may uncover important steps in the mechanism behind Pt2+-associated CIPN. There is little doubt that the efficacy of MnDPDP and its therapeutic improved counterpart Ca4Mn(DPDP)5 mainly depends on their MnSOD-mimetic activity when it comes to their potential use as rescue medicines during, e.g., acute myocardial infarction. However, pharmacokinetic considerations suggest that the efficacy of MnDPDP on Pt2+-associated neurotoxicity depends on another action of this drug. Electron paramagnetic resonance (EPR) studies have demonstrated that Pt2+ outcompetes Mn2+ and endogenous Zn2+ in binding to fodipir (DPDP), hence suggesting that the previously reported protective efficacy of MnDPDP against CIPN is a result of chelation and elimination of Pt2+ by DPDP, which in turn suggests that Mn2+ is unnecessary for efficacy when it comes to oxaliplatin-associated CIPN.


Subject(s)
Antineoplastic Agents , Manganese , Oxaliplatin , Peripheral Nervous System Diseases , Platinum , Humans , Antineoplastic Agents/adverse effects , Edetic Acid/analogs & derivatives , Manganese/adverse effects , Nitrosative Stress/drug effects , Oxaliplatin/adverse effects , Oxaliplatin/pharmacology , Oxidative Stress/drug effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/metabolism , Platinum/adverse effects , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Pyridoxal Phosphate/metabolism , Superoxide Dismutase/metabolism , Clinical Trials, Phase III as Topic
3.
Antioxidants (Basel) ; 12(3)2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36978857

ABSTRACT

Disappointing results from the POLAR A and M phase III trials involving colorectal cancer patients on chemotherapy with FOLFOX6 in curative (A) and palliative (M) settings have been reported by the principal investigators and the sponsor (PledPharma AB/Egetis Therapeutics AB). FOLFOX6, oxaliplatin in combination with 5-fluorouracil (5-FU), possesses superior tumoricidal activity in comparison to 5-FU alone, but suffers seriously from dose-limiting platinum-associated Chemotherapy-Induced Peripheral Neuropathy (CIPN). The aim of the POLAR trials was to demonstrate that PledOx [calmangafodipir; Ca4Mn(DPDP)5] reduced the incidence of persistent CIPN from 40% to 20%. However, this assumption was based on "explorative" data in the preceding PLIANT phase II trial, which did not mirror the expected incidence of unwanted toxicity in placebo patients. In POLAR A and M, the assessment of PledOx efficacy was conducted in patients that received at least six cycles of FOLFOX6, enabling analyses of efficacy in 239 A and 88 M patients. Instead of a hypothesized improvement from 40% to 20% incidence of persistent CIPN in the PledOx group, i.e., a 50% improvement, the real outcome was the opposite, i.e., an about 50% worsening in this bothersome toxicity. Mechanisms that may explain the disastrous outcome, with a statistically significant number of patients being seriously injured after having received PledOx, indicate interactions between two redox active metal cations, Pt2+ (oxaliplatin) and Mn2+ (PledOx). A far from surprising causal relationship that escaped prior detection by the study group and the sponsor. Most importantly, recently published data (ref 1) unequivocally indicate that the PLIANT study was not suited to base clinical phase III studies on. In conclusion, the POLAR and PLIANT trials show that PledOx and related manganese-containing compounds are unsuited for co-treatment with platinum-containing compounds. For use as a therapeutic adjunct in rescue treatment, like in ischemia-reperfusion of the heart or other organs, or in acetaminophen (paracetamol)-associated liver failure, there is little or nothing speaking against the use of PledOx or other PLED compounds. However, this must be thoroughly documented in more carefully designed clinical trials.

4.
Antioxidants (Basel) ; 10(12)2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34943040

ABSTRACT

On 2 July 2021, highly negative results were reported from the POLAR A and M phase III trials in patients with colorectal cancer, treated with an oxaliplatin-based regimen and co-treated with calmangafodipir (CaM; PledOx®; PledPharma AB/Egetis Therapeutics AB) or placebo. The results revealed persistent chemotherapy-induced peripheral neuropathy (CIPN) in 54.8% of the patients treated with PledOx, compared with 40.0% of the patients treated with the placebo (p < 0.05), i.e., a 37% increase in incidence of the side effect that the trial was aimed to prevent. The damaging outcome of the trials differed diametrically from an in-parallel conducted mice study and from a clinical trial with mangafodipir, the active ingredient of CaM. According to the authors of the POLAR report, the etiology of the profound increase in CIPN in the PledOx arm is unclear. However, these devastating effects are presumably explained by intravenous administrations of PledOx and oxaliplatin being too close in time and, thereby, causing unfavorable redox interactions between Mn2+ and Pt2-. In the mice study as well as in the preceding phase II clinical trial (PLIANT), PledOx was administered 10 min before the start of the oxaliplatin infusion; this was clearly an administration procedure, where the devastating interactions between PledOx and oxaliplatin could be avoided. However, when it comes to the POLAR trials, PledOx was administered, for incomprehensible reasons, "on Top of Modified FOLFOX6" at day one, i.e., after the two-hour oxaliplatin infusion instead of before oxaliplatin. This is a time point when the plasma concentration of oxaliplatin and Pt2+-metabolites is at its highest, and where the risk of devastating redox interactions between PledOx and oxaliplatin, in turn, is at its highest.

5.
Antioxidants (Basel) ; 9(10)2020 Oct 10.
Article in English | MEDLINE | ID: mdl-33050459

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is characterized by massive inflammation of the arterial endothelium accompanied by vasoconstriction and widespread pulmonary micro thrombi. As a result, due to the destruction of nitric oxide (•NO) by inflammatory superoxide (O2•-), pulmonary •NO concentration ceases, resulting in uncontrolled platelet aggregation and massive thrombosis, which kills the patients. Introducing •NO by inhalation (INO) may replace the loss of endothelium-derived •NO. The first results from clinical trials with INO in SARS-CoV-2 patients show a rapid and sustained improvement in cardiopulmonary function and decreased inflammation. An ongoing phase III study is expected to confirm the method's efficacy. INO may hence become a first line treatment in SARS-CoV-2 patients. However, due to the rapid inactivation of •NO by deoxyhemoglobin to nitrate, pulmonary administration of •NO will not protect remote organs. Another INO-related pharmacological approach to protect SARS-CoV-2 patients from developing life-threatening disease is to inhibit the O2•--driven destruction of •NO by neutralizing inflammatory O2•-. By making use of low molecular weight compounds that mimic the action of the enzyme manganese superoxide dismutase (MnSOD). The MnSOD mimetics of the so-called porphyrin type (e.g., AEOL 10150), salen type (e.g., EUK-8) and cyclic polyamine type (e.g., M40419, today known as GC4419 and avasopasem manganese) have all been shown to positively affect the inflammatory response in lung epithelial cells in preclinical models of chronic obstructive pulmonary disease. The Manganese diPyridoxyL EthylDiamine (MnPLED)-type mangafodipir (manganese dipyridoxyl diphosphate-MnDPDP), a magnetic resonance imaging (MRI) contrast agent that possesses MnSOD mimetic activity, has shown promising results in various forms of inflammation, in preclinical as well as clinical settings. Intravenously administration of mangafodipir will, in contrast to INO, reach remote organs and may hence become an important supplement to INO. From the authors' viewpoint, it appears logical to test mangafodipr in COVID-19 patients at risk of developing life-threatening SARS-CoV-2. Five days after submission of the current manuscript, Galera Pharmaceuticals Inc. announced the dosing of the first patient in a randomized, double-blind pilot phase II clinical trial with GC4419 for COVID-19. The study was first posted on ClinicalTrials.gov (Identifier: NCT04555096) 18 September 2020.

6.
Contrast Media Mol Imaging ; 2020: 3262835, 2020.
Article in English | MEDLINE | ID: mdl-32994754

ABSTRACT

The semistable chelate manganese (Mn) dipyridoxyl diphosphate (MnDPDP, mangafodipir), previously used as an intravenous (i.v.) contrast agent (Teslascan™, GE Healthcare) for Mn-ion-enhanced MRI (MEMRI), should be reappraised for clinical use but now as a diagnostic drug with cytoprotective properties. Approved for imaging of the liver and pancreas, MnDPDP enhances contrast also in other targets such as the heart, kidney, glandular tissue, and potentially retina and brain. Transmetallation releases paramagnetic Mn2+ for cellular uptake in competition with calcium (Ca2+), and intracellular (IC) macromolecular Mn2+ adducts lower myocardial T 1 to midway between native values and values obtained with gadolinium (Gd3+). What is essential is that T 1 mapping and, to a lesser degree, T 1 weighted imaging enable quantification of viability at a cellular or even molecular level. IC Mn2+ retention for hours provides delayed imaging as another advantage. Examples in humans include quantitative imaging of cardiomyocyte remodeling and of Ca2+ channel activity, capabilities beyond the scope of Gd3+ based or native MRI. In addition, MnDPDP and the metabolite Mn dipyridoxyl diethyl-diamine (MnPLED) act as catalytic antioxidants enabling prevention and treatment of oxidative stress caused by tissue injury and inflammation. Tested applications in humans include protection of normal cells during chemotherapy of cancer and, potentially, of ischemic tissues during reperfusion. Theragnostic use combining therapy with delayed imaging remains to be explored. This review updates MnDPDP and its clinical potential with emphasis on the working mode of an exquisite chelate in the diagnosis of heart disease and in the treatment of oxidative stress.


Subject(s)
Brain/diagnostic imaging , Contrast Media/metabolism , Edetic Acid/analogs & derivatives , Heart/physiology , Manganese/chemistry , Pyridoxal Phosphate/analogs & derivatives , Retina/diagnostic imaging , Brain/metabolism , Edetic Acid/metabolism , Heart/diagnostic imaging , Humans , Magnetic Resonance Imaging , Pyridoxal Phosphate/metabolism , Retina/metabolism
8.
Acta Oncol ; 57(6): 862-864, 2018 06.
Article in English | MEDLINE | ID: mdl-29243552
9.
Transl Oncol ; 10(4): 641-649, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28668762

ABSTRACT

Oxaliplatin, in combination with 5-fluorouracil plus folinate (or capecitabine), has increased survival substantially in stage III colorectal cancer and prolonged life in stage IV patients, but its use is compromised because of severe toxicity. Chemotherapy-induced peripheral neuropathy (CIPN) is the most problematic dose-limiting toxicity of oxaliplatin. Oncologists included for years calcium and magnesium infusion as part of clinical practice for preventing CIPN. Results from a phase III prospective study published in 2014, however, overturned this practice. No other treatments have been clinically proven to prevent this toxicity. There is a body of evidence that CIPN is caused by cellular oxidative stress. Clinical and preclinical data suggest that the manganese chelate and superoxide dismutase mimetic mangafodipir (MnDPDP) is an efficacious inhibitor of CIPN and other conditions caused by cellular oxidative stress, without interfering negatively with the tumoricidal activity of chemotherapy. MnPLED, the metabolite of MnDPDP, attacks cellular oxidative stress at several critical levels. Firstly, MnPLED catalyzes dismutation of superoxide (O2•-), and secondly, having a tremendous high affinity for iron (and copper), PLED binds and disarms redox active iron/copper, which is involved in several detrimental oxidative steps. A case report from 2009 and a recent feasibility study suggest that MnDPDP may prevent or even cure oxaliplatin-induced CIPN. Preliminary results from a phase II study (PLIANT) suggest efficacy also of calmangafodipir, but these results are according to available data obscured by a surprisingly low number of adverse events and a seemingly lower than expected efficacy of FOLFOX.

10.
Article in English | MEDLINE | ID: mdl-27533964

ABSTRACT

AIMS: The aim of the present study was to examine the feasibility of applying the catalytic antioxidant mangafodipir [MnDPDP, manganese (Mn) dipyridoxyl diphosphate] as a cardioprotective adjunct to primary percutaneous coronary intervention (pPCI) in patients with ST-segment elevation (STE) myocardial infarction (STEMI). Both MnDPDP and a metabolite (Mn dipyridoxyl ethyldiamine) possess properties as mitochondrial superoxide dismutase mimetics and iron chelators, and combat oxidative stress in various tissues and conditions. METHODS AND RESULTS: The study tested MnDPDP (n = 10) vs. saline placebo (n = 10), given as a brief intravenous (i.v.) infusion prior to balloon inflation during pPCI in patients with STEMI. Mangafodipir was well tolerated and did not affect heart rate or blood pressure. Despite longer ischaemic time (205 vs. 144 min, P = 0.019) in the MnDPDP group, plasma biomarker releases were identical for the two groups. With placebo vs. MnDPDP, mean STE resolutions were 69.8 vs. 81.9% (P = 0.224) at 6 h and 73.1 vs. 84.3% (P = 0.077) at 48 h. Cardiac magnetic resonance revealed mean infarct sizes of 32.5 vs. 26.2% (P = 0.406) and mean left ventricular (LV) ejection fractions of 41.8 vs. 47.7% (P = 0.617) with placebo vs. MnDPDP. More LV thrombi were detected in placebo hearts (5 of 8) than MnDPDP-treated hearts (1 of 10; P = 0.011). CONCLUSIONS: Mangafodipir is a safe drug for use as an adjunct to reperfusion therapy. A tendency to benefit of MnDPDP needs confirmation in a larger population. The study revealed important information for the design of a Phase II trial.


Subject(s)
Edetic Acid/analogs & derivatives , Heart Ventricles/diagnostic imaging , Percutaneous Coronary Intervention/methods , Pyridoxal Phosphate/analogs & derivatives , ST Elevation Myocardial Infarction/drug therapy , Ventricular Function, Left/physiology , Dose-Response Relationship, Drug , Edetic Acid/administration & dosage , Feasibility Studies , Female , Follow-Up Studies , Heart Ventricles/physiopathology , Humans , Infusions, Intravenous , Magnetic Resonance Imaging, Cine , Male , Middle Aged , Pyridoxal Phosphate/administration & dosage , Retrospective Studies , ST Elevation Myocardial Infarction/diagnosis , ST Elevation Myocardial Infarction/surgery , Treatment Outcome
11.
Drug Discov Today ; 20(4): 411-21, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25463039

ABSTRACT

Reactive oxygen species (ROS) and reactive nitrogen species (RNS) participate in pathological tissue damage. Mitochondrial manganese superoxide dismutase (MnSOD) normally keeps ROS and RNS in check. During development of mangafodipir (MnDPDP) as a magnetic resonance imaging (MRI) contrast agent, it was discovered that MnDPDP and its metabolite manganese pyridoxyl ethyldiamine (MnPLED) possessed SOD mimetic activity. MnDPDP has been tested as a chemotherapy adjunct in cancer patients and as an adjunct to percutaneous coronary intervention in patients with myocardial infarctions, with promising results. Whereas MRI contrast depends on release of Mn(2+), the SOD mimetic activity depends on Mn(2+) that remains bound to DPDP or PLED. Calmangafodipir [Ca4Mn(DPDP)5] is stabilized with respect to Mn(2+) and has superior therapeutic activity. Ca4Mn(DPDP)5 is presently being explored as a chemotherapy adjunct in a clinical multicenter Phase II study in patients with metastatic colorectal cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Antioxidants/therapeutic use , Biological Mimicry , Edetic Acid/analogs & derivatives , Ethylenediamines/therapeutic use , Manganese/metabolism , Pyridoxal Phosphate/analogs & derivatives , Superoxide Dismutase/metabolism , Animals , Antineoplastic Agents/chemistry , Antioxidants/chemistry , Antioxidants/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Edetic Acid/chemistry , Edetic Acid/metabolism , Edetic Acid/therapeutic use , Ethylenediamines/chemistry , Ethylenediamines/metabolism , Humans , Manganese/chemistry , Molecular Structure , Myocardial Infarction/therapy , Oxidative Stress/drug effects , Percutaneous Coronary Intervention , Pyridoxal Phosphate/chemistry , Pyridoxal Phosphate/metabolism , Pyridoxal Phosphate/therapeutic use , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Treatment Outcome
12.
Transl Oncol ; 5(1): 32-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22348174

ABSTRACT

Preclinical research suggests that the clinically approved magnetic resonance imaging contrast agent mangafodipir may protect against adverse events (AEs) caused by chemotherapy, without interfering negatively with the anticancer efficacy. The present translational study tested if pretreatment with mangafodipir lowers AEs during curative (adjuvant) FOLFOX6 chemotherapy in stage III colon cancer (Dukes' C). The study was originally scheduled to include 20 patients, but because of the unforeseen withdrawal of mangafodipir from the market, the study had to be closed after 14 patients had been included. The withdrawal of mangafodipir was purely based on commercial considerations from the producer and not on any safety concerns. The patients were treated throughout the first 3 of 12 scheduled cycles. Patients were randomized to a 5-minute infusion of either mangafodipir or placebo (7 in each group). AEs were evaluated according to the National Cancer Institute's (NCI) Common Terminology Criteria for Adverse Events and the Sanofi-NCI criteria. The primary end points were neutropenia and neurosensory toxicity. There were four AEs of grade 3 (severe) and one AE of grade 4 (life threatening) in four patients in the placebo group, whereas there were none in the mangafodipir group (P < .05). Of the grade 3 and 4 events, two were neutropenia and one was neurosensory toxicity. Furthermore, white blood cell count was statistically, significantly higher in the mangafodipir group than in the placebo group (P < .01) after treatment with FOLFOX. This small feasibility study seems to confirm what has been demonstrated preclinically, namely, that pretreatment with mangafodipir lowers AEs during adjuvant 5-fluorouracil plus oxaliplatin-based chemotherapy in colon cancer patients.

14.
Scand J Clin Lab Invest ; 68(2): 106-16, 2008.
Article in English | MEDLINE | ID: mdl-17852833

ABSTRACT

OBJECTIVE: Ischaemic preconditioning may protect the intestine from subsequent prolonged ischaemia. This study evaluates whether a much longer initial ischaemia, encountered clinically, may modify intestinal resistance to further ischaemia in a pig model. MATERIAL AND METHODS: After cross-clamping of the superior mesenteric artery for 1 h, the intestine was either reperfused for 8 h or a second cross-clamping for 1 h was performed at 4 h of reperfusion. Based on microarray analysis of intestinal samples at 1, 4 and 8 h of reperfusion, mRNA of selected genes was measured with QRT-PCR. RESULTS: The first ischaemic period caused exfoliation of surface epithelial cells from the basement membrane comprising about 90 % of the villi tips, a marked increase in permeability and depletion of ATP. The second ischaemic challenge caused about 30 % less denudation of the basement membrane (p = 0.008), no increase in permeability (p = 0.008) and less depletion of ATP (p = 0.039). mRNAs for superoxide dismutase 2, heat shock proteins and signal transducer and activator of transcription 3, which may protect against ischaemia/reperfusion injury, were up-regulated throughout the reperfusion period. mRNAs for matrix metalloproteinase 1, connexin 43 and peripheral myelin 22, which may be associated with cell migration or tight junctions, showed a particular up-regulation at 4 h of reperfusion. CONCLUSION: One hour of initial ischaemia followed by 4 h of reperfusion is associated with increased intestinal resistance to further ischaemia. The differential regulation of genes identified in this study provides working hypotheses for mechanisms behind this observation.


Subject(s)
Intestinal Mucosa/pathology , Ischemia/pathology , Animals , Intestinal Absorption , Intestinal Mucosa/blood supply , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Regional Blood Flow
15.
Magn Reson Med ; 58(3): 442-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17763343

ABSTRACT

Ischemic changes in excised rat myocardium were followed by series of T1 or T2 measurements from 1 to 60 min after isolated perfusion cessation, and the influence of manganese enhancement was investigated. An inverse Laplace transformation (ILT) of T1 or T2 data was used to resolve the number, time constants, and fractions of tissue water components in a continuous distribution. For T1 distributions, one single tissue component approximately 900 ms was significantly shortened and dispersed by manganese enhancement (25 and 200 microM MnCl2). For T2 distributions, three tissue components (approximately 30, approximately 100, and approximately 350 ms) were obtained initially. The two shortest components merged after approximately 10 min to one component (approximately 40 ms). Both T1 and T2 tissue components became shorter with time. In particular, the T2 distribution dynamics might be compatible with complex sequential changes in tissue water fractions during ischemia.


Subject(s)
Body Water/metabolism , Magnetic Resonance Spectroscopy , Myocardial Ischemia/metabolism , Myocardium/metabolism , Algorithms , Animals , Chlorides/pharmacokinetics , Extracellular Fluid/metabolism , Heart Ventricles/metabolism , Intracellular Fluid/metabolism , Male , Manganese Compounds/pharmacokinetics , Rats , Rats, Wistar , Time Factors
16.
Magn Reson Med ; 58(4): 674-86, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17899606

ABSTRACT

Water compartments were identified and equilibrium water exchange was studied in excised rat myocardium enriched with intracellular manganese (Mn(2+)). Standard relaxographic measurements were supplemented with diffusion-T(2) and T(1)-T(2) correlation measurements. In nonenriched myocardium, one T(1) component (800 ms) and three T(2) components (32, 120, and 350 ms) were identified. The correlation measurements revealed fast- and slow-diffusing water fractions with mean diffusion coefficients of 1.2 x 10(-5) and 3.0 x 10(-5) cm(2) s(-1). The two shortest T(2) components, which had different diffusivities, both originated from water in intracellular compartments. A component with longer relaxation time (T(1) approximately equal 2200 ms; T(2) approximately equal 1200 ms), originating from extra-tissue water, was also observed. The presence of this component may lead to erroneous estimations of water exchange rates from multiexponential relaxographic analyses of excised tissues. The tissue T(1) value is strongly reduced with increasing enrichment of Mn(2+), and eventually a second tissue T(1) component emerges, indicating a shift in the equilibrium water exchange between intra- and extracellular compartments from the fast-exchange limit to the slow-exchange regime. Using a two-site water exchange analysis, the lifetime of intracellular water, T(ic), was found to be 475 ms, with a fraction, p(ic), of 0.71.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Myocardium/metabolism , Water/metabolism , Animals , Extracellular Fluid/metabolism , Intracellular Fluid/metabolism , Male , Manganese/analysis , Rats , Rats, Wistar
17.
J Magn Reson Imaging ; 26(3): 720-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17729351

ABSTRACT

PURPOSE: To evaluate contrast accumulation in left ventricular (LV) myocardium after manganese dipyridoxyl-diphosphate (MnDPDP) administration in patients with recent first time myocardial infarction. MATERIALS AND METHODS: MnDPDP (5 micromol/kg) was administered to 10 patients with recent myocardial infarction (three to 12 weeks). One slice of interest (SOI) likely to traverse the infarction was chosen, and sectorial pre- and postcontrast longitudinal relaxivity rates (R(1)) and signal changes during infusion were estimated with a fast gradient echo sequence. LV volume and wall thickening were measured in short-axis cine recordings. Infarct localization from R(1) and wall thickening data were compared by vector analyses. RESULTS: Reduced wall thickening was associated with reduced precontrast R(1) and reduced contrast enhancement. Both remote and infarcted regions showed rapid initial contrast accumulation. In remote regions, this was followed by a continuing slow increase. Mean precontrast R(1) was 0.87 +/- 0.06 second(-1) in infarcted regions and 0.96 +/- 0.03 second(-1) in remote regions (P < 0.001). Mean R(1) change over one hour was 0.24 +/- 0.07 second(-1) in infarcted regions and 0.38 +/- 0.03 second(-1) in remote regions (P < 0.0001). CONCLUSION: Remote regions showed larger increases in R(1) than infarcted regions. This is most likely due to selective and slow Mn accumulation in viable myocytes.


Subject(s)
Contrast Media/pharmacology , Echocardiography/methods , Edetic Acid/analogs & derivatives , Heart Ventricles/pathology , Magnetic Resonance Imaging/methods , Myocardial Infarction/diagnosis , Myocardial Infarction/pathology , Pyridoxal Phosphate/analogs & derivatives , Adult , Aged , Biomarkers/chemistry , Coronary Vessels/pathology , Edetic Acid/pharmacology , Female , Humans , Male , Middle Aged , Myocardial Infarction/mortality , Pyridoxal Phosphate/pharmacology , Treatment Outcome
18.
J Magn Reson Imaging ; 24(5): 1047-55, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17024667

ABSTRACT

PURPOSE: To develop an in vivo MR method for evaluation of myocardial calcium channel activity through quantification of apparent unidirectional manganese influx constants following manganese dipyridoxyl-diphosphate (MnDPDP) infusions. MATERIALS AND METHODS: A total of 10 healthy volunteers were divided in two groups, and received 5 micromol of MnDPDP per kg of body weight intravenously in a 1.5 Tesla scanner over five or 30 minutes, respectively. A fast inversion recovery gradient echo sequence was used to estimate pre- and postcontrast R1 values and to measure signal changes following infusions. By assuming equal longitudinal relaxivity (r1) of the contrast in all tissue compartments, signal changes in blood and myocardial tissue yielded temporal input and tissue contrast concentrations respectively. Through a two-tissue compartment model, apparent unidirectional influx constants (Ki) for myocardial manganese accumulation were estimated. RESULTS: Consistent values for Ki in left ventricular wall were found, with a mean value of 5.96 mL/100 g/minute (SD=0.49; N=10). No statistical significant differences in Ki were found between the two infusion groups. CONCLUSION: Since unidirectional manganese accumulation depends upon intact myocyte membranes with functioning calcium channels, the use of unidirectional manganese influx rates may be a valuable research tool for in vivo studies of myocyte functioning in myocardial disease.


Subject(s)
Calcium Channels/metabolism , Echo-Planar Imaging/methods , Edetic Acid/analogs & derivatives , Image Interpretation, Computer-Assisted/methods , Ion Channel Gating/physiology , Myocardium/metabolism , Pyridoxal Phosphate/analogs & derivatives , Adult , Contrast Media/pharmacokinetics , Edetic Acid/pharmacokinetics , Female , Humans , Kinetics , Male , Metabolic Clearance Rate , Pyridoxal Phosphate/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Reference Values
19.
Scand Cardiovasc J ; 40(5): 291-7, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17012140

ABSTRACT

OBJECTIVE: To investigate oxidative stress and myocardial injury at different stages of coronary artery bypass grafting (CABG). DESIGN: Twenty patients underwent CABG with use of cardiopulmonary bypass (CPB) and with intermittent sampling of plasma and urine. Main markers were: 8-iso-PGF2alpha (oxidative stress); troponin T (myocardial injury); and 15-keto-dihydro-PGF2alpha and hsCRP (inflammation). RESULTS: Plasma 8-iso-PGF2alpha increased after start of surgery, but there was no further rise during CPB or after aortic cross-clamp release and no significant myocardial arterio-venous differences. An increase in troponin T was seen early after the operation, but no relationship was established between 8-iso-PGF2alpha and troponin T. 8-iso-PGF2alpha levels were elevated by preoperative withdrawal of acetylsalicylic acid (ASA) but reduced by intraoperative use of heparin. 15-keto-dihydro-PGF2alpha was elevated during operation and hsCRP following operation. CONCLUSIONS: In the present study oxidative stress was multifactorial in origin with main impacts from surgical trauma, less from CPB and little if any from myocardial ischemia-reperfusion events. In addition, cardiovascular drugs in common use like ASA and heparin seemed to influence the pro- and antioxidant balance, a finding that has to be confirmed in future studies.


Subject(s)
Coronary Artery Bypass/adverse effects , Coronary Artery Bypass/methods , Oxidative Stress , Aged , Biomarkers , Dinoprost/analogs & derivatives , Dinoprost/analysis , Drug Therapy , Female , Humans , Male , Middle Aged , Myocardial Reperfusion , Oxidative Stress/drug effects , Troponin T/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...