Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Bull Cancer ; 104(9): 727-734, 2017 Sep.
Article in French | MEDLINE | ID: mdl-28778339

ABSTRACT

INTRODUCTION: The increasing prescription of oral anticancer therapies has significantly changed inpatient care to outpatient care. This transformation requires an excellent coordination between different professionals to ensure healthcare channel security. METHOD: We performed a prospective study in 18 French cancer centers from March to April 2016. The aim of this study was to identify resources deployed to support patients receiving oral anticancer therapies and to assess pharmacist's involvement. RESULTS: More than half of the centers have developed patient education program and/or practice pharmaceutical consultations. In total, 54.5% have deployed an oral anticancer drugs program and the pharmacist is involved in multidisciplinary teams. In total, 44.4% of the centers have developed hospital-to-community coordination actions but all of them highlight the time-consuming character of those programs. DISCUSSION: Administrative burdens are seriously hindering patient education program's development. Multidisciplinary consultations can offer an attractive alternative because of easy implementation modalities. Finally, hospital-to-community coordination actions seem hard to implement and require harmonization of communication practices, and need more technical and financial means.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Patient Education as Topic/organization & administration , Pharmacists , Professional Role , Program Development , Administration, Oral , Health Education/methods , Humans , Program Development/statistics & numerical data , Prospective Studies , Surveys and Questionnaires
2.
Bull Cancer ; 103(7-8): 674-90, 2016.
Article in French | MEDLINE | ID: mdl-27286758

ABSTRACT

BACKGROUND: Patient education is the process by which health professionals impart information to patients and their caregivers that will alter their health behaviors; improve their health status to better manage their lives with a chronic disease. Patient education implies a profound paradigm shift in the conception of care among health professionals, and should result in structural care changes. Patient education has been promoted by the French Health system for 30years, including in the 2009 HPST law and Cancer Plan 2014-2019. A patient education program was designed in our hospital for breast cancer patients. MATERIAL AND METHODS: A multidisciplinary and transversal team of health professionals and resource patients was trained before grant application for funding of the program by the regional health care agency. Management of the project required that a functional unit be built for recording of all patient education related activities. A customized patient education program process was built under the leadership of a coordinator and several patient education project managers during bimonthly meetings, using an accurate timeline and a communication strategy to ensure full institutional support and team engagement. RESULTS: The grant was prepared in four months and the program started within the next four months with the aim to include 120 patients during year 1. The program includes a diagnosis of patient abilities and well-being resources, followed by collective and individual workshops undertaken in 4months for each patient. DISCUSSION: Patient education is positively evaluated by all participants and may contribute to better health care management in the long term but the financial and human resources allocated to such programs currently underestimate the needs. Sustainability of patient education programs requires that specific tools and more commitment be developed to support health care professionals and to promote patient coping and empowerment in the long term.


Subject(s)
Medical Oncology , Patient Education as Topic , Program Development , France , Humans , Medical Oncology/education , Neoplasms/epidemiology , Program Development/economics
3.
PLoS Pathog ; 9(11): e1003790, 2013.
Article in English | MEDLINE | ID: mdl-24278025

ABSTRACT

A new generation of strategies is evolving that aim to block malaria transmission by employing genetically modified vectors or mosquito pathogens or symbionts that express anti-parasite molecules. Whilst transgenic technologies have advanced rapidly, there is still a paucity of effector molecules with potent anti-malaria activity whose expression does not cause detrimental effects on mosquito fitness. Our objective was to examine a wide range of antimicrobial peptides (AMPs) for their toxic effects on Plasmodium and anopheline mosquitoes. Specifically targeting early sporogonic stages, we initially screened AMPs for toxicity against a mosquito cell line and P. berghei ookinetes. Promising candidate AMPs were fed to mosquitoes to monitor adverse fitness effects, and their efficacy in blocking rodent malaria infection in Anopheles stephensi was assessed. This was followed by tests to determine their activity against P. falciparum in An. gambiae, initially using laboratory cultures to infect mosquitoes, then culminating in preliminary assays in the field using gametocytes and mosquitoes collected from the same area in Mali, West Africa. From a range of 33 molecules, six AMPs able to block Plasmodium development were identified: Anoplin, Duramycin, Mastoparan X, Melittin, TP10 and Vida3. With the exception of Anoplin and Mastoparan X, these AMPs were also toxic to an An. gambiae cell line at a concentration of 25 µM. However, when tested in mosquito blood feeds, they did not reduce mosquito longevity or egg production at concentrations of 50 µM. Peptides effective against cultured ookinetes were less effective when tested in vivo and differences in efficacy against P. berghei and P. falciparum were seen. From the range of molecules tested, the majority of effective AMPs were derived from bee/wasp venoms.


Subject(s)
Anopheles/parasitology , Antimalarials , Antimicrobial Cationic Peptides , Bee Venoms , Bees/chemistry , Insect Proteins , Malaria, Falciparum/drug therapy , Oocysts , Plasmodium berghei , Plasmodium falciparum , Animals , Antimalarials/chemistry , Antimalarials/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Bee Venoms/chemistry , Bee Venoms/pharmacology , Cell Line , Female , Humans , Insect Proteins/chemistry , Insect Proteins/pharmacology , Male , Mice
4.
Malar J ; 11: 393, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23181931

ABSTRACT

BACKGROUND: Anopheles plumbeus has been recognized as a minor vector for human malaria in Europe since the beginning of the 20th century. In recent years this tree hole breeding mosquito species appears to have exploited novel breeding sites, including large and organically rich man-made containers, with consequently larger mosquito populations in close vicinity to humans. This lead to investigate whether current populations of An. plumbeus would be able to efficiently transmit Plasmodium falciparum, the parasite responsible for the most deadly form of malaria. METHODS: Anopheles plumbeus immatures were collected from a liquid manure pit in Switzerland and transferred as adults to the CEPIA (Institut Pasteur, France) where they were fed on P. falciparum gametocytes produced in vitro. Anopheles gambiae mosquitoes served as controls. Development of P. falciparum in both mosquito species was followed by microscopical detection of oocysts on mosquito midguts and by sporozoite detection in the head/thorax by PCR and microscopy. RESULTS: A total of 293 wild An. plumbeus females from four independent collections successfully fed through a membrane on blood containing P. falciparum gametocytes. Oocysts were observed in mosquito midguts and P. falciparum DNA was detected in head-thorax samples in all four experiments, demonstrating, on a large mosquito sample, that An. plumbeus is indeed receptive to P. falciparum NF54 and able to produce sporozoites. Importantly, the proportion of sporozoites-infected An. plumbeus was almost similar to that of An. gambiae (31 to 88% An. plumbeus versus 67 to 97% An. gambiae). However, the number of sporozoites produced was significantly lower in infected An. plumbeus. CONCLUSION: The results show that a sample of field-caught An. plumbeus has a moderate to high receptivity towards P. falciparum. Considering the increased mobility of humans between Europe and malaria endemic countries and changes in environment and climate, these data strongly suggest that An. plumbeus could act as a vector for malaria and thus significantly contribute to increasing the malaria transmission risk in Central-Western Europe. In locations showing high vulnerability to the presence of gametocyte carriers, the risk of transmission of malaria by An. plumbeus should be considered.


Subject(s)
Anopheles/growth & development , Anopheles/parasitology , Disease Vectors , Plasmodium falciparum/growth & development , Animal Structures/parasitology , Animals , Female , Humans , Microscopy , Switzerland
5.
Proc Natl Acad Sci U S A ; 109(28): E1922-30, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22689959

ABSTRACT

Anopheles stephensi mosquitoes expressing m1C3, m4B7, or m2A10 single-chain antibodies (scFvs) have significantly lower levels of infection compared to controls when challenged with Plasmodium falciparum, a human malaria pathogen. These scFvs are derived from antibodies specific to a parasite chitinase, the 25 kDa protein and the circumsporozoite protein, respectively. Transgenes comprising m2A10 in combination with either m1C3 or m4B7 were inserted into previously-characterized mosquito chromosomal "docking" sites using site-specific recombination. Transgene expression was evaluated at four different genomic locations and a docking site that permitted tissue- and sex-specific expression was researched further. Fitness studies of docking site and dual scFv transgene strains detected only one significant fitness cost: adult docking-site males displayed a late-onset reduction in survival. The m4B7/m2A10 mosquitoes challenged with P. falciparum had few or no sporozoites, the parasite stage infective to humans, in three of four experiments. No sporozoites were detected in m1C3/m2A10 mosquitoes in challenge experiments when both genes were induced at developmentally relevant times. These studies support the conclusion that expression of a single copy of a dual scFv transgene can completely inhibit parasite development without imposing a fitness cost on the mosquito.


Subject(s)
Anopheles/genetics , Anopheles/immunology , Anopheles/parasitology , Gene Expression Regulation, Developmental , Gene Expression Regulation , Plasmodium falciparum/metabolism , Single-Chain Antibodies/chemistry , Animals , Animals, Genetically Modified , Binding Sites , Culicidae , Female , Genetic Engineering/methods , In Situ Hybridization, Fluorescence , Male , Models, Genetic , Plasmids/metabolism , Plasmodium falciparum/genetics , Sporozoites/metabolism , Transgenes
7.
PLoS One ; 7(12): e52684, 2012.
Article in English | MEDLINE | ID: mdl-23285147

ABSTRACT

Functional studies have demonstrated a role for the Anopheles gambiae APL1A gene in resistance against the human malaria parasite, Plasmodium falciparum. Here, we exhaustively characterize the structure of the APL1 locus and show that three structurally different APL1A alleles segregate in the Ngousso colony. Genetic association combined with RNAi-mediated gene silencing revealed that APL1A alleles display distinct protective profiles against P. falciparum. One APL1A allele is sufficient to explain the protective phenotype of APL1A observed in silencing experiments. Epitope-tagged APL1A isoforms expressed in an in vitro hemocyte-like cell system showed that under assay conditions, the most protective APL1A isoform (APL1A(2)) localizes within large cytoplasmic vesicles, is not constitutively secreted, and forms only one protein complex, while a less protective isoform (APL1A(1)) is constitutively secreted in at least two protein complexes. The tested alleles are identical to natural variants in the wild A. gambiae population, suggesting that APL1A genetic variation could be a factor underlying natural heterogeneity of vector susceptibility to P. falciparum.


Subject(s)
Alleles , Anopheles/genetics , Genes, Insect , Amino Acid Sequence , Animals , Anopheles/immunology , Anopheles/parasitology , Gene Order , Gene Silencing , Haplotypes , Molecular Sequence Data , Plasmodium falciparum/immunology , Protein Transport , Quantitative Trait Loci , Sequence Alignment
8.
PLoS One ; 6(1): e14587, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21283619

ABSTRACT

Diseases transmitted by mosquitoes have a devastating impact on global health and this is worsening due to difficulties with existing control measures and climate change. Genetically modified mosquitoes that are refractory to disease transmission are seen as having great potential in the delivery of novel control strategies. Historically the genetic modification of insects has relied upon transposable elements which have many limitations despite their successful use. To circumvent these limitations the Streptomyces phage phiC31 integrase system has been successfully adapted for site-specific transgene integration in insects. Here, we present the first site-specific transformation of Anopheles gambiae, the principal vector of human malaria. Mosquitoes were initially engineered to incorporate the phiC31 targeting site at a defined genomic location. A second phase of genetic modification then achieved site-specific integration of Vida3, a synthetic anti-malarial gene. Expression of Vida3, specifically in the midgut of bloodfed females, offered consistent and significant protection against Plasmodium yoelii nigeriensis, reducing average parasite intensity by 85%. Similar protection was observed against Plasmodium falciparum in some experiments, although protection was inconsistent. In the fight against malaria, it is imperative to establish a broad repertoire of both anti-malarial effector genes and tissue-specific promoters for their expression, enabling those offering maximum effect with minimum fitness cost to be identified. In the future, this technology will allow effective comparisons and informed choices to be made, potentially leading to complete transmission blockade.


Subject(s)
Anopheles/genetics , Antimalarials/administration & dosage , Gene Targeting/methods , Malaria/prevention & control , Transgenes/genetics , Animals , Animals, Genetically Modified , Female , Humans , Insect Vectors , Malaria/therapy , Plasmodium falciparum/drug effects , Plasmodium yoelii/drug effects
9.
Malar J ; 9: 231, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20704707

ABSTRACT

BACKGROUND: The prevalence of Plasmodium falciparum and Plasmodium vivax malaria was very high in Corsica just before the Second World War. The last outbreak was in 1972 and the most recent indigenous case was in 2006. RESULTS: Analysis of historical data shows that anopheline vectors were abundant. Recent surveys demonstrated that potential vectors are still present in Corsica, despite the likely disappearance of Anopheles sacharovi. Moreover, P. falciparum can develop experimentally into these mosquitoes, notably Anopheles labranchiae, which is locally abundant, and parasites are regularly introduced into the island. DISCUSSION, CONCLUSIONS: The presence of vectors, the introduction of parasites and the conducive climate raise questions about the possibility of malaria re-emerging and becoming re-established in Corsica. Analysis of historic and current parasitological and entomological data shows that the current theoretical risk of indigenous cases or malaria foci is negligible, particularly since there is very little contact between humans and Anopheles mosquitoes, Plasmodium carriers are reliably treated and there is a widespread vector control on the island.


Subject(s)
Anopheles/parasitology , Insect Vectors/parasitology , Malaria/epidemiology , Animals , France/epidemiology , Humans , Malaria/parasitology , Malaria/transmission , Plasmodium falciparum/growth & development , Plasmodium vivax/growth & development , Polymerase Chain Reaction , Risk
10.
PLoS One ; 5(7): e11538, 2010 Jul 12.
Article in English | MEDLINE | ID: mdl-20634948

ABSTRACT

BACKGROUND: Invasion of the mosquito salivary glands by Plasmodium is a critical step for malaria transmission. From a SAGE analysis, we previously identified several genes whose expression in salivary glands was regulated coincident with sporozoite invasion of salivary glands. To get insights into the consequences of these salivary gland responses, here we have studied one of the genes, PRS1 (Plasmodium responsive salivary 1), whose expression was upregulated in infected glands, using immunolocalization and functional inactivation approaches. METHODOLOGY/PRINCIPAL FINDINGS: PRS1 belongs to a novel insect superfamily of genes encoding proteins with DM9 repeat motifs of uncharacterized function. We show that PRS1 is induced in response to Plasmodium, not only in the salivary glands but also in the midgut, the other epithelial barrier that Plasmodium has to cross to develop in the mosquito. Furthermore, this induction is observed using either the rodent parasite Plasmodium berghei or the human pathogen Plasmodium falciparum. In the midgut, PRS1 overexpression is associated with a relocalization of the protein at the periphery of invaded cells. We also find that sporozoite invasion of salivary gland cells occurs sequentially and induces intra-cellular modifications that include an increase in PRS1 expression and a relocalization of the corresponding protein into vesicle-like structures. Importantly, PRS1 knockdown during the onset of midgut and salivary gland invasion demonstrates that PRS1 acts as an agonist for the development of both parasite species in the two epithelia, highlighting shared vector/parasite interactions in both tissues. CONCLUSIONS/SIGNIFICANCE: While providing insights into potential functions of DM9 proteins, our results reveal that PRS1 likely contributes to fundamental interactions between Plasmodium and mosquito epithelia, which do not depend on the specific Anopheles/P. falciparum coevolutionary history.


Subject(s)
Anopheles/metabolism , Anopheles/parasitology , Digestive System/parasitology , Insect Proteins/metabolism , Salivary Glands/metabolism , Salivary Glands/parasitology , Animals , Anopheles/genetics , Blotting, Western , Digestive System/metabolism , Insect Proteins/classification , Insect Proteins/genetics , Microscopy, Confocal , Phylogeny , Plasmodium berghei/pathogenicity , Plasmodium falciparum/pathogenicity , Reverse Transcriptase Polymerase Chain Reaction
11.
PLoS Pathog ; 5(9): e1000576, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19750215

ABSTRACT

Genetically controlled resistance of Anopheles gambiae mosquitoes to Plasmodium falciparum is a common trait in the natural population, and a cluster of natural resistance loci were mapped to the Plasmodium-Resistance Island (PRI) of the A. gambiae genome. The APL1 family of leucine-rich repeat (LRR) proteins was highlighted by candidate gene studies in the PRI, and is comprised of paralogs APL1A, APL1B and APL1C that share > or =50% amino acid identity. Here, we present a functional analysis of the joint response of APL1 family members during mosquito infection with human and rodent Plasmodium species. Only paralog APL1A protected A. gambiae against infection with the human malaria parasite P. falciparum from both the field population and in vitro culture. In contrast, only paralog APL1C protected against the rodent malaria parasites P. berghei and P. yoelii. We show that anti-P. falciparum protection is mediated by the Imd/Rel2 pathway, while protection against P. berghei infection was shown to require Toll/Rel1 signaling. Further, only the short Rel2-S isoform and not the long Rel2-F isoform of Rel2 confers protection against P. falciparum. Protection correlates with the transcriptional regulation of APL1A by Rel2-S but not Rel2-F, suggesting that the Rel2-S anti-parasite phenotype results at least in part from its transcriptional control over APL1A. These results indicate that distinct members of the APL1 gene family display a mutually exclusive protective effect against different classes of Plasmodium parasites. It appears that a gene-for-pathogen-class system orients the appropriate host defenses against distinct categories of similar pathogens. It is known that insect innate immune pathways can distinguish between grossly different microbes such as Gram-positive bacteria, Gram-negative bacteria, or fungi, but the function of the APL1 paralogs reveals that mosquito innate immunity possesses a more fine-grained capacity to distinguish between classes of closely related eukaryotic pathogens than has been previously recognized.


Subject(s)
Anopheles/immunology , Insect Proteins/immunology , Malaria/immunology , Plasmodium/pathogenicity , Trans-Activators/immunology , Analysis of Variance , Animals , Anopheles/genetics , Caenorhabditis elegans Proteins , Child , Child, Preschool , Female , Humans , Insect Proteins/genetics , Membrane Proteins , Models, Immunological , Signal Transduction/immunology , Statistics, Nonparametric
12.
Eukaryot Cell ; 5(4): 672-82, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16607015

ABSTRACT

In eukaryotes, the high-mobility-group (HMG) nuclear factors are highly conserved throughout evolution and are divided into three families, including HGMB, characterized by an HMG box domain. Some HMGB factors are DNA structure specific and preferentially interact with distorted DNA sequences, trigger DNA bending, and hence facilitate the binding of nucleoprotein complexes that in turn activate or repress transcription. In Plasmodium falciparum, two HMGB factors were predicted: PfHMGB1 and PfHMGB2. They are small proteins, under 100 amino acids long, encompassing a characteristic HMG box domain closely related to box B of metazoan factors, which comprises two HMG box domains, A and B, in tandem. Computational analyses supported the conclusion that the Plasmodium proteins were genuine architectural HMGB factors, and in vitro analyses performed with both recombinant proteins established that they were able to interact with distorted DNA structures and bend linear DNA with different affinities. These proteins were detected in both asexual- and gametocyte-stage cells in Western blotting experiments and mainly in the parasite nuclei. PfHMGB1 is preferentially expressed in asexual erythrocytic stages and PfHMGB2 in gametocytes, in good correlation with transcript levels of expression. Finally, immunofluorescence studies revealed differential subcellular localizations: both factors were observed in the nucleus of asexual- and sexual-stage cells, and PfHMGB2 was also detected in the cytoplasm of gametocytes. In conclusion, in light of differences in their levels of expression, subcellular localizations, and capacities for binding and bending DNA, these factors are likely to play nonredundant roles in transcriptional regulation of Plasmodium development in erythrocytes.


Subject(s)
HMGB Proteins/genetics , Plasmodium falciparum/genetics , Amino Acid Sequence , Animals , Computational Biology , DNA/metabolism , Erythrocytes/parasitology , HMGB Proteins/classification , HMGB Proteins/metabolism , Humans , Life Cycle Stages , Molecular Sequence Data , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Regulatory Elements, Transcriptional , Sequence Alignment
13.
Malar J ; 5: 23, 2006 Mar 28.
Article in English | MEDLINE | ID: mdl-16569221

ABSTRACT

BACKGROUND: The Plasmodium species that infect rodents, particularly Plasmodium berghei and Plasmodium yoelii, are useful to investigate host-parasite interactions. The mosquito species that act as vectors of human plasmodia in South East Asia, Africa and South America show different susceptibilities to infection by rodent Plasmodium species. P. berghei and P. yoelii infect both Anopheles gambiae and Anopheles stephensi, which are found mainly in Africa and Asia, respectively. However, it was reported that P. yoelii can infect the South American mosquito, Anopheles albimanus, while P. berghei cannot. METHODS: P. berghei lines that express the green fluorescent protein were used to screen for mosquitoes that are susceptible to infection by P. berghei. Live mosquitoes were examined and screened for the presence of a fluorescent signal in the abdomen. Infected mosquitoes were then examined by time-lapse microscopy to reveal the dynamic behaviour of sporozoites in haemolymph and extracted salivary glands. RESULTS: A single fluorescent oocyst can be detected in live mosquitoes and P. berghei can infect A. albimanus. As in other mosquitoes, P. berghei sporozoites can float through the haemolymph and invade A. albimanus salivary glands and they are infectious in mice after subcutaneous injection. CONCLUSION: Fluorescent Plasmodium parasites can be used to rapidly screen susceptible mosquitoes. These results open the way to develop a laboratory model in countries where importation of A. gambiae and A. stephensi is not allowed.


Subject(s)
Anopheles/parasitology , Green Fluorescent Proteins/metabolism , Insect Vectors/parasitology , Malaria/transmission , Plasmodium berghei/metabolism , Plasmodium yoelii/metabolism , Animals , Green Fluorescent Proteins/genetics , Hemolymph/parasitology , Mice , Plasmodium berghei/genetics , Plasmodium yoelii/genetics , Species Specificity
14.
Microb Ecol ; 50(4): 475-87, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16328650

ABSTRACT

We studied the persistence of Bacillus thuringiensis serovar israelensis (Bti) in a typical breeding site of the mosquito Ochlerotatus caspius in a particularly sensitive salt marsh ecosystem following two Bti-based larvicidal applications (Vectobac 12AS, 1.95 L/ha). The treated area was composed of four larval biotopes that differed in terms of the most representative plant species (Sarcocornia fruticosa, Bolboschoenus maritimus, Phragmites australis, and Juncus maritimus) and the physical and chemical characteristics of the soil. We sampled water, soil, and plants at various times before and after the applications (from spring to autumn, 2001) and quantified the spores of B. thuringiensis (Bt) and Bacillus species. The B. cereus group accounted for between 0% and 20% of all Bacillus spp. before application depending on the larval biotope. No Bti were found before application. The variation in the quantity of bacilli during the mosquito breeding season depended more on the larval biotope than on the season or the larvicidal application. More bacilli were found in soil (10(4)-10(6) spores/g) than on plant samples (10(2)-10(4) spores/g). The abundance in water (10(5) to 10(7) spores/L) appeared to be correlated to the water level of the breeding site. The number of Bti spores increased just after application, after declining; no spores were detected in soil or water 3 months after application. However, low numbers of Bti spores were present on foliage from three of the four studied plant strata. In conclusion, the larvicidal application has very little impact on Bacillus spp. flora after one breeding season (two applications).


Subject(s)
Bacillus thuringiensis/physiology , Ecosystem , Pest Control, Biological/standards , Animals , Bacillus thuringiensis/classification , Bacillus thuringiensis/isolation & purification , France , Larva , Ochlerotatus , Plants/microbiology , Soil Microbiology , Spores, Bacterial/classification , Spores, Bacterial/isolation & purification , Spores, Bacterial/physiology , Time Factors , Water Microbiology
15.
Med Sci Monit ; 10(11): PI119-25, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15507863

ABSTRACT

BACKGROUND: Broad-spectrum beta-lactam is the standard therapy for febrile neutropenia (FN) in cancer patients. The aim of our study was to evaluate the treatment of FN by a once-daily administration of ceftriaxone (CFX) alone. MATERIAL/METHODS: From Jan. 1, 1997 to Dec. 31, 2001 we prospectively analyzed 100 episodes of FN in 94 patients. Inclusion criteria were: fever > or = 38.5 degrees C, neutrophil count (NC) <500/microl, presumed short duration (<7 days), and no antimicrobial treatment within the preceding 72 h. Treatment consisted of 2 g daily intravenous CFX alone until NC>500. Success criteria were: afebrile within 48 hours under CFX alone and without secondary infection. RESULTS: Twenty-seven episodes occurred in patients with performance status (PS)>2. The median duration of neutropenia was 3.5 days (range 1-22). Etiology of fever was: 75 of unknown origin (FUO), 6 clinically defined (CDI), and 19 microbiologically documented (MDI). Median CFX treatment duration was 5 days. Successful response was obtained in 87% of cases; no deaths occurred. Treatment efficacy differed between FUO, CDI, and MDI with, respectively, 92.0, 83.3, and 68.4% success rates (p=0.042). Treatment failure was mostly observed in patients with PS > or = 2 (p=0.0001). Among the 13 failures, 4 resolved in less than 4 days with CFX alone and 9 required additional or modified antimicrobial treatment. CONCLUSIONS: Considering the marked practical advantages of CFX alone (well-tolerated treatment with minimum side effects, once-daily administration, low cost, and high response rates), this single-agent regimen appears to be a valuable option in treatment of FN in patients with solid tumors.


Subject(s)
Ceftriaxone/therapeutic use , Neoplasms/complications , Neutropenia/drug therapy , Adult , Aged , Aged, 80 and over , Ceftriaxone/administration & dosage , Drug Therapy, Combination/therapeutic use , Female , Humans , Infections/complications , Infections/drug therapy , Infections/microbiology , Injections, Intravenous , Male , Middle Aged , Neutropenia/etiology , Neutropenia/microbiology
16.
EMBO J ; 21(24): 6673-80, 2002 Dec 16.
Article in English | MEDLINE | ID: mdl-12485988

ABSTRACT

Deciphering molecular interactions between the malaria parasite and its mosquito vector is an emerging area of research that will be greatly facilitated by the recent sequencing of the genomes of Anopheles gambiae mosquito and of various Plasmodium species. So far, most such studies have focused on Plasmodium berghei, a parasite species that infects rodents and is more amenable to studies. Here, we analysed the expression pattern of nine An.gambiae genes involved in immune surveillance during development of the human malaria parasite P.falciparum in mosquitoes fed on parasite-containing blood from patients in Cameroon. We found that P.falciparum ingestion triggers a midgut-associated, as well as a systemic, response in the mosquito, with three genes, NOS, defensin and GNBP, being regulated by ingestion of gametocytes, the infectious stage of the parasite. Surprisingly, we found a different pattern of expression of these genes in the An.gambiae-P.berghei model. Therefore, differences in mosquito reaction against various Plasmodium species may exist, which stresses the need to validate the main conclusions suggested by the P.berghei-An.gambiae model in the P.falciparum-An.gambiae system.


Subject(s)
Anopheles/genetics , Anopheles/immunology , Insect Proteins , Plasmodium falciparum/pathogenicity , Acute-Phase Proteins/biosynthesis , Animals , Anopheles/parasitology , Blood Proteins/biosynthesis , Defensins/biosynthesis , Escherichia coli/metabolism , Nitric Oxide Synthase/biosynthesis , Plasmodium berghei/metabolism , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases/metabolism , Time Factors , Transcription, Genetic , Up-Regulation
17.
Mem. Inst. Oswaldo Cruz ; 92(2): 257-62, Mar.-Apr. 1997. ilus, tab
Article in English | LILACS | ID: lil-184980

ABSTRACT

Bacillus thuringiensis (Bt) subsp. medellin (Btmed) produces parasporal crystalline inclusions which are toxic to mosquito larvae. It has been shown that the inclusions of this bacterium contain mainly proteins of 94, 68 and 28-30 kDa. EcoRI partially digested total DNA of Btmed was cloned by using the Lambda Zap II cloning kit. Recombinant plaques were screened with a mouse policlonal antibody raised against the 94 kDa crystal protein of Btmed. One of the positive plaques was selected, and by in vitro excision, a recombinant pBluescript SK(-) was obtained. The gene encoding the 94 kDa toxin of Btmed DNA was cloned in a 4.4 kb DNA fragment. Btmed DNA was then subcloned as a EcoRI/EcoRI fragment into the shuttle vector pBU4 producing the recombinant plasmid pBTM3 and used to transform by electroporation Bt subsp. israelensis (Bti) crystal negative strain 4Q2-81. Toxicity to mosquito larvae was estimated by using first instar laboratory reared Aedes aegypti, and Culex quinquefasciatus larvae challenged with whole crystals. Toxicity results indicate that the purified inclusions from the recombinant Bti strain were toxic to all mosquito species tested although the toxicity was not as high as one produced by the crystal of the Btmed wild type strain. Poliacrylamide gel electrophoresis indicate that the inclusions produced by the recombinant strain Bti (pBTM3) were mainly composed of the 94 kDa protein of Btmed, as it was determined by Western blot.


Subject(s)
Animals , Bacillus thuringiensis/genetics , Bacterial Toxins/pharmacology , Larva/drug effects , Cloning, Molecular , Culicidae/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...