11/9/156 (Item 7 from file: 73)

 10769043     EMBASE No: 2000247623

 Intravascular and endobronchial DNA delivery to murine lung tissue using

 a novel, nonviral vector

 

 Kukowska‑Latallo J.F.; Raczka E.; Quintana A.; Chen C.; Rymaszewski M.;

 Baker J.R. Jr.

 Dr. J.R. Baker Jr., Department of Internal Medicine, 9220 MSRB III, Univ.

 of Michigan Medical School, Ann Arbor, MI 48109‑0648   United States

 Human Gene Therapy ( HUM. GENE THER. ) ( United States )   01 JUL 2000 ,

 11/10 (1385‑1395)

 CODEN: HGTHE   ISSN: 1043‑0342

 Document Type: Journal ; Article

 Language: ENGLISH   Summary Language: ENGLISH

 Number Of References: 59

 

 Gene transfer to the lung can be achieved via either the airway or the

 pulmonary vasculature. We evaluated gene transfer and expression by

 intravascular and endobronchial routes, using DNA complexed with G9 PAMAM

 dendrimer or naked plasmid DNA. Intravascular tail vein delivery of

 dendrimer‑complexed pCF1CAT plasmid resulted in high levels of transgene

 expression in the lung at 12 and 24 hr, followed by a second peak of

 expression 3 to 5 days after administration. After intravenous

 administration of the complexes, CAT expression was never observed in

 organs other than the lung. There were only minimal levels of CAT protein

 expressed in the lung after intravenous administration of naked plasmid

 DNA. Repeated intravascular doses of the dendrimer‑complexed plasmid,

 administered four times at 4‑day intervals, maintained expression at

 15‑25% of peak concentrations achieved after the initial dose.

 Endobronchial delivery of naked pCF1CAT plasmid produced significant

 amounts of CAT protein in the lung. Comparison of intratracheal and

 intranasal routes resulted in similar expression levels of CAT in the

 lung and trachea. However, in juxtaposition to vascular delivery,

 intranasal delivery of dendrimer‑complexed plasmid DNA gave lower levels

 of CAT expression than that observed with naked plasmid DNA. In situ

 localization of CAT enzymatic activity suggested that vascular

 administration seemed to achieve expression in the lung parenchyma,

 mainly within the alveoli, while endobronchial administration primarily

 targeted bronchial epithelium. Our results show that intravenously

 administered G9 dendrimer is an effective vector for pulmonary gene

 transfer and that transgene expression can be prolonged by repeated

 administration of dendrimer‑complexed DNA.

 

 DRUG DESCRIPTORS:

 dendrimer; transmembrane conductance regulator; alpha 1 antitrypsin;

 liposome

 MEDICAL DESCRIPTORS:

 * virus vector; *lung parenchyma; *gene transfer; *lung

 disease‑‑therapy‑‑th

 gene expression; protein expression; enzyme activity; enzyme linked

 immunosorbent assay; histochemistry; transgene; plasmid; gene therapy;

 nonhuman; mouse; animal experiment; animal cell; article

 CAS Registry Number: 9041‑92‑3 (alpha 1 antitrypsin)

 Drug Descriptors:

 022 Human Genetics

    EMBASE (Dialog« File 73): (c) 2001 Elsevier Science B.V. All rights

                                 reserved.

 

 

 

 

11/9/5 (Item 5 from file: 155)

 10533454   20360072

   Enhanced gene expression in mouse lung after PEI‑DNA aerosol delivery.

 

   Gautam A; Densmore CL; Xu B; Waldrep JC

   Department of Molecular Physiology and Biophysics, Baylor College of

 Medicine, Houston, Texas 77030, USA.

   Mol Ther ( UNITED STATES )   Jul 2000 ,   2 (1) p63‑70 ,  ISSN

 1525‑0016   Journal Code: DRT

   Languages: ENGLISH

  Document type: JOURNAL ARTICLE

  Journal Announcement: 0011

  Subfile:   INDEX MEDICUS

   Aerosol gene delivery to the pulmonary system has vast potential for

 many diseases, including cystic fibrosis and lung cancer. We recently

 reported that polyethyleneimine (PEI), a cationic polymer, holds promise

 as a gene delivery vector for transfection in lung by aerosol. To further

 optimize the gene expression in the lung by aerosol, we utilized 5% CO(2)

 in air for the nebulization of PEI‑DNA complexes. Five percent

 CO(2)‑in‑air gave a threefold higher gene expression compared to normal

 air using the chloramphenicol acetyl transferase (CAT) reporter gene

 delivered by Aerotech II nebulizer. The delivery of DNA by PEI was dose

 dependent with the highest expression obtained when 2 mg of DNA in 10 ml

 was nebulized at a PEI nitrogen:DNA phosphate (N:P) ratio of 10:1. The

 optimal N:P ratio for lung transfection was found to be between 10:1 and

 20:1 using the CAT and luciferase reporter genes. The time‑course studies

 showed the highest expression at 24 h after aerosol delivery and 40‑50%

 of peak level was detectable even after a week. Tissue distribution

 indicates the expression to be specific to the lung with no detectable

 expression in any other tissue examined. Histological and biochemical

 analysis of lungs revealed no evidence of acute inflammation.

  Tags: Animal; Female; Support, Non‑U.S. Gov't

   Descriptors: *Aerosols; *Gene Therapy‑‑Methods‑‑MT; *Genetic

 Vectors‑‑Administration and Dosage‑‑AD; *Lung‑‑Metabolism‑‑ME;

 *Polyethyleneimine ; Carbon Dioxide‑‑Metabolism‑‑ME; Chloramphenicol

 O‑Acetyltransferase ‑‑Metabolism‑‑ME; Dose‑Response Relationship, Drug;

 DNA‑‑Administration and Dosage‑‑AD; Enzyme‑Linked Immunosorbent Assay;

 Gene Expression; Luciferase ‑‑Metabolism‑‑ME; Lung‑‑Pathology‑‑PA; Mice;

 Mice, Inbred BALB C; Peroxidase‑‑Metabolism‑‑ME;

 Plasmids‑‑Metabolism‑‑ME; Time Factors; Tissue Distribution; Transgenes

  CAS Registry No.: 0 (Aerosols); 0 (Genetic Vectors); 0 (Plasmids);

 124‑38‑9 (Carbon Dioxide); 9002‑98‑6 (Polyethyleneimine); 9007‑49‑2 (DNA)

 

  Enzyme No.: EC 1.11.1.7 (Peroxidase); EC 1.13.12.‑ (Luciferase); EC

 2.3.1.28 (Chloramphenicol O‑Acetyltransferase)

MEDLINE(R) (Dialog« File 155): (c) format only 2000 Dialog Corporation. All

                              rights reserved.

 

   ‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑

 

 11/9/20 (Item 20 from file: 155)

 10407316   20262156

   Gene transfer to adult human lung tissue ex vivo.

 

   McBride S; Rannie D; Harrison DJ

   CRC Laboratories, Pathology Department, University of Edinburgh Medical

 School, Teviot Place, Edinburgh EH8 9AG, Scotland, UK.

   Gene therapy ( ENGLAND )   Apr 2000 ,   7 (8) p675‑8 ,  ISSN 0969‑7128

   Journal Code: CCE

   Languages: ENGLISH

  Document type: JOURNAL ARTICLE

  Journal Announcement: 0008

  Subfile:   INDEX MEDICUS

   The potential of gene therapy for treatment of lung disease remains

 unrealised. Early model systems often resulted in promising efficiency of

 gene transfer, only to prove irreproducible in the clinic. While problems

 such as induction of host immune responses and duration of expression

 also need to be addressed, it is now widely believed that alternative,

 relevant models which more accurately reflect gene transfer efficiencies

 in human lungs are urgently required. We report here on a human lung

 slice culture system to assess gene transfer to adult lung epithelium. A

 lacZ‑expressing adenovirus (AdCA35lacZ) was used as a reporter vector. A

 solution of AdCA35lacZ was instilled via bronchioles into resected lung

 tissue, a route analogous to clinical administration. Following a 1 h

 incubation, the tissue was inflated with a 0.4% agarose solution,

 instilled via the same bronchioles. Once solidified, 500 &mgr;m slices of

 the tissue were prepared and cultured for 4 days. beta‑Galactosidase

 staining revealed lacZ transgene expression in bronchiolar and alveolar

 cells of the lung slices throughout the 4 days in culture. This system,

 which can also be used to study other viral and liposome vectors, could

 prove to be a useful alternative model for assessing gene delivery to

 adult human lung epithelium.

  Tags: Animal; Human; Support, Non‑U.S. Gov't

   Descriptors: *Adenoviridae‑‑Genetics‑‑GE; *Gene Therapy‑‑Methods‑‑MT;

 *Gene Transfer; *Genetic Vectors‑‑Administration and Dosage‑‑AD; *Lung ;

 Adult; Epithelium; Gene Expression; Lac Operon; Mice; Tissue Culture

  CAS Registry No.: 0 (Genetic Vectors)

MEDLINE(R) (Dialog« File 155): (c) format only 2000 Dialog Corporation. All

                              rights reserved.

 

   ‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑

 

 11/9/21 (Item 21 from file: 155)

 10400993   20220616

   Cationic lipid structure and formulation considerations for optimal

 gene transfection of the lung.

 

   Marshall J; Nietupski JB; Lee ER; Siegel CS; Rafter PW; Rudginsky SA;

 Chang CD; Eastman SJ; Harris DJ; Scheule RK; Cheng SH

   Genzyme Corporation, Framingham, MA 01701‑9322, USA.

 john.marshall@genzyme.com

   Journal of drug targeting ( SWITZERLAND )   2000 ,   7 (6) p453‑69 ,

  ISSN 1061‑186X   Journal Code: B3S

   Languages: ENGLISH

  Document type: JOURNAL ARTICLE

  Journal Announcement: 0008

  Subfile:   INDEX MEDICUS

   Enhanced gene transduction to the lung using cationic lipids could be

 attained through optimization of the structure of the lipids and the

 formulation of the cationic lipid:plasmid DNA (pDNA) complexes. We have

 expanded on our earlier observation of the importance of the structural

 orientation of the cationic lipid headgroup. Through the synthesis of a

 number of matched pairs of cationic lipids differing only in the

 configuration of their headgroup, we confirmed that those harboring a

 T‑shape headgroup are more active than their linear counterparts, at

 least when tested in the lungs of BALB/c mice. Additionally, we

 demonstrated that not only are the structural considerations of these

 cationic lipids important, but also their protonation state, the free

 base being invariably more active than its salt counterpart. The salt

 forms of cationic lipids bound pDNA with greater avidity, which may have

 affected their subsequent intracellular dissolution and transit of the

 pDNA to the nucleus. Inclusion of a number of frequently used solutes in

 the vehicle severely inhibited the gene transfection activity of the

 cationic lipids. The selection of neutral co‑lipids was also an important

 factor for overall transfection activity of the formulation, with

 significant gains in transfection activity realized when

 diphytanoylphosphatidylethanolamine or

 dilinoleoylphosphatidylethanolamine were used in lieu of

 dioleoylphosphatidylethanolamine. Finally, we showed that a

 transacylation reaction could occur between the cationic lipid and

 neutral co‑lipid which reduced the transfection activity of the

 complexes. It is the hope that as our understanding of the many factors

 that influence the activity of these cationic lipid:pDNA complexes

 improves, formulations with much greater potency can be realized for use

 in the treatment of pulmonary diseases.

  Tags: Animal; Female

   Descriptors: *Gene Therapy; *Lipids‑‑Administration and Dosage‑‑AD;

 *Lung‑‑Metabolism ‑‑ME; *Transfection ; Drug Stability;

 Excipients‑‑Pharmacology‑‑PD; Lipids‑‑Chemistry‑‑CH; Mice; Mice, Inbred

 BALB C

  CAS Registry No.: 0 (Excipients); 0 (Lipids)

MEDLINE(R) (Dialog« File 155): (c) format only 2000 Dialog Corporation. All

                              rights reserved.

 

   ‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑

 

 11/9/41 (Item 41 from file: 155)

 09954356   99316336

   Repeated administration of adenoviral vectors in lungs of human CD4

 transgenic mice treated with a nondepleting CD4 antibody.

 

   Chirmule N; Truneh A; Haecker SE; Tazelaar J; Gao Gp; Raper SE; Hughes

 JV; Wilson JM

   Institute for Human Gene Therapy, Department of Medicine, University of

 Pennsylvania, Wistar Institute, Philadelphia, PA 19104, USA.

   Journal of immunology ( UNITED STATES )   Jul 1 1999 ,   163 (1)

 p448‑55 ,  ISSN 0022‑1767   Journal Code: IFB

     Contract/Grant No.: P30DK47757‑06, DK, NIDDK; P50DK49136‑05, DK,

 NIDDK

   Languages: ENGLISH

  Document type: JOURNAL ARTICLE

  Journal Announcement: 9909

  Subfile:   AIM; INDEX MEDICUS

   The central role of CD4+ T cells in regulation of adenovirus

 vector‑mediated immune responses has been documented previously in murine

 models. We analyzed the effects of a nondepleting mAb to human CD4 (CD4

 mAb; Clenoliximab) on immune functions following intratracheal

 administration of adenoviral vectors in murine CD4‑deficient mice

 (muCD4KO) expressing a human CD4 transgene (HuCD4 mice). Treatment of

 HuCD4 mice with Clenoliximab inhibited both cell‑mediated and humoral

 immune responses to adenoviral Ags. Chronic treatment of HuCD4 mice with

 Clenoliximab permitted successful readministration of adenoviral vectors

 at least four times. The ability to readminister these vectors is

 associated with marked suppression of neutralizing Ab responses to viral

 capsid proteins. Clenoliximab also inhibited CTL and prolonged expression

 of the transgene. T or B cell responses to adenovirus did not emerge

 after the effects of a short course of Clenoliximab diminished. These

 data illustrate the potential utility of a nondepleting CD4 Ab in

 facilitating gene therapy using adenoviral vectors.

  Tags: Animal; Female; Human; Male; Support, Non‑U.S. Gov't; Support,

 U.S. Gov't, P.H.S.

   Descriptors: *Adenoviridae‑‑Genetics‑‑GE; *Antibodies,

 Monoclonal‑‑Administration and Dosage‑‑AD; *Antigens, CD4‑‑Genetics‑‑GE;

 *Antigens, CD4‑‑Immunology‑‑IM; *Genetic Vectors‑‑Administration and

 Dosage‑‑AD; *Lung‑‑Immunology‑‑IM ; Adenoviridae‑‑Immunology‑‑IM;

 Antibodies, Monoclonal‑‑Therapeutic Use‑‑TU; Cystic

 Fibrosis‑‑Genetics‑‑GE; Cystic Fibrosis‑‑Immunology‑‑IM; Cystic

 Fibrosis‑‑Therapy‑‑TH; CD4‑Positive T‑Lymphocytes‑‑Immunology‑‑IM;

 CD8‑Positive T‑Lymphocytes‑‑Immunology‑‑IM; Gene Therapy‑‑Methods‑‑MT;

 Genetic Vectors‑‑Immunology‑‑IM; Injections, Intraperitoneal; Intubation,

 Intratracheal; Lung‑‑Metabolism‑‑ME; Lymphocyte Depletion; Lymphocyte

 Transformation‑‑Genetics‑‑GE; Mice; Mice, Inbred C57BL; Mice, Inbred DBA;

 Mice, Knockout; Mice, Transgenic; Th1 Cells‑‑Metabolism‑‑ME; Th2 Cells

 ‑‑Metabolism‑‑ME

  CAS Registry No.: 0 (Antibodies, Monoclonal); 0 (Antigens, CD4); 0

 (Genetic Vectors)

MEDLINE(R) (Dialog« File 155): (c) format only 2000 Dialog Corporation. All

                              rights reserved.

 

 

11/9/110 (Item 110 from file: 155)

 08826974   96432153

   Systematic analysis of repeated gene delivery into animal lungs with a

 recombinant adenovirus vector.

 

   Dong JY; Wang D; Van Ginkel FW; Pascual DW; Frizzell RA

   Department of Laboratory Medicine, University of California, San

 Francisco 94143‑0724, USA.

   Human gene therapy ( UNITED STATES )   Feb 10 1996 ,   7 (3) p319‑31 ,

  ISSN 1043‑0342   Journal Code: A12

     Contract/Grant No.: DK/HL46177, DK, NIDDK; DK38518, DK, NIDDK;

 CA54430, CA, NCI

   Languages: ENGLISH

  Document type: JOURNAL ARTICLE

  Journal Announcement: 9702

  Subfile:   INDEX MEDICUS

   Adenovirus‑based vectors are promising candidates for genetic therapy

 of cystic fibrosis (CF). Because adenoviruses naturally infect airway

 cells, they grow to very high titers, and the transgenes carried by the

 adenoviruses are expressed at high levels. In addition, adenoviruses are

 relatively safe because the disease caused by the wild‑type virus is

 self‑limiting. One disadvantage of adenovirual vectors is that the

 transgene expression would be transient because adenoviruses do not

 integrate their DNA into the genome of the host cells. Adenoviral gene

 delivery into the lungs is also complicated by the anatomy of the airways

 and the defense mechanisms of the recipient. To assess the feasibility of

 adenovirus‑mediated gene therapy for CF, a recombinant adenovirus

 carrying a lacZ gene was delivered into animal lungs to study the

 efficiency and cellular distribution of gene transfer, the duration of

 gene expression, the possible histopathology of the lungs after gene

 transfer, and the efficacy of repeated administrations of the viral

 agent. The results of these studies demonstrate that (i) efficient gene

 transfer into animal lungs can be achieved; (ii) a near‑homogenous

 delivery of the vectors can be achieved by airway instillation, although

 the pattern of transduction varies among individual animals; (iii)

 pathological effects are generally mild in CD1 mice; (iv) gene expression

 is transient; (v) repetitive gene transfer is achievable, but becomes

 progressively less efficient, and (vi) immune responses are induced

 against both the viral and transgene products.

  Tags: Animal; Female; Male; Support, Non‑U.S. Gov't; Support, U.S.

 Gov't, P.H.S.

   Descriptors: *beta‑Galactosidase‑‑Genetics‑‑GE;

 *Adenoviridae‑‑Genetics‑‑GE; *Defective Viruses‑‑Genetics‑‑GE; *DNA,

 Recombinant‑‑Administration and Dosage‑‑AD; *Gene Therapy‑‑Methods‑‑MT;

 *Genetic Vectors‑‑Genetics‑‑GE; *Lung; *Recombinant Fusion

 Proteins‑‑Biosynthesis‑‑BI ; beta‑Galactosidase‑‑Biosynthesis‑‑BI;

 Adenoviridae‑‑Pathogenicity‑‑PY; Administration, Intranasal; Defective

 Viruses‑‑Pathogenicity‑‑PY; DNA, Recombinant‑‑Genetics‑‑GE;

 Epithelium‑‑Metabolism‑‑ME; Feasibility Studies; Gene Expression

 Regulation, Viral; Genes, Reporter; Genetic Vectors ‑‑Toxicity‑‑TO;

 Instillation, Drug; Lung‑‑Injuries‑‑IN; Lung‑‑Metabolism ‑‑ME;

 Lung‑‑Pathology‑‑PA; Mice; Pneumonia, Viral‑‑Etiology‑‑ET; Time Factors;

 Tissue Distribution; Trachea

  CAS Registry No.: 0 (DNA, Recombinant); 0 (Genetic Vectors); 0

 (Recombinant Fusion Proteins)

  Enzyme No.: EC 3.2.1.23 (beta‑Galactosidase)

MEDLINE(R) (Dialog« File 155): (c) format only 2000 Dialog Corporation. All

                              rights reserved.

 

‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑

 

 11/9/46 (Item 46 from file: 155)

 09916442   99261922

   Effect of immune response on gene transfer to the lung via systemic

 administration of cationic lipidic vectors.

 

   Li S; Wu SP; Whitmore M; Loeffert EJ; Wang L; Watkins SC; Pitt BR;

 Huang L

   Department of Pharmacology, University of Pittsburgh School of

 Medicine, Pittsburgh, Pennsylvania 15261, USA.

   American journal of physiology ( UNITED STATES )   May 1999 ,   276 (5

 Pt 1) pL796‑804 ,  ISSN 0002‑9513   Journal Code: 3U8

     Contract/Grant No.: CA‑59327, CA, NCI; CA‑64654, CA, NCI; CA‑71731,

 CA, NCI; +

   Languages: ENGLISH

  Document type: JOURNAL ARTICLE

  Journal Announcement: 9908

  Subfile:   INDEX MEDICUS

   Cationic lipid‑mediated intravenous gene delivery shows promise in

 treating pulmonary diseases including lung tumor metastases, pulmonary

 hypertension, and acute respiratory distress syndrome. Nevertheless,

 clinical applications of cationic lipidic vectors via intravenous

 administration are limited by their transient gene expression. In

 addition, repeated dosing is not effective at frequent intervals. In an

 effort to elucidate the mechanism of gene inactivation, we report in this

 study that cationic lipid‑protamine‑DNA (LPD) complexes, but not each

 component alone, can induce a high level of cytokine production,

 including interferon‑gamma and tumor necrosis factor‑alpha. Furthermore,

 we demonstrate that LPD administration triggers apoptosis in the lung, a

 phenomenon that may be mediated in part by the two cytokines. Treatment

 of mice with antibodies against the two cytokines prolongs the duration

 of gene expression and also improves lung transfection on a second

 administration of LPD. Although the mechanism underlying LPD‑induced

 cytokine production is unclear, methylation of the DNA significantly

 decreased the level of both interferon‑gamma and tumor necrosis

 factor‑alpha, suggesting that unmethylated CpG sequences in plasmid DNA

 play an important role. These data suggest that decreasing the

 CpG‑mediated immune response while not affecting gene expression may be a

 useful therapeutic strategy to improve cationic lipid‑mediated

 intravenous gene delivery to the lung.

  Tags: Animal; Female; Support, Non‑U.S. Gov't; Support, U.S. Gov't,

 P.H.S.

   Descriptors: *Cytokines‑‑Immunology‑‑IM; *DNA‑‑Immunology‑‑IM; *Gene

 Transfer; *Genetic Vectors; *Lipids; *Lung‑‑Metabolism‑‑ME ;

 Antibodies‑‑Pharmacology‑‑PD; Apoptosis; Cations; DNA‑‑Administration and

 Dosage‑‑AD; DNA Methylation; Gene Expression; Immunization, Passive;

 Injections, Intravenous; Interferon Type II‑‑Immunology‑‑IM; Lung

 Diseases ‑‑Therapy‑‑TH; Mice; Plasmids‑‑Genetics‑‑GE; Tumor Necrosis

 Factor ‑‑Immunology‑‑IM

  CAS Registry No.: 0 (Antibodies); 0 (Cations); 0 (Cytokines); 0 (Genetic

 Vectors); 0 (Lipids); 0 (Plasmids); 0 (Tumor Necrosis Factor); 82115‑62‑6

 (Interferon Type II); 9007‑49‑2 (DNA)

MEDLINE(R) (Dialog« File 155): (c) format only 2000 Dialog Corporation. All

                              rights reserved.

 

 

11/9/153 (Item 4 from file: 73)

 10905960     EMBASE No: 2000393474

 Biodistribution and transgene expression with nonviral cationic

 vector/DNA complexes in the lungs

 

 Bragonzi A.; Dina G.; Villa A.; Calori G.; Biffi A.; Bordignon C.; Assael

 B.M.; Conese M.

 M. Conese, Inst. for Experimen. Cystic Fibrosis, San Raffaele Scientific

 Institute, Via Olgettina 58, Milano 20132   Italy

 Gene Therapy ( GENE THER. ) ( United Kingdom )   2000 , 7/20 (1753‑1760)

 CODEN: GETHE   ISSN: 0969‑7128

 Document Type: Journal ; Article

 Language: ENGLISH   Summary Language: ENGLISH

 Number Of References: 36

 

 Biodistribution of nonviral cationic vector/DNA complexes was studied

 after systemic or intratracheal administration to the lungs and

 correlated with transgene expression. Intravenous injection in C57BI/6

 mice gave maximal and significant luciferase expression in the lungs with

 the cationic polymer PEI 22K/DNA complexes at the highest ratios of

 positive/negative charges versus DNA alone. While DOTAP/DNA complexes

 with high charge ratio determined lower but still significant luciferase

 activity versus uncomplexed DNA, GL‑67A and PEI 25K mediated negligible

 luciferase expression. Labelled PEI 22K and DOTAP complexes were evenly

 distributed in the alveolar region, where GFP expression was revealed,

 while PEI 25K and GL‑67A complexes were not detected, suggesting a

 different interaction of these complexes with the plasma membrane of

 endothelial cells. Following an intratracheal injection, the highest and

 significant levels of transfection were obtained with slightly positive

 PEI complexes as compared with DNA alone, whereas cationic lipid‑based

 vectors. DOTAP and GL‑67A, gave not significant luciferase activity. Both

 types of polyplexes gave similar levels of lung luciferase expression by

 targeting different airway cell populations. PEI 25K complexes determined

 high levels of GFP in the bronchial cells, confirming confocal data on

 fluorescent complexes internalization. PEI 22K complexes gave mainly high

 GFP signal in the distal tract of the bronchial tree, where tagged

 complexes were recovered. Fluorescent lipid complexes were found in

 aggregates in the lumen of bronchi totally (DOTAP) or partially (GL‑67A)

 co‑localizing with surfactant protein A. Results indicated that cationic

 polymers could overcome the surfactant barrier which inhibited airway

 cell transfection mediated by cationic lipids.

 

 Brand Name/Manufacturer Name: Exgen 500/Euromedex/France

 Manufacturer Names: Euromedex/France; Sigma Aldrich/United States;

 Hoffmann La Roche/Germany; Genzyme/United States

 DRUG DESCRIPTORS:

 * DNA‑‑intrathecal drug administration‑‑tl; *DNA‑‑intravenous drug

 administration‑‑iv; *polyethyleneimine‑‑intrathecal drug

 administration‑‑tl ; *polyethyleneimine‑‑intravenous drug

 administration‑‑iv

 luciferase‑‑endogenous compound‑‑ec; cation‑‑intrathecal drug

 administration‑‑tl; cation‑‑intravenous drug administration‑‑iv;

 liposome; macrogol; green fluorescent protein; surfactant protein

 A‑‑endogenous compound‑‑ec

 MEDICAL DESCRIPTORS:

 * lung; *transgene

 complex formation; gene expression; mouse strain; protein expression;

 enzyme activity; tissue distribution; lung alveolus; cell membrane;

 endothelium cell; genetic transfection; expression vector; gene

 targeting; cell population; bronchus; gene transfer; human; nonhuman;

 mouse; animal experiment; animal model; controlled study; human cell;

 animal tissue; article; priority journal

 Drug Terms (Uncontrolled): Exgen 500

 CAS Registry Number: 9007‑49‑2 (DNA); 74913‑72‑7 (polyethyleneimine);

 61970‑00‑1, 9014‑00‑0 ( luciferase); 25322‑68‑3 (macrogol)

 Drug Descriptors:

 015 Chest Diseases, Thoracic Surgery and Tuberculosis

  022 Human Genetics

  037 Drug Literature Index

    EMBASE (Dialog« File 73): (c) 2001 Elsevier Science B.V. All rights

                                 reserved

 

‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑

 

 11/9/47 (Item 47 from file: 155)

 09910534   99041640

   Direct gene transfer to the respiratory tract of mice with pure plasmid

 and lipid‑formulated DNA.

 

   McCluskie MJ; Chu Y; Xia JL; Jessee J; Gebyehu G; Davis HL

   Loeb Research Institute, Ottawa, Canada.

   Antisense & nucleic acid drug development ( UNITED STATES )   Oct 1998

 ,   8 (5) p401‑14 ,  ISSN 1087‑2906   Journal Code: CJY

   Languages: ENGLISH

  Document type: JOURNAL ARTICLE

  Journal Announcement: 9908

  Subfile:   INDEX MEDICUS

   Direct gene transfer into the respiratory system could be carried out

 for either therapeutic or immunization purposes. Here we demonstrate that

 cells in the lung can take up and express plasmid DNA encoding a

 luciferase reporter gene whether it is administered in naked form or

 formulated with cationic liposomes. Depending on the lipid used, the

 transfection efficiency with liposome‑formulated DNA may be higher, the

 same as, or less than that with pure plasmid DNA.

 Tetramethyltetraalkylspermine analogs with alkyl groups of 16 or 18

 carbons and DMRIE/cholesterol formulations proved particularly effective.

 Similar results for reporter gene expression in the lung were obtained

 whether the DNA (naked or lipid formulated) was administered by indirect,

 noninvasive intranasal delivery (inhaled or instilled) or by invasive,

 direct intratracheal delivery (injected or via a cannula). Reporter gene

 expression peaks around 4 days, then falls off dramatically by 9 days.

 The dose‑response is linear, at least up to 100 microg plasmid DNA,

 suggesting better transfection efficiencies might be realized if there

 was not a volume limitation. For a given dose of DNA, the best results

 are obtained when the DNA is mixed with the minimum amount of lipid that

 can complex it completely. These results are discussed in the context of

 direct gene transfer for either gene therapy or delivery of a mucosal DNA

 vaccine.

  Tags: Animal; Support, Non‑U.S. Gov't

   Descriptors: *DNA‑‑Metabolism‑‑ME; *Gene Transfer;

 *Liposomes‑‑Metabolism‑‑ME; *Plasmids ‑‑Genetics‑‑GE; *Respiratory

 System‑‑Metabolism‑‑ME ; beta‑Galactosidase‑‑Genetics‑‑GE;

 beta‑Galactosidase‑‑Immunology‑‑IM; beta‑Galactosidase‑‑Metabolism‑‑ME;

 Administration, Inhalation; Antibody Formation‑‑Immunology‑‑IM;

 Catheterization, Peripheral; Dose‑Response Relationship, Immunologic;

 DNA‑‑Administration and Dosage‑‑AD; DNA ‑‑Chemistry‑‑CH;

 DNA‑‑Genetics‑‑GE; Gene Expression‑‑Genetics‑‑GE; Genes,

 Reporter‑‑Genetics‑‑GE; Injections; Interferon Type II‑‑Genetics‑‑GE; Lac

 Operon‑‑Genetics‑‑GE; Liposomes‑‑Chemistry‑‑CH;

 Liposomes‑‑Immunology‑‑IM; Luciferase‑‑Genetics‑‑GE;

 Luciferase‑‑Metabolism‑‑ME; Lung‑‑Immunology‑‑IM ; Lung‑‑Metabolism‑‑ME;

 Mice; Mice, Inbred BALB C; Mice, Knockout; Plasmids‑‑Administration and

 Dosage‑‑AD; Plasmids‑‑Immunology‑‑IM; Respiratory System‑‑Immunology‑‑IM

  CAS Registry No.: 0 (Plasmids); 82115‑62‑6 (Interferon Type II);

 9007‑49‑2 (DNA)

  Enzyme No.: EC 1.13.12.‑ (Luciferase); EC 3.2.1.23 (beta‑Galactosidase)

MEDLINE(R) (Dialog« File 155): (c) format only 2000 Dialog Corporation. All

                              rights reserved.

 

   ‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑

 

 11/9/51 (Item 51 from file: 155)

 09842414   99145145

   Contribution of plasmid DNA to inflammation in the lung after

 administration of cationic lipid:pDNA complexes.

 

   Yew NS; Wang KX; Przybylska M; Bagley RG; Stedman M; Marshall J;

 Scheule RK ; Cheng SH

   Genzyme Corporation, Framingham, MA 01701‑9322, USA.

   Human gene therapy ( UNITED STATES )   Jan 20 1999 ,   10 (2) p223‑34 ,

  ISSN 1043‑0342   Journal Code: A12

   Languages: ENGLISH

  Document type: JOURNAL ARTICLE

  Journal Announcement: 9906

  Subfile:   INDEX MEDICUS

   Cationic lipid‑mediated gene transfer to the mouse lung induces a

 dose‑dependent inflammatory response that is characterized by an influx

 of leukocytes and elevated levels of the cytokines interleukin 6 (IL‑6),

 tumor necrosis factor alpha (TNF‑alpha), and interferon gamma

 (IFN‑gamma). We have examined the contribution of plasmid DNA (pDNA) to

 this observed toxicity, specifically the role of unmethylated CpG

 dinucleotides, which have been previously shown to be immunostimulatory.

 We report here that complexes of cationic lipid GL‑67 and unmethylated

 pDNA (pCF1‑CAT) instilled into the lungs of BALB/c mice induced highly

 elevated levels of the cytokines TNF‑alpha, IFN‑gamma, IL‑6, and IL‑12 in

 the bronchoalveolar lavage fluids (BALF). In contrast, BALF of animals

 administered either GL‑67 alone or GL‑67 complexed with SssI‑methylated

 pDNA contained low levels of these cytokines. Similar results were

 observed using a plasmid (pCF1‑null) that does not express a transgene,

 demonstrating that expression of chloramphenicol acetyltransferase (CAT)

 was not responsible for the observed inflammation. The response observed

 was dose dependent, with animals receiving increasingly higher amounts of

 unmethylated pDNA exhibiting progressively higher levels of the

 cytokines. Concomitant with this increase in cytokine levels were also

 elevated numbers of neutrophils in the BALF, suggesting a possible cause‑

 and‑effect relationship between neutrophil influx and generation of

 cytokines. Consistent with this proposal is the observation that

 reduction of neutrophils in the lung by administration of antibodies

 against Mac‑1alpha and LFA‑1 also diminished cytokine levels. This

 reduction in cytokine levels in the BALF was accompanied by an increase

 in transgene expression. In an attempt to abate the inflammatory

 response, sequences in the pDNA encoding the motif RRCGYY, shown to be

 most immunostimulatory, were selectively mutagenized. However,

 instillation of a plasmid in which 14 of the 17 CpG sites were altered

 into BALF/c mice did not reduce the levels of cytokines in the BALF

 compared with the unmodified vector. This suggests that other

 unmethylated motifs, in addition to RRCGYY, may also contribute to the

 inflammatory response. Together, these findings indicate that

 unmethylated CpG residues in pDNA are a major contributor to the

 induction of specific proinflammatory cytokines associated with

 instillation of cationic lipid:pDNA complexes into the lung. Strategies

 to abate this response are warranted to improve the efficacy of this

 nonviral gene delivery vector system for the treatment of chronic

 diseases.

  Tags: Animal

   Descriptors: *DNA‑‑Administration and Dosage‑‑AD; *Plasmids;

 *Pneumonia‑‑Genetics‑‑GE ; Bronchoalveolar Lavage Fluid; Cations; CpG

 Islands; DNA‑‑Metabolism‑‑ME; DNA Methylation; Interferon Type

 II‑‑Metabolism‑‑ME; Interleukin‑6 ‑‑Metabolism‑‑ME; Lung‑‑Metabolism‑‑ME;

 Mice; Mice, Inbred BALB C; Neutrophils‑‑Cytology‑‑CY;

 Pneumonia‑‑Metabolism‑‑ME; Tumor Necrosis Factor ‑‑Metabolism‑‑ME

  CAS Registry No.: 0 (Cations); 0 (Interleukin‑6); 0 (Plasmids); 0 (Tumor

 Necrosis Factor); 82115‑62‑6 (Interferon Type II); 9007‑49‑2 (DNA)

MEDLINE(R) (Dialog« File 155): (c) format only 2000 Dialog Corporation. All

                              rights reserved.

 

   ‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑‑