28(naveedjmpr-11-238

Upload: makram-tatri

Post on 03-Jun-2018

230 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/12/2019 28(naveedJMPR-11-238

    1/6

    Fly1Journal of Medicinal Plants Research Vol. 5(10), pp. 1812-1817, 18 May, 2011Available online at http://www.academicjournals.org/JMPRISSN 1996-0875 2011 Academic Journals

    Review

    Gene therapy: A review article

    Naveed Akhtar1, M. Akram2*, H. M. Asif1, Khan Usmanghani2, S. M. Ali Shah1,Saeed Ahmad Rao1, M. Uzair3, Ghazala Shaheen1and Khalil Ahmad1

    1Faculty of Pharmacy and Alternative Medicine, the Islamia University of Bahawalpur, Pakistan.2Faculty of Eastern Medicine, Hamdard University Karachi, Pakistan.

    3Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan.

    Accepted 14 March, 2011

    The introduction of nucleic acids into cells has as a purpose of medical condition or disease. Currently,gene therapy studies a broad range of potential therapeutic interventions, including the body's immunereaction to tumors, new blood vessels in the heart to alleviate heart attacks and to stop HIV-replicationin patients with AIDS (Coleman et al., 2003). There is also renewed emphasis on the gene therapy ofgenetic diseases, such as hemophilia A and B, and cystic fibrosis. Human gene therapyexperimentation raises many issues. In this review article, background of gene therapy, introduction,genetic diseases, gene function, germ line gene therapy, hurdles in gene therapy, methods for genetherapy, ex vivo, in vitroand in vivo-gene therapy, risks associated with gene therapy, have been given.

    Key words: Gene therapy, vectors, genetic diseases, methods for gene therapy.

    INTRODUCTION

    Advances in the molecular biology have been made earlyin the 1980. It has been already studied that humangenes can be sequenced and cloned. Scientists searchnew methods for easily producing of proteins, such asinsulin in diabetic patients. Modified bacteria, introducedin the body, can be harvested and injected in people, whocannot produce it naturally. Scientists try to introducegenes straight into human cells, focusing on diseases,caused by single-gene defects, such as cystic fibrosis,hemophilia, muscular dystrophy and sickle-cell anemia(Kay et al., 2000). Gene therapy for hemophilia B andother hereditary plasma protein deficiencies have shown

    great promise in pre-clinical and early clinical trials(Mannucci et al., 2001).Gene therapy can be broadly defined as a transfer of

    genetic material to cure a disease or at least to improvethe clinical status of a patient. One of the basic conceptsof gene therapy is to transform viruses into geneticshuttles, which would deliver the gene of interest into the

    *Corresponding author. E-mail: [email protected]: 92-021-6440083. Fax: 92-021-6440079.

    target cells. Based on the nature of the viral genomethese gene therapy vectors could be divided into RNAand/or DNA viral vectors. The majority of RNA virus-based vectors have been derived from simpleretroviruses like murine leukemia virus. A majoshortcoming of these vectors is that they are not able totransduce non-dividing cells. This problem may beovercome by use of novel retroviral vectors, derived fromlentiviruses, such as human immunodeficiency virus(HIV) (Ehmann et al., 1994). The most commonly usedDNA virus vectors are based on adenoviruses andadeno-associated viruses (AAVs). Although, the available

    vector systems are able to deliver genes in vivointo cellsthe ideal delivery vehicle has not been found. Thus, thepresent viral vectors should be used only with greacaution in human beings and further progress in vectordevelopment is necessary. Gene transfer technologiesare promising tools to manipulate donor T-cell immunityto enforce graft-versus-tumor/graft-versus-infection, whileprevention or control of graft versus host disease. For thispurpose, several cell and gene transfer approaches havebeen investigated at the pre-clinical level andimplemented in clinical trials (Mastaglio, 2010). Thenuclear envelope represents a key barrier to successful

  • 8/12/2019 28(naveedJMPR-11-238

    2/6

    non-viral transfection and gene therapy both in vitroandin vivo. Although the main purpose of the nuclearenvelope is to partite the cell to maintain cytoplasmiccomponents in the cytoplasm and nuclear components,the most notably genomic DNA in the nucleus, thisfunction poses a problem for transfections, in which

    exogenous DNA is delivered into the cytoplasm. Afterdelivery to the cytoplasm, nucleic acids rapidly becomemore complex, with cellular proteins that mediateinteractions with the cell machinery for their traffick. Thus,these proteins are that, in essence, which control thenuclear import of DNA, and we must also understandtheir activities in cells (Lam, 2010). Gene therapy forneurological, and in particular, neurodegenerative,diseases, is now a reality. A number of early phaseclinical trials have been completed and several arecurrently in progress. In view of this, it is criticallyimportant to be evaluated the immunological risk,associated with neurological gene therapy, which hasclear implications for trial safety and efficacy. Moreover, itis imperative in particular to identify factors, indicatingpotential high risk (McMenamin, 2010). Viral vectors arepotent gene-delivery platforms, used for the treatment ofgenetic and acquired diseases. However, just as viruseshave evolved to infect cells efficiently, the immunesystem has evolved to fight off what it perceives asinvading pathogens. Therefore, innate immunity andantigen-specific adaptive immune responses againstvector-derived antigens reduce the efficacy and stabilityof in vivo-gene transfer. In addition, a number of vectorsare derived from parent viruses that humans encounterthrough natural infection, resulting in pre-existingantibodies and possibly in memory responses against

    vector antigens. Similarly, antibody and T-cell responsesmight be directed against therapeutic gene products thatoften differ of the endogenous non-functional or absentprotein that is being replaced. As details andmechanisms of such immune reactions are uncovered,novel strategies are being developed, and vectors arebeing specifically engineered to avoid, suppress and/ormanipulate the response, ideally resulting in sustainedexpression and immune tolerance to the transgeneproduct (Nayak, 2010).

    Haematopoietic stem cell transplantation (HSCT) isnow widely used for treatment of primaryimmunodeficiencies (PID). For patients with specific

    disorders (severe combined immunodeficiency (SCID)-X1, adenosine deaminase deficiency (ADA)SCID, X-chronic granulomatous disease (CGD) and WiskottAldrich Syndrome (WAS), who lack a suitable humanleukocyte antigen- (HLA)-matched donor, gene therapyhas offered an important alternative treatment option(Qasim, 2009). Artificial chromosomes (ACs) are highlypromising vectors for use in gene therapy applications(Isman et al., 2008). They are able to maintainexpression of genomic-sized exogenous transgeneswithin target cells, without integrating into the host

    Akhtar et al. 1813

    genome. Although, these vectors have huge potentiaand benefits, in comparison with normal expressionconstructs, they are highly complex, technicallychallenging to construct and difficult to deliver to targetcells (Macnab, 2009). In the last two decadesremarkable advances have been made in the

    development of technologies used to engineer newaptamers and ribozymes. This has encouraged interesamong researchers, who seek to create new types ogene-control systems that could be made to respondspecifically to small-molecule signals. Validation of thefact that RNA-molecules can exhibit the characteristicsneeded to serve as precision genetic switches, has comefrom the discovery of numerous classes of natural ligand-sensing RNAs, called ribo-switches. Although a greadeal of progress has been made toward engineeringuseful designer ribo-switches, considerable advances areneeded before the performance characteristics of theseRNAs match those of protein systems that have been co-opted to regulate gene expression (Breaker, 2009)Pulmonary gene therapy cures diseases such as cysticfibrosis, 1-antitrypsin deficiency, lung cancer andpulmonary hypertension. Efficient expression of deliveredgenes in target cell types is essential for the achievementof this goal (Sinn, 2009).

    GENETIC DISEASES

    Cystic fibrosis, blood disorders, muscular dystrophy anddiabetes.

    Understanding gene function

    From the estimated 30 to 50,000 genes, we know thefunction of a very few. Attempting gene therapy howevery one of them works could address only some of thegenes, implicated in particular diseases. Likewise, genesmay have more than one function.

    Germ line gene therapy

    This technique involves the genetic modification of germcells. Such therapy would change the genetic make up of

    the egg or sperm of an individual and would be carried onto future generations. This would offer the possibility oremoving an inherited disorder from a family line forever.

    Hurdles in gene therapy

    The therapeutic genes are inserted into the body throughspecific constructs, called vectors, which delivetherapeutic genes to the patients cells. The moscommon vectors are viruses. Scientists try to manipulate

  • 8/12/2019 28(naveedJMPR-11-238

    3/6

    1814 J. Med. Plant. Res.

    the viral genome to remove the disease-causing genesand introduce therapeutic genes. The introduction ofviruses in the body might cause side effects like toxicity,immune and inflammatory responses, as well as genecontrol and targeting issues.

    METHODS FOR GENE THERAPY OF CANCER

    VirusesNaked DNA (vector-free)LiposomesProtein-DNA complexesGene gunCalcium phosphate precipitationElectroporationIntracellular microinjection

    Administration

    Ex vivo

    Cells are removed, genetically modified and transplantedback into a patient.

    In vivo

    Direct transfer of genetic material into patient.

    Choices of vectors

    Viral vectors:RetrovirusAdenovirusAdeno-associated virusHerpes Simplex Virus

    Non-viral vectors

    LiposomeDNApolymer conjugatesNaked DNA

    Gene therapy step by step

    Bone marrow from the patient is removed and grown inlaboratory conditions (Grunebaum et al., 2006). The cellsin culture are exposed to the virus, carrying the desiredgene. After infection and integration of the desired genein the cells DNA, the cells are returned in the patient byinjection into a vein. This technique is called ex vivo,because the gene is transferred to the cells, while theyare outside the patients body. In the in vivo-technique,

    the gene of interest is transferred to cells inside thepatients body by using of liposomes (fatty particles).

    Risks associated with current gene therapy

    Viruses can infect more than one cell types. Viral vectorsmight alter more than the intended cells. With othewords, the external gene might be inserted into the wronglocation in the DNA, causing cancer or other damageWhen DNA is injected directly into a tumor, there is a risksome DNA to be introduced into germ cells, producinginheritable changes. The gene might be over-expressed(toxicity); the viral vector could cause inflammation orimmune reaction; the virus could be transmitted to otheindividuals or the environment.

    RESEARCH STUDY

    Restorative effect of insulin-like growth factor-I genetherapy in the hypothalamus of senile rats withdopaminergic dysfunction

    Insulin-like growth factor-I (IGF-I) is a powerfuneuroprotective molecule that is strongly induced in thecentral nervous system after different insults. In onestudy a recombinant adenoviral vector (RAd-IGFIharboring the gene for rat IGF-I was constructed andused to implement IGF-I gene therapy in thehypothalamus of senile female rats, which displayhypothalamic dopaminergic (DA) neurodegeneration andas a consequence, chronic hyperprolactinemia

    Restorative IGF-I gene therapy was implemented inyoung (5 months) and senile (28 months) female ratswhich received a single intrahypothalamic injection of 3 x10(9) plaque-forming units of RAd-betagal (a controadenoviral vector expressing beta-galactosidase) or RAd-IGFI and were killed 17 days post-injection. In the younganimals, neither vector modified serum prolactin levelsbut in the RAd-IGFI-injected senile rats a nearly fulreversion of their hyperprolactinemic status wasrecorded. Morphometric analysis revealed a significanincrease in the total number of tyrosine hydroxylase-positive cells in the hypothalamus of experimental ascompared with control senile animals (5874486 and

    3390498, respectively). It was concluded that IGF-gene therapy in senile female rats is highly effective forestoring their hypothalamic DA dysfunction and thusreversing their chronic hyperprolactinemia (Here et al.2007).

    The ependimal route for Insuline-Like Growth Factor-1, Gene Therapy in the brain.

    In one study, it has been shown thaintracerebroventricular administration of the peptide

  • 8/12/2019 28(naveedJMPR-11-238

    4/6

    insulin-like growth factor-1 (IGF-1) is effectiveneuroprotective strategy in the brain of different animalmodels, a major advantage being the achievement ofhigh concentrations of IGF-1 in the brain without alteringserum levels of the peptide. In this study highperformance recombinant adenoviral (RAd) vectors

    expressing their transgene were used under the controlof the potent mouse cytomegalovirus immediate early(mCMV) promoter, to transduce brain ependymal cellswith high efficiency and to achieve effective release oftransgenic IGF-1 into the cerebrospinal fluid (CSF). RAdvectors expressing either the chimeric protein (TK/GFP)fus (green fluorescent protein fused to HSV1 thymidinekinase) or the cDNA encoding rat IGF-1, both driven bythe mCMV promoter were constructed. The vectors wereinjected into the lateral ventricles of young rats andchimeric GFP expression in brain sections was assessedby fluorescence microscopy. The ependymal cell markervimentin was detected by immunofluorescence andnuclei were labeled with the DNA dye DAPI. Blood andCSF samples were drawn at different times post vectorinjection. In all cerebral ventricles, vimentinimmunoreactive cells of the ependyma werepredominantly transduced by RAd-(TK/GFP)fus, showingnuclear and cytoplasmic expression of the transgene. Fortanycytes (TK/GFP)fus expression was evident in theircytoplasmic processes as they penetrated deep into thehypothalamic parenchyma. Intracerebroventricularinjection of RAd-IGF-1 induced high levels of IGF-1 in theCSF but not in serum. It was concluded that theependymal route constitutes an effective approach forimplementing experimental IGF-1 gene therapy in thebrain (Hereb et al., 2009).

    Protection and repair of the nigrostriataldopaminergic system by GDNF in vivo

    Glial cell line derived neurotrophic factor (GDNF), arecently cloned new member of the transforming growthfactor- superfamily, promotes survival of cultured fetalmesencephalic dopamine neurons and is expressed inthe developing striatum. Dopaminergic neurotoxin lmethyl4phenyl1,2,3,6tetrahydropyridine (MPTP)produces parkinsonian symptoms in man and GDNFexert protective or regenerative effects in vivoin the adult

    nigrostriatal dopamine system in C57/B1 mice. GDNFinjected over the substan-tia nigra or in striatum beforeMPTP potently protects the dopamine system, as shownby numbers of mesencephalic dopamine nerve cellbodies, dopamine nerve terminal densities and dopaminelevels.

    When GDNF is given after MPTP, dopamine levels andfibre densities are significantly restored. In both cases,motor behaviour is increased above normal levels. It wasconclude that intracerebral GDNF administration exertsboth protective and reparative effects on the nigrostriataldopamine system, which may have implications for

    Akhtar et al. 1815

    the development of new treatment strategies foParkinson's disease (Tomac et al., 1995).

    Glial cell line-derived neurotrophic factor supportssurvival of injured midbrain dopaminergic neurons

    Glial cell-lined derived neurotrophic factor (GDNF) hasbeen shown to promote survival of developingmesencephalic dopaminergic neurons in vitro. GDNF hasa positive effect on injured adult midbrain dopaminergicneurons in situ, in a study single dose of GDNF wasinjected into the substantia nigra following a unilateralesion of the nigrostriatal system. Rats were unilaterallylesioned by a single stereotaxic injection of 6hydroxydopamine (6-OHDA; 9 g/4 l normal saline with0.02% ascorbate) into the medial forebrain bundle andtested weekly for apomorphine-induced (0.05 mg/kg s. c.contralateral rotation behavior, Rats that manifested >300turns/hour received a nigral injection of 100 g GDNF, ocytochrome C as a control, 4 weeks following the 6OHDA lesion, Rotation behavior was quantified weeklyfor 5 weeks after GDNF. Rats were subsequentlyanesthetized, transcardially perfused, and processed fotyrosine hydroxylase immunohistochemistry. It was foundthat 100 g GDNF decreased apomorphine-inducedrotational behavior by more than 85%Immunohistochemical studies revealed that tyrosinehydroxylase immunoreactivity was equally reduced in thestriatum ipsilateral to the lesion in both cytochrome C andGDNF-injected animals. In contrast, large increments intyrosine hydroxylase immunoreactivity were observed inthe substantia nigra of animals treated with 100 g o

    GDNF, with a significant increase in numbers of tyrosinehydroxylase-immunoreactive cell bodies and neurites aswell as a small increase in the cell body area of theseneurons. It was concluded that GDNF can maintain thedopaminergic neuronal phenotype in a number of nigraneurons following a unilateral nigrostriatal lesion in the ra(Bowenkamp et al., 1995).

    Dopaminergic neurons protected from degenerationby GDNF gene therapy

    Glial cell line-derived neurotrophic factor (GDNF

    supports growth and survival of dopaminergic (DA)neurons. A replication-defective adenoviral (Ad) vectoencoding human GDNF injected near the rat substantianigra was found to protect DA neurons from theprogressive degeneration induced by the neurotoxin 6-hydroxydopamine (6-OHDA) injected into the striatum. AdGDNF gene therapy reduced loss of DA neuronsapproximately threefold 6 weeks after 6-OHDA lesion, ascompared with no treatment or injection of Ad lacZ or AdmGDNF (encoding a biologically inactive deletion mutanGDNF). It was concluded that Ad vector-mediated GDNFgene therapy may slow the DA neuronal cell loss in

  • 8/12/2019 28(naveedJMPR-11-238

    5/6

    1816 J. Med. Plant. Res.

    humans with Parkinson's disease (Choi-Lundberg et al.,1997)

    DISCUSSION

    Plasmid DNA (pDNA) expression vectors are funda-mental to all forms of non-viral gene transfer. Principlesof pDNA design and production include the impact ofbacterially-derived sequences on transgene expressionand minicircle approaches to minimize their effects.

    The impact of inclusion of DNA-elements such asscaffold matrix attachment regions (S/MARs),transcription factor (TF)-binding sites and tissue-specificpromoters are described. The benefits of eliminating CG-dinucleotides (CpGs) from the pDNA are also considered(Gill, 2009). In the past 2 years, new gene-targetingapproaches, using adeno-associated virus (AAV) anddesigner zinc-finger nucleases, have been successfully

    applied for production of genetically-modified ferrets,pigs, mice and zebrafish. Gene targeting, using thesetools, has been combined with somatic cell nucleartransfer and germ cell transplantation to generate gene-targeted animal models. These new technical advances,which do not require the generation of embryonic stemcell-derived chimeras, would greatly accelerate theproduction of non-mouse animal models for biomedicalresearch (Yan, 2009). In the last two years, significantadvances in understanding of the cellular-innateresponses, elicited or activated by the entry of ampliconparticles, have been seen, which might, in part, explainthe transient nature of the often observed in infected with

    helper-free amplicon stocks cells transgene expression.At the technological level, the most consistent progresshas been in strategies to enhance the stability oftransgene cassettes, either through integration into hostchromosomes or through the conversion of the amplicongenome into a replication-competent extra chromosomalelement (Epstein, 2009). Gene therapies, aiming thetreatment of arthritis and tissue repair, continue to be themost active areas of research for bone and jointdiseases. In the past 2 years, two trials in rheumatoidarthritis have been completed: A phase I study, reportingsafety and a phase I/II study, and that has not yet bepublished. An additional, small study has reported thefirst evidence of clinical efficacy.

    Two phase I trials of gene therapy for osteoarthritishave also been initiated. There is much pre-clinicalactivity in developing AAV vectors for future trials in thegene therapy of arthritis. The research onto the tissuerepair and regeneration remains at a pre-clinical stage,but a considerable volume of research attests to thepromise of gene transfer in this arena, especially in thecontext of bone healing, is necessary. For tissue repair,the major research questions are still which genes shouldbe used and how they could be best delivered (Evans,2009).

    CONCLUSION

    In the area of gene therapy, it is clear that many excitinginnovations are emerging. While many of these newgene-therapy and biotech products might yet haveunknown risks, they also have the potential fo

    tremendous patient benefit. In this review article, severacellular and gene transfer approaches have beeninvestigated at a pre-clinical level, and some clinical trialshave been made. A number of gene-therapy trials havebeen completed and several of them are currently inprogress. Viral vectors are potent gene-deliveryplatforms, used for treatment of genetic and acquireddiseases.

    REFERENCES

    Breaker R (2009) engineering ligand-responsive gene-control elements

    lessons learned from natural riboswitches, Gene Ther., 16:11891201.

    Bowenkamp KE, Hoffman A F, Gerhardt GA, Henry M A, Biddle PTHoffer B J, Granholm ACE(1995). Glial cell line-derived neurotrophicfactor supports survival of injured midbrain dopaminergic neurons. JComp. Neurol., 355: 479-489.

    Coleman JE, Huentelman MJ, Kasparov S, Metcalfe BL, Paton JFKatovich MJ, Rowland SL (2003). Ef ficient large-scale production andconcentration of HIV-1-based lentiviral vectors for use in vivoPhysiol. Genomics., 12: 221-228.

    Choi-Lundberg DL, Lin Q, Chang YN, Chiang YL, Hay CM, Mohajeri HDavidson BL, Bohn MC (1997). Dopaminergic neurons protectedfrom degeneration by GDNF gene therapy. Sci., 275: 838-841.

    Ehmann WC, Rabkin CS, Eyster, ME., Goedert JJ (1994)Thrombocytopenia in HIV-infected and uninfected hemophiliacsMulticenter Hemophilia Cohort Study. Am. J. Hematol., 54(4): 396

    300.Epstein A (2009). Progress and prospects: Biological properties andtechnological advances of herpes simplex virus type 1-basedamplicon vectors, Gene Ther., 16: 709-715.

    Evans C, Ghivizzani S, Robbins P (2009). Progress and Prospectsgenetic treatments for disorders of bones and joints. Gene Ther., 16944-952.

    Gill D, Pringle A, Hyde1 S (2009). Progress and Prospects: The designand production of plasmid vectors, Gene Ther., 16: 165-171.

    Grunebaum E, Mazzolari E, Porta F (2006). Bone marrowtransplantation for severe combined immune deficiency. JAMA, 295508-518.

    Hereb CB, Sonntag W, Morel G, Portiansky EL, Goya RG (2009).The ependimal route for Insuline-Like Growth Factor-1. GeneTherapy in the brain. Neurosci., 163: 442447.

    Here CB, Cristina C, Rimoldi OJ, Bec-Villalobos D, Cambiaggi VPortiansky EL, Goya RG (2007). Restorative effect of Insulin-like

    Growth Factor-I gene therapy in the hypothalamus of senile rats withdopaminergic dysfunction; Gene Ther., 14: 237-245.Isman O, Roberts ML, Morgan JE, Graham IR, Goldring K, Lawrence

    DJ, Dunckley G, Porter AC, Partridge TA, Dickson G (2008). HumGene Ther., 19(10): 1000-1008.

    Kay MA, Manno CS, Ragni MV (2000). Evidence for gene transfer andexpression of factor IX in hemophilia B patients treated with an AAVvector. Nat. Genet., 24: 257-261.

    Lam A, Dean A (2010). Progress and prospects: nuclear import ononviral vectors, Gene Ther., 17: 439-447.

    Macnab S, Whitehouse A (2009). Progress and prospects: humanartificial chromosomes. Gene Ther., 16: 1180-1188.

    Mannucci PM, Tuddenham EG (2001). The hemophilias--from royagenes to gene therapy. N. Engl. J. Med., 344: 1773.

  • 8/12/2019 28(naveedJMPR-11-238

    6/6

    Mastaglio S, Stanghellini Mt, Bordignon C, Bondanza A (2010).Progress and prospects: graft-versus-host disease.Gene Ther., 17(11): 1309-1317.

    McMenamin M, Wood M (2010). Progress and prospects:Immunobiology of gene therapy for neurodegenerative disease:prospects and risks, 17: 448-458.

    Nayak S, Herzog R (2010). Progress and prospects: immune responsesto viral vectors, Gene Ther., 17: 295-304.

    QasimW, Gaspar H, Thrasher A (2009). Progress and prospects: genetherapy for inherited immunodeficiencies, Gene Ther., 16: 1285-1291.

    Akhtar et al. 1817

    Sinn PL, Burnight E, McCray Jr (2009). Progress and Prospectsprospects of repeated pulmonary administration of viral vectorsGene Ther., 16: 1059-1065.

    Tomac A, Lindqvist E, Lin LFH, grenn SO, Young D, Hoffer BJ, OlsonL (1995). Protection and repair of the nigrostriatal dopaminergicsystem by GDNF in vivo. Nat., 373: 335-339.

    Yan Z, Sun X, Engelhardt JF (2009). Progress and prospectstechniques for site-directed mutagenesis in animal models, GeneTher., 16: 581-588.