Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Placenta ; 103: 50-52, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33075720

RESUMO

Umbilical cord blood is an important graft source in the treatment of many genetic, hematologic, and immunologic disorders by hematopoietic stem cell transplantation. Millions of cord blood units have been collected and stored for clinical use since the inception of cord blood banking in 1989. However, the use of cord blood in biomedical research has been limited by access to viable samples. Here, we present a cost-effective, self-sustaining model for the procurement of fresh umbilical cord blood components for research purposes within hospital-affiliated academic institutions.


Assuntos
Pesquisa Biomédica/organização & administração , Bancos de Sangue/organização & administração , Sangue Fetal , Modelos Organizacionais , Academias e Institutos/economia , Academias e Institutos/organização & administração , Academias e Institutos/normas , Pesquisa Biomédica/economia , Pesquisa Biomédica/métodos , Pesquisa Biomédica/normas , Bancos de Sangue/economia , Bancos de Sangue/normas , Coleta de Amostras Sanguíneas/economia , Coleta de Amostras Sanguíneas/métodos , Coleta de Amostras Sanguíneas/normas , California , Análise Custo-Benefício , Feminino , Sangue Fetal/citologia , Sangue Fetal/transplante , Transplante de Células-Tronco Hematopoéticas/economia , Transplante de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/normas , Humanos , Recém-Nascido , Gravidez
3.
Cytotherapy ; 20(12): 1486-1494, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30377039

RESUMO

A significant portion of the more than 1000 candidate cell and gene therapy products currently under clinical investigation (clinicaltrials.gov) are born out of academic research centers affiliated with universities, hospitals and non-profit research institutions. Supporting these efforts are myriad academic clinical materials production facilities with more than 40 such facilities currently operational in the United States alone. In March 2018, Stanford University's Laboratory for Cell and Gene Therapy held a symposium with the leaders and staff of more than 25 similar facilities to discuss the collective experience in developing, qualifying and operating cell and gene therapy manufacturing facilities according to current Good Manufacturing Practices. Topics included facility design, construction, staffing and operations and compliance. Leaders from several institutions gave overviews of the history of development of the facilities and discussed challenges and opportunities they had experienced over the past 10-20 years of operations. Working sessions were also held to discuss specific aspects of Process Development, Manufacturing, Quality Systems, Regulatory Affairs and Business Development with all participants contributing to the discussions. We summarize here the findings of this inaugural meeting with an emphasis on best practices and suggested guidelines for operations.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Terapia Genética , Instalações Industriais e de Manufatura , Centros Médicos Acadêmicos , Humanos , Instalações Industriais e de Manufatura/legislação & jurisprudência , Instalações Industriais e de Manufatura/organização & administração , Estados Unidos
4.
Mol Ther Methods Clin Dev ; 9: 99, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29601071

RESUMO

[This corrects the article DOI: 10.1016/j.omtm.2017.09.001.].

5.
Mol Ther Methods Clin Dev ; 8: 1-7, 2018 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-29034262

RESUMO

Lentiviral vectors are a common tool used to introduce new and corrected genes into cell therapy products for treatment of human diseases. Although lentiviral vectors are ideal for delivery and stable integration of genes of interest into the host cell genome, they potentially pose risks to human health, such as integration-mediated transformation and generation of a replication competent lentivirus (RCL) capable of infecting non-target cells. In consideration of the latter risk, all cell-based products modified by lentiviral vectors and intended for patient use must be tested for RCL prior to treatment of the patient. Current Food and Drug Administration (FDA) guidelines recommend use of cell-based assays to this end, which can take up to 6 weeks for results. However, qPCR-based assays are a quick alternative for rapid assessment of RCL in products intended for fresh infusion. We describe here the development and qualification of a qPCR assay based on detection of envelope gene sequences (vesicular stomatitis virus G glycoprotein [VSV-G]) for RCL in accordance with Minimum Information for Publication of Quantitative Real-Time PCR Experiments (MIQE) guidelines. Our results demonstrate the sensitivity, linearity, specificity, and reproducibility of detection of VSV-G sequences, with a low false-positive rate. These procedures are currently being used in our phase 1 clinical investigations.

6.
Mol Ther Methods Clin Dev ; 3: 16067, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27900346

RESUMO

Gene therapy for HIV-1 infection is a promising alternative to lifelong combination antiviral drug treatment. Chemokine receptor 5 (CCR5) is the coreceptor required for R5-tropic HIV-1 infection of human cells. Deletion of CCR5 renders cells resistant to R5-tropic HIV-1 infection, and the potential for cure has been shown through allogeneic stem cell transplantation with naturally occurring homozygous deletion of CCR5 in donor hematopoietic stem/progenitor cells (HSPC). The requirement for HLA-matched HSPC bearing homozygous CCR5 deletions prohibits widespread application of this approach. Thus, a strategy to disrupt CCR5 genomic sequences in HSPC using zinc finger nucleases was developed. Following discussions with regulatory agencies, we conducted IND-enabling preclinical in vitro and in vivo testing to demonstrate the feasibility and (preclinical) safety of zinc finger nucleases-based CCR5 disruption in HSPC. We report here the clinical-scale manufacturing process necessary to deliver CCR5-specific zinc finger nucleases mRNA to HSPC using electroporation and the preclinical safety data. Our results demonstrate effective biallelic CCR5 disruption in up to 72.9% of modified colony forming units from adult mobilized HSPC with maintenance of hematopoietic potential in vitro and in vivo. Tumorigenicity studies demonstrated initial product safety; further safety and feasibility studies are ongoing in subjects infected with HIV-1 (NCT02500849@clinicaltrials.gov).

8.
Curr HIV/AIDS Rep ; 12(1): 79-87, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25578054

RESUMO

Since the demonstration of a cure of an HIV+ patient with an allogeneic stem cell transplant using naturally HIV-resistant cells, significant interest in creating similar autologous products has fueled the development of a variety of "cell engineering" approaches to stem cell therapy for HIV. Among the more well-studied strategies is the inhibition of viral entry through disruption of expression of viral co-receptors or through competitive inhibitors of viral fusion with the cell membrane. Preclinical evaluation of these approaches often starts in vitro but ultimately is tested in animal models prior to clinical implementation. In this review, we trace the development of several key approaches (meganucleases, short hairpin RNA (shRNA), and fusion inhibitors) to modification of hematopoietic stem cells designed to impart resistance to HIV to their T-cell and monocytic progeny. The basic evolution of technologies through in vitro and in vivo testing is discussed as well as the pros and cons of each approach and how the addition of postentry inhibitors may enhance the overall antiviral efficacy of these approaches.


Assuntos
Terapia Genética , Infecções por HIV/terapia , HIV/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Replicação Viral/fisiologia , Humanos , Transplante Homólogo/métodos
9.
Front Genet ; 6: 357, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26858745

RESUMO

Rapid innovation in (epi)genetics and biomarker sciences is driving a new drug development and product development pathway, with the personalized medicine era dominated by biologic therapeutics and companion diagnostics. Companion diagnostics (CDx) are tests and assays that detect biomarkers and specific mutations to elucidate disease pathways, stratify patient populations, and target drug therapies. CDx can substantially influence the development and regulatory approval for certain high-risk biologics. However, despite the increasingly important role of companion diagnostics in the realization of personalized medicine, in the USA, there are only 23 Food and Drug Administration (FDA) approved companion diagnostics on the market for 11 unique indications. Personalized medicines have great potential, yet their use is currently constrained. A major factor for this may lie in the increased complexity of the companion diagnostic and corresponding therapeutic development and adoption pathways. Understanding the market dynamics of companion diagnostic/therapeutic (CDx/Rx) pairs is important to further development and adoption of personalized medicine. Therefore, data collected on a variety of factors may highlight incentives or disincentives driving the development of companion diagnostics. Statistical analysis for 36 hypotheses resulted in two significant relationships and 34 non-significant relationships. The sensitivity of the companion diagnostic was the only factor that significantly correlated with the price of the companion diagnostic. This result indicates that while there is regulatory pressure for the diagnostic and pharmaceutical industry to collaborate and co-develop companion diagnostics for the approval of personalized therapeutics, there seems to be a lack of parallel economic collaboration to incentivize development of companion diagnostics.

10.
Stem Cells Transl Med ; 3(10): 1199-208, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25107584

RESUMO

Genetic modification of adult human hematopoietic stem and progenitor cells (HSPCs) with lentiviral vectors leads to long-term gene expression in the progeny of the HSPCs and has been used to successfully treat several monogenic diseases. In some cases, the gene-modified cells have a selective growth advantage over nonmodified cells and eventually are the dominant engrafted population. However, in disease indications for which the gene-modified cells do not have a selective advantage, optimizing transduction of HSPC is paramount to successful stem cell-based gene therapy. We demonstrate here that transduction of adult CD34+ HSPCs with lentiviral vectors in the presence of rapamycin, a widely used mTORC1 inhibitor, results in an approximately threefold increase in stable gene marking with minimal effects on HSPC growth and differentiation. Using this approach, we have demonstrated that we can enhance the frequency of gene-modified HSPCs that give rise to clonogenic progeny in vitro without excessive increases in the number of vector copies per cell or changes in integration pattern. The genetic marking of HSPCs and expression of transgenes is durable, and transplantation of gene-modified HSPCs into immunodeficient mice results in high levels of gene marking of the lymphoid and myeloid progeny in vivo. The prior safe clinical history of rapamycin in other applications supports the use of this compound to generate gene-modified autologous HSPCs for our HIV gene therapy clinical trials.


Assuntos
Terapia Genética/métodos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Sirolimo/farmacologia , Transdução Genética/métodos , Animais , Técnicas de Cultura de Células/métodos , Citometria de Fluxo , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Técnicas In Vitro , Lentivirus , Camundongos , Camundongos Endogâmicos NOD , Reação em Cadeia da Polimerase
11.
Hum Gene Ther Methods ; 25(4): 221-31, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25003230

RESUMO

Hematopoietic stem cell gene therapy for HIV/AIDS is a promising alternative to lifelong antiretroviral therapy. One of the limitations of this approach is the number and quality of stem cells available for transplant following in vitro manipulations associated with stem cell isolation and genetic modification. The development of methods to increase the number of autologous, gene-modified stem cells available for transplantation would overcome this barrier. Hematopoietic stem and progenitor cells (HSPC) from adult growth factor-mobilized peripheral blood were cultured in the presence of an aryl hydrocarbon receptor antagonist (AhRA) previously shown to expand HSPC from umbilical cord blood. Qualitative and quantitative assessment of the hematopoietic potential of minimally cultured (MC-HSPC) or expanded HSPC (Exp-HSPC) was performed using an immunodeficient mouse model of transplantation. Our results demonstrate robust, multilineage engraftment of both MC-HSPC and Exp-HSPC although estimates of expansion based on stem cell phenotype were not supported by a corresponding increase in in vivo engrafting units. Bone marrow of animals transplanted with either MC-HSPC or Exp-HSPC contained secondary engrafting cells verifying the presence of primitive stem cells in both populations. However, the frequency of in vivo engrafting units among the more primitive CD34+/CD90+ HSPC population was significantly lower in Exp-HSPC compared with MC-HSPC. Exp-HSPC also produced fewer lymphoid progeny and more myeloid progeny than MC-HSPC. These results reveal that in vitro culture of adult HSPC in AhRA maintains but does not increase the number of in vivo engrafting cells and that HSPC expanded in vitro contain defects in lymphopoiesis as assessed in this model system. Further investigation is required before implementation of this approach in the clinical setting.


Assuntos
Células-Tronco Hematopoéticas/citologia , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Animais , Antígenos CD34/metabolismo , Compostos Azo/farmacologia , Linhagem da Célula , Células Cultivadas , Sangue Fetal/citologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Modelos Animais , Fenótipo , Pirazóis/farmacologia , Receptores de Hidrocarboneto Arílico/metabolismo , Antígenos Thy-1/metabolismo , Transplante Heterólogo
12.
Mol Ther ; 22(5): 952-63, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24576853

RESUMO

Gene therapy with hematopoietic stem and progenitor cells is a promising approach to engineering immunity to human immunodeficiency virus (HIV) that may lead to a functional cure for acquired immunodeficiency syndrome (AIDS). In support of this approach, we created lentiviral vectors with an engineered polycistronic platform derived from the endogenous MCM7 gene to express a diverse set of small antiviral RNAs and a drug resistance MGMT(P140K) marker. Multiple strategies for simultaneous expression of up to five RNA transgenes were tested. The placement and orientation of each transgene and its promoter were important determinants for optimal gene expression. Antiviral RNA expression from the MCM7 platform with a U1 promoter was sufficient to provide protection from R5-tropic HIV in macrophages and resulted in reduced hematopoietic toxicity compared with constructs expressing RNA from independent RNA polymerase III promoters. The addition of an HIV entry inhibitor and nucleolar TAR RNA decoy did not enhance antiviral potency over constructs that targeted only viral RNA transcripts. We also demonstrated selective enrichment of gene-modified cells in vivo using a humanized mouse model. The use of these less toxic, potent anti-HIV vectors expressing a drug selection marker is likely to enhance the in vivo efficacy of our stem cell gene therapy approach in treating HIV/AIDS.


Assuntos
Síndrome da Imunodeficiência Adquirida/terapia , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Terapia Genética , HIV/genética , Proteínas Supressoras de Tumor/genética , Síndrome da Imunodeficiência Adquirida/genética , Síndrome da Imunodeficiência Adquirida/patologia , Síndrome da Imunodeficiência Adquirida/virologia , Animais , Metilases de Modificação do DNA/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Resistência a Medicamentos/genética , Vetores Genéticos/uso terapêutico , HIV/imunologia , HIV/patogenicidade , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Humanos , Lentivirus/genética , Camundongos , Componente 7 do Complexo de Manutenção de Minicromossomo/genética , Estabilidade de RNA/genética , RNA Interferente Pequeno/genética , Proteínas Supressoras de Tumor/metabolismo
13.
Viruses ; 5(11): 2898-919, 2013 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-24284880

RESUMO

Over the past 15 years we have been investigating an alternative approach to treating HIV-1/AIDS, based on the creation of a disease-resistant immune system through transplantation of autologous, gene-modified (HIV-1-resistant) hematopoietic stem and progenitor cells (GM-HSPC). We propose that the expression of selected RNA-based HIV-1 inhibitors in the CD4+ cells derived from GM-HSPC will protect them from HIV-1 infection and results in a sufficient immune repertoire to control HIV-1 viremia resulting in a functional cure for HIV-1/AIDS. Additionally, it is possible that the subset of protected T cells will also be able to facilitate the immune-based elimination of latently infected cells if they can be activated to express viral antigens. Thus, a single dose of disease resistant GM-HSPC could provide an effective treatment for HIV-1+ patients who require (or desire) an alternative to lifelong antiretroviral chemotherapy. We describe herein the results from several pilot clinical studies in HIV-1 patients and our strategies to develop second generation vectors and clinical strategies for HIV-1+ patients with malignancy who require ablative chemotherapy as part of treatment and others without malignancy. The important issues related to stem cell source, patient selection, conditioning regimen and post-infusion correlative studies become increasingly complex and are discussed herein.


Assuntos
Terapia Genética , Infecções por HIV/genética , Infecções por HIV/terapia , HIV-1/fisiologia , Células-Tronco Hematopoéticas/imunologia , Adolescente , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD4-Positivos/virologia , Células Cultivadas , Criança , Pré-Escolar , Feminino , Infecções por HIV/imunologia , Infecções por HIV/virologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Lactente , Masculino , Pesquisa Translacional Biomédica , Adulto Jovem
14.
Mol Ther ; 21(6): 1259-69, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23587921

RESUMO

The HIV-1 coreceptor CCR5 is a validated target for HIV/AIDS therapy. The apparent elimination of HIV-1 in a patient treated with an allogeneic stem cell transplant homozygous for a naturally occurring CCR5 deletion mutation (CCR5(Δ32/Δ32)) supports the concept that a single dose of HIV-resistant hematopoietic stem cells can provide disease protection. Given the low frequency of naturally occurring CCR5(Δ32/Δ32) donors, we reasoned that engineered autologous CD34(+) hematopoietic stem/progenitor cells (HSPCs) could be used for AIDS therapy. We evaluated disruption of CCR5 gene expression in HSPCs isolated from granulocyte colony-stimulating factor (CSF)-mobilized adult blood using a recombinant adenoviral vector encoding a CCR5-specific pair of zinc finger nucleases (CCR5-ZFN). Our results demonstrate that CCR5-ZFN RNA and protein expression from the adenoviral vector is enhanced by pretreatment of HSPC with protein kinase C (PKC) activators resulting in >25% CCR5 gene disruption and that activation of the mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling pathway is responsible for this activity. Importantly, using an optimized dose of PKC activator and adenoviral vector we could generate CCR5-modified HSPCs which engraft in a humanized mouse model (albeit at a reduced level) and support multilineage differentiation in vitro and in vivo. Together, these data establish the basis for improved approaches exploiting adenoviral vector delivery in the modification of HSPCs.


Assuntos
Endonucleases/genética , Genômica/métodos , Células-Tronco Hematopoéticas/citologia , Receptores CCR5/genética , Dedos de Zinco/genética , Síndrome da Imunodeficiência Adquirida/terapia , Adenoviridae/genética , Animais , Antígenos CD34/genética , Antígenos CD34/metabolismo , Apoptose , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Endonucleases/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Deleção de Genes , Marcação de Genes , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos/genética , Fator Estimulador de Colônias de Granulócitos/metabolismo , HIV-1 , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Receptores CCR5/metabolismo
15.
Expert Opin Biol Ther ; 13(3): 437-45, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23394377

RESUMO

INTRODUCTION: HIV/AIDS continues to be a worldwide health problem and viral eradication has been an elusive goal. HIV+ patients are currently treated with combination antiretroviral therapy (cART) which is not curative. For many patients, cART is inaccessible, intolerable or unaffordable. Therefore, a new class of therapeutics for HIV is required to overcome these limitations. Cell and gene therapy for HIV has been proposed as a way to provide a functional cure for HIV in the form of a virus/infection resistant immune system. AREAS COVERED: In this review, the authors describe the standard therapy for HIV/AIDS, its limitations, current areas of investigation and the potential of hematopoietic stem cells modified with anti-HIV RNAs as a means to affect a functional cure for HIV. EXPERT OPINION: Cell and gene therapy for HIV/AIDS is a promising alternative to antiviral drug therapy and may provide a functional cure. In order to show clinical benefit, multiple mechanisms of inhibition of HIV entry and lifecycle are likely to be required. Among the most promising antiviral strategies is the use of transgenic RNA molecules that provide protection from HIV infection. When these molecules are delivered as gene-modified hematopoietic stem and progenitor cells, long-term repopulation of the patient's immune system with gene-modified progeny has been observed.


Assuntos
Terapia Genética , Infecções por HIV/terapia , HIV-1/fisiologia , Células-Tronco Hematopoéticas , RNA/genética , Terapia Baseada em Transplante de Células e Tecidos , Terapia Combinada , Infecções por HIV/genética , Humanos
16.
Hum Gene Ther ; 23(11): 1200-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22834872

RESUMO

Combinational therapy with small RNA inhibitory agents against multiple viral targets allows efficient inhibition of viral production by controlling gene expression at critical time points. Here we explore combinations of different classes of therapeutic anti-HIV-1 RNAs expressed from within the context of an intronic MCM7 (minichromosome maintenance complex component-7) platform that naturally harbors 3 microRNAs (miRNAs). We replaced the endogenous miRNAs with anti-HIV small RNAs, including small interfering RNAs (siRNAs) targeting HIV-1 tat and rev messages that function to induce post-transcriptional gene silencing by the RNA interference pathway, a nucleolar-localizing RNA ribozyme that targets the conserved U5 region of HIV-1 transcripts for degradation, and finally nucleolar trans-activation response (TAR) and Rev-binding element (RBE) RNA decoys designed to sequester HIV-1 Tat and Rev proteins inside the nucleolus. We demonstrate the versatility of the MCM7 platform in expressing and efficiently processing the siRNAs as miRNA mimics along with nucleolar small RNAs. Furthermore, three of the combinatorial constructs tested potently suppressed viral replication during a 1-month HIV challenge, with greater than 5-log inhibition compared with untransduced, HIV-1-infected CEM T lymphocytes. One of the most effective constructs contains an anti-HIV siRNA combined with a nucleolar-localizing U5 ribozyme and TAR decoy. This represents the first efficacious example of combining Drosha-processed siRNAs with small nucleolar ribonucleoprotein (snoRNP)-processed nucleolar RNA chimeras from a single intron platform for effective inhibition of viral replication. Moreover, we demonstrated enrichment/selection for cells expressing levels of the antiviral RNAs that provide optimal inhibition under the selective pressure of HIV. The combinations of si/snoRNAs represent a new paradigm for combinatorial RNA-based gene therapy applications.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Infecções por HIV/genética , HIV-1/genética , MicroRNAs/genética , Família Multigênica , Proteínas Nucleares/genética , RNA Catalítico/genética , RNA Interferente Pequeno/genética , Sequência de Bases , Linhagem Celular , Expressão Gênica , Ordem dos Genes , Terapia Genética , Vetores Genéticos/genética , Infecções por HIV/terapia , Humanos , Lentivirus/genética , Componente 7 do Complexo de Manutenção de Minicromossomo , Transdução Genética , Transgenes , Replicação Viral/genética
17.
Cytotherapy ; 14(7): 775-90, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22799276

RESUMO

Clinical trials over the last 15 years have demonstrated that cell and gene therapies for cancer, monogenic and infectious disease are feasible and can lead to long-term benefit for patients. However, these trials have been limited to proof-of-principle and were conducted on modest numbers of patients or over long periods of time. In order for these studies to move towards standard practice and commercialization, scalable technologies for the isolation, ex vivo manipulation and delivery of these cells to patients must be developed. Additionally, regulatory strategies and clinical protocols for the collection, creation and delivery of cell products must be generated. In this article we review recent progress in hematopoietic cell and gene therapy, describe some of the current issues facing the field and discuss clinical, technical and regulatory approaches used to navigate the road to product development.


Assuntos
Terapia Genética , Células-Tronco Hematopoéticas , Neoplasias/terapia , Terapia Baseada em Transplante de Células e Tecidos , Ensaios Clínicos como Assunto , Humanos , Imunoterapia Adotiva , Neoplasias/genética
18.
Sci Transl Med ; 2(36): 36ra43, 2010 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-20555022

RESUMO

AIDS patients who develop lymphoma are often treated with transplanted hematopoietic progenitor cells. As a first step in developing a hematopoietic cell-based gene therapy treatment, four patients undergoing treatment with these transplanted cells were also given gene-modified peripheral blood-derived (CD34(+)) hematopoietic progenitor cells expressing three RNA-based anti-HIV moieties (tat/rev short hairpin RNA, TAR decoy, and CCR5 ribozyme). In vitro analysis of these gene-modified cells showed no differences in their hematopoietic potential compared with nontransduced cells. In vitro estimates of successful expression of the anti-HIV moieties were initially as high as 22% but declined to approximately 1% over 4 weeks of culture. Ethical study design required that patients be transplanted with both gene-modified and unmanipulated hematopoietic progenitor cells obtained from the patient by apheresis. Transfected cells were successfully engrafted in all four infused patients by day 11, and there were no unexpected infusion-related toxicities. Persistent vector expression in multiple cell lineages was observed at low levels for up to 24 months, as was expression of the introduced small interfering RNA and ribozyme. Therefore, we have demonstrated stable vector expression in human blood cells after transplantation of autologous gene-modified hematopoietic progenitor cells. These results support the development of an RNA-based cell therapy platform for HIV.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/genética , Infecções por HIV/terapia , Linfoma Relacionado a AIDS/terapia , RNA/genética , Adulto , Feminino , Transplante de Células-Tronco Hematopoéticas , Humanos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
19.
J Immunother ; 30(6): 644-54, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17667528

RESUMO

We have developed an innovative system for ex vivo processing of patient-specific cell products to produce large numbers of T-lymphocytes in support of phase 2 adoptive immunotherapy trials for hematologic malignancies. Extensive efforts were undertaken to close the cell processing system to improve the safety profile of the process and comply with new federal regulations regarding cell and tissue processing. Our results demonstrate that apheresis products can be processed in a closed system (Cytomate) with similar yields (approximately 4 x 10(9) mononuclear cells/apheresis) and recoveries (approximately 60% of starting mononuclear cells) to manual cell processing. Cells processed with this system could be cryopreserved for up to 5 months without significant loss of recovery or viability. Additionally, we have evaluated the use of gas permeable bags and developed perfusion bioreactor protocols in which T cells can be rapidly produced in excess of 10(10) viable cells per liter of culture. Using similar methods for upfront processing, we have also developed methods for positive selection and ex vivo culture of CD4+ T cells that result in 200 to 800-fold expansion of fresh or cryopreserved samples. T cells produced in these systems were shown to retain activation-induced cytolytic capability and TH1/TH2 cytokine production as a measure of biologic potency. These new methods allow for more efficient production multiple patient-specific products by satisfying the basic tenants of safety and efficacy required for early phase clinical trials of cell products.


Assuntos
Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Técnicas de Cultura de Células , Imunoterapia Adotiva , Linfócitos T/imunologia , Reatores Biológicos , Remoção de Componentes Sanguíneos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Separação Celular , Citocinas/biossíntese , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Fenótipo , Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia
20.
Mol Ther ; 15(4): 825-33, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17299405

RESUMO

Metastatic neuroblastoma is a poor-prognosis malignancy arising during childhood that overexpresses the L1-cell adhesion molecule (CD171). We have previously described a tumor L1-cell adhesion molecule-specific, single chain antibody-derived, chimeric antigen receptor designated CE7R for re-directing the antigen-specific effector functioning of cytolytic T lymphocytes. Here, we report on the feasibility of isolating, and the safety of infusing, autologous CD8(+) cytolytic T lymphocyte clones co-expressing CE7R and the selection-suicide expression enzyme HyTK in children with recurrent/refractory neuroblastoma. The cytolytic T lymphocyte products were derived from peripheral blood mononuclear cells that were subjected to polyclonal activation, plasmid vector electrotransfer, limiting dilution hygromycin selection, and expansion to numbers sufficient for adoptive transfer. In total, 12 infusions (nine at 10(8) cells/m(2), three at 10(9) cells/m(2)) were administered to six patients. No overt toxicities to tissues known to express L1-cell adhesion molecule (e.g., central nervous system, adrenal medulla, and sympathetic ganglia) were observed. The persistence of cytolytic T lymphocyte clones in the circulation, measured by vector-specific quantitative polymerase chain reaction, was short (1-7 days) in patients with bulky disease, but significantly longer (42 days) in a patient with a limited disease burden. This first-in-humans pilot study sets the stage for clinical trials employing adoptive transfer in the context of minimal residual disease.


Assuntos
Imunoterapia Adotiva/métodos , Molécula L1 de Adesão de Célula Nervosa/imunologia , Neuroblastoma/terapia , Linfócitos T Citotóxicos/imunologia , Animais , Sequência de Bases , Sobrevivência Celular , Células Clonais/imunologia , Primers do DNA/genética , Vetores Genéticos , Humanos , Imunoterapia Adotiva/efeitos adversos , Camundongos , Molécula L1 de Adesão de Célula Nervosa/genética , Neuroblastoma/imunologia , Plasmídeos/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Segurança , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA