Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Front Immunol ; 14: 1288794, 2023.
Article in English | MEDLINE | ID: mdl-38022629

ABSTRACT

Introduction: The evolution of novel SARS-CoV-2 variants significantly affects vaccine effectiveness. While these effects can only be studied retrospectively, neutralizing antibody titers are most used as correlates of protection. However, studies assessing neutralizing antibody titers often show heterogeneous data. Methods: To address this, we investigated assay variance and identified virus infection time and dose as factors affecting assay robustness. We next measured neutralization against Omicron sub-variants in cohorts with hybrid or vaccine induced immunity, identifying a gradient of immune escape potential. To evaluate the effect of individual mutations on this immune escape potential of Omicron variants, we systematically assessed the effect of each individual mutation specific to Omicron BA.1, BA.2, BA.2.12.1, and BA.4/5. Results: We cloned a library of pseudo-viruses expressing spikes with single point mutations, and subjected it to pooled sera from vaccinated hosts, thereby identifying multiple mutations that independently affect neutralization potency. Discussion: These data might help to predict antigenic features of novel viral variants carrying these mutations and support the development of broad monoclonal antibodies.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Retrospective Studies , SARS-CoV-2/genetics , COVID-19/prevention & control , Mutation , Vaccination , Antibodies, Neutralizing
2.
Vox Sang ; 118(5): 384-391, 2023 May.
Article in English | MEDLINE | ID: mdl-36912154

ABSTRACT

BACKGROUND AND OBJECTIVES: Corrected count increment (CCI) measurements monitor the effectiveness of platelet transfusions in haemato-oncology, but they usually fail in patients undergoing cardiac surgery. We investigated whether polymerase chain reaction (PCR) of mitochondrial single-nucleotide polymorphisms (SNPs) is able to monitor the survival of transfused platelets in these patients. MATERIALS AND METHODS: Leukocyte-free, platelet-rich plasma was prepared from patients' blood to measure platelet counts based on patient-/donor-specific SNPs by digital PCR after DNA extraction. Platelet counts in samples from patients with severe thrombocytopenia were analysed by both PCR and flow cytometry. Ten patients undergoing cardiac surgery with the use of heart lung machine and without overt bleeding received a single apheresis platelet concentrate because of either dual platelet inhibition during a non-elective intervention or a complex procedure. Blood samples were collected at nine defined intervals (0-120 h) post transfusion. RESULTS: The digital PCR of the seven SNPs reliably quantified levels ≥0.6 G/L platelets, in good agreement with flow cytometry and without interference by other SNPs or by platelet activation. A mean 24-h CCI of 11.8 (range: 5.6-19.8) and a mean 120-h area under the curve (AUC) of 1386 (915-1821) hxG/L were observed for the transfused platelets. The mean AUC of 14,103 (3415-27,305) hxG/L for the patients' endogenous platelets indicates that transfused platelets represented only 11% (5-25) of the total platelet counts during 120 h post transfusion. CONCLUSION: PCR of mitochondrial SNPs offers a tool to assess the survival of platelets from apheresis concentrates in cardiac surgery patients to facilitate the implementation of improved transfusion strategies.


Subject(s)
Cardiac Surgical Procedures , Thrombocytopenia , Humans , Platelet Transfusion/methods , Blood Platelets/physiology , Polymerase Chain Reaction
3.
Cytotherapy ; 22(6): 337-342, 2020 06.
Article in English | MEDLINE | ID: mdl-32223996

ABSTRACT

BACKGROUND AIMS: At the frontier of transfusion medicine and transplantation, the field of cellular therapy is emerging. Most novel cellular therapy products are produced under investigational protocols with no clear standardization across cell processing centers. Thus, the purpose of this study was to uncover any variations in manufacturing practices for similar cellular therapy products across different cell processing laboratories worldwide. METHODS: An exploratory survey that was designed to identify variations in manufacturing practices in novel cellular therapy products was sent to cell processing laboratory directors worldwide. The questionnaire focused on the manufacturing life cycle of different cell therapies (i.e., collection, purification, in vitro expansion, freezing and storage, and thawing and washing), as well as the level of regulations followed to process each product type. RESULTS: The majority of the centers processed hematopoietic progenitor cells (HPCs) from peripheral blood (n = 18), bone marrow (n = 16) or cord blood (n = 19), making HPCs the most commonly processed cells. The next most commonly produced cellular therapies were lymphocytes (n = 19) followed by mesenchymal stromal cells (n = 14), dendritic cells (n = 9) and natural killer (NK) cells (n = 9). A minority of centers (<5) processed pancreatic islet cells (n = 4), neural cells (n = 3) and induced-pluripotent stem cells (n = 3). Thirty-two laboratories processed products under an investigational status, for either phase I/II (n = 27) or phase III (n = 17) clinical trials. If purification methods were used, these varied for the type of product processed and by institution. Environmental monitoring methods also varied by product type and institution. CONCLUSION: This exploratory survey shows a wide variation in cellular therapy manufacturing practices across different cell processing laboratories. A better understanding of the effect of these variations on the quality of these cell-based therapies will be important to assess for further process evaluation and development.


Subject(s)
Biotechnology/methods , Cell- and Tissue-Based Therapy/methods , Biotechnology/standards , Bone Marrow , Fetal Blood , Hematopoietic Stem Cells , Humans , Killer Cells, Natural , Laboratories/standards , Mesenchymal Stem Cells
4.
Transfusion ; 58(9): 2184-2191, 2018 09.
Article in English | MEDLINE | ID: mdl-30204955

ABSTRACT

BACKGROUND: Viability testing is a common practice in laboratories. The goal of this study was to ascertain current laboratory practices internationally for performing viability testing for cryopreserved cord blood (CB) products and glean information about how to standardize the method to improve interlaboratory reproducibility. STUDY DESIGN AND METHODS: A survey to evaluate current laboratory practices for viability testing was designed and distributed internationally. The question topics included sampling and testing methods, responses to unexpected results, and the rating of the reliability of the CB quality tests, together with expectations for standardization. RESULTS: There were 32 respondents to the survey, of whom 28 responded to the more detailed questionnaire about viability methods. Overall, responses indicated that various stains were used among the laboratories, and when multiple sites used the same viability stain the methods differed. The majority of the respondents were in favor of standardizing the viability testing methods. A wide variety of preferences were communicated about how to standardize the method, but a majority did advocate the use of 7-aminoactinomycin D (7-AAD) with flow cytometry. CONCLUSIONS: The survey results revealed a variety of tests and inconsistent interlaboratory practices for performing the viability assay. Flow cytometry with a 7-AAD dye was suggested as a first step toward standardization.


Subject(s)
Blood Preservation/methods , Blood Safety , Cord Blood Stem Cell Transplantation , Cryopreservation/methods , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Cell Nucleus/ultrastructure , Cell Separation/methods , Cell Survival , Cord Blood Stem Cell Transplantation/methods , Cord Blood Stem Cell Transplantation/standards , Dactinomycin/analogs & derivatives , Flow Cytometry/methods , Fluorescent Dyes , Health Care Surveys , Hematopoietic Stem Cells/ultrastructure , Humans , Infant, Newborn , International Cooperation , Internet , Laboratories/standards , Procedures and Techniques Utilization , Reproducibility of Results
5.
Transfusion ; 58(2): 461-469, 2018 02.
Article in English | MEDLINE | ID: mdl-29210068

ABSTRACT

BACKGROUND: Cell therapy products are often stored and transported between sites. The aim of this study was to determine the effect of storage temperature, solution, and cell concentration on nonmobilized, peripheral blood-derived mononuclear cells (MNCs). STUDY DESIGN AND METHODS: This was a multicenter prospective study involving healthy volunteers who underwent nonmobilized MNC collection by apheresis. Products were processed at local laboratories and concentrated to either 100 × 106 or 300 × 106 nucleated cells/mL in 5% human serum albumin (HSA) or HypoThermosol FRS (HT; BioLife Solutions). Products were stored at room temperature (RT; 20-25°C) or refrigerated temperatures (2-8°C) with assessment at 0, 24, 48, and 72 hours. NC and MNC concentration, viability, and flow cytometric analysis for CD3, CD4, CD8, CD14, CD19, CD25, and CD56 were measured. RESULTS: Viability decreased over time for all conditions tested. Refrigerated storage preserved viability greater than RT storage, especially for products with a higher cell concentration. RT maintenance with a high cell concentration was associated with a relative loss of CD14- and CD4-positive cells, whereas the concentration of cells positive for other markers tested did not vary. Finally, there was delayed decrease in pH when using HT compared with HSA; however, there was no difference in viability between the two solutions. CONCLUSION: Low cell concentrations (approx. 100 × 106 cells/mL), refrigerated temperatures, and HT storage solution appear to be the optimal conditions for storing nonmobilized, peripheral blood-derived MNC products.


Subject(s)
Antigens, CD/metabolism , Blood Component Removal , Blood Component Transfusion , Blood Preservation/methods , Leukocytes/cytology , Leukocytes/metabolism , Adult , Female , Humans , Male , Prospective Studies
6.
Transfusion ; 58(1): 151-157, 2018 01.
Article in English | MEDLINE | ID: mdl-29030954

ABSTRACT

BACKGROUND: Typical practice is to transfuse group-specific plasma units; however, there are situations where group AB plasma (universal donor) is issued to group A, B, or O recipients. If demand for group AB plasma exceeds collections, there is potential for shortage. This project explored the patterns of group AB plasma utilization at hospitals around the world. STUDY DESIGN AND METHODS: The study had two phases: a survey that inquired about hospital group AB plasma inventory, policies, and transfusion practices and a retrospective review of 2014 calendar year data where participants submitted information on plasma disposition including ABO group of unit and recipient, transfusion location, and select indications. Recruitment occurred through snowball sampling. Descriptive analyses were performed. RESULTS: Survey data were received from 25 centers across 10 countries; of those, 15 participants contributed to the data collection component. These 15 centers transfused a total of 43,369 AB plasma units during the study period. Only 1496 of 5541 (27%) group AB plasma units were transfused to group AB recipients. Transfusion policies, practices, and patterns were variable across sites. CONCLUSION: Group AB plasma units are frequently transfused to non-AB recipients. Whether transfusing 73% of group AB plasma units to non-AB recipients is the ideal inventory management strategy remains to be determined.


Subject(s)
ABO Blood-Group System , Blood Component Transfusion/statistics & numerical data , Inventories, Hospital/statistics & numerical data , Plasma , Adult , Americas , Blood Banks/statistics & numerical data , Blood Group Incompatibility , Child , Data Collection , Diagnosis-Related Groups , Europe , Health Care Surveys , Health Services Needs and Demand , Humans , Infant, Newborn , Internationality , Japan , New Zealand , Sampling Studies
7.
Transfusion ; 57(10): 2329-2337, 2017 10.
Article in English | MEDLINE | ID: mdl-28840943

ABSTRACT

BACKGROUND: Transfusion of group O blood to non-O recipients, or transfusion of D- blood to D+ recipients, can result in shortages of group O or D- blood, respectively. This study investigated RBC utilization patterns at hospitals around the world and explored the context and policies that guide ABO blood group and D type selection practices. STUDY DESIGN AND METHODS: This was a retrospective study on transfusion data from the 2013 calendar year. This study included a survey component that asked about hospital RBC selection and transfusion practices and a data collection component where participants submitted information on RBC unit disposition including blood group and D type of unit and recipient. Units administered to recipients of unknown ABO or D group were excluded. RESULTS: Thirty-eight hospitals in 11 countries responded to the survey, 30 of which provided specific RBC unit disposition data. Overall, 11.1% (21,235/191,397) of group O units were transfused to non-O recipients; 22.6% (8777/38,911) of group O D- RBC units were transfused to O D+ recipients, and 43.2% (16,800/38,911) of group O D- RBC units were transfused to recipients that were not group O D-. Disposition of units and hospital transfusion policy varied within and across hospitals of different sizes, with transfusion of group O D- units to non-group O D- patients ranging from 0% to 33%. CONCLUSION: A significant proportion of group O and D- RBC units were transfused to compatible, nonidentical recipients, although the frequency of this practice varied across sites.


Subject(s)
Erythrocyte Transfusion/statistics & numerical data , Erythrocytes/immunology , ABO Blood-Group System/immunology , Blood Group Incompatibility , Hospitals , Humans , Retrospective Studies , Rh-Hr Blood-Group System/immunology , Surveys and Questionnaires
8.
PLoS One ; 10(4): e0125622, 2015.
Article in English | MEDLINE | ID: mdl-25927683

ABSTRACT

Contaminations of cell cultures with microbiological organisms are well documented and can be managed in cell culture laboratories applying reliable detection, elimination and prevention strategies. However, the presence of viral contaminations in cell cultures is still a matter of debate and cannot be determined with general detection methods. In the present study we screened 577 human cell lines for the presence of murine leukemia viruses (MLV). Nineteen cell lines were found to be contaminated with MLV, including 22RV1 which is contaminated with the xenotropic murine leukemia virus-related virus variant of MLV. Of these, 17 cell lines were shown to produce active retroviruses determined by product enhanced reverse transcriptase PCR assay for reverse transcriptase activity. The contaminated cell lines derive from various solid tumor types as well as from leukemia and lymphoma types. A contamination of primary human cells from healthy volunteers could not be substantiated. Sequence analyses of 17 MLV PCR products and five complete MLV genomes of different infected cell lines revealed at least three groups of related MLV genotypes. The viruses harvested from the supernatants of infected cell cultures were infectious to uninfected cell cultures. In the course of the study we found that contamination of human genomic DNA preparations with murine DNA can lead to false-positive results. Presumably, xenotransplantations of the human tumor cells into immune-deficient mice to determine the tumorigenicity of the cells are mainly responsible for the MLV contaminations. Furthermore, the use of murine feeder layer cells during the establishment of human cell lines and a cross-contamination with MLV from infected cultures might be sources of infection. A screening of cell cultures for MLV contamination is recommended given a contamination rate of 3.3%.


Subject(s)
Leukemia Virus, Murine/isolation & purification , Cell Line , DNA, Viral/genetics , Genome, Viral/genetics , Genotype , Humans , Leukemia Virus, Murine/genetics , Polymerase Chain Reaction
9.
BMC Biotechnol ; 15: 10, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25888378

ABSTRACT

BACKGROUND: Antibody phage display is a proven key technology that allows the generation of human antibodies for diagnostics and therapy. From naive antibody gene libraries - in theory - antibodies against any target can be selected. Here we describe the design, construction and characterization of an optimized antibody phage display library. RESULTS: The naive antibody gene libraries HAL9 and HAL10, with a combined theoretical diversity of 1.5×10(10) independent clones, were constructed from 98 healthy donors using improved phage display vectors. In detail, most common phagemids employed for antibody phage display are using a combined His/Myc tag for detection and purification. We show that changing the tag order to Myc/His improved the production of soluble antibodies, but did not affect antibody phage display. For several published antibody libraries, the selected number of kappa scFvs were lower compared to lambda scFvs, probably due to a lower kappa scFv or Fab expression rate. Deletion of a phenylalanine at the end of the CL linker sequence in our new phagemid design increased scFv production rate and frequency of selected kappa antibodies significantly. The HAL libraries and 834 antibodies selected against 121 targets were analyzed regarding the used germline V-genes, used V-gene combinations and CDR-H3/-L3 length and composition. The amino acid diversity and distribution in the CDR-H3 of the initial library was retrieved in the CDR-H3 of selected antibodies showing that all CDR-H3 amino acids occurring in the human antibody repertoire can be functionally used and is not biased by E. coli expression or phage selection. Further, the data underline the importance of CDR length variations. CONCLUSION: The highly diverse universal antibody gene libraries HAL9/10 were constructed using an optimized scFv phagemid vector design. Analysis of selected antibodies revealed that the complete amino acid diversity in the CDR-H3 was also found in selected scFvs showing the functionality of the naive CDR-H3 diversity.


Subject(s)
Peptide Library , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics , Amino Acid Sequence , Autoantigens/chemistry , Autoantigens/immunology , Bacteriophages/genetics , Bacteriophages/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/immunology , Single-Chain Antibodies/chemistry
10.
Cytotherapy ; 15(3): 255-62, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23579058

ABSTRACT

BACKGROUND AIMS: Wide acceptance of the colony-forming unit (CFU) assay as a reliable potency test for stem cell products is hindered by poor inter-laboratory reproducibility. The goal of this study was to ascertain current laboratory practices for performing the CFU assay with an eye towards identifying practices that could be standardized to improve overall reproducibility. METHODS: A survey to evaluate current laboratory practices for performing CFU assays was designed and internationally distributed. RESULTS: There were 105 respondents to the survey, of whom 68% performed CFU assays. Most survey recipients specified that an automated rather than a manual cell count was performed on pre-diluted aliquots of stem cell products. Viability testing methods employed various stains, and when multiple sites used the same viability stain, the methods differed. Cell phenotype used to prepare working cell suspensions for inoculating the CFU assay differed among sites. Most respondents scored CFU assays at 14-16 days of incubation, but culture plates were read with various microscopes. Of 57 respondents, 42% had not performed a validation study or established assay linearity. Only 63% of laboratories had criteria for determining if a plate was overgrown with colonies. CONCLUSIONS: Survey results revealed inconsistent inter-laboratory practices for performing the CFU assay. The relatively low number of centers with validated CFU assays raises concerns about assay accuracy and emphasizes a need to establish central standards. The survey results shed light on numerous steps of the methodology that could be targeted for standardization across laboratories.


Subject(s)
Cell- and Tissue-Based Therapy , Colony-Forming Units Assay/standards , Hematopoietic Stem Cells , Stem Cells/cytology , Antigens, CD34/metabolism , Hematopoietic Stem Cell Transplantation , Humans , Reference Standards , Reproducibility of Results
11.
Mol Cell Proteomics ; 12(5): 1099-114, 2013 May.
Article in English | MEDLINE | ID: mdl-23315794

ABSTRACT

The recent Natural Killer (NK) cell maturation model postulates that CD34(+) hematopoietic stem cells (HSC) first develop into CD56(bright) NK cells, then into CD56(dim)CD57(-) and finally into terminally maturated CD56(dim)CD57(+). The molecular mechanisms of human NK cell differentiation and maturation however are incompletely characterized. Here we present a proteome analysis of distinct developmental stages of human primary NK cells, isolated from healthy human blood donors. Peptide sequencing was used to comparatively analyze CD56(bright) NK cells versus CD56(dim) NK cells and CD56(dim)CD57(-) NK cells versus CD56(dim)CD57(+) NK cells and revealed distinct protein signatures for all of these subsets. Quantitative data for about 3400 proteins were obtained and support the current differentiation model. Furthermore, 11 donor-independently, but developmental stage specifically regulated proteins so far undescribed in NK cells were revealed, which may contribute to NK cell development and may elucidate a molecular source for NK cell effector functions. Among those proteins, S100A4 (Calvasculin) and S100A6 (Calcyclin) were selected to study their dynamic subcellular localization. Upon activation of human primary NK cells, both proteins are recruited into the immune synapse (NKIS), where they colocalize with myosin IIa.


Subject(s)
Killer Cells, Natural/physiology , Proteome/metabolism , Amino Acid Sequence , CD56 Antigen/metabolism , CD57 Antigens/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Separation , Cells, Cultured , Humans , Immunological Synapses/metabolism , Lymphocyte Activation , Molecular Sequence Annotation , Molecular Sequence Data , Nonmuscle Myosin Type IIA/metabolism , Protein Transport , Proteome/chemistry , S100 Calcium Binding Protein A6 , S100 Calcium-Binding Protein A4 , S100 Proteins/chemistry , S100 Proteins/metabolism , Signal Transduction
12.
Transfus Med Hemother ; 40(6): 384-5, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24474887
13.
Tissue Eng Part A ; 19(3-4): 340-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22954487

ABSTRACT

There is a rise in the popularity of arthroscopic procedures in orthopedics. However, the majority of cell-based bone tissue-engineered constructs (TECs) rely on solid preformed scaffolding materials, which require large incisions and extensive dissections for placement at the defect site. Thus, they are not suitable for minimally invasive techniques. The aim of this study was to develop a clinically relevant, easily moldable, bone TEC, amenable to minimally invasive techniques, using human mesenchymal stromal cells (hMSCs) and calcium phosphate microparticles in combination with an in situ forming platelet-rich plasma gel obtained from human platelets. Most conventional TECs rely on seeding and culturing single-cell suspensions of hMSCs on scaffolds. However, for generating TECs amenable to the minimally invasive approach, it was essential to aggregate the hMSCs in vitro before seeding them on the scaffolds as unaggregated MSCs did not generate any bone. Twenty four hours of in vitro aggregation was determined to be optimal for maintaining cell viability in vitro and bone formation in vivo. Moreover, no statistically significant difference was observed in the amount of bone formed when the TECs were implanted via an open approach or a minimally invasive route. TECs generated using MSCs from three different human donors generated new bone through the minimally invasive route in a reproducible manner, suggesting that these TECs could be a viable alternative to preformed scaffolds employed through an open surgery for treating bone defects.


Subject(s)
Bone Development/physiology , Bone Substitutes/therapeutic use , Ceramics/therapeutic use , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Platelet-Rich Plasma , Tissue Engineering/methods , Animals , Cell Aggregation , Cells, Cultured , Combined Modality Therapy/methods , Gels/therapeutic use , Guided Tissue Regeneration/methods , Humans , Materials Testing , Mice , Minimally Invasive Surgical Procedures , Particle Size , Treatment Outcome
14.
Acta Biomater ; 8(8): 2969-77, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22522130

ABSTRACT

Numerous studies have shown that the physicochemical properties of biomaterials can control cell activity. Cell adhesion, proliferation, differentiation as well as tissue formation in vivo can be tuned by properties such as the porosity, surface micro- and nanoscale topography and chemical composition of biomaterials. This concept is very appealing for tissue engineering since instructive properties in bioactive materials can be more economical and time efficient than traditional strategies of cell pre-differentiation in vitro prior to implantation. The biomaterial surface, which is easy to modify due to its accessibility, may provide the necessary signals to elicit a certain cellular behavior. Here, we used gas plasma technology at atmospheric pressure to modify the physicochemical properties of polylactic acid and analyzed how this influenced pre-osteoblast proliferation and differentiation. Tetramethylsilane and 3-aminopropyl-trimethoxysilane with helium as a carrier gas or a mixture of nitrogen and hydrogen were discharged to polylactic acid discs to create different surface chemical compositions, hydrophobicity and microscale topographies. Such modifications influenced protein adsorption and pre-osteoblast cell adhesion, proliferation and osteogenic differentiation. Furthermore polylactic acid treated with tetramethylsilane enhanced osteogenic differentiation compared to the other surfaces. This promising surface modification could be further explored for potential development of bone graft substitutes.


Subject(s)
Cell Differentiation/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/drug effects , Plasma Gases/pharmacology , Adsorption/drug effects , Animals , Atmospheric Pressure , Biocompatible Materials/pharmacology , Cell Adhesion/drug effects , Cell Line , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Hydrophobic and Hydrophilic Interactions/drug effects , Lactic Acid/pharmacology , Mice , Microscopy, Atomic Force , Osteoblasts/metabolism , Polyesters , Polymers/pharmacology , Serum Albumin, Bovine/metabolism , Surface Properties/drug effects
15.
Hum Gene Ther Methods ; 23(1): 38-55, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22428979

ABSTRACT

SmartDCs (Self-differentiated Myeloid-derived Antigen-presenting-cells Reactive against Tumors) consist of highly viable dendritic cells (DCs) induced to differentiate with lentiviral vectors (LVs) after an overnight ex vivo transduction. Tricistronic vectors co-expressing cytokines (granulocyte-macrophage-colony stimulating factor [GM-CSF], interleukin [IL]-4) and a melanoma antigen (tyrosine related protein 2 [TRP2]) were used to transduce mouse bone marrow cells or human monocytes. Sixteen hours after transduction, the cells were dispensed in aliquots and cryopreserved for identity, potency, and safety analyses. Thawed SmartDCs readily differentiated into highly viable cells with a DC immunophenotype. Prime/boost subcutaneous administration of 1×10(6) thawed murine SmartDCs into C57BL/6 mice resulted into TRP2-specific CD8(+) T-cell responses and protection against lethal melanoma challenge. Human SmartDC-TRP2 generated with monocytes obtained from melanoma patients secreted endogenous cytokines associated with DC activation and stimulated TRP2-specific autologous T-cell expansion in vitro. Thawed human SmartDCs injected subcutaneously in NOD.Rag1(-/-).IL2rγ(-/-) mice maintained DC characteristics and viability for 1 month in vivo and did not cause any signs of pathology. For development of good manufacturing practices, CD14(+) monocytes selected by magnetic-activated cell separation were transduced in a closed bag system (multiplicity of infection of 5), washed, and cryopreserved. Fifty percent of the monocytes used for transduction were recovered for cryopreservation. Thawed SmartDCs produced in two independent runs expressed the endogenous cytokines GM-CSF and IL-4, and the resulting homogeneous SmartDCs that self-differentiated in vitro contained approximately 1.5-3.0 copies of integrated LVs per cell. Thus, this method facilitates logistics, standardization, and high recovery for the generation of viable genetically reprogrammed DCs for clinical applications.


Subject(s)
Biotechnology/methods , Dendritic Cells/immunology , Genetic Vectors/immunology , Immunotherapy/methods , Lentivirus/genetics , Melanoma/therapy , Animals , Blotting, Western , Cell Line , Cryopreservation , Cytokines/metabolism , DNA Primers/genetics , Dendritic Cells/virology , Flow Cytometry , Fluoresceins , Genes, RAG-1/genetics , Humans , Interleukin Receptor Common gamma Subunit/genetics , Melanoma/immunology , Membrane Proteins , Mice , Mice, Knockout , Peptide Fragments , Succinimides
16.
Anticancer Res ; 31(12): 4267-71, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22199290

ABSTRACT

BACKGROUND: Alterations of mitochondrial DNA (mtDNA) have been found in cancer patients, therefore informative mtDNA mutations could serve as biomarkers for the disease. MATERIALS AND METHODS: The two hypervariable regions HVR1 and HVR2 in the D-Loop region were sequenced in ten paired tissue and plasma samples from breast cancer patients. RESULTS: MtDNA mutations were found in all patients' samples, suggesting a 100% detection rate. Examining germline mtDNA mutations, a total of 85 mutations in the D-loop region were found; 31 of these mutations were detected in both tissues and matched plasma samples, the other 54 germline mtDNA mutations were found only in the plasma samples. Regarding somatic mtDNA mutations, a total of 42 mutations in the D-loop region were found in breast cancer tissues. CONCLUSION: Somatic mtDNA mutations in the D-loop region were detected in breast cancer tissues but not in the matched plasma samples, suggesting that more sensitive methods will be needed for such detection to be of clinical utility.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , DNA, Mitochondrial/genetics , Mutation , Aged , Breast Neoplasms/diagnosis , DNA/metabolism , DNA Primers/genetics , DNA, Mitochondrial/metabolism , Female , Germ-Line Mutation , Humans , Middle Aged , Mitochondria/metabolism , Protein Structure, Tertiary
17.
Pediatr Transplant ; 15(8): 809-18, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21967552

ABSTRACT

The presence of microchimerism in peripheral blood of solid organ transplant recipients has been postulated to be beneficial for allograft acceptance. Kinetics of donor cell trafficking and accumulation in pediatric allograft recipients are largely unknown. In this study, we implemented SNPs of the HVRs I and II of mitochondrial DNA to serve as molecular genetic markers to detect donor-specific cell chimerism after pediatric renal transplantation. Serial dilution of artificial chimeric DNA samples showed a linear correlation coefficient of R > 0.98 and a detection sensitivity of 0.01% with high reproducibility. Longitudinal semiquantitative analysis of donor-specific SNPs was then performed in peripheral blood mononuclear cells samples up to two yr post-transplant. Quantity of donor-specific cell chimerism in peripheral blood was highest in the early post-transplant period reaching values of ~10% after liver-kidney and 2.8% after renal transplantation. From one wk after transplantation, renal transplant patients exhibited an amount of donor-specific mtDNA ranging from 0.01% to 0.1%. We developed a highly accurate, sensitive, and rapid real-time quantitative PCR method using sequence-specific primers and fluorescent hydrolysis probes for the detection of at least 0.01% donor-specific cells in the recipient's peripheral blood after renal transplantation.


Subject(s)
DNA, Mitochondrial/genetics , Kidney Transplantation/immunology , Polymerase Chain Reaction/methods , Transplantation Chimera/genetics , DNA Mutational Analysis , Electron Transport Complex IV/genetics , Humans , Liver Transplantation/immunology , Liver-Specific Organic Anion Transporter 1 , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Tissue Donors
18.
Transfus Med Hemother ; 37(2): 56, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20737046
19.
Transfus Med Hemother ; 37(2): 57-64, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20737047

ABSTRACT

Cellular therapies that either use modifications of a patient's own cells or allogeneic cell lines are becoming in vogue. Besides the technical issues of optimal isolation, cultivation and modification, quality control of the generated cellular products are increasingly being considered to be more important. This is not only relevant for the cell's therapeutic application but also for cell science in general. Recent changes in editorial policies of respected journals, which now require proof of authenticity when cell lines are used, demonstrate that the subject of the present paper is not a virtual problem at all. In this article we provide 2 examples of contaminated cell lines followed by a review of the recent developments used to verify cell lines, stem cells and modifications of autologous cells. With relative simple techniques one can now prove the authenticity and the quality of the cellular material of interest and therefore improve the scientific basis for the development of cells for therapeutic applications. The future of advanced cellular therapies will require production and characterization of cells under GMP and GLP conditions, which include proof of identity, safety and functionality and absence of contamination.

20.
Transfusion ; 50(4): 843-55, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20003054

ABSTRACT

BACKGROUND: Dendritic cells (DCs) are applied worldwide in several clinical studies of immune therapy of malignancies, autoimmune diseases, and transplantations. Most legislative bodies are demanding high standards for cultivation and transduction of cells. Closed-cell cultivating systems like cell culture bags would simplify and greatly improve the ability to reach these cultivation standards. We investigated if a new polyolefin cell culture bag enables maturation and adenoviral modification of human DCs in a closed system and compare the results with standard polystyrene flasks. STUDY DESIGN AND METHODS: Mononuclear cells were isolated from HLA-A*0201-positive blood donors by leukapheresis. A commercially available separation system (CliniMACS, Miltenyi Biotec) was used to isolate monocytes by positive selection using CD14-specific immunomagnetic beads. The essentially homogenous starting cell population was cultivated in the presence of granulocyte-macrophage-colony-stimulating factor and interleukin-4 in a closed-bag system in parallel to the standard flask cultivation system. Genetic modification was performed on Day 4. After induction of maturation on Day 5, mature DCs could be harvested and cryopreserved on Day 7. During the cultivation period comparative quality control was performed using flow cytometry, gene expression profiling, and functional assays. RESULTS: Both flasks and bags generated mature genetically modified DCs in similar yields. Surface membrane markers, expression profiles, and functional testing results were comparable. The use of a closed-bag system facilitated clinical applicability of genetically modified DCs. CONCLUSIONS: The polyolefin bag-based culture system yields DCs qualitatively and quantitatively comparable to the standard flask preparation. All steps including cryopreservation can be performed in a closed system facilitating standardized, safe, and reproducible preparation of therapeutic cells.


Subject(s)
Cell Culture Techniques/methods , Dendritic Cells/physiology , Adult , Antigens, CD/analysis , Antigens, CD/genetics , Antigens, Surface/analysis , Cell Count , Cell Culture Techniques/instrumentation , Cell Division/physiology , Cellular Senescence/physiology , Child , Dendritic Cells/cytology , Dendritic Cells/ultrastructure , Female , Gene Transfer Techniques , Humans , Leukapheresis/methods , Leukocytes/cytology , Leukocytes/physiology , Lipopolysaccharide Receptors/analysis , Lipopolysaccharide Receptors/genetics , Male , Microscopy, Electron, Scanning , Middle Aged , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL