Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Transl Med ; 17(1): 258, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31391068

RESUMO

BACKGROUND: The rapid evolution of cell-based immunotherapies such as chimeric antigen receptor T-cells for treatment of hematological cancers has precipitated the need for a platform to expand these cells ex vivo in a safe, efficient, and reproducible manner. In the Quantum® Cell Expansion System (Quantum system) we evaluated the expansion of T-cells from healthy donors in a functionally-closed environment that reduces time and resources needed to produce a therapeutic dose. METHODS: Mononuclear cells from leukapheresis products from 5 healthy donors were activated with anti-CD3/CD28 Dynabeads® and expanded in the Quantum system for 8-9 days using xeno-free, serum-free medium and IL-2. Harvested cells were phenotyped by flow cytometry and evaluated for cytokine secretion by multiplex assays. RESULTS: From starting products of 30 or 85 × 106 mononuclear cells, CD3+ T-cell populations expanded over 500-fold following stimulation to provide yields up to 25 × 109 cells within 8 days. T-cell yields from all donors were similar in terms of harvest numbers, viability and doubling times. Functionality (secretion of IFN-γ, IL-2 and TNF-α) was retained in harvested T-cells upon restimulation in vitro and T-cells displayed therapeutically-relevant less-differentiated phenotypes of naïve and central memory T-cells, with low expression of exhaustion markers LAG-3 and PD-1. CONCLUSIONS: The Quantum system has been successfully used to produce large quantities of functional T-cells at clinical dosing scale and within a short timeframe. This platform could have wide applicability for autologous and allogeneic cellular immunotherapies for the treatment of cancer.


Assuntos
Complexo CD3/metabolismo , Técnicas de Cultura de Células/métodos , Linfócitos T/citologia , Biomarcadores/metabolismo , Proliferação de Células , Meios de Cultura , Citocinas/metabolismo , Humanos , Ácido Láctico/metabolismo , Fenótipo
2.
J Clin Med ; 12(17)2023 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-37685608

RESUMO

BACKGROUND: Prior studies have demonstrated improved efficacy when intra-articular (IA) therapeutics are injected using ultrasound (US) guidance. The aim of this study was to determine if clinical improvement in pain and function after IA hyaluronic acid injections using US is associated with changes in SF volumes and biomarker proteins at 3 months. METHODS: 49 subjects with symptomatic knee OA, BMI < 40, and KL radiographic grade II or III participated. Subjects with adequate aspirated synovial fluid (SF) volumes received two US-guided IA-HA injections of HYADD4 (24 mg/3 mL) 7 days apart. Clinical evaluations at 3, 6, and 12 months included WOMAC, VAS, PCS scores, 6 MWD, and US-measured SF depth. SF and blood were collected at 3 months and analyzed for four serum OA biomarkers and fifteen SF proteins. RESULTS: Statistical differences were observed at 3, 6, and 12 months compared to baseline values, with improvements at 12 months for WOMAC scores (50%), VAS (54%), and PCS scores (24%). MMP10 levels were lower at 3 months without changes in SF volumes, serum levels of C2C, COMP, HA, CPII, or SF levels of IL-1 ra, IL-4, 6, 7, 8, 15, 18, ILGFBP-1, 3, and MMP 1, 2, 3, 8, 9. Baseline clinical features or SF biomarker protein levels did not predict responsiveness at 3 months. CONCLUSIONS: Clinical improvements were observed at 12 months using US needle guidance for IA HA, whereas only one SF protein biomarker protein was different at 3 months. Larger studies are needed to identify which SF biomarkers will predict which individual OA patients will receive the greatest benefit from IA therapeutics.

3.
J Pancreat Cancer ; 7(1): 8-19, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33786412

RESUMO

Purpose: GI-4000, a series of recombinant yeast expressing four different mutated RAS proteins, was evaluated in subjects with resected ras-mutated pancreas cancer. Methods: Subjects (n = 176) received GI-4000 or placebo plus gemcitabine. Subjects' tumors were genotyped to identify which matched GI-4000 product to administer. Immune responses were measured by interferon-γ (IFNγ) ELISpot assay and by regulatory T cell (Treg) frequencies on treatment. Pretreatment plasma was retrospectively analyzed by matrix-assisted laser desorption/ionization-time-of-flight (MALDI-ToF) mass spectrometry for proteomic signatures predictive of GI-4000 responsiveness. Results: GI-4000 was well tolerated, with comparable safety findings between treatment groups. The GI-4000 group showed a similar pattern of median recurrence-free and overall survival (OS) compared with placebo. For the prospectively defined and stratified R1 resection subgroup, there was a trend in 1 year OS (72% vs. 56%), an improvement in OS (523.5 vs. 443.5 days [hazard ratio (HR) = 1.06 [confidence interval (CI): 0.53-2.13], p = 0.872), and increased frequency of immune responders (40% vs. 8%; p = 0.062) for GI-4000 versus placebo and a 159-day improvement in OS for R1 GI-4000 immune responders versus placebo (p = 0.810). For R0 resection subjects, no increases in IFNγ responses in GI-4000-treated subjects were observed. A higher frequency of R0/R1 subjects with a reduction in Tregs (CD4+/CD45RA+/Foxp3low) was observed in GI-4000-treated subjects versus placebo (p = 0.033). A proteomic signature was identified that predicted response to GI-4000/gemcitabine regardless of resection status. Conclusion: These results justify continued investigation of GI-4000 in studies stratified for likely responders or in combination with immune check-point inhibitors or other immunomodulators, which may provide optimal reactivation of antitumor immunity. ClinicalTrials.gov Number: NCT00300950.

4.
Cell Transplant ; 29: 963689720923578, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32662685

RESUMO

Absence or reduced frequency of human regulatory T cells (Tregs) can limit the control of inflammatory responses, autoimmunity, and the success of transplant engraftment. Clinical studies indicate that use of Tregs as immunotherapeutics would require billions of cells per dose. The Quantum® Cell Expansion System (Quantum system) is a hollow-fiber bioreactor that has previously been used to grow billions of functional T cells in a short timeframe, 8-9 d. Here we evaluated expansion of selected Tregs in the Quantum system using a soluble activator to compare the effects of automated perfusion with manual diffusion-based culture in flasks. Treg CD4+CD25+ cells from three healthy donors, isolated via column-free immunomagnetic negative/positive selection, were grown under static conditions and subsequently seeded into Quantum system bioreactors and into T225 control flasks in an identical culture volume of PRIME-XV XSFM medium with interleukin-2, for a 9-d expansion using a soluble anti-CD3/CD28/CD2 monoclonal antibody activator complex. Treg harvests from three parallel expansions produced a mean of 3.95 × 108 (range 1.92 × 108 to 5.58 × 108) Tregs in flasks (mean viability 71.3%) versus 7.00 × 109 (range 3.57 × 109 to 13.00 × 109) Tregs in the Quantum system (mean viability 91.8%), demonstrating a mean 17.7-fold increase in Treg yield for the Quantum system over that obtained in flasks. The two culture processes gave rise to cells with a memory Treg CD4+CD25+FoxP3+CD45RO+ phenotype of 93.7% for flasks versus 97.7% for the Quantum system. Tregs from the Quantum system demonstrated an 8-fold greater interleukin-10 stimulation index than cells from flask culture following restimulation. Quantum system-expanded Tregs proliferated, maintained their antigenic phenotype, and suppressed effector immune cells after cryopreservation. We conclude that an automated perfusion bioreactor can support the scale-up expansion of functional Tregs more efficiently than diffusion-based flask culture.


Assuntos
Imunoterapia/métodos , Linfócitos T Reguladores/imunologia , Humanos , Perfusão , Fenótipo
5.
Mater Sci Eng C Mater Biol Appl ; 96: 77-85, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30606590

RESUMO

The addition of a coating reagent to promote cell adherence is necessary to prepare the membrane surface of the Quantum® Cell Expansion System hollow-fiber bioreactor for the culture of mesenchymal stem cells. In this study, the efficacy of 8 potential coating reagents has been compared in terms of the doubling times of their cell populations, cell morphology, characterization via flow cytometry, and capacity for trilineage differentiation. Human fibronectin (FN), pooled human cryoprecipitate (CPPT), and recombinant human vitronectin (VN) were successful as coating reagents, and each product has advantages in different cell culture contexts. Mesenchymal stem cells harvested from Quantum cultured with each of these 3 compounds as coating reagents all met International Society for Cellular Therapy standards for plastic adherence, surface marker expression, and successful trilineage differentiation. No significant differences were observed among the doubling times from Quantum harvests using FN, CPPT, or VN as coating reagents (P = 0.31). Coating with gelatin, human serum albumin, collagen I, poly­l­lysine, and poly­d­lysine resulted in significantly lower harvest yield; these agents are not recommended for use as coating reagents in the Quantum system.


Assuntos
Reatores Biológicos , Técnicas de Cultura de Células/métodos , Materiais Revestidos Biocompatíveis/química , Membranas Artificiais , Células-Tronco Mesenquimais/metabolismo , Técnicas de Cultura de Células/instrumentação , Materiais Revestidos Biocompatíveis/análise , Humanos , Células-Tronco Mesenquimais/citologia
6.
Curr Stem Cell Rep ; 4(1): 46-51, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29600161

RESUMO

PURPOSE OF REVIEW: Recent developments in regenerative medicine have precipitated the need to expand gene-modified human T cells to numbers that exceed the capacity of well-plate-based, and flask-based processes. This review discusses the changes in process development that are needed to meet the cell expansion requirements by utilizing hollow-fiber bioreactors. Maintenance of cell proliferation over long periods can become limited by unfilled demands for nutrients and oxygen and by the accumulation of waste products in the local environment. RECENT FINDINGS: Perfusion feeding, improved gas exchange, and the efficient removal of lactate can increase the yield of T cells from an average of 10.8E +09 to more than 28E +09 in only 10 days. SUMMARY: Aggressively feeding cells and actively keeping cells in the bioreactor improves gas exchange and metabolite management over semi-static methods. The ability to remove the environmental constraints that can limit cell expansion by using a two-chamber hollow-fiber bioreactor will be discussed.

7.
J Immunother ; 41(3): 141-150, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29528991

RESUMO

We are developing whole, heat-killed, recombinant Saccharomyces cerevisiae yeast, engineered to encode target proteins, which stimulate immune responses against malignant cells expressing those targets. This phase 1 trial, enrolling patients with advanced colorectal or pancreas cancer, was designed to evaluate safety, immunogenicity, response, and overall survival of ascending doses of the GI-4000 series of products, which express 3 different forms of mutated Ras proteins. The study enrolled 33 heavily pretreated subjects (14 with pancreas and 19 with colorectal cancer), whose tumors were genotyped before enrollment to identify the specific ras mutation and thereby to identify which GI-4000 product to administer. No dose limiting toxicities were observed and no subject discontinued treatment due to a GI-4000 related adverse event (AE). The majority of AEs and all fatal events were due to underlying disease progression and AE frequencies were not significantly different among dose groups. GI-4000 was immunogenic, as Ras mutation-specific immune responses were detected on treatment in ∼60% of subjects. No objective tumor responses were observed but based on imaging, clinical status and/or biochemical markers, stable disease was observed in 6 subjects (18%) on day 29, while 1 subject had stable disease at days 57 and 85 follow-up visits. The median overall survival was 3.3 months (95% confidence interval, 2.3-5.3 mo), and 5 subjects survived past the 48-week follow-up period. No significant dose-dependent trends for survival were observed. This first clinical trial in humans with GI-4000 demonstrated a favorable safety profile and immunogenicity in the majority of subjects.


Assuntos
Terapia Biológica , Expressão Gênica , Mutação , Neoplasias/genética , Neoplasias/terapia , Saccharomyces cerevisiae/genética , Proteínas ras/genética , Adulto , Idoso , Terapia Biológica/métodos , Biomarcadores Tumorais , Ativação do Complemento , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imunidade , Masculino , Pessoa de Meia-Idade , Neoplasias/imunologia , Neoplasias/patologia , Saccharomyces cerevisiae/imunologia
8.
Vaccine ; 32(39): 4925-31, 2014 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-25045824

RESUMO

BACKGROUND: GS-4774 is a recombinant, heat-killed, yeast-based immunotherapy engineered to express hepatitis B virus (HBV)-specific antigens. GS-4774 is being developed as a therapeutic vaccine for chronic HBV infection. The aim of this study was to assess the safety, tolerability and immunogenicity of GS-4774 in healthy subjects. DESIGN: This was a randomized, open-label, dose-ascending study. Subjects were allocated to one of three dose groups (n=20 per group) to receive 10, 40 or 80 yeast units (YU; 1YU=10(7) yeast) of GS-4774 in two immunization regimens (five subcutaneous injections at weekly intervals with one monthly booster or three subcutaneous injections at monthly intervals). T-cell-mediated responses were determined by interferon (IFN)-γ enzyme-linked immunospot (ELISpot) assay and lymphocyte-proliferation assay (LPA). RESULTS: Adverse events were reported by 39 of 60 (65%) subjects; all were mild or moderate and none was serious. Adverse events occurred most frequently in the highest dose group, 80YU, and the number of individual events was higher after weekly immunization than monthly. The most common adverse events were injection-site reactions. Most (88%) subjects responded to GS-4774 by at least one of the T-cell assays. Following immunization with GS-4774, IFN-γ-producing T-cells specific for HBV antigens were detectable in 30 (51%) subjects. The ELISpot response was observed at all doses, with the highest frequency of responders occurring at the highest dose (10YU: 45%; 40YU: 35%; 80YU: 74%). Proliferative responses to HBV recombinant antigens were observed in 90% subjects; responses were mainly independent of GS-4774 dose and immunization regimen. CONCLUSIONS: GS-4774 was safe and well-tolerated in healthy subjects with injection-site reactions being the most frequently reported adverse events. With both weekly and monthly regimens, GS-4774 provided HBV-specific immune responses at all doses evaluated. Further evaluation of GS-4774 is ongoing in patients with chronic HBV infection. CLINICAL TRIAL REGISTRY: Clinicaltrials.gov (NCT01779505).


Assuntos
Vacinas contra Hepatite B/uso terapêutico , Hepatite B Crônica/terapia , Imunização Secundária , Adulto , Idoso , Feminino , Voluntários Saudáveis , Anticorpos Anti-Hepatite B/sangue , Vacinas contra Hepatite B/efeitos adversos , Vacinas contra Hepatite B/imunologia , Humanos , Imunidade Celular , Interferon gama/imunologia , Masculino , Pessoa de Meia-Idade , Saccharomyces cerevisiae , Linfócitos T/imunologia , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico , Adulto Jovem
9.
Clin Lung Cancer ; 15(6): 405-10, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25044103

RESUMO

INTRODUCTION: Patients with early-stage lung cancer have a high risk of recurrence despite multimodality therapy. KRAS-mutant lung adenocarcinomas are the largest genetically defined subgroup, representing 25% of patients. GI-4000 is a heat-killed recombinant Saccharomyces cerevisiae yeast-derived vaccine expressing mutant KRAS proteins. The present phase II study assessed the feasibility, immunogenicity, and safety of the GI-4000 vaccine in patients with early-stage, KRAS-mutant lung cancer. MATERIALS AND METHODS: Patients with stage I-III KRAS-mutant lung cancer who completed curative therapy were enrolled. The patients received the genotype matched GI-4000 vaccine for ≤ 3 years or until intolerance, disease recurrence, or death. The KRAS antigen T-cell response was assessed using the interferon-gamma enzyme-linked immunospot assay in peripheral blood mononuclear cells. The study was powered to detect an immune response in ≥ 25% of patients. RESULTS: A total of 24 patients were enrolled over 28 months. No vaccine-related serious adverse events occurred. One patient withdrew consent because of pain at the injection site. The study met its primary endpoint, with 50% of patients developing an immune response to mutant KRAS. The median number of vaccinations received was 15 (range, 1-19). Ten patients experienced disease recurrence, and 6 died. Compared with the genotypically matched historical controls, the recurrence rates were equivalent but overall survival showed a favorable trend. CONCLUSION: GI-4000 was well tolerated and immunogenic when used as consolidation therapy in patients with stage I-III KRAS-mutant lung cancer. The patterns of recurrence and death observed in the present study can be used to design a randomized study of GI-4000 with overall survival as the primary endpoint.


Assuntos
Adenocarcinoma/terapia , Vacinas Anticâncer/uso terapêutico , Neoplasias Pulmonares/terapia , Proteínas ras/uso terapêutico , Adenocarcinoma/genética , Adenocarcinoma/patologia , Idoso , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Células Cultivadas , Quimioterapia de Consolidação , Feminino , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Mutação/genética , Estadiamento de Neoplasias , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/imunologia , Linfócitos T/imunologia , Resultado do Tratamento , Proteínas ras/efeitos adversos , Proteínas ras/genética , Proteínas ras/imunologia
10.
PLoS One ; 9(7): e101904, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25051027

RESUMO

Chronic hepatitis B infection (CHB) is characterized by sub-optimal T cell responses to viral antigens. A therapeutic vaccine capable of restoring these immune responses could potentially improve HBsAg seroconversion rates in the setting of direct acting antiviral therapies. A yeast-based immunotherapy (Tarmogen) platform was used to make a vaccine candidate expressing hepatitis B virus (HBV) X, surface (S), and Core antigens (X-S-Core). Murine and human immunogenicity models were used to evaluate the type and magnitude of HBV-Ag specific T cell responses elicited by the vaccine. C57BL/6J, BALB/c, and HLA-A*0201 transgenic mice immunized with yeast expressing X-S-Core showed T cell responses to X, S and Core when evaluated by lymphocyte proliferation assay, ELISpot, intracellular cytokine staining (ICS), or tumor challenge assays. Both CD4+ and CD8+ T cell responses were observed. Human T cells transduced with HBc18-27 and HBs183-91 specific T cell receptors (TCRs) produced interferon gamma (IFNγ following incubation with X-S-Core-pulsed dendritic cells (DCs). Furthermore, stimulation of peripheral blood mononuclear cells (PBMCs) isolated from CHB patients or from HBV vaccine recipients with autologous DCs pulsed with X-S-Core or a related product (S-Core) resulted in pronounced expansions of HBV Ag-specific T cells possessing a cytolytic phenotype. These data indicate that X-S-Core-expressing yeast elicit functional adaptive immune responses and supports the ongoing evaluation of this therapeutic vaccine in patients with CHB to enhance the induction of HBV-specific T cell responses.


Assuntos
Vírus da Hepatite B/imunologia , Hepatite B Crônica/prevenção & controle , Linfócitos T/imunologia , Vacinação , Animais , Proliferação de Células , Células Cultivadas , Apresentação Cruzada , Células Dendríticas/imunologia , Células Dendríticas/virologia , Feminino , Vacinas contra Hepatite B/imunologia , Hepatite B Crônica/imunologia , Hepatite B Crônica/virologia , Humanos , Interferon gama/metabolismo , Interleucina-2/metabolismo , Neoplasias Hepáticas/virologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Saccharomyces cerevisiae/genética , Linfócitos T/virologia , Transativadores/genética , Transativadores/imunologia , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Proteínas Virais Reguladoras e Acessórias
11.
Vaccine ; 22(19): 2396-405, 2004 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-15193401

RESUMO

We have developed a vaccine delivery system based on the non-ionic block copolymer, Pluronic F127 (F127), combined with selected immunomodulators. F127-based matrices are characterized by a phenomenon known as reverse thermogelation, whereby the formulation undergoes a phase transition from liquid to gel upon reaching physiological temperatures. Protein antigens (tetanus toxoid (TT), diphtheria toxoid (DT) and anthrax recombinant protective antigen (rPA)) were formulated with F127 in combination with CpG motifs or chitosan, as examples of immunomodulators, and were compared to more traditional adjuvants in mice. IgG antibody responses were significantly enhanced by the F127/CpG and F127/chitosan combinations compared to antigens mixed with CpGs or chitosan alone. In addition, the responses were significantly greater than those elicited by aluminum salts. Furthermore, the functional activity of these antibodies was demonstrated using either in vivo tetanus toxin challenge or an anthrax lethal toxin neutralization assay. These studies suggest that a block-copolymer approach could enhance the delivery of a variety of clinically useful antigens in vaccination schemes.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Quitina/análogos & derivados , Quitina/administração & dosagem , Sistemas de Liberação de Medicamentos , Vacinas/administração & dosagem , Animais , Anticorpos Antibacterianos/biossíntese , Quitina/imunologia , Quitosana , Antitoxina Diftérica/biossíntese , Toxoide Diftérico/administração & dosagem , Toxoide Diftérico/imunologia , Sinergismo Farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Poloxâmero/administração & dosagem , Antitoxina Tetânica/biossíntese , Toxoide Tetânico/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA