Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cytotherapy ; 22(11): 642-652, 2020 11.
Article in English | MEDLINE | ID: mdl-32747299

ABSTRACT

Background aims: E3L is an immediate-early protein of vaccinia virus (VV) that is detected within 0.5 h of infection, potentially before the many immune evasion genes of vaccinia can exert their protective effects. E3L is highly conserved among orthopoxviruses and hence could provide important protective T-cell epitopes that should be retained in any subunit or attenuated vaccine. We have therefore evaluated the immunogenicity of E3L in healthy VV-vaccinated donors. Methods: Peripheral blood mononuclear cells from healthy volunteers (n = 13) who had previously received a smallpox vaccine (Dryvax) were activated and expanded using overlapping E3L peptides and their function, specificity and antiviral activity was analyzed. E3L-specific T cells were expanded from 7 of 12 (58.3%) vaccinated healthy donors. Twenty-five percent of these produced CD8+ T-cell responses and 87.5% produced CD4+ T cells. We identified epitopes restricted by HLA-B35 and HLA-DR15. Results: E3L-specific T cells killed peptide-loaded target cells as well as vaccinia-infected cells, but only CD8+ T cells could prevent the spread of infectious virus in virus inhibition assays. The epitopes recognized by E3L-specific T cells were shared with monkeypox, and although there was a single amino acid change in the variola epitope homolog, it was recognized by vaccinia-specific T-cells. Conclusions: It might be important to include E3L in any deletion mutant or subunit vaccine and E3L could provide a useful antigen to monitor protective immunity in humans.


Subject(s)
Antigens, Viral/immunology , Smallpox Vaccine/immunology , Smallpox/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/genetics , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Peptides/chemistry , Peptides/immunology , Smallpox/prevention & control , Tissue Donors , Vaccination , Vaccinia virus/genetics , Vaccinia virus/immunology , Virion/immunology , Virus Replication/physiology
2.
Mol Ther ; 23(3): 602-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25531693

ABSTRACT

Pexa-Vec (pexastimogene devacirepvec, JX-594) is an oncolytic and immunotherapeutic vaccinia virus designed to destroy cancer cells through viral lysis and induction of granulocyte-macrophage colony-stimulating factor (GM-CSF)-driven tumor-specific immunity. Pexa-Vec has undergone phase 1 and 2 testing alone and in combination with other therapies in adult patients, via both intratumoral and intravenous administration routes. We sought to determine the safety of intratumoral administration in pediatric patients. In a dose-escalation study using either 10(6) or 10(7) plaque-forming units per kilogram, we performed one-time injections in up to three tumor sites in five pediatric patients and two injections in one patient. Ages at study entry ranged from 4 to 21 years, and their cancer diagnoses included neuroblastoma, hepatocellular carcinoma, and Ewing sarcoma. All toxicities were ≤ grade 3. The most common side effects were sinus fever and sinus tachycardia. All three patients at the higher dose developed asymptomatic grade 1 treatment-related skin pustules that resolved within 3-4 weeks. One patient showed imaging evidence suggestive of antitumor biological activity. The two patients tested for cellular immunoreactivity to vaccinia antigens showed strong responses. Overall, our study suggests Pexa-Vec is safe to administer to pediatric patients by intratumoral administration and could be studied further in this patient population.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Cancer Vaccines/immunology , Gamma Rays , Immunotherapy/methods , Oncolytic Virotherapy/methods , Vaccinia virus/immunology , Adolescent , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cancer Vaccines/administration & dosage , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Child , Child, Preschool , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Humans , Injections, Intralesional , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Neoplasm Staging , Neuroblastoma/immunology , Neuroblastoma/pathology , Neuroblastoma/therapy , Sarcoma, Ewing/immunology , Sarcoma, Ewing/pathology , Sarcoma, Ewing/therapy , Vaccination , Vaccinia virus/genetics , Young Adult
3.
Cytotherapy ; 14(9): 1131-43, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22900959

ABSTRACT

BACKGROUND AIMS: Interest in natural killer (NK) cell-based immunotherapy has resurged since new protocols for the purification and expansion of large numbers of clinical-grade cells have become available. METHODS: We have successfully adapted a previously described NK expansion method that uses K562 cells expressing interleukin (IL)-15 and 4-1 BB Ligand (BBL) (K562-mb15-41BBL) to grow NK cells in novel gas-permeable static cell culture flasks (G-Rex). RESULTS: Using this system we produced up to 19 × 10(9) functional NK cells from unseparated apheresis products, starting with 15 × 10(7) CD3(-) CD56 (+) NK cells, within 8-10 days of culture. The G-Rex yielded a higher fold expansion of NK cells than conventional gas-permeable bags and required no cell manipulation or feeding during the culture period. We also showed that K562-mb15-41BBL cells up-regulated surface HLA class I antigen expression upon stimulation with the supernatants from NK cultures and stimulated alloreactive CD8 (+) T cells within the NK cultures. However, these CD3 (+) T cells could be removed successfully using the CliniMACS system. We describe our optimized NK cell cryopreservation method and show that the NK cells are viable and functional even after 12 months of cryopreservation. CONCLUSIONS: We have successfully developed a static culture protocol for large-scale expansion of NK cells in the gas permeable G-Rex system under good manufacturing practice (GMP) conditions. This strategy is currently being used to produce NK cells for cancer immunotherapy.


Subject(s)
Immunotherapy, Adoptive , K562 Cells/cytology , Killer Cells, Natural/cytology , T-Lymphocytes , 4-1BB Ligand/metabolism , Blood Component Removal , Cell Culture Techniques , Cell Survival , Cryopreservation , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
Hum Mol Genet ; 20(R1): R93-9, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21415041

ABSTRACT

Adoptive immunotherapy is an appealing approach to cancer treatment, with the potential for more precise targeting and reduced toxicity. While early clinical trial data using adoptive T cells against post-transplant virus-associated hematologic malignancies, lymphoma and melanoma have been promising, treating other solid tumors has proven to be more challenging. Adoptive lymphocytes have been genetically modified in many ways to improve activity and circumvent tumor evasion, including transfer of transgenic T-cell receptors and chimeric antigen receptors to redirect T cell and natural killer cell antigen specificity. Gene transfer may also allow expression of homeostatic cytokines or their receptors to overcome the lack of stimulatory signals or expression of dominant-negative receptors for inhibitory cytokines to compensate for an immunosuppressive tumor milieu. In addition, suicide genes can install a 'safety switch' on adoptively transferred cells to allow ablation if necessary. Although further refinement and validation are necessary, these genetic modification strategies offer hope for significant improvements in cancer immunotherapy.


Subject(s)
Gene Transfer Techniques , Immunotherapy, Adoptive/methods , Neoplasms/immunology , Neoplasms/therapy , Animals , Humans , Melanoma/immunology , Melanoma/metabolism , Melanoma/therapy , Neoplasms/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
5.
J Immunother ; 33(3): 305-15, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20445351

ABSTRACT

The clinical manufacture of antigen-specific cytotoxic T lymphocytes (CTLs) for adoptive immunotherapy is limited by the complexity and time required to produce large numbers with the desired function and specificity. The culture conditions required are rigorous, and in some cases only achieved in 2-cm wells in which cell growth is limited by gas exchange, nutrients, and waste accumulation. Bioreactors developed to overcome these issues tend to be complex, expensive, and not always conducive to CTL growth. We observed that antigen-specific CTLs undergo 7 to 10 divisions poststimulation. However, the expected CTL numbers were achieved only in the first week of culture. By recreating the culture conditions present during this first week-low frequency of antigen-specific T cells and high frequency of feeder cells-we were able to increase CTL expansion to expected levels that could be sustained for several weeks without affecting phenotype or function. However, the number of 24-well plates needed was excessive and cultures required frequent media changes, increasing complexity and manufacturing costs. Therefore, we evaluated novel gas-permeable culture devices (G-Rex) with a silicone membrane at the base allowing gas exchange to occur uninhibited by the depth of the medium above. This system effectively supports the expansion of CTL and actually increases output by up to 20-fold while decreasing the required technician time. Importantly, this amplified cell expansion is not because of more cell divisions but because of reduced cell death. This bioprocess optimization increased T-cell output while decreasing the complexity and cost of CTL manufacture, making cell therapy more accessible.


Subject(s)
Antigens/immunology , Cell Proliferation , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Line, Tumor , Humans , Immunotherapy, Adoptive/methods , K562 Cells , Lymphocyte Count , Reproducibility of Results , T-Lymphocytes, Cytotoxic/transplantation , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...