Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Ann Pharmacother ; 57(10): 1147-1153, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-36688289

RESUMEN

BACKGROUND: Recent evidence suggests tenecteplase at an intravenous dose of 0.25 mg/kg is as safe and efficacious as intravenous alteplase standard dose and demonstrates a more favorable pharmacokinetic profile for treatment of acute ischemic stroke. OBJECTIVE: The purpose was to compare the safety and efficacy of alteplase versus tenecteplase for the treatment of acute ischemic stroke at a large community hospital health system following conversion in the preferred formulary thrombolytic. METHODS: Prior to converting, medication safety and operationalization analyses were conducted. A multicenter, retrospective medical record review was performed for patients who received alteplase 6 months prior to formulary thrombolytic conversion and for tenecteplase 6 months post-conversion for the treatment of acute ischemic stroke. Primary outcomes included the rate of symptomatic intracranial and extracranial hemorrhage complications. Secondary outcomes included door-to-needle time, reduction in National Institute Health Stroke Scale at 24 hours and at discharge, order-to-administration time, and thrombolytic errors. The rates of hemorrhage were compared using binomial regression. RESULTS: Of the 287 patients reviewed, 115 received alteplase and 172 received tenecteplase. Symptomatic intracranial hemorrhagic complications occurred in 1 patient (1%) who received alteplase compared with 3 patients (2%) who received tenecteplase (P = 0.9). There was no statistical difference in rates of symptomatic intracranial or extracranial hemorrhagic complications. CONCLUSION AND RELEVANCE: Conversion from alteplase to tenecteplase can be safely and effectively achieved at a large community hospital health system with differing levels of stroke certification. There were also additional cost savings and practical advantages including workflow benefits.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Activador de Tejido Plasminógeno/efectos adversos , Tenecteplasa , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Hospitales Comunitarios , Estudios Retrospectivos , Isquemia Encefálica/tratamiento farmacológico , Fibrinolíticos/efectos adversos , Accidente Cerebrovascular/tratamiento farmacológico , Hemorragias Intracraneales/tratamiento farmacológico , Resultado del Tratamiento
2.
Nature ; 490(7420): 412-6, 2012 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-23051752

RESUMEN

Adoptive cell transfer therapies (ACTs) with cytotoxic T cells that target melanocytic antigens can achieve remissions in patients with metastatic melanomas, but tumours frequently relapse. Hypotheses explaining the acquired resistance to ACTs include the selection of antigen-deficient tumour cell variants and the induction of T-cell tolerance. However, the lack of appropriate experimental melanoma models has so far impeded clear insights into the underlying mechanisms. Here we establish an effective ACT protocol in a genetically engineered mouse melanoma model that recapitulates tumour regression, remission and relapse as seen in patients. We report the unexpected observation that melanomas acquire ACT resistance through an inflammation-induced reversible loss of melanocytic antigens. In serial transplantation experiments, melanoma cells switch between a differentiated and a dedifferentiated phenotype in response to T-cell-driven inflammatory stimuli. We identified the proinflammatory cytokine tumour necrosis factor (TNF)-α as a crucial factor that directly caused reversible dedifferentiation of mouse and human melanoma cells. Tumour cells exposed to TNF-α were poorly recognized by T cells specific for melanocytic antigens, whereas recognition by T cells specific for non-melanocytic antigens was unaffected or even increased. Our results demonstrate that the phenotypic plasticity of melanoma cells in an inflammatory microenvironment contributes to tumour relapse after initially successful T-cell immunotherapy. On the basis of our work, we propose that future ACT protocols should simultaneously target melanocytic and non-melanocytic antigens to ensure broad recognition of both differentiated and dedifferentiated melanoma cells, and include strategies to sustain T-cell effector functions by blocking immune-inhibitory mechanisms in the tumour microenvironment.


Asunto(s)
Desdiferenciación Celular , Inmunoterapia , Inflamación/patología , Melanoma/patología , Melanoma/terapia , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/trasplante , Traslado Adoptivo , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Melanoma/inmunología , Melanoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Microambiente Tumoral/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/farmacología , Antígeno gp100 del Melanoma/metabolismo
3.
Front Immunol ; 14: 1286688, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38077312

RESUMEN

Here, we describe the identification of two T-cell receptors (TRs) containing TRDV genes in their TRA chains, the first one in human and the second one in mouse. First, using 5'RACE on a mixed lymphocyte-tumor cell culture (MLTC), we identified TRDV1 5'-untranslated region (UTR) and complete coding sequence rearranged productively to TRAJ24. Single-cell TR RNA sequencing (RNA-seq) of the MLTC, conducted to identify additional clonotypes, revealed that the analysis software detected the hybrid TRDV-TRAJ TRA (TRA) chain but excluded it from the final results. In a separate project, we performed TR sequencing of tumor-infiltrating lymphocytes (TILs) in a murine tumor model. Here, the predominant clonotype contained a TRA chain with a TRDV2-2-TRAJ49 rearrangement. Again, the hybrid TRA chain was not reported in the final results. Transfection of both TR cDNAs resulted in cell surface localization of TR together with CD3, suggesting a productive protein in both cases. Tumor recognition of the Homo sapiens (Homsap) TRDV1-containing TR could be demonstrated by IFN Gamma ELISA ELISpot kit, whereas the Mus musculus (Musmus) TR did not recognize a tumor-derived cell line. To determine whether the TRDV-containing TRA chains we detected were rare events or whether TRDV genes are commonly incorporated into TRA chains, we queried the NCBI Sequence Read Archive for TR single-cell RNA-seq data and analyzed 21 human and 23 murine datasets. We found that especially Homsap TRDV1, Musmus TRDV1, and to some extent Musmus TRDV2-2 are more commonly incorporated into TRA chains than several TRAV genes, making those TRDV genes a relevant contribution to TRA diversity. TRDV-containing TRA chains are currently excluded from the final results of V-(D)-J dataset analyses with the CellRanger software. We provide a work-around to avoid exclusion of those hybrid TRA chains from the final analysis results.


Asunto(s)
Genes Codificadores de la Cadena delta de los Receptores de Linfocito T , Receptores de Antígenos de Linfocitos T , Humanos , Animales , Ratones , Receptores de Antígenos de Linfocitos T/genética , ADN Complementario
4.
Front Immunol ; 14: 1245559, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37849763

RESUMEN

Intorduction: Chondroitin sulfate proteoglycan 4 (CSPG4), also known as high molecular weight-melanoma associated antigen, is expressed in melanoma but also other tumor entities and constitutes an attractive target for immunotherapeutic approaches. While recent preclinical reports focused on anti-CSPG4 chimeric antigen receptors (CAR), we here explore T-cell receptor (TCR)-based approaches targeting CSPG4. Methods: The TCRs of two CSPG4-reactive T-cell clones (11C/73 and 2C/165) restricted by the highly prevalent HLA-C*07:01 allele were isolated and the respective αßTCR pairs were retrovirally expressed in CRISPR/Cas9-edited TCR-knockout T cells for functional testing. We also combined alpha and beta TCR chains derived from 11C/73 and 2C/165 in a cross-over fashion to assess for hemichain dominance. CSPG4+ melanoma, glioblastoma and lung cancer cell lines were identified and, if negative, retrovirally transduced with HLA-C*07:01. Results: Functional tests confirmed specific recognition of CSPG4+HLA-C*07:01+ target cells by the αßTCR retrieved from the parental T-cell clones and in part also by the cross-over TCR construct 2Cα-11Cß. Despite high surface expression, the 11Cα-2Cß combination, however, was not functional. Discussion: Collectively, 11C/73- and 2C/165-expressing T cells specifically and efficiently recognized CSPG4+HLA-C*07:01+ cancer cells which warrants further preclinical and clinical evaluation of these TCRs.


Asunto(s)
Antígenos HLA-C , Melanoma , Humanos , Antígenos HLA-C/genética , Receptores de Antígenos de Linfocitos T , Linfocitos T , Proteínas de la Membrana , Proteoglicanos Tipo Condroitín Sulfato
5.
J Med Case Rep ; 14(1): 135, 2020 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-32859260

RESUMEN

BACKGROUND: Rituximab is a well-established component of treatment regimens for B-cell non-Hodgkin lymphoma. Rituximab binds the CD20 antigen on the surface of B lymphocytes, causing an enhanced clearance of malignant and benign B cells. Thus, rituximab leads to depletion of normal B lymphocytes as well, which can cause substantial immunodeficiency. Ibrutinib inhibits the Bruton tyrosine kinase and thereby B-cell activity. It is used for the treatment of different B-lymphocyte malignancies, such as mantle cell lymphoma. Recently, the combination of both drugs has been tested in various clinical scenarios. CASE PRESENTATION: We present a case of disseminated enterovirus infection resulting from combined rituximab and ibrutinib maintenance treatment in a 57-year-old Caucasian patient. with mantle cell lymphoma. Initially presenting with myositis symptoms, further diagnostic investigation revealed myocarditis, enteritis, myeloencephalitis, and hepatitis. These organ manifestations led to potentially life-threatening complications such as rhabdomyolysis, delirium, and heart rhythm disturbances. After treatment with high-dose intravenous immunoglobulins, virus clearance was achieved and organ functions could be restored. CONCLUSIONS: This case emphasizes the risk of combined therapy with rituximab/ibrutinib for severe immune-related side effects with the necessity of continuous patient monitoring. High-dose intravenous therapy should be considered as treatment for severe enterovirus infection. In severe enterovirus infections, we recommend subtyping for the development of efficient preventive and therapeutic strategies.


Asunto(s)
Infecciones por Enterovirus , Linfoma de Células del Manto , Adenina/análogos & derivados , Adulto , Antígenos CD20 , Infecciones por Enterovirus/tratamiento farmacológico , Humanos , Linfoma de Células del Manto/tratamiento farmacológico , Persona de Mediana Edad , Piperidinas , Rituximab/efectos adversos
6.
PLoS One ; 15(9): e0238875, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32903281

RESUMEN

To facilitate preclinical testing of T-cell receptors (TCRs) derived from tumor-reactive T-cell clones it is necessary to develop convenient and rapid cloning strategies for the generation of TCR expression constructs. Herein, we describe a pDONR™221 vector backbone allowing to generate Gateway™ compatible entry clones encoding optimized bicistronic αßTCR constructs. It harbors P2A-linked TCR constant regions and head-to-head-oriented recognition sites of the Type IIS restriction enzymes BsmBI and BsaI for seamless cloning of the TCRα and TCRß V(D)J regions, respectively. Additional well-established TCR optimizations were incorporated to enhance TCR functionality. This included replacing of the human αßTCR constant regions with their codon-optimized murine counterparts for chimerization, addition of a second interchain disulfide bond and arrangement of the TCR chains in the order ß-P2A-α. We exemplified the utility of our vector backbone by cloning and functional testing of three melanoma-reactive TCRs in primary human T cells.


Asunto(s)
Clonación Molecular/métodos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Retroviridae/genética , Linfocitos T/citología , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Técnicas de Cocultivo , Vectores Genéticos/genética , Células HEK293 , Humanos , Linfocitos T/inmunología , Linfocitos T/virología , Transducción Genética , Recombinación V(D)J
7.
Exp Hematol ; 36(4): 451-63, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18261837

RESUMEN

OBJECTIVE: Current in vitro techniques for isolating leukemia-reactive cytotoxic T lymphocytes (CTLs) from healthy donors are of relatively low efficiency and yield responder populations with unknown biological significance. This study aimed at the development of a more reliable approach, allowing generation and expansion of acute myeloid leukemia (AML)-reactive CTLs using primary in vitro stimulation. MATERIALS AND METHODS: We established allogeneic mini-mixed lymphocyte-leukemia cultures (mini-MLLCs) by stimulating donor CD8(+) T cells with human leukocyte antigen (HLA) class I-matched AML blasts in microtiter plates. Before culture, CD8(+) T cells were separated into CD62L((high)+) and CD62L((low)+/neg) subsets enriched for naive/central memory and effector memory cells, respectively. RESULTS: In eight different related and unrelated donor/AML pairs, numerous CTL populations were isolated that specifically lysed myeloid leukemias in association with various HLA-A, -B, or -C alleles. These CTLs expressed T-cell receptors of single Vbeta-chain families, indicating their clonal origin. The majority of CTL clones were obtained from mini-MLLCs initiated with CD62L((high)+) cells. Using antigen-specific stimulation, multiple CTL populations were amplified to 10(8)-10(10) cells within 6 to 8 weeks. Three of four representative CTL clones were capable of completely preventing engraftment of human primary AML blasts in nonobese diabetic/severe combined immune deficient IL2Rgamma(null) mice. CONCLUSION: The mini-MLLC approach allows the efficient in vitro expansion of AML-reactive CTL clones from CD8(+)CD62L((high)+) precursors of healthy donors. These CTLs can inhibit leukemia engraftment in immunodeficient mice, suggesting their potential biological relevance.


Asunto(s)
Linfocitos T CD8-positivos/citología , Subunidad gamma Común de Receptores de Interleucina/genética , Selectina L/biosíntesis , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/prevención & control , Linfocitos T Citotóxicos/trasplante , Alelos , Animales , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Células Clonales , Pruebas Inmunológicas de Citotoxicidad , Genes MHC Clase I , Humanos , Leucemia Mieloide Aguda/sangre , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Trasplante de Neoplasias , Estándares de Referencia , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología
8.
Oncoimmunology ; 8(9): e1625688, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31428523

RESUMEN

The oncoantigen nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) induces cellular and humoral immune responses in patients with NPM-ALK-positive anaplastic large cell lymphoma (ALCL). We characterize the NPM-ALK-specific T-cell responses in a cohort of pediatric and adolescent ALCL-patients in remission without Human Leucocyte Antigen (HLA)-preselection. First, we assessed NPM-ALK-reactive T-cell responses and their HLA-class I restriction in patients by using dendritic cells (DCs) transfected with in vitro transcribed (IVT) NPM-ALK-RNA for CD8 (n = 20) or CD3 (n = 9) T-cell stimulation. NPM-ALK-specific T-cells were detected in twelve of 29 patients (nine of 20 with CD8-selected and three of nine with CD3-selected cells). Recognition of NPM-ALK was restricted by HLA-C alleles in six of eight, and by HLA-B alleles in four of eight analyzed patients. No NPM-ALK-reactivity was detected in 20 healthy individuals. Second, in order to define possible immunogenic NPM-ALK-epitope regions, DCs pulsed with pools of overlapping long NPM-ALK-peptides were used to stimulate T-cells in further 22 patients and ten controls. Responsive T-cells were detected in 15 patients and in five controls. A peptide pool located in the middle of the kinase domain induced ALK-reactive T-cells in 14 of 15 responsive patients. We could narrow to single peptides between p327-p370 of NPM-ALK in four patients. In conclusion, using IVT-RNA, 40% of NPM-ALK-positive ALCL-patients in remission had detectable NPM-ALK-specific T-cell responses which were mainly restricted by HLA-B and -C alleles. Peptide stimulation of T-cells revealed responses in almost 70% of patients and allowed describing an immunogenic region located in the ALK-kinase domain.

10.
Oncoimmunology ; 7(8): e1450127, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30221038

RESUMEN

The profound but frequently transient clinical responses to BRAFV600 inhibitor (BRAFi) treatment in melanoma emphasize the need for combinatorial therapies. Multiple clinical trials combining BRAFi and immunotherapy are under way to further enhance therapeutic responses. However, to which extent BRAFV600 inhibition may affect melanoma immunogenicity over time remains largely unknown. To support the development of an optimal treatment protocol, we studied the impact of prolonged BRAFi exposure on the recognition of melanoma cells by T cells in different patient models. We demonstrate that autologous CD8+ tumor-infiltrating lymphocytes (TILs) efficiently recognized short-term (3, 7 days) BRAFi-treated melanoma cells but were less responsive towards long-term (14, 21 days) exposed tumor cells. Those long-term BRAFi-treated melanoma cells showed a non-proliferative dedifferentiated phenotype and were less sensitive to four out of five CD8+ T cell clones, present in the preexisting TIL repertoire, of which three recognized shared antigens (Tyrosinase, Melan-A and CSPG4) and one being neoantigen-specific. Only a second neoantigen was steadily recognized independent of treatment duration. Notably, in all cases the impaired T cell activation was due to a time-dependent downregulation of their respective target antigens. Moreover, combinatorial treatment of melanoma cells with BRAFi and an inhibitor of its downstream kinase MEK had similar effects on T cell recognition. In summary, MAP kinase inhibitors (MAPKi) strongly alter the tumor antigen expression profile over time, favoring evolution of melanoma variants cross-resistant to both T cells and MAPKi. Our data suggest that simultaneous treatment with MAPKi and immunotherapy could be most effective for tumor elimination.

11.
Lung Cancer ; 58(1): 88-94, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17599645

RESUMEN

Mutations of the ras gene have been reported in 20-40% of NSCLC patients. If present, they are critical for the malignant phenotype of these tumors. Therefore, targeting them by specific vaccination is a promising therapeutic approach. In a clinical trial we screened for ras mutations in patients with NSCLC. Patients with ras-positive tumors were immunized six times intradermally with a mixture of seven peptides representing the most common ras mutations. Objectives of the study were the feasibility, efficacy and safety of the vaccination. In addition, the induction of a specific immune reaction was investigated by DTH tests, and the induction of peptide-specific T cells was tested in ex vivo IFN-gamma-ELISPOT assays. Five of 18 patients had ras mutations at codon 12. Four of these patients, all with adenocarcinomas (stage I: n=3, stage IV: n=1) entered the study. The patient with stage IV disease withdrew prematurely after the third application because of disease progression associated with pulmonary embolism. Ras-specific T cells were not detected ex vivo. However, one patient developed a positive DTH reaction after the fifth vaccination that increased after the sixth vaccination. Our results are in line with earlier trials reporting ras mutations in 20-40% of NSCLC patients. Vaccination with mutated ras peptides is feasible and well tolerated. One patient revealed a positive DTH test. An ex vivo detectable T cell response was not induced in any of the patients.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Proteínas ras , Anciano , Vacunas contra el Cáncer/efectos adversos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Codón , Terapia Combinada/efectos adversos , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Humanos , Inmunidad Celular , Inmunoterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Mutación , Estadificación de Neoplasias , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Linfocitos T/inmunología , Vacunación , Proteínas ras/genética , Proteínas ras/inmunología
12.
Oncotarget ; 8(17): 28312-28327, 2017 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-28423700

RESUMEN

T lymphocytes against tumor-specific mutated neoantigens can induce tumor regression. Also, the size of the immunogenic cancer mutanome is supposed to correlate with the clinical efficacy of checkpoint inhibition. Herein, we studied the susceptibility of tumor cell lines from lymph node metastases occurring in a melanoma patient over several years towards blood-derived, neoantigen-specific CD8+ T cells. In contrast to a cell line established during early stage III disease, all cell lines generated at later time points from stage IV metastases exhibited partial or complete loss of HLA class I expression. Whole exome and transcriptome sequencing of the four tumor lines and a germline control were applied to identify expressed somatic single nucleotide substitutions (SNS), insertions and deletions (indels). Candidate peptides encoded by these variants and predicted to bind to the patient's HLA class I alleles were synthesized and tested for recognition by autologous mixed lymphocyte-tumor cell cultures (MLTCs). Peptides from four mutated proteins, HERPUD1G161S, INSIG1S238F, MMS22LS437F and PRDM10S1050F, were recognized by MLTC responders and MLTC-derived T cell clones restricted by HLA-A*24:02 or HLA-B*15:01. Intracellular peptide processing was verified with transfectants. All four neoantigens could only be targeted on the cell line generated during early stage III disease. HLA loss variants of any kind were uniformly resistant. These findings corroborate that, although neoantigens represent attractive therapeutic targets, they also contribute to the process of cancer immunoediting as a serious limitation to specific T cell immunotherapy.


Asunto(s)
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Genes MHC Clase I , Melanoma/genética , Melanoma/inmunología , Mutación , Linfocitos T/inmunología , Linfocitos T/metabolismo , Alelos , Animales , Presentación de Antígeno , Línea Celular Tumoral , Biología Computacional/métodos , Modelos Animales de Enfermedad , Mapeo Epitopo , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación INDEL , Melanoma/patología , Ratones , Metástasis de la Neoplasia , Péptidos/química , Péptidos/genética , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Transcriptoma
13.
J Immunol Methods ; 308(1-2): 77-89, 2006 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-16336973

RESUMEN

The design and successful clinical implementation of cancer vaccines targeting the induction of T-cell mediated immunity is a rapidly evolving field that is hampered by an empirical selection of antigen and adjuvant. In particular, vaccines using defined tumor-associated peptide epitopes elicit only a restricted T-cell repertoire in a minority of patients. In this regard, vaccines comprising the whole spectrum of antigens presented by individual autologous tumors would be advantageous. In an in vitro model, we evaluated the capacity of naturally processed Epstein-Barr virus-transformed B-lymphoblastoid-cell line (LCL)-derived peptides to activate virus-specific CD8+ T cells of seropositive healthy individuals. While bulk peptides obtained by mild acid elution from LCL contained multiple T-cell epitopes, this complex mixture of peptides was poorly immunogenic, even when presented by mature dendritic cells (DC). Pretreatment of DC with proteasome inhibitors strongly enhanced the immunogenicity of single viral synthetic as well as bulk LCL peptides. This was most likely achieved by facilitating the loading of exogenous epitopes onto DC-associated HLA-class I complexes in the face of significant inter-peptide competition for such loading. Our results suggest that proteasome inhibitors may be used to increase the antigenicity of mature DC pulsed with exogenous synthetic or naturally processed peptide epitopes in vaccination trials.


Asunto(s)
Presentación de Antígeno/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Transformación Celular Viral , Células Clonales , Células Dendríticas/enzimología , Epítopos/metabolismo , Antígenos HLA/metabolismo , Herpesvirus Humano 4/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Técnicas In Vitro , Activación de Linfocitos , Prueba de Cultivo Mixto de Linfocitos , Monocitos/inmunología
14.
Cancer Res ; 76(15): 4347-58, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27261508

RESUMEN

Melanoma often recurs after a latency period of several years, presenting a T cell-edited phenotype that reflects a role for CD8(+) T cells in maintaining metastatic latency. Here, we report an investigation of a patient with multiple recurrent lesions, where poorly immunogenic melanoma phenotypes were found to evolve in the presence of autologous tumor antigen-specific CD8(+) T cells. Melanoma cells from two of three late recurrent metastases, developing within a 6-year latency period, lacked HLA class I expression. CD8(+) T cell-resistant, HLA class I-negative tumor cells became clinically apparent 1.5 and 6 years into stage IV disease. Genome profiling by SNP arrays revealed that HLA class I loss in both metastases originated from a shared chromosome 15q alteration and independently acquired focal B2M gene deletions. A third HLA class I haplotype-deficient lesion developed in year 3 of stage IV disease that acquired resistance toward dominant CD8(+) T-cell clonotypes targeting stage III tumor cells. At an early stage, melanoma cells showed a dedifferentiated c-Jun(high)/MITF(low) phenotype, possibly associated with immunosuppression, which contrasted with a c-Jun(low)/MITF(high) phenotype of T cell-edited tumor cells derived from late metastases. In summary, our work shows how tumor recurrences after long-term latency evolve toward T-cell resistance by independent genetic events, as a means for immune escape and immunotherapeutic resistance. Cancer Res; 76(15); 4347-58. ©2016 AACR.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Melanoma/genética , Proteínas de Neoplasias/inmunología , Humanos , Melanoma/patología , Metástasis de la Neoplasia
15.
J Immunol Methods ; 299(1-2): 165-75, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15914199

RESUMEN

Although the number of defined T cell epitopes of clinically relevant antigens is constantly increasing, there is still an enormous need to identify further peptides, processed from new antigens or presented by rare HLA molecules, respectively. Here we introduce a novel two-step approach for the rapid identification of T cell epitopes. It was established in the CMV infection model. From the peripheral blood of healthy donors sharing HLA-A1 according to HLA serotyping we isolated CD8+ T lymphocytes and generated dendritic cells (DCs). DCs were electroporated with CMV pp65 mRNA and tested for recognition by autologous CD8+ T lymphocytes in IFN-gamma ELISPOT assays. In all 10 CMV-seropositive donors, CMV pp65-specific CD8+ T cells were readily detectable ex vivo. In 7 of them the response was at least in part restricted by HLA-A1.1 as verified in IFN-gamma ELISPOT assays with pp65 mRNA-electroporated K562 cells stably transfected with HLA-A*0101 (K562/A*0101). In a subsequent step various 3'-deleted pp65 RNA fragments were rapidly generated by in vitro transcription of plasmid DNA-templates linearized with restriction enzymes at different sites within the pp65-coding sequence. Polyadenylated mRNA fragments were then electroporated into K562/A*0101 cells and tested for recognition by ex vivo CD8+ T cells in IFN-gamma ELISPOT assays. We thereby identified a 76 bp-long sequence as target of the HLA-A*0101-associated pp65-specific T cell response. From this region, 10 peptides predicted by current algorithms were synthesized and tested for recognition. Peptide pp65 364-373 (previously identified by a reverse immunology approach by [Hebart, H., Daginik, S., Stevanovic, S., Grigoleit, U., Dobler, A., Baur, M., Rauser, G., Sinzger, C., Jahn, G., Loeffler, J., Kanz, L., Rammensee, H. G., Einsele, H., 2002. Sensitive detection of human cytomegalovirus peptide-specific cytotoxic T-lymphocyte responses by interferon-gamma-enzyme-linked immunospot assay and flow cytometry in healthy individuals and in patients after allogeneic stem cell transplantation, Blood 99, 3830.]) was positively tested and found to be the dominant target epitope of the HLA-A1-restricted anti pp65 T cell response in all donors. We conclude that (i) the use of HLA-transfected K562 cells allows to dissect antigen-specific T cell responses to partial responses associated with defined HLA class I alleles and (ii) transfection of in vitro transcribed RNA fragments allows to identify immunogenic regions of a given antigen. The latter technique bypasses the need of prior cloning and sequencing of cDNA fragments, reduces the number of synthetic peptides to be tested and thus saves both costs and time.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Mapeo Epitopo/métodos , Epítopos de Linfocito T/análisis , ARN Mensajero/genética , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Epítopos de Linfocito T/genética , Antígeno HLA-A1/inmunología , Humanos , Péptidos/análisis , Péptidos/genética , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología
16.
Cancer Immunol Res ; 3(9): 992-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26048577

RESUMEN

The ability to use circulating peripheral blood cells and matched tumor sequencing data as a basis for neoantigen prediction has exciting possibilities for application in the personalized treatment of cancer patients. We have used a high-throughput screening approach, combining whole-exome sequence data, mRNA microarrays, and publicly available epitope prediction algorithm output to identify mutated proteins processed and displayed by patient tumors and recognized by circulating immune cells. Matched autologous melanoma cell lines and peripheral blood mononuclear cells were used to create mixed lymphocyte tumor cell cultures, resulting in an expansion of tumor-reactive T cells to use for mutated peptide screening. Five patients were investigated, three of whom had a durable complete response (CR; 15+ years) in an autologous melanoma-pulsed dendritic cell clinical trial. We identified seven mutated antigens in total that stimulated T-effector memory cells in two of the five patients. While the procedure did not result in clinically applicable neoantigens for all patients, those identified were likely important in tumor clearance, leading to durable CR. The nature of the screening process allows results to be obtained rapidly and is easily applicable to a wide variety of different tumor types.


Asunto(s)
Antígenos de Neoplasias/genética , Exoma/inmunología , Melanoma/genética , Melanoma/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Exoma/genética , Humanos , Interferón gamma/biosíntesis , Linfocitos Infiltrantes de Tumor/inmunología , Mutación
17.
J Immunol Methods ; 287(1-2): 125-36, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15099761

RESUMEN

Most assay systems for the quantification of antigen-specific T-cell responses in infectious, malignant and autoimmune disease depend on the peptide antigen format and are therefore restricted to known epitopes and their presenting HLA molecules. Here we tested in ELISPOT assays the application of in vitro-transcribed clonal mRNA as an alternative antigen format covering all potential epitopes of a given antigen. As model antigens, we chose pp65 of human cytomegalovirus (HCMV) and human tyrosinase (hTyr). Antigen-presenting cells (APC) were K562 cells stably transfected with single HLA class I alleles and autologous dendritic cells (DC). As effectors, we applied in vitro-generated anti-tyrosinase T-cell populations as well as ex vivo-CD8(+) lymphocytes from HCMV-seropositive donors. APC electroporated with clonal mRNA were efficient inducers of spot formation by antigen-experienced CD8(+) T cells. They were equivalent to peptide-loaded targets. mRNA electroporation did not induce non-specific spot formation. While the use of autologous mRNA-electroporated DC can uncover the complete individual T-cell response towards an antigen, mRNA-electroporated K562 cells stably transfected with single HLA class I alleles help to detect CD8(+) T-cell responses restricted by single HLA class I molecules.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Electroporación , Epítopos de Linfocito T/inmunología , Interferón gamma/análisis , ARN Mensajero/inmunología , Linfocitos T/inmunología , Clonación Molecular , Células Dendríticas , Genes MHC Clase I , Humanos , Técnicas para Inmunoenzimas , Interferón gamma/inmunología , Células K562 , Monofenol Monooxigenasa/inmunología , Fosfoproteínas/inmunología , Transfección , Proteínas de la Matriz Viral/inmunología
18.
J Immunol Methods ; 259(1-2): 95-110, 2002 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11730845

RESUMEN

ELISPOT assays are increasingly used for a direct detection and quantification of single antigen-specific T cells in freshly isolated peripheral blood mononuclear cells (PBMC). They are particularly attractive for the monitoring of specific T lymphocyte responses in clinical trials assessing antigen-specific immunizations in patients with cancer or chronic viral infections. However, one major limitation for the broad clinical implementation of ELISPOT assays is the lack of an inexhaustible source of suitable HLA-matched antigen-presenting cells (APC). Currently available allogeneic or xenogeneic APC (such as the human lymphoid hybrid T2 or HLA-transfected insect cells) can either lead to strong background spot production by APC-reactive T lymphocytes or have a low antigen presentation capability. Both phenomena can prevent the detection of low frequency T cell responses in PBMC. In search of alternative APC for ELISPOT assays, the human chronic myelogenous leukemia cell line K562 that per se does not express HLA class I and class II molecules on the cell surface was transfected with the HLA-A*0201 gene. Clonal HLA-A*0201-expressing K562 (K562/A*0201) cells were able to process and present endogenously expressed and exogenously loaded melanoma peptide antigens to HLA-A*0201-restricted cytolytic T lymphocyte clones in cytotoxicity and IFN-gamma ELISPOT assays. K562/A*0201 cells were then used as APC in IFN-gamma spot assays to detect ex vivo CD8(+) T lymphocytes responsive to known HLA-A*0201-binding peptide epitopes derived from cytomegalovirus, Epstein-Barr virus, influenza virus and melanoma in PBMC from HLA-A*0201-positive donors. In the majority of cases, peptide-pulsed K562/A*0201 cells were similarly efficient in the ability to visualize single antigen-specific CD8(+) T lymphocytes when compared to T2 cells. However, in contrast to T2, background reactivity of CD8(+) T cells responsive to unpulsed K562/A*0201 was regularly found to be negligible, thereby enhancing the sensitivity of the ELISPOT assay, particularly in donors with strong anti-T2 reactivity. K562 cells transfected with HLA-A*0201 or other HLA genes can serve as standard APC for monitoring T lymphocyte responses against tumor and viral peptide antigens.


Asunto(s)
Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Antígenos HLA-A/inmunología , Inmunoensayo/métodos , Interferón gamma/inmunología , Células K562/inmunología , Citotoxicidad Inmunológica , Antígenos HLA-A/genética , Humanos , Inmunoensayo/normas , Interferón gamma/análisis , Sensibilidad y Especificidad , Transfección
19.
J Cancer Res Clin Oncol ; 130(7): 367-74, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15067541

RESUMEN

Until the end of the 19th century the possibility that a tumor could be rejected merely by the body's immune defense was no more than a vision. After more than 100 years of preclinical and clinical research in the field, the vision of cancer immunotherapy became real and has, with multiple tools, successfully entered clinical standard practice. Non-specific mediators of immune defense, such as BCG for treatment of superficial bladder cancer or interferon-alpha for treatment of chronic myelogenous leukemia and hairy cell leukemia, can induce durable remissions. In particular, antigen-specific mediators of immune defense represent promising agents for targeted cancer therapies. Antibody-based treatment of B-cell lymphomas and breast cancer has dramatically improved disease response without major toxicity. A large number of new antibodies targeting different epitopes on a variety of malignant cells are now approaching clinical approval. Allogeneic stem cell transplantation for treatment of leukemia and certain cancers represents the first T cell-based adoptive immunotherapy with large-scale clinical application. Experimental T cell-based immunotherapies with promising clinical perspectives include tumor vaccines, adoptive transfer of autologous tumor-specific effector cells and the genetic transfer of tumor-specific T cell receptors into the patient's lymphocytes. These facts demonstrate that immunotherapy has now been established as the fourth column of cancer therapy besides surgery, radiotherapy, and chemotherapy. On the basis of its already proven efficacy, the usually favorable toxicity profiles and the development perspectives of the experimental approaches a further and more rapid expansion can be expected.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Traslado Adoptivo , Animales , Anticuerpos Antineoplásicos/inmunología , Anticuerpos Antineoplásicos/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Citocinas/farmacología , Citocinas/uso terapéutico , Epítopos , Humanos , Factores Inmunológicos/farmacología , Factores Inmunológicos/uso terapéutico , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Trasplante de Células Madre , Linfocitos T/inmunología , Trasplante Homólogo
20.
J Immunother ; 31(1): 52-62, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18157012

RESUMEN

Tumor cells that show downregulation of their tumor-associated antigens (TAAs) may be able to escape immune-mediated elimination. Therefore, efficient vaccine strategies attempt to target multiple TAAs simultaneously. This is easily achieved in dendritic cell (DC)-based vaccines by introducing antigens in the form of RNA. Although insufficient message may hinder adequate expression of individual TAAs when using total-tumor RNA, high amounts of individual RNAs as pools yield DCs presenting high numbers of specific peptide-major histocompatibility complex ligands with epitopes derived from different TAAs. We used the transfer of RNAs encoding the well-defined melanoma TAAs tyrosinase, Melan-A, CDK4mut, gp100, SNRP116mut, and GPNMBmut to characterize DCs at the levels of transfected RNA, expressed protein and peptide-major histocompatibility complex ligand presentation. TAA-encoding RNA was rapidly degraded in the DCs, allowing only a single surge in protein expression shortly after transfection. We compared the functional capacity of DCs transfected with pools of 3 versus 6 RNAs. Whereas functional assays demonstrated a decrease in stimulatory capacity of DCs transfected with a pool of 3 RNAs by only 30% as compared with single RNAs, a 60% loss was seen with 6 RNAs. We conclude that larger RNA pools result in diminished presentation of individual epitopes and suggest that smaller pools of RNA be transfected into separate DC populations which are then pooled to create multiplex vaccines.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos/inmunología , ARN/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Vacunas contra el Cáncer/genética , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/inmunología , Quinasa 4 Dependiente de la Ciclina/metabolismo , Células Dendríticas/metabolismo , Citometría de Flujo , Antígenos HLA-A/genética , Antígenos HLA-A/inmunología , Antígenos HLA-A/metabolismo , Antígeno HLA-A2 , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Cinética , Antígeno MART-1 , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Monofenol Monooxigenasa/genética , Monofenol Monooxigenasa/inmunología , Monofenol Monooxigenasa/metabolismo , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleoproteínas Nucleares Pequeñas/genética , Ribonucleoproteínas Nucleares Pequeñas/inmunología , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Transfección , Antígeno gp100 del Melanoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA