Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Blood Adv ; 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38885481

RESUMEN

Chimeric antigen receptor (CAR) T-cell therapy has emerged as a promising immunotherapeutic strategy for eradicating human cancers. Their therapeutic success and durability of clinical responses hinges, in large part, on their functional capacity, including the ability of these engineered cells to simultaneously expand and persist after infusion into patients. CD19 CAR T-cell polyfunctionality, assessing the simultaneous functions of cytokine production, proliferation, and cytotoxicity has been reported to correlate with clinical outcomes. Assay optimization is potentially limited by the heterogeneous nature of CAR T-cell infusion products and target specificity. We optimized a single-cell platform for polyfunctionality using CAR T cell products manufactured from healthy donors, engineered against a novel target, BAFF-R, and validated the protocol using CD19 CAR T cells. We observed distinct qualitative differences between BAFF-R and CD19 CAR T cells relative to the proportions of stimulatory vs. effector cytokines, based on target antigen density, and generally, CD19 CAR T cells exhibited lower indices of polyfunctionality. Finally, we applied our assay to the autologous BAFF-R CAR T-cell product generated from the first NHL patient treated on an ongoing clinical trial who had progressed after prior CD19 CAR T-cell therapy. We observed robust indicators of polyfunctionality, which correlated with successful CAR T cell expansion after infusion and achievement of durable complete remission ongoing after 18 months. The precise identification of factors determining the role of BAFF-R CAR T-cell fitness on toxicity and clinical outcome will require the application of this robust assay in the analysis of additional treated patients.

2.
Res Sq ; 2023 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-37790486

RESUMEN

Lymphoplasmacytic lymphoma (LPL) is an incurable low-grade B-cell lymphoma of the bone marrow. Despite a cumulative risk of progression, there is no approved therapy for patients in the asymptomatic phase. We conducted a first-in-human clinical trial of a novel therapeutic DNA idiotype neoantigen vaccine in nine patients with asymptomatic LPL. Treatment was well tolerated with no dose limiting toxicities. One patient achieved a minor response, and all remaining patients experienced stable disease, with median time to disease progression of 61+ months. Direct interrogation of the tumor microenvironment by single-cell transcriptome analysis revealed an unexpected dichotomous antitumor response, with significantly reduced numbers of clonal tumor mature B-cells, tracked by their unique BCR, and downregulation of genes involved in signaling pathways critical for B-cell survival post-vaccine, but no change in clonal plasma cell subpopulations. Downregulation of HLA class II molecule expression suggested intrinsic resistance by tumor plasma cell subpopulations and cell-cell interaction analyses predicted paradoxical upregulation of IGF signaling post vaccine by plasma cell, but not mature B-cell subpopulations, suggesting a potential mechanism of acquired resistance. Vaccine therapy induced dynamic changes in bone marrow T-cells, including upregulation of signaling pathways involved in T-cell activation, expansion of T-cell clonotypes, increased T-cell clonal diversity, and functional tumor antigen-specific cytokine production, with little change in co-inhibitory pathways or Treg. Vaccine therapy also globally altered cell-cell communication networks across various bone marrow cell types and was associated with reduction of protumoral signaling by myeloid cells, principally non-classical monocytes. These results suggest that this prototype neoantigen vaccine favorably perturbed the tumor immune microenvironment, resulting in reduction of clonal tumor mature B-cell, but not plasma cell subpopulations. Future strategies to improve clinical efficacy may require combinations of neoantigen vaccines with agents which specifically target LPL plasma cell subpopulations, or enable blockade of IGF-1 signaling or myeloid cell checkpoints.

3.
Biomedicines ; 11(3)2023 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-36979775

RESUMEN

Peripheral mononuclear blood cells (PBMCs) are the most widely used study materials for immunomonitoring and antigen-specific T-cell identification. However, limited patient PBMCs and low-frequency antigen-specific T cells remain as significant technical challenges. To address these limitations, we established a novel platform comprised of optimized HLA-matched immortalized B cells transfected with mRNA of a prototype viral or tumor antigen conjugated to MHC class-I trafficking domain protein (MITD) to increase the efficiency of epitope expression in antigen-presenting cells (APCs) essential to expanding antigen-specific T cells. When applied to CMV as a model, the IBMAM platform could successfully expand CMV-specific T cells from low-frequency CMV PBMCs from seropositive donors. Additionally, this platform can be applied to the validation of antigen specific TCRs. Together, compared to using APCs with synthesized peptides, this platform is an unlimited, highly efficient, and cost-effective resource in detecting and expanding antigen-specific T cells and validating antigen-specific TCRs.

4.
Blood Adv ; 7(6): 918-932, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36469551

RESUMEN

B-cell activating factor receptor (BAFF-R) is a mature B-cell survival receptor, which is highly expressed in a wide variety of B-cell malignancies but with minimal expression in immature B cells. These properties make BAFF-R an attractive target for therapy of B-cell lymphomas. We generated a novel humanized anti BAFF-R monoclonal antibody (mAb) with high specificity and potent in vitro and in vivo activity against B-cell lymphomas and leukemias. The humanized variants of an original chimeric BAFF-R mAb retained BAFF-R binding affinity and antibody-dependent cellular cytotoxicity (ADCC) against a panel of human cell lines and primary lymphoma samples. Furthermore, 1 humanized BAFF-R mAb clone and its afucosylated version, glycoengineered to optimize the primary mechanism of action, prolonged survival of immunodeficient mice bearing human tumor cell lines or patient-derived lymphoma xenografts in 3 separate models, compared with controls. Finally, the tissue specificity of this humanized mAb was confirmed against a broad panel of normal human tissues. Taken together, we have identified a robust lead-candidate BAFF-R mAb for clinical development.


Asunto(s)
Linfoma de Células B , Linfoma , Humanos , Ratones , Animales , Anticuerpos Monoclonales/uso terapéutico , Linfocitos B , Linfoma de Células B/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados , Linfoma/tratamiento farmacológico
5.
Leukemia ; 36(4): 1015-1024, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35039637

RESUMEN

Chimeric antigen receptor (CAR) T cells targeting CD19 mediate potent antitumor effects in B-cell malignancies including acute lymphoblastic leukemia (ALL), but antigen loss remains the major cause of treatment failure. To mitigate antigen escape and potentially improve the durability of remission, we developed a dual-targeting approach using an optimized, bispecific CAR construct that targets both CD19 and BAFF-R. CD19/BAFF-R dual CAR T cells exhibited antigen-specific cytokine release, degranulation, and cytotoxicity against both CD19-/- and BAFF-R-/- variant human ALL cells in vitro. Immunodeficient mice engrafted with mixed CD19-/- and BAFF-R-/- variant ALL cells and treated with a single dose of CD19/BAFF-R dual CAR T cells experienced complete eradication of both CD19-/- and BAFF-R-/- ALL variants, whereas mice treated with monospecific CD19 or BAFF-R CAR T cells succumbed to outgrowths of CD19-/BAFF-R+ or CD19+/BAFF-R- tumors, respectively. Further, CD19/BAFF-R dual CAR T cells showed prolonged in vivo persistence, raising the possibility that these cells may have the potential to promote durable remissions. Together, our data support clinical translation of BAFF-R/CD19 dual CAR T cells to treat ALL.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores Quiméricos de Antígenos , Animales , Antígenos CD19 , Humanos , Inmunoterapia Adoptiva , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores Quiméricos de Antígenos/genética , Linfocitos T
6.
Blood ; 139(9): 1289-1301, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-34521108

RESUMEN

We hypothesized that combining adoptively transferred autologous T cells with a cancer vaccine strategy would enhance therapeutic efficacy by adding antimyeloma idiotype (Id)-keyhole limpet hemocyanin (KLH) vaccine to vaccine-specific costimulated T cells. In this randomized phase 2 trial, patients received either control (KLH only) or Id-KLH vaccine, autologous transplantation, vaccine-specific costimulated T cells expanded ex vivo, and 2 booster doses of assigned vaccine. In 36 patients (KLH, n = 20; Id-KLH, n = 16), no dose-limiting toxicity was seen. At last evaluation, 6 (30%) and 8 patients (50%) had achieved complete remission in KLH-only and Id-KLH arms, respectively (P = .22), and no difference in 3-year progression-free survival was observed (59% and 56%, respectively; P = .32). In a 594 Nanostring nCounter gene panel analyzed for immune reconstitution (IR), compared with patients receiving KLH only, there was a greater change in IR genes in T cells in those receiving Id-KLH relative to baseline. Specifically, upregulation of genes associated with activation, effector function induction, and memory CD8+ T-cell generation after Id-KLH but not after KLH control vaccination was observed. Similarly, in responding patients across both arms, upregulation of genes associated with T-cell activation was seen. At baseline, all patients had greater expression of CD8+ T-cell exhaustion markers. These changes were associated with functional Id-specific immune responses in a subset of patients receiving Id-KLH. In conclusion, in this combination immunotherapy approach, we observed significantly more robust IR in CD4+ and CD8+ T cells in the Id-KLH arm, supporting further investigation of vaccine and adoptive immunotherapy strategies. This trial was registered at www.clinicaltrials.gov as #NCT01426828.


Asunto(s)
Traslado Adoptivo , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Vacunas contra el Cáncer/administración & dosificación , Células T de Memoria , Mieloma Múltiple , Vacunación , Autoinjertos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Vacunas contra el Cáncer/inmunología , Supervivencia sin Enfermedad , Femenino , Hemocianinas/administración & dosificación , Hemocianinas/inmunología , Humanos , Masculino , Células T de Memoria/inmunología , Células T de Memoria/trasplante , Mieloma Múltiple/inmunología , Mieloma Múltiple/mortalidad , Mieloma Múltiple/terapia , Tasa de Supervivencia , Trasplante Autólogo
8.
Cancer Immunol Immunother ; 67(8): 1181-1195, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29855694

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells with an immune suppressive phenotype. They represent a critical component of the immune suppressive niche described in cancer, where they support immune escape and tumor progression through direct effects on both the innate and adaptive immune responses, largely by contributing to maintenance of a high oxidative stress environment. The number of MDSCs positively correlates with protumoral activity, and often diminishes the effectiveness of immunotherapies, which is particularly problematic with the emergence of personalized medicine. Approaches targeting MDSCs showed promising results in preclinical studies and are under active investigation in clinical trials in combination with various immune checkpoint inhibitors. In this review, we discuss MDSC targets and therapeutic approaches targeting MDSC that have the aim of enhancing the existing tumor therapies.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoterapia , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/tratamiento farmacológico , Microambiente Tumoral/inmunología , Animales , Humanos , Neoplasias/inmunología
9.
BMC Cancer ; 18(1): 187, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29439670

RESUMEN

BACKGROUND: There is now a renewed interest in cancer vaccines. Patients responding to immune checkpoint blockade usually bear tumors that are heavily infiltrated by T cells and express a high load of neoantigens, indicating that the immune system is involved in the therapeutic effect of these agents; this finding strongly supports the use of cancer vaccine strategies. Lymphoplasmacytic lymphoma (LPL) is a low grade, incurable disease featuring an abnormal proliferation of Immunoglobulin (Ig)-producing malignant cells. Asymptomatic patients are currently managed by a "watchful waiting" approach, as available therapies provide no survival advantage if started before symptoms develop. Idiotypic determinants of a lymphoma surface Ig, formed by the interaction of the variable regions of heavy and light chains, can be used as a tumor-specific marker and effective vaccination using idiotypes was demonstrated in a positive controlled phase III trial. METHODS: These variable region genes can be cloned and used as a DNA vaccine, a delivery system holding tremendous potential for streamlining vaccine production. To increase vaccination potency, we are targeting antigen-presenting cells (APCs) by fusing the antigen with a sequence encoding a chemokine (MIP-3α), which binds an endocytic surface receptor on APCs. Asymptomatic phase LPL is an excellent model to test our vaccine since patients have not received chemotherapeutics that interfere with innate immune function and have low tumor burden. We are evaluating the safety of this next-generation DNA vaccine in a first-in-human clinical trial currently enrolling asymptomatic LPL patients. To elucidate the mode of action of this vaccine, we will assess its ability to generate tumor-specific immune responses and examine changes in the immune profile of both the peripheral blood and bone marrow. DISCUSSION: This vaccine could shift the current paradigm of clinical management for patients with asymptomatic LPL and inform development of other personalized approaches. TRIAL REGISTRATION: ClinicalTrials.gov identifier NCT01209871; registered on September 24, 2010.


Asunto(s)
Inmunoterapia Activa/métodos , Proteínas Recombinantes de Fusión/uso terapéutico , Vacunas de ADN/uso terapéutico , Macroglobulinemia de Waldenström/terapia , Adulto , Anciano , Antígenos/genética , Antígenos/inmunología , Antígenos/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/inmunología , Quimiocina CCL20/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Evaluación de Resultado en la Atención de Salud , Estudios Prospectivos , Proteínas Recombinantes de Fusión/inmunología , Vacunación/métodos , Vacunas de ADN/inmunología , Macroglobulinemia de Waldenström/inmunología , Macroglobulinemia de Waldenström/patología
10.
Oncoimmunology ; 5(11): e1232220, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27999743

RESUMEN

The B-cell receptor (BCR) expressed by a clonal B cell tumor is a tumor specific antigen (idiotype). However, the T-cell epitopes within human BCRs which stimulate protective immunity still lack detailed characterization. In this study, we identified 17 BCR peptide-specific CD4+ T-cell epitopes derived from BCR heavy and light chain variable region sequences. Detailed analysis revealed these CD4+ T-cell epitopes stimulated normal donors' and patients' Th1 CD4+ T cells to directly recognize the autologous tumors by secretion of IFNγ, indicating the epitopes are processed and presented by tumor cells. One BCR peptide-specific CD4+ T cell line was also cytotoxic and lysed autologous tumor cells through the perforin pathway. Sequence analysis of the epitopes revealed that 10 were shared by multiple primary patients' tumors, and 16 had the capacity to bind to more than one HLA DRB1 allele. T cells stimulated by shared epitopes recognized primary tumors expressing the same sequences on multiple HLA DRB1 alleles. In conclusion, we identified 17 BCR-derived CD4+ T-cell epitopes with promiscuous HLA DRB1 binding affinity that are shared by up to 36% of patients, suggesting a strategy to overcome the requirement for individual preparation of therapeutic agents targeting idiotype.

11.
Methods Mol Biol ; 1139: 289-303, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24619688

RESUMEN

Available therapies for lymphoplasmacytic lymphoma (LPL) provide no survival advantage if started before signs or symptoms of end-organ damage develop; hence, current recommendations are to follow a program of observation while patients are in the asymptomatic phase of disease. We hypothesize that using idiotypic determinants of a B-cell lymphoma's surface immunoglobulin as a tumor-specific marker, we can develop patient-specific chemokine-idiotype fusion DNA vaccines that induce an immune response against LPL. By activating the host immune system against the tumor antigen, we postulate that disease control of asymptomatic phase lymphoplasmacytic lymphoma can be maintained. These chemokine-idiotype fusion DNA vaccines provide protection in a lymphoma mouse model and have recently entered clinical trials. Herein, we describe procedures for the generation of therapeutic vaccines, particularly "second-generation" recombinant vaccines. Specifically, in the Methods section we describe how to identify lymphoma-associated immunoglobulin V (IgV) genes from patient biopsy and how to assemble these genes as single-chain variable gene fragment (scFv) in-frame with MIP-3α to generate novel DNA fusion vaccines.


Asunto(s)
Vacunas contra el Cáncer/genética , Idiotipos de Inmunoglobulinas/genética , Linfoma/inmunología , Linfoma/patología , Medicina de Precisión/métodos , Anticuerpos de Cadena Única/genética , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Secuencia de Aminoácidos , Vacunas contra el Cáncer/inmunología , Células Clonales/metabolismo , Clonación Molecular , ADN Complementario/genética , Humanos , Idiotipos de Inmunoglobulinas/química , Idiotipos de Inmunoglobulinas/inmunología , Datos de Secuencia Molecular , Plásmidos/genética , Análisis de Secuencia de ADN , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/inmunología
12.
J Immunol ; 190(9): 4887-98, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23536634

RESUMEN

Ag activation of the BCR may play a role in the pathogenesis of human follicular lymphoma (FL) and other B cell malignancies. However, the nature of the Ag(s) recognized by tumor BCRs has not been well studied. In this study, we used unbiased approaches to demonstrate that 42 (19.35%) of 217 tested FL Igs recognized vimentin as a shared autoantigen. The epitope was localized to the N-terminal region of vimentin for all vimentin-reactive tumor Igs. We confirmed specific binding to vimentin by using recombinant vimentin and by performing competitive inhibition studies. Furthermore, using indirect immunofluorescence staining, we showed that the vimentin-reactive tumor Igs colocalized with an anti-vimentin mAb in HEp-2 cells. The reactivity to N-terminal vimentin of IgG FL Igs was significantly higher than that of IgM FL Igs (30.4 versus 10%; p = 0.0022). However, vimentin-reactive FL Igs did not share CDR3 motifs and were not homologous. Vimentin was expressed in the T cell-rich regions of FL, suggesting that vimentin is available for binding with tumor BCRs within the tumor microenvironment. Vimentin was also frequently recognized by mantle cell lymphoma and multiple myeloma Igs. Our results demonstrate that vimentin is a shared autoantigen recognized by nonstereotyped FL BCRs and by the Igs of mantle cell lymphoma and multiple myeloma and suggest that vimentin may play a role in the pathogenesis of multiple B cell malignancies. These findings may lead to a better understanding of the biology and natural history of FL and other B cell malignancies.


Asunto(s)
Autoantígenos/inmunología , Linfoma de Células B/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Vimentina/inmunología , Secuencia de Aminoácidos , Anticuerpos Monoclonales/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Línea Celular Tumoral , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Linfoma de Células B/patología , Linfoma Folicular/inmunología , Linfoma Folicular/patología , Datos de Secuencia Molecular , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , Linfocitos T/inmunología , Linfocitos T/patología
13.
Clin Cancer Res ; 17(18): 5945-52, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21813633

RESUMEN

PURPOSE: The variable regions of Ig (idiotype, Id) expressed by malignant B cells can be used as tumor-specific antigens that induce humoral and cellular immunity. However, epitopes derived from Id that stimulate human CD8(+) T-cell immunity are incompletely characterized. EXPERIMENTAL DESIGN: The clonal Ig V(L) of human myeloma cell line U266 and five primary B-cell tumors were sequenced, and peptides corresponding to the Ig V(L) region were tested for their ability to stimulate CTLs from 10 HLA-A*0201-positive normal donors. The CTLs thus generated were tested against peptide-pulsed T2 cells and autologous tumor cells. RESULTS: Fourteen peptides derived from Ig light chain (V(L)) of U266 and primary B-cell tumors were used to generate 68 CTLs lines that specifically produced IFN-γ when cocultured with peptide-pulsed T2 cells. These CTLs lysed peptide-pulsed T2 cell as well as U266 or autologous tumor targets in an HLA class I-dependent manner. Sequence analysis revealed shared V(L) T-cell epitopes in U266 and primary B-cell tumors, not previously reported within Ig heavy chain (V(H)) sequences. CONCLUSION: This study thus identifies novel immunogenic CTLs epitopes from Id V(L), suggests that they are naturally presented on the surface of B-cell malignancies, and supports their inclusion in next-generation Id vaccines. The ability to prime T cells derived from normal HLA-matched donors, rather than patients, may also have direct application to current strategies, designed to generate allogeneic tumor-specific T cells for adoptive transfer.


Asunto(s)
Epítopos/inmunología , Cadenas Ligeras de Inmunoglobulina/inmunología , Leucemia Linfocítica Crónica de Células B/inmunología , Mieloma Múltiple/inmunología , Linfocitos T Citotóxicos/inmunología , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Epítopos de Linfocito T/inmunología , Humanos , Idiotipos de Inmunoglobulinas/inmunología , Péptidos/inmunología , Donantes de Tejidos
14.
Vaccine ; 28(50): 7970-8, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-20937320

RESUMEN

We recently reported that administration of low doses of myotoxins at vaccination sites potentiated antigen-specific T-cell immunity induced by genetic cancer vaccines in mice, an effect which was superior to TLR agonists. In the current study, we found unexpectedly that the mechanism of this potent adjuvant effect was immune-mediated. Myotoxins induced sterile inflammation at vaccination sites, associated with a predominant infiltration of dendritic cells (DC). Inhibition of DC recruitment abrogated the immune stimulation effect of myotoxins, suggesting the requirement for DC. Genetic profiling of myotoxin-treated tissues revealed characteristics of an immune microenvironment with up-regulation of chemokines, proinflammatory cytokines, Toll-like receptors (TLR) and their endogenous ligands, and activation of innate immunity. Mechanistic experiments in vivo also elucidated the requirement for genes triggering DC maturation including TLR signaling and CD40. These studies suggest that myotoxins-induced sterile inflammation generates a favorable microenvironment that promotes multiple stages in the development of adaptive immunity. This novel mechanism of immune potentiation may be exploited for development of adjuvants for genetic vaccines against infectious pathogens and cancer.


Asunto(s)
Inmunidad Adaptativa , Adyuvantes Inmunológicos/farmacología , Cardiotoxinas/farmacología , Células Dendríticas/inmunología , Inflamación/inmunología , Animales , Cardiotoxinas/inmunología , Citocinas/inmunología , Inmunidad Innata , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Receptores Toll-Like/inmunología , Regulación hacia Arriba , Vacunación/métodos , Vacunas de ADN/inmunología
15.
J Biomed Biotechnol ; 2010: 860160, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20454526

RESUMEN

HIV vaccine candidates with improved immunogenicity and induction of mucosal T-cell immunity are needed. A prime-boost strategy using a novel HIV glycoprotein 120 DNA vaccine was employed to immunize rhesus macaques. The DNA vaccine encoded a chimeric gp120 protein in fusion with monocyte chemoattractant protein-3, which was hypothesized to improve the ability of antigen-presenting cells to capture viral antigen through chemokine receptor-mediated endocytosis. DNA vaccination induced virus-reactive T cells in peripheral blood, detectable by T cell proliferation, INFgamma ELISPOT and sustained IL-6 production, without humoral responses. With a peptide-cocktail vaccine containing a set of conserved polypeptides of HIV-1 envelope protein, given by nasogastric administration, primed T-cell immunity was significantly boosted. Surprisingly, long-term and peptide-specific mucosal memory T-cell immunity was detected in both vaccinated macaques after one year. Therefore, data from this investigation offer proof-of-principle for potential effectiveness of the prime-boost strategy with a chemokine-fused gp120 DNA and warrant further testing in the nonhuman primate models for developing as a potential HIV vaccine candidate in humans.


Asunto(s)
Quimiocina CCL7/inmunología , Proteína gp120 de Envoltorio del VIH/inmunología , Inmunización Secundaria/métodos , Memoria Inmunológica/inmunología , Macaca mulatta/inmunología , Péptidos/inmunología , Vacunación/métodos , Animales , ADN , Inmunidad Celular/inmunología , Proteínas Recombinantes de Fusión/inmunología
16.
Blood ; 114(19): 4142-9, 2009 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-19749091

RESUMEN

Lymphoma idiotype protein vaccines have shown therapeutic potential in previous clinical studies, and results from a completed pivotal, phase 3 controlled trial are promising. However, streamlined production of these patient-specific vaccines is required for eventual clinical application. Here, we show that second-generation, chemokine-fused idiotype DNA vaccines, when combined with myotoxins that induced sterile inflammation with recruitment of antigen-presenting cells at vaccination sites, were exceptional in their ability to provoke memory antitumor immunity in mice, compared with several TLR agonists. The combined vaccination strategy elicited both antigen-specific T-cell responses and humoral immunity. Unexpectedly, vaccine-induced tumor protection was intact in B cell-deficient mice but was abrogated completely by T-cell depletion in vivo, suggesting T-cell dependence. Furthermore, the optimal effect of myotoxins was observed with fusion vaccines that specifically targeted antigen delivery to antigen-presenting cells and not with vaccines lacking a targeting moiety, suggesting that the rational vaccine design will require combination strategies with novel, proinflammatory agents and highly optimized molecular vaccine constructs. These studies also challenge the paradigm that antibody responses are the primary of idiotype-specific antitumor effects and support the optimization of idiotype vaccines designed to induce primarily T-cell immunity.


Asunto(s)
Linfocitos B/inmunología , Vacunas contra el Cáncer/administración & dosificación , Linfoma/inmunología , Linfoma/terapia , Linfocitos T/inmunología , Vacunas de ADN/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Animales , Células Presentadoras de Antígenos/inmunología , Línea Celular Tumoral , Proteínas Cardiotóxicas de Elápidos/administración & dosificación , Crotoxina/administración & dosificación , Sinergismo Farmacológico , Inmunidad Celular , Idiotipos de Inmunoglobulinas/administración & dosificación , Inflamación/inmunología , Ratones , Ratones Endogámicos BALB C
17.
Vaccine ; 25(46): 7955-61, 2007 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-17933439

RESUMEN

We investigated the antitumor effect of survivin DNA vaccine in murine pancreatic and lymphoma models, and if xenogenic survivin can generate stronger immune response. We found that mice vaccinated with either human or mouse survivin DNA have significantly slower tumor growth and longer survival than those vaccinated with vector DNA. There was no significant difference between groups that received human and mouse survivin DNA. Lymphocyte infiltration was greater in tumors of mice immunized with survivin DNA than in tumors of control mice. We conclude that survivin DNA vaccine generated specific antitumor effects with increased lymphocyte infiltration at the tumor sites.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Linfoma/prevención & control , Proteínas Asociadas a Microtúbulos/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias Pancreáticas/prevención & control , Vacunas de ADN/inmunología , Animales , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis , Linfocitos/inmunología , Linfoma/genética , Linfoma/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Proteínas Represoras , Survivin , Vacunas de ADN/genética
18.
Leuk Lymphoma ; 47(8): 1523-34, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16966263

RESUMEN

We observed novel transformations of follicular lymphoma (FL), first, a switch in immunoglobulin (Ig) light chain, and second, transformation of FL to acute lymphoblastic leukemia (ALL). Each set of tumors shared a common clonal origin, as demonstrated by expression of identical, unique CDR IIIH sequences, shared somatic mutations in JH, and identical bcl-2 translocation breakpoints of microdissected ALL cells. Molecular analysis of lambda V-gene expression demonstrated lambda-bearing cells in the original kappa tumor, while expansion of the lambda subclone at relapse occurred after active immunotherapy targeting the Ig receptor. These exceptional cases are compatible with a more contemporary model of lymphomagenesis in which critical events originate from genetic mechanisms which normally occur in germinal center (GC) B cells and challenge the current paradigm of parallel generation of subclones from an early, pre-GC precursor. It is also possible that the outgrowth of these variants was a consequence of immunoselection.


Asunto(s)
Transformación Celular Neoplásica/patología , Cambio de Clase de Inmunoglobulina , Cadenas Ligeras de Inmunoglobulina , Linfoma de Células B/etiología , Linfoma Folicular/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Adulto , Biopsia , Células Clonales/patología , Genes de Inmunoglobulinas/genética , Humanos , Cadenas kappa de Inmunoglobulina , Cadenas lambda de Inmunoglobulina , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiología
19.
Expert Rev Vaccines ; 5(3): 381-94, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16827622

RESUMEN

The unique antigenic determinants, termed idiotype, of the immunoglobulin expressed on a given B-cell malignancy can serve as a tumor-specific antigen for active immunotherapy. Administration of autologous tumor-derived idiotype protein conjugated to a carrier protein, keyhole limpet hemocyanin, together with granulocyte-macrophage colony-stimulating factor to follicular lymphoma patients in complete clinical remission was associated with induction of tumor-specific cellular and humoral immunity, molecular remissions, and prolonged disease-free survival. Idiotype vaccination in patients with mantle cell lymphoma following rituximab-containing chemotherapy induced tumor-specific T-cell immunity in the absence of B cells, suggesting that vaccines may be used in combination with rituximab. Three double-blind, randomized, Phase III idiotype vaccine trials are currently ongoing to definitively determine the clinical benefit of idiotype-keyhole limpet hemocyanin plus granulocyte-macrophage colony-stimulating factor vaccination in patients with lymphoma. Results from early clinical trials with idiotype vaccines suggested that both humoral and cellular immune responses may be independently associated with tumor regression and improved progression-free survival. With the increased use of rituximab for the treatment of follicular lymphoma and other B-cell non-Hodgkin's lymphomas, further improvement in the potency of the vaccines would require strategies to enhance T-cell responses, as rituximab depletes normal B cells and impairs the generation of antibody responses.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/uso terapéutico , Linfoma Folicular/tratamiento farmacológico , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales de Origen Murino , Antineoplásicos/uso terapéutico , Evaluación Preclínica de Medicamentos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Idiotipos de Inmunoglobulinas/inmunología , Linfoma Folicular/inmunología , Ensayos Clínicos Controlados Aleatorios como Asunto , Rituximab
20.
J Control Release ; 113(3): 261-70, 2006 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-16793161

RESUMEN

Idiotypic sequences, specific to the hypervariable regions of immunoglobulins expressed by malignant B cells offer a therapeutic target in B cell lymphoma. Efficient approaches have been described to clone a single chain fragment of the tumor immunoglobulin (Ig) comprising of heavy and light Ig chains (sFv) fused with proinflammatory chemokines. Tumor associated, poorly immunogenic self antigens encoded by plasmid DNA (pDNA) have been rendered immunogenic by chemokine fusion, thereby targeting to antigen presenting cells (APCs) which differentially express chemokine receptors. Here we present an injectable (parenteral) approach using synthetic polymer based cationic microparticle formulations for enhancing the potency of such chemokine/self antigen expressing plasmid construct. Branched and linear polyethyleneimine (PEI) were conjugated on poly (D, L lactide-co-glycolide) (PLGA) microparticles using carbodiimide chemistry followed by efficient loading of plasmid DNA. In addition to imparting significant buffering ability to these cationic microparticles, flow cytometry studies indicate that these DNA loaded microparticles significantly up regulate CD80 and MHC class II markers in phagocytic RAW264.7 cells, indicating intrinsic adjuvant effects. Intradermal injections in Balb/c mice with these formulations induced significant protection upon tumor challenge with 2.5 times the minimal lethal dose. Long term survival rates were significant (p < 0.05) in comparison with saline injected controls or blank microparticles. Further studies indicated that intramuscular delivery might provide better protection compared to intradermal injections and perform similar to gene gun mediated administration. We conclude, based on these promising in vivo results, that such surface-functionalized microparticles offer an attractive strategy to improve the potency of self antigen-based cancer DNA vaccines.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Ácido Láctico/química , Linfoma de Células B/inmunología , Microesferas , Polietileneimina/química , Ácido Poliglicólico/química , Polímeros/química , Vacunas de ADN/inmunología , Animales , Antígeno B7-1/metabolismo , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Región Variable de Inmunoglobulina/genética , Ácido Láctico/toxicidad , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Proteínas Quimioatrayentes de Monocitos/genética , Trasplante de Neoplasias , Polietileneimina/toxicidad , Ácido Poliglicólico/toxicidad , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Polímeros/toxicidad , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Vacunas de ADN/química , Vacunas de ADN/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...