Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 31(4): 527-8, 2009 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-19833080

RESUMEN

Immunotherapy, especially therapeutic vaccination, has a great deal of potential in the treatment of cancer and certain infectious diseases such as HIV (Allison et al., 2006; Fauci et al., 2008; Feldmann and Steinman, 2005). Numerous vaccine candidates have been tested in patients with a variety of tumor types and chronic viral diseases. Often, the best way to assess the clinical potential of these vaccines is to monitor the induced T cell response, and yet there are currently no standards for reporting these results. This letter is an effort to address this problem.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Inmunoensayo/normas , Monitorización Inmunológica/normas , Neoplasias/terapia , Guías de Práctica Clínica como Asunto/normas , Linfocitos T/inmunología , Vacunas Virales/uso terapéutico , Virosis/terapia , Vacunas contra el Cáncer/inmunología , Humanos , Inmunoterapia , Vacunas Virales/inmunología
2.
BMC Genomics ; 14: 869, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24325565

RESUMEN

BACKGROUND: Although transposable element (TE) derived DNA accounts for more than half of mammalian genomes and initiates a significant proportion of RNA transcripts, high throughput methods are rarely leveraged specifically to detect expression from interspersed repeats. RESULTS: To characterize the contribution of transposons to mammalian transcriptomes, we developed a custom microarray platform with probes covering known human and mouse transposons in both sense and antisense orientations. We termed this platform the "TE-array" and profiled TE repeat expression in a panel of normal mouse tissues. Validation with nanoString® and RNAseq technologies demonstrated that TE-array is an effective method. Our data show that TE transcription occurs preferentially from the sense strand and is regulated in highly tissue-specific patterns. CONCLUSIONS: Our results are consistent with the hypothesis that transposon RNAs frequently originate within genomic TE units and do not primarily accumulate as a consequence of random 'read-through' from gene promoters. Moreover, we find TE expression is highly dependent on the tissue context. This suggests that TE expression may be related to tissue-specific chromatin states or cellular phenotypes. We anticipate that TE-array will provide a scalable method to characterize transposable element RNAs.


Asunto(s)
Elementos Transponibles de ADN , Genómica/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Transcripción Genética , Animales , Línea Celular , Femenino , Expresión Génica , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos/normas , Especificidad de Órganos/genética , Reproducibilidad de los Resultados , Transfección
3.
Blood ; 117(2): 608-17, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-20935254

RESUMEN

Few published studies characterize early lymphocyte recovery after intensive chemotherapy for acute myelogenous leukemia (AML). To test the hypothesis that lymphocyte recovery mirrors ontogeny, we characterized early lymphocyte recovery in 20 consecutive patients undergoing induction timed sequential chemotherapy for newly diagnosed AML. Recovering T lymphocytes were predominantly CD4(+) and included a greatly expanded population of CD3(+)CD4(+)CD25(+)Foxp3(+) T cells. Recovering CD3(+)CD4(+)CD25(+)Foxp3(+) T cells were phenotypically activated regulatory T cells and showed suppressive activity on cytokine production in a mixed lymphocyte reaction. Despite an initial burst of thymopoiesis, most recovering regulatory T cells were peripherally derived. Furthermore, regulatory T cells showed marked oligoclonal skewing, suggesting that their peripheral expansion was antigen-driven. Overall, lymphocyte recovery after chemotherapy differs from ontogeny, specifically identifying a peripherally expanded oligoclonal population of activated regulatory T lymphocytes. These differences suggest a stereotyped immunologic recovery shared by patients with newly diagnosed AML after induction timed sequential chemotherapy. Further insight into this oligoclonal regulatory T-cell population will be fundamental toward developing effective immunomodulatory techniques to improve survival for patients with AML.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/inmunología , Subgrupos de Linfocitos T/citología , Linfocitos T Reguladores/citología , Adulto , Anciano , Separación Celular , Citarabina/administración & dosificación , Citosina/administración & dosificación , Daunorrubicina/administración & dosificación , Etopósido/administración & dosificación , Femenino , Flavonoides/administración & dosificación , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Mitoxantrona/administración & dosificación , Piperidinas/administración & dosificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Vidarabina/análogos & derivados , Adulto Joven
4.
J Immunol ; 186(4): 2148-55, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21242518

RESUMEN

Activated T cells can acquire membrane molecules from APCs through a process termed trogocytosis. The functional consequence of this event has been a subject of debate. Focusing on transfer of peptide-MHC class II (MHC-II) complexes from APCs to CD4(+) T cells after activation, in this study we investigated the molecule acquisition potential of naturally occurring regulatory T cells (Tregs) and CD4(+) Th cells. We show that acquisition of membrane molecules from APCs is an inherent feature of CD4(+) T cell activation. Triggering of the TCR enables CD4(+) T cells to acquire their agonist ligands as well as other irrelevant membrane molecules from the interacting APCs or bystander cells in a contact-dependent manner. Notably, trogocytosis is a continuous process during cell cycle progression, and Th cells and Tregs have comparable capacity for trogocytosis both in vitro and in vivo. The captured peptide-MHC-II molecules, residing in sequestered foci on the host cell surface, endow the host cells with Ag-presenting capability. Presentation of acquired peptide-MHC-II ligands by Th cells or Tregs has either stimulatory or regulatory effect on naive CD4(+) T cells, respectively. Furthermore, Th cells with captured peptide-MHC-II molecules become effector cells that manifest better recall responses, and Tregs with captured ligands exhibit enhanced suppression activity. These findings implicate trogocytosis in different subsets of CD4(+) T cells as an intrinsic mechanism for the fine tuning of Ag-specific CD4(+) T cell response.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos CD4/fisiología , Epítopos de Linfocito T/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Activación de Linfocitos/inmunología , Péptidos/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/patología , Antígenos CD4/biosíntesis , Línea Celular Tumoral , Pollos , Antígenos de Histocompatibilidad Clase II/inmunología , Memoria Inmunológica , Ligandos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Péptidos/inmunología , Mapeo de Interacción de Proteínas , Sarcoma Experimental/inmunología , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Colaboradores-Inductores/patología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología
5.
Blood ; 115(16): 3224-30, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20124511

RESUMEN

Because of its potent immunosuppressive yet stem cell-sparing activity, high-dose cyclophosphamide was tested as sole prophylaxis of graft-versus-host disease (GVHD) after myeloablative allogeneic bone marrow transplantation (alloBMT). We treated 117 patients (median age, 50 years; range, 21-66 years) with advanced hematologic malignancies; 78 had human leukocyte antigen (HLA)-matched related donors and 39 had HLA-matched unrelated donors. All patients received conventional myeloablation with busulfan/cyclophosphamide (BuCy) and T cell-replete bone marrow followed by 50 mg/kg/d of cyclophosphamide on days 3 and 4 after transplantation. The incidences of acute grades II through IV and grades III through IV GVHD for all patients were 43% and 10%, respectively. The nonrelapse mortality at day 100 and 2 years after transplantation were 9% and 17%, respectively. The actuarial overall survival and event-free survivals at 2 years after transplantation were 55% and 39%, respectively, for all patients and 63% and 54%, respectively, for patients who underwent transplantation while in remission. With a median follow-up of 26.3 months among surviving patients, the cumulative incidence of chronic GVHD is 10%. These results suggest that high-dose posttransplantation cyclophosphamide is an effective single-agent prophylaxis of acute and chronic GVHD after BuCy conditioning and HLA-matched BMT (clinicaltrials.gov no. NCT00134017).


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Ciclofosfamida/administración & dosificación , Enfermedad Injerto contra Huésped/prevención & control , Neoplasias Hematológicas/terapia , Inmunosupresores/administración & dosificación , Adulto , Anciano , Trasplante de Médula Ósea/mortalidad , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Neoplasias Hematológicas/mortalidad , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo , Resultado del Tratamiento , Adulto Joven
6.
Clin Dev Immunol ; 2012: 124187, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22778760

RESUMEN

The past decade has witnessed the evolvement of cancer immunotherapy as an increasingly effective therapeutic modality, evidenced by the approval of two immune-based products by the FDA, that is, the cancer vaccine Provenge (sipuleucel-T) for prostate cancer and the antagonist antibody against cytotoxic T-lymphocyte antigen-4 (CTLA-4) ipilimumab for advanced melanoma. In addition, the clinical evaluations of a variety of promising immunotherapy drugs are well under way. Benefiting from more efficacious immunotherapeutic agents and treatment strategies, a number of recent clinical studies have achieved unprecedented therapeutic outcomes in some patients with certain types of cancers. Despite these advances, however, the efficacy of most cancer immunotherapies currently under clinical development has been modest. A recurring scenario is that therapeutic maneuvers initially led to measurable antitumor immune responses in cancer patients but ultimately failed to improve patient outcomes. It is increasingly recognized that tumor cells can antagonize therapy-induced immune attacks through a variety of counterregulation mechanisms, which represent a fundamental barrier to the success of cancer immunotherapy. Herein we summarize the findings from some recent preclinical and clinical studies, focusing on how tumor cells advance their survival and expansion by hijacking therapy-induced immune effector mechanisms that would otherwise mediate their destruction.


Asunto(s)
Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Escape del Tumor/inmunología , Terapia Combinada , Humanos , Inmunomodulación
7.
Blood ; 114(9): 1736-45, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19556425

RESUMEN

Preclinical models have demonstrated the efficacy of granulocyte-macrophage colony-stimulating factor-secreting cancer immunotherapies (GVAX platform) accompanied by immunotherapy-primed lymphocytes after autologous stem cell transplantation in hematologic malignancies. We conducted a phase 2 study of this combination in adult patients with acute myeloid leukemia. Immunotherapy consisted of autologous leukemia cells admixed with granulocyte-macrophage colony-stimulating factor-secreting K562 cells. "Primed" lymphocytes were collected after a single pretransplantation dose of immunotherapy and reinfused with the stem cell graft. Fifty-four subjects were enrolled; 46 (85%) achieved a complete remission, and 28 (52%) received the pretransplantation immunotherapy. For all patients who achieved complete remission, the 3-year relapse-free survival (RFS) rate was 47.4% and overall survival was 57.4%. For the 28 immunotherapy-treated patients, the RFS and overall survival rates were 61.8% and 73.4%, respectively. Posttreatment induction of delayed-type hypersensitivity reactions to autologous leukemia cells was associated with longer 3-year RFS rate (100% vs 48%). Minimal residual disease was monitored by quantitative analysis of Wilms tumor-1 (WT1), a leukemia-associated gene. A decrease in WT1 transcripts in blood was noted in 69% of patients after the first immunotherapy dose and was also associated with longer 3-year RFS (61% vs 0%). In conclusion, immunotherapy in combination with primed lymphocytes and autologous stem cell transplantation shows encouraging signals of potential activity in acute myeloid leukemia (ClinicalTrials.gov: NCT00116467).


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Inmunoterapia/métodos , Leucemia Mieloide Aguda/terapia , Trasplante de Células Madre/métodos , Trasplante Autólogo/métodos , Adulto , Línea Celular Tumoral , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Inducción de Remisión , Resultado del Tratamiento
8.
Am J Hematol ; 86(1): 31-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21132730

RESUMEN

Although tyrosine kinase inhibitors have redefined the care of chronic myeloid leukemia (CML), these agents have not proved curative, likely due to resistance of the leukemia stem cells (LSC). While a number of potential therapeutic targets have emerged in CML, their expression in the LSC remains largely unknown. We therefore isolated subsets of CD34(+) stem/progenitor cells from normal donors and from patients with chronic phase or blast crisis CML. These cell subsets were then characterized based on ability to engraft immunodeficient mice and expression of candidate therapeutic targets. The CD34(+)CD38(-) CML cell population with high aldehyde dehydrogenase (ALDH) activity was the most enriched for immunodeficient mouse engrafting capacity. The putative targets: PROTEINASE 3, SURVIVIN, and hTERT were expressed only at relatively low levels by the CD34(+)CD38(-)ALDH(high) CML cells, similar to the normal CD34(+)CD38(-)ALDH(high) cells and less than in the total CML CD34(+) cells. In fact, the highest expression of these antigens was in normal, unfractionated CD34(+) cells. In contrast, PRAME and WT1 were more highly expressed by all CML CD34(+) subsets than their normal counterparts. Thus, ALDH activity appears to enrich for CML stem cells, which display an expression profile that is distinct from normal stem/progenitor cells and even the CML progenitors. Indeed, expression of a putative target by the total CD34(+) population in CML does not guarantee expression by the LSC. These expression patterns suggest that PROTEINASE 3, SURVIVIN, and hTERT are not optimal therapeutic targets in CML stem cells; whereas PRAME and WT1 seem promising.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , ADP-Ribosil Ciclasa 1/biosíntesis , Adulto , Anciano , Aldehído Deshidrogenasa/biosíntesis , Animales , Antígenos CD34/biosíntesis , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Femenino , Células Madre Hematopoyéticas/inmunología , Humanos , Proteínas Inhibidoras de la Apoptosis , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Persona de Mediana Edad , Mieloblastina/biosíntesis , Mieloblastina/genética , Trasplante de Neoplasias , ARN Mensajero/biosíntesis , Survivin , Telomerasa/biosíntesis , Telomerasa/genética , Proteínas WT1/biosíntesis , Proteínas WT1/genética
9.
J Immunol ; 183(1): 696-705, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19535642

RESUMEN

Previous work done in our laboratory, using mouse models, showed that soluble Fas ligand (sFasL) can efficiently delete donor anti-host T cells during their activation against irradiated host cells in MLCs. In the mouse models, this ex vivo sFasL treatment abrogated graft-vs-host disease (GVHD) while sparing donor T cells with antitumor reactivity. The present work was performed with human cells, to extend our work toward reduction of clinical GVHD. PBMC responders from a given individual (first party) were stimulated in vitro with irradiated PBMC stimulators from a second person (second party), in the presence of sFasL. In control MLCs without sFasL, alloreacting T cells began to up-regulate Fas (CD95) detectably and became sensitive to Fas-mediated apoptosis by as early as day 1-2. In MLCs containing sFasL, there were greatly reduced numbers of alloreacting CD3(+)CFSE(lo) cells, activation Ag-expressing CD4(hi) and CD8(hi) cells, IFN-gamma-producing CD4(+) and CD8(+) cells, and CD8(+)CD107a(+) CTLs. Furthermore, mice transplanted with the ex vivo sFasL/MLR-treated cells had prolonged time to fatal GVHD in an in vivo xenogeneic GVHD model. Responder cells harvested from primary MLCs containing sFasL had reduced proliferation in response to second party cells, but proliferated in response to CMV Ags, PHA, and third party cells. In addition, sFasL/MLR-treated cell populations contained influenza-specific T cells, CD4(+)FOXP3(+) T cells, and CD4(+)CD25(+) T cells. These data indicate that this ex vivo sFasL/MLR depletion of alloreacting human donor anti-host T cells was efficient and selective.


Asunto(s)
Proteína Ligando Fas/uso terapéutico , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Depleción Linfocítica/métodos , Subgrupos de Linfocitos T/inmunología , Animales , Proliferación Celular , Células Cultivadas , Femenino , Enfermedad Injerto contra Huésped/patología , Humanos , Activación de Linfocitos/inmunología , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Solubilidad , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/patología
10.
Nat Med ; 9(7): 952-8, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12778137

RESUMEN

A major focus of cancer immunotherapy is to develop strategies to induce T-cell responses through presentation of tumor antigens by dendritic cells (DCs). Current vaccines are limited in their ability to efficiently transfer antigens to DCs in vivo. Ex vivo-generated DCs can be efficiently loaded with antigen but after reinjection, few DCs traffic to secondary lymphoid organs, the critical sites for antigen presentation. To enhance efficiency and durability of antigen presentation by DCs, we transduced hematopoietic stem-progenitor cells (HSCs) with a model tumor antigen and then transplanted the gene-modified cells into irradiated recipient mice, which resulted in efficient expression of the transgene in a large proportion of donor derived DCs in lymphoid organs. The combination of bone marrow transplantation (BMT) using transduced HSCs, systemic agents that generate and activate DCs, and mature T-cell infusion resulted in substantial expansion and activation of antigen-specific T cells. This tripartite strategy provided potent antigen-specific immunotherapy for an aggressive established tumor.


Asunto(s)
Trasplante de Médula Ósea/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Inmunoterapia Adoptiva/métodos , Linfoma de Células B/inmunología , Linfoma de Células B/terapia , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Animales , Antígenos/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Antígenos CD40/genética , Antígenos CD40/inmunología , Antígenos CD40/metabolismo , Linfocitos T CD8-positivos/fisiología , Células Cultivadas , Células Dendríticas/fisiología , Hemaglutinación/genética , Células Madre Hematopoyéticas/fisiología , Interferón gamma/metabolismo , Lentivirus/genética , Linfoma de Células B/genética , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/genética , Bazo/citología , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/fisiología , Transducción Genética , Trasplante Autólogo
11.
Proc Natl Acad Sci U S A ; 105(38): 14342-6, 2008 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-18794521

RESUMEN

Antigen specific T cell migration to sites of infection or cancer is critical for an effective immune response. In mouse models of cancer, the number of lymphocytes reaching the tumor is typically only a few hundred, yet technology capable of imaging these cells using bioluminescence has yet to be achieved. A combination of codon optimization, removal of cryptic splice sites and retroviral modification was used to engineer an enhanced firefly luciferase (ffLuc) vector. Compared with ffLuc, T cells expressing our construct generated >100 times more light, permitting detection of as few as three cells implanted s.c. while maintaining long term coexpression of a reporter gene (Thy1.1). Expression of enhanced ffLuc in mouse T cells permitted the tracking of <3 x 10(4) adoptively transferred T cells infiltrating sites of vaccination and preestablished tumors. Penetration of light through deep tissues, including the liver and spleen, was also observed. Finally, we were able to enumerate infiltrating mouse lymphocytes constituting <0.3% of total tumor cellularity, representing a significant improvement over standard methods of quantitation including flow cytometry.


Asunto(s)
Inmunocompetencia , Luciferasas de Luciérnaga/metabolismo , Neoplasias/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Movimiento Celular , Células Cultivadas , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Vectores Genéticos/genética , Luciferasas de Luciérnaga/genética , Mediciones Luminiscentes , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Ingeniería de Proteínas , Retroviridae/genética , Linfocitos T/metabolismo , Transducción Genética
12.
J Exp Med ; 200(12): 1581-92, 2004 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-15596524

RESUMEN

Two seemingly incompatible models exist to explain the progression of cancers in immunocompetent hosts. The cancer immunosurveillance hypothesis posits that recognition of transformed cells by the immune system results in the generation of an effector response that may impede tumor growth. Clinically detectable cancer results from the emergence of tumor variants that escape this selective pressure. Alternatively, induction of immune tolerance to tumor antigens may enable cancer progression. We established a model where changes in the function of tumor-specific T cells and in tumor antigen expression could be followed during cancer progression. Early recognition of antigen led to activation, expansion, and effector function in tumor-specific CD4+ T cells resulting in the outgrowth of tumors expressing substantially reduced levels of antigen. Antigen loss was not complete, however, and levels remained above the threshold required for tumor-specific T cell recognition in vivo. In the face of persisting antigen, T cell tolerance ensued, leading to an impaired ability to mediate further antigen loss. Together, these studies establish that the processes of immunosurveillance and tumor editing coexist with a process in which the functional tumor-specific T cell repertoire is also "edited," reconciling two hypotheses historically central to our attempts to understand host antitumor immunity.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Carcinoma/inmunología , Neoplasias Renales/inmunología , Activación de Linfocitos/inmunología , Escape del Tumor , Animales , Carcinoma/patología , Modelos Animales de Enfermedad , Tolerancia Inmunológica/inmunología , Neoplasias Renales/patología , Masculino , Ratones , Ratones Endogámicos BALB C
13.
Sci Adv ; 5(7): eaaw5096, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31281894

RESUMEN

Chimeric antigen receptor (CAR) T cell therapy for hematologic malignancies is fraught with several unknowns, including number of functional T cells that engage target tumor, durability and subsequent expansion and contraction of that engagement, and whether toxicity can be managed. Non-invasive, serial imaging of CAR T cell therapy using a reporter transgene can address those issues quantitatively. We have transduced anti-CD19 CAR T cells with the prostate-specific membrane antigen (PSMA) because it is a human protein with restricted normal tissue expression and has an expanding array of positron emission tomography (PET) and therapeutic radioligands. We demonstrate that CD19-tPSMA(N9del) CAR T cells can be tracked with [18F]DCFPyL PET in a Nalm6 model of acute lymphoblastic leukemia. Divergence between the number of CD19-tPSMA(N9del) CAR T cells in peripheral blood and bone marrow and those in tumor was evident. These findings underscore the need for non-invasive repeatable monitoring of CAR T cell disposition clinically.


Asunto(s)
Antígenos de Superficie/metabolismo , Glutamato Carboxipeptidasa II/metabolismo , Inmunoterapia Adoptiva , Tomografía de Emisión de Positrones/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico por imagen , Animales , Antígenos CD19/metabolismo , Antígenos de Superficie/genética , Glutamato Carboxipeptidasa II/genética , Humanos , Leucemia Experimental/diagnóstico por imagen , Leucemia Experimental/patología , Lisina/análogos & derivados , Ratones Endogámicos NOD , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/fisiología , Urea/análogos & derivados
14.
Biol Blood Marrow Transplant ; 14(5): 499-509, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18410892

RESUMEN

The "allogeneic effect" refers to the induction of host B cell antibody synthesis or host T cell cytotoxicity, including tumoricidal activity, by an infusion of allogeneic lymphocytes. We show that treatment of mice with cyclophosphamide (Cy) followed by CD8(+) T cell-depleted allogeneic donor lymphocyte infusion (Cy + CD8(-) DLI) induces regression of established tumors with minimal toxicity in models of both hematologic and solid cancers, even though the donor cells are eventually rejected by the host immune system. The optimal antitumor effect of Cy + CD8(-) DLI required the presence of donor CD4(+) T cells, host CD8(+) T cells, and alloantigen expression by normal host but not tumor tissue. The results support a model in which a donor CD4(+) T cell-mediated graft-versus-host (GVH) reaction effectively awakens antitumor immunity among Cy-resistant host CD8(+) T cells. These events provide the cellular mechanism of the "allogeneic effect" in antitumor immunity. Cy + CD8(-) DLI may be an effective and minimally toxic strategy for awakening the host immune response to advanced cancers.


Asunto(s)
Reacción Injerto-Huésped/inmunología , Transfusión de Linfocitos/métodos , Animales , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Ciclofosfamida/uso terapéutico , Depleción Linfocítica , Ratones , Neoplasias/inmunología , Neoplasias/terapia , Trasplante Homólogo , Resultado del Tratamiento
15.
J Exp Med ; 215(3): 859-876, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29436396

RESUMEN

Depletion of immunosuppressive tumor-associated macrophages (TAMs) or reprogramming toward a proinflammatory activation state represent different strategies to therapeutically target this abundant myeloid population. In this study, we report that inhibition of colony-stimulating factor-1 receptor (CSF-1R) signaling sensitizes TAMs to profound and rapid reprogramming in the presence of a CD40 agonist before their depletion. Despite the short-lived nature of macrophage hyperactivation, combined CSF-1R+CD40 stimulation of macrophages is sufficient to create a proinflammatory tumor milieu that reinvigorates an effective T cell response in transplanted tumors that are either responsive or insensitive to immune checkpoint blockade. The central role of macrophages in regulating preexisting immunity is substantiated by depletion experiments, transcriptome analysis of ex vivo sorted TAMs, and gene expression profiling of whole tumor lysates at an early treatment time point. This approach enabled the identification of specific combination-induced changes among the pleiotropic activation spectrum of the CD40 agonist. In patients, CD40 expression on human TAMs was detected in mesothelioma and colorectal adenocarcinoma.


Asunto(s)
Inmunidad , Macrófagos/inmunología , Neoplasias/inmunología , Neoplasias/patología , Animales , Antígenos CD40/agonistas , Antígenos CD40/metabolismo , Linfocitos T CD8-positivos/inmunología , Femenino , Humanos , Inflamación/patología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Biológicos , Fenotipo , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo
16.
Cancer Res ; 65(5): 2026-34, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15753403

RESUMEN

A major limitation of adoptive immunotherapy is the availability of T cells specific for both terminally differentiated tumor cells and their clonogenic precursors. We show here that marrow-infiltrating lymphocytes (MILs) recognize myeloma cells after activation with anti-CD3/CD28 beads with higher frequency than activated peripheral blood lymphocytes from the same patients. Furthermore, activated MILs target both the terminally differentiated CD138+ plasma cells and the myeloma precursor as shown by profound inhibition in a tumor clonogenic assay. The presence of antigen in the marrow microenvironment seems to be important for the maintenance of tumor specificity. Taken together, these results highlight the intrinsic tumor specificity of MILs and describe a novel approach for the generation of tumor-specific T-cell populations suitable for adoptive immunotherapy of multiple myeloma.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Inmunoterapia Adoptiva , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Mieloma Múltiple/terapia , Células Plasmáticas/inmunología , Adulto , Anciano , Anticuerpos Monoclonales/uso terapéutico , Antígenos de Neoplasias/inmunología , Apoptosis , Antígenos CD28/inmunología , Complejo CD3/inmunología , Caspasas/metabolismo , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Femenino , Citometría de Flujo , Humanos , Masculino , Glicoproteínas de Membrana/inmunología , Persona de Mediana Edad , Mieloma Múltiple/inmunología , Proteoglicanos/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Sindecano-1 , Sindecanos , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
17.
Nanomedicine ; 2(2): 89-94, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17292120

RESUMEN

Cellular magnetic resonance imaging (MRI) relies on the use of intracellular contrast agents, primarily iron oxide compounds. Several techniques have been used to efficiently shuttle iron oxides into nonphagocytic cells, but all methods used until now require a prolonged incubation of cells. We hypothesized that instant magnetic labeling of cells could be achieved using electroporation. Neural stem cells (NSCs) and leukocytes from spleen and lymph nodes were suspended in a ferumoxide labeling solution, loaded into cuvettes, and subjected to electromechanical permeabilization using electroporation. Magnetically labeled cells were assayed for labeling efficiency, as well as for potential toxicity or altered function. To confirm the method's applicability to detect cells, MRI experiments were performed at 11.7 T. Magnetoelectroporation of NSCs, as demonstrated by Prussian blue staining, anti-dextran immunostaining, and a quantitative iron uptake assay, proved to be an efficient intracellular magnetic labeling method. Leukocytes including lymphocytes, which are notoriously difficult to label because of their membrane properties and small cytoplasmic volume, also demonstrated a pronounced uptake of ferumoxide. MRI experiments showed that labeled NSCs could be visualized as single cells and cell clusters in gelatin phantoms, and as proliferating cell masses in mouse brain. We have developed a convenient technique for instant magnetic labeling of cells. Because magnetoelectroporation allows the use of ferumoxides approved by the US Food and Drug Administration without additional agents, it has excellent potential for clinical translation.


Asunto(s)
Electroporación/métodos , Leucocitos/citología , Imagen por Resonancia Magnética/métodos , Magnetismo , Neuronas/citología , Células Madre/citología , United States Food and Drug Administration/legislación & jurisprudencia , Animales , Diferenciación Celular , Línea Celular , Supervivencia Celular , Ratones , Estados Unidos
18.
Cancer Res ; 63(24): 9007-15, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14695219

RESUMEN

A better understanding of how solid malignancies arise in an immunocompetent host, avoid immune recognition, and ultimately progress to widely disseminated cancer is essential to effectively harness the immune system against solid tumors. Because of their extra-lymphatic localization, it has been proposed that solid malignancies are just ignored by the immune system, thereby allowing their uncontrolled growth and dissemination. Alternatively, as most of the solid tumors are unable to express costimulatory molecules, the "signal one without signal two" model of tolerance induction has been frequently evoked to account for the failure of the immune system to reject antigenic tumors in vivo. In this study, we showed, however, that the extra-lymphatic growth of solid tumors is not immunologically ignored by the lymphoid compartment, resulting instead in the early induction of antigen-specific CD4(+) T-cell tolerance. Furthermore, analysis of parent-into-F1 bone marrow (BM) chimeras demonstrates that presentation of tumor antigens by BM-derived antigen-presenting cells represents the dominant mechanism in solid tumor-induced CD4(+) T-cell tolerance. Our findings of early development of antigen-specific T-cell unresponsiveness mediated by BM-derived antigen-presenting cells, not only provides a plausible explanation for the failure of the immune system to reject antigenic solid tumors in vivo, but more importantly, they have identified a barrier that, if appropriately manipulated, may lead to approaches to effectively harness the immune system against solid malignancies.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Carcinoma de Células Renales/inmunología , Anergia Clonal/inmunología , Neoplasias Renales/inmunología , Melanoma Experimental/inmunología , Animales , Presentación de Antígeno/inmunología , Carcinoma de Células Renales/patología , División Celular/inmunología , Inmunoterapia Adoptiva , Neoplasias Renales/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Masculino , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos
19.
Cancer Res ; 75(1): 51-61, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25388285

RESUMEN

Traditionally, cell-mediated immune responses to vaccination in animal models are evaluated by invasive techniques such as biopsy and organ extraction. We show here that by combining two noninvasive imaging technologies, MRI and bioluminescence imaging (BLI), we can visualize both the afferent and efferent arms of cellular events following vaccination longitudinally. To this end, we evaluated the immune response elicited by a novel Toll-like receptor 4 agonist vaccine adjuvant, glucopyranosyl lipid A (GLA), using a whole-cell tumor vaccine. After magnetovaccination, MRI was used to visualize antigen-presenting cell-mediated antigen capture and subsequent migration to draining lymph nodes (DLN). Paradoxically, we observed that the incorporation of GLA in the vaccine reduced these critical parameters of the afferent immune response. For the efferent arm, the magnitude of the ensuing antigen-specific T-cell response in DLN visualized using BLI correlated with antigen delivery to the DLN as measured by MRI. These findings were confirmed using flow cytometry. In spite of the GLA-associated reduction in antigen delivery to the DLN, however, the use of GLA as a vaccine adjuvant led to a massive proliferation of vaccine primed antigen-specific T cells in the spleen. This was accompanied by an enhanced tumor therapeutic effect of the vaccine. These findings suggest that GLA adjuvant changes the temporal and anatomical features of both the afferent and efferent arms of the vaccine response and illustrates the utility of quantitative noninvasive imaging as a tool for evaluating these parameters during vaccine optimization.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Ganglios Linfáticos/inmunología , Melanoma Experimental/terapia , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Femenino , Humanos , Mediciones Luminiscentes/métodos , Imagen por Resonancia Magnética/métodos , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Imagen Multimodal/métodos
20.
J Transl Med ; 2(1): 20, 2004 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-15212694

RESUMEN

The 6th annual Cancer Vaccines and Immunotherapy Colloquium at Walker's Cay was held under the auspices of the Albert B. Sabin Vaccine Institute on March 10-13, 2004. The Colloquium consisted of a select group of 34 scientists representing academia, biotechnology and pharmaceutical industry. The main goal of this gathering was to promote in a peaceful and comfortable environment exchanges between basic and clinical science. The secondary benefit was to inspire novel bench to bedside ventures and at the same time provide feed back about promising and/or disappointing clinical results that could help re-frame some scientific question or guide the design of future trials. Several topics were covered that included tumor antigen discovery and validation, platforms for vaccine development, tolerance, immune suppression and tumor escape mechanisms, adoptive T cell therapy and dendritic cell-based therapies, clinical trials and assessment of response. Here we report salient points raised by speakers or by the audience during animated discussion that followed each individual presentation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA