Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Oncoimmunology ; 9(1): 1758869, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32566387

RESUMEN

Ovarian cancer accounts for most deaths from gynecologic malignancies. Although more than 80% of patients respond to first-line standard of care, most of these responders present with recurrence and eventually succumb to carcinomatosis and chemotherapy-resistant disease. To improve patient survival, new modalities must, therefore, target or prevent recurrent disease. Here we describe for the first time a novel syngeneic mouse model of recurrent high-grade serous ovarian cancer (HGSOC), which allows immunotherapeutic interventions in a time course relevant to human carcinomatosis and disease course. Using this model, we demonstrate the efficacy of Transimmunization (TI), a dendritic cell (DC) vaccination strategy that uses autologous and physiologically derived DC loaded with autologous whole tumor antigens. TI has been proven successful in the treatment of human cutaneous T cell lymphoma and we report for the first time its in vivo efficacy against an intra-peritoneal solid tumor. Given as a single therapy, TI is able to elicit an effective anti-tumor immune response and inhibit immune-suppressive crosstalks with sufficient power to curtail tumor progression and establishment of carcinomatosis and recurrent disease. Specifically, TI is able to inhibit the expansion of tumor-associated macrophages as well as myeloid-derived suppressive cells consequently restoring T cell immune-surveillance. These results demonstrate the possible value of TI in the management of ovarian cancer and other intra-peritoneal tumors.


Asunto(s)
Neoplasias Ováricas , Animales , Carcinoma Epitelial de Ovario , Células Dendríticas , Femenino , Ratones , Recurrencia Local de Neoplasia/prevención & control , Neoplasias Ováricas/terapia , Neoplasias Cutáneas
2.
Yale J Biol Med ; 93(1): 145-159, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32226344

RESUMEN

Dendritic cells (DCs) are professional antigen-presenting cells, necessary for the initiation and maintenance of antigen-specific immunity and tolerance. Decades of research have been driven by hopes to harness the immunological capabilities of DCs and achieve physiological partnership with the immune system for therapeutic ends. Potential applications for DC-based immunotherapy include treatments for cancer, autoimmune disorders, and infectious diseases. However, DCs have poor availability in peripheral and lymphoid tissues and have poor survivability in culture, leading to the development of multiple strategies to generate and manipulate large numbers of DCs ex vivo. Among these is Extracorporeal Photopheresis (ECP), a widely used cancer immunotherapy. Recent advancements have uncovered that stimulation of monocyte-to-DC maturation via physiologic inflammatory signaling lies at the mechanistic core of ECP. Here, we describe the landscape of DC-based immunotherapy, the historical context of ECP, the current mechanistic understanding of ex vivo monocyte-to-DC maturation in ECP, and the implications of this understanding on making scientifically driven improvements to modern ECP protocols and devices.


Asunto(s)
Células Dendríticas/fisiología , Inmunoterapia/métodos , Neoplasias , Fotoféresis , Humanos , Neoplasias/inmunología , Neoplasias/terapia , Fotoféresis/instrumentación , Fotoféresis/métodos
3.
J Immunother Cancer ; 8(1)2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32209603

RESUMEN

Cells succumbing to stress via regulated cell death (RCD) can initiate an adaptive immune response associated with immunological memory, provided they display sufficient antigenicity and adjuvanticity. Moreover, multiple intracellular and microenvironmental features determine the propensity of RCD to drive adaptive immunity. Here, we provide an updated operational definition of immunogenic cell death (ICD), discuss the key factors that dictate the ability of dying cells to drive an adaptive immune response, summarize experimental assays that are currently available for the assessment of ICD in vitro and in vivo, and formulate guidelines for their interpretation.


Asunto(s)
Muerte Celular Inmunogénica/genética , Biología Molecular/métodos , Consenso , Guías como Asunto , Humanos
4.
Methods Mol Biol ; 2097: 173-195, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31776926

RESUMEN

Generation of large numbers of dendritic cells (DC) for research or immunotherapeutic purposes typically involves in vitro conversion of murine bone marrow precursors or human blood monocytes to DC via cultivation with supraphysiologic concentrations of cytokines such as GM-CSF and IL-4 for up to 7 days. Alternatively, our group has recently established a new approach, based on the underlying mechanism of action of a widely used cancer immunotherapy termed Extracorporeal Photochemotherapy (ECP). Our method of rapid and cytokine-free production of therapeutically relevant DC populations, leveraging the innate physiologic programs likely responsible for DC differentiation from blood monocytes in vivo, potentially offers a novel, inexpensive, and easily accessible source of DC for clinical and research uses. This approach involves ex vivo physiologic reprogramming of blood monocytes to immunologically tunable dendritic antigen-presenting cells, which we term "phDC," for physiological DC. To facilitate access and utilization of these new DC populations by the research community, in this chapter, we describe the use of a scaled-down version of the clinical ECP leukocyte-treatment device termed the Transimmunization (TI) chamber or plate, suitable for processing both mouse and human samples. We highlight the methodological sequences necessary to isolate mouse or human peripheral blood mononuclear cell (PBMC) from whole blood, and to expose those PBMC to the TI chamber for facilitating monocyte activation and conversion to physiological DC (phDC) through interaction with blood proteins and activated platelets under controlled flow conditions. We then provide sample protocols for potential applications of the generated DC, including their use as vaccinating antigen-presenting cells (APC) in murine in vivo antitumor models, and in human ex vivo T-cell stimulation and antigen cross-presentation assays which mimic clinical vaccination. We additionally highlight the technical aspects of loading mouse or human phDC with tumor-associated antigens (TAA) in the form of peptides or apoptotic tumor cells. We provide a simple and clinically relevant means to reprogram blood monocytes into functional APC, potentially replacing the comparatively expensive and clinically disappointing cytokine-derived DC which have previously dominated the dendritic cell landscape.


Asunto(s)
Células Dendríticas/citología , Inmunoterapia/métodos , Animales , Anticoagulantes/farmacología , Antígenos de Neoplasias/metabolismo , Donantes de Sangre , Células Cultivadas , Humanos , Masculino , Melanoma/inmunología , Melanoma/patología , Melanoma/terapia , Ratones Endogámicos C57BL , Neoplasias/inmunología , Neoplasias/patología , Péptidos/metabolismo , Fotoquimioterapia
5.
Int Rev Cell Mol Biol ; 349: 251-307, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31759433

RESUMEN

Dendritic cells (DCs) are professional antigen-presenting cells, required for the initiation of naïve and memory T cell responses and regulation of adaptive immunity. The discovery of DCs in 1973, which culminated in the Nobel Prize in Physiology or Medicine in 2011 for Ralph Steinman and colleagues, initially focused on the identification of adherent mononuclear cell fractions with uniquely stellate dendritic morphology, followed by key discoveries of their critical immunologic role in initiating and maintaining antigen-specific immunity and tolerance. The medical promise of marshaling these key capabilities of DCs for therapeutic modulation of antigen-specific immune responses has guided decades of research in hopes to achieve genuine physiologic partnership with the immune system. The potential uses of DCs in immunotherapeutic applications include cancer, infectious diseases, and autoimmune disorders; thus, methods for rapid and reliable large-scale production of DCs have been of great academic and clinical interest. However, difficulties in obtaining DCs from lymphoid and peripheral tissues, low numbers and poor survival in culture, have led to advancements in ex vivo production of DCs, both for probing molecular details of DC function as well as for experimenting with their clinical utility. Here, we review the development of a diverse array of DC production methodologies, ranging from cytokine-based strategies to genetic engineering tools devised for enhancing DC-specific immunologic functions. Further, we explore the current state of DC therapies in clinic, as well as emerging insights into physiologic production of DCs inspired by existing therapies.


Asunto(s)
Células Dendríticas/citología , Células Dendríticas/inmunología , Animales , Diferenciación Celular , Ingeniería Genética , Humanos , Inmunoterapia , Inflamación/inmunología , Vacunas/inmunología
6.
Cell Death Dis ; 10(8): 578, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31371700

RESUMEN

Extracorporeal photochemotherapy (ECP) is employed for the management of cutaneous T cell lymphoma (CTCL). ECP involves the extracorporeal exposure of white blood cells (WBCs) to a photosensitizer, 8-methoxypsoralen (8-MOP), in the context of ultraviolet A (UVA) radiation, followed by WBC reinfusion. Historically, the therapeutic activity of ECP has been attributed to selective cytotoxicity on circulating CTCL cells. However, only a fraction of WBCs is exposed to ECP, and 8-MOP is inactive in the absence of UVA light, implying that other mechanisms underlie the anticancer effects of ECP. Recently, ECP has been shown to enable the physiological differentiation of monocytes into dendritic cells (DCs) that efficiently cross-present tumor-associated antigens (TAAs) to CD8+ T lymphocytes to initiate cognate immunity. However, the source of TAAs and immunostimulatory signals for such DCs remains to be elucidated. Here, we demonstrate that 8-MOP plus UVA light reduces melanoma cell viability along with the emission of ICD-associated danger signals including calreticulin (CALR) exposure on the cell surface and secretion of ATP, high mobility group box 1 (HMGB1) and type I interferon (IFN). Consistently, melanoma cells succumbing to 8-MOP plus UVA irradiation are efficiently engulfed by monocytes, ultimately leading to cross-priming of CD8+ T cells against cancer. Moreover, malignant cells killed by 8-MOP plus UVA irradiation in vitro vaccinate syngeneic immunocompetent mice against living cancer cells of the same type, and such a protection is lost when cancer cells are depleted of calreticulin or HMGB1, as well as in the presence of an ATP-degrading enzyme or antibodies blocking type I IFN receptors. ECP induces bona fide ICD, hence simultaneously providing monocytes with abundant amounts of TAAs and immunostimulatory signals that are sufficient to initiate cognate anticancer immunity.


Asunto(s)
Antígenos de Neoplasias/genética , Linfoma Cutáneo de Células T/inmunología , Linfoma Cutáneo de Células T/terapia , Metoxaleno/farmacología , Adenosina Trifosfato/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de la radiación , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/efectos de la radiación , Proteína HMGB1/genética , Humanos , Muerte Celular Inmunogénica/efectos de los fármacos , Muerte Celular Inmunogénica/efectos de la radiación , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Leucocitos/efectos de la radiación , Linfoma Cutáneo de Células T/patología , Ratones , Monocitos/efectos de los fármacos , Monocitos/efectos de la radiación , Fotoféresis , Fármacos Fotosensibilizantes/farmacología , Receptor de Interferón alfa y beta/genética , Rayos Ultravioleta
7.
J Vis Exp ; (147)2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-31157760

RESUMEN

Extracorporeal photochemotherapy (ECP) is a widely used cancer immunotherapy for cutaneous T cell lymphoma (CTCL), operative in over 350 university centers worldwide. While ECP's clinical efficacy and exemplary safety profile have driven its widespread use, elucidation of the underlying mechanisms has remained a challenge, partly owing to lack of a laboratory ECP model. To overcome this obstacle and create a simple, user-friendly platform for ECP research, we developed a scaled-down version of the clinical ECP leukocyte-processing device, suitable for work with both mouse models, and small human blood samples. This device is termed the Transimmunization (TI) chamber, or plate. In a series of landmark experiments, the miniaturized device was used to produce a cellular vaccine that regularly initiated therapeutic anti-cancer immunity in several syngeneic mouse tumor models. By removing individual factors from the experimental system and ascertaining their contribution to the in vivo anti-tumor response, we then elucidated key mechanistic drivers of ECP immunizing potential. Collectively, our results revealed that anti-tumor effects of ECP are initiated by dendritic cells (DC), physiologically generated through blood monocyte interaction with platelets in the TI plate, and loaded with antigens from tumor cells whose apoptotic cell death is finely titrated by exposure to the photoactivatable DNA cross-linking agent 8-methoxypsoralen and UVA light (8-MOPA). When returned to the mouse, this cellular vaccine leads to specific and transferable anti-tumor T cell immunity. We verified that the TI chamber is also suitable for human blood processing, producing human DCs fully comparable in activation state and profile to those derived from the clinical ECP chamber. The protocols presented here are intended for ECP studies in mouse and man, controlled generation of apoptotic tumor cells with 8-MOPA, and rapid production of physiologic human and mouse monocyte-derived DCs for a variety of applications.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/fisiología , Animales , Apoptosis , Plaquetas/inmunología , Comunicación Celular , Humanos , Inmunización , Melanoma/patología , Ratones Endogámicos C57BL , Monocitos/inmunología , Fotoféresis , Neoplasias Cutáneas
8.
Cancer Res ; 78(14): 4045-4058, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29764863

RESUMEN

Extracorporeal photochemotherapy (ECP) is a cancer immunotherapy for cutaneous T-cell lymphoma (CTCL) operative in more than 350 centers worldwide. Although its efficacy and favorable safety profile have driven its widespread use, elucidation of its underlying mechanism has been difficult. In this study, we identify the principal contributors to the anticancer immunotherapeutic effects of ECP, with the goal of enhancing potency and broadening applicability to additional malignancies. First, we scaled down the clinical ECP leukocyte-processing device to mouse size. Second, we used that miniaturized device to produce a cellular vaccine that regularly initiated therapeutic antimelanoma immunity. Third, we individually subtracted key factors from either the immunizing inoculum or the treated animal to ascertain their contribution to the in vivo antimelanoma response. Platelet-signaled monocyte-to-dendritic cell (DC) differentiation followed by sorting/processing/presentation of tumor antigens derived from internalized apoptotic tumor cells were absolute requirements. As in clinical ECP, immunogenic cell death of tumor cells was finely titrated by DNA cross-linkage mediated by photoactivated 8-methoxypsoralen (8-MOPA). ECP-induced tumor-loaded DC were effective immunotherapeutic agents only if they were spared exposure to 8-MOPA, indicating that healthy DC are required for ECP. Infusion of responder T cells into naïve tumor-challenged mice established the protective role of stimulated T-cell antitumor immunity. Collectively, these results reveal that selective antitumor effects of ECP are initiated by tumor antigen-loaded, ECP-induced DC, which promote potent collaboration between CD4 and CD8 tumor-specific T cells. These mechanistic insights suggest potential therapeutic applicability of ECP to solid tumors in addition to CTCL.Significance: These findings identify principal cellular contributors to the anticancer immunotherapeutic impact of ECP and suggest this treatment may be applicable to a broad spectrum of immunogenic malignancies. Cancer Res; 78(14); 4045-58. ©2018 AACR.


Asunto(s)
Antineoplásicos/farmacología , Células Dendríticas/efectos de los fármacos , Linfoma Cutáneo de Células T/tratamiento farmacológico , Monocitos/efectos de los fármacos , Animales , Antígenos de Neoplasias/metabolismo , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Línea Celular Tumoral , Células Dendríticas/metabolismo , Inmunoterapia/métodos , Linfoma Cutáneo de Células T/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Fotoquimioterapia/métodos
9.
Plast Reconstr Surg Glob Open ; 4(9): e1032, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27757347

RESUMEN

We present a case of a 65-year-old man with cutaneous T-cell lymphoma treated with radiation therapy and an allogeneic hematopoietic stem cell transplant from his human leukocyte antigen-matched brother. Engraftment was successful, but the patient went on to develop painful, radiation-induced ulcers. The ulcers were fat-allografted using liposuctioned fat from his brother because of the patient's unique chimeric state. Postprocedure follow-up revealed epithelialization of the ulcer sites and significant improvement in neuropathic pain. Our unique case study supports the use of fat grafting for its restorative purposes and for its ability to alleviate chronic neuropathic pain. Additionally, it appears that our case provides a basis of a general approach to the treatment of radiation-induced ulcers in chimeric patients with lymphoid malignancies.

11.
J Biol Chem ; 291(17): 8931-50, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-26719339

RESUMEN

Here we report a "configuration-dependent" mechanism of action for IL-15:IL-15Rα (heterodimeric IL-15 or hetIL-15) where the manner by which IL-15:IL-15Rα molecules are presented to target cells significantly affects its function as a vaccine adjuvant. Although the cellular mechanism of IL-15 trans-presentation via IL-15Rα and its importance for IL-15 function have been described, the full effect of the IL-15:IL-15Rα configuration on responding cells is not yet known. We found that trans-presenting IL-15:IL-15Rα in a multivalent fashion on the surface of antigen-encapsulating nanoparticles enhanced the ability of nanoparticle-treated dendritic cells (DCs) to stimulate antigen-specific CD8(+) T cell responses. Localization of multivalent IL-15:IL-15Rα and encapsulated antigen to the same DC led to maximal T cell responses. Strikingly, DCs incubated with IL-15:IL-15Rα-coated nanoparticles displayed higher levels of functional IL-15 on the cell surface, implicating a mechanism for nanoparticle-mediated transfer of IL-15 to the DC surface. Using artificial antigen-presenting cells to highlight the effect of IL-15 configuration on DCs, we showed that artificial antigen-presenting cells presenting IL-15:IL-15Rα increased the sensitivity and magnitude of the T cell response, whereas IL-2 enhanced the T cell response only when delivered in a paracrine fashion. Therefore, the mode of cytokine presentation (configuration) is important for optimal immune responses. We tested the effect of configuration dependence in an aggressive model of murine melanoma and demonstrated significantly delayed tumor progression induced by IL-15:IL-15Rα-coated nanoparticles in comparison with monovalent IL-15:IL-15Rα. The novel mechanism of IL-15 transfer to the surface of antigen-processing DCs may explain the enhanced potency of IL-15:IL-15Rα-coated nanoparticles for antigen delivery.


Asunto(s)
Presentación de Antígeno/efectos de los fármacos , Antígenos de Neoplasias , Linfocitos T CD8-positivos/inmunología , Materiales Biocompatibles Revestidos/farmacología , Células Dendríticas/inmunología , Inmunidad Celular/efectos de los fármacos , Interleucina-15 , Nanopartículas , Neoplasias Experimentales , Receptores de Interleucina-15/inmunología , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/farmacología , Humanos , Interleucina-15/inmunología , Interleucina-15/farmacología , Ratones , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología
12.
Nat Genet ; 47(9): 1011-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26192916

RESUMEN

Cutaneous T cell lymphoma (CTCL) is a non-Hodgkin lymphoma of skin-homing T lymphocytes. We performed exome and whole-genome DNA sequencing and RNA sequencing on purified CTCL and matched normal cells. The results implicate mutations in 17 genes in CTCL pathogenesis, including genes involved in T cell activation and apoptosis, NF-κB signaling, chromatin remodeling and DNA damage response. CTCL is distinctive in that somatic copy number variants (SCNVs) comprise 92% of all driver mutations (mean of 11.8 pathogenic SCNVs versus 1.0 somatic single-nucleotide variant per CTCL). These findings have implications for new therapeutics.


Asunto(s)
Linfoma Cutáneo de Células T/genética , Neoplasias Cutáneas/genética , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Exoma , Expresión Génica , Frecuencia de los Genes , Estudios de Asociación Genética , Genómica , Humanos , Linfoma Cutáneo de Células T/metabolismo , Mutación Missense , Polimorfismo de Nucleótido Simple , Células Tumorales Cultivadas
13.
Int J Nanomedicine ; 9: 5231-46, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25419128

RESUMEN

Targeting antigen to dendritic cells (DCs) is a powerful and novel strategy for vaccination. Priming or loading DCs with antigen controls whether subsequent immunity will develop and hence whether effective vaccination can be achieved. The goal of our present work was to increase the potency of DC-based antitumor vaccines by overcoming inherent limitations associated with antigen stability and cross-presentation. Nanoparticles prepared from the biodegradable polymer poly(lactic-co-glycolic acid) have been extensively used in clinical settings for drug delivery and are currently the subject of intensive investigation as antigen delivery vehicles for vaccine applications. Here we describe a nanoparticulate delivery system with the ability to simultaneously carry a high density of protein-based antigen while displaying a DC targeting ligand on its surface. Utilizing a targeting motif specific for the DC-associated surface ligand DEC-205, we show that targeted nanoparticles encapsulating a MART-127-35 peptide are both internalized and cross-presented with significantly higher efficiency than isotype control-coated nanoparticles in human cells. In addition, the DEC-205-labeled nanoparticles rapidly escape from the DC endosomal compartment and do not colocalize with markers of early (EEA-1) or late endosome/lysosome (LAMP-1). This indicates that encapsulated antigens delivered by nanoparticles may have direct access to the class I cytoplasmic major histocompatibility complex loading machinery, overcoming the need for "classical" cross-presentation and facilitating heightened DC stimulation of anti-tumor CD8(+) T-cells. These results indicate that this delivery system provides a flexible and versatile methodology to deliver melanoma-associated antigen to DCs, with both high efficiency and heightened potency.


Asunto(s)
Antígenos CD/inmunología , Vacunas contra el Cáncer/administración & dosificación , Células Dendríticas/inmunología , Ácido Láctico/química , Lectinas Tipo C/inmunología , Antígeno MART-1/administración & dosificación , Melanoma/inmunología , Nanopartículas/química , Ácido Poliglicólico/química , Receptores de Superficie Celular/inmunología , Presentación de Antígeno/efectos de los fármacos , Vacunas contra el Cáncer/inmunología , Humanos , Ácido Láctico/inmunología , Antígeno MART-1/inmunología , Melanoma/terapia , Antígenos de Histocompatibilidad Menor , Terapia Molecular Dirigida , Nanopartículas/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
14.
Transfus Apher Sci ; 50(3): 379-87, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24215840

RESUMEN

Extracorporeal photochemotherapy (ECP) is a widely used method for either immunization against cutaneous T cell lymphoma or immunosuppression of graft-versus-host disease and organ transplant rejection (OTR). Leukapheresed blood is routed through a chamber, in which 8-methoxypsoralen is activated by ultraviolet energy (PUVA), thereby causing DNA crosslinks in processed leukocytes. Return of ECP-processed mononuclear leukocytes to the patient then modulates aberrant T cell immunity. Since interaction with the ECP flow chamber induces monocyte-to-dendritic antigen presenting cell (DC) maturation, we examined the possibility that PUVA may direct the most heavily exposed monocytes to differentiate into tolerogenic DC, while the least exposed DC might remain immunogenic. Expression of the glucocorticoid-induced leucine zipper (GILZ) gene is a distinguishing marker of tolerogenic DC. We report that PUVA directly stimulates GILZ expression. PUVA-exposed DC up-regulated GILZ, down-regulated costimulatory CD80 and CD86, became resistant to Toll-like receptor-induced maturation, increased IL-10 production and decreased IL-12p70 production, all features of immunosuppressive DC. Knockdown of GILZ with siRNA reduced IL-10 and increased IL-12p70 production, demonstrating that GILZ is critical for this profile. PUVA-induction of GILZ expression by DC may help explain how ECP suppresses GVHD and OTR. Conversely, those ECP-processed monocytes minimally exposed to PUVA may mediate ECP's immunogenic effects.


Asunto(s)
Células Dendríticas/inmunología , Inmunomodulación , Metoxaleno/farmacología , Fotoféresis , Fármacos Fotosensibilizantes/farmacología , Factores de Transcripción/inmunología , Adulto , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Células Dendríticas/citología , Femenino , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/efectos de la radiación , Inmunomodulación/efectos de los fármacos , Inmunomodulación/efectos de la radiación , Masculino , Terapia PUVA/métodos
15.
Transfus Apher Sci ; 50(3): 322-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23978554

RESUMEN

Extracorporeal photochemotherapy (ECP) is a widely used immunotherapy for cutaneous T cell lymphoma, as well as immunomodulation of graft-versus-host disease (GVHD) and transplanted organ rejection. ECP's mechanism encompasses large-scale physiologic platelet induction of dendritic cells (DCs). The normal bidirectional immunologic talents of DCs likely contribute heavily to ECP's capacity to immunize against tumor antigens, while also suppressing transplant immunopathology. Our understanding of how ECP physiologically induces monocyte-to-DC maturation can enhance the treatment's potency, potentially broaden its use to other cancers and autoimmune disorders and tailor its application to individual patients' diseases. ECP's next decade is filled with promise.


Asunto(s)
Células Dendríticas/inmunología , Monocitos/inmunología , Fotoféresis/métodos , Antígenos de Neoplasias/inmunología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/terapia , Células Dendríticas/patología , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Rechazo de Injerto/prevención & control , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/terapia , Humanos , Inmunización/métodos , Inmunomodulación , Monocitos/patología , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Trasplante de Órganos , Inmunología del Trasplante
16.
Transfus Apher Sci ; 50(3): 370-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24360371

RESUMEN

Extracorporeal Photochemotherapy (ECP) is a widely used therapy for cutaneous T cell lymphoma (CTCL). Although the mechanism of clinical action of ECP is not precisely established, previous studies have shown evidence of induction of dendritic cells (DCs). Here we show that, under flow conditions similar to those in post-capillary venules, ECP promotes platelet immobilization and activation, initiating stepwise receptor-ligand interactions with monocytes, which then differentiate into DC. These findings clarify how ECP directly stimulates DC maturation; suggest a new clinically applicable approach to the obtainment of DC; and identify a novel mechanism that may reflect physiological induction of DC.


Asunto(s)
Plaquetas/inmunología , Células Dendríticas/inmunología , Monocitos/inmunología , Fotoféresis , Activación Plaquetaria , Transducción de Señal , Adulto , Células Cultivadas , Femenino , Humanos , Masculino , Activación Plaquetaria/efectos de los fármacos , Activación Plaquetaria/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación
17.
Pharm Res ; 29(9): 2565-77, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22798259

RESUMEN

PURPOSE: In order to investigate Poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NP) as potential vehicles for efficient tumor antigen (TA) delivery to dendritic cells (DC), this study aimed to optimize encapsulation/release kinetics before determining immunogenicity of antigen-containing NP. METHODS: Various techniques were used to liberate TA from cell lines. Single (gp100) and multiple (B16-tumor lysate containing gp100) antigens were encapsulated within differing molecular weight PLGA co-polymers. Differences in morphology, encapsulation/release and biologic potency were studied. Findings were adopted to encapsulate fresh tumor lysate from patients with advanced tumors and compare stimulation of tumor infiltrating lymphocytes (TIL) against that achieved by soluble lysate. RESULTS: Four cycles of freeze-thaw + 15 s sonication resulted in antigen-rich lysates without the need for toxic detergents or protease inhibitors. The 80 KDa polymer resulted in maximal release of payload and favorable production of immunostimulatory IL-2 and IFN-γ. NP-mediated antigen delivery led to increased IFN-γ and decreased immunoinhibitory IL-10 synthesis when compared to soluble lysate. CONCLUSIONS: Four cycles of freeze-thaw followed by 15 s sonication is the ideal technique to obtain complex TA for encapsulation. The 80 KDa polymer has the most promising combination of release kinetics and biologic potency. Encapsulated antigens are immunogenic and evoke favorable TIL-mediated anti-tumor responses.


Asunto(s)
Antígenos de Neoplasias/inmunología , Ácido Láctico/química , Nanopartículas , Ácido Poliglicólico/química , Animales , Antígenos de Neoplasias/administración & dosificación , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Rastreo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
18.
J Invest Dermatol ; 132(1): 188-97, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21881587

RESUMEN

Cutaneous T-cell lymphoma (CTCL) is a heterogeneous non-Hodgkin's lymphoma that may variably involve the skin, lymph nodes, and peripheral blood. Malignant burden ranges from cutaneous patches and plaques with little evidence of blood involvement to erythroderma often in association with frank leukemia, as in Sézary syndrome. Toward a better understanding of the pathogenesis of this CD4+ T-cell malignancy, we conducted a high-resolution genomic analysis combining DNA (23 samples) and mRNA (12 samples) data of peripheral blood isolates from CTCL patients across a spectrum of stages. Strikingly, even patients with limited involvement, e.g., normal CD4 counts, contained significant copy-number alterations. Defining genomic characteristics of CTCL blood involvement included gains on 8q and 17q, and deletions on 17p and chromosome 10. A consensus analysis of 108 leukemic CTCL samples demonstrated global similarities among patients with varied blood involvement, narrowing 38 of 62 loci. Toward an annotated framework for in vitro testing, we also characterized genomic alterations in five CTCL cell lines (HH, HUT78, PNO, SeAx, and Sez4), revealing intact core features of leukemic CTCL. Together, these studies produce the most comprehensive view of the leukemic CTCL genome to date, with implications for pathogenesis, molecular classification, and potential future therapeutic developments.


Asunto(s)
Dosificación de Gen/genética , Genómica , Leucemia/genética , Linfoma Cutáneo de Células T/genética , Neoplasias Cutáneas/genética , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Femenino , Dosificación de Gen/inmunología , Genes Supresores de Tumor , Genoma Humano , Humanos , Leucemia/inmunología , Linfoma Cutáneo de Células T/inmunología , Masculino , Persona de Mediana Edad , Técnicas de Amplificación de Ácido Nucleico , Oncogenes/genética , Neoplasias Cutáneas/inmunología
19.
Nanomedicine ; 7(1): 1-10, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20692374

RESUMEN

Encapsulation of tumor-associated antigens in polymer nanoparticles (NP) is a promising approach to enhance efficiency of antigen delivery for anti-tumor vaccines. Head and neck squamous carcinoma (HNSCC) cell lines were initially used to generate tumor-associated antigens (TAA)-containing poly (lactic-co-glycolic acid) (PLGA) NP; encapsulation efficiency and release kinetics were profiled. Findings were adopted to entrap fresh tumor lysate from five patients with advanced HNSCC. To test the hypothesis that NP enhance antigen presentation, dendritic cell (DC) produced from patient blood monocyte precursors were loaded with either the un-encapsulated or NP-encapsulated versions of tumor lysates. These were used to stimulate freshly-isolated autologous CD8+ T cells. In four of five patients, anti-tumor CD8+ T cells showed significantly increased immunostimulatory IFN-γ (p=0.071) or decreased immmunoinhibitory IL-10 production (p=0.0004) associated with NP-mediated antigen delivery. The observations represent an enabling step in the production of clinically-translatable, inexpensive, highly-efficient, and personalized polymer-based immunotherapy for solid organ malignancies. FROM THE CLINICAL EDITOR: Enhancing the antigen presentation may be a viable approach to increase the efficiency of tumor cell directed cytotoxicity via immune mechanisms. This study presents an example for this using head and neck cancer cell lines and nanotechnology-based encapsulated antigen presentation to dendritic cells. The observed CD8+ T-cell response was significantly enhanced. This method may pave the way to a highly efficient cancer cell elimination method with minimal to no toxicity.


Asunto(s)
Células Dendríticas/citología , Inmunoterapia/métodos , Nanopartículas/química , Neoplasias/terapia , Polímeros/química , Línea Celular Tumoral , Células Cultivadas , Células Dendríticas/fisiología , Humanos , Técnicas In Vitro , Microscopía Electrónica de Rastreo , Nanopartículas/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA