Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 11: 832, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32457754

RESUMEN

Inhibitors that block the programmed cell death-1 (PD-1) pathway can potentiate endogenous antitumor immunity and have markedly improved cancer survival rates across a broad range of indications. However, these treatments work for only a minority of patients. The efficacy of anti-PD-1 inhibitors may be extended by cytokines, however, the incorporation of cytokines into therapeutic regimens has significant challenges. In their natural form when administered as recombinant proteins, cytokine treatments are often associated with low response rates. Most cytokines have a short half-life which limits their exposure and efficacy. In addition, cytokines can activate counterregulatory pathways, in the case of immune-potentiating cytokines this can lead to immune suppression and thereby diminish their potential efficacy. Improving the drug-like properties of natural cytokines using protein engineering can yield synthetic cytokines with improved bioavailability and tissue targeting, allowing for enhanced efficacy and reduced off-target effects. Using structure guided engineering we have designed a novel class of antibody-cytokine fusion proteins consisting of a PD-1 targeting antibody fused together with an interleukin-21 (IL-21) cytokine mutein. Our bifunctional fusion proteins can block PD-1/programmed death-ligand 1 (PD-L1) interaction whilst simultaneously delivering IL-21 cytokine to PD-1 expressing T cells. Targeted delivery of IL-21 can improve T cell function in a manner that is superior to anti-PD-1 monotherapy. Fusion of engineered IL-21 variants to anti-PD1 antibodies can improve the drug-like properties of IL-21 cytokine leading to improved cytokine serum half-life allowing for less frequent dosing. In addition, we show that targeted delivery of IL-21 can minimize any potential detrimental effect on local antigen-presenting cells. A highly attenuated IL-21 mutein variant (R9E:R76A) fused to a PD-1 antibody provides protection in a humanized mouse model of cancer that is refractory to anti-PD-1 monotherapy. Collectively, our preclinical data demonstrate that this approach may improve upon and extend the utility of anti-PD-1 therapeutics currently in the clinic.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Linfocitos T CD8-positivos/inmunología , Inmunoterapia , Interleucinas/uso terapéutico , Neoplasias/terapia , Animales , Anticuerpos Monoclonales/inmunología , Antígeno B7-H1/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucinas/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Neoplasias/inmunología , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico
2.
J Immunother Cancer ; 5: 33, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28428882

RESUMEN

BACKGROUND: Studies assessing immune parameters typically utilize human PBMCs or murine splenocytes to generate data that is interpreted as representative of immune status. Using splenocytes, we have shown memory CD4-T cells that expand following systemic immunostimulatory therapies undergo rapid IFNg-mediated activation induced cell death (AICD) resulting in a net loss of total CD4-T cells which correlates with elevated PD-1 expression. This is in contrast to CD8-T cells which expand with minimal PD-1 upregulation and apoptosis. In this study we expand upon our previous work by evaluating CD4 and CD8-T cell phenotype and distribution in peripheral organs which are more representative of immune responses occurring at metastatic sites following immunotherapy. METHODS: Phenotypic assessment of T cells in both lymphoid (spleen and LN) as well as peripheral organs (liver and lungs) in control and immunotherapy treated mice was performed to survey the impact of location on memory phenotype and activation marker status. Peripheral blood from patients undergoing systemic high dose IL-2 was also assessed for expression of PD-1 and memory phenotype. RESULTS: Here we reveal that, similar to what occurs in the spleen and lymph nodes, CD4-T cell numbers decreased while CD8-T cells expanded at these peripheral sites. In contrast to having differential expression of PD-1 as occurs in the spleen, both CD4 and CD8-T cells had significantly elevated levels of PD-1 in both the liver and lungs. Further analysis correlated PD-1 expression to CD62Llow (T effector/effector memory,TE/EM) expression which are more prevalent in CD4-T cells in general as well as CD8-T cells in peripheral organs. Similar elevated PD-1 expression on TE/EM cells was observed in patients undergoing systemic high-dose IL-2 therapy. CONCLUSIONS: These data highlight PD-1 expressing and/or TE/EM subsets of T cells in circulation as more representative of cells at immune sites and underscore the importance of valuation both in lymphoid as well as target organs when making determinations about immune status. TRIAL REGISTRATION: ClinicalTrials.gov NCT01416831. Registered August 12, 2011.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunoterapia/métodos , Bazo/inmunología , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Fenotipo
3.
Oncoimmunology ; 5(2): e1075114, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27057446

RESUMEN

Monoclonal antibodies (mAbs) targeting coinhibitory molecules such as PD-1, PD-L1 and CTLA-4 are increasingly used as targets of therapeutic intervention against cancer. While these targets have led to a critical paradigm shift in treatments for cancer, these approaches are also plagued with limitations owing to cancer immune evasion mechanisms and adverse toxicities associated with continuous treatment. It has been difficult to reproduce and develop interventions to these limitations preclinically due to poor reagent efficacy and reagent xenogenecity not seen in human trials. In this study, we investigated adverse effects of repeated administration of PD-1 and PD-L1 mAbs in the murine 4T1 mammary carcinoma model. We observed rapid and fatal hypersensitivity reactions in tumor bearing mice within 30-60 min after 4-5 administrations of PD-L1 or PD-1 mAb but not CTLA-4 antibody treatment. These events occurred only in mice bearing the highly inflammatory 4T1 tumor and did not occur in mice bearing non-inflammatory tumors. We observed that mortality was associated with systemic accumulation of IgG1 antibodies, antibodies specific to the PD-1 mAb, and accumulation of Gr-1high neutrophils in lungs which have been implicated in the IgG mediated pathway of anaphylaxis. Anti-PD-1 associated toxicities were alleviated when PD-1 blockade was combined with the therapeutic HSP90 inhibitor, ganetespib, which impaired immune responses toward the xenogeneic PD-1 mAb. This study highlights a previously uncharacterized fatal hypersensitivity exacerbated by the PD-1/PD-L1 axis in the broadly used 4T1 tumor model as well as an interesting relationship between this particular class of checkpoint blockade and tumor-dependent immunomodulation.

4.
Clin Cancer Res ; 22(17): 4328-40, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26979392

RESUMEN

PURPOSE: Previous studies demonstrate that intratumoral CpG immunotherapy in combination with radiotherapy acts as an in-situ vaccine inducing antitumor immune responses capable of eradicating systemic disease. Unfortunately, most patients fail to respond. We hypothesized that immunotherapy can paradoxically upregulate immunosuppressive pathways, a phenomenon we term "rebound immune suppression," limiting clinical responses. We further hypothesized that the immunosuppressive enzyme indolamine-2,3-dioxygenase (IDO) is a mechanism of rebound immune suppression and that IDO blockade would improve immunotherapy efficacy. EXPERIMENTAL DESIGN: We examined the efficacy and immunologic effects of a novel triple therapy consisting of local radiotherapy, intratumoral CpG, and systemic IDO blockade in murine models and a pilot canine clinical trial. RESULTS: In murine models, we observed marked increase in intratumoral IDO expression after treatment with radiotherapy, CpG, or other immunotherapies. The addition of IDO blockade to radiotherapy + CpG decreased IDO activity, reduced tumor growth, and reduced immunosuppressive factors, such as regulatory T cells in the tumor microenvironment. This triple combination induced systemic antitumor effects, decreasing metastases, and improving survival in a CD8(+) T-cell-dependent manner. We evaluated this novel triple therapy in a canine clinical trial, because spontaneous canine malignancies closely reflect human cancer. Mirroring our mouse studies, the therapy was well tolerated, reduced intratumoral immunosuppression, and induced robust systemic antitumor effects. CONCLUSIONS: These results suggest that IDO maintains immune suppression in the tumor after therapy, and IDO blockade promotes a local antitumor immune response with systemic consequences. The efficacy and limited toxicity of this strategy are attractive for clinical translation. Clin Cancer Res; 22(17); 4328-40. ©2016 AACR.


Asunto(s)
Inmunomodulación/efectos de los fármacos , Terapia de Inmunosupresión , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Neoplasias/inmunología , Neoplasias/metabolismo , Animales , Modelos Animales de Enfermedad , Perros , Activación Enzimática , Femenino , Melanoma Experimental , Ratones , Neoplasias/mortalidad , Neoplasias/terapia , Oligodesoxirribonucleótidos/administración & dosificación , Radioinmunoterapia/métodos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Resultado del Tratamiento , Microambiente Tumoral/inmunología
5.
Cancer Immunol Immunother ; 64(12): 1541-52, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26423422

RESUMEN

We have demonstrated that immunostimulatory therapies such as interleukin-2 (IL-2) and anti-CD40 (αCD40) can be combined to deliver synergistic anti-tumor effects. While this strategy has shown success, efficacy varies depending on a number of factors including tumor type and severe toxicities can be seen. We sought to determine whether blockade of negative regulators such as cytotoxic T lymphocyte antigen-4 (CTLA-4) could simultaneously prolong CD8(+) T cell responses and augment T cell anti-tumor effects. We devised a regimen in which anti-CTLA-4 was administered late so as to delay contraction and minimize toxicities. This late administration both enhanced and prolonged CD8 T cell activation without the need for additional IL-2. The quality of the T cell response was improved with increased frequency of effector/effector memory phenotype cells along with improved lytic ability and bystander expansion. This enhanced CD8 response translated to improved anti-tumor responses both at the primary and metastatic sites. Importantly, toxicities were not exacerbated with combination. This study provides a platform for rational design of immunotherapy combinations to maximize anti-tumor immunity while minimizing toxicities.


Asunto(s)
Antígenos CD8/metabolismo , Antígeno CTLA-4/antagonistas & inhibidores , Inmunoterapia , Neoplasias/inmunología , Proteínas Recombinantes de Fusión/farmacología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Interleucina-2/farmacología , Ratones , Ratones Endogámicos C57BL , Neoplasias/mortalidad , Neoplasias/terapia , Proteínas Recombinantes de Fusión/administración & dosificación
6.
J Immunol ; 195(8): 4010-9, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26363055

RESUMEN

Increasing evidence supports the hypothesis that cancer stem cells (CSCs) are resistant to antiproliferative therapies, able to repopulate tumor bulk, and seed metastasis. NK cells are able to target stem cells as shown by their ability to reject allogeneic hematopoietic stem cells but not solid tissue grafts. Using multiple preclinical models, including NK coculture (autologous and allogeneic) with multiple human cancer cell lines and dissociated primary cancer specimens and NK transfer in NSG mice harboring orthotopic pancreatic cancer xenografts, we assessed CSC viability, CSC frequency, expression of death receptor ligands, and tumor burden. We demonstrate that activated NK cells are capable of preferentially killing CSCs identified by multiple CSC markers (CD24(+)/CD44(+), CD133(+), and aldehyde dehydrogenase(bright)) from a wide variety of human cancer cell lines in vitro and dissociated primary cancer specimens ex vivo. We observed comparable effector function of allogeneic and autologous NK cells. We also observed preferential upregulation of NK activation ligands MICA/B, Fas, and DR5 on CSCs. Blocking studies further implicated an NKG2D-dependent mechanism for NK killing of CSCs. Treatment of orthotopic human pancreatic cancer tumor-bearing NSG mice with activated NK cells led to significant reductions in both intratumoral CSCs and tumor burden. Taken together, these data from multiple preclinical models, including a strong reliance on primary human cancer specimens, provide compelling preclinical evidence that activated NK cells preferentially target cancer cells with a CSC phenotype, highlighting the translational potential of NK immunotherapy as part of a combined modality approach for refractory solid malignancies.


Asunto(s)
Antígenos CD/inmunología , Inmunidad Celular , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Células Madre Neoplásicas/inmunología , Animales , Línea Celular Tumoral , Femenino , Humanos , Células Asesinas Naturales/patología , Ratones , Ratones Endogámicos NOD , Neoplasias/patología , Células Madre Neoplásicas/patología
7.
Immunity ; 43(2): 240-50, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26231116

RESUMEN

Primary T cell activation involves the integration of three distinct signals delivered in sequence: (1) antigen recognition, (2) costimulation, and (3) cytokine-mediated differentiation and expansion. Strong immunostimulatory events such as immunotherapy or infection induce profound cytokine release causing "bystander" T cell activation, thereby increasing the potential for autoreactivity and need for control. We show that during strong stimulation, a profound suppression of primary CD4(+) T-cell-mediated immune responses ensued and was observed across preclinical models and patients undergoing high-dose interleukin-2 (IL-2) therapy. This suppression targeted naive CD4(+) but not CD8(+) T cells and was mediated through transient suppressor of cytokine signaling-3 (SOCS3) inhibition of the STAT5b transcription factor signaling pathway. These events resulted in complete paralysis of primary CD4(+) T cell activation, affecting memory generation and induction of autoimmunity as well as impaired viral clearance. These data highlight the critical regulation of naive CD4(+) T cells during inflammatory conditions.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Herpesviridae/terapia , Inmunoterapia/métodos , Melanoma/terapia , Muromegalovirus/inmunología , Neoplasias Cutáneas/terapia , Animales , Antígenos/inmunología , Diferenciación Celular/genética , Proliferación Celular/genética , Anergia Clonal , Femenino , Infecciones por Herpesviridae/inmunología , Humanos , Inmunidad Celular , Memoria Inmunológica , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-2/administración & dosificación , Melanoma/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Ensayos Clínicos Controlados Aleatorios como Asunto , Transducción de Señal , Neoplasias Cutáneas/inmunología , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Carga Viral/inmunología
8.
J Exp Med ; 211(12): 2373-83, 2014 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-25366964

RESUMEN

Aging is a contributing factor in cancer occurrence. We recently demonstrated that systemic immunotherapy (IT) administration in aged, but not young, mice resulted in induction of rapid and lethal cytokine storm. We found that aging was accompanied by increases in visceral fat similar to that seen in young obese (ob/ob or diet-induced obese [DIO]) mice. Yet, the effects of aging and obesity on inflammatory responses to immunotherapeutics are not well defined. We determine the effects of adiposity on systemic IT tolerance in aged compared with young obese mice. Both young ob/ob- and DIO-generated proinflammatory cytokine levels and organ pathologies are comparable to those in aged ad libitum mice after IT, culminating in lethality. Young obese mice exhibited greater ratios of M1/M2 macrophages within the peritoneal and visceral adipose tissues and higher percentages of TNF(+) macrophages in response to αCD40/IL-2 as compared with young lean mice. Macrophage depletion or TNF blockade in conjunction with αCD40/IL-2 prevented cytokine storms in young obese mice and protected from lethality. Calorie-restricted aged mice contain less visceral fat and displayed reduced cytokine levels, protection from organ pathology, and protection from lethality upon αCD40/IL-2 administration. Our data demonstrate that adiposity is a critical factor in the age-associated pathological responses to systemic anti-cancer IT.


Asunto(s)
Adiposidad/inmunología , Envejecimiento/inmunología , Citocinas/inmunología , Inmunoterapia/métodos , Animales , Restricción Calórica , Citocinas/sangre , Femenino , Citometría de Flujo , Humanos , Mediadores de Inflamación/sangre , Mediadores de Inflamación/inmunología , Grasa Intraabdominal/inmunología , Grasa Intraabdominal/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Obesos , Neoplasias/inmunología , Neoplasias/terapia , Obesidad/genética , Obesidad/inmunología , Obesidad/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
9.
PLoS One ; 9(8): e102709, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25119341

RESUMEN

We have previously demonstrated that immunotherapy combining agonistic anti-CD40 and IL-2 (IT) results in synergistic anti-tumor effects. IT induces expansion of highly cytolytic, antigen-independent "bystander-activated" (CD8(+)CD44high) T cells displaying a CD25(-)NKG2D(+) phenotype in a cytokine dependent manner, which were responsible for the anti-tumor effects. While much attention has focused on CD4(+) T cell help for antigen-specific CD8(+) T cell expansion, little is known regarding the role of CD4(+) T cells in antigen-nonspecific bystander-memory CD8(+) T cell expansion. Utilizing CD4 deficient mouse models, we observed a significant expansion of bystander-memory T cells following IT which was similar to the non-CD4 depleted mice. Expanded bystander-memory CD8(+) T cells upregulated PD-1 in the absence of CD4(+) T cells which has been published as a hallmark of exhaustion and dysfunction in helpless CD8(+) T cells. Interestingly, compared to CD8(+) T cells from CD4 replete hosts, these bystander expanded cells displayed comparable (or enhanced) cytokine production, lytic ability, and in vivo anti-tumor effects suggesting no functional impairment or exhaustion and were enriched in an effector phenotype. There was no acceleration of the post-IT contraction phase of the bystander memory CD8(+) response in CD4-depleted mice. The response was independent of IL-21 signaling. These results suggest that, in contrast to antigen-specific CD8(+) T cell expansion, CD4(+) T cell help is not necessary for expansion and activation of antigen-nonspecific bystander-memory CD8(+) T cells following IT, but may play a role in regulating conversion of these cells from a central memory to effector phenotype. Additionally, the expression of PD-1 in this model appears to be a marker of effector function and not exhaustion.


Asunto(s)
Antígenos CD4/genética , Linfocitos T CD8-positivos/efectos de los fármacos , Interleucina-2/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Antígenos CD40/administración & dosificación , Antígenos CD40/farmacología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Citocinas/metabolismo , Citometría de Flujo , Inmunoterapia , Interleucina-2/administración & dosificación , Interleucinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Inmunológicos , Receptor de Muerte Celular Programada 1/metabolismo , Transducción de Señal
10.
J Immunol ; 193(4): 1709-16, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25000978

RESUMEN

Because of increasing interest in the removal of immunosuppressive pathways in cancer, the combination of IL-2 with Abs to neutralize TGF-ß, a potent immunosuppressive cytokine, was assessed. Combination immunotherapy resulted in significantly greater antitumor effects. These were correlated with significant increases in the numbers and functionality of NK cells, NK cell progenitors, and activated CD8 T cells, resulting in the observed antitumor effects. Combination immunotherapy also was accompanied by lesser toxicities than was IL-2 therapy alone. Additionally, we observed a dual competition between NK cells and activated CD8 T cells such that, after immunotherapy, the depletion of either effector population resulted in the increased total expansion of the other population and compensatory antitumor effects. This study demonstrates the efficacy of this combination immunotherapeutic regimen as a promising cancer therapy and illustrates the existence of potent competitive regulatory pathways between NK cells and CD8 T cells in response to systemic activation.


Asunto(s)
Anticuerpos Neutralizantes/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Interleucina-2/uso terapéutico , Células Asesinas Naturales/inmunología , Neoplasias/terapia , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Linfocitos T CD8-positivos/trasplante , Línea Celular Tumoral , Proteína Ligando Fas/inmunología , Femenino , Inmunoterapia/métodos , Células Asesinas Naturales/trasplante , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subfamilia A de Receptores Similares a Lectina de Células NK/biosíntesis , Subfamília C de Receptores Similares a Lectina de Células NK/biosíntesis , Subfamilia K de Receptores Similares a Lectina de Células NK/biosíntesis , Linfocitos T Reguladores/inmunología , Receptor fas/inmunología
11.
Clin Exp Immunol ; 175(1): 79-91, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23937663

RESUMEN

Primary viral infections induce activation of CD8(+) T cells responsible for effective resistance. We sought to characterize the nature of the CD8(+) T cell expansion observed after primary viral infection with influenza. Infection of naive mice with different strains of influenza resulted in the rapid expansion of memory CD8(+) T cells exhibiting a unique bystander phenotype with significant up-regulation of natural killer group 2D (NKG2D), but not CD25, on the CD44(high) CD8(+) T cells, suggesting an antigen non-specific phenotype. We further confirmed the non-specificity of this phenotype on ovalbumin-specific (OT-I) CD8(+) T cells, which are not specific to influenza. These non-specific CD8(+) T cells also displayed increased lytic capabilities and were observed primarily in the lung. Thus, influenza infection was shown to induce a rapid, antigen non-specific memory T cell expansion which is restricted to the specific site of inflammation. In contrast, CD8(+) T cells of a similar phenotype could be observed in other organs following administration of systemic agonistic anti-CD40 and interleukin-2 immunotherapy, demonstrating that bystander expansion in multiple sites is possible depending on whether the nature of activation is either acute or systemic. Finally, intranasal blockade of NKG2D resulted in a significant increase in viral replication early during the course of infection, suggesting that NKG2D is a critical mediator of anti-influenza responses prior to the initiation of adaptive immunity. These results characterize further the local bystander expansion of tissue-resident, memory CD8(+) T cells which, due to their early induction, may play an important NKG2D-mediated, antigen non-specific role during the early stages of viral infection.


Asunto(s)
Efecto Espectador/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Virus de la Influenza A/fisiología , Pulmón/inmunología , Infecciones por Orthomyxoviridae/inmunología , Replicación Viral/inmunología , Animales , Antígenos CD40/antagonistas & inhibidores , Antígenos CD40/genética , Antígenos CD40/inmunología , Linfocitos T CD8-positivos/patología , Inmunoterapia/métodos , Interleucina-2/genética , Interleucina-2/inmunología , Interleucina-2/farmacología , Pulmón/patología , Pulmón/virología , Ratones , Ratones Transgénicos , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/terapia , Replicación Viral/genética
12.
J Exp Med ; 210(11): 2223-37, 2013 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-24081947

RESUMEN

Cancer commonly occurs in the elderly and immunotherapy (IT) is being increasingly applied to this population. However, the majority of preclinical mouse tumor models assessing potential efficacy and toxicities of therapeutics use young mice. We assessed the impact of age on responses to systemic immune stimulation. In contrast to young mice, systemic cancer IT regimens or LPS given to aged mice resulted in rapid and lethal toxicities affecting multiple organs correlating with heightened proinflammatory cytokines systemically and within the parenchymal tissues. This inflammatory response and increased morbidity with age was independent of T cells or NK cells. However, prior in vivo depletion of macrophages in aged mice resulted in lesser cytokine levels, increased survival, and decreased liver histopathology. Furthermore, macrophages from aged mice and normal human elderly volunteers displayed heightened TNF and IL-6 production upon in vitro stimulation. Treatment of both TNF knockout mice and in vivo TNF blockade in aged mice resulted in significant increases in survival and lessened pathology. Importantly, TNF blockade in tumor-bearing, aged mice receiving IT displayed significant anti-tumor effects. These data demonstrate the critical role of macrophages in the age-associated hyper-inflammatory cytokine responses to systemic immunostimulation and underscore the importance of performing preclinical assessments in aged mice.


Asunto(s)
Envejecimiento/inmunología , Envejecimiento/patología , Inmunoterapia , Inflamación/patología , Neoplasias/inmunología , Neoplasias/patología , Animales , Antígenos CD40/inmunología , Citocinas/toxicidad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Neoplasias/terapia , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
13.
Front Oncol ; 3: 197, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23898464

RESUMEN

Cancer immunotherapy has emerged as a mainstream therapy option in the battle against cancer. Pre-clinical data demonstrates the ability of immunotherapy to harness the immune system to fight disseminated malignancy. Clinical translation has failed to recapitulate the promising results of pre-clinical studies although there have been some successes. In this review we explore some of the short-comings of cancer immunotherapy that have limited successful clinical translation. We will give special consideration to what we consider the most formidable hurdle to successful cancer immunotherapy: tumor-induced immune suppression and immune escape. We will discuss the need for antigen-specific immune responses for successful immunotherapy but also consider the need for antigen specificity as an Achilles heel of immunotherapy given tumor heterogeneity, immune editing, and antigen loss. Finally, we will discuss how combinatorial strategies may overcome some of the pitfalls of antigen specificity and highlight recent studies from our lab which suggest that the induction of antigen non-specific immune responses may also produce robust anti-tumor effects and bypass the need for antigen specificity.

14.
Oncoimmunology ; 1(7): 1208-1210, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23170282

RESUMEN

For a long time, cancer immunotherapy has focused on the induction of tumor-specific T cell-mediated immune responses. Now, a mounting body of evidence indicates that efficient anticancer immune responses also rely on innate immunity. Tietze et al. have recently elucidated an antigen-nonspecific role for memory CD8(+) T cells in cytokine-based cancer immunotherapy.

15.
PLoS One ; 7(10): e48049, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23133545

RESUMEN

The primary tumor represents a potential source of antigens for priming immune responses for disseminated disease. Current means of debulking tumors involves the use of cytoreductive conditioning that impairs immune cells or removal by surgery. We hypothesized that activation of the immune system could occur through the localized release of tumor antigens and induction of tumor death due to physical disruption of tumor architecture and destruction of the primary tumor in situ. This was accomplished by intratumor injection of magneto-rheological fluid (MRF) consisting of iron microparticles, in Balb/c mice bearing orthotopic 4T1 breast cancer, followed by local application of a magnetic field resulting in immediate coalescence of the particles, tumor cell death, slower growth of primary tumors as well as decreased tumor progression in distant sites and metastatic spread. This treatment was associated with increased activation of DCs in the draining lymph nodes and recruitment of both DCs and CD8(+)T cells to the tumor. The particles remained within the tumor and no toxicities were observed. The immune induction observed was significantly greater compared to cryoablation. Further anti-tumor effects were observed when MRF/magnet therapy was combined with systemic low dose immunotherapy. Thus, mechanical disruption of the primary tumor with MRF/magnetic field application represents a novel means to induce systemic immune activation in cancer.


Asunto(s)
Antineoplásicos/farmacología , Hierro/farmacología , Neoplasias Mamarias Animales/radioterapia , Animales , Linfocitos T CD8-positivos/citología , Células Dendríticas/citología , Progresión de la Enfermedad , Femenino , Citometría de Flujo/métodos , Sistema Inmunológico , Inmunoterapia/métodos , Campos Magnéticos , Magnetismo , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/terapia , Ratones , Ratones Endogámicos BALB C , Necrosis , Metástasis de la Neoplasia , Neoplasias/inmunología , Células Madre
16.
J Immunotoxicol ; 9(3): 248-58, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22734880

RESUMEN

Immunotherapy in the treatment of cancer is increasing, particularly with the recent FDA approval of sipuleucel-T and ipilimumab. The efficacy of anti-tumor immunotherapies has been modest compared to their theoretical and pre-clinical promise. This review evaluates the promise and pitfalls of immunotherapy and highlight some of the obstacles to improving anti-tumor immunotherapy: the need for technical refinement of therapies, the need for an increased understanding of how best to combine therapies with traditional cytotoxic therapies, the inability of patients to mount an effective immune response either due to disease burden or tumor induced immune suppression, the significant toxicities associated with many immunotherapies, and the lack of strongly immunogenic antigens required by many therapies. Further, antigen-non-specific immunotherapies, including cytokines such as interleukins and interferons, immuno-stimulatory agents such as CpG oligonucleotides, or BCG, antibodies targeted against receptors such as the agonistic CD40 or inhibitory CTLA-4 antibodies, and enzyme inhibitors such as those targeting cyclo-oxygenase or indolamine-2,3-dioxygenase are discussed. In addition, potential mechanisms of these therapies such as direct anti-tumor effects, reversal of immune suppression, activation of innate immunity, and antigen-non-specific T-cell activation are reviewed. We also appraise the potential of these antigen-non-specific therapies to overcome some of the previously described pitfalls of immunotherapy. Lastly, we discuss a recent series of studies from our laboratory demonstrating the importance of antigen-non-specific 'bystander activation' of memory T-lymphocytes by immunomodulatory therapies such as interleukin-2 and the antigen-non-specific anti-tumor effects of these cells.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Adyuvantes Inmunológicos/uso terapéutico , Anticuerpos Antineoplásicos/inmunología , Anticuerpos Antineoplásicos/uso terapéutico , Efecto Espectador/efectos de los fármacos , Efecto Espectador/inmunología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Citocinas/inmunología , Citocinas/uso terapéutico , Humanos , Memoria Inmunológica/efectos de los fármacos , Mycobacterium bovis/inmunología , Oligodesoxirribonucleótidos/uso terapéutico , Linfocitos T/inmunología
17.
Blood ; 119(13): 3073-83, 2012 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-22251483

RESUMEN

Memory T cells exhibit tremendous antigen specificity within the immune system and accumulate with age. Our studies reveal an antigen-independent expansion of memory, but not naive, CD8(+) T cells after several immunotherapeutic regimens for cancer resulting in a distinctive phenotype. Signaling through T-cell receptors (TCRs) or CD3 in both mouse and human memory CD8(+) T cells markedly up-regulated programmed death-1 (PD-1) and CD25 (IL-2 receptor α chain), and led to antigen-specific tumor cell killing. In contrast, exposure to cytokine alone in vitro or with immunotherapy in vivo did not up-regulate these markers but resulted in expanded memory CD8(+) T cells expressing NKG2D, granzyme B, and possessing broadly lytic capabilities. Blockade of NKG2D in mice also resulted in significantly diminished antitumor effects after immunotherapy. Treatment of TCR-transgenic mice bearing nonantigen expressing tumors with immunotherapy still resulted in significant antitumor effects. Human melanoma tissue biopsies obtained from patients after topically applied immunodulatory treatment resulted in increased numbers of these CD8(+) CD25(-) cells within the tumor site. These findings demonstrate that memory CD8(+) T cells can express differential phenotypes indicative of adaptive or innate effectors based on the nature of the stimuli in a process conserved across species.


Asunto(s)
Antígenos CD8/inmunología , Linfocitos T CD8-positivos/inmunología , Citocinas/uso terapéutico , Inmunoterapia/métodos , Neoplasias/terapia , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Animales , Células Cultivadas , Método Doble Ciego , Humanos , Memoria Inmunológica/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/inmunología , Placebos , Ensayos Clínicos Controlados Aleatorios como Asunto , Especificidad del Receptor de Antígeno de Linfocitos T/fisiología , Factores de Tiempo
18.
Int Rev Immunol ; 30(5-6): 238-93, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22053969

RESUMEN

Due to its immunogenecity and evidence of immune responses resulting in tumor regression, metastatic melanoma has been the target for numerous immunotherapeutic approaches. Unfortunately, based on the clinical outcomes, even the successful induction of tumor-specific responses does not correlate with efficacy. Immunotherapies can be divided into antigen-specific approaches, which seek to induce T cells specific to one or several known tumor associated antigens (TAA), or with antigen-nonspecific approaches, which generally activate T cells to become nonspecifically lytic effectors. Here the authors critically review the different immunotherapeutic approaches in melanoma.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia/métodos , Melanoma/inmunología , Melanoma/terapia , Linfocitos T/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Vacunas contra el Cáncer/inmunología , Niño , Humanos , Inmunoterapia Adoptiva , Melanoma/patología , Persona de Mediana Edad , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento , Adulto Joven
19.
Plant J ; 55(4): 580-95, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18435823

RESUMEN

SUMMARY: The plant hormone ethylene negatively regulates bacterial infection and nodule formation in legumes in response to symbiotic rhizobia, but the molecular mechanism(s) of ethylene action in symbiosis remain obscure. We have identified and characterized multiple mutant alleles of the MtSkl1 gene, which controls both ethylene sensitivity and nodule numbers. We show that this locus encodes the Medicago truncatula ortholog of the Arabidopsis ethylene signaling protein EIN2. In addition to the well-characterized role of MtSkl1 in rhizobial symbiosis, we show that MtSkl1 is involved in regulating early phases of the symbiotic interaction with mycorrhizal fungi, and in mediating root responses to cytokinin. MtSkl1 also functions in the defense against Rhizoctonia solani and Phytophthora medicaginis, with the latter interaction likely to involve positive feedback amplification of ethylene biosynthesis. Overexpression of the C-terminal domain of MtEIN2 is sufficient to block nodulation responses, consistent with previous reports in Arabidopsis on the activation of ethylene signaling. This same C-terminal region is uniquely conserved throughout the EIN2 homologs of angiosperms, which is consistent with its role as a higher plant-specific innovation essential to EIN2 function.


Asunto(s)
Proteínas de Arabidopsis/fisiología , Medicago truncatula/fisiología , Enfermedades de las Plantas/microbiología , Proteínas de Plantas/fisiología , Receptores de Superficie Celular/fisiología , Simbiosis/fisiología , Envejecimiento , Citocininas/metabolismo , Fabaceae/microbiología , Fabaceae/fisiología , Flores/fisiología , Homeostasis , Medicago truncatula/crecimiento & desarrollo , Medicago truncatula/microbiología , Raíces de Plantas/fisiología , Rhizobium/fisiología , Plantones/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...