Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 31(3): 686-700, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36641624

RESUMEN

Several studies have shown the influence of commensal microbes on T cell function, specifically in the setting of checkpoint immunotherapy for cancer. In this study, we investigated how vancomycin-induced gut microbiota dysbiosis affects chimeric antigen receptor (CAR) T immunotherapy using multiple preclinical models as well as clinical correlates. In two murine tumor models, hematopoietic CD19+-A20 lymphoma and CD19+-B16 melanoma, mice receiving vancomycin in combination with CD19-directed CAR T cell (CART-19) therapy displayed increased tumor control and tumor-associated antigens (TAAs) cross-presentation compared with CART-19 alone. Fecal microbiota transplant from human healthy donors to pre-conditioned mice recapitulated the results obtained in naive gut microbiota mice. Last, B cell acute lymphoblastic leukemia patients treated with CART-19 and exposed to oral vancomycin showed higher CART-19 peak expansion compared with unexposed patients. These results substantiate the role of the gut microbiota on CAR T cell therapy and suggest that modulation of the gut microbiota using vancomycin may improve outcomes after CAR T cell therapy across tumor types.


Asunto(s)
Microbioma Gastrointestinal , Receptores Quiméricos de Antígenos , Humanos , Ratones , Animales , Receptores de Antígenos de Linfocitos T/genética , Reactividad Cruzada , Vancomicina/farmacología , Inmunoterapia , Linfocitos T , Inmunoterapia Adoptiva/métodos , Receptores Quiméricos de Antígenos/genética , Antígenos CD19
2.
Oncologist ; 27(9): 799-808, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-35666292

RESUMEN

BACKGROUND: For patients with melanoma, gastrointestinal immune-related adverse events are common after receipt of anti-CTLA4 therapy. These present difficult decision points regarding whether to discontinue therapy. Detailing the situations in which colitis might predict for improved survival and how this is affected by discontinuation or resumption of therapy can help guide clinical decision-making. MATERIALS AND METHODS: Patients with stage IV melanoma receiving anti-CTLA4 therapy from 2008 to 2019 were analyzed. Immune-related colitis treated with ≥50 mg prednisone or equivalent daily or secondary immunosuppression was included. Moderate colitis was defined as receipt of oral glucocorticoids only; severe colitis was defined as requiring intravenous glucocorticoids or secondary immunosuppression. The primary outcome was overall survival (OS). RESULTS: In total, 171 patients received monotherapy, and 91 received dual checkpoint therapy. In the monotherapy group, 25 patients developed colitis and a nonsignificant trend toward improved OS was observed in this group. Notably, when colitis was categorized as none, moderate or severe, OS was significantly improved for moderate colitis only. This survival difference was not present after dual checkpoint therapy. There were no differences in known prognostic variables between groups, and on multivariable analysis neither completion of all ipilimumab cycles nor resumption of immunotherapy correlated with OS, while the development of moderate colitis did significantly affect OS. CONCLUSION: This single-institution retrospective series suggests moderate colitis correlates with improved OS for patients with stage IV melanoma treated with single-agent anti-CTLA4, but not dual agent, and that this is true regardless of whether the immune-checkpoint blockade is permanently discontinued.


Asunto(s)
Colitis , Melanoma , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/tratamiento farmacológico , Humanos , Ipilimumab/efectos adversos , Melanoma/terapia , Estudios Retrospectivos , Esteroides/uso terapéutico
3.
Semin Immunol ; 35: 35-47, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29490888

RESUMEN

Angiogenesis is a hallmark of cancer and a requisite that tumors must achieve to fulfill their metabolic needs of nutrients and oxygen. As a critical step in cancer progression, the 'angiogenic switch' allows tumor cells to survive and grow, and provides them access to vasculature resulting in metastatic progression and dissemination. Tumor-dependent triggering of the angiogenic switch has critical consequences on tumor progression which extends from an increased nutrient supply and relies instead on the ability of the tumor to hijack the host immune response for the generation of a local immunoprivileged microenvironment. Tumor angiogenic-mediated establishment of endothelial anergy is responsible for this process. However, tumor endothelium can also promote immune tolerance by unbalanced expression of co-stimulatory and co-inhibitory molecules and by releasing soluble factors that restrain T cell function and induce apoptosis. In this review, we discuss the molecular properties of the tumor endothelial barrier and endothelial anergy and discuss the main immunosuppressive mechanisms triggered by the tumor endothelium. Lastly, we describe the current anti-angiogenic therapeutic landscape and how targeting tumor angiogenesis can contribute to improve clinical benefits for patients.


Asunto(s)
Células Endoteliales/fisiología , Neoplasias/inmunología , Neovascularización Patológica , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Transformación Celular Neoplásica , Humanos , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
4.
J Immunol ; 195(8): 4020-7, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26378078

RESUMEN

Mitochondria provide energy for cells via oxidative phosphorylation. Reactive oxygen species, a byproduct of this mitochondrial respiration, can damage mitochondrial DNA (mtDNA), and somatic mtDNA mutations have been found in all colorectal, ovarian, breast, urinary bladder, kidney, lung, and pancreatic tumors studied. The resulting altered mitochondrial proteins or tumor-associated mitochondrial Ags (TAMAs) are potentially immunogenic, suggesting that they may be targetable Ags for cancer immunotherapy. In this article, we show that the RENCA tumor cell line harbors TAMAs that can drive an antitumor immune response. We generated a cellular tumor vaccine by pulsing dendritic cells with enriched mitochondrial proteins from RENCA cells. Our dendritic cell-based RENCA mitochondrial lysate vaccine elicited a cytotoxic T cell response in vivo and conferred durable protection against challenge with RENCA cells when used in a prophylactic or therapeutic setting. By sequencing mtDNA from RENCA cells, we identified two mutated molecules: COX1 and ND5. Peptide vaccines generated from mitochondrial-encoded COX1 but not from ND5 had therapeutic properties similar to RENCA mitochondrial protein preparation. Thus, TAMAs can elicit effective antitumor immune responses, potentially providing a new immunotherapeutic strategy to treat cancer.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Carcinoma de Células Renales/prevención & control , Ciclooxigenasa 1/inmunología , Neoplasias Renales/prevención & control , Proteínas de la Membrana/inmunología , Proteínas Mitocondriales/inmunología , NADH Deshidrogenasa/inmunología , Neoplasias Experimentales/prevención & control , Animales , Antígenos de Neoplasias/farmacología , Vacunas contra el Cáncer/farmacología , Carcinoma de Células Renales/inmunología , Ciclooxigenasa 1/farmacología , Neoplasias Renales/inmunología , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos BALB C , Proteínas Mitocondriales/farmacología , NADH Deshidrogenasa/farmacología , Neoplasias Experimentales/inmunología
5.
Nature ; 475(7355): 226-30, 2011 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-21753853

RESUMEN

Although immune mechanisms can suppress tumour growth, tumours establish potent, overlapping mechanisms that mediate immune evasion. Emerging evidence suggests a link between angiogenesis and the tolerance of tumours to immune mechanisms. Hypoxia, a condition that is known to drive angiogenesis in tumours, results in the release of damage-associated pattern molecules, which can trigger the rejection of tumours by the immune system. Thus, the counter-activation of tolerance mechanisms at the site of tumour hypoxia would be a crucial condition for maintaining the immunological escape of tumours. However, a direct link between tumour hypoxia and tolerance through the recruitment of regulatory cells has not been established. We proposed that tumour hypoxia induces the expression of chemotactic factors that promote tolerance. Here we show that tumour hypoxia promotes the recruitment of regulatory T (T(reg)) cells through induction of expression of the chemokine CC-chemokine ligand 28 (CCL28), which, in turn, promotes tumour tolerance and angiogenesis. Thus, peripheral immune tolerance and angiogenesis programs are closely connected and cooperate to sustain tumour growth.


Asunto(s)
Hipoxia de la Célula , Quimiocinas CC/metabolismo , Tolerancia Inmunológica/inmunología , Neovascularización Patológica , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Hipoxia de la Célula/genética , Línea Celular Tumoral , Quimiocinas CC/genética , Medios de Cultivo Condicionados/farmacología , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Receptores CCR10/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Cancer Immunol Immunother ; 64(10): 1339-48, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26267042

RESUMEN

Targeting the tumor vasculature with anti-angiogenesis modalities is a bona fide validated approach that has complemented cancer treatment paradigms. Tumor vasculature antigens (TVA) can be immunologically targeted and offers multiple theoretical advantages that may enhance existing strategies against cancer. We focused on tumor endothelial marker 1 (TEM1/CD248) as a model TVA since it is broadly expressed on many different cancers. Our DNA-based vaccine approach demonstrated that CD248 can be effectively targeted immunologically; anti-tumor responses were generated in several mouse models; and CD8(+)/CD4(+) T cell responses were elicited against peptides derived from CD248 protein. Our work supports our contention that CD248 is a novel immunotherapeutic target for cancer treatment and highlights the efficient, safe and translatable use of DNA-based immunotherapy. We next briefly highlight ongoing investigations targeting CD248 with antibodies as a diagnostic imaging agent and as a therapeutic antibody in an early clinical trial. The optimal approach for generating effective DNA-based cancer vaccines for several tumor types may be a combinatorial approach that enhances immunogenicity such as combination with chemotherapy. Additional combination approaches are discussed and include those that alleviate the immunosuppressive tumor microenvironment induced by myeloid-derived suppressor cells and T regulatory cells. Targeting the tumor vasculature by CD248-based immunological modalities expands the armamentarium against cancer.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Vacunas contra el Cáncer/uso terapéutico , Endotelio Vascular/metabolismo , Inmunoterapia/métodos , Neoplasias/terapia , Neovascularización Patológica/terapia , Linfocitos T/inmunología , Animales , Antígenos CD/genética , Antígenos de Neoplasias/genética , Terapia Combinada , Modelos Animales de Enfermedad , Endotelio Vascular/inmunología , Humanos , Neoplasias/inmunología , Neovascularización Patológica/inmunología , Vacunas de ADN/uso terapéutico
7.
Cancer J ; 29(2): 84-88, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36957978

RESUMEN

ABSTRACT: It is increasingly recognized that heterogeneities in tumor response and severity of adverse effects in irradiated patients can be attributed to the tumor microenvironment and host-related factors. Among the latter, a growing body of literature in recent years has demonstrated the role of the patient's microbiome in modulating both tumor and normal tissue response to radiotherapy (RT). Upon contact with the environment after birth, the infant's gastrointestinal tract is rapidly colonized by microbiota, which is low in diversity and predominantly characterized by 2 dominant species, Actinobacteria and Proteobacteria. With time, intestinal microbiota diversity increases, and colonization of Firmicutes and Bacteroidetes becomes dominant. By the time a child reaches 3 years, the gut microbiota composition has been reshaped and is relatively similar to that of an adult. The microbiome colonizing the different body organs comprises various species and abundances, which may impact human health. Although the adult microbiome composition is thought to remain stable in health, microbiome diversity and composition respond to different environmental and pathological conditions, including pharmaceutical interventions and RT. Our review focuses on how the gut microbiota modulates normal tissue toxicity and tumor control. Readers who want to learn more about how RT shapes gut microbiome diversity and composition are referred to several excellent recently published reviews.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Adulto , Lactante , Niño , Humanos , Firmicutes , Proteobacteria , Bacteroidetes
8.
Clin Transl Radiat Oncol ; 39: 100587, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36718252

RESUMEN

Background and purpose: CD19-targeting chimeric antigen receptor T-cell (CART) therapy is a promising treatment for relapsed/refractory non-Hodgkin lymphoma, but most patients experience post-CART progression. We describe our institutional experience of salvage radiotherapy (SRT) in this setting. Materials and methods: Of 94 patients who received CART therapy from 2018 to 2020, 21 received SRT for post-CART progression. Patients were divided into two groups: locoregional disease (n = 9 [43 %], all disease encompassable within an RT field) and advanced disease (n = 12 [57 %]). Patterns of failure, progression-free survival (PFS), overall survival (OS), and toxicity were assessed. Results: Median time from CART infusion to SRT was 4.0 months (range, 0.6-11.5 months). In the locoregional disease group, 8/9 patients (89 %) were treated with comprehensive SRT to a median dose of 37.5 Gy in a median of 15 fractions. In the advanced disease group, all patients (n = 12) were treated with focal SRT to a median dose of 20.8 Gy in a median of 5 fractions. Median follow-up post-SRT was 15.2 months. In-field response was observed in 8/9 (89 %) in the locoregional disease and 8/9 (89 %) evaluable patients in the advanced disease groups. 17/18 evaluable patients (94 %) patients experienced post-SRT progression, all with a distant component. Median OS was 7.4 months; 21 months for locoregional disease versus 2.4 months for advanced disease (p = 0.0002). Median PFS was 1.1 month, and similarly poor regardless of group. No grade ≥ 3 toxicities occurred. Conclusions: SRT post-CART therapy appears safe with encouraging in-field response but high rates of out-of-field progression, even for those presenting with locoregional disease, highlighting the need for integration of novel systemic agents.

9.
Cancer Res ; 83(21): 3562-3576, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37578274

RESUMEN

Parkin is an E3 ubiquitin ligase, which plays a key role in the development of Parkinson disease. Parkin defects also occur in numerous cancers, and a growing body of evidence indicates that Parkin functions as a tumor suppressor that impedes a number of cellular processes involved in tumorigenesis. Here, we generated murine and human models that closely mimic the advanced-stage tumors where Parkin deficiencies are found to provide deeper insights into the tumor suppressive functions of Parkin. Loss of Parkin expression led to aggressive tumor growth, which was associated with poor tumor antigen presentation and limited antitumor CD8+ T-cell infiltration and activation. The effect of Parkin deficiency on tumor growth was lost following depletion of CD8+ T cells. In line with previous findings, Parkin deficiency was linked with mitochondria-associated metabolic stress, PTEN degradation, and enhanced Akt activation. Increased Akt signaling led to dysregulation of antigen presentation, and treatment with the Akt inhibitor MK2206-2HCl restored antigen presentation in Parkin-deficient tumors. Analysis of data from patients with clear cell renal cell carcinoma indicated that Parkin expression was downregulated in tumors and that low expression correlated with reduced overall survival. Furthermore, low Parkin expression correlated with reduced patient response to immunotherapy. Overall, these results identify a role for Parkin deficiency in promoting tumor immune evasion that may explain the poor prognosis associated with loss of Parkin across multiple types of cancer. SIGNIFICANCE: Parkin prevents immune evasion by regulating tumor antigen processing and presentation through the PTEN/Akt network, which has important implications for immunotherapy treatments in patients with Parkin-deficient tumors.


Asunto(s)
Presentación de Antígeno , Neoplasias , Animales , Humanos , Ratones , Inmunoterapia , Neoplasias/genética , Neoplasias/terapia , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-akt , Escape del Tumor , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
10.
Nat Med ; 28(4): 713-723, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35288695

RESUMEN

Anti-CD19 chimeric antigen receptor (CAR) T cell therapy has led to unprecedented responses in patients with high-risk hematologic malignancies. However, up to 60% of patients still experience disease relapse and up to 80% of patients experience CAR-mediated toxicities, such as cytokine release syndrome or immune effector cell-associated neurotoxicity syndrome. We investigated the role of the intestinal microbiome on these outcomes in a multicenter study of patients with B cell lymphoma and leukemia. We found in a retrospective cohort (n = 228) that exposure to antibiotics, in particular piperacillin/tazobactam, meropenem and imipenem/cilastatin (P-I-M), in the 4 weeks before therapy was associated with worse survival and increased neurotoxicity. In stool samples from a prospective cohort of CAR T cell recipients (n = 48), the fecal microbiome was altered at baseline compared to healthy controls. Stool sample profiling by 16S ribosomal RNA and metagenomic shotgun sequencing revealed that clinical outcomes were associated with differences in specific bacterial taxa and metabolic pathways. Through both untargeted and hypothesis-driven analysis of 16S sequencing data, we identified species within the class Clostridia that were associated with day 100 complete response. We concluded that changes in the intestinal microbiome are associated with clinical outcomes after anti-CD19 CAR T cell therapy in patients with B cell malignancies.


Asunto(s)
Microbioma Gastrointestinal , Síndromes de Neurotoxicidad , Receptores Quiméricos de Antígenos , Antígenos CD19 , Humanos , Inmunoterapia Adoptiva/efectos adversos , Síndromes de Neurotoxicidad/etiología , Estudios Prospectivos , Estudios Retrospectivos
11.
J Transl Med ; 9: 77, 2011 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-21609494

RESUMEN

BACKGROUND: Time-dependent chemotherapeutic agents can selectively target tumor cells in susceptible phases of the cell cycle however a fraction of tumor cells in non-vulnerable cell cycle phases remain drug-resistant. Immunotherapy represents a promising approach to overcome the limitation of phase-specific drugs and improve their clinical efficacy. Here, we investigated the potential use of anticancer chemotherapeutic drugs in combination with IL-18, a cytokine with strong immunostimulatory properties. METHODS: Four chemotherapeutic drugs commonly used in ovarian cancer were first tested for the ability to increase the immunogenicity and killing of the murine ovarian cancer cell line ID8 in vitro. Chemotherapeutric agents with measured time-dependent immune-enhancing effects were then tested for antitumor effectiveness in vivo in combination with IL-18 immunotherapy using the ID8-Vegf ovarian cancer model. RESULTS: Paclitaxel or topotecan exposure alone mediated incomplete, time-dependent killing against the murine ovarian cancer cell line ID8 in vitro, whereas carboplatin or gemcitabine mediated comprehensive, dose-dependent killing. In the plateau phase of the time-dependent killing by topotecan or paclitaxel, drug-resistant ID8 cells were more immunogenic with elevated expression of MHC-I and Fas, and increased sensitivity to CTL and Fas agonistic antibody in vitro. Moreover, the antitumor effectiveness of time-dependent agents in vivo was significantly improved with the addition of IL-18 through a T cell-dependent mechanism, while the effectiveness of drugs without significant phase specificity were not. CONCLUSIONS: Tumor immunotherapy with IL-18 can significantly augment the killing fraction of phase-specific chemotherapeutic drugs and provide survival benefit. The safety profile of IL-18 and its positive interactions with select anticancer chemotherapeutic agents strongly supports the clinical investigation of this combinatorial approach.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoterapia , Interleucina-18/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Animales , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Interleucina-18/farmacología , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Paclitaxel/farmacología , Linfocitos T/efectos de los fármacos , Factores de Tiempo , Topotecan/farmacología , Regulación hacia Arriba/efectos de los fármacos , Receptor fas/metabolismo
12.
J Immunother Cancer ; 9(2)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33563772

RESUMEN

BACKGROUND: Tumor endothelial marker 1 (TEM1) is a protein expressed in the tumor-associated endothelium and/or stroma of various types of cancer. We previously demonstrated that immunization with a plasmid-DNA vaccine targeting TEM1 reduced tumor progression in three murine cancer models. Radiation therapy (RT) is an established cancer modality used in more than 50% of patients with solid tumors. RT can induce tumor-associated vasculature injury, triggering immunogenic cell death and inhibition of the irradiated tumor and distant non-irradiated tumor growth (abscopal effect). Combination treatment of RT with TEM1 immunotherapy may complement and augment established immune checkpoint blockade. METHODS: Mice bearing bilateral subcutaneous CT26 colorectal or TC1 lung tumors were treated with a novel heterologous TEM1-based vaccine, in combination with RT, and anti-programmed death-ligand 1 (PD-L1) antibody or combinations of these therapies, tumor growth of irradiated and abscopal tumors was subsequently assessed. Analysis of tumor blood perfusion was evaluated by CD31 staining and Doppler ultrasound imaging. Immunophenotyping of peripheral and tumor-infiltrating immune cells as well as functional analysis was analyzed by flow cytometry, ELISpot assay and adoptive cell transfer (ACT) experiments. RESULTS: We demonstrate that addition of RT to heterologous TEM1 vaccination reduces progression of CT26 and TC1 irradiated and abscopal distant tumors as compared with either single treatment. Mechanistically, RT increased major histocompatibility complex class I molecule (MHCI) expression on endothelial cells and improved immune recognition of the endothelium by anti-TEM1 T cells with subsequent severe vascular damage as measured by reduced microvascular density and tumor blood perfusion. Heterologous TEM1 vaccine and RT combination therapy boosted tumor-associated antigen (TAA) cross-priming (ie, anti-gp70) and augmented programmed cell death protein 1 (PD-1)/PD-L1 signaling within CT26 tumor. Blocking the PD-1/PD-L1 axis in combination with dual therapy further increased the antitumor effect and gp70-specific immune responses. ACT experiments show that anti-gp70 T cells are required for the antitumor effects of the combination therapy. CONCLUSION: Our findings describe novel cooperative mechanisms between heterologous TEM1 vaccination and RT, highlighting the pivotal role that TAA cross-priming plays for an effective antitumor strategy. Furthermore, we provide rationale for using heterologous TEM1 vaccination and RT as an add-on to immune checkpoint blockade as triple combination therapy into early-phase clinical trials.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias Colorrectales/terapia , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Neoplasias Pulmonares/terapia , Proteínas de Neoplasias/metabolismo , Vacunas de ADN/administración & dosificación , Adenoviridae/genética , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/farmacología , Línea Celular Tumoral , Neoplasias Colorrectales/diagnóstico por imagen , Terapia Combinada , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Pulmonares/diagnóstico por imagen , Ratones , Hipofraccionamiento de la Dosis de Radiación , Resultado del Tratamiento , Ultrasonografía Doppler , Vacunas de ADN/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Natl Cancer Inst ; 113(2): 162-170, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32294209

RESUMEN

BACKGROUND: Gut microbial diversity is associated with improved response to immune checkpoint inhibitors (ICI). Based on the known detrimental impact that antibiotics have on microbiome diversity, we hypothesized that antibiotic receipt prior to ICI would be associated with decreased survival. METHODS: Patients with stage III and IV melanoma treated with ICI between 2008 and 2019 were selected from an institutional database. A window of antibiotic receipt within 3 months prior to the first infusion of ICI was prespecified. The primary outcome was overall survival (OS), and secondary outcomes were melanoma-specific mortality and immune-mediated colitis requiring intravenous steroids. All statistical tests were two-sided. RESULTS: There were 568 patients in our database of which 114 received antibiotics prior to ICI. Of the patients, 35.9% had stage III disease. On multivariable Cox proportional hazards analysis of patients with stage IV disease, the antibiotic-exposed group had statistically significantly worse OS (hazard ratio [HR] = 1.81, 95% confidence interval [CI] = 1.27 to 2.57; P <.001). The same effect was observed among antibiotic-exposed patients with stage III disease (HR = 2.78, 95% CI = 1.31 to 5.87; P =.007). When limited to only patients who received adjuvant ICI (n = 89), antibiotic-exposed patients also had statistically significantly worse OS (HR = 4.84, 95% CI = 1.09 to 21.50; P =.04). The antibiotic group had a greater incidence of colitis (HR = 2.14, 95% CI = 1.02 to 4.52; P =.046). CONCLUSION: Patients with stage III and IV melanoma exposed to antibiotics prior to ICI had statistically significantly worse OS than unexposed patients. Antibiotic exposure was associated with greater incidence of moderate to severe immune-mediated colitis. Given the large number of antibiotics prescribed annually, physicians should be judicious with their use in cancer populations likely to receive ICI.


Asunto(s)
Antibacterianos/efectos adversos , Microbioma Gastrointestinal/genética , Variación Genética/efectos de los fármacos , Melanoma/tratamiento farmacológico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antibacterianos/uso terapéutico , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/efectos adversos , Supervivencia sin Enfermedad , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Variación Genética/genética , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Factores Inmunológicos/antagonistas & inhibidores , Factores Inmunológicos/genética , Inmunoterapia/efectos adversos , Masculino , Melanoma/microbiología , Melanoma/mortalidad , Melanoma/patología , Persona de Mediana Edad , Estadificación de Neoplasias , Adulto Joven
14.
Gynecol Oncol ; 116(2): 222-33, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19959212

RESUMEN

In the past decade we have witnessed important advances in the treatment of gynecological cancers and have recognized their potential immunogenicity. This has opened the door to explore immune therapy not only in HPV-induced cancers but also in ovarian and endometrial cancers. Here we will review the off-target immune effects of select chemotherapy drugs commonly used to treat gynecologic cancers and novel tools that can stimulate both the adaptive and innate immune mechanisms such as novel pleiotropic cytokines, Toll-like receptors, and powerful antibodies that can target inhibitory checkpoints, thereby activating effector cellular immune mechanisms and neutralizing suppressor cells. We will also review how existing drugs can be used for combinatorial tumor therapy.


Asunto(s)
Quimioterapia/métodos , Neoplasias de los Genitales Femeninos/terapia , Inmunoterapia/métodos , Terapia Combinada , Femenino , Neoplasias de los Genitales Femeninos/tratamiento farmacológico , Neoplasias de los Genitales Femeninos/inmunología , Humanos
15.
JCI Insight ; 5(16)2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32814714

RESUMEN

Granulosa cell tumors (GCT) are rare ovarian malignancies. Due to the lack of effective treatment in late relapse, there is a clear unmet need for novel therapies. Forkhead Box L2 (FOXL2) is a protein mainly expressed in granulosa cells (GC) and therefore is a rational therapeutic target. Since we identified tumor infiltrating lymphocytes (TILs) as the main immune population within GCT, TILs from 11 GCT patients were expanded, and their phenotypes were interrogated to determine that T cells acquired late antigen-experienced phenotypes and lower levels of PD1 expression. Importantly, TILs maintained their functionality after ex vivo expansion as they vigorously reacted against autologous tumors (100% of patients) and against FOXL2 peptides (57.1% of patients). To validate the relevance of FOXL2 as a target for immune therapy, we developed a plasmid DNA vaccine (FoxL2-tetanus toxin; FoxL2-TT) by fusing Foxl2 cDNA with the immune-enhancing domain of TT. Mice immunization with FoxL2-TT controlled growth of FOXL2-expressing ovarian (BR5) and breast (4T1) cancers in a T cell-mediated manner. Combination of anti-PD-L1 with FoxL2-TT vaccination further reduced tumor progression and improved mouse survival without affecting the female reproductive system and pregnancy. Together, our results suggest that FOXL2 immune targeting can produce substantial long-term clinical benefits. Our study can serve as a foundation for trials testing immunotherapeutic approaches in patients with ovarian GCT.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Proteína Forkhead Box L2/inmunología , Tumor de Células de la Granulosa/inmunología , Linfocitos Infiltrantes de Tumor/patología , Adulto , Animales , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Epítopos , Femenino , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Tumor de Células de la Granulosa/patología , Tumor de Células de la Granulosa/terapia , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Tolerancia Inmunológica/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos , Persona de Mediana Edad , Embarazo , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Microambiente Tumoral , Vacunas de ADN/inmunología , Vacunas de ADN/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Oncogene ; 39(22): 4344-4357, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32335582

RESUMEN

We explore a novel strategy of activating immune signaling through increased micronuclei formation utilizing a cell cycle checkpoint inhibitor to drive cell cycle progression following ionizing radiation. The Chk1/2 inhibitor AZD7762 is used to abrogate radiation therapy (RT)-induced G2/M cell cycle arrest in multiple cell lines and, we find that this therapeutic combination promotes increased micronuclei formation in vitro and subsequently drives increased type I interferon signaling and cytotoxic T-cell activation. In vivo studies using B16-F10 melanoma cancer cells implanted in C57/BL6 mice demonstrate improved rates of tumor control at the abscopal (unirradiated) site, located outside of the radiation field, only in the AZD7762 + RT group, with a corresponding reduction in mean tumor volume, increase in the CD8 T-cell population, and immune activated gene signaling. Our results demonstrate that targeted inhibition of cell cycle checkpoint activation following ionizing radiation drives increased production of immunogenic micronuclei, leading to systemic tumor response with potential future clinical benefit.


Asunto(s)
Núcleo Celular/efectos de los fármacos , Núcleo Celular/efectos de la radiación , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Quinasa de Punto de Control 2/antagonistas & inhibidores , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Melanoma Experimental/inmunología , Proteínas de Neoplasias/antagonistas & inhibidores , Tiofenos/farmacología , Urea/análogos & derivados , Animales , Línea Celular Tumoral , Femenino , Humanos , Interferón beta/biosíntesis , Interferón beta/genética , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Melanoma Experimental/radioterapia , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Pruebas de Micronúcleos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Factor de Transcripción STAT1/biosíntesis , Factor de Transcripción STAT1/genética , Carga Tumoral/efectos de los fármacos , Carga Tumoral/efectos de la radiación , Urea/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Clin Invest ; 130(1): 466-479, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31815742

RESUMEN

Alterations in gut microbiota impact the pathophysiology of several diseases, including cancer. Radiotherapy (RT), an established curative and palliative cancer treatment, exerts potent immune modulatory effects, inducing tumor-associated antigen (TAA) cross-priming with antitumor CD8+ T cell elicitation and abscopal effects. We tested whether the gut microbiota modulates antitumor immune response following RT distal to the gut. Vancomycin, an antibiotic that acts mainly on gram-positive bacteria and is restricted to the gut, potentiated the RT-induced antitumor immune response and tumor growth inhibition. This synergy was dependent on TAA cross presentation to cytolytic CD8+ T cells and on IFN-γ. Notably, butyrate, a metabolite produced by the vancomycin-depleted gut bacteria, abrogated the vancomycin effect. In conclusion, depletion of vancomycin-sensitive bacteria enhances the antitumor activity of RT, which has important clinical ramifications.


Asunto(s)
Presentación de Antígeno/efectos de la radiación , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Microbioma Gastrointestinal , Neoplasias Experimentales , Animales , Presentación de Antígeno/genética , Antígenos de Neoplasias/genética , Butiratos/inmunología , Linfocitos T CD8-positivos/patología , Células Dendríticas/patología , Femenino , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/efectos de la radiación , Ratones , Ratones Noqueados , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/radioterapia
18.
J Immunother Cancer ; 8(2)2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32817208

RESUMEN

BACKGROUND: Novel therapeutic strategies in ovarian cancer (OC) are needed as the survival rate remains dismally low. Although dendritic cell-based cancer vaccines are effective in eliciting therapeutic responses, their complex and costly manufacturing process hampers their full clinical utility outside specialized clinics. Here, we describe a novel approach of generating a rapid and effective cancer vaccine using ascites-derived monocytes for treating OC. METHODS: Using the ID8 mouse ovarian tumor model and OC patient samples, we isolated ascites monocytes and evaluated them with flow cytometry, Luminex cytokine and chemokine array analysis, ex vivo cocultures with T cells, in vivo tumor challenge and T cell transfer experiments, RNA-sequencing and mass spectrometry. RESULTS: We demonstrated the feasibility of isolating ascites monocytes and restoring their ability to function as bona fide antigen-presenting cells (APCs) with Toll-like receptor (TLR) 4 lipopolysaccharide and TLR9 CpG-oligonucleotides, and a blocking antibody to interleukin-10 receptor (IL-10R Ab) in the ID8 model. The ascites monocytes were laden with tumor antigens at a steady state in vivo. After a short 48 hours activation, they upregulated maturation markers (CD80, CD86 and MHC class I) and demonstrated strong ex vivo T cell stimulatory potential and effectively suppressed tumor and malignant ascites in vivo. They also induced protective long-term T cell memory responses. To evaluate the translational potential of this approach, we isolated ascites monocytes from stage III/IV chemotherapy-naïve OC patients. Similarly, the human ascites monocytes presented tumor-associated antigens (TAAs), including MUC1, ERBB2, mesothelin, MAGE, PRAME, GPC3, PMEL and TP53 at a steady state. After a 48-hour treatment with TLR4 and IL-10R Ab, they efficiently stimulated oligoclonal tumor-associated lymphocytes (TALs) with strong reactivity against TAAs. Importantly, the activated ascites monocytes retained their ability to activate TALs in the presence of ascitic fluid. CONCLUSIONS: Ascites monocytes are naturally loaded with tumor antigen and can perform as potent APCs following short ex vivo activation. This novel ascites APC vaccine can be rapidly prepared in 48 hours with a straightforward and affordable manufacturing process, and would be an attractive therapeutic vaccine for OC.


Asunto(s)
Ascitis/fisiopatología , Vacunas contra el Cáncer/inmunología , Monocitos/metabolismo , Neoplasias Ováricas/inmunología , Receptores Toll-Like/inmunología , Animales , Femenino , Humanos , Mesotelina , Ratones , Neoplasias Ováricas/mortalidad , Análisis de Supervivencia
19.
J Transl Med ; 7: 104, 2009 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-20003308

RESUMEN

BACKGROUND: Liposomal doxorubicin (Doxil) is a cytotoxic chemotherapy drug with a favorable hematologic toxicity profile. Its active drug, doxorubicin, has interesting immunomodulatory properties. Here, the effects of Doxil on surviving tumor cell immunophenotype were investigated. METHODS: Using ID8 murine ovarian cancer cells, the immunomodulatory effects of Doxil were studied by measuring its impact on ovarian cancer cell expression of MHC class-I and Fas, and susceptibility to immune attack in vitro. To evaluate the ability of Doxil to cooperate with cancer immunotherapy, the interaction between Doxil and Interleukin 18 (IL-18), a pleiotropic immunostimulatory cytokine, was investigated in vivo in mice bearing ID8-Vegf tumors. RESULTS: While Doxil killed ID8 tumor cells in a dose-dependent manner, tumor cells escaping Doxil-induced apoptosis upregulated surface expression of MHC-I and Fas, and were sensitized to CTL killing and Fas-mediated death in vitro. We therefore tested the hypothesis that the combination of immunotherapy with Doxil provides positive interactions. Combination IL-18 and Doxil significantly suppressed tumor growth compared with either monotherapy in vivo and uniquely resulted in complete tumor regression and long term antitumor protection in a significant proportion of mice. CONCLUSION: These data demonstrate that Doxil favorably changes the immunophenotype of a large fraction of the tumor that escapes direct killing thus creating an opportunity to expand tumor killing by immunotherapy, which can be capitalized through addition of IL-18 in vivo.


Asunto(s)
Antibióticos Antineoplásicos , Doxorrubicina , Interleucina-18 , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/inmunología , Animales , Antibióticos Antineoplásicos/inmunología , Antibióticos Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Doxorrubicina/inmunología , Doxorrubicina/uso terapéutico , Femenino , Humanos , Inmunofenotipificación , Interleucina-18/inmunología , Interleucina-18/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neoplasias Ováricas/patología , Tasa de Supervivencia
20.
Cancer J ; 25(3): 165-177, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31135523

RESUMEN

The clear cell subtype of kidney cancer encompasses most renal cell carcinoma cases and is associated with the loss of von Hippel-Lindau gene function or expression. Subsequent loss or mutation of the other allele influences cellular stress responses involving nutrient and hypoxia sensing. Autophagy is an important regulatory process promoting the disposal of unnecessary or degraded cellular components, tightly linked to almost all cellular processes. Organelles and proteins that become damaged or that are no longer needed in the cell are sequestered and digested in autophagosomes upon fusing with lysosomes, or alternatively, released via vesicular exocytosis. Tumor development tends to disrupt the regulation of the balance between this process and apoptosis, permitting prolonged cell survival and increased replication. Completed trials of autophagic inhibitors using hydroxychloroquine in combination with other anticancer agents including rapalogues and high-dose interleukin 2 have now been reported. The complex nature of autophagy and the unique biology of clear cell renal cell carcinoma warrant further understanding to better develop the next generation of relevant anticancer agents.


Asunto(s)
Autofagia , Carcinoma de Células Renales/metabolismo , Endotelio/metabolismo , Neoplasias Renales/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Autofagia/genética , Biomarcadores , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/etiología , Carcinoma de Células Renales/patología , Ensayos Clínicos como Asunto , Citoesqueleto/metabolismo , Susceptibilidad a Enfermedades , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Endotelio/efectos de los fármacos , Endotelio/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/etiología , Neoplasias Renales/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Mitofagia/efectos de los fármacos , Mitofagia/genética , Imagen Molecular , Terapia Molecular Dirigida , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA