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
2.
Nat Med ; 29(8): 2121-2132, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37414899

RESUMEN

Fecal microbiota transplantation (FMT) represents a potential strategy to overcome resistance to immune checkpoint inhibitors in patients with refractory melanoma; however, the role of FMT in first-line treatment settings has not been evaluated. We conducted a multicenter phase I trial combining healthy donor FMT with the PD-1 inhibitors nivolumab or pembrolizumab in 20 previously untreated patients with advanced melanoma. The primary end point was safety. No grade 3 adverse events were reported from FMT alone. Five patients (25%) experienced grade 3 immune-related adverse events from combination therapy. Key secondary end points were objective response rate, changes in gut microbiome composition and systemic immune and metabolomics analyses. The objective response rate was 65% (13 of 20), including four (20%) complete responses. Longitudinal microbiome profiling revealed that all patients engrafted strains from their respective donors; however, the acquired similarity between donor and patient microbiomes only increased over time in responders. Responders experienced an enrichment of immunogenic and a loss of deleterious bacteria following FMT. Avatar mouse models confirmed the role of healthy donor feces in increasing anti-PD-1 efficacy. Our results show that FMT from healthy donors is safe in the first-line setting and warrants further investigation in combination with immune checkpoint inhibitors. ClinicalTrials.gov identifier NCT03772899 .


Asunto(s)
Trasplante de Microbiota Fecal , Melanoma , Animales , Ratones , Trasplante de Microbiota Fecal/métodos , Inhibidores de Puntos de Control Inmunológico , Heces/microbiología , Melanoma/terapia , Inmunoterapia , Resultado del Tratamiento
3.
J Immunol ; 210(10): 1598-1606, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37000461

RESUMEN

Tumor-specific CD8+ T cells are critical components of antitumor immunity; however, factors that modulate their phenotype and function have not been completely elucidated. Cytokines IL-12 and IL-27 have recognized roles in promoting CD8+ T cells' effector function and mediated antitumor responses. Tumor-specific CD8+ tumor-infiltrating lymphocytes (TILs) can be identified based on surface expression of CD39, whereas bystander CD8+ TILs do not express this enzyme. It is currently unclear how and why tumor-specific CD8+ T cells uniquely express CD39. Given the important roles of IL-12 and IL-27 in promoting CD8+ T cell functionality, we investigated whether these cytokines could modulate CD39 expression on these cells. Using in vitro stimulation assays, we identified that murine splenic CD8+ T cells differentially upregulate CD39 in the presence of IL-12 and IL-27. Subsequently, we assessed the exhaustion profile of IL-12- and IL-27-induced CD39+CD8+ T cells. Despite the greatest frequency of exhausted CD39+CD8+ T cells after activation with IL-12, as demonstrated by the coexpression of TIM-3+PD-1+LAG-3+ and reduced degranulation capacity, these cells retained the ability to produce IFN-γ. IL-27-induced CD39+CD8+ T cells expressed PD-1 but did not exhibit a terminally exhausted phenotype. IL-27 was able to attenuate IL-12-mediated inhibitory receptor expression on CD39+CD8+ T cells but did not rescue degranulation ability. Using an immunogenic neuro-2a mouse model, inhibiting IL-12 activity reduced CD39+CD8+ TIL frequency compared with controls without changing the overall CD8+ TIL frequency. These results provide insight into immune regulators of CD39 expression on CD8+ T cells and further highlight the differential impact of CD39-inducing factors on the phenotype and effector functions of CD8+ T cells.


Asunto(s)
Interleucina-12 , Interleucina-27 , Animales , Ratones , Interleucina-12/metabolismo , Interleucina-27/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T CD8-positivos , Linfocitos Infiltrantes de Tumor , Citocinas/metabolismo , Fenotipo
4.
Cancer Immunol Immunother ; 72(5): 1247-1260, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36396738

RESUMEN

Strategies to modify the gut microbiome in cancer patients using fecal microbiota transplantation (FMT) have gained momentum as a therapeutic intervention. However, how FMT impacts innate-like, antimicrobial T lymphocytes is unclear. In this study, we assessed peripheral blood (PB) mucosa-associated invariant T (MAIT) cell frequencies and functions in patients with metastatic renal cell carcinoma (mRCC) before and seven days after they received FMT as part of a clinical trial. We found comparable MAIT cell frequencies in healthy controls and mRCC patients. In contrast, γδ T cells exhibited a numerical decline in mRCC, which was partially reversed by FMT. We also found a significant increase in the PB CD4+ MAIT cell compartment of mRCC patients with or without FMT. Paired sample analyses revealed CD69 upregulation on MAIT cells accompanied by decreased PD-1 levels post-FMT. These changes were unique to MAIT cells as non-MAIT T lymphocytes showed either no trend or a trend in the opposite direction. Importantly, FMT did not render MAIT cells exhausted as also judged by their stable expression of TIM-3, LAG-3, BTLA, CTLA-4, TIGIT and VISTA. These findings were corroborated in functional assays in which MAIT cells were stimulated with MR1 ligands or with a combination of IL-12 and IL-18 to produce inflammatory cytokines and granzyme B. Indeed, when stimulated ex vivo with IL-12 and IL-18, MAIT cells mounted a more rigorous TNF-α response post-FMT. In conclusion, FMT improves MAIT cell functions, which should serve patients well in subsequent microbial challenges in the face of cancer-elicited immunosuppression. Trial Registration: https://clinicaltrials.gov/ Identifier: NCT04163289 (registration date: November 14, 2019).


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Células T Invariantes Asociadas a Mucosa , Humanos , Interleucina-18/metabolismo , Carcinoma de Células Renales/terapia , Carcinoma de Células Renales/metabolismo , Células T Invariantes Asociadas a Mucosa/metabolismo , Trasplante de Microbiota Fecal , Neoplasias Renales/terapia , Neoplasias Renales/metabolismo , Interleucina-12/metabolismo
5.
Mol Ther ; 31(2): 535-551, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36068918

RESUMEN

Immune checkpoint blockade can induce potent and durable responses in patients with highly immunogenic mismatch repair-deficient tumors; however, these drugs are ineffective against immune-cold neuroblastoma tumors. To establish a role for a T cell-based therapy against neuroblastoma, we show that T cell and memory T cell-dependent gene expression are associated with improved survival in high-risk neuroblastoma patients. To stimulate anti-tumor immunity and reproduce this immune phenotype in neuroblastoma tumors, we used CRISPR-Cas9 to knockout MLH1-a crucial molecule in the DNA mismatch repair pathway-to induce mismatch repair deficiency in a poorly immunogenic murine neuroblastoma model. Induced mismatch repair deficiency increased the expression of proinflammatory genes and stimulated T cell infiltration into neuroblastoma tumors. In contrast to adult cancers with induced mismatch repair deficiency, neuroblastoma tumors remained unresponsive to anti-PD1 treatment. However, anti-CTLA4 therapy was highly effective against these tumors. Anti-CTLA4 therapy promoted immune memory and T cell epitope spreading in cured animals. Mechanistically, the effect of anti-CTLA4 therapy against neuroblastoma tumors with induced mismatch repair deficiency is CD4+ T cell dependent, as depletion of these cells abolished the effect. Therefore, a therapeutic strategy involving mismatch repair deficiency-based T cell infiltration of neuroblastoma tumors combined with anti-CTLA4 can serve as a novel T cell-based treatment strategy for neuroblastoma.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Colorrectales , Neuroblastoma , Ratones , Animales , Memoria Inmunológica , Neoplasias Colorrectales/patología , Neuroblastoma/genética , Neuroblastoma/terapia
6.
Eur J Immunol ; 50(5): 695-704, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31943150

RESUMEN

CD5 is expressed on T cells and a subset of B cells (B1a). It can attenuate TCR signalling and impair CTL activation and is a therapeutic targetable tumour antigen expressed on leukemic T and B cells. However, the potential therapeutic effect of functionally blocking CD5 to increase T cell anti-tumour activity against tumours (including solid tumours) has not been explored. CD5 knockout mice show increased anti-tumour immunity: reducing CD5 on CTLs may be therapeutically beneficial to enhance the anti-tumour response. Here, we show that ex vivo administration of a function-blocking anti-CD5 MAb to primary mouse CTLs of both tumour-naïve mice and mice bearing murine 4T1 breast tumour homografts enhanced their capacity to respond to activation by treatment with anti-CD3/anti-CD28 MAbs or 4T1 tumour cell lysates. Furthermore, it enhanced TCR signalling (ERK activation) and increased markers of T cell activation, including proliferation, CD69 levels, IFN-γ production, apoptosis and Fas receptor and Fas ligand levels. Finally, CD5 function-blocking MAb treatment enhanced the capacity of CD8+ T cells to kill 4T1-mouse tumour cells in an ex vivo assay. These data support the potential of blockade of CD5 function to enhance T cell-mediated anti-tumour immunity.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Antígenos CD28/inmunología , Antígenos CD5/inmunología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Linfocitos T Citotóxicos/efectos de los fármacos , Animales , Antineoplásicos Inmunológicos , Antígenos CD28/antagonistas & inhibidores , Antígenos CD28/genética , Antígenos CD5/antagonistas & inhibidores , Antígenos CD5/genética , Extractos Celulares/farmacología , Línea Celular Tumoral , Proliferación Celular , Proteína Ligando Fas/genética , Proteína Ligando Fas/inmunología , Femenino , Regulación de la Expresión Génica , Interferón gamma/genética , Interferón gamma/inmunología , Activación de Linfocitos/efectos de los fármacos , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Transducción de Señal , Linfocitos T Citotóxicos/inmunología , Receptor fas/genética , Receptor fas/inmunología
7.
Clin Cancer Res ; 23(4): 1068-1079, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-27856600

RESUMEN

Purpose: The ubiquitously expressed transmembrane glycoprotein CD47 delivers an anti-phagocytic (do not eat) signal by binding signal-regulatory protein α (SIRPα) on macrophages. CD47 is overexpressed in cancer cells and its expression is associated with poor clinical outcomes. TTI-621 (SIRPαFc) is a fully human recombinant fusion protein that blocks the CD47-SIRPα axis by binding to human CD47 and enhancing phagocytosis of malignant cells. Blockade of this inhibitory axis using TTI-621 has emerged as a promising therapeutic strategy to promote tumor cell eradication.Experimental Design: The ability of TTI-621 to promote macrophage-mediated phagocytosis of human tumor cells was assessed using both confocal microscopy and flow cytometry. In vivo antitumor efficacy was evaluated in xenograft and syngeneic models and the role of the Fc region in antitumor activity was evaluated using SIRPαFc constructs with different Fc tails.Results: TTI-621 enhanced macrophage-mediated phagocytosis of both hematologic and solid tumor cells, while sparing normal cells. In vivo, TTI-621 effectively controlled the growth of aggressive AML and B lymphoma xenografts and was efficacious in a syngeneic B lymphoma model. The IgG1 Fc tail of TTI-621 plays a critical role in its antitumor activity, presumably by engaging activating Fcγ receptors on macrophages. Finally, TTI-621 exhibits minimal binding to human erythrocytes, thereby differentiating it from CD47 blocking antibodies.Conclusions: These data indicate that TTI-621 is active across a broad range of human tumors. These results further establish CD47 as a critical regulator of innate immune surveillance and form the basis for clinical development of TTI-621 in multiple oncology indications. Clin Cancer Res; 23(4); 1068-79. ©2016 AACR.


Asunto(s)
Antígenos de Diferenciación/genética , Antígeno CD47/genética , Inmunoglobulina G/inmunología , Neoplasias/tratamiento farmacológico , Receptores Inmunológicos/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Animales , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Bloqueadores/inmunología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Antígeno CD47/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Ratones , Neoplasias/inmunología , Neoplasias/patología , Fagocitosis/efectos de los fármacos , Unión Proteica , Mapas de Interacción de Proteínas/efectos de los fármacos , Receptores Inmunológicos/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Oncotarget ; 7(15): 20825-39, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-26959114

RESUMEN

Human tumor heterogeneity promotes therapeutic failure by increasing the likelihood of resistant cell subpopulations. The PARP-1 inhibitor olaparib is approved for use in BRCA-mutated ovarian cancers but BRCA2-reversion mutations lead to functional homologous recombination repair (HRR) and olaparib resistance. To overcome that resistance and expand use of PARP1 inhibition to cancers with functional HRR, we developed an antisense strategy to render the majority of tumor cells in a population BRCA2-deficient. We predicted that this strategy would render HRR-proficient tumor cells sensitive to olaparib and prevent emergence of resistance in a tumor cell population heterogeneous for HRR proficiency. We report that BRCA2 downregulation sensitized multiple human tumor cell lines (but not non-cancer human kidney cells) to olaparib and, combined with olaparib, increased aneuploidy and chromosomal translocations in human tumor cells. In a mixed HRR-proficient and HRR-deficient cell population, olaparib monotherapy allowed outgrowth of HRR-proficient cells resistant to subsequent olaparib treatment. Combined BRCA2 inhibition and olaparib treatment prevented selection of HRR-proficient cells and inhibited proliferation of the entire population. Treatment with BRCA2 siRNA and olaparib decreased ovarian xenograft growth in mice more effectively than either treatment alone. In vivo use of BRCA2 antisense oligonucleotides may be a viable option to expand clinical use of olaparib and prevent resistance.


Asunto(s)
Antineoplásicos/farmacología , Proteína BRCA2/antagonistas & inhibidores , Resistencia a Antineoplásicos/genética , Neoplasias Ováricas/tratamiento farmacológico , Ftalazinas/farmacología , Piperazinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteína BRCA2/genética , Biomarcadores de Tumor/genética , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , ARN Interferente Pequeño/genética , Reparación del ADN por Recombinación/efectos de los fármacos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Tumori ; 102(1): 31-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26429639

RESUMEN

AIMS AND BACKGROUND: In light of the need for more selective anticancer therapy, much work has been directed at developing compounds or biological agents that target functions specific to cancer cells. To this end, numerous viruses have been engineered to exploit the dependence of cancer cells on particular anomalies that contribute to their rogue proliferative activity, such as dysfunctional p53, overactive mitogenic signaling, or a defective interferon response. The oncolytic human adenovirus dl1520 (ONYX-015) was engineered to propagate specifically in p53-deficient tumors, which comprise over half of all tumors. Based on successes in clinical trials, the full potential of dl1520 and other oncolytic viruses may be even better realized by using them in combination with conventional chemotherapy drugs. METHODS: As a model system in which to test this potential, representative cell lines from 2 common cancer types, oral squamous cell carcinoma (HN-5a) and colon adenocarcinoma (HT-29), were chosen, as well as platinum-drug-resistant variants of each. RESULTS: Following preliminary screening of virus and drug combinations, dl1520 and melphalan were found to synergistically inhibit proliferation of all the cancer cell lines. Melphalan pretreatment or cotreatment with dl1520 enhanced inhibition of proliferation by dl1520 by up to 60% and increased apoptosis by up to 25%. The tight-junction protein CAR (coxsackie and adenovirus receptor), via which adenovirus enters cells, was not upregulated by treatment with melphalan, suggesting that other mechanisms contribute to synergy. CONCLUSIONS: The synergy between melphalan and dl1520 suggests that tumor-selective cell killing by oncolytic viruses may be augmented by combining with cytotoxic drugs.


Asunto(s)
Adenoviridae , Antineoplásicos/farmacología , Melfalán/farmacología , Neoplasias/tratamiento farmacológico , Virus Oncolíticos , Vacunas Virales/farmacología , Adenocarcinoma/tratamiento farmacológico , Antineoplásicos Alquilantes/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Sinergismo Farmacológico , Humanos , Neoplasias de la Boca/tratamiento farmacológico
10.
PLoS One ; 10(11): e0143435, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26579709

RESUMEN

Indoleamine 2,3-dioxygenase-1 (IDO) is an immune regulatory enzyme expressed by most human tumors. IDO levels in tumor cells correlate with increased metastasis and poor patient outcome and IDO is linked to tumor cell resistance to immunotherapy, radiation therapy, and chemotherapy. Knowledge of tumor cell-autonomous effects of IDO, independent of its well-known role in regulating and suppressing anti-tumor immune responses, is limited. Clonal populations of A549 human lung adenocarcinoma cells stably transfected with anti-IDO shRNA or scrambled control shRNA were used to study IDO effects on drug sensitivity and resistance. IFNγ was used to induce IDO in those cells. We show, for the first time, that IDO mediates human tumor cell resistance to the candidate anticancer drugs FK866 (an NAD+ inhibitor), methoxyamine (MX, a base excision repair [BER] inhibitor) and approved anticancer drugs pemetrexed (a folate anti-metabolite) and gemcitabine (a nucleoside analogue), and combined treatment with pemetrexed and MX, in the absence of immune cells. Concurrent knockdown of IDO and thymidylate synthase (TS, a key rate-limiting enzyme in DNA synthesis and repair) sensitizes human lung cancer cells to pemetrexed and 5FUdR to a greater degree than knockdown of either target alone. We conclude that BER in IDO-expressing A549 cells plays a major role in mediating resistance to a range of approved and candidate anticancer drugs. IDO inhibitors are undergoing clinical trials primarily to improve antitumor immune responses. We show that targeting IDO alone or in combination with TS is a potentially valuable therapeutic strategy for cancer treatment, independent of immune activity and in combination with conventional chemotherapy.


Asunto(s)
Acrilamidas/farmacología , Desoxicitidina/análogos & derivados , Regulación hacia Abajo/efectos de los fármacos , Hidroxilaminas/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Pemetrexed/farmacología , Piperidinas/farmacología , Animales , Proteína BRCA2/metabolismo , Línea Celular Tumoral , Células Clonales , Reparación del ADN/efectos de los fármacos , Desoxicitidina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Floxuridina/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Ratones SCID , NAD/metabolismo , ARN Interferente Pequeño/metabolismo , Timidilato Sintasa/antagonistas & inhibidores , Timidilato Sintasa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
11.
Oncotarget ; 6(26): 22397-409, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26087398

RESUMEN

Nucleoside metabolism enzymes are determinants of chemotherapeutic drug activity. The nucleoside salvage enzyme deoxycytidine kinase (dCK) activates gemcitabine (2', 2'-difluoro-2'-deoxycytidine) and is negatively regulated by deoxycytidine triphosphate (dCTP). Reduction of dCTP in tumor cells could, therefore, enhance gemcitabine activity. Mitochondrial thymidine kinase 2 (TK2) phosphorylates deoxycytidine to generate dCTP. We hypothesized that: (1) TK2 modulates human tumor cell sensitivity to gemcitabine, and (2) antisense knockdown of TK2 would decrease dCTP and increase dCK activity and gemcitabine activation. siRNA downregulation of TK2 sensitized MCF7 and HeLa cells (high and moderate TK2) but not A549 cells (low TK2) to gemcitabine. Combined treatment with TK2 siRNA and gemcitabine increased dCK. We also hypothesized that TK2 siRNA-induced drug sensitization results in mitochondrial damage that enhances gemcitabine effectiveness. TK2 siRNA and gemcitabine decreased mitochondrial redox status, DNA content, and activity. This is the first demonstration of a direct role for TK2 in gemcitabine resistance, or any independent role in cancer drug resistance, and further distinguishes TK2 function from that of other dTMP-producing enzymes [cytosolic TK1 and thymidylate synthase (TS)]. siRNA knockdown of TK1 and/or TS did not sensitize cancer cells to gemcitabine indicating that, among the 3 enzymes, only TK2 is a candidate therapeutic target for combination with gemcitabine.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Desoxicitidina Quinasa/metabolismo , Desoxicitidina/análogos & derivados , Neoplasias/terapia , ARN Interferente Pequeño/administración & dosificación , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Línea Celular Tumoral , Desoxicitidina/farmacología , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Células MCF-7 , Mitocondrias/enzimología , Neoplasias/tratamiento farmacológico , Neoplasias/genética , ARN Interferente Pequeño/genética , Transfección , Gemcitabina
12.
Mol Oncol ; 8(8): 1429-40, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24974076

RESUMEN

Tumor cells have unstable genomes relative to non-tumor cells. Decreased DNA integrity resulting from tumor cell instability is important in generating favorable therapeutic indices, and intact DNA repair mediates resistance to therapy. Targeting DNA repair to promote the action of anti-cancer agents is therefore an attractive therapeutic strategy. BRCA2 is involved in homologous recombination repair. BRCA2 defects increase cancer risk but, paradoxically, cancer patients with BRCA2 mutations have better survival rates. We queried TCGA data and found that BRCA2 alterations led to increased survival in patients with ovarian and endometrial cancer. We developed a BRCA2-targeting second-generation antisense oligonucleotide (ASO), which sensitized human lung, ovarian, and breast cancer cells to cisplatin by as much as 60%. BRCA2 ASO treatment overcame acquired cisplatin resistance in head and neck cancer cells, but induced minimal cisplatin sensitivity in non-tumor cells. BRCA2 ASO plus cisplatin reduced respiration as an early event preceding cell death, concurrent with increased glucose uptake without a difference in glycolysis. BRCA2 ASO and cisplatin decreased metastatic frequency in vivo by 77%. These results implicate BRCA2 as a regulator of metastatic frequency and cellular metabolic response following cisplatin treatment. BRCA2 ASO, in combination with cisplatin, is a potential therapeutic anti-cancer agent.


Asunto(s)
Proteína BRCA2/metabolismo , Cisplatino/farmacología , Animales , Proteína BRCA2/genética , Línea Celular , Línea Celular Tumoral , Proliferación Celular/genética , Proliferación Celular/fisiología , Embrión de Pollo , Humanos , Metástasis de la Neoplasia/genética , Oligorribonucleótidos Antisentido/farmacología , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo
13.
Oncotarget ; 5(9): 2778-91, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24784564

RESUMEN

Indoleamine 2,3-dioxygenase-1 (IDO) is an immunosuppressive molecule expressed by most human tumors. IDO levels correlate with poor prognosis in cancer patients and IDO inhibitors are under investigation to enhance endogenous anticancer immunosurveillance. Little is known of immune-independent functions of IDO relevant to cancer therapy. We show, for the first time, that IDO mediates human tumor cell resistance to a PARP inhibitor (olaparib), gamma radiation, cisplatin, and combined treatment with olaparib and radiation, in the absence of immune cells. Antisense-mediated reduction of IDO, alone and (in a synthetic lethal approach) in combination with antisense to the DNA repair protein BRCA2 sensitizes human lung cancer cells to olaparib and cisplatin. Antisense reduction of IDO decreased NAD+ in human tumor cells. NAD+ is essential for PARP activity and these data suggest that IDO mediates treatment resistance independent of immunity and at least partially due to a previously unrecognized role for IDO in DNA repair. Furthermore, IDO levels correlated with accumulation of tumor cells in G1 and depletion of cells in G2/M of the cell cycle, suggesting that IDO effects on cell cycle may also modulate sensitivity to radiation and chemotherapeutic agents. IDO is a potentially valuable therapeutic target in cancer treatment, independent of immune function and in combination with other therapies.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Rayos gamma , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Tolerancia a Radiación , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Western Blotting , Ciclo Celular , Proliferación Celular , Cisplatino/administración & dosificación , Terapia Combinada , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Neoplasias/patología , Ftalazinas/administración & dosificación , Piperazinas/administración & dosificación , ARN Interferente Pequeño/genética , Células Tumorales Cultivadas
14.
Mol Imaging Biol ; 16(1): 63-73, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23836502

RESUMEN

PURPOSE: The bacterial gene MagA imparts magnetic properties to mammalian cells and provides a basis for cell tracking by magnetic resonance imaging (MRI). In a mouse model of tumor growth from transplanted cells, we used repetitive MRI to demonstrate the in vivo imaging potential of MagA expression relative to a modified ferritin overexpression system, lacking regulation through iron response elements (HF + LF). PROCEDURES: Subcutaneous tumor xenografts were monitored weekly from days 2 to 34 post-injection. Small animal MRI employed balanced steady-state free precession. Imaging was correlated with tumor histology using hematoxylin, Prussian Blue, Ki-67, and BS-1 lectin. RESULTS: Tumor heterogeneity with respect to tissue morphology and magnetic resonance (MR) contrast was apparent within a week of cell transplantation. In MagA- and HF + LF-expressing tumors, MR contrast enhancement was recorded up to day 20 post-injection and 0.073-cm(3) tumor volumes. MagA-expressing tumors showed increases in both quantity and quality of MR contrast as measured by fractional void volume and contrast-to-noise ratio, respectively. MR contrast in both MagA- and HF + LF-expressing tumors was maximal by day 13, doubling fractional void volume 1 week ahead of controls. CONCLUSIONS: MagA- and HF + LF-expressing tumor xenografts augment MR contrast after 1 week of growth. MagA expression increases MR contrast within days of cell transplantation and provides MR contrast comparable to HF + LF. MagA has utility for monitoring cell growth and differentiation, with potential for in vivo detection of reporter gene expression using MRI.


Asunto(s)
Medios de Contraste , Ferritinas/metabolismo , Genes Bacterianos/genética , Imagen por Resonancia Magnética/métodos , Neoplasias/diagnóstico , Neoplasias/patología , Subunidades de Proteína/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Espacio Intracelular/metabolismo , Ratones , Ratones Desnudos , Neoplasias/genética , Relación Señal-Ruido , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Mol Ther Nucleic Acids ; 2: e78, 2013 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-23481354

RESUMEN

A high mutation rate leading to tumor cell heterogeneity is a driver of malignancy in human cancers. Paradoxically, however, genomic instability can also render tumors vulnerable to therapeutic attack. Thus, targeting DNA repair may induce an intolerable level of DNA damage in tumor cells. BRCA2 mediates homologous recombination repair, and BRCA2 polymorphisms increase cancer risk. However, tumors with BRCA2 mutations respond better to chemotherapy and are associated with improved patient prognosis. Thymidylate synthase (TS) is also involved in DNA maintenance and generates cellular thymidylate. We determined that antisense downregulation of BRCA2 synergistically potentiated drugs with mechanisms of action related to BRCA2 function (cisplatin, melphalan), a phenomenon we named "complementary lethality." TS knockdown induced complementary lethality to TS-targeting drugs (5-FUdR and pemetrexed) but not DNA cross-linking agents. Combined targeting of BRCA2 and TS induced complementary lethality to both DNA-damaging and TS-targeting agents, thus creating multidrug sensitive tumors. In addition, we demonstrated for the first time that simultaneous downregulation of both targets induced combined complementary lethality to multiple mechanistically different drugs in the same cell population. In this study, we propose and define the concept of "complementary lethality" and show that actively targeting BRCA2 and TS is of potential therapeutic benefit in multidrug treatment of human tumors. This work has contributed to the development of a BRCA2-targeting antisense oligdeoxynucleotide (ASO) "BR-1" which we will test in vivo in combination with our TS-targeting ASO "SARI 83" and attempt early clinical trials in the future.Molecular Therapy - Nucleic Acids (2013) 2, e78; doi:10.1038/mtna.2013.7 published online 12 March 2013.

16.
Mol Imaging ; 8(3): 129-39, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19723470

RESUMEN

Molecular imaging with magnetic resonance imaging (MRI) may benefit from the ferrimagnetic properties of magnetosomes, membrane-enclosed iron biominerals whose formation in magnetotactic bacteria is encoded by multiple genes. One such gene is MagA, a putative iron transporter. We have examined expression of MagA in mouse neuroblastoma N2A cells and characterized their response to iron loading and cellular imaging by MRI. MagA expression augmented both Prussian blue staining and the elemental iron content of N2A cells, without altering cell proliferation, in cultures grown in the presence of iron supplements. Despite evidence for iron incorporation in both MagA and a variant, MagAE137V, only MagA expression produced intracellular contrast detectable by MRI at 11 Tesla. We used this stable expression system to model a new sequence for cellular imaging with MRI, using the difference between gradient and spin echo images to distinguish cells from artifacts in the field of view. Our results show that MagA activity in mammalian cells responds to iron supplementation and functions as a contrast agent that can be deactivated by a single point mutation. We conclude that MagA is a candidate MRI reporter gene that can exploit more fully the superior resolution of MRI in noninvasive medical imaging.


Asunto(s)
Proteínas Bacterianas/análisis , Neoplasias de la Mama/patología , Proteínas de Transporte de Catión/análisis , Medios de Contraste/administración & dosificación , Proteínas Fluorescentes Verdes/análisis , Imagen por Resonancia Magnética/métodos , Neuroblastoma/patología , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proteínas de Transporte de Catión/biosíntesis , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Medios de Contraste/metabolismo , Femenino , Expresión Génica , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Miembro Posterior , Humanos , Hierro/administración & dosificación , Hierro/metabolismo , Espectrometría de Masas , Ratones , Trasplante de Neoplasias , Neuroblastoma/genética , Neuroblastoma/metabolismo , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Transfección , Zinc/metabolismo
17.
DNA Cell Biol ; 27(5): 229-40, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18358073

RESUMEN

Thymidylate synthase (TS) is essential for DNA replication and is a target for cancer chemotherapy. However, toxicity to normal cells and tumor cell drug resistance necessitate development of new therapeutic strategies. One such strategy is to use antisense (AS) technology to reduce TS mRNA and protein levels in treated cells. We have developed oligodeoxynucleotides (ODNs) that target different regions of TS mRNA, inhibit human tumor cell proliferation as single agents, and enhance cytotoxicity of clinically useful TS protein-targeting drugs. Here we describe ODN 491, a novel 20mer AS ODN complementary to a previously untargeted portion of the TS mRNA coding region. AS ODN 491 decreased TS mRNA levels to different degrees in a panel of human tumor-derived cell lines, and induced different physiological effects in a tumor cell line-dependent manner. ODN 491 (like AS TS ODN 83, previously shown to be effective) decreased TS protein levels in HeLa cells with a concomitant increase in sensitivity to TS-targeting chemotherapeutics. However (and contrary to HeLa cell response to an AS ODN 83), it did not, as a single agent, inhibit HeLa cell proliferation. In MCF-7 cells, ODN 491 treatment was less effective at reducing TS mRNA and did not reduce TS protein, nor did it enhance sensitivity to TS-targeting or other chemotherapeutics. Moreover, specifically in MCF-7 cells but not HeLa cells, ODN 491 as a single agent induced apoptosis. These data indicate that AS TS ODN 491 is an effective AS reagent targeting a novel TS mRNA region. However, treatment of tumor cell lines with AS TS ODNs targeting different TS mRNA regions results in a pattern of physiological effects that varies in a tumor cell line-specific fashion. In addition, the capacity of different AS TS ODNs to induce physiological effects does not correlate well with their capacity to reduce TS mRNA and/or protein and, further, depends on the region of TS mRNA selected for targeting. Recognition of tumor cell-specific and mRNA region-specific variability in response to AS TS ODNs will be important in designing AS TS ODNs for potential clinical use.


Asunto(s)
Antineoplásicos/farmacología , Oligonucleótidos Antisentido/farmacología , Timidilato Sintasa/genética , Apoptosis/fisiología , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Células HeLa/efectos de los fármacos , Humanos , ARN Mensajero/metabolismo , Transcripción Genética , Transfección
18.
Exp Toxicol Pathol ; 56(4-5): 313-9, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15816360

RESUMEN

h-R3 is a humanized anti-epidermal growth factor receptor (EGFR) monoclonal antibody (MAb). This receptor is over-expressed in the majority of tumors of epithelial origin, including glioblastomas. 188Rhenium (188Re) constitutes an ideal radionuclide for imagining and radioimmunotherapy, and its toxicity is known, nevertheless, it is unknown if 188Os, as 188Re's daughter, has any local or systemic toxicity effect when it is administered intracerebrally for treating intracranial tumors. For this reason we decided to assess the toxicity of stable 188Os once the complete decay of 188Re has occurred, by administering intracerebrally to rats the h-R3 labeled with 188Os. Forty rats (20 each sex) were distributed randomly into four experimental groups (ten per group): control group received 5microL of glucoheptonate solution vehicle; two other groups were treated with unlabeled or labeled h-R3 with 188Os. The remaining group served as a non-treated control group. A single 5 microL dose (2.5 microL into each lateral ventricle) of neutral solution containing 50 microg of h-R3 labeled initially with 13.25 microCi of 188Re was stereotactically administered into lateral ventricles 8 days after the conjugation with the radionuclide was done. Each animal was observed daily for detection of toxicity signs. Body weights were recorded on days 0, 7 and 14. Blood samples for analysis of hematological and clinical chemistry parameters were taken on days 0 and 14. Necropsy and histopathological studies were carried out at the end of the study. All animals gained weight by day 14. There were no changes in hematological and clinical chemistry, but minimal histopathological changes were observed at the application sites. This study shows that single doses of 188Os-h-R3 is tolerable and causes minimal local and no systemic toxicity effects in rats.


Asunto(s)
Anticuerpos Monoclonales/toxicidad , Encéfalo/efectos de los fármacos , Osmio/toxicidad , Radioisótopos/administración & dosificación , Radioisótopos/toxicidad , Animales , Anticuerpos Monoclonales/administración & dosificación , Encéfalo/patología , Receptores ErbB/inmunología , Femenino , Inyecciones Intraventriculares , Masculino , Osmio/administración & dosificación , Ratas , Ratas Sprague-Dawley
19.
J Clin Oncol ; 22(9): 1646-54, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15117987

RESUMEN

PURPOSE: To evaluate safety and preliminary efficacy of the humanized anti-epidermal growth factor receptor monoclonal antibody h-R3 in combination with radiotherapy (RT) in unresectable head and neck cancer patients. Secondary end points were the measurement of h-R3 serum levels and the assessment of the potential mechanisms of antitumor effect on patient biopsies. Anti-idiotypic response to h-R3 was assessed. To predict pharmacologic effect, a mathematical model for antibodies recognizing antigens expressed in tumors and normal tissues was built. PATIENTS AND METHODS: Twenty-four patients with advanced carcinomas of the head and neck received six once-weekly infusions of h-R3 at four dose levels in combination with RT. Pretreatment tumor biopsies were obtained to evaluate epidermal growth factor receptor expression as an enrollment criterion. Second biopsies were taken to evaluate the proliferative activity and angiogenesis in comparison with the pretreatment samples. Patient serum samples were collected to measure h-R3 levels and anti-idiotypic response. RESULTS: The combination of h-R3 and RT was well tolerated. Antibody-related adverse events consisted in infusion reactions. No skin or allergic toxicity appeared. Overall survival significantly increased after the use of the higher antibody doses. Immunohistochemistry studies of tumor specimens before and after treatment revealed that antitumor response correlated with antiproliferative and antiangiogenic effect. One patient developed antibodies to h-R3. The mathematical model predicted that the maximum difference between the area under the curve in tumors and normal tissues is reached when the antibody has intermediate affinity. CONCLUSION: h-R3 is a well-tolerated drug that may enhance radiocurability of unresectable head and neck neoplasms.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antineoplásicos/inmunología , Carcinoma/inmunología , Carcinoma/radioterapia , Receptores ErbB/inmunología , Receptores ErbB/uso terapéutico , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/radioterapia , Anciano , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Antineoplásicos/análisis , Biopsia , Terapia Combinada , Femenino , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Neovascularización Patológica , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...