Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Neurooncol ; 147(3): 599-605, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32274629

RESUMEN

PURPOSE: Glioblastoma (GBM) is the most common and malignant primary adult brain tumor. Current care includes surgical resection, radiation, and chemotherapy. Recent clinical trials for GBM have demonstrated extended survival using interventions such as tumor vaccines or tumor-treating fields. However, prognosis generally remains poor, with expected survival of 20 months after randomization. Chemokine-based immunotherapy utilizing CCL21 locally recruits lymphocytes and dendritic cells to enhance host antitumor response. Here, we report a preliminary study utilizing CPZ-vault nanoparticles as a vehicle to package, protect, and steadily deliver therapy to optimize CCL21 therapy in a murine flank model of GBM. METHODS: GL261 cells were subcutaneously injected into the left flank of eight-week-old female C57BL/6 mice. Mice were treated with intratumoral injections of either: (1) CCL21-packaged vault nanoparticles (CPZ-CCL21), (2) free recombinant CCL21 chemokine empty vault nanoparticles, (3) empty vault nanoparticles, or 4) PBS. RESULTS: The results of this study showed that CCL21-packaged vault nanoparticle injections can decrease the tumor volume in vivo. Additionally, this study showed mice injected with CCL21-packaged vault nanoparticle had the smallest average tumor volume and remained the only treatment group with a negative percent change in tumor volume. CONCLUSIONS: This preliminary study establishes vault nanoparticles as a feasible vehicle to increase drug delivery and immune response in a flank murine model of GBM. Future animal studies involving an intracranial orthotopic tumor model are required to fully evaluate the potential for CCL21-packaged vault nanoparticles as a strategy to bypass the blood brain barrier, enhance intracranial immune activity, and improve intracranial tumor control and survival.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Quimiocina CCL21/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Glioblastoma/inmunología , Glioblastoma/patología , Inmunoterapia/métodos , Animales , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Quimiocina CCL21/inmunología , Femenino , Glioblastoma/terapia , Ratones Endogámicos C57BL , Nanopartículas
2.
Sci Rep ; 9(1): 5165, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30914733

RESUMEN

Juvenile tissue healing is capable of extensive scarless healing that is distinct from the scar-forming process of the adult healing response. Although many growth factors can be found in the juvenile healing process, the molecular mechanisms of juvenile tissue healing are poorly understood. Here we show that juvenile mice deficient in the chemokine receptor CCR7 exhibit diminished large-scale healing potential, whereas CCR7-depleted adult mice undergo normal scar-forming healing similar to wild type mice. In addition, the CCR7 ligand CCL21 was transiently expressed around damaged cartilage in juvenile mice, whereas it is rarely expressed in adults. Notably, exogenous CCL21 administration to adults decreased scar-forming healing and enhanced hyaline-cartilage repair in rabbit osteochondral defects. Our data indicate that the CCL21/CCR7 axis may play a role in the molecular control mechanism of juvenile cartilage repair, raising the possibility that agents modulating the production of CCL21 in vivo can improve the quality of cartilage repair in adults. Such a strategy may prevent post-traumatic arthritis by mimicking the self-repair in juvenile individuals.


Asunto(s)
Envejecimiento/metabolismo , Cartílago/patología , Quimiocina CCL21/metabolismo , Receptores CCR7/metabolismo , Transducción de Señal , Cicatrización de Heridas , Animales , Huesos/efectos de los fármacos , Huesos/patología , Cartílago/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL21/administración & dosificación , Quimiocina CCL21/farmacología , Condrocitos/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Femenino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/patología , Ratones Endogámicos C57BL , Conejos , Cicatrización de Heridas/efectos de los fármacos
3.
Cancer Immunol Immunother ; 67(12): 1853-1862, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30209589

RESUMEN

The GM.CD40L vaccine, which recruits and activates dendritic cells, migrates to lymph nodes, activating T cells and leading to systemic tumor cell killing. When combined with the CCL21 chemokine, which recruits T cells and enhances T-cell responses, additive effects have been demonstrated in non-small cell lung cancer mouse models. Here, we compared GM.CD40L versus GM.CD40L plus CCL21 (GM.CD40L.CCL21) in lung adenocarcinoma patients with ≥ 1 line of treatment. In this phase I/II randomized trial (NCT01433172), patients received intradermal vaccines every 14 days (3 doses) and then monthly (3 doses). A two-stage minimax design was used. During phase I, no dose-limiting toxicities were shown in three patients who received GM.CD40L.CCL21. During phase II, of evaluable patients, 5/33 patients (15.2%) randomized for GM.DCD40L (p = .023) and 3/32 patients (9.4%) randomized for GM.DCD40L.CCL21 (p = .20) showed 6-month progression-free survival. Median overall survival was 9.3 versus 9.5 months with GM.DCD40L versus GM.DCD40L.CCL21 (95% CI 0.70-2.25; p = .44). For GM.CD40L versus GM.CD40L.CCL21, the most common treatment-related adverse events (TRAEs) were grade 1/2 injection site reaction (51.4% versus 61.1%) and grade 1/2 fatigue (35.1% versus 47.2%). Grade 1 immune-mediated TRAEs were isolated to skin. No patients showed evidence of pseudo-progression or immune-related TRAEs of grade 1 or greater of pneumonitis, endocrinopathy, or colitis, and none discontinued treatment due to toxicity. Although we found no significant associations between vaccine immunogenicity and outcomes, in limited biopsies, one patient treated with GMCD40L.CCL21 displayed abundant tumor-infiltrating lymphocytes. This possible effectiveness warrants further investigation of GM.CD40L in combination approaches.


Asunto(s)
Adenocarcinoma/terapia , Ligando de CD40/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/terapia , Quimiocina CCL21/administración & dosificación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Inmunoterapia , Adenocarcinoma/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia
4.
Otolaryngol Head Neck Surg ; 158(1): 110-117, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28895464

RESUMEN

Objective To evaluate the adverse effects and therapeutic efficacy of our biocompatible polymer platform delivering targeted local therapy of cytokine CCL21 and cisplatin in a partially resected xenograft animal model of head and neck squamous cell carcinoma. In addition, to evaluate the efficacy of cotreatment with radiotherapy and assess the biocompatibility of the cisplatin-eluting polymer in the murine neck. Study Design Experimental animal study. Setting Academic research laboratory. Subjects and Methods SCCVII/SF cell injection established head and neck squamous cell carcinoma tumors in C3H/HeJ mice. Subjects underwent surgery, and a chemokine-eluting polymer was implanted into the resected site. Subjects treated with cisplatin received radiation or no radiation, and tissue was harvested after 8 weeks to assess polymer biocompatibility. Results Our results with the polymer platform significantly ( P < .05) reduced SCCVII/SF tumor size in C3H/HeJ mice with cisplatin (49% ± 8.7%, Δ3.4 ± 0.6 cm3 [95% CI]), CCL21 (42% ± 4.8%, Δ3.5 ± 0.4 cm3), and cisplatin/CCL21 dual-agent polymer (82% ± 4.4%, Δ8.0 ± 0.4 cm3) as compared with controls. Cisplatin polymer with high-dose (16 Gy) and low-dose (4 Gy) radiation reduced tumor mass (respectively, 92% ± 7.2%, Δ6.1 ± 0.5 cm3; 85% ± 7.4%, Δ5.7 ± 0.5 cm3) as compared with the reduction from high-dose radiotherapy alone (70% ± 7.9%, Δ4.7 ± 0.5 cm3). No significant toxicity or inflammation was noted on histopathology after radiotherapy and cisplatin-eluting polymer treatment. Conclusion Cisplatin, CCL21, and cisplatin/CCL21 dual-agent polymer all exhibit significant antitumor effects and decrease tumor burden. Moreover, combination cisplatin polymer with radiotherapy may permit a decrease in intensity of radiation therapy in patients having received the cisplatin polymer. Histopathologic analysis suggests that the polymer is free from significant adverse effects in this model and warrants clinical trial.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Quimiocina CCL21/administración & dosificación , Cisplatino/administración & dosificación , Sistemas de Liberación de Medicamentos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Polímeros/farmacología , Animales , Materiales Biocompatibles/farmacología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Ratones , Poliésteres/farmacología
5.
Blood ; 124(2): 296-304, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-24876562

RESUMEN

Development of T cells in the thymus requires continuous importation of T-lineage progenitors from the bone marrow via the circulation. Following bone marrow transplant, recovery of a normal peripheral T-cell pool depends on production of naïve T cells in the thymus; however, delivery of progenitors to the thymus limits T-lineage reconstitution. Here, we examine homing of intravenously delivered progenitors to the thymus following irradiation and bone marrow reconstitution. Surprisingly, following host conditioning by irradiation, we find that homing of lymphoid-primed multipotent progenitors and common lymphoid progenitors to the thymus decreases more than 10-fold relative to unirradiated mice. The reduction in thymic homing in irradiated mice is accompanied by a significant reduction in CCL25, an important chemokine ligand for thymic homing. We show that pretreatment of bone marrow progenitors with CCL25 and CCL21 corrects the defect in thymic homing after irradiation and promotes thymic reconstitution. These data suggest new therapeutic approaches to promote T-cell regeneration.


Asunto(s)
Trasplante de Médula Ósea , Movimiento Celular/efectos de los fármacos , Quimiocina CCL21/administración & dosificación , Quimiocinas CC/administración & dosificación , Células Progenitoras Linfoides/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Acondicionamiento Pretrasplante , Animales , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/inmunología , Células Cultivadas , Femenino , Células Progenitoras Linfoides/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T/fisiología , Timo/citología , Timo/efectos de los fármacos , Timo/efectos de la radiación
6.
Mol Cancer ; 12(1): 153, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24304581

RESUMEN

BACKGROUND: Secondary lymphoid tissue chemokine (SLC) is a key CC chemokine for chemotaxis of immune cells and has been an attractive candidate for anti-tumor treatments. However, among the immune cells recruited by SLC to tumors, the CD25+ Foxp3+ regulatory T cells (Tregs) compromise the anti-tumor effects. In this study, we proposed the combination therapy of intratumoral co-administration of SLC and anti-CD25 monoclonal antibodies (mAbs). We hypothesized that the intratumoral injections of SLC and depletion of Tregs would have stronger inhibition effects on the progression of hepatocellular carcinoma (HCC) in mice. METHODS: C57BL/6 mice were inoculated subcutaneously with the murine HCC cell line, and mice with visible tumors were treated intratumorally with SLC, SLC plus anti-CD25 mAbs or the control antibodies. The percentages of Tregs, effector CD8+ T cells and CD4+ T cells were checked in the tumors, lymph nodes, spleen and liver at regular intervals. The levels of intratumoral IL-12, IFN-γ, IL-10 and TGF-ß1 were evaluated. The final anti-tumor effects were measured by the tumor volume and weight as well as the intratumoral activity of MMP2 and MMP9. Bone-marrow-derived dendritic cells were used to explore the mechanisms of maturation induced by SLC in vitro. RESULTS: Our experiments showed the combination therapy significantly decreased the frequency of Tregs, and increased CD8+ T cells and CD4+ T cells at tumor sites. These alterations were accompanied by an increased level of IL-12 and IFN-γ, and decreased level of IL-10 and TGF-ß1. Unexpectedly, we observed a significantly decreased percentage of Tregs, and increased CD8+ T cells and CD4+ T cells in the lymph nodes, spleen and liver after the combination therapy. The growth and invasiveness of HCC was also maximally inhibited in the combination therapy compared with the SLC alone. Furthermore, we confirmed SLC induced the maturation of DCs via NF-κB p65 and this maturation would benefit the combination therapy. CONCLUSIONS: Our data demonstrated that intratumoral co-administration of SLC and anti-CD25 mAbs was an effective treatment for HCC, which was correlated with the altered tumor microenvironment and systemically optimized percentages of Tregs, CD8+ T cells and CD4+ T cells in peripheral immune organs.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma Hepatocelular/terapia , Quimiocina CCL21/administración & dosificación , Neoplasias Hepáticas Experimentales/terapia , Depleción Linfocítica , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Carcinoma Hepatocelular/inmunología , Terapia Combinada , Células Dendríticas/metabolismo , Factores de Transcripción Forkhead/metabolismo , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Hígado/inmunología , Neoplasias Hepáticas Experimentales/inmunología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores CCR7/metabolismo , Bazo/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Korean Med Sci ; 26(10): 1270-6, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22022177

RESUMEN

Secondary lymphoid tissue chemokine (SLC), which is expressed in T cell zones of secondary lymphoid organs, including the spleen and lymph nodes, strongly recruits both T lymphocytes and mature dendritic cells. As appropriate interaction of tumor-specific T cells and mature dendritic cells, equipped with tumor antigens, is a prerequisite for effective T cell immunity against established tumors, we mobilized lymphocytes and dendritic cells to tumor sites by intratumoral injection of secondary lymphoid tissue chemokine-Fc (SLC-Fc) fusion protein using the B16F10 murine melanoma model. Activation of dendritic cells, another prerequisite for the effective activation of naïve tumor-specific T cells, was achieved by the addition of immunostimulatory cytosine-phosphorothioate-guanine oligodeoxynucleotide (CpG-ODN) into the tumor site. Intratumoral administration of SLC-Fc or CpG-ODN revealed antitumor effects against B16F10 murine melanoma grown in the subcutaneous space. Co-treatment of SLC-Fc and CpG-ODN displayed synergistic effects in reducing the tumor size. The synergistic antitumor effect in co-treatment group was correlated with the synergistic/additive increase in the infiltration of CD4(+) T cells and CD11c(+) dendritic cells in the tumor mass compared to the single treatment groups. These results suggest that the combined use of chemokines and adjuvant molecules may be a possible strategy in clinical tumor immunotherapy.


Asunto(s)
Quimiocina CCL21/administración & dosificación , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Oligodesoxirribonucleótidos/administración & dosificación , Animales , Antígeno CD11c/inmunología , Linfocitos T CD4-Positivos/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocina CCL21/farmacología , Quimiotaxis de Leucocito , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inmunoterapia , Inyecciones Intralesiones , Ratones , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/farmacología , Linfocitos T/inmunología , Linfocitos T/metabolismo
8.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-127701

RESUMEN

Secondary lymphoid tissue chemokine (SLC), which is expressed in T cell zones of secondary lymphoid organs, including the spleen and lymph nodes, strongly recruits both T lymphocytes and mature dendritic cells. As appropriate interaction of tumor-specific T cells and mature dendritic cells, equipped with tumor antigens, is a prerequisite for effective T cell immunity against established tumors, we mobilized lymphocytes and dendritic cells to tumor sites by intratumoral injection of secondary lymphoid tissue chemokine-Fc (SLC-Fc) fusion protein using the B16F10 murine melanoma model. Activation of dendritic cells, another prerequisite for the effective activation of naive tumor-specific T cells, was achieved by the addition of immunostimulatory cytosine-phosphorothioate-guanine oligodeoxynucleotide (CpG-ODN) into the tumor site. Intratumoral administration of SLC-Fc or CpG-ODN revealed antitumor effects against B16F10 murine melanoma grown in the subcutaneous space. Co-treatment of SLC-Fc and CpG-ODN displayed synergistic effects in reducing the tumor size. The synergistic antitumor effect in co-treatment group was correlated with the synergistic/additive increase in the infiltration of CD4+ T cells and CD11c+ dendritic cells in the tumor mass compared to the single treatment groups. These results suggest that the combined use of chemokines and adjuvant molecules may be a possible strategy in clinical tumor immunotherapy.


Asunto(s)
Animales , Ratones , Antígeno CD11c/inmunología , Linfocitos T CD4-Positivos/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocina CCL21/administración & dosificación , Quimiotaxis de Leucocito , Células Dendríticas/inmunología , Inmunoterapia , Inyecciones Intralesiones , Melanoma Experimental/inmunología , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/administración & dosificación , Linfocitos T/inmunología
9.
J Ethnopharmacol ; 120(2): 215-9, 2008 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-18790041

RESUMEN

AIM OF STUDY: This study was performed to elicit the effectiveness of bee venom (BV), a traditional immunosuppressive Korean acupuncture agent, on the maturation of dendrtic cells (DCs). MATERIALS AND METHODS: Immature dendritic cells (iDCs) were generated from mouse bone marrow cells with GM-CSF. After 10 days of initial differentiation, DCs were activated with lipopolysaccharides (LPS) for another 48h in the presence or absence of BV. Surface molecule analysis, intracytoplasmic staining of cytokines, FITC-conjugated antigen uptake, and transwell migration assays were conducted with iDCs and activated DCs. RESULTS: Up-regulation of costimulatory molecules, typical of mature DCs (mDCs) was inhibited by addition of BV. Pro-inflammatory cytokines were also found to be reduced with BV treatment in LPS-stimulated DC. A decrease in antigen uptake upon the maturation of DC was reversed in low dose BV treated mDC. In addition, BV treated mDC demonstrated reduced directional migration in response to CCL21, a lymphoid chemokine which directs mDC. CONCLUSIONS: BV may have a therapeutic effect an on abnormally activated immune status, such as autoimmune rheumatoid arthritis, through an immune-modulatory effect on DC.


Asunto(s)
Antígenos CD/efectos de los fármacos , Venenos de Abeja/farmacología , Células Dendríticas/efectos de los fármacos , Inmunosupresores/farmacología , Animales , Antígenos CD/inmunología , Venenos de Abeja/administración & dosificación , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Quimiocina CCL21/administración & dosificación , Células Dendríticas/inmunología , Relación Dosis-Respuesta a Droga , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Inmunosupresores/administración & dosificación , Mediadores de Inflamación/metabolismo , Lipopolisacáridos , Medicina Tradicional Coreana , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba/efectos de los fármacos
10.
Zhonghua Yi Xue Za Zhi ; 88(44): 3112-5, 2008 Dec 02.
Artículo en Chino | MEDLINE | ID: mdl-19159591

RESUMEN

OBJECTIVE: To explore the impact of secondary lymphoid tissue chemokine (SLC) on lymphocyte migration and the significance thereof in the pathogenesis of ulcerative colitis (UC). METHODS: Sixty SD rats were randomly divided into 3 equal groups: model group undergoing dripping of 40% acetone solution of dinitro-chlorobenzene (DNCB) on the back for 2 weeks and then enema of 6% DNCB acetone solution so as to establish models of UC, and then intravenous injection of normal saline (NS) for 5 days; SLC antibody intervention group undergoing intravenous injection of SLC antibody 15 microg x ml(-1) x kg(-1) immediately after the establishing of model; and control group undergoing enema of NS nly and then intravenous injection of NS for 5 days. Six days after the establishing of model venous blood samples were collected from the portal veins of the 3 groups. Lymphocytes were isolated and cultured. RT-PCR was used to detect the mRNA expression of the SLC receptor CCR7. Boyden chamber system was used to examine the migration ability of the lymphocytes exposed to SLC of 20, 40, 60, 80, and 100 ng/ml respectively. ELISA was used to detect the expression of interleukin (IL)-10 and interferon (IFN)-gamma in the supernatants of the lymphocytes of different groups. RESULTS: RT-PCR showed that the CCR7 mRNA expression level of the model group was (0.792 +/- 0.108), significantly higher than that of the intervention group (0.386 +/- 0.115, P = 0.0429), and the CCR7 mRNA expression levels of these 2 groups were both significantly higher than that of the control group (0.106 +/- 0.029, both P < 0.01). SLC dose-dependently promoted the migration ability of the lymphocytes, but there existed a saturation phenomenon. Exposed to 80 ng/ml SLC the migration level of the lymphocytes of the model group peaked to (85.9 +/- 16.0), 3.7 times as high as that of the control group (20.5 +/- 1.8, P < 0.01), and the migration level of the lymphocytes of the intervention group was 38.2 +/- 6.3, significantly higher than that of the control group too (P < 0.05). SLC enhanced the expression of IFN-gamma of the lymphocytes of the model group, while reduced the IL-10 level, and both effects peaked at the concentration of 80 ng/ml (P = 0.042, P = 0.036). CONCLUSION: SLC promotes the lymphocyte migration and boosts the differentiation of lymphocytes, thus participating in the pathogenesis of UC.


Asunto(s)
Quimiocina CCL21/metabolismo , Colitis Ulcerosa/metabolismo , Linfocitos/citología , Animales , Movimiento Celular , Células Cultivadas , Quimiocina CCL21/administración & dosificación , Colitis Ulcerosa/inmunología , Femenino , Interferón gamma/metabolismo , Interleucina-10/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores CCR7/metabolismo
11.
Cancer Biol Ther ; 6(8): 1206-10, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17617742

RESUMEN

In previous studies, the chemokine CCL21 has shown biological activities that include T cell, natural killer (NK) cell, and dendritic cell (DC) chemoattraction. The goal of this study was to determine the effects of administering CCL21 to orthotopic mammary tumors in terms of impact on tumor growth rate, immune cell infiltration of the primary tumor and survival. We found that a single intratumoral administration of CCL21 slowed the growth of orthotopic mammary tumors and increased intratumoral infiltration by T cells, NK cells and DCs. CCL21 intratumoral administration also prolonged the survival of tumor-earing mice. Furthermore, mice that received intratumoral neoadjuvant CCL21 ior to surgical resection of tumors survived significantly longer than control mice. The urviving neoadjuvant CCL21-reated mice, when challenged again with cl-6, had significantly slower rate of tumor growth than challenged control mice. Thus, our ata indicate that CCL21 treatment prior to mammary tumor resection can significantly rolong survival and increase resistance to subsequent tumor challenge. Overall, our indings suggest that intratumoral administration of CCL21 has potential as a neoadjuvant mmunotherapy for breast cancer.


Asunto(s)
Quimiocina CCL21/uso terapéutico , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/inmunología , Terapia Neoadyuvante/métodos , Animales , Quimiocina CCL21/administración & dosificación , Células Dendríticas/efectos de los fármacos , Femenino , Células Asesinas Naturales/efectos de los fármacos , Linfocitos Infiltrantes de Tumor , Neoplasias Mamarias Animales/cirugía , Ratones , Ratones Endogámicos BALB C , Linfocitos T/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...