Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Breast Cancer Res Treat ; 204(3): 521-530, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38194131

RESUMO

PURPOSE: Boswellic acids, active components of frankincense, suppress tumor proliferation in vitro with a strong clinical trial safety profile in patients with inflammatory diseases. We performed a Phase Ia window of opportunity trial of Boswellia serrata (B. serrata) in patients with breast cancer to evaluate its biologic activity and safety. METHODS: Patients with invasive breast cancer were treated pre-operatively with B. Serrata (2400 mg/day PO) until the night before surgery for a median of 11 days (SD 6 days; range: 5-23 days). Paraffin-embedded sections from pretreatment diagnostic core biopsies and post-treatment surgical excisions were evaluated using a tunnel assay and immunohistochemistry staining with Ki-67 antibodies. A non-intervention retrospective control arm consisting of core and surgical tissue specimens from untreated patients was used to compare patients treated with B. Serrata. The change in proliferation and apoptosis between diagnostic core specimens and surgical specimens was compared between the control and treatment groups using a two-tailed paired t-test. RESULTS: Twenty-two patients were enrolled, of which 20 received treatment, and 18 had sufficient tissue for IHC. There was an increase in percent change in proliferation from core biopsy to surgical excision in the control group (n = 18) of 54.6 ± 21.4%. In the B. serrata-treated group there was a reduction in proliferation between core biopsy and excision (n = 18) of 13.8 ± 11.7%. This difference was statistically significant between the control and B. serrata-treated groups (p = 0.008). There was no difference in change in apoptosis. There were no serious adverse events related to the drug. CONCLUSION: Boswellia serrata inhibited breast cancer proliferation and was well-tolerated in a Phase Ia window of opportunity trial.


Assuntos
Boswellia , Neoplasias da Mama , Franquincenso , Triterpenos , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Estudos Retrospectivos , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico
2.
Breast Cancer Res Treat ; 202(1): 129-137, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37584883

RESUMO

PURPOSE: High-risk breast pathology is a breast cancer risk factor for which timely treatment is crucial. Nurse navigation programs have been implemented to minimize delays in patient care. This study evaluated nurse navigation in terms of timeliness to surgery for patients with high-risk breast pathology. METHODS: This was a single-institution, retrospective review of patients with identified high-risk breast pathology undergoing lumpectomy between January 2017 and June 2019. Patients were stratified into cohorts based on periods with and without nurse navigation. Preoperative and postoperative time to care as well as demographic and tumor characteristics were compared using univariate and multivariate analysis. RESULTS: 100 patients had assigned nurse navigators and 29 patients did not. Nurse navigation was associated with reduced time from referral to date of surgery (DOS) by 16.9 days (p = 0.003). Patients > 75 years had a shorter time to first appointment (p = 0.03), and patients with Medicare insurance had a reduced time from referral to DOS (p = 0.005). 20% of all patients were upstaged to cancer on final surgical pathology. CONCLUSION: Nurse navigation was significantly associated with decreased time to care for patients with high-risk breast pathology undergoing lumpectomy. We recommend nurse navigation programs as part of a comprehensive approach for patients with high-risk breast pathology.


Assuntos
Neoplasias da Mama , Navegação de Pacientes , Humanos , Idoso , Estados Unidos , Feminino , Medicare , Neoplasias da Mama/cirurgia , Encaminhamento e Consulta , Estudos Retrospectivos
3.
Breast Cancer Res Treat ; 189(2): 471-481, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34132936

RESUMO

PURPOSE: Shave margins have been shown to decrease positive final margins in partial mastectomy. We investigated prognostic factors associated with residual disease in shave margins. METHODS: Patients with invasive breast carcinoma and ductal carcinoma in situ (DCIS) who had circumferential shave margins excised during lumpectomy were abstracted from a retrospective database from 2015 to 2018. We defined residual occult disease (ROD) as either (1) residual disease in a shave margin when the initial lumpectomy specimen had negative margins or (2) residual disease in a shave margin that did not correspond with the positive lumpectomy margin. We identified the frequency of ROD and conducted logistic regression analysis to identify associated prognostic factors. RESULTS: 166 Patients (139 invasive carcinoma, 27 DCIS) were included with median follow-up of 28 months (9-50 months). Residual occult disease existed in 34 (24.5%) with invasive carcinoma and 8 (29.6%) with DCIS. In univariate analyses of the invasive group, invasive lobular carcinoma and a positive initial, non-corresponding lumpectomy margin were predictive of ROD (OR 3.63, p = 0.04, OR 3.48, p = 0.003 respectively). In multivariate analysis, a positive lumpectomy margin remained significant, p = 0.007. No variables were associated with ROD in DCIS. CONCLUSION: Residual occult disease was shown to be a frequent event in this analysis of lumpectomy with circumferential shave margins. Having a positive initial lumpectomy margin was predictive of ROD in a non-corresponding margin. Surgeons should consider not being selective in their shave margins or margin of re-excision if shave margins were not obtained in their initial surgery.


Assuntos
Neoplasias da Mama , Carcinoma Ductal de Mama , Carcinoma Intraductal não Infiltrante , Neoplasias da Mama/epidemiologia , Neoplasias da Mama/cirurgia , Carcinoma Ductal de Mama/cirurgia , Carcinoma Intraductal não Infiltrante/cirurgia , Feminino , Humanos , Mastectomia , Mastectomia Segmentar , Neoplasia Residual , Prognóstico , Reoperação , Estudos Retrospectivos
4.
J Hepatol ; 66(1): 75-85, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27520877

RESUMO

BACKGROUND & AIMS: We have established a clinically relevant animal model of hepatocellular cancer (HCC) in immune competent mice to elucidate the complex dialog between host immunity and tumors during HCC initiation and progression. Mechanistic findings have been leveraged to develop a clinically feasible anti-tumor chemoimmunotherapeutic strategy. METHODS: Intraperitoneal injection of carbon tetrachloride and intrasplenic inoculation of oncogenic hepatocytes were combined to induce progressive HCCs in fibrotic livers of immunocompetent mice. Immunization and adoptive cell transfer (ACT) were used to dissect the tumor antigen-specific immune response. The ability of the tyrosine kinase inhibitor sunitinib to enhance immunotherapy in the setting of HCC was evaluated. RESULTS: This new mouse model mimics human HCC and reflects its typical features. Tumor-antigen-specific CD8+ T cells maintained a naïve phenotype and remained responsive during early-stage tumor progression. Late tumor progression produced circulating tumor cells, tumor migration into draining lymph nodes, and profound exhaustion of tumor-antigen-specific CD8+ T cells associated with accumulation of programmed cell death protein 1 (PD-1)hi CD8+ T cells and regulatory T cells (Tregs). Sunitinib-mediated tumoricidal effect and Treg suppression synergized with antibody-mediated blockade of PD-1 to powerfully suppress tumor growth and activate anti-tumor immunity. CONCLUSION: Treg accumulation and upregulation of PD-1 provide two independent mechanisms to induce profound immune tolerance in HCC. Chemoimmunotherapy using Food and Drug Administration-approved sunitinib with anti-PD-1 antibodies achieved significant tumor control, supporting translation of this approach for the treatment of HCC patients. LAY SUMMARY: In the current study, we have established a clinically relevant mouse model which mimics human liver cancer. Using this unique model, we studied the response of the immune system to this aggressive cancer. Findings from this trial have led to the development of an innovative and clinically feasible chemoimmunotherapeutic strategy.


Assuntos
Carcinoma Hepatocelular , Imunoterapia/métodos , Indóis/farmacologia , Neoplasias Hepáticas , Pirróis/farmacologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/imunologia , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Citotoxicidade Imunológica/fisiologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Antígenos de Histocompatibilidade Classe II/imunologia , Tolerância Imunológica , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Camundongos , Estadiamento de Neoplasias , Receptor de Morte Celular Programada 1/metabolismo , Sunitinibe , Linfócitos T Reguladores/imunologia
5.
Cancer Immunol Immunother ; 66(6): 737-751, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28280853

RESUMO

Adoptive cellular therapy (ACT) with the Th17 subset of CD4+ T cells can cure established melanoma in preclinical models and holds promise for treating human cancer. However, little is known about the growth factors necessary for optimal engraftment and anti-tumor activity of Th17 cells. Due to the central role of IL-2 receptor gamma chain (IL2Rγ-chain) cytokines (IL-2, IL-7, and IL-15) in the activity and persistence of many T cell subsets after adoptive transfer, we hypothesized that these cytokines are important for Th17 cells. We found that Th17 cells proliferated in response to IL-2, IL-7, and IL-15 in vitro. However, in contrast to many other T cell subsets, including conventionally activated CD8+ T cells, we found that Th17 cells were resistant to apoptosis in the absence of IL2Rγ-chain cytokines. To determine whether Th17 cells utilize IL2Rγ-chain cytokines in vivo, we tracked Th17 cell engraftment after adoptive transfer with or without cytokine depletion. Depletion of IL-7 and/or IL-2 decreased initial engraftment, while depletion of IL-15 did not. Supplementation of IL-2 increased initial Th17 engraftment. To assess the clinical relevance of these findings, we treated melanoma-bearing mice with Th17 cell adoptive transfer and concurrent cytokine depletion or supplementation. We found that simultaneous depletion of IL-2 and IL-7 decreased therapeutic efficacy, depletion of IL-15 had no effect, and IL-2 supplementation increased therapeutic efficacy. Our results show that Th17 cells are responsive to IL2Rγ-chain cytokines, and provide insight into the application of these cytokines for Th17-based therapeutic strategies.


Assuntos
Apoptose/efeitos dos fármacos , Subunidade gama Comum de Receptores de Interleucina/imunologia , Interleucina-15/farmacologia , Interleucina-2/farmacologia , Interleucina-7/farmacologia , Melanoma Experimental/imunologia , Células Th17/imunologia , Animais , Antineoplásicos/farmacologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Imunoterapia Adotiva , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Melanoma Experimental/patologia , Melanoma Experimental/prevenção & controle , Camundongos , Células Th17/metabolismo
6.
J Immunol ; 194(4): 1737-47, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25576595

RESUMO

The inducible costimulator (ICOS) plays a key role in the development of Th17 cells, but its role in the development and antitumor activity of IL-17-producing CD8(+) T cells (Tc17) remains unknown. We found that ICOS costimulation was important for the functional maintenance, but not differentiation, of Tc17 cells in vitro. Blocking the ICOS pathway using an antagonist mAb or by using recipient mice genetically deficient in the ICOS ligand reduced the antitumor activity of adoptively transferred Tc17 cells. Conversely, activating Tc17 cells with an ICOS agonist in vitro enhanced their capacity to eradicate melanoma and induce autoimmune vitiligo when infused into mice. However, ICOS stimulation did not augment the antitumor activity of IL-2 expanded T cells. Additional investigation revealed that ICOS stimulation not only increased IL-2Rα, CXCR3, and IL-23R expression on Tc17 cells, but also dampened their expression of suppressive molecule CD39. Although Tc17 cells activated with an ICOS agonist cosecreted heightened IL-17A, IL-9, and IFN-γ, their therapeutic effectiveness was critically dependent on IFN-γ production. Depletion of IL-17A and IL-9 had little impact on antitumor Tc17 cells activated with an ICOS agonist. Collectively, our work reveals that the ICOS pathway potentiates the antitumor activity of adoptively transferred Tc17 cells. This work has major implications for the design of vaccine, Ab and cell-based therapies for autoimmunity, infectious disease, and cancer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunoterapia Adotiva/métodos , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Melanoma/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Diferenciação Celular/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Interleucina-17/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL
7.
Biol Cybern ; 110(2-3): 91-116, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27086133

RESUMO

In comparison with the high level of knowledge about vehicle dynamics which exists nowadays, the role of the driver in the driver-vehicle system is still relatively poorly understood. A large variety of driver models exist for various applications; however, few of them take account of the driver's sensory dynamics, and those that do are limited in their scope and accuracy. A review of the literature has been carried out to consolidate information from previous studies which may be useful when incorporating human sensory systems into the design of a driver model. This includes information on sensory dynamics, delays, thresholds and integration of multiple sensory stimuli. This review should provide a basis for further study into sensory perception during driving.


Assuntos
Condução de Veículo/psicologia , Cibernética , Humanos , Percepção/fisiologia
8.
Cancer Immunol Immunother ; 64(5): 539-49, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25676709

RESUMO

Mouse CD8(+) T cells conditioned with interleukin (IL)-12 ex vivo mediate the potent regression of established melanoma when transferred into lymphodepleted mice. However, the quantitative and qualitative changes induced by IL-12 in the responding mouse CD8(+) T cells have not been well defined. Moreover, the mechanisms by which IL-12-conditioning impacts human CD8(+) T cells, and how such cells might be expanded prior to infusion into patients is not known. We found that ex vivo IL-12-conditioning of mouse CD8(+) T cells led to a tenfold-100-fold increase in persistence and anti-tumor efficacy upon adoptive transfer into lymphodepleted mice. The enhancing effect of IL-12 was associated with maintenance of functional avidity. Importantly, in the context of ongoing ACT clinical trials, human CD8(+) T cells genetically modified with a tyrosinase-specific T cell receptor (TCR) exhibited significantly enhanced functional activity when conditioned with IL-12 as indicated by heightened granzyme B expression and elevated peptide-specific CD107a degranulation. This effect was sustainable despite the 20 days of in vitro cellular expansion required to expand cells over 1,000-fold allowing adequate cell numbers for administration to cancer patients. Overall, these findings support the efficacy and feasibility of ex vivo IL-12-conditioning of TCR-modified human CD8(+) T cells for adoptive transfer and cancer therapy.


Assuntos
Transferência Adotiva , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/transplante , Interleucina-12/farmacologia , Melanoma/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Neoplasias Cutâneas/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Granzimas/biossíntese , Humanos , Interleucina-12/imunologia , Depleção Linfocítica , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Cutâneas/imunologia
9.
Blood ; 120(1): 1-2, 2012 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-22767572

RESUMO

In this issue of Blood, Bergamaschi and colleagues demonstrate that effectively all circulating IL-15 found in the serum of humans and mice is associated with the soluble IL-15Rα.(1)


Assuntos
Subunidade alfa de Receptor de Interleucina-15/sangue , Subunidade alfa de Receptor de Interleucina-15/imunologia , Interleucina-15/sangue , Interleucina-15/química , Animais , Feminino , Humanos
10.
J Immunol ; 188(8): 3639-47, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22430740

RESUMO

CD8(+) T cell responses have been shown to be regulated by dendritic cells (DCs) and CD4(+) T cells, leading to the tenet that CD8(+) T cells play a passive role in their own differentiation. In contrast, by using a DNA vaccination model, to separate the events of vaccination from those of CD8(+) T cell priming, we demonstrate that CD8(+) T cells, themselves, actively limit their own memory potential through CD8(+) T cell-derived IFN-γ-dependent modification of the IL-12/IL-15Rα axis on DCs. Such CD8(+) T cell-driven cytokine alterations result in increased T-bet and decreased Bcl-2 expression, and thus decreased memory progenitor formation. These results identify an unrecognized role for CD8(+) T cells in the regulation of their own effector differentiation fate and a previously uncharacterized relationship between the balance of inflammation and memory formation.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Memória Imunológica , Receptores de Interleucina-12/imunologia , Receptores de Interleucina-15/imunologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/imunologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/imunologia , Humanos , Interferon gama , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-15/genética , Interleucina-15/imunologia , Ativação Linfocitária , Melanoma Experimental/imunologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interleucina-12/genética , Receptores de Interleucina-15/genética , Transdução de Sinais/imunologia , Vacinação , Vacinas de DNA/imunologia
11.
J Immunol ; 189(4): 1627-38, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22798675

RESUMO

Recent advancements in T cell immunotherapy suggest that T cells engineered with high-affinity TCR can offer better tumor regression. However, whether a high-affinity TCR alone is sufficient to control tumor growth, or the T cell subset bearing the TCR is also important remains unclear. Using the human tyrosinase epitope-reactive, CD8-independent, high-affinity TCR isolated from MHC class I-restricted CD4(+) T cells obtained from tumor-infiltrating lymphocytes (TIL) of a metastatic melanoma patient, we developed a novel TCR transgenic mouse with a C57BL/6 background. This HLA-A2-restricted TCR was positively selected on both CD4(+) and CD8(+) single-positive cells. However, when the TCR transgenic mouse was developed with a HLA-A2 background, the transgenic TCR was primarily expressed by CD3(+)CD4(-)CD8(-) double-negative T cells. TIL 1383I TCR transgenic CD4(+), CD8(+), and CD4(-)CD8(-) T cells were functional and retained the ability to control tumor growth without the need for vaccination or cytokine support in vivo. Furthermore, the HLA-A2(+)/human tyrosinase TCR double-transgenic mice developed spontaneous hair depigmentation and had visual defects that progressed with age. Our data show that the expression of the high-affinity TIL 1383I TCR alone in CD3(+) T cells is sufficient to control the growth of murine and human melanoma, and the presence or absence of CD4 and CD8 coreceptors had little effect on its functional capacity.


Assuntos
Autoimunidade , Imunoterapia Adotiva/métodos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Complexo CD3/imunologia , Citometria de Fluxo , Antígeno HLA-A2/imunologia , Humanos , Linfócitos do Interstício Tumoral/imunologia , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Ann Surg ; 258(6): 879-86, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23657083

RESUMO

OBJECTIVE: To determine whether 1 of 2 vaccines based on dendritic cells (DCs) and poxvectors encoding CEA (carcinoembryonic antigen) and MUC1 (PANVAC) would lengthen survival in patients with resected metastases of colorectal cancer (CRC). BACKGROUND: Recurrences after complete resections of metastatic CRC remain frequent. Immune responses to CRC are associated with fewer recurrences, suggesting a role for cancer vaccines as adjuvant therapy. Both DCs and poxvectors are potent stimulators of immune responses against cancer antigens. METHODS: Patients, disease-free after CRC metastasectomy and perioperative chemotherapy (n = 74), were randomized to injections of autologous DCs modified with PANVAC (DC/PANVAC) or PANVAC with per injection GM-CSF (granulocyte-macrophage colony-stimulating factor). Endpoints were recurrence-free survival overall survival, and rate of CEA-specific immune responses. Clinical outcome was compared with that of an unvaccinated, contemporary group of patients who had undergone CRC metastasectomy, received similar perioperative therapy, and would have otherwise been eligible for the study. RESULTS: Recurrence-free survival at 2 years was similar (47% and 55% for DC/PANVAC and PANVAC/GM-CSF, respectively) (χ P = 0.48). At a median follow-up of 35.7 months, there were 2 of 37 deaths in the DC/PANVAC arm and 5 of 37 deaths in the PANVAC/GM-CSF arm. The rate and magnitude of T-cell responses against CEA was statistically similar between study arms. As a group, vaccinated patients had superior survival compared with the contemporary unvaccinated group. CONCLUSIONS: Both DC and poxvector vaccines have similar activity. Survival was longer for vaccinated patients than for a contemporary unvaccinated group, suggesting that a randomized trial of poxvector vaccinations compared with standard follow-up after metastasectomy is warranted. (NCT00103142).


Assuntos
Vacinas Anticâncer , Antígeno Carcinoembrionário , Neoplasias Colorretais/prevenção & controle , Células Dendríticas , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Imunização/métodos , Glicoproteínas de Membrana , Mucina-1 , Recidiva Local de Neoplasia/prevenção & controle , Adulto , Idoso , Antígeno Carcinoembrionário/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/cirurgia , Intervalo Livre de Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mucina-1/genética , Metástase Neoplásica , Poxviridae/genética
13.
IEEE Trans Cybern ; 53(8): 4791-4804, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35077382

RESUMO

Automated vehicle steering control systems have great potential to improve road safety. The development of such systems calls for mathematical driver models able to represent human drivers' steering behavior in response to automated steering intervention. This article concerns the experimental evaluation of a game-theoretic driver steering control model. The driver model centers on a steering control strategy developed based on the Nash equilibrium of a theoretic noncooperative game between the driver and automated steering controller. The key parameters of the game-theoretic driver model are identified by fitting the model to real driver steering behavior measured from six driver subjects in an experiment using a driving simulator. The game-theoretic driver model is evaluated by compared to a "conventional" optimal-control-theoretic driver model, and analyzing their model fitting errors. Results from the analysis demonstrate that the game-theoretic driver model is statistically significantly better than the conventional driver model for representing three out of the six subjects' steering behavior. For the other three subjects, both the two models perform statistically equivalently well.


Assuntos
Condução de Veículo , Teoria dos Jogos , Humanos
14.
Cell Immunol ; 276(1-2): 67-74, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22560674

RESUMO

Recently, we showed that post cyclophosphamide (CTX) microenvironment benefits the function of transferred T cells. Analysis of the kinetics of cellular recovery after CTX treatment showed that a single 4 mg/mouse CTX treatment decreased the absolute number of leukocytes in the peripheral blood (PBL) at days 3-15, and in the spleen and bone marrow (BM) at days 3-6. The absolute numbers of CD11c(+)CD11b(-) and CD11c(+)CD11b(+) dendritic cells (DCs), CD11b(+) and Ly6G(+) myeloid cells, T and B cells, CD4(+)CD25(+) T regulatory (T(reg)) cells, and NK1.1(+) cells also decreased. The cell numbers returned to control levels during the recovery phase. The absolute numbers of B cells remained low for 3 weeks. The numbers of DCs increased in PBL and spleen at day 9 but returned to control levels at day 15. These data indicate that CTX alters the cellular microenvironment in kinetics that might be precisely targeted to benefit the host.


Assuntos
Linfócitos B/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Ciclofosfamida/farmacologia , Células Mieloides/efeitos dos fármacos , Baço/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Medula Óssea/imunologia , Sobrevivência Celular/efeitos dos fármacos , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/imunologia , Baço/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia
15.
J Surg Res ; 176(2): 359-66, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22221605

RESUMO

BACKGROUND: The development of novel therapeutics for pancreatic cancer has been hindered by a lack of relevant preclinical models. The purpose of this study was to evaluate the clinical relevancy of two pancreatic cancer models using standard-of-care therapeutic agent gemcitabine. MATERIALS AND METHODS: Murine Panc02 cells were injected directly into the spleen or pancreas of C57BL/6 mice to respectively create models of metastatic and locally advanced pancreatic cancer. Beginning 7 d post-Panc02 injection, treated mice received 20 mg/kg gemcitabine i.p. every 3 d. Animals were sacrificed when the untreated mice became moribund and tumor/liver weight used to assess tumor burden. RESULTS: Untreated mice became moribund 22 d after pancreatic Panc02 injection. Gross analysis revealed localized pancreatic tumors weighing 1.063 g. Intrasplenic Panc02 injection produced extensive liver metastasis by d 15 when the untreated mice first became moribund. Liver weights at this time averaged 3.6 g compared with the average non-tumor-bearing weight of 1.23 g. Gemcitabine therapy resulted in a 54% decrease in localized pancreatic tumor weight and 62.5% decrease in metastatic liver weight. Additionally, gemcitabine therapy extended animal survival to 20.5 d compared with 18.0 d average for the untreated mice. CONCLUSIONS: We describe two models depicting both locally advanced and metastatic pancreatic cancer in immunocompetent mice. In efforts to establish baseline therapeutic efficacy, we determined that gemcitabine reduces tumor burden in both models and enhances survival in the metastatic model. These clinically relevant models provide valuable tools to evaluate novel therapeutics in pancreatic cancer.


Assuntos
Modelos Animais de Doenças , Imunocompetência/imunologia , Camundongos , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/secundário , Animais , Antimetabólitos Antineoplásicos/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Camundongos Endogâmicos C57BL , Transplante de Neoplasias/métodos , Pâncreas/imunologia , Neoplasias Pancreáticas/tratamento farmacológico , Índice de Gravidade de Doença , Baço/imunologia , Taxa de Sobrevida , Gencitabina
16.
J Immunol ; 184(4): 1737-47, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20083664

RESUMO

Preconditioning a recipient host with lymphodepletion can markedly augment adoptive T cell therapy. However, the precise mechanisms involved are poorly understood. In a recent study, we observed a significant increase in the circulating levels of dendritic cells (DCs; CD11c(+)CD11b(+)) during the recovery from cyclophosphamide (CTX)-induced lymphodepletion. Herein, we demonstrate that the CTX-induced DC expansion was not altered by adjuvant chemotherapy or tumor burden but was augmented by coadministration of granulocyte-colony stimulating factor. Although the increase in the number of DCs was preceded by a systemic expansion of a population expressing the phenotype of myeloid-derived suppressor cells (Gr-1(+)CD11b(+)), depletion of these Gr-1(+) cells had no effect on the noted expansion. Moreover, when Gr-1(high)CD11b(high) cells were sorted from CTX-treated mice and adoptively transferred into control or CTX-treated recipients, they did not differentiate into DCs. Post-CTX expansion of DCs was associated with proliferation of DCs in bone marrow (BM) during the lymphopenic phase and in the blood and spleen during the recovery phase. Furthermore, adoptive transfer of BM cells from CTX-treated mice produced equal numbers of DCs in the blood of either CTX-treated or untreated recipients. CTX induced a dynamic surge in the expression of growth factors and chemokines in BM, where CCR2 and Flt3 signaling pathways were critical for DC expansion. In sum, our data suggest that CTX induces proliferation of DCs in BM prior to their expansion in the periphery. Targeting DCs at these phases would significantly improve their contribution to the clinical application of lymphodepletion to adoptive immunotherapy.


Assuntos
Proliferação de Células , Ciclofosfamida/farmacologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Proteínas de Membrana/fisiologia , Transferência Adotiva , Animais , Antígeno CD11b/biossíntese , Linhagem Celular Tumoral , Ciclofosfamida/administração & dosagem , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Leucopenia/imunologia , Leucopenia/patologia , Ligantes , Depleção Linfocítica , Melanoma Experimental , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Receptores de Quimiocinas/biossíntese
17.
J Immunol ; 182(4): 2030-40, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19201856

RESUMO

Recent preclinical studies suggest that vaccination following adoptive transfer of CD8(+) T cells into a lymphopenic host can augment the therapeutic antitumor responses of the transferred cells. However, the mechanism by which the lymphopenic microenvironment benefits Ag-specific CD8(+) T cell responses remains elusive. We show herein that induction of lymphodepletion by a single 4 mg cyclophosphamide (CTX) treatment induces a marked expansion of immature dendritic cells (DCs) in the peripheral blood on days 8-16 post-CTX (termed restoration phase). In vitro, these DCs were functional, because they showed normal phagocytosis and effective Ag presentation capability upon activation. In vivo, administration of the TLR3 agonist poly(I:C) at the peak of DC expansion (day 12 postlymphopenia) induced inflammatory cytokine production and increases in the number of activated DCs in lymph nodes. Importantly, boosting with gp100(25-33) melanoma peptide combined with poly(I:C) 12 days after an initial priming with the same regimen significantly increased the expansion and the antitumor efficacy of adoptively transferred pmel-1 CD8(+) T cells. These responses were abrogated after depletion of activated DCs during Ag boosting. In conclusion, our data show that CTX treatment induces, during the restoration phase, expansion of immature DCs, which are functional and can be exploited in vivo to foster more effective antitumor adoptive immunotherapy strategies.


Assuntos
Vacinas Anticâncer/imunologia , Ciclofosfamida/toxicidade , Células Dendríticas/imunologia , Imunossupressores/toxicidade , Melanoma Experimental/terapia , Condicionamento Pré-Transplante/métodos , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/transplante , Células Dendríticas/efeitos dos fármacos , Citometria de Fluxo , Imunoterapia Adotiva/métodos , Indutores de Interferon/imunologia , Linfopenia/induzido quimicamente , Melanoma Experimental/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Poli I-C/imunologia , Receptor 3 Toll-Like/imunologia , Antígeno gp100 de Melanoma
18.
Adv Surg ; 45: 249-63, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21954692

RESUMO

PH caused by anesthesia-induced thermoregulatory inhibition and exposure to cold operating room environments still occurs in a significant proportion of patients undergoing major surgery. Although the association between specific perioperative temperatures (in and of themselves) and postoperative morbidity remains unclear, there is fair evidence to suggest that perioperative active warming may reduce the risk of postoperative cardiac events, bleeding, and SSIs. As such, proactive efforts by surgical teams to prevent PH are warranted and have become the standard of care at many institutions. Continued intraoperative monitoring of core temperature (ideally using esophageal probes) is recommended in all cases lasting more than 30 minutes, both to detect malignant hyperthermia and to maintain normothermia. Preoperative and/or intraoperative use of warmed forced-air devices is an effective way to minimize redistribution hypothermia following induction, whereas intraoperative use of warmed i.v. fluids helps reduce the potential for fluid-induced hypothermia and, in turn, optimizes rates of perioperative normothermia.


Assuntos
Hipotermia/fisiopatologia , Procedimentos Cirúrgicos Operatórios , Coagulação Sanguínea/fisiologia , Regulação da Temperatura Corporal/fisiologia , Humanos , Hipotermia/prevenção & controle , Período Perioperatório , Infecção da Ferida Cirúrgica/fisiopatologia , Termômetros
19.
Ann Surg ; 252(4): 696-704, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20881777

RESUMO

OBJECTIVE: To analyze the association between perioperative normothermia (temperature ≥36°C) and surgical site infections (SSIs) after gastrointestinal (GI) surgery. SUMMARY OF BACKGROUND DATA: Although active warming during colorectal surgery reduces SSIs, there is limited evidence that perioperative normothermia is associated with lower rates of SSI. Nonetheless, hospitals participating in the Surgical Care Improvement Project must report normothermia rates during major surgery. METHODS: We conducted a nested, matched, case-control study; cases consisted of GI surgery patients enrolled in our National Surgical Quality Improvement Program database between March 2006 and March 2009 who developed SSIs. Patient/surgery risk factors for SSI were obtained from the National Surgical Quality Improvement Program database. Perioperative temperature/antibiotic/glucose data were obtained from medical records. Cases/controls were compared using univariate/random effects/logistic regression models. Independent risk factors for SSIs were identified using multivariate/random effects/logistic regression models. RESULTS: A total of 146 cases and 323 matched controls were identified; 82% of patients underwent noncolorectal surgery. Cases were more likely to have final intraoperative normothermia compared with controls (87.6% vs. 77.8%, P = 0.015); rates of immediate postoperative normothermia were similar (70.6% vs. 65.3%, respectively, P = 0.19). Emergent surgery/higher wound class were associated with higher rates of intraoperative normothermia. Independent risk factors for SSI were diabetes, surgical complexity, small bowel surgery, and nonlaparoscopic surgery. There was no independent association between perioperative normothermia and SSI (adjusted odds ratio, 1.05; 95% confidence interval, 0.48-2.33; P = 0.90). CONCLUSIONS: Pay-for-reporting measures focusing on perioperative normothermia may be of limited value in preventing SSI after GI surgery. Studies to define the benefit of active warming after noncolorectal GI surgery are warranted.


Assuntos
Temperatura Corporal , Trato Gastrointestinal/cirurgia , Infecção da Ferida Cirúrgica/prevenção & controle , Estudos de Casos e Controles , Feminino , Humanos , Período Intraoperatório , Masculino , Pessoa de Meia-Idade , Análise de Regressão , Infecção da Ferida Cirúrgica/epidemiologia
20.
Cancer Immunol Immunother ; 59(3): 341-53, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19921513

RESUMO

Adoptive transfer of autologous tumor-reactive T cells holds promise as a cancer immunotherapy. In this approach, T cells are harvested from a tumor-bearing host, expanded in vitro and infused back to the same host. Conditioning of the recipient host with a lymphodepletion regimen of chemotherapy or radiotherapy before adoptive T cell transfer has been shown to substantially improve survival and anti-tumor responses of the transferred cells. These effects are further enhanced when the adoptive T cell transfer is followed by vaccination with tumor antigens in combination with a potent immune adjuvant. Although significant progress has been made toward an understanding of the reasons underlying the beneficial effects of lymphodepletion to T cell adoptive therapy, the precise mechanisms remain poorly understood. Recent studies, including ours, would indicate a more central role for antigen presenting cells, in particular dendritic cells. Unraveling the exact role of these important cells in mediation of the beneficial effects of lymphodepletion could provide novel pathways toward the rational design of more effective anti-cancer immunotherapy. This article focuses on how the frequency, phenotype, and functions of dendritic cells are altered during the lymphopenic and recovery phases post-induction of lymphodepletion, and how they affect the anti-tumor responses of adoptively transferred T cells.


Assuntos
Transferência Adotiva , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Depleção Linfocítica , Neoplasias/terapia , Linfócitos T/imunologia , Animais , Antígenos de Neoplasias/uso terapêutico , Humanos , Depleção Linfocítica/métodos , Vacinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA