Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Oncologist ; 28(1): e1-e8, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36342104

RESUMO

INTRODUCTION: Patients with gastrointestinal (GI) cancers have an increased risk of serious complications and death from SARS-CoV-2 infection. The immunogenicity of vaccines in patients with GI cancers receiving anti-cancer therapies is unclear. We conducted a prospective study to evaluate the prevalence of neutralizing antibodies in a cohort of GI cancer patients receiving chemotherapy following SARS-CoV-2 vaccination. MATERIALS AND METHODS: Between September 2020 and April 2021, patients with cancer undergoing chemotherapy were enrolled. At baseline (day 0), days 28, 56, and 84, we assessed serum antibodies to SARS-CoV-2 spike (anti-S) and anti-nucleocapsid (anti-NP) and concomitantly assessed virus neutralization using a pseudovirus neutralization assay. Patients received either the Pfizer/BioNTech BNT162b2, or the Oxford/AstraZeneca ChAdOx1 vaccine. RESULTS: All 152 patients enrolled had a prior diagnosis of cancer; colorectal (n = 80, 52.6%), oesophagogastric (n = 38, 25.0%), and hepato pancreatic biliary (n = 22, 12.5%). Nearly all were receiving systemic anti-cancer therapy (99.3%). Of the 51 patients who did not receive a vaccination prior to, or during the study, 5 patients had detectable anti-NP antibodies. Ninety-nine patients received at least one dose of vaccine prior to, or during the study. Within 19 days following the first dose of vaccine, 30.0% had anti-S detected in serum which increased to 70.2% at days 20-39. In the 19 days following a second dose, anti-S positivity was 84.2% (32/38). However, pseudovirus neutralization titers (pVNT80) decreased from days 20 to 39. CONCLUSION: Despite the immunosuppressive effects of chemotherapy, 2 doses of SARS-CoV-2 vaccines are able to elicit a protective immune response in patients' ongoing treatment for gastrointestinal cancers. Decreases in pseudoviral neutralization were observed after 20-39 days, re-affirming the current recommendation for vaccine booster doses. CLINICAL TRIAL REGISTRATION NUMBER: NCT04427280.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Neoplasias Gastrointestinais , Imunogenicidade da Vacina , Humanos , Anticorpos , Vacina BNT162 , Neoplasias Gastrointestinais/tratamento farmacológico , Estudos Prospectivos , SARS-CoV-2
2.
Front Oncol ; 13: 1258365, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38094609

RESUMO

Background: This study aimed to identify microRNAs (miRs) as circulating biomarkers of resistance to first-line trastuzumab-based therapy in advanced HER2-positive oesophago-gastric cancer patients. Methods: A high-throughput 1015 Exiqon miRCURY LNA™ microRNA inhibitor library screen was performed in trastuzumab-treated HER2-positive NCI-N87 and HER2-negative FLO-1 oesophago-gastric cancer cell lines. NanoString nCounter® miR analysis was performed in NCI-N87, FLO-1, and MAGIC trial (ISRCTN93793971) formalin-fixed paraffin-embedded (FFPE) oesophago-gastric cancer patient samples. MiR-148a-3p copies in plasma samples were quantified using digital droplet polymerase chain reaction (ddPCR) from HER2-positive oesophago-gastric cancer patients treated with standard-of-care trastuzumab-based therapy within the FOrMAT (NCT02112357) and PLATFORM (NCT02678182) clinical trials. The primary endpoints were overall survival (OS) for plasma miR-148a-3p HIGH (>median) versus LOW (≤median). The secondary endpoints were progression-free survival (PFS) and 3-month progression-free rates (PFRs) miR-148a-3p HIGH versus LOW. PLATFORM sensitivity analysis normalised miR-148a-3p (NmiR-148a-3p). Results: The inhibition of miR-148a-3p reduced NCI-N87 relative cell viability (<0.6) and expression was high (>242) in NCI-N87 and HER2-positive MAGIC trial patients (n=5). Normalised-miR-148a-3p (NmiR-148a-3p) LOW versus HIGH demonstrated a statistically significant difference in 3-month PFRs (n=23; OR, 0.11 [0.02-0.78]; p=0.027; aOR, 0.03 [0.001-0.71], p=0.029) but no difference in OS or PFS. There was no statistically significant relationship between miR-148-3p LOW versus HIGH for OS (PLATFORM, n=62; hazard ratio [HR], 0.98 [0.57-1.66]; p=0.933; FOrMAT, n=8; HR, 0.54 [0.13-2.31]; p=0.322), PFS (n=62; HR, 1.08 [0.65-1.81]; p=0.759; FOrMAT, n=8; HR, 1.26 [0.31-5.07]; p=0.714), or PFRs (PLATFORM, n=31; odds ratio [OR], 0.67 [0.2-2.8]; p=0.577). Conclusion: Normalised miR-148a-3p may be a relevant biomarker for trastuzumab-based therapy in advanced HER2-positive oesophago-gastric cancer patients.

3.
Stem Cells ; 27(10): 2614-23, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19650040

RESUMO

Multipotent mesenchymal stromal/stem cells (MSC) have shown potential clinical utility. However, previous assessments of MSC behavior in recipients have relied on visual detection in host tissue following sacrifice, failing to monitor in vivo MSC dispersion in a single animal and limiting the number of variables that can be observed concurrently. In this study, we used noninvasive, in vivo bioluminescent imaging to determine conditions under which MSC selectively engraft in sites of inflammation. MSC modified to express firefly luciferase (ffLuc-MSC) were injected into healthy mice or mice bearing inflammatory insults, and MSC localization was followed with bioluminescent imaging. The inflammatory insults investigated included cutaneous needle-stick and surgical incision wounds, as well as xenogeneic and syngeneic tumors. We also compared tumor models in which MSC were i.v. or i.p. delivered. Our results demonstrate that ffLuc-expressing human MSC (hMSC) systemically delivered to nontumor-bearing animals initially reside in the lungs, then egress to the liver and spleen, and decrease in signal over time. However, hMSC in wounded mice engraft and remain detectable only at injured sites. Similarly, in syngeneic and xenogeneic breast carcinoma-bearing mice, bioluminescent detection of systemically delivered MSC revealed persistent, specific colocalization with sites of tumor development. This pattern of tropism was also observed in an ovarian tumor model in which MSC were i.p. injected. In this study, we identified conditions under which MSC tropism and selective engraftment in sites of inflammation can be monitored by bioluminescent imaging over time. Importantly, these consistent findings were independent of tumor type, immunocompetence, and route of MSC delivery.


Assuntos
Biomarcadores Tumorais/metabolismo , Quimiotaxia/fisiologia , Sobrevivência de Enxerto/fisiologia , Inflamação/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Neoplasias/metabolismo , Animais , Biomarcadores Tumorais/análise , Neoplasias da Mama/metabolismo , Carcinoma/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Humanos , Inflamação/fisiopatologia , Proteínas Luminescentes/metabolismo , Células-Tronco Mesenquimais/citologia , Microscopia de Fluorescência/métodos , Neoplasias/fisiopatologia , Neoplasias Ovarianas/metabolismo , Vísceras/citologia , Vísceras/metabolismo , Ferimentos e Lesões/metabolismo , Ferimentos e Lesões/fisiopatologia
4.
Cytotherapy ; 12(5): 615-25, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20230221

RESUMO

BACKGROUND AIMS: Because of the inflammatory nature and extensive stromal compartment in pancreatic tumors, we investigated the role of mesenchymal stromal cells (MSC) to engraft selectively in pancreatic carcinomas and serve as anti-tumor drug delivery vehicles to control pancreatic cancer progression. METHODS: Human pancreatic carcinoma cells, PANC-1, expressing renilla luciferase were orthotopically implanted into SCID mice and allowed to develop for 10 days. Firefly luciferase-transduced MSC or MSC expressing interferon (IFN)-beta were then injected intraperitoneally weekly for 3 weeks. Mice were monitored by bioluminescent imaging for expression of renilla (PANC-1) and firefly (MSC) luciferase. RESULTS: MSC selectively homed to sites of primary and metastatic pancreatic tumors and inhibited tumor growth (P=0.032). The production of IFN-beta within the tumor site by MSC-IFN-beta further suppressed tumor growth (P=0.0000083). Prior studies indicated that MSC home to sites of inflammation; therefore, we sought to alter the tumor microenvironment through treatment with a potent anti-inflammatory agent. After treatment, inflammation-associated mediators were effectively down-regulated, including NFkappaB, vascular endothelial growth factor (VEGF) and interleukin (IL)-6 as well as chemokines involved in MSC migration (CCL3 and CCL25). Treatment with the anti-inflammatory agent CDDO-Me before and after MSC-IFN-beta injections resulted in reduction of MSC in the tumors and reversed the positive effect of tumor inhibition by MSC-IFN-beta alone (P=0.041). CONCLUSIONS: These results suggest that MSC exhibit innate anti-tumor effects against PANC-1 cells and can serve as delivery vehicles for IFN-beta for the treatment of pancreatic cancer. However, these beneficial effects may be lost in therapies combining MSC with anti-inflammatory agents.


Assuntos
Interferon beta/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Neoplasias Pancreáticas/terapia , Células Estromais/metabolismo , Animais , Antineoplásicos/imunologia , Antineoplásicos/uso terapêutico , Processos de Crescimento Celular/imunologia , Linhagem Celular Tumoral , Terapia Genética , Inibidores do Crescimento/imunologia , Inibidores do Crescimento/uso terapêutico , Humanos , Terapia de Imunossupressão , Inflamação , Interferon beta/genética , Interferon beta/imunologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Células Estromais/patologia , Células Estromais/transplante , Transgenes/genética
5.
Transfusion ; 48(12): 2638-44, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18798803

RESUMO

BACKGROUND: Hematopoietic stem cells (HSCs) are routinely obtained from marrow, mobilized peripheral blood, and umbilical cord blood. Mesenchymal stem cells (MSCs) are traditionally isolated from marrow. Bone marrow-derived MSCs (BM-MSCs) have previously demonstrated their ability to act as a feeder layer in support of ex vivo cord blood expansion. However, the use of BM-MSCs to support the growth, differentiation, and engraftment of cord blood may not be ideal for transplant purposes. Therefore, the potential of MSCs from a novel source, the Wharton's jelly of umbilical cords, to act as stromal support for the long-term culture of cord blood HSC was evaluated. STUDY DESIGN AND METHODS: Umbilical cord-derived MSCs (UC-MSCs) were cultured from the Wharton's jelly of umbilical cord segments. The UC-MSCs were then profiled for expression of 12 cell surface receptors and tested for their ability to support cord blood HSCs in a long-term culture-initiating cell (LTC-IC) assay. RESULTS: Upon culture, UC-MSCs express a defined set of cell surface markers (CD29, CD44, CD73, CD90, CD105, CD166, and HLA-A) and lack other markers (CD45, CD34, CD38, CD117, and HLA-DR) similar to BM-MSCs. Like BM-MSCs, UC-MSCs effectively support the growth of CD34+ cord blood cells in LTC-IC assays. CONCLUSION: These data suggest the potential therapeutic application of Wharton's jelly-derived UC-MSCs to provide stromal support structure for the long-term culture of cord blood HSCs as well as the possibility of cotransplantation of genetically identical, HLA-matched, or unmatched cord blood HSCs and UC-MSCs in the setting of HSC transplantation.


Assuntos
Separação Celular/métodos , Sangue Fetal/citologia , Células-Tronco Mesenquimais/citologia , Forma Celular , Células Cultivadas , Humanos , Fatores de Tempo
6.
Exp Hematol ; 34(8): 1060-8, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16863912

RESUMO

OBJECTIVE: Cytokine treatment with granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), and stem cell factor (SCF) is a mainstay of current and future clinical and research protocols for peripheral blood stem cell mobilization, therapeutic care after hematopoietic stem cell transplantation (HSCT), and ex vivo hematopoietic stem and progenitor cell (HSC/HPC) expansion. We have previously shown that the peptidase CD26 (DPPIV/dipeptidylpeptidase IV) negatively regulates HSC/HPC and that inhibition of CD26 improves the chemotactic ability and trafficking of HSC/HPC. We set out to establish whether short-term in vitro G-CSF, GM-CSF, or SCF treatment upregulates CD26 and thereby has a detrimental effect on the chemotactic potential of HSC/HPC that could be reversed by CD26 inhibitor treatment. MATERIALS AND METHODS: CD34+ or CD34+CD38- cells, a population enriched in HSC, were isolated from human umbilical cord blood and subjected to G-CSF, GM-CSF, or SCF treatment. We then evaluated CD26 expression, CD26 activity, and CXCL12 (SDF-1)-induced migration in the presence or absence of a CD26 inhibitor, Diprotin A. RESULTS: Treatment with G-CSF and GM-CSF but not SCF upregulates CD26 expression and activity resulting in a CD26 inhibitor-reversible downregulation of CXCL12-induced chemotactic response. CONCLUSIONS: Short-term in vitro G-CSF and GM-CSF treatment upregulates the peptidase CD26, resulting in downregulation of the functional ability of CD34+CD38- cells to respond to the chemokine CXCL12. This suggests that current and future clinical protocols utilizing G-CSF and GM-CSF may have unforeseen detrimental effects on the trafficking of HSC/HPC during HSCT that can be overcome through the use of CD26 inhibitors.


Assuntos
ADP-Ribosil Ciclase 1/análise , Antígenos CD34/análise , Quimiotaxia/efeitos dos fármacos , Dipeptidil Peptidase 4/genética , Sangue Fetal/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células-Tronco Hematopoéticas/fisiologia , Quimiocina CXCL12 , Quimiocinas CXC/fisiologia , Humanos , Regulação para Cima
7.
Methods Mol Biol ; 904: 173-90, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22890932

RESUMO

The act of migration is similar for many cell types. The migratory mechanism of mesenchymal stem cells (MSC) is not completely elucidated, yet many of the initial studies have been based on current understanding of the leukocyte migration. A normal function of MSC is the ability of the cell to migrate to and repair wounded tissue. This wound healing property of MSC originates with migration towards inflammatory signals produced by the wounded environment [1]. A tumor and its microenvironment are capable of eliciting a similar inflammatory response from the MSC, thus resulting in migration of the MSC towards the tumor microenvironment. We have shown MSC migration both in vitro and in vivo. In this chapter, we elucidate several in vivo methods to study MSC migration and mobilization to the tumor microenvironment. The first model is an exogenous model of MSC migration that can be performed in both xenograft and syngenic systems with in vitro expanded MSC. The second model utilizes transgenic-fluorescent--colored mice to follow endogenous bone marrow components including MSC. The third technique enables us to analyze data from the transgenic model through multispectral imaging. Furthermore, the migratory phenotype of MSC can be exploited for use in tumor-targeted gene delivery therapy has been efficacious in animal model studies and is an anticipated therapeutic device in clinical trials.


Assuntos
Movimento Celular/fisiologia , Rastreamento de Células/métodos , Inflamação/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Adipócitos/citologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular , Linhagem Celular , Separação Celular/métodos , Condrócitos/citologia , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Camundongos , Osteócitos/citologia , Microambiente Tumoral
8.
PLoS One ; 7(2): e30563, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22363446

RESUMO

To meet the requirements for rapid tumor growth, a complex array of non-neoplastic cells are recruited to the tumor microenvironment. These cells facilitate tumor development by providing matrices, cytokines, growth factors, as well as vascular networks for nutrient and waste exchange, however their precise origins remain unclear. Through multicolored tissue transplant procedures; we have quantitatively determined the contribution of bone marrow-derived and adipose-derived cells to stromal populations within syngeneic ovarian and breast murine tumors. Our results indicate that subpopulations of tumor-associated fibroblasts (TAFs) are recruited from two distinct sources. The majority of fibroblast specific protein (FSP) positive and fibroblast activation protein (FAP) positive TAFs originate from mesenchymal stem/stromal cells (MSC) located in bone marrow sources, whereas most vascular and fibrovascular stroma (pericytes, α-SMA(+) myofibroblasts, and endothelial cells) originates from neighboring adipose tissue. These results highlight the capacity for tumors to utilize multiple sources of structural cells in a systematic and discriminative manner.


Assuntos
Tecido Adiposo/patologia , Células da Medula Óssea/patologia , Células-Tronco Mesenquimais/patologia , Microambiente Tumoral , Animais , Biomarcadores/metabolismo , Células da Medula Óssea/metabolismo , Linhagem Celular Tumoral , Feminino , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos
9.
Exp Hematol ; 39(5): 580-590.e1, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21291952

RESUMO

OBJECTIVE: Megakaryopoiesis involves commitment of hematopoietic stem cells (HSC) toward the myeloid lineage in combination with the proliferation, maturation, and terminal differentiation of progenitors into megakaryocytes. The exact mechanism of megakaryocyte development from HSC is unknown, but growth factors such as thrombopoietin have been identified as critical. Additionally, it has been suggested that the chemokine CXCL12/stromal-cell derived factor-1α has a role in regulating megakaryopoiesis and thrombopoiesis. We recently reported the importance of the extracellular protease CD26 (dipeptidylpeptidase IV) in regulating HSC responses to CXCL12, as well as modulating HSC trafficking into and out of the bone marrow. However, the importance of CD26 for megakaryopoiesis has not been reported. We therefore compared megakaryocyte development between CD26-deficient (CD26(-/-)) mice and C57BL/6 control mice. MATERIALS AND METHODS: Adult CD26(-/-) mice and C57BL/6 control mice were evaluated using blood differentials, histological analysis, flow cytometric analysis, and progenitor colony assays. RESULTS: Bone marrow from CD26(-/-) mice has a significantly expanded megakaryocyte and megakaryocyte progenitor population compared to control C57BL/6 mice bone marrow. CONCLUSIONS: Our results indicate that endogenous CD26 normally suppresses megakaryopoiesis and that loss of CD26 activity results in expansion of the megakaryocyte progenitor population in vivo. This suggests the potential use of CD26 inhibitors to improve megakaryocyte progenitor function and/or reconstitution of the megakaryocyte cell population.


Assuntos
Dipeptidil Peptidase 4/deficiência , Células Progenitoras de Megacariócitos/citologia , Animais , Divisão Celular , Dipeptidil Peptidase 4/metabolismo , Citometria de Fluxo , Células Progenitoras de Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise Multivariada
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA