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
1.
Med Dosim ; 48(4): 312-314, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37716851

RESUMEN

Comprehensive nodal breast irradiation is traditionally treated utilizing 3DCRT planning with a three or four field technique. While the three or four field photon technique may be the standard of care for comprehensive nodal breast treatment, the ipsilateral lung dose and heart mean can often be of concern dependent on patient's anatomy. Standard dose constraints (200cGy x25) per the Alliance A221505 trial strive to maintain the ipsilateral V20<35% and heart mean <3Gy. Double scattering proton therapy treatment has a significant advantage over photons in reducing the ipsilateral lung and heart dose due the Spread-Out Bragg Peak and rapid dose drop off. Proton therapy in comprehensive nodal breast irradiation can be beneficial when dose constraints are exceeding in a photon plan or in the re-irradiation setting. Within this study, a comparison of ipsilateral lung and heart mean doses will be evaluated using both photon three/four field technique and double scattering proton therapy technique. A decreased ipsilateral lung V20 can be correlated to all proton plans that were conducted in the study. In all but one patient there was a decrease in the heart mean as well, which was due to patient anatomy.


Asunto(s)
Neoplasias de la Mama , Terapia de Protones , Radioterapia Conformacional , Humanos , Femenino , Protones , Planificación de la Radioterapia Asistida por Computador/métodos , Radioterapia Conformacional/métodos , Terapia de Protones/métodos , Dosificación Radioterapéutica , Neoplasias de la Mama/radioterapia
2.
Cancers (Basel) ; 15(13)2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-37444636

RESUMEN

Historically, the role of radiation in gynecological metastatic disease involved palliation for pain or bleeding. Stereotactic Body Radiation Therapy (SBRT) has shown survival benefits in oligometastatic disease from varying primary histologies in recent randomized trials. However, gynecologic primary oligometastases have been underrepresented in these trials. Recent studies across gynecological malignancy types have similarly shown favorable outcomes and acceptable toxicities from treating recurrent or oligometastatic gynecologic cancer (ROMGC) patients with definitive radiation therapy. The largest body of literature reported on the use of SBRT in ovarian cancer, which was found to be an effective option, especially in the setting of chemo-resistant disease. Despite the encouraging outcomes using SBRT in oligometastatic gynecologic malignancies, SBRT remains underutilized given the lack of randomized studies studying ROMGC with long term follow-up. While waiting for future prospective trials to establish the role of SBRT as the standard of care in ROMGC patients, this review focuses on reporting the advantages and drawbacks of this technique and examines the current literature to help guide patient centered treatment decisions.

3.
PLoS One ; 18(6): e0287052, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37379309

RESUMEN

Human CD34+ hematopoietic stem and progenitor cells (HSPCs) are a standard source of cells for clinical HSC transplantations as well as experimental xenotransplantation to generate "humanized mice". To further extend the range of applications of these humanized mice, we developed a protocol to efficiently edit the genomes of human CD34+ HSPCs before transplantation. In the past, manipulating HSPCs has been complicated by the fact that they are inherently difficult to transduce with lentivectors, and rapidly lose their stemness and engraftment potential during in vitro culture. However, with optimized nucleofection of sgRNA:Cas9 ribonucleoprotein complexes, we are now able to edit a candidate gene in CD34+ HSPCs with almost 100% efficiency, and transplant these modified cells in immunodeficient mice with high engraftment levels and multilineage hematopoietic differentiation. The result is a humanized mouse from which we knocked out a gene of interest from their human immune system.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Humanos , Ratones , Animales , Antígenos CD34 , Técnicas de Inactivación de Genes , Sistema Inmunológico , Trasplante de Células Madre Hematopoyéticas/métodos , Ratones SCID
4.
J Immunol ; 209(3): 606-620, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35817516

RESUMEN

Despite recent therapeutic progress, advanced melanoma remains lethal for many patients. The composition of the immune tumor microenvironment (TME) has decisive impacts on therapy response and disease outcome, and high-dimensional analyses of patient samples reveal the heterogeneity of the immune TME. Macrophages infiltrate TMEs and generally associate with tumor progression, but the underlying mechanisms are incompletely understood. Because experimental systems are needed to elucidate the functional properties of these cells, we developed a humanized mouse model reconstituted with human immune cells and human melanoma. We used two strains of recipient mice, supporting or not supporting the development of human myeloid cells. We found that human myeloid cells favored metastatic spread of the primary tumor, thereby recapitulating the cancer-supportive role of macrophages. We next analyzed the transcriptome of human immune cells infiltrating tumors versus other tissues. This analysis identified a cluster of myeloid cells present in the TME, but not in other tissues, which do not correspond to canonical M2 cells. The transcriptome of these cells is characterized by high expression of glycolytic enzymes and multiple chemokines and by low expression of gene sets associated with inflammation and adaptive immunity. Compared with humanized mouse results, we found transcriptionally similar myeloid cells in patient-derived samples of melanoma and other cancer types. The humanized mouse model described here thus complements patient sample analyses, enabling further elucidation of fundamental principles in melanoma biology beyond M1/M2 macrophage polarization. The model can also support the development and evaluation of candidate antitumor therapies.


Asunto(s)
Macrófagos , Melanoma , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Activación de Macrófagos , Melanoma/patología , Ratones , Microambiente Tumoral
6.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34257155

RESUMEN

Trastuzumab, a targeted anti-human epidermal-growth-factor receptor-2 (HER2) monoclonal antibody, represents a mainstay in the treatment of HER2-positive (HER2+) breast cancer. Although trastuzumab treatment is highly efficacious for early-stage HER2+ breast cancer, the majority of advanced-stage HER2+ breast cancer patients who initially respond to trastuzumab acquire resistance to treatment and relapse, despite persistence of HER2 gene amplification/overexpression. Here, we sought to leverage HER2 overexpression to engage antibody-dependent cellular phagocytosis (ADCP) through a combination of trastuzumab and anti-CD47 macrophage checkpoint immunotherapy. We have previously shown that blockade of CD47, a surface protein expressed by many malignancies (including HER2+ breast cancer), is an effective anticancer therapy. CD47 functions as a "don't eat me" signal through its interaction with signal regulatory protein-α (SIRPα) on macrophages to inhibit phagocytosis. Hu5F9-G4 (magrolimab), a humanized monoclonal antibody against CD47, blocks CD47's "don't eat me" signal, thereby facilitating macrophage-mediated phagocytosis. Preclinical studies have shown that combining Hu5F9-G4 with tumor-targeting antibodies, such as rituximab, further enhances Hu5F9-G4's anticancer effects via ADCP. Clinical trials have additionally demonstrated that Hu5F9-G4, in combination with rituximab, produced objective responses in patients whose diffuse large B cell lymphomas had developed resistance to rituximab and chemotherapy. These studies led us to hypothesize that combining Hu5F9-G4 with trastuzumab would produce an anticancer effect in antibody-dependent cellular cytotoxicity (ADCC)-tolerant HER2+ breast cancer. This combination significantly suppressed the growth of ADCC-tolerant HER2+ breast cancers via Fc-dependent ADCP. Our study demonstrates that combining trastuzumab and Hu5F9-G4 represents a potential new treatment option for HER2+ breast cancer patients, even for patients whose tumors have progressed after trastuzumab.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Antineoplásicos Inmunológicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Antígeno CD47/inmunología , Trastuzumab/administración & dosificación , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Antígeno CD47/antagonistas & inhibidores , Antígeno CD47/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos , Quimioterapia Combinada , Femenino , Humanos , Inmunoterapia , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología
7.
J Palliat Med ; 24(11): 1689-1696, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34160293

RESUMEN

Background: The COVID-19 pandemic has spurred unprecedented need for specialty palliative care. The Palliative Care Quality Collaborative (PCQC) provides unique infrastructure for rapid data collection and analysis. Objectives: To capture and describe real-time, real-world experiences of specialty palliative care professionals caring for patients with COVID-19 through a rapid reporting tool and registry. Setting: Palliative care clinicians consulted for patients either positive for COVID-19, under investigation for COVID-19, or recovered from COVID-19. Design: The PCQC created a 13-item COVID-19 case report form (CRF), modeled after the PCQC core dataset for specialty palliative care quality measurement. Twelve items offered discrete answer choices and one was open-ended. The CRF was publicized widely (e.g., social media, e-mail list serves) and completed through a link on the PCQC website. Results: Three hundred six reports (298 adult, 8 pediatric) were submitted between April 6, 2020 and October 7, 2020. The majority of patients (83%) were 50 years or older; 25% were 80 or older, and 78% were COVID-19 positive. Male gender identity was significantly more prevalent than female (58% vs. 40%, p < 0.002). The most common comorbidity was cardiovascular disease (23%). Of adult hospital-based patients, 69% were full code before palliative care consultation versus 28% after (p < 0.05). All pediatric patients were full code before and after palliative care consult. Qualitative themes were strained communication with patients, family visitation challenges, communication barriers between clinicians and families, rapid changes in palliative care medical management, community care options difficult to find, lack of testing in community-based settings, and guardianship and legal challenges. Conclusion: Preliminary data from the first 306 patients reported to the PCQC COVID-19 Registry describe palliative care use concentrated among older and higher risk patients and challenges to the provision of palliative care during this pandemic.


Asunto(s)
COVID-19 , Cuidados Paliativos , Adulto , COVID-19/terapia , Niño , Femenino , Humanos , Masculino , Pandemias
8.
Front Immunol ; 12: 643852, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33692812

RESUMEN

Since the late 1980s, mice have been repopulated with human hematopoietic cells to study the fundamental biology of human hematopoiesis and immunity, as well as a broad range of human diseases in vivo. Multiple mouse recipient strains have been developed and protocols optimized to efficiently generate these "humanized" mice. Here, we review three guiding principles that have been applied to the development of the currently available models: (1) establishing tolerance of the mouse host for the human graft; (2) opening hematopoietic niches so that they can be occupied by human cells; and (3) providing necessary support for human hematopoiesis. We then discuss four remaining challenges: (1) human hematopoietic lineages that poorly develop in mice; (2) limited antigen-specific adaptive immunity; (3) absent tolerance of the human immune system for its mouse host; and (4) sub-functional interactions between human immune effectors and target mouse tissues. While major advances are still needed, the current models can already be used to answer specific, clinically-relevant questions and hopefully inform the development of new, life-saving therapies.


Asunto(s)
Inmunidad Adaptativa , Modelos Animales de Enfermedad , Hematopoyesis/inmunología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/inmunología , Animales , Xenoinjertos , Humanos , Ratones
9.
Med Hypotheses ; 147: 110482, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33460993

RESUMEN

The disease presentation of primary hyperparathyroidism (PHPT) is more severe in children. We hypothesize that this difference in disease presentation is a result of several factors including a delay in diagnosis, age-related differences in calcium metabolism and bone turnover, and the influence of the growth hormone (GH)/insulin growth factor-1 (IGF-1) axis. Only the first two explanations for the heightened disease severity of PHPT in children have been previously discussed in the literature. In regards to the potential role GH and IGF-1 may play in this disparity, previous studies have documented decreases in GH and IGF-1 secretion in symptomatic adult PHPT patients potentially influencing the severity of the patients' disease presentation. While these studies have yet to be replicated in the pediatric population, given that both GH and IGF-1 are important for the achievement of peak bone mass in young individuals, it is logical to infer that the GH/IGF-1 axis may be partly responsible for the severe presentation of PHPT in children.


Asunto(s)
Hiperparatiroidismo Primario , Adulto , Densidad Ósea , Remodelación Ósea , Niño , Humanos , Hiperparatiroidismo Primario/complicaciones , Hiperparatiroidismo Primario/diagnóstico , Factor I del Crecimiento Similar a la Insulina
10.
Med Hypotheses ; 144: 109982, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32531542

RESUMEN

Primary hyperparathyroidism (PHPT) is a common endocrine condition which disrupts physiologic calcium regulation. PHPT causes persistent hypercalcemia via the elevated and constant secretion of parathyroid hormone. Due to the effects of parathyroid hormone on target organs such as the bones, kidneys and gastrointestinal tract, untreated PHPT can lead to complications such as decreased bone mineral density, nephrolithiasis, and chronic abdominal pain, respectively. Given PHPT's drastic effect on calcium metabolism, it is likely that the condition also affects other organ systems, such as the inner ear, which rely on calcium for normal structure and function. Specifically, the saccule and utricle have otoconia made of calcium carbonate deposited on a protein framework. We hypothesize that PHPT, the epitome calcium disorder, can cause otoconia degeneration, one manifestation of which is benign paroxysmal positional vertigo (BPPV). As a preliminarily test of this hypothesis, we measured the levels of otolin-1, an inner ear glycoprotein found in the otoconia protein framework and a proposed biomarker for otoconia degeneration in patients with PHPT. We found a positive linear relationship between PTH (parathyroid hormone) and otolin-1 levels (R2 = 0.53) and total calcium and PTH levels (R2 = 0.32). These findings suggest that both PTH and total calcium levels affect otolin-1 levels, implying that the calcium dysregulation caused by PHPT may contribute to the otoconia breakdown and may be associated with inner ear disorders such as BPPV.


Asunto(s)
Hipercalcemia , Hiperparatiroidismo Primario , Vértigo Posicional Paroxístico Benigno , Calcio , Humanos , Hiperparatiroidismo Primario/complicaciones , Membrana Otolítica , Hormona Paratiroidea
11.
Clin Cancer Res ; 25(12): 3561-3571, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30890547

RESUMEN

PURPOSE: Near-infrared photoimmunotherapy (NIR-PIT) is a localized molecular cancer therapy combining a photosensitizer-conjugated mAb and light energy. CD47 is an innate immune checkpoint widely expressed on bladder cancer cells, but absent from luminal normal urothelium. Targeting CD47 for NIR-PIT has the potential to selectively induce cancer cell death and minimize damage to normal urothelium. EXPERIMENTAL DESIGN: The cytotoxic effect of NIR-PIT with anti-CD47-IR700 was investigated in human bladder cancer cell lines and primary human bladder cancer cells derived from fresh surgical samples. Phagocytosis assays were performed to evaluate macrophage activity after NIR-PIT. Anti-CD47-IR700 was administered to murine xenograft tumor models of human bladder cancer for in vivo molecular imaging and NIR-PIT. RESULTS: Cytotoxicity in cell lines and primary bladder cancer cells significantly increased in a light-dose-dependent manner with CD47-targeted NIR-PIT. Phagocytosis of cancer cells significantly increased with NIR-PIT compared with antibody alone (P = 0.0002). In vivo fluorescence intensity of anti-CD47-IR700 in tumors reached a peak 24-hour postinjection and was detectable for at least 14 days. After a single round of CD47-targeted NIR-PIT, treated animals showed significantly slower tumor growth compared with controls (P < 0.0001). Repeated CD47-targeted NIR-PIT treatment further slowed tumor growth (P = 0.0104) and improved survival compared with controls. CONCLUSIONS: CD47-targeted NIR-PIT increased direct cancer cell death and phagocytosis resulting in inhibited tumor growth and improved survival in a murine xenograft model of human bladder cancer.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/farmacología , Antígeno CD47/antagonistas & inhibidores , Inmunoterapia/métodos , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Neoplasias de la Vejiga Urinaria/terapia , Animales , Femenino , Humanos , Masculino , Ratones , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Front Oncol ; 9: 1380, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32038992

RESUMEN

In recent years, immunotherapies have been clinically investigated in AML and other myeloid malignancies. While most of these are focused on stimulating the adaptive immune system (including T cell checkpoint inhibitors), several key approaches targeting the innate immune system have been identified. Macrophages are a key cell type in the innate immune response with CD47 being identified as a dominant macrophage checkpoint. CD47 is a "do not eat me" signal, overexpressed in myeloid malignancies that leads to tumor evasion of phagocytosis by macrophages. Blockade of CD47 leads to engulfment of leukemic cells and therapeutic elimination. Pre-clinical data has demonstrated robust anti-cancer activity in multiple hematologic malignancies including AML and myelodysplastic syndrome (MDS). In addition, clinical studies have been underway with CD47 targeting agents in both AML and MDS as monotherapy and in combination. This review will describe the role of CD47 in myeloid malignancies and pre-clinical data supporting CD47 targeting. In addition, initial clinical data of CD47 targeting in AML/MDS will be reviewed, and including the first-in-class anti-CD47 antibody magrolimab.

13.
Sci Transl Med ; 10(469)2018 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-30487249

RESUMEN

Peritoneal adhesions are fibrous tissues that tether organs to one another or to the peritoneal wall and are a major cause of postsurgical and infectious morbidity. The primary molecular chain of events leading to the initiation of adhesions has been elusive, chiefly due to the lack of an identifiable cell of origin. Using clonal analysis and lineage tracing, we have identified injured surface mesothelium expressing podoplanin (PDPN) and mesothelin (MSLN) as a primary instigator of peritoneal adhesions after surgery in mice. We demonstrate that an anti-MSLN antibody diminished adhesion formation in a mouse model where adhesions were induced by surgical ligation to form ischemic buttons and subsequent surgical abrasion of the peritoneum. RNA sequencing and bioinformatics analyses of mouse mesothelial cells from injured mesothelium revealed aspects of the pathological mechanism of adhesion development and yielded several potential regulators of this process. Specifically, we show that PDPN+MSLN+ mesothelium responded to hypoxia by early up-regulation of hypoxia-inducible factor 1 alpha (HIF1α) that preceded adhesion development. Inhibition of HIF1α with small molecules ameliorated the injury program in damaged mesothelium and was sufficient to diminish adhesion severity in a mouse model. Analyses of human adhesion tissue suggested that similar surface markers and signaling pathways may contribute to surgical adhesions in human patients.


Asunto(s)
Anticuerpos/farmacología , Biomarcadores/metabolismo , Epitelio/patología , Adherencias Tisulares/patología , Animales , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/ultraestructura , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mesotelina , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Peritoneo/efectos de los fármacos , Peritoneo/lesiones , Peritoneo/patología , Adherencias Tisulares/genética , Transcripción Genética
14.
Nat Immunol ; 19(1): 76-84, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29180808

RESUMEN

Exciting progress in the field of cancer immunotherapy has renewed the urgency of the need for basic studies of immunoregulation in both adaptive cell lineages and innate cell lineages. Here we found a central role for major histocompatibility complex (MHC) class I in controlling the phagocytic function of macrophages. Our results demonstrated that expression of the common MHC class I component ß2-microglobulin (ß2M) by cancer cells directly protected them from phagocytosis. We further showed that this protection was mediated by the inhibitory receptor LILRB1, whose expression was upregulated on the surface of macrophages, including tumor-associated macrophages. Disruption of either MHC class I or LILRB1 potentiated phagocytosis of tumor cells both in vitro and in vivo, which defines the MHC class I-LILRB1 signaling axis as an important regulator of the effector function of innate immune cells, a potential biomarker for therapeutic response to agents directed against the signal-regulatory protein CD47 and a potential target of anti-cancer immunotherapy.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Receptor Leucocitario Tipo Inmunoglobulina B1/inmunología , Macrófagos/inmunología , Neoplasias/inmunología , Fagocitosis/inmunología , Animales , Línea Celular Tumoral , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunoterapia/métodos , Receptor Leucocitario Tipo Inmunoglobulina B1/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/metabolismo , Neoplasias/terapia , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/terapia
15.
Proc Natl Acad Sci U S A ; 114(49): E10578-E10585, 2017 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-29158380

RESUMEN

Cancer immunotherapy has emerged as a promising therapeutic intervention. However, complete and durable responses are only seen in a fraction of patients who have cancer. A key factor that limits therapeutic success is the infiltration of tumors by cells of the myeloid lineage. The inhibitory receptor signal regulatory protein-α (SIRPα) is a myeloid-specific immune checkpoint that engages the "don't eat me" signal CD47 expressed on tumors and normal tissues. We therefore developed the monoclonal antibody KWAR23, which binds human SIRPα with high affinity and disrupts its binding to CD47. Administered by itself, KWAR23 is inert, but given in combination with tumor-opsonizing monoclonal antibodies, KWAR23 greatly augments myeloid cell-dependent killing of a collection of hematopoietic and nonhematopoietic human tumor-derived cell lines. Following KWAR23 antibody treatment in a human SIRPA knockin mouse model, both neutrophils and macrophages infiltrate a human Burkitt's lymphoma xenograft and inhibit tumor growth, generating complete responses in the majority of treated animals. We further demonstrate that a bispecific anti-CD70/SIRPα antibody outperforms individually delivered antibodies in specific types of cancers. These studies demonstrate that SIRPα blockade induces potent antitumor activity by targeting multiple myeloid cell subsets that frequently infiltrate tumors. Thus, KWAR23 represents a promising candidate for combination therapy.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/farmacología , Antígenos de Diferenciación/inmunología , Linfoma de Burkitt/terapia , Fagocitosis/efectos de los fármacos , Receptores Inmunológicos/inmunología , Animales , Antígenos de Diferenciación/genética , Linfoma de Burkitt/genética , Linfoma de Burkitt/inmunología , Linfoma de Burkitt/patología , Ligando CD27/genética , Ligando CD27/inmunología , Antígeno CD47/genética , Antígeno CD47/inmunología , Línea Celular Tumoral , Terapia Combinada/métodos , Expresión Génica , Técnicas de Sustitución del Gen , Humanos , Inmunoterapia/métodos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Unión Proteica , Receptores Inmunológicos/genética , Transgenes , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Nat Commun ; 8: 14802, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28378740

RESUMEN

CD47 is a cell surface molecule that inhibits phagocytosis of cells that express it by binding to its receptor, SIRPα, on macrophages and other immune cells. CD47 is expressed at different levels by neoplastic and normal cells. Here, to reveal mechanisms by which different neoplastic cells generate this dominant 'don't eat me' signal, we analyse the CD47 regulatory genomic landscape. We identify two distinct super-enhancers (SEs) associated with CD47 in certain cancer cell types. We show that a set of active constituent enhancers, located within the two CD47 SEs, regulate CD47 expression in different cancer cell types and that disruption of CD47 SEs reduces CD47 gene expression. Finally we report that the TNF-NFKB1 signalling pathway directly regulates CD47 by interacting with a constituent enhancer located within a CD47-associated SE specific to breast cancer. These results suggest that cancers can evolve SE to drive CD47 overexpression to escape immune surveillance.


Asunto(s)
Neoplasias de la Mama/metabolismo , Antígeno CD47/fisiología , Elementos de Facilitación Genéticos , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Transducción de Señal , Regulación hacia Arriba , Animales , Neoplasias de la Mama/patología , Antígeno CD47/genética , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Subunidad p50 de NF-kappa B/metabolismo , Fagocitosis , Unión Proteica , Factor de Necrosis Tumoral alfa/metabolismo
17.
Cancer Immunol Res ; 4(12): 1072-1087, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27856424

RESUMEN

Cancer immunotherapies hold much promise, but their potential in veterinary settings has not yet been fully appreciated. Canine lymphomas are among the most common tumors of dogs and bear remarkable similarity to human disease. In this study, we examined the combination of CD47 blockade with anti-CD20 passive immunotherapy for canine lymphoma. The CD47/SIRPα axis is an immune checkpoint that regulates macrophage activation. In humans, CD47 is expressed on cancer cells and enables evasion from phagocytosis. CD47-blocking therapies are now under investigation in clinical trials for a variety of human cancers. We found the canine CD47/SIRPα axis to be conserved biochemically and functionally. We identified high-affinity SIRPα variants that antagonize canine CD47 and stimulate phagocytosis of canine cancer cells in vitro When tested as Fc fusion proteins, these therapeutic agents exhibited single-agent efficacy in a mouse xenograft model of canine lymphoma. As robust synergy between CD47 blockade and tumor-specific antibodies has been demonstrated for human cancer, we evaluated the combination of CD47 blockade with 1E4-cIgGB, a canine-specific antibody to CD20. 1E4-cIgGB could elicit a therapeutic response against canine lymphoma in vivo as a single agent. However, augmented responses were observed when combined with CD47-blocking therapies, resulting in synergy in vitro and in vivo and eliciting cures in 100% of mice bearing canine lymphoma. Our findings support further testing of CD47-blocking therapies alone and in combination with CD20 antibodies in the veterinary setting. Cancer Immunol Res; 4(12); 1072-87. ©2016 AACR.


Asunto(s)
Antígenos CD20/inmunología , Antígeno CD47/inmunología , Inmunoterapia , Linfoma de Células B Grandes Difuso/terapia , Animales , Línea Celular Tumoral , Perros , Femenino , Inmunoglobulina G/uso terapéutico , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/veterinaria , Macrófagos/inmunología , Masculino , Ratones , Fagocitosis , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nature ; 536(7614): 86-90, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27437576

RESUMEN

Atherosclerosis is the disease process that underlies heart attack and stroke. Advanced lesions at risk of rupture are characterized by the pathological accumulation of diseased vascular cells and apoptotic cellular debris. Why these cells are not cleared remains unknown. Here we show that atherogenesis is associated with upregulation of CD47, a key anti-phagocytic molecule that is known to render malignant cells resistant to programmed cell removal, or 'efferocytosis'. We find that administration of CD47-blocking antibodies reverses this defect in efferocytosis, normalizes the clearance of diseased vascular tissue, and ameliorates atherosclerosis in multiple mouse models. Mechanistic studies implicate the pro-atherosclerotic factor TNF-α as a fundamental driver of impaired programmed cell removal, explaining why this process is compromised in vascular disease. Similar to recent observations in cancer, impaired efferocytosis appears to play a pathogenic role in cardiovascular disease, but is not a fixed defect and may represent a novel therapeutic target.


Asunto(s)
Anticuerpos Bloqueadores/inmunología , Anticuerpos Bloqueadores/farmacología , Aterosclerosis/prevención & control , Antígeno CD47/inmunología , Fagocitosis/efectos de los fármacos , Animales , Apoptosis , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/terapia , Antígeno CD47/biosíntesis , Antígeno CD47/metabolismo , Arterias Carótidas/patología , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , FN-kappa B/metabolismo , Biosíntesis de Proteínas , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
19.
Proc Natl Acad Sci U S A ; 112(7): 2145-50, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25646432

RESUMEN

Macrophage-mediated programmed cell removal (PrCR) is an important mechanism of eliminating diseased and damaged cells before programmed cell death. The induction of PrCR by eat-me signals on tumor cells is countered by don't-eat-me signals such as CD47, which binds macrophage signal-regulatory protein α to inhibit phagocytosis. Blockade of CD47 on tumor cells leads to phagocytosis by macrophages. Here we demonstrate that the activation of Toll-like receptor (TLR) signaling pathways in macrophages synergizes with blocking CD47 on tumor cells to enhance PrCR. Bruton's tyrosine kinase (Btk) mediates TLR signaling in macrophages. Calreticulin, previously shown to be an eat-me signal on cancer cells, is activated in macrophages for secretion and cell-surface exposure by TLR and Btk to target cancer cells for phagocytosis, even if the cancer cells themselves do not express calreticulin.


Asunto(s)
Calreticulina/fisiología , Macrófagos/inmunología , Neoplasias/patología , Proteínas Tirosina Quinasas/metabolismo , Receptores Toll-Like/fisiología , Agammaglobulinemia Tirosina Quinasa , Humanos , Neoplasias/enzimología , Neoplasias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...