Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Virol ; 96(7): e0005722, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35319225

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused over 5 million deaths worldwide. Pneumonia and systemic inflammation contribute to its high mortality. Many viruses use heparan sulfate proteoglycans as coreceptors for viral entry, and heparanase (HPSE) is a known regulator of both viral entry and inflammatory cytokines. We evaluated the heparanase inhibitor Roneparstat, a modified heparin with minimum anticoagulant activity, in pathophysiology and therapy for COVID-19. We found that Roneparstat significantly decreased the infectivity of SARS-CoV-2, SARS-CoV-1, and retroviruses (human T-lymphotropic virus 1 [HTLV-1] and HIV-1) in vitro. Single-cell RNA sequencing (scRNA-seq) analysis of cells from the bronchoalveolar lavage fluid of COVID-19 patients revealed a marked increase in HPSE gene expression in CD68+ macrophages compared to healthy controls. Elevated levels of HPSE expression in macrophages correlated with the severity of COVID-19 and the expression of inflammatory cytokine genes, including IL6, TNF, IL1B, and CCL2. In line with this finding, we found a marked induction of HPSE and numerous inflammatory cytokines in human macrophages challenged with SARS-CoV-2 S1 protein. Treatment with Roneparstat significantly attenuated SARS-CoV-2 S1 protein-mediated inflammatory cytokine release from human macrophages, through disruption of NF-κB signaling. HPSE knockdown in a macrophage cell line also showed diminished inflammatory cytokine production during S1 protein challenge. Taken together, this study provides a proof of concept that heparanase is a target for SARS-CoV-2-mediated pathogenesis and that Roneparstat may serve as a dual-targeted therapy to reduce viral infection and inflammation in COVID-19. IMPORTANCE The complex pathogenesis of COVID-19 consists of two major pathological phases: an initial infection phase elicited by SARS-CoV-2 entry and replication and an inflammation phase that could lead to tissue damage, which can evolve into acute respiratory failure or even death. While the development and deployment of vaccines are ongoing, effective therapy for COVID-19 is still urgently needed. In this study, we explored HPSE blockade with Roneparstat, a phase I clinically tested HPSE inhibitor, in the context of COVID-19 pathogenesis. Treatment with Roneparstat showed wide-spectrum anti-infection activities against SARS-CoV-2, HTLV-1, and HIV-1 in vitro. In addition, HPSE blockade with Roneparstat significantly attenuated SARS-CoV-2 S1 protein-induced inflammatory cytokine release from human macrophages through disruption of NF-κB signaling. Together, this study provides a proof of principle for the use of Roneparstat as a dual-targeting therapy for COVID-19 to decrease viral infection and dampen the proinflammatory immune response mediated by macrophages.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Heparina/análogos & derivados , Línea Celular , Citocinas/metabolismo , Fenofibrato , Técnicas de Silenciamiento del Gen , Glucuronidasa/genética , Glucuronidasa/metabolismo , Heparina/uso terapéutico , Humanos , Inmunidad/efectos de los fármacos , Inflamación , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , FN-kappa B , SARS-CoV-2
2.
Breast Cancer Res ; 23(1): 63, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34088357

RESUMEN

BACKGROUND: Breast cancer mortality is principally due to tumor recurrence, which can occur following extended periods of clinical remission that may last decades. While clinical latency has been postulated to reflect the ability of residual tumor cells to persist in a dormant state, this hypothesis remains unproven since little is known about the biology of these cells. Consequently, defining the properties of residual tumor cells is an essential goal with important clinical implications for preventing recurrence and improving cancer outcomes. METHODS: To identify conserved features of residual tumor cells, we modeled minimal residual disease using inducible transgenic mouse models for HER2/neu and Wnt1-driven tumorigenesis that recapitulate cardinal features of human breast cancer progression, as well as human breast cancer cell xenografts subjected to targeted therapy. Fluorescence-activated cell sorting was used to isolate tumor cells from primary tumors, residual lesions following oncogene blockade, and recurrent tumors to analyze gene expression signatures and evaluate tumor-initiating cell properties. RESULTS: We demonstrate that residual tumor cells surviving oncogenic pathway inhibition at both local and distant sites exist in a state of cellular dormancy, despite adequate vascularization and the absence of adaptive immunity, and retain the ability to re-enter the cell cycle and give rise to recurrent tumors after extended latency periods. Compared to primary or recurrent tumor cells, dormant residual tumor cells possess unique features that are conserved across mouse models for human breast cancer driven by different oncogenes, and express a gene signature that is strongly associated with recurrence-free survival in breast cancer patients and similar to that of tumor cells in which dormancy is induced by the microenvironment. Although residual tumor cells in both the HER2/neu and Wnt1 models are enriched for phenotypic features associated with tumor-initiating cells, limiting dilution experiments revealed that residual tumor cells are not enriched for cells capable of giving rise to primary tumors, but are enriched for cells capable of giving rise to recurrent tumors, suggesting that tumor-initiating populations underlying primary tumorigenesis may be distinct from those that give rise to recurrence following therapy. CONCLUSIONS: Residual cancer cells surviving targeted therapy reside in a well-vascularized, desmoplastic microenvironment at both local and distant sites. These cells exist in a state of cellular dormancy that bears little resemblance to primary or recurrent tumor cells, but shares similarities with cells in which dormancy is induced by microenvironmental cues. Our observations suggest that dormancy may be a conserved response to targeted therapy independent of the oncogenic pathway inhibited or properties of the primary tumor, that the mechanisms underlying dormancy at local and distant sites may be related, and that the dormant state represents a potential therapeutic target for preventing cancer recurrence.


Asunto(s)
Terapia Molecular Dirigida , Neoplasia Residual/patología , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Supervivencia sin Enfermedad , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Ratones , Ratones Transgénicos , Terapia Molecular Dirigida/efectos adversos , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Neoplasia Residual/irrigación sanguínea , Neoplasia Residual/etiología , Neoplasia Residual/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neovascularización Patológica/patología , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Proteína Wnt1/antagonistas & inhibidores , Proteína Wnt1/genética
3.
Nanomedicine ; 13(4): 1353-1362, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28115246

RESUMEN

Nanoparticle drug carriers hold potential to improve current cancer therapy by delivering payload to the tumor environment and decreasing toxic side effects. Challenges in nanotechnology drug delivery include plasma instability, site-specific delivery, and relevant biomarkers. We have developed a triblock polymer comprising a hydroxamic acid functionalized center block that chelates iron to form a stabilized micelle that physically entraps chemotherapeutic drugs in the hydrophobic core. The iron-imparted stability significantly improves the integrity of the micelle and extends circulation pharmacokinetics in plasma over that of free drug. Furthermore, the paramagnetic properties of the iron-crosslinking exhibits contrast in the tumors for imaging by magnetic resonance. Three separate nanoparticle formulations demonstrate improved anti-tumor efficacy in xenograft models and decreased toxicity. We report a stabilized polymer micelle that improves the tolerability and efficacy of chemotherapeutic drugs, and holds potential for non-invasive MRI to image drug delivery and deposition in the tumor.


Asunto(s)
Antineoplásicos/farmacocinética , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias Colorrectales/tratamiento farmacológico , Portadores de Fármacos/química , Hierro/química , Micelas , Animales , Línea Celular Tumoral , Portadores de Fármacos/farmacocinética , Femenino , Humanos , Imagen por Resonancia Magnética , Ratones Desnudos , Polímeros/química , Ratas Sprague-Dawley , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Adv Ther ; 41(2): 492-508, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38104037

RESUMEN

Following the clinical perspective and concept that a healthy body will not develop chronic wounds, the central approach for the treatment described here is based on an understanding of how the body transforms the wound microenvironment from a non-healing to a healing state. As part of a comprehensive treatment regimen that includes OCM™ (complete matrix), wound preparation, and skin protectant formulations, the OCM contains components for complete wound healing by attending to the individual needs required to promote the closure of each unique chronic wound. During application of the comprehensive treatment regimen in independent investigator-led trials, the total wound percentage average reduction over the first 4 weeks of treatment was 60% across multiple wound types; median time to total wound closure was 6.9 weeks. Safety testing of the OCM formulation shows no potential allergenicity, no potential sensitization, and no known product-related adverse events. Clinical trials evaluating the OCM formulation as part of the comprehensive treatment regimen of multiple wound types are underway. Results of clinical trials and real-world experiences will expand current knowledge of the wound-healing potential of this novel product.


Asunto(s)
Traumatismo Múltiple , Cicatrización de Heridas , Humanos
5.
Mol Cancer Ther ; 20(6): 1183-1198, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33785647

RESUMEN

Breast cancer bone metastases are common and incurable. Tumoral integrin ß3 (ß3) expression is induced through interaction with the bone microenvironment. Although ß3 is known to promote bone colonization, its functional role during therapy of established bone metastases is not known. We found increased numbers of ß3+ tumor cells in murine bone metastases after docetaxel chemotherapy. ß3+ tumor cells were present in 97% of post-neoadjuvant chemotherapy triple-negative breast cancer patient samples (n = 38). High tumoral ß3 expression was associated with worse outcomes in both pre- and postchemotherapy triple-negative breast cancer groups. Genetic deletion of tumoral ß3 had minimal effect in vitro, but significantly enhanced in vivo docetaxel activity, particularly in the bone. Rescue experiments confirmed that this effect required intact ß3 signaling. Ultrastructural, transcriptomic, and functional analyses revealed an alternative metabolic response to chemotherapy in ß3-expressing cells characterized by enhanced oxygen consumption, reactive oxygen species generation, and protein production. We identified mTORC1 as a candidate for therapeutic targeting of this ß3-mediated, chemotherapy-induced metabolic response. mTORC1 inhibition in combination with docetaxel synergistically attenuated murine bone metastases. Furthermore, micelle nanoparticle delivery of mTORC1 inhibitor to cells expressing activated αvß3 integrins enhanced docetaxel efficacy in bone metastases. Taken together, we show that ß3 integrin induction by the bone microenvironment promotes resistance to chemotherapy through an altered metabolic response that can be defused by combination with αvß3-targeted mTORC1 inhibitor nanotherapy. Our work demonstrates the importance of the metastatic microenvironment when designing treatments and presents new, bone-specific strategies for enhancing chemotherapeutic efficacy.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Neoplasias de la Mama/tratamiento farmacológico , Integrina beta3/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Docetaxel/farmacología , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Análisis de Supervivencia
6.
J Clin Invest ; 131(20)2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34520398

RESUMEN

Tumor-infiltrating myeloid cells contribute to the development of the immunosuppressive tumor microenvironment. Myeloid cell expression of arginase 1 (ARG1) promotes a protumor phenotype by inhibiting T cell function and depleting extracellular l-arginine, but the mechanism underlying this expression, especially in breast cancer, is poorly understood. In breast cancer clinical samples and in our mouse models, we identified tumor-derived GM-CSF as the primary regulator of myeloid cell ARG1 expression and local immune suppression through a gene-KO screen of breast tumor cell-produced factors. The induction of myeloid cell ARG1 required GM-CSF and a low pH environment. GM-CSF signaling through STAT3 and p38 MAPK and acid signaling through cAMP were required to activate myeloid cell ARG1 expression in a STAT6-independent manner. Importantly, breast tumor cell-derived GM-CSF promoted tumor progression by inhibiting host antitumor immunity, driving a significant accumulation of ARG1-expressing myeloid cells compared with lung and melanoma tumors with minimal GM-CSF expression. Blockade of tumoral GM-CSF enhanced the efficacy of tumor-specific adoptive T cell therapy and immune checkpoint blockade. Taken together, we show that breast tumor cell-derived GM-CSF contributes to the development of the immunosuppressive breast cancer microenvironment by regulating myeloid cell ARG1 expression and can be targeted to enhance breast cancer immunotherapy.


Asunto(s)
Arginasa/fisiología , Neoplasias de la Mama/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Tolerancia Inmunológica , Células Mieloides/enzimología , Microambiente Tumoral , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , AMP Cíclico/fisiología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL
7.
Oncotarget ; 9(51): 29698-29714, 2018 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-30038714

RESUMEN

In many cancers, combination therapy regimens are successfully improving response and survival rates, but the challenges of toxicity remain. GRP78, the master regulator of the unfolded protein response, is emerging as a target that is upregulated in tumors, specifically following treatment, and one that impacts tumor cell survival and disease recurrence. Here, we show IT-139, an antitumor small molecule inhibitor, suppresses induction of GRP78 from different types of endoplasmic reticulum (ER) stress in a variety of cancer cell lines, including those that have acquired therapeutic resistance, but not in the non-cancer cells being tested. We further determined that IT-139 treatment exacerbates ER stress while at the same time suppresses GRP78 induction at the transcriptional level. Our studies revealed a differential effect of IT-139 on chaperone protein family expression at multiple levels in different cancer cell lines. In xenograft studies, IT-139 decreased BRAF inhibitor upregulation of GRP78 expression in the tumor, while having minimal effect on GRP78 expression in the adjacent normal cells. The preferential decrease in GRP78 levels in tumor cells over normal cells, supported by the manageable safety profile seen in the Phase 1 clinical trial, reinforce the value IT-139 brings to combination therapies as it continues its clinical development.

8.
Rev Endocr Metab Disord ; 8(3): 199-213, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17657606

RESUMEN

Breast cancer begins as a localized disease, but has the potential to spread to distant sites within the body. This process--known as metastasis--is the leading cause of death from breast cancer. Whether the ability of cancer cells to metastasize is an intrinsic or acquired feature is currently a topic of considerable debate. Nevertheless, the key cellular events required for metastasis are generally accepted. These include invasion of the surrounding stromal tissue, intravasation, evasion of programmed cell death, arrest within the vasculature at a distant site, extravasation, and establishment and growth within a new microenvironment. The development of mouse models that faithfully mimic critical aspects of human neoplasia has been instrumental in framing our current understanding of multistage carcinogenesis. This review examines the advantages and limitations of existing murine models for mammary carcinogenesis for probing the molecular mechanisms that contribute to metastasis, as well as non-invasive tumor imaging approaches to facilitate these investigations.


Asunto(s)
Neoplasias de la Mama/genética , Metástasis de la Neoplasia/genética , Animales , Neoplasias de la Mama/patología , Adhesión Celular/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Ratones , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología
9.
Proc Natl Acad Sci U S A ; 100(24): 14205-10, 2003 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-14612570

RESUMEN

Mice with a targeted deletion of beta3 integrin were used to examine the process by which tumor cells metastasize and destroy bone. Injection of B16 melanoma cells into the left cardiac ventricle resulted in osteolytic bone metastasis in 74% of beta3+/+ mice by 14 days. In contrast, only 4% of beta3-/- mice developed bone lesions. Direct intratibial inoculation of tumor resulted in marrow replacement by tumor in beta3-/- mice, but no associated trabecular bone resorption as seen inbeta3+/+ mice. Bone marrow transplantation studies showed that susceptibility to bone metastasis was conferred by a bone marrow-derived cell. To dissect the roles of osteoclast and platelet beta3 integrins in this model of bone metastasis, osteoclast-defective src-/- mice were used. Src-null mice were protected from tumor-associated bone destruction but were not protected from tumor cell metastasis to bone. In contrast, a highly specific platelet aggregation inhibitor of activated alphaIIbbeta3 prevented B16 metastases. These data demonstrate a critical role for platelet alphaIIbbeta3 in tumor entry into bone and suggest a mechanism by which antiplatelet therapy may be beneficial in preventing the metastasis of solid tumors.


Asunto(s)
Plaquetas/inmunología , Neoplasias Óseas/inmunología , Neoplasias Óseas/secundario , Integrina beta3/fisiología , Melanoma Experimental/inmunología , Melanoma Experimental/secundario , Osteoclastos/inmunología , Animales , Trasplante de Médula Ósea , Neoplasias Óseas/patología , Genes src , Integrina beta3/genética , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoclastos/patología , Osteólisis/inmunología , Osteólisis/patología , Glicoproteína IIb de Membrana Plaquetaria/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA