Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Surg Endosc ; 38(9): 5160-5168, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39039297

RESUMO

BACKGROUND: Space travel is experiencing a renaissance with expanding commercial and international efforts. Space surgery will have growing relevance as mission frequency and distances increase beyond low Earth orbit. METHODS: This white paper from the SAGES Space Surgery Task Force raises awareness among the SAGES membership regarding the challenges and opportunities surrounding this emerging field that anticipates surgical care in the most extreme, austere environments. RESULTS: Innovation in technology and preventive medicine principles will enhance the effectiveness of space surgical care when the need arises. The impact of advancements in space and terrestrial medicine to support space exploration indicates the need for a surgeon to oversee medical/surgical invasive treatment to ensure astronaut health and mission success. Advanced technology, including semi- and autonomous robotic systems, may be a preferred way to deliver this care in the foreseeable future. There is currently a need to develop training curricula and flight-compatible supplies and technology for physicians that deliver surgical care to this special patient population. The protocols and technology developed to address the unique challenges of space travel will provide value for care in space as well as in extreme, austere terrestrial environments on Earth. CONCLUSION: Space surgery will continue to evolve as commercial and government programs explore further into space. The SAGES Space Surgery Task Force is favorably positioned to significantly contribute to addressing some capability gaps in delivering surgical care in space.


Assuntos
Voo Espacial , Humanos , Medicina Aeroespacial , Procedimentos Cirúrgicos Robóticos/educação
2.
Surg Innov ; 28(5): 573-581, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33339485

RESUMO

Introduction. Prophylactic surgery before spaceflight may eliminate the risk of appendicitis and cholecystitis in astronauts on deep space missions. However, even minimally invasive surgery increases the risk of small bowel obstruction (SBO). Probabilistic risk assessment (PRA) is a method that can be used to estimate the benefits and risks of prophylactic surgery. Methods. Risks of appendicitis and cholecystitis during a 2.5-year Mars mission are compared to the risk of SBO after laparoscopic removal of the appendix, gallbladder, or both. A PRA model using Monte Carlo methodology was used to forecast the risks. Results. Prophylactic appendectomy and cholecystectomy combined, conferred an increased probability of medical evacuation (pEVAC) due to SBO as compared to the no surgery group. A slightly higher probability for the loss of crew life (pLOCL) was found in the no surgery group when compared to the cases in which either prophylactic appendectomy alone, or appendectomy plus cholecystectomy are performed. Discussion. The need for medical evacuation can be viewed as a potential risk for death in the context of a space mission where evacuation is not possible. Because of the higher pEVAC due to SBO and relatively small benefit in the reduction of pLOCL in the prophylactic surgery groups, this analysis does not support the prophylactic removal of appendix and/or gallbladder for spaceflight. Future advances in surgical or medical technique or mission medical capabilities may change these results. This work demonstrates the utility of PRA in providing quantitative answers to "what if" questions where limited information is available.


Assuntos
Apendicite , Voo Espacial , Apendicectomia/efeitos adversos , Astronautas , Humanos , Medição de Risco
3.
Cancer Immunol Immunother ; 69(6): 1015-1027, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32088771

RESUMO

Oncolytic virus (OV) therapy is an emerging approach with the potential to redefine treatment options across a range of cancer indications and in patients who remain resistant to existing standards of care, including immuno-oncology (IO) drugs. MEDI5395, a recombinant Newcastle disease virus (NDV), engineered to express granulocyte-macrophage colony-stimulating factor (GM-CSF), exhibits potent oncolytic activity. It was hypothesized that activation of immune cells by MEDI5395, coupled with its oncolytic activity, would enhance the priming of antitumor immunity. Using MEDI5395 and recombinant NDVs encoding fluorescent reporter genes, we demonstrated preferential virus uptake and non-productive infection in myeloid cells, including monocytes, macrophages, and dendritic cells (DCs). Infection resulted in immune-cell activation, with upregulation of cell surface activation markers (e.g., CD80, PD-L1, HLA-DR) and secretion of proinflammatory cytokines (IFN-α2a, IL-6, IL-8, TNF-α). Interestingly, in vitro M2-polarized macrophages were more permissive to virus infection than were M1-polarized macrophages. In a co-culture system, infected myeloid cells were effective virus vectors and mediated the transfer of infectious NDV particles to tumor cells, resulting in cell death. Furthermore, NDV-infected DCs stimulated greater proliferation of allogeneic T cells than uninfected DCs. Antigens released after NDV-induced tumor cell lysis were cross-presented by DCs and drove activation of tumor antigen-specific autologous T cells. MEDI5395 therefore exhibited potent immunostimulatory activity and an ability to enhance antigen-specific T-cell priming. This, coupled with its tumor-selective oncolytic capacity, underscores the promise of MEDI5395 as a multimodal therapeutic, with potential to both enhance current responding patient populations and elicit de novo responses in resistant patients.


Assuntos
Vírus da Doença de Newcastle/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Linhagem Celular Tumoral , Vetores Genéticos , Humanos , Imunidade Inata
4.
J Virol ; 91(16)2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28592535

RESUMO

Newcastle disease virus (NDV) is an oncolytic virus being developed for the treatment of cancer. Following infection of a human ovarian cancer cell line (OVCAR3) with a recombinant low-pathogenic NDV, persistent infection was established in a subset of tumor cells. Persistently infected (PI) cells exhibited resistance to superinfection with NDV and established an antiviral state, as demonstrated by upregulation of interferon and interferon-induced genes such as myxoma resistance gene 1 (Mx1) and retinoic acid-inducing gene-I (RIG-I). Viruses released from PI cells induced higher cell-to-cell fusion than the parental virus following infection in two tumor cell lines tested, HT1080 and HeLa, and remained attenuated in chickens. Two mutations, one in the fusion (F) protein cleavage site, F117S (F117S), and another in hemagglutinin-neuraminidase (HN), G169R (HN169R), located in the second sialic acid binding region, were responsible for the hyperfusogenic phenotype. F117S improves F protein cleavage efficiency, facilitating cell-to-cell fusion, while HN169R possesses a multifaceted role in contributing to higher fusion, reduced receptor binding, and lower neuraminidase activity, which together result in increased fusion and reduced viral replication. Thus, establishment of persistent infection in vitro involves viral genetic changes that facilitate efficient viral spread from cell to cell as a potential mechanism to escape host antiviral responses. The results of our study also demonstrate a critical role in the viral life cycle for the second receptor binding region of the HN protein, which is conserved in several paramyxoviruses.IMPORTANCE Oncolytic Newcastle disease virus (NDV) could establish persistent infection in a tumor cell line, resulting in a steady antiviral state reflected by constitutively expressed interferon. Viruses isolated from persistently infected cells are highly fusogenic, and this phenotype has been mapped to two mutations, one each in the fusion (F) and hemagglutinin-neuraminidase (HN) proteins. The F117S mutation in the F protein cleavage site improved F protein cleavage efficiency while the HN169R mutation located at the second receptor binding site of the HN protein contributed to a complex phenotype consisting of a modest increase in fusion and cell killing, lower neuraminidase activity, and reduced viral growth. This study highlights the intricate nature of these two mutations in the glycoproteins of NDV in the establishment of persistent infection. The data also shed light on the critical balance between the F and HN proteins required for efficient NDV infection and their role in avian pathogenicity.


Assuntos
Proteína HN/metabolismo , Vírus da Doença de Newcastle/crescimento & desenvolvimento , Proteínas Virais de Fusão/metabolismo , Animais , Sítios de Ligação , Fusão Celular , Linhagem Celular Tumoral , Galinhas , Proteína HN/genética , Humanos , Mutação de Sentido Incorreto , Ácido N-Acetilneuramínico/metabolismo , Ligação Proteica , Proteínas Virais de Fusão/genética
5.
J Virol ; 90(11): 5343-5352, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27009956

RESUMO

UNLABELLED: Clinical development of a mesogenic strain of Newcastle disease virus (NDV) as an oncolytic agent for cancer therapy has been hampered by its select agent status due to its pathogenicity in avian species. Using reverse genetics, we have generated a lead candidate oncolytic NDV based on the mesogenic NDV-73T strain that is no longer classified as a select agent for clinical development. This recombinant NDV has a modification at the fusion protein (F) cleavage site to reduce the efficiency of F protein cleavage and an insertion of a 198-nucleotide sequence into the HN-L intergenic region, resulting in reduced viral gene expression and replication in avian cells but not in mammalian cells. In mammalian cells, except for viral polymerase (L) gene expression, viral gene expression is not negatively impacted or increased by the HN-L intergenic insertion. Furthermore, the virus can be engineered to express a foreign gene while still retaining the ability to grow to high titers in cell culture. The recombinant NDV selectively replicates in and kills tumor cells and is able to drive potent tumor growth inhibition following intratumoral or intravenous administration in a mouse tumor model. The candidate is well positioned for clinical development as an oncolytic virus. IMPORTANCE: Avian paramyxovirus type 1, NDV, has been an attractive oncolytic agent for cancer virotherapy. However, this virus can cause epidemic disease in poultry, and concerns about the potential environmental and economic impact of an NDV outbreak have precluded its clinical development. Here we describe generation and characterization of a highly potent oncolytic NDV variant that is unlikely to cause Newcastle disease in its avian host, representing an essential step toward moving NDV forward as an oncolytic agent. Several attenuation mechanisms have been genetically engineered into the recombinant NDV that reduce chicken pathogenicity to a level that is acceptable worldwide without impacting viral production in cell culture. The selective tumor replication of this recombinant NDV, both in vitro and in vivo, along with efficient tumor cell killing makes it an attractive oncolytic virus candidate that may provide clinical benefit to patients.


Assuntos
Neoplasias Experimentais/terapia , Neoplasias Experimentais/virologia , Neoplasias/terapia , Vírus da Doença de Newcastle/genética , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Animais , DNA Intergênico/genética , Expressão Gênica , Terapia Genética , Humanos , Injeções Intravenosas , Camundongos , Vírus da Doença de Newcastle/crescimento & desenvolvimento , Vírus Oncolíticos/crescimento & desenvolvimento , Recombinação Genética , Genética Reversa/métodos , Replicação Viral/genética
6.
Sci Rep ; 14(1): 12129, 2024 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-38802399

RESUMO

Many targeted cancer therapies rely on biomarkers assessed by scoring of immunohistochemically (IHC)-stained tissue, which is subjective, semiquantitative, and does not account for expression heterogeneity. We describe an image analysis-based method for quantitative continuous scoring (QCS) of digital whole-slide images acquired from baseline human epidermal growth factor receptor 2 (HER2) IHC-stained breast cancer tissue. Candidate signatures for patient stratification using QCS of HER2 expression on subcellular compartments were identified, addressing the spatial distribution of tumor cells and tumor-infiltrating lymphocytes. Using data from trastuzumab deruxtecan-treated patients with HER2-positive and HER2-negative breast cancer from a phase 1 study (NCT02564900; DS8201-A-J101; N = 151), QCS-based patient stratification showed longer progression-free survival (14.8 vs 8.6 months) with higher prevalence of patient selection (76.4 vs 56.9%) and a better cross-validated log-rank p value (0.026 vs 0.26) than manual scoring based on the American Society of Clinical Oncology / College of American Pathologists guidelines. QCS-based features enriched the HER2-negative subgroup by correctly predicting 20 of 26 responders.


Assuntos
Neoplasias da Mama , Seleção de Pacientes , Receptor ErbB-2 , Trastuzumab , Humanos , Feminino , Receptor ErbB-2/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Trastuzumab/uso terapêutico , Pessoa de Meia-Idade , Biomarcadores Tumorais/metabolismo , Adulto , Imunoconjugados/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Idoso , Imuno-Histoquímica , Camptotecina/análogos & derivados
7.
Nat Cell Biol ; 8(6): 551-61, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16715076

RESUMO

p63 is critical for epithelial development yet little is known about the transcriptional programmes it regulates. By characterising transcriptional changes and cellular effects following modulation of p63 expression, we have defined a vital role for p63 in cellular adhesion. Knockdown of p63 expression caused downregulation of cell adhesion-associated genes, cell detachment and anoikis in mammary epithelial cells and keratinocytes. Conversely, overexpression of the TAp63gamma or deltaNp63alpha isoforms of p63 upregulated cell adhesion molecules, increased cellular adhesion and conferred resistance to anoikis. Apoptosis induced by loss of p63 was rescued by signalling downstream of beta4 integrin. Our results implicate p63 as a key regulator of cellular adhesion and survival in basal cells of the mammary gland and other stratified epithelial tissues.


Assuntos
Células Epiteliais/citologia , Proteínas de Membrana/fisiologia , Anoikis , Adesão Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Regulação da Expressão Gênica , Humanos , Queratinócitos/citologia , Proteínas de Membrana/genética , Isoformas de Proteínas
8.
Proc Natl Acad Sci U S A ; 107(11): 5012-7, 2010 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-20194747

RESUMO

Aberrant activation of Notch receptors has been implicated in breast cancer; however, the mechanisms contributing to Notch-dependent transformation remain elusive because Notch displays dichotomous functional activities, promoting both proliferation and growth arrest. We investigated the cellular basis for the heterogeneous responses to Notch pathway activation in 3D cultures of MCF-10A mammary epithelial cells. Expression of a constitutively active Notch-1 intracellular domain (NICD) was found to induce two distinct types of 3D structures: large, hyperproliferative structures and small, growth-arrested structures with reduced cell-to-matrix adhesion. Interestingly, we found that these heterogeneous phenotypes reflect differences in Notch pathway activation levels; high Notch activity caused down-regulation of multiple matrix-adhesion genes and inhibition of proliferation, whereas low Notch activity maintained matrix adhesion and provoked a strong hyperproliferative response. Moreover, microarray analyses implicated NICD-induced p63 down-regulation in loss of matrix adhesion. In addition, a reverse-phase protein array-based analysis and subsequent loss-of-function studies identified STAT3 as a dominant downstream mediator of the NICD-induced outgrowth. These results indicate that the phenotypic responses to Notch are determined by the dose of pathway activation; and this dose affects the balance between growth-stimulative and growth-suppressive effects. This unique feature of Notch signaling provides insights into mechanisms that contribute to the dichotomous effects of Notch during development and tumorigenesis.


Assuntos
Células Epiteliais/metabolismo , Glândulas Mamárias Humanas/citologia , Receptor Notch1/metabolismo , Transdução de Sinais , Adesão Celular , Proliferação de Células , Células Cultivadas , Células Epiteliais/citologia , Matriz Extracelular/metabolismo , Feminino , Humanos , Fenótipo , Estrutura Terciária de Proteína , Receptor Notch1/química , Fator de Transcrição STAT3/metabolismo , Transativadores/metabolismo , Fatores de Transcrição , Proteínas Supressoras de Tumor/metabolismo
9.
NPJ Microgravity ; 8(1): 55, 2022 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-36460679

RESUMO

Lunar dust (LD), the component of lunar regolith with particle sizes less than 20 µm, covers the surface of the Moon. Due to its fineness, jagged edges, and electrostatic charge, LD adheres to and coats almost any surface it contacts. As a result, LD poses known risks to the proper functioning of electronic and mechanical equipment on the lunar surface. However, its mechanical irritancy and chemical reactivity may also pose serious health risks to humans by a number of mechanisms. While Apollo astronauts reported mild short-lived respiratory symptoms, the spectrum of health effects associated with high-dose acute exposure or chronic low-dose exposure are not yet well-understood. This paper explores known and potential human risks of exposure to LD which are thought to be important in planning upcoming lunar missions and planetary surface work.

10.
Sci Rep ; 12(1): 2699, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35177674

RESUMO

The development of covalent inhibitors against KRAS G12C represents a major milestone in treatment of RAS-driven cancers, especially in non-small cell lung cancer (NSCLC), where KRAS G12C is one of the most common oncogenic driver. Here we investigated if additional KRAS mutations co-occur with KRAS G12C (c.34G>T) in NSCLC tumours and if such mutation co-occurrence affects cellular response to G12C-specific inhibitors. Analysis of a large cohort of NSCLC patients whose tumours harboured KRAS mutations revealed co-occurring KRAS mutations in up to 8% of tumours with the KRAS c.34G>T mutation. KRAS c.35G>T was the most frequently co-occurring mutation, and could occur on the same allele (in cis) translating to a single mutant KRAS G12F protein, or on the other allele (in trans), translating to separate G12C and G12V mutant proteins. Introducing KRAS c.35G>T in trans in the KRAS G12C lung cancer model NCI-H358, as well as the co-occurrence in cis in the KRAS G12F lung cancer model NCI-H2291 led to cellular resistance to the G12C-specific inhibitor AZ'8037 due to continuing active MAPK and PI3K cascades in the presence of the inhibitor. Overall, our study provides a comprehensive assessment of co-occurring KRAS mutations in NSCLC and in vitro evidence of the negative impact of co-occurring KRAS mutations on cellular response to G12C inhibitors, highlighting the need for a comprehensive KRAS tumour genotyping for optimal patient selection for treatment with a KRAS G12C inhibitor.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Mutação/genética , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Taxa de Mutação , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Estudos Retrospectivos
11.
J Clin Oncol ; 40(17): 1870-1878, 2022 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-35192411

RESUMO

PURPOSE: Selumetinib can increase radioactive iodine (RAI) avidity in RAI-refractory tumors. We investigated whether selumetinib plus adjuvant RAI improves complete remission (CR) rates in patients with differentiated thyroid cancer (DTC) at high risk of primary treatment failure versus RAI alone. METHODS: ASTRA (ClinicalTrials.gov identifier: NCT01843062) is an international, phase III, randomized, placebo-controlled, double-blind trial. Patients with DTC at high risk of primary treatment failure (primary tumor > 4 cm; gross extrathyroidal extension outside the thyroid gland [T4 disease]; or N1a/N1b disease with ≥ 1 metastatic lymph node(s) ≥ 1 cm or ≥ 5 lymph nodes [any size]) were randomly assigned 2:1 to selumetinib 75 mg orally twice daily or placebo for approximately 5 weeks (no stratification). On treatment days 29-31, recombinant human thyroid-stimulating hormone (0.9 mg)-stimulated RAI (131I; 100 mCi/3.7 GBq) was administered, followed by 5 days of selumetinib/placebo. The primary end point (CR rate 18 months after RAI) was assessed in the intention-to-treat population. RESULTS: Four hundred patients were enrolled (August 27, 2013-March 23, 2016) and 233 randomly assigned (selumetinib, n = 155 [67%]; placebo, n = 78 [33%]). No statistically significant difference in CR rate 18 months after RAI was observed (selumetinib n = 62 [40%]; placebo n = 30 [38%]; odds ratio 1.07 [95% CI, 0.61 to 1.87]; P = .8205). Treatment-related grade ≥ 3 adverse events were reported in 25/154 patients (16%) with selumetinib and none with placebo. The most common adverse event with selumetinib was dermatitis acneiform (n = 11 [7%]). No treatment-related deaths were reported. CONCLUSION: Postoperative pathologic risk stratification identified patients with DTC at high risk of primary treatment failure, although the addition of selumetinib to adjuvant RAI failed to improve the CR rate for these patients. Future strategies should focus on tumor genotype-tailored drug selection and maintaining drug dosing to optimize RAI efficacy.


Assuntos
Adenocarcinoma , Neoplasias da Glândula Tireoide , Adenocarcinoma/tratamento farmacológico , Benzimidazóis/efeitos adversos , Método Duplo-Cego , Humanos , Radioisótopos do Iodo/efeitos adversos , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/radioterapia
12.
Mol Oncol ; 16(10): 1969-1985, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34866317

RESUMO

Nearly all estrogen receptor (ER)-positive (POS) metastatic breast cancers become refractory to endocrine (ET) and other therapies, leading to lethal disease presumably due to evolving genomic alterations. Timely monitoring of the molecular events associated with response/progression by serial tissue biopsies is logistically difficult. Use of liquid biopsies, including circulating tumor cells (CTC) and circulating tumor DNA (ctDNA), might provide highly informative, yet easily obtainable, evidence for better precision oncology care. Although ctDNA profiling has been well investigated, the CTC precision oncology genomic landscape and the advantages it may offer over ctDNA in ER-POS breast cancer remain largely unexplored. Whole-blood (WB) specimens were collected at serial time points from patients with advanced ER-POS/HER2-negative (NEG) advanced breast cancer in a phase I trial of AZD9496, an oral selective ER degrader (SERD) ET. Individual CTC were isolated from WB using tandem CellSearch® /DEPArray™ technologies and genomically profiled by targeted single-cell DNA next-generation sequencing (scNGS). High-quality CTC (n = 123) from 12 patients profiled by scNGS showed 100% concordance with ctDNA detection of driver estrogen receptor α (ESR1) mutations. We developed a novel CTC-based framework for precision medicine actionability reporting (MI-CTCseq) that incorporates novel features, such as clonal predominance and zygosity of targetable alterations, both unambiguously identifiable in CTC compared to ctDNA. Thus, we nominated opportunities for targeted therapies in 73% of patients, directed at alterations in phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), fibroblast growth factor receptor 2 (FGFR2), and KIT proto-oncogene, receptor tyrosine kinase (KIT). Intrapatient, inter-CTC genomic heterogeneity was observed, at times between time points, in subclonal alterations. Our analysis suggests that serial monitoring of the CTC genome is feasible and should enable real-time tracking of tumor evolution during progression, permitting more combination precision medicine interventions.


Assuntos
Neoplasias da Mama , DNA Tumoral Circulante , Células Neoplásicas Circulantes , Biomarcadores Tumorais/genética , Neoplasias da Mama/patologia , DNA Tumoral Circulante/genética , Antagonistas de Estrogênios , Estudos de Viabilidade , Feminino , Genômica , Humanos , Mutação/genética , Células Neoplásicas Circulantes/patologia , Medicina de Precisão
13.
Mol Cancer Ther ; 20(9): 1723-1734, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34224361

RESUMO

A recombinant Newcastle Disease Virus (NDV), encoding either a human (NDVhuGM-CSF, MEDI5395) or murine (NDVmuGM-CSF) GM-CSF transgene, combined broad oncolytic activity with the ability to significantly modulate genes related to immune functionality in human tumor cells. Replication in murine tumor lines was significantly diminished relative to human tumor cells. Nonetheless, intratumoral injection of NDVmuGM-CSF conferred antitumor effects in three syngeneic models in vivo; with efficacy further augmented by concomitant treatment with anti-PD-1/PD-L1 or T-cell agonists. Ex vivo immune profiling, including T-cell receptor sequencing, revealed profound immune-contexture changes consistent with priming and potentiation of adaptive immunity and tumor microenvironment (TME) reprogramming toward an immune-permissive state. CRISPR modifications rendered CT26 tumors significantly more permissive to NDV replication, and in this setting, NDVmuGM-CSF confers immune-mediated effects in the noninjected tumor in vivo Taken together, the data support the thesis that MEDI5395 primes and augments cell-mediated antitumor immunity and has significant utility as a combination partner with other immunomodulatory cancer treatments.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Imunomodulação , Imunoterapia/métodos , Vírus da Doença de Newcastle/genética , Terapia Viral Oncolítica/instrumentação , Microambiente Tumoral , Animais , Apoptose , Proliferação de Células , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Neoplasias do Colo/terapia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Nat Med ; 27(5): 793-801, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33941921

RESUMO

Durvalumab is a programmed death-ligand 1 (PD-L1) inhibitor with clinical activity in advanced urothelial cancer (AUC)1. AUC is characterized by several recurrent targetable genomic alterations2-5. This study ( NCT02546661 , BISCAY) combined durvalumab with relevant targeted therapies in biomarker-selected chemotherapy-refractory AUC populations including: (1) fibroblast growth factor receptor (FGFR) inhibitors in tumors with FGFR DNA alterations (FGFRm); (2) pharmacological inhibitor of the enzyme poly-ADP ribose polymerase (PARP) in tumors with and without DNA homologous recombination repair deficiency (HRRm); and (3) TORC1/2 inhibitors in tumors with DNA alteration to the mTOR/PI3K pathway3-5.This trial adopted a new, biomarker-driven, multiarm adaptive design. Safety, efficacy and relevant biomarkers were evaluated. Overall, 391 patients were screened of whom 135 were allocated to one of six study arms. Response rates (RRs) ranged 9-36% across the study arms, which did not meet efficacy criteria for further development. Overall survival (OS) and progression-free survival (PFS) were similar in the combination arms and durvalumab monotherapy arm. Biomarker analysis showed a correlation between circulating plasma-based DNA (ctDNA) and tissue for FGFRm. Sequential circulating tumor DNA analysis showed that changes to FGFRm correlated with clinical outcome. Our data support the clinical activity of FGFR inhibition and durvalumab monotherapy but do not show increased activity for any of the combinations. These findings question the targeted/immune therapy approach in AUC.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Terapia de Alvo Molecular/métodos , Neoplasias Urológicas/tratamento farmacológico , Antígeno B7-H1/antagonistas & inibidores , Benzamidas/uso terapêutico , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 2 de Rapamicina/antagonistas & inibidores , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Morfolinas/uso terapêutico , Fosfatidilinositol 3-Quinases/genética , Ftalazinas/uso terapêutico , Piperazinas/uso terapêutico , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/genética , Intervalo Livre de Progressão , Pirimidinas/uso terapêutico , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores de Fatores de Crescimento de Fibroblastos/genética , Serina-Treonina Quinases TOR/genética , Neoplasias Urológicas/genética , Neoplasias Urológicas/patologia , Urotélio/patologia
15.
Mol Cancer Ther ; 20(6): 1080-1091, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33785652

RESUMO

Suppressive myeloid cells mediate resistance to immune checkpoint blockade. PI3Kγ inhibition can target suppressive macrophages, and enhance efficacy of immune checkpoint inhibitors. However, how PI3Kγ inhibitors function in different tumor microenvironments (TME) to activate specific immune cells is underexplored. The effect of the novel PI3Kγ inhibitor AZD3458 was assessed in preclinical models. AZD3458 enhanced antitumor activity of immune checkpoint inhibitors in 4T1, CT26, and MC38 syngeneic models, increasing CD8+ T-cell activation status. Immune and TME biomarker analysis of MC38 tumors revealed that AZD3458 monotherapy or combination treatment did not repolarize the phenotype of tumor-associated macrophage cells but induced gene signatures associated with LPS and type II INF activation. The activation biomarkers were present across tumor macrophages that appear phenotypically heterogenous. AZD3458 alone or in combination with PD-1-blocking antibodies promoted an increase in antigen-presenting (MHCII+) and cytotoxic (iNOS+)-activated macrophages, as well as dendritic cell activation. AZD3458 reduced IL-10 secretion and signaling in primary human macrophages and murine tumor-associated macrophages, but did not strongly regulate IL-12 as observed in other studies. Therefore, rather than polarizing tumor macrophages, PI3Kγ inhibition with AZD3458 promotes a cytotoxic switch of macrophages into antigen-presenting activated macrophages, resulting in CD8 T-cell-mediated antitumor activity with immune checkpoint inhibitors associated with tumor and peripheral immune activation.


Assuntos
Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Checkpoint Imunológico/uso terapêutico , Animais , Modelos Animais de Doenças , Feminino , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos
16.
Clin Cancer Res ; 26(16): 4242-4249, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32234755

RESUMO

PURPOSE: Fulvestrant, the first-in-class selective estrogen receptor (ER) degrader (SERD), is clinically effective in patients with ER+ breast cancer, but it has administration and pharmacokinetic limitations. Pharmacodynamic data suggest complete ER degradation is not achieved at fulvestrant's clinically feasible dose. This presurgical study (NCT03236974) compared the pharmacodynamic effects of fulvestrant with AZD9496, a novel, orally bioavailable, nonsteroidal, potent SERD, in treatment-naïve patients with ER+ HER2- primary breast cancer awaiting curative intent surgery. PATIENTS AND METHODS: Patients were randomized 1:1 to receive AZD9496 250 mg twice daily from day 1 for 5-14 days, or fulvestrant 500 mg on day 1. On-treatment imaging-guided core tumor biopsies were taken between day 5 and 14 and compared with pretreatment diagnostic biopsies. The primary objective was to compare the effects of AZD9496 and fulvestrant on ER expression. Secondary objectives included changes in progesterone receptor (PR) and Ki-67 pharmacokinetic/pharmacodynamic relationships and safety. RESULTS: Forty-six women received treatment (AZD9496 n = 22; fulvestrant n = 24); 35 paired biopsies were evaluable (AZD9496 n = 15; fulvestrant n = 20). The least square mean estimate for ER H-score reduction was 24% after AZD9496 versus 36% after fulvestrant treatment (P = 0.86). AZD9496 also reduced PR H-scores (-33.3%) and Ki-67 levels (-39.9%) from baseline, but was also not superior to fulvestrant (PR: -68.7%, P = 0.97; Ki-67: -75.4%, P = 0.98). No new safety findings were identified. CONCLUSIONS: This was the first presurgical study to demonstrate that an oral SERD affects its key biological targets. However, AZD9496 was not superior to fulvestrant at the dose tested.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Cinamatos/administração & dosagem , Receptor alfa de Estrogênio/genética , Fulvestranto/administração & dosagem , Indóis/administração & dosagem , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Cinamatos/efeitos adversos , Estradiol/genética , Feminino , Fulvestranto/efeitos adversos , Humanos , Indóis/efeitos adversos , Pessoa de Meia-Idade , Receptor ErbB-2/genética , Receptores de Progesterona/genética
17.
Elife ; 72018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30543181

RESUMO

Optical super-resolution microscopy techniques enable high molecular specificity with high spatial resolution and constitute a set of powerful tools in the investigation of the structure of supramolecular assemblies such as viruses. Here, we report on a new methodology which combines Structured Illumination Microscopy (SIM) with machine learning algorithms to image and classify the structure of large populations of biopharmaceutical viruses with high resolution. The method offers information on virus morphology that can ultimately be linked with functional performance. We demonstrate the approach on viruses produced for oncolytic viriotherapy (Newcastle Disease Virus) and vaccine development (Influenza). This unique tool enables the rapid assessment of the quality of viral production with high throughput obviating the need for traditional batch testing methods which are complex and time consuming. We show that our method also works on non-purified samples from pooled harvest fluids directly from the production line.


Assuntos
Aprendizado de Máquina , Microscopia de Fluorescência/métodos , Vírus da Doença de Newcastle/química , Orthomyxoviridae/química , Algoritmos , Automação , Processamento de Imagem Assistida por Computador , Vacinas contra Influenza/imunologia , Vírus da Doença de Newcastle/ultraestrutura , Vacinas Atenuadas/imunologia
18.
Acad Radiol ; 22(2): 139-48, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25572926

RESUMO

RATIONALE AND OBJECTIVES: To develop and test an algorithm that outlines the breast boundaries using information from fat and water magnetic resonance images. MATERIALS AND METHODS: Three algorithms were implemented and tested using registered fat and water magnetic resonance images. Two of the segmentation algorithms are simple extensions of the techniques used for contrast-enhanced images: one algorithm uses clustering and local gradient (CLG) analysis and the other algorithm uses a Hessian-based sheetness filter (HSF). The third segmentation algorithm uses k-means++ and dynamic programming (KDP) for finding the breast pixels. All three algorithms separate the left and right breasts using either a fixed region or a morphological method. The performance is quantified using a mutual overlap (Dice) metric and a pectoral muscle boundary error. The algorithms are evaluated against three manual tracers using 266 breast images from 14 female subjects. RESULTS: The KDP algorithm has a mean overlap percentage improvement that is statistically significant relative to the HSF and CLG algorithms. When using a fixed region to remove the tissue between breasts with tracer 1 as a reference, the KDP algorithm has a mean overlap of 0.922 compared to 0.864 (P < .01) for HSF and 0.843 (P < .01) for CLG. The performance of KDP is very similar to tracers 2 (0.926 overlap) and 3 (0.929 overlap). The performance analysis in terms of pectoral muscle boundary error showed that the fraction of the muscle boundary within three pixels of reference tracer 1 is 0.87 using KDP compared to 0.578 for HSF and 0.617 for CLG. Our results show that the performance of the KDP algorithm is independent of breast density. CONCLUSIONS: We developed a new automated segmentation algorithm (KDP) to isolate breast tissue from magnetic resonance fat and water images. KDP outperforms the other techniques that focus on local analysis (CLG and HSF) and yields a performance similar to human tracers.


Assuntos
Tecido Adiposo/patologia , Água Corporal , Neoplasias da Mama/patologia , Interpretação de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética/métodos , Técnica de Subtração , Algoritmos , Mama , Feminino , Humanos , Aumento da Imagem/métodos , Reconhecimento Automatizado de Padrão/métodos , Programação Linear , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
19.
MAbs ; 6(6): 1425-38, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25484064

RESUMO

Generation of functional antibodies against integral membrane proteins such as the G-protein coupled receptor CXCR2 is technically challenging for several reasons, including limited epitope accessibility, the requirement for a lipid environment to maintain structure and their existence in dynamic conformational states. Antibodies to human CXCR2 were generated by immunization in vivo and by in vitro selection methods. Whole cell immunization of transgenic mice and screening of phage display libraries using CXCR2 magnetic proteoliposomes resulted in the isolation of antibodies with distinct modes of action. The hybridoma-derived antibody fully inhibited IL-8 and Gro-α responses in calcium flux and ß-arrestin recruitment assays. The phage-display derived antibodies were allosteric antagonists that showed ligand dependent differences in functional assays. The hybridoma and phage display antibodies did not cross-compete in epitope competition assays and mapping using linear and CLIPS peptides confirmed that they recognized distinct epitopes of human CXCR2. This illustrates the benefits of using parallel antibody isolation approaches with different antigen presentation methods to successfully generate functionally and mechanistically diverse antagonistic antibodies to human CXCR2. The method is likely to be broadly applicable to other complex membrane proteins.


Assuntos
Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos/imunologia , Epitopos/imunologia , Receptores de Interleucina-8B/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Arrestinas/imunologia , Arrestinas/metabolismo , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/imunologia , Cálcio/imunologia , Cálcio/metabolismo , Linhagem Celular , Técnicas de Visualização da Superfície Celular/métodos , Quimiocina CXCL1/imunologia , Quimiocina CXCL1/farmacologia , Mapeamento de Epitopos/métodos , Epitopos/metabolismo , Células HEK293 , Humanos , Hibridomas , Imunização , Interleucina-8/imunologia , Interleucina-8/farmacologia , Camundongos Transgênicos , Dados de Sequência Molecular , Biblioteca de Peptídeos , Ligação Proteica/imunologia , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo , Transdução de Sinais/imunologia , beta-Arrestinas
20.
NMR Biomed ; 22(1): 114-27, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19016452

RESUMO

Tumor choline metabolites have potential for use as diagnostic indicators of breast cancer phenotype and can be non-invasively monitored in vivo by MRS. Extract studies have determined that the principle diagnostic component of these peaks is phosphocholine (PCho), the biosynthetic precursor to the membrane phospholipid, phosphatidylcholine (PtdCho). The ability to resolve and quantify PCho in vivo would improve the accuracy of this putative diagnostic tool. In addition, determining the biochemical mechanisms underlying these metabolic perturbations will improve the understanding of breast cancer and may suggest potential molecular targets for drug development. Reported herein is the in vivo resolution and quantification of PCho and glycerophosphocholine (GPC) in breast cancer xenografts in SCID mice via image-guided 31P MRS, localized to a single voxel. Tumor metabolites are also detected using ex vivo extracts and high-resolution NMR spectroscopy and are quantified in the metastatic tumor line, MDA-mb-231. Also reported is the quantification of cytosolic and lipid metabolites in breast cells of differing cancer phenotype, and the identification of metabolites that differ among these cell lines. In cell extracts, PCho and the PtdCho breakdown products, lysophosphatidylcholine, GPC and glycerol 3-phosphate, are all raised in breast cancer lines relative to an immortalized non-malignant line. These metabolic differences are in direct agreement with differences in expression of genes encoding enzymes in the choline metabolic pathway. Results of this study are consistent with previous studies, which have concluded that increased choline uptake, increased choline kinase activity, and increased phosholipase-mediated turnover of PtdCho contribute to the observed increase in PCho in breast cancer. In addition, this study presents evidence suggesting a specific role for phospholipase A2-mediated PtdCho catabolism. Gene expression changes following taxane therapy are also reported and are consistent with previously reported changes in choline metabolites after the same therapy in the same tumor model.


Assuntos
Colina/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Espectroscopia de Ressonância Magnética , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/genética , Redes e Vias Metabólicas/genética , Animais , Extratos Celulares , Linhagem Celular Tumoral , Docetaxel , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Neoplásicos , Humanos , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Redes e Vias Metabólicas/efeitos dos fármacos , Camundongos , Camundongos SCID , Fenótipo , Fosfatidilcolinas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Taxoides/farmacologia , Taxoides/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA