Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-37772709

RESUMO

The mTOR inhibitor, everolimus, is an important clinical management component of metastatic ER+ breast cancer (BC). However, most patients develop resistance and progress on therapy, highlighting the need to discover strategies that increase mTOR inhibitor effectiveness. We developed ER+ BC cell lines, sensitive or resistant to everolimus, and discovered that combination treatment of ONC201/TIC10 with everolimus inhibited cell growth in 2D/3D in vitro studies. We confirmed increased therapeutic response in primary patient cells progressing on everolimus, supporting clinical relevance. We show that ONC201/TIC10 mechanism in metastatic ER+ BC cells involves oxidative phosphorylation inhibition and stress response activation. Transcriptomic analysis in everolimus resistant breast patient tumors and mitochondrial functional assays in resistant cell lines demonstrated increased mitochondrial respiration dependency, contributing to ONC201/TIC10 sensitivity. We propose that ONC201/TIC10 and modulation of mitochondrial function may provide an effective add-on therapy strategy for patients with metastatic ER+ BCs resistant to mTOR inhibitors.


Breast cancer is one of the most frequently diagnosed cancers globally, particularly among women. The most common type of breast cancer expresses a receptor for the hormone estrogen. Many treatments block the activity of estrogen and therefore slow or block the development and spread of this type of breast cancer. For patients with advanced breast cancer, hormone-blocking treatments work best in combination with other drugs, including one called everolimus. However, in many patients the cancer cells become resistant to these therapies, leading to disease progression and decreased survival. To explore treatment strategies that could enhance the effectiveness of existing therapies for breast cancer, Farmaki et al. studied how cancer cells which had become resistant to everolimus responded when treated with an experimental drug called ONC201/TIC10. A combination of everolimus and ONC201/TIC10 inhibited growth of resistant cancer cells that had been grown in a three-dimensional arrangement to mimic human tumors. Moreover, the drug combination effectively targeted breast cancer cells collected from patients whose cancer had progressed while being treated with everolimus, suggesting that ONC201/TIC10 could be relevant in a clinical setting. Finally, molecular and biochemical experiments revealed that the drug ONC201/TIC10 works by disrupting the pathways that everolimus-resistant cancer cells use to generate the energy required to grow and proliferate. Taken together these findings suggest that ONC201/TIC10 may provide an effective add-on therapy for patients with certain types of advanced breast cancer that are no longer responding to everolimus. Before this becomes a reality for patients, however, there will have to be more experimental testing of ONC201/TIC10 to determine optimal dosing and timing strategy for future clinical trials.


Assuntos
Antineoplásicos , Neoplasias da Mama , Imidazóis , Piridinas , Pirimidinas , Humanos , Feminino , Everolimo/farmacologia , Everolimo/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Inibidores de MTOR , Linhagem Celular Tumoral , Serina-Treonina Quinases TOR , Resistencia a Medicamentos Antineoplásicos
2.
Front Mol Biosci ; 9: 981962, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36304922

RESUMO

Endocrine therapy remains the primary treatment choice for ER+ breast cancers. However, most advanced ER+ breast cancers ultimately develop resistance to endocrine. This acquired resistance to endocrine therapy is often driven by the activation of the PI3K/AKT/mTOR signaling pathway. Everolimus, a drug that targets and inhibits the mTOR complex has been shown to improve clinical outcomes in metastatic ER+ breast cancers. However, there are no biomarkers currently available to guide the use of everolimus in the clinic for progressive patients, where multiple therapeutic options are available. Here, we utilized gene expression signatures from 9 ER+ breast cancer cell lines and 23 patients treated with everolimus to develop and validate an integrative machine learning biomarker of mTOR inhibitor response. Our results show that the machine learning biomarker can successfully distinguish responders from non-responders and can be applied to identify patients that will most likely benefit from everolimus treatment.

3.
Neurotherapeutics ; 19(2): 635-648, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35226341

RESUMO

Resection of brain tumors frequently causes injury to the surrounding brain tissue that exacerbates cerebral edema by activating an inflammatory cascade. Although corticosteroids are often utilized peri-operatively to alleviate the symptoms associated with brain edema, they increase operative morbidities and suppress the efficacy of immunotherapy. Thus, novel approaches to minimize cerebral edema caused by neurosurgical procedures will have significant utility in the management of patients with brain tumors. We have studied the role of the receptor for advanced glycation end products (RAGE) and its ligands on inflammatory responses to neurosurgical injury in mice and humans. Blood-brain barrier (BBB) integrity and neuroinflammation were characterized by Nanostring, flow cytometry, qPCR, and immunoblotting of WT and RAGE knockout mice brains subjected to surgical brain injury (SBI). Human tumor tissue and fluid collected from the resection cavity of patients undergoing craniotomy were also analyzed by single-cell RNA sequencing and ELISA. Genetic ablation of RAGE significantly abrogated neuroinflammation and BBB disruption in the murine SBI model. The inflammatory responses to SBI were associated with infiltration of S100A9-expressing myeloid-derived cells into the brain. Local release of pro-inflammatory S100A9 was confirmed in patients following tumor resection. RAGE and S100A9 inhibitors were as effective as dexamethasone in attenuating neuroinflammation. However, unlike dexamethasone and S100A9 inhibitor, RAGE inhibition did not diminish the efficacy of anti-PD-1 immunotherapy in glioma-bearing mice. These observations confirm the role of the RAGE axis in surgically induced neuroinflammation and provide an alternative therapeutic option to dexamethasone in managing post-operative cerebral edema.


Assuntos
Anti-Inflamatórios , Edema Encefálico , Neoplasias Encefálicas , Receptor para Produtos Finais de Glicação Avançada , Animais , Anti-Inflamatórios/farmacologia , Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Lesões Encefálicas/complicações , Neoplasias Encefálicas/cirurgia , Dexametasona/uso terapêutico , Modelos Animais de Doenças , Humanos , Camundongos , Ratos , Ratos Sprague-Dawley , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores
4.
Nat Cancer ; 2(6): 658-671, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34712959

RESUMO

Combining cyclin-dependent kinase (CDK) inhibitors with endocrine therapy improves outcomes for metastatic estrogen receptor positive (ER+) breast cancer patients but its value in earlier stage patients is unclear. We examined evolutionary trajectories of early-stage breast cancer tumors, using single cell RNA sequencing (scRNAseq) of serial biopsies from the FELINE clinical trial (#NCT02712723) of endocrine therapy (letrozole) alone or combined with the CDK inhibitor ribociclib. Despite differences in subclonal diversity evolution across patients and treatments, common resistance phenotypes emerged. Resistant tumors treated with combination therapy showed accelerated loss of estrogen signaling with convergent up-regulation of JNK signaling through growth factor receptors. In contrast, cancer cells maintaining estrogen signaling during mono- or combination therapy showed potentiation of CDK4/6 activation and ERK upregulation through ERBB4 signaling. These results indicate that combination therapy in early-stage ER+ breast cancer leads to emergence of resistance through a shift from estrogen to alternative growth signal-mediated proliferation.


Assuntos
Neoplasias da Mama , Neoplasias da Mama/tratamento farmacológico , Ensaios Clínicos como Assunto , Quinase 4 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/genética , Estrogênios/uso terapêutico , Feminino , Genômica , Humanos , Receptores de Estrogênio/genética
5.
Nat Commun ; 12(1): 3039, 2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-34031395

RESUMO

The evolution of resistance in high-grade serous ovarian cancer (HGSOC) cells following chemotherapy is only partially understood. To understand the selection of factors driving heterogeneity before and through adaptation to treatment, we profile single-cell RNA-sequencing (scRNA-seq) transcriptomes of HGSOC tumors collected longitudinally during therapy. We analyze scRNA-seq data from two independent patient cohorts to reveal that HGSOC is driven by three archetypal phenotypes, defined as oncogenic states that describe the majority of the transcriptome variation. Using a multi-task learning approach to identify the biological tasks of each archetype, we identify metabolism and proliferation, cellular defense response, and DNA repair signaling as consistent cell states found across patients. Our analysis demonstrates a shift in favor of the metabolism and proliferation archetype versus cellular defense response archetype in cancer cells that received multiple lines of treatment. While archetypes are not consistently associated with specific whole-genome driver mutations, they are closely associated with subclonal populations at the single-cell level, indicating that subclones within a tumor often specialize in unique biological tasks. Our study reveals the core archetypes found in progressive HGSOC and shows consistent enrichment of subclones with the metabolism and proliferation archetype as resistance is acquired to multiple lines of therapy.


Assuntos
Cistadenocarcinoma Seroso/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Fenótipo , Linhagem Celular Tumoral , Reparo do DNA , Resistencia a Medicamentos Antineoplásicos , Feminino , Heterogeneidade Genética , Humanos , Mutação , Análise de Sequência de RNA , Transcriptoma
6.
Transl Oncol ; 14(1): 100946, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33221681

RESUMO

Cancer cell phenotypes evolve during a tumor's treatment. In some cases, tumor cells acquire cancer stem cell-like (CSL) traits such as resistance to chemotherapy and diminished differentiation; therefore, targeting these cells may be therapeutically beneficial. In this study we show that in progressive estrogen receptor positive (ER+) metastatic breast cancer tumors, resistant subclones that emerge following chemotherapy have increased CSL abundance. Further, in vitro organoid growth of ER+ patient cancer cells also shows that chemotherapy treatment leads to increased abundance of ALDH+/CD44+ CSL cells. Chemotherapy induced CSL abundance is blocked by treatment with a pan-HDAC inhibitor, belinostat. Belinostat treatment diminished both mammosphere formation and size following chemotherapy, indicating a decrease in progenitor CSL traits. HDAC inhibitors specific to class IIa (HDAC4, HDAC5) and IIb (HDAC6) were shown to primarily reverse the chemo-resistant CSL state. Single-cell RNA sequencing analysis with patient samples showed that HDAC targets and MYC signaling were promoted by chemotherapy and inhibited upon HDAC inhibitor treatment. In summary, HDAC inhibition can block chemotherapy-induced drug resistant phenotypes with 'one-two punch' strategy in refractory breast cancer cells.

7.
Cancer Cell Int ; 20: 253, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32565737

RESUMO

BACKGROUND: CDK4/6 inhibitors such as ribociclib are becoming widely used targeted therapies in hormone-receptor-positive (HR+) human epidermal growth factor receptor 2-negative (HER2-) breast cancer. However, cancers can advance due to drug resistance, a problem in which tumor heterogeneity and evolution are key features. METHODS: Ribociclib-resistant HR+/HER2- CAMA-1 breast cancer cells were generated through long-term ribociclib treatment. Characterization of sensitive and resistant cells were performed using RNA sequencing and whole exome sequencing. Lentiviral labeling with different fluorescent proteins enabled us to track the proliferation of sensitive and resistant cells under different treatments in a heterogeneous, 3D spheroid coculture system using imaging microscopy and flow cytometry. RESULTS: Transcriptional profiling of sensitive and resistant cells revealed the downregulation of the G2/M checkpoint in the resistant cells. Exploiting this acquired vulnerability; resistant cells exhibited collateral sensitivity for the Wee-1 inhibitor, adavosertib (AZD1775). The combination of ribociclib and adavosertib achieved additional antiproliferative effect exclusively in the cocultures compared to monocultures, while decreasing the selection for resistant cells. CONCLUSIONS: Our results suggest that optimal antiproliferative effects in heterogeneous cancers can be achieved via an integrative therapeutic approach targeting sensitive and resistant cancer cell populations within a tumor, respectively.

8.
Proc Natl Acad Sci U S A ; 117(27): 16072-16082, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32571915

RESUMO

The extent to which immune cell phenotypes in the peripheral blood reflect within-tumor immune activity prior to and early in cancer therapy is unclear. To address this question, we studied the population dynamics of tumor and immune cells, and immune phenotypic changes, using clinical tumor and immune cell measurements and single-cell genomic analyses. These samples were serially obtained from a cohort of advanced gastrointestinal cancer patients enrolled in a trial with chemotherapy and immunotherapy. Using an ecological population model, fitted to clinical tumor burden and immune cell abundance data from each patient, we find evidence of a strong tumor-circulating immune cell interaction in responder patients but not in those patients that progress on treatment. Upon initiation of therapy, immune cell abundance increased rapidly in responsive patients, and once the peak level is reached tumor burden decreases, similar to models of predator-prey interactions; these dynamic patterns were absent in nonresponder patients. To interrogate phenotype dynamics of circulating immune cells, we performed single-cell RNA sequencing at serial time points during treatment. These data show that peripheral immune cell phenotypes were linked to the increased strength of patients' tumor-immune cell interaction, including increased cytotoxic differentiation and strong activation of interferon signaling in peripheral T cells in responder patients. Joint modeling of clinical and genomic data highlights the interactions between tumor and immune cell populations and reveals how variation in patient responsiveness can be explained by differences in peripheral immune cell signaling and differentiation soon after the initiation of immunotherapy.


Assuntos
Comunicação Celular/imunologia , Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Fenótipo , Microambiente Tumoral/imunologia , Regulação da Expressão Gênica , Humanos , Fatores Imunológicos/genética , Fatores Imunológicos/imunologia , Monócitos/imunologia , Análise de Sequência de RNA , Análise de Célula Única , Linfócitos T/imunologia
9.
Drug Metab Dispos ; 44(12): 1940-1948, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27621203

RESUMO

Traditional in vitro human liver cell culture models lose key hepatic functions such as metabolic activity during short-term culture. Advanced three-dimensional (3D) liver coculture platforms offer the potential for extended hepatocyte functionality and allow for the study of more complex biologic interactions, which can improve and refine human drug safety evaluations. Here, we use a perfusion flow 3D microreactor platform for the coculture of cryopreserved primary human hepatocytes and Kupffer cells to study the regulation of cytochrome P450 3A4 isoform (CYP3A4) activity by chronic interleukin 6 (IL-6)-mediated inflammation over 2 weeks. Hepatocyte cultures remained stable over 2 weeks, with consistent albumin production and basal IL-6 levels. Direct IL-6 stimulation that mimics an inflammatory state induced a dose-dependent suppression of CYP3A4 activity, an increase in C-reactive protein (CRP) secretion, and a decrease in shed soluble interleukin-6 receptor (IL-6R) levels, indicating expected hepatic IL-6 bioactivity. Tocilizumab, an anti-IL-6R monoclonal antibody used to treat rheumatoid arthritis, has been demonstrated clinically to impact small molecule drug pharmacokinetics by modulating cytochrome P450 enzyme activities, an effect not observed in traditional hepatic cultures. We have now recapitulated the clinical observation in a 3D bioreactor system. Tocilizumab was shown to desuppress CYP3A4 activity while reducing the CRP concentration after 72 hours in the continued presence of IL-6. This change in CYP3A4 activity decreased the half-life and area under the curve up to the last measurable concentration (AUClast) of the small molecule CYP3A4 substrate simvastatin hydroxy acid, measured before and after tocilizumab treatment. We conclude that next-generation in vitro liver culture platforms are well suited for these types of long-term treatment studies and show promise for improved drug safety assessment.


Assuntos
Anticorpos Monoclonais/metabolismo , Interações Medicamentosas/fisiologia , Fígado/metabolismo , Bibliotecas de Moléculas Pequenas/metabolismo , Anticorpos Monoclonais Humanizados/metabolismo , Proteína C-Reativa/metabolismo , Células Cultivadas , Técnicas de Cocultura/métodos , Citocromo P-450 CYP3A/metabolismo , Meia-Vida , Hepatócitos/metabolismo , Humanos , Inflamação/metabolismo , Interleucina-6/metabolismo , Células de Kupffer/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA