Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Transl Med ; 22(1): 85, 2024 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-38246995

RESUMEN

The extracellular matrix (ECM) plays critical roles in cytoskeletal support, biomechanical transduction and biochemical signal transformation. Tumor-associated macrophage (TAM) function is regulated by matrix stiffness in solid tumors and is often associated with poor prognosis. ECM stiffness-induced mechanical cues can activate cell membrane mechanoreceptors and corresponding mechanotransducers in the cytoplasm, modulating the phenotype of TAMs. Currently, tuning TAM polarization through matrix stiffness-induced mechanical stimulation has received increasing attention, whereas its effect on TAM fate has rarely been summarized. A better understanding of the relationship between matrix stiffness and macrophage function will contribute to the development of new strategies for cancer therapy. In this review, we first introduced the overall relationship between macrophage polarization and matrix stiffness, analyzed the changes in mechanoreceptors and mechanotransducers mediated by matrix stiffness on macrophage function and tumor progression, and finally summarized the effects of targeting ECM stiffness on tumor prognosis to provide insight into this new field.


Asunto(s)
Macrófagos , Macrófagos Asociados a Tumores , Membrana Celular , Citoplasma , Matriz Extracelular
2.
Cancer Sci ; 113(8): 2849-2861, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35661486

RESUMEN

Although resistance to poly(ADP-ribose) polymerase inhibitors (PARPi) has gradually become a major challenge in the maintenance therapy for high-grade serous ovarian carcinoma (HGSOC), there are no universal indicators for resistance monitoring in patients. A key resistance mechanism to PARPi is the restoration of homologous recombination repair (HRR), including BRCA reversion mutations and changes in DNA damage repair proteins. To explore mutation profiles associated with PARPi resistance, we undertook targeted 42-gene deep sequencing of circulating cell-free DNA (cfDNA) extracted from HGSOC patients pre- and post-treatment with olaparib maintenance therapy. We found that pathogenic germline mutations in the HRR pathway, including BRCA1/2, were strongly associated with improved clinical outcomes, and newly acquired MRE11A mutations significantly shortened the progression-free survival (PFS) of patients. Furthermore, dynamic fluctuations of somatic mutation sites in CHEK2:p.K373E and CHEK2:p.R406H can be used for evaluating the therapeutic efficacy of patients. MRE11A:p.K464R might be a vital driving factor of olaparib resistance, as patients with newly acquired MRE11A:p.K464R in post-treatment cfDNA had significantly shorter PFS than those without it. These findings provide potential noninvasive biomarkers for efficacy evaluation and resistance monitoring of olaparib treatment, and lay the foundation for developing combination treatment after olaparib resistance.


Asunto(s)
Ácidos Nucleicos Libres de Células , Neoplasias Ováricas , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Mutación , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Ftalazinas/farmacología , Ftalazinas/uso terapéutico , Piperazinas , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico
3.
Microb Ecol ; 84(1): 302-313, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34405250

RESUMEN

The vaginal microbiota is closely related to HPV infection and cervical cancer (CC), but its relationship with platinum-based chemotherapy responsiveness is unknown. The study aimed to investigate the vaginal microbiota diversity of women with locally advanced cervical cancer (LACC) and compare the differences between responders and nonresponders. We characterized the 16S rRNA gene sequencing of vaginal microbiome of 66 vaginal samples, including 26 LACC patients before neoadjuvant chemotherapy and 40 healthy controls. Compared with the healthy controls, alpha diversity was significantly increased in CC patients (p <0.05) with more unconventionality bacterial colonization. Beta diversity also significantly differed between cervical cancer patients and controls (p <0.01). Within the CC patients, alpha diversity in vaginal samples was significantly higher in the nonresponders versus the responders (p <0.01), and the Ace index and Chao index were negatively correlated with mass reduction (p <0.001). Moreover, the Bacteroides genus enriched in the nonresponders had a ROC-plot AUC value reaching 0.84. The study suggests the vaginal microbiota in LACC patients is associated with platinum-based chemotherapy responsiveness. Alpha diversity and Bacteroides abundance have the potential of identifying platinum-resistant patients at an early time. These findings provide a basis for further research on the relationship between vaginal microbiome and chemotherapy effect in LACC.


Asunto(s)
Microbiota , Neoplasias del Cuello Uterino , Femenino , Humanos , Microbiota/genética , Terapia Neoadyuvante , ARN Ribosómico 16S/genética , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/microbiología , Vagina/microbiología
4.
Ann Intern Med ; 174(4): 453-461, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33284684

RESUMEN

BACKGROUND: The understanding of viral positivity and seroconversion during the course of coronavirus disease 2019 (COVID-19) is limited. OBJECTIVE: To describe patterns of viral polymerase chain reaction (PCR) positivity and evaluate their correlations with seroconversion and disease severity. DESIGN: Retrospective cohort study. SETTING: 3 designated specialty care centers for COVID-19 in Wuhan, China. PARTICIPANTS: 3192 adult patients with COVID-19. MEASUREMENTS: Demographic, clinical, and laboratory data. RESULTS: Among 12 780 reverse transcriptase PCR tests for severe acute respiratory syndrome coronavirus 2 that were done, 24.0% had positive results. In 2142 patients with laboratory-confirmed COVID-19, the viral positivity rate peaked within the first 3 days. The median duration of viral positivity was 24.0 days (95% CI, 18.9 to 29.1 days) in critically ill patients and 18.0 days (CI, 16.8 to 19.1 days) in noncritically ill patients. Being critically ill was an independent risk factor for longer viral positivity (hazard ratio, 0.700 [CI, 0.595 to 0.824]; P < 0.001). In patients with laboratory-confirmed COVID-19, the IgM-positive rate was 19.3% in the first week, peaked in the fifth week (81.5%), and then decreased steadily to around 55% within 9 to 10 weeks. The IgG-positive rate was 44.6% in the first week, reached 93.3% in the fourth week, and then remained high. Similar antibody responses were seen in clinically diagnosed cases. Serum inflammatory markers remained higher in critically ill patients. Among noncritically ill patients, a higher proportion of those with persistent viral positivity had low IgM titers (<100 AU/mL) during the entire course compared with those with short viral positivity. LIMITATION: Retrospective study and irregular viral and serology testing. CONCLUSION: The rate of viral PCR positivity peaked within the initial few days. Seroconversion rates peaked within 4 to 5 weeks. Dynamic laboratory index changes corresponded well to clinical signs, the recovery process, and disease severity. Low IgM titers (<100 AU/mL) are an independent risk factor for persistent viral positivity. PRIMARY FUNDING SOURCE: None.


Asunto(s)
Prueba de COVID-19 , COVID-19/diagnóstico , Neumonía Viral/diagnóstico , Neumonía Viral/virología , SARS-CoV-2 , Carga Viral , Adulto , Anciano , Anticuerpos Antivirales/sangre , COVID-19/epidemiología , China/epidemiología , Enfermedad Crítica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pandemias , Neumonía Viral/epidemiología , Estudios Retrospectivos , Seroconversión , Índice de Severidad de la Enfermedad
5.
Gynecol Oncol ; 163(2): 358-363, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34507827

RESUMEN

BACKGROUND: Gynecologic cancers seriously threaten women's life and health. This study aims to assess the long-term trends of mortality from the three major gynecologic cancers in China and to examine the age-, period-, and cohort-specific effects behind them during the period 1990 to 2019. METHODS: The mortality data of cervical, ovarian, and uterine cancer in China were obtained from the Global Burden of Disease Study 2019 and were analyzed with the age-period-cohort framework. RESULTS: It was found that the net drift for cervical cancer mortality was -0.19% (95% CI, -0.46% to 0.08%) per year, for ovarian cancer was 0.76% (95% CI, 0.57% to 0.95%) per year, and for uterine cancer was -3.09% (95% CI, -3.44% to -2.76%) per year from 1990 to 2019. During this period, while cervical cancer remained the most common cause of death among gynecologic cancers among Chinese women, ovarian cancer replaced uterine cancer as the second leading cause of death in gynecologic cancers after about 2005. Significant age, cohort, and period effects were found for the mortality trends of all three major gynecologic cancers. CONCLUSIONS: The secular trends of mortality from the three major gynecologic cancers in China and their underlying age, period, and cohort effects are likely to reflect the progress of diagnosis and treatment, rapid socio-economic transitions, and the accompanying lifestyle and behavior changes. More priorities of further epidemiology studies and efforts on the prevention and control should be given to three major gynecologic cancers.


Asunto(s)
Neoplasias Ováricas/mortalidad , Neoplasias del Cuello Uterino/mortalidad , Neoplasias Uterinas/mortalidad , Adulto , Factores de Edad , Anciano , Pueblo Asiatico/estadística & datos numéricos , China/epidemiología , Efecto de Cohortes , Femenino , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Persona de Mediana Edad , Mortalidad/historia , Mortalidad/tendencias , Neoplasias Ováricas/historia , Neoplasias del Cuello Uterino/historia , Neoplasias Uterinas/historia , Adulto Joven
6.
J Cancer Res Clin Oncol ; 150(1): 13, 2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38231277

RESUMEN

PURPOSE: WEE1 is a crucial kinase involved in the regulation of G2/M checkpoint within the cell cycle. This article aims to comprehensively review the existing knowledge on the implication of WEE1 as a therapeutic target in tumor progression and drug resistance. Furthermore, we summarize the current predictive biomarkers employed to treat cancer with WEE1 inhibitors. METHODS: A systematic review of the literature was conducted to analyze the association between WEE1 inhibition and cancer progression, including tumor advancement and drug resistance. Special attention was paid to the identification and utilization of predictive biomarkers related to therapeutic response to WEE1 inhibitors. RESULTS: The review highlights the intricate involvement of WEE1 in tumor progression and drug resistance. It synthesizes the current knowledge on predictive biomarkers employed in WEE1 inhibitor treatments, offering insights into their prognostic significance. Notably, the article elucidates the potential for precision medicine by understanding these biomarkers in the context of tumor treatment outcomes. CONCLUSION: WEE1 plays a pivotal role in tumor progression and is a promising therapeutic target. Distinguishing patients that would benefit from WEE1 inhibition will be a major direction of future research.


Asunto(s)
Neoplasias , Medicina de Precisión , Humanos , Biomarcadores , Proteínas de Ciclo Celular , División Celular , Puntos de Control de la Fase G2 del Ciclo Celular , Neoplasias/tratamiento farmacológico , Proteínas Tirosina Quinasas
7.
Nat Commun ; 15(1): 2089, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38453961

RESUMEN

Hyperthermic intraperitoneal chemotherapy's role in ovarian cancer remains controversial, hindered by limited understanding of hyperthermia-induced tumor cellular changes. This limits developing potent combinatory strategies anchored in hyperthermic intraperitoneal therapy (HIPET). Here, we perform a comprehensive multi-omics study on ovarian cancer cells under hyperthermia, unveiling a distinct molecular panorama, primarily characterized by rapid protein phosphorylation changes. Based on the phospho-signature, we pinpoint CDK1 kinase is hyperactivated during hyperthermia, influencing the global signaling landscape. We observe dynamic, reversible CDK1 activity, causing replication arrest and early mitotic entry post-hyperthermia. Subsequent drug screening shows WEE1 inhibition synergistically destroys cancer cells with hyperthermia. An in-house developed miniaturized device confirms hyperthermia and WEE1 inhibitor combination significantly reduces tumors in vivo. These findings offer additional insights into HIPET, detailing molecular mechanisms of hyperthermia and identifying precise drug combinations for targeted treatment. This research propels the concept of precise hyperthermic intraperitoneal therapy, highlighting its potential against ovarian cancer.


Asunto(s)
Hipertermia Inducida , Neoplasias Ováricas , Femenino , Humanos , Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Multiómica , Mitosis , Neoplasias Ováricas/terapia , Neoplasias Ováricas/patología
8.
Cell Rep Med ; 5(7): 101631, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38986623

RESUMEN

Ovarian cancer (OC) manifests as a complex disease characterized by inter- and intra-patient heterogeneity. Despite enhanced biological and genetic insights, OC remains a recalcitrant malignancy with minimal survival improvement. Based on multi-site sampling and a multi-lineage patient-derived xenograft (PDX) establishment strategy, we present herein the establishment of a comprehensive PDX biobank from histologically and molecularly heterogeneous OC patients. Comprehensive profiling of matched PDX and patient samples demonstrates that PDXs closely recapitulate parental tumors. By leveraging multi-lineage models, we reveal that the previously reported genomic disparities of PDX could be mainly attributed to intra-patient spatial heterogeneity instead of substantial model-independent genomic evolution. Moreover, DNA damage response pathway inhibitor (DDRi) screening uncovers heterogeneous responses across models. Prolonged iterative drug exposure recapitulates acquired drug resistance in initially sensitive models. Meanwhile, interrogation of induced drug-resistant (IDR) models reveals that suppressed interferon (IFN) response and activated Wnt/ß-catenin signaling contribute to acquired DDRi drug resistance.


Asunto(s)
Neoplasias Ováricas , Humanos , Femenino , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , Animales , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto , Vía de Señalización Wnt/genética , Resistencia a Antineoplásicos/genética , Genómica/métodos , Bancos de Muestras Biológicas , Heterogeneidad Genética , Daño del ADN/genética , Interferones/metabolismo , Interferones/genética , Linaje de la Célula/genética
9.
Cell Biosci ; 13(1): 178, 2023 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-37759323

RESUMEN

BACKGROUND: Although the clinical application of PARP inhibitors has brought hope to ovarian cancer, the problem of its resistance has become increasingly prominent. Therefore, clinical experts have been focused on finding specific indicators and therapeutic targets that can be used for resistance monitoring of PARP inhibitors. RESULTS: By cfDNA detecting during Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer, we found the presence of MRE11:p.K464R mutation was strongly associated with acquired Olaparib resistance. Structural analysis revealed that the MRE11:p.K464R mutation is situated at a critical site where the MRE11 protein interacts with other biomolecules, leading to potential structural and functional abnormalities of MRE11 protein. Functionally, MRE11:p.K464R mutation enhanced the tolerance of Olaparib by reducing the DNA damage. Mechanistically, MRE11:p.K464R mutation improved the efficiency of DNA damage repair and induce Olaparib resistance by enhancing its binding activity with the interacting proteins (including RAD50 and RPS3). Among them, the enhanced binding of MRE11:p.K464R mutation to RAD50/RPS3 facilitated non-homologous end joining (NHEJ) repair in tumor cells, thereby expanding the scope of research into acquired resistance to PARP inhibitors. CONCLUSIONS: Our findings provide a theoretical basis for MRE11:p.K464R mutation as a specific indicator of resistance monitoring in Olaparib treatment, and the exploration of its resistance mechanism provides a novel insights for the formulation of combination ther therapies after Olaparib resistance.

10.
Acta Biomater ; 157: 428-441, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36549633

RESUMEN

Ovarian cancer (OC) ranks first among gynecologic malignancies in terms of mortality. The benefits of poly (ADP-ribose) polymerase (PARP) inhibitors appear to be limited to OC with BRCA mutations. Concurrent administration of WEE1 inhibitors (eg, adavosertib (Ada)) and PARP inhibitors (eg, olaparib (Ola)) effectively suppress ovarian tumor growth regardless of BRCA mutation status, but is poorly tolerated. Henceforth, we aimed to seek a strategy to reduce the toxic effects of this combination by taking advantage of the mesoporous polydopamine (MPDA) nanoparticles with good biocompatibility and high drug loading capacity. In this work, we designed a tumor-targeting peptide TMTP1 modified MPDA-based nano-drug delivery system (TPNPs) for targeted co-delivery of Ada and Ola to treat OC. Ada and Ola could be effectively loaded into MPDA nanoplatform and showed tumor microenvironment triggered release behavior. The nanoparticles induced more apoptosis in OC cells, and significantly enhanced the synergy of combination therapy with Ada plus Ola in murine OC models. Moreover, the precise drug delivery of TPNPs towards tumor cells significantly diminished the toxic side effects caused by concurrent administration of Ada and Ola. Co-delivery of WEE1 inhibitors and PARP inhibitors via TPNPs represents a promising approach for the treatment of OC. STATEMENT OF SIGNIFICANCE: Combination therapy of WEE1 inhibitors (eg, Ada) with PARP inhibitors (eg, Ola) effectively suppress ovarian tumor growth regardless of BRCA mutation status. However, poor tolerability limits its clinical application. To address this issue, we construct a tumor-targeting nano-drug delivery system (TPNP) for co-delivery of Ada and Ola. The nanoparticles specifically target ovarian cancer and effectively enhance the antitumor effect while minimizing undesired toxic side effects. As the first nanomedicine co-loaded with a WEE1 inhibitor and a PARP inhibitor, TPNP-Ada-Ola may provide a promising and generally applicable therapeutic strategy for ovarian cancer patients.


Asunto(s)
Nanopartículas , Neoplasias Ováricas , Animales , Femenino , Humanos , Ratones , Línea Celular Tumoral , Sistema de Administración de Fármacos con Nanopartículas/efectos adversos , Neoplasias Ováricas/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/efectos adversos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Microambiente Tumoral
11.
J Immunother Cancer ; 11(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37072347

RESUMEN

BACKGROUND: Mismatch repair deficiency (dMMR) is a well-recognized biomarker for response to immune checkpoint blockade (ICB). Strategies to convert MMR-proficient (pMMR) to dMMR phenotype with the goal of sensitizing tumors to ICB are highly sought. The combination of bromodomain containing 4 (BRD4) inhibition and ICB provides a promising antitumor effect. However, the mechanisms underlying remain unknown. Here, we identify that BRD4 inhibition induces a persistent dMMR phenotype in cancers. METHODS: We confirmed the correlation between BRD4 and mismatch repair (MMR) by the bioinformatic analysis on The Cancer Genome Atlas and Clinical Proteomic Tumor Analysis Consortium data, and the statistical analysis on immunohistochemistry (IHC) scores of ovarian cancer specimens. The MMR genes (MLH1,MSH2,MSH6,PMS2) were measured by quantitative reverse transcription PCR, western blot, and IHC. The MMR status was confirmed by whole exome sequencing, RNA sequencing, MMR assay and hypoxanthine-guanine phosphoribosyl transferase gene mutation assay. The BRD4i AZD5153 resistant models were induced both in vitro and in vivo. The transcriptional effects of BRD4 on MMR genes were investigated by chromatin immunoprecipitation among cell lines and data from the Cistrome Data Browser. The therapeutic response to ICB was testified in vivo. The tumor immune microenvironment markers, such as CD4, CD8, TIM-3, FOXP3, were measured by flow cytometry. RESULTS: We identified the positive correlation between BRD4 and MMR genes in transcriptional and translational aspects. Also, the inhibition of BRD4 transcriptionally reduced MMR genes expression, resulting in dMMR status and elevated mutation loads. Furthermore, prolonged exposure to AZD5153 promoted a persistent dMMR signature both in vitro and in vivo, enhancing tumor immunogenicity, and increased sensitivity to α-programmed death ligand-1 therapy despite the acquired drug resistance. CONCLUSIONS: We demonstrated that BRD4 inhibition suppressed expression of genes critical to MMR, dampened MMR, and increased dMMR mutation signatures both in vitro and in vivo, sensitizing pMMR tumors to ICB. Importantly, even in BRD4 inhibitors (BRD4i)-resistant tumor models, the effects of BRD4i on MMR function were maintained rendering tumors sensitive to ICB. Together, these data identified a strategy to induce dMMR in pMMR tumors and further, indicated that BRD4i sensitive and resistant tumors could benefit from immunotherapy.


Asunto(s)
Neoplasias Colorrectales , Proteínas Nucleares , Humanos , Proteínas Nucleares/genética , Inhibidores de Puntos de Control Inmunológico , Reparación de la Incompatibilidad de ADN/genética , Factores de Transcripción/genética , Proteómica , Neoplasias Colorrectales/patología , Mutación , Microambiente Tumoral , Proteínas de Ciclo Celular/genética
12.
Cell Genom ; 3(1): 100211, 2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36777180

RESUMEN

Cervical cancer (CC) that is caused by high-risk human papillomavirus (HPV) remains a significant public health problem worldwide. HPV integration sites can be silent or actively transcribed, leading to the production of viral-host fusion transcripts. Herein, we demonstrate that only productive HPV integration sites were nonrandomly distributed across both viral and host genomes, suggesting that productive integration sites are under selection and likely to contribute to CC pathophysiology. Furthermore, using large-scale, multi-omics (clinical, genomic, transcriptional, proteomic, phosphoproteomic, and single-cell) data, we demonstrate that tumors with productive HPV integration are associated with higher E6/E7 proteins and enhanced tumor aggressiveness and immunoevasion. Importantly, productive HPV integration increases from carcinoma in situ to advanced disease. This study improves our understanding of the functional consequences of HPV fusion transcripts on the biology and pathophysiology of HPV-driven CCs, suggesting that productive HPV integration should be evaluated as an indicator of high risk for progression to aggressive cancers.

13.
Cell Rep ; 41(13): 111890, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36577369

RESUMEN

Ovarian cancer (OC) remains a clinical challenge for its difficulty in early diagnosis and insensitivity to treatments. Gut microbiota modulate multiple carcinoma progression through immunoregulation. The relationship between OC and gut microbiota has not been fully characterized. We find that the feces of patients with OC demonstrate different characteristics from benign controls. After fecal microbiota transplantation (FMT) from patients with OC into OC-bearing mice, the tumor development accelerates. Further, an Akkermansia supplementation with FMT significantly suppresses OC progression in mice. RNA sequencing of tumors shows that T cell activation pathways are upregulated after Akkermansia supplementation with FMT. Moreover, acetate accumulation accompanies Akkermansia abundance elevation, which is associated with enhanced interferon γ (IFNγ) secretion of CD8+ T cells and also its tumor-killing property. This work highlights the importance of protective gut microbiome in immune surveillance of OC, which connects accumulation of acetate and the cytotoxic function of CD8+ T cells by increasing IFNγ secretion.


Asunto(s)
Trasplante de Microbiota Fecal , Neoplasias Ováricas , Ratones , Animales , Femenino , Humanos , Akkermansia , Linfocitos T CD8-positivos , Heces , Neoplasias Ováricas/terapia , Suplementos Dietéticos
14.
J Exp Med ; 219(1)2022 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-34825915

RESUMEN

Targeted therapies represent attractive combination partners with immune checkpoint blockade (ICB) to increase the population of patients who benefit or to interdict the emergence of resistance. We demonstrate that targeting WEE1 up-regulates immune signaling through the double-stranded RNA (dsRNA) viral defense pathway with subsequent responsiveness to immune checkpoint blockade even in cGAS/STING-deficient tumors, which is a typical phenotype across multiple cancer types. WEE1 inhibition increases endogenous retroviral elements (ERVs) expression by relieving SETDB1/H3K9me3 repression through down-regulating FOXM1. ERVs trigger dsRNA stress and interferon response, increasing recruitment of anti-tumor T cells with concurrent PD-L1 elevation in multiple tumor models. Furthermore, combining WEE1 inhibition and PD-L1 blockade induced striking tumor regression in a CD8+ T cell-dependent manner. A WEE1 inhibition-induced viral defense signature provides a potentially informative biomarker for patient selection for combination therapy with WEE1 and ICB. WEE1 inhibition stimulates anti-tumor immunity and enhances sensitivity to ICB, providing a rationale for the combination of WEE1 inhibitors and ICB in clinical trials.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Retrovirus Endógenos/genética , Neoplasias Experimentales/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , ARN Bicatenario/genética , Transducción de Señal/efectos de los fármacos , Células A549 , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Retrovirus Endógenos/metabolismo , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Pirazoles/administración & dosificación , Pirazoles/farmacología , Pirimidinonas/administración & dosificación , Pirimidinonas/farmacología , ARN Bicatenario/metabolismo , Transducción de Señal/genética , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Carga Tumoral/inmunología
15.
Cancer Lett ; 528: 31-44, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34942306

RESUMEN

The CDK4/6 inhibitor, palbociclib has recently entered clinic-trial stage for breast cancer treatment. However, translating its efficacy to other solid tumors has been challenging, especially for aggressive solid tumors. We found that the effect of palbociclib as a single agent was limited due to primary and acquired resistance in multiple ovarian cancer (OC) models. Among these, patient-derived organoid and xenograft models are two most representative models of drug responsiveness in patients with OC. In preclinical models, this study demonstrated that activated MAPK/PI3K-AKT pathway and cell cycle-related proteins induced the resistance to palbociclib, which was overcome by the addition of the bromodomain protein 4 (BRD4) inhibitor AZD5153. Moreover, this study revealed that AZD5153 and palbociclib had a synergistic lethal effect on inducing the cell cycle arrest and increasing apoptosis, even in RB-deficient cell lines. Based on these results, it is anticipated that this class of drugs, including AZD5153, which inhibit the cell cycle-related protein and MAPK/PI3K-AKT pathway, will exhibit synergistic effects with palbociclib in OC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proteínas de Ciclo Celular/efectos de los fármacos , Compuestos Heterocíclicos con 2 Anillos/uso terapéutico , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/uso terapéutico , Piridazinas/uso terapéutico , Piridinas/uso terapéutico , Animales , Antineoplásicos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Femenino , Compuestos Heterocíclicos con 2 Anillos/farmacología , Humanos , Ratones , Piperazinas/farmacología , Pirazoles/farmacología , Piridazinas/farmacología , Piridinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cell Rep Med ; 3(12): 100856, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36543113

RESUMEN

Tumor-infiltrating lymphocytes (TILs), especially CD8+ TILs, represent a favorable prognostic factor in high-grade serous ovarian cancer (HGSOC) and other tumor lineages. Here, we analyze the spatial heterogeneity of different TIL subtypes in HGSOC. We integrated RNA sequencing, whole-genome sequencing, bulk T cell receptor (TCR) sequencing, as well as single-cell RNA/TCR sequencing to investigate the characteristics and differential composition of TILs across different HGSOC sites. Two immune "cold" patterns in ovarian cancer are identified: (1) ovarian lesions with low infiltration of mainly dysfunctional T cells and immunosuppressive Treg cells and (2) omental lesions infiltrated with non-tumor-specific bystander cells. Exhausted CD8 T cells that are preferentially enriched in ovarian tumors exhibit evidence for expansion and cytotoxic activity. Inherent tumor immune microenvironment characteristics appear to be the main contributor to the spatial differences in TIL status. The landscape of spatial heterogeneity of TILs may inform potential strategies for therapeutic manipulation in HGSOC.


Asunto(s)
Quistes Ováricos , Neoplasias Ováricas , Humanos , Femenino , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Pronóstico , Multiómica , Receptores de Antígenos de Linfocitos T/genética , Microambiente Tumoral/genética
17.
Adv Sci (Weinh) ; 9(21): e2105469, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35619328

RESUMEN

Targeting the G2/M checkpoint mediator WEE1 has been explored as a novel treatment strategy in ovarian cancer, but mechanisms underlying its efficacy and resistance remains to be understood. Here, it is demonstrated that the WEE1 inhibitor AZD1775 induces endoplasmic reticulum stress and activates the protein kinase RNA-like ER kinase (PERK) and inositol-required enzyme 1α (IRE1α) branches of the unfolded protein response (UPR) in TP53 mutant (mtTP53) ovarian cancer models. This is facilitated through NF-κB mediated senescence-associated secretory phenotype. Upon AZD1775 treatment, activated PERK promotes apoptotic signaling via C/EBP-homologous protein (CHOP), while IRE1α-induced splicing of XBP1 (XBP1s) maintains cell survival by repressing apoptosis. This leads to an encouraging synergistic antitumor effect of combining AZD1775 and an IRE1α inhibitor MKC8866 in multiple cell lines and preclinical models of ovarian cancers. Taken together, the data reveal an important dual role of the UPR signaling network in mtTP53 ovarian cancer models in response to AZD1775 and suggest that inhibition of the IRE1α-XBP1s pathway may enhance the efficacy of AZD1775 in the clinics.


Asunto(s)
Endorribonucleasas , Neoplasias Ováricas , Proteínas Serina-Treonina Quinasas , Benzopiranos , Endorribonucleasas/antagonistas & inhibidores , Endorribonucleasas/metabolismo , Femenino , Humanos , Inositol/metabolismo , Morfolinas , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Pirazoles/farmacología , Pirimidinonas/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Respuesta de Proteína Desplegada/genética , Respuesta de Proteína Desplegada/fisiología
18.
Aging (Albany NY) ; 13(23): 24943-24962, 2021 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-34862879

RESUMEN

Ongoing pandemic and potential resurgence of Coronavirus disease 2019 (COVID-19) has prompted urgent efforts to investigate the immunological memory of convalescent patients, especially in patients with active cancers. Here we performed single-cell RNA sequencing in peripheral blood samples of 3 healthy donors (HDs), 4 COVID-19 patients (Covs) and 4 COVID-19 patients with active gynecological tumor (TCs) pre- and post- anti-tumor treatment. All Covs patients had recovered from their acute infection. Interestingly, the molecular features of PBMCs in TCs are similar to that in Covs, suggesting that convalescent COVID-19 with gynecologic tumors do not have major immunological changes and may be protected against reinfection similar to COVID-19 patients without tumors. Moreover, the chemotherapy given to these patients mainly caused neutropenia, while having little effect on the proportion and functional phenotype of T and B cells, and T cell clonal expansion. Notably, anti-PD-L1 treatment massively increased cytotoxic scores of NK cells, and T cells, and facilitated clonal expansion of T cells in these patients. It is likely that T cells could protect patients from SARS-CoV-2 virus reinfection and anti-PD-L1 treatment can enhance the anti-viral activity of the T cells.


Asunto(s)
COVID-19/complicaciones , Neoplasias de los Genitales Femeninos/complicaciones , Neoplasias de los Genitales Femeninos/terapia , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Anticuerpos Antivirales/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , COVID-19/inmunología , COVID-19/prevención & control , Femenino , Neoplasias de los Genitales Femeninos/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Análisis de la Célula Individual , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
19.
Aging (Albany NY) ; 13(5): 7589-7607, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33658399

RESUMEN

Vulvovaginal atrophy (VVA) is a common menopause-related symptom affecting more than 50% of midlife and older women and cancer patients whose ovarian function are lost or damaged. Regardless of estrogen deficiency, whether other factors such as the gut microbiota play role in VVA have not been thoroughly investigated. To this end, we performed ovariectomy on 12-weeks' old mice and follow-up at 4 weeks after ovariectomy, and observed atrophied vagina and an altered gut microbiota in ovariectomized mice.. We further performed fecal microbiota transplantation with feces from another cohort of ovary-intact fecund female mice to the ovariectomized ones, and found that the vaginal epithelial atrophy was significantly alleviated as well as the gut microbiota was pointedly changed. All these results suggest that ovarian activity has some influence on the gut microbiota, and the latter from the ovary-intact female mice can somehow make the vagina of mice deficient in ovarian function healthier maybe by up-expressing ESR1 in vaginal cells and enhancing regeneration in vagina. This kind of association between gut microbiota and vaginal health need further exploration such that it may provide an alternative treatment by modulating gut microbiota in patients suffering from VVA but may be reluctant to hormone therapy.


Asunto(s)
Trasplante de Microbiota Fecal , Ovariectomía/efectos adversos , Vagina/patología , Animales , Atrofia , Femenino , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética
20.
Cancer Res ; 81(10): 2714-2729, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33589518

RESUMEN

Mutant KRAS tumors are associated with poor outcomes, at least in part, due to decreased therapeutic sensitivity. Here, we show that KRAS mutations are associated with resistance to monotherapy and combination therapy with PARP inhibitors (PARPi) and immune checkpoint blockade with anti-PD-L1 antibodies. In mutant KRAS tumors, inhibition of KRAS signaling with MEK inhibitors (MEKi) triggered and amplified PARPi-induced DNA damage, cytosolic double-stranded DNA accumulation, STING pathway activation, and CD8+ T-cell recruitment. Moreover, MEKi decreased myeloid-derived suppressor cell infiltration, in part, by inhibiting IL6 and GMCSF production. Importantly, addition of MEKi to PARPi and anti-PD-L1 resulted in marked tumor inhibition in immunocompetent mutant KRAS tumor models. This study provides the underlying mechanistic data to support evaluation of PARPi, MEKi, and anti-PD-L1 combination in clinical trials of mutant KRAS tumors. SIGNIFICANCE: This study provides key insights into the potential for using MEKi combined with PARPi and anti-PD-L1 for the treatment of all mutant KRAS tumors. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/10/2714/F1.large.jpg.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Apoptosis , Antígeno B7-H1/antagonistas & inhibidores , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Quimioterapia Combinada , Femenino , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Mutación , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA