Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.207
Filtrar
1.
J Am Chem Soc ; 146(37): 25462-25466, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39240652

RESUMO

Near-infrared (NIR) aggregation-induced emission luminogens (AIEgens) are excellent probes for tumor imaging, but there still is space to improve their imaging specificity and sensitivity. In this work, a strategy of tandem targeting and dual aggregation of an AIEgen is proposed to achieve these two purposes. An AIEgen, ß-tBu-Ala-Cys(StBu)-Lys(Biotin)-Pra(QMT)-CBT (Ala-Biotin-QMT), is designed to tandem target the biotin receptor and leucine aminopeptidase of a cancer cell and thereafter undergo CBT-Cys click reaction-mediated dual aggregations in the cell. Experimental results show that Ala-Biotin-QMT renders 4.8-fold and 7.9-fold higher NIR fluorescence signals over those in the "biotin + LAP inhibitor"-treated control groups in living HepG2 cells and HepG2 tumor-bearing mice, respectively. We anticipate that Ala-Biotin-QMT, which has the tandem targeting and dual aggregation property to simultaneously achieve enhanced tumor enrichment and fluorescence onset, could be applied for accurate cancer diagnosis in the clinic in the future.


Assuntos
Corantes Fluorescentes , Imagem Óptica , Humanos , Animais , Camundongos , Células Hep G2 , Corantes Fluorescentes/química , Corantes Fluorescentes/síntese química , Raios Infravermelhos , Leucil Aminopeptidase/metabolismo , Biotina/química , Neoplasias/diagnóstico por imagem , Receptores de Fatores de Crescimento
2.
Hippocampus ; 34(9): 491-502, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39091158

RESUMO

Hippocampal area CA2 has garnered attention in recent times owing to its significant involvement in social memory and distinctive plasticity characteristics. Research has revealed that the CA2 region demonstrates a remarkable resistance to plasticity, particularly in the Schaffer Collateral (SC)-CA2 pathway. In this study we investigated the role of Nogo-A, a well-known axon growth inhibitor and more recently discovered plasticity regulator, in modulating plasticity within the CA2 region. The findings demonstrate that blocking Nogo-A in male rat hippocampal slices facilitates the establishment of both short-term and long-term plasticity in the SC-CA2 pathway, while having no impact on the Entorhinal Cortical (EC)-CA2 pathway. Additionally, the study reveals that inhibiting Nogo-A enables association between the SC and EC pathways. Mechanistically, we confirm that Nogo-A operates through its well-known co-receptor, p75 neurotrophin receptor (p75NTR), and its downstream signaling factor such as Rho-associated protein kinase (ROCK), as their inhibition also allows plasticity induction in the SC-CA2 pathway. Additionally, the induction of long-term depression (LTD) in both the EC and SC-CA2 pathways led to persistent LTD, which was not affected by Nogo-A inhibition. Our study demonstrates the involvement of Nogo-A mediated signaling mechanisms in limiting synaptic plasticity within the CA2 region.


Assuntos
Região CA2 Hipocampal , Plasticidade Neuronal , Proteínas Nogo , Sinapses , Animais , Proteínas Nogo/metabolismo , Masculino , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Região CA2 Hipocampal/fisiologia , Região CA2 Hipocampal/metabolismo , Região CA2 Hipocampal/efeitos dos fármacos , Ratos Sprague-Dawley , Ratos , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Córtex Entorrinal/fisiologia , Córtex Entorrinal/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Vias Neurais/fisiologia , Proteínas da Mielina/metabolismo , Proteínas da Mielina/genética , Proteínas do Tecido Nervoso , Receptores de Fatores de Crescimento
3.
Transgenic Res ; 33(4): 255-266, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38981975

RESUMO

The p75NTR neurotrophin receptor has positive and negative roles regulating cell survival in the nervous system. Unambiguous interpretation of p75NTR function in vivo has been complicated, however, by residual expression of alternate forms of p75NTR protein in initial p75NTR knock-out mouse models. As rats are the preferred rodent for studying brain and behaviour, and to simplify interpretation of the knock-out phenotype, we report here the generation of a mutant rat devoid of the p75NTR protein. TALEN-mediated recombination in embryonic stem cells (ESCs) was used to flank exon 2 of p75NTR with Lox P sites and produce transgenic rats carrying either un-recombined floxed p75NTREx2-fl, or recombined, exon-2 deleted p75NTREx2-Δ alleles. Crossing p75NTREx2-fl rats with a Cre-deleter strain efficiently removed exon 2 in vivo. Excision of exon 2 causes a frameshift after p75NTR Gly23 and eliminated p75NTR protein expression. Rats lacking p75NTR were healthy, fertile, and histological analysis did not reveal significant changes in cellular density or overall structure in their brains. p75NTR function is therefore largely dispensable for normal development, growth and basal homeostasis in the rat. However, the availability of constitutive and conditional p75NTREx2-Δ rats provides new opportunities to investigate specific roles of p75NTR upon injury and during tissue repair.


Assuntos
Ratos Transgênicos , Animais , Ratos , Receptores de Fator de Crescimento Neural/genética , Receptores de Fator de Crescimento Neural/metabolismo , Fertilidade/genética , Feminino , Encéfalo/metabolismo , Encéfalo/crescimento & desenvolvimento , Masculino , Éxons/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Embrionárias/metabolismo , Receptores de Fatores de Crescimento
4.
CNS Neurosci Ther ; 30(7): e14875, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39072998

RESUMO

BACKGROUND: Angiogenesis is crucial in neuroprotection of secondary thalamic injury after cortical infarction. The p75 neurotrophin receptor (p75NTR) plays a key role in activating angiogenesis. However, the effects of p75NTR on angiogenesis in the thalamus after cortical infarction are largely unknown. Herein we investigate whether p75NTR facilitates angiogenesis to attenuate secondary thalamic damage via activating hypoxia-inducible factor 1α (HIF-1α)/vascular endothelial growth factor (VEGF) pathway mediated by Von Hippel-Lindau (VHL) after distal middle cerebral artery occlusion (dMCAO). METHODS: The male rat model of dMCAO was established. The effects of p75NTR on the angiogenesis was evaluated using RNA-sequencing, immunohistochemistry, western blot, quantitative real-time polymerase chain reaction, magnetic resonance imaging, behavior tests, viral and pharmacological interventions. RESULTS: We found that the p75NTR and vessel density were decreased in ipsilateral thalamus after dMCAO. The p75NTR-VHL interaction was reduced, which promoted the ubiquitination degradation of HIF-1α and reduced VEGF expression after dMCAO. Notably, p75NTR overexpression restrained the ubiquitination degradation of HIF-1α by inhibiting VHL-HIF-1α interaction, further promoted angiogenesis, increased cerebral blood flow of ipsilateral thalamus and improved neurological function after dMCAO. CONCLUSION: For the first time, we highlighted that the enhancement of p75NTR-VHL interaction promoted angiogenesis in attenuating secondary thalamic damage after dMCAO.


Assuntos
Infarto da Artéria Cerebral Média , Neovascularização Fisiológica , Ratos Sprague-Dawley , Tálamo , Animais , Masculino , Ratos , Tálamo/metabolismo , Tálamo/patologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Neovascularização Fisiológica/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Receptores de Fator de Crescimento Neural/metabolismo , Receptores de Fator de Crescimento Neural/genética , Infarto Cerebral/patologia , Angiogênese , Proteínas do Tecido Nervoso , Receptores de Fatores de Crescimento
5.
Cell Commun Signal ; 22(1): 319, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858728

RESUMO

Several different signaling pathways that regulate cell proliferation and differentiation are initiated by binding of ligands to cell-surface and membrane-bound enzyme-linked receptors, such as receptor tyrosine kinases and serine-threonine kinases. They prompt phosphorylation of tyrosine and serine-threonine residues and initiate downstream signaling pathways and priming of intracellular molecules that convey the signal in the cytoplasm and nucleus, with transcriptional activation of specific genes enriching cell growth and survival-related cascades. These cell processes are rhythmically driven by molecular clockworks endowed in every cell type and when deregulated play a crucial role in cancer onset and progression. Growth factors and their matching receptor-dependent signaling are frequently overexpressed and/or dysregulated in many cancer types. In this review we focus on the interplay between biological clocks and Growth Factor Receptor-dependent signaling in the context of carcinogenesis.


Assuntos
Carcinogênese , Transdução de Sinais , Humanos , Carcinogênese/metabolismo , Carcinogênese/genética , Carcinogênese/patologia , Animais , Receptores de Fatores de Crescimento/metabolismo , Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/genética
6.
Int J Mol Sci ; 25(11)2024 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-38891978

RESUMO

Binge alcohol consumption during adolescence can produce lasting deficits in learning and memory while also increasing the susceptibility to substance use disorders. The adolescent intermittent ethanol (AIE) rodent model mimics human adolescent binge drinking and has identified the nucleus basalis magnocellularis (NbM) as a key site of pathology. The NbM is a critical regulator of prefrontal cortical (PFC) cholinergic function and attention. The cholinergic phenotype is controlled pro/mature neurotrophin receptor activation. We sought to determine if p75NTR activity contributes to the loss of cholinergic phenotype in AIE by using a p75NTR modulator (LM11A-31) to inhibit prodegenerative signaling during ethanol exposure. Male and female rats underwent 5 g/kg ethanol (AIE) or water (CON) exposure following 2-day-on 2-day-off cycles from postnatal day 25-57. A subset of these groups also received a protective dose of LM11A-31 (50 mg/kg) during adolescence. Rats were trained on a sustained attention task (SAT) and behaviorally relevant acetylcholine (ACh) activity was recorded in the PFC with a fluorescent indicator (AChGRAB 3.0). AIE produced learning deficits on the SAT, which were spared with LM11A-31. In addition, PFC ACh activity was blunted by AIE, which LM11A-31 corrected. Investigation of NbM ChAT+ and TrkA+ neuronal expression found that AIE led to a reduction of ChAT+TrkA+ neurons, which again LM11A-31 protected. Taken together, these findings demonstrate the p75NTR activity during AIE treatment is a key regulator of cholinergic degeneration.


Assuntos
Acetilcolina , Neurônios Colinérgicos , Etanol , Córtex Pré-Frontal , Animais , Feminino , Masculino , Ratos , Acetilcolina/metabolismo , Atrofia , Comportamento Animal/efeitos dos fármacos , Neurônios Colinérgicos/metabolismo , Neurônios Colinérgicos/efeitos dos fármacos , Modelos Animais de Doenças , Etanol/toxicidade , Proteínas do Tecido Nervoso , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Ratos Sprague-Dawley , Receptores de Fatores de Crescimento , Receptores de Fator de Crescimento Neural/metabolismo
7.
Front Biosci (Landmark Ed) ; 29(5): 184, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38812320

RESUMO

This review article explores the intricate correlation between growth factors and bone metastases, which play a crucial role in the development of several types of malignancies, namely breast, prostate, lung, and renal cancers. The focal point of our discussion is on crucial receptors for growth factors, including Epidermal Growth Factor Receptor (EGFR), Transforming Growth Factor-ß (TGFß), Vascular Endothelial Growth Factor Receptor (VEGFR), and Fibroblast Growth Factor Receptor (FGFR). These receptors, which are essential for cellular activities including growth, differentiation, and survival, have important involvement in the spread of cancer and the interactions between tumors and the bone environment. We discuss the underlying mechanisms of bone metastases, with a specific emphasis on the interaction between growth factor receptors and the bone microenvironment. EGFR signaling specifically enhances the process of osteoclast development and the formation of osteolytic lesions, especially in breast and lung malignancies. TGFß receptors have a role in both osteolytic and osteoblastic metastases by releasing TGFß, which attracts cancer cells and promotes bone remodeling. This is a crucial element in the spread of prostate cancer to the bones. The functions of FGFR and VEGFR in the processes of bone formation and tumor angiogenesis, respectively, highlight the complex and diverse nature of these interactions. The review emphasizes the possibility of targeted therapeutics targeting these receptors to interrupt the cycle of tumor development and bone degradation. Therapeutic approaches include focusing on the VEGF/VEGFR, EGF/EGFR, FGF/FGFR, and TGFß/TGFßR pathways. These include a variety of compounds, such as small molecule inhibitors and monoclonal antibodies, which have shown potential to interfere with tumor-induced alterations in bone. The text discusses clinical trials and preclinical models, offering insights into the effectiveness and constraints of various treatments. Ultimately, this study provides a succinct but thorough summary of the present knowledge and treatment strategies focused on growth factor receptors in bone metastases. This highlights the significance of comprehending the signaling of growth factor receptors in the microenvironment where tumors spread to the bones, as well as the possibility of using targeted therapies to enhance the results for cancer patients with bone metastases. The advancement of treating bone metastases hinges on the development of treatments that specifically target the intricate relationships between malignancies and bone.


Assuntos
Neoplasias Ósseas , Humanos , Neoplasias Ósseas/secundário , Neoplasias Ósseas/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Receptores ErbB/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Animais , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
8.
Biomed Pharmacother ; 175: 116615, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38663101

RESUMO

Estrogens play a critical role in the initiation and progression of breast cancer. Estrogen receptor (ER)α, ERß, and G protein-coupled estrogen receptor are the primary receptors for estrogen in breast cancer. These receptors are mainly activated by binding with estrogens. The crosstalk between ERs and membrane growth factor receptors creates additional pathways that amplify the effects of their ligands and promote tumor growth. This crosstalk may cause endocrine therapy resistance in ERα-positive breast cancer. Furthermore, this may explain the resistance to anti-human epidermal growth factor receptor-2 (HER2) treatment in ERα-/HER2-positive breast cancer and chemotherapy resistance in triple-negative breast cancer. Accordingly, it is necessary to understand the complex crosstalk between ERs and growth factor receptors. In this review, we delineate the crosstalk between ERs and membrane growth factor receptors in breast cancer. Moreover, this review highlights the current progress in clinical treatment and discusses how pharmaceuticals target the crosstalk. Lastly, we discuss the current challenges and propose potential solutions regarding the implications of targeting crosstalk via pharmacological inhibition. Overall, the present review provides a landscape of the crosstalk between ERs and membrane growth factor receptors in breast cancer, along with valuable insights for future studies and clinical treatments using a chemotherapy-sparing regimen to improve patient quality of life.


Assuntos
Neoplasias da Mama , Receptores de Estrogênio , Humanos , Receptores de Estrogênio/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Animais , Receptor Cross-Talk/efeitos dos fármacos , Receptores de Fatores de Crescimento/metabolismo , Receptores de Fatores de Crescimento/antagonistas & inibidores , Terapia de Alvo Molecular , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos/uso terapêutico , Antineoplásicos/farmacologia
9.
Lung Cancer ; 190: 107529, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38452600

RESUMO

OBJECTIVES: Osimertinib is largely used as first-line therapy for metastatic epithelial growth factor receptor (EGFR) mutant lung cancers based on the FLAURA clinical trial. Real-world patient outcomes often differ from clinical trial outcomes. This study evaluated the efficacy of first-line osimertinib in patients treated in British Columbia (BC), Canada. Furthermore, we compared the outcomes of patients who would and would not have been eligible for the original FLAURA trial. METHODS: Consecutive patients receiving first-line osimertinib for metastatic EGFR exon19 or L858R lung cancer were identified using the BC Cancer Pharmacy Database. Patient eligibility for the FLAURA clinical trial were retrospectively reviewed based on the following criteria: ECOG ≥ 2, symptomatic brain metastases or on steroids, hemoglobin < 90 g/L, platelets < 100x109/L, or a creatinine clearance < 50 mL/min. mOS was assessed for the entire population and compared between patients who would have been eligible and ineligible for FLAURA. RESULTS: From January 2020 to October 2021, 311 patients received first-line osimertinib; 44 % (137/311) were deemed FLAURA ineligible, predominantly due to low ECOG (n = 120). After a median follow-up of 26.5 months, the mOS for the entire cohort was 27.4 months (95 %CI 23.8-30.1). The mOS for ineligible patients was 18 months shorter than eligible patients (15.8 vs 34.2, p < 0.001). Ineligible patients had higher rates of de novo stage IV disease, higher rates of stage IVB disease, and more sites of disease than eligible patients. CONCLUSION: In this real-world population, nearly half of patients would have been ineligible for FLAURA. The mOS was one year shorter than reported in FLAURA. However, patients who would have been eligible for the FLAURA clinical trial had similar OS to patients enrolled in FLAURA. Trial ineligible patients had a higher burden of disease at baseline which may have led to inferior outcomes. Further research is needed to improve outcomes in these patients.


Assuntos
Acrilamidas , Carcinoma Pulmonar de Células não Pequenas , Indóis , Neoplasias Pulmonares , Pirimidinas , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/induzido quimicamente , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Estudos Retrospectivos , Inibidores de Proteínas Quinases/efeitos adversos , Receptores ErbB/genética , Compostos de Anilina/uso terapêutico , Compostos de Anilina/efeitos adversos , Receptores de Fatores de Crescimento/uso terapêutico , Mutação/genética
10.
Traffic ; 25(1): e12928, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38272447

RESUMO

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant disorder affecting 1 in 5000-8000 individuals. Hereditary hemorrhagic telangiectasia type 1 (HHT1) is the most common HHT and manifests as diverse vascular malformations ranging from mild symptoms such as epistaxis and mucosal and cutaneous telangiectases to severe arteriovenous malformations (AVMs) in the lungs, brain or liver. HHT1 is caused by heterozygous mutations in the ENG gene, which encodes endoglin, the TGFß homodimeric co-receptor. It was previously shown that some endoglin HHT1-causing variants failed to traffic to the plasma membrane due to their retention in the endoplasmic reticulum (ER) and consequent degradation by ER-associated degradation (ERAD). Endoglin is a homodimer formed in the ER, and we therefore hypothesized that mixed heterodimers might form between ER-retained variants and WT protein, thus hampering its maturation and trafficking to the plasma membrane causing dominant negative effects. Indeed, HA-tagged ER-retained mutants formed heterodimers with Myc-tagged WT endoglin. Moreover, variants L32R, V105D, P165L, I271N and C363Y adversely affected the trafficking of WT endoglin by reducing its maturation and plasma membrane localization. These results strongly suggest dominant negative effects exerted by these ER-retained variants aggravating endoglin loss of function in patients expressing them in the heterozygous state with the WT allele. Moreover, this study may help explain some of the variability observed among HHT1 patients due to the additional loss of function exerted by the dominant negative effects in addition to that due to haploinsufficiency. These findings might also have implications for some of the many conditions impacted by ERAD.


Assuntos
Telangiectasia Hemorrágica Hereditária , Humanos , Alelos , Endoglina/genética , Retículo Endoplasmático/metabolismo , Mutação , Receptores de Superfície Celular/genética , Receptores de Fatores de Crescimento , Telangiectasia Hemorrágica Hereditária/genética , Telangiectasia Hemorrágica Hereditária/diagnóstico , Telangiectasia Hemorrágica Hereditária/metabolismo
11.
Anticancer Drugs ; 35(1): 76-80, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37067984

RESUMO

Lung cancer is the malignancy with the highest morbidity and mortality worldwide. Approximately 60% of non-small cell lung cancer (NSCLC) presents driver alterations most of which are targetable. Nowadays, limited clinical data are available regarding the efficacy of epithelial growth factor receptor (EGFR) tyrosine kinase inhibitors in patients with NSCLC harboring uncommon EGFR mutations, considering their heterogeneity. Herein, we report a rare case of EGFR-mutated lung adenocarcinoma which has developed into squamous cell carcinoma with uncommon EGFR (Ex18) compound mutations and phosphatidylinositol 3-kinase mutation receiving afatinib at the forefront.


Assuntos
Adenocarcinoma de Pulmão , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Inibidores de Proteínas Quinases/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Receptores ErbB/genética , Mutação , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Receptores de Fatores de Crescimento/genética
12.
Cells ; 12(23)2023 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-38067157

RESUMO

The corneal epithelium is the first anatomical barrier between the environment and the cornea; it is critical for proper light refraction onto the retina and prevents pathogens (e.g., bacteria, viruses) from entering the immune-privileged eye. Trauma to the highly innervated corneal epithelium is extremely painful and if not resolved quickly or properly, can lead to infection and ultimately blindness. The healthy eye produces its own growth factors and is continuously bathed in tear fluid that contains these proteins and other nutrients to maintain the rapid turnover and homeostasis of the ocular surface. In this article, we review the roles of growth factors in corneal epithelial homeostasis and regeneration and some of the limitations to their use therapeutically.


Assuntos
Córnea , Epitélio Corneano , Receptores de Fatores de Crescimento , Transdução de Sinais , Peptídeos e Proteínas de Sinalização Intercelular , Homeostase
15.
JCO Precis Oncol ; 7: e2200422, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37487150

RESUMO

PURPOSE: Activating mutations in KRAS, NRAS, and BRAF are known to cause resistance to anti-epidermal growth factor receptor (EGFR) therapy; however, only approximately 40% of patients with colorectal cancer (CRC) with RASWT tumors respond to anti-EGFR treatment. We sought to discover novel biomarkers to predict response to anti-EGFR antibody treatment in CRC and to understand mechanisms of resistance to anti-EGFR therapy. MATERIALS AND METHODS: Transcriptomic profiles from three clinical and two preclinical cohorts treated with cetuximab were used to assign consensus molecular subtypes (CMS) to each sample and correlated with outcomes. RESULTS: Restricting to RASWT patients, we observed that CMS2 tumors (canonical subtype) had significantly higher response rates relative to other CMS when treated with cetuximab combination with doublet chemotherapy (Okita et al cohort: 92% disease control rate (DCR) for CMS2, chi-square P = .04; CALGB/SWOG 80405 cohort: 90% objective response rate (ORR) for CMS2, chi-square P < .001) and with single-agent cetuximab (68%, chi-square P = .01). CMS2 tumors showed best response among right-sided (ORR = 80%) and left-sided (ORR = 92%) tumors in the CALGB/SWOG 80405 cohort. CMS2 cells lines were most likely to be sensitive to cetuximab (60%) and CMS2 patient-derived xenograft had the highest DCR (84%). We found Myc, E2F, and mammalian target of rapamycin pathways were consistently upregulated in resistant samples (enrichment score >1, false discovery rate <0.25). Inhibitors of these pathways in resistant cell lines exhibited additive effects with cetuximab. CONCLUSION: These data suggest that CRC transcriptional profiles, when used to assign CMS, provide additional ability to predict response to anti-EGFR therapy relative to using tumor sidedness alone. Notably both right-sided and left-sided CMS2 tumors had excellent response, suggesting that anti-EGFR therapy be included as a treatment option for right-sided CMS2 tumors.


Assuntos
Neoplasias Colorretais , Humanos , Cetuximab/farmacologia , Cetuximab/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Receptores de Fatores de Crescimento/uso terapêutico
16.
Cell Biochem Biophys ; 81(3): 395-408, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37395856

RESUMO

Several decades of research and clinical trials have conclusively provided proof of concept on the usefulness of monoclonal antibodies in the armamentarium against cancer. There are numerous mAbs approved for both, the treatment of solid tumors as well as hematological malignancies. These have ranked in the top ten best-selling drugs in recent years and one such mAb, pembrolizumab, is slated to be the highest revenue-generating drug by 2024. A large proportion of the mAbs in oncology have been approved by regulatory agencies in just the past decade and many professionals working in the field have been unable to keep abreast with the latest mAbs available and their mechanism of action. In this review, we aim to provide a systematic compilation of the various mAbs in oncology, approved by the US FDA in the past decade. It also elaborates on the mechanism of action of the newly approved mAbs to provide an overall update of the same. For this purpose, we have referred to the Drugs at FDA and relevant articles from PubMed from the year 2010 to date.


Assuntos
Antineoplásicos Imunológicos , Neoplasias , Antineoplásicos Imunológicos/metabolismo , Antineoplásicos Imunológicos/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Antígenos de Neoplasias/metabolismo , Microambiente Tumoral , Receptores de Fatores de Crescimento/antagonistas & inibidores , Antígenos CD/metabolismo , Proteínas de Checkpoint Imunológico/metabolismo , Animais
17.
Artigo em Inglês | MEDLINE | ID: mdl-37316420

RESUMO

OBJECTIVE: The aim of this study was to report a case series of patients with metastatic colorectal cancer (mCRC) undergoing panitumumab-containing regimens affected by oral lesions and to review the current literature. STUDY DESIGN: Electronic medical records of mCRC patients referred to treat mouth sores during the treatment with the anti-epithelial growth factor receptor (EGFR)-panitumumab-were retrospectively reviewed. Patients' characterization, clinical profile of oral lesions, and management outcomes were documented. Additionally, modifications or discontinuation of the antineoplastic treatment as well as the occurrence of other adverse events (AEs) were analyzed. RESULTS: A total of 7 patients were included. The oral lesions appeared in a median time of 10 days (range 7-11 days) after the drug administration. The median reported pain score was 5 (range 1-9), causing feeding discomfort. Oral lesions with a marked aphthous-like appearance, among others, occurred in all cases and involved nonkeratinized mucosa more likely. At least 1 patient had dose reduction of the treatment and 1 patient needed discontinuation due to panitumumab-associated stomatitis. Dermatologic AEs were the most prevalent. Clinical improvement was obtained with topical corticosteroid therapy and/or photobiomodulation. CONCLUSIONS: In summary, panitumumab-containing regimens were associated with a particular pattern of oral lesions consistent with stomatitis. This event may eventually affect the tolerability of the treatment in patients with mCRC.


Assuntos
Neoplasias Colorretais , Estomatite , Humanos , Panitumumabe/uso terapêutico , Anticorpos Monoclonais , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Estudos Retrospectivos , Receptores ErbB/metabolismo , Receptores de Fatores de Crescimento/uso terapêutico , Estomatite/induzido quimicamente , Estomatite/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica
18.
Int J Oncol ; 62(6)2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37165911

RESUMO

We previously reported that radiotherapy­resistant (RT­R) triple negative breast cancer (TNBC) cells upregulate the expression of endothelial­specific molecule­1 (ESM­1) compared with TNBC cells. In addition, ESM­1 is involved in an increased proliferation and invasion of RT­R­TNBC cells compared with TNBC cells. It was further identified that, in RT­R­TNBC cells, P2Y2 purinergic receptor (P2Y2R)­mediated activation of p21­activated kinase 1 (PAK1), protein kinase C (PKC), c­Jun N­terminal kinase (JNK) and p38 MAPKs is related to ESM­1 expression via forkhead box O1 (FoxO1) regulation. Notably, it has been reported that P2Y2R mediates the transactivation of vascular epithelial growth factor receptor 2 (VEGFR2), and VEGFR2 is known to be involved in ESM­1 expression. Therefore, in the present study, the involvement of VEGFR2 in the P2Y2R­mediated ESM­1 upregulation in RT­R­TNBC cells and the relationship between P2Y2R and VEGFR2 activation was further examined. Western blotting and reverse transcription­PCR were used to monitor the expression of ESM­1, and the results demonstrated that extracellular ATP treatment regulated the expression of ESM­1 in a P2Y2R­dependent manner in RT­R­MDA­MB­231 cells. In addition, extracellular ATP activated Src and VEGFR2 after 5 min of incubation, which was abolished by knockdown of P2Y2R expression. VEGFR2 activation in response to ATP was also decreased by inhibiting Src activity, suggesting that ATP­activated P2Y2R regulates VEGFR2 phosphorylation via Src activation. Furthermore, ATP­induced ESM­1 expression was decreased by transfection with VEGFR2 small interfering RNA (siRNA). ESM­1­related signaling molecules, PAK1, PKC, JNK and p38 MAPKs, and the transcriptional regulator, FoxO1, which were activated by ATP, were also decreased following transfection with VEGFR2 siRNA. These results suggest that P2Y2R­mediated transactivation of VEGFR2 through Src phosphorylation is associated with ESM­1 overexpression in RT­R­TNBC cells.


Assuntos
Receptores Purinérgicos P2Y2 , Neoplasias de Mama Triplo Negativas , Receptor 2 de Fatores de Crescimento do Endotélio Vascular , Humanos , Trifosfato de Adenosina/metabolismo , Fosforilação , Proteína Quinase C/metabolismo , Receptores de Fatores de Crescimento/metabolismo , RNA Interferente Pequeno/metabolismo , Ativação Transcricional , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/radioterapia , Receptores Purinérgicos P2Y2/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
Sci Rep ; 13(1): 6639, 2023 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-37095146

RESUMO

Endoglin (ENG) is a single-pass transmembrane protein highly expressed on vascular endothelial cells, although low expression levels can be detected in many other cell types. Its extracellular domain can be found in circulation known as soluble endoglin (sENG). Levels of sENG are elevated in many pathological conditions, in particular preeclampsia. We have shown that while loss of cell surface ENG decreases BMP9 signaling in endothelial cells, knocking down ENG in blood cancer cells enhances BMP9 signaling. Despite sENG binding to BMP9 with high affinity and blocking the type II receptor binding site on BMP9, sENG did not inhibit BMP9 signaling in vascular endothelial cells, but the dimeric form of sENG inhibited BMP9 signaling in blood cancer cells. Here we report that in non-endothelial cells such as human multiple myeloma cell lines and the mouse myoblast cell line C2C12, both monomeric and dimeric forms of sENG inhibit BMP9 signaling when present at high concentrations. Such inhibition can be alleviated by the overexpression of ENG and ACVRL1 (encoding ALK1) in the non-endothelial cells. Our findings suggest that the effects of sENG on BMP9 signaling is cell-type specific. This is an important consideration when developing therapies targeting the ENG and ALK1 pathway.


Assuntos
Células Endoteliais , Receptores de Fatores de Crescimento , Camundongos , Gravidez , Animais , Feminino , Humanos , Endoglina/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Fosforilação , Ligação Proteica , Células Endoteliais/metabolismo , Receptores de Activinas Tipo II/metabolismo
20.
J Control Release ; 357: 333-341, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37028450

RESUMO

Pulmonary delivery offers a non-invasive route for the administration of biotherapeutics. In this context, understanding and control of a transport into, and across cellular barriers is central to the design of delivery systems. Here, we report our study on receptor mediated delivery of protein cargo by a formulation comprising sub-300 nm sized non-covalent protein complexes with biotin-conjugated PEG-poly(glutamic acid) (biotin-PEG2k-b-GA10) and PEG2k-b-GA30 copolymers blend as targeting and complexing functionalities. Designed complexes achieve intracellular delivery of the cargo in lung derived A549 epithelial cells in vitro via sodium-dependent multivitamin transporter (biotin receptor). We further show that biotin receptor driven endocytosis preferentially involves dynamin- and caveolae-dependent vesicular internalization, switching the transport pathway away from predominantly clathrin-dependent entry of free protein. Significantly for a protective intracellular delivery of biotherapeutics based on non-covalent complexation with polymeric excipients, the study provides evidence of intracellular presence of the complexing copolymer; demonstrated exploiting biotin in biotin-PEG2k-b-GA10 copolymer as a tag for binding with fluorescently labelled avidin. Moreover, analysis of intracellular localization of constitutive species shortly following cellular internalization suggests a co-localization of biotin-PEG2k-b-GA10 copolymer and protein constitutive species. The study demonstrates intracellular delivery of biotin targeted non-covalent complexes with a protein cargo, the result with important implications in a design of enabling technology platforms for protective, receptor mediated intracellular delivery of biotherapeutics.


Assuntos
Biotina , Receptores de Fatores de Crescimento , Biotina/química , Peptídeos , Avidina , Endocitose
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA