Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.142
Filtrar
1.
Lung Cancer ; 190: 107529, 2024 Apr.
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
2.
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
3.
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
4.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
Adv Healthc Mater ; 12(15): e2202989, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36740892

RESUMO

Invasion and metastasis are the leading causes of death of patients with CRC. 5-Fluorouracil is widely used in clinic practice as the basic chemotherapy drug for CRC. However, it is inefficient in inhibiting tumor metastasis. MicroRNA-10b is uninvolved in regulating the growth of primary tumors; however, it could induce early tumor metastases and is a key regulator of chemotherapeutic resistance to 5-FU. A multifunctional nanovehicle that can carry small molecule drugs not only through the hydrophobic pockets of conjugated ß-cyclodextrin but also through electrostatic interaction between the conjugated peptides and siRNA to target functional genes is previously developed. In this study, a nanovehicle, named GCD, with epithelium growth factor receptor (EGFR)-targeted characteristics to simultaneously deliver chemotherapeutic and nucleotide drugs to distinct targets in CRC, is employed. These data show that co-delivery of 5-FU and anti-miR-10b can be effectively applied to targeted therapy of EGFR-overexpressed CRC, particularly inhibiting the metastasis of CRC. Furthermore, the therapeutic effect of this combination on tumor xenograft models derived from patients with CRC is evaluated. Taken together, this study may provide insights into the inhibition of tumor growth and metastasis simultaneously.


Assuntos
MicroRNAs , Neoplasias , Humanos , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , MicroRNAs/metabolismo , Neoplasias/tratamento farmacológico , Receptores de Fatores de Crescimento , Receptores ErbB/genética , Receptores ErbB/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos
14.
Autophagy ; 19(3): 1034-1035, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-35980802

RESUMO

Aberrant growth factor receptor signaling is among the most common oncogenic drivers in cancer biology. Receptor signaling classically induces cancer growth through signaling cascades that mediate effects largely through transcriptional control. Recently, post-transcriptional RNA modifications, collectively designated as epitranscriptomics, have emerged as a critical layer of dysregulation in cancer biology. We recently reported that PDGFR (platelet-derived growth factor receptor) activity in cancer stem cells (CSCs) derived from glioblastoma patients displays increased post-transcriptional mRNA methylation (N6-methyladenosine [m6A]), which promotes CSC maintenance through regulation of mitophagy. Specifically, PDGF-PDGFRB signaling upregulates the expression of the m6A methyltransferase METTL3, which then decorates the mitophagy regulator OPTN (optineurin) mRNA with m6A, thereby promoting OPTN mRNA degradation. Glioblastomas express lower levels of OPTN than normal brain, and forced expression of OPTN reduces tumor growth, supporting a tumor suppressive role for OPTN. Pharmacological targeting of METTL3 with PDGFR or activation of mitophagy demonstrates a combinatorial benefit. Collectively, our results suggest that upstream regulation of mitophagy in lethal cancers is mediated through growth factor receptor control of post-transcriptional RNA regulation, offering novel therapeutic paradigms.


Assuntos
Autofagia , Mitofagia , Humanos , Transdução de Sinais , Receptores de Fatores de Crescimento , RNA Mensageiro/genética , Metiltransferases/metabolismo
15.
Turk J Gastroenterol ; 34(2): 118-127, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36445057

RESUMO

BACKGROUND: Rat sarcoma virus mutational status guides first-line treatment in metastatic colorectal cancer. This study was a multi center, multi-country ambispective, observational study in the Middle East and North Africa assessing regional rat sarcoma virus testing practices in newly diagnosed patients. METHODS: The retrospective arm (2011-2014) included adults with metastatic colorectal cancer who had initiated first-line therapy with ≥1 post-baseline visit and survival data. The prospective arm (2014-2019) enrolled newly diagnosed patients with histologically proven metastatic colorectal cancer with ≥1 measurable lesion per Response Evaluation Criteria in Solid Tumors, and tissue availability for biomarker analysis. Data look-back and follow-up were 2 years; the rate of RAS mutation was evaluated. RESULTS: RAS testing was ordered for patients in retrospective (326/417) and prospective (407/500) studies. In the former, testing was typically prescribed after first-line treatment initiation, significantly more in patients with stage IV disease (P < .005), resulting in the addition of targeted therapy (41.8% anti-epidermal growth factor receptor, 30.2% anti-vascular endothelial growth factor) in wild-type metastatic colorectal cancer, and significantly impacted the treatment of left-sided tumors (P = .037). In the latter, 58.4% were RAS wild-type; 41.6% were RAS mutant. Non-prescription of RAS testing was attributed to test unavailability, financial, or medical rea sons; predictors of testing prescription were older age, primary tumor in ascending colon, and high tumor grade. RAS status knowledge resulted in the addition of anti-vascular endothelial growth factor (20.4%) or anti-epidermal growth factor receptor therapy (21.2%). CONCLUSION: Before 2014, RAS testing in patients with colorectal cancer in the Middle East and North Africa was often performed after first-line treatment. Testing is more routine in newly diagnosed patients, potentially shifting early treatment patterns.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Neoplasias Retais , Humanos , Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias Colorretais/genética , Fatores de Crescimento Endotelial/genética , Fatores de Crescimento Endotelial/uso terapêutico , Mutação , Estudos Prospectivos , Proteínas Proto-Oncogênicas p21(ras)/genética , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento/uso terapêutico , Estudos Retrospectivos , Sistema de Registros
16.
Gan To Kagaku Ryoho ; 50(13): 1504-1506, 2023 Dec.
Artigo em Japonês | MEDLINE | ID: mdl-38303322

RESUMO

The patient was a 75-year-old man who had undergone potentially curative surgery for Stage Ⅲb rectal cancer followed by resection of liver metastases. Two years after the resection of liver metastases, lung and remnant liver metastases were found. He received chemotherapy for unresectable metastatic tumors. Based on the findings of molecular and pathological examinations(RAS: wild type; BRAF: wild type; MSI: negative; HER2: negative), the following chemotherapy regimens were administered: first-line, FOLFIRI plus panitumumab(PANI); second-line, mFOLFOX6; third-line, trifluridine/tipiracil; fourth- line, regorafenib. After fourth-line treatment, he was judged to have disease progression due to the increase in his lung and liver metastases and the elevation of tumor markers. All standard regimens were refractory, but the Eastern Cooperative Oncology Group performance status was zero and a liquid biopsy for RAS still showed wild type. Therefore, rechallenge therapy with anti-epidermal growth factor receptor(EGFR)drugs, cetuximab(CET)and irinotecan(IRI), was administered 13 months after the final course of FOLFIRI plus PANI treatment. After 4 courses of CET plus IRI, the size of the 2 metastatic tumors markedly decreased and his tumor marker levels normalized.


Assuntos
Neoplasias Hepáticas , Neoplasias Retais , Idoso , Humanos , Masculino , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cetuximab , Receptores ErbB , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/cirurgia , Neoplasias Hepáticas/secundário , Recidiva Local de Neoplasia/tratamento farmacológico , Receptores de Fatores de Crescimento/uso terapêutico , Neoplasias Retais/tratamento farmacológico , Neoplasias Retais/cirurgia , Neoplasias Retais/patologia
17.
J Exp Clin Cancer Res ; 41(1): 363, 2022 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-36578014

RESUMO

BACKGROUND: Sushi domain-containing protein 4 (SUSD4) is a recently discovered protein with unknown cellular functions. We previously revealed that SUSD4 can act as complement inhibitor and as a potential tumor suppressor. METHODS: In a syngeneic mouse model of breast cancer, tumors expressing SUSD4 had a smaller volume compared with the corresponding mock control tumors. Additionally, data from three different expression databases and online analysis tools confirm that for breast cancer patients, high mRNA expression of SUSD4 in the tumor tissue correlates with a better prognosis. In vitro experiments utilized triple-negative breast cancer cell lines (BT-20 and MDA-MB-468) stably expressing SUSD4. Moreover, we established a cell line based on BT-20 in which the gene for EGFR was knocked out with the CRISPR-Cas9 method. RESULTS: We discovered that the Epithelial Growth Factor Receptor (EGFR) interacts with SUSD4. Furthermore, triple-negative breast cancer cell lines stably expressing SUSD4 had higher autophagic flux. The initiation of autophagy required the expression of EGFR but not phosphorylation of the receptor. Expression of SUSD4 in the breast cancer cells led to activation of the tumor suppressor LKB1 and consequently to the activation of AMPKα1. Finally, autophagy was initiated after stimulation of the ULK1, Atg14 and Beclin-1 axis in SUSD4 expressing cells. CONCLUSIONS: In this study we provide novel insight into the molecular mechanism of action whereby SUSD4 acts as an EGFR inhibitor without affecting the phosphorylation of the receptor and may potentially influence the recycling of EGFR to the plasma membrane.


Assuntos
Neoplasias de Mama Triplo Negativas , Humanos , Animais , Camundongos , Neoplasias de Mama Triplo Negativas/metabolismo , Fosforilação , Receptores ErbB/genética , Receptores ErbB/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Autofagia , Linhagem Celular Tumoral
18.
Exp Clin Transplant ; 20(11): 1016-1021, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36524888

RESUMO

OBJECTIVES: Stem cells from human exfoliated deciduous teeth are a population of multipotent mesenchymal stem cells that can self-renew and actively secrete a broad spectrum of trophic and immunomodulatory factors.Brain-derivedneurotrophic factor is able to induce stem cells to neural differentiation to repair the nervous system. However,the mechanism ofbrain-derivedneurotrophic factor-induced neural differentiation in stem cells from human exfoliated deciduous teeth remains unclear. MATERIALS AND METHODS: In this study, we focused on brain-derived neurotrophic factor and investigated its effects on neural differentiation in stem cells from human exfoliated deciduous teeth. We cultured stem cells from human exfoliated deciduous teeth under various different brain-derived neurotrophic factor concentrations. We then analyzed the effects of brain-derived neurotrophic factor to the neural differen-tiation and associated signaling pathways in stem cells from human exfoliated deciduous teeth. RESULTS: We demonstrated that a high concentration of brain-derived neurotrophic factor could induce neural differentiation in stem cells from human exfoliated deciduous teeth, and brain-derived neurotrophic factor also increased the expression levels of neural differentiation-related proteins in stem cells from human exfoliated deciduous teeth, which was associated with the suppression of growth factor receptor-bound protein 2/extracellular signal-regulated kinase 1 and 2 signaling pathways. CONCLUSIONS: Knockdown of growth factor receptor- bound protein 2 inhibited the neural differentiation of stem cells from human exfoliated deciduous teeth via changes in growth factor receptor-bound protein 2/extracellular signal-regulated kinase signaling pathways, but this phenotype could be rescued by overexpression of extracellular signal-regulated kinase. Our findings suggested that brain-derived neurotrophic factor can regulate the differentiation of stem cells from human exfoliated deciduous teeth through the growth factor receptor-bound protein 2/extracellular signal-regulated kinase signaling pathway.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , MAP Quinases Reguladas por Sinal Extracelular , Humanos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/farmacologia , Dente Decíduo , Resultado do Tratamento , Células-Tronco , Diferenciação Celular , Transdução de Sinais , Receptores de Fatores de Crescimento/metabolismo , Células Cultivadas
19.
PLoS One ; 17(10): e0275648, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36206298

RESUMO

Dramatic improvements in cancer survival have occurred in the last decade, but the quality of life for many survivors is compromised due to severe, long-lasting, and often irreversible side effects of chemotherapy. The neurological side effects, chemotherapy induced peripheral neuropathy (CIPN) and cancer related/induced cognitive impairment (CRCI/CICI), are under-recognized and can occur after chemotherapy, immunotherapy, or radiation. The cellular mechanisms underlying these neurological side effects are poorly understood and there are no effective treatments or preventions, other than reduction or termination of cancer therapy. In our preliminary prospective, non-interventional study to examine the side effects of chemotherapy in patients with breast cancer (NCT03872141), patients with breast cancer who received standard of care single agent weekly taxane-based chemotherapy were assessed at baseline, midpoint, and end of treatment for neurological and cognitive changes and for blood levels of potential protein biomarkers (n = 13). CIPN and CRCI both showed an increase in severity with accumulating taxane and these changes were compared to protein alternations over the course of treatment. Using peripheral blood collected from patients (n = 10) during chemotherapy and tested with an antibody array curated by the MD Anderson RPPA Core), we found that 19 proteins were increased, and 12 proteins decreased over 12 weeks of treatment. Among those downregulate were proteins known to be critical for neuronal viability and function including GRB2 (growth factor receptor-bound protein 2) and NCS1 (neuronal calcium sensor 1). Concurrently, proteins associated with apoptosis, including BAK1 (Bcl-1 homologous antagonist/killer), were upregulated. These results support the proposal that CIPN and CRCI increase with increasing taxane exposure, and identified several proteins that are altered with taxane exposure that could be implicated in their pathogenesis. In conclusion, our study provides evidence for progressive neurological changes and the rationale to investigate the molecular basis for these changes with the goal of target identification for mitigation of these neurological side effects.


Assuntos
Antineoplásicos , Neoplasias da Mama , Doenças do Sistema Nervoso Periférico , Antineoplásicos/efeitos adversos , Neoplasias da Mama/patologia , Hidrocarbonetos Aromáticos com Pontes , Cognição , Feminino , Humanos , Estudos Prospectivos , Qualidade de Vida , Receptores de Fatores de Crescimento , Taxoides/efeitos adversos
20.
Biol Pharm Bull ; 45(10): 1531-1536, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36184512

RESUMO

Acne-like eruption caused by anti-epidermal growth factor receptor (EGFR) antibodies such as panitumumab reduces treatment adherence and patient QOL; an alternative therapy is desired. Meanwhile, the usefulness of oral Non-steroidal Anti-inflammatory Drugs (NSAIDs) for acne-like eruptions caused by low-molecular-weight EGFR inhibitors such as erlotinib has been reported in the treatment of lung cancer. This study aimed to investigate whether the combined use of oral NSAIDs and panitumumab for colorectal cancer patients helps prevent acne-like eruption. We retrospectively investigated 167 colorectal cancer patients who had been treated with panitumumab for three cycles or more. The observation period was set from the start of panitumumab treatment to the end of three cycles. Within this period, the incidence and severity of acne-like eruptions were compared. A total of 59 and 108 patients were in the NSAIDs use and non-use groups, respectively, showing differences in the incidence of acne-like eruption rates (78.0 vs. 90.7%, respectively; p = 0.033). In the use group, eruption severity grades 0, 1, 2, and 3 were observed in 13, 33, 13, and 0 patients, respectively; the corresponding values in the non-use group were 10, 60, 36, and 2, respectively (p = 0.007). Oral NSAIDs may help prevent acne-like eruptions caused by panitumumab.


Assuntos
Acne Vulgar , Neoplasias Colorretais , Acne Vulgar/induzido quimicamente , Acne Vulgar/tratamento farmacológico , Administração Oral , Anti-Inflamatórios/uso terapêutico , Anti-Inflamatórios não Esteroides/efeitos adversos , Anticorpos Monoclonais , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/tratamento farmacológico , Receptores ErbB , Cloridrato de Erlotinib/uso terapêutico , Humanos , Panitumumabe/uso terapêutico , Qualidade de Vida , Receptores de Fatores de Crescimento/uso terapêutico , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...