Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Cell Physiol ; 323(4): C1325-C1332, 2022 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-36094434

RESUMO

Chemotherapy is a common therapy to treat patients with breast cancer but also leads to skeletal muscle deconditioning. Skeletal muscle deconditioning is multifactorial and intermuscular adipose tissue (IMAT) accumulation is closely linked to muscle dysfunction. To date, there is no clinical study available investigating IMAT development through a longitudinal protocol and the underlying mechanisms remain unknown. Our study was dedicated to investigating IMAT content in patients with early breast cancer who were treated with chemotherapy and exploring the subsequent cellular mechanisms involved in its development. We included 13 women undergoing chemotherapy. Muscle biopsies and ultrasonography assessment were performed before and after chemotherapy completion. Histological and Western blotting analyses were conducted. We found a substantial increase in protein levels of three mature adipocyte markers (perilipin, +901%; adiponectin, +135%; FABP4, +321%; P < 0.05). These results were supported by an increase in oil red O-positive staining (+358%; P < 0.05). A substantial increase in PDGFRα protein levels was observed (+476%; P < 0.05) highlighting an increase in fibro-adipogenic progenitors (FAPs) content. The cross-sectional area of the vastus lateralis muscle fibers substantially decreased (-21%; P < 0.01), and muscle architecture was altered, as shown by a decrease in fascicle length (-15%; P < 0.05) and a decreasing trend in muscle thickness (-8%; P = 0.08). We demonstrated both IMAT development and muscle atrophy in patients with breast cancer who were treated with chemotherapy. FAPs, critical stem cells inducing both IMAT development and skeletal muscle atrophy, also increased, suggesting that FAPs likely play a critical role in the skeletal muscle deconditioning observed in patients with breast cancer who were treated with chemotherapy.


Assuntos
Neoplasias da Mama , Adiponectina/metabolismo , Tecido Adiposo/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Músculo Esquelético/metabolismo , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/diagnóstico por imagem , Atrofia Muscular/metabolismo , Perilipinas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo
2.
J Proteome Res ; 19(1): 292-299, 2020 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-31679342

RESUMO

Meningiomas are in most cases benign brain tumors. The WHO 2016 classification defines three grades of meningiomas. This classification had a prognosis value because grade III meningiomas have a worse prognosis value compared to grades I and II meningiomas. However, some benign or atypical meningiomas can have a clinical aggressive behavior. There are currently no reliable markers which allow distinguishing between the meningiomas with a good prognosis and those which may recur. High-resolution magic angle spinning (HRMAS) spectrometry is a noninvasive method able to determine the metabolite profile of a tissue sample. We retrospectively analyzed 62 meningioma samples by using HRMAS spectrometry (43 metabolites). We described a metabolic profile defined by a high concentration for acetate, threonine, N-acetyl-lysine, hydroxybutyrate, myoinositol, ascorbate, scylloinositol, and total choline and a low concentration for aspartate, glucose, isoleucine, valine, adenosine, arginine, and alanine. This metabolomic signature was associated with poor prognosis histological markers [Ki-67 ≥ 40%, high histological grade and negative progesterone receptor (PR) expression]. We also described a similar metabolomic spectrum between grade III and grade I meningiomas. Moreover, all grade I meningiomas with a low Ki-67 expression and a positive PR expression did not have the same metabolomic profile. Metabolomic analysis could be used to determine an aggressive meningioma in order to discuss a personalized treatment. Further studies are needed to confirm these results and to correlate this metabolic profile with survival data.


Assuntos
Neoplasias Encefálicas/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Meningioma/metabolismo , Aminoácidos/análise , Aminoácidos/metabolismo , Biópsia , Neoplasias Encefálicas/cirurgia , Proliferação de Células , Humanos , Antígeno Ki-67/metabolismo , Meningioma/patologia , Meningioma/cirurgia , Metabolômica/métodos
3.
World J Surg Oncol ; 17(1): 70, 2019 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-30992070

RESUMO

BACKGROUND: Meningioma is the most common adult primary intracranial tumor. Malignant meningioma is a rare variant of meningioma. The prognosis for the patients with these tumors is poor, due to the tumor's capacity for relapse and to develop distant metastases. These tumors can present the same evolutionary course as aggressive carcinoma. CASE DESCRIPTION: We report the case of distant brain and gastro-intestinal tract (GIT) metastases. A 78-year-old patient developed malignant meningioma with a Ki-67 proliferative index of 40%. According to guidelines, surgery followed by postoperative radiotherapy (RT) was performed. Three months after the end of RT, he presented histologically proven meningioma distant brain and GIT metastases. CONCLUSIONS: To our knowledge, this is the first case of meningioma GIT metastases. Also, we report the difficulty to confirm the diagnosis of meningioma metastases. Indeed, malignant meningioma has the same histopathological features as melanoma or carcinoma. The standard of care for the management of malignant meningioma is gross total surgery followed by postoperative radiotherapy. Metastatic meningioma is uncommon and no guidelines for the management of recurrent or metastatic meningioma have yet been published. However, several studies reported systemic therapeutic options such as antibody against VEGF, somatostatin analogs, PDGF-R, and VEGF-R tyrosine kinase inhibitors, in the case of recurrent or metastatic meningioma. We also made a review of the actual literature of systemic treatment options for metastatic meningioma.


Assuntos
Neoplasias Encefálicas/secundário , Neoplasias Gastrointestinais/secundário , Neoplasias Meníngeas/patologia , Meningioma/patologia , Idoso , Neoplasias Encefálicas/terapia , Neoplasias Gastrointestinais/terapia , Humanos , Masculino , Neoplasias Meníngeas/terapia , Meningioma/terapia , Gradação de Tumores
5.
J Cachexia Sarcopenia Muscle ; 15(1): 292-305, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38183352

RESUMO

BACKGROUND: Breast cancer patients are commonly treated with sequential administrations of epirubicin-cyclophosphamide (EC) and paclitaxel (TAX). The chronic effect of this treatment induces skeletal muscle alterations, but the specific effect of each chemotherapy agent is unknown. This study aimed to investigate the effect of EC or TAX administration on skeletal muscle homeostasis in breast cancer patients. METHODS: Twenty early breast cancer patients undergoing EC followed by TAX chemotherapies were included. Two groups of 10 women were established and performed vastus lateralis skeletal muscle biopsies either before the first administration (pre) of EC (50 ± 14 years) or TAX (50 ± 16 years) and 4 days later (post). Mitochondrial respiratory capacity recording, reactive oxygen species production, western blotting and histological analyses were performed. RESULTS: Decrease in muscle fibres cross-sectional area was only observed post-EC (-25%; P < 0.001), associated with a reduction in mitochondrial respiratory capacity for the complex I (CI)-linked substrate state (-32%; P = 0.001), oxidative phosphorylation (OXPHOS) by CI (-35%; P = 0.002), CI&CII (-26%; P = 0.022) and CII (-24%; P = 0.027). If H2 O2 production was unchanged post-EC, an increase was observed post-TAX for OXPHOS by CII (+25%; P = 0.022). We found a decrease in makers of mitochondrial content, as shown post-EC by a decrease in the protein levels of citrate synthase (-53%; P < 0.001) and VDAC (-39%; P < 0.001). Despite no changes in markers of mitochondrial fission, a decrease in the expression of a marker of mitochondrial inner-membrane fusion was found post-EC (OPA1; -60%; P < 0.001). We explored markers of mitophagy and found reductions post-EC in the protein levels of PINK1 (-63%; P < 0.001) and Parkin (-56%; P = 0.005), without changes post-TAX. An increasing trend in Bax protein level was found post-EC (+96%; P = 0.068) and post-TAX (+77%; P = 0.073), while the Bcl-2 level was decreased only post-EC (-52%; P = 0.007). If an increasing trend in TUNEL-positive signal was observed post-EC (+68%; P = 0.082), upregulation was highlighted post-TAX (+86%; P < 0.001), suggesting activation of the apoptosis process. CONCLUSIONS: We demonstrated that a single administration of EC induced, in only 4 days, skeletal muscle atrophy and mitochondrial alterations in breast cancer patients. These alterations were characterized by reductions in mitochondrial function and content as well as impairment of mitochondrial dynamics and an increase in apoptosis. TAX administration did not worsen these alterations as this group had already received EC during the preceding weeks. However, it resulted in an increased apoptosis, likely in response to the increased H2 O2 production.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/metabolismo , Mitocôndrias/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Complexo I de Transporte de Elétrons/metabolismo , Apoptose
6.
Transl Lung Cancer Res ; 12(11): 2342-2346, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-38090529

RESUMO

Background: Anaplastic lymphoma kinase (ALK) rearrangement confers sensitivity to second- and third-generation ALK inhibitors, which have become the standard of care for ALK-positive non-small cell lung carcinoma (NSCLC). However, primary resistance to these inhibitors remains a rare and poorly understood phenomenon, especially in cases involving kinesin light chain 1 (KLC1)/ALK-rearranged metastatic NSCLC. Case Description: In this report, we present a unique and challenging case of primary resistance to second- and third-generation ALK tyrosine kinase inhibitors (TKIs) attributed to KLC1/ALK gene fusion partners in a patient with ALK-positive pleural metastatic NSCLC. The patient's disease progression was rapid and unresponsive to multiple lines of ALK-targeted therapies, including alectinib, brigatinib, and lorlatinib, underscoring the need for a deeper understanding of primary resistance mechanisms in such cases. Conclusions: The occurrence of primary resistance to ALK inhibitors in metastatic NSCLC with ALK rearrangement is an infrequent occurrence, and its underlying mechanisms remain elusive. This case emphasizes the importance of further research to elucidate the specific mechanisms of primary resistance to ALK TKIs in non-canonical ALK fusion partners like KLC1. Developing targeted therapies for such rare cases is a clinical challenge that warrants continued investigation and innovation in the field of precision oncology.

7.
Clin Nucl Med ; 48(2): 176-178, 2023 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-36607366

RESUMO

ABSTRACT: A falcine meningioma was diagnosed in a 66-year-old woman and was treated by surgery and 2 times by radiotherapy during 9 years of follow-up with the diagnosis of atypical meningioma. Three months after the last radiotherapy, incidental liver lesions were detected on chest CT realized for suspected pneumonia. In view of the predisposing factors for meningioma metastases, 68Ga-DOTATOC hepatic and cerebral PET/MRI was performed and completed by total body PET/CT demonstrating a somatostatin receptor 2 overexpression of the multiple liver lesions and several bone lesions. Biopsies from the liver and iliac bone confirmed the metastatic origin of meningioma.


Assuntos
Neoplasias Hepáticas , Neoplasias Meníngeas , Meningioma , Compostos Organometálicos , Feminino , Humanos , Idoso , Meningioma/diagnóstico , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Neoplasias Meníngeas/diagnóstico , Compostos Radiofarmacêuticos , Tomografia por Emissão de Pósitrons , Octreotida , Neoplasias Hepáticas/diagnóstico por imagem
8.
Ann Transl Med ; 11(5): 223, 2023 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-37007549

RESUMO

Background: Osimertinib is approved in first line metastatic epidermal growth factor receptor (EGFR) mutated non-small cell lung cancer (NSCLC). Acquired EGFR L718V mutation is a rare mechanism of resistance towards osimertinib in L858R+ NSCLC with potential sensibility to afatinib. This case reported an acquired EGFR L718V/TP53 V727M resistance co-mutation to osimertinib with discordant molecular pattern between plasmatic and cerebral fluid in a leptomeningeal and bone metastatic EGFR L858R mutant NSCLC. Case Description: A 52-year-old female, diagnosed with a bone metastatic EGFR L858R-mutated NSCLC, was treated with osimertinib as second line treatment for a leptomeningeal progression. She developed an acquired EGFR L718V/TP53 V272M resistance co-mutation after seventeen months of treatment. Discordant molecular status was observed between plasmatic (L718V+/TP53+/L858R+) and cerebrospinal fluid (CSF) (L718V-/TP53+/L858R+). Afatinib as third line did not prevent neurological progression. Conclusions: Acquired EGFR L718V mutation mediate a rare mechanism of resistance to osimertinib. Some cases reported sensibility to afatinib in patients with EGFR L718V mutation. In this described case, afatinib had no efficacy against neurological progression. This could be explained by the absence of EGFR L718V mutation in CSF tumor cells and concomitant TP53 V272M mutation as negative survival prognostic. Identify resistance mechanisms against osimertinib and develop specific therapeutic approaches remain a challenge in clinical routine.

9.
Cancers (Basel) ; 15(20)2023 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-37894307

RESUMO

Lung cancers with ALK rearrangement represent less than 5% of all lung cancers. ALK inhibitors are currently used to treat first-line metastatic non-small cell lung cancer with ALK rearrangement. Compared to chemotherapy, ALK inhibitors have improved progression-free survival, overall survival, and quality of life for patients. The results of several phase 3 studies with a follow-up of over 6 years suggest that the life expectancy of these patients treated with targeted therapies is significantly higher than 5 years and could approach 10 years. Nevertheless, these treatments induce haematological toxicities, including neutropenia. Few data are available on neutropenia induced by ALK inhibitors and on the pathophysiological mechanism and therapeutic adaptations necessary to continue the treatment. Given the high efficacy of these treatments, managing side effects to avoid treatment interruptions is essential. Here, we have reviewed the data from published clinical studies and case reports to provide an overview of neutropenia induced by ALK inhibitors.

10.
J Neuroimmune Pharmacol ; 17(3-4): 377-379, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-34687000

RESUMO

Immune checkpoint inhibitors (ICIs) have led to a revolution in cancer management, mainly due to lasting long-term durable responses in a subset of patients with metastatic solid tumours (Gettinger et al. in JCO 36(17):1675-1684, 2018). As immunotherapy is gradually being applied for the treatment of a large range of solid tumours, the incidence of neurological immune-related adverse events (irAEs) has increased (2). Neurologic toxicities that result in high morbidity rates and even mortality have emerged as serious complications of ICIs (Johnson et al. in J Immuno Cancer 7(1):134, 2019; Wang et al. in JAMA Oncol 4(12):1721, 2018). Small-cell lung cancer (SCLC) is common cause of neurologic paraneoplastic syndrome (Sebastian et al. in J Thorac Oncol 14(11):1878-1880, 2019). Nevertheless, the distinction between neurologic iRAEs and paraneoplastic neurological syndromes (PNSs) in patients with SCLC treated by ICIs remains challenging (Williams et al. JAMA Neurol 73(8):928, 2016). As immunotherapy is gradually being applied for the treatment of a large range of solid tumours, the incidence of neurological autoimmune adverse events has increased. Neurologic toxicities that result in high morbidity rates and even mortality have emerged as serious complications of ICIs and have yet to be fully understood. We report a case of an immune induced cerebellar ataxia in a 47 year-old small-cell neuroendocrine carcinoma patient undergoing checkpoint blockade by atezolizumab, a programmed cell death-1 ligand (PDL-1) inhibitor. After 4 cycles of immunotherapy, the patient presented with kinetic and static cerebellar syndrome leading to the diagnosis of TRIM9-Abs ICI-related cerebellar irAE. Therapeutic management was discussed in multidisciplinary meetings in the lack of therapeutic guidelines. There was no clinical improvement. Because of high morbidity and no treatment evidence, neurologic symptoms developing under ICI require early diagnosis and may indicate the need for definitive treatment discontinuation.


Assuntos
Ataxia Cerebelar , Neoplasias , Humanos , Pessoa de Meia-Idade , Ataxia Cerebelar/induzido quimicamente , Ataxia Cerebelar/terapia , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Imunoterapia/efeitos adversos
11.
J Cachexia Sarcopenia Muscle ; 13(3): 1896-1907, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35373507

RESUMO

BACKGROUND: Chemotherapy is extensively used to treat breast cancer and is associated with skeletal muscle deconditioning, which is known to reduce patients' quality of life, treatment efficiency, and overall survival. To date, skeletal muscle mitochondrial alterations represent a major aspect explored in breast cancer patients; nevertheless, the cellular mechanisms remain relatively unknown. This study was dedicated to investigating overall skeletal muscle mitochondrial homeostasis in early breast cancer patients undergoing chemotherapy, including mitochondrial quantity, function, and dynamics. METHODS: Women undergoing (neo)adjuvant anthracycline-cyclophosphamide and taxane-based chemotherapy participated in this study (56 ± 12 years). Two muscle biopsies were collected from the vastus lateralis muscle before the first and after the last chemotherapy administration. Mitochondrial respiratory capacity, reactive oxygen species production, and western blotting analyses were performed. RESULTS: Among the 11 patients, we found a decrease in key markers of mitochondrial quantity, reaching -52.0% for citrate synthase protein levels (P = 0.02) and -38.2% for VDAC protein levels (P = 0.04). This mitochondrial content loss is likely explained by reduced mitochondrial biogenesis, as evidenced by a decrease in PGC-1α1 protein levels (-29.5%; P = 0.04). Mitochondrial dynamics were altered, as documented by a decrease in MFN2 protein expression (-33.4%; P = 0.01), a key marker of mitochondrial outer membrane fusion. Mitochondrial fission is a prerequisite for mitophagy activation, and no variation was found in either key markers of mitochondrial fission (Fis1 and DRP1) or mitophagy (Parkin, PINK1, and Mul1). Two contradictory hypotheses arise from these results: defective mitophagy, which probably increases the number of damaged and fragmented mitochondria, or a relative increase in mitophagy through elevated mitophagic potential (Parkin/VDAC ratio; +176.4%; P < 0.02). Despite no change in mitochondrial respiratory capacity and COX IV protein levels, we found an elevation in H2 O2 production (P < 0.05 for all substrate additions) without change in antioxidant enzymes. We investigated the apoptosis pathway and found an increase in the protein expression of the apoptosis initiation marker Bax (+72.0%; P = 0.04), without variation in the anti-apoptotic protein Bcl-2. CONCLUSIONS: This study demonstrated major mitochondrial alterations subsequent to chemotherapy in early breast cancer patients: (i) a striking reduction in mitochondrial biogenesis, (ii) altered mitochondrial dynamics and potential mitophagy defects, (iii) exacerbated H2 O2 production, and (iv) increased initiation of apoptosis. All of these alterations likely explain, at least in part, the high prevalence of skeletal muscle and cardiorespiratory deconditioning classically observed in breast cancer patients.


Assuntos
Neoplasias da Mama , Neoplasias da Mama/metabolismo , Feminino , Homeostase , Humanos , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Qualidade de Vida , Ubiquitina-Proteína Ligases/metabolismo
12.
Int J Cancer ; 129(11): 2744-55, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21538350

RESUMO

Withaferin A (WFA) is purified from the plant Withania somnifera and inhibits the vimentin cytoskeleton. Vimentin overexpression in cancer correlates with metastatic disease, induction of epithelial to mesenchymal transition and reduced patient survival. As vimentin functions in cell motility, we wanted to test the hypothesis that WFA inhibits cancer metastasis by disrupting vimentin function. These data showed that WFA had weak cytotoxic and apoptotic activity at concentrations less than or equal to 500 nM, but retained potent anti-invasive activity at these low doses. Imaging of breast cancer cell lines revealed that WFA induces perinuclear vimentin accumulation followed by rapid vimentin depolymerization. A concomitant induction of vimentin ser56 phosphorylation was observed, which is consistent with vimentin disassembly. Structure activity relationships were established using a set of chemically modified WFA analogs and showed that the predicted vimentin-binding region of WFA is necessary to induce vimentin ser56 phosphorylation and for its anti-invasive activity. Pharmacokinetic studies in mice revealed that WFA reaches peak concentrations up to 2 µM in plasma with a half-life of 1.36 hr following a single 4 mg/kg dose. In a breast cancer metastasis mouse model, WFA showed dose-dependent inhibition of metastatic lung nodules and induced vimentin ser56 phosphorylation, with minimal toxicity to lung tissue. Based upon these studies, we conclude that WFA is a potent breast cancer anti-metastatic agent and the anti-metastatic activity of WFA is, at least in part, mediated through its effects on vimentin and vimentin ser56 phosphorylation.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Movimento Celular/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Serina/química , Vimentina/metabolismo , Vitanolídeos/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Western Blotting , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Cancer ; 117(17): 4049-59, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21365629

RESUMO

BACKGROUND: Lonafarnib (LNF) is a protein farnesyl transferase (FTase) inhibitor that has shown synergistic activity with taxanes in preclinical models and early stage clinical trials. Preclinical findings suggested tubulin acetylation and FTase expression levels may be important determinants of drug sensitivity that would help identify patient populations more likely to benefit from this regimen. This pilot study evaluated the biological effects of LNF and docetaxel (DTX) combination therapy in refractory solid tumors by comparing pretreatment and post-treatment tumor biopsies. METHODS: Patients with histologically confirmed locally advanced or metastatic solid malignancies refractory to standard therapies or with no effective therapies available were eligible. Patients were randomized to 1 of 4 dosing cohorts: 1) 30 mg/m², 100 mg; 2) 36 mg/m², 100 mg; 3) 30 mg/m², 150 mg; or 4) 36 mg/m², 150 mg of DTX intravenously weekly, LNF orally twice daily, respectively. RESULTS: Of the 38 patients enrolled, 36 were treated, and 29 were evaluable for toxicity and response assessment. The combination of LNF and DTX was tolerated in all cohorts with the exception of a 28% incidence of grade 3/4 diarrhea, which was manageable with aggressive antidiarrheal regimens. Seven patients derived clinically meaningful benefit from this combination treatment; these patients had significantly lower basal FTase-beta mRNA expression levels than the mean study population level (P < .05). Correlation of clinical benefit with tubulin acetylation content as well as basal acetyl-tubulin content were evaluated. However, no significant correlation was found. CONCLUSIONS: Despite the small number of patients, these findings support our preclinical mechanistic studies and warrant further clinical investigations using FTase-beta mRNA expression as a potential predictive biomarker to select for an enriched patient population to study the effects of taxane and FTase inhibitor combination therapies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Farnesiltranstransferase/sangue , Neoplasias/tratamento farmacológico , Piperidinas/administração & dosagem , Piridinas/administração & dosagem , Taxoides/administração & dosagem , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Docetaxel , Inibidores Enzimáticos/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto
14.
Clin Nucl Med ; 46(10): e515-e517, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-33782316

RESUMO

ABSTRACT: Succinate dehydrogenase (SDH)-deficient gastrointestinal stromal tumors (GISTs) are associated with loss of function of SDH complex and represent 5% to 7.5% of GISTs. SDH-deficient GISTs usually develop in the stomach of children and young adults, and could be part of Carney triad or Carney-Stratakis syndromes including paraganglioma. SDH-deficient GISTs are often indolent despite the high rate of distant metastasis, and overall unresponsive to tyrosine kinase inhibitors. However, epigenetic inactivation of MGMT leads to potential effectiveness of alkylating agents. We report the 18F-FDG PET/CT results for monitoring response to TKI and alkylating drugs in a patient with refractory SDHB-deficient GIST.


Assuntos
Tumores do Estroma Gastrointestinal , Preparações Farmacêuticas , Alquilantes , Criança , Fluordesoxiglucose F18 , Tumores do Estroma Gastrointestinal/diagnóstico por imagem , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Tumores do Estroma Gastrointestinal/genética , Humanos , Mutação , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Inibidores de Proteínas Quinases/uso terapêutico , Succinato Desidrogenase/genética , Adulto Jovem
15.
Chin Clin Oncol ; 10(3): 26, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33752411

RESUMO

BACKGROUND: Atypical meningioma is a variant of meningioma with a high risk of recurrence. Gross total resection is the standard of treatment, while no consensus on optimal adjuvant management has been found. METHODS: Between 2008 and 2018, a retrospective search identified 216 grade II meningiomas treated in six centers. Clinical, histological, and therapeutic data were analyzed to determine the prognostic factors of recurrence and survival. RESULTS: In total, 216 patients underwent surgical resection. Among these, 122 patients (56%) underwent gross total resection, and 21% of the patients received adjuvant radiotherapy. Univariate analysis reported subtotal resection, high Ki-67, negative progesterone receptor (PR) and histological grade evolution as unfavorable prognosis factors. According to multivariate analysis, the Ki-67 proliferative index (cut-off value of 17.5%) was the only prognostic factor of recurrence (HR 1.1; 95% CI, 1.0-1.2, P=0.048). Gross total resection improved progression-free survival (PFS) (P=0.03) but without impact on overall survival (OS) (P=0.2). Median PFS and OS times were longer for patients receiving adjuvant radiotherapy than those who did not receive adjuvant radiotherapy. PFS (P=0.3) and OS (P=0.7) were associated with adjuvant RT by trend only. After a median follow-up time of 6.7 years, 99 (46%) patients relapsed. Median progression-free and OS rates were 4.5 (95% CI, 3.5-5.5) and 14.7 years (11.4-NA), respectively. CONCLUSIONS: In this study, Ki-67 proliferative index was significantly associated with recurrence. Gross total resection significantly improved PFS without impacting OS. Adjuvant radiotherapy delayed recurrence and improved OS, but a longer follow-up time is needed to distinguish a statistically significant difference. Large prospective studies are needed to determine postoperative treatment guidelines.


Assuntos
Neoplasias Meníngeas , Meningioma , Recidiva Local de Neoplasia , Humanos , Neoplasias Meníngeas/diagnóstico , Neoplasias Meníngeas/terapia , Meningioma/diagnóstico , Meningioma/terapia , Recidiva Local de Neoplasia/diagnóstico , Prognóstico , Radioterapia Adjuvante , Estudos Retrospectivos
16.
Cancer Imaging ; 20(1): 70, 2020 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-33023662

RESUMO

Contrast-enhanced magnetic resonance imaging is currently the standard of care in the management of primary brain tumors, although certain limitations remain. Metabolic imaging has proven useful for an increasing number of indications in oncology over the past few years, most particularly 18F-FDG PET/CT. In neuro-oncology, 18F-FDG was insufficient to clearly evaluate brain tumors. Amino-acid radiotracers such as 18F-FDOPA were then evaluated in the management of brain diseases, notably tumoral diseases. Even though European guidelines on the use of amino-acid PET in gliomas have been published, it is crucial that future studies standardize acquisition and interpretation parameters. The aim of this article was to systematically review the potential effect of this metabolic imaging technique in numerous steps of the disease: primary and recurrence diagnosis, grading, local and systemic treatment assessment, and prognosis. A total of 41 articles were included and analyzed in this review. It appears that 18F-FDOPA PET holds promise as an effective additional tool in the management of gliomas. More consistent prospective studies are still needed.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Glioma/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Di-Hidroxifenilalanina/análogos & derivados , Humanos , Tomografia por Emissão de Pósitrons/normas , Guias de Prática Clínica como Assunto , Compostos Radiofarmacêuticos
17.
Mol Cancer Ther ; 7(7): 1900-8, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18645000

RESUMO

The majority of patients with HER2-overexpressing metastatic breast cancer who initially respond to the HER2-targeted antibody trastuzumab show disease progression within 1 year. The identification of novel agents that effectively inhibit survival of cancer cells that have progressed on trastuzumab is critical for improving outcome for this patient population. In the current study, we show that the phenolic compound nordihydroguaiaretic acid (NDGA) promoted cell death of trastuzumab-naive and trastuzumab-refractory HER2-overexpressing breast cancer cells. NDGA induced DNA fragmentation, cleavage of poly(ADP-ribose) polymerase and caspase-3, and inhibition of colony formation. In addition, NDGA inhibited insulin-like growth factor-I and HER2 signaling in trastuzumab-refractory cells, with reduced downstream phosphatidylinositol-3 kinase/Akt signaling. Importantly, combination treatment with NDGA and trastuzumab suppressed proliferation and survival of trastuzumab-refractory cells to a greater degree than either agent alone, suggesting that NDGA increases the sensitivity of refractory cells to trastuzumab. Derivatives of NDGA are currently in clinical trial for other solid tumors. Our data strongly support further study of NDGA as a potential therapeutic against breast cancers that have progressed on trastuzumab.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Masoprocol/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Receptor IGF Tipo 1/antagonistas & inibidores , Anticorpos Monoclonais Humanizados , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trastuzumab , Resultado do Tratamento
18.
Ann Transl Med ; 7(5): 106, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31019956

RESUMO

Anaplastic lymphoma kinase inhibitors (ALKi) like ceritinib are considered standard for front-line treatment of non-small cell lung cancers (NSCLC) harboring a translocation of the anaplastic lymphoma kinase (ALK) gene. We report herein a case of interstitial lung disease (ILD) that developed following a 7-month ceritinib treatment without recurrence under either crizotinib or brigatinib, two others ALKi.

19.
Front Biosci ; 13: 3906-12, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18508484

RESUMO

The HER2 receptor tyrosine kinase is amplified and/or overexpressed in approximately 30 percent of metastatic breast cancers. Interactions and cross signaling from the HER2 receptor to other growth factor receptors may potentially contribute to therapeutic resistance. In this review, we discuss HER2 receptor cross talk with the estrogen receptor and implications toward resistance to endocrine therapies. We also review mechanisms of resistance to the HER2-targeted antibody trastuzumab, including signaling from other members of the HER family, increased signaling through the PI3-kinase pathway, and cross talk from the insulin-like growth factor-I receptor to HER2. Finally, we will provide perspective on how HER2 receptor cross talk may provide critical information for developing novel therapeutic options for HER2-overexpressing breast cancers.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Receptor Cross-Talk/fisiologia , Receptor ErbB-2/fisiologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Antineoplásicos/uso terapêutico , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/fisiopatologia , Linhagem Celular Tumoral , Feminino , Humanos , Fosfatidilinositol 3-Quinases/fisiologia , Receptor ErbB-2/antagonistas & inibidores , Receptor IGF Tipo 1/fisiologia , Receptores de Estrogênio/fisiologia , Transdução de Sinais , Tamoxifeno/uso terapêutico , Trastuzumab
20.
Mol Biol Cell ; 14(1): 67-77, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12529427

RESUMO

The adapter protein FADD consists of two protein interaction domains: a death domain and a death effector domain. The death domain binds to activated death receptors such as Fas, whereas the death effector domain binds to procaspase 8. An FADD mutant, which consists of only the death domain (FADD-DD), inhibits death receptor-induced apoptosis. FADD-DD can also activate a mechanistically distinct, cell type-specific apoptotic pathway that kills normal but not cancerous prostate epithelial cells. Here, we show that this apoptosis occurs through activation of caspases 9, 3, 6, and 7 and a serine protease. Simultaneous inhibition of caspases and serine proteases prevents FADD-DD-induced death. Inhibition of either pathway alone does not prevent cell death but does affect the morphology of the dying cells. Normal prostate epithelial cells require both the caspase and serine protease inhibitors to efficiently prevent apoptosis in response to TRAIL. In contrast, the serine protease inhibitor does not affect TRAIL-induced death in prostate tumor cells suggesting that the FADD-DD-dependent pathway can be activated by TRAIL. This apoptosis pathway is activated in a cell type-specific manner that is defective in cancer cells, suggesting that this pathway may be targeted during cancer development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose/fisiologia , Proteínas de Transporte/metabolismo , Caspases/metabolismo , Serina Endopeptidases/metabolismo , Adenoviridae , Proteínas Reguladoras de Apoptose , Epitélio/metabolismo , Proteína de Domínio de Morte Associada a Fas , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Masculino , Glicoproteínas de Membrana/metabolismo , Próstata/metabolismo , Estrutura Terciária de Proteína , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA