Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Exp Med ; 219(12)2022 12 05.
Article in English | MEDLINE | ID: mdl-36169652

ABSTRACT

Lung cancer remains the leading cause of cancer-related death worldwide. We identify DSTYK, a dual serine/threonine and tyrosine non-receptor protein kinase, as a novel actionable target altered in non-small cell lung cancer (NSCLC). We also show DSTYK's association with a lower overall survival (OS) and poorer progression-free survival (PFS) in multiple patient cohorts. Abrogation of DSTYK in lung cancer experimental systems prevents mTOR-dependent cytoprotective autophagy, impairs lysosomal biogenesis and maturation, and induces accumulation of autophagosomes. Moreover, DSTYK inhibition severely affects mitochondrial fitness. We demonstrate in vivo that inhibition of DSTYK sensitizes lung cancer cells to TNF-α-mediated CD8+-killing and immune-resistant lung tumors to anti-PD-1 treatment. Finally, in a series of lung cancer patients, DSTYK copy number gain predicts lack of response to the immunotherapy. In summary, we have uncovered DSTYK as new therapeutic target in lung cancer. Prioritization of this novel target for drug development and clinical testing may expand the percentage of NSCLC patients benefiting from immune-based treatments.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Serine , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/metabolism , Threonine , Tumor Necrosis Factor-alpha/metabolism , Tyrosine
2.
Cancer Lett ; 529: 70-84, 2022 03 31.
Article in English | MEDLINE | ID: mdl-34971753

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) play a major role in cancer progression. In this study, we investigated the mechanisms by which complement C5a increases the capacity of polymorphonuclear MDSCs (PMN-MDSCs) to promote tumor growth and metastatic spread. Stimulation of PMN-MDSCs with C5a favored the invasion of cancer cells via a process dependent on the formation of neutrophil extracellular traps (NETs). NETosis was dependent on the production of high mobility group box 1 (HMGB1) by cancer cells. Moreover, C5a induced the surface expression of the HMGB1 receptors TLR4 and RAGE in PMN-MDSCs. In a mouse lung metastasis model, inhibition of C5a, C5a receptor-1 (C5aR1) or NETosis reduced the number of circulating-tumor cells (CTCs) and the metastatic burden. In support of the translational relevance of these findings, C5a was able to stimulate migration and NETosis in PMN-MDSCs obtained from lung cancer patients. Furthermore, myeloperoxidase (MPO)-DNA complexes, as markers of NETosis, were elevated in lung cancer patients and significantly correlated with C5a levels. In conclusion, C5a induces the formation of NETs from PMN-MDSCs in the presence of cancer cells, which may facilitate cancer cell dissemination and metastasis.


Subject(s)
Complement C5a/immunology , Extracellular Traps/immunology , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Heterografts , Humans , Immunophenotyping , Mice , Models, Biological , Neoplasm Metastasis , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Receptor, Anaphylatoxin C5a/metabolism
4.
Cancer Med ; 7(7): 3474-3483, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29766673

ABSTRACT

Single nucleotide polymorphisms (SNPs) may modulate individual susceptibility to carcinogens. We designed a genome-wide association study to characterize individuals presenting extreme phenotypes of high and low risk to develop tobacco-induced non-small cell lung cancer (NSCLC), and we validated our results. We hypothesized that this strategy would enrich the frequencies of the alleles that contribute to the observed traits. We genotyped 2.37 million SNPs in 95 extreme phenotype individuals, that is: heavy smokers that either developed NSCLC at an early age (extreme cases); or did not present NSCLC at an advanced age (extreme controls), selected from a discovery set (n = 3631). We validated significant SNPs in 133 additional subjects with extreme phenotypes selected from databases including >39,000 individuals. Two SNPs were validated: rs12660420 (pcombined  = 5.66 × 10-5 ; ORcombined  = 2.80), mapping to a noncoding transcript exon of PDE10A; and rs6835978 (pcombined  = 1.02 × 10-4 ; ORcombined  = 2.57), an intronic variant in ATP10D. We assessed the relevance of both proteins in early-stage NSCLC. PDE10A and ATP10DmRNA expressions correlated with survival in 821 stage I-II NSCLC patients (p = 0.01 and p < 0.0001). PDE10A protein expression correlated with survival in 149 patients with stage I-II NSCLC (p = 0.002). In conclusion, we validated two variants associated with extreme phenotypes of high and low risk of developing tobacco-induced NSCLC. Our findings may allow to identify individuals presenting high and low risk to develop tobacco-induced NSCLC and to characterize molecular mechanisms of carcinogenesis and resistance to develop NSCLC.

5.
Cancer Lett ; 402: 43-51, 2017 08 28.
Article in English | MEDLINE | ID: mdl-28549790

ABSTRACT

Id1 promotes carcinogenesis and metastasis, and predicts prognosis of non-small cell lung cancer (NSCLC)-adenocarcionoma patients. We hypothesized that Id1 may play a critical role in lung cancer colonization of the liver by affecting both tumor cells and the microenvironment. Depleted levels of Id1 in LLC (Lewis lung carcinoma cells, LLC shId1) significantly reduced cell proliferation and migration in vitro. Genetic loss of Id1 in the host tissue (Id1-/- mice) impaired liver colonization and increased survival of Id1-/- animals. Histologically, the presence of Id1 in tumor cells of liver metastasis was responsible for liver colonization. Microarray analysis comparing liver tumor nodules from Id1+/+ mice and Id1-/- mice injected with LLC control cells revealed that Id1 loss reduces the levels of EMT-related proteins, such as vimentin. In tissue microarrays containing 532 NSCLC patients' samples, we found that Id1 significantly correlated with vimentin and other EMT-related proteins. Id1 loss decreased the levels of vimentin, integrinß1, TGFß1 and snail, both in vitro and in vivo. Therefore, Id1 enables both LLC and the host microenvironment for an effective liver colonization, and may represent a novel therapeutic target to avoid NSCLC liver metastasis.


Subject(s)
Carcinoma, Lewis Lung/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Inhibitor of Differentiation Protein 1/metabolism , Liver Neoplasms/metabolism , Lung Neoplasms/metabolism , Tumor Microenvironment , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/secondary , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/secondary , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Differentiation Protein 1/drug effects , Inhibitor of Differentiation Protein 1/genetics , Integrin beta1/genetics , Integrin beta1/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Time Factors , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Burden , Vimentin/genetics , Vimentin/metabolism
6.
Oncologist ; 21(2): 220-32, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26668254

ABSTRACT

UNLABELLED: Despite much investment and progress, oncology is still an area with significant unmet medical needs, with new therapies and more effective use of current therapies needed. The emergent field of pharmacometrics combines principles from pharmacology (pharmacokinetics [PK] and pharmacodynamics [PD]), statistics, and computational modeling to support drug development and optimize the use of already marketed drugs. Although it has gained a role within drug development, its use in clinical practice remains scarce. The aim of the present study was to review the principal pharmacometric concepts and provide some examples of its use in oncology. Integrated population PK/PD/disease progression models as part of the pharmacometrics platform provide a powerful tool to predict outcomes so that the right dose can be given to the right patient to maximize drug efficacy and reduce drug toxicity. Population models often can be developed with routinely collected medical record data; therefore, we encourage the application of such models in the clinical setting by generating close collaborations between physicians and pharmacometricians. IMPLICATIONS FOR PRACTICE: The present review details how the emerging field of pharmacometrics can integrate medical record data with predictive pharmacological and statistical models of drug response to optimize and individualize therapies. In order to make this routine practice in the clinic, greater awareness of the potential benefits of the field is required among clinicians, together with closer collaboration between pharmacometricians and clinicians to ensure the requisite data are collected in a suitable format for pharmacometrics analysis.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Medical Oncology/trends , Neoplasms/drug therapy , Pharmacokinetics , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Computer Simulation , Drug-Related Side Effects and Adverse Reactions , Humans , Neoplasms/pathology
7.
Cancer Res ; 75(12): 2416-25, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25939602

ABSTRACT

Predictive biomarkers can play a key role in individualized disease monitoring. Unfortunately, the use of biomarkers in clinical settings has thus far been limited. We have previously shown that mechanism-based pharmacokinetic/pharmacodynamic modeling enables integration of nonvalidated biomarker data to provide predictive model-based biomarkers for response classification. The biomarker model we developed incorporates an underlying latent variable (disease) representing (unobserved) tumor size dynamics, which is assumed to drive biomarker production and to be influenced by exposure to treatment. Here, we show that by integrating CT scan data, the population model can be expanded to include patient outcome. Moreover, we show that in conjunction with routine medical monitoring data, the population model can support accurate individual predictions of outcome. Our combined model predicts that a change in disease of 29.2% (relative standard error 20%) between two consecutive CT scans (i.e., 6-8 weeks) gives a probability of disease progression of 50%. We apply this framework to an external dataset containing biomarker data from 22 small cell lung cancer patients (four patients progressing during follow-up). Using only data up until the end of treatment (a total of 137 lactate dehydrogenase and 77 neuron-specific enolase observations), the statistical framework prospectively identified 75% of the individuals as having a predictable outcome in follow-up visits. This included two of the four patients who eventually progressed. In all identified individuals, the model-predicted outcomes matched the observed outcomes. This framework allows at risk patients to be identified early and therapeutic intervention/monitoring to be adjusted individually, which may improve overall patient survival.


Subject(s)
Lung Neoplasms/pathology , Models, Biological , Models, Statistical , Precision Medicine/methods , Small Cell Lung Carcinoma/pathology , Aged , Biomarkers, Tumor/analysis , Disease Progression , Female , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/therapy , Male , Middle Aged , Predictive Value of Tests , Small Cell Lung Carcinoma/diagnosis , Small Cell Lung Carcinoma/therapy
10.
Eur J Nucl Med Mol Imaging ; 41(11): 2058-65, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24990403

ABSTRACT

PURPOSE: The tumour molecular profile predicts the activity of epidermal growth factor receptor (EGFR) inhibitors in non-small-cell lung cancer (NSCLC). However, tissue availability and tumour heterogeneity limit its assessment. We evaluated whether [(18)F]FDG PET might help predict KRAS and EFGR mutation status in NSCLC. METHODS: Between January 2005 and October 2011, 340 NSCLC patients were tested for KRAS and EGFR mutation status. We identified patients with stage III and IV disease who had undergone [(18)F]FDG PET/CT scanning for initial staging. SUVpeak, SUVmax and SUVmean of the single hottest tumour lesions were calculated, and their association with KRAS and EGFR mutation status was assessed. A receiver operator characteristic (ROC) curve analysis and a multivariate analysis (including SUVmean, gender, age and AJCC stage) were performed to identify the potential value of [(18)F]FDG PET/CT for predicting KRAS mutation. RESULTS: From 102 patients staged using [(18)F]FDG PET/CT, 28 (27%) had KRAS mutation (KRAS+), 22 (22%) had EGFR mutation (EGFR+) and 52 (51%) had wild-type KRAS and EGFR profiles (WT). KRAS+ patients showed significantly higher [(18)F]FDG uptake than EGFR+ and WT patients (SUVmean 9.5, 5.7 and 6.6, respectively; p < 0.001). No significant differences were observed in [(18)F]FDG uptake between EGFR+ patients and WT patients. ROC curve analysis for KRAS mutation status discrimination yielded an area under the curve of 0.740 for SUVmean (p < 0.001). The multivariate analysis showed a sensitivity and specificity of 78.6% and 62.2%, respectively, and the AUC was 0.773. CONCLUSION: NSCLC patients with tumours harbouring KRAS mutations showed significantly higher [(18)F]FDG uptake than WT patients, as assessed in terms of SUVpeak, SUVmax and SUVmean. A multivariate model based on age, gender, AJCC stage and SUVmean might be used as a predictive marker of KRAS mutation status in patients with stage III or IV NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnostic imaging , ErbB Receptors/genetics , Fluorodeoxyglucose F18 , Lung Neoplasms/diagnostic imaging , Mutation , Positron-Emission Tomography , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Multivariate Analysis , Proto-Oncogene Proteins p21(ras) , Tomography, X-Ray Computed
11.
Med. paliat ; 21(2): 79-88, abr.-jun. 2014. tab
Article in Spanish | IBECS | ID: ibc-124733

ABSTRACT

OBJETIVO: Sugerir recomendaciones para ayudar al paciente con cáncer avanzado, o al familiar, que plantea el uso de marihuana u otras formas de cannabis medicinal como tratamiento sintomático o de soporte. MÉTODO: A partir de un ejemplo real, se presenta un resumen de la farmacología de los cannabinoides naturales y sintéticos, de la evidencia sobre su eficacia como tratamiento sintomático en cáncer avanzado, y de la actitud y expectativas del paciente o la familia que plantea el uso medicinal del cannabis. Resultado: El cannabis medicinal contiene más de 60 cannabinoides naturales (de los cuales el delta-9-tetrahidrocannabinol es el más importante) y otras sustancias. En nuestro medio, los pacientes que plantean el empleo de cannabis como tratamiento sintomático no solicitan cannabinoides sintéticos aprobados en otros países sino la hierba de cannabis o sus derivados. Lo suelen consumir fumado (lo que favorece la aparición más temprana de niveles plasmáticos) o en infusión. El consumo de cannabis medicinal se presta a una gran variabilidad en las concentraciones plasmáticas de delta-9-tetrahidrocannabinol. Existen pocos estudios que evalúen científicamente la eficacia del cannabis medicinal en el control de síntomas del paciente con cáncer avanzado. Los estudios realizados con cannabinoides sintéticos son metodológicamente muy limitados, pero aportan cierta evidencia sobre el efecto de los cannabinoides en el alivio del dolor (también como coadyuvante), de las náuseas y de los vómitos inducidos por quimioterapia en el enfermo oncológico. No hay evidencia suficiente para afirmar su eficacia en el tratamiento de la hiporexia. Los efectos secundarios de los cannabinoides en dosis moderadas (como en el uso de cannabis medicinal) son en su mayoría leves y de perfil neuropsicológico. La actitud y las expectativas del paciente, junto con el modo de empleo del cannabis medicinal, pueden favorecer que parte de su beneficio se deba a un efecto placebo. CONCLUSIONES: El cannabis medicinal no parece ser tan activo como esperan los pacientes ni tan tóxico como suponen muchos profesionales. Para responder al paciente con cáncer avanzado que plantea su uso como tratamiento sintomático es aconsejable evitar prejuicios, actuar con respeto y prudencia y buscar el beneficio sintomático del paciente


OBJECTIVE: To contribute ideas to answer advanced cancer patients asking about the use of medicinal cannabis as a symptomatic treatment. Method: Based on a real example, we present a summary of the pharmacology of natural and synthetic cannabinoids, the evidence on its effectiveness as a symptomatic treatment in advanced cancer, and the patient's attitude and expectations raised by the medicinal use of cannabis. RESULT: There are more than 60 different cannabinoids (the most relevant: delta-9-tetrahydrocannabinol) and other substances in cannabis. In our setting, patients who ask about the use of cannabis as a symptomatic treatment do not look for synthetic cannabinoids approved in other countries, but rather prefer herbal cannabis or its derivatives. They usually consume smoked cannabis (favoring the earlier onset of plasma levels) or, rarely, in infusion. Use of medicinal cannabis causes a wide variation in plasma concentrations of delta-9-tetrahydrocannabinol. There is evidence supporting a certain effect of synthetic cannabinoids in pain relief (also as an adjuvant), and as antiemetic for chemotherapy-induced nausea and vomiting in cancer PATIENTS: This evidence is based primarily on results of studies with limited methodological quality. There is insufficient evidence to assert its effectiveness in the treatment of anorexia. Side effects of moderate doses of synthetic cannabinoids or medicinal cannabis are mostly mild, as well as their psycho-neurological profile. The attitude and expectations of the patient, and the way to consume of medical cannabis may favor that part of its benefit may be due to a placebo effect. CONCLUSIONS: Medicinal cannabis does not seem as active as patients expect, or as toxic as many professionals suspect. To respond to the advanced cancer patient asking about their use as symptomatic treatment is advisable to avoid prejudice, to respect and to act with caution seeking the patient's symptomatic Benefit


Subject(s)
Humans , Neoplasms/drug therapy , Pain/drug therapy , Cannabinoids/therapeutic use , Cannabis , Palliative Care/methods , Pain Management/methods
12.
AAPS J ; 16(3): 609-19, 2014 May.
Article in English | MEDLINE | ID: mdl-24740245

ABSTRACT

The development of individualized therapies poses a major challenge in oncology. Significant hurdles to overcome include better disease monitoring and early prediction of clinical outcome. Current clinical practice consists of using Response Evaluation Criteria in Solid Tumors (RECIST) to categorize response to treatment. However, the utility of RECIST is restricted due to limitations on the frequency of measurement and its categorical rather than continuous nature. We propose a population modeling framework that relates circulating biomarkers in plasma, easily obtained from patients, to tumor progression levels assessed by imaging scans (i.e., RECIST categories). We successfully applied this framework to data regarding lactate dehydrogenase (LDH) and neuron specific enolase (NSE) concentrations in patients diagnosed with small cell lung cancer (SCLC). LDH and NSE have been proposed as independent prognostic factors for SCLC. However, their prognostic and predictive value has not been demonstrated in the context of standard clinical practice. Our model incorporates an underlying latent variable ("disease level") representing (unobserved) tumor size dynamics, which is assumed to drive biomarker production and to be influenced by exposure to treatment; these assumptions are in agreement with the known physiology of SCLC and these biomarkers. Our model predictions of unobserved disease level are strongly correlated with disease progression measured by RECIST criteria. In conclusion, the proposed framework enables prediction of treatment outcome based on circulating biomarkers and therefore can be a powerful tool to help clinicians monitor disease in SCLC.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Small Cell/diagnosis , L-Lactate Dehydrogenase/analysis , Lung Neoplasms/diagnosis , Phosphopyruvate Hydratase/analysis , Antineoplastic Agents , Antineoplastic Agents, Phytogenic , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin , Carcinoma, Small Cell/drug therapy , Cisplatin , Disease Progression , Disease-Free Survival , Etoposide , Humans , Lung Neoplasms/drug therapy , Models, Biological , Population , Predictive Value of Tests
13.
PLoS One ; 9(2): e89747, 2014.
Article in English | MEDLINE | ID: mdl-24587006

ABSTRACT

BACKGROUND: Palonosetron is a potent second generation 5- hydroxytryptamine-3 selective antagonist which can be administered by either intravenous (IV) or oral routes, but subcutaneous (SC) administration of palonosetron has never been studied, even though it could have useful clinical applications. In this study, we evaluate the bioavailability of SC palonosetron. PATIENTS AND METHODS: Patients treated with platinum-based chemotherapy were randomized to receive SC or IV palonosetron, followed by the alternative route in a crossover manner, during the first two cycles of chemotherapy. Blood samples were collected at baseline and 10, 15, 30, 45, 60, 90 minutes and 2, 3, 4, 6, 8, 12 and 24 h after palonosetron administration. Urine was collected during 12 hours following palonosetron. We compared pharmacokinetic parameters including AUC0-24h, t1/2, and Cmax observed with each route of administration by analysis of variance (ANOVA). RESULTS: From October 2009 to July 2010, 25 evaluable patients were included. AUC0-24h for IV and SC palonosetron were respectively 14.1 and 12.7 ng × h/ml (p=0.160). Bioavalability of SC palonosetron was 118% (95% IC: 69-168). Cmax was lower with SC than with IV route and was reached 15 minutes following SC administration. CONCLUSIONS: Palonosetron bioavailability was similar when administered by either SC or IV route. This new route of administration might be specially useful for outpatient management of emesis and for administration of oral chemotherapy. TRIAL REGISTRATION: ClinicalTrials.gov NCT01046240.


Subject(s)
Antiemetics/administration & dosage , Isoquinolines/administration & dosage , Isoquinolines/pharmacokinetics , Neoplasms/complications , Quinuclidines/administration & dosage , Quinuclidines/pharmacokinetics , Vomiting/drug therapy , Vomiting/etiology , Administration, Intravenous , Adult , Aged , Antiemetics/adverse effects , Antiemetics/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Female , Humans , Injections, Subcutaneous , Isoquinolines/adverse effects , Male , Middle Aged , Neoplasms/drug therapy , Palonosetron , Platinum/administration & dosage , Quinuclidines/adverse effects , Treatment Outcome
14.
PLoS One ; 9(1): e86263, 2014.
Article in English | MEDLINE | ID: mdl-24475095

ABSTRACT

PURPOSE: To identify tissue microRNAs predictive of sunitinib activity in patients with metastatic renal-cell-carcinoma (MRCC) and to evaluate in vitro their mechanism of action in sunitinib resistance. METHODS: We screened 673 microRNAs using TaqMan Low-density-Arrays (TLDAs) in tumors from MRCC patients with extreme phenotypes of marked efficacy and resistance to sunitinib, selected from an identification cohort (n = 41). The most relevant differentially expressed microRNAs were selected using bioinformatics-based target prediction analysis and quantified by qRT-PCR in tumors from patients presenting similar phenotypes selected from an independent cohort (n = 101). In vitro experiments were conducted to study the role of miR-942 in sunitinib resistance. RESULTS: TLDAs identified 64 microRNAs differentially expressed in the identification cohort. Seven candidates were quantified by qRT-PCR in the independent series. MiR-942 was the most accurate predictor of sunitinib efficacy (p = 0.0074). High expression of miR-942, miR-628-5p, miR-133a, and miR-484 was significantly associated with decreased time to progression and overall survival. These microRNAs were also overexpressed in the sunitinib resistant cell line Caki-2 in comparison with the sensitive cell line. MiR-942 overexpression in Caki-2 up-regulates MMP-9 and VEGF secretion which, in turn, promote HBMEC endothelial migration and sunitinib resistance. CONCLUSIONS: We identified differentially expressed microRNAs in MRCC patients presenting marked sensitivity or resistance to sunitinib. MiR-942 was the best predictor of efficacy. We describe a novel paracrine mechanism through which high miR-942 levels in MRCC cells up-regulates MMP-9 and VEGF secretion to enhance endothelial migration and sunitinib resistance. Our results support further validation of these miRNA in clinical confirmatory studies.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Indoles/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , MicroRNAs/genetics , Pyrroles/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Humans , Indoles/pharmacology , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Male , Matrix Metalloproteinase 9/metabolism , Middle Aged , Models, Biological , Neoplasm Metastasis , Paracrine Communication , Prognosis , Pyrroles/pharmacology , Reproducibility of Results , Sunitinib , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
15.
J Transl Med ; 11: 13, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23311395

ABSTRACT

BACKGROUND: Inhibitor of DNA binding 1 (Id1) and 3 (Id3) genes have been related with the inhibition of cell differentiation, cell growth promotion and tumor metastasis. Recently, Id1 has been identified as an independent prognostic factor in patients with lung adenocarcinoma, regardless of the stage. Furthermore, Id1 may confer resistance to treatment (both, radiotherapy and chemotherapy). METHODS: We have studied, using monoclonal antibodies for immunohistochemistry, the Id1 and Id3 tumor epithelial expression in 17 patients with stage III-N2 non-small cell lung cancer (NSCLC) treated with definitive chemoradiotherapy. RESULTS: Id1 expression is observed in 82.4% of the tumors, whereas Id3 expression is present in 41.2% of the samples. Interestingly, Id1 and Id3 expression are mutually correlated (R = 0.579, p = 0.015). In a subgroup analysis of patients with the most locally advanced disease (T4N2 stage), co-expression of Id1 and Id3 showed to be related with a worse overall survival (45 vs 6 months, p = 0.002). A trend towards significance for a worse progression free survival (30 vs 1 months, p = 0.219) and a lower response rate to the treatment (RR = 50% vs 87.5%, p = 0.07) were also observed. CONCLUSIONS: A correlation between Id1 and Id3 protein expression is observed. Id1 and Id3 co-expression seems associated with a poor clinical outcome in patients with locally advanced NSCLC treated with definitive chemoradiotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Proteins/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Neoplasm Proteins/genetics , Adult , Aged , Carcinoma, Non-Small-Cell Lung/genetics , Cohort Studies , Combined Modality Therapy , Female , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Male , Middle Aged , Treatment Outcome
16.
Support Care Cancer ; 21(2): 649-52, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23207922

ABSTRACT

Within an oncology setting, certain chemotherapy drugs, such as cisplatin, may lead to magnesium loss causing nephropathy. Neurological and cardiovascular symptoms caused by hypomagnesaemia are well known. The relationship between serious hypomagnesemia and severe pain is not well documented but nevertheless, when faced with unexplained episodes of pain which do not respond to powerful analgesics, it is important to review blood magnesium levels. We present two cases of opioid-refractory pain attacks. Patients received drugs which have been linked to hypomagnesemia. In both cases, endovenous magnesium replacement led to a drastic improvement in pain management.


Subject(s)
Antineoplastic Agents/adverse effects , Kidney/drug effects , Lymphoma, Non-Hodgkin/complications , Magnesium Deficiency/complications , Nasopharyngeal Neoplasms/complications , Pain/etiology , Adult , Aged , Antineoplastic Agents/therapeutic use , Female , Humans , Lymphoma, Non-Hodgkin/drug therapy , Magnesium Deficiency/chemically induced , Magnesium Deficiency/etiology , Male , Nasopharyngeal Neoplasms/drug therapy , Neoplasm Metastasis , Palliative Care
17.
Clin Transl Oncol ; 14(11): 835-41, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22855163

ABSTRACT

BACKGROUND: The management of operable locally advanced N2 non-small cell lung cancer (NSCLC) is a controversial topic. Concurrent chemoradiation (CT-RT) is considered the standard of care for inoperable or unresectable patients, but the role of trimodality treatment remains controversial. We present our institution's experience with the management of stage III (N2) NSCLC patients, analyzing whether the addition of surgery improves survival when compared with definitive CT-RT alone. METHODS: From 1996 to 2006, 72 N2 NSCLC patients were treated. Thirty-four patients received cisplatin-based induction chemotherapy, followed by paclitaxel-cisplatin CT-RT, and 38 patients underwent surgery preceded by induction and/or followed by adjuvant therapy. Survival curves were estimated by Kaplan-Meier analysis, and the differences were assessed with the log-rank test. RESULTS: Most of the patients (87 %) were men. The median age was 59 years. A statistically significant association between T3-T4c and definitive CT-RT as well as between T1-T2c and surgery was noted (p < 0.0001). After a median follow-up period of 35 months, the median overall survival (OS) was 42 months for the surgery group versus 41 months for the CT-RT patients (p = 0.590). The median progression-free survival (PFS) was 14 months after surgery and 25 months after CT-RT (p = 0.933). Responders to radical CT-RT had a better OS than non-responders (43 vs. 17 months, respectively, p = 0.011). No significant differences were found in the OS or PFS between the pN0 [14 (37.8 %) patients] and non-pN0 patients at thoracotomy. Three treatment-related deaths (7.8 %) were observed in the surgical cohort and none in the CT-RT group. CONCLUSIONS: The addition of surgery did not render a median OS or PFS benefit when compared with CT-RT alone in our series of stage III-N2 NSCLC patients, in accordance with previously published data. However, responses to CT-RT had a greater impact in terms of OS and PFS. Although the patients selected for management including surgery showed a favorable T clinical staging in comparison to patients exclusively treated with definitive CT-RT, similar survival outcomes were found.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Carcinoma, Non-Small-Cell Lung/surgery , Chemotherapy, Adjuvant , Combined Modality Therapy , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/surgery , Male , Middle Aged
18.
Am J Clin Oncol ; 33(2): 137-43, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19786847

ABSTRACT

PURPOSE: To determine feasibility and efficacy of concurrent paclitaxel and cisplatin with definitive hyperfractionated radiotherapy (HFRT) in locally advanced head-and-neck squamous cell carcinoma (HNSCC). PATIENTS AND METHODS: Forty-two patients stages III to IV head-and-neck squamous cell carcinoma were enrolled in 2 consecutive prospective trials from August 1998 to January 2006. In study 1, 16 patients received HFRT in 2 courses of 39.6 Gy each with a split of 2 weeks with concurrent paclitaxel (175 mg/m) and cisplatin (100 mg/m) on days 1, 21, 36, and 57. In study 2, 26 patients received a continuous course of 74.4 Gy of HFRT with concurrent weekly paclitaxel (50 mg/m) and cisplatin (30 mg/m). RESULTS: Tumor locations included oropharynx 48%, hypopharynx 24%, larynx 12%, paranasal sinuses 7%, salivary gland 2%, oral cavity 2% and unknown primary 5%. In study 1, all patients received 3 to 4 cycles of chemotherapy and completed the programmed radiotherapy course. In study 2, 69% received 5 to 6 cycles of chemotherapy and 92% completed the irradiation. Overall, 93% of objective responses were observed (complete 76%, partial 17%). Median follow-up was 50 months (range: 12-97). Pattern of recurrence was local 8%, distant 13%, and combined 3%. Acute toxicity grades 3 to 4 in studies 1 and 2 was 75% and 88%, respectively (P = ns). Globally, 5-year overall survival were 68%, with a median of 71 months (range: 50-91). On multivariate analysis, male gender (P = 0.04) and complete response (P = 0.01) were predictive of improved survival. CONCLUSION: HFRT combined with cisplatin and paclitaxel is very active but at the expense of severe toxicity. Efficacy and toxicity in studies.1 and 2 were not different despite completely different treatment strategies (chemotherapy dose intensity vs. radiotherapy dose intensity).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Adult , Aged , Carcinoma, Squamous Cell/pathology , Cisplatin/administration & dosage , Combined Modality Therapy , Dose Fractionation, Radiation , Drug Administration Schedule , Female , Follow-Up Studies , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Neoplasm Staging , Paclitaxel/administration & dosage , Prospective Studies , Survival Rate , Treatment Outcome
19.
Exp Dermatol ; 18(9): 771-80, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19552768

ABSTRACT

Pemphigus vulgaris (PV) is an autoimmune blistering disease characterized by the presence of IgG autoantibodies against Dsg3. Our aim was to investigate the molecular events implicated in the development and localization of apoptosis and acantholysis in PV. We used a passive transfer mouse model together with immunohistochemical (IHC) techniques and the TUNEL assay, with quantification analysis in the basal layer of the epidermis. The activated signalling molecules analysed and apoptotic cells detected showed an identical localization. Herein, we found for the first time in vivo an increased expression of activated HER receptor isoforms in the basal layer in PV lesions. Besides, we observed the almost total lack of activated Akt compared with a higher level of activated mTOR within the basal cells of the epidermis. Our observations strongly support that the restriction of acantholysis to the basal layer may be due, at least in part, to the selective and increased presence of activated HER receptor isoforms in these cells. After phosphorylation of HER receptor isoforms, intracellular signalling pathways are activated in the basal layer. In addition, the imbalance in Akt/mTOR that takes place in the basal cells may provide intracellular signals necessary for the development of apoptosis and acantholysis.


Subject(s)
Acantholysis , Apoptosis , Carrier Proteins/metabolism , ErbB Receptors/metabolism , Pemphigus/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Betacellulin , Disease Models, Animal , Enzyme Activation , Epidermal Growth Factor/metabolism , Epidermis/metabolism , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Humans , Immunoglobulin G , Intercellular Signaling Peptides and Proteins/metabolism , Intradermal Tests , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Pemphigus/physiopathology , Pyrazoles , Pyrimidines , Quinazolines , Sirolimus , TOR Serine-Threonine Kinases , Transforming Growth Factor alpha/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
20.
Lung Cancer ; 64(1): 124-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19008012

ABSTRACT

Pemetrexed is a multitargeted antifolate approved for the second-line treatment of locally advanced or metastatic non-small cell lung cancer. The combination of pemetrexed with gemcitabine has been studied in several clinical trials showing a promising antitumor activity with a mild toxicity profile. We present the case of a patient who experienced fever, arthralgia, skin rash and high serum ferritin levels after first cycle of this chemotherapy combination, compatible with an adult onset Still's disease. This adverse event has not been previously reported.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Still's Disease, Adult-Onset/chemically induced , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Glutamates/administration & dosage , Guanine/administration & dosage , Guanine/analogs & derivatives , Humans , Male , Middle Aged , Pemetrexed , Still's Disease, Adult-Onset/drug therapy , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...