9 research outputs found
The dynamic use of EGFR mutation analysis in cell-free DNA as a follow-up biomarker during different treatment lines in non-small-cell lung cancer patients
Epidermal growth factor receptor (EGFR) mutational testing in advanced non-small-cell lung cancer (NSCLC) is usually performed
in tumor tissue, although cfDNA (cell-free DNA) could be an alternative. We evaluated EGFR mutations in cfDNA as a
complementary tool in patients, who had already known EGFR mutations in tumor tissue and were treated with either
EGFR-tyrosine kinase inhibitors (TKIs) or chemotherapy. We obtained plasma samples from 21 advanced NSCLC patients with
known EGFR tumor mutations, before and during therapy with EGFR-TKIs and/or chemotherapy. cfDNA was isolated and
EGFR mutations were analyzed with the multiple targeted cobas EGFR Mutation Test v2. EGFR mutations were detected at
baseline in cfDNA from 57% of patients. The semiquantitative index (SQI) significantly decreased from the baseline
(median = 11, IQR = 9 5-13) to the best response (median = 0, IQR = 0-0, p < 0 01), followed by a significant increase at
progression (median = 11, IQR = 11-15, p < 0 01) in patients treated with either EGFR-TKIs or chemotherapy. The SQI obtained
with the cobas EGFR Mutation Test v2 did not correlate with the concentration in copies/mL determined by droplet digital
PCR. Resistance mutation p.T790M was observed at progression in patients with either type of treatment. In conclusion, cfDNA
multiple targeted EGFR mutation analysis is useful for treatment monitoring in tissue of EGFR-positive NSCLC patients
independently of the drug received
Genomic characterization of individuals presenting extreme phenotypes of high and low risk to develop tobacco-induced lung cancer
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 (p(combined)=5.66x10(-5); ORcombined=2.80), mapping to a noncoding transcript exon of PDE10A; and rs6835978 (p(combined)=1.02x10(-4); ORcombined=2.57), an intronic variant in ATP10D. We assessed the relevance of both proteins in early-stage NSCLC. PDE10A and ATP10D mRNA 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
The dynamic use of EGFR mutation analysis in cell-free DNA as a follow-up biomarker during different treatment lines in non-small-cell lung cancer patients
Epidermal growth factor receptor (EGFR) mutational testing in advanced non-small-cell lung cancer (NSCLC) is usually performed
in tumor tissue, although cfDNA (cell-free DNA) could be an alternative. We evaluated EGFR mutations in cfDNA as a
complementary tool in patients, who had already known EGFR mutations in tumor tissue and were treated with either
EGFR-tyrosine kinase inhibitors (TKIs) or chemotherapy. We obtained plasma samples from 21 advanced NSCLC patients with
known EGFR tumor mutations, before and during therapy with EGFR-TKIs and/or chemotherapy. cfDNA was isolated and
EGFR mutations were analyzed with the multiple targeted cobas EGFR Mutation Test v2. EGFR mutations were detected at
baseline in cfDNA from 57% of patients. The semiquantitative index (SQI) significantly decreased from the baseline
(median = 11, IQR = 9 5-13) to the best response (median = 0, IQR = 0-0, p < 0 01), followed by a significant increase at
progression (median = 11, IQR = 11-15, p < 0 01) in patients treated with either EGFR-TKIs or chemotherapy. The SQI obtained
with the cobas EGFR Mutation Test v2 did not correlate with the concentration in copies/mL determined by droplet digital
PCR. Resistance mutation p.T790M was observed at progression in patients with either type of treatment. In conclusion, cfDNA
multiple targeted EGFR mutation analysis is useful for treatment monitoring in tissue of EGFR-positive NSCLC patients
independently of the drug received
CXCR1 and CXCR2 chemokine receptor agonists produced by tumors induce neutrophil extracellular traps that interfere with immune cytotoxicity
Neutrophils are expanded and abundant in cancer-bearing hosts. Under the influence of CXCR1 and CXCR2
chemokine receptor agonists and other chemotactic factors produced by tumors, neutrophils, and granulocytic myeloid-derived suppressor cells (MDSCs) from cancer patients extrude their neutrophil extracellular
traps (NETs). In our hands, CXCR1 and CXCR2 agonists proved to be the major mediators of cancer-promoted NETosis. NETs wrap and coat tumor cells and shield them from cytotoxicity, as mediated by CD8+
T cells and natural killer (NK) cells, by obstructing contact between immune cells and the surrounding target
cells. Tumor cells protected from cytotoxicity by NETs underlie successful cancer metastases in mice and the
immunotherapeutic synergy of protein arginine deiminase 4 (PAD4) inhibitors, which curtail NETosis with immune checkpoint inhibitors. Intravital microscopy provides evidence of neutrophil NETs interfering cytolytic
cytotoxic T lymphocytes (CTLs) and NK cell contacts with tumor cells
Characterization of the perioperative changes of exosomal immune-related cytokines induced by prostatectomy in early-stage prostate cancer patients
Background: Myeloid-derived suppressor cells (MDSCs) are relevant in prostate cancer microenvironment
collaborating in tumor development. The main tumor marker used in this disease, prostate-specific antigen
(PSA), does not provide information related to this tumor microenvironment. Cancer cells secrete exosomes
carrying bioactive molecules contributing to MDSCs recruitment and induction. The aim of this study was to
characterize the perioperative changes of exosomal cytokines relevant in MDSCs recruitment induced by pros-
tatectomy in prostate cancer patients.
Methods: Blood was drawn from 26 early-stage prostate cancer patients before and after radical prostatectomy
and from 16 healthy volunteers. Serum exosomes were separated by precipitation. Cytokines related with MDSC
cell recruitment and activation CCL2, CXCL2, CXCL5, CXCL8, CXCL12, MIF, S100A9 and TGF-Ă were measured
in serum and serum-derived exosomes using immunometric assays.
Results: All cytokines were detected both in serum and exosomes, except for CXCL12, which was detected only in serum. Exosomes were enriched specially in MIF, TGF-Ă and CXCL2. Presurgical MIF levels in exosomes correlated negatively with serum PSA. Also, presurgical TGF-Ă decreased both in serum and exosomes as Gleason score rises. PatientsÌ presurgical exosomes had increased CCL2, CXCL5 and TGF-Ă levels than exosomes from healthy controls. These differences were not observed when cytokines were analyzed in serum, except for TGF-Ă.Cytokine levels of CCL2, CXCL5 decreased in patientsâ postsurgical exosomes, while TGF-Ă further increased. On the contrary, S100A9 levels were lower in patientsÌ presurgical exosomes but increased after radical prostatectomy.
Conclusions: Blood exosomal content in cytokines constitute an attractive source to evaluate MDSCs immuno-
modulators providing additional information related to tumor microenvironment in prostate cance
Characterization of the perioperative changes of exosomal immune-related cytokines induced by prostatectomy in early-stage prostate cancer patients
Background: Myeloid-derived suppressor cells (MDSCs) are relevant in prostate cancer microenvironment
collaborating in tumor development. The main tumor marker used in this disease, prostate-specific antigen
(PSA), does not provide information related to this tumor microenvironment. Cancer cells secrete exosomes
carrying bioactive molecules contributing to MDSCs recruitment and induction. The aim of this study was to
characterize the perioperative changes of exosomal cytokines relevant in MDSCs recruitment induced by pros-
tatectomy in prostate cancer patients.
Methods: Blood was drawn from 26 early-stage prostate cancer patients before and after radical prostatectomy
and from 16 healthy volunteers. Serum exosomes were separated by precipitation. Cytokines related with MDSC
cell recruitment and activation CCL2, CXCL2, CXCL5, CXCL8, CXCL12, MIF, S100A9 and TGF-Ă were measured
in serum and serum-derived exosomes using immunometric assays.
Results: All cytokines were detected both in serum and exosomes, except for CXCL12, which was detected only in serum. Exosomes were enriched specially in MIF, TGF-Ă and CXCL2. Presurgical MIF levels in exosomes correlated negatively with serum PSA. Also, presurgical TGF-Ă decreased both in serum and exosomes as Gleason score rises. PatientsÌ presurgical exosomes had increased CCL2, CXCL5 and TGF-Ă levels than exosomes from healthy controls. These differences were not observed when cytokines were analyzed in serum, except for TGF-Ă.Cytokine levels of CCL2, CXCL5 decreased in patientsâ postsurgical exosomes, while TGF-Ă further increased. On the contrary, S100A9 levels were lower in patientsÌ presurgical exosomes but increased after radical prostatectomy.
Conclusions: Blood exosomal content in cytokines constitute an attractive source to evaluate MDSCs immuno-
modulators providing additional information related to tumor microenvironment in prostate cance
Serum interleukin-8 reflects tumor burden and treatment response across malignancies of multiple tissue origins
Purpose: Interleukin-8 (IL8) is a chemokine produced by malignant cells of multiple cancer types. It
exerts various functions in shaping protumoral vascularization and inflammation/immunity. We evaluated
sequential levels of serum IL8 in preclinical tumor models and in patients to assess its ability to estimate
tumor burden.
Experimental Design:IL8levels were monitored by sandwich ELISAsin cultured tumor cells supernatants,
tumor-xenografted mice serum, and in samples from 126 patients with cancer. We correlated IL8 serum levels
with baseline tumor burden and with treatment-induced changes in tumor burden, as well as with prognosis.
Results: IL8 concentrations correlated with the number of IL8-producing tumor cells in culture. In
xenografted neoplasms, IL8 serum levels rapidly dropped after surgical excision, indicating an accurate
correlation with tumor burden. In patients with melanoma (n Œ 16), renal cell carcinoma (RCC; n Œ 23),
nonâsmall cell lung cancer (NSCLC; n ÂŒ 21), or hepatocellular carcinoma (HCC; n ÂŒ 30), serum IL8
concentrations correlated with tumor burden and stage, survival (melanoma, n Œ 16; RCC, n Œ 23; HCC,
n Œ 33), and objective responses to therapy, including those to BRAF inhibitors (melanoma, n Œ 16) and
immunomodulatory monoclonal antibodies (melanoma, n Œ 8). IL8 concentrations in urine (n Œ 18) were
mainly elevated in tumors with direct contact with the urinary tract.
Conclusions: IL8 levels correlate with tumor burden in preclinical models and in patients with cancer.
IL8 is a potentially useful biomarker to monitor changes in tumor burden following anticancer therapy, and
has prognostic significance
Genomic characterization of individuals presenting extreme phenotypes of high and low risk to develop tobacco-induced lung cancer
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 (p(combined)=5.66x10(-5); ORcombined=2.80), mapping to a noncoding transcript exon of PDE10A; and rs6835978 (p(combined)=1.02x10(-4); ORcombined=2.57), an intronic variant in ATP10D. We assessed the relevance of both proteins in early-stage NSCLC. PDE10A and ATP10D mRNA 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
Identification of mutations associated with acquired resistance to sunitinib in renal cell cancer
Sunitinib is one of the most widely used targeted therapeutics for renal cell carcinoma (RCC), but acquired resistance against
targeted therapies remains a major clinical challenge. To dissect mechanisms of acquired resistance and unravel reliable
predictive biomarkers for sunitinib in RCC, we sequenced the exons of 409 tumor-suppressor genes and oncogenes in paired
tumor samples from an RCC patient, obtained at baseline and after development of acquired resistance to sunitinib. From
newly arising mutations, we selected, using in silico prediction models, six predicted to be deleterious, located in G6PD,
LRP1B, SETD2, TET2, SYNE1, and DCC. Consistently, immunoblotting analysis of lysates derived from sunitinib-desensitized
RCC cells and their parental counterparts showed marked differences in the levels and expression pattern of the proteins
encoded by these genes. Our further analysis demonstrates essential roles for these proteins in mediating sunitinib
cytotoxicity and shows that their loss of function renders tumor cells resistant to sunitinib in vitro and in vivo. Finally, sunitinib
resistance induced by continuous exposure or by inhibition of the six proteins was overcome by treatment with cabozantinib or
a low-dose combination of lenvatinib and everolimus. Collectively, our results unravel novel markers of acquired resistance to
sunitinib and clinically relevant approaches for overcoming this resistance in RCC