Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Biomed Pharmacother ; 176: 116857, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38850664

ABSTRACT

Metastatic colorectal cancer (mCRC) currently lacks reliable biomarkers for precision medicine, particularly for chemotherapy-based treatments. This study examines the behavior of 11 CXC chemokines in the blood of 104 mCRC patients undergoing first-line oxaliplatin-based treatment to pinpoint predictive and prognostic markers. Serum samples were collected before treatment, at response evaluation (EVAR), and at disease progression or last follow-up. Chemokines were assessed in all samples using a Luminex® custom panel. CXCL13 levels increased at EVAR in responders, while in non-responders it decreased. Increasing levels of CXCL13 at EVAR, independently correlated with improved progression-free survival (PFS) and overall survival (OS). Nanostring® analysis in primary tumor samples showed CXCL13 gene expression's positive correlation not only with gene profiles related to an immunogenic tumor microenvironment, increased B cells and T cells (mainly CD8+) but also with extended OS. In silico analysis using RNAseq data from liver metastases treated or not with neoadjuvant oxaliplatin-based combinations, and deconvolution analysis using the MCP-counter algorithm, confirmed CXCL13 gene expression's association with increased immune infiltration, improved OS, and Tertiary Lymphoid Structures (TLSs) gene signatures, especially in neoadjuvant-treated patients. CXCL13 analysis in serum from 36 oxaliplatin-treated patients from the METIMMOX study control arm, reported similar findings. In conclusion, the increase of CXCL13 levels in peripheral blood and its association with the formation of TLSs within the metastatic lesions, emerges as a potential biomarker indicative of the therapeutic efficacy in mCRC patients undergoing oxaliplatin-based treatment.

2.
Mol Oncol ; 17(9): 1908-1916, 2023 09.
Article in English | MEDLINE | ID: mdl-37097008

ABSTRACT

Efficiency of expanded genomic profiling (EGP) programmes in terms of final inclusion of patients in genomically matched therapies is still unknown. Fit patients with advanced and refractory colorectal cancer (CRC) were selected for an EGP programme. Next-generation sequencing (NGS) analysis from formalin-fixed paraffin-embedded tumour samples was performed. The purpose was to describe the prevalence of genomic alterations defined by the ESMO Scale for Clinical Actionability of Molecular Targets (ESCAT), as well as the percentage of patients finally included in genomically guided clinical trials. In total, 187 patients were recruited. Mutational profile was obtained in 177 patients (10 patients were failure due to insufficient tumour sample), copy number alterations in 41 patients and fusions in 31 patients. ESCAT-defined alterations were detected in 28.8% of the intention-to-analyse population. BRAF V600E was clustered in ESCAT I, with a prevalence of 3.7%, KRAS G12C and ERBB2 amplification were clustered in ESCAT II, whose prevalence was 4.2% and 1.6%, respectively. Most alterations were classified in ESCAT III (mutations in ERBB2, PIK3CA or FGFR genes and MET amplification) and IV (mutations in BRAF non-V600E, ERBB3, FBXW7, NOTCH, RNF43), with a single prevalence under 5%, except for PIK3CA mutation (9%). The final rate of inclusion into genomically guided clinical trials was 2.7%, including therapies targeting BRAF V600E or RNF43 mutations in two patients each, and ERBB2 mutation in one patient. In conclusion, EGP programmes in patients with advanced CRC are feasible and identify a subset of patients with potentially druggable genomic alterations. However, further efforts must be made to increase the rate of patients treated with genomically guided therapies.


Subject(s)
Colorectal Neoplasms , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Colorectal Neoplasms/genetics , Mutation/genetics , Genomics , High-Throughput Nucleotide Sequencing
3.
Cancers (Basel) ; 15(4)2023 Feb 06.
Article in English | MEDLINE | ID: mdl-36831367

ABSTRACT

About 5 to 15% of all colorectal cancers harbor mismatch repair deficient/microsatellite instability-high status (dMMR/MSI-H) that associates with high tumor mutation burden and increased immunogenicity. As a result, and in contrast to other colorectal cancer phenotypes, a significant subset of dMMR/MSI-H cancer patients strongly benefit from immunotherapy. Yet, a large proportion of these tumors remain unresponsive to any immuno-modulating treatment. For this reason, current efforts are focused on the characterization of resistance mechanisms and the identification of predictive biomarkers to guide therapeutic decision-making. Here, we provide an overview on the new advances related to the diagnosis and definition of dMMR/MSI-H status and focus on the distinct clinical, functional, and molecular cues that associate with dMMR/MSI-H colorectal cancer. We review the development of novel predictive factors of response or resistance to immunotherapy and their potential application in the clinical setting. Finally, we discuss current and emerging strategies applied to the treatment of localized and metastatic dMMR/MSI-H colorectal tumors in the neoadjuvant and adjuvant setting.

4.
Cancers (Basel) ; 14(17)2022 Aug 30.
Article in English | MEDLINE | ID: mdl-36077748

ABSTRACT

The gut microbiome is a potential modifiable risk factor for colorectal cancer (CRC). We re-analyzed all eight previously published stool sequencing data and conducted an MWAS meta-analysis. We used cross-validated LASSO predictive models to identify a microbiome signature for predicting the risk of CRC and precancerous lesions. These models were validated in a new study, Colorectal Cancer Screening (COLSCREEN), including 156 participants that were recruited in a CRC screening context. The MWAS meta-analysis identified 95 bacterial species that were statistically significantly associated with CRC (FDR < 0.05). The LASSO CRC predictive model obtained an area under the receiver operating characteristic curve (aROC) of 0.81 (95%CI: 0.78−0.83) and the validation in the COLSCREEN dataset was 0.75 (95%CI: 0.66−0.84). This model selected a total of 32 species. The aROC of this CRC-trained model to predict precancerous lesions was 0.52 (95%CI: 0.41−0.63). We have identified a signature of 32 bacterial species that have a good predictive accuracy to identify CRC but not precancerous lesions, suggesting that the identified microbes that were enriched or depleted in CRC are merely a consequence of the tumor. Further studies should focus on CRC as well as precancerous lesions with the intent to implement a microbiome signature in CRC screening programs.

5.
Int J Mol Sci ; 24(1)2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36613564

ABSTRACT

The search for immunotherapy biomarkers in Microsatellite Instability High/Deficient Mismatch Repair system (MSI-H/dMMR) metastatic colorectal cancer (mCRC) is an unmet need. Sixteen patients with mCRC and MSI-H/dMMR (determined by either immunohistochemistry or polymerase chain reaction) treated with PD-1/PD-L1 inhibitors at our institution were included. According to whether the progression-free survival with PD-1/PD-L1 inhibitors was longer than 6 months or shorter, patients were clustered into the IT-responder group (n: 9 patients) or IT-resistant group (n: 7 patients), respectively. In order to evaluate determinants of benefit with PD-1/PD-L1 inhibitors, we performed multimodal analysis including genomics (through NGS panel tumour-only with 431 genes) and the immune microenvironment (using CD3, CD8, FOXP3 and PD-L1 antibodies). The following mutations were more frequent in IT-resistant compared with IT-responder groups: B2M (4/7 versus 2/9), CTNNB1 (2/7 versus 0/9), and biallelic PTEN (3/7 versus 1/9). Biallelic ARID1A mutations were found exclusively in the IT-responder group (4/9 patients). Tumour mutational burden did not correlate with immunotherapy benefit, neither the rate of indels in homopolymeric regions. Of note, biallelic ARID1A mutated tumours had the highest immune infiltration and PD-L1 scores, contrary to tumours with CTNNB1 mutation. Immune microenvironment analysis showed higher densities of different T cell subpopulations and PD-L1 expression in IT-responders. Misdiagnosis of MSI-H/dMMR inferred by discordances between immunohistochemistry and polymerase chain reaction was only found in the IT-resistant population (3/7 patients). Biallelic ARID1A mutations and Wnt signalling activation through CTNNB1 mutation were associated with high and low T cell immune infiltrates, respectively, and deserve special attention as determinants of response to PD-1/PD-L1 inhibitors. The non-MSI-H phenotype in dMMR is associated with poor benefit to immunotherapy. Our results suggest that mechanisms of resistance to immunotherapy are multi-factorial.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Humans , B7-H1 Antigen/genetics , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , DNA Mismatch Repair , Programmed Cell Death 1 Receptor/genetics , Colonic Neoplasms/genetics , Colorectal Neoplasms/therapy , Colorectal Neoplasms/drug therapy , Microsatellite Repeats , Microsatellite Instability , Immunotherapy , Tumor Microenvironment/genetics
6.
Int J Mol Sci ; 22(3)2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33535583

ABSTRACT

The study of the human microbiome in oncology is a growing and rapidly evolving field. In the past few years, there has been an exponential increase in the number of studies investigating associations of microbiome and cancer, from oncogenesis and cancer progression to resistance or sensitivity to specific anticancer therapies. The gut microbiome is now known to play a significant role in antitumor immune responses and in predicting the efficacy of immune-checkpoint inhibitors in cancer patients. Beyond the gut, the tumor-associated microbiome-microbe communities located either in the tumor or within its body compartment-seems to interact with the local microenvironment and the tumor immune contexture, ultimately impacting cancer progression and treatment outcome. However, pre-clinical research focusing on causality and mechanistic pathways as well as proof-of-concept studies are still needed to fully understand the potential clinical utility of microbiome in cancer patients. Moreover, there is a need for the standardization of methodology and the implementation of quality control across microbiome studies to allow for a better interpretation and greater comparability of the results reported between them. This review summarizes the accumulating evidence in the field and discusses the current and upcoming challenges of microbiome studies.


Subject(s)
Gene Expression Regulation, Bacterial , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Microbiota/physiology , Neoplasms/microbiology , Tumor Microenvironment , Animals , Antineoplastic Agents/pharmacology , Carcinogenesis , Cell Transformation, Neoplastic , Computational Biology , Cytoplasm/metabolism , Disease Progression , Dysbiosis , Gastrointestinal Microbiome/physiology , Humans , Immunity , Metagenome , Metagenomics , Mice , RNA, Ribosomal, 16S/metabolism
7.
Sci Rep ; 9(1): 3816, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30846724

ABSTRACT

Soft-tissue sarcomas (STS) are an uncommon and heterogeneous group of malignancies that result in high mortality. Metastatic STS have very bad prognosis due to the lack of effective treatments. Dinaciclib is a model drug for the family of CDK inhibitors. Its main targets are cell cycle regulator CDK1 and protein synthesis controller CDK9. We present data supporting Dinaciclib ability to inactivate in vitro different STS models at nanomolar concentrations. Moreover, the different rhythms of cell death induction allow us to further study into the mechanism of action of the drug. Cell death was found to respond to the mitochondrial pathway of apoptosis. Anti-apoptotic Bcl-xL was identified as the key regulator of this process. Already natural low levels of pro-apoptotic proteins BIM and PUMA in tolerant cell lines were insufficient to inhibit Bcl-xL as this anti-apoptotic protein showed a slow decay curve after Dinaciclib-induced protein synthesis disruption. Combination of Dinaciclib with BH3-mimetics led to quick and massive apoptosis induction in vitro, but in vivo assessment was prevented due to liver toxicity. Additionally, Bcl-xL inhibitor A-1331852 also synergized with conventional chemotherapy drugs as Gemcitabine. Thus, Bcl-xL targeted therapy arises as a major opportunity to the treatment of STS.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Death/drug effects , Pyridinium Compounds/pharmacology , Sarcoma/pathology , bcl-X Protein/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cyclic N-Oxides , Humans , Indolizines , Mice
8.
Expert Opin Drug Saf ; 17(9): 945-951, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30073902

ABSTRACT

INTRODUCTION: Ramucirumab, a human monoclonal antibody against vascular endothelial growth factor receptor 2 (VEGFR-2), is an antiangiogenic therapy that has been approved in combination with FOLFIRI in second-line treatment of metastatic colorectal cancer (mCRC), after progression on or after therapy with bevacizumab, oxaliplatin, and a fluoropyrimidine. A thorough review of the safety of ramucirumab in this setting and in the context of other antiangiogenic agents is merited. Areas covered: We provide an overview of activity and summarize in detail the overall safety and tolerability profile of ramucirumab in mCRC patients on the basis of a literature review of all published clinical trials in this setting, including both single-agent and combination studies. A focus on adverse events of interest and specific populations is included, as well as a critical comparison with other antiangiogenic therapies. Expert opinion: As an effective agent in pretreated mCRC patients, the toxicity profile of ramucirumab is similar to those of other angiogenesis inhibitors used in the second-line mCRC setting. The next challenge will be to find biomarkers of response and toxicity to antiangiogenic therapies in order to more effectively implement personalized medicine in these patients.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/pathology , Humans , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Ramucirumab
9.
Target Oncol ; 13(1): 81-87, 2018 02.
Article in English | MEDLINE | ID: mdl-29177953

ABSTRACT

BACKGROUND: Gemcitabine plus sirolimus enhances apoptosis in vitro and increases anti-tumor efficacy in vivo in soft-tissue sarcoma (STS) models. OBJECTIVE: The objective of this study was to evaluate the activity and toxicity of the combination of gemcitabine plus sirolimus in patients with STS after failure of standard chemotherapy. PATIENTS AND METHODS: Advanced STS patients, previously treated with doxorubicin and/or ifosfamide, were included in this single-arm phase II study. Patients received gemcitabine 800 mg/m2 intravenously (iv) at 10 mg/m2/min on days 1 and 8 every 3 weeks plus sirolimus 5 mg daily orally (po). After enrolment of the first 12 patients, the study protocol was amended due to toxicity and the starting dose of sirolimus was reduced to 3 mg daily po. Archival tumor samples were analyzed for extracellular signal-regulated kinase 1 and 2 (ERK1/2) expression and correlated with outcome. The primary endpoint was progression-free rate (PFR) at 3 months. RESULTS: From May 2012 to May 2013, 28 patients were enrolled at eight centers. PFR at 3 and 6 months was 44% and 20%, respectively, with 12 patients being free of progression at 3 months. Median progression-free survival (PFS) was 1.85 months (95% confidence interval [CI] 0.73-2.97) and median overall survival (OS) was 9.2 months (95% CI 5.8-12.5). No responses were observed. The most common grade 3-4 hematologic toxicities were neutropenia (48%) and leukopenia (41%) and the most frequent grade 3 non-hematologic toxicities were infection (18.5%), transaminitis (15%), fatigue (11%), and pneumonitis (11%). ERK1/2 expression was significantly correlated with PFS (p = 0.026). CONCLUSIONS: The combination of gemcitabine and sirolimus is an active treatment in STS. Further investigation is warranted. ClinicalTrials.gov identifier: NCT01684449.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Sarcoma/drug therapy , Sirolimus/therapeutic use , Adult , Aged , Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Female , Hispanic or Latino , Humans , Male , Middle Aged , Research Design , Sarcoma/pathology , Sirolimus/pharmacology , Young Adult , Gemcitabine
10.
Onco Targets Ther ; 9: 7573-7582, 2016.
Article in English | MEDLINE | ID: mdl-28008275

ABSTRACT

Gastrointestinal stromal tumor (GIST) is the most common mesenchymal tumor of the gastrointestinal tract. In advanced setting and after progression to imatinib, the multi-targeted receptor tyrosine kinase inhibitor sunitinib has clearly demonstrated a clinical benefit in terms of response rate and progression-free survival with an acceptable toxicity profile. The recommended schedule for sunitinib administration is 50 mg per day 4 weeks ON and 2 weeks OFF; however, potential alternative schedules are also reviewed in the present article. Several biomarkers have been explored to better select candidates for sunitinib therapy, such as the value of early changes in standardized uptake value assessed by positron emission tomography with 18F-fluorodeoxyglucose, circulating biomarkers, clinical biomarkers such as the appearance of arterial hypertension during treatment that correlates with better outcomes, and the GIST genotype. GISTs with KIT mutations at exon 9 and the so-called wild-type GISTs seem to better respond to sunitinib. Nonetheless, further investigation is required to confirm these findings as well as to understand the mechanisms of sunitinib resistance such as the development of new KIT mutations or conformational changes in KIT receptor.

11.
Cancer Chemother Pharmacol ; 77(1): 133-46, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26563256

ABSTRACT

Soft tissue sarcomas (STS) constitute an uncommon and heterogeneous group of tumours, which require a complex and specialized multidisciplinary management. The diagnostic approach should include imaging studies and core needle biopsy performed prior to undertaking surgery. Wide excision is the mainstay of treatment for localized sarcoma, and associated preoperative or postoperative radiotherapy should be administered in high-risk patients. Adjuvant chemotherapy was associated with a modest improvement in survival in a meta-analysis and constitutes a standard option in selected patients with high-risk STS. In metastatic patients, surgery must be evaluated in selected cases. In the rest of patients, chemotherapy and, in some subtypes, targeted therapy often used in a sequential strategy constitutes the treatment of election. Despite important advances in the understanding of the pathophysiology of the disease, the advances achieved in therapeutic results may be deemed still insufficient. Moreover, due to the rarity and complexity of the disease, the results in clinical practice are not always optimal. For this reason, the Spanish Group for Research on Sarcoma (GEIS) has developed a multidisciplinary clinical practice guidelines document, with the aim of facilitating the diagnosis and treatment of these patients in Spain. In the document, each practical recommendation is accompanied by level of evidence and grade of recommendation on the basis of the available data.


Subject(s)
Sarcoma/therapy , Soft Tissue Neoplasms/therapy , Antineoplastic Agents/therapeutic use , Chemotherapy, Adjuvant/methods , Humans , Sarcoma/diagnosis , Sarcoma/pathology , Soft Tissue Neoplasms/diagnosis , Soft Tissue Neoplasms/pathology , Spain
12.
Front Oncol ; 5: 82, 2015.
Article in English | MEDLINE | ID: mdl-25905041

ABSTRACT

Cell death can occur through different mechanisms, defined by their nature and physiological implications. Correct assessment of cell death is crucial for cancer therapy success. Sarcomas are a large and diverse group of neoplasias from mesenchymal origin. Among cell death types, apoptosis is by far the most studied in sarcomas. Albeit very promising in other fields, regulated necrosis and other cell death circumstances (as so-called "autophagic cell death" or "mitotic catastrophe") have not been yet properly addressed in sarcomas. Cell death is usually quantified in sarcomas by unspecific assays and in most cases the precise sequence of events remains poorly characterized. In this review, our main objective is to put into context the most recent sarcoma cell death findings in the more general landscape of different cell death modalities.

13.
Endocrinol. nutr. (Ed. impr.) ; 59(7): 438-451, ago. 2012. ilus, tab
Article in Spanish | IBECS | ID: ibc-104069

ABSTRACT

Los tumores neuroendocrinos gastroenteropancreáticos (TNE-GEP) constituyen el segundo tumor avanzado más prevalente del tracto digestivo. En los últimos años, los recursos invertidos para la investigación en esta población de pacientes se han visto aumentados exponencialmente convirtiéndose en uno de los escenarios más atractivos para la investigación oncológica. Varias proteínas proangiogénicas han sido identificadas como sobreexpresadas en los TNE-GEP, incluyendo el factor de crecimiento del endotelio vascular y sus receptores, y las vías de señalización intracelular más relacionadas como la del receptor del factor de crecimiento epidérmico, el receptor tipo i del factor de crecimiento similar a la insulina y la vía de PI3K-(PTEN)-AKT-mTOR. Los resultados recientes de los 3 estudios fase iii más importantes en TNE-GEP han permitido la aprobación de 2 terapias dirigidas, sunitinib y everolimus, para el tratamiento de los pacientes con tumores neuroendocrinos pancreáticos después de décadas de mínimos avances en esta población (AU)


Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are the second most prevalent group of advanced gastrointestinal tract tumors. Resources invested in research on this patient population have exponentially increased in recent years, and this has become one of the most attractive fields for oncological research. Several proangiogenic proteins have been found to be overexpressed in GEP-NETs, including vascular endothelial growth factor and its receptors and the more closely related intracellular signaling pathways such as the epidermal growth factor pathway, type I insulin-like growth factor receptor, and the PI3K-(PTEN)-AKTmTOR pathway. The recent results of the three most important Phase III studies in GEP-NET have allowed for approval of two targeted agents, sunitinib and everolimus, for the treatment of patients with pancreatic neuroendocrine tumors after decades of minimal advances in this population (AU)


Subject(s)
Humans , Neuroendocrine Tumors/therapy , Gastrointestinal Neoplasms/therapy , Pancreatic Neoplasms/therapy , Biological Therapy/methods , Antibodies, Monoclonal/therapeutic use , Angiogenesis Modulating Agents/analysis , Biomarkers, Tumor/analysis
14.
Endocrinol Nutr ; 59(7): 438-51, 2012.
Article in Spanish | MEDLINE | ID: mdl-22565119

ABSTRACT

Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are the second most prevalent group of advanced gastrointestinal tract tumors. Resources invested in research on this patient population have exponentially increased in recent years, and this has become one of the most attractive fields for oncological research. Several proangiogenic proteins have been found to be overexpressed in GEP-NETs, including vascular endothelial growth factor and its receptors and the more closely related intracellular signaling pathways such as the epidermal growth factor pathway, type I insulin-like growth factor receptor, and the PI3K-(PTEN)-AKT-mTOR pathway. The recent results of the three most important Phase III studies in GEP-NETs have allowed for approval of two targeted agents, sunitinib and everolimus, for the treatment of patients with pancreatic neuroendocrine tumors after decades of minimal advances in this population.


Subject(s)
Gastrointestinal Neoplasms/drug therapy , Molecular Targeted Therapy , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Everolimus , Humans , Immunosuppressive Agents/therapeutic use , Sirolimus/analogs & derivatives , Sirolimus/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...