Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Ann Oncol ; 33(8): 750-768, 2022 08.
Article in English | MEDLINE | ID: mdl-35809752

ABSTRACT

Circulating tumour DNA (ctDNA) assays conducted on plasma are rapidly developing a strong evidence base for use in patients with cancer. The European Society for Medical Oncology convened an expert working group to review the analytical and clinical validity and utility of ctDNA assays. For patients with advanced cancer, validated and adequately sensitive ctDNA assays have utility in identifying actionable mutations to direct targeted therapy, and may be used in routine clinical practice, provided the limitations of the assays are taken into account. Tissue-based testing remains the preferred test for many cancer patients, due to limitations of ctDNA assays detecting fusion events and copy number changes, although ctDNA assays may be routinely used when faster results will be clinically important, or when tissue biopsies are not possible or inappropriate. Reflex tumour testing should be considered following a non-informative ctDNA result, due to false-negative results with ctDNA testing. In patients treated for early-stage cancers, detection of molecular residual disease or molecular relapse, has high evidence of clinical validity in anticipating future relapse in many cancers. Molecular residual disease/molecular relapse detection cannot be recommended in routine clinical practice, as currently there is no evidence for clinical utility in directing treatment. Additional potential applications of ctDNA assays, under research development and not recommended for routine practice, include identifying patients not responding to therapy with early dynamic changes in ctDNA levels, monitoring therapy for the development of resistance mutations before clinical progression, and in screening asymptomatic people for cancer. Recommendations for reporting of results, future development of ctDNA assays and future clinical research are made.


Subject(s)
Circulating Tumor DNA , Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , Humans , Mutation , Neoplasm Recurrence, Local , Precision Medicine/methods
2.
ESMO Open ; 6(2): 100060, 2021 04.
Article in English | MEDLINE | ID: mdl-33647598

ABSTRACT

Precision medicine is now pivotal to design patients' specific treatment strategies with the aim of prolonging progression and overall survival. In this regard, invasive tumor tissue testing has so far been the golden standard for making cancer diagnosis, but has limitations. Cell-free tumor DNA (ctDNA), a form of liquid biopsy, is a noninvasive biomarker that can be isolated from patients' blood and other biofluids. An increasing body of evidence has demonstrated clinical utility of plasma ctDNA profiling to select patients for genomic-driven therapies. Analyses of mutations in plasma ctDNA have shown high accuracy and more rapid identification of mutations, allowing matching patients for specific therapies with equivalent clinical efficacy to that of the tissue profiling. In the clinical setting, ctDNA has been recently implemented to select patients with specific genomic alterations to targeted treatments, and a few molecular tests have been approved for use in non-small-cell lung, prostate, ovarian, and breast cancers. However, standardization of ctDNA collection, storage, and analysis methods would be critical to facilitate the wide adoption of ctDNA technology in routine clinical practice. This review summarizes how we can exploit ctDNA analysis to treat cancer patients, and explains how the results should be interpreted. In addition, we focus on how ctDNA could be used in the future as a marker of minimal residual disease to guide adjuvant therapy, as an immuno-oncology biomarker in patients treated with immune checkpoint blockade drugs, and as an early cancer detection marker to screen the asymptomatic population.


Subject(s)
Circulating Tumor DNA , Neoplasms/diagnosis , Circulating Tumor DNA/genetics , Genomics , Humans , Liquid Biopsy , Neoplasms/genetics , Neoplasms/therapy
3.
Ann Oncol ; 31(8): 978-990, 2020 08.
Article in English | MEDLINE | ID: mdl-32610166

ABSTRACT

BACKGROUND: The use of next-generation sequencing technologies has enabled the rapid identification of non-synonymous somatic mutations in cancer cells. Neoantigens are mutated peptides derived from somatic mutations not present in normal tissues that may result in the presentation of tumour-specific peptides capable of eliciting antitumour T-cell responses. Personalised neoantigen-based cancer vaccines and adoptive T-cell therapies have been shown to prime host immunity against tumour cells and are under clinical trial development. However, the optimisation and standardisation of neoantigen identification, as well as its delivery as immunotherapy are needed to increase tumour-specific T-cell responses and, thus, the clinical efficacy of current cancer immunotherapies. METHODS: In this recommendation article, launched by the European Society for Medical Oncology (ESMO), we outline and discuss the available framework for neoantigen prediction and present a systematic review of the current scientific evidence. RESULTS: A number of computational pipelines for neoantigen prediction are available. Most of them provide peptide major histocompatibility complex (MHC) binding affinity predictions, but more recent approaches incorporate additional features like variant allele fraction, gene expression, and clonality of mutations. Neoantigens can be predicted in all cancer types with high and low tumour mutation burden, in part by exploiting tumour-specific aberrations derived from mutational frameshifts, splice variants, gene fusions, endogenous retroelements and other tumour-specific processes that could yield more potently immunogenic tumour neoantigens. Ongoing clinical trials will highlight those cancer types and combinations of immune therapies that would derive the most benefit from neoantigen-based immunotherapies. CONCLUSIONS: Improved identification, selection and prioritisation of tumour-specific neoantigens are needed to increase the scope of benefit from cancer vaccines and adoptive T-cell therapies. Novel pipelines are being developed to resolve the challenges posed by high-throughput sequencing and to predict immunogenic neoantigens.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Antigens, Neoplasm/genetics , Immunotherapy , Medical Oncology , Neoplasms/genetics , Neoplasms/therapy , Precision Medicine , Practice Guidelines as Topic
5.
Ann Oncol ; 30(2): 211-218, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30576421

ABSTRACT

Challenges in obtaining tissue specimens from patients with brain tumours limit the diagnosis and molecular characterisation and impair the development of better therapeutic approaches. The analysis of cell-free tumour DNA in plasma (considered a liquid biopsy) has facilitated the characterisation of extra-cranial tumours. However, cell-free tumour DNA in plasma is limited in quantity and may not reliably capture the landscape of genomic alterations of brain tumours. Here, we review recent work assessing the relevance of cell-free tumour DNA from cerebrospinal fluid in the characterisation of brain cancer. We focus on the advances in the use of the cerebrospinal fluid as a source of cell-free tumour DNA to facilitate diagnosis, reveal actionable genomic alterations, monitor responses to therapy, and capture tumour heterogeneity in patients with primary brain tumours and brain and leptomeningeal metastases. Profiling cerebrospinal fluid cell-free tumour DNA provides the opportunity to precisely acquire and monitor genomic information in real time and guide precision therapies.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/pathology , Central Nervous System Neoplasms/secondary , Circulating Tumor DNA/genetics , DNA, Neoplasm/genetics , Liquid Biopsy/methods , Biomarkers, Tumor/cerebrospinal fluid , Brain Neoplasms/cerebrospinal fluid , Brain Neoplasms/genetics , Central Nervous System Neoplasms/cerebrospinal fluid , Central Nervous System Neoplasms/genetics , Circulating Tumor DNA/cerebrospinal fluid , DNA, Neoplasm/cerebrospinal fluid , Humans , Neoplasm Staging
7.
Ann Oncol ; 28(7): 1590-1596, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28449049

ABSTRACT

BACKGROUND: Burnout in health care professionals could have serious negative consequences on quality of patient care, professional satisfaction and personal life. Our aim was to investigate the burnout prevalence, work and lifestyle factors potentially affecting burnout amongst European oncologists ≤40 (YOs). METHODS: A survey was conducted using the validated Maslach Burnout Inventory (MBI) and additional questions exploring work/lifestyle factors. Statistical analyses were carried out to identify factors associated with burnout. RESULTS: Total of 737 surveys (all ages) were collected from 41 European countries. Countries were divided into six regions. Results from 595 (81%) YOs were included (81% medical oncologists; 52% trainees, 62% women). Seventy-one percent of YOs showed evidence of burnout (burnout subdomains: depersonalization 50%; emotional exhaustion 45; low accomplishment 35%). Twenty-two percent requested support for burnout during training and 74% reported no hospital access to support services. Burnout rates were significantly different across Europe (P < 0.0001). Burnout was highest in central European (84%) and lowest in Northern Europe (52%). Depersonalization scores were higher in men compared with women (60% versus 45% P = 0.0001) and low accomplishment was highest in the 26-30 age group (P < 0.01). In multivariable linear regression analyses, European region, work/life balance, access to support services, living alone and inadequate vacation time remained independent burnout factors (P < 0.05). CONCLUSIONS: This is the largest burnout survey in European Young Oncologists. Burnout is common amongst YOs and rates vary across Europe. Achieving a good work/life balance, access to support services and adequate vacation time may reduce burnout levels. Raising awareness, support and interventional research are needed.


Subject(s)
Burnout, Professional/epidemiology , Occupational Health , Oncologists , Adult , Age Factors , Attitude of Health Personnel , Burnout, Professional/diagnosis , Burnout, Professional/psychology , Burnout, Professional/therapy , Chi-Square Distribution , Depersonalization , Emotions , Europe/epidemiology , Female , Health Knowledge, Attitudes, Practice , Health Surveys , Humans , Job Satisfaction , Linear Models , Logistic Models , Male , Multivariate Analysis , Oncologists/psychology , Patient Acceptance of Health Care , Quality of Life , Risk Factors , Sex Factors , Work-Life Balance
8.
Br J Cancer ; 112(4): 650-9, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25625276

ABSTRACT

BACKGROUND: Heat-shock protein 990 (HSP990) is a potent and selective synthetic small-molecule HSP90 inhibitor. The primary objectives of this phase I first-in-human study were to determine dose-limiting toxicities (DLTs), maximum-tolerated dose (MTD) and recommended phase II dose (RP2D). Secondary objectives included characterisation of the safety profile, pharmacokinetics (PKs) and pharmacodynamics (PDs). METHODS: Heat-shock protein 990 was administered orally once or two times weekly on a 28-day cycle schedule in patients with advanced solid tumours. Dose escalation was guided by a Bayesian logistic regression model with overdose control. RESULTS: A total of 64 patients were enrolled. Fifty-three patients received HSP990 once weekly at 2.5, 5, 10, 20, 30, 50 or 60 mg, whereas 11 patients received HSP990 two times weekly at 25 mg. Median duration of exposure was 8 weeks (range 1-116 weeks) and 12 patients remained on treatment for >16 weeks. Dose-limiting toxicities occurred in seven patients and included diarrhoea, QTc prolongation, ALT/AST elevations and central neurological toxicities. The most common drug-related adverse events were diarrhoea, fatigue and decreased appetite. Further dose escalation beyond 60 mg once weekly was not possible owing to neurological toxicity. Rapid absorption, no drug accumulation and large interpatient variability in PK exposures were observed. No objective responses were seen; 25 patients had a best overall response of stable disease. CONCLUSIONS: Heat-shock protein 990 is relatively well tolerated, with neurological toxicity being the most relevant DLT. The single agent MTD/RP2D of HSP990 was declared at 50 mg once weekly.


Subject(s)
Antineoplastic Agents/administration & dosage , Heat-Shock Proteins, Small/administration & dosage , Neoplasms/drug therapy , Pyridones/administration & dosage , Pyrimidines/administration & dosage , Administration, Oral , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Disease Progression , Dose-Response Relationship, Drug , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins, Small/adverse effects , Heat-Shock Proteins, Small/pharmacokinetics , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/pathology , Pyridones/adverse effects , Pyridones/pharmacokinetics , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Treatment Outcome
9.
Clin Transl Oncol ; 16(11): 937-41, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25060567

ABSTRACT

Breast cancers and most malignant tumors are composed of heterogeneous tumor cells both at genetic and morphological levels; intra-tumor heterogeneity can be one underlying cause of therapeutic resistance. Classical studies have focused on analyses of the relationship between primary tumors and metastatic dissemination, and on subclone evolution. However, it should be noted that tumor heterogeneity at the level of protein expression (proteomics) has not been yet studied in depth. The differences in protein expression also can play an important role in elucidating the relationship between intra-tumor heterogeneity and resistance to systemic therapy. In fact, in human tumors there is not always a homogeneous expression of many of the crucial factors involved in cell signaling, such as pMAPK, pAKt, pMTOR, even with constitutive oncogenic alterations upstream, such as HER2, PI3 K. Conversely, two of these factors, peIF4E and p4E-BP1, which are downstream, and control protein translation, show a diffuse and strong protein expression. In summary, most of cell signaling factors show a heterogeneous expression, regardless of oncogenic alterations. Tissue heterogeneity could be driven by local factors, including hypoxia. The fact that the phosphorylation of crucial proteins such as 4E-BP1 and eIF4E is observed homogeneously throughout most tumors and are druggable opens the chance to get real potential targets in cancer therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Phosphoproteins/metabolism , Signal Transduction , Animals , Cell Cycle Proteins , Female , Humans , Phosphorylation , Proteomics , Signal Transduction/physiology
10.
Ann Oncol ; 25(9): 1729-1735, 2014 09.
Article in English | MEDLINE | ID: mdl-25009010

ABSTRACT

BACKGROUND: Plasma-derived cell-free tumor DNA (ctDNA) constitutes a potential surrogate for tumor DNA obtained from tissue biopsies. We posit that massively parallel sequencing (MPS) analysis of ctDNA may help define the repertoire of mutations in breast cancer and monitor tumor somatic alterations during the course of targeted therapy. PATIENT AND METHODS: A 66-year-old patient presented with synchronous estrogen receptor-positive/HER2-negative, highly proliferative, grade 2, mixed invasive ductal-lobular carcinoma with bone and liver metastases at diagnosis. DNA extracted from archival tumor material, plasma and peripheral blood leukocytes was subjected to targeted MPS using a platform comprising 300 cancer genes known to harbor actionable mutations. Multiple plasma samples were collected during the fourth line of treatment with an AKT inhibitor. RESULTS: Average read depths of 287x were obtained from the archival primary tumor, 139x from the liver metastasis and between 200x and 900x from ctDNA samples. Sixteen somatic non-synonymous mutations were detected in the liver metastasis, of which 9 (CDKN2A, AKT1, TP53, JAK3, TSC1, NF1, CDH1, MML3 and CTNNB1) were also detected in >5% of the alleles found in the primary tumor sample. Not all mutations identified in the metastasis were reliably identified in the primary tumor (e.g. FLT4). Analysis of ctDNA, nevertheless, captured all mutations present in the primary tumor and/or liver metastasis. In the longitudinal monitoring of the patient, the mutant allele fractions identified in ctDNA samples varied over time and mirrored the pharmacodynamic response to the targeted therapy as assessed by positron emission tomography-computed tomography. CONCLUSIONS: This proof-of-principle study is one of the first to demonstrate that high-depth targeted MPS of plasma-derived ctDNA constitutes a potential tool for de novo mutation identification and monitoring of somatic genetic alterations during the course of targeted therapy, and may be employed to overcome the challenges posed by intra-tumor genetic heterogeneity. REGISTERED CLINICAL TRIAL: www.clinicaltrials.gov, NCT01090960.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , DNA, Neoplasm/blood , DNA, Neoplasm/genetics , Base Sequence , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell-Free System , Female , Genetic Heterogeneity , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Middle Aged , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Sequence Analysis, DNA
11.
J Intern Med ; 276(1): 41-51, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24661605

ABSTRACT

Cells within tumours have diverse genomes and epigenomes and interact differentially with their surrounding microenvironment generating intratumour heterogeneity, which has critical implications for treating cancer patients. Understanding the cellular and microenvironment composition and characteristics in individual tumours is critical to stratify the patient population that is likely to benefit from specific treatment regimens. Here, we will review the current understanding of intratumour heterogeneity at the genomic, epigenomic and microenvironmental levels. We will also discuss the clinical implications and the challenges posed by intratumour heterogeneity and evaluate noninvasive methods such as circulating biomarkers to characterize the cellular diversity of tumours. Comprehensive assessment of the molecular features of patients based on tumour specimen characterization (including intratumour spatial and temporal variations), ancillary noninvasive methods (such as circulating biomarkers and molecular imaging approaches) and the correct design of clinical trials are required to guide administration of targeted therapy and to control therapeutic resistance. Finding the means to accurately determine and effectively control tumour heterogeneity and translate these achievements into patient benefit are major goals in modern oncology.


Subject(s)
Neoplasms/genetics , Precision Medicine/methods , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic/genetics , Genetic Heterogeneity , Humans , Neoplasms/pathology , Neoplasms/therapy , Pilot Projects , Tumor Microenvironment
12.
Ann Oncol ; 24(5): 1163-70, 2013 May.
Article in English | MEDLINE | ID: mdl-23165977

ABSTRACT

Management of the axilla in early breast cancer (EBC) patients has dramatically evolved in recent years from more radical to increasingly conservative approaches. Classically, the EBC patients with a clinically positive axilla are offered axillary lymph node dissection (ALND) and those with a clinically negative axilla (cN0) are offered sentinel lymph node (SLN) biopsy, which obviates the complications related to ALND and provides adequate surgical staging and comparable locoregional control and survival. The need for performing ALND when the SLN is positive and contemporary adjuvant treatment is delivered has been questioned in recent years. On the other hand, ongoing trials are testing whether node-positive patients can be spared chemotherapy, based on intrinsic primary tumor biology. Because the integration of novel surgical management and tumor biology is needed, this article provides an overview of the current challenges that a more detailed knowledge of tumor biology has brought to EBC staging and treatment. We propose that breast cancer oncologists (surgeons, radiation therapists, and medical oncologists) should focus their efforts on offering therapy tailored to each patient's needs in such a way that no matter which treatment is used, no overtreatment occurs.


Subject(s)
Breast Neoplasms/pathology , Lymph Node Excision , Sentinel Lymph Node Biopsy , Axilla , Breast Neoplasms/surgery , Female , Humans , Lymph Nodes/surgery , Lymphatic Metastasis , Neoplasm Recurrence, Local/surgery , Treatment Outcome
13.
Ann Oncol ; 23(4): 897-902, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21828361

ABSTRACT

BACKGROUND: Elderly breast cancer patients are usually excluded from clinical trials. Nevertheless, with the increasing use of trastuzumab, there is a need to address trastuzumab-related cardiotoxicity in this population. PATIENTS AND METHODS: Records for patients ≥ 70 years treated with trastuzumab since 2005 were reviewed. New York Heart Association classification was used to document symptomatic cardiotoxicity. Asymptomatic cardiotoxicity was defined as an absolute drop ≥ 10% with a final left ventricular ejection fraction < 50% or an absolute drop > 20%. RESULTS: Forty-five patients, median age 75.9 years (range 70-92), were identified. Three of 24 (12.5%) early breast cancer patients and 5 of 21 (23.8%) with advanced disease experienced asymptomatic cardiotoxicity. Four of 45 patients (8.9%), all with advanced breast cancer, developed symptomatic congestive heart failure. All but one of them recovered in a median time of 5 weeks (range 3-21). Patients with trastuzumab-related cardiotoxicity presented more often with cardiovascular risk factors, such as history of cardiac disease (33% versus 9.1%, P = 0.017) and diabetes (33.3% versus 6.1%, P = 0.010), compared with those without. CONCLUSIONS: Elderly breast cancer patients with a history of cardiac disease and/or diabetes treated with trastuzumab have an increased incidence of cardiotoxicity. Continuous cardiac monitoring is especially advised in this population.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , Breast Neoplasms/drug therapy , Carcinoma, Ductal, Breast/drug therapy , Heart Failure/chemically induced , Aged , Aged, 80 and over , Breast Neoplasms/complications , Carcinoma, Ductal, Breast/complications , Diabetes Complications/chemically induced , Female , Humans , Multivariate Analysis , Risk Factors , Trastuzumab
SELECTION OF CITATIONS
SEARCH DETAIL
...