Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Oncogene ; 41(10): 1445-1455, 2022 03.
Article in English | MEDLINE | ID: mdl-35039635

ABSTRACT

Prostate cancer (PCa) is the fifth leading cause of cancer related deaths worldwide, in part due to a lack of molecular stratification tools that can distinguish primary tumours that will remain indolent from those that will metastasise. Amongst potential molecular biomarkers, microRNAs (miRs) have attracted particular interest because of their high stability in body fluids and fixed tissues. These small non-coding RNAs modulate several physiological and pathological processes, including cancer progression. Herein we explore the prognostic potential and the functional role of miRs in localised PCa and their relation to nodal metastasis. We define a 7-miR signature that is associated with poor survival independently of age, Gleason score, pathological T state, N stage and surgical margin status and that is also prognostic for disease-free survival in patients with intermediate-risk localised disease. Within our 7-miR signature, we show that miR-378a-3p (hereafter miR-378a) levels are low in primary tumours compared to benign prostate tissue, and also lower in Gleason score 8-9 compared to Gleason 6-7 PCa. We demonstrate that miR-378a impairs glucose metabolism and reduces proliferation in PCa cells through independent mechanisms, and we identify glucose transporter 1 (GLUT1) messenger RNA as a direct target of miR-378a. We show that GLUT1 inhibition hampers glycolysis, leading to cell death. Our data provides a rational for a new PCa stratification strategy based on miR expression, and it reveals that miR-378a and GLUT1 are potential therapeutic targets in highly aggressive glycolytic PCa.


Subject(s)
MicroRNAs , Prostatic Neoplasms , Cell Line, Tumor , Cell Proliferation/genetics , Glucose , Glucose Transporter Type 1/genetics , Humans , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Prostatic Neoplasms/genetics
2.
Ann R Coll Surg Engl ; 103(1): e17-e19, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32969264

ABSTRACT

Hepatic herniation through the diaphragm is a rare finding. It generally occurs due to a congenital diaphragmatic abnormality or blunt trauma resulting in a diaphragmatic defect. Making the diagnosis is difficult, as there are few definitive clinical signs and chest radiograph (CXR) findings may be non-specific. To our knowledge, only a single case report exists of penetrating right diaphragm injury leading to hepatic herniation. A 42-year-old man presented to the emergency department of a regional hospital with hyperglycaemia and exertional dyspnoea. He was diagnosed with diabetes mellitus type 2. He gave a history of smoking for 15 pack-years, was negative for retroviral disease and had no history of pulmonary tuberculosis. He had no significant surgical history but reported being stabbed with a knife in 1995. The point of entry was below the level of the nipple in the right anterior axillary line. At the time, he was treated with an intercostal drain and discharged home. CXR showed a right-sided chest mass. We considered a differential diagnosis of pulmonary consolidation, diaphragm eventration or hepatothorax. Computerized tomography of the chest and abdomen demonstrated apparent intrathoracic extension of the right liver lobe and partial attenuation of the superior vena cava and right atrium due to a mass effect. The upper border of the liver abutted the aortic arch. Surgical treatment options were discussed. The patient declined surgery and will be followed up as an outpatient.


Subject(s)
Diaphragm/injuries , Dyspnea/etiology , Hernia, Diaphragmatic/diagnosis , Liver/diagnostic imaging , Wounds, Stab/complications , Adult , Diagnosis, Differential , Diaphragm/diagnostic imaging , Drainage , Dyspnea/surgery , Hernia, Diaphragmatic/etiology , Hernia, Diaphragmatic/surgery , Humans , Male , Radiography, Thoracic , Time Factors , Tomography, X-Ray Computed
3.
Ann Oncol ; 32(2): 269-278, 2021 02.
Article in English | MEDLINE | ID: mdl-33130216

ABSTRACT

BACKGROUND: KRAS is mutated in ∼90% of pancreatic ductal adenocarcinomas, ∼35% of colorectal cancers and ∼20% of non-small-cell lung cancers. There has been recent progress in targeting G12CKRAS specifically, but therapeutic options for other mutant forms of KRAS are limited, largely because the complexity of downstream signaling and feedback mechanisms mean that targeting individual pathway components is ineffective. DESIGN: The protein kinases RAF and SRC are validated therapeutic targets in KRAS-mutant pancreatic ductal adenocarcinomas, colorectal cancers and non-small-cell lung cancers and we show that both must be inhibited to block growth of these cancers. We describe CCT3833, a new drug that inhibits both RAF and SRC, which may be effective in KRAS-mutant cancers. RESULTS: We show that CCT3833 inhibits RAF and SRC in KRAS-mutant tumors in vitro and in vivo, and that it inhibits tumor growth at well-tolerated doses in mice. CCT3833 has been evaluated in a phase I clinical trial (NCT02437227) and we report here that it significantly prolongs progression-free survival of a patient with a G12VKRAS spindle cell sarcoma who did not respond to a multikinase inhibitor and therefore had limited treatment options. CONCLUSIONS: New drug CCT3833 elicits significant preclinical therapeutic efficacy in KRAS-mutant colorectal, lung and pancreatic tumor xenografts, demonstrating a treatment option for several areas of unmet clinical need. Based on these preclinical data and the phase I clinical unconfirmed response in a patient with KRAS-mutant spindle cell sarcoma, CCT3833 requires further evaluation in patients with other KRAS-mutant cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , src-Family Kinases/genetics
4.
Ann Oncol ; 30(5): 804-814, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30838379

ABSTRACT

BACKGROUND: The advent of effective adjuvant therapies for patients with resected melanoma has highlighted the need to stratify patients based on risk of relapse given the cost and toxicities associated with treatment. Here we assessed circulating tumor DNA (ctDNA) to predict and monitor relapse in resected stage III melanoma. PATIENTS AND METHODS: Somatic mutations were identified in 99/133 (74%) patients through tumor tissue sequencing. Personalized droplet digital PCR (ddPCR) assays were used to detect known mutations in 315 prospectively collected plasma samples from mutation-positive patients. External validation was performed in a prospective independent cohort (n = 29). RESULTS: ctDNA was detected in 37 of 99 (37%) individuals. In 81 patients who did not receive adjuvant therapy, 90% of patients with ctDNA detected at baseline and 100% of patients with ctDNA detected at the postoperative timepoint relapsed at a median follow up of 20 months. ctDNA detection predicted patients at high risk of relapse at baseline [relapse-free survival (RFS) hazard ratio (HR) 2.9; 95% confidence interval (CI) 1.5-5.6; P = 0.002] and postoperatively (HR 10; 95% CI 4.3-24; P < 0.001). ctDNA detection at baseline [HR 2.9; 95% CI 1.3-5.7; P = 0.003 and postoperatively (HR 11; 95% CI 4.3-27; P < 0.001] was also associated with inferior distant metastasis-free survival (DMFS). These findings were validated in the independent cohort. ctDNA detection remained an independent predictor of RFS and DMFS in multivariate analyses after adjustment for disease stage and BRAF mutation status. CONCLUSION: Baseline and postoperative ctDNA detection in two independent prospective cohorts identified stage III melanoma patients at highest risk of relapse and has potential to inform adjuvant therapy decisions.


Subject(s)
Circulating Tumor DNA/blood , Melanoma/blood , Neoplasm Recurrence, Local/blood , Skin Neoplasms/blood , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , Female , GTP Phosphohydrolases/genetics , Humans , Male , Melanoma/genetics , Melanoma/pathology , Membrane Proteins/genetics , Middle Aged , Mutation , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/genetics , Neoplasm Staging , Prognosis , Prospective Studies , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Survival Rate , Young Adult , Melanoma, Cutaneous Malignant
5.
Ann Oncol ; 29(2): 490-496, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29112704

ABSTRACT

Background: Patients with high-risk stage II/III resected melanoma commonly develop distant metastases. At present, we cannot differentiate between patients who will recur or those who are cured by surgery. We investigated if circulating tumor DNA (ctDNA) can predict relapse and survival in patients with resected melanoma. Patients and methods: We carried out droplet digital polymerase chain reaction to detect BRAF and NRAS mutations in plasma taken after surgery from 161 stage II/III high-risk melanoma patients enrolled in the AVAST-M adjuvant trial. Results: Mutant BRAF or NRAS ctDNA was detected (≥1 copy of mutant ctDNA) in 15/132 (11%) BRAF mutant patient samples and 4/29 (14%) NRAS mutant patient samples. Patients with detectable ctDNA had a decreased disease-free interval [DFI; hazard ratio (HR) 3.12; 95% confidence interval (CI) 1.79-5.47; P < 0.0001] and distant metastasis-free interval (DMFI; HR 3.22; 95% CI 1.80-5.79; P < 0.0001) versus those with undetectable ctDNA. Detectable ctDNA remained a significant predictor after adjustment for performance status and disease stage (DFI: HR 3.26, 95% CI 1.83-5.83, P < 0.0001; DMFI: HR 3.45, 95% CI 1.88-6.34, P < 0.0001). Five-year overall survival rate for patients with detectable ctDNA was 33% (95% CI 14%-55%) versus 65% (95% CI 56%-72%) for those with undetectable ctDNA. Overall survival was significantly worse for patients with detectable ctDNA (HR 2.63; 95% CI 1.40-4.96); P = 0.003) and remained significant after adjustment for performance status (HR 2.50, 95% CI 1.32-4.74, P = 0.005). Conclusion: ctDNA predicts for relapse and survival in high-risk resected melanoma and could aid selection of patients for adjuvant therapy. Clinical trial number: ISRCTN 81261306.


Subject(s)
Circulating Tumor DNA/blood , Melanoma/blood , Neoplasm Recurrence, Local/blood , Skin Neoplasms/blood , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , GTP Phosphohydrolases/genetics , Humans , Kaplan-Meier Estimate , Male , Melanoma/genetics , Melanoma/mortality , Membrane Proteins/genetics , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/mortality , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins B-raf/genetics , Retrospective Studies , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Young Adult , Melanoma, Cutaneous Malignant
6.
Eur J Cancer ; 88: 1-9, 2018 01.
Article in English | MEDLINE | ID: mdl-29175734

ABSTRACT

INTRODUCTION: Tumour burden is a prognostic biomarker in metastatic melanoma. However, tumour burden is difficult to measure and there are currently no reliable surrogate biomarkers to easily and reliably determine it. The aim of this study was to assess the potential of plasma total cell free DNA as biomarker of tumour burden and prognosis in metastatic melanoma patients. MATERIALS AND METHODS: A prospective biomarker cohort study for total plasma circulating cell-free DNA (cfDNA) concentration was performed in 43 metastatic melanoma patients. For 38 patients, paired blood collections and scan assessments were available before treatment and at first response evaluation. Tumour burden was calculated as the sum of volumes from three-dimensional radiological measurements of all metastatic lesions in individual patients. RESULTS: Baseline cfDNA concentration correlated with pre-treatment tumour burden (ρ = 0.52, P < 0.001). Baseline cfDNA levels correlated significantly with hazard of death and overall survival, and a cut off value of 89 pg/µl identified two distinct prognostic groups (HR = 2.22 for high cfDNA, P = 0.004). Patients with cfDNA ≥89 pg/µl had shorter OS (10.0 versus 22.7 months, P = 0.009; HR = 2.22 for high cfDNA, P = 0.004) and the significance was maintained when compared with lactic dehydrogenase (LDH) in a multivariate analysis. We also found a correlation between the changes of cfDNA and treatment-related changes in tumour burden (ρ = 0.49, P = 0.002). In addition, the ratio between baseline cfDNA and tumour burden was prognostic (HR = 2.7 for cfDNA/tumour volume ≥8 pg/(µl*cm3), P = 0.024). CONCLUSIONS: We have demonstrated that cfDNA is a surrogate marker of tumour burden in metastatic melanoma patients, and that it is prognostic for overall survival.


Subject(s)
Biomarkers, Tumor/genetics , Cell-Free Nucleic Acids/genetics , Melanoma/genetics , Tumor Burden/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Cell-Free Nucleic Acids/blood , Female , Humans , Kaplan-Meier Estimate , Male , Melanoma/blood , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Prognosis , Prospective Studies , Young Adult
7.
Ann Oncol ; 29(1): 30-35, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29140430

ABSTRACT

Background: Precision medicine is rapidly evolving within the field of oncology and has brought many new concepts and terminologies that are often poorly defined when first introduced, which may subsequently lead to miscommunication within the oncology community. The European Society for Medical Oncology (ESMO) recognises these challenges and is committed to support the adoption of precision medicine in oncology. To add clarity to the language used by oncologists and basic scientists within the context of precision medicine, the ESMO Translational Research and Personalised Medicine Working Group has developed a standardised glossary of relevant terms. Materials and methods: Relevant terms for inclusion in the glossary were identified via an ESMO member survey conducted in Autumn 2016, and by the ESMO Translational Research and Personalised Medicine Working Group members. Each term was defined by experts in the field, discussed and, if necessary, modified by the Working Group before reaching consensus approval. A literature search was carried out to determine which of the terms, 'precision medicine' and 'personalised medicine', is most appropriate to describe this field. Results: A total of 43 terms are included in the glossary, grouped into five main themes-(i) mechanisms of decision, (ii) characteristics of molecular alterations, (iii) tumour characteristics, (iv) clinical trials and statistics and (v) new research tools. The glossary classes 'precision medicine' or 'personalised medicine' as technically interchangeable but the term 'precision medicine' is favoured as it more accurately reflects the highly precise nature of new technologies that permit base pair resolution dissection of cancer genomes and is less likely to be misinterpreted. Conclusions: The ESMO Precision Medicine Glossary provides a resource to facilitate consistent communication in this field by clarifying and raising awareness of the language employed in cancer research and oncology practice. The glossary will be a dynamic entity, undergoing expansion and refinement over the coming years.


Subject(s)
Medical Oncology , Precision Medicine , Dictionaries, Medical as Topic , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy
8.
Ann Oncol ; 27(10): 1959-65, 2016 10.
Article in English | MEDLINE | ID: mdl-27502704

ABSTRACT

BACKGROUND: The application of precision medicine in oncology requires in-depth characterisation of a patient's tumours and the dynamics of their responses to treatment. PATIENTS AND METHODS: We used next-generation sequencing of circulating cell-free DNA (cfDNA) to monitor the response of a KIT p.L576P-mutant metastatic vaginal mucosal melanoma to sequential targeted, immuno- and chemotherapy. RESULTS: Despite a KIT mutation, the response to imatinib was mixed. Unfortunately, tumours were not accessible for molecular analysis. To study the mechanism underlying the mixed clinical response, we carried out whole-exome sequencing and targeted longitudinal analysis of cfDNA. This revealed two tumour subclones; one with a KIT mutation that responded to imatinib and a second KIT-wild-type subclone that did not respond to imatinib. Notably, the subclones also responded differently to immunotherapy. However, both subclones responded to carboplatin/paclitaxel, and although the KIT-wild-type subclone progressed after chemotherapy, it responded to subsequent re-administration of paclitaxel. CONCLUSION: We show that cfDNA can reveal tumour evolution and subclonal responses to therapy even when biopsies are not available.


Subject(s)
Cell-Free Nucleic Acids/genetics , Melanoma/drug therapy , Proto-Oncogene Proteins c-kit/genetics , Vaginal Neoplasms/drug therapy , Adult , Aged , Biomarkers, Pharmacological , Carboplatin/administration & dosage , Cell-Free Nucleic Acids/drug effects , DNA, Neoplasm/drug effects , DNA, Neoplasm/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Imatinib Mesylate/administration & dosage , Melanoma/genetics , Melanoma/pathology , Middle Aged , Mutation , Paclitaxel/administration & dosage , Precision Medicine , Vaginal Neoplasms/genetics , Vaginal Neoplasms/pathology , Exome Sequencing
9.
Ann Oncol ; 27(8): 1443-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27143638

ABSTRACT

Recent advances in biotechnologies have led to the development of multiplex genomic and proteomic analyses for clinical use. Nevertheless, guidelines are currently lacking to determine which molecular assays should be implemented in metastatic cancers. The first MAP conference was dedicated to exploring the use of genomics to better select therapies in the treatment of metastatic cancers. Sixteen consensus items were covered. There was a consensus that new technologies like next-generation sequencing of tumors and ddPCR on circulating free DNA have convincing analytical validity. Further work needs to be undertaken to establish the clinical utility of liquid biopsies and the added clinical value of expanding from individual gene tests into large gene panels. Experts agreed that standardized bioinformatics methods for biological interpretation of genomic data are needed and that precision medicine trials should be stratified based on the level of evidence available for the genomic alterations identified.


Subject(s)
Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Proteomics , Genomics , High-Throughput Nucleotide Sequencing , Humans , Neoplasms/pathology , Precision Medicine
10.
Cell Death Dis ; 7: e2177, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27054332

ABSTRACT

B-cell acute lymphoblastic leukemia (B-ALL) is an aggressive hematological disease that kills ~50% of adult patients. With the exception of some BCR-ABL1(+) patients who benefit from tyrosine kinase inhibitors, there are no effective targeted therapies for adult B-ALL patients and chemotherapy remains first-line therapy despite adverse side effects and poor efficacy. We show that, although the MEK/ERK pathway is activated in B-ALL cells driven by different oncogenes, MEK inhibition does not suppress B-ALL cell growth. However, MEK inhibition synergized with BCL-2/BCL-XL family inhibitors to suppress proliferation and induce apoptosis in B-ALL cells. We show that this synergism is mediated by the pro-apoptotic factor BIM, which is dephosphorylated as a result of MEK inhibition, allowing it to bind to and neutralize MCL-1, thereby enhancing BCL-2/BCL-XL inhibitor-induced cell death. This cooperative effect is observed in B-ALL cells driven by a range of genetic abnormalities and therefore has significant therapeutic potential.


Subject(s)
Bcl-2-Like Protein 11/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , Apoptosis/drug effects , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , RNA Interference , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Thioglycolates/pharmacology , Thioglycolates/therapeutic use , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics , bcl-X Protein/metabolism
11.
Ann Oncol ; 26(6): 1180-1187, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25712455

ABSTRACT

BACKGROUND: Chronic myeloid leukaemia (CML) is characterised by the presence of a fusion driver oncogene, BCR-ABL1, which is a constitutive tyrosine kinase. Tyrosine kinase inhibitors (TKIs) are the central treatment strategy for CML patients and have significantly improved survival rates, but the T315I mutation in the kinase domain of BCR-ABL1 confers resistance to all clinically approved TKIs, except ponatinib. However, compound mutations can mediate resistance even to ponatinib and remain a clinical challenge in CML therapy. Here, we investigated a ponatinib-resistant CML patient through whole-genome sequencing (WGS) to identify the cause of resistance and to find alternative therapeutic targets. PATIENTS AND METHODS: We carried out WGS on a ponatinib-resistant CML patient and demonstrated an effective combination therapy against the primary CML cells derived from this patient in vitro. RESULTS: Our findings demonstrate the emergence of compound mutations in the BCR-ABL1 kinase domain following ponatinib treatment, and chromosomal structural variation data predicted amplification of BCL2. The primary CD34(+) CML cells from this patient showed increased sensitivity to the combination of ponatinib and ABT-263, a BCL2 inhibitor with a negligible effect against the normal CD34(+) cells. CONCLUSION: Our results show the potential of personalised medicine approaches in TKI-resistant CML patients and provide a strategy that could improve clinical outcomes for these patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Imidazoles/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Precision Medicine , Protein Kinase Inhibitors/therapeutic use , Pyridazines/therapeutic use , Aged , Aniline Compounds/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , DNA Mutational Analysis , Drug Resistance, Neoplasm/genetics , Drug Screening Assays, Antitumor , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Genome-Wide Association Study , Humans , Imidazoles/adverse effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Molecular Targeted Therapy , Mutation , Predictive Value of Tests , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Pyridazines/adverse effects , Sulfonamides/therapeutic use , Treatment Failure , Tumor Cells, Cultured
12.
Ann Oncol ; 25(5): 959-67, 2014 May.
Article in English | MEDLINE | ID: mdl-24504448

ABSTRACT

BACKGROUND: BRAF is mutated in ∼42% of human melanomas (COSMIC. http://www.sanger.ac.uk/genetics/CGP/cosmic/) and pharmacological BRAF inhibitors such as vemurafenib and dabrafenib achieve dramatic responses in patients whose tumours harbour BRAF(V600) mutations. Objective responses occur in ∼50% of patients and disease stabilisation in a further ∼30%, but ∼20% of patients present primary or innate resistance and do not respond. Here, we investigated the underlying cause of treatment failure in a patient with BRAF mutant melanoma who presented primary resistance. METHODS: We carried out whole-genome sequencing and single nucleotide polymorphism (SNP) array analysis of five metastatic tumours from the patient. We validated mechanisms of resistance in a cell line derived from the patient's tumour. RESULTS: We observed that the majority of the single-nucleotide variants identified were shared across all tumour sites, but also saw site-specific copy-number alterations in discrete cell populations at different sites. We found that two ubiquitous mutations mediated resistance to BRAF inhibition in these tumours. A mutation in GNAQ sustained mitogen-activated protein kinase (MAPK) signalling, whereas a mutation in PTEN activated the PI3 K/AKT pathway. Inhibition of both pathways synergised to block the growth of the cells. CONCLUSIONS: Our analyses show that the five metastases arose from a common progenitor and acquired additional alterations after disease dissemination. We demonstrate that a distinct combination of mutations mediated primary resistance to BRAF inhibition in this patient. These mutations were present in all five tumours and in a tumour sample taken before BRAF inhibitor treatment was administered. Inhibition of both pathways was required to block tumour cell growth, suggesting that combined targeting of these pathways could have been a valid therapeutic approach for this patient.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Melanoma/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/genetics , Adult , Antineoplastic Agents/therapeutic use , DNA Mutational Analysis , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gq-G11 , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Male , Melanoma/drug therapy , Melanoma/secondary , Mutation, Missense , PTEN Phosphohydrolase/genetics , Polymorphism, Single Nucleotide , Precision Medicine , Proto-Oncogene Proteins B-raf/genetics , Sequence Deletion , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Treatment Outcome , Tumor Cells, Cultured
14.
Oncogene ; 32(26): 3207-12, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-22926515

ABSTRACT

The RAF proteins are cytosolic protein kinases that regulate cell responses to extracellular signals. There are three RAF proteins in cells, ARAF, BRAF and CRAF, and recent studies have shown that the formation of complexes by these different isoforms has an important role in their activation, particularly in response to RAF inhibitors. Here, we investigated the role of ARAF in cancer cell signaling and examined the role of ARAF in mediating paradoxical activation of the MAPK pathway in cells treated with RAF inhibitors. We show that two mutations that occur in ARAF in cancer inactivate the kinase. We also show that ARAF is not functionally redundant with CRAF and cannot substitute for CRAF downstream of RAS. We further show that ARAF binds to and is activated by BRAF and that ARAF also forms complexes with CRAF. Critically, ARAF seems to stabilize BRAF:CRAF complexes in cells treated with RAF inhibitors and thereby regulate cell signaling in a subtle manner to ensure signaling efficiency.


Subject(s)
Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins c-raf/metabolism , raf Kinases/chemistry , Cell Line, Tumor , Enzyme Activation , Humans , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Protein Isoforms , Protein Multimerization , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/genetics , Signal Transduction , raf Kinases/antagonists & inhibitors , raf Kinases/genetics , raf Kinases/metabolism
15.
Br J Cancer ; 105(3): 353-9, 2011 Jul 26.
Article in English | MEDLINE | ID: mdl-21750549

ABSTRACT

METHOD: The safety of oral sorafenib up to a maximum protocol-specified dose combined with dacarbazine in patients with metastatic, histologically confirmed melanoma was investigated in a phase I dose-escalation study and the activity of the combination was explored in an open-label phase II study. RESULTS: In the phase I study, three patients were treated with sorafenib 200 mg twice daily (b.i.d.) plus 1000 mg m(-2) dacarbazine on day 1 of a 21-day cycle and 15 patients had the sorafenib dose escalated to 400 mg b.i.d. without reaching the maximum tolerated dose of the combination. In the phase II study (n=83), the overall response rate was 12% (95% CI: 6, 21): one complete and nine partial, with median response duration of 46.7 weeks. Stable disease was the best response in 37%; median duration was 13.3 weeks. Median overall survival (OS) was 37.0 weeks (95% CI: 33.9, 46.0). CONCLUSION: Oral sorafenib combined with dacarbazine had acceptable toxicity and some antineoplastic activity against metastatic melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzenesulfonates/administration & dosage , Dacarbazine/administration & dosage , Melanoma/drug therapy , Pyridines/administration & dosage , Skin Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Disease-Free Survival , Drug Administration Schedule , Female , Humans , Male , Maximum Tolerated Dose , Melanoma/mortality , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Niacinamide/analogs & derivatives , Phenylurea Compounds , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Sorafenib
16.
Oncogene ; 29(36): 4989-5005, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20639898

ABSTRACT

Hepatocellular carcinoma (HCC) is a highly prevalent, treatment-resistant malignancy with a multifaceted molecular pathogenesis. Current evidence indicates that during hepatocarcinogenesis, two main pathogenic mechanisms prevail: (1) cirrhosis associated with hepatic regeneration after tissue damage caused by hepatitis infection, toxins (for example, alcohol or aflatoxin) or metabolic influences, and (2) mutations occurring in single or multiple oncogenes or tumor suppressor genes. Both mechanisms have been linked with alterations in several important cellular signaling pathways. These pathways are of interest from a therapeutic perspective, because targeting them may help to reverse, delay or prevent tumorigenesis. In this review, we explore some of the major pathways implicated in HCC. These include the RAF/MEK/ERK pathway, phosphatidylinositol-3 kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, WNT/beta-catenin pathway, insulin-like growth factor pathway, hepatocyte growth factor/c-MET pathway and growth factor-regulated angiogenic signaling. We focus on the role of these pathways in hepatocarcinogenesis, how they are altered, and the consequences of these abnormalities. In addition, we also review the latest preclinical and clinical data on the rationally designed targeted agents that are now being directed against these pathways, with early evidence of success.


Subject(s)
Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/ethnology , Liver Neoplasms/etiology , Liver Neoplasms/therapy , Signal Transduction/physiology , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , ErbB Receptors/genetics , ErbB Receptors/physiology , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/physiology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MAP Kinase Signaling System/physiology , Models, Biological , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/physiology , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/physiology , Signal Transduction/genetics , Somatomedins/genetics , Somatomedins/physiology
17.
Br J Cancer ; 95(5): 581-6, 2006 Sep 04.
Article in English | MEDLINE | ID: mdl-16880785

ABSTRACT

The effects of sorafenib--an oral multikinase inhibitor targeting the tumour and tumour vasculature--were evaluated in patients with advanced melanoma enrolled in a large multidisease Phase II randomised discontinuation trial (RDT). Enrolled patients received a 12-week run-in of sorafenib 400 mg twice daily (b.i.d.). Patients with changes in bi-dimensional tumour measurements <25% from baseline were then randomised to sorafenib or placebo for a further 12 weeks (ie to week 24). Patients with > or =25% tumour shrinkage after the run-in continued on open-label sorafenib, whereas those with > or =25% tumour growth discontinued treatment. This analysis focussed on secondary RDT end points: changes in bi-dimensional tumour measurements from baseline after 12 weeks and overall tumour responses (WHO criteria) at week 24, progression-free survival (PFS), safety and biomarkers (BRAF, KRAS and NRAS mutational status). Of 37 melanoma patients treated during the run-in phase, 34 were evaluable for response: one had > or =25% tumour shrinkage and remained on open-label sorafenib; six (16%) had <25% tumour growth and were randomised (placebo, n=3; sorafenib, n=3); and 27 had > or =25% tumour growth and discontinued. All three randomised sorafenib patients progressed by week 24; one remained on sorafenib for symptomatic relief. All three placebo patients progressed by week-24 and were re-started on sorafenib; one experienced disease re-stabilisation. Overall, the confirmed best responses for each of the 37 melanoma patients who received sorafenib were 19% stable disease (SD) (ie n=1 open-label; n=6 randomised), 62% (n=23) progressive disease (PD) and 19% (n=7) unevaluable. The overall median PFS was 11 weeks. The six randomised patients with SD had overall PFS values ranging from 16 to 34 weeks. The most common drug-related adverse events were dermatological (eg rash/desquamation, 51%; hand-foot skin reaction, 35%). There was no relationship between V600E BRAF status and disease stability. DNA was extracted from the biopsies of 17/22 patients. Six had V600E-positive tumours (n=4 had PD; n=1 had SD; n=1 unevaluable for response), and 11 had tumours containing wild-type BRAF (n=9 PD; n=1 SD; n=1 unevaluable for response). In conclusion, sorafenib is well tolerated but has little or no antitumour activity in advanced melanoma patients as a single agent at the dose evaluated (400 mg b.i.d.). Ongoing trials in advanced melanoma are evaluating sorafenib combination therapies.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Benzenesulfonates/therapeutic use , Melanoma/drug therapy , Pyridines/therapeutic use , Adult , Aged , Aged, 80 and over , Angiogenesis Inhibitors/toxicity , Benzenesulfonates/toxicity , DNA Primers , Female , Genes, ras , Humans , Male , Melanoma/blood supply , Melanoma/genetics , Melanoma/pathology , Middle Aged , Neoplasm Staging , Niacinamide/analogs & derivatives , Phenylurea Compounds , Polymerase Chain Reaction , Proto-Oncogene Proteins B-raf/genetics , Pyridines/toxicity , Safety , Sorafenib
18.
Br J Cancer ; 95(4): 496-505, 2006 Aug 21.
Article in English | MEDLINE | ID: mdl-16880792

ABSTRACT

Cellular senescence, the irreversible proliferative arrest seen in somatic cells after a limited number of divisions, is considered a crucial barrier to cancer, but direct evidence for this in vivo was lacking until recently. The best-known form of human cell senescence is attributed to telomere shortening and a DNA-damage response through p53 and p21. There is also a more rapid form of senescence, dependent on the p16-retinoblastoma pathway. p16 (CDKN2A) is a known melanoma susceptibility gene. Here, we use retrovirally mediated gene transfer to confirm that the normal form of senescence in cultured human melanocytes involves p16, since disruption of the p16/retinoblastoma pathway is required as well as telomerase activation for immortalisation. Expression (immunostaining) patterns of senescence mediators and markers in melanocytic lesions provide strong evidence that cell senescence occurs in benign melanocytic naevi (moles) in vivo and does not involve p53 or p21 upregulation, although p16 is widely expressed. In comparison, dysplastic naevi and early (radial growth-phase, RGP) melanomas show less p16 and some p53 and p21 immunostaining. All RGP melanomas expressed p21, suggesting areas of p53-mediated senescence, while most areas of advanced (vertical growth-phase) melanomas lacked both p16 and p21, implying escape from both forms of senescence (immortalisation). Moreover, nuclear p16 but not p21 expression can be induced in human melanocytes by oncogenic BRAF, as found in around 80% of naevi. We conclude that cell senescence can form a barrier to melanoma development. This also provides a potential explanation of why p16 is a melanoma suppressor gene.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/physiology , Melanoma/pathology , Nevus/pathology , Skin Neoplasms/pathology , Cell Survival , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Progression , Humans , Melanocytes/metabolism , Proto-Oncogene Proteins B-raf/pharmacology , Tumor Suppressor Protein p53/metabolism
19.
J Med Chem ; 47(10): 2651-8, 2004 May 06.
Article in English | MEDLINE | ID: mdl-15115406

ABSTRACT

Nine new nitrogen mustard compounds derived from 2,6-difluoro-4-hydroxy- (3a-e) and 2,6-difluoro-4-amino- (4a-d) aniline were synthesized as potential prodrugs. They were designed to be activated to their corresponding 3,5-difluorophenol and -aniline (4)-nitrogen mustards by the enzyme carboxypeptidase G2 (CPG2) in gene-directed enzyme prodrug therapy (GDEPT) models. The compounds were tested for cytotoxicity in the MDA MB-361 breast adenocarcinoma. The cell line was engineered to express stably either CPG2 tethered to the cell surface stCPG2-(Q)3 or beta-galactosidase (beta-Gal) as control. The cytotoxicity differentials were calculated between CPG 2-expressing and -nonexpressing cells and yielded different results for the two series of prodrugs despite their structural similarities. While the phenol compounds are ineffective as prodrugs, their aniline counterparts exhibit outstanding activity in the tumor cell lines expressing CPG2. [3,5-Difluoro-4-[bis(2-chloroethyl)amino]phenyl]carbamoyl-l-glutamic acid gave a differential of >227 in MDA MB361 cells as compared with 19 exhibited by 4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoyl-l-glutamic acid, 1a, which has been in clinical trials.


Subject(s)
Antineoplastic Agents/metabolism , Glutamic Acid/metabolism , Nitrogen Mustard Compounds/metabolism , Prodrugs/metabolism , gamma-Glutamyl Hydrolase/metabolism , Aniline Mustard/analogs & derivatives , Aniline Mustard/chemical synthesis , Aniline Mustard/metabolism , Aniline Mustard/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Benzene Derivatives/chemical synthesis , Benzene Derivatives/metabolism , Benzene Derivatives/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Genetic Therapy , Glutamic Acid/analogs & derivatives , Glutamic Acid/chemical synthesis , Glutamic Acid/pharmacology , Half-Life , Humans , Mice , Neoplasm Transplantation , Nitrogen Mustard Compounds/chemical synthesis , Nitrogen Mustard Compounds/pharmacology , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Structure-Activity Relationship , Transplantation, Heterologous , gamma-Glutamyl Hydrolase/chemistry , gamma-Glutamyl Hydrolase/genetics
20.
Br J Cancer ; 88(10): 1622-30, 2003 May 19.
Article in English | MEDLINE | ID: mdl-12771932

ABSTRACT

We have generated fusion proteins between vascular endothelial growth factor (VEGF) and the bacterial enzyme carboxypeptidase G2 (CPG2) that can activate the prodrug 4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoyl-L-glutamic acid (CMDA). Three asparagine residues of CPG2 were mutated to glutamine (CPG2(Q)3) to prevent glycosylation during secretion, and truncations of VEGF(165) were fused to either the C- or N-terminal of CPG2. The K(m) of the fusion proteins (37.5 microM) was similar to that of secreted CPG2(Q)3 (29.5 microM) but greater than that of wild-type CPG2 (8 microM). The affinity of the fusion proteins for VEGF receptor-2 (VEGFR2) (K(d)=0.5-1.1 nM) was similar to that of [(125)I]VEGF (K(d)=0.5 nM) (ELISA) or slightly higher (K(d)=1.3-9.6 nM) (competitive RIA). One protein, VEGF(115)-CPG2(Q)3-H(6), possessed 140% of the enzymic activity of secreted CPG2(Q)3, and had a faster half-maximal binding time for VEGFR2 (77 s), than the other candidates (330 s). In vitro, VEGF(115)-CPG2(Q)3-H(6) targeted CMDA cytotoxicity only towards VEGFR-expressing cells. The plasma half-life of VEGF(115)-CPG2(Q)3-H(6) in vivo was 3 h, comparable to equivalent values observed in ADEPT. We conclude that enzyme prodrug therapy using VEGF as a targeting moiety represents a promising novel antitumour therapy, with VEGF(115)-CPG2(Q)3-H(6) being a lead candidate.


Subject(s)
Endothelial Growth Factors/pharmacology , Glutamates/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Lymphokines/pharmacology , Nitrogen Mustard Compounds/pharmacology , Prodrugs/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , gamma-Glutamyl Hydrolase/pharmacology , Adenocarcinoma/pathology , Endothelial Growth Factors/genetics , Endothelium/cytology , Female , Glutamine , Humans , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/genetics , Mutagenesis, Site-Directed , Neovascularization, Pathologic , Ovarian Neoplasms/pathology , Plasmids , Point Mutation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-2/drug effects , Vascular Endothelial Growth Factors , gamma-Glutamyl Hydrolase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL