Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 7270, 2024 03 27.
Article in English | MEDLINE | ID: mdl-38538606

ABSTRACT

Cancer risk is associated with the widely debated measure body mass index (BMI). Fat mass and fat-free mass measurements from bioelectrical impedance may further clarify this association. The UK Biobank is a rare resource in which bioelectrical impedance and BMI data was collected on ~ 500,000 individuals. Using this dataset, a comprehensive analysis using regression, principal component and genome-wide genetic association, provided multiple levels of evidence that increasing whole body fat (WBFM) and fat-free mass (WBFFM) are both associated with increased post-menopausal breast cancer risk, and colorectal cancer risk in men. WBFM was inversely associated with prostate cancer. We also identified rs615029[T] and rs1485995[G] as associated in independent analyses with both PMBC (p = 1.56E-17 and 1.78E-11) and WBFFM (p = 2.88E-08 and 8.24E-12), highlighting splice variants of the intriguing long non-coding RNA CUPID1 (LINC01488) as a potential link between PMBC risk and fat-free mass.


Subject(s)
Body Composition , Neoplasms , Male , Humans , Body Composition/genetics , Body Mass Index , Genetic Predisposition to Disease , Neoplasms/etiology , Neoplasms/genetics , Electric Impedance
3.
Nat Commun ; 14(1): 4444, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37488191

ABSTRACT

Samuraciclib is a selective oral CDK7-inhibitor. A multi-modular, open-label Phase I study to evaluate safety and tolerability of samuraciclib in patients with advanced malignancies was designed (ClinicalTrials.gov: NCT03363893). Here we report results from dose escalation and 2 expansion cohorts: Module 1A dose escalation with paired biopsy cohort in advanced solid tumor patients, Module 1B-1 triple negative breast cancer (TNBC) monotherapy expansion, and Module 2A fulvestrant combination in HR+/HER2- breast cancer patients post-CDK4/6-inhibitor. Core study primary endpoints are safety and tolerability, and secondary endpoints are pharmacokinetics (PK), pharmacodynamic (PD) activity, and anti-tumor activity. Common adverse events are low grade nausea, vomiting, and diarrhea. Maximum tolerated dose is 360 mg once daily. PK demonstrates dose proportionality (120 mg-480 mg), a half-life of approximately 75 hours, and no fulvestrant interaction. In dose escalation, one partial response (PR) is identified with disease control rate of 53% (19/36) and reduction of phosphorylated RNA polymerase II, a substrate of CDK7, in circulating lymphocytes and tumor tissue. In TNBC expansion, one PR (duration 337 days) and clinical benefit rate at 24 weeks (CBR) of 20.0% (4/20) is achieved. In combination with fulvestrant, 3 patients achieve PR with CBR 36.0% (9/25); in patients without detectable TP53-mutation CBR is 47.4% (9/19). In this study, samuraciclib exhibits tolerable safety and PK is supportive of once-daily oral administration. Clinical activity in TNBC and HR+/HER2-breast cancer post-CDK4/6-inhibitor settings warrants further evaluation.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Fulvestrant , Administration, Oral , Biopsy , Cyclin-Dependent Kinase Inhibitor Proteins , Cyclin-Dependent Kinases , Enzyme Inhibitors
4.
Cancer Cell ; 41(7): 1222-1241.e7, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37433281

ABSTRACT

For inoperable esophageal adenocarcinoma (EAC), identifying patients likely to benefit from recently approved immunochemotherapy (ICI+CTX) treatments remains a key challenge. We address this using a uniquely designed window-of-opportunity trial (LUD2015-005), in which 35 inoperable EAC patients received first-line immune checkpoint inhibitors for four weeks (ICI-4W), followed by ICI+CTX. Comprehensive biomarker profiling, including generation of a 65,000-cell single-cell RNA-sequencing atlas of esophageal cancer, as well as multi-timepoint transcriptomic profiling of EAC during ICI-4W, reveals a novel T cell inflammation signature (INCITE) whose upregulation correlates with ICI-induced tumor shrinkage. Deconvolution of pre-treatment gastro-esophageal cancer transcriptomes using our single-cell atlas identifies high tumor monocyte content (TMC) as an unexpected ICI+CTX-specific predictor of greater overall survival (OS) in LUD2015-005 patients and of ICI response in prevalent gastric cancer subtypes from independent cohorts. Tumor mutational burden is an additional independent and additive predictor of LUD2015-005 OS. TMC can improve patient selection for emerging ICI+CTX therapies in gastro-esophageal cancer.


Subject(s)
Adenocarcinoma , Esophageal Neoplasms , Stomach Neoplasms , Humans , Monocytes , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Immunotherapy
5.
Br J Cancer ; 128(6): 958-966, 2023 04.
Article in English | MEDLINE | ID: mdl-36823364

ABSTRACT

Over the past 15 years, there has been great interest in the potential to repurpose the diabetes drug, metformin, as a cancer treatment. However, despite considerable efforts being made to investigate its efficacy in a number of large randomised clinical trials in different tumour types, results have been disappointing to date. This perspective article summarises how interest initially developed in the oncological potential of metformin and the diverse clinical programme of work to date including our contribution to establishing the intra-tumoral pharmacodynamic effects of metformin in the clinic. We also discuss the lessons that can be learnt from this experience and whether a further clinical investigation of metformin in cancer is warranted.


Subject(s)
Metformin , Neoplasms , Humans , Metformin/therapeutic use , Drug Repositioning , Hypoglycemic Agents/therapeutic use , Neoplasms/drug therapy
6.
Cancer Metastasis Rev ; 41(3): 491-515, 2022 09.
Article in English | MEDLINE | ID: mdl-36038791

ABSTRACT

Obesity-related cancers account for 40% of the cancer cases observed in the USA and obesity is overtaking smoking as the most widespread modifiable risk factor for carcinogenesis. Here, we use the hallmarks of cancer framework to delineate how obesity might influence the carcinogenic hallmarks in somatic cells. We discuss the effects of obesity on (a) sustaining proliferative signaling; (b) evading growth suppressors; (c) resisting cell death; (d) enabling replicative immortality; (e) inducing angiogenesis; (f) activating invasion and metastasis; (g) reprogramming energy metabolism; and (h) avoiding immune destruction, together with its effects on genome instability and tumour-promoting inflammation. We present the current understanding and controversies in this evolving field, and highlight some areas in need of further cross-disciplinary focus. For instance, the relative importance of the many potentially causative obesity-related factors is unclear for each type of malignancy. Even within a single tumour type, it is currently unknown whether one obesity-related factor consistently plays a predominant role, or if this varies between patients or, even in a single patient with time. Clarifying how the hallmarks are affected by obesity may lead to novel prevention and treatment strategies for the increasingly obese population.


Subject(s)
Carcinogenesis , Neoplasms , Humans , Neoplasms/metabolism , Neovascularization, Pathologic/pathology , Obesity/complications , Signal Transduction
7.
NPJ Breast Cancer ; 8(1): 45, 2022 Apr 07.
Article in English | MEDLINE | ID: mdl-35393425

ABSTRACT

Platinum derivatives are commonly used for the treatment of patients with metastatic triple-negative breast cancer (TNBC). However, resistance often develops, leading to treatment failure. This expansion cohort (part C2) of the previously reported phase 1b trial (NCT02157792) is based on the recommended phase 2 dose of the combination of the ataxia-telangiectasia and Rad3-related (ATR) inhibitor berzosertib and cisplatin observed in patients with advanced solid tumors, including TNBC. Forty-seven patients aged ≥18 years with advanced TNBC received cisplatin (75 mg/m2; day 1) and berzosertib (140 mg/m2; days 2 and 9), in 21-day cycles. Berzosertib was well tolerated, with a similar toxicity profile to that reported previously for this combination. The overall response rate (90% confidence interval) was 23.4% (13.7, 35.8). No relevant associations were observed between response and gene alterations. Further studies combining ATR inhibitors with platinum compounds may be warranted in highly selected patient populations.

8.
Br J Cancer ; 122(1): 45-51, 2020 01.
Article in English | MEDLINE | ID: mdl-31819180

ABSTRACT

Window of opportunity trials exploit the 'window' of time after cancer diagnosis, typically prior to initiation of cancer therapy. In recent years this study design has become a more regular feature of drug development, as this 'window' provides an opportunity to carry out a thorough pharmacodynamic assessment of a therapy of interest in tumours that are unperturbed by prior treatment. Many of the first window trials interrogated the bioactivity of drugs being repurposed for cancer treatment, in particular the anti-mitochondrial agent, metformin. In this review, we describe examples of window study designs that have been used to assess drugs that target cancer metabolism with a focus on metformin. In addition, we discuss how window studies may aid the development of molecular metabolic cancer imaging.


Subject(s)
Clinical Trials as Topic/methods , Neoplasms/metabolism , Research Design , Time-to-Treatment , Drug Repositioning/methods , Humans , Metabolomics , Metformin/therapeutic use , Neoplasms/drug therapy
9.
Br J Cancer ; 122(2): 258-265, 2020 01.
Article in English | MEDLINE | ID: mdl-31819193

ABSTRACT

BACKGROUND: Epidemiological studies suggest that metformin may reduce the incidence of cancer in patients with diabetes and multiple late phase clinical trials assessing the potential of repurposing this drug are underway. Transcriptomic profiling of tumour samples is an excellent tool to understand drug bioactivity, identify candidate biomarkers and assess for mechanisms of resistance to therapy. METHODS: Thirty-six patients with untreated primary breast cancer were recruited to a window study and transcriptomic profiling of tumour samples carried out before and after metformin treatment. RESULTS: Multiple genes that regulate fatty acid oxidation were upregulated at the transcriptomic level and there was a differential change in expression between two previously identified cohorts of patients with distinct metabolic responses. Increase in expression of a mitochondrial fatty oxidation gene composite signature correlated with change in a proliferation gene signature. In vitro assays showed that, in contrast to previous studies in models of normal cells, metformin reduces fatty acid oxidation with a subsequent accumulation of intracellular triglyceride, independent of AMPK activation. CONCLUSIONS: We propose that metformin at clinical doses targets fatty acid oxidation in cancer cells with implications for patient selection and drug combinations. CLINICAL TRIAL REGISTRATION: NCT01266486.


Subject(s)
Breast Neoplasms/drug therapy , Fatty Acids/metabolism , Metformin/pharmacology , Protein Kinases/genetics , AMP-Activated Protein Kinase Kinases , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Diabetes Mellitus/drug therapy , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Lipid Metabolism/drug effects , Lipid Peroxidation/drug effects , Mice , Mitochondria/drug effects , Oxidation-Reduction/drug effects , Transcriptome/drug effects
10.
Eur J Cancer ; 124: 131-141, 2020 01.
Article in English | MEDLINE | ID: mdl-31765988

ABSTRACT

BACKGROUND: AZD8931 has equipotent activity against epidermal growth factor receptor, erbB2, and erbB3. Primary objectives were to determine the recommended phase II dose (RP2D) of AZD8931 + chemotherapy, and subsequently assess safety/preliminary clinical activity in patients with operable oesophagogastric cancer (OGC). METHODS: AZD8931 (20 mg, 40 mg or 60 mg bd) was given with Xelox (oxaliplatin + capecitabine) for eight 21-day cycles, continuously or with intermittent schedule (4 days on/3 off every week; 14 days on/7 off, per cycle) in a rolling-six design. Subsequently, patients with OGC were randomised 2:1 to AZD8931 + Xelox at RP2D or Xelox only for two cycles, followed by radical oesophagogastric surgery. Secondary outcomes were safety, complete resection (R0) rate, six-month progression-free survival (PFS) and overall survival. RESULTS: During escalation, four dose-limiting toxicities were observed among 24 patients: skin rash (1) and failure to deliver 100% of Xelox because of treatment-associated grade III-IV adverse events (AEs) (3: diarrhoea and vomiting; vomiting; fatigue). Serious adverse events (SAE) occurred in 15 of 24 (63%) patients. RP2D was 20-mg bd with the 4/3 schedule. In the expansion phase, 2 of 20 (10%) patients in the Xelox + AZD8931 group and 5/10 (50%) patients in the Xelox group had grade III-IV AEs. Six-month PFS was 85% (90% CI: 66%-94%) in Xelox + AZD8931 and 100% in Xelox alone. Seven deaths (35%) occurred with Xelox + AZD8931 and one (10%) with Xelox. R0 rate was 45% (9/20) with Xelox + AZD8931 and 90% (9/10) with Xelox-alone (P = 0.024). CONCLUSION: Xelox + AZD8931 (20 mg bd 4/3 days) has an acceptable safety profile administered as neoadjuvant therapy in operable patients with OGC. (Trial registration: EudraCT 2011-003169-13, ISRCTN-68093791).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Capecitabine/administration & dosage , Esophageal Neoplasms/therapy , Oxaloacetates/administration & dosage , Quinazolines/administration & dosage , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-3/antagonists & inhibitors , Stomach Neoplasms/therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Capecitabine/adverse effects , Diarrhea/chemically induced , Diarrhea/epidemiology , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophagogastric Junction/pathology , Esophagogastric Junction/surgery , Exanthema/chemically induced , Exanthema/epidemiology , Fatigue/chemically induced , Fatigue/epidemiology , Female , Humans , Male , Margins of Excision , Maximum Tolerated Dose , Middle Aged , Neoadjuvant Therapy/adverse effects , Neoadjuvant Therapy/methods , Oxaliplatin/administration & dosage , Oxaliplatin/adverse effects , Oxaloacetates/adverse effects , Progression-Free Survival , Quinazolines/adverse effects , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Vomiting/chemically induced , Vomiting/epidemiology
11.
Cell Metab ; 28(5): 679-688.e4, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30244975

ABSTRACT

Late-phase clinical trials investigating metformin as a cancer therapy are underway. However, there remains controversy as to the mode of action of metformin in tumors at clinical doses. We conducted a clinical study integrating measurement of markers of systemic metabolism, dynamic FDG-PET-CT, transcriptomics, and metabolomics at paired time points to profile the bioactivity of metformin in primary breast cancer. We show metformin reduces the levels of mitochondrial metabolites, activates multiple mitochondrial metabolic pathways, and increases 18-FDG flux in tumors. Two tumor groups are identified with distinct metabolic responses, an OXPHOS transcriptional response (OTR) group for which there is an increase in OXPHOS gene transcription and an FDG response group with increased 18-FDG uptake. Increase in proliferation, as measured by a validated proliferation signature, suggested that patients in the OTR group were resistant to metformin treatment. We conclude that mitochondrial response to metformin in primary breast cancer may define anti-tumor effect.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Hypoglycemic Agents/pharmacology , Metabolic Networks and Pathways/drug effects , Metformin/pharmacology , Adult , Aged , Antineoplastic Agents/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Glucose/analogs & derivatives , Glucose/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Middle Aged , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Positron Emission Tomography Computed Tomography , Transcriptome/drug effects
12.
Br J Hosp Med (Lond) ; 76(7): 409-13, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26140560

ABSTRACT

The main role of early phase clinical trials in cancer is to determine the dose to take forward for future clinical study. However, study design is changing in order to account for the change in focus of drug development toward molecularly targeted agents.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Clinical Trials, Phase I as Topic/ethics , Clinical Trials, Phase I as Topic/methods , Neoplasms/drug therapy , Antineoplastic Agents/pharmacokinetics , Dose-Response Relationship, Drug , Humans , Maximum Tolerated Dose
13.
J Natl Cancer Inst Monogr ; 2015(51): 81-6, 2015 May.
Article in English | MEDLINE | ID: mdl-26063894

ABSTRACT

There has been growing interest in the potential of the altered metabolic state typical of cancer cells as a drug target. The antidiabetes drug, metformin, is now under intense investigation as a safe method to modify cancer metabolism. Several studies have used window of opportunity in breast cancer patients before neoadjuvant chemotherapy to correlate gene expression analysis, metabolomics, immunohistochemical markers, and metabolic serum markers with those likely to benefit. We review the role metabolite measurement, functional imaging and gene sequencing analysis play in elucidating the effects of metabolically targeted drugs in cancer treatment and determining patient selection.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Metformin/therapeutic use , Neoadjuvant Therapy/methods , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Chemotherapy, Adjuvant/methods , Female , Humans , Hypoglycemic Agents/therapeutic use , Metabolome/drug effects , Transcriptome/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL