Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Nat Med ; 30(3): 785-796, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38365950

ABSTRACT

Multiple clinical trials targeting the gut microbiome are being conducted to optimize treatment outcomes for immune checkpoint blockade (ICB). To improve the success of these interventions, understanding gut microbiome changes during ICB is urgently needed. Here through longitudinal microbiome profiling of 175 patients treated with ICB for advanced melanoma, we show that several microbial species-level genome bins (SGBs) and pathways exhibit distinct patterns from baseline in patients achieving progression-free survival (PFS) of 12 months or longer (PFS ≥12) versus patients with PFS shorter than 12 months (PFS <12). Out of 99 SGBs that could discriminate between these two groups, 20 were differentially abundant only at baseline, while 42 were differentially abundant only after treatment initiation. We identify five and four SGBs that had consistently higher abundances in patients with PFS ≥12 and <12 months, respectively. Constructing a log ratio of these SGBs, we find an association with overall survival. Finally, we find different microbial dynamics in different clinical contexts including the type of ICB regimen, development of immune-related adverse events and concomitant medication use. Insights into the longitudinal dynamics of the gut microbiome in association with host factors and treatment regimens will be critical for guiding rational microbiome-targeted therapies aimed at enhancing ICB efficacy.


Subject(s)
Gastrointestinal Microbiome , Melanoma , Microbiota , Humans , Gastrointestinal Microbiome/genetics , Melanoma/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Cognition
2.
Ther Adv Med Oncol ; 15: 17588359231160140, 2023.
Article in English | MEDLINE | ID: mdl-36970111

ABSTRACT

Tebentafusp is a first-in-class immunotherapy agent that comprises an engineered T-cell receptor targeting a gp100 epitope presented by human leukocyte antigen-A*02:01 cells, fused to an anti-CD3 single-chain variable fragment. Tebentafusp is both the first bispecific T-cell engager to show efficacy in the treatment of advanced solid cancer and the first anti-cancer treatment to demonstrate an overall survival benefit in patients with uveal melanoma (UM). This review article will focus on the clinical development of tebentafusp, the mechanism of action and resultant evolution of the management of advanced UM.

3.
BMC Cancer ; 23(1): 166, 2023 Feb 18.
Article in English | MEDLINE | ID: mdl-36805683

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of melanoma and other cancers. However, no reliable biomarker of survival or response has entered the clinic to identify those patients with melanoma who are most likely to benefit from ICIs. Glycosylation affects proteins and lipids' structure and functions. Tumours are characterized by aberrant glycosylation which may contribute to their progression and hinder an effective antitumour immune response. METHODS: We aim at identifying novel glyco-markers of response and survival by leveraging the N-glycome of total serum proteins collected in 88 ICI-naive patients with advanced melanoma from two European countries. Samples were collected before and during ICI treatment. RESULTS: We observe that responders to ICIs present with a pre-treatment N-glycome profile significantly shifted towards higher abundancy of low-branched structures containing lower abundances of antennary fucose, and that this profile is positively associated with survival and a better predictor of response than clinical variables alone. CONCLUSION: While changes in serum protein glycosylation have been previously implicated in a pro-metastatic melanoma behaviour, we show here that they are also associated with response to ICI, opening new avenues for the stratification of patients and the design of adjunct therapies aiming at improving immune response.


Subject(s)
Immune Checkpoint Inhibitors , Melanoma , Humans , Melanoma/drug therapy , Ambulatory Care Facilities , Europe , Polysaccharides
4.
EBioMedicine ; 83: 104235, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36007304

ABSTRACT

BACKGROUND: Inflammation can modulate tumour growth and progression, and influence clinical response to treatment. We investigated the potential of circulating inflammatory proteins for response stratification of immune checkpoint inhibitor (ICI) therapy for advanced melanoma. METHODS: Study subjects were 87 patients with unresectable stage III or IV cutaneous melanoma from the multiple centres across the United Kingdom (UK) and the Netherlands (NL) who received ipilimumab, nivolumab, or pembrolizumab, or a combination of ipilimumab and nivolumab. Serum samples were collected before and during ICI therapy at follow-up visits scheduled every third week over a 12-week period. We performed targeted quantification of 92 proteins involved in inflammation and tested for association of their pre-treatment and on-treatment levels, as well as longitudinal changes, with overall response rate, progression-free survival, and overall survival. FINDINGS: We observed consistently higher pre-treatment levels of interleukin-6 (IL-6), hepatocyte growth factor (HGF), and monocyte chemotactic protein 2 (MCP-2), in non-responders compared to responders (meta-analysis p=3.31 × 10-4, 2.29 × 10-4, and 1.02 × 10-3, respectively). Patients' stratification according to the median value of IL-6, HGF, and MCP-2 highlighted a cumulative negative effect of pre-treatment levels of the three proteins on response (p=1.13 × 10-2), with overall response rate among patients presenting with combined elevated IL-6, HGF, and MCP-2 levels being three-fold lower (26.7%) compared to patients with none of the three proteins elevated (80.0%, p=9.22 × 10-3). Longitudinal data analysis showed that on-treatment changes in circulating inflammatory proteins are not correlated with response. INTERPRETATION: Our findings are in line with an increasing body of evidence that the pro-inflammatory cytokine IL-6 can influence response to ICI in advanced melanoma, and further support a role of circulating HGF and MCP-2 levels as prognostic biomarkers as suggested by previous smaller studies. Inflammatory proteins may serve as predictive biomarkers of ICI response and valuable targets for combination therapy. FUNDING: This work was supported by the Seerave Foundation and Dutch Cancer Society.


Subject(s)
Melanoma , Skin Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemokine CCL8 , Hepatocyte Growth Factor , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Inflammation/drug therapy , Interleukin-6 , Ipilimumab/therapeutic use , Melanoma/pathology , Nivolumab , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology
5.
Nat Med ; 28(3): 535-544, 2022 03.
Article in English | MEDLINE | ID: mdl-35228751

ABSTRACT

The composition of the gut microbiome has been associated with clinical responses to immune checkpoint inhibitor (ICI) treatment, but there is limited consensus on the specific microbiome characteristics linked to the clinical benefits of ICIs. We performed shotgun metagenomic sequencing of stool samples collected before ICI initiation from five observational cohorts recruiting ICI-naive patients with advanced cutaneous melanoma (n = 165). Integrating the dataset with 147 metagenomic samples from previously published studies, we found that the gut microbiome has a relevant, but cohort-dependent, association with the response to ICIs. A machine learning analysis confirmed the link between the microbiome and overall response rates (ORRs) and progression-free survival (PFS) with ICIs but also revealed limited reproducibility of microbiome-based signatures across cohorts. Accordingly, a panel of species, including Bifidobacterium pseudocatenulatum, Roseburia spp. and Akkermansia muciniphila, associated with responders was identified, but no single species could be regarded as a fully consistent biomarker across studies. Overall, the role of the human gut microbiome in ICI response appears more complex than previously thought, extending beyond differing microbial species simply present or absent in responders and nonresponders. Future studies should adopt larger sample sizes and take into account the complex interplay of clinical factors with the gut microbiome over the treatment course.


Subject(s)
Gastrointestinal Microbiome , Melanoma , Skin Neoplasms , Gastrointestinal Microbiome/genetics , Humans , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/drug therapy , Melanoma/genetics , Reproducibility of Results , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics
6.
Eur J Cancer ; 138: 149-155, 2020 10.
Article in English | MEDLINE | ID: mdl-32889369

ABSTRACT

Immune-checkpoint inhibitors (ICIs) have revolutionised the therapeutic landscape for multiple malignancies and the health of the gut microbiome (GM) is strongly linked with therapeutic responses to ICI. This review explores the implications of diet and medication on the GM for patients receiving ICI. Clinical trials are underway to explore the impact of factors such as faecal microbiota transfer, probiotics, prebiotics, bacteria consortia and a number of dietary interventions on patients receiving ICI. Randomised controlled trials are lacking, and inferences are currently based on short-term clinical and observational studies. Antibiotics should be avoided before ICI initiation, and depending on prospective data, future consideration may be given to temporary delay of initiation of non-urgent ICI if patient has had broad spectrum antibiotics within 1 month of planned treatment initiation. Proton pump inhibitor use should be discontinued when not clearly indicated and potential switch to a histamine H2-receptor antagonist considered. Patients should be advised to minimise animal meat intake and maximise plants, aiming to consume ≥30 plant types weekly. A high fibre intake (>30 g/day) has been seen to be beneficial in increasing the chance of ICI response. Fermented foods may have a beneficial effect on the GM and should be introduced where possible. Ideally, all patients should be referred to a nutritionist or dietician with knowledge of GM before commencing ICI.


Subject(s)
Bacteria/drug effects , Diet , Gastrointestinal Microbiome/drug effects , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Neoplasms/drug therapy , Animals , Anti-Bacterial Agents/adverse effects , Bacteria/immunology , Diet/adverse effects , Dietary Supplements , Dysbiosis , Fecal Microbiota Transplantation , Host-Pathogen Interactions , Humans , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Neoplasms/immunology , Neoplasms/microbiology , Nutritional Status , Proton Pump Inhibitors/adverse effects
7.
Melanoma Res ; 29(1): 102-106, 2019 02.
Article in English | MEDLINE | ID: mdl-30395075

ABSTRACT

Immune checkpoint inhibitor (IO) induced colitis is primarily managed with corticosteroids. Most patients have a rapid resolution of symptoms and do not require additional immunosuppressants. Many patients, however, require prolonged corticosteroid courses to maintain control of toxicity. Mycophenolate mofetil (MMF) is the prodrug of mycophenolic acid; which in turn directly inhibits activated T and B lymphocytes. MMF, in addition to corticosteroids, may enable reduction of corticosteroids without precipitating resurgence of colitis. Metastatic melanoma patients between 1 January 2017 and 31 December 2017 with combination IO-induced colitis were managed with a novel treatment algorithm: upfront oral enteric-coated MMF alongside high-dose corticosteroids. Outcome measures included incidence of colitis flare, time to grade 1 colitis, time to patient-reported normal bowel habit and overall cumulative corticosteroid exposure. Thirteen patients developed high-grade combination IO-induced colitis; 11 were managed with the combination of high-dose corticosteroid and MMF. Median patient age was 59 (range: 28-73) years. Four (36%) developed flare of colitis; flares occurred at a median of 11 (interquartile range: 4.5-16.75) days. All colitis flares responded fully to infliximab (5 mg/kg). The remaining seven patients did not develop colitis flare during corticosteroid wean. All patients were successfully weaned from corticosteroids and none had a resurgence of colitis at 8 weeks following discontinuation of MMF. Concomitant enteric-coated MMF alongside high-dose corticosteroids may hasten the improvement of high-grade colitis to normal bowel habit and reduce the incidence of colitis flare.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Colitis/drug therapy , Melanoma/drug therapy , Mycophenolic Acid/therapeutic use , Skin Neoplasms/drug therapy , Adult , Aged , Colitis/chemically induced , Disease Management , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Ipilimumab/administration & dosage , Male , Melanoma/pathology , Middle Aged , Nivolumab/administration & dosage , Prognosis , Prospective Studies , Skin Neoplasms/secondary
8.
Onco Targets Ther ; 6: 1197-206, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-24043945

ABSTRACT

Olaparib has shown promising anticancer activity as a single agent in the treatment and maintenance of recurrent ovarian cancer in early clinical trials, but it is far from standard therapy. This article outlines the problem of relapsed ovarian cancer and the mechanisms of poly(ADP-ribose) polymerase inhibitors and reviews the recent literature pertaining to olaparib in ovarian cancer.

9.
Expert Rev Anticancer Ther ; 13(5): 513-22, 2013 May.
Article in English | MEDLINE | ID: mdl-23617343

ABSTRACT

The incidence of malignant melanoma is increasing annually. Early stages can be cured with surgical intervention but metastatic disease has generally had a dismal prognosis with few effective interventions. A half of all melanomas possess a BRAF mutation, which can be targeted by specific inhibitors. Vemurafenib is an orally active, purposely designed mutant BRAF inhibitor, which has recently been shown to have a survival benefit measured in months in metastatic patients. In this article, the authors discuss the scientific rationale, drug development process and clinical trials that have led to vemurafenib becoming the first BRAF inhibitor approved for the treatment of patients with mutant BRAF metastatic melanoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Indoles/therapeutic use , Melanoma/drug therapy , Sulfonamides/therapeutic use , Administration, Oral , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Drug Design , Humans , Indoles/adverse effects , Indoles/pharmacology , Melanoma/genetics , Melanoma/pathology , Neoplasm Metastasis , Neoplasm Staging , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/adverse effects , Sulfonamides/pharmacology , Vemurafenib
10.
J Invest Dermatol ; 133(5): 1269-77, 2013 May.
Article in English | MEDLINE | ID: mdl-23190890

ABSTRACT

Approximately 50% of melanomas require oncogenic B-RAF(V600E) signaling for proliferation, survival, and metastasis, and the use of highly selective B-RAF inhibitors has yielded remarkable, although short-term, clinical responses. Reactivation of signaling downstream of B-RAF is frequently associated with acquired resistance to B-RAF inhibitors, and the identification of B-RAF targets may therefore provide new strategies for managing melanoma. In this report, we applied whole-genome expression analyses to reveal that oncogenic B-RAF(V600E) regulates genes associated with epithelial-mesenchymal transition in normal cutaneous human melanocytes. Most prominent was the B-RAF-mediated transcriptional repression of E-cadherin, a keratinocyte-melanoma adhesion molecule whose loss is intimately associated with melanoma invasion and metastasis. Here we identify a link between oncogenic B-RAF, the transcriptional repressor Tbx3, and E-cadherin. We show that B-RAF(V600E) induces the expression of Tbx3, which potently represses E-cadherin expression in melanocytes and melanoma cells. Tbx3 expression is normally restricted to developmental embryonic tissues and promoting cell motility, but it is also aberrantly increased in various cancers and has been linked to tumor cell invasion and metastasis. We propose that this B-RAF/Tbx3/E-cadherin pathway has a critical role in promoting the metastasis of B-RAF-mutant melanomas.


Subject(s)
Cadherins/antagonists & inhibitors , Down-Regulation/physiology , Melanoma/pathology , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction/physiology , Skin Neoplasms/pathology , T-Box Domain Proteins/metabolism , Cadherins/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement/physiology , Cells, Cultured , Epithelial-Mesenchymal Transition/physiology , Humans , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/metabolism , Melanoma/physiopathology , Neoplasm Invasiveness , Skin Neoplasms/metabolism , Skin Neoplasms/physiopathology , Transcription, Genetic/physiology
11.
Mol Biol Cell ; 21(15): 2770-9, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20534814

ABSTRACT

Tbx2 is a member of a large family of transcription factors defined by homology to the T-box DNA-binding domain. Tbx2 plays a key role in embryonic development, and in cancer through its capacity to suppress senescence and promote invasiveness. Despite its importance, little is known of how Tbx2 is regulated or how it achieves target gene specificity. Here we show that Tbx2 specifically associates with active hypophosphorylated retinoblastoma protein (Rb1), a known regulator of many transcription factors involved in cell cycle progression and cellular differentiation, but not with the Rb1-related proteins p107 or p130. The interaction with Rb1 maps to a domain immediately carboxy-terminal to the T-box and enhances Tbx2 DNA binding and transcriptional repression. Microarray analysis of melanoma cells expressing inducible dominant-negative Tbx2, comprising the T-box and either an intact or mutated Rb1 interaction domain, shows that Tbx2 regulates the expression of many genes involved in cell cycle control and that a mutation which disrupts the Rb1-Tbx2 interaction also affects Tbx2 target gene selectivity. Taken together, the data show that Rb1 is an important determinant of Tbx2 functional specificity.


Subject(s)
Retinoblastoma Protein/metabolism , T-Box Domain Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Cell Line , DNA/metabolism , Humans , Melanoma/metabolism , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Repressor Proteins/metabolism , Retinoblastoma Protein/chemistry , T-Box Domain Proteins/chemistry , Transcription, Genetic
12.
Clin Cancer Res ; 15(15): 4978-85, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19638462

ABSTRACT

PURPOSE: To determine the maximum tolerated dose, dose-limiting toxicity, pharmacokinetics, and preliminary therapeutic activity profile of CHR-2797 (tosedostat), a novel, orally bioavailable inhibitor of the M1 family of aminopeptidases with antiproliferative and antiangiogenic activity in vitro. EXPERIMENTAL DESIGN: A phase I study of accelerated titration design that escalated through nine doses (10-320 mg) in patients (Eastern Cooperative Oncology Group performance status, < or =2) with advanced solid tumors. CHR-2797 was administered once daily. RESULTS: Forty patients (median age, 60 years; range, 24-80 years; male, 27; female, 13) were treated in 12 cohorts with once daily doses (10-320 mg). Dose-limiting toxicities were thrombocytopenia, dizziness, and visual abnormalities in one patient, and anemia, blurred vision, and vomiting in a second patient at 320 mg, resulting in an inability to complete 28 days of study drug. The most commonly observed toxicities were fatigue, diarrhea, peripheral edema, nausea, dizziness, and constipation. One patient had a partial response (renal cell carcinoma) and four patients had stable disease for >6 months. CHR-2797 and its active metabolite, CHR-79888, show dose-proportional increases in plasma AUC and C(max). The terminal half-life for CHR-2797 is approximately 1 to 3.5 hours and between 6 and 11 hours for CHR-79888. Intracellular (packed blood cells) exposure to CHR-79888 is consistent with intracellular levels that proved to be efficacious in xenograft models. CONCLUSION: CHR-2797 is well tolerated and can be safely administered at doses that result in intracellular levels of CHR-79888 that are associated with activity in preclinical models. The recommended dose for single agent therapy in solid tumors is 240 mg/d.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Enzyme Inhibitors/pharmacokinetics , Glycine/analogs & derivatives , Hydroxamic Acids/pharmacokinetics , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Female , Glycine/administration & dosage , Glycine/adverse effects , Glycine/pharmacokinetics , Half-Life , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/adverse effects , Male , Middle Aged , Neoplasms/diagnostic imaging , Radiography
13.
Clin Cancer Res ; 13(19): 5834-40, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17908976

ABSTRACT

PURPOSE: This phase I study was undertaken to define the maximum tolerated dose, safety, and pharmacokinetic profile of CP-751,871. EXPERIMENTAL DESIGN: Using a rapid dose escalation design, patients with advanced nonhematologic malignancies were treated with CP-751,871 in four dose escalation cohorts. CP-751,871 was administered i.v. on day 1 of each 21-day cycle. Pharmacokinetic evaluation was done in all treatment cohorts during cycles 1 and 4. RESULTS: Twenty-four patients received 110 cycles at four dose levels. The maximum tolerated dose exceeded the maximal feasible dose of 20 mg/kg and, thus, was not identified. Treatment-related toxicities were generally mild. The most common adverse events were hyperglycemia, anorexia, nausea, elevated aspartate aminotransferase, elevated gamma-glutamyltransferase, diarrhea, hyperuracemia, and fatigue. At 20 mg/kg, 10 of 15 patients experienced stability of disease. Two of these patients experienced long-term stability. There were no objective responses. Pharmacokinetic analysis revealed a dose-dependent increase in CP-751,871 exposure and approximately 2-fold accumulation on repeated dosing in 21-day cycles. Plasma concentrations of CP-751,871 attained were several log-fold greater than the biologically active concentration. Treatment with CP-751,871 increased serum insulin and human growth hormone levels, with modest increases in serum glucose levels. CONCLUSIONS: CP-751,871 has a favorable safety profile and was well tolerated when given in continuous cycles. At the maximal feasible dose of 20 mg/kg, there was a moderate accumulation in plasma exposure, and most of the treated patients experienced stability of disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Neoplasms/drug therapy , Receptor, IGF Type 1/chemistry , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/therapeutic use , Cohort Studies , Female , Growth Hormone/metabolism , Humans , Immunoglobulins, Intravenous , Insulin/metabolism , Male , Maximum Tolerated Dose , Middle Aged , Receptor, IGF Type 1/immunology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...