Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Tom J. G. Gevers is active.

Publication


Featured researches published by Tom J. G. Gevers.


Nature Reviews Gastroenterology & Hepatology | 2013

Diagnosis and management of polycystic liver disease

Tom J. G. Gevers; Joost P. H. Drenth

Polycystic liver disease (PLD) is arbitrarily defined as a liver that contains >20 cysts. The condition is associated with two genetically distinct diseases: as a primary phenotype in isolated polycystic liver disease (PCLD) and as an extrarenal manifestation in autosomal dominant polycystic kidney disease (ADPKD). Processes involved in hepatic cystogenesis include ductal plate malformation with concomitant abnormal fluid secretion, altered cell–matrix interaction and cholangiocyte hyperproliferation. PLD is usually a benign disease, but can cause debilitating abdominal symptoms in some patients. The main risk factors for growth of liver cysts are female sex, exogenous oestrogen use and multiple pregnancies. Ultrasonography is very useful for achieving a correct diagnosis of a polycystic liver and to differentiate between ADPKD and PCLD. Current radiological and surgical therapies for symptomatic patients include aspiration–sclerotherapy, fenestration, segmental hepatic resection and liver transplantation. Medical therapies that interact with regulatory mechanisms controlling expansion and growth of liver cysts are under investigation. Somatostatin analogues are promising; several clinical trials have shown that these drugs can reduce the volume of polycystic livers. The purpose of this Review is to provide an update on the diagnosis and management of PLD with a focus on literature published in the past 4 years.


Alimentary Pharmacology & Therapeutics | 2012

The long-term outcome of patients with polycystic liver disease treated with lanreotide

Melissa Chrispijn; Frederik Nevens; Tom J. G. Gevers; Ragna Vanslembrouck; M.G.H. van Oijen; Walter Coudyzer; Aswin L. Hoffmann; Helena M. Dekker; R.A. de Man; L. van Keimpema; Joost P. H. Drenth

Aliment Pharmacol Ther 2012; 35: 266–274


Journal of Hepatology | 2013

Everolimus does not further reduce polycystic liver volume when added to long acting octreotide: Results from a randomized controlled trial

Melissa Chrispijn; Tom J. G. Gevers; Jeroen C. Hol; René Monshouwer; Heleen M. Dekker; Joost P. H. Drenth

BACKGROUND & AIMS Polycystic liver disease (PLD) is associated with autosomal dominant polycystic kidney disease (ADPKD) or autosomal dominant polycystic liver disease (PCLD). The resulting hepatomegaly compromises quality of life. Somatostatin analogues reduce PLD volume by approximately 5% when given for 6-12 months. A pilot trial in 16 ADPKD patients demonstrated that sirolimus, an mTOR inhibitor, reduced PLD volume by 26%. The aim of this study was to assess the PLD volume reducing effect of everolimus and octreotide relative to octreotide monotherapy. METHODS We designed a randomized controlled trial that compared 48 weeks of everolimus 2.5 mg daily, combined with octreotide 40 mg intramuscularly every 4 weeks, to octreotide monotherapy. We included PCLD and ADPKD patients. Exclusion criteria were MDRD-GFR <60 ml/min/1.73 m(2) and liver volume <2500 ml. Primary outcome was change in liver volume measured with CT-volumetry. RESULTS We randomized 44 PLD patients (29 PCLD, 15 ADPKD, 89% female) to treatment with octreotide (n=23) or octreotide-everolimus (n=21). Liver volume decreased by 3.5% (p<0.01) in the monotherapy arm, compared to 3.8% with combination therapy (p<0.01). The difference between treatment arms was not significant (p=0.73). CONCLUSIONS Adding everolimus to octreotide in PLD does not increase the liver volume reducing effect of octreotide.


Acta Neuropathologica | 2010

Sulfation of heparan sulfate associated with amyloid-beta plaques in patients with Alzheimer's disease.

Ilona B. Bruinsma; Luuk te Riet; Tom J. G. Gevers; Gerdy B. ten Dam; Toin H. van Kuppevelt; Guido David; Benno Küsters; Robert M.W. de Waal; Marcel M. Verbeek

Alzheimer’s disease (AD) is characterized by pathological lesions such as amyloid-β (Aβ) plaques and cerebral amyloid angiopathy. Both these lesions consist mainly of aggregated Aβ protein and this aggregation is affected by macromolecules such as heparan sulfate (HS) proteoglycans. Previous studies demonstrated that HS enhances fibrillogenesis of Aβ and that this enhancement is dependent on the degree of sulfation of HS. In addition, it has been reported that these sulfation epitopes do not occur randomly but have a defined tissue distribution. Until now, the distribution of sulfation epitopes of HS has not yet been studied in human brain. We investigated whether a specific HS epitope is associated with Aβ plaques by performing immunohistochemistry on occipital neocortical and hippocampal tissue sections from AD patients using five HS epitope-specific phage display antibodies. Antibodies recognizing highly N-sulfated HS demonstrated the highest level of staining in both fibrillar Aβ plaques and non-fibrillar Aβ plaques, whereas antibodies recognizing HS regions with a lower degree of N-sulfate modifications were only immunoreactive with fibrillar Aβ plaques. Thus, our results suggest that a larger variety of HS epitopes is associated with fibrillar Aβ plaques, but the HS epitopes associated with non-fibrillar Aβ plaques seem to be more restricted, selectively consisting of highly N-sulfated epitopes.


American Journal of Kidney Diseases | 2014

Rationale and design of the DIPAK 1 study: A randomized controlled clinical trial assessing the efficacy of lanreotide to halt disease progression in autosomal dominant polycystic kidney disease

Esther Meijer; Joost P. H. Drenth; Hedwig M.A. D'Agnolo; Niek F. Casteleijn; Johan W. de Fijter; Tom J. G. Gevers; Peter Kappert; Dorien J.M. Peters; Mahdi Salih; Darius Soonawala; Edwin M. Spithoven; Vicente E. Torres; Folkert W. Visser; Jack F.M. Wetzels; Robert Zietse; Ron T. Gansevoort

BACKGROUND There are limited therapeutic options to slow the progression of autosomal dominant polycystic kidney disease (ADPKD). Recent clinical studies indicate that somatostatin analogues are promising for treating polycystic liver disease and potentially also for the kidney phenotype. We report on the design of the DIPAK 1 (Developing Interventions to Halt Progression of ADPKD 1) Study, which will examine the efficacy of the somatostatin analogue lanreotide on preservation of kidney function in ADPKD. STUDY DESIGN The DIPAK 1 Study is an investigator-driven, randomized, multicenter, controlled, clinical trial. SETTING & PARTICIPANTS We plan to enroll 300 individuals with ADPKD and estimated glomerular filtration rate (eGFR) of 30-60 mL/min/1.73 m(2) who are aged 18-60 years. INTERVENTION Patients will be randomly assigned (1:1) to standard care or lanreotide, 120 mg, subcutaneously every 28 days for 120 weeks, in addition to standard care. OUTCOMES Main study outcome is the slope through serial eGFR measurements starting at week 12 until end of treatment for lanreotide versus standard care. Secondary outcome parameters include change in eGFR from pretreatment versus 12 weeks after treatment cessation, change in kidney volume, change in liver volume, and change in quality of life. MEASUREMENTS Blood and urine will be collected and questionnaires will be filled in following a fixed scheme. Magnetic resonance imaging will be performed for assessment of kidney and liver volume. RESULTS Assuming an average change in eGFR of 5.2 ± 4.3 (SD) mL/min/1.73 m(2) per year in untreated patients, 150 patients are needed in each group to detect a 30% reduction in the rate of kidney function loss between treatment groups with 80% power, 2-sided α = 0.05, and 20% protocol violators and/or dropouts. LIMITATIONS The design is an open randomized controlled trial and measurement of our primary end point does not begin at randomization. CONCLUSIONS The DIPAK 1 Study will show whether subcutaneous administration of lanreotide every 4 weeks attenuates disease progression in patients with ADPKD.


World Journal of Gastroenterology | 2013

Evaluation of hepatic cystic lesions

Marten A Lantinga; Tom J. G. Gevers; Joost P. H. Drenth

Hepatic cysts are increasingly found as a mere coincidence on abdominal imaging techniques, such as ultrasonography (USG), computed tomography (CT) and magnetic resonance imaging (MRI). These cysts often present a diagnostic challenge. Therefore, we performed a review of the recent literature and developed an evidence-based diagnostic algorithm to guide clinicians in characterising these lesions. Simple cysts are the most common cystic liver disease, and diagnosis is based on typical USG characteristics. Serodiagnostic tests and microbubble contrast-enhanced ultrasound (CEUS) are invaluable in differentiating complicated cysts, echinococcosis and cystadenoma/cystadenocarcinoma when USG, CT and MRI show ambiguous findings. Therefore, serodiagnostic tests and CEUS reduce the need for invasive procedures. Polycystic liver disease (PLD) is arbitrarily defined as the presence of > 20 liver cysts and can present as two distinct genetic disorders: autosomal dominant polycystic kidney disease (ADPKD) and autosomal dominant polycystic liver disease (PCLD). Although genetic testing for ADPKD and PCLD is possible, it is rarely performed because it does not affect the therapeutic management of PLD. USG screening of the liver and both kidneys combined with extensive family history taking are the cornerstone of diagnostic decision making in PLD. In conclusion, an amalgamation of these recent advances results in a diagnostic algorithm that facilitates evidence-based clinical decision making.


Liver International | 2014

Evaluating health‐related quality of life in patients with polycystic liver disease and determining the impact of symptoms and liver volume

Titus F. M. Wijnands; Myrte K. Neijenhuis; Wietske Kievit; Frederik Nevens; Marie C. Hogan; Vicente E. Torres; Tom J. G. Gevers; Joost P. H. Drenth

Polycystic liver disease (PLD) follows a progressive course ultimately leading to severe hepatomegaly and mechanical complaints in a subset of patients. It is still unknown to what extent this compromises health‐related quality of life (HRQL). Our aim was to determine HRQL in PLD patients and investigate its association with concurrent abdominal symptoms and liver volume.


Current Opinion in Gastroenterology | 2011

Somatostatin analogues for treatment of polycystic liver disease

Tom J. G. Gevers; Joost P. H. Drenth

Purpose of review The present review summarizes the existing knowledge on polycystic liver disease (PCLD) and highlights the progress made in medical treatment for this condition in the past year. Recent findings PCLD is associated with autosomal dominant polycystic kidney disease (ADPKD) and autosomal dominant PCLD. Signaling pathways of adenosine 3′,5′-cyclic monophosphate (cAMP) and mammalian target of rapamycin (mTOR) are aberrantly regulated in polycystic livers and promote hepatic cystogenesis. Somatostatin analogues reduce intracellular cAMP, and this might prevent fluid accumulation in hepatic cysts. Several clinical trials published over the last year now show that somatostatin analogues when given for 6–12 months in patients with ADPKD and PCLD decrease total liver volume, attenuate polycystic kidney volume, and improve perception of health. In two recent studies mTOR inhibitors failed to halt the progression of ADPKD. It is still too early to recommend to start somatostatin analogues in PCLD and definitive answers should come from future clinical trials. Summary Somatostatin analogues are promising new medical drug options in the treatment of PCLD. However, more needs to be elucidated with regard to molecular mechanisms in hepatic cystogenesis, the uncertainty who will respond to therapy and long-term outcomes.


Alimentary Pharmacology & Therapeutics | 2013

Safety and efficacy of different lanreotide doses in the treatment of polycystic liver disease: pooled analysis of individual patient data

Frederik Temmerman; Tom J. G. Gevers; Thien Anh Ho; Ragna Vanslembrouck; Walter Coudyzer; J. van Pelt; Bert Bammens; Yves Pirson; Joost P. H. Drenth; Frederik Nevens

Long‐acting lanreotide (LAN) 120 mg every 4 weeks reduces liver volume (LV) in patients with polycystic liver diseases (PCLD). Animal studies demonstrated that the inhibition of hepatic and renal cystogenesis is dose dependent.


Nephrology Dialysis Transplantation | 2014

A stepwise approach for effective management of chronic pain in autosomal-dominant polycystic kidney disease

Niek F. Casteleijn; Folkert W. Visser; Joost P. H. Drenth; Tom J. G. Gevers; Gerbrand J. Groen; Marie C. Hogan; Ron T. Gansevoort

Chronic pain, defined as pain existing for >4-6 weeks, affects >60% of patients with autosomal-dominant polycystic disease (ADPKD). It can have various causes, indirectly or directly related to the increase in kidney and liver volume in these patients. Chronic pain in ADPKD patients is often severe, impacting physical activity and social relationships, and frequently difficult to manage. This review provides an overview of pathophysiological mechanisms that can lead to pain and discusses the sensory innervation of the kidneys and the upper abdominal organs, including the liver. In addition, the results of a systematic literature search of ADPKD-specific treatment options are presented. Based on pathophysiological knowledge and evidence derived from the literature an argumentative stepwise approach for effective management of chronic pain in ADPKD is proposed.

Collaboration


Dive into the Tom J. G. Gevers's collaboration.

Top Co-Authors

Avatar

Joost P. H. Drenth

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

J.P.H. Drenth

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Frederik Nevens

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Ron T. Gansevoort

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Wietske Kievit

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Darius Soonawala

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Folkert W. Visser

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

T.F. Wijnands

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Jack F.M. Wetzels

Radboud University Nijmegen

View shared research outputs
Researchain Logo
Decentralizing Knowledge