Network


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

Hotspot


Dive into the research topics where E. Meershoek-Klein Kranenbarg is active.

Publication


Featured researches published by E. Meershoek-Klein Kranenbarg.


Annals of Oncology | 2015

Adjuvant chemotherapy for rectal cancer patients treated with preoperative (chemo)radiotherapy and total mesorectal excision: a Dutch Colorectal Cancer Group (DCCG) randomized phase III trial

Anne J Breugom; W. van Gijn; E. W. Muller; Åke Berglund; C.B.M. van den Broek; Tone Fokstuen; Hans Gelderblom; Ellen Kapiteijn; Jan Willem Leer; Corrie A.M. Marijnen; Hendrik Martijn; E. Meershoek-Klein Kranenbarg; Iris D. Nagtegaal; Lars Påhlman; C.J.A. Punt; Hein Putter; A. G. H. Roodvoets; H.J.T. Rutten; W. H. Steup; Bengt Glimelius; C.J.H. van de Velde

BACKGROUND The discussion on the role of adjuvant chemotherapy for rectal cancer patients treated according to current guidelines is still ongoing. A multicentre, randomized phase III trial, PROCTOR-SCRIPT, was conducted to compare adjuvant chemotherapy with observation for rectal cancer patients treated with preoperative (chemo)radiotherapy and total mesorectal excision (TME). PATIENTS AND METHODS The PROCTOR-SCRIPT trial recruited patients from 52 hospitals. Patients with histologically proven stage II or III rectal adenocarcinoma were randomly assigned (1:1) to observation or adjuvant chemotherapy after preoperative (chemo)radiotherapy and TME. Radiotherapy consisted of 5 × 5 Gy. Chemoradiotherapy consisted of 25 × 1.8-2 Gy combined with 5-FU-based chemotherapy. Adjuvant chemotherapy consisted of 5-FU/LV (PROCTOR) or eight courses capecitabine (SCRIPT). Randomization was based on permuted blocks of six, stratified according to centre, residual tumour, time between last irradiation and surgery, and preoperative treatment. The primary end point was overall survival. RESULTS Of 470 enrolled patients, 437 were eligible. The trial closed prematurely because of slow patient accrual. Patients were randomly assigned to observation (n = 221) or adjuvant chemotherapy (n = 216). After a median follow-up of 5.0 years, 5-year overall survival was 79.2% in the observation group and 80.4% in the chemotherapy group [hazard ratio (HR) 0.93, 95% confidence interval (CI) 0.62-1.39; P = 0.73]. The HR for disease-free survival was 0.80 (95% CI 0.60-1.07; P = 0.13). Five-year cumulative incidence for locoregional recurrences was 7.8% in both groups. Five-year cumulative incidence for distant recurrences was 38.5% and 34.7%, respectively (P = 0.39). CONCLUSION The PROCTOR-SCRIPT trial could not demonstrate a significant benefit of adjuvant chemotherapy with fluoropyrimidine monotherapy after preoperative (chemo)radiotherapy and TME on overall survival, disease-free survival, and recurrence rate. However, this trial did not complete planned accrual. REGISTRATION NUMBER Dutch Colorectal Cancer group, CKTO 2003-16, ISRCTN36266738.


Ejso | 2009

Transanal endoscopic microsurgery versus total mesorectal excision of T1 rectal adenocarcinomas with curative intention

E. J. R. de Graaf; Pascal G. Doornebosch; R.A.E.M. Tollenaar; E. Meershoek-Klein Kranenbarg; A.C. de Boer; F.C. Bekkering; C.J.H. van de Velde

PURPOSE After total mesorectal excision (TME) for rectal cancer, pathology is standardized with margin status as a predictor for recurrence. This has yet to be implemented after transanal endoscopic microsurgery (TEM) and was investigated prospectively for T1 rectal adenocarcinomas. PATIENTS AND METHODS Eighty patients after TEM were compared to 75 patients after TME. The study protocol included standardized pathology. TEM patients were eligible when excision margins were negative. RESULTS TEM was safer than TME as reflected by operating time, blood loss, hospital stay, morbidity, re-operation rate and stoma formation (all P<0.001). Mortality after TEM was 0% and after TME 4%. At 5 years after TEM and TME, both overall survival (TEM 75% versus TME 77%, P=0.9) and cancer-specific survival (TEM 90% versus TME 87%, P=0.5) were comparable. Local recurrence rate after TEM was 24% and after TME 0% (HR 79.266, 95% CI, 1.208 to 5202, P<0.0001). CONCLUSION For T1 rectal adenocarcinomas TEM is much saver than TME and survival is comparable. After TEM local recurrence rate is substantial, despite negative excision margins.


Ejso | 2009

Improved survival after resectable non-cardia gastric cancer in the Netherlands: The importance of surgical training and quality control

P. Krijnen; M. den Dulk; E. Meershoek-Klein Kranenbarg; M.L.E.A. Jansen-Landheer; C.J.H. van de Velde

BACKGROUND In The Netherlands, standardised limited D1 and extended D2 lymph node dissections in the treatment of resectable gastric cancer were introduced nationwide within the framework of the Dutch D1-D2 Gastric Cancer Trial between 1989 and 1993. In a population-based study, we evaluated whether the survival of patients with resectable gastric cancer improved over time on a regional level. METHODS We compared 5-year overall and relative survival of patients with curatively resected non-cardia gastric cancer in the regional cancer registry of the Comprehensive Cancer Centre West in The Netherlands before the Dutch D1-D2 trial (1986 to mid 1989; n = 273), during the trial period (mid 1989 to mid 1993; n = 255), and after the trial (mid 1993 to 1999; n = 219), adjusting for prognostic variables. RESULTS Unadjusted survival was highest in the post-trial period: 5-year overall and relative survival were 42% and 52%, respectively, compared to 34% and 41% in the pre-trial period, and 39% and 46% in the trial period (p = 0.31 and p = 0.06, respectively). After adjustment for age, gender, tumour site, pT-stage, nodal status and hospital volume, the effect of period on survival was more apparent (p = 0.009). Compared to the pre-trial period, the hazard ratio was 0.83 (95% confidence interval, 0.68-1.02) for the trial period, and 0.72 (0.58-0.89) after the trial. Less than 1% of the patients received adjuvant therapy. CONCLUSION Survival of patients with curatively resected non-cardia gastric cancer has improved. Standardisation and surgical training in D1 and D2 lymph node dissection are the most likely explanation for this improvement.


Annals of Oncology | 2014

Addition of zoledronic acid to neoadjuvant chemotherapy does not enhance tumor response in patients with HER2 negative stage II/III breast cancer: the NEOZOTAC trial (BOOG 2010-01)

Ayoub Charehbili; S van de Ven; Vincent T.H.B.M. Smit; E. Meershoek-Klein Kranenbarg; Neveen A. T. Hamdy; Hein Putter; Joan B. Heijns; Lj van Warmerdam; Lw Kessels; M.W. Dercksen; Mj Pepels; E. Maartense; H. W. M. van Laarhoven; Birgit E.P.J. Vriens; Mnjm Wasser; A.E. van Leeuwen-Stok; G.J. Liefers; C.J.H. van de Velde; J. W. R. Nortier; Judith R. Kroep

BACKGROUND The role of zoledronic acid (ZA) when added to the neoadjuvant treatment of breast cancer (BC) in enhancing the clinical and pathological response of tumors is unclear. The effect of ZA on the antitumor effect of neoadjuvant chemotherapy has not prospectively been studied before. PATIENTS AND METHODS NEOZOTAC is a national, multicenter, randomized study comparing the efficacy of TAC (docetaxel, adriamycin and cyclophosphamide i.v.) followed by granulocyte colony-stimulating factor on day 2 with or without ZA 4 mg i.v. q 3 weeks inpatients withstage II/III, HER2-negative BC. We present data on the pathological complete response (pCR in breast and axilla), on clinical response using MRI, and toxicity. Post hoc subgroup analyses were undertaken to address the predictive value of menopausal status. RESULTS Addition of ZA to chemotherapy did not improve pCR rates (13.2% for TAC+ZA versus 13.3% for TAC). Postmenopausal women (N = 96) had a numerical benefit from ZA treatment (pCR 14.0% for TAC+ZA versus 8.7% for TAC, P = 0.42). Clinical objective response did not differ between treatment arms (72.9% versus 73.7%). There was no difference in grade III/IV toxicity between treatment arms. CONCLUSIONS Addition of ZA to neoadjuvant chemotherapy did not improve pathological or clinical response to chemotherapy. Further investigations are warranted in postmenopausal women with BC, since this subgroup might benefit from ZA treatment.


Ejso | 2013

An increasing use of defunctioning stomas after low anterior resection for rectal cancer. Is this the way to go

H.S. Snijders; C.B.M. van den Broek; Michel W.J.M. Wouters; E. Meershoek-Klein Kranenbarg; T. Wiggers; H.J.T. Rutten; C.J.H. van de Velde; R.A.E.M. Tollenaar; J.W.T. Dekker

BACKGROUND The last decade there has been an increased awareness of the problem of anastomotic leakage after low anterior resection for rectal cancer, which may have led to more defunctioning stomas. In this study, current use of defunctioning stomas was assessed and compared to the use of defunctioning stomas at the time of the TME-trial together with associated outcomes. METHODS Eligible patients with rectal cancer undergoing low anterior resection were selected from the Dutch Surgical Colorectal Audit (DSCA, n = 988). Similar patients were selected from the TME-trial (n = 891). The percentages of patients with a defunctioning stoma, anastomotic leakage and postoperative mortality rates were studied. Multivariable models were used to study possible confounding on the outcomes. RESULTS At the time of the TME-trial, 57% of patients received a defunctioning stoma. At the time of the DSCA, 70% of all patients received a defunctioning stoma (p < 0.001). Anastomotic leakage rates were similar (11.4% and 12.1%; p = 0.640). The postoperative mortality rate differed (3.9% in the TME-trial vs. 1.1% in the DSCA; p < 0.001), but was not associated with a more frequent use of a stoma (OR 1.80, 95% CI 0.91-3.58). CONCLUSION In current surgical practice, 70% of patients undergoing LAR for rectal cancer receives a defunctioning stomas. This percentage seems increased when compared to data from the TME-trial. Clinically relevant anastomotic leakage rates remained similar. Therefore, current routine use of defunctioning stomas should be questioned.


Molecular Oncology | 2015

Disorganised stroma determined on pre-treatment breast cancer biopsies is associated with poor response to neoadjuvant chemotherapy: Results from the NEOZOTAC trial

Tim J. A. Dekker; Ayoub Charehbili; Vincent T.H.B.M. Smit; P. ten Dijke; E. Meershoek-Klein Kranenbarg; C.J.H. van de Velde; J. W. R. Nortier; R.A.E.M. Tollenaar; Wilma E. Mesker; Judith R. Kroep

The tumor‐associated stroma is of importance for tumor progression and is generally accepted to have a significant influence on patient prognosis. However, little is known regarding specific features of tumor‐associated stromal tissues and response to (neoadjuvant) chemotherapy. This study investigated the predictive value of extracellular matrix organization on response to chemotherapy in patients treated in the NEOZOTAC trial.


Annals of Oncology | 2012

Relationship between specific adverse events and efficacy of exemestane therapy in early postmenopausal breast cancer patients

D.B.Y. Fontein; Danny Houtsma; Elysée T.M. Hille; C. Seynaeve; Hein Putter; E. Meershoek-Klein Kranenbarg; H.-J. Guchelaar; Hans Gelderblom; L Dirix; Robert Paridaens; John M.S. Bartlett; J. W. R. Nortier; C.J.H. van de Velde

BACKGROUND Many adverse events (AEs) associated with aromatase inhibitors (AIs) involve symptoms related to the depletion of circulating estrogens, and may be related to efficacy. We assessed the relationship between specific AEs [hot flashes (HF) and musculoskeletal AEs (MSAE)] and survival outcomes in Dutch and Belgian patients treated with exemestane (EXE) in the Tamoxifen Exemestane Adjuvant Multinational (TEAM) trial. Additionally, the relationship between hormone receptor expression and AEs was assessed. METHODS Efficacy end points were relapse-free survival (RFS), overall survival (OS) and breast cancer-specific mortality (BCSM), starting at 6 months after starting EXE treatment. AEs reported in the first 6 months of treatment were included. Specific AEs comprised HF and/or MSAE. Landmark analyses and Cox proportional hazards models assessed survival differences up to 5 years. RESULTS A total of 1485 EXE patients were included. Patients with HF had a better RFS than patients without HF [multivariate hazard ratio (HR) 0.393, 95% confidence interval (CI) 0.19-0.813; P = 0.012]. The occurrence of MSAE versus no MSAE did not relate to better RFS (multivariate HR 0.677, 95% CI 0.392-1.169; P = 0.162). Trends were maintained for OS and BCSM. Quantitative hormone receptor expression was not associated with specific AEs. CONCLUSIONS Some AEs associated with estrogen depletion are related to better outcomes and may be valuable biomarkers in AI treatment.


Cancer Research | 2017

Abstract S1-04: Optimal duration of extended letrozole treatment after 5 years of adjuvant endocrine therapy; results of the randomized phase III IDEAL trial (BOOG 2006-05)

Ej Blok; Cjh van de Velde; E. Meershoek-Klein Kranenbarg; Hein Putter; J. van den Bosch; E. Maartense; A.E. van Leeuwen-Stok; G. J. Liefers; J. W. R. Nortier; Ejt Rutgers; Jr Kroep

Despite the success of adjuvant endocrine therapy in early breast cancer, approximately 50% of all recurrences still occur after the initial 5 years of follow-up. Earlier studies confirmed that endocrine therapy extension after 5 years of adjuvant tamoxifen with either tamoxifen or aromatase inhibitors up to 10 years leads to an improvement in survival. However, aromatase inhibitors are currently standard of care in the initial 5 years of adjuvant therapy, and the benefit of extended use beyond 5 years of AI based therapy is still debated. The randomized phase III IDEAL trial was designed to study the optimal duration of extended adjuvant endocrine therapy after the initial 5 years of any endocrine therapy. Between April 2007 and November 2011, 1824 postmenopausal, HR-positive early breast cancer patients were included by 74 hospitals in the Netherlands. Enrolled patients earlier received 5 years of any endocrine therapy (87.9% AI based),completed this treatment no longer than 2 years before randomization, and did not have any recurrence at the moment of inclusion. The included patients were randomly allocated to either 2.5 or 5 years of extended letrozole (daily 2.5mg). Primary outcome was disease free survival (DFS), secondary endpoints were overall survival (OS), distant disease free survival (DDFS), contralateral breast cancer and safety. Results & Discussion: Results will become available soon. Citation Format: Blok EJ, van de Velde CJH, Meershoek-Klein Kranenbarg EM, Putter H, van den Bosch J, Maartense E, van Leeuwen-Stok AE, Liefers G-J, Nortier JWR, Rutgers EJT, Kroep JR. Optimal duration of extended letrozole treatment after 5 years of adjuvant endocrine therapy; results of the randomized phase III IDEAL trial (BOOG 2006-05) [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr S1-04.


Cancer Research | 2010

Abstract S2-4: Final Results of a Prospectively Planned Biomarker Analysis: HER1-3 as Predictive Markers of Benefit from Early Treatment with Aromatase Inhibitors Versus Tamoxifen in the TEAM Pathology Sub-Study.

Jms Bartlett; Cl Brookes; Cjh van de Velde; Deborah D. Stocken; Fm Campbell; Annette Hasenburg; Charlene Kay; D Kiebeck; Christos Markopoulos; E. Meershoek-Klein Kranenbarg; Elizabeth Mallon; L Dirix; T Robson; C. Seynaeve; D. Rea

Background: The Tamoxifen and Exemestane Adjuvant Multinational (TEAM) trial included prospectively planned biomarker studies to identify predictive biomarkers for patients receiving endocrine therapy. Quantitative IHC data for ER/PgR, HER1, HER2, HER3 and FISH analysis of HER2 in all cases was available for the current analysis relative to outcome of estrogen receptor-positive (ER+) early breast cancer patients treated with exemestane versus tamoxifen. Patients & Methods: Pathology blocks from 4598 TEAM patients were collected and tissue microarrays constructed. Quantitative analysis of receptors (HER1/2/3) by conventional IHC, and FISH (for HER2 only) were analysed relative to disease-free survival and treatment on an intent to treat basis using survival data for the first 2.75 years of the TEAM trial. Results: Of 4595 eligible cases samples received, 16 were excluded, and 4010 had complete biomarker data for all markers (HER1/HER2 & HER3) for the final biomarker analysis, 3.5% were HER1 positive, 13% HER2 positive & 21% HER3 positive. 1248 (31%) cases were HER1or2or3 positive (HER1-3+ve). HER1-3 positivity was associated with poor outcome (HR=1.6 95%CI=1.3-2.0). In HER1-3 negative patients the hazard ratio (for risk of relapse on exemestane versus tamoxifen in the first 2.75 years) was 0.68 (95% CI = 0.52-0.89), for the HER1-3 positive cases the hazard ratio was 1.14 (95% CI = 0.83-1.56) with a significant treatment by marker interaction (HR=1.68 95%CI=1.1-2.5; p=0.0014 in multivariate analysis). Trends for similar effects were seen for HER1 negative (-ve) vs HER1 positive (+ve) (HRs 0.80, 95%CI=0.65-0.99 vs 1.63 95%CI=0.74-3.59), HER2-ve vs HER2+ve (HRs 0.71, 95%CI=0.57- 0.9 vs 1.69 95%CI=108-2.63) and HER3-ve vs HER3+ve (HRs 0.78 95%CI=0.62-0.99 vs 1.04 95%CI=0.67-1.62) breast cancers. Conclusion: Preferential exemestane versus tamoxifen treatment benefit was seen in HER1/2/3 negative cases, whilst HER1/2/3 positive cases had a poor prognosis in this endocrine treated population (suggesting a degree of resistance to endocrine therapy), and no evidence of additional benefit from AIs versus tamoxifen. These three Type I receptor tyrosine kinases appear to identify breast cancers with relative resistance to all forms of endocrine therapy. This prospectively planned and powered treatment by marker analysis provides high level scientific evidence which may assist clinicians and patients in determining optimal AI schedules for women with early breast cancer. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr S2-4.


Colorectal Disease | 2018

Introduction of a colorectal cancer screening programme: results from a single-centre study

N. C. A. Vermeer; R. R. Bahadoer; E. Bastiaannet; Fabian A. Holman; E. Meershoek-Klein Kranenbarg; G. J. Liefers; C.J.H. van de Velde; Koen C.M.J. Peeters

In 2014, a national colorectal cancer (CRC) screening programme was launched in the Netherlands. It is difficult to assess for the individual patients with CRC whether the oncological benefits of surgery will outweigh the morbidity of the procedure, especially in early lesions. This study compares patient and tumour characteristics between screen‐detected and nonscreen‐detected patients. Also, we present an overview of treatment options and clinical dilemmas when treating patients with early‐stage colorectal disease.

Collaboration


Dive into the E. Meershoek-Klein Kranenbarg's collaboration.

Top Co-Authors

Avatar

C.J.H. van de Velde

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hein Putter

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

J. W. R. Nortier

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Cjh van de Velde

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ayoub Charehbili

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Judith R. Kroep

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jr Kroep

VU University Amsterdam

View shared research outputs
Top Co-Authors

Avatar

Ej Blok

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

G.J. Liefers

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge