Network


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

Hotspot


Dive into the research topics where Winald R. Gerritsen is active.

Publication


Featured researches published by Winald R. Gerritsen.


The New England Journal of Medicine | 2013

Abiraterone in Metastatic Prostate Cancer without Previous Chemotherapy

Charles J. Ryan; Matthew R. Smith; Johann S. de Bono; Arturo Molina; Christopher J. Logothetis; Paul de Souza; Karim Fizazi; Paul N. Mainwaring; Josep M. Piulats; Siobhan Ng; Joan Carles; Peter Mulders; Ethan Basch; Eric J. Small; Fred Saad; Dirk Schrijvers; Hendrik Van Poppel; Som D. Mukherjee; Henrik Suttmann; Winald R. Gerritsen; Thomas W. Flaig; Daniel J. George; Evan Y. Yu; Allan J. Pantuck; Eric Winquist; Celestia S. Higano; Mary-Ellen Taplin; Youn C. Park; Thian Kheoh; Thomas W. Griffin

BACKGROUND Abiraterone acetate, an androgen biosynthesis inhibitor, improves overall survival in patients with metastatic castration-resistant prostate cancer after chemotherapy. We evaluated this agent in patients who had not received previous chemotherapy. METHODS In this double-blind study, we randomly assigned 1088 patients to receive abiraterone acetate (1000 mg) plus prednisone (5 mg twice daily) or placebo plus prednisone. The coprimary end points were radiographic progression-free survival and overall survival. RESULTS The study was unblinded after a planned interim analysis that was performed after 43% of the expected deaths had occurred. The median radiographic progression-free survival was 16.5 months with abiraterone-prednisone and 8.3 months with prednisone alone (hazard ratio for abiraterone-prednisone vs. prednisone alone, 0.53; 95% confidence interval [CI], 0.45 to 0.62; P<0.001). Over a median follow-up period of 22.2 months, overall survival was improved with abiraterone-prednisone (median not reached, vs. 27.2 months for prednisone alone; hazard ratio, 0.75; 95% CI, 0.61 to 0.93; P=0.01) but did not cross the efficacy boundary. Abiraterone-prednisone showed superiority over prednisone alone with respect to time to initiation of cytotoxic chemotherapy, opiate use for cancer-related pain, prostate-specific antigen progression, and decline in performance status. Grade 3 or 4 mineralocorticoid-related adverse events and abnormalities on liver-function testing were more common with abiraterone-prednisone. CONCLUSIONS Abiraterone improved radiographic progression-free survival, showed a trend toward improved overall survival, and significantly delayed clinical decline and initiation of chemotherapy in patients with metastatic castration-resistant prostate cancer. (Funded by Janssen Research and Development, formerly Cougar Biotechnology; ClinicalTrials.gov number, NCT00887198.).


Lancet Oncology | 2014

Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial

Eugene D. Kwon; Charles G. Drake; Howard I. Scher; Karim Fizazi; Alberto Bossi; Alfons J.M. van den Eertwegh; Michael Krainer; Nadine Houédé; Ricardo Santos; Hakim Mahammedi; Siobhan Ng; Michele Maio; Fabio A. Franke; Santhanam Sundar; Neeraj Agarwal; Andries M. Bergman; Tudor Ciuleanu; Ernesto Korbenfeld; Lisa Sengeløv; Steinbjørn Hansen; Christopher J. Logothetis; Tomasz M. Beer; M. Brent McHenry; Paul Gagnier; David Liu; Winald R. Gerritsen

BACKGROUND Ipilimumab is a fully human monoclonal antibody that binds cytotoxic T-lymphocyte antigen 4 to enhance antitumour immunity. Our aim was to assess the use of ipilimumab after radiotherapy in patients with metastatic castration-resistant prostate cancer that progressed after docetaxel chemotherapy. METHODS We did a multicentre, randomised, double-blind, phase 3 trial in which men with at least one bone metastasis from castration-resistant prostate cancer that had progressed after docetaxel treatment were randomly assigned in a 1:1 ratio to receive bone-directed radiotherapy (8 Gy in one fraction) followed by either ipilimumab 10 mg/kg or placebo every 3 weeks for up to four doses. Non-progressing patients could continue to receive ipilimumab at 10 mg/kg or placebo as maintenance therapy every 3 months until disease progression, unacceptable toxic effect, or death. Patients were randomly assigned to either treatment group via a minimisation algorithm, and stratified by Eastern Cooperative Oncology Group performance status, alkaline phosphatase concentration, haemoglobin concentration, and investigator site. Patients and investigators were masked to treatment allocation. The primary endpoint was overall survival, assessed in the intention-to-treat population. This trial is registered with ClinicalTrials.gov, number NCT00861614. FINDINGS From May 26, 2009, to Feb 15, 2012, 799 patients were randomly assigned (399 to ipilimumab and 400 to placebo), all of whom were included in the intention-to-treat analysis. Median overall survival was 11·2 months (95% CI 9·5-12·7) with ipilimumab and 10·0 months (8·3-11·0) with placebo (hazard ratio [HR] 0·85, 0·72-1·00; p=0·053). However, the assessment of the proportional hazards assumption showed that it was violated (p=0·0031). A piecewise hazard model showed that the HR changed over time: the HR for 0-5 months was 1·46 (95% CI 1·10-1·95), for 5-12 months was 0·65 (0·50-0·85), and beyond 12 months was 0·60 (0·43-0·86). The most common grade 3-4 adverse events were immune-related, occurring in 101 (26%) patients in the ipilimumab group and 11 (3%) of patients in the placebo group. The most frequent grade 3-4 adverse events included diarrhoea (64 [16%] of 393 patients in the ipilimumab group vs seven [2%] of 396 in the placebo group), fatigue (40 [11%] vs 35 [9%]), anaemia (40 [10%] vs 43 [11%]), and colitis (18 [5%] vs 0). Four (1%) deaths occurred because of toxic effects of the study drug, all in the ipilimumab group. INTERPRETATION Although there was no significant difference between the ipilimumab group and the placebo group in terms of overall survival in the primary analysis, there were signs of activity with the drug that warrant further investigation. FUNDING Bristol-Myers Squibb.


The New England Journal of Medicine | 2017

Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma

Joaquim Bellmunt; Ronald de Wit; David J. Vaughn; Yves Fradet; Jae Lyun Lee; Lawrence Fong; Nicholas J. Vogelzang; Miguel Angel Climent; Daniel P. Petrylak; Toni K. Choueiri; Andrea Necchi; Winald R. Gerritsen; Howard Gurney; David I. Quinn; S. Culine; Cora N. Sternberg; Yabing Mai; Christian Heinrich Poehlein; Rodolfo F. Perini; Dean F. Bajorin

Background Patients with advanced urothelial carcinoma that progresses after platinum‐based chemotherapy have a poor prognosis and limited treatment options. Methods In this open‐label, international, phase 3 trial, we randomly assigned 542 patients with advanced urothelial cancer that recurred or progressed after platinum‐based chemotherapy to receive pembrolizumab (a highly selective, humanized monoclonal IgG4κ isotype antibody against programmed death 1 [PD‐1]) at a dose of 200 mg every 3 weeks or the investigators choice of chemotherapy with paclitaxel, docetaxel, or vinflunine. The coprimary end points were overall survival and progression‐free survival, which were assessed among all patients and among patients who had a tumor PD‐1 ligand (PD‐L1) combined positive score (the percentage of PD‐L1–expressing tumor and infiltrating immune cells relative to the total number of tumor cells) of 10% or more. Results The median overall survival in the total population was 10.3 months (95% confidence interval [CI], 8.0 to 11.8) in the pembrolizumab group, as compared with 7.4 months (95% CI, 6.1 to 8.3) in the chemotherapy group (hazard ratio for death, 0.73; 95% CI, 0.59 to 0.91; P=0.002). The median overall survival among patients who had a tumor PD‐L1 combined positive score of 10% or more was 8.0 months (95% CI, 5.0 to 12.3) in the pembrolizumab group, as compared with 5.2 months (95% CI, 4.0 to 7.4) in the chemotherapy group (hazard ratio, 0.57; 95% CI, 0.37 to 0.88; P=0.005). There was no significant between‐group difference in the duration of progression‐free survival in the total population (hazard ratio for death or disease progression, 0.98; 95% CI, 0.81 to 1.19; P=0.42) or among patients who had a tumor PD‐L1 combined positive score of 10% or more (hazard ratio, 0.89; 95% CI, 0.61 to 1.28; P=0.24). Fewer treatment‐related adverse events of any grade were reported in the pembrolizumab group than in the chemotherapy group (60.9% vs. 90.2%); there were also fewer events of grade 3, 4, or 5 severity reported in the pembrolizumab group than in the chemotherapy group (15.0% vs. 49.4%). Conclusions Pembrolizumab was associated with significantly longer overall survival (by approximately 3 months) and with a lower rate of treatment‐related adverse events than chemotherapy as second‐line therapy for platinum‐refractory advanced urothelial carcinoma. (Funded by Merck; KEYNOTE‐045 ClinicalTrials.gov number, NCT02256436.)


Nature Reviews Clinical Oncology | 2014

Therapeutic vaccines for cancer: an overview of clinical trials

Ignacio Melero; Gustav Gaudernack; Winald R. Gerritsen; Christoph Huber; Giorgio Parmiani; Suzy Scholl; Nicholas Thatcher; John Wagstaff; Christoph Zielinski; Ian Faulkner; Håkan Mellstedt

The therapeutic potential of host-specific and tumour-specific immune responses is well recognized and, after many years, active immunotherapies directed at inducing or augmenting these responses are entering clinical practice. Antitumour immunization is a complex, multi-component task, and the optimal combinations of antigens, adjuvants, delivery vehicles and routes of administration are not yet identified. Active immunotherapy must also address the immunosuppressive and tolerogenic mechanisms deployed by tumours. This Review provides an overview of new results from clinical studies of therapeutic cancer vaccines directed against tumour-associated antigens and discusses their implications for the use of active immunotherapy.


Lancet Oncology | 2012

Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial

Alfons J.M. van den Eertwegh; Jurjen Versluis; H. Pieter van den Berg; Saskia J. A. M. Santegoets; R. Jeroen A. van Moorselaar; Tim M. van der Sluis; Helen Gall; Thomas C. Harding; Karin Jooss; Israel Lowy; Rik J. Scheper; A. G. M. Stam; B. Mary E. von Blomberg; Tanja D. de Gruijl; Kristen Hege; Natalie Sacks; Winald R. Gerritsen

BACKGROUND The granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells vaccine (GVAX) has antitumour activity against prostate cancer; preclinical studies have shown potent synergy when combined with ipilimumab, an antibody that blocks cytotoxic T-lymphocyte antigen 4. We aimed to assess the safety of combined treatment with GVAX and ipilimumab in patients with metastatic castration-resistant prostate cancer (mCRPC). METHODS We did an open-labelled, single-centre, dose-escalation study of ipilimumab concurrent with a fixed dose of GVAX, with a subsequent expansion phase, both at the VU University Medical Centre (Amsterdam, Netherlands). Eligible patients had documented mCRPC and had not been previously treated with chemotherapy. All patients received a 5×10(8) cell priming dose of GVAX intradermally on day 1 with subsequent intradermal injections of 3×10(8) cells every 2 weeks for 24 weeks. The vaccinations were combined with intravenous ipilimumab every 4 weeks. We enrolled patients in cohorts of three; each cohort received an escalating dose of ipilimumab at 0·3, 1·0, 3·0, or 5·0 mg/kg. Our primary endpoint was safety. This study is registered with ClinicalTrials.gov, number NCT01510288. FINDINGS We enrolled 12 patients into our dose-escalation cohort. We did not record any severe immune-related adverse events at the first two dose levels. At the 3·0 mg/kg dose level, one patient had grade 2 and two patients grade 3 hypophysitis; at the 5·0 mg/kg dose level, two patients had grade 3 hypophysitis and one patient developed grade 4 sarcoid alveolitis (a dose-limiting toxic effect). Due to observed clinical activity and toxic events, we decided to expand the 3·0 mg/kg dose level, rather than enrol a further three patients at the 5·0 mg/kg level. 16 patients were enrolled in the expansion cohort, two of whom developed grade 2 hypophysitis, three colitis (one grade 1 and two grade 2), and one grade 3 hepatitis--all immune-related adverse events. The most common adverse events noted in all 28 patients were injection-site reactions (grade 1-2 events seen in all patients), fatigue (grade 1-2 in 20 patients, grade 3 in two), and pyrexia (grade 1-2 in 15 patients, grade 3 in one). 50% or greater declines in prostate-specific antigen from baseline was recorded in seven patients (25%); all had received 3·0 mg/kg or 5·0 mg/kg ipilimumab. INTERPRETATION GVAX combined with 3·0 mg/kg ipilimumab is tolerable and safe for patients with mCRPC. Further research on the combined treatment of patients with mCRPC with vaccination and ipilimumab is warranted. FUNDING Cell Genesys Inc, Prostate Cancer Foundation, Dutch Cancer Society (KWF-VU 2006-3697), and Foundation Stichting VUmc Cancer Center Amsterdam.


Cancer Gene Therapy | 2000

Targeting of adenoviral vectors through a bispecific single-chain antibody

Hidde J. Haisma; Jacques Grill; David T. Curiel; Susan Hoogeland; Victor W. van Beusechem; Winald R. Gerritsen

Recombinant adenoviral vectors are attractive in the context of cancer gene therapy because they are capable of delivering genes to a wide variety of tissues. The utility of adenoviruses is limited by their lack of specificity and by the absence of the receptor(s) for these viruses on many tumor cells. Redirecting adenoviral vectors to tissue- or tumor-specific targets can be achieved by using bispecific conjugates produced by chemical linkage of an anti-adenovirus antibody (Ab) and a ligand or Ab directed toward a specific target. To avoid the limitations of chemical conjugates, molecular conjugates of anti-fiber knob and ligand have been proposed. We present here a novel strategy that allows the production of recombinant bispecific single-chain Abs directed at cell surface molecules. A construct was made that encodes a neutralizing anti-adenovirus fiber single-chain Fv (scFv) Ab (S11) fused to a scFv Ab (425) directed against the epidermal growth factor receptor. The fusion protein markedly enhanced the infection efficiency of adenoviral vectors in epidermal growth factor receptor-expressing cell lines. The bispecific scFv could be purified and concentrated after binding of its 6His tag to a nickel column without significant loss of activity. This approach should permit the production of high quantities of active bispecific scFv for in vivo use. The universal design of the construct allows rapid screening for relevant specific scFv directed at cell surface antigens that can be incorporated into adenoviral targeting strategies.


Gene Therapy | 1999

Tumor-specific gene transfer via an adenoviral vector targeted to the pan-carcinoma antigen EpCAM

Hidde J. Haisma; H.M. Pinedo; A van Rijswijk; I.H. van der Meulen-Muileman; B A Sosnowski; W Ying; V W van Beusechem; B W Tillman; Winald R. Gerritsen; David T. Curiel

The utility of adenoviral vectors for cancer therapy is limited due to their lack of specificity for tumor cells. In order to target adenovirus to tumor, the natural tropism of the adenovirus should be ablated and replaced by a tumor-specific binding domain. To this end, a neutralizing anti-fiber antibody conjugated to an anti-EpCAM antibody was created that targets the adenovirus to the EpCAM antigen present on tumor cells. The EpCAM antigen was chosen as the target because this antigen is highly expressed on a variety of adenocarcinomas of different origin such as breast, ovary, colon and lung, whereas EpCAM expression is limited in normal tissues. In these studies, the EpCAM-targeted adenovirus was shown to infect specifically cancer cell lines of different origin expressing EpCAM such as ovary, colon and head and neck. Gene transfer was blocked by excess anti-EpCAM antibody and dramatically reduced in EpCAM negative cell lines, thus showing the specificity of the EpCAM-targeted adenovirus. Importantly, infection with targeted adenovirus was independent of CAR, which is the natural receptor for adenovirus binding, since blocking of CAR with recombinant fiber knob did not affect infection with targeted adenovirus. Apart from the cancer cell lines, the efficacy of targeted viral infection was studied in freshly isolated primary human colon cancer cells. As colon cancer predominantly metastasizes to liver, and adenovirus has a high tropism for hepatocytes, we also sought to determine if the EpCAM-targeted adenovirus showed reduced infectivity of human liver cells. The bispecific antibody could successfully mediate gene transfer to primary human colon cancer cells, whereas it almost completely abolished infection of liver cells. This work thus demonstrates that EpCAM-targeted adenoviral vectors can be specifically directed to a wide variety of adenocarcinomas. This approach may prove to be useful for selective gene therapy of cancer.


Blood | 2011

Blood platelets contain tumor-derived RNA biomarkers

R. Jonas A. Nilsson; Leonora Balaj; Esther Hulleman; Sjoerd van Rijn; D. Michiel Pegtel; Maudy Walraven; Anders Widmark; Winald R. Gerritsen; Henk M.W. Verheul; W. Peter Vandertop; David P. Noske; Johan Skog; Thomas Wurdinger

Diagnostic platforms providing biomarkers that are highly predictive for diagnosing, monitoring, and stratifying cancer patients are key instruments in the development of personalized medicine. We demonstrate that tumor cells transfer (mutant) RNA into blood platelets in vitro and in vivo, and show that blood platelets isolated from glioma and prostate cancer patients contain the cancer-associated RNA biomarkers EGFRvIII and PCA3, respectively. In addition, gene-expression profiling revealed a distinct RNA signature in platelets from glioma patients compared with normal control subjects. Because platelets are easily accessible and isolated, they may form an attractive platform for the companion diagnostics of cancer.


Journal of Clinical Oncology | 2017

Randomized, Double-Blind, Phase III Trial of Ipilimumab Versus Placebo in Asymptomatic or Minimally Symptomatic Patients With Metastatic Chemotherapy-Naive Castration-Resistant Prostate Cancer

Tomasz M. Beer; Eugene D. Kwon; Charles G. Drake; Karim Fizazi; Christopher J. Logothetis; Gwenaelle Gravis; Vinod Ganju; Jonathan Polikoff; Fred Saad; Piotr Humanski; Josep M. Piulats; Pablo Fernando Gonzalez Mella; Siobhan Ng; Dirk Jaeger; Francis Parnis; Fabio A. Franke; Javier Puente; Roman Carvajal; Lisa Sengeløv; M. Brent McHenry; Arvind Varma; Alfonsus Johannes Maria van den Eertwegh; Winald R. Gerritsen

Purpose Ipilimumab increases antitumor T-cell responses by binding to cytotoxic T-lymphocyte antigen 4. We evaluated treatment with ipilimumab in asymptomatic or minimally symptomatic patients with chemotherapy-naive metastatic castration-resistant prostate cancer without visceral metastases. Patients and Methods In this multicenter, double-blind, phase III trial, patients were randomly assigned (2:1) to ipilimumab 10 mg/kg or placebo every 3 weeks for up to four doses. Ipilimumab 10 mg/kg or placebo maintenance therapy was administered to nonprogressing patients every 3 months. The primary end point was overall survival (OS). Results Four hundred patients were randomly assigned to ipilimumab and 202 to placebo; 399 were treated with ipilimumab and 199 with placebo. Median OS was 28.7 months (95% CI, 24.5 to 32.5 months) in the ipilimumab arm versus 29.7 months (95% CI, 26.1 to 34.2 months) in the placebo arm (hazard ratio, 1.11; 95.87% CI, 0.88 to 1.39; P = .3667). Median progression-free survival was 5.6 months in the ipilimumab arm versus 3.8 with placebo arm (hazard ratio, 0.67; 95.87% CI, 0.55 to 0.81). Exploratory analyses showed a higher prostate-specific antigen response rate with ipilimumab (23%) than with placebo (8%). Diarrhea (15%) was the only grade 3 to 4 treatment-related adverse event (AE) reported in ≥ 10% of ipilimumab-treated patients. Nine (2%) deaths occurred in the ipilimumab arm due to treatment-related AEs; no deaths occurred in the placebo arm. Immune-related grade 3 to 4 AEs occurred in 31% and 2% of patients, respectively. Conclusion Ipilimumab did not improve OS in patients with metastatic castration-resistant prostate cancer. The observed increases in progression-free survival and prostate-specific antigen response rates suggest antitumor activity in a patient subset.


Gene Therapy | 2000

Recombinant adenovirus vectors with knobless fibers for targeted gene transfer

V W van Beusechem; Alct van Rijswijk; Hhg van Es; Hidde J. Haisma; H.M. Pinedo; Winald R. Gerritsen

Adenoviral vector systems for gene therapy can be much improved by targeting vectors to specific cell types. This requires both the complete ablation of native adenovirus tropism and the introduction of a novel binding affinity in the viral capsid. We reasoned that these requirements could be fulfilled by deleting the entire knob domain of the adenovirus fiber protein and replacing it with two distinct moieties that provide a trimerization function for the knobless fiber and specific binding to the target cell, respectively. To test this concept, we constructed adenoviral vectors carrying knobless fibers comprising the α-helix trimerization domain from MoMuLV envelope glycoprotein. Two mimic targeting ligands, a Myc-epitope and a 6His-tag, were attached via a flexible linker peptide. The targeted knobless fiber molecules were properly expressed and imported into the nucleus of adenovirus packaging cells, where they were incorporated as functional trimers into the adenovirus capsid. Both ligands were exposed on the surface of the virion and were available for specific binding to their target molecules. Moreover, the knobless fibers mediated gene delivery into cells displaying receptors for the coupled ligand. Hence, these knobless fibers are prototype substrates for versatile addition of targeting ligands to generate truly targeted adenoviruses.

Collaboration


Dive into the Winald R. Gerritsen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David T. Curiel

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Harm Westdorp

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Tanja D. de Gruijl

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rik J. Scheper

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerty Schreibelt

Radboud University Nijmegen

View shared research outputs
Researchain Logo
Decentralizing Knowledge