Network


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

Hotspot


Dive into the research topics where A. Rob P. M. Valentijn is active.

Publication


Featured researches published by A. Rob P. M. Valentijn.


The New England Journal of Medicine | 2009

Vaccination against HPV-16 Oncoproteins for Vulvar Intraepithelial Neoplasia

Gemma G. Kenter; Marij J. P. Welters; A. Rob P. M. Valentijn; Margriet J. G. Löwik; Dorien M. A. Berends-van der Meer; Annelies P.G. Vloon; Farah Essahsah; Lorraine M. Fathers; Rienk Offringa; Jan W. Drijfhout; Amon R. Wafelman; Jaap Oostendorp; Gert Jan Fleuren; Sjoerd H. van der Burg; Cornelis J. M. Melief

BACKGROUND Vulvar intraepithelial neoplasia is a chronic disorder caused by high-risk types of human papillomavirus (HPV), most commonly HPV type 16 (HPV-16). Spontaneous regression occurs in less than 1.5% of patients, and the rate of recurrence after treatment is high. METHODS We investigated the immunogenicity and efficacy of a synthetic long-peptide vaccine in women with HPV-16-positive, high-grade vulvar intraepithelial neoplasia. Twenty women with HPV-16-positive, grade 3 vulvar intraepithelial neoplasia were vaccinated three or four times with a mix of long peptides from the HPV-16 viral oncoproteins E6 and E7 in incomplete Freunds adjuvant. The end points were clinical and HPV-16-specific T-cell responses. RESULTS The most common adverse events were local swelling in 100% of the patients and fever in 64% of the patients; none of these events exceeded grade 2 of the Common Terminology Criteria for Adverse Events of the National Cancer Institute. At 3 months after the last vaccination, 12 of 20 patients (60%; 95% confidence interval [CI], 36 to 81) had clinical responses and reported relief of symptoms. Five women had complete regression of the lesions, and HPV-16 was no longer detectable in four of them. At 12 months of follow-up, 15 of 19 patients had clinical responses (79%; 95% CI, 54 to 94), with a complete response in 9 of 19 patients (47%; 95% CI, 24 to 71). The complete-response rate was maintained at 24 months of follow-up. All patients had vaccine-induced T-cell responses, and post hoc analyses suggested that patients with a complete response at 3 months had a significantly stronger interferon-gamma-associated proliferative CD4+ T-cell response and a broad response of CD8+ interferon-gamma T cells than did patients without a complete response. CONCLUSIONS Clinical responses in women with HPV-16-positive, grade 3 vulvar intraepithelial neoplasia can be achieved by vaccination with a synthetic long-peptide vaccine against the HPV-16 oncoproteins E6 and E7. Complete responses appear to be correlated with induction of HPV-16-specific immunity.


Clinical Cancer Research | 2008

Induction of Tumor-Specific CD4+ and CD8+ T-Cell Immunity in Cervical Cancer Patients by a Human Papillomavirus Type 16 E6 and E7 Long Peptides Vaccine

Marij J. P. Welters; Gemma G. Kenter; Sytse J. Piersma; Annelies P.G. Vloon; Margriet J. G. Löwik; Dorien M. A. Berends-van der Meer; Jan W. Drijfhout; A. Rob P. M. Valentijn; Amon R. Wafelman; Jaap Oostendorp; Gert Jan Fleuren; Rienk Offringa; Cornelis J. M. Melief; Sjoerd H. van der Burg

Purpose: The study aims to evaluate the effect of a human papillomavirus type 16 (HPV16) E6 and E7 synthetic long peptides vaccine on the antigen-specific T-cell response in cervical cancer patients. Experimental Design: Patients with resected HPV16-positive cervical cancer were vaccinated with an overlapping set of long peptides comprising the sequences of the HPV16 E6 and E7 oncoproteins emulsified in Montanide ISA-51. HPV16-specific T-cell immune responses were analyzed by evaluating the magnitude, breadth, type, and polarization by proliferation assays, IFNγ-ELISPOT, and cytokine production and phenotyped by the T-cell markers CD4, CD8, CD25, and Foxp3. Results: Vaccine-induced T-cell responses against HPV16 E6 and E7 were detected in six of six and five of six patients, respectively. These responses were broad, involved both CD4+ and CD8+ T cells, and could be detected up to 12 months after the last vaccination. The vaccine-induced responses were dominated by effector type CD4+CD25+Foxp3− type 1 cytokine IFNγ-producing T cells but also included the expansion of T cells with a CD4+CD25+Foxp3+ phenotype. Conclusions: The HPV16 E6 and E7 synthetic long peptides vaccine is highly immunogenic, in that it increases the number and activity of HPV16-specific CD4+ and CD8+ T cells to a broad array of epitopes in all patients. The expansion of CD4+ and CD8+ tumor-specific T cells, both considered to be important in the antitumor response, indicates the immunotherapeutic potential of this vaccine. Notably, part of the vaccine-induced T cells display a CD4+CD25+Foxp3+ phenotype that is frequently associated with regulatory T-cell function, suggesting that strategies to disarm this subset of T cells should be considered as components of immunotherapeutic modalities against HPV-induced cancers.


Clinical Cancer Research | 2008

Phase I Immunotherapeutic Trial with Long Peptides Spanning the E6 and E7 Sequences of High-Risk Human Papillomavirus 16 in End-Stage Cervical Cancer Patients Shows Low Toxicity and Robust Immunogenicity

Gemma G. Kenter; Marij J. P. Welters; A. Rob P. M. Valentijn; Margriet J. G. Löwik; Dorien M. A. Berends-van der Meer; Annelies P.G. Vloon; Jan W. Drijfhout; Amon R. Wafelman; Jaap Oostendorp; Gert Jan Fleuren; Rienk Offringa; Sjoerd H. van der Burg; Cornelis J. M. Melief

Purpose: To determine the toxicity, safety, and immunogenicity of a human papillomavirus 16 (HPV16) E6 and E7 long peptide vaccine administered to end-stage cervical cancer patients. Experimental Design: Three groups of end-stage cervical cancer patients (in total n = 35) were s.c. vaccinated with HPV16 E6 combined with or separated from HPV16 E7 overlapping long peptides in Montanide ISA-51 adjuvant, four times at 3-week intervals. Group 1 received 300 μg/peptide at a single site and group 2 received 100 μg/peptide of the E6 peptides in one limb and 300 μg/peptide of the E7 peptides in a second limb. Group 3 received separate injections of E6 and E7 peptides, each at a dose of 50 μg/peptide. The primary end point was to determine safety and toxicity of the HPV16 long peptides vaccine. In addition, the vaccine-induced T-cell response was assessed by IFNγ enzyme-linked immunospot. Results: No toxicity beyond grade 2 was observed during and after four vaccinations. In a few patients, transient flu-like symptoms were observed. Enzyme-linked immunospot analysis of the vaccine-induced immune response revealed that coinjection of the E6 and E7 peptides resulted in a strong and broad T-cell response dominated by immunity against E6. Injection of the E6 and E7 peptides at two different sites increased the E7 response but did not affect the magnitude of the E6-induced immune response. Conclusions: The HPV16 E6 and E7 long peptide-based vaccine is well tolerated and capable of inducing a broad IFNγ-associated T-cell response even in end-stage cervical cancer patients.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Success or failure of vaccination for HPV16-positive vulvar lesions correlates with kinetics and phenotype of induced T-cell responses

Marij J. P. Welters; Gemma G. Kenter; Peggy J. de Vos van Steenwijk; Margriet J. G. Löwik; Dorien M. A. Berends-van der Meer; Farah Essahsah; Linda F. M. Stynenbosch; Annelies P.G. Vloon; Tamara H. Ramwadhdoebe; Sytse J. Piersma; Jeanette M. van der Hulst; A. Rob P. M. Valentijn; Lorraine M. Fathers; Jan W. Drijfhout; Kees L. M. C. Franken; Jaap Oostendorp; Gert Jan Fleuren; Cornelis J. M. Melief; Sjoerd H. van der Burg

One half of a group of 20 patients with human papillomavirus type 16 (HPV16)-induced vulvar intraepithelial neoplasia grade 3 displayed a complete regression (CR) after therapeutic vaccination with HPV16 E6/E7 synthetic long peptides. Patients with relatively larger lesions generally did not display a CR. To investigate immune correlates of treatment failure, patients were grouped according to median lesion size at study entry, and HPV16-specific immunity was analyzed at different time points by complementary immunological assays. The group of patients with smaller lesions displayed stronger and broader vaccine-prompted HPV16-specific proliferative responses with higher IFNγ (P = 0.0003) and IL-5 (P < 0.0001) levels than patients with large lesions. Characteristically, this response was accompanied by a distinct peak in cytokine levels after the first vaccination. In contrast, the patient group with larger lesions mounted higher frequencies of HPV16-specific CD4+CD25+Foxp3+ T cells (P = 0.005) and displayed a lower HPV16-specific IFNγ/IL-10 ratio after vaccination (P < 0.01). No disparity in T memory immunity to control antigens was found, indicating that the differences in HPV-specific immunity did not reflect general immune failure. We observed a strong correlation between a defined set of vaccine-prompted specific immune responses and the clinical efficacy of therapeutic vaccination. Notably, a high ratio of HPV16-specific vaccine-prompted effector T cells to HPV16-specific CD4+CD25+Foxp3+ T cells was predictive of clinical success. Foxp3+ T cells have been associated previously with impaired immunity in malignancies. Here we demonstrate that the vaccine-prompted level of this population is associated with early treatment failure.


Clinical Cancer Research | 2009

Induction of p53-Specific Immunity by a p53 Synthetic Long Peptide Vaccine in Patients Treated for Metastatic Colorectal Cancer

Frank M. Speetjens; Peter J. K. Kuppen; Marij J. P. Welters; Farah Essahsah; Anne Marie E.G. Voet van den Brink; M. Graziella Kallenberg Lantrua; A. Rob P. M. Valentijn; Jaap Oostendorp; Lorraine M. Fathers; Hans W. Nijman; Jan W. Drijfhout; Cornelis J. H. van de Velde; Cornelis J. M. Melief; Sjoerd H. van der Burg

Purpose: The tumor-associated self-antigen p53 is commonly overexpressed in cancer, including colorectal cancer, and can serve as a target for immunotherapy. The safety and immunogenicity of a p53 synthetic long peptide (p53-SLP) vaccine were investigated in patients treated for metastatic colorectal cancer. Experimental Design: Ten patients were vaccinated twice with a set of 10 overlapping p53-SLP in a phase I/II trial. Both the safety and the breadth, magnitude, and polarization of vaccine-induced p53-specific T cells was evaluated in blood samples drawn before and after vaccination by IFN-γ enzyme-linked immunospot, proliferation, cytokine secretion, and multiparameter flow cytometry. The migratory capacity of p53-specific T cells was evaluated by assessing their presence in a biopsy of the second vaccination site. Results: Toxicity was limited to grade 1/2, mostly at the vaccination site. p53-specific T-cell responses were induced in 9 of 10 colorectal cancer patients as measured by IFN-γ enzyme-linked immunospot, proliferation, and cytokine bead array. In 6 of 9 tested patients, p53-specific T-cell reactivity persisted at least 6 months. Furthermore, p53-specific T cells isolated from the vaccination site were characterized as CD4+ T cells producing both T-helper types 1 and 2 cytokines on stimulation with p53 peptide and p53 protein. Multiparameter flow cytometry revealed that only a minor population of the p53-specific CD4+ T cells was optimally polarized. Conclusions: The p53-SLP vaccine is safe and capable to induce p53-specific T-cell responses in patients treated for colorectal cancer. New trials should focus on improving the polarization of the p53-SLP vaccine-induced T-cell response.


International Journal of Cancer | 2009

Immunization with a P53 synthetic long peptide vaccine induces P53-specific immune responses in ovarian cancer patients, a phase II trial

Ninke Leffers; Annechien Lambeck; Marloes Gooden; Baukje-Nynke Hoogeboom; Rinze Wolf; Ineke E. Hamming; Bouke G. Hepkema; Pax H.B. Willemse; Barbara H. W. Molmans; Harry Hollema; Jan W. Drijfhout; Willem Sluiter; A. Rob P. M. Valentijn; Loraine M. Fathers; Jaap Oostendorp; Ate G.J. van der Zee; Cornelis Joseph Melief; Sjoerd H. van der Burg; Toos Daemen; Hans W. Nijman

The prognosis of ovarian cancer, the primary cause of death from gynecological malignancies, has only modestly improved over the last decades. Immunotherapy is one of the new treatment modalities explored for this disease. To investigate safety, tolerability, immunogenicity and obtain an impression of clinical activity of a p53 synthetic long peptide (p53‐SLP) vaccine, twenty patients with recurrent elevation of CA‐125 were included, eighteen of whom were immunized 4 times with 10 overlapping p53‐SLP in Montanide ISA51. The first 5 patients were extensively monitored for toxicity, but showed no ≥ grade 3 toxicity, thus accrual was continued. Overall, toxicity was limited to grade 1 and 2, mostly locoregional, inflammatory reactions. IFN‐γ producing p53‐specific T‐cell responses were induced in all patients who received all 4 immunizations as measured by IFN‐γ ELISPOT. An IFN‐γ secretion assay showed that vaccine‐induced p53‐specific T‐cells were CD4+, produced both Th1 and Th2 cytokines as analyzed by cytokine bead array. Notably, Th2 cytokines dominated the p53‐specific response. P53‐specific T‐cells were present in a biopsy of the last immunization site of at least 9/17 (53%) patients, reflecting the migratory capacity of p53‐specific T‐cells. As best clinical response, stable disease evaluated by CA‐125 levels and CT‐scans, was observed in 2/20 (10%) patients, but no relationship was found with vaccine‐induced immunity. This study shows that the p53‐SLP vaccine is safe, well tolerated and induces p53‐specific T‐cell responses in ovarian cancer patients. Upcoming trials will focus on improving T helper‐1 polarization and clinical efficacy.


Journal of Translational Medicine | 2013

HPV16 synthetic long peptide (HPV16-SLP) vaccination therapy of patients with advanced or recurrent HPV16-induced gynecological carcinoma, a phase II trial.

Mariette I.E. van Poelgeest; Marij J. P. Welters; Edith M.G. van Esch; Linda F. M. Stynenbosch; Gijs Kerpershoek; Els L. van Persijn van Meerten; Muriel van den Hende; Margriet J. G. Löwik; Dorien M. A. Berends-van der Meer; Lorraine M. Fathers; A. Rob P. M. Valentijn; Jaap Oostendorp; Gert Jan Fleuren; Cornelis J. M. Melief; Gemma G. Kenter; Sjoerd H. van der Burg

BackgroundHuman papilloma virus type 16 (HPV16)-induced gynecological cancers, in particular cervical cancers, are found in many women worldwide. The HPV16 encoded oncoproteins E6 and E7 are tumor-specific targets for the adaptive immune system permitting the development of an HPV16-synthetic long peptide (SLP) vaccine with an excellent treatment profile in animal models. Here, we determined the toxicity, safety, immunogenicity and efficacy of the HPV16 SLP vaccine in patients with advanced or recurrent HPV16-induced gynecological carcinoma.MethodsPatients with HPV16-positive advanced or recurrent gynecological carcinoma (n = 20) were subcutaneously vaccinated with an HPV16-SLP vaccine consisting of a mix of 13 HPV16 E6 and HPV16 E7 overlapping long peptides in Montanide ISA-51 adjuvant. The primary endpoints were safety, toxicity and tumor regression as determined by RECIST. In addition, the vaccine-induced T-cell response was assessed by proliferation and associated cytokine production as well as IFNγ-ELISPOT.ResultsNo systemic toxicity beyond CTCAE grade II was observed. In a few patients transient flu-like symptoms were observed. In 9 out of 16 tested patients vaccine-induced HPV16-specific proliferative responses were detected which were associated with the production of IFNγ, TNFα, IL-5 and/or IL-10. ELISPOT analysis revealed a vaccine-induced immune response in 11 of the 13 tested patients. The capacity to respond to the vaccine was positively correlated to the patient’s immune status as reflected by their response to common recall antigens at the start of the trial. Median survival was 12.6 ± 9.1 months. No regression of tumors was observed among the 12 evaluable patients. Nineteen patients died of progressive disease.ConclusionsThe HPV16-SLP vaccine was well tolerated and induced a broad IFNγ-associated T-cell response in patients with advanced or recurrent HPV16-induced gynecological carcinoma but neither induced tumor regression nor prevented progressive disease. We, therefore, plan to use this vaccine in combination with chemotherapy and immunomodulation.


Cancer Immunology, Immunotherapy | 2012

A placebo-controlled randomized HPV16 synthetic long-peptide vaccination study in women with high-grade cervical squamous intraepithelial lesions

Peggy J. de Vos van Steenwijk; Tamara H. Ramwadhdoebe; Margriet J. G. Löwik; Caroline E. van der Minne; Dorien M. A. Berends-van der Meer; Lorraine M. Fathers; A. Rob P. M. Valentijn; Jaap Oostendorp; Gert Jan Fleuren; Bart W. J. Hellebrekers; Marij J. P. Welters; Mariette I.E. van Poelgeest; Cornelis J. M. Melief; Gemma G. Kenter; Sjoerd H. van der Burg

The aim of this study was to investigate the capacity of an HPV16 E6/E7 synthetic overlapping long-peptide vaccine to stimulate the HPV16-specific T-cell response, to enhance the infiltration of HPV16-specific type 1 T cells into the lesions of patients with HPV16+ high-grade cervical squamous intraepithelial lesion (HSIL) and HPV clearance. This was a placebo-controlled randomized phase II study in patients with HPV16-positive HSIL. HPV16-specific T-cell responses were determined pre- and post-vaccination by ELISPOT, proliferation assay and cytokine assays in PBMC and HSIL-infiltrating lymphocytes, and delayed-type hypersensitivity skin tests. Motivational problems of this patient group to postpone treatment of their premalignant lesions affected the inclusion rates and caused the study to stop prematurely. Of the accrued patients, 4 received a placebo and 5 received 1–2 vaccinations. Side effects mainly were flu-like symptoms and injection site reactions. A strong HPV-specific IFNγ-associated T-cell response was detected by ELISPOT in all vaccinated patients. The outcome of the skin tests correlated well with the ELISPOT analysis. The cytokine profile associated with HPV16-specific proliferation varied from robust type 1 to dominant type 2 responses. No conclusions could be drawn on vaccine-enhanced T-cell infiltration of the lesion, and there was no HPV clearance at the time of LEEP excision. Thus, vaccination of HSIL patients results in increased HPV16-specific T-cell immunity. Further development of this type of treatment relies on the ability to motivate patients and in the reduction in the side effects.


Tetrahedron | 1997

Solid-phase synthesis of lysine-based cluster galactosides with high affinity for the asialoglycoprotein Receptor

A. Rob P. M. Valentijn; Gijgs A. van der Marel; Leo A. J. M. Sliedregt; Theo J.C. van Berkel; Erik A.L. Biessen; Jacques H. van Boom

Abstract Structurally well defined di- and tri-antennary lysine-based galactose- and N-acetylgalactosamine-containing ligands for the hepatic asialoglycoprotein receptor (ASGP-R) could be assembled on a solid support, using a combined Fmoc/Alloc protecting group strategy for the amino functions of lysine. This methodology allowed easy introduction of spacers, the length of which could be readily accommodated for optimal binding to the ASGP-R.


International Journal of Cancer | 2013

Addition of interferon-α to the p53-SLP® vaccine results in increased production of interferon-γ in vaccinated colorectal cancer patients: a phase I/II clinical trial.

Eliane C.M. Zeestraten; Frank M. Speetjens; Marij J. P. Welters; Sepideh Saadatmand; Linda F. M. Stynenbosch; Rogier Jongen; Ellen Kapiteijn; Hans Gelderblom; Hans W. Nijman; A. Rob P. M. Valentijn; Jaap Oostendorp; Lorraine M. Fathers; Jan W. Drijfhout; Cornelis J. H. van de Velde; Peter J. K. Kuppen; Sjoerd H. van der Burg; Cornelis J. M. Melief

We previously established safety and immunogenicity of a p53 synthetic long peptides (p53‐SLP®) vaccine. In the current trial, we investigated whether combination of interferon‐alpha (IFN‐α) with p53‐SLP® is both safe and able to improve the induced p53‐specific IFN‐γ response. Eleven colorectal cancer patients successfully treated for metastatic disease were enrolled in this study. Of these, nine patients completed follow‐up after two injections with p53‐SLP® together with IFN‐α. Safety and p53‐specific immune responses were determined before and after vaccination. Furthermore, cryopreserved PBMCs were compared head‐to‐head to cryopreserved PBMCs obtained in our previous trial with p53‐SLP® only. Toxicity of p53‐SLP® vaccination in combination with IFN‐α was limited to Grade 1 or 2, with predominantly small ongoing swellings at the vaccination site. All patients harbored p53‐specific T cells after vaccination and most patients showed p53‐specific antibodies. Compared to the previous trial, addition of IFN‐α significantly improved the frequency of p53‐specific T cells in IFN‐γ ELISPOT. Moreover, in this trial, p53‐specific T cells were detectable in blood samples of all patients in a direct ex vivo multiparameter flowcytometric assay, opposed to only 2 of 10 patients vaccinated with p53‐SLP® only. Finally, patients in this trial displayed a broader p53‐specific immunoglobulin‐G response, indicating an overall better p53‐specific T‐helper response. Our study shows that p53‐SLP® vaccination combined with IFN‐α injection is safe and capable of inducing p53‐specific immunity. When compared to a similar trial with p53‐SLP® vaccination alone the combination was found to induce significantly more IFN‐γ producing p53‐specific T cells.

Collaboration


Dive into the A. Rob P. M. Valentijn's collaboration.

Top Co-Authors

Avatar

Sjoerd H. van der Burg

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Cornelis J. M. Melief

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jan W. Drijfhout

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Marij J. P. Welters

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jaap Oostendorp

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gemma G. Kenter

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gert Jan Fleuren

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge