20 results on '"Vliet, H.J. van der"'
Search Results
2. Epidermal growth factor receptor as target for perioperative elimination of circulating colorectal cancer cells
- Author
-
Gruijs, M., Braster, R., Overdijk, M.B., Hellingman, T., Verploegen, S., Korthouwer, R., Wilk, B.J. van der, Parren, P.W.H.I., Vliet, H.J. van der, Boegels, M., and Egmond, M. van
- Abstract
Surgical resection of the tumor is the primary treatment of colorectal cancer patients. However, we previously demonstrated that abdominal surgery promotes the adherence of circulating tumor cells (CTC) in the liver and subsequent liver metastasis development. Importantly, preoperative treatment with specific tumor-targeting monoclonal antibodies (mAb) prevented surgery-induced liver metastasis development in rats. This study investigated whether the epidermal growth factor receptor (EGFR) represents a suitable target for preoperative antibody treatment of colorectal cancer patients undergoing surgery. The majority of patients with resectable colorectal liver metastases were shown to have EGFR + CTCs. Three different anti-EGFR mAbs (cetuximab, zalutumumab, and panitumumab) were equally efficient in the opsonization of tumor cell lines. Additionally, all three mAbs induced antibody-dependent cellular phagocytosis (ADCP) of tumor cells by macrophages at low antibody concentrations in vitro, independent of mutations in EGFR signaling pathways. The plasma of cetuximab-treated patients efficiently opsonized tumor cells ex vivo and induced phagocytosis. Furthermore, neither proliferation nor migration of epithelial cells was affected in vitro, supporting that wound healing will not be hampered by treatment with low anti-EGFR mAb concentrations. These data support the use of a low dose of anti-EGFR mAbs prior to resection of the tumor to eliminate CTCs without interfering with the healing of the anastomosis. Ultimately, this may reduce the risk of metastasis development, consequently improving long-term patient outcome significantly.
- Published
- 2022
3. A Phase I Open-Label Clinical Trial Evaluating the Therapeutic Vaccine hVEGF26-104/RFASE in Patients with Advanced Solid Malignancies
- Author
-
Goedegebuure, R.S.A., Wentink, M.Q., Vliet, H.J. van der, Timmerman, P, Griffioen, A.W., Gruijl, T.D. de, Verheul, H.M.W., Goedegebuure, R.S.A., Wentink, M.Q., Vliet, H.J. van der, Timmerman, P, Griffioen, A.W., Gruijl, T.D. de, and Verheul, H.M.W.
- Abstract
Contains fulltext : 232008.pdf (Publisher’s version ) (Open Access), LESSONS LEARNED: The novel therapeutic vaccine hVEGF(26-104) /RFASE was found to be safe and well tolerated in patients with cancer. hVEGF(26-104) /RFASE failed to induce seroconversion against native hVEGF(165) and, accordingly, neither a decrease in circulating vascular endothelial growth factor (VEGF) levels nor clinical benefit was observed. Remarkably, hVEGF(26-104) /RFASE induced VEGF(165) -neutralizing antibodies in a nonhuman primate model. The absence of seroconversion in human calls for caution in the interpretation of efficacy of human vaccines in nonhuman primates. BACKGROUND: Targeting vascular endothelial growth factor-A (VEGF) is a well-established anticancer therapy. We designed a first-in-human clinical trial to investigate the safety and immunogenicity of the novel vaccine hVEGF(26-104) /RFASE. METHODS: Patients with advanced solid malignancies with no standard treatment options available were eligible for this phase I study with a 3+3 dose-escalation design. On days 0, 14, and 28, patients received intramuscular hVEGF(26-104) , a truncated synthetic three-dimensional (3D)-structured peptide mimic covering the amino acids 26-104 of the human VEGF(165) isoform, emulsified in the novel adjuvant Raffinose Fatty Acid Sulphate Ester (RFASE), a sulpholipopolysaccharide. Objectives were to determine safety, induction of VEGF-neutralizing antibodies, and the maximum tolerated dose. Blood was sampled to measure VEGF levels and antibody titers. RESULTS: Eighteen of 27 enrolled patients received three immunizations in six different dose-levels up to 1,000 μg hVEGF(26-104) and 40 mg RFASE. No dose-limiting toxicity was observed. Although in four patients an antibody titer against hVEGF(26-104) was induced (highest titer: 2.77 (10) log), neither a reduction in VEGF levels nor neutralizing antibodies against native VEGF(165) were detected. CONCLUSION: Despite having an attractive safety profile, hVEGF(26-104) /RFASE was not able to elicit seroconversions against
- Published
- 2021
4. Survival Benefit of Repeat Local Treatment in Patients Suffering From Early Recurrence of Colorectal Cancer Liver Metastases.
- Author
-
Hellingman, T., Kuiper, B.I., Buffart, L.M., Meijerink, M.R., Versteeg, K.S., Swijnenburg, R.J., Delden, O.M. van, Haasbeek, C.J.A., Vries, J.J.J. de, Waesberghe, J.H. van, Zonderhuis, B.M., Vliet, H.J. van der, Kazemier, G., Hellingman, T., Kuiper, B.I., Buffart, L.M., Meijerink, M.R., Versteeg, K.S., Swijnenburg, R.J., Delden, O.M. van, Haasbeek, C.J.A., Vries, J.J.J. de, Waesberghe, J.H. van, Zonderhuis, B.M., Vliet, H.J. van der, and Kazemier, G.
- Abstract
01 december 2021, Contains fulltext : 244864.pdf (Publisher’s version ) (Open Access), BACKGROUND: A uniform treatment strategy for patients suffering from early recurrence after local treatment of CRLM is currently lacking. The aim of this observational cohort study was to assess the potential survival benefit of repeat local treatment compared to systemic therapy in patients suffering from early recurrence of CRLM. PATIENTS AND METHODS: Patients who developed recurrent CRLM within 12 months after initial local treatment with curative intent were retrospectively identified in Amsterdam University Medical Centers between 2009-2019. Differences in overall and progression-free survival among treatment strategies were assessed using multivariable Cox regression analyses. RESULTS: A total of 135 patients were included. Median overall survival of 41 months [range 4-135] was observed in patients who received repeat local treatment, consisting of upfront or repeat local treatment after neoadjuvant systemic therapy, compared to 24 months [range 1-55] in patients subjected to systemic therapy alone (adjusted HR = 0.42 [95%-CI: 0.25-0.72]; P = .002). Prolonged progression-free survival was observed after neoadjuvant systemic therapy followed by repeat local treatment, as compared to upfront repeat local treatment in patients with recurrent CRLM within 4 months following initial local treatment of CRLM (adjusted HR = 0.36 [95%-CI: 0.15-0.86]; P = .021). CONCLUSION: Patients with early recurrence of CRLM should be considered for repeat local treatment strategies. A multimodality approach, consisting of neoadjuvant systemic therapy followed by repeat local treatment, appeared favorable in patients with recurrence within 4 months following initial local treatment of CRLM.
- Published
- 2021
5. Natural Killer Cells and Anti-Cancer Therapies: Reciprocal Effects on Immune Function and Therapeutic Response
- Author
-
Toffoli, E.C., Sheikhi, A., Höppner, Y.D., Kok, P de, Yazdanpanah-Samani, M., Spanholtz, J., Verheul, H.M.W., Vliet, H.J. van der, Gruijl, T.D. de, Toffoli, E.C., Sheikhi, A., Höppner, Y.D., Kok, P de, Yazdanpanah-Samani, M., Spanholtz, J., Verheul, H.M.W., Vliet, H.J. van der, and Gruijl, T.D. de
- Abstract
Contains fulltext : 232069.pdf (Publisher’s version ) (Open Access), Natural Killer (NK) cells are innate immune cells with the unique ability to recognize and kill virus-infected and cancer cells without prior immune sensitization. Due to their expression of the Fc receptor CD16, effector NK cells can kill tumor cells through antibody-dependent cytotoxicity, making them relevant players in antibody-based cancer therapies. The role of NK cells in other approved and experimental anti-cancer therapies is more elusive. Here, we review the possible role of NK cells in the efficacy of various anti-tumor therapies, including radiotherapy, chemotherapy, and immunotherapy, as well as the impact of these therapies on NK cell function.
- Published
- 2021
6. The prognostic impact of circulating miRNAs in patients with advanced esophagogastric cancer during palliative chemotherapy
- Author
-
Zweeden, A.A. van, Opperman, R.C.M., Honeywell, R.J., Peters, G.J., Verheul, H.M.W., Vliet, H.J. van der, Poel, D., Zweeden, A.A. van, Opperman, R.C.M., Honeywell, R.J., Peters, G.J., Verheul, H.M.W., Vliet, H.J. van der, and Poel, D.
- Abstract
Contains fulltext : 235502.pdf (Publisher’s version ) (Open Access), The prognosis of patients with advanced oesophageal cancer (EC) and gastric cancer (GC) is poor. Circulating microRNAs (ci-miRNAs) may have prognostic and predictive value to improve patient selection for palliative treatment. The purpose of this study is to assess the prognostic and predictive value of specific ci-miRNAs in plasma of patients with EC and GC treated with first-line palliative gemcitabine and cisplatin. Droplet digital PCR (ddPCR) was used to quantify miR-200c-3p, miR-375, miR-21-5p, miR-148a-3p, miR-146a-5p, miR-141-3p and miR-218-5p in plasma from 68 patients. ci-miRNA expression was analyzed in relation to overall survival (OS), progression-free survival (PFS), and response to chemotherapy. ci-miRNA levels were detectable in 36 baseline (71%) samples and in 14 (47%) follow-up samples. Increased circulating miR-200c-3p in GC showed a trend (p = 0.06) towards a shorter OS. High circulating miR-375 was associated with a longer OS (p = 0.02) in patients with esophageal adenocarcinoma (EAC). No significant difference was observed in ci-miRNA expression between paired pre- and on-treatment samples. ci-miRNA expression was not associated with response to chemotherapy. ci-miRNAs can be measured in plasma samples of patients treated with first-line palliative chemotherapy using ddPCR despite prolonged storage in heparin. Elevated circulating miR-375 might be a prognostic marker for patients with EAC.
- Published
- 2021
7. A specific microRNA profile as predictive biomarker for systemic treatment in patients with metastatic colorectal cancer
- Author
-
Poel, D., Gootjes, E.C., Bakkerus, L., Trypsteen, W., Dekker, H., Vliet, H.J. van der, Grieken, N.C. van, Verhoef, C., Buffart, T.E., Verheul, H.M.W., Poel, D., Gootjes, E.C., Bakkerus, L., Trypsteen, W., Dekker, H., Vliet, H.J. van der, Grieken, N.C. van, Verhoef, C., Buffart, T.E., and Verheul, H.M.W.
- Abstract
Contains fulltext : 229550.pdf (publisher's version ) (Open Access), BACKGROUND: Palliative systemic therapy is currently standard of care for patients with extensive metastatic colorectal cancer (mCRC). A biomarker predicting chemotherapy benefit which prevents toxicity from ineffective treatment is urgently needed. Therefore, a previously developed tissue-derived microRNA profile to predict clinical benefit from chemotherapy was evaluated in tissue biopsies and serum from patients with mCRC. METHODS: Samples were prospectively collected from patients (N = 132) who were treated with capecitabine or 5-FU/LV with oxaliplatin ± bevacizumab. Response evaluation was performed according to RECIST 1.1 after three or four cycles, respectively. Baseline tissue and serum miRNAs expression levels of miR-17-5p, miR-20a-5p, miR-30a-5p, miR-92a-3p, miR-92b-3p, and miR-98-5p were quantified with RT-qPCR and droplet digital PCR, respectively. Combined predictive performance of selected variables was tested using logistic regression analysis. RESULTS: From 132 patients, 81 fresh frozen tissue biopsies from metastases and 93 serum samples were available. Based on expression levels of miRNAs in tissue, progressive disease could be predicted with an AUC of 0.85 (95% CI:0.72-0.91) and response could be predicted with an AUC of 0.70 (95% CI:0.56-0.80). This did not outperform clinical parameters alone (respectively P = .14 and P = .27). Expression levels of miR-92a-3p and miR-98-5p in serum significantly improved the predictive value of clinical parameters for response to chemotherapy (AUC 0.74, 95% CI:0.64-0.84, P = .003) in this cohort. CONCLUSIONS: The additive predictive value to clinical parameters of the tissue-derived six miRNA profile for clinical benefit could not be validated in patients with mCRC treated with first-line systemic therapy. Although miR-92a-3p and miR-98-5p serum levels improved the predictive value of clinical parameters, it remained insufficient for clinical decision-making.
- Published
- 2020
8. The effect of nutritional counseling on muscle mass and treatment outcome in patients with metastatic colorectal cancer undergoing chemotherapy: A randomized controlled trial
- Author
-
Werf, A. van der, Langius, J.A.E., Beeker, A., Tije, A.J. Ten, Vulink, A.J., Haringhuizen, A., Berkhof, J., Vliet, H.J. van der, Verheul, H.M.W., Schueren, M.A.E. de van der, Werf, A. van der, Langius, J.A.E., Beeker, A., Tije, A.J. Ten, Vulink, A.J., Haringhuizen, A., Berkhof, J., Vliet, H.J. van der, Verheul, H.M.W., and Schueren, M.A.E. de van der
- Abstract
Contains fulltext : 229520.pdf (publisher's version ) (Closed access), BACKGROUND & AIMS: A low muscle mass before start of treatment and loss of muscle mass during chemotherapy is related to adverse outcomes in patients with cancer. In this randomized controlled trial, the effect of nutritional counseling on change in muscle mass and treatment outcome in patients with metastatic colorectal cancer during first-line chemotherapy was studied. METHODS: Patients scheduled for first-line chemotherapy (n = 107) were randomly assigned to individualized nutritional counseling by a dietitian (NC) or usual care (UC). NC was aimed at sufficient protein- and energy intake, supported by oral supplements or enteral feeding if indicated. Furthermore, physical activity was encouraged. Outcomes were assessed at baseline (T0) and the time of the first (T1) and second (T2) regular follow-up computed tomography scans. The proportion of patients with a clinically relevant decrease in skeletal muscle area of ≥6.0 cm(2), measured by computed tomography, was the primary outcome. Secondary outcomes included body weight, quality of life, treatment toxicity and progression free and overall survival. RESULTS: A total of 107 patients were enrolled (mean age, 65 years (SD, 11 years), 63% male). Mean change in skeletal muscle area from T0 till T1 was -2.5 (SD, 9.5) cm(2), with no difference between NC versus UC (p = 0.891). The proportion of patients with a clinically relevant decrease in skeletal muscle area of ≥6.0 cm(2) did not differ (NC 30% versus UC 31%, p = 0.467). NC compared with UC had a significant positive effect on body weight (B coefficient 1.7, p = 0.045), progression free survival (p = 0.039) and overall survival (p = 0.046). CONCLUSIONS: NC of patients undergoing chemotherapy for metastatic colorectal cancer had no effect on muscle mass. However, we found that NC may increase body weight and improve progression free survival and overall survival compared to UC in this group of patients. These findings need further evaluation in future clinical trial
- Published
- 2020
9. Optimal treatment of opioid induced constipation in daily clinical practice - an observational study
- Author
-
Neefjes, E.C.W., Wijngaart, H. van der, Vorst, M.J.D.L. van der, Oever, D. Ten, Vliet, H.J. van der, Beeker, A., Rhodius, C.A., Berg, H.Pieter van den, Berkhof, J., Verheul, H.M.W., Neefjes, E.C.W., Wijngaart, H. van der, Vorst, M.J.D.L. van der, Oever, D. Ten, Vliet, H.J. van der, Beeker, A., Rhodius, C.A., Berg, H.Pieter van den, Berkhof, J., and Verheul, H.M.W.
- Abstract
Contains fulltext : 207395.pdf (publisher's version ) (Open Access), BACKGROUND: Opioids are prescribed in over 40% of patients with advanced cancer, but side effects occur frequently. In this study we evaluated the development and treatment of opioid induced constipation (OIC), and OIC resolving effect of methylnaltrexone for different opioid subtypes in daily clinical practice. METHODS: Patients with cancer using opioids were included in a retrospective chart analysis. Baseline characteristics, data on opioid use, laxative use, and OIC were collected. Patients with OIC who were prescribed methylnaltrexone, were included in a prospective observational trial (NCT01955213). RESULTS: Thirty-nine of 327 patients (pts) with cancer who were treated with opioids suffered from OIC (overall prevalence 12%; 95%-CI: 8-15%). The prevalence of OIC was similar in patients treated with oxycodone or fentanyl (12 of 81 pts. vs. 18 of 110 pts., RR 0.9; 95%CI 0.4-2.0). The morphine equivalent daily dose did not significantly differ between opioid subtypes (fentanyl 89 mg (IQR 60-180) vs. oxycodone 40 mg (40-80), P = 0.231). Twenty-two individual patients (7%) were admitted for OIC. Most effective laxatives in admitted patients were enemas, methylnaltrexone, or 4-l polyethylene-glycol solution. In the prospective observational study, the effect of methylnaltrexone could be evaluated in 23 patients. Eleven patients achieved the primary endpoint of >/=2 laxation responses out of the first four doses methylnaltrexone, independent of opioid subtype. CONCLUSIONS: OIC is a burdensome clinical problem independent of opioid subtype. Timely intensification of prophylactic laxative treatment, especially when opioid doses increase, may help to prevent OIC. Clinically overt OIC requires a more intensive laxative regimen with for example methylnaltrexone. TRIAL REGISTRATION: NCT01955213 .
- Published
- 2019
10. The effect of everolimus and low-dose cyclophosphamide on immune cell subsets in patients with metastatic renal cell carcinoma: results from a phase I clinical trial
- Author
-
Huijts, C.M., Lougheed, S.M., Bodalal, Z., Herpen, C.M.L. van, Hamberg, P., Tascilar, M., Haanen, J.B.A.G., Verheul, H.M., Gruijl, T.D. de, Vliet, H.J. van der, Huijts, C.M., Lougheed, S.M., Bodalal, Z., Herpen, C.M.L. van, Hamberg, P., Tascilar, M., Haanen, J.B.A.G., Verheul, H.M., Gruijl, T.D. de, and Vliet, H.J. van der
- Abstract
Contains fulltext : 203064.pdf (publisher's version ) (Open Access), For the treatment of metastatic renal cell cancer several strategies are used among which the mTOR inhibitor everolimus. As mTOR plays an important role in the immune system, e.g., by controlling the expression of the transcription factor FoxP3 thereby regulating regulatory T cells (Tregs), it plays a key role in the balance between tolerance and inflammation. Previous reports showed stimulatory effects of mTOR inhibition on the expansion of Tregs, an effect that can be considered detrimental in terms of cancer control. Since metronomic cyclophosphamide (CTX) was shown to selectively deplete Tregs, a phase 1 clinical trial was conducted to comprehensively investigate the immune-modulating effects of several dosages and schedules of CTX in combination with the standard dose of everolimus, with the explicit aim to achieve selective Treg depletion. Our data show that 50 mg of CTX once daily and continuously administered, in combination with the standard dose of 10 mg everolimus once daily, not only results in depletion of Tregs, but also leads to a reduction in MDSC, a sustained level of the CD8(+) T-cell population accompanied by an increased effector to suppressor ratio, and reversal of negative effects on three peripheral blood DC subsets. These positive effects on the immune response may contribute to improved survival, and therefore this combination therapy is further evaluated in a phase II clinical trial.
- Published
- 2019
11. Perioperative systemic therapy and cytoreductive surgery with HIPEC versus upfront cytoreductive surgery with HIPEC alone for isolated resectable colorectal peritoneal metastases: protocol of a multicentre, open-label, parallel-group, phase II-III, randomised, superiority study (CAIRO6)
- Author
-
Rovers, K.P., Bakkers, C., Simkens, G., Burger, J.W.A., Nienhuijs, S.W., Creemers, G.M., Thijs, A.M.J., Brandt-Kerkhof, A.R.M., Madsen, E.V., Ayez, N., Boer, N.L. de, Meerten, E. van, Tuynman, J.B., Kusters, M., Sluiter, N.R., Verheul, H.M.W., Vliet, H.J. van der, Wiezer, M.J., Boerma, D., Wassenaar, E.C.E., Los, M., Hunting, C.B., Aalbers, A.G., Kok, N.F., Kuhlmann, K.F., Boot, H., Chalabi, M., Kruijff, S., Been, L.B., Ginkel, R.J. van, Groot, D.J.A. de, Fehrmann, R.S.N., Wilt, J.H.W. de, Bremers, A.J.A., Reuver, P.R. de, Radema, S.A., Herbschleb, K.H., Grevenstein, W.M. van, Witkamp, A.J., Koopman, M., Mohammad, N. Haj, Duyn, E.B. van, Mastboom, W.J.B., Mekenkamp, L.J., Nederend, J., Lahaye, M.J., Snaebjornsson, P., Verhoef, C., Laarhoven, H.W.M. van, Zwinderman, A.H., Bouma, J.M., Kranenburg, O., Erve, I. van 't, Fijneman, R.J., Dijkgraaf, M.G., Hemmer, P.H., Punt, C.J.A., Tanis, P.J., Hingh, I. de, Rovers, K.P., Bakkers, C., Simkens, G., Burger, J.W.A., Nienhuijs, S.W., Creemers, G.M., Thijs, A.M.J., Brandt-Kerkhof, A.R.M., Madsen, E.V., Ayez, N., Boer, N.L. de, Meerten, E. van, Tuynman, J.B., Kusters, M., Sluiter, N.R., Verheul, H.M.W., Vliet, H.J. van der, Wiezer, M.J., Boerma, D., Wassenaar, E.C.E., Los, M., Hunting, C.B., Aalbers, A.G., Kok, N.F., Kuhlmann, K.F., Boot, H., Chalabi, M., Kruijff, S., Been, L.B., Ginkel, R.J. van, Groot, D.J.A. de, Fehrmann, R.S.N., Wilt, J.H.W. de, Bremers, A.J.A., Reuver, P.R. de, Radema, S.A., Herbschleb, K.H., Grevenstein, W.M. van, Witkamp, A.J., Koopman, M., Mohammad, N. Haj, Duyn, E.B. van, Mastboom, W.J.B., Mekenkamp, L.J., Nederend, J., Lahaye, M.J., Snaebjornsson, P., Verhoef, C., Laarhoven, H.W.M. van, Zwinderman, A.H., Bouma, J.M., Kranenburg, O., Erve, I. van 't, Fijneman, R.J., Dijkgraaf, M.G., Hemmer, P.H., Punt, C.J.A., Tanis, P.J., and Hingh, I. de
- Abstract
Contains fulltext : 207201.pdf (publisher's version ) (Open Access), BACKGROUND: Upfront cytoreductive surgery with HIPEC (CRS-HIPEC) is the standard treatment for isolated resectable colorectal peritoneal metastases (PM) in the Netherlands. This study investigates whether addition of perioperative systemic therapy to CRS-HIPEC improves oncological outcomes. METHODS: This open-label, parallel-group, phase II-III, randomised, superiority study is performed in nine Dutch tertiary referral centres. Eligible patients are adults who have a good performance status, histologically or cytologically proven resectable PM of a colorectal adenocarcinoma, no systemic colorectal metastases, no systemic therapy for colorectal cancer within six months prior to enrolment, and no previous CRS-HIPEC. Eligible patients are randomised (1:1) to perioperative systemic therapy and CRS-HIPEC (experimental arm) or upfront CRS-HIPEC alone (control arm) by using central randomisation software with minimisation stratified by a peritoneal cancer index of 0-10 or 11-20, metachronous or synchronous PM, previous systemic therapy for colorectal cancer, and HIPEC with oxaliplatin or mitomycin C. At the treating physician's discretion, perioperative systemic therapy consists of either four 3-weekly neoadjuvant and adjuvant cycles of capecitabine with oxaliplatin (CAPOX), six 2-weekly neoadjuvant and adjuvant cycles of 5-fluorouracil/leucovorin with oxaliplatin (FOLFOX), or six 2-weekly neoadjuvant cycles of 5-fluorouracil/leucovorin with irinotecan (FOLFIRI) followed by four 3-weekly (capecitabine) or six 2-weekly (5-fluorouracil/leucovorin) adjuvant cycles of fluoropyrimidine monotherapy. Bevacizumab is added to the first three (CAPOX) or four (FOLFOX/FOLFIRI) neoadjuvant cycles. The first 80 patients are enrolled in a phase II study to explore the feasibility of accrual and the feasibility, safety, and tolerance of perioperative systemic therapy. If predefined criteria of feasibility and safety are met, the study continues as a phase III study with 3-year overall sur
- Published
- 2019
12. Metronomic cyclophosphamide attenuates mTOR-mediated expansion of regulatory T cells, but does not impact clinical outcome in patients with metastatic renal cell cancer treated with everolimus
- Author
-
Werter, I.M., Huijts, C.M., Lougheed, S.M., Hamberg, P., Polee, M.B., Tascilar, M., Los, M., Haanen, J., Helgason, H.H., Verheul, H.M.W., Gruijl, T.D. de, Vliet, H.J. van der, Werter, I.M., Huijts, C.M., Lougheed, S.M., Hamberg, P., Polee, M.B., Tascilar, M., Los, M., Haanen, J., Helgason, H.H., Verheul, H.M.W., Gruijl, T.D. de, and Vliet, H.J. van der
- Abstract
Contains fulltext : 207393.pdf (publisher's version ) (Open Access), INTRODUCTION: Metastatic renal cell cancer (mRCC) patients have a median overall survival (mOS) of approximately 28 months. Until recently, mammalian target of rapamycin (mTOR) inhibition with everolimus was the standard second-line treatment regimen for mRCC patients, improving median progression-free survival (mPFS). Treatment with everolimus supports the expansion of immunosuppressive regulatory T cells (Tregs), which exert a negative effect on antitumor immune responses. In a phase 1 dose-escalation study, we have recently demonstrated that a low dose of 50 mg oral cyclophosphamide once daily can be safely combined with everolimus in mRCC patients and prevents the everolimus-induced increase in Tregs. MATERIALS AND METHODS: In a multicenter phase 2 study, performed in patients with mRCC not amenable to or progressive on a vascular endothelial growth factor (VEGF)-receptor tyrosine kinase inhibitor (TKI) containing treatment regimen, we assessed whether the addition of this metronomic dosing schedule of cyclophosphamide to therapy with everolimus could result in an improvement of progression-free survival (PFS) after 4 months of treatment. RESULTS: Though results from this study confirmed that combination treatment effectively lowered circulating levels of Tregs, addition of cyclophosphamide did not improve the PFS rate at 4 months. For this reason, the study was abrogated at the predefined interim analysis. CONCLUSION: Although the comprehensive immunomonitoring analysis performed in this study provides relevant information for the design of future immunotherapeutic approaches, the addition of metronomic cyclophosphamide to mRCC patients receiving everolimus cannot be recommended.
- Published
- 2019
13. Mechanisms of intimate and long-distance cross-talk between glioma and myeloid cells: How to break a vicious cycle
- Author
-
Wurdinger, T., Deumelandt, K., Vliet, H.J. van der, Wesseling, P., Gruijl, T.D. de, Wurdinger, T., Deumelandt, K., Vliet, H.J. van der, Wesseling, P., and Gruijl, T.D. de
- Abstract
Item does not contain fulltext, Glioma-associated microglia and macrophages (GAMs) and myeloid-derived suppressor cells (MDSCs) condition the glioma microenvironment to generate an immunosuppressed niche for tumour expansion. This immunosuppressive microenvironment is considered to be shaped through a complex multi-step interactive process between glioma cells, GAMs and MDSCs. Glioma cells recruit GAMs and MDSCs to the tumour site and block their maturation. Glioma cell-derived factors subsequently skew these cells towards an immunosuppressive, tumour-promoting phenotype. Finally, GAMs and MDSCs enhance immune suppression in the glioma microenvironment and promote glioma growth, invasiveness, and neovascularization. The local and distant cross-talk between glioma cells and GAMs and MDSCs is regulated by a plethora of soluble proteins and cell surface-bound factors, and possibly via extracellular vesicles and platelets. Importantly, GAMs and MDSCs have been reported to impair the efficacy of glioma therapy, in particular immunotherapeutic approaches. Therefore, advancing our understanding of the function of GAMs and MDSCs in brain tumours and targeted intervention of their immunosuppressive function may benefit the treatment of glioma.
- Published
- 2014
14. Preoperative chemoradiation therapy in combination with panitumumab for patients with resectable esophageal cancer: the PACT study
- Author
-
Kordes, S., Berge Henegouwen, M.I. van, Hulshof, M.C.C., Bergman, J.J., Vliet, H.J. van der, Kapiteijn, E., Laarhoven, H.W.M. van, Richel, D.J., Klinkenbijl, J.H.G., Meijer, S.L., Wilmink, J.W., Kordes, S., Berge Henegouwen, M.I. van, Hulshof, M.C.C., Bergman, J.J., Vliet, H.J. van der, Kapiteijn, E., Laarhoven, H.W.M. van, Richel, D.J., Klinkenbijl, J.H.G., Meijer, S.L., and Wilmink, J.W.
- Abstract
Item does not contain fulltext, PURPOSE: Preoperative chemoradiation therapy (CRT) has become the standard treatment strategy for patients with resectable esophageal cancer. This multicenter phase 2 study investigated the efficacy of the addition of the epidermal growth factor receptor (EGFR) inhibitor panitumumab to a preoperative CRT regimen with carboplatin, paclitaxel, and radiation therapy in patients with resectable esophageal cancer. METHODS AND MATERIALS: Patients with resectable cT1N1M0 or cT2-3N0 to -2M0 tumors received preoperative CRT consisting of panitumumab (6 mg/kg) on days 1, 15, and 29, weekly administrations of carboplatin (area under the curve [AUC] = 2), and paclitaxel (50 mg/m(2)) for 5 weeks and concurrent radiation therapy (41.4 Gy in 23 fractions, 5 days per week), followed by surgery. Primary endpoint was pathologic complete response (pCR) rate. We aimed at a pCR rate of more than 40%. Furthermore, we explored the predictive value of biomarkers (EGFR, HER 2, and P53) for pCR. RESULTS: From January 2010 until December 2011, 90 patients were enrolled. Patients were diagnosed predominantly with adenocarcinoma (AC) (80%), T3 disease (89%), and were node positive (81%). Three patients were not resected due to progressive disease. The primary aim was unmet, with a pCR rate of 22%. Patients with AC and squamous cell carcinoma reached a pCR of 14% and 47%, respectively. R0 resection was achieved in 95% of the patients. Main grade 3 toxicities were rash (12%), fatigue (11%), and nonfebrile neutropenia (11%). None of the biomarkers was predictive for response. CONCLUSIONS: The addition of panitumumab to CRT with carboplatin and paclitaxel was safe and well tolerated but could not improve pCR rate to the preset criterion of 40%.
- Published
- 2014
15. Taking The Great Commission Online - A Study Of American Evangelicals And Their Appropriation of the Internet
- Author
-
Vliet, H.J. van der, Bos, D. (Thesis Advisor), Verheul, J., Vliet, H.J. van der, Bos, D. (Thesis Advisor), and Verheul, J.
- Abstract
This thesis addresses an important topic that is relevant to our understanding of American society: the way American evangelical communities used the internet as a media platform to reach audiences and mobilize political support for their views. Although the political mobilization of religious groups through “televangelism” has been on the academic agenda for years, the use of the Internet is a relatively new field which Harrison van der Vliet opens up by exploring two case studies; Focus on the Family and the Southern Baptist Convention.
- Published
- 2012
16. Attenuation of invariant natural killer T-cell anergy induction through intradermal delivery of alpha-galactosylceramide.
- Author
-
Bontkes, H.J., Moreno, M., Hangalapura, B.N., Lindenberg, J.J., Groot, J. de, Lougheed, S., Vliet, H.J. van der, Eertwegh, A.J. van den, Gruijl, T.D. de, Blomberg, B.M.E. von, Scheper, R.J., Bontkes, H.J., Moreno, M., Hangalapura, B.N., Lindenberg, J.J., Groot, J. de, Lougheed, S., Vliet, H.J. van der, Eertwegh, A.J. van den, Gruijl, T.D. de, Blomberg, B.M.E. von, and Scheper, R.J.
- Abstract
1 september 2010, Item does not contain fulltext, CD1d restricted, alpha-galactosylceramide (alphaGC) responsive invariant (i)NKT cells positively regulate immune responses. Both intravenous and intradermal administered alphaGC are known to activate iNKT cells. iNKT cells become unresponsive to a second intravenous alphaGC injection, whereas no data are available regarding potential anergy upon intradermal administration. Here, comparative analysis of two intradermal versus two intravenous injections in mice demonstrated that iNKT cell anergy was prevented by intradermal injection and when combined with a vaccine, superior tumor protection afforded by intradermally administered alphaGC. Moreover, human skin dendritic cells (DC) took up intradermally injected alphaGC and activated iNKT cells upon migration, while iNKT cells in human skin-draining lymph nodes expanded in response to alphaGC presented either by exogenously added DC or by CD1d positive antigen presenting cells in the lymph nodes. In conclusion, glycolipids such as alphaGC may greatly improve the efficacy of skin immunization strategies, targeting cutaneous and lymph node DC.
- Published
- 2010
17. Chronically stimulated mouse invariant NKT cell lines have a preserved capacity to enhance protection against experimental tumor metastases.
- Author
-
Molling, J.W., Moreno, M., Groot, J. de, Vliet, H.J. van der, Blomberg, B.M.E. von, Eertwegh, A.J. van den, Scheper, R.J., Bontkes, H.J., Molling, J.W., Moreno, M., Groot, J. de, Vliet, H.J. van der, Blomberg, B.M.E. von, Eertwegh, A.J. van den, Scheper, R.J., and Bontkes, H.J.
- Abstract
Contains fulltext : 69635.pdf (publisher's version ) (Closed access), In pre-clinical models, CD1d restricted invariant Natural Killer T (iNKT) cells play a pivotal role in natural anti-tumor immune responses, mainly by trans-activating cells of both the innate and adaptive arms via swift and potent cytokine secretion. We have previously reported that patients with a severely reduced circulating iNKT cell pool have a poor clinical response to radio therapy of head and neck squamous cell carcinoma. Therefore, these patients might benefit from an immunotherapeutic approach aimed at the increase of circulating levels of iNKT cells. Furthermore, we have generated both human and mouse iNKT cell lines, and demonstrated that they had retained the capacity to release both Th1 and Th2 type cytokines even after long-term in vitro expansion using alpha-galactosylceramide (alphaGalCer) pulsed dendritic cells (DC). Here, we establish, in a pre-clinical tumor model that the large scale long lived polyclonal iNKT cell lines we generated have a preserved capacity to evoke an in vivo cytokine storm upon adoptive transfer, independently of supplemental alphaGalCer administration. This results in an augmented NK cell mediated protection against B16.F10 experimental lung metastases in vivo. These findings underscore the potential of autologous adoptive transfer of ex vivo expanded iNKT cells as a strategy to enhance immunotherapeutic modalities for the treatment of cancer patients.
- Published
- 2008
18. Invariant natural killer T cells and immunotherapy of cancer.
- Author
-
Molling, J.W., Moreno, M., Vliet, H.J. van der, Eertwegh, A.J. van den, Scheper, R.J., Blomberg, B.M.E. von, Bontkes, H.J., Molling, J.W., Moreno, M., Vliet, H.J. van der, Eertwegh, A.J. van den, Scheper, R.J., Blomberg, B.M.E. von, and Bontkes, H.J.
- Abstract
Contains fulltext : 69606.pdf (publisher's version ) (Closed access), Invariant CD1d restricted natural killer T (iNKT) cells are regulatory cells that express a canonical TCR-Valpha-chain (Valpha24.Jalpha18 in humans and Valpha14.Jalpha18 in mice) which recognizes glycolipid antigens presented by the monomorphic CD1d molecule. They can secrete a wide variety of both pro-inflammatory and anti-inflammatory cytokines very swiftly upon their activation. Evidence for the significance of iNKT cells in human cancer has been ambiguous. Still, the (pre-)clinical findings reviewed here, provide evidence for a distinct contribution of iNKT cells to natural anti-tumor immune responses in humans. Furthermore, clinical phase I studies that are discussed here have revealed that the infusion of cancer patients with ligand-loaded dendritic cells or cultured iNKT cells is well tolerated. We thus underscore the potential of iNKT cell based immunotherapy in conjunction with established modalities such as surgery and radiotherapy, as adjuvant therapy against carcinomas.
- Published
- 2008
19. Low levels of circulating invariant natural killer T cells predict poor clinical outcome in patients with head and neck squamous cell carcinoma.
- Author
-
Molling, J.W., Langius, J.A.E., Langendijk, J.A., Leemans, C.R., Bontkes, H.J., Vliet, H.J. van der, Blomberg, B.M.E. von, Scheper, R.J., Eertwegh, A.J. van den, Molling, J.W., Langius, J.A.E., Langendijk, J.A., Leemans, C.R., Bontkes, H.J., Vliet, H.J. van der, Blomberg, B.M.E. von, Scheper, R.J., and Eertwegh, A.J. van den
- Abstract
Contains fulltext : 53110.pdf (publisher's version ) (Closed access), PURPOSE: Evading antitumor immune responses is an important aspect of the pathogenesis of head and neck squamous cell carcinoma (HNSCC). Invariant CD1d-restricted natural killer T (iNKT) cells play an allegedly pivotal role in such responses via transactivation of immune effector cells. It has been reported that iNKT cells are reduced in peripheral blood of cancer patients compared with healthy controls. Here, we investigated whether the extent of this deficiency affected disease outcome in HNSCC patients. PATIENTS AND METHODS: In a prospective study, circulating iNKT cell numbers were evaluated in 47 patients before radiotherapy. Patients were stratified in three groups based on iNKT cell levels, and clinical data were obtained during a median follow-up period of 31 months. RESULTS: A small, compared with an intermediate or large, circulating iNKT cell fraction was significantly associated with decreased 3-year overall survival rate (39% v 75% and 92%, respectively), disease-specific survival rate (43% v 87% and 92%, respectively), and locoregional control rate (31% v 74% and 92%, respectively) in HNSCC patients. Cox regression revealed that the iNKT cell level, as well as clinical T stage, was an independent prognostic parameter even after correction for the confounding effect of age. CONCLUSION: A severe circulating iNKT cell deficiency was related to poor clinical outcome in HNSCC patients, suggesting their critical contribution to antitumor immune responses. Furthermore, screening for iNKT cell levels may be useful for determining which patients can benefit from immunotherapeutic adjuvant therapies aimed at reconstitution of the circulating iNKT cell pool.
- Published
- 2007
20. Generation and sustained expansion of mouse spleen invariant NKT cell lines with preserved cytokine releasing capacity.
- Author
-
Molling, J.W., Moreno, M., Vliet, H.J. van der, Blomberg, B.M.E. von, Eertwegh, A.J. van den, Scheper, R.J., Bontkes, H.J., Molling, J.W., Moreno, M., Vliet, H.J. van der, Blomberg, B.M.E. von, Eertwegh, A.J. van den, Scheper, R.J., and Bontkes, H.J.
- Abstract
Contains fulltext : 53448.pdf (publisher's version ) (Closed access), Invariant Natural Killer T (iNKT) cells are CD1d restricted innate lymphoid cells with an invariant T cell receptor (TCR) alpha chain gene rearrangement (Valpha24-Jalpha18 in human and Valpha14-Jalpha18 in mouse). iNKT cells play a pivotal role in anti-tumor immune responses via cytokine mediated transactivation of various cells which mediate innate and adaptive immune responses. Here we describe, to our knowledge for the first time, the generation of long-term mouse spleen derived iNKT cell lines. We found that dendritic cells (DC) derived from the D1 line, but not Mf4/4 macrophages, loaded with the artificial iNKT cell ligand alpha-Galactosylceramide (alphaGalCer) could be employed to expand iNKT cells in vitro. Furthermore, exogenously added IL-7, but not IL-2 or IL-15 had a pronounced additive effect on iNKT cell expansion. Using this method up to 10(8) iNKT cells could be obtained from one spleen within 12 to 14 weeks, and cell lines could be continued for up to 24 months. Importantly, the iNKT cell lines had retained the capacity to swiftly secrete substantial amounts of both T helper (Th) 1 and Th2 cytokines upon activation. In conclusion we have generated iNKT cell lines with high yields that can be maintained for up to 24 months, by repeated stimulation using alpha-GalCer loaded D1.DC and IL-7. These in vitro expanded iNKT cells preserved the capacity to swiftly produce both Th1 and Th2 type cytokines and are currently being utilized in pre-clinical adoptive transfer models to identify and optimize the characteristics of therapeutically effective iNKT cells in an anti-tumor setting.
- Published
- 2007
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.