40 results on '"Stuurman D"'
Search Results
2. Darolutamide does not interfere with OATP-mediated uptake of docetaxel
- Author
-
Buck, SAJ, Talebi, Z, Drabison, T, Jin, Y, Gibson, AA, Hu, P, de Bruijn, P, de Ridder, CMA, Stuurman, D, Hu, SY, van Weerden, WM, Koolen, SLW, de Wit, R, Sparreboom, A, Mathijssen, RHJ, Eisenmann, ED, Buck, SAJ, Talebi, Z, Drabison, T, Jin, Y, Gibson, AA, Hu, P, de Bruijn, P, de Ridder, CMA, Stuurman, D, Hu, SY, van Weerden, WM, Koolen, SLW, de Wit, R, Sparreboom, A, Mathijssen, RHJ, and Eisenmann, ED
- Abstract
The addition of darolutamide, an androgen receptor signalling inhibitor, to therapy with docetaxel has recently been approved as a strategy to treat metastatic prostate cancer. OATP1B3 is an SLC transporter that is highly expressed in prostate cancer and is responsible for the accumulation of substrates, including docetaxel, into tumours. Given that darolutamide inhibits OATP1B3 in vitro, we sought to characterise the impact of darolutamide on docetaxel pharmacokinetics. We investigated the influence of darolutamide on OATP1B3 transport using in vitro and in vivo models. We assessed the impact of darolutamide on the tumour accumulation of docetaxel in a patient-derived xenograft (PDX) model and on an OATP1B biomarker in patients. Darolutamide inhibited OATP1B3 in vitro at concentrations higher than the reported Cmax. Consistent with these findings, in vivo studies revealed that darolutamide does not influence the pharmacokinetics of Oatp1b substrates, including docetaxel. Docetaxel accumulation in PDX tumours was not decreased in the presence of darolutamide. Metastatic prostate cancer patients had similar levels of OATP1B biomarkers, regardless of treatment with darolutamide. Consistent with a low potential to inhibit OATP1B3-mediated transport in vitro, darolutamide does not significantly impede the transport of Oatp1b substrates in vivo or in patients. Our findings support combined treatment with docetaxel and darolutamide, as no OATP1B3 transporter based drug–drug interaction was identified.
- Published
- 2024
3. Side-by-side comparison of the two widely studied GRPR radiotracers, radiolabeled NeoB and RM2, in a preclinical setting
- Author
-
Damiana, T. S.T., Paraïso, P., de Ridder, C., Stuurman, D., Seimbille, Y., Dalm, S. U., Damiana, T. S.T., Paraïso, P., de Ridder, C., Stuurman, D., Seimbille, Y., and Dalm, S. U.
- Abstract
Introduction: NeoB and RM2 are the most investigated gastrin-releasing peptide receptor (GRPR)–targeting radiotracers in preclinical and clinical studies. Therefore, an extensive side-by-side comparison of the two radiotracers is valuable to demonstrate whether one has advantages over the other. Accordingly, this study aims to compare the in vitro and in vivo characteristics of radiolabeled NeoB and RM2 to guide future clinical studies. Method: The stability of the radiolabeled GRPR analogs was determined in phosphate buffered saline (PBS), and commercially available mouse and human serum. Target affinity was determined by incubating human prostate cancer PC-3 cells with [177Lu]Lu-NeoB or [177Lu]Lu-RM2, + / − increasing concentrations of unlabeled NeoB, RM2, or Tyr4-bombesin (BBN). To determine uptake and specificity cells were incubated with [177Lu]Lu-NeoB or [177Lu]Lu-RM2 + / − Tyr4-BBN. Moreover, in vivo studies were performed to determine biodistribution and pharmacokinetics. Finally, radiotracer binding to various GRPR-expressing human cancer tissues was investigated. Results: Both radiotracers demonstrated high stability in PBS and human serum, but stability in mouse serum decreased substantially over time. Moreover, both radiotracers demonstrated high GRPR affinity and specificity, but a higher uptake of [177Lu]Lu-NeoB was observed in in vitro studies. In vivo, no difference in tumor uptake was seen. The most prominent difference in uptake in physiological organs was observed in the GRPR-expressing pancreas; [177Lu]Lu-RM2 had less pancreatic uptake and a shorter pancreatic half-life than [177Lu]Lu-NeoB. Furthermore, [177Lu]Lu-RM2 presented with a lower tumor-to-kidney ratio, while the tumor-to-blood ratio was lower for [177Lu]Lu-NeoB. The autoradiography studies revealed higher bind
- Published
- 2023
4. Standardization of viability assays and high-content live-cell imaging protocols for large-scale drug testing in prostate cancer PDX-derived organoids
- Author
-
Van Hemelryk, A., primary, Erkens-Schulze, S., additional, Lam, L., additional, Stuurman, D., additional, de Ridder, C.M.A., additional, French, P.J., additional, van Royen, M.E., additional, and van Weerden, W.M., additional
- Published
- 2022
- Full Text
- View/download PDF
5. Patient-derived xenografts and organoids recapitulate castration-resistant prostate cancer with sustained androgen receptor signaling
- Author
-
Van Hemelryk, A., primary, Tomljanovic, I., additional, Stuurman, D., additional, de Ridder, C.M.A., additional, Teubel, W.J., additional, Erkens-Schulze, S., additional, van de Werken, H.J.G., additional, van Royen, M., additional, Grudniewska, M., additional, Jenster, G.W., additional, and van Weerden, W.M., additional
- Published
- 2022
- Full Text
- View/download PDF
6. Determining the ideal mass and clinical potential of a bimodal probe for image-guided surgery of GRPR-positive cancer
- Author
-
Verhoeven, M., Handula, M., Van den Brink, L., De Ridder, C. M. A., Stuurman, D. C., Seimbille, Y., Dalm, S. U., Radiology & Nuclear Medicine, and Urology
- Subjects
SDG 3 - Good Health and Well-being - Published
- 2022
7. Reovirus mutant jin-3 exhibits lytic and immune-stimulatory effects in preclinical human prostate cancer models
- Author
-
Merbel, A.F. van de, Horst, G. ter, Mark, M.H. van der, Bots, S.T.F., Wollenberg, D.J.M. van den, Ridder, C.M., Stuurman, D., Aalders, T., Erkens-Schulz, S., Montfoort, N. van, Karthaus, W.R., Mehra, N., Smits, M., Schalken, J.A., Weerden, W.M. van, Hoeben, R.C., Pluijm, G. van der, Merbel, A.F. van de, Horst, G. ter, Mark, M.H. van der, Bots, S.T.F., Wollenberg, D.J.M. van den, Ridder, C.M., Stuurman, D., Aalders, T., Erkens-Schulz, S., Montfoort, N. van, Karthaus, W.R., Mehra, N., Smits, M., Schalken, J.A., Weerden, W.M. van, Hoeben, R.C., and Pluijm, G. van der
- Abstract
Contains fulltext : 251831.pdf (Publisher’s version ) (Open Access), Treatment of castration-resistant prostate cancer remains a challenging clinical problem. Despite the promising effects of immunotherapy in other solid cancers, prostate cancer has remained largely unresponsive. Oncolytic viruses represent a promising therapeutic avenue, as oncolytic virus treatment combines tumour cell lysis with activation of the immune system and mounting of effective anti-tumour responses. Mammalian Orthoreoviruses are non-pathogenic human viruses with a preference of lytic replication in human tumour cells. In this study, we evaluated the oncolytic efficacy of the bioselected oncolytic reovirus mutant jin-3 in multiple human prostate cancer models. The jin-3 reovirus displayed efficient infection, replication, and anti-cancer responses in 2D and 3D prostate cancer models, as well as in ex vivo cultured human tumour slices. In addition, the jin-3 reovirus markedly reduced the viability and growth of human cancer cell lines and patient-derived xenografts. The infection induced the expression of mediators of immunogenic cell death, interferon-stimulated genes, and inflammatory cytokines. Taken together, our data demonstrate that the reovirus mutant jin-3 displays tumour tropism, and induces potent oncolytic and immunomodulatory responses in human prostate cancer models. Therefore, jin-3 reovirus represents an attractive candidate for further development as oncolytic agent for treatment of patients with aggressive localised or advanced prostate cancer.
- Published
- 2022
8. Cabazitaxel efficacy is strongly enhanced by continued Androgen Receptor Targeted Agents (ARTA) in Castration-Resistant Prostate Cancer (CRPC)
- Author
-
Mout, L., primary, Van Royen, M.E., additional, De Ridder, C.M.A., additional, Stuurman, D., additional, Marques, R.B., additional, De Geer, W.S., additional, Van De Werken, H.J.G., additional, Mathijssen, R.H.J., additional, De Wit, R., additional, Lolkema, M.P., additional, and Van Weerden, W.M., additional
- Published
- 2021
- Full Text
- View/download PDF
9. Patient-derived preclinical models of castration resistant prostate cancer: Xenografts and PDX-derived organoids to recapitulate patients’ disease profiles
- Author
-
Van Hemelryk, A., primary, Tomljanovic, I., additional, De Ridder, C.M.A., additional, Stuurman, D., additional, Erkens-Schulze, S., additional, Teubel, W.J., additional, Verhoef, E.I., additional, Van Leenders, G.J.L.H., additional, Cangiano, M., additional, Beumer, I.J., additional, Van De Werken, H.J.G., additional, Van Royen, M.E., additional, Grudniewska, M., additional, and Van Weerden, W.M., additional
- Published
- 2021
- Full Text
- View/download PDF
10. Androgen receptor signalling impairs docetaxel efficacy in castration-resistant prostate cancer
- Author
-
Mout, L. (Lisanne), Moll, J.M. (Jan), Chen, M. (Min), de Morrée, E.S., Ridder, C.M.A. (Corrina) de, Gibson, A., Stuurman, D., Aghai, A., Erkens-Schulze, S. (Sigrun), Mathijssen, A.H.J. (Ron), Sparreboom, A, Wit, R. (Ronald) de, Lolkema, M.P.J.K., Weerden, W.M. (Wytske) van, Mout, L. (Lisanne), Moll, J.M. (Jan), Chen, M. (Min), de Morrée, E.S., Ridder, C.M.A. (Corrina) de, Gibson, A., Stuurman, D., Aghai, A., Erkens-Schulze, S. (Sigrun), Mathijssen, A.H.J. (Ron), Sparreboom, A, Wit, R. (Ronald) de, Lolkema, M.P.J.K., and Weerden, W.M. (Wytske) van
- Published
- 2020
- Full Text
- View/download PDF
11. In Vivo Evaluation of Indium-111–Labeled 800CW as a Necrosis-Avid Contrast Agent
- Author
-
Stroet, M.C.M. (Marcus C. M.), Blois, E. (Erik) de, Stuurman, D. (Debra), Ridder, C.M.A. (Corrina) de, Haeck, J.C. (Joost), Seimbille, Y. (Yann), Mezzanotte, L. (Laura), Jong, M. (Marion) de, Löwik, C.W.G.M. (Clemens), Panth, K.M. (Kranthi M.), Stroet, M.C.M. (Marcus C. M.), Blois, E. (Erik) de, Stuurman, D. (Debra), Ridder, C.M.A. (Corrina) de, Haeck, J.C. (Joost), Seimbille, Y. (Yann), Mezzanotte, L. (Laura), Jong, M. (Marion) de, Löwik, C.W.G.M. (Clemens), and Panth, K.M. (Kranthi M.)
- Abstract
Purpose: Current clinical measurements for tumor treatment efficiency rely often on changes in tumor volume measured as shrinkage by CT or MRI, which become apparent after multiple lines of treatment and pose a physical and psychological burden on the patient. Detection of therapy-induced cell death in the tumor can be a fast measure for treatment efficiency. However, there are no reliable clinical tools for detection of tumor necrosis. Previously, we studied the necrosis avidity of cyanine-based fluorescent dyes, which suffered long circulation times before tumor necrosis could be imaged due to low hydrophilicity. We now present the application of radiolabeled 800CW, a commercially available cyanine with high hydrophilicity, to image tumor necrosis in a mouse model. Procedures: We conjugated 800CW to DOTA via a PEG linker, for labeling with single-photon emission-computed tomography isotope indium-111, yielding [111In]In-DOTA-PEG4-800CW. We then investigated specific [111In]In-DOTA-PEG4-800CW uptake by dead cells in vitro, using both fluorescence and radioactivity as detection modalities. Finally, we investigated [111In]In-DOTA-PEG4-800CW uptake into necrotic tumor regions of a 4T1 breast tumor model in mice. Results: We successfully prepared a precursor and developed a reliable procedure for labeling 800CW with indium-111. We detected specific [111In]In-DOTA-PEG4-800CW uptake by dead cells, using both fluorescence and radioactivity. Albeit with a tumor uptake of only 0.37%ID/g at 6 h post injection, we were able to image tumor necrosis with a tumor to background ratio of 7:4. Fluorescence and radioactivity in cryosections from the dissected tumors were colocalized with tumor necrosis, confirmed by TUNEL staining. Conclusions: [111In]In-DOTA-PEG4-800CW can be used to image tumor necrosis in vitro and in vivo. Further research will elucidate the application of [111In]In-DOTA-PEG4-800CW or other radiolabeled hydrophilic cyanines for the detection of necrosis caused b
- Published
- 2020
- Full Text
- View/download PDF
12. 119 (PB109) - Patient-derived xenografts and organoids recapitulate castration-resistant prostate cancer with sustained androgen receptor signaling
- Author
-
Van Hemelryk, A., Tomljanovic, I., Stuurman, D., de Ridder, C.M.A., Teubel, W.J., Erkens-Schulze, S., van de Werken, H.J.G., van Royen, M., Grudniewska, M., Jenster, G.W., and van Weerden, W.M.
- Published
- 2022
- Full Text
- View/download PDF
13. 118 (PB108) - Standardization of viability assays and high-content live-cell imaging protocols for large-scale drug testing in prostate cancer PDX-derived organoids
- Author
-
Van Hemelryk, A., Erkens-Schulze, S., Lam, L., Stuurman, D., de Ridder, C.M.A., French, P.J., van Royen, M.E., and van Weerden, W.M.
- Published
- 2022
- Full Text
- View/download PDF
14. Radiosensitivity of patient-derived xenograft tissue slices versus organoids in prostate cancer
- Author
-
Zhang, W., primary, Van Hemelryk, A., additional, De Ridder, C.M.A., additional, Erkens-Schulze, S., additional, Stuurman, D., additional, Van Gent, D.C., additional, Nonnekens, J., additional, and Van Weerden, W.M., additional
- Published
- 2019
- Full Text
- View/download PDF
15. A novel panel of patient-derived preclinical models of prostate cancer: xenografts and PDX-derived organoids to recapitulate patients disease profiles
- Author
-
Van Hemelryk, A., primary, De Ridder, C.M.A., additional, Erkens-Schulze, S., additional, Teubel, W.J., additional, Stuurman, D., additional, Mc Kinney, C., additional, Cangiano, M., additional, Beumer, I.J., additional, and Van Weerden, W.M., additional
- Published
- 2019
- Full Text
- View/download PDF
16. Docetaxel efficacy in prostate cancer is affected by testosterone; unraveling clinical observations
- Author
-
Mout, L., primary, Moll, J.M., additional, Chen, M., additional, De Ridder, C., additional, Gibson, A.A., additional, Stuurman, D., additional, Aghai, A., additional, Mathijssen, R.H.J, additional, Sparreboom, A., additional, De Wit, R., additional, Lolkema, M.P., additional, and Van Weerden, W.M., additional
- Published
- 2019
- Full Text
- View/download PDF
17. Testosterone Diminishes Cabazitaxel Efficacy and Intratumoral Accumulation in a Prostate Cancer Xenograft Model
- Author
-
Mout, L. (Lisanne), Wit, R. (Ronald) de, Stuurman, D. (Debra), Verhoef, E.I. (Esther), Mathijssen, A.H.J. (Ron), Ridder, C.M.A. (Corrina) de, Lolkema, M.P. (Martijn), Weerden, W.M. (Wytske) van, Mout, L. (Lisanne), Wit, R. (Ronald) de, Stuurman, D. (Debra), Verhoef, E.I. (Esther), Mathijssen, A.H.J. (Ron), Ridder, C.M.A. (Corrina) de, Lolkema, M.P. (Martijn), and Weerden, W.M. (Wytske) van
- Abstract
Inactivation of the androgen receptor (AR) pathway by androgen deprivation therapy (ADT) is the mainstay of (metastatic) prostate cancer therapy. Ultimately, the AR pathway will be re-activated despite castrate levels of circulating androgens. Thereby, maintaining its role even in castration resistant prostate cancer (CRPC). The recent STAMPEDE and CHAARTED trials showed that docetaxel in combination with ADT increased survival in hormone sensitive prostate cancer patients, suggesting cross-talk between AR signaling and chemotherapy efficacy. We hypothesized that a similar interaction may also apply for CRPC that is treated with cabazitaxel. We studied the impact of androgen status on the efficacy, pharmacodynamics and -kinetics of cabazitaxel in a unique and clinically relevant patient derived xenograft model of castration resistant disease. We found that cabazitaxel is highly effective in a castrate setting with strongly reduced AR activation, while tumor growth inhibition by cabazitaxel was completely abolished in the presence of high AR pathway activity. Moreover, additional experiments showed that intratumoral cabazitaxel levels were 3.5 times higher in tumors from castrated mice as compared to tumors from androgen-supplemented animals. We confirmed that cabazitaxel pharmacokinetics were not affected by testosterone, suggesting that androgen status might influence cabazitaxel tumor uptake directly. This study reveals the impact of androgen status on cabazitaxel efficacy and supports the potential of combination of taxane chemotherapeutics with AR axis targeting agents.
- Published
- 2017
- Full Text
- View/download PDF
18. P64 - Radiosensitivity of patient-derived xenograft tissue slices versus organoids in prostate cancer
- Author
-
Zhang, W., Van Hemelryk, A., De Ridder, C.M.A., Erkens-Schulze, S., Stuurman, D., Van Gent, D.C., Nonnekens, J., and Van Weerden, W.M.
- Published
- 2019
- Full Text
- View/download PDF
19. P58 - Docetaxel efficacy in prostate cancer is affected by testosterone; unraveling clinical observations
- Author
-
Mout, L., Moll, J.M., Chen, M., De Ridder, C., Gibson, A.A., Stuurman, D., Aghai, A., Mathijssen, R.H.J, Sparreboom, A., De Wit, R., Lolkema, M.P., and Van Weerden, W.M.
- Published
- 2019
- Full Text
- View/download PDF
20. P50 - A novel panel of patient-derived preclinical models of prostate cancer: xenografts and PDX-derived organoids to recapitulate patients disease profiles
- Author
-
Van Hemelryk, A., De Ridder, C.M.A., Erkens-Schulze, S., Teubel, W.J., Stuurman, D., Mc Kinney, C., Cangiano, M., Beumer, I.J., and Van Weerden, W.M.
- Published
- 2019
- Full Text
- View/download PDF
21. 48 Lack of Clinical Efficacy of the Combination of Lenalidomide Plus Docetaxel is Predicted by the Lack of Effect in the Orthotopic PC346C Xenograft Model
- Author
-
van Weerden, W.M., primary, Meulenbeld, H., additional, Boer, A., additional, Stuurman, D., additional, Aghai, A., additional, de Ridder, C.M.A., additional, and de Wit, R., additional
- Published
- 2012
- Full Text
- View/download PDF
22. eSOMA-DM1, a Maytansinoid-Based Theranostic Small-Molecule Drug Conjugate for Neuroendocrine Tumors.
- Author
-
Chapeau D, Beekman S, Piet A, Li L, de Ridder C, Stuurman D, and Seimbille Y
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Receptors, Somatostatin metabolism, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Antineoplastic Agents pharmacology, Tissue Distribution, Mice, Nude, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors diagnostic imaging, Maytansine chemistry, Maytansine therapeutic use, Maytansine pharmacology, Maytansine pharmacokinetics
- Abstract
Background: The main challenges of conventional chemotherapy lie in its lack of selectivity and specificity, leading to significant side effects. Using a small-molecule drug conjugate (SMDC) ensures specific delivery of a cytotoxic drug to the tumor site by coupling it to a targeting vector. This promising strategy can be applied to neuroendocrine tumors (NETs) by choosing a targeting vector that binds specifically to somatostatin receptor subtype 2 (SSTR2). Additionally, incorporation of a bifunctional chelate into the molecule enables complexation of both diagnostic and therapeutic radionuclides. Thus, it facilitates monitoring of the distribution of the SMDC in the body and allows for the implementation of combination therapy. In our study, we designed eSOMA-DM1, a SMDC combining the SSTR2-targeted octreotate peptide and the cytotoxic agent DM1 via a chelate-bridged linker (N
3 -Py-DOTAGA). This approach warrants conjugation of the targeting vector and the drug at opposite sites to avoid undesired steric hindrance effects. Methods: Synthesis of the DM1 moiety ( 4 ) involved a three-step synthetic route, followed by the conjugation to the cyclic peptide, N3 -Py-DOTAGA-d-Phe-cyclo[Cys-Tyr-d-Trp-Lys-Thr-Cys]-Thr-OH, through a copper-free click reaction, resulting in eSOMA-DM1. Subsequent labeling with [111 In]InCl3 gave a high radiochemical yield and purity. In vitro assessments of eSOMA-DM1 binding, uptake, and internalization were conducted in SSTR2-transfected U2OS cells. Ex vivo biodistribution and fluorescence imaging were performed in H69-tumor bearing mice. Results: eSOMA-DM1 exhibited an IC50 value for SSTR2 similar to the gold standard DOTA-TATE. The uptake of [111 In]In-eSOMA-DM1 in U2OS.SSTR2 cells was 1.2-fold lower than that of [111 In]In-DOTA-TATE. Tumor uptake in H69-xenografted mice was higher for [111 In]In-eSOMA-DM1 at all-time points compared to [111 In]In-DOTA-TATE. Prolonged blood circulation led to increased accumulation of [111 In]In-eSOMA-DM1 in highly vascularized tissues, such as the lungs, skin, and heart. Excretion through the kidneys, liver, and spleen was also observed. Conclusion: eSOMA-DM1 is a SMDC developed for NET showing promising characteristics in vitro. However, the in vivo results obtained with [111 In]In-eSOMA-DM1 suggest the need for adjustments to optimize its distribution.- Published
- 2024
- Full Text
- View/download PDF
23. Darolutamide does not interfere with OATP-mediated uptake of docetaxel.
- Author
-
Buck SAJ, Talebi Z, Drabison T, Jin Y, Gibson AA, Hu P, de Bruijn P, de Ridder CMA, Stuurman D, Hu S, van Weerden WM, Koolen SLW, de Wit R, Sparreboom A, Mathijssen RHJ, and Eisenmann ED
- Subjects
- Humans, Male, Animals, Mice, Drug Interactions, Cell Line, Tumor, HEK293 Cells, Docetaxel pharmacology, Docetaxel pharmacokinetics, Solute Carrier Organic Anion Transporter Family Member 1B3 metabolism, Prostatic Neoplasms drug therapy, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Xenograft Model Antitumor Assays, Pyrazoles pharmacology, Pyrazoles pharmacokinetics
- Abstract
The addition of darolutamide, an androgen receptor signalling inhibitor, to therapy with docetaxel has recently been approved as a strategy to treat metastatic prostate cancer. OATP1B3 is an SLC transporter that is highly expressed in prostate cancer and is responsible for the accumulation of substrates, including docetaxel, into tumours. Given that darolutamide inhibits OATP1B3 in vitro, we sought to characterise the impact of darolutamide on docetaxel pharmacokinetics. We investigated the influence of darolutamide on OATP1B3 transport using in vitro and in vivo models. We assessed the impact of darolutamide on the tumour accumulation of docetaxel in a patient-derived xenograft (PDX) model and on an OATP1B biomarker in patients. Darolutamide inhibited OATP1B3 in vitro at concentrations higher than the reported C
max . Consistent with these findings, in vivo studies revealed that darolutamide does not influence the pharmacokinetics of Oatp1b substrates, including docetaxel. Docetaxel accumulation in PDX tumours was not decreased in the presence of darolutamide. Metastatic prostate cancer patients had similar levels of OATP1B biomarkers, regardless of treatment with darolutamide. Consistent with a low potential to inhibit OATP1B3-mediated transport in vitro, darolutamide does not significantly impede the transport of Oatp1b substrates in vivo or in patients. Our findings support combined treatment with docetaxel and darolutamide, as no OATP1B3 transporter based drug-drug interaction was identified., (© 2024 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)- Published
- 2024
- Full Text
- View/download PDF
24. eTFC-01: a dual-labeled chelate-bridged tracer for SSTR2-positive tumors.
- Author
-
Chapeau D, Beekman S, Handula M, Murce E, de Ridder C, Stuurman D, and Seimbille Y
- Abstract
Background: Integrating radioactive and optical imaging techniques can facilitate the prognosis and surgical guidance for cancer patients. Using a single dual-labeled tracer ensures consistency in both imaging modalities. However, developing such molecule is challenging due to the need to preserve the biochemical properties of the tracer while introducing bulky labeling moieties. In our study, we designed a trifunctional chelate that facilitates the coupling of the targeting vector and fluorescent dye at opposite sites to avoid undesired steric hindrance effects. The synthesis of the trifunctional chelate N
3 -Py-DOTAGA-(tBu)3 (7) involved a five-step synthetic route, followed by conjugation to the linear peptidyl-resin 8 through solid-phase synthesis. After deprotection and cyclization, the near-infrared fluorescent dye sulfo-Cy.5 was introduced using copper free click chemistry, resulting in eTFC-01. Subsequently, eTFC-01 was labeled with [111 In]InCl3 . In vitro assessments of eTFC-01 binding, uptake, and internalization were conducted in SSTR2-transfected U2OS cells. Ex-vivo biodistribution and fluorescence imaging were performed in H69-tumor bearing mice., Results: eTFC-01 demonstrated a two-fold higher IC50 value for SSTR2 compared to the gold standard DOTA-TATE. Labeling of eTFC-01 with [111 In]InCl3 gave a high radiochemical yield and purity. The uptake of [111 In]In-eTFC-01 in U2OS.SSTR2 cells was two-fold lower than the uptake of [111 In]In-DOTA-TATE, consistent with the binding affinity. Tumor uptake in H69-xenografted mice was lower for [111 In]In-eTFC-01 at all-time points compared to [111 In]In-DOTA-TATE. Prolonged blood circulation led to increased accumulation of [111 In]In-eTFC-01 in highly vascularized tissues, such as lungs, skin, and heart. Fluorescence measurements in different organs correlated with the radioactive signal distribution., Conclusion: The successful synthesis and coupling of the trifunctional chelate to the peptide and fluorescent dye support the potential of this synthetic approach to generate dual labeled tracers. While promising in vitro, the in vivo results obtained with [111 In]In-eTFC-01 suggest the need for adjustments to enhance tracer distribution., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
25. Darolutamide Added to Docetaxel Augments Antitumor Effect in Models of Prostate Cancer through Cell Cycle Arrest at the G1-S Transition.
- Author
-
Buck SAJ, Van Hemelryk A, de Ridder C, Stuurman D, Erkens-Schulze S, van 't Geloof S, Teubel WJ, Koolen SLW, Martens-Uzunova ES, van Royen ME, de Wit R, Mathijssen RHJ, and van Weerden WM
- Subjects
- Male, Humans, Animals, Mice, Cell Proliferation drug effects, Cell Line, Tumor, Antineoplastic Combined Chemotherapy Protocols pharmacology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Pyrazoles pharmacology, Cell Cycle Checkpoints drug effects, Receptors, Androgen metabolism, Drug Synergism, Docetaxel pharmacology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology, Xenograft Model Antitumor Assays
- Abstract
Resistance to taxane chemotherapy is frequently observed in metastatic prostate cancer. The androgen receptor (AR) is a major driver of prostate cancer and a key regulator of the G1-S cell-cycle checkpoint, promoting cancer cell proliferation by irreversible passage to the S-phase. We hypothesized that AR signaling inhibitor (ARSi) darolutamide in combination with docetaxel could augment antitumor effect by impeding the proliferation of taxane-resistant cancer cells. We monitored cell viability in organoids, tumor volume, and PSA secretion in patient-derived xenografts (PDX) and analyzed cell cycle and signaling pathway alterations. Combination treatment increased antitumor effect in androgen-sensitive, AR-positive prostate cancer organoids and PDXs. Equally beneficial effects of darolutamide added to docetaxel were observed in a castration-resistant model, progressive on docetaxel, enzalutamide, and cabazitaxel. In vitro studies showed that docetaxel treatment with simultaneous darolutamide resulted in a reduction of cells entering the S-phase in contrast to only docetaxel. Molecular analysis in the prostate cancer cell line LNCaP revealed an upregulation of cyclin-dependent kinase inhibitor p21, supporting blockade of S-phase entry and cell proliferation. Our results provide a preclinical support for combining taxanes and darolutamide as a multimodal treatment strategy in patients with metastatic prostate cancer progressive on ARSi and taxane chemotherapy., (©2023 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
26. Synthesis and Evaluation of ePSMA-DM1: A New Theranostic Small-Molecule Drug Conjugate (T-SMDC) for Prostate Cancer.
- Author
-
Murce E, Spaan E, Beekman S, van den Brink L, Handula M, Stuurman D, de Ridder C, Dalm SU, and Seimbille Y
- Abstract
Small-molecule drug conjugates (SMDCs) are compounds in which a therapeutic payload is conjugated to a targeting vector, for specific delivery to the tumor site. This promising approach can be translated to the treatment of prostate cancer by selecting a targeting vector which binds to the prostate-specific membrane antigen (PSMA). Moreover, the addition of a bifunctional chelator to the molecule allows for the use of both diagnostic and therapeutic radionuclides. In this way, the distribution of the SMDC in the body can be monitored, and combination therapy regimes can be implemented. We combined a glutamate-urea-lysine vector to the cytotoxic agent DM1 and a DOTA chelator via an optimized linker to obtain the theranostic SMDC (T-SMDC) ePSMA-DM1. ePSMA-DM1 retained a high binding affinity to PSMA and demonstrated PSMA-specific uptake in cells. Glutathione stability assays showed that the half-life of the T-SMDC in a reducing environment was 2 h, and full drug release was obtained after 6 h. Moreover, 100 nM of ePSMA-DM1 reduced the cell viability of the human PSMA-positive LS174T cells by >85% after 72 h of incubation, which was comparable to a 10-fold higher dose of free DM1. [
111 In]In-ePSMA-DM1 and [177 Lu]Lu-ePSMA-DM1 were both obtained in high radiochemical yields and purities (>95%), with >90% stability in PBS and >80% stability in mouse serum for up to 24 h post incubation at 37 °C. SPECT/CT imaging studies allowed for a faint tumor visualization of [111 In]In-ePSMA-DM1 at 1 h p.i., and the ex vivo biodistribution showed tumor uptake (2.39 ± 0.29% ID/g) at 1 h p.i., with the compound retained in the tumor for up to 24 h. Therefore, ePSMA-DM1 is a promising T-SMDC candidate for prostate cancer, and the data obtained so far warrant further investigations, such as therapeutic experiments, after further optimization.- Published
- 2023
- Full Text
- View/download PDF
27. [ 212 Pb]Pb-eSOMA-01: A Promising Radioligand for Targeted Alpha Therapy of Neuroendocrine Tumors.
- Author
-
Chapeau D, Koustoulidou S, Handula M, Beekman S, de Ridder C, Stuurman D, de Blois E, Buchatskaya Y, van der Schilden K, de Jong M, Konijnenberg MW, and Seimbille Y
- Abstract
Peptide receptor radionuclide therapy (PRRT) has been applied to the treatment of neuroendocrine tumors (NETs) for over two decades. However, improvement is still needed, and targeted alpha therapy (TAT) with alpha emitters such as lead-212 (
212 Pb) represents a promising avenue. A series of ligands based on octreotate was developed. Lead-203 was used as an imaging surrogate for the selection of the best candidate for the studies with lead-212.203/212 Pb radiolabeling and in vitro assays were carried out, followed by SPECT/CT imaging and ex vivo biodistribution in NCI-H69 tumor-bearing mice. High radiochemical yields (≥99%) and purity (≥96%) were obtained for all ligands. [203 Pb]Pb-eSOMA-01 and [203 Pb]Pb-eSOMA-02 showed high stability in PBS and mouse serum up to 24 h, whereas [203 Pb]Pb-eSOMA-03 was unstable in those conditions. All compounds exhibited a nanomolar affinity (2.5-3.1 nM) for SSTR2. SPECT/CT images revealed high tumor uptake at 1, 4, and 24 h post-injection of [203 Pb]Pb-eSOMA-01/02. Ex vivo biodistribution studies confirmed that the highest uptake in tumors was observed with [212 Pb]Pb-eSOMA-01. [212 Pb]Pb-eESOMA-01 displayed the highest absorbed dose in the tumor (35.49 Gy/MBq) and the lowest absorbed dose in the kidneys (121.73 Gy/MBq) among the three tested radioligands. [212 Pb]Pb-eSOMA-01 is a promising candidate for targeted alpha therapy of NETs. Further investigations are required to confirm its potential.- Published
- 2023
- Full Text
- View/download PDF
28. First preclinical evaluation of [ 225 Ac]Ac-DOTA-JR11 and comparison with [ 177 Lu]Lu-DOTA-JR11, alpha versus beta radionuclide therapy of NETs.
- Author
-
Handula M, Beekman S, Konijnenberg M, Stuurman D, de Ridder C, Bruchertseifer F, Morgenstern A, Denkova A, de Blois E, and Seimbille Y
- Abstract
Background: The [
177 Lu]Lu-DOTA-TATE mediated peptide receptor radionuclide therapy (PRRT) of neuroendocrine tumors (NETs) is sometimes leading to treatment resistance and disease recurrence. An interesting alternative could be the somatostatin antagonist, [177 Lu]Lu-DOTA-JR11, that demonstrated better biodistribution profile and higher tumor uptake than [177 Lu]Lu-DOTA-TATE. Furthermore, treatment with alpha emitters showed improvement of the therapeutic index of PRRT due to the high LET offered by the alpha particles compared to beta emitters. Therefore, [225 Ac]Ac-DOTA-JR11 can be a potential candidate to improve the treatment of NETs (Graphical abstract). DOTA-JR11 was radiolabeled with [225 Ac]Ac(NO3 )3 and [177 Lu]LuCl3 . Stability studies were performed in phosphate buffered saline (PBS) and mouse serum. In vitro competitive binding assay has been carried out in U2OS-SSTR2 + cells fornat La-DOTA-JR11,nat Lu-DOTA-JR11 and DOTA-JR11. Ex vivo biodistribution studies were performed in mice inoculated with H69 cells at 4, 24, 48 and 72 h after injection of [225 Ac]Ac-DOTA-JR11. A blocking group was included to verify uptake specificity. Dosimetry of selected organs was determined for [225 Ac]Ac-DOTA-JR11 and [177 Lu]Lu-DOTA-JR11., Results: [225 Ac]Ac-DOTA-JR11 has been successfully prepared and obtained in high radiochemical yield (RCY; 95%) and radiochemical purity (RCP; 94%). [225 Ac]Ac-DOTA-JR11 showed reasonably good stability in PBS (77% intact radiopeptide at 24 h after incubation) and in mouse serum (~ 81% intact radiopeptide 24 h after incubation). [177 Lu]Lu-DOTA-JR11 demonstrated excellent stability in both media (> 93%) up to 24 h post incubation. Competitive binding assay revealed that complexation of DOTA-JR11 withnat La andnat Lu did not affect its binding affinity to SSTR2. Similar biodistribution profiles were observed for both radiopeptides, however, higher uptake was noticed in the kidneys, liver and bone for [225 Ac]Ac-DOTA-JR11 than [177 Lu]Lu-DOTA-JR11., Conclusion: [225 Ac]Ac-DOTA-JR11 showed a higher absorbed dose in the kidneys compared to [177 Lu]Lu-DOTA-JR11, which may limit further studies with this radiopeptide. However, several strategies can be explored to reduce nephrotoxicity and offer opportunities for future clinical investigations with [225 Ac]Ac-DOTA-JR11., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
29. Radiochemical and Biological Evaluation of 3p- C -NETA-ePSMA-16, a Promising PSMA-Targeting Agent for Radiotheranostics.
- Author
-
Murce E, Ahenkorah S, Beekman S, Handula M, Stuurman D, de Ridder C, Cleeren F, and Seimbille Y
- Abstract
Bifunctional chelators (BFCs) are a key element in the design of radiopharmaceuticals. By selecting a BFC that efficiently complexes diagnostic and therapeutic radionuclides, a theranostic pair possessing almost similar biodistribution and pharmacokinetic properties can be developed. We have previously reported 3p- C -NETA as a promising theranostic BFC, and the encouraging preclinical outcomes obtained with [
18 F]AlF-3p- C -NETA-TATE led us to conjugate this chelator to a PSMA-targeting vector for imaging and treatment of prostate cancer. In this study, we synthesized 3p- C -NETA-ePSMA-16 and radiolabeled it with different diagnostic (111 In,18 F) and therapeutic (177 Lu,213 Bi) radionuclides. 3p- C -NETA-ePSMA-16 showed high affinity to PSMA (IC50 = 4.61 ± 1.33 nM), and [111 In]In-3p- C -NETA-ePSMA-16 showed specific cell uptake (1.41 ± 0.20% ID/106 cells) in PSMA expressing LS174T cells. Specific tumor uptake of [111 In]In-3p- C -NETA-ePSMA-16 was observed up to 4 h p.i. (1.62 ± 0.55% ID/g at 1 h p.i.; 0.89 ± 0.58% ID/g at 4 h p.i.) in LS174T tumor-bearing mice. Only a faint signal could be seen at 1 h p.i. in the SPECT/CT scans, whereas dynamic PET/CT scans performed after administration of [18 F]AlF-3p- C -NETA-ePSMA-16 in PC3-Pip tumor xenografted mice resulted in a better tumor visualization and imaging contrast. Therapy studies with short-lived radionuclides such as213 Bi could further elucidate the therapeutic potential of 3p- C -NETA-ePSMA-16 as a radiotheranostic.- Published
- 2023
- Full Text
- View/download PDF
30. DNA-PKcs inhibitors sensitize neuroendocrine tumor cells to peptide receptor radionuclide therapy in vitro and in vivo .
- Author
-
Reuvers TGA, Verkaik NS, Stuurman D, de Ridder C, Groningen MCC, de Blois E, and Nonnekens J
- Subjects
- Humans, Mice, Animals, DNA-Activated Protein Kinase metabolism, Creatinine, Quality of Life, Radioisotopes metabolism, DNA, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors radiotherapy
- Abstract
Background: Peptide receptor radionuclide therapy (PRRT) increases progression-free survival and quality of life of neuroendocrine tumor (NET) patients, however complete cures are rare and dose-limiting toxicity has been reported. PRRT induces DNA damage of which DNA double strand breaks (DSBs) are the most cytotoxic. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a key player in DSB repair and its inhibition therefore is a potential way to enhance PRRT efficacy without increasing the dosage. Methods: We analyzed effects of combining PRRT and DNA-PKcs inhibitor AZD7648 on viability, cell death and clonogenic survival on SSTR2-expressing cell lines BON1-SSTR2, GOT1 and NCI-H69. Therapy-induced DNA damage response was assessed by analyzing DSB foci levels and cell cycle distributions. In vivo efficacy was investigated in BON1-SSTR2 and NCI-H69 xenografted mice and hematologic and renal toxicity were monitored by blood counts, creatinine levels and analyzing renal morphology. Results: Combining PRRT and AZD7648 significantly decreased viability of BON1-SSTR2, GOT1 and NCI-H69 cells and induced cell death in GOT1 and BON1-SSTR2 cells. A strong effect of AZD7648 on PRRT-induced DSB repair was found. In GOT1 cells, this was accompanied by induction of cell cycle blocks. However, BON1-SSTR2 cells were unable to fully arrest their cell cycle and polyploid cells with high DNA damage levels were detected. In vivo , AZD7648 significantly sensitized BON1-SSTR2 and NCI-H69 xenograft models to PRRT. In addition, combination therapy did not induce significant changes in body weight, blood composition, plasma creatinine levels and renal morphology, indicating the absence of severe acute hematologic and renal toxicity. Conclusion: These results highlight that the potentiation of the therapeutic effect of PRRT by DNA-PKcs inhibition is a highly effective and well-tolerated therapeutic strategy. Based on our findings, we recommend initiation of phase I/II studies in patients to find a safe and effective combination regimen., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)
- Published
- 2023
- Full Text
- View/download PDF
31. Preclinical Evaluation of a PSMA-Targeting Homodimer with an Optimized Linker for Imaging of Prostate Cancer.
- Author
-
Murce E, Beekman S, Spaan E, Handula M, Stuurman D, de Ridder C, and Seimbille Y
- Subjects
- Male, Humans, Animals, Mice, Tissue Distribution, Single Photon Emission Computed Tomography Computed Tomography, Radiopharmaceuticals chemistry, Cell Line, Tumor, Antigens, Surface metabolism, Prostatic Neoplasms diagnostic imaging, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology
- Abstract
Prostate-specific membrane antigen (PSMA) targeting radiopharmaceuticals have been successfully used for diagnosis and therapy of prostate cancer. Optimization of the available agents is desirable to improve tumor uptake and reduce side effects to non-target organs. This can be achieved, for instance, via linker modifications or multimerization approaches. In this study, we evaluated a small library of PSMA-targeting derivatives with modified linker residues, and selected the best candidate based on its binding affinity to PSMA. The lead compound was coupled to a chelator for radiolabeling, and subject to dimerization. The resulting molecules, 22 and 30 , were highly PSMA specific (IC
50 = 1.0-1.6 nM) and stable when radiolabeled with indium-111 (>90% stable in PBS and mouse serum up to 24 h). Moreover, [111 In]In- 30 presented a high uptake in PSMA expressing LS174T cells, with 92.6% internalization compared to 34.1% for PSMA-617. Biodistribution studies in LS174T mice xenograft models showed that [111 In]In- 30 had a higher tumor and kidney uptake compared to [111 In]In-PSMA-617, but increasing T/K and T/M ratios at 24 h p.i. Tumors could be clearly visualized at 1 h p.i. by SPECT/CT after administration of [111 In]In- 22 and [111 In]In-PSMA-617, while [111 In]In- 30 showed a clear signal at later time-points (e.g., 24 h p.i.).- Published
- 2023
- Full Text
- View/download PDF
32. Synthesis and Evaluation of Two Long-Acting SSTR2 Antagonists for Radionuclide Therapy of Neuroendocrine Tumors.
- Author
-
Koustoulidou S, Handula M, de Ridder C, Stuurman D, Beekman S, de Jong M, Nonnekens J, and Seimbille Y
- Abstract
Somatostatin receptor subtype 2 (SSTR2) has become an essential target for radionuclide therapy of neuroendocrine tumors (NETs). JR11 was introduced as a promising antagonist peptide to target SSTR2. However, due to its rapid blood clearance, a better pharmacokinetic profile is necessary for more effective treatment. Therefore, two JR11 analogs (8a and 8b), each carrying an albumin binding domain, were designed to prolong the blood residence time of JR11. Both compounds were labeled with lutetium-177 and evaluated via in vitro assays, followed by in vivo SPECT/CT imaging and ex vivo biodistribution studies. [177Lu]Lu-8a and [177Lu]Lu-8b were obtained with high radiochemical purity (>97%) and demonstrated excellent stability in PBS and mouse serum (>95%). [177Lu]Lu-8a showed better affinity towards human albumin compared to [177Lu]Lu-8b. Further, 8a and 8b exhibited binding affinities 30- and 48-fold lower, respectively, than that of the parent peptide JR11, along with high cell uptake and low internalization rate. SPECT/CT imaging verified high tumor accumulation for [177Lu]Lu-8a and [177Lu]Lu-JR11 at 4, 24, 48, and 72 h post-injection, but no tumor uptake was observed for [177Lu]Lu-8b. Ex vivo biodistribution studies revealed high and increasing tumor uptake for [177Lu]Lu-8a. However, its extended blood circulation led to an unfavorable biodistribution profile for radionuclide therapy.
- Published
- 2022
- Full Text
- View/download PDF
33. Preclinical Assessment of the Combination of PSMA-Targeting Radionuclide Therapy with PARP Inhibitors for Prostate Cancer Treatment.
- Author
-
Ruigrok EAM, Verkaik NS, de Blois E, de Ridder C, Stuurman D, Roobol SJ, Van Gent DC, de Jong M, Van Weerden WM, and Nonnekens J
- Subjects
- Animals, Cell Line, Tumor, Humans, Male, Mice, Prostate pathology, Radioisotopes therapeutic use, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology, Prostatic Neoplasms radiotherapy
- Abstract
Prostate specific membrane antigen targeted radionuclide therapy (PSMA-TRT) is a promising novel treatment for prostate cancer (PCa) patients. However, PSMA-TRT cannot be used for curative intent yet, thus additional research on how to improve the therapeutic efficacy is warranted. A potential way of achieving this, is combining TRT with poly ADP-ribosylation inhibitors (PARPi), which has shown promising results for TRT of neuroendocrine tumor cells. Currently, several clinical trials have been initiated for this combination for PCa, however so far, no evidence of synergism is available for PCa. Therefore, we evaluated the combination of PSMA-TRT with three classes of PARPi in preclinical PCa models. In vitro viability and survival assays were performed using PSMA-expressing PCa cell lines PC3-PIP and LNCaP to assess the effect of increasing concentrations of PARPi veliparib, olaparib or talazoparib in combination with PSMA-TRT compared to single PARPi treatment. Next, DNA damage analyses were performed by quantifying the number of DNA breaks by immunofluorescent stainings. Lastly, the potential of the combination treatments was studied in vivo in mice bearing PC3-PIP xenografts. Our results show that combining PSMA-TRT with PARPi did not synergistically affect the in vitro clonogenic survival or cell viability. DNA-damage analysis revealed only a significant increase in DNA breaks when combining PSMA-TRT with veliparib and not in the other combination treatments. Moreover, PSMA-TRT with PARPi treatment did not improve tumor control compared to PSMA-TRT monotherapy. Overall, the data presented do not support the assumption that combining PSMA-TRT with PARPi leads to a synergistic antitumor effect in PCa. These results underline that extensive preclinical research using various PCa models is imperative to validate the applicability of the combination strategy for PCa, as it is for other cancer types.
- Published
- 2022
- Full Text
- View/download PDF
34. Reovirus mutant jin-3 exhibits lytic and immune-stimulatory effects in preclinical human prostate cancer models.
- Author
-
van de Merbel AF, van der Horst G, van der Mark MH, Bots STF, van den Wollenberg DJM, de Ridder CMA, Stuurman D, Aalders T, Erkens-Schulz S, van Montfoort N, Karthaus WR, Mehra N, Smits M, Schalken JA, van Weerden WM, Hoeben RC, and van der Pluijm G
- Subjects
- Animals, Cell Line, Tumor, Humans, Male, Mammals, Mammalian orthoreovirus 3, Oncolytic Virotherapy, Oncolytic Viruses genetics, Prostatic Neoplasms genetics, Prostatic Neoplasms therapy, Reoviridae genetics
- Abstract
Treatment of castration-resistant prostate cancer remains a challenging clinical problem. Despite the promising effects of immunotherapy in other solid cancers, prostate cancer has remained largely unresponsive. Oncolytic viruses represent a promising therapeutic avenue, as oncolytic virus treatment combines tumour cell lysis with activation of the immune system and mounting of effective anti-tumour responses. Mammalian Orthoreoviruses are non-pathogenic human viruses with a preference of lytic replication in human tumour cells. In this study, we evaluated the oncolytic efficacy of the bioselected oncolytic reovirus mutant jin-3 in multiple human prostate cancer models. The jin-3 reovirus displayed efficient infection, replication, and anti-cancer responses in 2D and 3D prostate cancer models, as well as in ex vivo cultured human tumour slices. In addition, the jin-3 reovirus markedly reduced the viability and growth of human cancer cell lines and patient-derived xenografts. The infection induced the expression of mediators of immunogenic cell death, interferon-stimulated genes, and inflammatory cytokines. Taken together, our data demonstrate that the reovirus mutant jin-3 displays tumour tropism, and induces potent oncolytic and immunomodulatory responses in human prostate cancer models. Therefore, jin-3 reovirus represents an attractive candidate for further development as oncolytic agent for treatment of patients with aggressive localised or advanced prostate cancer., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
35. Continued androgen signalling inhibition improves cabazitaxel efficacy in prostate cancer.
- Author
-
Mout L, van Royen ME, de Ridder C, Stuurman D, van de Geer WS, Marques R, Buck SAJ, French PJ, van de Werken HJG, Mathijssen RHJ, de Wit R, Lolkema MP, and van Weerden WM
- Subjects
- Androgen Receptor Antagonists pharmacology, Animals, Apoptosis drug effects, Benzamides pharmacology, Cell Line, Tumor, Disease Models, Animal, Drug Resistance, Neoplasm drug effects, Fluorescent Antibody Technique, Humans, Male, Mice, Microtubules metabolism, Nitriles pharmacology, Phenylthiohydantoin pharmacology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms etiology, Prostatic Neoplasms pathology, Receptors, Androgen metabolism, Taxoids therapeutic use, Xenograft Model Antitumor Assays, Androgens metabolism, Prostatic Neoplasms metabolism, Signal Transduction drug effects, Taxoids pharmacology
- Abstract
Background: The androgen receptor (AR) pathway is a key driver of neoplastic behaviour in the different stages of metastatic prostate cancer (mPCa). Targeting the AR therefore remains the cornerstone for mPCa treatment. We have previously reported that activation of AR signalling affects taxane chemo-sensitivity in preclinical models of castration resistant PCa (CRPC). Here, we explored the anti-tumour efficacy of the AR targeted inhibitor enzalutamide combined with cabazitaxel., Methods: We used the AR positive CRPC model PC346C-DCC-K to assess the in vitro and in vivo activity of combining enzalutamide with cabazitaxel. Subsequent validation studies were performed using an enzalutamide resistant VCaP model. To investigate the impact of AR signalling on cabazitaxel activity we used quantitative live-cell imaging of tubulin stabilization and apoptosis related nuclear fragmentation., Findings: Enzalutamide strongly amplified cabazitaxel anti-tumour activity in the patient-derived xenograft models PC346C-DCC-K (median time to humane endpoint 77 versus 48 days, P<0.0001) and VCaP-Enza-B (median time to humane endpoint 80 versus 53 days, P<0.001). Although enzalutamide treatment by itself was ineffective in reducing tumour growth, it significantly suppressed AR signalling in PC346C-DCC-K tumours as shown by AR target gene expression. The addition of enzalutamide enhanced cabazitaxel induced apoptosis as shown by live-cell imaging (P<0.001)., Interpretation: Our study demonstrates that cabazitaxel efficacy can be improved by simultaneous blocking of AR signalling by enzalutamide, even if AR targeted treatment no longer affects tumour growth. These findings support clinical studies that combine AR targeted inhibitors with cabazitaxel in CRPC., Competing Interests: Declaration of Competing Interest RDW—Advisory role/speaker fees; Sanofi, Merck, Lilly, Roche, Bayer, Janssen, Clovis and research funding (Institutional); Sanofi, Bayer. MPL—Advisory role/speaker fees; Incyte, Amgen, Janssen Cilag B.V., Bayer, Servier, Roche, Pfizer Sanofi Aventis Netherlands BV, Astellas and has received research funding (Institutional) from Sanofi, JnJ, Merck and Astellas. RHJM—has received research funding (Institutional) from Sanofi and Astellas. WvW—has received research funding (Institutional) from Sanofi, Millennium, Janssen Pharmaceuticals, and Servier. LM—Advisory role/speaker fees from Sanofi. HvdW — Advisory role/speaker fees from Bayer., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
36. Extensive preclinical evaluation of lutetium-177-labeled PSMA-specific tracers for prostate cancer radionuclide therapy.
- Author
-
Ruigrok EAM, van Vliet N, Dalm SU, de Blois E, van Gent DC, Haeck J, de Ridder C, Stuurman D, Konijnenberg MW, van Weerden WM, de Jong M, and Nonnekens J
- Subjects
- Animals, Antigens, Surface metabolism, Cell Line, Tumor, Humans, Lutetium therapeutic use, Male, Mice, Radioisotopes, Tissue Distribution, Glutamate Carboxypeptidase II metabolism, Prostatic Neoplasms diagnostic imaging, Prostatic Neoplasms radiotherapy
- Abstract
Purpose: Various radiolabeled prostate-specific membrane antigen (PSMA)-targeting tracers are clinically applied for prostate cancer (PCa) imaging and targeted radionuclide therapy. The PSMA binding affinities, biodistribution, and DNA-damaging capacities of these radiotracers have not yet been compared in detail. A major concern of PSMA-targeting radiotracers is the toxicity in other PSMA-expressing organs, such as the salivary glands, thus demanding careful evaluation of the most optimal and safest radiotracer. In this extensive preclinical study, we evaluated the clinically applied PSMA-targeting small molecule inhibitors DOTA-PSMA-617 (PSMA-617) and DOTAGA-PSMA-I&T (PSMA-I&T) and the PSMA nanobody DOTA-JVZ-007 (JVZ-007) using PSMA-expressing cell lines, a unique set of PCa patient-derived xenografts (PDX) and healthy human tissues., Methods and Results: In vitro displacement studies on PSMA-expressing cells and cryosections of a PSMA-positive PDX revealed high and specific binding affinity for all three tracers labeled with lutetium-177 with IC
50 values in the nanomolar range. Interestingly, [177 Lu]Lu-JVZ-007 could not be displaced by PSMA-617 or PSMA-I&T, suggesting that this tracer targets an alternative binding site. Autoradiography assays on cryosections of human salivary and renal tissues revealed [177 Lu]Lu-PSMA-617 to have the lowest binding to these healthy organs compared with [177 Lu]Lu-PSMA-I&T. In vivo biodistribution assays confirmed the in vitro results with comparable tumor uptake of [177 Lu]Lu-PSMA-617 and [177 Lu]Lu-PSMA-I&T at all timepoints, resulting in induction of similar levels of DNA double-strand breaks in the tumors. However, [177 Lu]Lu-PSMA-I&T demonstrated approximately 40× higher renal uptake at 4 and 8 h post injection resulting in an unfavorable tumor-to-kidney ratio., Conclusion: [177 Lu]Lu-PSMA-617 has the most favorable biodistribution in mice as well as more favorable binding characteristics in vitro in PSMA-positive cells and human kidney and salivary gland specimens compared with [177 Lu]Lu-PSMA-I&T and [177 Lu]Lu-JVZ-007. Based on our preclinical evaluation, [177 Lu]Lu-PSMA-617 is the best performing tracer to be taken further into clinical evaluation for PSMA-targeted radiotherapeutic development although with careful evaluation of the tracer binding to PSMA-expressing organs.- Published
- 2021
- Full Text
- View/download PDF
37. Androgen receptor signalling impairs docetaxel efficacy in castration-resistant prostate cancer.
- Author
-
Mout L, Moll JM, Chen M, de Morrée ES, de Ridder CMA, Gibson A, Stuurman D, Aghai A, Erkens-Schulze S, Mathijssen RHJ, Sparreboom A, de Wit R, Lolkema MP, and van Weerden WM
- Subjects
- Acetylation, Androgen Antagonists pharmacokinetics, Androgen Receptor Antagonists therapeutic use, Animals, Antineoplastic Agents pharmacokinetics, Cell Death, Cell Line, Tumor, Cell Nucleus metabolism, Cell Survival, Disease Progression, Docetaxel pharmacokinetics, Drug Interactions, Humans, In Situ Nick-End Labeling, Male, Mice, Mice, Nude, Neoplasm Proteins metabolism, Neoplasm Transplantation, Prostate-Specific Antigen biosynthesis, Prostatic Neoplasms, Castration-Resistant metabolism, Prostatic Neoplasms, Castration-Resistant pathology, Receptors, Androgen drug effects, Signal Transduction drug effects, Solute Carrier Organic Anion Transporter Family Member 1B3 metabolism, Testosterone administration & dosage, Testosterone antagonists & inhibitors, Testosterone metabolism, Tubulin drug effects, Tubulin metabolism, Androgen Antagonists therapeutic use, Antineoplastic Agents therapeutic use, Docetaxel therapeutic use, Drug Resistance, Neoplasm physiology, Prostatic Neoplasms, Castration-Resistant drug therapy, Receptors, Androgen metabolism, Testosterone pharmacology
- Abstract
Androgen receptor (AR) signalling drives neoplastic growth and therapy resistance in prostate cancer. Recent clinical data show that docetaxel combined with androgen deprivation therapy improves outcome in hormone-sensitive disease. We studied whether testosterone and AR signalling interferes with docetaxel treatment efficacy in castration-resistant prostate cancer (CRPC). We found that testosterone supplementation significantly impaired docetaxel tumour accumulation in a CRPC model, resulting in decreased tubulin stabilisation and antitumour activity. Furthermore, testosterone competed with docetaxel for uptake by the drug transporter OATP1B3. Irrespective of docetaxel-induced tubulin stabilisation, AR signalling by testosterone counteracted docetaxel efficacy. AR-pathway activation could also reverse long-term tumour regression by docetaxel treatment in vivo. These results indicate that to optimise docetaxel efficacy, androgen levels and AR signalling need to be suppressed. This study lends evidence for continued maximum suppression of AR signalling by combining targeted therapeutics with docetaxel in CRPC.
- Published
- 2020
- Full Text
- View/download PDF
38. Testosterone Diminishes Cabazitaxel Efficacy and Intratumoral Accumulation in a Prostate Cancer Xenograft Model.
- Author
-
Mout L, de Wit R, Stuurman D, Verhoef E, Mathijssen R, de Ridder C, Lolkema M, and van Weerden W
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Humans, Male, Mice, Nude, Prostatic Neoplasms pathology, Treatment Outcome, Prostatic Neoplasms drug therapy, Taxoids pharmacokinetics, Taxoids therapeutic use, Testosterone therapeutic use, Xenograft Model Antitumor Assays
- Abstract
Inactivation of the androgen receptor (AR) pathway by androgen deprivation therapy (ADT) is the mainstay of (metastatic) prostate cancer therapy. Ultimately, the AR pathway will be re-activated despite castrate levels of circulating androgens. Thereby, maintaining its role even in castration resistant prostate cancer (CRPC). The recent STAMPEDE and CHAARTED trials showed that docetaxel in combination with ADT increased survival in hormone sensitive prostate cancer patients, suggesting cross-talk between AR signaling and chemotherapy efficacy. We hypothesized that a similar interaction may also apply for CRPC that is treated with cabazitaxel. We studied the impact of androgen status on the efficacy, pharmacodynamics and -kinetics of cabazitaxel in a unique and clinically relevant patient derived xenograft model of castration resistant disease. We found that cabazitaxel is highly effective in a castrate setting with strongly reduced AR activation, while tumor growth inhibition by cabazitaxel was completely abolished in the presence of high AR pathway activity. Moreover, additional experiments showed that intratumoral cabazitaxel levels were 3.5 times higher in tumors from castrated mice as compared to tumors from androgen-supplemented animals. We confirmed that cabazitaxel pharmacokinetics were not affected by testosterone, suggesting that androgen status might influence cabazitaxel tumor uptake directly. This study reveals the impact of androgen status on cabazitaxel efficacy and supports the potential of combination of taxane chemotherapeutics with AR axis targeting agents., (Copyright © 2017 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
39. High Efficacy of Combination Therapy Using PI3K/AKT Inhibitors with Androgen Deprivation in Prostate Cancer Preclinical Models.
- Author
-
Marques RB, Aghai A, de Ridder CMA, Stuurman D, Hoeben S, Boer A, Ellston RP, Barry ST, Davies BR, Trapman J, and van Weerden WM
- Subjects
- Aniline Compounds pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Castration methods, Cell Line, Tumor, Chromones pharmacology, Humans, Male, Prostatic Neoplasms metabolism, Pyrimidines pharmacology, Pyrroles pharmacology, Receptors, Androgen metabolism, Treatment Outcome, Xenograft Model Antitumor Assays methods, Cell Proliferation drug effects, PTEN Phosphohydrolase deficiency, Phosphoinositide-3 Kinase Inhibitors, Prostatic Neoplasms drug therapy, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Signal Transduction drug effects
- Abstract
Background: The phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT pathway is frequently activated during prostate cancer (PCa) progression through loss or mutation of the phosphatase and tensin homolog (PTEN) gene. Following the androgen receptor (AR) pathway, it is the second major driver of PCa growth., Objective: To assess efficacy of novel PI3K/AKT-targeted therapies in PCa models, as a single agent and in combination with androgen deprivation., Design, Setting, and Participants: Twelve human PCa cell lines were tested in vitro for sensitivity to the AKT inhibitor AZD5363 and the PI3K beta/delta inhibitor AZD8186. The combination of AZD5363 and AZD8186 with castration was evaluated in vivo in PTEN-negative versus PTEN-positive patient-derived xenografts. Tumors and plasma were collected for biomarker analysis., Outcome Measurements and Statistical Analysis: In vitro growth inhibition was determined by methylthiazolyldiphenyl-tetrazolium bromide assay. In vivo efficacy was monitored by caliper measurements of subcutaneous tumor volume. PI3K/AKT and AR pathway activity was analyzed by Western blot, enzyme-linked immunosorbent assay, and real-time polymerase chain reaction., Results and Limitations: AZD5363 and AZD8186 inhibited in vitro growth of 10 of 12 and 7 of 12 PCa cell lines, respectively, with increased sensitivity under androgen depletion. In vivo, AZD5363 and AZD8186 as single agents significantly inhibited growth of PTEN-negative PC346C xenografts compared to placebo by 60% and 66%, respectively. Importantly, combination of either agent with castration resulted in long-lasting tumor regression, which persisted after treatment cessation. Expression of AR-target genes kallikrein-related peptidase 3 (KLK3, also known as PSA); transmembrane protease, serine 2 (TMPRSS2); and FK506 binding protein 5 (FKBP5) was upregulated after PI3K/AKT inhibition. Neither compound inhibited tumor growth in the PTEN-positive PC310 model., Conclusions: Combination with hormonal therapy improved efficacy of PI3K/AKT-targeted agents in PTEN-negative PCa models. Upregulation of AR-target genes upon PI3K/AKT inhibition suggests a compensatory crosstalk between the PI3K-AR pathways. These data strongly advocate for further clinical evaluation., Patient Summary: Inactivation of the PTEN gene is a common event promoting prostate cancer (PCa) progression. This preclinical study illustrates the potent anticancer activity of novel PTEN-targeted drugs on PCa models, particularly in combination with hormonal therapy., (Copyright © 2014 European Association of Urology. Published by Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
40. [INCREASE IN EXPENDITURES OF PUBLIC HEALTH SERVICE].
- Author
-
STUURMAN D
- Subjects
- Humans, Netherlands, United States, Economics, Medical, Health Expenditures, Public Health Administration, United States Public Health Service
- Published
- 1964
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.