Morten Mau-Sorensen
Copenhagen University Hospital
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Morten Mau-Sorensen.
Neuro-oncology | 2015
Ulrik Lassen; Olivier Chinot; Catherine McBain; Morten Mau-Sorensen; Vibeke Andrée Larsen; Maryline Barrie; Patrick Roth; Oliver Krieter; Ka Wang; Kai Habben; Jean Tessier; Angelika Lahr; Michael Weller
BACKGROUND We conducted a phase 1 dose-escalation study of RO5323441, a novel antiplacental growth factor (PlGF) monoclonal antibody, to establish the recommended dose for use with bevacizumab and to investigate the pharmacokinetics, pharmacodynamics, safety/tolerability, and preliminary clinical efficacy of the combination. METHODS Twenty-two participants with histologically confirmed glioblastoma in first relapse were treated every 2 weeks with RO5323441 (625 mg, 1250 mg, or 2500 mg) plus bevacizumab (10 mg/kg). A standard 3 + 3 dose-escalation trial design was used. RESULTS RO5323441 combined with bevacizumab was generally well tolerated, and the maximum tolerated dose was not reached. Two participants experienced dose-limiting toxicities (grade 3 meningitis associated with spinal fluid leak [1250 mg] and grade 3 cerebral infarction [2500 mg]). Common adverse events included hypertension (14 participants, 64%), headache (12 participants, 55%), dysphonia (11 participants, 50%) and fatigue (6 participants, 27%).The pharmacokinetics of RO5323441 were linear, over-the-dose range, and bevacizumab exposure was unaffected by RO5323441 coadministration. Modulation of plasmatic angiogenic proteins, with increases in VEGFA and decreases in FLT4, was observed. Dynamic contrast-enhanced/diffusion-weighted MRI revealed large decreases in vascular parameters that were maintained through the dosing period. Combination therapy achieved an overall response rate of 22.7%, including one complete response, and median progression-free and overall survival of 3.5 and 8.5 months, respectively. CONCLUSION The toxicity profile of RO5323441 plus bevacizumab was acceptable and manageable. The observed clinical activity of the combination does not appear to improve on that obtained with single-agent bevacizumab in patients with recurrent glioblastoma.
Clinical Cancer Research | 2015
Markus Hansson; Peter Gimsing; Ashraf Badros; Titti Martinsson Niskanen; Hareth Nahi; Fritz Offner; Morten Salomo; Elisabeth Sonesson; Morten Mau-Sorensen; Yvonne Stenberg; Annika Sundberg; Ingrid Teige; Jan Van Droogenbroeck; Stina Wichert; Maurizio Zangari; Björn Frendéus; Magnus Korsgren; Martine Poelman; Guido Tricot
Purpose: This multicenter, first-in-human study evaluated safety, tolerability, pharmacokinetics, and pharmacodynamics of BI-505, a human anti-ICAM-1 monoclonal antibody, in advanced relapsed/refractory multiple myeloma patients. Experimental Design: BI-505 was given intravenously, every 2 weeks, at escalating doses from 0.0004 to 20 mg/kg, with extension of therapy until disease progression for responding or stable patients receiving 0.09 mg/kg or higher doses. Results: A total of 35 patients were enrolled. The most common adverse events were fatigue, pyrexia, headache, and nausea. Adverse events were generally mild to moderate, and those attributed to study medication were mostly limited to the first dose and manageable with premedication and slower infusion. No maximum tolerated dose was identified. BI-505′s half-life increased with dose while clearance decreased, suggesting target-mediated clearance. The ICAM-1 epitopes on patient bone marrow myeloma were completely saturated at 10 mg/kg doses. Using the International Myeloma Working Group criteria, 7 patients on extended therapy had stable disease for more than 2 months. Conclusions: BI-505 can be safely administered at doses that saturate myeloma cell ICAM-1 receptors in patients. This study was registered at www.clinicaltrials.gov (NCT01025206). Clin Cancer Res; 21(12); 2730–6. ©2015 AACR.
Clinical Cancer Research | 2015
Ulrik Niels Lassen; Didier Meulendijks; Lilian L Siu; Vaios Karanikas; Morten Mau-Sorensen; Jan H. M. Schellens; Derek J. Jonker; Aaron Richard Hansen; Mary Ellen Simcox; Kathleen Schostack; Dean Bottino; Hua Zhong; Markus Roessler; Suzana Vega-Harring; Tiantom Jarutat; David Geho; Ka Wang; Mark DeMario; Glenwood D. Goss
Purpose: Tumor necrosis factor (TNF)–like weak inducer of apoptosis (TWEAK) and fibroblast growth factor-inducible molecule 14 (Fn14) are a ligand–receptor pair frequently overexpressed in solid tumors. TWEAK:Fn14 signaling regulates multiple oncogenic processes through MAPK, AKT, and NFκB pathway activation. A phase I study of RG7212, a humanized anti-TWEAK IgG1κ monoclonal antibody, was conducted in patients with advanced solid tumors expressing Fn14. Experimental Design: Dose escalations, over a 200- to 7,200-mg range, were performed with patients enrolled in weekly (QW), bi-weekly (Q2W), or every-three-week (Q3W) schedules. Primary objectives included determination of dose and safety profile. Secondary endpoints included assessments related to inhibition of TWEAK:Fn14 signaling, tumor proliferation, tumor immune cell infiltration, and pharmacokinetics. Results: In 192 treatment cycles administered to 54 patients, RG7212 was well-tolerated with no dose-limiting toxicities observed. More than 95% of related adverse events were limited to grade 1/2. Pharmacokinetics were dose proportional for all cohorts, with a t1/2 of 11 to 12 days. Pharmacodynamic changes included clearance of free and total TWEAK ligand and reductions in tumor Ki-67 and TRAF1. A patient with BRAF wild-type melanoma who received 36 weeks of RG7212 therapy had tumor regression and pharmacodynamic changes consistent with antitumor effects. Fifteen patients (28%) received 16 or more weeks of RG7212 treatment. Conclusion: RG7212 demonstrated excellent tolerability and favorable pharmacokinetics. Pharmacodynamic endpoints were consistent with reduced TWEAK:Fn14 signaling. Tumor regression was observed and prolonged stable disease was demonstrated in multiple heavily pretreated patients with solid tumors. These encouraging results support further study of RG7212. Clin Cancer Res; 21(2); 258–66. ©2014 AACR.
Clinical Cancer Research | 2017
Ying Chen; Sandra Catalina Camacho; Thomas Silvers; Albiruni R. A. Razak; Nashat Y. Gabrail; John F. Gerecitano; Eva Kalir; Elena Pereira; Brad R. Evans; Susan J. Ramus; Fei Huang; Nolan Priedigkeit; Estefania Rodriguez; Michael J. Donovan; Faisal M. Khan; Tamara Kalir; Robert Sebra; Andrew V. Uzilov; Rong Chen; Rileen Sinha; Richard Halpert; Jean-Noel Billaud; Sharon Shacham; Dilara McCauley; Yosef Landesman; Tami Rashal; Michael Kauffman; Mansoor Raza Mirza; Morten Mau-Sorensen; Peter Dottino
Purpose: The high fatality-to-case ratio of ovarian cancer is directly related to platinum resistance. Exportin-1 (XPO1) is a nuclear exporter that mediates nuclear export of multiple tumor suppressors. We investigated possible clinicopathologic correlations of XPO1 expression levels and evaluated the efficacy of XPO1 inhibition as a therapeutic strategy in platinum-sensitive and -resistant ovarian cancer. Experimental Design: XPO1 expression levels were analyzed to define clinicopathologic correlates using both TCGA/GEO datasets and tissue microarrays (TMA). The effect of XPO1 inhibition, using the small-molecule inhibitors KPT-185 and KPT-330 (selinexor) alone or in combination with a platinum agent on cell viability, apoptosis, and the transcriptome was tested in immortalized and patient-derived ovarian cancer cell lines (PDCL) and platinum-resistant mice (PDX). Seven patients with late-stage, recurrent, and heavily pretreated ovarian cancer were treated with an oral XPO1 inhibitor. Results: XPO1 RNA overexpression and protein nuclear localization were correlated with decreased survival and platinum resistance in ovarian cancer. Targeted XPO1 inhibition decreased cell viability and synergistically restored platinum sensitivity in both immortalized ovarian cancer cells and PDCL. The XPO1 inhibitor–mediated apoptosis occurred through both p53-dependent and p53-independent signaling pathways. Selinexor treatment, alone and in combination with platinum, markedly decreased tumor growth and prolonged survival in platinum-resistant PDX and mice. In selinexor-treated patients, tumor growth was halted in 3 of 5 patients, including one with a partial response, and was safely tolerated by all. Conclusions: Taken together, these results provide evidence that XPO1 inhibition represents a new therapeutic strategy for overcoming platinum resistance in women with ovarian cancer. Clin Cancer Res; 23(6); 1552–63. ©2016 AACR.
European Journal of Cancer | 2015
Mie Grunnet; Ib Jarle Christensen; Ulrik Lassen; Lars Henrik Jensen; Magnus Lydolph; Jennifer J. Knox; Mairead Mcnamara; Mark Jitlal; Harpreet Wasan; John Bridgewater; Juan W. Valle; Morten Mau-Sorensen
BACKGROUND Carbohydrate associated antigen (CA19-9) has been approved by the FDA as a biomarker for monitoring treatment effect in pancreatic cancer. However, the value of serum CA19-9 as a biomarker of response to chemotherapy in bile duct cancer is unclear. The aim of this study was to determine if a decline in CA19-9 (CA19-9 response) during chemotherapy is predictive of survival in patients with inoperable bile duct cancer. METHODS Consecutive patients with inoperable bile duct cancer treated at a University Hospital were retrospectively included in an investigational cohort (n = 212). Three validation cohorts were established including patients 1) participating in phase I/II trials at a Danish Hospital (n = 71), 2) identified retrospectively in a Canadian cohort (n = 196) and 3) randomized in the ABC-02 trial (n = 410). Patients with a baseline CA19-9 and at least one CA19-9 value measured 10-12 weeks after the start of chemotherapy were included. Multivariate Cox regression analyses were performed. RESULTS Patients meeting the criteria to be included were 54 in the investigational cohort and 34, 68 and 148 in the three validation sets, respectively. Multivariate analysis included radiological response, performance status, bilirubin, gender, site of cancer, extend of disease, CA19-9 at baseline and age. A hazard ratio (HR) of 0.60 (95%CI: 0.44-0.80, p = 0.0005) for death in CA19-9 responders was reached in the investigational cohort. The predictive value of CA 19-9 response was confirmed in all three validation cohorts. CONCLUSIONS CA19-9 response is a robust predictor of survival in patients with inoperable bile duct cancer in four independent data sets.
International Journal of Neuroscience | 2016
Mikkel Christian Alanin; Camilla Klausen; Per Cayé-Thomasen; Carsten Thomsen; Kaare Fugleholm; Lars Poulsgaard; Ulrik Lassen; Morten Mau-Sorensen; Kenneth Francis Hofland
Purpose: The hallmark of neurofibromatosis type 2 (NF2) is bilateral vestibular schwannomas (VS). Approximately 80% of NF2 patients also have intracranial meningiomas. Vascular endothelial growth factor (VEGF) is expressed in both NF2-related and sporadic occurring meningiomas and anti-VEGF therapy (bevacizumab) may, therefore, be beneficial in NF2-related meningiomas. The purpose of the study was to report the effect of bevacizumab on meningiomas in NF2 patients. Materials and methods: We retrospectively reviewed the effect of bevacizumab on the cross-sectional area (CSA) of 14 intracranial meningiomas in 7 NF2 patients. Bevacizumab 10 mg/kg was administered intravenously every two weeks for six months and 15 mg/kg every three weeks thereafter. Patients were evaluated according to the modified Macdonald criteria with repeated magnetic resonance (MR) scans. Results: The median duration of therapy was 27 months (range 16–34) and 42 MR scans (median 8, range 4–11) were reviewed. The median annual change in meningioma CSA prior to bevacizumab was 2% (range –4%–+76%). During treatment, a decrease in meningioma CSA was observed in 5 of 14 meningiomas (36%) in 5 of 7 patients (71%). The median decrease in CSA was –10% (range –3%––25%). One meningioma (7%) progressed and the remaining (93%) had stable disease. Conclusions: Bevacizumab may slow or reverse the growth of some NF-related meningiomas. However, we have previously reported a fatal case of intracerebral hemorrhage following bevacizumab in NF2 patients, wherefore, this effect needs to be balanced carefully against the risk of side effects.
Clinical Cancer Research | 2016
Didier Meulendijks; Ulrik Niels Lassen; Lillian L. Siu; Alwin D. R. Huitema; Vaios Karanikas; Morten Mau-Sorensen; Derek J. Jonker; Aaron Richard Hansen; Mary Ellen Simcox; Kathleen Schostack; Dean Bottino; Hua Zhong; Markus Roessler; Suzana Vega-Harring; Tiantom Jarutat; David Geho; Karen Wang; Mark DeMario; Glenwood D. Goss; Jan H. M. Schellens
Purpose: The TWEAK–Fn14 pathway represents a novel anticancer target that is being actively investigated. Understanding the relationship between pharmacokinetics of anti-TWEAK therapeutics and tumor pharmacodynamics is critical. We investigated exposure-response relationships of RG7212, an anti-TWEAK mAb, in patients with Fn14-expressing tumors. Experimental Design: Patients with Fn14-positive tumors (IHC≥1+) treated in a phase I first-in-human study with ascending doses of RG7212 were the basis for this analysis. Pharmacokinetics of RG7212 and dynamics of TWEAK were determined, as were changes in tumor TWEAK–Fn14 signaling in paired pre- and posttreatment tumor biopsies. The objectives of the analysis were to define exposure-response relationships and the relationship between pretreatment tumor Fn14 expression and pharmacodynamic effect. Associations between changes in TWEAK–Fn14 signaling and clinical outcome were explored. Results: Thirty-six patients were included in the analysis. RG7212 reduced plasma TWEAK to undetectable levels at all observed RG7212 exposures. In contrast, reductions in tumor Fn14 and TRAF1 protein expression were observed only at higher exposure (≥300 mg*h/mL). Significant reductions in tumor Ki-67 expression and early changes in serum concentrations of CCL-2 and MMP-9 were observed exclusively in patients with higher drug exposure who had high pretreatment tumor Fn14 expression. Pretreatment tumor Fn14 expression was not associated with outcome, but a trend toward longer time on study was observed with high versus low RG7212 exposure. Conclusions: RG7212 reduced tumor TWEAK–Fn14 signaling in a systemic exposure-dependent manner. In addition to higher exposure, relatively high Fn14 expression might be required for pharmacodynamic effect of anti-TWEAK monoclonal antibodies. Clin Cancer Res; 22(4); 858–67. ©2015 AACR.
Scandinavian Journal of Gastroenterology | 2015
Mie Grunnet; Dan Calatayud; Nicolai Aa Schultz; Jane Preuss Hasselby; Morten Mau-Sorensen; Nils Brünner; Jan Stenvang
Abstract Background. Bile duct and pancreatic cancer (PC) have poor prognoses and treatment options for inoperable patients are scarce. In order to improve outcome for these patients, there is an urgent need for biomarkers predictive of treatment effect. Irinotecan is a topoisomerase 1 (Top1) poison. Top1 protein, TOP1 gene copy number and mRNA expression, respectively, have been proposed as predictive biomarkers of response to irinotecan in other cancers. Here we investigate the occurrence of TOP1 gene aberrations in cancers of the bile ducts and pancreas. Material and methods. TOP1 and centromere 20 (CEN-20) numbers were investigated by fluorescence in situ hybridization analyses in tumor tissue from 226 patients. The frequencies of aberration in the TOP1 gene copy number, the CEN-20 copy number and the TOP1/CEN-20 ratio were analyzed. As TOP1 is located on chromosome 20, the CEN-20 probe was included to distinguish between chromosomal and gene amplifications. Results. In PC, 29.8% had an increased TOP1 copy number (≥3.5n gene copies per cell) and 10.8% had a TOP1/CEN-20 ratio >1.5. In bile duct cancer, 12.8 % had an increased TOP1 copy number and 6.4% had a TOP1/CEN-20 ratio >1.5. Neither the TOP1 copy number nor the TOP1/CEN-20 ratios could predict overall survival. Conclusion. We here report that a substantial number of patients with bile duct or PC have increased TOP1 copy number and increased TOP1/CEN-20 ratio making further analyses on the association between TOP1 gene copy number and irinotecan efficacy clinically relevant.
Clinical Biochemistry | 2014
Mie Grunnet; Ib Jarle Christensen; Ulrik Lassen; Lars Henrik Jensen; M. Lydolph; I.K. Lund; T. Thurison; Gunilla Høyer-Hansen; Morten Mau-Sorensen
BACKGROUND High levels of intact and cleaved forms of the urokinase-type plasminogen activator receptor (uPAR) in both tissue and blood are associated with poor survival in several cancer diseases. The prognostic significance of uPAR in cholangiocarcinoma is unknown. The aims of this study were to determine if pre-treatment serum levels of uPAR forms and a decrease in levels during chemotherapy are predictive of survival in patients with inoperable cholangiocarcinoma. DESIGN AND METHODS Patients with inoperable cholangiocarcinoma were consecutively included in the training set (n=108). A test set included patients from a different hospital using similar treatment guidelines (n=60). Serum levels of the different uPAR forms were determined using time-resolved fluorescence immunoassays (TR-FIA). The Cox proportional hazards model was used for the uni- and multivariate survival analyses. RESULTS Baseline level of uPAR(I-III)+uPAR(II-III) was an independent predictor of survival (HR=2.08, 95% CI:1.46-2.97, p<0.0001). Applying the linear predictor from the training set to the test set, it was validated that uPAR(I-III)+uPAR(II-III) predicted overall survival (p=0.049). A high level of uPAR(I-III)+uPAR(II-III) after 2cycles of chemotherapy was associated with poor survival (HR=1.79, 95% CI:1.08-2.97, p=0.023, n=57). This predictor, however, was not significant in the test set (p=0.21, 26 events in 27 patients). CONCLUSION The baseline level of uPAR(I-III)+uPAR(II-III) is a predictor of survival in inoperable cholangiocarcinoma patients.
Annals of Oncology | 2014
A. Mahipal; Nashat Y. Gabrail; A. Sukari; Hemchandra Mahaseth; Morten Mau-Sorensen; Sharon Shacham; Jean-Richard Saint-Martin; S. Friedlander; Yosef Landesman; J. Ellis; Eran Shacham; J. McCartney; T. Marshall; D. Vincent; Tami Rashal; Michael Kauffman; Mansoor Raza Mirza; Albiruni R. A. Razak
ABSTRACT Aim: The nuclear export of most tumor suppressor proteins (TSPs) inactivates their anti-cancer functions. Exportin 1 (XPO1) is the exclusive nuclear exporter of nearly all TSPs. Multiple TSP pathways are altered in HN-SCC including p53, CDKN2A, pRB and others. Selinexor (KPT-330) is a novel, orally bioavailable, selective inhibitor of nuclear export (SINE) that blocks XPO1, leading to the nuclear accumulation and re-activation of TSPs. Selinexor has shown potent in vitro and in vivo activity in numerous models of solid tumors with squamous histology. Methods: Oral selinexor was given at 8-10 doses (2-3 times/week)/28-day cycle as part of an ongoing Phase 1 study in advanced solid tumors (KCP-330-002, NCT01607905). Response evaluation was done every 2 cycles (RECIST 1.1). Over 130 patients (pts) were enrolled, including 17 pts with heavily pretreated HN-SCC with objectively progressive disease (PD) on study entry. Results: Seventeen pts with HN-SCC (14 M, 3 F; median age 56 yrs; ECOG PS 0/1: 4/13; median prior regimens: 2.6 (range 1-4)) received selinexor across 6 dose levels (23 to 65 mg/m2). Grade 4 AEs included thrombocytopenia without bleeding in 3 pts. The most common AEs (Grade 1/2/3) include: fatigue (12%/41%/6%), nausea (41%/6%/6%), anorexia (12%/41%/0%), diarrhea (35%/12%/0%), weight loss (18%/24%/0%) and thrombocytopenia (18%/0%/12%). Supportive care with glucocorticoids, appetite stimulants and anti-emetics improved constitutional symptoms. Pharmacokinetics and pharmacodynamics showed dose-dependent increases in Cmax/AUC0-inf and in XPO1 mRNA. Tumor biopsies showed nuclear localization of TSPs (p53, FOXO3A, IkB) and apoptosis induction. Fourteen pts with PD on study entry were evaluable for efficacy. Nine patients (64%) had stable disease and 5 pts (36%) had progressive disease. 5 pts have remained on therapy for ≥4 months (4-13). Conclusions: Oral selinexor is generally well tolerated and can be administered over prolonged periods. Durable single agent disease control was observed in heavily pretreated HN-SCC pts. The recommended dose is 65 mg/m2 twice weekly and a Phase 2 study in patients with HN-SCC is expected to begin in mid 2014. Disclosure: All authors have declared no conflicts of interest.