Network


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

Hotspot


Dive into the research topics where Ray Valencia is active.

Publication


Featured researches published by Ray Valencia.


Clinical Cancer Research | 2013

In Vivo Imaging of Prostate Cancer Using [68Ga]-Labeled Bombesin Analog BAY86-7548

Esa Kähkönen; Ivan Jambor; Jukka Kemppainen; Kaisa Lehtiö; Tove Grönroos; Anna Kuisma; Pauliina Luoto; Henri J Sipila; Tuula Tolvanen; Kalle Alanen; Jonna Silén; Markku Kallajoki; Anne Roivainen; Niklaus Schäfer; Roger Schibli; Martina Dragic; Anass Johayem; Ray Valencia; Sandra Borkowski; Heikki Minn

Purpose: A novel [68Ga]-labeled DOTA-4-amino-1-carboxymethyl-piperidine-D-Phe-Gln-Trp-Ala-Val-Gly-His-Sta-Leu-NH2 peptide (BAY86-7548) having high affinity to bombesin receptor subtype II to detect primary and metastatic prostate carcinoma using positron emission tomography/computed tomography (PET/CT) was synthesized and evaluated for prostate cancer. Experimental Design: In this first human study with BAY86-7548, 14 men scheduled for radical prostatectomy (n = 11) or with biochemical recurrence after surgery or hormonal therapy (n = 3) were enrolled. The patients received an intravenous injection of BAY86-7548 followed by over 60-minute dynamic imaging of prostate gland (n = 10) and/or subsequent whole-body imaging (n = 14). The visual assessment of PET/CT images included evaluation of intraprostatic (12 subsextants) and pelvic nodal uptake of BAY86-7548 in 11 surgical patients and detection of potential metastatic foci in all patients. In patients with biochemical recurrence, results were compared with those of either [11C]-acetate (n = 2) or [18F]-fluoromethylcholine (n = 1) PET/CT. Results: We found a sensitivity, specificity, and accuracy of 88%, 81% and 83%, respectively, for detection of primary PCa and sensitivity of 70% for metastatic lymph nodes using histology as gold standard. BAY86-7548 correctly detected local recurrence in prostate bed and showed nodal relapse in accordance with [11C]-acetate PET/CT in 2 patients with biochemical relapse. In the third hormone refractory patient, BAY86-7548 failed to show multiple bone metastases evident on [18F]-fluoromethylcholine PET/CT. Conclusion: BAY86-7548 PET/CT is a promising molecular imaging technique for detecting intraprostatic prostate cancer. Clin Cancer Res; 19(19); 5434–43. ©2013 AACR.


The Journal of Nuclear Medicine | 2013

Plasma Pharmacokinetics, Whole-Body Distribution, Metabolism, and Radiation Dosimetry of 68Ga Bombesin Antagonist BAY 86-7548 in Healthy Men

Anne Roivainen; Esa Kähkönen; Pauliina Luoto; Sandra Borkowski; Birte Hofmann; Ivan Jambor; Kaisa Lehtiö; Tuija Rantala; Antje Rottmann; Henri Sipilä; Rick Sparks; Sami Suilamo; Tuula Tolvanen; Ray Valencia; Heikki Minn

This first-in-human study investigated the safety, tolerability, metabolism, pharmacokinetics, biodistribution, and radiation dosimetry of 68Ga-bombesin antagonist 68Ga-DOTA-4-amino-1-carboxymethylpiperidine-d-Phe-Gln-Trp-Ala-Val-Gly-His-Sta-Leu-NH2 (BAY 86-7548). Methods: Five healthy men underwent dynamic whole-body PET/CT after an intravenous injection of BAY 86-7548 (138 ± 5 MBq). Besides total radioactivity, plasma samples were analyzed by radio–high-performance liquid chromatography for metabolism of the tracer. Dosimetry was calculated using the OLINDA/EXM software. Results: Three radioactive plasma metabolites were detected. The proportion of unchanged BAY 86-7548 decreased from 92% ± 9% at 1 min after injection to 19% ± 2% at 65 min. The organs with the highest absorbed doses were the urinary bladder wall (0.62 mSv/MBq) and the pancreas (0.51 mSv/MBq). The mean effective dose was 0.051 mSv/MBq. BAY 86-7548 was well tolerated by all subjects. Conclusion: Intravenously injected BAY 86-7548 is safe, and rapid metabolism is demonstrated. A 150-MBq injection of BAY 86-7548 results in an effective dose of 7.7 mSv, which could be reduced to 5.7 mSv with frequent bladder voids.


Journal of Cerebral Blood Flow and Metabolism | 2015

Quantification of [18F]DPA-714 binding in the human brain: initial studies in healthy controls and Alzheimer's disease patients.

Sandeep S.V. Golla; Ronald Boellaard; Vesa Oikonen; Anja Hoffmann; Bart N.M. van Berckel; Albert D. Windhorst; Jere Virta; Merja Haaparanta-Solin; Pauliina Luoto; Nina Savisto; Olof Solin; Ray Valencia; Andrea Thiele; Jonas Eriksson; Robert C. Schuit; Adriaan A. Lammertsma; Juha O. Rinne

Fluorine-18 labelled N,N-diethyl-2-(2-[4-(2-fluoroethoxy)phenyl]-5,7-dimethylpyrazolo[1,5-α]pyrimidine-3-yl)acetamide ([18F] DPA-714) binds to the 18-kDa translocator protein (TSPO) with high affinity. The aim of this initial methodological study was to develop a plasma input tracer kinetic model for quantification of [18F]DPA-714 binding in healthy subjects and Alzheimers disease (AD) patients, and to provide a preliminary assessment whether there is a disease-related signal. Ten AD patients and six healthy subjects underwent a dynamic positron emission tomography (PET) study along with arterial sampling and a scan protocol of 150 minutes after administration of 250 ± 10 MBq [18F]DPA-714. The model that provided the best fits to tissue time activity curves (TACs) was selected based on Akaike Information Criterion and F-test. The reversible two tissue compartment plasma input model with blood volume parameter was the preferred model for quantification of [18F]DPA-714 kinetics, irrespective of scan duration, volume of interest, and underlying volume of distribution (VT). Simplified reference tissue model (SRTM)-derived binding potential (BPND) using cerebellar gray matter as reference tissue correlated well with plasma input-based distribution volume ratio (DVR). These data suggest that [18F]DPA-714 cannot be used for separating individual AD patients from heathy subjects, but further studies including TSPO binding status are needed to substantiate these findings.


The Journal of Nuclear Medicine | 2015

Dosimetry and first clinical evaluation of the new 18F-radiolabeled bombesin analogue BAY 864367 in patients with prostate cancer.

Bert-Ram Sah; Irene A. Burger; Roger Schibli; Matthias Friebe; Ludger Dinkelborg; Keith Graham; Sandra Borkowski; Claudia Bacher-Stier; Ray Valencia; Ananth Srinivasan; Thomas F. Hany; Linjing Mu; Peter Wild; Niklaus Schaefer

The aim of this first-in-man study was to demonstrate the feasibility, safety, and tolerability, as well as provide dosimetric data and evaluate the imaging properties, of the bombesin analogue BAY 864367 for PET/CT in a small group of patients with primary and recurrent prostate cancer (PCa). Methods: Ten patients with biopsy-proven PCa (5 with primary PCa and 5 with prostate-specific antigen recurrence after radical prostatectomy) were prospectively selected for this exploratory clinical trial with BAY 864367, a new 18F-labeled bombesin analogue. PET scans were assessed at 6 time points, up to 110 min after intravenous administration of 302 ± 11 MBq of BAY 864367. Imaging results were compared with 18F-fluorocholine PET/CT scans. Dosimetry was calculated using the OLINDA/EXM software. Results: Three of 5 patients with primary disease showed positive tumor delineation in the prostate, and 2 of 5 patients with biochemical relapse showed a lesion suggestive of recurrence on the BAY 864367 scan. Tumor-to-background ratio averaged 12.9 ± 7.0. The ratio of malignant prostate tissue to normal prostate tissue was 4.4 ± 0.6 in 3 patients with tracer uptake in the primary PCa. Mean effective dose was 4.3 ± 0.3 mSv/patient (range, 3.7–4.9 mSv). Conclusion: BAY 864367, a novel 18F-labeled bombesin tracer, was successfully investigated in a first-in-man clinical trial of PCa and showed favorable dosimetric values. Additionally, the application was safe and well tolerated. The tracer delineated tumors in a subset of patients, demonstrating the potential of gastrin-releasing-peptide receptor imaging.


Cancer Research | 2015

Abstract DDT02-02: BAY 1143572: A first-in-class, highly selective, potent and orally available inhibitor of PTEFb/CDK9 currently in Phase I, inhibits MYC and shows convincing anti-tumor activity in multiple xenograft models by the induction of apoptosis

Arne Scholz; Ulrich Luecking; Gerhard Siemeister; Philip Lienau; Ulf Boemer; Peter Ellinghaus; Annette O. Walter; Ray Valencia; Stuart Ince; Franz von Nussbaum; Dominik Mumberg; Michael Brands; Karl Ziegelbauer

PTEFb/CDK9 mediated transcription of short-lived anti-apoptotic survival proteins like MYC, a key oncogene in multiple tumors, plays a critical role in cancer cell growth and survival. In addition, these survival proteins exhibit important functions in the development of resistance to chemotherapy. In contrast to pan-CDK inhibitors which are currently evaluated in Phase I and II clinical trials, to our knowledge PTEFb selective inhibitors have not been explored for clinical utility. We report for the first time the preclinical profile and structure of BAY 1143572, a novel selective PTEFb/CDK9 inhibitor currently being investigated in a Phase I clinical trial. BAY 1143572 had potent and highly selective PTEFb-kinase inhibitory activity in the low nanomolar range against PTEFb/CDK9 and an at least 50-fold selectivity against other CDKs in enzymatic assays. Furthermore, BAY 1143572 showed a favorable selectivity against a panel of non-CDK kinases in vitro. The potent enzymatic activity on PTEFb translated into broad antiproliferative activity against a panel of tumor cell lines with sub-micromolar IC-50 values. In line with the proposed mode of action, a concentration-dependent inhibition of the phosphorylation of the RNA polymerase II and downstream reduction of MYC mRNA and protein levels was observed in vitro. This inhibition was accompanied by an induction of apoptosis in cellular assays. BAY 1143572 also showed single agent in vivo efficacy at tolerated doses in various xenograft tumor models in mice and rats upon once daily oral administration. Potent anti-tumor activity characterized with partial or even complete remissions could be documented in models showing different MYC gene alterations like amplifications and translocations. Treatment with BAY 1143572 resulted in a transient inhibition of intratumoral MYC mRNA and protein levels and an induction of apoptosis in these models. The inhibition of MYC mRNA was also observed in blood cells of BAY 1143572-treated rats indicating the potential clinical utility of MYC in blood cells as a pharmacodynamic marker in clinical development. The in vivo efficacy of BAY 1143572 was significantly enhanced in combination with several chemotherapeutics in different solid tumor models. These pharmacology data provided the rationale for the initiation of clinical development of BAY 1143572 in advanced cancer patients (NCT01938638). In conclusion, our data provide preclinical proof of concept for BAY 1143572 as a potent and highly selective inhibitor of PTEFb/CDK9 with first-in-class potential. Further clinical evaluation of BAY 1143572 for the treatment of cancers dependent on the transcription of the key oncogene MYC and other short-lived survival proteins is warranted. Citation Format: Arne Scholz, Ulrich Luecking, Gerhard Siemeister, Philip Lienau, Ulf Boemer, Peter Ellinghaus, Annette O. Walter, Ray Valencia, Stuart Ince, Franz von Nussbaum, Dominik Mumberg, Michael Brands, Karl Ziegelbauer. BAY 1143572: A first-in-class, highly selective, potent and orally available inhibitor of PTEFb/CDK9 currently in Phase I, inhibits MYC and shows convincing anti-tumor activity in multiple xenograft models by the induction of apoptosis. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr DDT02-02. doi:10.1158/1538-7445.AM2015-DDT02-02


The Journal of Nuclear Medicine | 2016

Parametric Binding Images of the TSPO Ligand 18F-DPA-714

Sandeep S.V. Golla; Ronald Boellaard; Vesa Oikonen; Anja Hoffmann; Bart N.M. van Berckel; Albert D. Windhorst; Jere Virta; Erik te Beek; Geert Jan Groeneveld; Merja Haaparanta-Solin; Pauliina Luoto; Nina Savisto; Olof Solin; Ray Valencia; Andrea Thiele; Jonas Eriksson; Robert C. Schuit; Adriaan A. Lammertsma; Juha O. Rinne

18F-labeled N,N-diethyl-2-(2-[4-(2-fluoroethoxy)phenyl]-5,7-dimethylpyrazolo[1,5-α]pyrimidine-3-yl)acetamide (DPA-714) is a radioligand for the 18-kDa translocator protein. The purpose of the present study was to identify the best method for generating quantitative parametric images of 18F-DPA-714 binding. Methods: Ninety-minute dynamic 18F-DPA-714 PET scans with full arterial sampling from 6 healthy subjects and 9 Alzheimer disease (AD) patients were used. Plasma-input–based Logan graphical analysis and spectral analysis were used to generate parametric volume of distribution (VT) images. Five versions of Ichise, reference Logan, and 2 basis function implementations (receptor parametric mapping and simplified reference tissue model 2 [SRTM2]) of SRTM, all using gray matter cerebellum as the reference region, were applied to generate nondisplaceable binding potential (BPND) images. Results: Plasma-input Logan analysis (r2 = 0.99; slope, 0.88) and spectral analysis (r2 = 0.99, slope, 0.93) generated estimates of VT that correlated well with values obtained using nonlinear regression. BPND values generated using SRTM2 (r2 = 0.83; slope, 0.95) and reference Logan analysis (r2 = 0.88; slope, 1.01) correlated well with nonlinear regression–based estimates. Conclusion: Both Logan analysis and spectral analysis can be used to obtain quantitatively accurate VT images of 18F-DPA-714. In addition, SRTM2 and reference Logan analysis can provide accurate BPND images. These parametric images could be used for voxel-based comparisons.


Cancer Research | 2016

Abstract 3022: BAY 1143572, a first-in-class, highly selective, potent and orally available inhibitor of PTEFb/CDK9 currently in Phase I, shows convincing anti-tumor activity in preclinical models of acute myeloid leukemia (AML)

Arne Scholz; Thomas Oellerich; Akhtar Hussain; Sarah Lindner; Ulrich Luecking; Annette O. Walter; Peter Ellinghaus; Ray Valencia; Franz von Nussbaum; Dominik Mumberg; Michael Brands; Stuart Ince; Hubert Serve; Karl Ziegelbauer

PTEFb/CDK9 mediated transcription of short-lived anti-apoptotic survival proteins like Mcl-1 and Myc, plays a critical role in cancer cell growth and survival in various tumor entities including AML. In addition, these survival proteins exhibit important functions in the development of resistance to chemotherapy. In contrast to pan-CDK inhibitors, to our knowledge PTEFb selective inhibitors have not been explored for clinical utility. We report the preclinical activity of BAY 1143572, a novel selective PTEFb/CDK9 inhibitor (AACR; Cancer Res 2015;75(15 Suppl):Abstract nr DDT02-02) currently being investigated in Phase I clinical trials in advanced cancer (NCT01938638) and acute leukemia (NCT02345382) in various in vitro, ex vivo and in vivo models of AML. BAY 1143572 inhibited the proliferation of 7 MLL-rearrangements positive and negative AML cell lines with a median IC50 of 385 nM (range 230-1100 nM) and induced apoptosis. Furthermore, BAY 1143572 showed potent in vitro activity in 8 out of 10 non-MLL-rearranged patient derived AML samples incl. NPM1 mutant and Flt3-ITD positive samples derived from intermediate and high risk patients. Moreover, we elucidated the dynamic changes of the cellular proteome/phosphoproteome upon pharmacological and genetic PTEFb inhibition and identified PTEFb interaction partners in various AML in vitro models. These analyses uncover the oncogenic PTEFb-dependent signaling networks and substantiate the molecular rationale for the use of PTEFb inhibitors in this indication. When applied in vivo, BAY 1143572 exhibited single agent efficacy at tolerated doses in 4 out of 5 AML xenograft tumor models in mice and in 2 out of 2 AML xenograft tumor models in rats upon once daily oral administration. Of note, partial or even complete remissions could be achieved in several models. Furthermore, intermittent dosing schedules with up to 4 days treatment pauses were feasible in terms of efficacy and tolerability. Using MOLM-13 xenografts in mice and rats to address the in vivo MoA of BAY 1143572, a transient inhibition of RNA polymerase II phosphorylation, MYC mRNA and protein levels, MCL-1 mRNA and protein levels, and an induction of apoptosis was documented. In conclusion, our data provide the rationale for the initiation of clinical development of BAY 1143572 as a potent and highly selective inhibitor of PTEFb/CDK9 with first-in-class potential for the treatment of AML patients. A phase I clinical trial to determine the safety, tolerability and recommended Phase 2 dose in this indication is ongoing (NCT02345382). Citation Format: Arne Scholz, Thomas Oellerich, Akhtar Hussain, Sarah Lindner, Ulrich Luecking, Annette O. Walter, Peter Ellinghaus, Ray Valencia, Franz von Nussbaum, Dominik Mumberg, Michael Brands, Stuart Ince, Hubert Serve, Karl Ziegelbauer. BAY 1143572, a first-in-class, highly selective, potent and orally available inhibitor of PTEFb/CDK9 currently in Phase I, shows convincing anti-tumor activity in preclinical models of acute myeloid leukemia (AML). [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3022.


Cancer Research | 2015

Abstract 2828: Rapid identification of potent and highly selective, oral PTEFb Inhibitor BAY 1143572 with first in class potential

Ulrich Luecking; Arne Scholz; Philip Lienau; Gerhard Siemeister; Dirk Kosemund; Rolf Bohlmann; Knut Eis; Mark Jean Gnoth; Ildiko Terebesi; Kirstin Meyer; Katja Prelle; Ray Valencia; Stuart Ince; Franz von Nussbaum; Dominik Mumberg; Karl Ziegelbauer; Bert Klebl; Axel Choidas; Peter Nussbaumer; Matthias Baumann; Carsten Schultz-Fademrecht; Gerd Ruehter; Jan Eickhoff; Michael Brands

PTEFb (positive transcription elongation factor b) is a heterodimer of the transcriptional control kinase CDK9 (Cyclin-dependent kinase 9) and Cyclin T. PTEFb phosphorylates and activates RNA polymerase II. PTEFb inhibition causes rapid depletion of short-lived mRNA transcripts and their associated protein products involved in proliferation and survival like Myc, or Mcl-1 which results in cell death of addicted tumor cells. We previously disclosed the profile of the lead compound PTEFb BAY1, a nanomolar PTEFb inhibitor with 50-fold selectivity within the CDK family and cellular potency of about 1 μM in proliferation assays on various human tumor cell lines [1]. PTEFb BAY1 also revealed in vivo efficacy in a human acute myeloid leukemia (AML) xenograft model in nude mice. However, the lead compound also displayed certain limitations in ADME properties like low aqueous solubility and a strong recognition by efflux transporters in the Caco2 assay. Based on these findings, extensive lead optimisation efforts led to the rapid identification of BAY 1143572 which is a more potent and highly selective, orally available PTEFb inhibitor with first-in-class potential. BAY 1143572 has a high aqueous solubility, reduced drug efflux and a moderate oral bioavailability across species that allows daily as well as intermittent dosing schedules in animal models. BAY 1143572 revealed strong in vitro and in vivo anti-tumor efficacy with various cell-lines. BAY 1143572 is currently being evaluated in a Phase I study to determine the safety, tolerability, pharmacokinetics and initial pharmacodynamic biomarker response in patients with advanced cancer. This presentation will highlight the key learnings from our PTEFb lead optimization program. [1]: AACR, April 5-9, 2014, San Diego, Poster Presentation, Abstract 4538, Cancer Res October 1, 2014, 74:4538; doi:10.1158/1538-7445.AM2014-4538 Citation Format: Ulrich TJ Luecking, Arne Scholz, Philip Lienau, Gerhard Siemeister, Dirk Kosemund, Rolf Bohlmann, Knut Eis, Mark Gnoth, Ildiko Terebesi, Kirstin Meyer, Katja Prelle, Ray Valencia, Stuart Ince, Franz von Nussbaum, Dominik Mumberg, Karl Ziegelbauer, Bert Klebl, Axel Choidas, Peter Nussbaumer, Matthias Baumann, Carsten Schultz-Fademrecht, Gerd Ruehter, Jan Eickhoff, Michael Brands. Rapid identification of potent and highly selective, oral PTEFb Inhibitor BAY 1143572 with first in class potential. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2828. doi:10.1158/1538-7445.AM2015-2828


Cancer Research | 2017

Abstract 984: Identification of potent and highly selective PTEFb inhibitor BAY 1251152 for the treatment of cancer: from p.o. to i.v. application via scaffold hops

Ulrich Luecking; Arne Scholz; Dirk Kosemund; Rolf Bohlmann; Hans Briem; Philip Lienau; Gerhard Siemeister; Ildiko Terebesi; Kirstin Meyer; Katja Prelle; Ray Valencia; Stuart Ince; Franz von Nussbaum; Dominik Mumberg; Karl Ziegelbauer; Michael Brands

PTEFb/CDK9 mediated transcription of short-lived anti-apoptotic survival proteins like Mcl-1 and Myc plays a critical role in cancer cell growth and survival in various tumor entities including AML. In addition, these survival proteins play important roles in the development of resistance to chemotherapy. We previously disclosed the preclinical profile of BAY 1143572, the first selective, orally available PTEFb/CDK9 inhibitor that entered clinical development [1-3]. BAY 1143572 had low nanomolar activity against PTEFb/CDK9, an at least 50-fold selectivity against other CDKs in enzymatic assays and broad anti-proliferative activity against a panel of tumour cell lines with sub-micromolar IC50 values. BAY 1143572 also showed single agent in vivo efficacy at tolerated doses in various xenograft tumour models in mice and rats upon once daily oral administration. To fully explore future treatment options using selective PTEFb/CDK9 inhibitors we initiated a follow-up program to identify novel PTEFb/CDK9 inhibitors for treatment of cancer with increased potency enabling i.v. treatment of patients. Extensive lead optimisation efforts, including various scaffold hops, led to the identification of BAY 1251152. In comparison to oral BAY 1143572, BAY 1251152 shows significantly increased biochemical (IC50 CDK9 = 3 nM) and cellular potency (IC50 MOLM13 = 29 nM), increased selectivity against CDK2 as well as high permeability and no efflux. The significantly reduced therapeutic dose and high solubility of BAY 1251152 enable the desired i.v. application. BAY 1251152 demonstrated excellent efficacy upon i.v. treatment in xenograft models (e.g. MOLM13) in mice and rats. BAY 1251152 is currently being evaluated in Phase I studies (NCT02635672; NCT02745743) to determine the safety, tolerability, pharmacokinetics and initial pharmacodynamic biomarker response in patients with advanced cancer. This presentation will highlight the key learnings from our PTEFb/CDK9 i.v. lead optimization program and disclose the structure of BAY 1251152 for the first time. [1]: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3022. [2]: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr DDT02-02. doi:10.1158/1538-7445.AM2015-DDT02-02. [3]: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2828. doi:10.1158/1538-7445.AM2015-2828 Citation Format: Ulrich T. Luecking, Arne Scholz, Dirk Kosemund, Rolf Bohlmann, Hans Briem, Philip Lienau, Gerhard Siemeister, Ildiko Terebesi, Kirstin Meyer, Katja Prelle, Ray Valencia, Stuart Ince, Franz von Nussbaum, Dominik Mumberg, Karl Ziegelbauer, Michael Brands. Identification of potent and highly selective PTEFb inhibitor BAY 1251152 for the treatment of cancer: from p.o. to i.v. application via scaffold hops [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 984. doi:10.1158/1538-7445.AM2017-984


Cancer Research | 2017

Abstract CT094: Phase I study of fibroblast growth factor receptor 2 antibody-drug conjugate (FGFR2-ADC) BAY 1187982 in patients with advanced cancer

Sung-Bae Kim; Funda Meric-Bernstam; Jordan Berlin; Sabine Wittemer-Rump; Motonobu Osada; Ray Valencia; Aleksei Babich; Rong Liu; Andy Hwang; Takahiko Tanigawa; Frank Reetz; Dirk Laurent; Aparna Kalyan

Background: FGFR2-ADC (BAY 1187982) consists of an anti-FGFR2 antibody (BAY 1179470) and a stably linked, highly potent and non-cell permeable microtubule-binding auristatin toxophore. A phase I study was conducted in patients (pts) with advanced solid tumors focusing on tumor types with a described expression of FGFR2. Methods: Eligible pts with solid tumors, known to express FGFR2 and refractory to standard treatment, adequate organ function, and ECOG performance status (PS) ≤ 1 were enrolled. BAY 1187982 was administered by an one hour intravenous infusion on an every 21 days (q3w) schedule in 20 pts (5 cholangiocarcinoma, 5 colon, 2 gastric, 2 breast cancer and 6 others) at doses ranging from 0.1 to 1.3 mg/kg. Dose escalation followed the continous reassessment design treating 3-4 pts per cohort. Pts were assessed for AEs weekly in C1-C3 and on D1 in subsequent cycles. Tumor response was assessed every 6 weeks. Blood samples for a full pharmacokinetic profile were collected after dosing on D1 of C1 and C3 and thereafter every second cycle pre-dose and at the end of infusion. Results: A total of 20 pts were evaluable: 55% female, mean age 52 yrs (range 24-76), PS 0/1 75% / 25%. No drug-related serious adverse events were reported at dose levels 0.1 and 0.2 mg/kg. At 0.4 mg/kg drug-related serious adverse events were similar to other ADCs, including reversible asymptomatic liver function test increases, and gastrointestinal disorders. Dose limiting toxicities (DLTs) were observed at dose level 0.8 and 1.3 mg/kg, respectively: 0.8 mg/kg cohort grade 3 thrombocytopenia (N=1); 1.3 mg/kg cohort grade 3 corneal epitheliopathy (N=1), grade 4 thrombocytopenia (N=1) and proteinuria progressing to nephrotic syndrome after treatment in C2 or C3 (N=2) with signs of reversibility after discontinuation. In dose level 0.4 and 0.8 mg/kg the majority of pts discontinued study drug treatment already in C1 or C2 due to disease progression and data on proteinuria at these dose levels were absent or spare. As a consequence only doses below 0.4 mg/kg were considered safe. Exposures of BAY 1187982 increased in a dose-proportional manner in the investigated dose range from 0.1 to 1.3 mg/kg. Conclusion: While this drug was tolerated well at lower dose levels, dose-limiting toxicities occurred below the expected therapeutic dose level. As the predicted human efficacious dose level based on the majority of animal models was above 0.4 mg/kg (Wittemer-Rump et al. AACR; Cancer Res 2015;75, 15 Suppl: Abstract nr 1683) a Phase II dose cannot be recommended and a successful clinical development of the compound was regarded as unlikely. The data from the preclinical safety analysis in monkey and rat will be submitted as abstract to AACR 2017. Citation Format: Sung-Bae Kim, Funda Meric-Bernstam, Jordan Berlin, Sabine Wittemer-Rump, Motonobu Osada, Ray Valencia, Aleksei Babich, Rong Liu, Andy Hwang, Takahiko Tanigawa, Frank Reetz, Dirk Laurent, Aparna Kalyan. Phase I study of fibroblast growth factor receptor 2 antibody-drug conjugate (FGFR2-ADC) BAY 1187982 in patients with advanced cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr CT094. doi:10.1158/1538-7445.AM2017-CT094

Collaboration


Dive into the Ray Valencia's collaboration.

Top Co-Authors

Avatar

Pauliina Luoto

Turku University Hospital

View shared research outputs
Top Co-Authors

Avatar

Sandra Borkowski

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Stuart Ince

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Juha O. Rinne

Turku University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Olof Solin

University of Helsinki

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arne Scholz

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Dominik Mumberg

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Franz von Nussbaum

Bayer HealthCare Pharmaceuticals

View shared research outputs
Researchain Logo
Decentralizing Knowledge