Network


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

Hotspot


Dive into the research topics where Roger Ulrich is active.

Publication


Featured researches published by Roger Ulrich.


Blood | 2011

CAL-101, a p110δ selective phosphatidylinositol-3-kinase inhibitor for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability

Brian Lannutti; Sarah Meadows; Sarah E.M. Herman; Adam Kashishian; Bart H. Steiner; Amy J. Johnson; John C. Byrd; Jeffrey W. Tyner; Marc Loriaux; Mike Deininger; Brian J. Druker; Kamal D. Puri; Roger Ulrich; Neill A. Giese

Phosphatidylinositol-3-kinase p110δ serves as a central integration point for signaling from cell surface receptors known to promote malignant B-cell proliferation and survival. This provides a rationale for the development of small molecule inhibitors that selectively target p110δ as a treatment approach for patients with B-cell malignancies. We thus identified 5-fluoro-3-phenyl-2-[(S)-1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one (CAL-101), a highly selective and potent p110δ small molecule inhibitor (half-maximal effective concentration [EC(50)] = 8nM). Using tumor cell lines and primary patient samples representing multiple B-cell malignancies, we have demonstrated that constitutive phosphatidylinositol-3-kinase pathway activation is p110δ-dependent. CAL-101 blocked constitutive phosphatidylinositol-3-kinase signaling, resulting in decreased phosphorylation of Akt and other downstream effectors, an increase in poly(ADP-ribose) polymerase and caspase cleavage and an induction of apoptosis. These effects have been observed across a broad range of immature and mature B-cell malignancies, thereby providing a rationale for the ongoing clinical evaluation of CAL-101.


Blood | 2014

Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110δ, for relapsed/refractory chronic lymphocytic leukemia

Jennifer R. Brown; John C. Byrd; Steven Coutre; Don M. Benson; Ian W. Flinn; Nina D. Wagner-Johnston; Stephen E. Spurgeon; Brad S. Kahl; Celeste M. Bello; Heather K. Webb; Dave Johnson; Sissy Peterman; Daniel Li; Thomas M. Jahn; Brian Lannutti; Roger Ulrich; Albert S. Yu; Langdon L. Miller; Richard R. Furman

In a phase 1 trial, idelalisib (GS-1101, CAL-101), a selective inhibitor of the lipid kinase PI3Kδ, was evaluated in 54 patients with relapsed/refractory chronic lymphocytic leukemia (CLL) with adverse characteristics including bulky lymphadenopathy (80%), extensive prior therapy (median 5 [range 2-14] prior regimens), treatment-refractory disease (70%), unmutated IGHV (91%), and del17p and/or TP53 mutations (24%). Patients were treated at 6 dose levels of oral idelalisib (range 50-350 mg once or twice daily) and remained on continuous therapy while deriving clinical benefit. Idelalisib-mediated inhibition of PI3Kδ led to abrogation of Akt phosphorylation in patient CLL cells and significantly reduced serum levels of CLL-related chemokines. The most commonly observed grade ≥3 adverse events were pneumonia (20%), neutropenic fever (11%), and diarrhea (6%). Idelalisib treatment resulted in nodal responses in 81% of patients. The overall response rate was 72%, with 39% of patients meeting the criteria for partial response per IWCLL 2008 and 33% meeting the recently updated criteria of PR with treatment-induced lymphocytosis.(1,2) The median progression-free survival for all patients was 15.8 months. This study demonstrates the clinical utility of inhibiting the PI3Kδ pathway with idelalisib. Our findings support the further development of idelalisib in patients with CLL. These trials were registered at clinicaltrials.gov as #NCT00710528 and #NCT01090414.


Blood | 2014

Idelalisib, a selective inhibitor of phosphatidylinositol 3-kinase-δ, as therapy for previously treated indolent non-Hodgkin lymphoma

Ian W. Flinn; Brad S. Kahl; John P. Leonard; Richard R. Furman; Jennifer R. Brown; John C. Byrd; Nina D. Wagner-Johnston; Steven Coutre; Don M. Benson; Sissy Peterman; Yoonjin Cho; Heather K. Webb; David Michael Johnson; Albert S. Yu; Roger Ulrich; Wayne R. Godfrey; Langdon L. Miller; Stephen E. Spurgeon

Idelalisib (GS-1101, CAL-101), an oral inhibitor of phosphatidylinositol 3-kinase-δ, was evaluated in a phase I study in 64 patients with relapsed indolent non-Hodgkin lymphoma (iNHL). Patients had a median (range) age of 64 (32-91) years, 34 (53%) had bulky disease (≥1 lymph nodes ≥5 cm), and 37 (58%) had refractory disease. Patients had received a median (range) of 4 (1-10) prior therapies. Eight dose regimens of idelalisib were evaluated; idelalisib was taken once or twice daily continuously at doses ranging from 50 to 350 mg. After 48 weeks, patients still benefitting (n = 19; 30%) enrolled into an extension study. Adverse events (AEs) occurring in 20% or more patients (total%/grade ≥3%) included diarrhea (36/8), fatigue (36/3), nausea (25/3), rash (25/3), pyrexia (20/3), and chills (20/0). Laboratory abnormalities included neutropenia (44/23), anemia (31/5), thrombocytopenia (25/11), and serum transaminase elevations (48/25). Twelve (19%) patients discontinued therapy due to AEs. Idelalisib induced disease regression in 46/54 (85%) of evaluable patients achieving an overall response rate of 30/64 (47%), with 1 patient having a complete response (1.6%). Median duration of response was 18.4 months, median progression-free survival was 7.6 months. Idelalisib is well tolerated and active in heavily pretreated, relapsed/refractory patients with iNHL. These trials were registered at clinicaltrials.gov as NCT00710528 and NCT01090414.


Clinical Cancer Research | 2017

The Bruton's tyrosine kinase (BTK) inhibitor acalabrutinib demonstrates potent on-target effects and efficacy in two mouse models of chronic lymphocytic leukemia

Sarah E.M. Herman; Arnau Montraveta; Carsten U. Niemann; Helena Mora-Jensen; Michael Gulrajani; Fanny Krantz; Rose Mantel; Lisa L. Smith; Fabienne McClanahan; Bonnie K. Harrington; Dolors Colomer; Todd Covey; John C. Byrd; Raquel Izumi; Allard Kaptein; Roger Ulrich; Amy J. Johnson; Brian Lannutti; Adrian Wiestner; Jennifer A. Woyach

Purpose: Acalabrutinib (ACP-196) is a novel, potent, and highly selective Bruton tyrosine kinase (BTK) inhibitor, which binds covalently to Cys481 in the ATP-binding pocket of BTK. We sought to evaluate the antitumor effects of acalabrutinib treatment in two established mouse models of chronic lymphocytic leukemia (CLL). Experimental Design: Two distinct mouse models were used, the TCL1 adoptive transfer model where leukemic cells from Eμ-TCL1 transgenic mice are transplanted into C57BL/6 mice, and the human NSG primary CLL xenograft model. Mice received either vehicle or acalabrutinib formulated into the drinking water. Results: Utilizing biochemical assays, we demonstrate that acalabrutinib is a highly selective BTK inhibitor as compared with ibrutinib. In the human CLL NSG xenograft model, treatment with acalabrutinib demonstrated on-target effects, including decreased phosphorylation of PLCγ2, ERK, and significant inhibition of CLL cell proliferation. Furthermore, tumor burden in the spleen of the mice treated with acalabrutinib was significantly decreased compared with vehicle-treated mice. Similarly, in the TCL1 adoptive transfer model, decreased phosphorylation of BTK, PLCγ2, and S6 was observed. Most notably, treatment with acalabrutinib resulted in a significant increase in survival compared with mice receiving vehicle. Conclusions: Treatment with acalabrutinib potently inhibits BTK in vivo, leading to on-target decreases in the activation of key signaling molecules (including BTK, PLCγ2, S6, and ERK). In two complementary mouse models of CLL, acalabrutinib significantly reduced tumor burden and increased survival compared with vehicle treatment. Overall, acalabrutinib showed increased BTK selectivity compared with ibrutinib while demonstrating significant antitumor efficacy in vivo on par with ibrutinib. Clin Cancer Res; 23(11); 2831–41. ©2016 AACR.


Journal of Pharmacology and Experimental Therapeutics | 2017

Acalabrutinib (ACP-196): A Covalent Bruton Tyrosine Kinase Inhibitor with a Differentiated Selectivity and In Vivo Potency Profile

Tjeerd Barf; Todd Covey; Raquel Izumi; Bas van de Kar; Michael Gulrajani; Bart Van Lith; Maaike van Hoek; Edwin de Zwart; Diana Mittag; Dennis Demont; Saskia Verkaik; Fanny Krantz; Paul G. Pearson; Roger Ulrich; Allard Kaptein

Several small-molecule Bruton tyrosine kinase (BTK) inhibitors are in development for B cell malignancies and autoimmune disorders, each characterized by distinct potency and selectivity patterns. Herein we describe the pharmacologic characterization of BTK inhibitor acalabrutinib [compound 1, ACP-196 (4-[8-amino-3-[(2S)-1-but-2-ynoylpyrrolidin-2-yl]imidazo[1,5-a]pyrazin-1-yl]-N-(2-pyridyl)benzamide)]. Acalabrutinib possesses a reactive butynamide group that binds covalently to Cys481 in BTK. Relative to the other BTK inhibitors described here, the reduced intrinsic reactivity of acalabrutinib helps to limit inhibition of off-target kinases having cysteine-mediated covalent binding potential. Acalabrutinib demonstrated higher biochemical and cellular selectivity than ibrutinib and spebrutinib (compounds 2 and 3, respectively). Importantly, off-target kinases, such as epidermal growth factor receptor (EGFR) and interleukin 2-inducible T cell kinase (ITK), were not inhibited. Determination of the inhibitory potential of anti-immunoglobulin M–induced CD69 expression in human peripheral blood mononuclear cells and whole blood demonstrated that acalabrutinib is a potent functional BTK inhibitor. In vivo evaluation in mice revealed that acalabrutinib is more potent than ibrutinib and spebrutinib. Preclinical and clinical studies showed that the level and duration of BTK occupancy correlates with in vivo efficacy. Evaluation of the pharmacokinetic properties of acalabrutinib in healthy adult volunteers demonstrated rapid absorption and fast elimination. In these healthy individuals, a single oral dose of 100 mg showed approximately 99% median target coverage at 3 and 12 hours and around 90% at 24 hours in peripheral B cells. In conclusion, acalabrutinib is a BTK inhibitor with key pharmacologic differentiators versus ibrutinib and spebrutinib and is currently being evaluated in clinical trials.


Clinical Cancer Research | 2017

Combined BTK and PI3Kδ Inhibition with Acalabrutinib and ACP-319 Improves Survival and Tumor Control in CLL Mouse Model

Carsten U. Niemann; Helena Mora-Jensen; Eman L. Dadashian; Fanny Krantz; Todd Covey; Shih-Shih Chen; Nicholas Chiorazzi; Raquel Izumi; Roger Ulrich; Brian Lannutti; Adrian Wiestner; Sarah E.M. Herman

Purpose: Targeting the B-cell receptor (BCR) pathway with inhibitors of Bruton tyrosine kinase (BTK) and PI3Kδ is highly effective for the treatment of chronic lymphocytic leukemia (CLL). However, deep remissions are uncommon, and drug resistance with single-agent therapy can occur. In vitro studies support the effectiveness of combing PI3Kδ and BTK inhibitors. Experimental Design: As CLL proliferation and survival depends on the microenvironment, we used murine models to assess the efficacy of the BTK inhibitor acalabrutinib combined with the PI3Kδ inhibitor ACP-319 in vivo. We compared single-agent with combination therapy in TCL1-192 cell–injected mice, a model of aggressive CLL. Results: We found significantly larger reductions in tumor burden in the peripheral blood and spleen of combination-treated mice. Although single-agent therapy improved survival compared with control mice by a few days, combination therapy extended survival by over 2 weeks compared with either single agent. The combination reduced tumor proliferation, NF-κB signaling, and expression of BCL-xL and MCL-1 more potently than single-agent therapy. Conclusions: The combination of acalabrutinib and ACP-319 was superior to single-agent treatment in a murine CLL model, warranting further investigation of this combination in clinical studies. Clin Cancer Res; 23(19); 5814–23. ©2017 AACR.


Cancer Research | 2015

Abstract 408: ACP-196, an orally bioavailable covalent selective inhibitor of Btk, modulates the innate tumor microenvironment, exhibits antitumor efficacy and enhances gemcitabine activity in pancreatic cancer

Brian Lannutti; Michael Gulrajani; Fanny Krantz; Elena Bibikova; Todd Covey; Katti Jessen; Wayne Rothbaum; David Michael Johnson; Roger Ulrich

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Pancreatic ductal adenocarcinoma exists in a complex desmoplastic microenvironment that provides stromal support for tumor growth and conceals the tumor from immune surveillance. Tumor-associated stroma comprises a mix of fibroblasts, immunosuppressive T regulatory cells (Tregs), myeloid suppressive monocytes (MDSCs) and tumor-associated macrophages (TAMs) that promote tumor growth and restrain immune-mediated tumor cell killing. The targeting of immune infiltrates may impair stromal support and enhance immune-mediated killing of pancreatic cancer cells. Brutons tyrosine kinase (Btk) is a nonreceptor enzyme in the Tec kinase family expressed among cells of hematopoietic origin including B cells, myeloid cells, mast cells and platelets, but not T cells, where it regulates multiple cellular processes. Here we describe an unexpected finding of ACP-196, a potent, novel, second generation Btk inhibitor with improved selectivity and target coverage that binds covalently to a cysteine residue (Cys481) in the front position of the ATP-binding pocket. In an orthotopic mouse model of pancreatic cancer, KPC derived pancreatic cancer cells (KrasG12D; Trp53R172H; Pdx1-Cre) were injected into the pancreases. Vehicle, single agent ACP-196 (15 mg/kg/BID, gavage), single agent gemcitabine (50 mg/kg, IV) and combination ACP-196 with gemcitabine were evaluated for efficacy. By 4 weeks of treatment, mice in the vehicle group showed signs of health deterioration and all mice were euthanized, tumors were collected and measured. Relative to the vehicle treatment, ACP-196 monotherapy resulted in a >2-fold reduction in tumor growth compared with less than a 2-fold reduction with gemcitabine alone. The combination of ACP-196 and gemcitabine resulted in a further reduction in tumor growth when compared to each single agent. Interestingly, analysis of tumor tissues showed that single agent ACP-196 inhibited immunosuppressive populations of TAMs and MDSCs. Surprisingly, Treg populations were also reduced with a robust expansion of CD8+ T cells in the tumors. None of these effects were observed with gemcitabine alone. Although Btk is not expressed in T cells, this finding maybe the result of inhibiting the MDSC and TAM populations within the tumor microenvironment, a mechanism of action which is currently under investigation. Taken together, these data identify Btk as a novel target for modulating tumor immune escape and suggest that pharmacologic targeting of suppressive myeloid cells by ACP-196 induces therapeutic benefit. ACP-196 is currently being evaluated in clinical trials including frontline and salvage pancreatic cancer. Citation Format: Brian J. Lannutti, Michael Gulrajani, Fanny Krantz, Elena Bibikova, Todd Covey, Katti Jessen, Wayne Rothbaum, David M. Johnson, Roger Ulrich. ACP-196, an orally bioavailable covalent selective inhibitor of Btk, modulates the innate tumor microenvironment, exhibits antitumor efficacy and enhances gemcitabine activity in pancreatic cancer. [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 408. doi:10.1158/1538-7445.AM2015-408


Molecular Cancer Therapeutics | 2009

Abstract B136: CAL‐120, a novel dual p110β/p110δ phosphatidylinositol‐3‐kinase (PI3K) inhibitor, attenuates PI3K signaling and demonstrates potent in vivo antitumor activity against solid tumors

Brian Lannutti; Adam Kashishian; Sarah Meadows; Bart H. Steiner; Lynn Ueno; Heather K. Webb; Kamal D. Puri; Roger Ulrich; Peter K. Vogt; Neill A. Giese

Phosphatidylinositide 3‐kinases (PI3K) are a family of lipid kinases that are involved in signaling events which control a diverse number of cellular processes. The activation of the PI3K pathway by cell surface receptors is directly mediated by the class I isoforms (α, β, δ, and γ). Aberrant regulation of the PI3K signaling pathway is frequently observed in a wide range of human malignancies including gain‐of‐function mutations in PI3K p110α isoform and/or loss‐of‐function mutations in PTEN phosphatase, which is responsible for down regulation of PI3K signaling. In purified enzyme assays, CAL‐120, a dual p110β/p110δ PI3K inhibitor, was inactive against class II and III PI3K family members,the PI3K‐related protein kinases mTOR and DNA‐PK as well as an additional ∼350 protein kinases in a genome wide screen. The ability of CAL‐120 to block oncogenic transformation mediated by individual PI3K class Ia isoforms was evaluated in primary cells using viral transduction. Foci formation mediated by p110α and p110δ was inhibited at 15–200 nM whereas little or no inhibition was observed against oncogenic forms of p110α at 20‐fold higher concentrations. To further demonstrate p110 isoform selectivity, AKT phosphorylation was induced in embryonic fibroblasts with PDGF or LPA that is mediated by p110α and p110β respectively. CAL‐120 inhibited the p110β response with an IC50 of 1.2 µM whereas the p110α IC50 was greater than 20 µM. The antitumor activity of CAL‐120 was evaluated in a panel of 23 human tumor cell lines representing different tissues and PI3K pathway mutations. Constitutive PI3K pathway activation as measured by AKT phosphorylation was observed in 50% of the cell lines and was highly correlated with PTEN mutations. In all cases, CAL‐120 blocked AKT phosphorylation at concentrations of 0.1–1.0 µM. In most cases inhibition of the phosphorylation of downstream effectors Akt, GSK‐3 , and S6 ribosomal protein was also observed over this concentration range. These effects of CAL‐120 on PI3K pathway inhibition correlated with G1 cell cycle arrest leading to inhibition of tumor cell proliferation and in a number of cases induced apoptosis. Of note was a lack of PI3K pathway activation in cell lines with K‐RAS mutations and their insensitivity to CAL‐120 treatment. In mice bearing xenografts of MCF‐7 breast adenocarcinoma (p110α mutation), PC‐3 prostate adenocarcinoma (PTEN deficient), or OVCAR‐3 ovarian adenocarcinoma (no pathway mutation), oral administration of CAL‐120 significantly inhibited tumor growth or caused tumor regression in each of these models. These data are the first to demonstrate that p110β/δ inhibition in the absence of effects on p110α is an effective strategy for the treatment of solid tumors. The antitumor activity was not restricted to cells with PTEN loss and was surprisingly observed even when p110α mutations were present. Collectively, these preclinical data support clinical evaluation of CAL‐120, an oral dual p110α/p110δ inhibitor, for the treatment of patients with solid tumors. (L. U. and P.K.V. are supported by grants from the National Cancer Institute. This is manuscript number 20362 of The Scripps Research Institute). Citation Information: Mol Cancer Ther 2009;8(12 Suppl):B136.


Blood | 2010

Clinical Safety and Activity In a Phase 1 Study of CAL-101, An Isoform-Selective Inhibitor of Phosphatidylinositol 3-Kinase P110δ, In Patients with Relapsed or Refractory Non-Hodgkin Lymphoma

Brad S. Kahl; John C. Byrd; Ian W. Flinn; Nina D. Wagner-Johnston; Stephen E. Spurgeon; Don M. Benson; Richard R. Furman; Jennifer R. Brown; Steven Coutre; Brian Lannutti; Neil A. Giese; Roger Ulrich; Heather K. Webb; Sissy Peterman; Leanne Holes; Albert S. Yu


Archive | 2009

Therapies for hematologic malignancies

W. Michael Gallatin; Roger Ulrich; Neill A. Giese

Collaboration


Dive into the Roger Ulrich's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ahmed Hamdy

Hamad Medical Corporation

View shared research outputs
Top Co-Authors

Avatar

John C. Byrd

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Neill A. Giese

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge