Network


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

Hotspot


Dive into the research topics where Kai H. Barck is active.

Publication


Featured researches published by Kai H. Barck.


Nature Chemical Biology | 2011

Specific Btk inhibition suppresses B cell– and myeloid cell–mediated arthritis

Julie Di Paolo; Tao Huang; Mercedesz Balazs; James Barbosa; Kai H. Barck; Brandon J. Bravo; Richard A. D. Carano; James W. Darrow; Douglas R. Davies; Laura DeForge; Lauri Diehl; Ronald E. Ferrando; Steven L. Gallion; Anthony M. Giannetti; Peter Gribling; Vincent Hurez; Sarah G. Hymowitz; Randall Jones; Jeffrey E. Kropf; Wyne P. Lee; Patricia Maciejewski; Scott Mitchell; Hong Rong; Bart L. Staker; J. Andrew Whitney; Sherry Yeh; Wendy B. Young; Christine Yu; Juan Zhang; Karin Reif

Brutons tyrosine kinase (Btk) is a therapeutic target for rheumatoid arthritis, but the cellular and molecular mechanisms by which Btk mediates inflammation are poorly understood. Here we describe the discovery of CGI1746, a small-molecule Btk inhibitor chemotype with a new binding mode that stabilizes an inactive nonphosphorylated enzyme conformation. CGI1746 has exquisite selectivity for Btk and inhibits both auto- and transphosphorylation steps necessary for enzyme activation. Using CGI1746, we demonstrate that Btk regulates inflammatory arthritis by two distinct mechanisms. CGI1746 blocks B cell receptor-dependent B cell proliferation and in prophylactic regimens reduces autoantibody levels in collagen-induced arthritis. In macrophages, Btk inhibition abolishes FcγRIII-induced TNFα, IL-1β and IL-6 production. Accordingly, in myeloid- and FcγR-dependent autoantibody-induced arthritis, CGI1746 decreases cytokine levels within joints and ameliorates disease. These results provide new understanding of the function of Btk in both B cell- or myeloid cell-driven disease processes and provide a compelling rationale for targeting Btk in rheumatoid arthritis.


Nature Medicine | 2009

Targeted depletion of lymphotoxin-alpha-expressing TH1 and TH17 cells inhibits autoimmune disease.

Eugene Y. Chiang; Ganesh Kolumam; Xin Yu; Michelle Francesco; Sinisa Ivelja; Ivan Peng; Peter Gribling; Jean Shu; Wyne P. Lee; Canio J. Refino; Mercedesz Balazs; Andres Paler-Martinez; Allen Nguyen; Judy Young; Kai H. Barck; Richard A. D. Carano; Ron Ferrando; Lauri Diehl; Devavani Chatterjea; Jane L. Grogan

Uncontrolled T helper type 1 (TH1) and TH17 cells are associated with autoimmune responses. We identify surface lymphotoxin-α (LT-α) as common to TH0, TH1 and TH17 cells and employ a unique strategy to target these subsets using a depleting monoclonal antibody (mAb) directed to surface LT-α. Depleting LT-α–specific mAb inhibited T cell–mediated models of delayed-type hypersensitivity and experimental autoimmune encephalomyelitis. In collagen-induced arthritis (CIA), preventive and therapeutic administration of LT-α–specific mAb inhibited disease, and immunoablated T cells expressing interleukin-17 (IL-17), interferon-γ and tumor necrosis factor-α (TNF-α), whereas decoy lymphotoxin-β receptor (LT-βR) fusion protein had no effect. A mutation in the Fc tail, rendering the antibody incapable of Fcγ receptor binding and antibody-dependent cellular cytotoxicity activity, abolished all in vivo effects. Efficacy in CIA was preceded by a loss of rheumatoid-associated cytokines IL-6, IL-1β and TNF-α within joints. These data indicate that depleting LT-α–expressing lymphocytes with LT-α–specific mAb may be beneficial in the treatment of autoimmune disease.


PLOS Pathogens | 2010

Host-Detrimental Role of Esx-1-Mediated Inflammasome Activation in Mycobacterial Infection

Fredric Carlsson; Janice Kim; Calin Dumitru; Kai H. Barck; Richard A. D. Carano; Mei Sun; Lauri Diehl; Eric J. Brown

The Esx-1 (type VII) secretion system is a major virulence determinant of pathogenic mycobacteria, including Mycobacterium marinum. However, the molecular events and host-pathogen interactions underlying Esx-1-mediated virulence in vivo remain unclear. Here we address this problem in a non-lethal mouse model of M. marinum infection that allows detailed quantitative analysis of disease progression. M. marinum established local infection in mouse tails, with Esx-1-dependent formation of caseating granulomas similar to those formed in human tuberculosis, and bone deterioration reminiscent of skeletal tuberculosis. Analysis of tails infected with wild type or Esx-1-deficient bacteria showed that Esx-1 enhanced generation of proinflammatory cytokines, including the secreted form of IL-1β, suggesting that Esx-1 promotes inflammasome activation in vivo. In vitro experiments indicated that Esx-1-dependent inflammasome activation required the host NLRP3 and ASC proteins. Infection of wild type and ASC-deficient mice demonstrated that Esx-1-dependent inflammasome activation exacerbated disease without restricting bacterial growth, indicating a host-detrimental role of this inflammatory pathway in mycobacterial infection. These findings define an immunoregulatory role for Esx-1 in a specific host-pathogen interaction in vivo, and indicate that the Esx-1 secretion system promotes disease and inflammation through its ability to activate the inflammasome.


Cancer Cell | 2013

A Rare Population of CD24+ITGB4+Notchhi Cells Drives Tumor Propagation in NSCLC and Requires Notch3 for Self-Renewal

Yanyan Zheng; Cecile de la Cruz; Leanne C. Sayles; Chris Alleyne-Chin; Dedeepya Vaka; Tim D. Knaak; Marty Bigos; Yue Xu; Chuong D. Hoang; Joseph B. Shrager; Hans Joerg Fehling; Dorothy French; William F. Forrest; Zhaoshi Jiang; Richard A. D. Carano; Kai H. Barck; Erica Jackson; E. Alejandro Sweet-Cordero

Sustained tumor progression has been attributed to a distinct population of tumor-propagating cells (TPCs). To identify TPCs relevant to lung cancer pathogenesis, we investigated functional heterogeneity in tumor cells isolated from Kras-driven mouse models of non-small-cell lung cancer (NSCLC). CD24(+)ITGB4(+)Notch(hi) cells are capable of propagating tumor growth in both a clonogenic and an orthotopic serial transplantation assay. While all four Notch receptors mark TPCs, Notch3 plays a nonredundant role in tumor cell propagation in two mouse models and in human NSCLC. The TPC population is enriched after chemotherapy, and the gene signature of mouse TPCs correlates with poor prognosis in human NSCLC. The role of Notch3 in tumor propagation may provide a therapeutic target for NSCLC.


Journal of Experimental Medicine | 2007

A novel inhibitor of the alternative pathway of complement reverses inflammation and bone destruction in experimental arthritis

Kenneth J. Katschke; Karim Y. Helmy; Micah Steffek; Hongkang Xi; JianPing Yin; Wyne P. Lee; Peter Gribling; Kai H. Barck; Richard A. D. Carano; Robin Taylor; Linda Rangell; Lauri Diehl; Philip E. Hass; Christian Wiesmann; Menno van Lookeren Campagne

Complement is an important component of the innate and adaptive immune response, yet complement split products generated through activation of each of the three complement pathways (classical, alternative, and lectin) can cause inflammation and tissue destruction. Previous studies have shown that complement activation through the alternative, but not classical, pathway is required to initiate antibody-induced arthritis in mice, but it is unclear if the alternative pathway (AP) plays a role in established disease. Previously, we have shown that human complement receptor of the immunoglobulin superfamily (CRIg) is a selective inhibitor of the AP of complement. Here, we present the crystal structure of murine CRIg and, using mutants, provide evidence that the structural requirements for inhibition of the AP are conserved in human and mouse. A soluble form of CRIg reversed inflammation and bone loss in two experimental models of arthritis by inhibiting the AP of complement in the joint. Our data indicate that the AP of complement is not only required for disease induction, but also disease progression. The extracellular domain of CRIg thus provides a novel tool to study the effects of inhibiting the AP of complement in established disease and constitutes a promising therapeutic with selectivity for a single complement pathway.


The Journal of Pathology | 2012

Anti-VEGF antibody therapy does not promote metastasis in genetically engineered mouse tumour models†

Mallika Singh; Suzana S. Couto; William F. Forrest; Anthony Lima; Jason H. Cheng; Rafael Molina; Jason E. Long; Patricia Hamilton; Angela McNutt; Ian Kasman; Michelle Nannini; Hani Bou Reslan; Tim C. Cao; Calvin C K Ho; Kai H. Barck; Richard A. D. Carano; Oded Foreman; Jeffrey Eastham-Anderson; Adrian M. Jubb; Napoleone Ferrara; Leisa Johnson

Resistance to anti‐angiogenic therapy can occur via several potential mechanisms. Unexpectedly, recent studies showed that short‐term inhibition of either VEGF or VEGFR enhanced tumour invasiveness and metastatic spread in preclinical models. In an effort to evaluate the translational relevance of these findings, we examined the consequences of long‐term anti‐VEGF monoclonal antibody therapy in several well‐validated genetically engineered mouse tumour models of either neuroendocrine or epithelial origin. Anti‐VEGF therapy decreased tumour burden and increased overall survival, either as a single agent or in combination with chemotherapy, in all four models examined. Importantly, neither short‐ nor long‐term exposure to anti‐VEGF therapy altered the incidence of metastasis in any of these autochthonous models, consistent with retrospective analyses of clinical trials. In contrast, we observed that sunitinib treatment recapitulated previously reported effects on tumour invasiveness and metastasis in a pancreatic neuroendocrine tumour (PNET) model. Consistent with these results, sunitinib treatment resulted in an up‐regulation of the hypoxia marker GLUT1 in PNETs, whereas anti‐VEGF did not. These results indicate that anti‐VEGF mediates anti‐tumour effects and therapeutic benefits without a paradoxical increase in metastasis. Moreover, these data underscore the concept that drugs targeting VEGF ligands and receptors may affect tumour metastasis in a context‐dependent manner and are mechanistically distinct from one another. Copyright


Clinical Cancer Research | 2012

Targeting the PI3K Pathway in the Brain - Efficacy of a PI3K Inhibitor Optimized to Cross the Blood-Brain Barrier

Laurent Salphati; Timothy P. Heffron; Bruno Alicke; Merry Nishimura; Kai H. Barck; Richard A. D. Carano; Jonathan Cheong; Kyle A. Edgar; Joan M. Greve; Samir Kharbanda; Hartmut Koeppen; Shari Lau; Leslie Lee; Jodie Pang; Emile Plise; Jenny L. Pokorny; Hani Bou Reslan; Jann N. Sarkaria; Jeffrey Wallin; Xiaolin Zhang; Stephen E. Gould; Alan G. Olivero; Heidi S. Phillips

Purpose: Glioblastoma (GBM), the most common primary brain tumor in adults, presents a high frequency of alteration in the PI3K pathway. Our objectives were to identify a dual PI3K/mTOR inhibitor optimized to cross the blood–brain barrier (BBB) and characterize its brain penetration, pathway modulation in the brain and efficacy in orthotopic xenograft models of GBM. Experimental Design: Physicochemical properties of PI3K inhibitors were optimized using in silico tools, leading to the identification of GNE-317. This compound was tested in cells overexpressing P-glycoprotein (P-gp) or breast cancer resistance protein (BCRP). Following administration to mice, GNE-317 plasma and brain concentrations were determined, and phosphorylated biomarkers (pAkt, p4EBP1, and pS6) were measured to assess PI3K pathway suppression in the brain. GNE-317 efficacy was evaluated in the U87, GS2, and GBM10 orthotopic models of GBM. Results: GNE-317 was identified as having physicochemical properties predictive of low efflux by P-gp and BCRP. Studies in transfected MDCK cells showed that GNE-317 was not a substrate of either transporter. GNE-317 markedly inhibited the PI3K pathway in mouse brain, causing 40% to 90% suppression of the pAkt and pS6 signals up to 6-hour postdose. GNE-317 was efficacious in the U87, GS2, and GBM10 orthotopic models, achieving tumor growth inhibition of 90% and 50%, and survival benefit, respectively. Conclusions: These results indicated that specific optimization of PI3K inhibitors to cross the BBB led to potent suppression of the PI3K pathway in healthy brain. The efficacy of GNE-317 in 3 intracranial models of GBM suggested that this compound could be effective in the treatment of GBM. Clin Cancer Res; 18(22); 6239–48. ©2012 AACR.


Developmental Biology | 2012

Chondroitin sulfate synthase 1 (Chsy1) is required for bone development and digit patterning.

Deanna Grant Wilson; Khanhky Phamluong; Wei Yu Lin; Kai H. Barck; Richard A. D. Carano; Lauri Diehl; Andrew S. Peterson; Flavius Martin; Mark Solloway

Joint and skeletal development is highly regulated by extracellular matrix (ECM) proteoglycans, of which chondroitin sulfate proteoglycans (CSPGs) are a major class. Despite the requirement of joint CSPGs for skeletal flexibility and structure, relatively little is understood regarding their role in establishing joint positioning or in modulating signaling and cell behavior during joint formation. Chondroitin sulfate synthase 1 (Chsy1) is one of a family of enzymes that catalyze the extension of chondroitin and dermatan sulfate glycosaminoglycans. Recently, human syndromic brachydactylies have been described to have loss-of-function mutations at the CHSY1 locus. In concordance with these observations, we demonstrate that mice lacking Chsy1, though viable, display chondrodysplasia and decreased bone density. Notably, Chsy1(-/-) mice show a profound limb patterning defect in which orthogonally shifted ectopic joints form in the distal digits. Associated with the digit-patterning defect is a shift in cell orientation and an imbalance in chondroitin sulfation. Our results place Chsy1 as an essential regulator of joint patterning and provide a mouse model of human brachydactylies caused by mutations in CHSY1.


Magnetic Resonance in Medicine | 2008

Quantification of viable tumor microvascular characteristics by multispectral analysis

Leanne Berry; Kai H. Barck; Mary Ann Go; Jed Ross; Xiumin Wu; Simon Williams; Alvin Gogineni; Mary J. Cole; Nicholas van Bruggen; Germaine Fuh; Frank Peale; Napoleone Ferrara; Sarajane Ross; Ralph Schwall; Richard A. D. Carano

Tumor heterogeneity complicates the quantification of tumor microvascular characteristics assessed by dynamic contrast‐enhanced MRI (DCE‐MRI). To address this issue a novel approach was developed that combines DCE‐MRI with diffusion‐based multispectral (MS) analysis to quantify the microvascular characteristics of specific tumor tissue populations. Diffusion‐based MS segmentation (feature space: apparent diffusion coefficient, T2 and proton density) was performed to identify tumor tissue populations and the DCE‐MRI characteristics were determined for each tissue class. The ability of this MS DCE‐MRI technique to detect microvascular changes due to treatment with an antibody (G6‐31) to vascular endothelial growth factor‐A (VEGF) was evaluated in a tumor xenograft mouse model. Anti‐VEGF treatment resulted in a significant reduction in Ktrans for the MS viable tumor tissue class (−0.0034 ± 0.0022 min−1, P < 0.01) at 24 hr posttreatment that differ significantly from the change observed in the control group (0.0002 ± 0.0025 min−1). Viable tumor Ktrans for the anti‐VEGF group was also reduced 62% relative to the pretreatment values (P < 0.01). Necrotic tissue classes were found to add only noise to DCE‐MRI estimates. This approach provides a means to measure physiological parameters within the viable tumor and address the issue of tumor heterogeneity that complicates DCE‐MRI analysis. Magn Reson Med, 2008.


Clinical Cancer Research | 2013

Development and Preclinical Characterization of a Humanized Antibody Targeting CXCL12

Cuiling Zhong; Jianyong Wang; Bing Li; Hong Xiang; Mark Ultsch; Mary Coons; Terence Wong; Nan Chiang; Suzy Clark; Robyn Clark; Leah Quintana; Peter Gribling; Eric Suto; Kai H. Barck; Racquel Corpuz; Jenny Yao; Rashi Takkar; Wyne P. Lee; Lisa A. Damico-Beyer; Richard D. Carano; Camellia W. Adams; Robert F. Kelley; Weiru Wang; Napoleone Ferrara

Purpose: Our goal was to develop a potent humanized antibody against mouse/human CXCL12. This report summarized its in vitro and in vivo activities. Experimental Design: Cell surface binding and cell migration assays were used to select neutralizing hamster antibodies, followed by testing in several animal models. Monoclonal antibody (mAb) 30D8 was selected for humanization based on its in vitro and in vivo activities. Results: 30D8, a hamster antibody against mouse and human CXCL12α, CXCL12β, and CXCL12γ, was shown to dose-dependently block CXCL12α binding to CXCR4 and CXCR7, and CXCL12α-induced Jurkat cell migration in vitro. Inhibition of primary tumor growth and/or metastasis was observed in several models. 30D8 alone significantly ameliorated arthritis in a mouse collagen-induced arthritis model (CIA). Combination with a TNF-α antagonist was additive. In addition, 30D8 inhibited 50% of laser-induced choroidal neovascularization (CNV) in mice. Humanized 30D8 (hu30D8) showed similar in vitro and in vivo activities as the parental hamster antibody. A crystal structure of the hu30D8 Fab/CXCL12α complex in combination with mutational analysis revealed a “hot spot” around residues Asn44/Asn45 of CXCL12α and part of the RFFESH region required for CXCL12α binding to CXCR4 and CXCR7. Finally, hu30D8 exhibited fast clearance in cynomolgus monkeys but not in rats. Conclusion: CXCL12 is an attractive target for treatment of cancer and inflammation-related diseases; hu30D8 is suitable for testing this hypothesis in humans. Clin Cancer Res; 19(16); 4433–45. ©2013 AACR.

Collaboration


Dive into the Kai H. Barck's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge