Network


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

Hotspot


Dive into the research topics where Marna Williams is active.

Publication


Featured researches published by Marna Williams.


Clinical Cancer Research | 2008

CS1, a Potential New Therapeutic Antibody Target for the Treatment of Multiple Myeloma

Eric D. Hsi; Roxanne Steinle; Balaji Balasa; Susann Szmania; Aparna Draksharapu; Benny Shum; Mahrukh Huseni; David B. Powers; Amulya Nanisetti; Yin Zhang; Audie Rice; Anne van Abbema; Melanie Wong; Gao Liu; Fenghuang Zhan; Myles Dillon; Shihao Chen; Susan Rhodes; Franklin Fuh; Naoya Tsurushita; Shankar Kumar; Vladimir Vexler; John D. Shaughnessy; Bart Barlogie; Frits van Rhee; Mohamad A. Hussein; Daniel E. H. Afar; Marna Williams

Purpose: We generated a humanized antibody, HuLuc63, which specifically targets CS1 (CCND3 subset 1, CRACC, and SLAMF7), a cell surface glycoprotein not previously associated with multiple myeloma. To explore the therapeutic potential of HuLuc63 in multiple myeloma, we examined in detail the expression profile of CS1, the binding properties of HuLuc63 to normal and malignant cells, and the antimyeloma activity of HuLuc63 in preclinical models. Experimental Design: CS1 was analyzed by gene expression profiling and immunohistochemistry of multiple myeloma samples and numerous normal tissues. HuLuc63-mediated antimyeloma activity was tested in vitro in antibody-dependent cellular cytotoxicity (ADCC) assays and in vivo using the human OPM2 xenograft model in mice. Results: CS1 mRNA was expressed in >90% of 532 multiple myeloma cases, regardless of cytogenetic abnormalities. Anti-CS1 antibody staining of tissues showed strong staining of myeloma cells in all plasmacytomas and bone marrow biopsies. Flow cytometric analysis of patient samples using HuLuc63 showed specific staining of CD138+ myeloma cells, natural killer (NK), NK-like T cells, and CD8+ T cells, with no binding detected on hematopoietic CD34+ stem cells. HuLuc63 exhibited significant in vitro ADCC using primary myeloma cells as targets and both allogeneic and autologous NK cells as effectors. HuLuc63 exerted significant in vivo antitumor activity, which depended on efficient Fc-CD16 interaction as well as the presence of NK cells in the mice. Conclusions: These results suggest that HuLuc63 eliminates myeloma cells, at least in part, via NK-mediated ADCC and shows the therapeutic potential of targeting CS1 with HuLuc63 for the treatment of multiple myeloma.


The Lancet | 2014

Etrolizumab as induction therapy for ulcerative colitis: a randomised, controlled, phase 2 trial

Severine Vermeire; Sharon O'Byrne; Mary E. Keir; Marna Williams; Timothy Lu; John C. Mansfield; Christopher A. Lamb; Brian G. Feagan; Julián Panés; Azucena Salas; Daniel C. Baumgart; Stefan Schreiber; Iris Dotan; William J. Sandborn; Gaik Wei Tew; Diana Luca; Meina T Tang; Lauri Diehl; Jeffrey Eastham-Anderson; Gert De Hertogh; Clémentine Perrier; Jackson G. Egen; John A. Kirby; Gert Van Assche; Paul Rutgeerts

BACKGROUND Etrolizumab is a humanised monoclonal antibody that selectively binds the β7 subunit of the heterodimeric integrins α4β7 and αEβ7. We aimed to assess etrolizumab in patients with moderately-to-severely active ulcerative colitis. METHODS In this double-blind, placebo-controlled, randomised, phase 2 study, patients with moderately-to-severely active ulcerative colitis who had not responded to conventional therapy were recruited from 40 referral centres in 11 countries. Eligible patients (aged 18-75 years; Mayo Clinic Score [MCS] of 5 of higher [or ≥6 in USA]; and disease extending 25 cm or more from anal verge) were randomised (1:1:1) to one of two dose levels of subcutaneous etrolizumab (100 mg at weeks 0, 4, and 8, with placebo at week 2; or 420 mg loading dose [LD] at week 0 followed by 300 mg at weeks 2, 4, and 8), or matching placebo. The primary endpoint was clinical remission at week 10, defined as MCS of 2 or less (with no individual subscore of >1), analysed in the modified intention-to-treat population (mITT; all randomly assigned patients who had received at least one dose of study drug, had at least one post-baseline disease-activity assessment, and had a centrally read screening endoscopic subscore of ≥2). This study is registered with ClinicalTrials.gov, number NCT01336465. FINDINGS Between Sept 2, 2011, and July 11, 2012, 124 patients were randomly assigned, of whom five had a endoscopic subscore of 0 or 1 and were excluded from the mITT population, leaving 39 patients in the etrolizumab 100 mg group, 39 in the etrolizumab 300 mg plus LD group, and 41 in the placebo group for the primary analyses. No patients in the placebo group had clinical remission at week 10, compared with eight (21% [95% CI 7-36]) patients in the etrolizumab 100 mg group (p=0·0040) and four (10% [0·2-24]) patients in the 300 mg plus LD group (p=0·048). Adverse events occurred in 25 (61%) of 41 patients in the etrolizumab 100 mg group (five [12%] of which were regarded as serious), 19 (48%) of 40 patients in the etrolizumab 300 mg plus LD group (two [5%] serious), and 31 (72%) of 43 patients in the placebo group (five [12%] serious). INTERPRETATION Etrolizumab was more likely to lead to clinical remission at week 10 than was placebo. Therefore, blockade of both α4β7 and αEβ7 might provide a unique therapeutic approach for the treatment of ulcerative colitis, and phase 3 studies have been planned. FUNDING Genentech.


Gut | 2013

A randomised phase I study of etrolizumab (rhuMAb β7) in moderate to severe ulcerative colitis

Paul Rutgeerts; Richard N. Fedorak; Daan W. Hommes; Andreas Sturm; Daniel C. Baumgart; Brian Bressler; Stefan Schreiber; John C. Mansfield; Marna Williams; Meina Tang; Jennifer Visich; Xiaohui Wei; Mary E. Keir; Diana Luca; Dimitri Danilenko; Jackson G. Egen; Sharon O'Byrne

Objective Etrolizumab (rhuMAb β7, anti-β7, PRO145223) is a humanised monoclonal antibody targeting the β7 subunit of the heterodimeric integrins α4β7 and αEβ7, which are implicated in leucocyte migration and retention in ulcerative colitis (UC). This randomised phase I study evaluated the safety and pharmacology of etrolizumab in patients with moderate to severe UC. Design In the single ascending dose (SAD) stage, etrolizumab (0.3, 1.0, 3.0, 10 mg/kg intravenous, 3.0 mg/kg subcutaneous (SC) or placebo) was administered 4:1 (n=25) in each cohort. In the multiple dose (MD) stage, new patients received monthly etrolizumab (0.5 mg/kg SC (n=4), 1.5 mg/kg SC (n=5), 3.0 mg/kg SC (n=4), 4.0 mg/kg intravenous (n=5)) or placebo (n=5). The pharmacokinetics was studied and Mayo Clinic Score evaluated at baseline, day 29 (SAD), and days 43 and 71 (MD). Results In the SAD stage, there were no dose limiting toxicities, infusion or injection site reactions. Two impaired wound healing serious adverse events occurred in two patients receiving etrolizumab. In the MD stage, there were no dose limiting toxicities, and no infusion or injection site reactions. Headache was the most common adverse event, occurring more often in etrolizumab patients. Antietrolizumab antibodies were detected in two subjects. The duration of β7 receptor full occupancy was dose related. A clinical response was observed in 12/18 patients, and clinical remission in 3/18 patients treated with etrolizumab in the MD stage, compared with 4/5 and 1/5 placebo patients, respectively. Conclusion Etrolizumab is well tolerated in moderate to severe UC. Further investigation is warranted.


Leukemia | 2010

Anti-CD22-MCC-DM1: An antibody-drug conjugate with a stable linker for the treatment of non-Hodgkin's lymphoma

Andrew G. Polson; Marna Williams; A. M. Gray; Reina N. Fuji; Kirsten Achilles Poon; Jacqueline McBride; Helga Raab; T. Januario; MaryAnn Go; Jeffrey Lau; Shang-Fan Yu; Changchun Du; Franklin Fuh; C. Tan; Y. Wu; W.-C. Liang; Saileta Prabhu; J.-P. Stephan; J.-A. Hongo; Randall Dere; Rong Deng; M. Cullen; R de Tute; Fiona Bennett; Andy C. Rawstron; Andrew Jack; Allen Ebens

Antibody-drug conjugates (ADCs) are potent cytotoxic drugs linked to antibodies through chemical linkers, and allow specific targeting of drugs to neoplastic cells. The expression of CD22 is limited to B-cells, and we show that CD22 is expressed on the vast majority of non-Hodgkins lymphomas (NHLs). An ideal target for an ADC for the treatment of NHL would have limited expression outside the B-cell compartment and be highly effective against NHL. We generated an ADC consisting of a humanized anti-CD22 antibody conjugated to the anti-mitotic agent maytansine with a stable linker (anti-CD22-MCC-DM1). Anti-CD22-MCC-DM1 was broadly effective in in vitro killing assays on NHL B-cell lines. We did not find a strong correlation between in vitro potency and CD22 surface expression, internalization of ADC or sensitivity to free drug. We show that anti-CD22-MCC-DM1 was capable of inducing complete tumor regression in NHL xenograft mouse models. Further, anti-CD22-MCC-DM1 was well tolerated in cynomolgus monkeys and substantially decreased circulating B-cells as well as follicle size and germinal center formation in lymphoid organs. These results suggest that anti-CD22-MCC-DM1 has an efficacy, safety and pharmacodynamic profile that support its use as a treatment for NHL.


British Journal of Pharmacology | 2011

A humanized monoclonal antibody targeting the β7 integrin selectively blocks intestinal homing of T lymphocytes

Eric Stefanich; Dimitry M. Danilenko; Hong Wang; Sharon O'Byrne; R Erickson; Thomas Gelzleichter; H Hiraragi; H Chiu; S Ivelja; S Jeet; S Gadkari; O Hwang; Franklin Fuh; Caroline Looney; Kathy Howell; V Albert; Mercedesz Balazs; C Refino; S Fong; S Iyer; Marna Williams

BACKGROUND AND PURPOSE rhuMAb Beta7 is a humanized anti‐human β7 monoclonal antibody currently in phase I in inflammatory bowel disease. rhuMAb Beta7 binds the β7 subunit of the integrins α4β7 and αEβ7, blocking interaction with their ligands. These integrins play key roles in immune cell homing to and retention in mucosal sites, and are associated with chronic inflammatory diseases of the gastrointestinal tract. The goal of this study was to evaluate the mucosal specificity of rhuMAb Beta7.


Molecular Cancer Therapeutics | 2013

DCDT2980S, an Anti-CD22-Monomethyl Auristatin E Antibody–Drug Conjugate, Is a Potential Treatment for Non-Hodgkin Lymphoma

Dongwei Li; Kirsten Achilles Poon; Shang-Fan Yu; Randall Dere; MaryAnn Go; Jeffrey Lau; Bing Zheng; Kristi Elkins; Dimitry M. Danilenko; Katherine R. Kozak; Pamela Chan; Josefa Chuh; Xiaoyan Shi; Denise Nazzal; Franklin Fuh; Jacqueline McBride; Vanitha Ramakrishnan; Ruth de Tute; Andy C. Rawstron; Andrew Jack; Rong Deng; Yu-Waye Chu; David Dornan; Marna Williams; William Ho; Allen Ebens; Saileta Prabhu; Andrew G. Polson

Antibody–drug conjugates (ADC), potent cytotoxic drugs linked to antibodies via chemical linkers, allow specific targeting of drugs to neoplastic cells. We have used this technology to develop the ADC DCDT2980S that targets CD22, an antigen with expression limited to B cells and the vast majority of non-Hodgkin lymphomas (NHL). DCDT2980S consists of a humanized anti-CD22 monoclonal IgG1 antibody with a potent microtubule-disrupting agent, monomethyl auristatin E (MMAE), linked to the reduced cysteines of the antibody via a protease cleavable linker, maleimidocaproyl-valine-citrulline-p-aminobenzoyloxycarbonyl (MC-vc-PAB). We describe the efficacy, safety, and pharmacokinetics of DCDT2980S in animal models to assess its potential as a therapeutic for the treatment of B-cell malignancies. We did not find a strong correlation between in vitro or in vivo efficacy and CD22 surface expression, nor a correlation of sensitivity to free drug and in vitro potency. We show that DCDT2980S was capable of inducing complete tumor regression in xenograft mouse models of NHL and can be more effective than rituximab plus combination chemotherapy at drug exposures that were well tolerated in cynomolgus monkeys. These results suggest that DCDT2980S has an efficacy, safety, and pharmacokinetics profile that support potential treatment of NHL. Mol Cancer Ther; 12(7); 1255–65. ©2013 AACR.


Arthritis Research & Therapy | 2012

Safety, pharmacokinetics, and biologic activity of pateclizumab, a novel monoclonal antibody targeting lymphotoxin α: results of a phase I randomized, placebo-controlled trial

Brinda Emu; Diana Luca; Carolyn Offutt; Jane L. Grogan; Bernadette Rojkovich; Marna Williams; Meina T Tang; Jim Xiao; John C. Davis

IntroductionPateclizumab (MLTA3698A) is a humanized mAb against lymphotoxin α (LTα), a transiently expressed cytokine on activated B and T cells (Th1, Th17), which are implicated in rheumatoid arthritis (RA) pathogenesis. This study was conducted to assess the safety, tolerability, < NOTE: For clarity and per AMA/S-W Style, please restore the use of Oxford/serial commas (ie: David likes vanilla, strawberry, and chocolate ice cream) throughout. and biologic activity of single and multiple doses of intravenous (IV) or subcutaneous (SC) pateclizumab in RA patients.MethodsThe single ascending dose (SAD) phase in patients with stable RA consisted of six cohorts (4:1 active:placebo at 0.3 mg/kg IV, 1.0 mg/kg IV, 1.0 mg/kg SC, 3.0 mg/kg IV, 3.0 mg/kg SC, and 5.0 mg/kg IV; n = 5/cohort). In the multiple ascending dose (MAD) phase, patients with prespecified RA disease activity received three doses of pateclizumab or placebo (4:1) every 2 weeks (1.0 mg/kg SC, n = 10; 3.0 mg/kg SC, n = 20; or 5.0 mg/kg IV, n = 5). Safety and tolerability were assessed throughout, and clinical activity was determined after three doses (Week 6).ResultsWe observed no serious adverse events (AEs) or dose-limiting toxicities, and the majority of AEs were mild to moderate. The pharmacokinetic profiles were linear, and clearance was independent of dose. Reductions in levels of serum CXCL13 were observed, supporting the biologic activity of pateclizumab on the LTα pathway. Patients receiving pateclizumab in the 3.0 mg/kg MAD group (3.0 mg/kg SC) demonstrated ACR20, ACR50, and ACR70 response rates at week 6 of 75%, 56% and 25%, respectively, compared with 57%, 29%, and 0% in the placebo group. The median Disease Activity Score in 28 joints, C-reactive protein, reduction was 28% for pateclizumab, versus 8.4% for placebo.ConclusionsPateclizumabwas generally well-tolerated in RA patients. Preliminary evidence of clinical activity was observed in active RA patients at the dose level targeted for clinical effect.


British Journal of Pharmacology | 2017

Anti-CD22 and anti-CD79b antibody-drug conjugates preferentially target proliferating B cells.

Franklin Fuh; Caroline Looney; Dongwei Li; Kirsten Achilles Poon; Randall Dere; Dimitry M. Danilenko; Jacqueline McBride; Chae Reed; Shan Chung; Bing Zheng; William Mathews; Andrew G. Polson; Saileta Prabhu; Marna Williams

CD22 and CD79b are cell‐surface receptors expressed on B‐cell‐derived malignancies such as non‐Hodgkins lymphoma (NHL). An anti‐mitotic agent, monomethyl auristatin E, was conjugated to anti‐CD22 and anti‐CD79b antibodies to develop target‐specific therapies for NHL. The mechanism of action (MOA) and pharmacological and pharmacokinetic (PK) profiles of these antibody‐drug conjugates (ADCs) were investigated in cynomolgus monkeys.


Archive | 2004

Therapeutic use of anti-CS1 antibodies

Marna Williams; J. Jun Tso; Nicolas F. Landolfi; Gao Liu


Molecular Cancer Therapeutics | 2009

In vivo effects of targeting CD79b with antibodies and antibody-drug conjugates

Bing Zheng; Reina N. Fuji; Kristi Elkins; Shang-Fan Yu; Franklin Fuh; Josefa Chuh; Christine Tan; Jo-Anne Hongo; Helga Raab; Katherine R. Kozak; Marna Williams; Elena McDorman; Dan L. Eaton; Allen Ebens; Andrew Polson

Collaboration


Dive into the Marna Williams's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gao Liu

University of Cincinnati

View shared research outputs
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