8 results on '"Annika Sundberg"'
Search Results
2. Data from A Phase I Dose-Escalation Study of Antibody BI-505 in Relapsed/Refractory Multiple Myeloma
- Author
-
Guido Tricot, Martine Poelman, Magnus Korsgren, Björn Frendeus, Maurizio Zangari, Stina Wichert, Jan Van Droogenbroeck, Ingrid Teige, Annika Sundberg, Yvonne Stenberg, Morten Mau-Sorensen, Elisabeth Sonesson, Morten Salomo, Fritz Offner, Hareth Nahi, Titti Martinsson Niskanen, Ashraf Badros, Peter Gimsing, and Markus Hansson
- Abstract
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.
- Published
- 2023
- Full Text
- View/download PDF
3. A platform for phenotypic discovery of therapeutic antibodies and targets applied on Chronic Lymphocytic Leukemia
- Author
-
Anne Ljungars, Linda Mårtensson, Markus Hansson, Mathilda Kovacek, Björn Frendéus, Ingrid Teige, Bo Jansson, Ulla-Carin Tornberg, V. Kuci Emruli, Jenny Mattsson, Mikael Mattsson, Annika Sundberg, Sara Ek, Nina Persson, Mats Ohlin, Gunnar Juliusson, and Mats Jerkeman
- Subjects
0301 basic medicine ,Cancer Research ,Phage display ,medicine.drug_class ,Chronic lymphocytic leukemia ,Brief Communication ,Monoclonal antibody ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,medicine ,Cytotoxicity ,biology ,business.industry ,CD23 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,biology.protein ,Antibody ,business - Abstract
Development of antibody drugs against novel targets and pathways offers great opportunities to improve current cancer treatment. We here describe a phenotypic discovery platform enabling efficient identification of therapeutic antibody-target combinations. The platform utilizes primary patient cells throughout the discovery process and includes methods for differential phage display cell panning, high-throughput cell-based specificity screening, phenotypic in vitro screening, target deconvolution, and confirmatory in vivo screening. In this study the platform was applied on cancer cells from patients with Chronic Lymphocytic Leukemia resulting in discovery of antibodies with improved cytotoxicity in vitro compared to the standard of care, the CD20-specific monoclonal antibody rituximab. Isolated antibodies were found to target six different receptors on Chronic Lymphocytic Leukemia cells; CD21, CD23, CD32, CD72, CD200, and HLA-DR of which CD32, CD200, and HLA-DR appeared as the most potent targets for antibody-based cytotoxicity treatment. Enhanced antibody efficacy was confirmed in vivo using a patient-derived xenograft model.
- Published
- 2018
- Full Text
- View/download PDF
4. A Phase I Dose-Escalation Study of Antibody BI-505 in Relapsed/Refractory Multiple Myeloma
- Author
-
Markus Hansson, Titti Martinsson Niskanen, Guido Tricot, Morten Mau-Sørensen, Morten Salomo, Ashraf Badros, Annika Sundberg, Fritz Offner, Peter Gimsing, Yvonne Stenberg, Stina Wichert, Björn Frendéus, Elisabeth Sonesson, Maurizio Zangari, Magnus Korsgren, Ingrid Teige, Hareth Nahi, Martine Poelman, and Jan Van Droogenbroeck
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Antineoplastic Agents ,Gastroenterology ,Siltuximab ,chemistry.chemical_compound ,Recurrence ,Internal medicine ,medicine ,Humans ,Elotuzumab ,Adverse effect ,Multiple myeloma ,Aged ,Bortezomib ,business.industry ,Antibodies, Monoclonal ,Middle Aged ,medicine.disease ,Surgery ,Treatment Outcome ,Oncology ,Tolerability ,chemistry ,Pharmacodynamics ,Female ,Premedication ,Drug Monitoring ,Multiple Myeloma ,business ,medicine.drug - Abstract
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.
- Published
- 2015
- Full Text
- View/download PDF
5. A Human ICAM-1 Antibody Isolated by a Function-First Approach Has Potent Macrophage-Dependent Antimyeloma Activity In Vivo
- Author
-
Niina Veitonmäki, Andrea Lundqvist, Markus Hansson, Linda Mårtensson, Annika Sundberg, Guido Tricot, Ming Zeng, Mathilda Kovacek, Ingrid Teige, Lena Danielsson, Björn Frendéus, Anne Ljungars, Fenghuang Zhan, Zhan-Chun Li, Ye Yang, Tobias Löfstedt, and Titti Martinsson-Niskanen
- Subjects
Adult ,Male ,Cancer Research ,Mice, SCID ,Pharmacology ,Epitope ,Epitopes ,Mice ,Peptide Library ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Receptor ,B cell ,Multiple myeloma ,Aged ,Aged, 80 and over ,B-Lymphocytes ,ICAM-1 ,biology ,Macrophages ,Receptors, IgG ,Antibodies, Monoclonal ,Cell Biology ,Middle Aged ,Intercellular Adhesion Molecule-1 ,medicine.disease ,Xenograft Model Antitumor Assays ,medicine.anatomical_structure ,Oncology ,Cell culture ,biology.protein ,Female ,Antibody ,Multiple Myeloma - Abstract
SummaryWe isolated a tumor B-cell-targeting antibody, BI-505, from a highly diversified human phage-antibody library, using a pioneering “function-first” approach involving screening for (1) specificity for a tumor B cell surface receptor, (2) induction of tumor programmed cell death, and (3) enhanced in vivo antitumor activity compared to currently used treatments. BI-505 bound to intercellular adhesion molecule-1, identifying a previously unrecognized role for this receptor as a therapeutic target in cancer. The BI-505 epitope was strongly expressed on the surface of multiple myeloma cells from both newly diagnosed and relapsed patients. BI-505 had potent macrophage-dependent antimyeloma activity and conferred enhanced survival compared to currently used treatments in advanced experimental models of multiple myeloma.
- Published
- 2013
- Full Text
- View/download PDF
6. Antagonistic human FcγRIIB (CD32B) antibodies have anti-tumor activity and overcome resistance to antibody therapy in vivo
- Author
-
Markus Hansson, C. Ian Mockridge, Robert J. Oldham, Björn Frendéus, Björn Nilsson, Neil Smyth, Ali Roghanian, Vallari Shah, Lekh N. Dahal, Jenny Mattson, Stephen A. Beers, Zhan-Chun Li, Mathilda Kovacek, Peter Johnson, Kerry L. Cox, Annika Sundberg, Linda Mårtensson, Ingrid Teige, H.T. Claude Chan, Khiyam Hussain, Anne Ljungars, Andrew T M Vaughan, Sonya James, Martin J. Glennie, Bhavwanti Sheth, J. Sjef Verbeek, Mats Jerkeman, Giusi Manfredi, Emily L Williams, Gunnar Juliusson, Andrew Davies, and Mark S. Cragg
- Subjects
Cancer Research ,Stromal cell ,media_common.quotation_subject ,Cell ,Biology ,Antibodies, Monoclonal, Murine-Derived ,Mice ,Immune system ,Neoplasms ,medicine ,Animals ,Humans ,Internalization ,media_common ,Antibody-dependent cell-mediated cytotoxicity ,Receptors, IgG ,Antibodies, Monoclonal ,Drug Synergism ,Cell Biology ,medicine.anatomical_structure ,Oncology ,Immunology ,Monoclonal ,biology.protein ,Rituximab ,Antibody ,medicine.drug - Abstract
SummaryTherapeutic antibodies have transformed cancer therapy, unlocking mechanisms of action by engaging the immune system. Unfortunately, cures rarely occur and patients display intrinsic or acquired resistance. Here, we demonstrate the therapeutic potential of targeting human (h) FcγRIIB (CD32B), a receptor implicated in immune cell desensitization and tumor cell resistance. FcγRIIB-blocking antibodies prevented internalization of the CD20-specific antibody rituximab, thereby maximizing cell surface accessibility and immune effector cell mediated antitumor activity. In hFcγRIIB-transgenic (Tg) mice, FcγRIIB-blocking antibodies effectively deleted target cells in combination with rituximab, and other therapeutic antibodies, from resistance-prone stromal compartments. Similar efficacy was seen in primary human tumor xenografts, including with cells from patients with relapsed/refractory disease. These data support the further development of hFcγRIIB antibodies for clinical assessment.
- Published
- 2014
7. Hydrography, near-bottom currents, and grazing impact of the filter-feeding ascidianCiona intestinalisin a Danish fjord
- Author
-
Hans Ulrik Riisgård, Carsten Jürgensen, and Annika Sundberg Jensen
- Subjects
geography ,geography.geographical_feature_category ,Stratification (water) ,Fjord ,Aquatic Science ,Biology ,biology.organism_classification ,Water column ,Oceanography ,Benthic zone ,Phytoplankton ,Ciona intestinalis ,Hydrography ,Cove - Abstract
Grazing exerted by a bed of filter-feeding ascidians, Ciona intestinalis, was examined in Kertinge Nor, the innermost shallow cove of a Danish fjord connected to the Great Belt. The aim was to study the dynamic interactions between the water column and benthic Filter feeders in order to account for the variability of phytoplankton biomass seen in the fjord. The moderate tidal- and density-driven near-bottom currents were both estimated and directly measured by means of an underwater video technique, and the downstream reduction in algal concentration over the ascidian bed documented and mathematically modelled. Relatively high algal concentrations developing in the surface layers during stratification may only be available to benthic grazers if a density-driven circulation, due to a salinity change in the Great Belt, carries these algae down to the bottom. Though wind is a crucial force for vertical mixing and transport of phytoplankton to the bottom, the frequent salinity changes in the Danish s...
- Published
- 1998
- Full Text
- View/download PDF
8. Hydrography, near-bottom currents, and grazing impact of the filter-feeding ascidianCiona intestinalisin a Danish fjord
- Author
-
Riisgård, Hans Ulrik, primary, Jensen, Annika Sundberg, additional, and Jürgensen, Carsten, additional
- Published
- 1998
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.