1
|
Jiang H, Acharya C, An G, Zhong M, Feng X, Wang L, Dasilva N, Song Z, Yang G, Adrian F, Qiu L, Richardson P, Munshi NC, Tai YT, Anderson KC. Retraction Note: SAR650984 directly induces multiple myeloma cell death via lysosomal-associated and apoptotic pathways, which is further enhanced by pomalidomide. Leukemia 2024:10.1038/s41375-024-02443-z. [PMID: 39438590 DOI: 10.1038/s41375-024-02443-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Affiliation(s)
- H Jiang
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Myeloma and Lymphoma Centre, Department of Hematology, Chang Zheng Hospital, The Second Military Medical University, Shanghai, China
| | - C Acharya
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - G An
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - M Zhong
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - X Feng
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - L Wang
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - N Dasilva
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Z Song
- Sanofi Oncology, Cambridge, MA, USA
| | - G Yang
- Sanofi Oncology, Cambridge, MA, USA
| | - F Adrian
- Sanofi Oncology, Cambridge, MA, USA
| | - L Qiu
- Institute of Hematology, CAMS & PUMC, Tianjin, China
| | - P Richardson
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - N C Munshi
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Y -T Tai
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - K C Anderson
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Fagnano E, Pendharkar S, Colton M, Jones PN, Sallan MC, Klymenko T, Braun A, Klein C, Honeychurch J, Cheadle EJ, Illidge TM. Stromal cell inhibition of anti-CD20 antibody mediated killing of B-cell malignancies. Front Cell Dev Biol 2023; 11:1270398. [PMID: 38020903 PMCID: PMC10646167 DOI: 10.3389/fcell.2023.1270398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: The glycoengineered type II anti-CD20 monoclonal antibody obinutuzumab has been licensed for treatment in follicular non-Hodgkin lymphoma and B-CLL following clinical trials demonstrating superior outcomes to standard of care treatment. However, ultimately many patients still relapse, highlighting the need to understand the mechanisms behind treatment failure to improve patient care. Resistance to chemotherapy is often caused by the ability of malignant B-cells to migrate to the bone marrow and home into the stromal layer. Therefore, this study aimed to investigate whether stromal cells were also able to inhibit type II anti-CD20 antibody mechanisms of action, contributing to resistance to therapy. Methods: A stromal-tumor co-culture was established in vitro between Raji or Daudi B-cell tumor cells and M210B4 stromal cells in 24 well plates. Results: Contact with stromal cells was able to protect tumor cells from obinutuzumab mediated programmed cell death (PCD), antibody dependent cellular phagocytosis and antibody dependent cellular cytotoxicity. Furthermore, such protection required direct contact between stroma and tumor cells. Stromal cells appeared to interfere with obinutuzumab mediated B-cell homotypic adhesion through inhibiting and reversing actin remodelling, potentially as a result of stromal-tumor cell contact leading to downregulation of CD20 on the surface of tumor cells. Further evidence for the potential role of CD20 downregulation comes through the reduction in surface CD20 expression and inhibition of obinutuzumab mediated PCD when tumor cells are treated with Ibrutinib in the presence of stromal cells. The proteomic analysis of tumor cells after contact with stromal cells led to the identification of a number of altered pathways including those involved in cell adhesion and the actin cytoskeleton and remodeling. Discussion: This work demonstrates that contact between tumor cells and stromal cells leads to inhibition of Obinutuzumab effector functions and has important implications for future therapies to improve outcomes to anti-CD20 antibodies. A deeper understanding of how anti-CD20 antibodies interact with stromal cells could prove a useful tool to define better strategies to target the micro-environment and ultimately improve patient outcomes in B-cell malignancies.
Collapse
Affiliation(s)
- Ester Fagnano
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Swati Pendharkar
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Madyson Colton
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Philip N. Jones
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Marta Crespi Sallan
- Centre for Haemato-Oncology, John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Tetyana Klymenko
- Department of Biosciences and Chemistry, Sheffield Hallam University, Sheffield, United Kingdom
| | - Andrejs Braun
- Centre for Haemato-Oncology, John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Christian Klein
- Roche Innovation Center Zurch, Roche Glycart AG, Schlieren, Switzerland
| | - Jamie Honeychurch
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Eleanor J. Cheadle
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Timothy M. Illidge
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
3
|
Deng L, Xu G. Update on the Application of Monoclonal Antibody Therapy in Primary Membranous Nephropathy. Drugs 2023; 83:507-530. [PMID: 37017915 DOI: 10.1007/s40265-023-01855-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
When first introduced, rituximab (RTX), a chimeric anti-CD20 monoclonal antibody, brought about an alternative therapeutic paradigm for primary membranous nephropathy (PMN). Rituximab was shown to be effective and safe in PMN patients with kidney dysfunction, with. patients receiving second-line rituximab therapy achieving remission as effectively as those patients who had not previously received immunotherapy. No safety issues were reported. The B cell-driven protocol seems to be as efficient as the 375 mg/m2 × 4 regimen or 1 g × 2 regimen in achieving B cell depletion and remission, but patients with high M-type phospholipase A2 receptor (PLA2R) antibody levels may benefit from a higher dose of rituximab. While rituximab added another therapeutic option to the treatment regimen, it does have limitations as 20 to 40% of patients do not respond. Not all patients respond to RTX therapy for lymphoproliferative disorders either, therefore further novel anti-CD20 monoclonal antibodies have been developed and these may provide alternative therapeutic options for PMN. Ofatumumab, a fully human monoclonal antibody, specifically recognizes an epitope encompassing both the small and large extracellular loops of the CD20 molecule, resulting in increased complement-dependent cytotoxic activity. Ocrelizumab binds an alternative but overlapping epitope region to rituximab and displays enhanced antibody-dependent cellular cytotoxic (ADCC) activities. Obinutuzumab is designed to have a modified elbow-hinge amino acid sequence, leading to increased direct cell death induction and ADCC activities. In PMN clinical studies, ocrelizumab and obinutuzumab showed promising results, while ofatumumab displayed mixed results. However, there is a lack of randomized controlled trials with large samples, especially direct head-to-head comparisons. Alternative molecular mechanisms have been suggested in this context to explore novel therapeutic strategies. B cell activator-targeted, plasma cell-targeted and complement-directed treatments may lead to novel therapy paradigms for PMN. Exploratory strategies for the use of drugs with different mechanisms, such as a combination of rituximab and cyclophosphamide and a steroid, a combination of rituximab and a calcineurin inhibitor, may provide more rapid and efficient remission, but the combination of standard immunosuppression with rituximab could increase infection risk.
Collapse
Affiliation(s)
- Le Deng
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
4
|
ARHGAP-RhoA signaling provokes homotypic adhesion-triggered cell death of metastasized diffuse-type gastric cancer. Oncogene 2022; 41:4779-4794. [PMID: 36127398 DOI: 10.1038/s41388-022-02469-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/09/2022]
Abstract
Genetic alteration of Rho GTPase-activating proteins (ARHGAP) and GTPase RhoA is a hallmark of diffuse-type gastric cancer and elucidating its biological significance is critical to comprehensively understanding this malignancy. Here, we report that gene fusions of ARHGAP6/ARHGAP26 are frequent genetic events in peritoneally-metastasized gastric and pancreatic cancer. From the malignant ascites of patients, we established gastric cancer cell lines that spontaneously gain hotspot RHOA mutations or four different ARHGAP6/ARHGAP26 fusions. These alterations critically downregulate RhoA-ROCK-MLC2 signaling, which elicits cell death. Omics and functional analyses revealed that the downstream signaling initiates actin stress fibers and reinforces intercellular junctions via several types of catenin. E-cadherin-centered homotypic adhesion followed by lysosomal membrane permeabilization is a pivotal mechanism in cell death. These findings support the tumor-suppressive nature of ARHGAP-RhoA signaling and might indicate a new avenue of drug discovery against this refractory cancer.
Collapse
|
5
|
Panaampon J, Kariya R, Okada S. Efficacy and mechanism of the anti-CD38 monoclonal antibody Daratumumab against primary effusion lymphoma. Cancer Immunol Immunother 2021; 71:1017-1031. [PMID: 34545416 DOI: 10.1007/s00262-021-03054-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 09/06/2021] [Indexed: 02/08/2023]
Abstract
Primary effusion lymphoma (PEL) is a rare, aggressive B cell non-Hodgkin's lymphoma of the body cavities with malignant effusions. The prognosis is poor, and no optimal treatment has been established. CD38 is a type II transmembrane glycoprotein known to overexpress in multiple myeloma (MM). Daratumumab (DARA), a human CD38-targeting monoclonal antibody (mAb), is approved for MM treatment. In this study, we found expression of CD38 on PEL cells and assessed the anti-PEL activity of DARA. We found that both KHYG-1 and N6 (CD16-transfected KHYG-1) NK cell lines showed direct killing activity against PEL cells with induction of CD107a, and NK-mediated cytotoxicity by N6NK (CD16+) cells increased with DARA treatment. We confirmed direct NK activity and antibody-dependent cell cytotoxicity (ADCC) by expanded NK cells, indicating that DARA has high ADCC activity. We elucidated the antibody-dependent cell phagocytosis (ADCP) by using human monocyte-derived macrophages (MDMs) and mouse peritoneal macrophages. DARA also showed potent complement-dependent cytolysis (CDC) toward PEL. DARA also induced PEL cell death in the presence of a cross-linking antibody. Moreover, treatment with DARA inhibited tumor growth in a PEL xenograft mouse model. These results provide preclinical evidence that Ab targeting of CD38 could be an effective therapeutic strategy for the treatment of PEL.
Collapse
Affiliation(s)
- Jutatip Panaampon
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Ryusho Kariya
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan.
| |
Collapse
|
6
|
Sopp JM, Peters SJ, Rowley TF, Oldham RJ, James S, Mockridge I, French RR, Turner A, Beers SA, Humphreys DP, Cragg MS. On-target IgG hexamerisation driven by a C-terminal IgM tail-piece fusion variant confers augmented complement activation. Commun Biol 2021; 4:1031. [PMID: 34475514 PMCID: PMC8413284 DOI: 10.1038/s42003-021-02513-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 08/02/2021] [Indexed: 12/20/2022] Open
Abstract
The majority of depleting monoclonal antibody (mAb) drugs elicit responses via Fc-FcγR and Fc-C1q interactions. Optimal C1q interaction is achieved through hexameric Fc:Fc interactions at the target cell surface. Herein is described an approach to exploit the tailpiece of the naturally multimeric IgM to augment hexamerisation of IgG. Fusion of the C-terminal tailpiece of IgM promoted spontaneous hIgG hexamer formation, resulting in enhanced C1q recruitment and complement-dependent cytotoxicity (CDC) but with off-target complement activation and reduced in-vivo efficacy. Mutation of the penultimate tailpiece cysteine to serine (C575S) ablated spontaneous hexamer formation, but facilitated reversible hexamer formation after concentration in solution. C575S mutant tailpiece antibodies displayed increased complement activity only after target binding, in-line with the concept of 'on-target hexamerisation', whilst retaining efficient in-vivo efficacy and augmented target cell killing in the lymph node. Hence, C575S-tailpiece technology represents an alternative format for promoting on-target hexamerisation and enhanced CDC.
Collapse
Affiliation(s)
- Joshua M Sopp
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | | | - Robert J Oldham
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Sonya James
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Ian Mockridge
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Ruth R French
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Stephen A Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Mark S Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.
| |
Collapse
|
7
|
Nowicka M, Hilton LK, Ashton-Key M, Hargreaves CE, Lee C, Foxall R, Carter MJ, Beers SA, Potter KN, Bolen CR, Klein C, Knapp A, Mir F, Rose-Zerilli M, Burton C, Klapper W, Scott DW, Sehn LH, Vitolo U, Martelli M, Trneny M, Rushton CK, Slack GW, Farinha P, Strefford JC, Oestergaard MZ, Morin RD, Cragg MS. Prognostic significance of FCGR2B expression for the response of DLBCL patients to rituximab or obinutuzumab treatment. Blood Adv 2021; 5:2945-2957. [PMID: 34323958 PMCID: PMC8361458 DOI: 10.1182/bloodadvances.2021004770] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/08/2021] [Indexed: 01/16/2023] Open
Abstract
Fc γ receptor IIB (FcγRIIB) is an inhibitory molecule capable of reducing antibody immunotherapy efficacy. We hypothesized its expression could confer resistance in patients with diffuse large B-cell lymphoma (DLBCL) treated with anti-CD20 monoclonal antibody (mAb) chemoimmunotherapy, with outcomes varying depending on mAb (rituximab [R]/obinutuzumab [G]) because of different mechanisms of action. We evaluated correlates between FCGR2B messenger RNA and/or FcγRIIB protein expression and outcomes in 3 de novo DLBCL discovery cohorts treated with R plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) reported by Arthur, Schmitz, and Reddy, and R-CHOP/G-CHOP-treated patients in the GOYA trial (NCT01287741). In the discovery cohorts, higher FCGR2B expression was associated with significantly shorter progression-free survival (PFS; Arthur: hazard ratio [HR], 1.09; 95% confidence interval [CI], 1.01-1.19; P = .0360; Schmitz: HR, 1.13; 95% CI, 1.02-1.26; P = .0243). Similar results were observed in GOYA with R-CHOP (HR, 1.26; 95% CI, 1.00-1.58; P = .0455), but not G-CHOP (HR, 0.91; 95% CI, 0.69-1.20; P = .50). A nonsignificant trend that high FCGR2B expression favored G-CHOP over R-CHOP was observed (HR, 0.67; 95% CI, 0.44-1.02; P = .0622); however, low FCGR2B expression favored R-CHOP (HR, 1.58; 95% CI, 1.00-2.50; P = .0503). In Arthur and GOYA, FCGR2B expression was associated with tumor FcγRIIB expression; correlating with shorter PFS for R-CHOP (HR, 2.17; 95% CI, 1.04-4.50; P = .0378), but not G-CHOP (HR, 1.37; 95% CI, 0.66-2.87; P = .3997). This effect was independent of established prognostic biomarkers. High FcγRIIB/FCGR2B expression has prognostic value in R-treated patients with DLBCL and may confer differential responsiveness to R-CHOP/G-CHOP.
Collapse
Affiliation(s)
| | - Laura K Hilton
- BC Cancer Centre for Lymphoid Cancer, Vancouver, BC, Canada
- Canada's Michael Smith Genome Sciences Centre, Vancouver, BC, Canada
| | - Margaret Ashton-Key
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
- Southampton University Hospitals NHS Foundation Trust, Southampton, United Kingdom
| | - Chantal E Hargreaves
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Chern Lee
- Southampton University Hospitals NHS Foundation Trust, Southampton, United Kingdom
| | - Russell Foxall
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton, Faculty of Medicine, Southampton, United Kingdom
| | - Matthew J Carter
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton, Faculty of Medicine, Southampton, United Kingdom
| | - Stephen A Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton, Faculty of Medicine, Southampton, United Kingdom
| | - Kathleen N Potter
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | | | | | | | - Farheen Mir
- Royal Marsden Hospital, Sutton, United Kingdom
| | - Matthew Rose-Zerilli
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Cathy Burton
- St James's Institute of Oncology, Leeds, United Kingdom
| | - Wolfram Klapper
- Department of Hematopathology, University of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - David W Scott
- BC Cancer Centre for Lymphoid Cancer, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Laurie H Sehn
- BC Cancer Centre for Lymphoid Cancer, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Umberto Vitolo
- Multidisciplinary Oncology Outpatient Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Maurizio Martelli
- Department of Translational and Precision Medicine, Hematology, Sapienza University, Rome, Italy
| | - Marek Trneny
- 1st Medical Faculty, Charles University, Prague, Czech Republic; and
| | - Christopher K Rushton
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada; and
| | - Graham W Slack
- BC Cancer Centre for Lymphoid Cancer, Vancouver, BC, Canada
| | - Pedro Farinha
- BC Cancer Centre for Lymphoid Cancer, Vancouver, BC, Canada
| | - Jonathan C Strefford
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | | | - Ryan D Morin
- Canada's Michael Smith Genome Sciences Centre, Vancouver, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada; and
| | - Mark S Cragg
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton, Faculty of Medicine, Southampton, United Kingdom
| |
Collapse
|
8
|
Bondza S, Ten Broeke T, Nestor M, Leusen JHW, Buijs J. Bivalent binding on cells varies between anti-CD20 antibodies and is dose-dependent. MAbs 2021; 12:1792673. [PMID: 32744151 PMCID: PMC7531561 DOI: 10.1080/19420862.2020.1792673] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Based on their mechanism of action, two types of anti-CD20 antibodies are distinguished: Type I, which efficiently mediate complement-dependent cytotoxicity, and Type II, which instead are more efficient in inducing direct cell death. Several molecular characteristics of these antibodies have been suggested to underlie these different biological functions, one of these being the manner of binding to CD20 expressed on malignant B cells. However, the exact binding model on cells is unclear. In this study, the binding mechanism of the Type I therapeutic antibodies rituximab (RTX) and ofatumumab (OFA) and the Type II antibody obinutuzumab (OBI) were established by real-time interaction analysis on live cells. It was found that the degree of bivalent stabilization differed for the antibodies: OFA was stabilized the most, followed by RTX and then OBI, which had the least amount of bivalent stabilization. Bivalency inversely correlated with binding dynamics for the antibodies, with OBI displaying the most dynamic binding pattern, followed by RTX and OFA. For RTX and OBI, bivalency and binding dynamics were concentration dependent; at higher concentrations the interactions were more dynamic, whereas the percentage of antibodies that bound bivalent was less, resulting in concentration-dependent apparent affinities. This was barely noticeable for OFA, as almost all molecules bound bivalently at the tested concentrations. We conclude that the degree of bivalent binding positively correlates with the complement recruiting capacity of the investigated CD20 antibodies.
Collapse
Affiliation(s)
- Sina Bondza
- Department of Immunology, Genetics and Pathology, Uppsala University , Uppsala, Sweden.,Ridgeview Instruments AB , Uppsala, Sweden
| | - Toine Ten Broeke
- Center for Translational Immunology, University Medical Center Utrecht , Utrecht, The Netherlands.,Current: Department of Pathology, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Marika Nestor
- Department of Immunology, Genetics and Pathology, Uppsala University , Uppsala, Sweden
| | - Jeanette H W Leusen
- Center for Translational Immunology, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Jos Buijs
- Department of Immunology, Genetics and Pathology, Uppsala University , Uppsala, Sweden.,Ridgeview Instruments AB , Uppsala, Sweden
| |
Collapse
|
9
|
Lee DSW, Rojas OL, Gommerman JL. B cell depletion therapies in autoimmune disease: advances and mechanistic insights. Nat Rev Drug Discov 2021; 20:179-199. [PMID: 33324003 PMCID: PMC7737718 DOI: 10.1038/s41573-020-00092-2] [Citation(s) in RCA: 316] [Impact Index Per Article: 105.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 01/30/2023]
Abstract
In the past 15 years, B cells have been rediscovered to be not merely bystanders but rather active participants in autoimmune aetiology. This has been fuelled in part by the clinical success of B cell depletion therapies (BCDTs). Originally conceived as a method of eliminating cancerous B cells, BCDTs such as those targeting CD20, CD19 and BAFF are now used to treat autoimmune diseases, including systemic lupus erythematosus and multiple sclerosis. The use of BCDTs in autoimmune disease has led to some surprises. For example, although antibody-secreting plasma cells are thought to have a negative pathogenic role in autoimmune disease, BCDT, even when it controls the disease, has limited impact on these cells and on antibody levels. In this Review, we update our understanding of B cell biology, review the results of clinical trials using BCDT in autoimmune indications, discuss hypotheses for the mechanism of action of BCDT and speculate on evolving strategies for targeting B cells beyond depletion.
Collapse
Affiliation(s)
- Dennis S. W. Lee
- grid.17063.330000 0001 2157 2938Department of Immunology, University of Toronto, Toronto, ON Canada
| | - Olga L. Rojas
- grid.17063.330000 0001 2157 2938Department of Immunology, University of Toronto, Toronto, ON Canada
| | - Jennifer L. Gommerman
- grid.17063.330000 0001 2157 2938Department of Immunology, University of Toronto, Toronto, ON Canada
| |
Collapse
|
10
|
Crossland RE, Perutelli F, Bogunia-Kubik K, Mooney N, Milutin Gašperov N, Pučić-Baković M, Greinix H, Weber D, Holler E, Pulanić D, Wolff D, Dickinson AM, Inngjerdingen M, Grce M. Potential Novel Biomarkers in Chronic Graft-Versus-Host Disease. Front Immunol 2020; 11:602547. [PMID: 33424849 PMCID: PMC7786047 DOI: 10.3389/fimmu.2020.602547] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Prognostic, diagnostic or predictive biomarkers are urgently needed for assessment of chronic graft-versus-host disease (cGvHD), a major risk for patients undergoing allogeneic hematopoietic stem cell transplantation. The main goal of this review generated within the COST Action EUROGRAFT "Integrated European Network on Chronic Graft Versus Host Disease" was to identify potential novel biomarkers for cGvHD besides the widely accepted molecular and cellular biomarkers. Thus, the focus was on cellular biomarkers, alloantibodies, glycomics, endothelial derived particles, extracellular vesicles, microbiome, epigenetic and neurologic changes in cGvHD patients. Both host-reactive antibodies in general, and particularly alloantibodies have been associated with cGvHD and require further consideration. Glycans attached to IgG modulate its activity and represent a promising predictive and/or stratification biomarker for cGVHD. Furthermore, epigenetic changes such as microRNAs and DNA methylation represent potential biomarkers for monitoring cGvHD patients and novel targets for developing new treatment approaches. Finally, the microbiome likely affects the pathophysiology of cGvHD; bacterial strains as well as microbial metabolites could display potential biomarkers for dysbiosis and risk for the development of cGvHD. In summary, although there are no validated biomarkers currently available for clinical use to better inform on the diagnosis, prognosis or prediction of outcome for cGvHD, many novel sources of potential markers have shown promise and warrant further investigation using well characterized, multi-center patient cohorts.
Collapse
Affiliation(s)
- Rachel E. Crossland
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Francesca Perutelli
- Department of Molecular Biotechnology and Health Sciences, School of Medicine, University of Torino, Torino, Italy
| | - Katarzyna Bogunia-Kubik
- Department of Clinical Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Nuala Mooney
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapies, Hôpital Saint Louis, Paris, France
| | | | | | - Hildegard Greinix
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Daniela Weber
- Department of Internal Medicine III, Faculty of Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Ernst Holler
- Department of Internal Medicine III, Faculty of Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Dražen Pulanić
- Division of Hematology, Department of Internal Medicine, University Hospital Centre Zagreb, Medical School, University of Zagreb, Zagreb, Croatia
| | - Daniel Wolff
- Department of Internal Medicine III, Faculty of Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Anne M. Dickinson
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Marit Inngjerdingen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Magdalena Grce
- Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
11
|
Kozlova V, Ledererova A, Ladungova A, Peschelova H, Janovska P, Slusarczyk A, Domagala J, Kopcil P, Vakulova V, Oppelt J, Bryja V, Doubek M, Mayer J, Pospisilova S, Smida M. CD20 is dispensable for B-cell receptor signaling but is required for proper actin polymerization, adhesion and migration of malignant B cells. PLoS One 2020; 15:e0229170. [PMID: 32210425 PMCID: PMC7094844 DOI: 10.1371/journal.pone.0229170] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 01/31/2020] [Indexed: 11/19/2022] Open
Abstract
Surface protein CD20 serves as the critical target of immunotherapy in various B-cell malignancies for decades, however its biological function and regulation remain largely elusive. Better understanding of CD20 function may help to design improved rational therapies to prevent development of resistance. Using CRISPR/Cas9 technique, we have abrogated CD20 expression in five different malignant B-cell lines. We show that CD20 deletion has no effect upon B-cell receptor signaling or calcium flux. Also B-cell survival and proliferation is unaffected in the absence of CD20. On the contrary, we found a strong defect in actin cytoskeleton polymerization and, consequently, defective cell adhesion and migration in response to homeostatic chemokines SDF1α, CCL19 and CCL21. Mechanistically, we could identify a reduction in chemokine-triggered PYK2 activation, a calcium-activated signaling protein involved in activation of MAP kinases and cytoskeleton regulation. These cellular defects in consequence result in a severely disturbed homing of B cells in vivo.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- Antigens, CD20/genetics
- Antigens, CD20/metabolism
- Antigens, CD20/physiology
- B-Lymphocytes/pathology
- B-Lymphocytes/physiology
- Cell Adhesion/physiology
- Cell Line, Tumor
- Cell Movement/physiology
- Gene Knockdown Techniques
- Humans
- Leukemia, B-Cell/metabolism
- Leukemia, B-Cell/pathology
- Lymphoma, B-Cell/metabolism
- Lymphoma, B-Cell/pathology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Polymerization
- Protein Multimerization/physiology
- Receptors, Antigen, B-Cell/metabolism
- Signal Transduction/immunology
Collapse
Affiliation(s)
- Veronika Kozlova
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Hematology and Oncology, Medical Faculty of Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Aneta Ledererova
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Hematology and Oncology, Medical Faculty of Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Adriana Ladungova
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Helena Peschelova
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Pavlina Janovska
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | | | - Joanna Domagala
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Pavel Kopcil
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Viera Vakulova
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Jan Oppelt
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Vitezslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Michael Doubek
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Hematology and Oncology, Medical Faculty of Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Jiri Mayer
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Hematology and Oncology, Medical Faculty of Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Sarka Pospisilova
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Hematology and Oncology, Medical Faculty of Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Michal Smida
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Hematology and Oncology, Medical Faculty of Masaryk University and University Hospital Brno, Brno, Czech Republic
- * E-mail:
| |
Collapse
|
12
|
Luan C, Chen B. Clinical application of obinutuzumab for treating chronic lymphocytic leukemia. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:2899-2909. [PMID: 31692500 PMCID: PMC6707935 DOI: 10.2147/dddt.s212500] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/14/2019] [Indexed: 12/02/2022]
Abstract
Alkylators and nucleoside analogs were the main drugs for treatingchronic lymphoblastic leukemia (CLL), which have been replaced by monoclonal antibodies, such as rituximab in the past 10 years for refractory or relapsed CLL. The first-line immunochemotherapy regimen, rituximab combined with nucleoside analogs, significantly increased CLL patients’ first-reaction rate and improved progression-free survival. Despite the long-lasting remissions by the use of chemoimmunotherapy, most CLL patients will relapse eventually. The obinutuzumab (GA101), an updated CD20 antibody, that is thought to achieve a more durable response with unique molecular and functional characteristics. Obinutuzumab is a humanized, monoclonal type II CD20 antibody modified by glycoengineering. The glycoengineered Fc portion enhances the binding affinity to the FcγRIII receptor on immune effector cells, resulting in increased antibody-dependent cellular cytotoxicity and phagocytosis. In addition, the type II antibody binding characteristics of obinutuzumab to CD20 lead to an efficient induction of direct non-apoptotic cell death. This review summarizes the results of clinical studies using obinutuzumab and looks forward to its further application in treating CLL clinically.
Collapse
Affiliation(s)
- Chunyan Luan
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| |
Collapse
|
13
|
Aljabri A, Vijayan V, Stankov M, Nikolin C, Figueiredo C, Blasczyk R, Becker JU, Linkermann A, Immenschuh S. HLA class II antibodies induce necrotic cell death in human endothelial cells via a lysosomal membrane permeabilization-mediated pathway. Cell Death Dis 2019; 10:235. [PMID: 30850581 PMCID: PMC6408495 DOI: 10.1038/s41419-019-1319-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/30/2018] [Accepted: 12/06/2018] [Indexed: 12/16/2022]
Abstract
Antibody-mediated rejection (AMR) is the major cause of allograft loss after solid organ transplantation. Circulating donor-specific antibodies against human leukocyte antigen (HLA), in particular HLA class II antibodies are critical for the pathogenesis of AMR via interactions with endothelial cells (ECs). To investigate the effects of HLA class II antibody ligation to the graft endothelium, a model of HLA-DR antibody-dependent stimulation was utilized in primary human ECs. Antibody ligation of HLA class II molecules in interferon-γ-treated ECs caused necrotic cell death without complement via a pathway that was independent of apoptosis and necroptosis. HLA-DR-mediated cell death was blocked by specific neutralization of antibody ligation with recombinant HLA class II protein and by lentiviral knockdown of HLA-DR in ECs. Importantly, HLA class II-mediated cytotoxicity was also induced by relevant native allele-specific antibodies from human allosera. Necrosis of ECs in response to HLA-DR ligation was mediated via hyperactivation of lysosomes, lysosomal membrane permeabilization (LMP), and release of cathepsins. Notably, LMP was caused by reorganization of the actin cytoskeleton. This was indicated by the finding that LMP and actin stress fiber formation by HLA-DR antibodies were both downregulated by the actin polymerization inhibitor cytochalasin D and inhibition of Rho GTPases, respectively. Finally, HLA-DR-dependent actin stress fiber formation and LMP led to mitochondrial stress, which was revealed by decreased mitochondrial membrane potential and generation of reactive oxygen species in ECs. Taken together, ligation of HLA class II antibodies to ECs induces necrotic cell death independent of apoptosis and necroptosis via a LMP-mediated pathway. These findings may enable novel therapeutic approaches for the treatment of AMR in solid organ transplantation.
Collapse
Affiliation(s)
- Abid Aljabri
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany.,King Saud Medical City, Riyadh, Saudi Arabia
| | - Vijith Vijayan
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Metodi Stankov
- Department for Clinical Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Christoph Nikolin
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | | | - Rainer Blasczyk
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | | | - Andreas Linkermann
- Department of Internal Medicine III, Division of Nephrology, University Carl Gustav Carus, Dresden, Germany
| | - Stephan Immenschuh
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
14
|
Mature IgD low/- B cells maintain tolerance by promoting regulatory T cell homeostasis. Nat Commun 2019; 10:190. [PMID: 30643147 PMCID: PMC6331566 DOI: 10.1038/s41467-018-08122-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 12/19/2018] [Indexed: 12/31/2022] Open
Abstract
A number of different B cell subsets have been shown to exhibit regulatory activity using a variety of mechanisms to attenuate inflammatory diseases. Here we show, using anti-CD20-mediated partial B cell depletion in mice, that a population of mature B cells distinguishable by IgDlow/- expression maintains tolerance by, at least in part, promoting CD4+Foxp3+ regulatory T cell homeostatic expansion via glucocorticoid-induced tumor necrosis factor receptor ligand, or GITRL. Cell surface phenotyping, transcriptome analysis and developmental study data show that B cells expressing IgD at a low level (BDL) are a novel population of mature B cells that emerge in the spleen from the transitional-2 stage paralleling the differentiation of follicular B cells. The cell surface phenotype and regulatory function of BDL are highly suggestive that they are a new B cell subset. Human splenic and peripheral blood IgDlow/- B cells also exhibit BDL regulatory activity, rendering them of therapeutic interest.
Collapse
|
15
|
Typical and Atypical Inducers of Lysosomal Cell Death: A Promising Anticancer Strategy. Int J Mol Sci 2018; 19:ijms19082256. [PMID: 30071644 PMCID: PMC6121368 DOI: 10.3390/ijms19082256] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 07/30/2018] [Accepted: 07/30/2018] [Indexed: 12/22/2022] Open
Abstract
Lysosomes are conservative organelles with an indispensable role in cellular degradation and the recycling of macromolecules. However, in light of recent findings, it has emerged that the role of lysosomes in cancer cells extends far beyond cellular catabolism and includes a variety of cellular pathways, such as proliferation, metastatic potential, and drug resistance. It has been well described that malignant transformation leads to alterations in lysosomal structure and function, which, paradoxically, renders cancer cells more sensitive to lysosomal destabilization. Furthermore, lysosomes are implicated in the regulation and execution of cell death in response to diverse stimuli and it has been shown that lysosome-dependent cell death can be utilized to overcome apoptosis and drug resistance. Thus, the purpose of this review is to characterize the role of lysosome in cancer therapy and to describe how these organelles impact treatment resistance. We summarized the characteristics of typical inducers of lysosomal cell death, which exert its function primarily via alterations in the lysosomal compartment. The review also presents other anticancer agents with the predominant mechanism of action different from lysosomal destabilization, the activity of which is influenced by lysosomal signaling, including classical chemotherapeutics, kinase inhibitors, monoclonal antibodies, as well as photodynamic therapy.
Collapse
|
16
|
Pierpont TM, Limper CB, Richards KL. Past, Present, and Future of Rituximab-The World's First Oncology Monoclonal Antibody Therapy. Front Oncol 2018; 8:163. [PMID: 29915719 PMCID: PMC5994406 DOI: 10.3389/fonc.2018.00163] [Citation(s) in RCA: 213] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/30/2018] [Indexed: 12/13/2022] Open
Abstract
Rituximab is a chimeric mouse/human monoclonal antibody (mAb) therapy with binding specificity to CD20. It was the first therapeutic antibody approved for oncology patients and was the top-selling oncology drug for nearly a decade with sales reaching $8.58 billion in 2016. Since its initial approval in 1997, it has improved outcomes in all B-cell malignancies, including diffuse large B-cell lymphoma, follicular lymphoma, and chronic lymphocytic leukemia. Despite widespread use, most mechanistic data have been gathered from in vitro studies while the roles of the various response mechanisms in humans are still largely undetermined. Polymorphisms in Fc gamma receptor and complement protein genes have been implicated as potential predictors of differential response to rituximab, but have not yet shown sufficient influence to impact clinical decisions. Unlike most targeted therapies developed today, no known biomarkers to indicate target engagement/tumor response have been identified, aside from reduced tumor burden. The lack of companion biomarkers beyond CD20 itself has made it difficult to predict which patients will respond to any given anti-CD20 antibody. In the past decade, two new anti-CD20 antibodies have been approved: ofatumumab, which binds a distinct epitope of CD20, and obinutuzumab, a mAb derived from rituximab with modifications to the Fc portion and to its glycosylation. Both are fully humanized and have biological activity that is distinct from that of rituximab. In addition to these new anti-CD20 antibodies, another imminent change in targeted lymphoma treatment is the multitude of biosimilars that are becoming available as rituximab's patent expires. While the widespread use of rituximab itself will likely continue, its biosimilars will increase global access to the therapy. This review discusses current research into mechanisms and potential biomarkers of rituximab response, as well as its biosimilars and the newer CD20 binding mAb therapies. Increased ability to assess the effectiveness of rituximab in an individual patient, along with the availability of alternative anti-CD20 antibodies will likely lead to dramatic changes in how we use CD20 antibodies going forward.
Collapse
Affiliation(s)
- Timothy M. Pierpont
- Richards Laboratory, Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States
| | - Candice B. Limper
- Richards Laboratory, Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States
| | - Kristy L. Richards
- Richards Laboratory, Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States
- Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
17
|
Katikaneni DS, Jin L. B cell MHC class II signaling: A story of life and death. Hum Immunol 2018; 80:37-43. [PMID: 29715484 DOI: 10.1016/j.humimm.2018.04.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/08/2018] [Accepted: 04/25/2018] [Indexed: 01/17/2023]
Abstract
MHC class II regulates B cell activation, proliferation, and differentiation during cognate B cell-T cell interaction. This is, in part, due to the MHC class II signaling in B cells. Activation of MHC Class II in human B cells or "primed" murine B cells leads to tyrosine phosphorylation, calcium mobilization, AKT, ERK, JNK activation. In addition, crosslinking MHC class II with monoclonal Abs kill malignant human B cells. Several humanized anti-HLA-DR/MHC class II monoclonal Abs entered clinical trials for lymphoma/leukemia and MHC class II-expressing melanomas. Mechanistically, MHC class II is associated with a wealth of transmembrane proteins including the B cell-specific signaling proteins CD79a/b, CD19 and a group of four-transmembrane proteins including tetraspanins and the apoptotic protein MPYS/STING. Furthermore, MHC class II signals are compartmentalized in the tetraspanin-enriched microdomains. In this review, we discuss our current understanding of MHC class II signaling in B cells focusing on its physiological significance and the therapeutic potential.
Collapse
Affiliation(s)
- Divya Sai Katikaneni
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610, United States
| | - Lei Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610, United States.
| |
Collapse
|
18
|
Palazzo A, Herter S, Grosmaire L, Jones R, Frey CR, Limani F, Bacac M, Umana P, Oldham RJ, Marshall MJE, Cox KL, Turaj AH, Cragg MS, Klein C, Carter MJ, Tannheimer S. The PI3Kδ-Selective Inhibitor Idelalisib Minimally Interferes with Immune Effector Function Mediated by Rituximab or Obinutuzumab and Significantly Augments B Cell Depletion In Vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:2304-2312. [PMID: 29453281 PMCID: PMC5857647 DOI: 10.4049/jimmunol.1700323] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 01/22/2018] [Indexed: 12/23/2022]
Abstract
Idelalisib is a highly selective oral inhibitor of PI3Kδ indicated for the treatment of patients with relapsed chronic lymphocytic leukemia in combination with rituximab. Despite additive clinical effects, previous studies have paradoxically demonstrated that targeted therapies potentially negatively affect anti-CD20 mAb effector mechanisms. To address these potential effects, we investigated the impact of PI3Kδ inhibition by idelalisib on the effector mechanisms of rituximab and obinutuzumab. At clinically relevant concentrations, idelalisib minimally influenced rituximab- and obinutuzumab-mediated Ab-dependent cellular cytotoxicity and phagocytosis on human lymphoma cell lines, while maintaining the superiority of obinutuzumab-mediated Ab-dependent cellular cytotoxicity. Consistent with this, idelalisib did not influence obinutuzumab-mediated B cell depletion in whole-blood B cell-depletion assays. Further, idelalisib significantly enhanced obinutuzumab-mediated direct cell death of chronic lymphocytic leukemia cells. In murine systems, in vivo inhibition of PI3Kδ minimally interfered with maximal rituximab- or obinutuzumab-mediated depletion of leukemic targets. In addition, the duration of rituximab- and obinutuzumab-mediated depletion of leukemia cells was extended by combination with PI3Kδ inhibition. Collectively, these data demonstrate that PI3Kδ inhibition does not significantly affect the effector mechanisms induced by rituximab or obinutuzumab and provides an effective in vivo therapeutic combination. Therefore, combinations of obinutuzumab and idelalisib are currently being assessed in clinical studies.
Collapse
Affiliation(s)
| | - Sylvia Herter
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, CH-8952 Zurich, Switzerland; and
| | | | - Randy Jones
- Gilead Sciences, Inc., Foster City, CA 98102
| | | | - Florian Limani
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, CH-8952 Zurich, Switzerland; and
| | - Marina Bacac
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, CH-8952 Zurich, Switzerland; and
| | - Pablo Umana
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, CH-8952 Zurich, Switzerland; and
| | - Robert J Oldham
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Michael J E Marshall
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Kerry L Cox
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Anna H Turaj
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Mark S Cragg
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Christian Klein
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, CH-8952 Zurich, Switzerland; and
| | - Matthew J Carter
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | | |
Collapse
|
19
|
Ghosh S, Banerjee S. Mathematical modeling of cancer-immune system, considering the role of antibodies. Theory Biosci 2018; 137:67-78. [PMID: 29572780 DOI: 10.1007/s12064-018-0261-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/09/2018] [Indexed: 11/30/2022]
Abstract
A mathematical model for the quantitative analysis of cancer-immune interaction, considering the role of antibodies has been proposed in this paper. The model is based on the clinical evidence, which states that antibodies can directly kill cancerous cells (Ivano et al. in J Clin Investig 119(8):2143-2159, 2009). The existence of transcritical bifurcation, which has been proved using Sotomayor theorem, provides strong biological implications. Through numerical simulations, it has been illustrated that under certain therapy (like monoclonal antibody therapy), which is capable of altering the parameters of the system, cancer-free state can be obtained.
Collapse
Affiliation(s)
- Sumana Ghosh
- Department of Mathematics, Indian Institute of Technology Roorkee, Roorkee, Uttaranchal, 247667, India
| | - Sandip Banerjee
- Department of Mathematics, Indian Institute of Technology Roorkee, Roorkee, Uttaranchal, 247667, India.
| |
Collapse
|
20
|
Association of rituximab with graphene oxide confers direct cytotoxicity for CD20-positive lymphoma cells. Oncotarget 2017; 7:12806-22. [PMID: 26859679 PMCID: PMC4914323 DOI: 10.18632/oncotarget.7230] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 01/26/2016] [Indexed: 11/29/2022] Open
Abstract
Non-Hodgkin lymphoma (NHL) is one of the most common hematologic malignancies among adults for which the chimeric monoclonal anti-CD20 antibody (Ab) rituximab (RTX) is used as first-line therapy. As RTX itself is not directly cytotoxic but relies on host immune effector mechanisms or chemotherapeutic agents to attack target cells, its therapeutic capacity may become limited when host effector mechanisms are compromised. Currently, refractory disease and relapse with NHL are still common, highlighting the need for novel anti-CD20 antibody strategies with superior therapeutic efficacy over current protocols. We hypothesized that making RTX directly cytotoxic might improve the therapeutic efficacy. Graphene oxide (GO) has recently emerged as a highly attractive nanomaterial for biomedical applications; and several studies have reported cytotoxic effect of GO on benign and malignant cells in vitro. Herein, we report that RTX can be stably associated with GO, and that GO-associated RTX (RTX/GO) demonstrates remarkably high avidity for CD20. Binding of GO-associated RTX to CD20-positive lymphoma cells induces CD20 capping and target cell death through an actin dependent mechanism. In vivo, GO-associated RTX, but not free RTX, quickly eliminates high-grade lymphomas in the absence of host effector mechanisms in a xenograft lymphoma mouse model. Our findings represent the first demonstration of using GO-associated antibody as effective cytotoxic therapy for human B cell malignancies in the absence of chemotherapy, and these findings could have important clinical implications.
Collapse
|
21
|
Sarraf Yazdy M, Cheson BD. Impact of obinutuzumab alone and in combination for follicular lymphoma. BLOOD AND LYMPHATIC CANCER-TARGETS AND THERAPY 2017; 7:73-83. [PMID: 31360086 PMCID: PMC6467363 DOI: 10.2147/blctt.s114173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Although rituximab-based chemoimmunotherapy prolongs the survival of patients with follicular lymphoma (FL), this disease is considered incurable in most patients. Thus, new therapies are needed not only for those in the relapsed/refractory setting, but also for initial treatment. Obinutuzumab (G, GA101) is a third-generation, fully humanized type II glycoengineered, anti-CD20 monoclonal antibody that results in increased direct cell death and antibody-dependent, cell-mediated cytotoxicity/phagocytosis compared to rituximab. Obinutuzumab has significant antitumor activity when used alone or in combinations in untreated or relapsed refractory FL patients. Studies have demonstrated its ability to prolong progression-free survival and, in some cases, overall survival, and to eliminate minimal residual disease. Several ongoing trials are investigating combinations with chemotherapy, immunomodulators, targeted drugs, and immunotherapy agents. G is generally well tolerated, with associated adverse effects including infusion-related reactions, neutropenia, thrombocytopenia, and reactivation of hepatitis B virus. Future studies with this antibody should focus on identifying predictive markers and developing chemotherapy-free combinations that will improve the outcome of patients with FL.
Collapse
Affiliation(s)
- Maryam Sarraf Yazdy
- Division of Hematology-Oncology, Georgetown University Hospital, Lombardi Comprehensive Cancer Center, Washington, DC, USA,
| | - Bruce D Cheson
- Division of Hematology-Oncology, Georgetown University Hospital, Lombardi Comprehensive Cancer Center, Washington, DC, USA,
| |
Collapse
|
22
|
Bohra C, Sokol L, Dalia S. Progressive Multifocal Leukoencephalopathy and Monoclonal Antibodies: A Review. Cancer Control 2017; 24:1073274817729901. [PMID: 28975841 PMCID: PMC5937251 DOI: 10.1177/1073274817729901] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 07/03/2017] [Indexed: 11/27/2022] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is a viral infection predominantly seen in patients with HIV infection. However, with the increased use of monoclonal antibodies (MAB) for various lymphoproliferative disorders, we are now seeing this infection in non-HIV patients on drugs such as natalizumab, rituximab, and so on. The aim of this article is to review the relationship between the occurrence of PML and MAB used in the treatment of hematological malignancies and autoimmune diseases. Review of articles from PubMed-indexed journals which study PML in relation to the use of MAB. Relevant literature demonstrated an increased risk of reactivation of latent John Cunningham polyomavirus (JCV) resulting in development of PML in patients on long-term therapy with MAB. The highest incidence of 1 PML case per 1000 treated patients and 1 case per 32 000 was observed in patients treated with natalizumab and rituximab, respectively. Serological and polymerase chain reaction tests for the detection of JCV can be helpful in risk stratification of patients for the development of PML before and during therapy with MAB. Treatment with MAB can result in development of PML. Clinicians should include PML in differential diagnosis in patients treated with these agents if they manifest central nervous system symptoms.
Collapse
Affiliation(s)
- Chandrashekar Bohra
- Internal Medicine Program, University of South Florida, Tampa, FL, USA
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center &
Research Institute, Tampa, FL, USA
- Mercy Oncology and Hematology–Joplin, Joplin, MO, USA
| | - Lubomir Sokol
- Internal Medicine Program, University of South Florida, Tampa, FL, USA
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center &
Research Institute, Tampa, FL, USA
- Mercy Oncology and Hematology–Joplin, Joplin, MO, USA
| | - Samir Dalia
- Internal Medicine Program, University of South Florida, Tampa, FL, USA
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center &
Research Institute, Tampa, FL, USA
- Mercy Oncology and Hematology–Joplin, Joplin, MO, USA
| |
Collapse
|
23
|
Cleary KLS, Chan HTC, James S, Glennie MJ, Cragg MS. Antibody Distance from the Cell Membrane Regulates Antibody Effector Mechanisms. THE JOURNAL OF IMMUNOLOGY 2017; 198:3999-4011. [PMID: 28404636 DOI: 10.4049/jimmunol.1601473] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 03/16/2017] [Indexed: 01/04/2023]
Abstract
Immunotherapy using mAbs, such as rituximab, is an established means of treating hematological malignancies. Abs can elicit a number of mechanisms to delete target cells, including complement-dependent cytotoxicity, Ab-dependent cellular cytotoxicity, and Ab-dependent cellular phagocytosis. The inherent properties of the target molecule help to define which of these mechanisms are more important for efficacy. However, it is often unclear why mAb binding to different epitopes within the same target elicits different levels of therapeutic activity. To specifically address whether distance from the target cell membrane influences the aforementioned effector mechanisms, a panel of fusion proteins consisting of a CD20 or CD52 epitope attached to various CD137 scaffold molecules was generated. The CD137 scaffold was modified through the removal or addition of cysteine-rich extracellular domains to produce a panel of chimeric molecules that held the target epitope at different distances along the protein. It was shown that complement-dependent cytotoxicity and Ab-dependent cellular cytotoxicity favored a membrane-proximal epitope, whereas Ab-dependent cellular phagocytosis favored an epitope positioned further away. These findings were confirmed using reagents targeting the membrane-proximal or -distal domains of CD137 itself before investigating these properties in vivo, where a clear difference in the splenic clearance of transfected tumor cells was observed. Together, this work demonstrates how altering the position of the Ab epitope is able to change the effector mechanisms engaged and facilitates the selection of mAbs designed to delete target cells through specific effector mechanisms and provide more effective therapeutic agents.
Collapse
Affiliation(s)
- Kirstie L S Cleary
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - H T Claude Chan
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - Sonja James
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - Martin J Glennie
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - Mark S Cragg
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| |
Collapse
|
24
|
Feng JJ, Ontaneda D. Treating primary-progressive multiple sclerosis: potential of ocrelizumab and review of B-cell therapies. Degener Neurol Neuromuscul Dis 2017; 7:31-45. [PMID: 30050376 PMCID: PMC6053100 DOI: 10.2147/dnnd.s100096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multiple sclerosis (MS) therapy has evolved rapidly with an increased availability of several immunomodulating therapies over the past two decades. Disease-modifying therapies have proven to be effective in treating relapse-remitting MS (RRMS). However, clinical trials involving some of the same agents for secondary-progressive and primary-progressive MS (SPMS and PPMS) have been largely negative. The pathogenesis of progressive MS remains unclear, but B-cells may play a significant role in chronic compartmentalized inflammation, likely contributing to disease progression. Biologics targeted at B-cells, such as rituximab, are effective in treating RRMS. Ocrelizumab is a humanized monoclonal antibody to CD20+ B-cells that has shown positive results in PPMS with a significant reduction in disease progression. This review aims to discuss in detail the involvement of B-cells in MS pathogenesis, current progress of currently available and investigational biologics, with focus on ocrelizumab, and future prospects for B-cell therapy in PPMS.
Collapse
Affiliation(s)
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Cleveland Clinic, Cleveland, OH, USA,
| |
Collapse
|
25
|
Al-Sawaf O, Fischer K, Engelke A, Pflug N, Hallek M, Goede V. Obinutuzumab in chronic lymphocytic leukemia: design, development and place in therapy. Drug Des Devel Ther 2017; 11:295-304. [PMID: 28182141 PMCID: PMC5279834 DOI: 10.2147/dddt.s104869] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
For decades, treatment of chronic lymphocytic leukemia (CLL) has been based on chemotherapy. This changed when the first CD20 antibody rituximab was introduced. Since 2008, the combination of chemotherapy and CD20 antibodies has become the standard of care for most patients, and a significant fraction of patients had very long-lasting remissions after chemoimmunotherapy. Despite the improvement of response rates and overall survival (OS) by the use of chemoimmunotherapy, most CLL patients will relapse eventually. One approach to achieve more durable responses was the development of obinutuzumab (GA101), a new type of CD20 antibody that has unique molecular and functional characteristics. Obinutuzumab is a type II fully humanized CD20 antibody that binds to a partly different epitope of the CD20 protein than rituximab and due to its glycoengineered design induces greater antibody-dependent cell-mediated cytotoxicity (ADCC). Initial preclinical observations of a more effective B-cell depletion have been successfully reproduced in clinical trials with CLL patients. This review summarizes results of preclinical as well as clinical studies with obinutuzumab and provides an outlook on its future role in the therapy of CLL.
Collapse
Affiliation(s)
- Othman Al-Sawaf
- German CLL Study Group, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Kirsten Fischer
- German CLL Study Group, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Anja Engelke
- German CLL Study Group, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Natali Pflug
- German CLL Study Group, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Michael Hallek
- German CLL Study Group, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Valentin Goede
- German CLL Study Group, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
26
|
Zheng JY, Tan HL, Matsudaira PT, Choo A. Excess reactive oxygen species production mediates monoclonal antibody-induced human embryonic stem cell death via oncosis. Cell Death Differ 2017; 24:546-558. [PMID: 28106884 DOI: 10.1038/cdd.2016.164] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/03/2016] [Accepted: 11/25/2016] [Indexed: 12/29/2022] Open
Abstract
Antibody-mediated cell killing has significantly facilitated the elimination of undesired cells in therapeutic applications. Besides the well-known Fc-dependent mechanisms, pathways of antibody-induced apoptosis were also extensively studied. However, with fewer studies reporting the ability of antibodies to evoke an alternative form of programmed cell death, oncosis, the molecular mechanism of antibody-mediated oncosis remains underinvestigated. In this study, a monoclonal antibody (mAb), TAG-A1 (A1), was generated to selectively kill residual undifferentiated human embryonic stem cells (hESC) so as to prevent teratoma formation upon transplantation of hESC-derived products. We revealed that A1 induces hESC death via oncosis. Aided with high-resolution scanning electron microscopy (SEM), we uncovered nanoscale morphological changes in A1-induced hESC oncosis, as well as A1 distribution on hESC surface. A1 induces hESC oncosis via binding-initiated signaling cascade, most likely by ligating receptors on surface microvilli. The ability to evoke excess reactive oxygen species (ROS) production via the Nox2 isoform of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase is critical in the cell death pathway. Excess ROS production occurs downstream of microvilli degradation and homotypic adhesion, but upstream of actin reorganization, plasma membrane damage and mitochondrial membrane permeabilization. To our knowledge, this is the first mechanistic model of mAb-induced oncosis on hESC revealing a previously unrecognized role for NAPDH oxidase-derived ROS in mediating oncotic hESC death. These findings in the cell death pathway may potentially be exploited to improve the efficiency of A1 in eliminating undifferentiated hESC and to provide insights into the study of other mAb-induced cell death.
Collapse
Affiliation(s)
- Ji Yun Zheng
- Mechanobiology Institute (MBI), National University of Singapore (NUS), T-Lab, No. 10-01, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Heng Liang Tan
- Stem Cell 1 Group, Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, No. 06-01 Centros, Singapore 138668, Singapore
| | - Paul Thomas Matsudaira
- Mechanobiology Institute (MBI), National University of Singapore (NUS), T-Lab, No. 10-01, 5A Engineering Drive 1, Singapore 117411, Singapore.,Department of Biological Science, Faculty of Science, National University of Singapore (NUS), 14 Science Drive 4, Singapore 117543, Singapore
| | - Andre Choo
- Stem Cell 1 Group, Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, No. 06-01 Centros, Singapore 138668, Singapore.,Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore (NUS), 9 Engineering Drive 1, Singapore 117575, Singapore
| |
Collapse
|
27
|
Zhao T, Ren H, Wang X, Liu P, Yan F, Jiang W, Li Y, Li J, Gribben JG, Jia L, Hao J. Rituximab-induced HMGB1 release is associated with inhibition of STAT3 activity in human diffuse large B-cell lymphoma. Oncotarget 2016; 6:27816-31. [PMID: 26315113 PMCID: PMC4695028 DOI: 10.18632/oncotarget.4816] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 07/20/2015] [Indexed: 11/25/2022] Open
Abstract
Treatment with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) has greatly improved clinical outcomes in patients with diffuse large B-cell lymphoma (DLBCL) compared with CHOP. The mechanism of rituximab-induced cell death is poorly understood. We found that rituximab does not enhance the directly killing efficacy of CHOP, as tested on a panel of DLBCL cell lines. Rituximab induced a rapid release of HMGB1 (High mobility group protein B 1). This release is independent of cell death but significantly correlated with an inhibition on STAT3 activity. In the resting state, HMGB1 co-localizes and interacts with STAT3 in the nucleus of DLBCL cells. Treatment with rituximab breaks this binding and triggers HMGB1 release. Treatment with R-CHOP but not CHOP significantly increased plasma HMGB1 and decreased IL-10 concentrations in DLBCL patients compared with controls. The conditioned medium from rituximab-treated DLBCL cells is able to trigger dendritic cell maturation, phagocytosis, and IFN-g secretion by cytotoxic T cells. In conclusion, our results demonstrate that rituximab induces an inhibition on STAT3 activity, leading to increased HMGB1 release and decreased IL-10 secretion, which elicits immune responses, suggesting that indirect effects on the immune system rather than direct killing contribute to elimination of DLBCL.
Collapse
Affiliation(s)
- Tiansuo Zhao
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - He Ren
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Xiuchao Wang
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Pengfei Liu
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Fan Yan
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Wenna Jiang
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Yang Li
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Jing Li
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - John G Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Li Jia
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Jihui Hao
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
28
|
Li H, Zhang G, Jiang C, Zhang F, Ke C, Zhao H, Sun Y, Zhao M, Chen D, Zhu X, Zhang L, Li B, Dai J, Li W. Suppression of Rituximab-resistant B-cell lymphoma with a novel multi-component anti-CD20 mAb nanocluster. Oncotarget 2016; 6:24192-204. [PMID: 26284588 PMCID: PMC4695179 DOI: 10.18632/oncotarget.4206] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/18/2015] [Indexed: 01/02/2023] Open
Abstract
Although the anti-CD20 antibody Rituximab has revolutionized the treatment of Non-Hodgkin Lymphoma (NHL), resistance to treatment still existed. Thus, strategies for suppressing Rituximab-resistant NHLs are urgently needed. Here, an anti-CD20 nanocluster (ACNC) is successfully constructed from its type I and type II mAb (Rituximab and 11B8). These distinct anti-CD20 mAbs are mass grafted to a short chain polymer (polyethylenimine). Compared with parental Rituximab and 11B8, the ACNC had a reduced “off-rate”. Importantly, ACNC efficiently inhibited Rituximab-resistant lymphomas in both disseminated and localized human NHL xenograft models. Further results revealed that ACNC is significantly potent in inducing caspase-dependent apoptosis and lysosome-mediated programmed cell death (PCD). This may help explain why ACNC is effective in suppressing rituximab-resistant lymphoma while Rituximab and 11B8 are not. Additionally, ACNC experienced low clearance from peripheral blood and high intratumor accumulation. This improved pharmacokinetics is attributed to the antibody-antigen reaction (active targeting) and enhanced permeability and retention (ERP) effect (passive targeting). This study suggested that ACNC might be a promising therapeutic agent for treatment of rituximab-resistant lymphomas.
Collapse
Affiliation(s)
- Huafei Li
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - Ge Zhang
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - Cheng Jiang
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - Fulei Zhang
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - Changhong Ke
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - He Zhao
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - Yun Sun
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - Mengxin Zhao
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - Di Chen
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - Xiandi Zhu
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - Li Zhang
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - Bohua Li
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - Jianxin Dai
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China
| | - Wei Li
- International Joint Cancer Institute, the Second Military Medical University, Shanghai, China.,State Key Laboratory of Antibody Medicine and Targeting Therapy and Shanghai Key Laboratory of Cell Engineering, Shanghai, China.,PLA General Hospital Cancer Center, PLA Graduate School of Medicine, Beijing, China
| |
Collapse
|
29
|
Abstract
Obinutuzumab is a humanized, type II anti-CD20 monoclonal antibody designed for strong induction of direct cell death and antibody-dependent cell-mediated cytotoxicity. The Phase III GADOLIN trial tested the clinical efficacy of obinutuzumab plus bendamustine followed by obinutuzumab monotherapy in rituximab-refractory indolent non-Hodgkin lymphoma versus treatment with bendamustine alone. It demonstrated significantly longer progression-free survival for the obinutuzumab-containing regimen in this difficult to treat patient group. Based on the results of this trial, US FDA approval was most recently granted for obinutuzumab in the treatment of follicular lymphoma that has relapsed after or was refractory to a rituximab-containing regimen. This article summarizes the available data on chemistry, pharmacokinetics, clinical efficacy and safety of obinutuzumab in the treatment of indolent non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Jennifer Edelmann
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - John G Gribben
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| |
Collapse
|
30
|
Establishment of a Therapeutic Anti-Pan HLA-Class II Monoclonal Antibody That Directly Induces Lymphoma Cell Death via Large Pore Formation. PLoS One 2016; 11:e0150496. [PMID: 27028595 PMCID: PMC4814124 DOI: 10.1371/journal.pone.0150496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 02/15/2016] [Indexed: 02/04/2023] Open
Abstract
To develop a new therapeutic monoclonal Antibody (mAb) for Hodgkin lymphoma (HL), we immunized a BALB/c mouse with live HL cell lines, alternating between two HL cell lines. After hybridization, we screened the hybridoma clones by assessing direct cytotoxicity against a HL cell line not used for immunization. We developed this strategy for establishing mAb to reduce the risk of obtaining clonotypic mAb specific for single HL cell line. A newly established mouse anti-human mAb (4713) triggered cytoskeleton-dependent, but complement- and caspase-independent, cell death in HL cell lines, Burkitt lymphoma cell lines, and advanced adult T-cell leukemia cell lines. Intravenous injection of mAb 4713 in tumor-bearing SCID mice improved survival significantly. mAb 4713 was revealed to be a mouse anti-human pan-HLA class II mAb. Treatment with this mAb induced the formation of large pores on the surface of target lymphoma cells within 30 min. This finding suggests that the cell death process induced by this anti-pan HLA-class II mAb may involve the same death signals stimulated by a cytolytic anti-pan MHC class I mAb that also induces large pore formation. This multifaceted study supports the therapeutic potential of mAb 4713 for various forms of lymphoma.
Collapse
|
31
|
Zhang F, Yang J, Li H, Liu M, Zhang J, Zhao L, Wang L, LingHu R, Feng F, Gao X, Dong B, Liu X, Zi J, Zhang W, Hu Y, Pan J, Tian L, Hu Y, Han Z, Zhang H, Wang X, Zhao L. Combating rituximab resistance by inducing ceramide/lysosome-involved cell death through initiation of CD20-TNFR1 co-localization. Oncoimmunology 2016; 5:e1143995. [PMID: 27467962 DOI: 10.1080/2162402x.2016.1143995] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 12/31/2022] Open
Abstract
Despite the success of CD20 antibody rituximab in immunotherapy, acquired resistance is one of the prime obstacles for the successful treatment of B-cell malignancies. There is an urgent need to intensify efforts against resistance in cancer treatment. Growing evidence indicated that lysosomes may form an "Achilles heel" for cancer cells by sensitizing them to death pathways. Here, we uncover an important role of CD20 in initiation of ceramide/lysosomal membrane permeabilization (LMP)-mediated cell death, showing that colocalization of CD20-TNFR1 after type II CD20 antibody ligation can stimulate de novo ceramide synthesis by ceramide synthase and consequently induce remarkable lysosomal permeabilization (LMP) and lysosome-mediated cell death. Further studies show that the potent lysosome-mediated cell death induced by CD20 antibodies exhibits a profound killing effect against both rituximab-sensitive and -resistant (RR) lymphoma. Furthermore, engineering of rituximab by introducing a point mutation endows it with the ability to induce potent ceramide/LMP-mediated cell death in both RR lymphoma and primary B-cell malignancies from patients with rituximab-refractory, suggesting the potential clinical application to combat rituximab resistance.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Oncology, PLA General Hospital Cancer Center, PLA School of Medicine and Key Laboratory of Cell Engineering & Antibody, Beijing and Institute for Translational Medicine, Second Military Medical University , Shanghai, People's Republic of China
| | - Junlan Yang
- Department of Oncology, PLA General Hospital Cancer Center, PLA School of Medicine and Key Laboratory of Cell Engineering & Antibody, Beijing and Institute for Translational Medicine, Second Military Medical University , Shanghai, People's Republic of China
| | - Huafei Li
- Department of Oncology, PLA General Hospital Cancer Center, PLA School of Medicine and Key Laboratory of Cell Engineering & Antibody, Beijing and Institute for Translational Medicine, Second Military Medical University , Shanghai, People's Republic of China
| | - Moyan Liu
- Department of Nephrology, General Hospital of Jinan Military Command , Jinan, People's Republic of China
| | - Jie Zhang
- Nursing Department, PLA General Hospital, PLA School of Medicine , Beijing, People's Republic of China
| | - Lichao Zhao
- Medical Department, General Hospital of Jinan Military Command , Jinan, China
| | - Lingxiong Wang
- Department of Oncology, PLA General Hospital Cancer Center, PLA School of Medicine and Key Laboratory of Cell Engineering & Antibody, Beijing and Institute for Translational Medicine, Second Military Medical University , Shanghai, People's Republic of China
| | - RuiXia LingHu
- National Clinical Research Center for Normal Aging and Geriatric & Institute of Geriatric, PLA General Hospital and The Key Lab of Normal Aging and Geriatric , Beijing, People's Republic of China
| | - Fan Feng
- Department of Pharmacy, General Hospital of Shenyang Military Command , Shenyang, People's Republic of China
| | - Xudong Gao
- Department of Gastroenterology, PLA 302 Hospital , Beijing, People's Republic of China
| | - Biqin Dong
- Department of Physics, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education) , and Key Laboratory of Surface Physics, Fudan University , Shanghai, People's Republic of China
| | - Xiaohan Liu
- Department of Physics, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education) , and Key Laboratory of Surface Physics, Fudan University , Shanghai, People's Republic of China
| | - Jian Zi
- Department of Physics, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education) , and Key Laboratory of Surface Physics, Fudan University , Shanghai, People's Republic of China
| | - Weijing Zhang
- Department of Lymphoma, Affiliated Hospital of Academy of Military Medical Science , Beijing, People's Republic of China
| | - Yi Hu
- Department of Oncology, PLA General Hospital Cancer Center, PLA School of Medicine and Key Laboratory of Cell Engineering & Antibody, Beijing and Institute for Translational Medicine, Second Military Medical University , Shanghai, People's Republic of China
| | - Jingkun Pan
- National Clinical Research Center for Normal Aging and Geriatric & Institute of Geriatric, PLA General Hospital and The Key Lab of Normal Aging and Geriatric , Beijing, People's Republic of China
| | - Lei Tian
- National Clinical Research Center for Normal Aging and Geriatric & Institute of Geriatric, PLA General Hospital and The Key Lab of Normal Aging and Geriatric , Beijing, People's Republic of China
| | - Yazuo Hu
- National Clinical Research Center for Normal Aging and Geriatric & Institute of Geriatric, PLA General Hospital and The Key Lab of Normal Aging and Geriatric , Beijing, People's Republic of China
| | - Zhitao Han
- National Clinical Research Center for Normal Aging and Geriatric & Institute of Geriatric, PLA General Hospital and The Key Lab of Normal Aging and Geriatric , Beijing, People's Republic of China
| | - Honghong Zhang
- National Clinical Research Center for Normal Aging and Geriatric & Institute of Geriatric, PLA General Hospital and The Key Lab of Normal Aging and Geriatric , Beijing, People's Republic of China
| | - Xiaoning Wang
- National Clinical Research Center for Normal Aging and Geriatric & Institute of Geriatric, PLA General Hospital and The Key Lab of Normal Aging and Geriatric , Beijing, People's Republic of China
| | - Lei Zhao
- Department of Oncology, PLA General Hospital Cancer Center, PLA School of Medicine and Key Laboratory of Cell Engineering & Antibody, Beijing and Institute for Translational Medicine, Second Military Medical University, Shanghai, People's Republic of China; National Clinical Research Center for Normal Aging and Geriatric & Institute of Geriatric, PLA General Hospital and The Key Lab of Normal Aging and Geriatric, Beijing, People's Republic of China
| |
Collapse
|
32
|
Jiang H, Acharya C, An G, Zhong M, Feng X, Wang L, Dasilva N, Song Z, Yang G, Adrian F, Qiu L, Richardson P, Munshi NC, Tai YT, Anderson KC. SAR650984 directly induces multiple myeloma cell death via lysosomal-associated and apoptotic pathways, which is further enhanced by pomalidomide. Leukemia 2016; 30:399-408. [PMID: 26338273 DOI: 10.1038/leu.2015.240] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/20/2015] [Accepted: 08/21/2015] [Indexed: 12/22/2022]
Abstract
The anti-CD38 monoclonal antibody SAR650984 (SAR) is showing promising clinical activity in treatment of relapsed and refractory multiple myeloma (MM). Besides effector-mediated antibody-dependent cellular cytotoxicity and complement-mediated cytotoxicity, we here define molecular mechanisms of SAR-directed MM cell death and enhanced anti-MM activity triggered by SAR with Pomalidomide (Pom). Without Fc-cross-linking agents or effector cells, SAR specifically induces homotypic aggregation (HA)-associated cell death in MM cells dependent on the level of cell surface CD38 expression, actin cytoskeleton and membrane lipid raft. SAR and its F(ab)'2 fragments trigger caspase 3/7-dependent apoptosis in MM cells highly expressing CD38, even with p53 mutation. Importantly, SAR specifically induces lysosome-dependent cell death (LCD) by enlarging lysosomes and increasing lysosomal membrane permeabilization associated with leakage of cathepsin B and LAMP-1, regardless of the presence of interleukin-6 or bone marrow stromal cells. Conversely, the lysosomal vacuolar H+-ATPase inhibitor blocks SAR-induced LCD. SAR further upregulates reactive oxygen species. Pom enhances SAR-induced direct and indirect killing even in MM cells resistant to Pom/Len. Taken together, SAR is the first therapeutic monoclonal antibody mediating direct cytotoxicity against MM cells via multiple mechanisms of action. Our data show that Pom augments both direct and effector cell-mediated MM cytotoxicity of SAR, providing the framework for combination clinical trials.
Collapse
Affiliation(s)
- H Jiang
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Myeloma and Lymphoma Centre, Department of Hematology, Chang Zheng Hospital, The Second Military Medical University, Shanghai, China
| | - C Acharya
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - G An
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - M Zhong
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - X Feng
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - L Wang
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - N Dasilva
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Z Song
- Sanofi Oncology, Cambridge, MA, USA
| | - G Yang
- Sanofi Oncology, Cambridge, MA, USA
| | - F Adrian
- Sanofi Oncology, Cambridge, MA, USA
| | - L Qiu
- Institute of Hematology, CAMS & PUMC, Tianjin, China
| | - P Richardson
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - N C Munshi
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Y-T Tai
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - K C Anderson
- LeBow Institute for Myeloma Therapeutics and the Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
Könitzer JD, Sieron A, Wacker A, Enenkel B. Reformatting Rituximab into Human IgG2 and IgG4 Isotypes Dramatically Improves Apoptosis Induction In Vitro. PLoS One 2015; 10:e0145633. [PMID: 26713448 PMCID: PMC4694715 DOI: 10.1371/journal.pone.0145633] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/06/2015] [Indexed: 12/31/2022] Open
Abstract
The direct induction of cell death, or apoptosis, in target cells is one of the effector mechanisms for the anti CD20 antibody Rituximab. Here we provide evidence that Rituximab’s apoptotic ability is linked to the antibody IgG isotype. Reformatting Rituximab from the standard human IgG1 heavy chain into IgG2 or IgG4 boosted in vitro apoptosis induction in the Burkitt’s lymphoma B cell line Ramos five and four-fold respectively. The determinants for this behavior are located in the hinge region and CH1 domain of the heavy chain. By transplanting individual IgG2 or IgG4 specific amino acid residues onto otherwise IgG1 like backbones, thereby creating hybrid antibodies, the same enhancement of apoptosis induction could be achieved. The cysteines at position 131 of the CH1 domain and 219 in the hinge region, involved in IgG2 and IgG4 disulfide formation, were found to be of particular structural importance. Our data indicates that the hybrid antibodies possess a different CD20 binding mode than standard Rituximab, which appears to be key in enhancing apoptotic ability. The presented work opens up an interesting engineering route for enhancing the direct cytotoxic ability of therapeutic antibodies.
Collapse
Affiliation(s)
- Jennifer D. Könitzer
- Boehringer Ingelheim, Division Research Germany, Immune Modulation and Biotherapeutics Discovery, Biberach/Riß, Germany
- * E-mail:
| | - Annette Sieron
- Boehringer Ingelheim, Biopharma Operations Germany, Biberach/Riß, Germany
| | - Angelika Wacker
- Boehringer Ingelheim, Bioprocess and Pharmaceutical Development Germany, Biberach/Riß, Germany
| | - Barbara Enenkel
- Boehringer Ingelheim, Bioprocess and Pharmaceutical Development Germany, Biberach/Riß, Germany
| |
Collapse
|
34
|
Synergistic anti-tumor therapy by a comb-like multifunctional antibody nanoarray with exceptionally potent activity. Sci Rep 2015; 5:15712. [PMID: 26508306 PMCID: PMC4623742 DOI: 10.1038/srep15712] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 09/10/2015] [Indexed: 01/10/2023] Open
Abstract
Simultaneously blocking multiple mediators offers new hope for the treatment of complex diseases. However, the curative potential of current combination therapy by chronological administration of separate monoclonal antibodies (mAbs) or multi-specific mAbs is still moderate due to inconvenient manipulation, low cooperative effectors, poor pharmacokinetics and insufficient tumor accumulation. Here, we describe a facile strategy that arms distinct mAbs with cooperative effectors onto a long chain to form a multicomponent comb-like nano mAb. Unlike dissociative parental mAbs, the multifunctional mAb nanoarray (PL-RB) constructed from type I/II anti-CD20 mAbs shows good pharmacokinetics. This PL-RB simultaneously targets distinct epitopes on a single antigen (Ag) and neighboring Ags on different lymphocytes. This unique intra- and intercellular Ag cross-linking endows the multifunctional mAb nanoarray with potent apoptosis activity. The exceptional apoptosis, complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC) that are synchronously evoked by the nano PL-RB are further synergistically promoted via enhanced permeability and retention (EPR), which resulted in high intratumor accumulation and excellent anti-lymphoma efficiency.
Collapse
|
35
|
Hill BT, Kalaycio M. Profile of obinutuzumab for the treatment of patients with previously untreated chronic lymphocytic leukemia. Onco Targets Ther 2015; 8:2391-7. [PMID: 26366093 PMCID: PMC4562745 DOI: 10.2147/ott.s68770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a hematologic malignancy derived from a clonal population of mature B-lymphocytes characterized by relatively low CD20 antigen expression. Although the disease often takes an indolent course, the majority of patients will eventually require therapy. Standard treatment for medically fit patients includes purine analogs and/or alkylating agents in addition to the type I anti-CD20 monoclonal antibody, rituximab. This therapy is inherently myelosuppressive and can result in significant morbidity and even mortality in patients with impaired performance status due to age and/or medical comorbidities. Historically, treatment options for the elderly or frail patient population were limited to mono-therapy with the oral alkylating agent, chlorambucil, rituximab, or another type I anti-CD20 monoclonal antibody ofatumumab. Recently, a newer-generation anti-CD20 monoclonal antibody, obinutuzumab, was developed for patients with CLL. Obinutuzumab is a humanized type II monoclonal antibody that appears to have more direct antibody-dependent cell-mediated cytotoxicity (ADCC) and possibly more direct cytotoxicity in vitro than previously available type I antibodies. A large Phase III prospective randomized clinical trial for older patients with impaired renal function and/or significant medical comorbidities demonstrated that when compared to conventionally-dosed rituximab and chlorambucil, the combination of chlorambucil and obinutuzumab administered at a dose and schedule involving early loading doses improved response rates and progression-free survival without significantly increasing toxicity. Results of this pivotal trial led to the FDA (US Food and Drug Administration) approval of obinutuzumab in combination with chlorambucil for frontline treatment of CLL. Obinutuzumab expands the armamentarium of active and less-toxic targeted agents in the evolving treatment landscape of CLL, providing physicians and patients with an additional therapeutic option.
Collapse
Affiliation(s)
- Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Matt Kalaycio
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
36
|
Sehn LH, Goy A, Offner FC, Martinelli G, Caballero MD, Gadeberg O, Baetz T, Zelenetz AD, Gaidano G, Fayad LE, Buckstein R, Friedberg JW, Crump M, Jaksic B, Zinzani PL, Padmanabhan Iyer S, Sahin D, Chai A, Fingerle-Rowson G, Press OW. Randomized Phase II Trial Comparing Obinutuzumab (GA101) With Rituximab in Patients With Relapsed CD20+ Indolent B-Cell Non-Hodgkin Lymphoma: Final Analysis of the GAUSS Study. J Clin Oncol 2015; 33:3467-74. [PMID: 26282650 DOI: 10.1200/jco.2014.59.2139] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Obinutuzumab (GA101), a novel glycoengineered type II anti-CD20 monoclonal antibody, demonstrated responses in single-arm studies of patients with relapsed/refractory non-Hodgkin lymphoma. This is the first prospective, randomized study comparing safety and efficacy of obinutuzumab with rituximab in relapsed indolent lymphoma. The primary end point of this study was the overall response rate (ORR) in patients with follicular lymphoma after induction and safety in patients with indolent lymphoma. PATIENTS AND METHODS A total of 175 patients with relapsed CD20(+) indolent lymphoma requiring therapy and with previous response to a rituximab-containing regimen were randomly assigned (1:1) to four once-per-week infusions of either obinutuzumab (1,000 mg) or rituximab (375 mg/m(2)). Patients without evidence of disease progression after induction therapy received obinutuzumab or rituximab maintenance therapy every 2 months for up to 2 years. RESULTS Among patients with follicular lymphoma (n = 149), ORR seemed higher for obinutuzumab than rituximab (44.6% v 33.3%; P = .08). This observation was also demonstrated by a blinded independent review panel that measured a higher ORR for obinutuzumab (44.6% v 26.7%; P = .01). However, this difference did not translate into an improvement in progression-free survival. No new safety signals were observed for obinutuzumab, and the incidence of adverse events was balanced between arms, with the exception of infusion-related reactions and cough, which were higher in the obinutuzumab arm. CONCLUSION Obinutuzumab demonstrated a higher ORR without appreciable differences in safety compared with rituximab. However, the clinical benefit of obinutuzumab in this setting remains unclear and should be evaluated within phase III trials.
Collapse
Affiliation(s)
- Laurie H Sehn
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA.
| | - Andre Goy
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Fritz C Offner
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Giovanni Martinelli
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - M Dolores Caballero
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Ole Gadeberg
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Tara Baetz
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Andrew D Zelenetz
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Gianluca Gaidano
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Luis E Fayad
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Rena Buckstein
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jonathan W Friedberg
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Michael Crump
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Branimir Jaksic
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Pier Luigi Zinzani
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Swaminathan Padmanabhan Iyer
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Deniz Sahin
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Akiko Chai
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Günter Fingerle-Rowson
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Oliver W Press
- Laurie H. Sehn, Centre for Lymphoid Cancer, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia; Tara Baetz, Queen's University, Kingston General Hospital, Kingston; Rena Buckstein, Sunnybrook Health Sciences Center; Michael Crump, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada; Andre Goy and Luis E. Fayad, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; Fritz C. Offner, Institute of Hematology and Medical Oncology, University of Bologna; Pier Luigi Zinzani, Institute of Hematology "Seràgnoli" University of Bologna, Bologna; Giovanni Martinelli, European Institute of Oncology, Milano; Gianluca Gaidano, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy; M. Dolores Caballero, University Hospital of Salamanca, Salamanca, Spain; Ole Gadeberg, Vejle Hospital, Vejle, Denmark; Andrew D. Zelenetz, Memorial Sloan Kettering Cancer Center, New York; Jonathan Friedberg, James P. Wilmot Cancer Center, University of Rochester, Rochester, NY; Branimir Jadkisic, Clinical Hospital Merkur, University of Zagreb, Zagreb, Croatia; Swaminathan Padmanabhan Iyer, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, TX; Deniz Sahin and Günter Fingerle-Rowson, Roche, Basel, Switzerland; Akiko Chai, Genentech BioOncology, South San Francisco, CA; Oliver Press, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
37
|
Hartley JM, Chu TW, Peterson EM, Zhang R, Yang J, Harris J, Kopeček J. Super-Resolution Imaging and Quantitative Analysis of Membrane Protein/Lipid Raft Clustering Mediated by Cell-Surface Self-Assembly of Hybrid Nanoconjugates. Chembiochem 2015; 16:1725-9. [PMID: 26097072 PMCID: PMC4551445 DOI: 10.1002/cbic.201500278] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Indexed: 11/12/2022]
Abstract
Super-resolution imaging was used to quantify organizational changes in the plasma membrane after treatment with hybrid nanoconjugates. The nanoconjugates crosslinked CD20 on the surface of malignant B cells, thereby inducing apoptosis. Super-resolution images were analyzed by using pair-correlation analysis to determine cluster size and to count the average number of molecules in the clusters. The role of lipid rafts was investigated by pre-treating cells with a cholesterol chelator and actin destabilizer to prevent lipid raft formation. Lipid raft cluster size correlated with apoptosis induction after treatment with the nanoconjugates. Lipid raft clusters had radii of ∼ 200 nm in cells treated with the hybrid nanoconjugates. Super-resolution images provided precise molecule location coordinates that could be used to determine density of bound conjugates, cluster size, and number of molecules per cluster.
Collapse
Affiliation(s)
- Jonathan M Hartley
- Department of Bioengineering, University of Utah, 20 S. 2030 E., Room 108, Salt Lake City, UT 84112 (USA)
| | - Te-Wei Chu
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 S. 2000 E Room 301, Salt Lake City, UT 84112 (USA)
| | - Eric M Peterson
- Department of Chemistry, University of Utah, 315 S. 1400 E, Room 2020, Salt Lake City, UT 84112 (USA)
| | - Rui Zhang
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 S. 2000 E Room 301, Salt Lake City, UT 84112 (USA)
| | - Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 S. 2000 E Room 301, Salt Lake City, UT 84112 (USA)
| | - Joel Harris
- Department of Chemistry, University of Utah, 315 S. 1400 E, Room 2020, Salt Lake City, UT 84112 (USA)
| | - Jindřich Kopeček
- Department of Bioengineering, University of Utah, 20 S. 2030 E., Room 108, Salt Lake City, UT 84112 (USA).
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 S. 2000 E Room 301, Salt Lake City, UT 84112 (USA).
| |
Collapse
|
38
|
Owen CJ, Stewart DA. Obinutuzumab for the treatment of patients with previously untreated chronic lymphocytic leukemia: overview and perspective. Ther Adv Hematol 2015; 6:161-70. [PMID: 26288711 PMCID: PMC4530370 DOI: 10.1177/2040620715586528] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is the most common lymphoproliferative disorder in the Western world and predominantly affects older people. Until recently, most studies in CLL focused on younger patients in whom intensive therapy with the addition of rituximab to fludarabine and cyclophosphamide was shown to improve survival. Obinutuzumab is a novel type II anti-CD20 monoclonal antibody (mAb) that recently demonstrated an overall survival advantage when combined with chemotherapy in previously untreated older patients with CLL and comorbidities. Obinutuzumab was superior to rituximab in this same study in terms of response rates and progression-free survival. Several preclinical and early phase clinical studies also support the efficacy of obinutuzumab. The most frequent adverse event noted with obinutuzumab is infusion-related reactions, which occur more frequently than with rituximab and are typically restricted to the first cycle of therapy. Based on these results, obinutuzumab should be considered the gold standard mAb for combination with chemotherapy in previously untreated patients with CLL and comorbidities. The marked efficacy of obinutuzumab with a weak chemotherapy backbone implies significant potency of this mAb, making it the ideal partner for combination studies with other agents in CLL.
Collapse
Affiliation(s)
- Carolyn J Owen
- Departments of Medicine and Oncology, University of Calgary, 603 South Tower, Foothills Medical Centre, Calgary, Alberta T2N 2T9, Canada
| | - Douglas A Stewart
- Departments of Medicine and Oncology, University of Calgary, Alberta, Canada
| |
Collapse
|
39
|
Li HF, Wu C, Chen T, Zhang G, Zhao H, Ke CH, Xu Z. Construction and characterization of an anti-CD20 mAb nanocomb with exceptionally excellent lymphoma-suppressing activity. Int J Nanomedicine 2015; 10:4783-96. [PMID: 26257518 PMCID: PMC4525799 DOI: 10.2147/ijn.s80129] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The CD20-directed monoclonal antibody rituximab (RTX) established a new era in the treatment of non-Hodgkin lymphoma (NHL); however, suboptimal response and/or resistance to RTX still limit its clinical merits. Although four effector mechanisms are validated to participate in CD20-based immunotherapy, including complement-dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity, caspase-dependent apoptosis, and lysosome-mediated programmed cell death (PCD), they could hardly be synchronously activated by any anti-CD20 mAb or mAb derivative until now. Herein, a novel mAb nanocomb (polyethylenimine polymer–RTX–tositumomab [PPRT nanocomb]) was firstly constructed through mass arming two different anti-CD20 mAbs (RTX and tositumomab) to one polymer by nanotechnology. Comparing with free mAbs, PPRT nanocomb possesses a comparable binding ability and reduced “off-rate” to surface CD20 of NHL cells. When treated by PPRT nanocomb, the caspase-dependent apoptosis was remarkably enhanced except for concurrently eliciting complement-dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity, and lysosome-mediated PCD. Besides, “cross-cell link”-assisted homotypic adhesion by PPRT nanocomb further enhanced the susceptibility to PCD of lymphoma cells. Pharmacokinetic assays revealed that PPRT nanocomb experienced a relatively reduced clearance from peripheral blood compared with free antibodies. With the cooperation of all the abovementioned superiorities, PPRT nanocomb exhibits exceptionally excellent in vivo antitumor activities in both disseminated and localized human NHL xenotransplant models.
Collapse
Affiliation(s)
- Hua-Fei Li
- International Joint Cancer Institute, Translation Medicine Institute, the Second Military Medical University, Shanghai, People's Republic of China ; Planning Division, Scientific Research Department, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, People's Republic of China ; Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, People's Republic of China
| | - Cong Wu
- Department of Laboratory Diagnosis, Changhai Hospital, the Second Military Medical University, Shanghai, People's Republic of China
| | - Ting Chen
- Department of Cardiology, Changhai Hospital, the Second Military Medical University, Shanghai, People's Republic of China
| | - Ge Zhang
- International Joint Cancer Institute, Translation Medicine Institute, the Second Military Medical University, Shanghai, People's Republic of China
| | - He Zhao
- International Joint Cancer Institute, Translation Medicine Institute, the Second Military Medical University, Shanghai, People's Republic of China
| | - Chang-Hong Ke
- International Joint Cancer Institute, Translation Medicine Institute, the Second Military Medical University, Shanghai, People's Republic of China
| | - Zheng Xu
- Planning Division, Scientific Research Department, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, People's Republic of China
| |
Collapse
|
40
|
Abstract
Antibodies against surface molecules of human tumors are now frequently administered in combination with strong chemotherapy, increasing therapeutic efficacy but making the task of elucidating immunological events more difficult. Experiments on genetically manipulated mice indicate that antibody efficacy is greatest when IgG antibody coating tumor cells is engaged by the Fcγ-receptors of effector cells, chiefly the monocyte/macrophage lineage. Evidence suggests lesser roles for NK cells, neutrophils, receptor-mediated cytotoxicity and complement-mediated cytotoxicity. The classical mode of killing employed by macrophages is phagocytosis, but much has to be learned about optimally activating macrophages for this task, and about any other modes of cytotoxicity used. There is renewed interest in antigenic modulation, which implies removal of therapeutic antibody linked with antigen from target-cell surfaces. It is now apparent that this removal of immune complexes can be achieved either by internalization by the target cell, or by transfer of the complexes to another cell by trogocytosis. In trials, anti-idiotype antibodies surprisingly proved therapeutically more effective than anti-CD20, despite anti-idiotype being more effectively removed from target-cell surfaces by antigenic modulation. This anomalous result might reflect the fact that persistence of anti-CD20 immune complexes in large amounts induces serious effector modulation, which paralyzes macrophage attacks on antibody-coated cells. The case for effector modulation is argued by analogy with the therapeutic suppression of autoimmune inflammation by effector modulation, achieved by infusion either of normal IgG in large amounts, or of anti-red cell IgG in relatively small amounts.
Collapse
Affiliation(s)
- George T Stevenson
- University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| |
Collapse
|
41
|
Goede V, Klein C, Stilgenbauer S. Obinutuzumab (GA101) for the treatment of chronic lymphocytic leukemia and other B-cell non-hodgkin's lymphomas: a glycoengineered type II CD20 antibody. Oncol Res Treat 2015; 38:185-92. [PMID: 25877943 DOI: 10.1159/000381524] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 02/25/2015] [Indexed: 12/26/2022]
Abstract
Obinutuzumab (GA101) is a humanized, monoclonal type II CD20 antibody modified by glycoengineering. The glycoengineered Fc portion enhances the binding affinity to the FcγRIII receptor on immune effector cells, resulting in increased antibody-dependent cellular cytotoxicity and phagocytosis. In addition, the type II antibody binding characteristics of obinutuzumab to CD20 lead to an efficient induction of direct non-apoptotic cell death. Preclinical data demonstrated more efficient B-cell depletion in whole blood and superior antitumor activity in xenograft models of obinutuzumab as compared to the type I CD20 antibody rituximab. In previously untreated patients with chronic lymphocytic leukemia (CLL) and comorbidities, obinutuzumab plus chlorambucil increased response rates and prolonged progression-free survival compared with rituximab plus chlorambucil. Obinutuzumab had an acceptable and manageable safety profile, with infusion-related reactions during the first infusion as the most common adverse event. Further phase I/II clinical trials have also shown promising activity in other CD20-positive B-cell non-Hodgkin's lymphomas (NHL). Therefore, several clinical studies are planned or ongoing to investigate obinutuzumab with different combination partners in both untreated and relapsed/refractory patients with different B-cell NHL entities, which in addition to CLL include diffuse large B-cell lymphoma and follicular lymphoma. © 2015 S. Karger GmbH, Freiburg.
Collapse
Affiliation(s)
- Valentin Goede
- German CLL Study Group, Department I of Internal Medicine, University Hospital Cologne, Germany
| | | | | |
Collapse
|
42
|
Autophagy collaborates with ubiquitination to downregulate oncoprotein E2A/Pbx1 in B-cell acute lymphoblastic leukemia. Blood Cancer J 2015; 5:e274. [PMID: 25615280 PMCID: PMC4314458 DOI: 10.1038/bcj.2014.96] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 12/12/2014] [Indexed: 12/16/2022] Open
Abstract
B-cell acute lymphoblastic leukemia (B-ALL) accounts for the most cancer incidences in children. We present here that autophagy is downregulated in pediatric B-ALL, suggesting a possible link between autophagy failure and pediatric B-ALL leukemogenesis. With a pediatric t(1;19) B-ALL xenograft mouse model, we show here that activation of autophagy by preventive administration of rapamycin improved the survival of leukemia animals by partial restoration of hematopoietic stem/progenitor cells, whereas treatment of the animals with rapamycin caused leukemia bone marrow cell-cycle arrest. Activation of autophagy in vitro or in vivo by rapamycin or starvation downregulated oncogenic fusion protein E2A/Pbx1. Furthermore, E2A/Pbx1 was found to be colocalized with autophagy marker LC3 in autolysosomes and with ubiquitin in response to autophagy stimuli, whereas autophagy or ubiquitination inhibitor blocked these colocalizations. Together, our data suggest a collaborative action between autophagy and ubiquitination in the degradation of E2A/Pbx1, thereby revealing a novel strategy for targeted preventive or treatment therapy on the pediatric ALL.
Collapse
|
43
|
Vaughan AT, Chan CHT, Klein C, Glennie MJ, Beers SA, Cragg MS. Activatory and inhibitory Fcγ receptors augment rituximab-mediated internalization of CD20 independent of signaling via the cytoplasmic domain. J Biol Chem 2015; 290:5424-37. [PMID: 25568316 DOI: 10.1074/jbc.m114.593806] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Type I anti-CD20 mAb such as rituximab and ofatumumab engage with the inhibitory FcγR, FcγRIIb on the surface of B cells, resulting in immunoreceptor tyrosine-based inhibitory motif (ITIM) phosphorylation. Internalization of the CD20·mAb·FcγRIIb complex follows, the rate of which correlates with FcγRIIb expression. In contrast, although type II anti-CD20 mAb such as tositumomab and obinutuzumab also interact with and activate FcγRIIb, this interaction fails to augment the rate of CD20·mAb internalization, raising the question of whether ITIM phosphorylation plays any role in this process. We have assessed the molecular requirements for the internalization process and demonstrate that in contrast to internalization of IgG immune complexes, FcγRIIb-augmented internalization of rituximab-ligated CD20 occurs independently of the FcγRIIb ITIM, indicating that signaling downstream of FcγRIIb is not required. In transfected cells, activatory FcγRI, FcγRIIa, and FcγRIIIa augmented internalization of rituximab-ligated CD20 in a similar manner. However, FcγRIIa mediated a slower rate of internalization than cells expressing equivalent levels of the highly homologous FcγRIIb. The difference was maintained in cells expressing FcγRIIa and FcγRIIb lacking cytoplasmic domains and in which the transmembrane domains had been exchanged. This difference may be due to increased degradation of FcγRIIa, which traffics to lysosomes independently of rituximab. We conclude that the cytoplasmic domain of FcγR is not required for promoting internalization of rituximab-ligated CD20. Instead, we propose that FcγR provides a structural role in augmenting endocytosis that differs from that employed during the endocytosis of immune complexes.
Collapse
Affiliation(s)
- Andrew T Vaughan
- From the Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, General Hospital, Southampton SO16 6YD, United Kingdom and
| | - Claude H T Chan
- From the Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, General Hospital, Southampton SO16 6YD, United Kingdom and
| | - Christian Klein
- the Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, CH-8952 Schlieren, Switzerland
| | - Martin J Glennie
- From the Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, General Hospital, Southampton SO16 6YD, United Kingdom and
| | - Stephen A Beers
- From the Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, General Hospital, Southampton SO16 6YD, United Kingdom and
| | - Mark S Cragg
- From the Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, General Hospital, Southampton SO16 6YD, United Kingdom and
| |
Collapse
|
44
|
Winiarska M, Bojarczuk K, Pyrzynska B, Bil J, Siernicka M, Dwojak M, Bobrowicz M, Miazek N, Zapala P, Zagozdzon A, Krol M, Syta A, Podszywalow-Bartnicka P, Pilch Z, Dabrowska-Iwanicka A, Juszczynski P, Efremov DG, Slabicki M, Zenz T, Le Roy A, Olive D, Rygiel TP, Leusen JHW, Golab J. Inhibitors of SRC kinases impair antitumor activity of anti-CD20 monoclonal antibodies. MAbs 2014; 6:1300-13. [PMID: 25517315 PMCID: PMC4622538 DOI: 10.4161/mabs.32106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Clinical trials with SRC family kinases (SFKs) inhibitors used alone or in a combination with anti-CD20 monoclonal antibodies (mAbs) are currently underway in the treatment of B-cell tumors. However, molecular interactions between these therapeutics have not been studied so far. A transcriptional profiling of tumor cells incubated with SFKs inhibitors revealed strong downregulation of MS4A1 gene encoding CD20 antigen. In a panel of primary and established B-cell tumors we observed that SFKs inhibitors strongly affect CD20 expression at the transcriptional level, leading to inhibition of anti-CD20 mAbs binding and increased resistance of tumor cells to complement-dependent cytotoxicity. Activation of the AKT signaling pathway significantly protected cells from dasatinib-triggered CD20 downregulation. Additionally, SFKs inhibitors suppressed antibody-dependent cell-mediated cytotoxicity by direct inhibition of natural killer cells. Abrogation of antitumor activity of rituximab was also observed in vivo in a mouse model. Noteworthy, the effects of SFKs inhibitors on NK cell function are largely reversible. The results of our studies indicate that development of optimal combinations of novel treatment modalities with anti-CD20 mAbs should be preceded by detailed preclinical evaluation of their effects on target cells.
Collapse
Affiliation(s)
- Magdalena Winiarska
- a Department of Immunology; Center for Biostructure Research ; Medical University of Warsaw ; Warsaw , Poland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Da Roit F, Engelberts PJ, Taylor RP, Breij ECW, Gritti G, Rambaldi A, Introna M, Parren PWHI, Beurskens FJ, Golay J. Ibrutinib interferes with the cell-mediated anti-tumor activities of therapeutic CD20 antibodies: implications for combination therapy. Haematologica 2014; 100:77-86. [PMID: 25344523 DOI: 10.3324/haematol.2014.107011] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The novel Bruton tyrosine kinase inhibitor ibrutinib and phosphatidyl-4-5-biphosphate 3-kinase-δ inhibitor idelalisib are promising drugs for the treatment of chronic lymphocytic leukemia and B-cell non-Hodgkin lymphoma, either alone or in combination with anti-CD20 antibodies. We investigated the possible positive or negative impact of these drugs on all known mechanisms of action of both type I and type II anti-CD20 antibodies. Pretreatment with ibrutinib for 1 hour did not increase direct cell death of cell lines or chronic lymphocytic leukemia samples mediated by anti-CD20 antibodies. Pre-treatment with ibrutinib did not inhibit complement activation or complement-mediated lysis. In contrast, ibrutinib strongly inhibited all cell-mediated mechanisms induced by anti-CD20 antibodies rituximab, ofatumumab or obinutuzumab, either in purified systems or whole blood assays. Activation of natural killer cells, and antibody-dependent cellular cytotoxicity by these cells, as well as phagocytosis by macrophages or neutrophils were inhibited by ibrutinib with a half maximal effective concentration of 0.3-3 μM. Analysis of anti-CD20 mediated activation of natural killer cells isolated from patients on continued oral ibrutinib treatment suggested that repeated drug dosing inhibits these cells in vivo. Finally we show that the phosphatidyl-4-5-biphosphate 3-kinase-δ inhibitor idelalisib similarly inhibited the immune cell-mediated mechanisms induced by anti-CD20 antibodies, although the effects of this drug at 10 μM were weaker than those observed with ibrutinib at the same concentration. We conclude that the design of combined treatment schedules of anti-CD20 antibodies with these kinase inhibitors should consider the multiple negative interactions between these two classes of drugs.
Collapse
Affiliation(s)
- Fabio Da Roit
- Center of Cellular Therapy "G. Lanzani", Division of Hematology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | | | - Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | | | - Giuseppe Gritti
- Center of Cellular Therapy "G. Lanzani", Division of Hematology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Alessandro Rambaldi
- Center of Cellular Therapy "G. Lanzani", Division of Hematology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Martino Introna
- Center of Cellular Therapy "G. Lanzani", Division of Hematology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Paul W H I Parren
- Genmab, Utrecht, the Netherlands Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Josée Golay
- Center of Cellular Therapy "G. Lanzani", Division of Hematology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
46
|
Vire B, Skarzynski M, Thomas JD, Nelson CG, David A, Aue G, Burke TR, Rader C, Wiestner A. Harnessing the fcμ receptor for potent and selective cytotoxic therapy of chronic lymphocytic leukemia. Cancer Res 2014; 74:7510-7520. [PMID: 25344228 DOI: 10.1158/0008-5472.can-14-2030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is a B-cell malignancy in need of new, effective, and safe therapies. The recently identified IgM receptor FcμR is overexpressed on malignant B cells in CLL and mediates the rapid internalization and lysosomal shuttling of IgM via its Fc fragment (Fcμ). To exploit this internalization and trafficking pathway for targeted drug delivery, we engineered an IgM-derived protein scaffold (Fcμ) and linked it with the cytotoxic agent monomethylauristatin F. This Fcμ-drug conjugate was selectively toxic for FcμR-expressing cell lines in vitro and for CLL cells but not autologous normal T cells ex vivo. Notably, the cytotoxic activity of the Fcμ-drug conjugate was maintained in CLL cells carrying a 17p deletion, which predicts resistance to standard chemotherapy. Next, we tested the possible therapeutic application of the Fcμ-drug conjugate in immunodeficient NOD/SCID/IL-2Rγ(null) (NSG) mice engrafted with peripheral blood cells from patients with leukemia. Three intravenous injections of the Fcμ-drug conjugate over a 10-day period were well tolerated and selectively killed the human CLL cells but not the coengrafted autologous human T cells. In summary, we developed a novel strategy for targeted cytotoxic therapy of CLL based on the unique properties of FcμR. FcμR-targeted drug delivery showed potent and specific therapeutic activity in CLL, thus providing proof of concept for FcμR as a valuable therapeutic target in CLL and for IgM-based antibody-drug conjugates as a new targeting platform.
Collapse
Affiliation(s)
- Bérengère Vire
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Martin Skarzynski
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joshua D Thomas
- Chemical Biology Laboratory, Molecular Discovery Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Christopher G Nelson
- Chemical Biology Laboratory, Molecular Discovery Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Alexandre David
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Georg Aue
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Terrence R Burke
- Chemical Biology Laboratory, Molecular Discovery Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Christoph Rader
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Department of Cancer Biology and Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
47
|
Breton CS, Nahimana A, Aubry D, Macoin J, Moretti P, Bertschinger M, Hou S, Duchosal MA, Back J. A novel anti-CD19 monoclonal antibody (GBR 401) with high killing activity against B cell malignancies. J Hematol Oncol 2014; 7:33. [PMID: 24731302 PMCID: PMC4021825 DOI: 10.1186/1756-8722-7-33] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/07/2014] [Indexed: 01/16/2023] Open
Abstract
Background CD19 is a B cell lineage specific surface receptor whose broad expression, from pro-B cells to early plasma cells, makes it an attractive target for the immunotherapy of B cell malignancies. In this study we present the generation of a novel humanized anti-CD19 monoclonal antibody (mAb), GBR 401, and investigate its therapeutic potential on human B cell malignancies. Methods GBR 401 was partially defucosylated in order to enhance its cytotoxic function. We analyzed the in vitro depleting effects of GBR 401 against B cell lines and primary malignant B cells from patients in the presence or in absence of purified NK cells isolated from healthy donors. In vivo, the antibody dependent cellular cytotoxicity (ADCC) efficacy of GBR 401 was assessed in a B cell depletion model consisting of SCID mice injected with healthy human donor PBMC, and a malignant B cell depletion model where SCID mice are xenografted with both primary human B-CLL tumors and heterologous human NK cells. Furthermore, the anti-tumor activity of GBR 401 was also evaluated in a xenochimeric mouse model of human Burkitt lymphoma using mice xenografted intravenously with Raji cells. Pharmacological inhibition tests were used to characterize the mechanism of the cell death induced by GBR 401. Results GBR 401 exerts a potent in vitro and in vivo cytotoxic activity against primary samples from patients representing various B-cell malignancies. GBR 401 elicits a markedly higher level of ADCC on primary malignant B cells when compared to fucosylated similar mAb and to Rituximab, the current anti-CD20 mAb standard immunotherapeutic treatment for B cell malignancies, showing killing at 500 times lower concentrations. Of interest, GBR 401 also exhibits a potent direct killing effect in different malignant B cell lines that involves homotypic aggregation mediated by actin relocalization. Conclusion These results contribute to consolidate clinical interest in developing GBR 401 for treatment of hematopoietic B cell malignancies, particularly for patients refractory to anti-CD20 mAb therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Michel A Duchosal
- Service and Central Laboratory of Hematology, University Hospital of Lausanne, Rue du Bugnon 46, 1011- CHUV, Lausanne, Switzerland.
| | | |
Collapse
|
48
|
Methuosis: nonapoptotic cell death associated with vacuolization of macropinosome and endosome compartments. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1630-42. [PMID: 24726643 DOI: 10.1016/j.ajpath.2014.02.028] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 01/30/2014] [Accepted: 02/04/2014] [Indexed: 12/19/2022]
Abstract
Apoptosis is the most widely recognized form of physiological programmed cell death. During the past three decades, various nonapoptotic forms of cell death have gained increasing attention, largely because of their potential importance in pathological processes, toxicology, and cancer therapy. A recent addition to the panoply of cell death phenotypes is methuosis. The neologism is derived from the Greek methuo (to drink to intoxication) because the hallmark of this form of cell death is displacement of the cytoplasm by large fluid-filled vacuoles derived from macropinosomes. The demise of the cell resembles many forms of necrosis, insofar as there is a loss of metabolic capacity and plasma membrane integrity, without the cell shrinkage and nuclear fragmentation associated with apoptosis. Methuosis was initially defined in glioblastoma cells after ectopic expression of activated Ras, but recent reports have described small molecules that can induce the features of methuosis in a broad spectrum of cancer cells, including those that are resistant to conventional apoptosis-inducing drugs. This review summarizes the available information about the distinguishing morphological characteristics and underlying mechanisms of methuosis. We compare and contrast methuosis with other cytopathological conditions in which accumulation of clear cytoplasmic vacuoles is a prominent feature. Finally, we highlight key questions that need to be answered to determine whether methuosis truly represents a unique form of regulated cell death.
Collapse
|
49
|
Zhao L, Xie F, Tong X, Li H, Chen Y, Qian W, Duan S, Zheng J, Zhao Z, Li B, Zhang D, Zhao J, Dai J, Wang H, Hou S, Guo Y. Combating non-Hodgkin lymphoma by targeting both CD20 and HLA-DR through CD20-243 CrossMab. MAbs 2014; 6:740-8. [PMID: 24670986 DOI: 10.4161/mabs.28613] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Although rituximab has revolutionized the treatment of hematological malignancies, the acquired resistance is one of the prime obstacles for cancer treatment, and development of novel CD20-targeting antibodies with potent anti-tumor activities and specificities is urgently needed. Emerging evidence has indicated that lysosomes can be considered as an "Achilles heel" for cancer cells, and might serve as an effective way to kill resistant cancer cells. HLA-DR antibody L243 has been recently reported to elicit potent lysosome-mediated cell death in lymphoma and leukemia cells, suggesting that HLA-DR could be used as a potential target against lymphoma. In this study, we generated a bispecific immunoglobulin G-like antibody targeting both CD20 and HLA-DR (CD20-243 CrossMab) through CrossMab technology. We found that the CrossMab could induce remarkably high levels of complement-dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity and anti-proliferative activity. Notably, although HLA-DR is expressed on normal and malignant cells, the CrossMab exhibited highly anti-tumor specificity, showing efficient eradication of hematological malignancies both in vitro and in vivo. Our data indicated that combined targeting of CD20 and HLA-DR could be an effective approach against malignancies, suggesting that CD20-243 CrossMab would be a promising therapeutic agent against lymphoma.
Collapse
Affiliation(s)
- Lei Zhao
- International Joint Cancer Institute; Second Military Medical University; Shanghai, PR China; PLA General Hospital Cancer Center; PLA School of Medicine; Beijing, PR China; The State Key Laboratory of Antibody Medicine & Targeting Therapy and Shanghai Key Laboratory of Cell Engineering & Antibody; Shanghai, PR China
| | - Feiyue Xie
- PLA General Hospital Cancer Center; PLA School of Medicine; Beijing, PR China
| | | | - Huafei Li
- International Joint Cancer Institute; Second Military Medical University; Shanghai, PR China
| | - Yaling Chen
- International Joint Cancer Institute; Second Military Medical University; Shanghai, PR China
| | - Weizhu Qian
- International Joint Cancer Institute; Second Military Medical University; Shanghai, PR China
| | - Shuyan Duan
- School of Pharmaceutical; Liaocheng University; Liaocheng, PR China
| | - Juan Zheng
- School of Pharmaceutical; Liaocheng University; Liaocheng, PR China
| | - Ziye Zhao
- School of Pharmaceutical; Liaocheng University; Liaocheng, PR China
| | - Bohua Li
- International Joint Cancer Institute; Second Military Medical University; Shanghai, PR China
| | - Dapeng Zhang
- International Joint Cancer Institute; Second Military Medical University; Shanghai, PR China; The State Key Laboratory of Antibody Medicine & Targeting Therapy and Shanghai Key Laboratory of Cell Engineering & Antibody; Shanghai, PR China
| | - Jian Zhao
- International Joint Cancer Institute; Second Military Medical University; Shanghai, PR China; PLA General Hospital Cancer Center; PLA School of Medicine; Beijing, PR China; The State Key Laboratory of Antibody Medicine & Targeting Therapy and Shanghai Key Laboratory of Cell Engineering & Antibody; Shanghai, PR China
| | - Jianxin Dai
- International Joint Cancer Institute; Second Military Medical University; Shanghai, PR China; PLA General Hospital Cancer Center; PLA School of Medicine; Beijing, PR China; The State Key Laboratory of Antibody Medicine & Targeting Therapy and Shanghai Key Laboratory of Cell Engineering & Antibody; Shanghai, PR China; School of Pharmaceutical; Liaocheng University; Liaocheng, PR China
| | - Hao Wang
- International Joint Cancer Institute; Second Military Medical University; Shanghai, PR China; PLA General Hospital Cancer Center; PLA School of Medicine; Beijing, PR China; The State Key Laboratory of Antibody Medicine & Targeting Therapy and Shanghai Key Laboratory of Cell Engineering & Antibody; Shanghai, PR China; School of Pharmaceutical; Liaocheng University; Liaocheng, PR China
| | - Sheng Hou
- International Joint Cancer Institute; Second Military Medical University; Shanghai, PR China; PLA General Hospital Cancer Center; PLA School of Medicine; Beijing, PR China; The State Key Laboratory of Antibody Medicine & Targeting Therapy and Shanghai Key Laboratory of Cell Engineering & Antibody; Shanghai, PR China; School of Pharmaceutical; Liaocheng University; Liaocheng, PR China
| | - Yajun Guo
- International Joint Cancer Institute; Second Military Medical University; Shanghai, PR China; PLA General Hospital Cancer Center; PLA School of Medicine; Beijing, PR China; The State Key Laboratory of Antibody Medicine & Targeting Therapy and Shanghai Key Laboratory of Cell Engineering & Antibody; Shanghai, PR China; School of Pharmaceutical; Liaocheng University; Liaocheng, PR China
| |
Collapse
|
50
|
Tomkins-Netzer O, Taylor SRJ, Lightman S. Can rituximab induce long-term disease remission in patients with intra-ocular non-infectious inflammation? ACTA ACUST UNITED AC 2013; 230:109-15. [PMID: 23948944 DOI: 10.1159/000351426] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Treatment of non-infectious uveitis is based primarily on the use of systemic corticosteroids and second-line immunosuppressive drugs. However, their extensive side effect profile, particularly for steroids, has led to the increased use of other immunosuppressive drugs, as sparing capacity agents. Rituximab is an anti-CD20 chimeric antibody, often given as a single course of 2 infusions, resulting in complete depletion of peripheral mature B cells. While it is licensed to treat refractory systemic lymphoma patients, it has also shown promising results in systemic auto-immune diseases, where a single course of treatment is able to achieve long-term clinical remission. Treatment with rituximab has been reported for various ocular conditions, suggesting it may be effective in inducing long-term disease control and other systemic immunosuppressive agents can be reduced or discontinued. When disease relapse occurs, a further course or courses can be given with good results. This review summarizes the current evidence regarding the role of rituximab in treating non-infectious uveitis.
Collapse
|