201
|
Suresh T, Lee LX, Joshi J, Barta SK. New antibody approaches to lymphoma therapy. J Hematol Oncol 2014; 7:58. [PMID: 25355407 PMCID: PMC4172963 DOI: 10.1186/s13045-014-0058-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/29/2014] [Indexed: 01/01/2023] Open
Abstract
The CD20-directed monoclonal antibody rituximab established a new era in lymphoma therapy. Since then other epitopes on the lymphoma surface have been identified as potential targets for monoclonal antibodies (mAb). While most mAbs eliminate lymphoma cells mainly by antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity or direct cell death, others counter mechanisms utilized by malignant cells to evade immune surveillance. Expression of PD-L1 on malignant or stromal cells in the tumor environment for example leads to T-cell anergy. Targeting either PD-1 or PD-L1 via mAbs can indirectly eliminate cancer cells by unblocking the host intrinsic immune response. Yet another mechanism of targeted therapy with mAbs are bi-specific T-cell engagers (BiTE) such as blinatumomab, which directly engages the host immune cells. These examples highlight the broad spectrum of available therapies targeting the lymphoma surface with mAbs utilizing both passive and active immune pathways. Many of these agents have already demonstrated significant activity in clinical trials. In this review we will focus on novel CD20-directed antibodies as well as mAbs directed against newer targets like CD19, CD22, CD40, CD52 and CCR4. In addition we will review mAbs unblocking immune checkpoints and the BiTE blinatumomab. Given the success of mAbs and the expansion in active and passive immunotherapies, these agents will play an increasing role in the treatment of lymphomas.
Collapse
|
202
|
Illidge T, Cheadle EJ, Donaghy C, Honeychurch J. Update on obinutuzumab in the treatment of B-cell malignancies. Expert Opin Biol Ther 2014; 14:1507-17. [PMID: 25190612 DOI: 10.1517/14712598.2014.948414] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION The anti-CD20 mAb rituximab has revolutionized the treatment of B-cell malignancies, improving outcome for patients. Despite these improvements, the majority of patients still relapse and become refractory to rituximab. Further efforts to improve anti-CD20 mAb efficacy have recently focused on obinutuzumab /GA101, a novel anti-CD20 mAb glycoengineered to display enhanced Fc-mediated effector mechanisms and induce direct cell death. AREAS COVERED We provide an overview of the current insights into the mechanisms of action of obinutuzumab focusing on how structural modifications and differences to rituximab led to designation of obinutuzumab as a type II antibody. We summarize data from preclinical studies and recent clinical trials including the Phase III trial in chronic lymphocytic leukemia (CLL), which led to FDA approval in November 2013. EXPERT OPINION Clinical data are now emerging confirming the promise of the initial preclinical data that demonstrated superior efficacy of obinutuzumab over rituximab at similar dosing. The emerging randomized Phase III data from older comorbid patients with previously untreated CLL demonstrated significant improvements in molecular remission rates and median progression-free survival of obinutuzumab plus chlorambucil versus rituximab plus chlorambucil. This emerging data provide reasons to be optimistic that outcomes for patients with B-cell malignancies can be further improved with obinutuzumab.
Collapse
Affiliation(s)
- Tim Illidge
- University of Manchester, Institute of Cancer Sciences, The Christie Hospital, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre , Manchester M20 4BX , UK +44 0 161 446 8110 ; +44 0 161 446 8001 ;
| | | | | | | |
Collapse
|
203
|
|
204
|
Obinutuzumab (GA101) in relapsed/refractory chronic lymphocytic leukemia: final data from the phase 1/2 GAUGUIN study. Blood 2014; 124:2196-202. [PMID: 25143487 DOI: 10.1182/blood-2014-07-586610] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
GAUGUIN evaluated the safety and efficacy of obinutuzumab (GA101) monotherapy in patients with relapsed/refractory chronic lymphocytic leukemia (CLL). In phase 1 (dose escalation), 13 patients received obinutuzumab 400 to 1200 mg (days 1 and 8 of cycle 1; day 1 of cycles 2-8). In phase 2, 20 patients received a fixed dose of 1000 mg (days 1, 8, and 15 of cycle 1; day 1 of cycles 2-8). Infusion-related reactions occurred in nearly all patients, but few were grade 3/4. Grade 3/4 neutropenia occurred in 7 patients in phase 1 (but was not dose-related) and in 4 patients in phase 2. Overall end-of-treatment response (all partial responses) was 62% (phase 1) and 15% (phase 2); best overall response was 62% and 30%, respectively. Phase 2 median progression-free survival was 10.7 months and median duration of response was 8.9 months. In summary, obinutuzumab monotherapy is active in patients with heavily pretreated relapsed/refractory CLL.
Collapse
|
205
|
Lim SH, Levy R. Translational medicine in action: anti-CD20 therapy in lymphoma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:1519-24. [PMID: 25086174 DOI: 10.4049/jimmunol.1490027] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The introduction of rituximab for B cell lymphoma in the late 1990s inaugurated a new era of cancer therapy showcasing mAbs. mAbs are in principle an amalgamation of two characteristics of a perfect anticancer drug. First, rituximab is a therapy targeted to the tumor cell, but it carries fewer side effects than does chemotherapy. Second, with its ability to directly engage the host immune system, it could potentially elicit longer lasting anticancer immunity, although this remains to be proven. This review highlights the fundamental scientific discoveries that allowed the development of clinically successful anti-CD20 mAbs. Since the approval of rituximab, a considerable amount of work has been undertaken by different groups trying to understand the workings and limitations of anti-CD20s. All of these efforts will be critical in designing new mAbs to CD20 and other targets and, ultimately, of anticancer mAbs that will improve on, or even replace, chemotherapy.
Collapse
Affiliation(s)
- Sean H Lim
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305
| | - Ronald Levy
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305
| |
Collapse
|
206
|
Li M, Xiao X, Zhang W, Liu L, Xi N, Wang Y. AFM analysis of the multiple types of molecular interactions involved in rituximab lymphoma therapy on patient tumor cells and NK cells. Cell Immunol 2014; 290:233-44. [PMID: 25117605 DOI: 10.1016/j.cellimm.2014.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/12/2014] [Accepted: 07/12/2014] [Indexed: 10/25/2022]
Abstract
Rituximab is a monoclonal antibody drug approved for the treatment of patients with lymphomas. Rituximab's main killing mechanism is antibody-dependent cellular cytotoxicity (ADCC). During ADCC, rituximab's fragment antigen binding (Fab) region binds to the CD20 antigen on the tumor cell and its fragment crystallizable (Fc) region binds to the Fc receptor (FcR) on the natural killer (NK) cells. In this study, two types of molecular interactions (CD20-rituximab, FcR-rituximab) involved in ADCC were measured simultaneously on cells prepared from biopsy specimens of lymphoma patients by utilizing atomic force microscopy (AFM) with functionalized tips carrying rituximab. NK cells were detected by specific NKp46 fluorescent labeling and tumor cells were detected by specific ROR1 fluorescent labeling. Based on the fluorescence recognition, the binding affinity and distribution of FcRs on NK cells, and CD20 on tumor cells, were quantitatively measured and mapped. The binding affinity and distribution of FcRs (on NK cells) and CD20 (on tumor cells) were associated with rituximab clinical efficacy. The experimental results provide a new approach to simultaneously quantify the multiple types of molecular interactions involved in rituximab ADCC mechanism on patient biopsy cells, which is of potential clinical significance to predict rituximab efficacy for personalized medicine.
Collapse
Affiliation(s)
- Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiubin Xiao
- Department of Lymphoma, Affiliated Hospital of Military Medical Academy of Sciences, Beijing 100071, China
| | - Weijing Zhang
- Department of Lymphoma, Affiliated Hospital of Military Medical Academy of Sciences, Beijing 100071, China
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China.
| | - Ning Xi
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China; Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China.
| | - Yuechao Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China
| |
Collapse
|
207
|
Casulo C, Vose JM, Ho WY, Kahl B, Brunvand M, Goy A, Kasamon Y, Cheson B, Friedberg JW. A phase I study of PRO131921, a novel anti-CD20 monoclonal antibody in patients with relapsed/refractory CD20+ indolent NHL: correlation between clinical responses and AUC pharmacokinetics. Clin Immunol 2014; 154:37-46. [PMID: 24928323 DOI: 10.1016/j.clim.2014.06.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 05/28/2014] [Accepted: 06/03/2014] [Indexed: 12/11/2022]
Abstract
PRO131921 is a third-generation, humanized anti-CD20 monoclonal antibody with increased antibody-dependent cytotoxicity and complement-dependent cytotoxicity compared to rituximab. In this phase I study, PRO131921 was administered as a single agent to patients with CD20+, relapsed or refractory, indolent non-Hodgkin lymphoma (NHL) who had been treated with a prior rituximab-containing regimen. The primary aim of this study was safety and tolerability of PRO131921. The secondary aim of the study, and focus of this report, was to determine the pharmacokinetics (PK) profile of PRO131921 and establish a correlation between drug exposure and clinical efficacy. Patients were treated with PRO131921 by intravenous infusion weekly for 4 weeks and the dose was escalated based on safety in a 3+3 design. Twenty-four patients were treated with PRO131921 at doses from 25mg/m(2) to 800 mg/m(2). Analysis of PK data demonstrated a correlation between higher normalized drug exposure (normalized AUC) and tumor shrinkage (p = .0035). Also, normalized AUC levels were higher among responders and subjects displaying tumor shrinkage versus subjects progressing or showing no regression (p = 0.030). In conclusion, PRO131921 demonstrated clinical activity in rituximab-relapsed and refractory indolent NHL patients. The observation that higher normalized AUC may be associated with improved clinical responses has potential implications in future trials of monoclonal antibody-based therapies, and emphasizes the importance of early PK studies to optimize antibody efficacy.
Collapse
Affiliation(s)
- Carla Casulo
- James P. Wilmot Cancer Center, University of Rochester, Rochester, NY, USA.
| | - Julie M Vose
- University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Brad Kahl
- Department of Medicine, University of Wisconsin School of Medicine, Madison, WI, USA
| | | | - Andre Goy
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Yvette Kasamon
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Bruce Cheson
- Division of Hematology and Oncology, Georgetown University Hospital, Washington, DC, USA
| | | |
Collapse
|
208
|
Reddy V, Leandro M. Variability in clinical and biological response to rituximab in autoimmune diseases: an opportunity for personalized therapy? ACTA ACUST UNITED AC 2014. [DOI: 10.2217/ijr.14.18] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
209
|
Abstract
INTRODUCTION We analyse data for the use of obinutuzumab in the treatment of CD20(+) lymphoproliferative disorders with a focus on chronic lymphocytic leukaemia (CLL). Targeted therapy against CD20 with the mAb rituximab led to significant improvements in survival for patients with B-cell non-Hodgkin lymphoma (NHL) and is the current mainstay of treatment for CD20(+) malignancies. Despite this, many patients relapse or become refractory after rituximab-containing therapies, so efforts have been made to develop better anti-CD20 mAbs. Obinutuzumab recently demonstrated superiority over rituximab in the only published Phase III study comparing the two antibodies. AREAS COVERED Obinutuzumab is a humanised, anti-CD20 mAb being compared to rituximab in several Phase III studies. An overview of obinutuzumab, its mechanisms of action and results of Phase I-III studies are presented. EXPERT OPINION The demonstration of superiority of obinutuzumab over rituximab in the CLL11 Phase III study is potentially practice-changing. Obinutuzumab has also proven safe and efficacious in CD20(+) NHL in Phase I/II studies and results of Phase III studies in NHL are eagerly awaited. The potential implications of improved outcomes for CLL and NHL with the introduction of this more potent anti-CD20 antibody are tremendous given the impressive results obtained after the introduction of rituximab over a decade ago.
Collapse
Affiliation(s)
- Carolyn J Owen
- University of Calgary, Tom Baker Cancer Centre, Division of Hematology and Hematological Malignancies , 603 South Tower, Foothills Medical Centre, 1403-29th St NW, Calgary, Alberta, T2N 2T9 , Canada
| | | |
Collapse
|
210
|
Al-Zoobi L, Salti S, Colavecchio A, Jundi M, Nadiri A, Hassan GS, El-Gabalawy H, Mourad W. Enhancement of Rituximab-induced cell death by the physical association of CD20 with CD40 molecules on the cell surface. Int Immunol 2014; 26:451-65. [PMID: 24894009 DOI: 10.1093/intimm/dxu046] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
CD20 is an attractive therapeutic target given the success of its monoclonal antibody, Rituximab, in the treatment of B-cell malignancies and B-cell-mediated autoimmune diseases. Treatment with Rituximab causes a rapid depletion of B cells and a decrease in disease symptoms. Despite the clinical efficiency of Rituximab, its mechanism of action is not completely understood. In this study, we aimed at further investigating the Rituximab-induced cell death and the factors affecting such responses. Our results indicate that Rituximab-induced cell death depends on the nature of the cells and levels of CD20 expression on the cell surface. Coexpression of CD20 with CD40, a member of the TNF receptor family that is known to be physically associated with CD20 on the cell surface, enhances the apoptotic response induced by Rituximab. Inhibiting the formation of CD40 disulfide-bound-homodimers, a process required for some CD40 signaling, further enhances Rituximab-induced cell death. Cell death induced by anti-CD40 mAb is also upregulated by the presence of CD20, suggesting a bidirectional influence of the CD20/CD40 association. Moreover, treating cells with both anti-CD20 and anti-CD40 antibodies improves the cell death response induced by a single-agent treatment. These results highlight the role of the CD20/CD40 association in triggering B-cell depletion and may pave the way for an alternative more efficient therapeutic strategy in treating B-cell-mediated disorders.
Collapse
Affiliation(s)
- Loubna Al-Zoobi
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Suzanne Salti
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Anna Colavecchio
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Malek Jundi
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Amal Nadiri
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Ghada S Hassan
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Hani El-Gabalawy
- Arthritis Centre, University of Manitoba Arthritis Centre, RR149-800 Sherbrooke Street, Winnipeg, Manitoba R3A 1M4, Canada
| | - Walid Mourad
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| |
Collapse
|
211
|
Baer Ii WH, Maini A, Jacobs I. Barriers to the Access and Use of Rituximab in Patients with Non-Hodgkin's Lymphoma and Chronic Lymphocytic Leukemia: A Physician Survey. Pharmaceuticals (Basel) 2014; 7:530-44. [PMID: 24810947 PMCID: PMC4035768 DOI: 10.3390/ph7050530] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/25/2014] [Accepted: 04/29/2014] [Indexed: 02/08/2023] Open
Abstract
Biologics such as rituximab are an important component of oncology treatment strategies, although access to such therapies is challenging in countries with limited resources. This study examined access to rituximab and identified potential barriers to its use in the United States, Mexico, Turkey, Russia, and Brazil. The study also examined whether availability of a biosimilar to rituximab would improve access to, and use of, rituximab. Overall, 450 hematologists and oncologists completed a survey examining their use of rituximab in patients with non-Hodgkin’s lymphoma (NHL) and chronic lymphocytic leukemia (CLL). Less than 40% of physicians considered rituximab as easy to access from a cost perspective. Furthermore, many physicians chose not to treat, were unable to treat, or had to modify treatment with rituximab despite guidelines recommending its use in NHL and CLL patients. Insurance coverage, reimbursement, and cost to patient were commonly reported as barriers to the use of rituximab. Across all markets, over half of physicians reported that they would increase use of rituximab if a biosimilar was available. We conclude that rituximab use would increase across all therapy types and markets if a biosimilar was available, although a biosimilar would have the greatest impact in Brazil, Mexico, and Russia.
Collapse
Affiliation(s)
- William H Baer Ii
- MercyHealth-ClinXus LLC, 260 Jefferson St., Grand Rapids, MI 49503, USA.
| | - Archana Maini
- Broward Health Medical Center, 1625 SE 3rd Ave. #525, Ft. Lauderdale, FL 33314, USA.
| | - Ira Jacobs
- Pfizer Inc., 235 East 42nd St., New York, NY 10017, USA.
| |
Collapse
|
212
|
Abstract
The complement terminal pathway clears pathogens by generating cytotoxic membrane attack complex (MAC) pores on target cells. For more than 40 years, biochemical and cellular assays have been used to characterize the lytic nature of the MAC and to define its protein composition. Although models for pore formation have been inferred from structures of bacterial cytolysins, it was only recently that we were able to visualize how complement components come together during MAC assembly. This review highlights structural analyses of terminal pathway complexes to explore molecular mechanisms underlying MAC formation.
Collapse
Affiliation(s)
- Doryen Bubeck
- Department of Life Sciences, Sir Ernst Chain Building, South Kensington Campus, Imperial College London , London SW7 2AZ, U.K
| |
Collapse
|
213
|
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: 1.3] [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
|
214
|
Abstract
In this issue of Blood, Vaughan et al demonstrate that certain antibodies that are used therapeutically in lymphoma treatment (e.g., rituximab) undergo Fcg receptor IIb (FcgRIIb)–mediated internalization from the B-cell surface in a manner that is independent of activation of FcgRIIb with important implications for the design of antibody-based therapeutics (see figure).
Collapse
|
215
|
Blair BG, Bardelli A, Park BH. Somatic alterations as the basis for resistance to targeted therapies. J Pathol 2014; 232:244-54. [PMID: 24114654 DOI: 10.1002/path.4278] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 08/23/2013] [Accepted: 09/21/2013] [Indexed: 12/20/2022]
Abstract
Recent advances in genetics and genomics have revealed new genes and pathways that are somatically altered in human malignancies. This wealth of knowledge has translated into molecularly defined targets for therapy over the past two decades, serving as key examples that translation of laboratory findings can have great impact on the ability to treat patients with cancer. However, given the genetic instability and heterogeneity that are characteristic of all human cancers, drug resistance to virtually all therapies has emerged, posing further and future challenges for clinical oncology. Here we review the history of targeted therapies, including examples of genetically defined cancer targets and their approved therapies. We also discuss resistance mechanisms that have been uncovered, with an emphasis on somatic genetic alterations that lead to these phenotypes.
Collapse
Affiliation(s)
- Brian G Blair
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | | | | |
Collapse
|
216
|
Li M, Xiao X, Zhang W, Liu L, Xi N, Wang Y. Nanoscale distribution of CD20 on B-cell lymphoma tumour cells and its potential role in the clinical efficacy of rituximab. J Microsc 2014; 254:19-30. [PMID: 24499016 DOI: 10.1111/jmi.12112] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 01/07/2014] [Indexed: 12/22/2022]
Abstract
Rituximab is an exciting monoclonal antibody drug approved for treating B-cell lymphomas and its target is the CD20 antigen which is expressed on the surface of B cells. In recent years, the variable efficacies of rituximab among different lymphoma patients have become an important clinical issue and urgently need to be solved for further development of antibodies with enhanced efficacies. In this work, atomic force microscopy (AFM) was used to investigate the nanoscale distribution of CD20 on the surface of tumour B cells from lymphoma patients to examine its potential role in the clinical therapeutic effects of rituximab. By performing ROR1 fluorescence labelling (ROR1 is a specific tumour cell surface marker) on the bone marrow cells prepared from B-cell lymphoma patients, the tumour B cells were recognized, and then AFM tips carrying rituximabs via polyethylene glycol crosslinkers were moved to the tumour cells to probe the specific CD20-rituximab interactions. By applying AFM single-molecule force spectroscopy (SMFS) at the local areas (500×500 nm²) on the surface of tumour B cells, the nanoscale distributions of CD20 on the surface of tumour B cells were mapped, visually showing that CD20 distributed heterogeneously on the cell surface. Bone marrow cell samples from three clinical B-cell lymphoma cases were collected to analyze the binding affinity and nanoscale distribution of CD20 on tumour cells. The experimental results showed that CD20 distribution on tumour cells were to some extent related to the clinical therapeutic outcomes while the CD20-rituximab binding forces did not have distinct effects to the clinical outcomes. These results can provide novel insights in understanding the rituximab's clinical efficacies from the nanoscale distribution of CD20 on the tumour cells at single-cell and single-molecule levels.
Collapse
Affiliation(s)
- M Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China.,University of Chinese Academy of Sciences, Beijing, China
| | - X Xiao
- Department of Lymphoma, Affiliated Hospital of Military Medical Academy of Sciences, Beijing, China
| | - W Zhang
- Department of Lymphoma, Affiliated Hospital of Military Medical Academy of Sciences, Beijing, China
| | - L Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
| | - N Xi
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China.,Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Y Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
| |
Collapse
|
217
|
Baig NA, Taylor RP, Lindorfer MA, Church AK, LaPlant BR, Pettinger AM, Shanafelt TD, Nowakowski GS, Zent CS. Induced resistance to ofatumumab-mediated cell clearance mechanisms, including complement-dependent cytotoxicity, in chronic lymphocytic leukemia. THE JOURNAL OF IMMUNOLOGY 2014; 192:1620-9. [PMID: 24431228 DOI: 10.4049/jimmunol.1302954] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ofatumumab (OFA), a human CD20-targeting mAb, kills B lymphocytes using the innate immune system including complement-dependent cytotoxicity (CDC). The efficacy of OFA in patients with chronic lymphocytic leukemia (CLL) is limited by drug resistance, which is not well characterized. To better understand mechanisms of resistance, we prospectively studied CLL cells isolated from blood samples collected before and after in vivo exposure to the initial dose of OFA therapy in 25 patients undergoing their first treatment for progressive CLL. As previously reported, OFA therapy rapidly decreased the absolute lymphocyte count, CD20 expression by CLL cells, and serum complement levels. We now show that after administration of the first dose of OFA, there was a modest rebound in the absolute lymphocyte count and serum complement levels, but substantial ongoing loss of CD20 expression by CLL cells. These post-OFA treatment CLL cells were highly resistant to OFA-mediated CDC but retained sensitivity to alemtuzumab-mediated CDC in vitro. Posttherapy serum OFA levels correlated inversely with both the amount of pretreatment circulating cell-bound CD20 and with the decrease in this value following treatment. In vitro OFA-mediated CDC did not predict clinical responses, and the patients with first-dose reactions to OFA did not have markers of increased complement activation in vivo. We propose that optimal efficacy of CD20- targeted therapy for CLL requires determining an mAb dose size and frequency that optimizes CLL killing without exceeding the capacity of the cytotoxic mechanisms and thus minimizes loss of CD20 expression in the surviving CLL cells.
Collapse
Affiliation(s)
- Nisar A Baig
- Division of Hematology, Mayo Clinic, Rochester, MN 55905
| | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Abstract
Multiple sclerosis (MS) is a life-long, potentially debilitating disease of the central nervous system (CNS). MS is considered to be an immune-mediated disease, and the presence of autoreactive peripheral lymphocytes in CNS compartments is believed to be critical in the process of demyelination and tissue damage in MS. Although MS is not currently a curable disease, several disease-modifying therapies (DMTs) are now available, or are in development. These DMTs are all thought to primarily suppress autoimmune activity within the CNS. Each therapy has its own mechanism of action (MoA) and, as a consequence, each has a different efficacy and safety profile. Neurologists can now select therapies on a more individual, patient-tailored basis, with the aim of maximizing potential for long-term efficacy without interruptions in treatment. The MoA and clinical profile of MS therapies are important considerations when making that choice or when switching therapies due to suboptimal disease response. This article therefore reviews the known and putative immunological MoAs alongside a summary of the clinical profile of therapies approved for relapsing forms of MS, and those in late-stage development, based on published data from pivotal randomized, controlled trials.
Collapse
|
219
|
Bakema JE, van Egmond M. Fc receptor-dependent mechanisms of monoclonal antibody therapy of cancer. Curr Top Microbiol Immunol 2014; 382:373-92. [PMID: 25116109 DOI: 10.1007/978-3-319-07911-0_17] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Targeted therapies like treatment with monoclonal antibodies (mAbs) have entered the arsenal of modern anticancer drugs. mAbs combine specificity with multiple effector functions that can lead to reduction of tumour burden. Direct mechanisms of action, including induction of apoptosis or growth inhibition, depend on the biology of the target antigen. Fc tails of mAbs have furthermore the potential to initiate complement-dependent lysis as well as immune effector cell-mediated tumour cell killing via binding to Fc receptors. Natural killer cells can induce apoptosis via antibody-dependent cellular cytotoxicity (ADCC), whereas macrophages are able to phagocytose mAb-opsonized tumour cells (antibody-dependent cellular phagocytosis; ADCP). Finally, neutrophils can induce non-apoptotic tumour cell death, especially in the presence of immunoglobulin A (IgA) antitumour mAbs. In spite of promising clinical successes in some malignancies, improvement of mAb immunotherapy is required to achieve overall complete remission in cancer patients. New strategies to enhance Fc receptor-mediated mechanisms of action or to overcome the immunosuppressive microenvironment of the tumour in mAb therapy of cancer are therefore currently being explored and will be addressed in this chapter.
Collapse
Affiliation(s)
- Jantine E Bakema
- Tumor Biology Section, Department of Otolaryngology/Head-Neck Surgery, VU University Medical Center, De Boelelaan 1117, 1007 MB, Amsterdam, The Netherlands
| | | |
Collapse
|
220
|
Wu C, Li H, Zhao H, Zhang W, Chen Y, Yue Z, Lu Q, Wan Y, Tian X, Deng A. Potentiating antilymphoma efficacy of chemotherapy using a liposome for integration of CD20 targeting, ultra-violet irradiation polymerizing, and controlled drug delivery. NANOSCALE RESEARCH LETTERS 2014; 9:447. [PMID: 25221463 PMCID: PMC4151082 DOI: 10.1186/1556-276x-9-447] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 08/23/2014] [Indexed: 05/06/2023]
Abstract
Unlike most malignancies, chemotherapy but not surgery plays the most important role in treating non-Hodgkin lymphoma (NHL). Currently, liposomes have been widely used to encapsulate chemotherapeutic drugs in treating solid tumors. However, higher in vivo stability owns a much more important position for excellent antitumor efficacy in treating hematological malignancies. In this study, we finely fabricated a rituximab Fab fragment-decorated liposome based on 1,2-bis(10,12-tricosadiynoyl)-sn-glycero-3-phosphocholine (DC8,9PC), which can form intermolecular cross-linking through the diacetylenic group by ultra-violet (UV) irradiation. Our experimental results demonstrated that after the UV irradiation, the liposomes exhibit better serum stability and slower drug release with a decreased mean diameter of approximately 285 nm. The cellular uptake of adriamycin (ADR) by this Fab-navigated liposome was about four times of free drugs. Cytotoxicity assays against CD20(+) lymphoma cells showed that the half maximal (50%) inhibitory concentration (IC50) of ADR-loaded immunoliposome was only one fourth of free ADR at the same condition. In vivo studies were evaluated in lymphoma-bearing SCID mice. With the high serum stability, finely regulated structure, active targeting strategy via antigen-antibody reaction and passive targeting strategy via enhanced permeability and retention (EPR) effect, our liposome exhibits durable and potent antitumor activities both in the disseminated and localized human NHL xeno-transplant models.
Collapse
Affiliation(s)
- Cong Wu
- Department of Laboratory Diagnosis, Changhai Hospital affiliated to the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Huafei Li
- Department of Laboratory Diagnosis, Changhai Hospital affiliated to the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
- International Joint Cancer Institute, the Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - He Zhao
- Institute of Pediatric Research, Children's Hospital affiliated to Soochow University, 303 Jingde Road, Suzhou 215000, China
| | - Weiwei Zhang
- Department of Laboratory Diagnosis, Changhai Hospital affiliated to the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yan Chen
- Department of Laboratory Diagnosis, Changhai Hospital affiliated to the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Zhanyi Yue
- Department of Laboratory Diagnosis, Changhai Hospital affiliated to the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Qiong Lu
- Department of Laboratory Diagnosis, Changhai Hospital affiliated to the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yuxiang Wan
- Department of Laboratory Diagnosis, Changhai Hospital affiliated to the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Xiaoyu Tian
- Department of Laboratory Diagnosis, Changhai Hospital affiliated to the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Anmei Deng
- Department of Laboratory Diagnosis, Changhai Hospital affiliated to the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
221
|
CD20 mAb-Mediated Complement Dependent Cytotoxicity of Tumor Cells is Enhanced by Blocking the Action of Factor I. Antibodies (Basel) 2013. [DOI: 10.3390/antib2040598] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
222
|
Inhibitory FcγRIIb (CD32b) becomes activated by therapeutic mAb in both cis and trans and drives internalization according to antibody specificity. Blood 2013; 123:669-77. [PMID: 24227819 DOI: 10.1182/blood-2013-04-490821] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A major feature that distinguishes type I from type II anti-CD20 monoclonal antibodies (mAbs) and reduces their therapeutic efficacy is the tendency to internalize from the cell surface. We have shown previously that the extent of internalization correlates with the capacity of type I mAb to simultaneously engage both CD20 and the inhibitory Fcγ receptor, FcγRIIb, in a bipolar configuration. Here, we investigated whether mAbs directed at other B-cell surface receptors also engaged FcγRIIb and whether this interaction promoted internalization. Most mAbs engaged and activated FcγRIIb, with the strength of activation related to the level of mAb bound to the cell surface. However, engagement did not affect internalization of most mAb-ligated receptors, either in cell lines or primary chronic lymphocytic leukemia cells with the exception of CD19 and CD38. Furthermore, at high cell concentrations/density both cis and trans interactions between cell-surface bound mAb and FcγRIIb were evident, but trans interactions did not inhibit type I anti-CD20 mAb-mediated internalization. These data identify that FcγRIIb is engaged by many mAbs in both cis and trans configurations, triggering its activation, but that internalization via FcγRIIb occurs for only a select subset. These findings have implications when designing new antibody-based therapeutics.
Collapse
|
223
|
Mao CP, Brovarney MR, Dabbagh K, Birnböck HF, Richter WF, Del Nagro CJ. Subcutaneous versus intravenous administration of rituximab: pharmacokinetics, CD20 target coverage and B-cell depletion in cynomolgus monkeys. PLoS One 2013; 8:e80533. [PMID: 24265828 PMCID: PMC3827219 DOI: 10.1371/journal.pone.0080533] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 10/15/2013] [Indexed: 12/27/2022] Open
Abstract
The CD20-specific monoclonal antibody rituximab (MabThera®, Rituxan®) is widely used as the backbone of treatment for patients with hematologic disorders. Intravenous administration of rituximab is associated with infusion times of 4–6 hours, and can be associated with infusion-related reactions. Subcutaneous administration of rituximab may reduce this and facilitate administration without infusion-related reactions. We sought to determine the feasibility of achieving equivalent efficacy (measured by endogenous B-cell depletion) and long-term durability of CD20 target coverage for subcutaneously administered rituximab compared with intravenous dosing. In these preclinical studies, male cynomolgus monkeys were treated with either intravenous rituximab or novel subcutaneous formulation of rituximab containing human recombinant DNA-derived hyaluronidase enzyme. Peripheral blood samples were analyzed for serum rituximab concentrations, peripheral B-cell depletion, and CD20 target coverage, including subset analysis according to CD21+ status. Distal lymph node B-cell depletion and CD20 target coverage were also measured. Initial peak serum concentrations of rituximab were significantly higher following intravenous administration than subcutaneous. However, the mean serum rituximab trough concentrations were comparable at 2 and 7 days post-first dose and 9 and 14 days post-second dose. Efficacy of B-cell depletion in both peripheral blood and distal lymph nodes was comparable for both methods. In lymph nodes, 9 days after the second dose with subcutaneous and intravenous rituximab, B-cell levels were decreased by 57% and 42% respectively. Similarly, levels of peripheral blood B cells were depleted by >94% for both subcutaneous and intravenous dosing at all time points. Long-term recovery of free unbound surface CD20 levels was similar, and the duration of B-cell depletion was equally sustained over 2 months for both methods. These results demonstrate that, despite initial peak serum drug level differences, subcutaneous rituximab has similar durability, pharmacodynamics, and efficacy compared with intravenous rituximab.
Collapse
Affiliation(s)
- Cheng-Ping Mao
- Discovery Inflammation, Roche Palo Alto, Palo Alto, California, United States of America
| | - Martin R. Brovarney
- Discovery Inflammation, Roche Palo Alto, Palo Alto, California, United States of America
| | - Karim Dabbagh
- Discovery Inflammation, Roche Palo Alto, Palo Alto, California, United States of America
| | | | | | - Christopher J. Del Nagro
- Discovery Inflammation, Roche Palo Alto, Palo Alto, California, United States of America
- Discovery Oncology, Genentech Incorporated, South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
224
|
Abstract
Despite major progress in the treatment of rheumatoid arthritis (RA), strong unmet medical need remains, as only a minor proportion of patients reach sustained clinical remission. New approaches are therefore necessary, and include manipulation of regulatory T cells, which might be able to restore the disturbed immune system and could even lead to a cure if this restored regulation were to prove sustainable. Logistical and conceptual problems, however, beset this attractive therapeutic approach, including difficulties with ex vivo expansion of cells, specificity of targeting and the optimal time point of administration. Therefore, alternative avenues are being investigated, such as targeting B-cell effector functions and newly identified proinflammatory cytokines. On the basis of success with B-cell depleting therapy using anti-CD20 agents, further treatment modalities are now exploring direct or indirect interference in B-cell-mediated immunity with the use of agents directed against other B-cell surface molecules. Novel approaches target intracellular B-cell signalling and regulatory B cells. New cytokine-directed therapies target important proinflammatory mediators such as GM-CSF, new members of the IL-1 family, IL-6 and its receptor, IL-17, IL-20, IL-21, IL-23 as well as synovium-specific targets. This article reviews these emerging cell and cytokine targets with special focus on biologic agents, some of which might reach the clinic soon whereas others will require considerable time in development. Nevertheless, these exciting new approaches will considerably enhance our repertoire in the battle against this potentially devastating disease.
Collapse
|
225
|
Li M, Xiao X, Liu L, Xi N, Wang Y, Dong Z, Zhang W. Nanoscale mapping and organization analysis of target proteins on cancer cells from B-cell lymphoma patients. Exp Cell Res 2013; 319:2812-21. [DOI: 10.1016/j.yexcr.2013.07.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 07/21/2013] [Accepted: 07/23/2013] [Indexed: 01/20/2023]
|
226
|
Montalvao F, Garcia Z, Celli S, Breart B, Deguine J, Van Rooijen N, Bousso P. The mechanism of anti-CD20-mediated B cell depletion revealed by intravital imaging. J Clin Invest 2013; 123:5098-103. [PMID: 24177426 DOI: 10.1172/jci70972] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 08/22/2013] [Indexed: 12/14/2022] Open
Abstract
Anti-CD20 Ab therapy has proven successful for treating B cell malignancies and a number of autoimmune diseases. However, how anti-CD20 Abs operate in vivo to mediate B cell depletion is not fully understood. In particular, the anatomical location, the type of effector cells, and the mechanism underlying anti-CD20 therapy remain uncertain. Here, we found that the liver is a major site for B cell depletion and that recirculation accounts for the decrease in B cell numbers observed in secondary lymphoid organs. Using intravital imaging, we established that, upon anti-CD20 treatment, Kupffer cells (KCs) mediate the abrupt arrest and subsequent engulfment of B cells circulating in the liver sinusoids. KCs were also effective in depleting malignant B cells in a model of spontaneous lymphoma. Our results identify Ab-dependent cellular phagocytosis by KCs as a primary mechanism of anti-CD20 therapy and provide an experimental framework for optimizing the efficacy of therapeutic Abs.
Collapse
|
227
|
Eradication of non-Hodgkin lymphoma through the induction of tumor-specific T-cell immunity by CD20-Flex BiFP. Blood 2013; 122:4230-6. [PMID: 24178967 DOI: 10.1182/blood-2013-04-496554] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Although monoclonal antibodies, including CD20 antibody rituximab, are standard therapeutics for several cancers, their efficacy remains variable and often modest. There is an urgent need to enhance the efficacy of the current generation of anticancer antibodies. Flt3 ligand, a soluble protein, has the ability to induce substantial expansion of dendritic cells (DCs). In this study, we constructed a bispecific immunoglobulin G-like bispecific fusion protein (BiFP) targeting both CD20 and Flt3 (CD20-Flex) by using CrossMab technology. We found that the BiFP exhibited stabilities that were comparable with the parental antibody rituximab and were able to bind to both targets with unaltered binding affinity. Notably, our data indicated that CD20-Flex BiFP could not only eliminate lymphoma temporarily but also potentiate tumor-specific T-cell immunity, which affords a long-lasting protection from tumor recurrence. The results showed that the expansion and infiltration of DCs into tumor tissues by CD20-Flex BiFP could be an effective way to generate protective immune responses against cancer, suggesting that the CD20-Flex BiFP could be a promising therapeutic agent against B-cell lymphomas.
Collapse
|
228
|
Okroj M, Österborg A, Blom AM. Effector mechanisms of anti-CD20 monoclonal antibodies in B cell malignancies. Cancer Treat Rev 2013; 39:632-9. [DOI: 10.1016/j.ctrv.2012.10.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 10/01/2012] [Accepted: 10/16/2012] [Indexed: 11/25/2022]
|
229
|
Williams EL, Tutt AL, Beers SA, French RR, Chan CHT, Cox KL, Roghanian A, Penfold CA, Butts CL, Boross P, Verbeek JS, Cragg MS, Glennie MJ. Immunotherapy Targeting Inhibitory Fcγ Receptor IIB (CD32b) in the Mouse Is Limited by Monoclonal Antibody Consumption and Receptor Internalization. THE JOURNAL OF IMMUNOLOGY 2013; 191:4130-40. [DOI: 10.4049/jimmunol.1301430] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
230
|
Laprevotte E, Voisin G, Ysebaert L, Klein C, Daugrois C, Laurent G, Fournie JJ, Quillet-Mary A. Recombinant human IL-15 trans-presentation by B leukemic cells from chronic lymphocytic leukemia induces autologous NK cell proliferation leading to improved anti-CD20 immunotherapy. THE JOURNAL OF IMMUNOLOGY 2013; 191:3634-40. [PMID: 23997218 DOI: 10.4049/jimmunol.1300187] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Recombinant human IL-15 (rhIL-15) is one of the most promising cytokines for antitumor immunotherapy. In physiology IL-15 trans-presentation by accessory cells leads to pleiotropic activities, including activation of immune cells, such as NK cells. NK cells are largely involved in Ab-dependent cellular cytotoxicity mediated by therapeutic mAbs, such as rituximab, in chronic lymphocytic leukemia (CLL). Nevertheless, in CLL, Ab-dependent cellular cytotoxicity is relatively impaired by the low E:T ratio (NK/B leukemic cells). Thus, any strategy leading to an increase in NK cell number and activation status can offer new strategies for CLL treatment. To this end, we evaluated the effect of rhIL-15 on autologous NK cell stimulation in CLL samples. We show that rhIL-15 induces NK cell activation and proliferation, leading to improved B leukemic cell depletion. This phenomenon is significantly increased in the presence of anti-CD20 mAbs. In addition, the greater effect of obinutuzumab versus rituximab suggests a cooperative role between rhIL-15 signaling and CD16 signaling in the induction of NK cell proliferation. Moreover, rhIL-15-induced proliferation of autologous NK cells is strictly dependent on their interaction with B leukemic cells, identified in this study as new accessory cells for rhIL-15 trans-presentation. Thus, rhIL-15 is able to promote NK cell-based activity in Ab immunotherapy of CLL.
Collapse
Affiliation(s)
- Emilie Laprevotte
- INSERM Unité Mixte de Recherche 1037, Cancer Research Center of Toulouse, BP3028 Centre Hospitalier de l'Université Purpan, F-31300 Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
231
|
Chao MP. Treatment challenges in the management of relapsed or refractory non-Hodgkin's lymphoma - novel and emerging therapies. Cancer Manag Res 2013; 5:251-69. [PMID: 24049458 PMCID: PMC3775637 DOI: 10.2147/cmar.s34273] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Over the last few decades, advances in immunochemotherapy have led to dramatic improvement in the prognosis of non-Hodgkin's lymphoma (NHL). Despite these advances, relapsed and refractory disease represents a major treatment challenge. For both aggressive and indolent subtypes of NHL, there is no standard of care for salvage regimens, with prognosis after relapse remaining relatively poor. Nevertheless, there are multiple emerging classes of targeted therapies for relapsed/refractory disease, including monoclonal antibodies, antibody- drug conjugates, radioimmunotherapy, small-molecule inhibitors of cell-growth pathways, and novel chemotherapy agents. This review will discuss treatment challenges of NHL, current available salvage regimens for relapsed/refractory NHL, and the safety and efficacy of novel emerging therapies.
Collapse
Affiliation(s)
- Mark P Chao
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
232
|
Preferential splenic CD8(+) T-cell activation in rituximab-nonresponder patients with immune thrombocytopenia. Blood 2013; 122:2477-86. [PMID: 23963041 DOI: 10.1182/blood-2013-03-491415] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The pathogenic role of B cells in immune thrombocytopenia (ITP) has justified the therapeutic use of anti-CD20 antibodies such as rituximab (RTX). However, 60% of ITP patients do not respond to RTX. To decipher the mechanisms implicated in the failure of RTX, and because the spleen plays a well-recognized role in ITP pathogenesis, 12 spleens from ITP patients who had been nonresponders to RTX therapy were compared with 11 spleens from RTX-untreated ITP patients and 9 controls. We here demonstrate that in RTX-nonresponder ITP patients, preferential Th1 and Tc1 T lymphocyte polarizations occur, associated with an increase in splenic effector memory CD8(+) T-cell frequency. Moreover, in the RTX- nonresponder patient group, the CD8(+) T-cell repertoire displays a restricted pattern. In the blood, the phenotype of CD8(+) T cells before and after RTX treatment is not modified in responders or nonresponders. Altogether, these results demonstrate for the first time an activation of splenic CD8(+) T cells in ITP patients who did not respond to RTX and suggest their involvement in platelet destruction in these patients.
Collapse
|
233
|
McKenzie CGJ, Guo L, Freedman J, Semple JW. Cellular immune dysfunction in immune thrombocytopenia (ITP). Br J Haematol 2013; 163:10-23. [DOI: 10.1111/bjh.12480] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
234
|
Li M, Liu L, Xi N, Wang Y, Xiao X, Zhang W. Imaging and measuring the biophysical properties of Fc gamma receptors on single macrophages using atomic force microscopy. Biochem Biophys Res Commun 2013; 438:709-14. [PMID: 23916706 DOI: 10.1016/j.bbrc.2013.07.114] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 07/28/2013] [Indexed: 12/22/2022]
Abstract
Fc gamma receptors (FcγR), widely expressed on effector cells (e.g., NK cells, macrophages), play an important role in clinical cancer immunotherapy. The binding of FcγRs to the Fc portions of antibodies that are attached to the target cells can activate the antibody-dependent cell-mediated cytotoxicity (ADCC) killing mechanism which leads to the lysis of target cells. In this work, we used atomic force microscopy (AFM) to observe the cellular ultra-structures and measure the biophysical properties (affinity and distribution) of FcγRs on single macrophages in aqueous environments. AFM imaging was used to obtain the topographies of macrophages, revealing the nanoscale cellular fine structures. For molecular interaction recognition, antibody molecules were attached onto AFM tips via a heterobifunctional polyethylene glycol (PEG) crosslinker. With AFM single-molecule force spectroscopy, the binding affinities of FcγRs were quantitatively measured on single macrophages. Adhesion force mapping method was used to localize the FcγRs, revealing the nanoscale distribution of FcγRs on local areas of macrophages. The experimental results can improve our understanding of FcγRs on macrophages; the established approach will facilitate further research on physiological activities involved in antibody-based immunotherapy.
Collapse
Affiliation(s)
- Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China
| | | | | | | | | | | |
Collapse
|
235
|
Taylor RP, Lindorfer MA. The role of complement in mAb-based therapies of cancer. Methods 2013; 65:18-27. [PMID: 23886909 DOI: 10.1016/j.ymeth.2013.07.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 11/26/2022] Open
Abstract
The ability of complement to promote lysis of antibody-opsonized cells is well-established. Virtually all of the molecular details of this reaction have been elucidated and numerous points of regulation have also been delineated. Use of this information, along with the techniques that were first applied in the fundamental studies of complement, has allowed for investigations of the role of complement in mAb-based immunotherapies of cancer. These studies, which have often combined in vitro investigations with parallel correlative clinical measurements, have revealed that several FDA-approved mAbs make use of complement as an effector function in promoting opsonization and killing of targeted malignant cells. We describe the key methods used in this work, and discuss how the results of these studies provide rational approaches for making more effective use of complement in mAb-based cancer immunotherapy.
Collapse
Affiliation(s)
- Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
236
|
Affiliation(s)
- Michael H Kershaw
- Cancer Immunology Program, Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3010 Victoria, Australia
| | | |
Collapse
|
237
|
Li M, Xiao X, Liu L, Xi N, Wang Y, Dong Z, Zhang W. Atomic force microscopy study of the antigen-antibody binding force on patient cancer cells based on ROR1 fluorescence recognition. J Mol Recognit 2013; 26:432-8. [PMID: 23836471 DOI: 10.1002/jmr.2287] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/15/2013] [Accepted: 05/24/2013] [Indexed: 12/12/2022]
Affiliation(s)
| | - Xiubin Xiao
- Department of Lymphoma, Affiliated Hospital of Military Medical Academy of Sciences; Beijing; 100071; China
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences; Shenyang; 110016; China
| | | | - Yuechao Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences; Shenyang; 110016; China
| | - Zaili Dong
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences; Shenyang; 110016; China
| | - Weijing Zhang
- Department of Lymphoma, Affiliated Hospital of Military Medical Academy of Sciences; Beijing; 100071; China
| |
Collapse
|
238
|
Salles GA, Morschhauser F, Solal-Céligny P, Thieblemont C, Lamy T, Tilly H, Gyan E, Lei G, Wenger M, Wassner-Fritsch E, Cartron G. Obinutuzumab (GA101) in patients with relapsed/refractory indolent non-Hodgkin lymphoma: results from the phase II GAUGUIN study. J Clin Oncol 2013; 31:2920-6. [PMID: 23835715 DOI: 10.1200/jco.2012.46.9718] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The phase II part of the phase I/II GAUGUIN study evaluated the efficacy and safety of two different doses of obinutuzumab (GA101), a type II, glycoengineered, humanized anti-CD20 monoclonal antibody, in patients with relapsed/refractory indolent non-Hodgkin lymphoma. PATIENTS AND METHODS Patients were randomly assigned to receive eight cycles of obinutuzumab (GA101) as a flat dose of 400 mg on days 1 and 8 of cycle 1 and also on day 1 of cycles 2 to 8 (400/400 mg) or 1,600 mg on days 1 and 8 of cycle 1 and 800 mg on day 1 of cycles 2 to 8 (1,600/800 mg). RESULTS Forty patients were enrolled, including 34 with follicular lymphoma; 38 of 40 patients had previously received rituximab and 22 of 40 were rituximab refractory. The overall response rate at the end of treatment was 55% (95% CI, 32% to 76%) in the 1,600/800-mg group (9% complete responders) and 17% (95% CI, 4% to 41%) in the 400/400-mg group (no complete responders). Five of 10 rituximab-refractory patients had an end-of-treatment response in the 1,600/800-mg group versus one of 12 in the 400/400-mg group. Median progression-free survival was 11.9 months in the 1,600/800-mg group (range, 1.8 to 33.9+ months) and 6.0 months in the 400/400-mg group (range, 1.0 to 33.9+ months). The most common adverse events were infusion-related reactions (IRRs) seen in 73% of patients, but only two patients had grade 3 to 4 IRRs (both in the 1,600/800-mg group). No IRRs were considered serious, and no patients withdrew for IRRs. CONCLUSION The 1,600/800-mg dose schedule of obinutuzumab (GA101) has encouraging activity with an acceptable safety profile in relapsed/refractory indolent non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Gilles A Salles
- Hospices Civils de Lyon-Université de Lyon, 165 Chemin du Grand Revoyet, 69495 Pierre-Bénite cedex, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Morschhauser FA, Cartron G, Thieblemont C, Solal-Céligny P, Haioun C, Bouabdallah R, Feugier P, Bouabdallah K, Asikanius E, Lei G, Wenger M, Wassner-Fritsch E, Salles GA. Obinutuzumab (GA101) monotherapy in relapsed/refractory diffuse large b-cell lymphoma or mantle-cell lymphoma: results from the phase II GAUGUIN study. J Clin Oncol 2013; 31:2912-9. [PMID: 23835718 DOI: 10.1200/jco.2012.46.9585] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Obinutuzumab (GA101), a type II, glycoengineered, humanized anti-CD20 monoclonal antibody, was superior to rituximab in human diffuse large B-cell lymphoma (DLBCL) and mantle-cell lymphoma (MCL) xenograft models. In phase I of our study, obinutuzumab (GA101) exhibited encouraging activity but no clear dose-response relationship, and few patients had aggressive histologies. The efficacy and safety of two doses of obinutuzumab (GA101) were explored in our randomized phase II trial in patients with heavily pretreated DBLCL and MCL. PATIENTS AND METHODS Patients were randomly assigned to receive eight cycles of obinutuzumab (GA101) either as a flat dose of 400 mg for all infusions (days 1 and 8 of cycle 1; day 1 of cycles 2 to 8) or 1,600 mg on days 1 and 8 of cycle 1 and 800 mg on day 1 of cycles 2 to 8. RESULTS Forty patients were enrolled: 21 patients in the 400/400-mg treatment arm (DLBCL, n = 10; MCL, n = 11) and 19 patients in the 1,600/800-mg arm (DLBCL, n = 15; MCL, n = 4). End-of-treatment response was 28% (32% and 24% in the 1,600/800-mg and 400/400-mg study arms, respectively). Best overall response rates were 37% in the 1,600/800-mg arm and 24% in the 400/400-mg study arm (DLBCL, eight [32%] of 25 patients; MCL, four [27%] of 15 patients). Five (20%) of 25 rituximab-refractory patients exhibited treatment response, including four of 12 in the 1,600/800-mg group. The most common adverse events were infusion-related reactions (IRRs), which were manageable. Three patients had grade 3/4 IRRs. Grade 3/4 neutropenia was seen in only one patient. CONCLUSION Obinutuzumab (GA101) 1,600/800 mg achieves early steady-state concentration and clinical activity with an acceptable safety profile in relapsed/refractory DLBCL and MCL, supporting further exploration.
Collapse
Affiliation(s)
- Franck Andre Morschhauser
- Hematology Department, EA 4481 GRIIOT, Centre Hospitalier Régional Universitaire de Lille, Lille, France.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Abstract
Epratuzumab, a humanized anti-CD22 antibody, is currently in clinical trials of B-cell lymphomas and autoimmune diseases, demonstrating therapeutic activity in non-Hodgkin lymphoma (NHL) and systemic lupus erythematosus (SLE). Thus, epratuzumab offers a promising option for CD22-targeted immunotherapy, yet its mechanism of action remains poorly understood. Here we report for the first time that epratuzumab promptly induces a marked decrease of CD22 (>80%), CD19 (>50%), CD21 (>50%), and CD79b (>30%) on the surface of B cells in peripheral blood mononuclear cells (PBMCs) obtained from normal donors or SLE patients, and of NHL cells (Daudi and Raji) spiked into normal PBMCs. Although some Fc-independent loss of CD22 is expected from internalization by epratuzumab, the concurrent and prominent reduction of CD19, CD21, and CD79b is Fc dependent and results from their transfer from epratuzumab-opsonized B cells to FcγR-expressing monocytes, natural killer cells, and granulocytes via trogocytosis. The findings of reduced levels of CD19 are implicative for the efficacy of epratuzumab in autoimmune diseases because elevated CD19 has been correlated with susceptibility to SLE in animal models as well as in patients. This was confirmed herein by the finding that SLE patients receiving epratuzumab immunotherapy had significantly reduced CD19 compared with treatment-naïve patients.
Collapse
|
241
|
Abstract
PURPOSE OF REVIEW This manuscript reviews current knowledge and recent findings regarding antibody-independent functions of B cells in transplantation. RECENT FINDINGS Until recently the functions of B cells in transplantation have been attributed almost entirely to the antibodies they produce. However, the results of recent trials of B-cell-depleting agents for treatment of antibody-mediated rejection as well as auto-immune disease raised awareness that B cells mediate functions independent of antibody synthesis. SUMMARY These 'nonclassical' functions place B cells at the center of immune regulation with the power to enhance or inhibit immunity.
Collapse
|
242
|
van Egmond M, Bakema JE. Neutrophils as effector cells for antibody-based immunotherapy of cancer. Semin Cancer Biol 2013; 23:190-9. [DOI: 10.1016/j.semcancer.2012.12.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 12/21/2012] [Indexed: 12/24/2022]
|
243
|
Kamburova EG, Koenen HJPM, Borgman KJE, ten Berge IJ, Joosten I, Hilbrands LB. A single dose of rituximab does not deplete B cells in secondary lymphoid organs but alters phenotype and function. Am J Transplant 2013; 13:1503-11. [PMID: 23570303 DOI: 10.1111/ajt.12220] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/31/2013] [Accepted: 02/18/2013] [Indexed: 01/25/2023]
Abstract
A single dose of the anti-CD20 monoclonal antibody rituximab induces a nearly complete B cell depletion in peripheral blood, but not in secondary lymphoid organs. Modulation of this remaining B cell population due to rituximab treatment may contribute to the therapeutic effects of rituximab. To assess the in vivo effects of rituximab we used lymph nodes (LNs) collected during renal transplant surgery in patients who had received rituximab 4 weeks earlier in preparation for an ABO-incompatible transplantation. Rituximab treatment resulted in a lower percentage of naïve (IgD(+)CD27(-)) and a higher percentage of switched memory (IgD(-)CD27(+)) B cells. Remarkably, transitional (CD24(++)CD38(++)) B cells were virtually lacking in the LNs of rituximab-treated patients. Moreover, LN-derived B cells from rituximab-treated patients produced different amounts of various Ig-subclasses after anti-CD40/IL-21 stimulation ex vivo. Finally, after stimulation of allogeneic T cells with LN-derived B cells from rituximab-treated patients, the proliferated T cells showed a decreased production of IL-17. In conclusion, after treatment with rituximab there remains a B cell population with different functional capacities. Consequently, the effect of rituximab on the immune response will not only be determined by the extent of B cell depletion, but also by the functional properties of the remaining B cells.
Collapse
Affiliation(s)
- E G Kamburova
- Department of Laboratory Medicine, Medical Immunology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
244
|
Kurrasch R, Brown JC, Chu M, Craigen J, Overend P, Patel B, Wolfe S, Chang DJ. Subcutaneously administered ofatumumab in rheumatoid arthritis: a phase I/II study of safety, tolerability, pharmacokinetics, and pharmacodynamics. J Rheumatol 2013; 40:1089-96. [PMID: 23729801 DOI: 10.3899/jrheum.121118] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To investigate the safety and tolerability of a single subcutaneous (SC) dose of ofatumumab, a fully human anti-CD20 monoclonal antibody, in patients with rheumatoid arthritis (RA) taking background methotrexate (MTX). Secondary objectives included characterizing pharmacokinetics and pharmacodynamics. METHODS In this single-blind, phase I/II study, 35 patients with RA were randomized in 5 cohorts to receive a single subcutaneous (SC) ofatumumab dose ranging from 0.3 to 100 mg, or placebo, following premedication with oral acetaminophen and antihistamine. Patients were followed for 24 weeks with extended followup to monitor B cell and immunoglobulin recovery for up to 2 years if required. RESULTS Thirty-five patients received the following treatment: 0.3 mg, n = 4; 3 mg, n = 6; 30 mg, n = 8; 60 mg, n = 6; 100 mg, n = 3; placebo, n = 8. The most common adverse events in the combined ofatumumab groups were headache, nausea, and upper respiratory tract infection. Because of tolerability concerns, only 3 patients were given 100 mg. For the 30-100 mg doses, median maximum plasma concentration values ranged from 4.02 to 4.49 days. Mean elimination half-life values ranged from 5.20 to 6.83 days. Increasing peripheral median B cell depletion was observed from 0.3 mg up to 30 mg, and full target B cell depletion was achieved with 30 mg, 60 mg, and 100 mg. CONCLUSION Treatment of RA patients with SC ofatumumab doses of 30 mg or higher resulted in profound and prolonged B cell depletion in blood. Single doses up to 60 mg were tolerated without glucocorticoid premedication. (ClinicalTrials.gov identifier NCT00686868).
Collapse
|
245
|
Raitses Gurevich M, Fishelson Z. Construction and characterization of recombinant human C9 or C7 linked to single chain Fv directed to CD25. Mol Immunol 2013; 55:400-8. [PMID: 23582305 DOI: 10.1016/j.molimm.2013.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 03/13/2013] [Accepted: 03/16/2013] [Indexed: 01/17/2023]
Abstract
Complement-dependent cytotoxicity (CDC) is a potent promoter of tumor clearance during monoclonal antibody therapy. Complement activation on antibody-bearing tumor cells results in formation of the membrane attack complex (MAC), which activates cell death. The complement activation cascade that bridges between antibody binding and MAC formation is regulated by complement inhibitors that are over-expressed on tumor cells. In order to bypass those complement regulators, we have designed an immunoconjugate composed of a humanized single chain Fv of an anti-Tac (CD25) monoclonal antibody fused at its C terminus either to complement protein C9 (scFv-C9) or to complement C7 (scFv-C7) and tagged with six histidines at the C terminal end. Recombinant scFv-C9 and scFv-C7 were expressed in 293T cells and purified. Both are shown to efficiently bind to CD25-positive tumor cells. In addition, scFv-C9, but not scFv-C7, increases MAC deposition on the cells and enhances complement-mediated cell death of target CD25-positive cells. Thus, scFv-C9 fusion protein is potentially a novel reagent for application in cancer immunotherapy.
Collapse
Affiliation(s)
- Maria Raitses Gurevich
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | |
Collapse
|
246
|
Ricklin D, Lambris JD. Complement in immune and inflammatory disorders: pathophysiological mechanisms. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:3831-8. [PMID: 23564577 PMCID: PMC3623009 DOI: 10.4049/jimmunol.1203487] [Citation(s) in RCA: 364] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although acute or chronic inflammation is a common component of many clinical disorders, the underlying processes can be highly distinct. In recent years, the complement system has been associated with a growing number of immunological and inflammatory conditions that include degenerative diseases, cancer, and transplant rejection. It becomes evident that excessive activation or insufficient control of complement activation on host cells can cause an immune imbalance that may fuel a vicious cycle between complement, inflammatory cells, and tissue damage that exacerbates clinical complications. Although the exact involvement of complement needs to be carefully investigated for each disease, therapeutic modulation of complement activity emerges as an attractive target for upstream inhibition of inflammatory processes. This review provides an update about the functional and collaborative capabilities of complement, highlights major disease areas with known complement contribution, and indicates the potential for complement as a focal point in immunomodulatory strategies for treating inflammatory diseases.
Collapse
Affiliation(s)
- Daniel Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
247
|
Blüml S, McKeever K, Ettinger R, Smolen J, Herbst R. B-cell targeted therapeutics in clinical development. Arthritis Res Ther 2013; 15 Suppl 1:S4. [PMID: 23566679 PMCID: PMC3624127 DOI: 10.1186/ar3906] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
B lymphocytes are the source of humoral immunity and are thus a critical component of the adaptive immune system. However, B cells can also be pathogenic and the origin of disease. Deregulated B-cell function has been implicated in several autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, and multiple sclerosis. B cells contribute to pathological immune responses through the secretion of cytokines, costimulation of T cells, antigen presentation, and the production of autoantibodies. DNA-and RNA-containing immune complexes can also induce the production of type I interferons, which further promotes the inflammatory response. B-cell depletion with the CD20 antibody rituximab has provided clinical proof of concept that targeting B cells and the humoral response can result in significant benefit to patients. Consequently, the interest in B-cell targeted therapies has greatly increased in recent years and a number of new biologics exploiting various mechanisms are now in clinical development. This review provides an overview on current developments in the area of B-cell targeted therapies by describing molecules and subpopulations that currently offer themselves as therapeutic targets, the different strategies to target B cells currently under investigation as well as an update on the status of novel therapeutics in clinical development. Emerging data from clinical trials are providing critical insight regarding the role of B cells and autoantibodies in various autoimmune conditions and will guide the development of more efficacious therapeutics and better patient selection.
Collapse
Affiliation(s)
- Stephan Blüml
- MedImmune, LLC, Department of Research, One MedImmune Way, Gaithersburg, MD 20854, USA
| | | | | | | | | |
Collapse
|
248
|
Colliou N, Picard D, Caillot F, Calbo S, Le Corre S, Lim A, Lemercier B, Le Mauff B, Maho-Vaillant M, Jacquot S, Bedane C, Bernard P, Caux F, Prost C, Delaporte E, Doutre MS, Dreno B, Franck N, Ingen-Housz-Oro S, Chosidow O, Pauwels C, Picard C, Roujeau JC, Sigal M, Tancrede-Bohin E, Templier I, Eming R, Hertl M, D'Incan M, Joly P, Musette P. Long-Term Remissions of Severe Pemphigus After Rituximab Therapy Are Associated with Prolonged Failure of Desmoglein B Cell Response. Sci Transl Med 2013; 5:175ra30. [DOI: 10.1126/scitranslmed.3005166] [Citation(s) in RCA: 164] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
249
|
Wang K, Jiang Y, Zheng W, Liu Z, Li H, Lou J, Gu M, Wang X. Silencing of human phosphatidylethanolamine-binding protein 4 enhances rituximab-induced death and chemosensitization in B-cell lymphoma. PLoS One 2013; 8:e56829. [PMID: 23451095 PMCID: PMC3581549 DOI: 10.1371/journal.pone.0056829] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/15/2013] [Indexed: 01/10/2023] Open
Abstract
Rituximab is the first line drug to treat non Hodgkin's lymphoma (B-NHL) alone or in combination with chemotherapy. However, 30-40% of B-NHL patients are unresponsive to rituximab or resistant after therapy. Human phosphatidylethanolamine-binding protein 4 (hPEBP4) is a novel member of PEBP family and functions as an anti-apoptotic molecule. In this study, we found hPEBP4 to be expressed in up to 90% of B-cell lymphoma patients, but in only 16.7% of normal lymph nodes. Interestingly, hPEBP4 overexpression inhibited rituximab-mediated complement dependent cytotoxicity (R-CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC) in B-NHL cells while downregulation of hPEBP4 augmented the therapeutic efficacy of rituximab both in vitro and in vivo. Furthermore, hPEBP4 silencing sensitized the primary B-acute lymphocytic leukemia (B-ALL) cells to R-CDC. During rituximab-mediated complement dependent cytotoxicity, hPEBP4 was recruited to the cell membrane in a PE-binding domain dependent manner and inhibited R-CDC induced calcium flux and reactive oxygen species (ROS) generation. These events contributed to the decrease of cell death induced by R-CDC in B-cell lymphomas. Meanwhile, hPEBP4 knockdown potentiated the chemosensitization of the rituximab in B-cell lymphoma cells by regulating the expression of Bcl-xl, Cycline E, p21(waf/cip1) and p53 and the activation of caspase-3 and caspase-9. Considering that hPEBP4 conferred cellular resistance to rituximab treatment and was preferentially expressed in lymphoma tissue, it could be a potential valuable target for adjuvant therapy for B-cell lymphoma.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Murine-Derived/pharmacology
- Antibodies, Monoclonal, Murine-Derived/therapeutic use
- Apoptosis/drug effects
- Apoptosis/genetics
- Cell Line, Tumor
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Silencing
- Humans
- Immunohistochemistry
- In Vitro Techniques
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Microscopy, Confocal
- Phosphatidylethanolamine Binding Protein/genetics
- Phosphatidylethanolamine Binding Protein/metabolism
- Reactive Oxygen Species/metabolism
- Rituximab
Collapse
Affiliation(s)
- Kai Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yu Jiang
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Weiyan Zheng
- Department of Hematology, First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Zhiyong Liu
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Hui Li
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Jianzhou Lou
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Meidi Gu
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xiaojian Wang
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- * E-mail:
| |
Collapse
|
250
|
Natarajan A, Gowrishankar G, Nielsen CH, Wang S, Iagaru A, Goris ML, Gambhir SS. Positron emission tomography of 64Cu-DOTA-Rituximab in a transgenic mouse model expressing human CD20 for clinical translation to image NHL. Mol Imaging Biol 2013; 14:608-16. [PMID: 22231277 DOI: 10.1007/s11307-011-0537-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
PURPOSE This study aims to evaluate (64)Cu-DOTA-rituximab (PETRIT) in a preclinical transgenic mouse model expressing human CD20 for potential clinical translation. PROCEDURES (64)Cu was chelated to DOTA-rituximab. Multiple radiolabeling, quality assurance, and imaging experiments were performed. The human CD20 antigen was expressed in B cells of transgenic mice (CD20TM). The mice groups studied were: (a) control (nude mice, n = 3) that received 7.4 MBq/dose, (b) with pre-dose (CD20TM, n = 6) received 2 mg/kg pre-dose of cold rituximab prior to PETRIT of 7.4 MBq/dose, and (c) without pre-dose (CD20TM, n = 6) PETRIT alone received 7.4 MBq/dose. Small animal PET was used to image mice at various time points (0, 1, 2, 4, 24, 48, and 72 h). The OLINDA/EXM software was used to determine the human equivalent dose for individual organs. RESULTS PETRIT was obtained with a specific activity of 545 ± 38.91 MBq/nmole, radiochemical purity >95%, and immunoreactivity >75%. At 24 h, spleenic uptake of PETRIT%ID/g (mean ± STD) with and without pre-dose was 1.76 ± 0.43% and 16.5 ± 0.45%, respectively (P value = 0.01). Liver uptake with and without pre-dose was 0.41 ± 0.51% and 0.52 ± 0.17% (P value = 0.86), respectively. The human equivalents of highest dose organs with and without pre-dose are osteogenic cells at 30.8 ± 0.4 μSv/MBq and the spleen at 99 ± 4 μSv/MBq, respectively. CONCLUSIONS PET imaging with PETRIT in huCD20 transgenic mice provided human dosimetry data for eventual applications in non-Hodgkins lymphoma patients.
Collapse
Affiliation(s)
- Arutselvan Natarajan
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA.
| | | | | | | | | | | | | |
Collapse
|