1
|
Forró B, Kajtár B, Lacza Á, Kereskai L, Vida L, Kőszegi B, Urbán P, Kun J, Gyenesei A, Kosztolányi S, Kehl D, Jáksó P. Multiparameter flow cytometric and transcriptional analyis of CD20 positive T-cells in bone marrow in patients of multiple myeloma and monoclonal gammopathy of undetermined significance. Front Immunol 2025; 16:1464940. [PMID: 40079005 PMCID: PMC11896981 DOI: 10.3389/fimmu.2025.1464940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction CD20+ T-cells were described firstly in peripheral blood and later in bone marrow in patients with hematological tumors, and certain immune-mediated diseases. During our hematological diagnostic work, this peculiar subgroup of lymphocytes has been consistently observed associated with untreated monoclonal gammopathy of undetermined significance (MGUS) and myeloma (MM). Despite the expanding literature data, the exact function of CD20+ T cells remains unclear. Methods We investigated the incidence of CD20+ T-cells in MGUS (n=27), and MM using a larger cohort (n=125) and compared it with control bone marrow samples (n=39). We examined their presence before and after treatment in 32 cases with flow cytometry. Comprehensive flow cytometric analysis included the examination of functional (T-cell activation, cytotoxic molecules and T-cell exhaustion) and maturation markers in a large number of cases. In addition RNA sequencing and subsequent bioinformatics analyses were carried out to detect differentially expressed (DE) genes of FACS sorted CD20+ T-cells versus CD20- T-cells. Results and discussion We found that CD20+ T-cells are phenotypically and transcriptionally different from CD20- T-cells. Elevated incidence of CD20+ T-cells in MGUS and MM and the expression of CD8, NKG2D, and CD28 suggests anti-tumor functionality. Increased PD-1 expression indicates T-cell exhaustion which was mostly detected in the samples of patients with a higher tumor percentage. The majority of CD20+ T-cells are effector or effector memory T-cells. Some of the differentially expressed genes suggest antitumor function via regulating T-cell activation pathways, while other genes involved in tumor escape from immune surveillance by suppressing T-cells or by reprogramming T-cells toward T-cell exhaustion. Our findings suggest that CD20+ T-cells may play a vital role both in immune surveillance and immune escape contributing to progression of multiple myeloma.
Collapse
Affiliation(s)
- Barbara Forró
- Department of Pathology, University of Pécs Medical School, Clinical Centre, Pécs, Hungary
| | - Béla Kajtár
- Department of Pathology, University of Pécs Medical School, Clinical Centre, Pécs, Hungary
| | - Ágnes Lacza
- Department of Pathology, University of Pécs Medical School, Clinical Centre, Pécs, Hungary
| | - László Kereskai
- Department of Pathology, University of Pécs Medical School, Clinical Centre, Pécs, Hungary
| | - Livia Vida
- Department of Pathology, University of Pécs Medical School, Clinical Centre, Pécs, Hungary
| | - Balázs Kőszegi
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Péter Urbán
- Genomics and Bioinformatics Core Facility, Szentágothai Research Centre of the University of Pécs, Pécs, Hungary
| | - József Kun
- Genomics and Bioinformatics Core Facility, Szentágothai Research Centre of the University of Pécs, Pécs, Hungary
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs, Hungary
| | - Attila Gyenesei
- Genomics and Bioinformatics Core Facility, Szentágothai Research Centre of the University of Pécs, Pécs, Hungary
| | - Szabolcs Kosztolányi
- 1st Department of Internal Medicine, University of Pécs Medical School, Pécs, Hungary
| | - Dániel Kehl
- Faculty of Business and Economics, University of Pécs, Pécs, Hungary
| | - Pál Jáksó
- Department of Pathology, University of Pécs Medical School, Clinical Centre, Pécs, Hungary
| |
Collapse
|
2
|
Mok TC, Mok CC. Non-TNF biologics and their biosimilars in rheumatoid arthritis. Expert Opin Biol Ther 2024; 24:599-613. [PMID: 38766765 DOI: 10.1080/14712598.2024.2358165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic inflammatory rheumatic disease that affects both the articular and extra-articular structures, leading to significant joint damage, disability and excess mortality. The treatment algorithm of RA has changed tremendously in the past 1-2 decades because of the emergence of novel biological therapies that target different mechanisms of action in addition to TNFα. AREAS COVERED This article summarizes the evidence and safety of the non-TNF biological DMARDs in the treatment of RA, including those that target B cells, T-cell co-stimulation, interleukin (IL)-6 and granulocyte-monocyte colony-stimulating factor (GM-CSF). The targeted synthetic DMARDs such as the Janus kinase inhibitors are not included. The availability of the less costly biosimilars has enabled more patients to receive biological therapy earlier in the course of the disease. The evidence for the non-TNF biosimilar compounds in RA is also reviewed. EXPERT OPINION There are unmet needs of developing novel therapeutic agents to enhance the response rate and provide more options for difficult-to-treat RA. These include the newer generation biologic and targeted synthetic DMARDs. A personalized treatment strategy in RA requires evaluation of the cellular, cytokine, genomic and transcriptomic profile that would predict treatment response to biologic or targeted DMARDs of different mechanisms of action.
Collapse
Affiliation(s)
- Tsz Ching Mok
- Department of Medicine, Ruttonjee Hospital, Hong Kong, China
| | - Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, Hong Kong, China
| |
Collapse
|
3
|
Fernandes DA. Review on Metal-Based Theranostic Nanoparticles for Cancer Therapy and Imaging. Technol Cancer Res Treat 2023; 22:15330338231191493. [PMID: 37642945 PMCID: PMC10467409 DOI: 10.1177/15330338231191493] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 08/31/2023] Open
Abstract
Theranostic agents are promising due to their ability to diagnose, treat and monitor different types of cancer using a variety of imaging modalities. The advantage specifically of nanoparticles is that they can accumulate easily at the tumor site due to the large gaps in blood vessels near tumors. Such high concentration of theranostic agents at the target site can lead to enhancement in both imaging and therapy. This article provides an overview of nanoparticles that have been used for cancer theranostics, and the different imaging, treatment options and signaling pathways that are important when using nanoparticles for cancer theranostics. In particular, nanoparticles made of metal elements are emphasized due to their wide applications in cancer theranostics. One important aspect discussed is the ability to combine different types of metals in one nanoplatform for use as multimodal imaging and therapeutic agents for cancer.
Collapse
|
4
|
Lindorfer MA, Taylor RP. FcγR-Mediated Trogocytosis 2.0: Revisiting History Gives Rise to a Unifying Hypothesis. Antibodies (Basel) 2022; 11:antib11030045. [PMID: 35892705 PMCID: PMC9326535 DOI: 10.3390/antib11030045] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 12/25/2022] Open
Abstract
There is increasing interest in the clinical implications and immunology of trogocytosis, a process in which the receptors on acceptor cells remove and internalize cognate ligands from donor cells. We have reported that this phenomenon occurs in cancer immunotherapy, in which cells that express FcγR remove and internalize CD20 and bound mAbs from malignant B cells. This process can be generalized to include other reactions including the immune adherence phenomenon and antibody-induced immunosuppression. We discuss in detail FcγR-mediated trogocytosis and the evidence supporting a proposed predominant role for liver sinusoidal endothelial cells via the action of the inhibitory receptor FcγRIIb2. We describe experiments to test the validity of this hypothesis. The elucidation of the details of FcγR-mediated trogocytosis has the potential to allow for the development of novel therapies that can potentially block or enhance this reaction, depending upon whether the process leads to unfavorable or positive biological effects.
Collapse
|
5
|
The Multiple Roles of Trogocytosis in Immunity, the Nervous System, and Development. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1601565. [PMID: 34604381 PMCID: PMC8483919 DOI: 10.1155/2021/1601565] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022]
Abstract
Trogocytosis is a general biological process that involves one cell physically taking small parts of the membrane and other components from another cell. In trogocytosis, one cell seems to take little “bites” from another cell resulting in multiple outcomes from these cell-cell interactions. Trogocytosis was first described in protozoan parasites, which by taking pieces of host cells, kill them and cause tissue damage. Now, it is known that this process is also performed by cells of the immune system with important consequences such as cell communication and activation, elimination of microbial pathogens, and even control of cancer cells. More recently, trogocytosis has also been reported to occur in cells of the central nervous system and in various cells during development. Some of the molecules involved in phagocytosis also participate in trogocytosis. However, the molecular mechanisms that regulate trogocytosis are still a mystery. Elucidating these mechanisms is becoming a research area of much interest. For example, why neutrophils can engage trogocytosis to kill Trichomonas vaginalis parasites, but neutrophils use phagocytosis to eliminate already death parasites? Thus, trogocytosis is a significant process in normal physiology that multiple cells from different organisms use in various scenarios of health and disease. In this review, we present the basic principles known on the process of trogocytosis and discuss the importance in this process to host-pathogen interactions and to normal functions in the immune and nervous systems.
Collapse
|
6
|
Tsao LC, Force J, Hartman ZC. Mechanisms of Therapeutic Antitumor Monoclonal Antibodies. Cancer Res 2021; 81:4641-4651. [PMID: 34145037 PMCID: PMC8448950 DOI: 10.1158/0008-5472.can-21-1109] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/24/2021] [Accepted: 06/16/2021] [Indexed: 11/16/2022]
Abstract
Monoclonal antibodies (mAb) are a major component of cancer therapy. In this review, we summarize the different therapeutic mAbs that have been successfully developed against various tumor-expressed antigens and examine our current understanding of their different mechanisms of antitumor action. These mechanisms of action (MOA) largely center on the stimulation of different innate immune effector processes, which appear to be principally responsible for the efficacy of most unconjugated mAb therapies against cancer. This is evident in studies of mAbs targeting antigens for hematologic cancers, with emerging data also demonstrating the critical nature of innate immune-mediated mechanisms in the efficacy of anti-HER2 mAbs against solid HER2+ cancers. Although HER2-targeted mAbs were originally described as inhibitors of HER2-mediated signaling, multiple studies have since demonstrated these mAbs function largely through their engagement with Fc receptors to activate innate immune effector functions as well as complement activity. Next-generation mAbs are capitalizing on these MOAs through improvements to enhance Fc-activity, although regulation of these mechanisms may vary in different tumor microenvironments. In addition, novel antibody-drug conjugates have emerged as an important means to activate different MOAs. Although many unknowns remain, an improved understanding of these immunologic MOAs will be essential for the future of mAb therapy and cancer immunotherapy.
Collapse
Affiliation(s)
- Li-Chung Tsao
- Department of Surgery, Duke University, Durham, North Carolina
| | - Jeremy Force
- Department of Medicine, Duke University, Durham, North Carolina
| | - Zachary C Hartman
- Department of Surgery, Duke University, Durham, North Carolina.
- Department of Pathology, Duke University, Durham, North Carolina
| |
Collapse
|
7
|
McBride HJ, Jassem S, Chow V, Kanakaraj P, Lebrec H, Kuhns S, Ferbas J, Wong M, Thway TM. Non-clinical similarity of biosimilar ABP 798 with rituximab reference product. Biologicals 2021; 72:42-53. [PMID: 34303595 DOI: 10.1016/j.biologicals.2021.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 05/13/2021] [Accepted: 05/20/2021] [Indexed: 01/02/2023] Open
Abstract
ABP 798 is a biosimilar to Rituxan® (rituximab reference product [RP]). Non-clinical assessments relevant to the primary and secondary mechanisms of action (MOA) contribute to the totality of the evidence (TOE) in supporting biosimilarity and are critical in providing scientific evidence for extrapolation of indications. Similarity of ABP 798 with rituximab RP was investigated across a range of biological activities which have potential impact on pharmacokinetics and clinical efficacy with non-clinical assessments relevant to MOA such as CD20 internalization, trogocytosis, binding to primary human natural killer (NK) cells as well as the ability to induce antibody-dependent cellular phagocytosis (ADCP) in peripheral blood mononuclear cells. Additionally, in vitro synergy of ABP 798 or RP with chemotherapeutic agents, in vivo xenograft studies in mice, and toxicological assessments in cynomolgus monkeys (including B cell depletion and toxicokinetics) were also conducted. Results from these non-clinical assessments contribute to the TOE supporting the biosimilarity between ABP 798 and rituximab RP across a range of primary and secondary MOAs and support justification for extrapolation to all indications of use for ABP 798 for which the RP is approved.
Collapse
Affiliation(s)
- Helen J McBride
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | - Shea Jassem
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | - Vincent Chow
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | | | - Herve Lebrec
- Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA.
| | - Scott Kuhns
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | - John Ferbas
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | - Min Wong
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | - Theingi M Thway
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| |
Collapse
|
8
|
Desai S, Mo C, Gaglione EM, Yuan CM, Stetler-Stevenson M, Tian X, Maric I, Wake L, Farooqui MZ, Drinkwater DC, Soto S, Valdez J, Hughes TE, Nierman P, Lotter J, Marti GE, Pleyer C, Sun C, Superata J, Nichols C, Herman SEM, Lindorfer MA, Taylor RP, Wiestner A, Ahn IE. Risk-adapted, ofatumumab-based chemoimmunotherapy and consolidation in treatment-naïve chronic lymphocytic leukemia: a phase 2 study. Leuk Lymphoma 2021; 62:1816-1827. [PMID: 33653216 DOI: 10.1080/10428194.2021.1888379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
High-risk cytogenetics and minimal residual disease (MRD) after chemoimmunotherapy (CIT) predict unfavorable outcome in chronic lymphocytic leukemia (CLL). This phase 2 study investigated risk-adapted CIT in treatment-naïve CLL (NCT01145209). Patients with high-risk cytogenetics received induction with fludarabine, cyclophosphamide, and ofatumumab. Those without high-risk cytogenetics received fludarabine and ofatumumab. After induction, MRD positive (MRD+) patients received 4 doses of ofatumumab consolidation. MRD negative (MRD-) patients had no intervention. Of 28 evaluable for response, all responded to induction and 10 (36%) achieved MRD-. Two-year progression-free survival (PFS) was 71.4% (CI95, 56.5-90.3%). There was no significant difference in median PFS between the high-risk and the standard-risk groups. Ofatumumab consolidation didn't convert MRD + to MRD-. In the MRD + group, we saw selective loss of CD20 antigens during therapy. In conclusion, risk-adapted CIT is feasible in treatment-naïve CLL. Ofatumumab consolidation didn't improve depth of response in MRD + patients. Loss of targetable CD20 likely reduces efficacy of consolidation therapy.
Collapse
Affiliation(s)
- Sanjal Desai
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA.,Medstar Washington Hospital Center, Washington, D.C., USA
| | - Clifton Mo
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA.,Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Erika M Gaglione
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA.,Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Constance M Yuan
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Xin Tian
- Office of Biostatistics Research, NHLBI, NIH, Bethesda, MD, USA
| | - Irina Maric
- Department of Laboratory Medicine, Clinical Research Center, NIH, Bethesda, MD, USA
| | - Laura Wake
- Department of Laboratory Medicine, Clinical Research Center, NIH, Bethesda, MD, USA
| | - Mohammed Z Farooqui
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | - Susan Soto
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Janet Valdez
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Thomas E Hughes
- Department of Pharmacy, Clinical Research Center, NIH, Bethesda, MD, USA
| | - Pia Nierman
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jennifer Lotter
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Gerald E Marti
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Christopher Pleyer
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Clare Sun
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jeanine Superata
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Cydney Nichols
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Sarah E M Herman
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Inhye E Ahn
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
9
|
Felberg A, Taszner M, Urban A, Majeranowski A, Jaskuła K, Jurkiewicz A, Stasiłojć G, Blom AM, Zaucha JM, Okrój M. Monitoring of the Complement System Status in Patients With B-Cell Malignancies Treated With Rituximab. Front Immunol 2020; 11:584509. [PMID: 33329558 PMCID: PMC7710700 DOI: 10.3389/fimmu.2020.584509] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
Rituximab is a pioneering anti-CD20 monoclonal antibody that became the first-line drug used in immunotherapy of B-cell malignancies over the last twenty years. Rituximab activates the complement system in vitro, but there is an ongoing debate on the exact role of this effector mechanism in therapeutic effect. Results of both in vitro and in vivo studies are model-dependent and preclude clear clinical conclusions. Additional confounding factors like complement inhibition by tumor cells, loss of target antigen and complement depletion due to excessively applied immunotherapeutics, intrapersonal variability in the concentration of main complement components and differences in tumor burden all suggest that a personalized approach is the best strategy for optimization of rituximab dosage and therapeutic schedule. Herein we critically review the existing knowledge in support of such concept and present original data on markers of complement activation, complement consumption, and rituximab accumulation in plasma of patients with chronic lymphocytic leukemia (CLL) and non-Hodgkin’s lymphomas (NHL). The increase of markers such as C4d and terminal complement complex (TCC) suggest the strongest complement activation after the first administration of rituximab, but not indicative of clinical outcome in patients receiving rituximab in combination with chemotherapy. Both ELISA and complement-dependent cytotoxicity (CDC) functional assay showed that a substantial number of patients accumulate rituximab to the extent that consecutive infusions do not improve the cytotoxic capacity of their sera. Our data suggest that individual assessment of CDC activity and rituximab concentration in plasma may support clinicians’ decisions on further drug infusions, or instead prescribing a therapy with anti-CD20 antibodies like obinutuzumab that more efficiently activate effector mechanisms other than complement.
Collapse
Affiliation(s)
- Anna Felberg
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Michał Taszner
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Urban
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Alan Majeranowski
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Kinga Jaskuła
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Jurkiewicz
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Grzegorz Stasiłojć
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Anna M Blom
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Jan M Zaucha
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Marcin Okrój
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
10
|
Zhukova OV, Kovaleva TF, Arkhipova EV, Ryabov SA, Mukhina IV. Tumor-associated macrophages: Role in the pathological process of tumorigenesis and prospective therapeutic use (Review). Biomed Rep 2020; 13:47. [PMID: 32934819 DOI: 10.3892/br.2020.1354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 07/10/2020] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to evaluate the current body of knowledge regarding tumor-associated macrophages (TAMs) and their potential use in antitumor therapy, based on their role in the pathological process of tumorigenesis. For this purpose, a critical analysis of published data and summarization of the findings available from original studies, focusing on the role of TAMs in the pathological process, and their potential therapeutic application was performed. Promising key avenues of research were identified in this field. The following issues seem the most promising and thus worth further investigation: i) The process of M1/M2 macrophage polarization, macrophage characteristics at intermediate polarization steps and their role in the tumor process; ii) determining the conditions necessary for transitions between the M1 and M2 macrophage phenotypes and the role of signals from the microenvironment in this process; iii) cause-and-effect associations between the quantity and quality of macrophages, and the prognosis and outcome of the pathological process; iv) modulation of macrophages and stimulation of their phagocytic activity with drugs; v) targeted vector-based systems for drug delivery to macrophages; and vi) targeted drug delivery systems with macrophages as carriers, thus potentially combining chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Olga V Zhukova
- Department of Pharmaceutical Technology, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| | - Tatiana F Kovaleva
- Department of Molecular and Cellular Technologies, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| | - Evgenia V Arkhipova
- Pre-Clinical Research Center, Central Research Laboratory, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| | - Sergey A Ryabov
- Department of High-Molecular and Colloid Chemistry, National Research Lobachevsky State University, Nizhny Novgorod 603950, Russia
| | - Irina V Mukhina
- Fundamental Medicine Institute and Physiology Department, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| |
Collapse
|
11
|
Abstract
Trogocytosis is part of an emerging, exciting theme of cell-cell interactions both within and between species, and it is relevant to host-pathogen interactions in many different contexts. Trogocytosis is a process in which one cell physically extracts and ingests "bites" of cellular material from another cell. It was first described in eukaryotic microbes, where it was uncovered as a mechanism by which amoebae kill cells. Trogocytosis is potentially a fundamental form of eukaryotic cell-cell interaction, since it also occurs in multicellular organisms, where it has functions in the immune system, in the central nervous system, and during development. There are numerous scenarios in which trogocytosis occurs and an ever-evolving list of functions associated with this process. Many aspects of trogocytosis are relevant to microbial pathogenesis. It was recently discovered that immune cells perform trogocytosis to kill Trichomonas vaginalis parasites. Additionally, through trogocytosis, Entamoeba histolytica acquires and displays human cell membrane proteins, enabling immune evasion. Intracellular bacteria seem to exploit host cell trogocytosis, since they can use it to spread from cell to cell. Thus, a picture is emerging in which trogocytosis plays critical roles in normal physiology, infection, and disease.
Collapse
|
12
|
Mechanisms of Resistance to Anti-CD38 Daratumumab in Multiple Myeloma. Cells 2020; 9:cells9010167. [PMID: 31936617 PMCID: PMC7017193 DOI: 10.3390/cells9010167] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023] Open
Abstract
Daratumumab (Dara) is the first-in-class human-specific anti-CD38 mAb approved for the treatment of multiple myeloma (MM). Although recent data have demonstrated very promising results in clinical practice and trials, some patients do not achieve a partial response, and ultimately all patients undergo progression. Dara exerts anti-MM activity via antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), complement-dependent cytotoxicity (CDC), and immunomodulatory effects. Deregulation of these pleiotropic mechanisms may cause development of Dara resistance. Knowledge of this resistance may improve the therapeutic management of MM patients.
Collapse
|
13
|
Calcein release assay as a method for monitoring serum complement activity during monoclonal antibody therapy in patients with B-cell malignancies. J Immunol Methods 2019; 476:112675. [PMID: 31629742 DOI: 10.1016/j.jim.2019.112675] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/09/2019] [Accepted: 09/23/2019] [Indexed: 11/23/2022]
Abstract
Monoclonal antibodies ofatumumab (anti-CD20) and alemtuzumab (anti-CD52) which are approved for usage in patients with chronic lymphocytic leukemia (CLL), efficiently activate the classical complement pathway. However complement is an exhaustible component and high doses of its activators may deplete complement-dependent cytotoxicity (CDC) potential, thus reducing the effect of repeated mAb dosing. Widely used method to measure CDC activity of patients' serum is hemolytic assay (CH50) on sheep erythrocytes. Despite its simplicity, such CH50 assay may not reflect pivotal interactions between patient serum and human complement inhibitors on the surface of target cells. We propose calcein release assay performed on tumor cells similar to those targeted by therapeutic antibodies as an alternative method. We analyzed serum samples collected from 12 patients participating in the clinical study, receiving s.c. 30 mg alemtuzumab three times per week combined with i.v. ofatumumab at an initial dose of 300 mg in week 3 further escalated to 2000 mg every other week. All serum samples were measured by hemolytic assay on sheep erythrocytes as well as using calcein release assay on CD20-positive Raji cells. Our data show that results obtained from both assays are related to each other at the level of the whole group (n = 96 samples, Spearman r = 0.504, p < .001) but may substantially differ when analyzing individual patients. Furthermore, by using CDC assay on Raji cells, we found that in the presented clinical study CDC serum potential was not significantly affected when measured before consecutive administrations in most of the patients.
Collapse
|
14
|
Lei B, He A, Cao X, Chen Y, Ma X, Zhou F, Zhang P, Qian L, Zhang W. Regulation of expression of MLAA-34 gene through transcriptional factors E2F1 and MZF-1. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2100-2110. [PMID: 31934032 PMCID: PMC6949652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 04/23/2019] [Indexed: 06/10/2023]
Abstract
Approximately 20% of adult patients with acute myeloid leukemia fail to achieve remission with initial induction chemotherapy, and around half ultimately experience relapse after achieving complete remission. Relapse continues to be a major hurdle in achieving cure after obtaining remission with induction chemotherapy in patients with acute myeloid leukemia. In last two decades, the immunogenic vaccine, involving peptide, protein, or DNA, has brought new perspectives for tumor immunotherapy. MLAA-34 is a newly identified monocytic leukemia-associated antigen. Downregulation of MLAA-34 expression significantly suppressed the proliferation of U937 cells in vitro and increased the spontaneous apoptosis of leukemia. However, the regulatory mechanisms of MLAA-34 gene are still unknown at present. Analysis of the promoter region of the MLAA-34 gene and reporter gene assays revealed that 600 bp core region was responsible for its regulation. In addition, our study indicated that E2F1 acts as a transcription repressor and MZF-1 acts as a transcription activator of the MLAA-34 gene.
Collapse
Affiliation(s)
- Bo Lei
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi Province, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi Province, China
| | - Xingmei Cao
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi Province, China
| | - Yinxia Chen
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi Province, China
| | - Xiaorong Ma
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi Province, China
| | - Fuling Zhou
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi Province, China
| | - Pengyu Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi Province, China
| | - Lu Qian
- Department of Hematology, Xi’an No.4 HospitalXi’an, Shaanxi Province, China
| | - Wanggang Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi Province, China
| |
Collapse
|
15
|
Oostindie SC, van der Horst HJ, Lindorfer MA, Cook EM, Tupitza JC, Zent CS, Burack R, VanDerMeid KR, Strumane K, Chamuleau MED, Mutis T, de Jong RN, Schuurman J, Breij ECW, Beurskens FJ, Parren PWHI, Taylor RP. CD20 and CD37 antibodies synergize to activate complement by Fc-mediated clustering. Haematologica 2019; 104:1841-1852. [PMID: 30792198 PMCID: PMC6717598 DOI: 10.3324/haematol.2018.207266] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/19/2019] [Indexed: 11/24/2022] Open
Abstract
CD20 monoclonal antibody therapies have significantly improved the outlook for patients with B-cell malignancies. However, many patients acquire resistance, demonstrating the need for new and improved drugs. We previously demonstrated that the natural process of antibody hexamer formation on targeted cells allows for optimal induction of complement-dependent cytotoxicity. Complement-dependent cytotoxicity can be potentiated by introducing a single point mutation such as E430G in the IgG Fc domain that enhances intermolecular Fc-Fc interactions between cell-bound IgG molecules, thereby facilitating IgG hexamer formation. Antibodies specific for CD37, a target that is abundantly expressed on healthy and malignant B cells, are generally poor inducers of complement-dependent cytotoxicity. Here we demonstrate that introduction of the hexamerization-enhancing mutation E430G in CD37-specific antibodies facilitates highly potent complement-dependent cytotoxicity in chronic lymphocytic leukemia cells ex vivo. Strikingly, we observed that combinations of hexamerization-enhanced CD20 and CD37 antibodies cooperated in C1q binding and induced superior and synergistic complement-dependent cytotoxicity in patient-derived cancer cells compared to the single agents. Furthermore, CD20 and CD37 antibodies colocalized on the cell membrane, an effect that was potentiated by the hexamerization-enhancing mutation. Moreover, upon cell surface binding, CD20 and CD37 antibodies were shown to form mixed hexameric antibody complexes consisting of both antibodies each bound to their own cognate target, so-called hetero-hexamers. These findings provide novel insights into the mechanisms of synergy in antibody-mediated complement-dependent cytotoxicity and provide a rationale to explore Fc-engineering and antibody hetero-hexamerization as a tool to enhance the cooperativity and therapeutic efficacy of antibody combinations.
Collapse
Affiliation(s)
- Simone C Oostindie
- Genmab, Utrecht, the Netherlands .,Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Hilma J van der Horst
- Department of Hematology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Erika M Cook
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jillian C Tupitza
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Clive S Zent
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Richard Burack
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Karl R VanDerMeid
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Martine E D Chamuleau
- Department of Hematology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Tuna Mutis
- Department of Hematology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | | | | | | | | | - Paul W H I Parren
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands.,Lava Therapeutics, Utrecht, the Netherlands
| | - Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
16
|
Felberg A, Urban A, Borowska A, Stasiłojć G, Taszner M, Hellmann A, Blom AM, Okrój M. Mutations resulting in the formation of hyperactive complement convertases support cytocidal effect of anti-CD20 immunotherapeutics. Cancer Immunol Immunother 2019; 68:587-598. [PMID: 30725204 PMCID: PMC6447516 DOI: 10.1007/s00262-019-02304-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 01/17/2019] [Indexed: 12/16/2022]
Abstract
Anti-CD20 monoclonal antibodies (mAbs) rituximab and ofatumumab are potent activators of the classical complement pathway, and have been approved for the treatment of B-cell malignancies. However, complement exhaustion and overexpression of complement inhibitors by cancer cells diminish their therapeutic potential. The strategies of targeting membrane complement inhibitors by function-blocking antibodies and the supplementation with fresh frozen plasma have been proposed to overcome tumour cell resistance. We present a novel approach, which utilizes gain-of-function variants of complement factor B (FB), a component of alternative C3/C5 convertases, which augment mAb-activated reactions through a positive feedback mechanism called an amplification loop. If complement concentration is limited, an addition of quadruple gain-of-function FB mutant p.D279G p.F286L p.K323E p.Y363A (or selected single mutants) results in significantly increased complement-mediated lysis of ofatumumab-resistant tumour cells, as well as the complete lysis of moderately sensitive cells. Importantly, this effect cannot be achieved by further increasing ofatumumab concentration. Potentiation of cytotoxic effect towards moderately sensitive cells was less apparent at physiological serum concentration. However, an addition of hyperactive FB could compensate the loss of cytotoxic potential of serum collected from the NHL and CLL patients after infusion of rituximab. Residual levels of rituximab in such sera, in combination with added FB, were able to efficiently lyse tumour cells. We suggest that the administration of gain-of-function variants of FB can restore cytotoxic potential of complement-exhausted serum and maximize the therapeutic effect of circulating anti-CD20 mAbs.
Collapse
Affiliation(s)
- Anna Felberg
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1 Street, 80-211, Gdańsk, Poland
| | - Aleksandra Urban
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1 Street, 80-211, Gdańsk, Poland
| | - Anna Borowska
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1 Street, 80-211, Gdańsk, Poland
| | - Grzegorz Stasiłojć
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1 Street, 80-211, Gdańsk, Poland
| | - Michał Taszner
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Andrzej Hellmann
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Anna Maria Blom
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Marcin Okrój
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1 Street, 80-211, Gdańsk, Poland.
| |
Collapse
|
17
|
Krejcik J, Frerichs KA, Nijhof IS, van Kessel B, van Velzen JF, Bloem AC, Broekmans MEC, Zweegman S, van Meerloo J, Musters RJP, Poddighe PJ, Groen RWJ, Chiu C, Plesner T, Lokhorst HM, Sasser AK, Mutis T, van de Donk NWCJ. Monocytes and Granulocytes Reduce CD38 Expression Levels on Myeloma Cells in Patients Treated with Daratumumab. Clin Cancer Res 2017; 23:7498-7511. [PMID: 29025767 PMCID: PMC5732844 DOI: 10.1158/1078-0432.ccr-17-2027] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/01/2017] [Accepted: 09/28/2017] [Indexed: 12/21/2022]
Abstract
Purpose: Daratumumab treatment results in a marked reduction of CD38 expression on multiple myeloma cells. The aim of this study was to investigate the clinical implications and the underlying mechanisms of daratumumab-mediated CD38 reduction.Experimental Design: We evaluated the effect of daratumumab alone or in combination with lenalidomide-dexamethasone, on CD38 levels of multiple myeloma cells and nontumor immune cells in the GEN501 study (daratumumab monotherapy) and the GEN503 study (daratumumab combined with lenalidomide-dexamethasone). In vitro assays were also performed.Results: In both trials, daratumumab reduced CD38 expression on multiple myeloma cells within hours after starting the first infusion, regardless of depth and duration of the response. In addition, CD38 expression on nontumor immune cells, including natural killer cells, T cells, B cells, and monocytes, was also reduced irrespective of alterations in their absolute numbers during therapy. In-depth analyses revealed that CD38 levels of multiple myeloma cells were only reduced in the presence of complement or effector cells, suggesting that the rapid elimination of CD38high multiple myeloma cells can contribute to CD38 reduction. In addition, we discovered that daratumumab-CD38 complexes and accompanying cell membrane were actively transferred from multiple myeloma cells to monocytes and granulocytes. This process of trogocytosis was also associated with reduced surface levels of some other membrane proteins, including CD49d, CD56, and CD138.Conclusions: Daratumumab rapidly reduced CD38 expression levels, at least in part, through trogocytosis. Importantly, all these effects also occurred in patients with deep and durable responses, thus excluding CD38 reduction alone as a mechanism of daratumumab resistance.The trials were registered at www.clinicaltrials.gov as NCT00574288 (GEN501) and NCT1615029 (GEN503). Clin Cancer Res; 23(24); 7498-511. ©2017 AACR.
Collapse
Affiliation(s)
- Jakub Krejcik
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
- Vejle Hospital and University of Southern Denmark, Vejle, Denmark
| | - Kris A Frerichs
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Inger S Nijhof
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Berris van Kessel
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Jeroen F van Velzen
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Andries C Bloem
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Johan van Meerloo
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - René J P Musters
- Department of Physiology, VU University, Amsterdam, the Netherlands
| | - Pino J Poddighe
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands
| | - Richard W J Groen
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | | | - Torben Plesner
- Vejle Hospital and University of Southern Denmark, Vejle, Denmark
| | - Henk M Lokhorst
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - A Kate Sasser
- Janssen Research and Development, Spring House, Pennsylvania
| | - Tuna Mutis
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | | |
Collapse
|
18
|
Saesoo S, Sathornsumetee S, Anekwiang P, Treetidnipa C, Thuwajit P, Bunthot S, Maneeprakorn W, Maurizi L, Hofmann H, Rungsardthong RU, Saengkrit N. Characterization of liposome-containing SPIONs conjugated with anti-CD20 developed as a novel theranostic agent for central nervous system lymphoma. Colloids Surf B Biointerfaces 2017; 161:497-507. [PMID: 29128836 DOI: 10.1016/j.colsurfb.2017.11.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/19/2017] [Accepted: 11/01/2017] [Indexed: 11/29/2022]
Abstract
Despite advances in neuroscience cancer research during the past decades, the survival of cancer patients has only marginally improved and the cure remains unlikely. The blood-brain barrier (BBB) is a major obstacle protecting the entry of therapeutic agents to central nervous system, especially for primary central nervous system lymphoma (PCNSL). Thus, the use of small nanoparticle as a drug carrier may be new strategies to overcome this problem. In this study, we fabricated liposome consisting of superparamagnetic iron oxide nanoparticles (SPIONs) functionalized with anti-CD20 (Rituximab; RTX). The designed nanoparticles have a theranostic property which is not only to improve drug delivery, but also to offer diagnostic and monitoring capabilities. TEM images revealed the spherical shape of liposome with the approximately average diameters about 140-190nm with slightly negatively charge surfaces. Superparamagnetic property of SPIONs-loaded liposomes was confirmed by VSM. Liposome colloidal could be prolonged at 4°C and 25°C storages. RTX conjugated liposome induced cell internalization and apoptosis effect in B-lymphoma cells. Drug targeting and therapeutic effect was investigated in BBB model. The result confirmed that liposome nanocarrier is required as a drug carrier for effectively RTX across the BBB.
Collapse
Affiliation(s)
- S Saesoo
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand
| | - S Sathornsumetee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand; Departments of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University 2 Wanglang Road, Bangkoknoi, Bangkok 10700, Thailand, Thailand
| | - P Anekwiang
- NANOTEC-Mahidol University Center of Excellence in Nanotechnology for Cancer Diagnosis and Treatment, Faculty of Medicine Siriraj Hospital, Mahidol University 2 Wanglang Road, Bangkoknoi, Bangkok 10700, Thailand, Thailand
| | - C Treetidnipa
- NANOTEC-Mahidol University Center of Excellence in Nanotechnology for Cancer Diagnosis and Treatment, Faculty of Medicine Siriraj Hospital, Mahidol University 2 Wanglang Road, Bangkoknoi, Bangkok 10700, Thailand, Thailand
| | - P Thuwajit
- NANOTEC-Mahidol University Center of Excellence in Nanotechnology for Cancer Diagnosis and Treatment, Faculty of Medicine Siriraj Hospital, Mahidol University 2 Wanglang Road, Bangkoknoi, Bangkok 10700, Thailand, Thailand; Departments of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University 2 Wanglang Road, Bangkoknoi, Bangkok 10700, Thailand, Thailand
| | - S Bunthot
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand
| | - W Maneeprakorn
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand
| | - L Maurizi
- Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR 6303 CNRS - Université Bourgogne Franche-Comté, BP 47870, F-21078 Dijon cedex, France
| | - H Hofmann
- Powder Technology Laboratory, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | | | - N Saengkrit
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand.
| |
Collapse
|
19
|
Schmitt C, Marie-Cardine A, Bensussan A. Therapeutic Antibodies to KIR3DL2 and Other Target Antigens on Cutaneous T-Cell Lymphomas. Front Immunol 2017; 8:1010. [PMID: 28912774 PMCID: PMC5582066 DOI: 10.3389/fimmu.2017.01010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/07/2017] [Indexed: 11/13/2022] Open
Abstract
KIR3DL2 is a member of the killer cell immunoglobulin-like receptor (KIR) family that was initially identified at the surface of natural killer (NK) cells. KIR3DL2, also known as CD158k, is expressed as a disulfide-linked homodimer. Each chain is composed of three immunoglobulin-like domains and a long cytoplasmic tail containing two immunoreceptor tyrosine-based inhibitory motifs. Beside its expression on NK cells, it is also found on rare circulating T lymphocytes, mainly CD8+. Although the KIR gene number varies between haplotype, KIR3DL2 is a framework gene present in all individuals. Together with the presence of genomic regulatory sequences unique to KIR3DL2, this suggests some particular functions for the derived protein in comparison with other KIR family members. Several ligands have been identified for KIR3DL2. As for other KIRs, binding to HLA class I molecules is essential for NK development by promoting phenomena such as licensing and driving NK cell maturation. For KIR3DL2, this includes binding to HLA-A3 and -A11 and to the free heavy chain form of HLA-B27. In addition, KIR3DL2 binds to CpG oligonucleotides (ODN) and ensures their transport to endosomal toll-like receptor 9 that promotes cell activation. These characteristics have implicated KIR3DL2 in several pathologies: ankylosing spondylitis and cutaneous T-cell lymphomas such as Sézary syndrome, CD30+ cutaneous lymphoma, and transformed mycosis fungoides. Consequently, a new generation of humanized monoclonal antibodies (mAbs) directed against KIR3DL2 has been helpful in the diagnosis, follow-up, and treatment of these diseases. In addition, preliminary clinical studies of a novel targeted immunotherapy for cutaneous T-cell lymphomas using the anti-KIR3DL2 mAb IPH4102 are now underway. In this review, we discuss the various aspects of KIR3DL2 on the functions of CD4+ T cells and how targeting this receptor helps to develop innovative therapeutic strategies.
Collapse
Affiliation(s)
- Christian Schmitt
- INSERM U976, Hôpital Saint-Louis, Paris, France.,Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Anne Marie-Cardine
- INSERM U976, Hôpital Saint-Louis, Paris, France.,Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Armand Bensussan
- INSERM U976, Hôpital Saint-Louis, Paris, France.,Paris Diderot University, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
20
|
Pers YM, Jorgensen C. Perspectives of ofatumumab as CD20 targeted therapy in rheumatoid arthritis and other autoimmune diseases. Immunotherapy 2017; 8:1091-6. [PMID: 27485081 DOI: 10.2217/imt-2016-0003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune condition viewed as a severe destructive disease. The treatment strategies include anti-CD20 monoclonal antibody (mAb)-targeting B cells. Ofatumumab specifically targets a membrane-proximal epitope on the CD20 molecule distinct from other anti-CD20 antibodies including rituximab and ocrelizumab, and bind the epitope located on the large loop of CD20. This explains a more durable B-cell depletion and a different pharmacodynamic. We review the pharmacodynamic of B-cell depletion and analyze the results in RA and other B-cell-mediated autoimmune diseases. The randomized trial in RA showed clinical efficacy comparable to rituximab at week 24. However, structural impact has not been demonstrated. Studies including RA patients refractory to rituximab would be useful to define the optimal strategy of ofatumumab therapy.
Collapse
Affiliation(s)
- Yves Marie Pers
- Inserm, U1183, Hôpital Saint-Eloi, Montpellier, F-34295 France.,Université MONTPELLIER, UFR de Médecine, Montpellier, F-34000 France.,Service d'immuno-Rhumatologie, CHU Lapeyronie, Montpellier, F-34295 France
| | - Christian Jorgensen
- Inserm, U1183, Hôpital Saint-Eloi, Montpellier, F-34295 France.,Université MONTPELLIER, UFR de Médecine, Montpellier, F-34000 France.,Service d'immuno-Rhumatologie, CHU Lapeyronie, Montpellier, F-34295 France
| |
Collapse
|
21
|
Golay J. Direct targeting of cancer cells with antibodies: What can we learn from the successes and failure of unconjugated antibodies for lymphoid neoplasias? J Autoimmun 2017; 85:6-19. [PMID: 28666691 DOI: 10.1016/j.jaut.2017.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/11/2017] [Indexed: 12/26/2022]
Abstract
Following approval in 1997 of the anti-CD20 antibody rituximab for the treatment of B-NHL and CLL, many other unconjugated IgG1 MAbs have been tested in pre-clinical and clinical trials for the treatment of lymphoid neoplasms. Relatively few have been approved however and these are directed against a limited number of target antigens (CD20, CD52, CCR4, CD38, CD319). We review here the known biological properties of these antibodies and discuss which factors may have led to their success or may, on the contrary, limit their clinical application. Common factors of the approved MAbs are that the target antigen is expressed at relatively high levels on the neoplastic targets and their mechanism of action is mostly immune-mediated. Indeed most of these MAbs induce ADCC and phagocytosis by macrophages, and many also activate complement, leading to target cell lysis. In contrast direct cell death induction is not a common feature but may enhance efficacy in some cases. Interestingly, a key factor for the success of several MAbs appears to be their capacity to skew immunity towards an anti-tumour mode, by inhibiting/depleting suppressor cells and/or activating immune cells within the microenvironment, independently of FcγRs. We also expose here some of the strategies employed by industry to expand the clinical use of these molecules beyond their original indication. Interestingly, due to the central role of lymphocytes in the control of the immune response, several of the antibodies are now successfully used to treat many different autoimmune diseases and have also been formally approved for some of these new indications. There is little doubt that this trend will continue and that the precise mechanisms of therapeutic MAbs will be further dissected and better understood in the context of both tumour immunology and autoimmunity.
Collapse
Affiliation(s)
- Josée Golay
- Center of Cellular Therapy "G. Lanzani", USC Haematology, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Via Garibaldi 11-13, 24128, Bergamo, Italy.
| |
Collapse
|
22
|
Ngambenjawong C, Gustafson HH, Pun SH. Progress in tumor-associated macrophage (TAM)-targeted therapeutics. Adv Drug Deliv Rev 2017; 114:206-221. [PMID: 28449873 DOI: 10.1016/j.addr.2017.04.010] [Citation(s) in RCA: 587] [Impact Index Per Article: 73.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/13/2017] [Accepted: 04/14/2017] [Indexed: 02/06/2023]
Abstract
As an essential innate immune population for maintaining body homeostasis and warding off foreign pathogens, macrophages display high plasticity and perform diverse supportive functions specialized to different tissue compartments. Consequently, aberrance in macrophage functions contributes substantially to progression of several diseases including cancer, fibrosis, and diabetes. In the context of cancer, tumor-associated macrophages (TAMs) in tumor microenvironment (TME) typically promote cancer cell proliferation, immunosuppression, and angiogenesis in support of tumor growth and metastasis. Oftentimes, the abundance of TAMs in tumor is correlated with poor disease prognosis. Hence, significant attention has been drawn towards development of cancer immunotherapies targeting these TAMs; either depleting them from tumor, blocking their pro-tumoral functions, or restoring their immunostimulatory/tumoricidal properties. This review aims to introduce readers to various aspects in development and evaluation of TAM-targeted therapeutics in pre-clinical and clinical stages.
Collapse
Affiliation(s)
- Chayanon Ngambenjawong
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, United States
| | - Heather H Gustafson
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, United States
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
23
|
Chen YCE, Mapp S, Blumenthal A, Burgess ML, Mazzieri R, Mattarollo SR, Mollee P, Gill D, Saunders NA. The duality of macrophage function in chronic lymphocytic leukaemia. Biochim Biophys Acta Rev Cancer 2017; 1868:176-182. [PMID: 28347751 DOI: 10.1016/j.bbcan.2017.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/09/2017] [Accepted: 03/21/2017] [Indexed: 12/23/2022]
Abstract
Chronic lymphocytic leukaemia (CLL) is the most common adult leukaemia and, in some patients, is accompanied by resistance to both chemotherapeutics and immunotherapeutics. In this review we will discuss the role of tumour associated macrophages (TAMs) in promoting CLL cell survival and resistance to immunotherapeutics. In addition, we will discuss mechanisms by which TAMs suppress T-cell mediated antitumour responses. Thus, targeting macrophages could be used to i) reduce the leukaemic burden via the induction of T-cell-mediated antitumour responses, ii) to reduce pro-survival signalling and enhance response to conventional chemotherapeutics or iii) enhance the response to therapeutic antibodies in current clinical use.
Collapse
Affiliation(s)
- Y C E Chen
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - S Mapp
- Department of Haematology, Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - A Blumenthal
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - M L Burgess
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia; Department of Haematology, Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - R Mazzieri
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - S R Mattarollo
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - P Mollee
- Department of Haematology, Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - D Gill
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia; Department of Haematology, Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - N A Saunders
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia.
| |
Collapse
|
24
|
Cook EM, Lindorfer MA, van der Horst H, Oostindie S, Beurskens FJ, Schuurman J, Zent CS, Burack R, Parren PWHI, Taylor RP. Antibodies That Efficiently Form Hexamers upon Antigen Binding Can Induce Complement-Dependent Cytotoxicity under Complement-Limiting Conditions. THE JOURNAL OF IMMUNOLOGY 2016; 197:1762-75. [PMID: 27474078 DOI: 10.4049/jimmunol.1600648] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/28/2016] [Indexed: 11/19/2022]
Abstract
Recently, we demonstrated that IgG Abs can organize into ordered hexamers after binding their cognate Ags expressed on cell surfaces. This process is dependent on Fc:Fc interactions, which promote C1q binding, the first step in classical pathway complement activation. We went on to engineer point mutations that stimulated IgG hexamer formation and complement-dependent cytotoxicity (CDC). The hexamer formation-enhanced (HexaBody) CD20 and CD38 mAbs support faster, more robust CDC than their wild-type counterparts. To further investigate the CDC potential of these mAbs, we used flow cytometry, high-resolution digital imaging, and four-color confocal microscopy to examine their activity against B cell lines and primary chronic lymphocytic leukemia cells in sera depleted of single complement components. We also examined the CDC activity of alemtuzumab (anti-CD52) and mAb W6/32 (anti-HLA), which bind at high density to cells and promote substantial complement activation. Although we observed little CDC for mAb-opsonized cells reacted with sera depleted of early complement components, we were surprised to discover that the Hexabody mAbs, as well as ALM and W6/32, were all quite effective at promoting CDC in sera depleted of individual complement components C6 to C9. However, neutralization studies conducted with an anti-C9 mAb verified that C9 is required for CDC activity against cell lines. These highly effective complement-activating mAbs efficiently focus activated complement components on the cell, including C3b and C9, and promote CDC with a very low threshold of MAC binding, thus providing additional insight into their enhanced efficacy in promoting CDC.
Collapse
Affiliation(s)
- Erika M Cook
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908
| | | | | | | | | | - Clive S Zent
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642
| | - Richard Burack
- Department of Pathology, University of Rochester Medical Center, Rochester, NY 14642; and
| | - Paul W H I Parren
- Genmab, 3584 CM Utrecht, the Netherlands; Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908;
| |
Collapse
|
25
|
Velmurugan R, Challa DK, Ram S, Ober RJ, Ward ES. Macrophage-Mediated Trogocytosis Leads to Death of Antibody-Opsonized Tumor Cells. Mol Cancer Ther 2016; 15:1879-89. [PMID: 27226489 DOI: 10.1158/1535-7163.mct-15-0335] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 05/17/2016] [Indexed: 12/16/2022]
Abstract
Understanding the complex behavior of effector cells such as monocytes or macrophages in regulating cancerous growth is of central importance for cancer immunotherapy. Earlier studies using CD20-specific antibodies have demonstrated that the Fcγ receptor (FcγR)-mediated transfer of the targeted receptors from tumor cells to these effector cells through trogocytosis can enable escape from antibody therapy, leading to the viewpoint that this process is protumorigenic. In the current study, we demonstrate that persistent trogocytic attack results in the killing of HER2-overexpressing breast cancer cells. Further, antibody engineering to increase FcγR interactions enhances this tumoricidal activity. These studies extend the complex repertoire of activities of macrophages to trogocytic-mediated cell death of HER2-overexpressing target cells and have implications for the development of effective antibody-based therapies. Mol Cancer Ther; 15(8); 1879-89. ©2016 AACR.
Collapse
Affiliation(s)
- Ramraj Velmurugan
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas. Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, Texas. Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas. Department of Immunology, UT Southwestern Medical Center, Dallas, Texas
| | - Dilip K Challa
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas. Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, Texas. Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas. Department of Immunology, UT Southwestern Medical Center, Dallas, Texas
| | - Sripad Ram
- Department of Immunology, UT Southwestern Medical Center, Dallas, Texas
| | - Raimund J Ober
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas. Department of Biomedical Engineering, Texas A&M University, College Station, Texas. Department of Immunology, UT Southwestern Medical Center, Dallas, Texas.
| | - E Sally Ward
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas. Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, Texas. Department of Immunology, UT Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
26
|
Gallagher S, Turman S, Yusuf I, Akhgar A, Wu Y, Roskos LK, Herbst R, Wang Y. Pharmacological profile of MEDI-551, a novel anti-CD19 antibody, in human CD19 transgenic mice. Int Immunopharmacol 2016; 36:205-212. [PMID: 27163209 DOI: 10.1016/j.intimp.2016.04.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/15/2016] [Accepted: 04/21/2016] [Indexed: 10/21/2022]
Abstract
B cell depletion therapy is beneficial for patients with B cell malignancies and autoimmune diseases. CD19, a transmembrane protein, is expressed on a vast majority of normal and neoplastic B cells, making it a suitable target for monoclonal antibody (MAb) mediated immunotherapy. We have developed MEDI-551, an affinity optimized and afucosylated IgG1 MAb targeting human CD19 for B cell depletion. MEDI-551 is currently under investigation in multiple clinical trials. Because MEDI-551 does not cross react with rodent and non-human primate CD19, the pharmacological characteristics of the MAb were evaluated in human CD19 transgenic mice (hCD19 Tg). Here we show that MEDI-551 potently depletes tissue and circulating B cells in hCD19 Tg mice and is more efficacious than the anti-CD19 MAb with intact fucose. The length of B cell depletion depends on MEDI-551 dose; and, B cell recovery in the circulation follows stepwise phenotypic maturation. Furthermore, intravenous (IV) and subcutaneous (SC) administration of MEDI-551 results in comparable efficacy. Lastly, the combination of MEDI-551 with the anti-CD20 MAb, rituximab, further prolongs the duration of B cell depletion. In summary, the pharmacological profile of MEDI-551 presented in hCD19 Tg mice supports further testing of MEDI-551 in clinical trials involving B cell malignancies and autoimmune diseases.
Collapse
Affiliation(s)
- Sandra Gallagher
- Respiratory, Inflammation and Autoimmune Research, Gaithersburg, MD, United States
| | - Sean Turman
- Respiratory, Inflammation and Autoimmune Research, Gaithersburg, MD, United States
| | - Isharat Yusuf
- Respiratory, Inflammation and Autoimmune Research, Gaithersburg, MD, United States
| | - Ahmad Akhgar
- Translational Sciences, MedImmune LLC, Gaithersburg, MD, United States
| | - Yuling Wu
- Translational Sciences, MedImmune LLC, Gaithersburg, MD, United States
| | - Lorin K Roskos
- Translational Sciences, MedImmune LLC, Gaithersburg, MD, United States
| | - Ronald Herbst
- Respiratory, Inflammation and Autoimmune Research, Gaithersburg, MD, United States
| | - Yue Wang
- Respiratory, Inflammation and Autoimmune Research, Gaithersburg, MD, United States.
| |
Collapse
|
27
|
Stasiłojć G, Österborg A, Blom AM, Okrój M. New perspectives on complement mediated immunotherapy. Cancer Treat Rev 2016; 45:68-75. [DOI: 10.1016/j.ctrv.2016.02.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 12/25/2022]
|
28
|
Taylor RP, Lindorfer MA. Cytotoxic mechanisms of immunotherapy: Harnessing complement in the action of anti-tumor monoclonal antibodies. Semin Immunol 2016; 28:309-16. [PMID: 27009480 DOI: 10.1016/j.smim.2016.03.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/07/2016] [Indexed: 01/02/2023]
Abstract
Several mAbs that have been approved for the treatment of cancer make use of complement-dependent cytotoxicity (CDC) to eliminate tumor cells. Comprehensive investigations, based on in vitro studies, mouse models and analyses of patient blood samples after mAb treatment have provided key insights into the details of individual steps in the CDC reaction. Based on the lessons learned from these studies, new and innovative approaches are now being developed to increase the clinical efficacy of next generation mAbs with respect to CDC. These improvements include engineering changes in the mAbs to enhance their ability to activate complement. In addition, mAb dosing paradigms are being developed that take into account the capacity as well as the limitations of the complement system to eliminate a substantial burden of mAb-opsonized cells. Over the next few years it is likely these approaches will lead to mAbs that are far more effective in the treatment of cancer.
Collapse
Affiliation(s)
- Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, United States.
| | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| |
Collapse
|
29
|
Zent CS, Wang XV, Ketterling RP, Hanson CA, Libby EN, Barrientos JC, Call TG, Chang JE, Liu JJ, Calvo AR, Lazarus HM, Rowe JM, Luger SM, Litzow MR, Tallman MS. A phase II randomized trial comparing standard and low dose rituximab combined with alemtuzumab as initial treatment of progressive chronic lymphocytic leukemia in older patients: a trial of the ECOG-ACRIN cancer research group (E1908). Am J Hematol 2016; 91:308-12. [PMID: 26662208 PMCID: PMC4806550 DOI: 10.1002/ajh.24265] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 11/28/2015] [Indexed: 12/31/2022]
Abstract
Chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL) patients requiring initial therapy are often older and frailer and unsuitable candidates for standard chemoimmunotherapy regimens. Shorter duration combination monoclonal antibody (mAb) therapy using alemtuzumab and rituximab has been shown to be effective and tolerable treatment for CLL. Standard dose anti-CD20 mAb therapy causes loss of CD20 expression by surviving CLL cells, which can be minimized by decreasing the mAb dose. We report a randomized phase II clinical trial enrolling older (≥ 65 years) patients (median age 76 years, n = 31) with treatment naïve progressive CLL. Patients received 8-12 weeks of standard subcutaneous alemtuzumab with either intravenous standard (375 mg/m(2) weekly)(n = 16) or low dose (20 mg/m(2) 3x week)(n = 15) rituximab. This study was closed before full accrual because the manufacturer withdrew alemtuzumab for treatment of CLL. The overall response rate was 90% with an 45% complete response rate, median progression-free survival of 17.9 months and no significant differences in outcome between the low and standard dose rituximab arms. The major toxicities were cytopenia and infection with one treatment fatality caused by progressive multifocal leukoencephalopathy but no other opportunistic infections. Combination mAb therapy was effective and tolerable treatment for older and frailer patients with progressive CLL, achieving a high rate of complete remissions. These data support the role of mAb in therapy for less fit CLL patients and the further study of low dose higher frequency anti-CD20 mAb therapy as a potentially more effective use of anti-CD20 mAb in the treatment of CLL.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Alemtuzumab
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Disease-Free Survival
- Dose-Response Relationship, Drug
- Drug Administration Schedule
- Female
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Male
- Rituximab/administration & dosage
- Rituximab/adverse effects
- Rituximab/therapeutic use
Collapse
Affiliation(s)
- Clive S. Zent
- University of Rochester Medical Center, Rochester, New York (current location) Mayo Clinic, Rochester, Minnesota (former location)
| | - Xin Victoria Wang
- Dana Farber Cancer Institute – ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | | | | | - Edward N. Libby
- Seattle Cancer Care Alliance, Seattle, Washington (current location) University of New Mexico Cancer Center, Albuquerque, NM
| | | | | | | | | | | | | | | | | | | | - Martin S. Tallman
- Memorial Sloan Kettering Cancer Center, New York, New York (current location) Northwestern University School of Medicine, Chicago, Illinois (former location)
| |
Collapse
|
30
|
Church AK, VanDerMeid KR, Baig NA, Baran AM, Witzig TE, Nowakowski GS, Zent CS. Anti-CD20 monoclonal antibody-dependent phagocytosis of chronic lymphocytic leukaemia cells by autologous macrophages. Clin Exp Immunol 2015; 183:90-101. [PMID: 26307241 DOI: 10.1111/cei.12697] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2015] [Indexed: 01/01/2023] Open
Abstract
Unconjugated monoclonal antibodies (mAbs) are an important component of effective combination therapies for chronic lymphocytic leukaemia (CLL). Antibody-dependent phagocytosis (ADP) is a major mediator of mAb cytotoxicity, but there is limited knowledge of the determinants of ADP efficacy. We used macrophages derived in vitro from autologous circulating monocytes to test the effects of mAb structure and concentration, target : effector cell ratio, duration of co-incubation and CLL cell CD20 expression on ADP. Next-generation anti-CD20 mAbs (ofatumumab, ublituximab, obinutuzumab, ocaratuzumab) were significantly more effective at inducing ADP compared to rituximab, but none were as effective as the anti-CD52 mAb alemtuzumab. Ofatumumab (10 μg/ml) used as a representative next-generation anti-CD20 mAb achieved an ADP plateau at 3 h co-incubation with a target : effector ratio of 10 : 1 (mean = 2.1 CLL cells/macrophage, range = 1.5-3.5). At 0.156 μg/ml (the lowest concentration tested) ofatumumab ADP was significantly higher than alemtuzumab. However, ofatumumab-induced ADP did not increase significantly at higher mAb concentrations. We show that anti-CD20 mAb ADP efficacy is determined by the mAb characteristics, target : effector ratio and incubation time. We suggest that preclinical evaluation of anti-CD20 mAbs to understand the determinants of ADP could be useful in designing future combination therapies for CLL.
Collapse
Affiliation(s)
- A K Church
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - K R VanDerMeid
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - N A Baig
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - A M Baran
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - T E Witzig
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | - C S Zent
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
31
|
Ying HQ, Wang F, Chen XL, He BS, Pan YQ, Chen J, Liu X, Cao WJ, Peng HX, Lin K, Wang SK. FCGR2A, FCGR3A polymorphisms and therapeutic efficacy of anti-EGFR monoclonal antibody in metastatic colorectal cancer. Oncotarget 2015; 6:28071-28083. [PMID: 26363448 PMCID: PMC4695045 DOI: 10.18632/oncotarget.4872] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/30/2015] [Indexed: 01/05/2023] Open
Abstract
Anti-EGFR monoclonal antibodies (mAb) such as cetuximab, panitumumab are one kind of efficacious targeted drugs in treatment of metastatic colorectal cancer (mCRC). However, only a small proportion of patients harbored wild-KRAS genotype can benefit from it. We hypothesized that personal genetic heterogeneity might be the main cause leading to obvious difference in its clinical efficacy. A retrospective study including 82 mCRC patients treated with chemotherapy plus cetuximab and a comprehensive meta-analysis containing 2831 cases within sixteen eligible studies were conducted to investigate the possible association between FCGR2A H131R and FCGR3A V158F and clinical outcome of mCRC patients treated with anti-EGFR mAb based therapy. Results of the retrospective study showed that H131R within FCGR2A or V158F within FCGR3A were not associated with clinical outcome in 82 KRAS wild chemorefractory mCRC patients in co-dominant, dominant, recessive, over-dominant, allele genetic models. However, the comprehensive meta-analysis with the largest of sample size obtained the significant result between FCGR3A V158F and PFS (FV/VV vs. FF: Ph = 0.027, MSR = 0.680, 95%CI = 0.549-0.842 in overall population; Ph = 0.12, MSR = 0.728, 95%CI = 0.648-0.818 in KRAS wild population) and OS (VV vs. FF: Ph < 0.001, MSR = 0.733, 95%CI = 0.578-0.930 in overall population). These findings indicate that KRAS wild chemorefractory mCRC individual harbored genotype FF of V158Fcan benefit from anti-EGFR mAb adjuvant therapy in terms of PFS and OS, and it may be useful genetic biomarker to predict clinical survival of mCRC individuals with anti-EGFR mAb based therapy.
Collapse
Affiliation(s)
- Hou-Qun Ying
- Medical College, Southeast University, Nanjing 210009, Jiangsu, China
- Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Feng Wang
- Medical College, Southeast University, Nanjing 210009, Jiangsu, China
| | - Xiao-Lin Chen
- Department of Clinical Laboratory, Pingxiang People's Hospital, Pingxiang, 337055, Jiangxi, China
| | - Bang-Shun He
- Medical College, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yu-Qin Pan
- Medical College, Southeast University, Nanjing 210009, Jiangsu, China
| | - Jie Chen
- Medical College, Southeast University, Nanjing 210009, Jiangsu, China
- Life Scientific College, Nanjing Normal University, Nanjing 210046, Jiangsu, China
| | - Xian Liu
- Medical College, Southeast University, Nanjing 210009, Jiangsu, China
| | - Wei-Jun Cao
- Department of Digestion, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu China
| | - Hong-Xin Peng
- Medical College, Southeast University, Nanjing 210009, Jiangsu, China
| | - Kang Lin
- Medical College, Southeast University, Nanjing 210009, Jiangsu, China
| | - Shu-Kui Wang
- Medical College, Southeast University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
32
|
Antibodies reactive to cleaved sites in complement proteins enable highly specific measurement of soluble markers of complement activation. Mol Immunol 2015; 66:164-70. [DOI: 10.1016/j.molimm.2015.02.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/27/2015] [Accepted: 02/27/2015] [Indexed: 01/11/2023]
|
33
|
Capuano C, Romanelli M, Pighi C, Cimino G, Rago A, Molfetta R, Paolini R, Santoni A, Galandrini R. Anti-CD20 Therapy Acts via FcγRIIIA to Diminish Responsiveness of Human Natural Killer Cells. Cancer Res 2015; 75:4097-108. [DOI: 10.1158/0008-5472.can-15-0781] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 07/21/2015] [Indexed: 11/16/2022]
|
34
|
Battella S, Cox MC, Santoni A, Palmieri G. Natural killer (NK) cells and anti-tumor therapeutic mAb: unexplored interactions. J Leukoc Biol 2015; 99:87-96. [PMID: 26136506 DOI: 10.1189/jlb.5vmr0415-141r] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/16/2015] [Indexed: 12/11/2022] Open
Abstract
Tumor-targeting mAb are widely used in the treatment of a variety of solid and hematopoietic tumors and represent the first immunotherapeutic approach successfully arrived to the clinic. Nevertheless, the role of distinct immune mechanisms in contributing to their therapeutic efficacy is not completely understood and may vary depending on tumor- or antigen/antibody-dependent characteristics. Availability of next-generation, engineered, tumor-targeting mAb, optimized in their capability to recruit selected immune effectors, re-enforces the need for a deeper understanding of the mechanisms underlying anti-tumor mAb functionality. NK cells participate with a major role to innate anti-tumor responses, by exerting cytotoxic activity and producing a vast array of cytokines. As the CD16 (low-affinity FcγRIIIA)-activating receptor is expressed on the majority of NK cells, its effector functions can be ideally recruited against therapeutic mAb-opsonized tumor cells. The exact role of NK cells in determining therapeutic efficacy of tumor-targeting mAb is still unclear and much sought after. This knowledge will be instrumental to design innovative combination schemes with newly validated immunomodulatory agents. We will summarize what is known about the role of NK cells in therapeutic anti-tumor mAb therapy, with particular emphasis on RTX chimeric anti-CD20 mAb, the first one used in clinical practice for treating B cell malignancies.
Collapse
Affiliation(s)
- Simone Battella
- Departments of *Experimental Medicine and Molecular Medicine, Hematology Unit, Sant'Andrea Hospital, and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Maria Christina Cox
- Departments of *Experimental Medicine and Molecular Medicine, Hematology Unit, Sant'Andrea Hospital, and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Angela Santoni
- Departments of *Experimental Medicine and Molecular Medicine, Hematology Unit, Sant'Andrea Hospital, and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Gabriella Palmieri
- Departments of *Experimental Medicine and Molecular Medicine, Hematology Unit, Sant'Andrea Hospital, and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| |
Collapse
|
35
|
|
36
|
Zhang PW, Chen L, Huang T, Zhang N, Kong XY, Cai YD. Classifying ten types of major cancers based on reverse phase protein array profiles. PLoS One 2015; 10:e0123147. [PMID: 25822500 PMCID: PMC4378934 DOI: 10.1371/journal.pone.0123147] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 02/24/2015] [Indexed: 12/20/2022] Open
Abstract
Gathering vast data sets of cancer genomes requires more efficient and autonomous procedures to classify cancer types and to discover a few essential genes to distinguish different cancers. Because protein expression is more stable than gene expression, we chose reverse phase protein array (RPPA) data, a powerful and robust antibody-based high-throughput approach for targeted proteomics, to perform our research. In this study, we proposed a computational framework to classify the patient samples into ten major cancer types based on the RPPA data using the SMO (Sequential minimal optimization) method. A careful feature selection procedure was employed to select 23 important proteins from the total of 187 proteins by mRMR (minimum Redundancy Maximum Relevance Feature Selection) and IFS (Incremental Feature Selection) on the training set. By using the 23 proteins, we successfully classified the ten cancer types with an MCC (Matthews Correlation Coefficient) of 0.904 on the training set, evaluated by 10-fold cross-validation, and an MCC of 0.936 on an independent test set. Further analysis of these 23 proteins was performed. Most of these proteins can present the hallmarks of cancer; Chk2, for example, plays an important role in the proliferation of cancer cells. Our analysis of these 23 proteins lends credence to the importance of these genes as indicators of cancer classification. We also believe our methods and findings may shed light on the discoveries of specific biomarkers of different types of cancers.
Collapse
Affiliation(s)
- Pei-Wei Zhang
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, P.R. China
| | - Tao Huang
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
- * E-mail: (TH); (NZ); (XYK); (YDC)
| | - Ning Zhang
- Department of Biomedical Engineering, Tianjin Key Lab of BME Measurement, Tianjin University, Tianjin, P.R. China
- * E-mail: (TH); (NZ); (XYK); (YDC)
| | - Xiang-Yin Kong
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
- * E-mail: (TH); (NZ); (XYK); (YDC)
| | - Yu-Dong Cai
- College of Life Science, Shanghai University, Shanghai, P.R. China
- * E-mail: (TH); (NZ); (XYK); (YDC)
| |
Collapse
|
37
|
Zent CS, Call TG, Bowen DA, Conte MJ, LaPlant BR, Witzig TE, Ansell SM, Weiner GJ. Early treatment of high risk chronic lymphocytic leukemia with alemtuzumab, rituximab and poly-(1-6)-beta-glucotriosyl-(1-3)- beta-glucopyranose beta-glucan is well tolerated and achieves high complete remission rates. Leuk Lymphoma 2015; 56:2373-8. [PMID: 25676035 PMCID: PMC4573786 DOI: 10.3109/10428194.2015.1016932] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Poly-[1-6]-β-glucopyranosyl-[1-3]-β-glucopyranose (PGG) beta glucan is a Saccharomyces cerevisiae derived 1,3/1,6 glucose polymer with innate immune system activation potential. This phase I/II clinical trial enrolled 20 eligible patients with chronic lymphocytic leukemia with high-risk biological markers for early initial treatment with alemtuzumab, rituximab and PGG beta glucan (1-2-4 mg/kg/dose) over 31 days. PGG beta glucan at 4 mg/kg was well tolerated and used for the phase II study. There were three grade 3-4 toxicities at least possibly attributed to treatment. Nineteen (95%) patients responded to treatment with 13 (65%) complete responses. All patients were alive at a median follow-up of 24.4 months (range: 9.5-37). Eleven patients had progressive disease (median 17.6 months, 95% confidence interval [CI]: 9.7, 32.1) and eight patients were retreated (median 35.3 months, 95% CI: 17.9, not reached). We conclude that PGG beta glucan, alemtuzumab and rituximab treatment is tolerable and results in a high complete response rate.
Collapse
MESH Headings
- Aged
- Alemtuzumab
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor
- Chromosome Aberrations
- Disease Progression
- Female
- Follow-Up Studies
- Humans
- Kaplan-Meier Estimate
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Male
- Middle Aged
- Remission Induction
- Rituximab/administration & dosage
- Treatment Outcome
- beta-Glucans/administration & dosage
Collapse
Affiliation(s)
| | | | | | | | - Betsy R. LaPlant
- Department of Health Sciences Research, Mayo Clinic, Rochester MN
| | | | | | - George J. Weiner
- Holden Comprehensive Cancer Center and Department of Internal Medicine, University of Iowa, Iowa City IA
| |
Collapse
|
38
|
Melis JPM, Strumane K, Ruuls SR, Beurskens FJ, Schuurman J, Parren PWHI. Complement in therapy and disease: Regulating the complement system with antibody-based therapeutics. Mol Immunol 2015; 67:117-30. [PMID: 25697848 DOI: 10.1016/j.molimm.2015.01.028] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/26/2015] [Accepted: 01/27/2015] [Indexed: 12/23/2022]
Abstract
Complement is recognized as a key player in a wide range of normal as well as disease-related immune, developmental and homeostatic processes. Knowledge of complement components, structures, interactions, and cross-talk with other biological systems continues to grow and this leads to novel treatments for cancer, infectious, autoimmune- or age-related diseases as well as for preventing transplantation rejection. Antibodies are superbly suited to be developed into therapeutics with appropriate complement stimulatory or inhibitory activity. Here we review the design, development and future of antibody-based drugs that enhance or dampen the complement system.
Collapse
Affiliation(s)
| | | | | | | | | | - Paul W H I Parren
- Genmab, Utrecht, The Netherlands; Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
39
|
Fcγ-receptor-mediated trogocytosis impacts mAb-based therapies: historical precedence and recent developments. Blood 2014; 125:762-6. [PMID: 25498911 DOI: 10.1182/blood-2014-10-569244] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
A specialized form of trogocytosis occurs when Fcγ receptors on acceptor cells take up and internalize donor cell-associated immune complexes composed of specific monoclonal antibodies (mAbs) bound to target antigens on donor cells. This trogocytosis reaction, an example of antigenic modulation, has been described in recent clinical correlative studies and in vitro investigations for several mAbs used in cancer immunotherapy, including rituximab and ofatumumab. We discuss the impact of Fcγ-receptor-mediated trogocytosis on the efficacy of cancer immunotherapy and other mAb-based therapies.
Collapse
|