101
|
Rotondi M, Molteni M, Leporati P, Capelli V, Marinò M, Chiovato L. Autoimmune Thyroid Diseases in Patients Treated with Alemtuzumab for Multiple Sclerosis: An Example of Selective Anti-TSH-Receptor Immune Response. Front Endocrinol (Lausanne) 2017; 8:254. [PMID: 29033895 PMCID: PMC5626941 DOI: 10.3389/fendo.2017.00254] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/15/2017] [Indexed: 01/14/2023] Open
Abstract
Alemtuzumab, a humanized anti-CD52 monoclonal antibody, is approved for the treatment of active relapsing-remitting multiple sclerosis (MS). Alemtuzumab induces a rapid and prolonged depletion of lymphocytes from the circulation, which results in a profound immuno-suppression status followed by an immune reconstitution phase. Secondary to reconstitution autoimmune diseases represent the most common side effect of Alemtuzumab treatment. Among them, Graves' disease (GD) is the most frequent one with an estimated prevalence ranging from 16.7 to 41.0% of MS patients receiving Alemtuzumab. Thyrotropin (TSH) receptor (R)-reactive B cells are typically observed in GD and eventually present this autoantigen to T-cells, which, in turn, secrete several pro-inflammatory cytokines and chemokines. Given that reconstitution autoimmunity is more frequently characterized by autoantibody-mediated diseases rather than by destructive Th1-mediated disorders, it is not surprising that GD is the most commonly reported side effect of Alemtuzumab treatment in patients with MS. On the other hand, immune reconstitution GD was not observed in a large series of patients with rheumatoid arthritis treated with Alemtuzumab. This negative finding supports the view that patients with MS are intrinsically more at risk for developing Alemtuzumab-related thyroid dysfunctions and in particular of GD. From a clinical point of view, Alemtuzumab-induced GD is characterized by a surprisingly high rate of remission, both spontaneous and after antithyroid drugs, as well as by a spontaneous shift to hypothyroidism, which is supposed to result from a change from stimulating to blocking TSH-receptor antibodies. These immune and clinical peculiarities support the concept that antithyroid drugs should be the first-line treatment in Alemtuzumab-induced Graves' hyperthyroidism.
Collapse
Affiliation(s)
- Mario Rotondi
- Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, ICS-Maugeri IRCCS, University of Pavia, Pavia, Italy
| | - Martina Molteni
- Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, ICS-Maugeri IRCCS, University of Pavia, Pavia, Italy
| | - Paola Leporati
- Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, ICS-Maugeri IRCCS, University of Pavia, Pavia, Italy
| | - Valentina Capelli
- Department of Internal Medicine and Therapeutics, and Department of Medical and Surgical Sciences, University of Pavia, Pavia, Italy
| | - Michele Marinò
- Endocrinology Unit I, Department of Clinical and Experimental Medicine, University of Pisa, University Hospital of Pisa, Pisa, Italy
| | - Luca Chiovato
- Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, ICS-Maugeri IRCCS, University of Pavia, Pavia, Italy
- *Correspondence: Luca Chiovato,
| |
Collapse
|
102
|
Gross CC, Ahmetspahic D, Ruck T, Schulte-Mecklenbeck A, Schwarte K, Jörgens S, Scheu S, Windhagen S, Graefe B, Melzer N, Klotz L, Arolt V, Wiendl H, Meuth SG, Alferink J. Alemtuzumab treatment alters circulating innate immune cells in multiple sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2016; 3:e289. [PMID: 27766281 PMCID: PMC5063395 DOI: 10.1212/nxi.0000000000000289] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/12/2016] [Indexed: 11/15/2022]
Abstract
Objective: To characterize changes in myeloid and lymphoid innate immune cells in patients with relapsing-remitting multiple sclerosis (MS) during a 6-month follow-up after alemtuzumab treatment. Methods: Circulating innate immune cells including myeloid cells and innate lymphoid cells (ILCs) were analyzed before and 6 and 12 months after onset of alemtuzumab treatment. Furthermore, a potential effect on granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)–23 production by myeloid cells and natural killer (NK) cell cytolytic activity was determined. Results: In comparison to CD4+ T lymphocytes, myeloid and lymphoid innate cell subsets of patients with MS expressed significantly lower amounts of CD52 on their cell surface. Six months after CD52 depletion, numbers of circulating plasmacytoid dendritic cells (DCs) and conventional DCs were reduced compared to baseline. GM-CSF and IL-23 production in DCs remained unchanged. Within the ILC compartment, the subset of CD56bright NK cells specifically expanded under alemtuzumab treatment, but their cytolytic activity did not change. Conclusions: Our findings demonstrate that 6 months after alemtuzumab treatment, specific DC subsets are reduced, while CD56bright NK cells expanded in patients with MS. Thus, alemtuzumab specifically restricts the DC compartment and expands the CD56bright NK cell subset with potential immunoregulatory properties in MS. We suggest that remodeling of the innate immune compartment may promote long-term efficacy of alemtuzumab and preserve immunocompetence in patients with MS.
Collapse
Affiliation(s)
- Catharina C Gross
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Diana Ahmetspahic
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Tobias Ruck
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Andreas Schulte-Mecklenbeck
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Kathrin Schwarte
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Silke Jörgens
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Stefanie Scheu
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Susanne Windhagen
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Bettina Graefe
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Nico Melzer
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Luisa Klotz
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Volker Arolt
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Heinz Wiendl
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Sven G Meuth
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| | - Judith Alferink
- Department of Neurology (C.C.G., T.R., A.S.-M., N.M., L.K., H.W., S.G.M.), University Hospital Münster; Department of Psychiatry (D.A., K.S., S.J., V.A., J.A.) and Cluster of Excellence EXC 1003, Cells in Motion (D.A., K.S., H.W., S.G.M.), University of Münster; Institute of Medical Microbiology and Hospital Hygiene (S.S.), University of Düsseldorf; and Department of Neurology (S.W., B.G.), Clinics Osnabrück, Germany
| |
Collapse
|
103
|
Alemtuzumab-induced remission of multiple sclerosis-associated uveitis. Int Ophthalmol 2016; 37:1229-1233. [DOI: 10.1007/s10792-016-0370-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 09/30/2016] [Indexed: 12/18/2022]
|
104
|
Tati K, Yazdanpanah-Samani M, Ramezani A, Mahmoudi Maymand E, Ghaderi A. Establishment a CHO Cell Line Expressing Human CD52 Molecule. Rep Biochem Mol Biol 2016; 5:56-61. [PMID: 28070536 PMCID: PMC5214685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 05/22/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND CD52 is a small glycoprotein with a GPI anchor at its C-terminus. CD52 is expressed by Normal and malignant T and B lymphocytes and monocytes. There are detectable amounts of soluble CD52 in plasma of patients with CLL and could be used as a tumor marker. Although the biological function of CD52 is unknown but it seems that CD52 may be involved in migration and activation of T-cells .The aim of this study was to clone and express human CD52 gene in CHO cell line and studying its function in more details. METHODS Based on GenBank databases two specific primers were designed for amplification of cd52 gene. Total RNA was extracted from Raji cell line and cDNA synthesized. Amplified fragment was cloned in pBudCE4.1 vector. The new construct was transfected to CHO-K1 cell line using electroporation method. Expression of recombinant CD52 protein was evaluated by Real time PCR and flow cytometry methods. RESULTS Amplification of CD52 gene using specific primers on Raji cDNA showed a 209 bp band. New construct was confirmed by PCR and restriction pattern and sequence analysis. The new construct was designated as pBudKT1. RT-PCR analysis detected cd52 mRNAs in transfected cells and Flow cytometry Results showed that 78.4 % of cells represented CD52 in their surfaces. CONCLUSION In conclusion, we established a human CD52 positive cell line, CHO-CD52, and the protein was expressed on the membrane. Cloning of the CD52 gene could be the first step for the production of therapeutic monoclonal antibodies and detection systems for soluble CD52 in biological fluids.
Collapse
Affiliation(s)
- Khadijeh Tati
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical sciences, Shiraz, Iran
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Yazdanpanah-Samani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical sciences, Shiraz, Iran
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Mahmoudi Maymand
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical sciences, Shiraz, Iran
| |
Collapse
|
105
|
CD52-Negative NK Cells Are Abundant in the Liver and Less Susceptible to Alemtuzumab Treatment. PLoS One 2016; 11:e0161618. [PMID: 27560943 PMCID: PMC4999148 DOI: 10.1371/journal.pone.0161618] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 08/09/2016] [Indexed: 01/30/2023] Open
Abstract
Background T-cell depleting strategies have become an integral part of immunosuppressive regimens in organ transplantation. Alemtuzumab is a humanized monoclonal antibody against CD52, a cell-surface antigen on several immune cells. It has been suggested that lymphocyte depletion increases the risk of serious infections. However, this has not been observed with short-term alemtuzumab treatment in an organ transplant setting. For induction therapy using alemtuzumab following liver transplantation, we found that T- and B-cell numbers declined rapidly after alemtuzumab therapy; however, the natural killer (NK) cell number was sustained. NK cells are important effectors of innate immunity. Since the effects of alemtuzumab on NK cell functions, especially those of liver NK cells, are unknown, this study aimed to investigate this in detail. Methods To assess the effect of alemtuzumab on NK cells, samples were obtained from 7 organ donors and examined by flow cytometry using Annexin V and propidium iodide. Phenotypical and functional differences within subsets of NK cells with different levels of CD52 expression were determined by flow cytometry and in vitro cytotoxicity assays. Results CD52 expression on NK cells was lower than that on other lymphocyte subsets. The liver contained a large number of CD52− NK cells compared with the peripheral blood. In vitro treatment of liver-derived NK cells with alemtuzumab did not result in cell death. In contrast, co-incubation with alemtuzumab induced cell death in peripheral blood mononuclear cells and non-NK cells in the liver. Furthermore, CD52− liver NK cells were more cytotoxic and produced more IFN-γ than CD52+ NK cells after cytokine activation. Conclusion The liver contains a large number of CD52− NK cells. These cells are refractory to alemtuzumab and have robust activity. These findings indicate that CD52− NK cells persist and could protect against infection after alemtuzumab-based lymphocyte depletion.
Collapse
|
106
|
CD38 expression and complement inhibitors affect response and resistance to daratumumab therapy in myeloma. Blood 2016; 128:959-70. [DOI: 10.1182/blood-2016-03-703439] [Citation(s) in RCA: 224] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/08/2016] [Indexed: 12/25/2022] Open
Abstract
Key Points
Response to the CD38-targeting antibody daratumumab is significantly associated with CD38 expression levels on the tumor cells. Resistance to daratumumab is accompanied by increased expression of complement-inhibitory proteins.
Collapse
|
107
|
Sharma K, Janik JE, O'Mahony D, Stewart D, Pittaluga S, Stetler-Stevenson M, Jaffe ES, Raffeld M, Fleisher TA, Lee CC, Steinberg SM, Waldmann TA, Morris JC. Phase II Study of Alemtuzumab (CAMPATH-1) in Patients with HTLV-1-Associated Adult T-cell Leukemia/lymphoma. Clin Cancer Res 2016; 23:35-42. [PMID: 27486175 DOI: 10.1158/1078-0432.ccr-16-1022] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/30/2016] [Accepted: 07/12/2016] [Indexed: 12/13/2022]
Abstract
PURPOSE Therapeutic regimens for adult T-cell leukemia/lymphoma (ATL) are limited with unsatisfactory results, thereby warranting development of novel therapies. This study investigated antitumor activity and toxicity of alemtuzumab with regard to response, duration of response, progression-free survival, and overall survival in patients with human T-cell lymphotropic virus-1 (HTLV-1)-associated ATL. EXPERIMENTAL DESIGN Twenty-nine patients with chronic, acute, and lymphomatous types of ATL were enrolled in a single-institution, nonrandomized, open-label phase II trial wherein patients received intravenous alemtuzumab 30 mg three times weekly for a maximum of 12 weeks. RESULTS Twenty-nine patients were evaluable for response and toxicity. The overall objective response was 15 of 29 patients [95% confidence interval (CI), 32.5%-70.6%]. The 15 patients who responded manifested a median time to response of 1.1 months. Median response duration was 1.4 months for the whole group and 14.5 months among responders. Median progression-free survival was 2.0 months. Median overall survival was 5.9 months. The most common adverse events were 2 with vasovagal episodes (7%) and 3 with hypotensive episodes (10%), leukopenia (41%) grade 3 and (17%) grade 4, lymphocytopenia (59%) grade 3, neutropenia (31%) grade 3, anemia (24%), and thrombocytopenia (10%). All patients developed cytomegalovirus antigenemia (CMV). Three were symptomatic and all responded to antiviral therapy. Grade 3 or 4 infections were reported in 4 (14%) of patients. CONCLUSIONS Alemtuzumab induced responses in patients with acute HTLV-1-associated ATL with acceptable toxicity, but with short duration of responses. These studies support inclusion of alemtuzumab in novel multidrug therapies for ATL. Clin Cancer Res; 23(1); 35-42. ©2016 AACR.
Collapse
Affiliation(s)
- Kamal Sharma
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - John E Janik
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Deirdre O'Mahony
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Donn Stewart
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Stefania Pittaluga
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | | | - Elaine S Jaffe
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Mark Raffeld
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Thomas A Fleisher
- Clinical Pathology Department, Mark O. Hatfield Clinical Research Center, NIH, Bethesda, Maryland
| | - Cathryn C Lee
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| | - John C Morris
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
108
|
Willis MD, Harding KE, Pickersgill TP, Wardle M, Pearson OR, Scolding NJ, Smee J, Robertson NP. Alemtuzumab for multiple sclerosis: Long term follow-up in a multi-centre cohort. Mult Scler 2016; 22:1215-23. [DOI: 10.1177/1352458515614092] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 10/01/2015] [Indexed: 11/15/2022]
Abstract
Background: Alemtuzumab has recently been approved for treatment of relapsing MS, but concerns remain about its use since long-term studies of adverse events remain limited. Furthermore, a clear understanding of its application and durability of effect in clinical practice has yet to evolve. Objectives: To investigate long-term efficacy and safety outcomes in a multicentre cohort of patients treated with alemtuzumab. Methods: Patients treated from 2000 and followed-up at three regional centres were identified. Baseline and prospective data were obtained and validated by clinical record review. Results: One hundred patients were identified with a mean follow-up of 6.1 years (range 1–13). Forty patients were retreated with at least one further treatment cycle. Annualized relapse rates fell from 2.1 to 0.2 ( p<0.0001) post-treatment and were sustained for up to eight years of follow-up. Mean change in EDSS score was +0.14. Forty-seven patients developed secondary autoimmunity. Conclusion: Observed reduction in relapse rates reflected those reported in clinical trials, but we were unable to corroborate previous observations of disability reversal. 40% of patients required additional treatment cycles. Autoimmune adverse events were common, occurring at a higher rate than previously reported, but were largely predictable, and could be managed effectively within a rigorous monitoring regime.
Collapse
Affiliation(s)
- MD Willis
- Institute of Psychological Medicine and Clinical Neuroscience, Cardiff University, University Hospital of Wales, UK/Department of Neurology, University Hospital of Wales, UK
| | - KE Harding
- Institute of Psychological Medicine and Clinical Neuroscience, Cardiff University, University Hospital of Wales, UK/Department of Neurology, University Hospital of Wales, UK
| | - TP Pickersgill
- Department of Neurology, University Hospital of Wales, UK
| | - M Wardle
- Department of Neurology, University Hospital of Wales, UK
| | - OR Pearson
- Department of Neurology, Morriston Hospital, UK
| | - NJ Scolding
- Department of Neurology, Southmead Hospital, UK
| | - J Smee
- Department of Neurology, University Hospital of Wales, UK
| | - NP Robertson
- Institute of Psychological Medicine and Clinical Neuroscience, Cardiff University, University Hospital of Wales, UK/Department of Neurology, University Hospital of Wales, UK
| |
Collapse
|
109
|
Milo R. Therapeutic strategies targeting B-cells in multiple sclerosis. Autoimmun Rev 2016; 15:714-8. [DOI: 10.1016/j.autrev.2016.03.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 01/08/2023]
|
110
|
Luessi F, Zipp F, Witsch E. Dendritic cells as therapeutic targets in neuroinflammation. Cell Mol Life Sci 2016; 73:2425-50. [PMID: 26970979 PMCID: PMC11108452 DOI: 10.1007/s00018-016-2170-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 02/02/2016] [Accepted: 02/25/2016] [Indexed: 12/23/2022]
Abstract
Multiple sclerosis (MS) is the most common chronic inflammatory demyelinating disorder of the central nervous system characterized by infiltration of immune cells and progressive damage to myelin sheaths and neurons. There is still no cure for the disease, but drug regimens can reduce the frequency of relapses and slightly delay progression. Myeloid cells or antigen-presenting cells (APCs) such as dendritic cells (DC), macrophages, and resident microglia, are key players in both mediating immune responses and inducing immune tolerance. Mounting evidence indicates a contribution of these myeloid cells to the pathogenesis of multiple sclerosis and to the effects of treatment, the understanding of which might provide strategies for more potent novel therapeutic interventions. Here, we review recent insights into the role of APCs, with specific focus on DCs in the modulation of neuroinflammation in MS.
Collapse
Affiliation(s)
- Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University of Mainz,Rhine Main Neuroscience Network (rmn2), Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University of Mainz,Rhine Main Neuroscience Network (rmn2), Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Esther Witsch
- Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University of Mainz,Rhine Main Neuroscience Network (rmn2), Langenbeckstrasse 1, 55131, Mainz, Germany.
| |
Collapse
|
111
|
Abstract
BACKGROUND Multiple sclerosis (MS) is an autoimmune, T-cell-dependent, inflammatory, demyelinating disease of the central nervous system, with an unpredictable course. Current MS therapies focus on treating exacerbations, preventing new exacerbations and avoiding the progression of disability. However, at present there is no effective treatment that is capable of safely and effectively reaching these objectives. This has led to the development and investigation of new drugs. Recent clinical trials suggest that alemtuzumab, a humanised monoclonal antibody against cell surface CD52, could be a promising option for MS. OBJECTIVES To assess the safety and effectiveness of alemtuzumab used alone or associated with other treatments to decrease disease activity in patients with any form of MS. SEARCH METHODS We searched the Trials Register of the Cochrane Multiple Sclerosis and Rare Diseases of the CNS Group (30 April 2015), which contains trials from the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, EMBASE, CINAHL, LILACS and the trial registry databases ClinicalTrials.gov and WHO International Clinical Trials Registry Platform. There was no restriction on the source, publication date or language. SELECTION CRITERIA All randomised clinical trials (RCTs) involving adults diagnosed with any form of MS according to the McDonald criteria, comparing alemtuzumab alone or associated with other medications, at any dose and for any duration, versus placebo or any other active drug therapy or alemtuzumab in other dose, regimen or duration. The co-primary outcomes were relapse-free survival, sustained disease progression and number of participants with at least one of any adverse events, including serious adverse events. DATA COLLECTION AND ANALYSIS Two independent review authors performed study selection, data extraction and 'Risk of bias' assessment. A third review author checked the process for accuracy. We used the Cochrane 'Risk of bias' tool to assess the risk of bias of the studies included in the review. We used the GRADE system to assess the quality of the body of evidence. To measure the treatment effect on dichotomous outcomes we used the risk ratio (RR); for the treatment effect on continuous outcomes, we used the mean difference (MD) and for time-to-event outcomes we used hazard ratio (HR). We calculated 95% confidence intervals (CI) for these measures. When there was no heterogeneity, we used a fixed-effect model to pool data. MAIN RESULTS Three RCTs (1713 participants) fulfilled the selection criteria and we included them in the review. All three trials compared alemtuzumab versus subcutaneous interferon beta-1a for patients with relapsing-remitting MS. Patients were treatment-naive in the CARE-MS and CAMMS223 studies. The CARE-MS II study included patients with at least one relapse while being treated with interferon beta or glatiramer acetate. Alemtuzumab was given for 12 or 24 months; for some outcomes, the follow-up period reached 36 months. The regimens were (a) 12 mg or 24 mg per day administered intravenously, once a day for five consecutive days at month 0 and 12 or (b) 24 mg per day, intravenously, once a day for three consecutive days at month 12 and 24. The patients in the other arm of the trials received interferon beta-1a 44 μg subcutaneously three times weekly after dose titration.At 24 months, alemtuzumab 12 mg was associated with: (a) higher relapse-free survival (hazard ratio (HR) 0.50, 95% CI 0.41 to 0.60; 1248 participants, two studies, moderate quality evidence); (b) higher sustained disease progression-free survival (HR 0.62, 95% CI 0.44 to 0.87; 1191 participants; two studies; moderate quality evidence); (c) a slightly higher number of participants with at least one adverse event (RR 1.04, 95% CI 1.01 to 1.06; 1248 participants; two studies; moderate quality evidence); (d) a lower number of participants with new or enlarging T2-hyperintense lesions on magnetic resonance imaging (MRI) (RR 0.74, 95% CI 0.59 to 0.91; 1238 participants; two studies; I(2) = 80%); and (e) a lower number of dropouts (RR 0.31, 95% CI 0.23 to 0.41; 1248 participants; two studies, I(2) = 29%; low quality evidence).At 36 months, alemtuzumab 24 mg was associated with: (a) higher relapse-free survival (45 versus 17; HR 0.21, 95% CI 0.11 to 0.40; one study; 221 participants); (b) a higher sustained disease progression-free survival (HR 0.33, 95% CI 0.16 to 0.69; one study; 221 participants); and (c) no statistical difference in the rate of participants with at least one adverse event. We did not find any study that reported any of the following outcomes: rate of participants free of clinical disease activity, quality of life, fatigue or change in the numbers of MRI T2- and T1-weighted lesions after treatment. It was not possible to perform subgroup analyses according to disease type and disability at baseline due to lack of data. AUTHORS' CONCLUSIONS In patients with relapsing-remitting MS, alemtuzumab 12 mg was better than subcutaneous interferon beta-1a for the following outcomes assessed at 24 months: relapse-free survival, sustained disease progression-free survival, number of participants with at least one adverse event and number of participants with new or enlarging T2-hyperintense lesions on MRI. The quality of the evidence for these results was low to moderate. Alemtuzumab 24 mg seemed to be better than subcutaneous interferon beta-1a for relapse-free survival and sustained disease progression-free survival, at 36 months.More randomised clinical trials are needed to evaluate the effects of alemtuzumab on other forms of MS and compared with other therapeutic options. These new studies should assess additional relevant outcomes such as the rate of participants free of clinical disease activity, quality of life, fatigue and adverse events (individual rates, serious adverse events and long-term adverse events). Moreover, these new studies should evaluate other doses and durations of alemtuzumab course.
Collapse
Affiliation(s)
- Rachel Riera
- Brazilian Cochrane CentreCentro de Estudos em Medicina Baseada em Evidências e Avaliação Tecnológica em SaúdeRua Borges Lagoa, 564 cj 63São PauloSPBrazil04038‐000
| | - Gustavo JM Porfírio
- Brazilian Cochrane CentreCentro de Estudos em Medicina Baseada em Evidências e Avaliação Tecnológica em SaúdeRua Borges Lagoa, 564 cj 63São PauloSPBrazil04038‐000
| | - Maria R Torloni
- Centro de Estudos de Saúde Baseada em Evidências e Avaliação Tecnológica em SaúdeBrazilian Cochrane CentreRua Borges Lagoa, 564 cj 63São PauloSão PauloBrazil04038‐000
| | | |
Collapse
|
112
|
Obermann M, Ruck T, Pfeuffer S, Baum J, Wiendl H, Meuth SG. Simultaneous early-onset immune thrombocytopenia and autoimmune thyroid disease following alemtuzumab treatment in relapsing-remitting multiple sclerosis. Mult Scler 2016; 22:1235-41. [PMID: 26980848 DOI: 10.1177/1352458516638558] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 02/11/2016] [Indexed: 11/15/2022]
Abstract
OBJECTIVE We report two cases of patients with relapsing-remitting multiple sclerosis with early-onset thrombocytopenia and autoimmune thyroid disease after the first treatment course with 60-mg alemtuzumab. METHODS Case series and review of the literature. RESULTS Both patients showed severe thrombocytopenia with platelet counts of 2 × 10(9) and 11 × 10(9)/L, respectively, as well as increased thyroid antibodies within only a few months after initiating alemtuzumab treatment (11 and 9 months). Both patients responded considerably well to medical therapy including corticosteroids and intravenous immunoglobulins with slow platelet recovery over several weeks. Interestingly, both patients were previously treated with fingolimod and showed a marked lymphocytopenia that led to discontinuation. CONCLUSION These cases emphasize the necessity of careful clinical surveillance and proper education of patients treated with alemtuzumab as proposed by the safety-monitoring program. Previous severe lymphocytopenia under therapy with other disease-modifying therapies may be a risk factor for the development of immune thrombocytopenia.
Collapse
Affiliation(s)
- Mark Obermann
- Center for Neurology, Asklepios Hospitals Schildautal, Seesen, Germany/Departments of Neurology and Haematology, University of Duisburg-Essen, Duisburg, Germany
| | - Tobias Ruck
- Department of Neurology, University of Münster, Münster, Germany
| | - Steffen Pfeuffer
- Department of Neurology, University of Münster, Münster, Germany
| | - Julia Baum
- Departments of Neurology and Haematology, University of Duisburg-Essen, Duisburg, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Münster, Germany
| | - Sven G Meuth
- Department of Neurology, University of Münster, Münster, Germany
| |
Collapse
|
113
|
Ruck T, Afzali AM, Lukat KF, Eveslage M, Gross CC, Pfeuffer S, Bittner S, Klotz L, Melzer N, Wiendl H, Meuth SG. ALAIN01--Alemtuzumab in autoimmune inflammatory neurodegeneration: mechanisms of action and neuroprotective potential. BMC Neurol 2016; 16:34. [PMID: 26966029 PMCID: PMC4785638 DOI: 10.1186/s12883-016-0556-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 03/02/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Alemtuzumab (Lemtrada®) is a newly approved therapeutic agent for relapsing-remitting multiple sclerosis (RRMS). In previous phase II and III clinical trials, alemtuzumab has proven superior efficacy to subcutaneous interferon beta-1a concerning relapse rate and disability progression with unprecedented durability and long-lasting freedom of disease activity. The humanized monoclonal antibody targets CD52, leading to a rapid and long-lasting depletion, especially of B and T cells. Arising from hematopoietic precursor cells a fundamental reprogramming of the immune system restores tolerogenic networks effectively suppressing autoimmune inflammatory responses in the central nervous system (CNS). Despite its favourable effects alemtuzumab holds a severe risk of side effects with secondary autoimmunity being the most considerable. Markers for risk stratification and treatment response improving patient selection and therapy guidance are a big unmet need for MS patients and health care providers. METHODS/DESIGN This is a mono center, single arm, explorative phase IV study including 15 patients with highly active RRMS designed for 3 years. Patients will be studied by a high-resolution analysis comprising a repertoire of various immunological assays for the detection of immune cells and their function in peripheral blood as well as the cerebrospinal fluid (CSF). These assays encompass a number of experiments investigating immune cell subset composition, activation status, cytokine secretion, migratory capacity, potential neuroprotective properties and cytolytic activity complemented by instrument-based diagnostics like MRI scans, evoked potentials and optical coherence tomography (OCT). DISCUSSION Our study represents the first in-depth and longitudinal functional analysis of key immunological parameters in the periphery and the CNS compartment underlying the fundamental effects of alemtuzumab in MS patients. By combining clinical, experimental and MRI data our study will provide a deeper understanding of alemtuzumab's mechanisms of action (MOA) potentially identifying immune signatures associated with treatment response or the development of secondary autoimmunity. After validation in larger cohorts this might help to improve efficacy and safety of alemtuzumab therapy in RRMS patients. TRIAL REGISTRATION NCT02419378 (clinicaltrials.gov), registered 31 March 2015.
Collapse
Affiliation(s)
- Tobias Ruck
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| | - Ali Maisam Afzali
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | | | - Maria Eveslage
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Catharina C Gross
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Steffen Pfeuffer
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Stefan Bittner
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Luisa Klotz
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Nico Melzer
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Sven G Meuth
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| |
Collapse
|
114
|
EXP CLIN TRANSPLANTExp Clin Transplant 2016; 14. [DOI: 10.6002/ect.2015.0254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
115
|
Klebanoff CA, Rosenberg SA, Restifo NP. Prospects for gene-engineered T cell immunotherapy for solid cancers. Nat Med 2016; 22:26-36. [PMID: 26735408 PMCID: PMC6295670 DOI: 10.1038/nm.4015] [Citation(s) in RCA: 269] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 11/20/2015] [Indexed: 02/08/2023]
Abstract
Adoptive transfer of receptor-engineered T cells has produced impressive results in treating patients with B cell leukemias and lymphomas. This success has captured public imagination and driven academic and industrial researchers to develop similar 'off-the-shelf' receptors targeting shared antigens on epithelial cancers, the leading cause of cancer-related deaths. However, the successful treatment of large numbers of people with solid cancers using this strategy is unlikely to be straightforward. Receptor-engineered T cells have the potential to cause lethal toxicity from on-target recognition of normal tissues, and there is a paucity of truly tumor-specific antigens shared across tumor types. Here we offer our perspective on how expanding the use of genetically redirected T cells to treat the majority of patients with solid cancers will require major technical, manufacturing and regulatory innovations centered around the development of autologous gene therapies targeting private somatic mutations.
Collapse
Affiliation(s)
- Christopher A Klebanoff
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Steven A Rosenberg
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Nicholas P Restifo
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
116
|
Al-Khamis FA. The use of immune modulating drugs for the treatment of multiple sclerosis. NEUROSCIENCES (RIYADH, SAUDI ARABIA) 2016; 21:4-9. [PMID: 26818160 PMCID: PMC5224410 DOI: 10.17712/nsj.2016.1.20150252] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review discusses the mechanisms of action of 4 immune modulating drugs currently used in the treatment of multiple sclerosis (MS), including Alemtuzumab, a humanized monoclonal antibody that functions by targeting CD52, an antigen primarily expressed on T and B lymphocytes and monocytes/macrophages, resulting in their depletion and subsequent repopulation; Dimethyl fumarate that switches cytokine production toward a T helper 2 profile and enhances cytosolic levels of nuclear factor erythroid 2-related factor 2, which has immune regulatory and cytoprotective effects on oligodendrocytes, neurons, and glial cells; Fingolimod functions by blocking the release of activated lymphocytes from lymph nodes by targeting sphingosin-1-phosphate receptors; Natalizumab a humanized monoclonal antibody binds α4β1-integrin resulting in reduced migration of immune cells from blood across the blood-brain barrier into the CNS. This review presents the most up to date information on mechanisms of action, safety, and efficacy of these immune modulators and provides future perspectives for the treatment of MS.
Collapse
Affiliation(s)
- Fahd A. Al-Khamis
- From the Department of Neurology, Faculty of Medicine, Deanship for Scientific Research, University of Dammam, Dammam, Kingdom of Saudi Arabia,Address correspondence and reprint request to: Dr. Fahd A. Al-Khamis, Department of Neurology, Faculty of Medicine, Deanship for Scientific Research, University of Dammam, Dammam, Kingdom of Saudi Arabia. E-mail:
| |
Collapse
|
117
|
Abstract
Alemtuzumab is a humanized monoclonal antibody against CD52 (cluster of differentiation 52) and is approved for the therapy of relapsing-remitting multiple sclerosis. The application of alemtuzumab leads to a rapid, but long-lasting depletion predominantly of CD52-bearing B and T cells with reprogramming effects on immune cell composition resulting in the restoration of tolerogenic networks. Alemtuzumab has proven high efficacy in clinical phase II and III trials, where interferon β-1a was used as active comparator. However, alemtuzumab is associated with frequent and considerable risks. Most importantly secondary autoimmune disease affects 30%-40% of patients, predominantly impairing thyroid function. Extensive monitoring and early intervention allow for an appropriate risk management. However, new and reliable biomarkers for individual risk stratification and treatment response to improve patient selection and therapy guidance are a significant unmet need. Only a deeper understanding of the underlying mechanisms of action (MOA) will reveal such markers, maximizing the best potential risk-benefit ratio for the individual patient. This review provides and analyses the current knowledge on the MOA of alemtuzumab. Most recent data on efficacy and safety of alemtuzumab are presented and future research opportunities are discussed.
Collapse
|
118
|
Turner MJ, Pang PT, Chretien N, Havari E, LaMorte MJ, Oliver J, Pande N, Masterjohn E, Carter K, Reczek D, Brondyk W, Roberts BL, Kaplan JM, Siders WM. Reduction of inflammation and preservation of neurological function by anti-CD52 therapy in murine experimental autoimmune encephalomyelitis. J Neuroimmunol 2015. [PMID: 26198912 DOI: 10.1016/j.jneuroim.2015.05.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Alemtuzumab, a monoclonal antibody directed against human CD52, is used in the treatment of MS. To characterize the impact of anti-CD52 administration, a monoclonal antibody to mouse CD52 (anti-muCD52) was generated and evaluated in EAE mouse models of MS. A single course of anti-muCD52 provided a therapeutic benefit accompanied by a reduction in the frequency of autoreactive T lymphocytes and production of pro-inflammatory cytokines. Examination of the CNS revealed a decrease in infiltrating lymphocytes, demyelination and axonal loss. Electrophysiological assessment showed preservation of axonal conductance in the spinal cord. These findings suggest that anti-CD52 therapy may help preserve CNS integrity.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antigens, CD/immunology
- Antigens, Neoplasm/immunology
- Axons/drug effects
- Axons/immunology
- Axons/pathology
- CD52 Antigen
- Demyelinating Diseases/drug therapy
- Demyelinating Diseases/immunology
- Demyelinating Diseases/pathology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Glycoproteins/antagonists & inhibitors
- Glycoproteins/immunology
- Humans
- Inflammation/drug therapy
- Inflammation/immunology
- Inflammation/pathology
- Inflammation Mediators/antagonists & inhibitors
- Inflammation Mediators/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Sequence Data
Collapse
Affiliation(s)
- Michael J Turner
- Neuroimmunology Research, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States.
| | - Petti T Pang
- Neuroimmunology Research, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| | - Nathalie Chretien
- Neuroimmunology Research, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| | - Evis Havari
- Neuroimmunology Research, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| | - Michael J LaMorte
- Neuroimmunology Research, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| | - Julian Oliver
- Pathology, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| | - Nilesh Pande
- Neuroimmunology Research, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| | - Elizabeth Masterjohn
- Biologics Discovery, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| | - Karen Carter
- Biologics Discovery, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| | - David Reczek
- Biologics Discovery, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| | - William Brondyk
- Biologics Discovery, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| | - Bruce L Roberts
- Neuroimmunology Research, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| | - Johanne M Kaplan
- Neuroimmunology Research, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| | - William M Siders
- Neuroimmunology Research, Genzyme, a Sanofi company, 49 New York Avenue, Framingham, MA 01701, United States
| |
Collapse
|
119
|
Upregulation of CD38 expression on multiple myeloma cells by all-trans retinoic acid improves the efficacy of daratumumab. Leukemia 2015; 29:2039-49. [PMID: 25975191 DOI: 10.1038/leu.2015.123] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/21/2015] [Accepted: 05/05/2015] [Indexed: 12/21/2022]
Abstract
Daratumumab is an anti-CD38 monoclonal antibody with lytic activity against multiple myeloma (MM) cells, including ADCC (antibody-dependent cellular cytotoxicity) and CDC (complement-dependent cytotoxicity). Owing to a marked heterogeneity of response to daratumumab therapy in MM, we investigated determinants of the sensitivity of MM cells toward daratumumab-mediated ADCC and CDC. In bone marrow samples from 144 MM patients, we observed no difference in daratumumab-mediated lysis between newly diagnosed or relapsed/refractory patients. However, we discovered, next to an expected effect of effector (natural killer cells/monocytes) to target (MM cells) ratio on ADCC, a significant association between CD38 expression and daratumumab-mediated ADCC (127 patients), as well as CDC (56 patients). Similarly, experiments with isogenic MM cell lines expressing different levels of CD38 revealed that the level of CD38 expression is an important determinant of daratumumab-mediated ADCC and CDC. Importantly, all-trans retinoic acid (ATRA) increased CD38 expression levels but also reduced expression of the complement-inhibitory proteins CD55 and CD59 in both cell lines and primary MM samples. This resulted in a significant enhancement of the activity of daratumumab in vitro and in a humanized MM mouse model as well. Our results provide the preclinical rationale for further evaluation of daratumumab combined with ATRA in MM patients.
Collapse
|
120
|
Wang H, Dong J, Shi P, Liu J, Zuo L, Li Y, Gong J, Gu L, Zhao J, Zhang L, Zhang W, Zhu W, Li N, Li J. Anti-mouse CD52 monoclonal antibody ameliorates intestinal epithelial barrier function in interleukin-10 knockout mice with spontaneous chronic colitis. Immunology 2015; 144:254-62. [PMID: 25087772 DOI: 10.1111/imm.12366] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/30/2014] [Accepted: 07/30/2014] [Indexed: 01/01/2023] Open
Abstract
Intestinal inflammation causes tight junction changes and death of epithelial cells, and plays an important role in the development of Crohn's disease (CD). CD52 monoclonal antibody (CD52 mAb) directly targets the cell surface CD52 and is effective in depleting mature lymphocytes by cytolytic effects in vivo, leading to long-lasting changes in adaptive immunity. The aim of this study was to investigate the therapeutic effect of CD52 mAb on epithelial barrier function in animal models of IBD. Interleukin-10 knockout mice (IL-10(-/-) ) of 16 weeks with established colitis were treated with CD52 mAb once a week for 2 weeks. Severity of colitis, CD4(+) lymphocytes and cytokines in the lamina propria, epithelial expression of tight junction proteins, morphology of tight junctions, tumour necrosis factor-α (TNF-α)/TNF receptor 2 (TNFR2) mRNA expression, myosin light chain kinase (MLCK) expression and activity, as well as epithelial apoptosis in proximal colon were measured at the end of the experiment. CD52 mAb treatment effectively attenuated colitis associated with decreased lamina propria CD4(+) lymphocytes and interferon-γ/IL-17 responses in colonic mucosa in IL-10(-/-) mice. After CD52 mAb treatment, attenuation of colonic permeability, increased epithelial expression and correct localization of tight junction proteins (occludin and zona occludens protein-1), as well as ameliorated tight junction morphology were observed in IL-10(-/-) mice. CD52 mAb treatment also effectively suppressed the epithelial apoptosis, mucosa TNF-α mRNA expression, epithelial expression of long MLCK, TNFR2 and phosphorylation of MLC. Our results indicated that anti-CD52 therapy may inhibit TNF-α/TNFR2-mediated epithelial apoptosis and MLCK-dependent tight junction permeability by depleting activated T cells in the gut mucosa.
Collapse
Affiliation(s)
- Honggang Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Affiliation(s)
- Mark D Willis
- Institute of Psychological Medicine and Clinical Neuroscience, Cardiff University, University Hospital of Wales, Heath Park, Cardiff, UK
| | - Neil P Robertson
- Institute of Psychological Medicine and Clinical Neuroscience, Cardiff University, University Hospital of Wales, Heath Park, Cardiff, UK
| |
Collapse
|
122
|
Havrdova E, Horakova D, Kovarova I. Alemtuzumab in the treatment of multiple sclerosis: key clinical trial results and considerations for use. Ther Adv Neurol Disord 2015; 8:31-45. [PMID: 25584072 DOI: 10.1177/1756285614563522] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alemtuzumab is a humanized monoclonal antibody therapy that has recently been approved in over 30 countries for patients with active relapsing-remitting multiple sclerosis. It acts by targeting CD52, an antigen primarily expressed on T and B lymphocytes, resulting in their depletion and subsequent repopulation. The alemtuzumab clinical development program used an active comparator, subcutaneous interferon beta-1a, to show that alemtuzumab is a highly efficacious disease-modifying therapy, with benefits on relapses, disability outcomes, and freedom from clinical disease and magnetic resonance imaging activity. The safety profile was consistent across studies and no new safety signals have emerged during follow-up in the extension study. Infusion-associated reactions are common with alemtuzumab, but rarely serious. Infection incidence was elevated with alemtuzumab in clinical studies; most infections were mild or moderate in severity. Autoimmune adverse events occurred in approximately a third of patients, manifesting mainly as thyroid disorders, and less frequently as immune thrombocytopenia or nephropathy. A comprehensive monitoring program lasting at least 4 years after the last alemtuzumab dose allows early detection and effective management of autoimmune adverse events. Further experience with alemtuzumab in the clinic will provide needed long-term data.
Collapse
Affiliation(s)
- Eva Havrdova
- Department of Neurology and Center for Clinical Neuroscience, First Medical Faculty, Charles University in Prague, Katerinska 30, Prague, 120 00, Czech Republic
| | - Dana Horakova
- Department of Neurology and Center for Clinical Neuroscience, First Medical Faculty, Charles University in Prague, Prague, Czech Republic
| | - Ivana Kovarova
- Department of Neurology and Center for Clinical Neuroscience, First Medical Faculty, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
123
|
Ni X, Jorgensen JL, Goswami M, Challagundla P, Decker WK, Kim YH, Duvic MA. Reduction of Regulatory T Cells by Mogamulizumab, a Defucosylated Anti-CC Chemokine Receptor 4 Antibody, in Patients with Aggressive/Refractory Mycosis Fungoides and Sézary Syndrome. Clin Cancer Res 2014; 21:274-85. [DOI: 10.1158/1078-0432.ccr-14-0830] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
124
|
Caspase-8 polymorphisms result in reduced Alemtuzumab-induced T-cell apoptosis and worse survival after transplantation. Bone Marrow Transplant 2014; 50:237-43. [PMID: 25347010 DOI: 10.1038/bmt.2014.238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/11/2014] [Accepted: 09/13/2014] [Indexed: 11/08/2022]
Abstract
Allo-SCT using unrelated donors is a curative treatment for patients with hematological disorders. The best donor is one matched for 10/10 HLA alleles, however studies have shown an additional survival benefit when considering other genetic factors. It has been shown that a six-nucleotide insertion/deletion polymorphism in the CASP8 gene promoter results in reduced susceptibility of T lymphocytes to undergo apoptosis. In 186 SCT recipients, we found a significantly better OS in those who received a transplant from a WT/WT donor compared with donors with a deletion (3 years: 52 vs 34%; P=0.03; multivariate analysis; RR 0.61; 95% CI 0.38-0.98, P=0.04). This was more marked when both the patient and the donor had a deletion (3 years OS: 62% compared with 36%, P=0.01). As the majority of these patients received Alemtuzumab during conditioning, we went on to analyze the in vitro effect of the polymorphism on Alemtuzumab-induced apoptosis. We showed statistically significantly higher percentages of apoptotic naïve CD4 (P<0.0005) and CD8 (P<0.0005) T cells in WT/WT donors in comparison with donors with a deletion. These data imply an unrecognized role for the CASP8 promoter polymorphism on survival following unrelated SCT particularly in the context of T-cell depletion with Alemtuzumab.
Collapse
|
125
|
Abstract
Alemtuzumab is a humanized monoclonal antibody directed against CD52 to deplete circulating T and B lymphocytes; lymphocyte depletion is followed by a distinctive pattern of T- and B-cell repopulation, changing the balance of the immune system. This review reports the efficacy and safety findings of the phase 2 CAMMS223 trial and the phase 3 CARE-MS I and II trials investigating alemtuzumab for the treatment of active relapsing–remitting MS. Alemtuzumab, administered intravenously, was shown to improve relapse rate versus subcutaneous interferon beta-1a in patients who were treatment-naive (CAMMS223 and CARE-MS I) or had relapsed on prior therapy (CARE-MS II), and to reduce sustained accumulation of disability (CAMMS223 and CARE-MS II). Important adverse events were infusion-associated reactions, serious infections and autoimmune events. A safety monitoring program allowed for early detection and management of autoimmune events. Recommendations for the monitoring of adverse events are made. Alemtuzumab’s mechanism of action, pharmacodynamics and opportunities for future research are discussed.
Collapse
Affiliation(s)
- Hans-Peter Hartung
- Department of Neurology and Center for Neuropsychiatry, Medical Faculty, Heinrich-Heine University, Germany
| | - Orhan Aktas
- Department of Neurology and Center for Neuropsychiatry, Medical Faculty, Heinrich-Heine University, Germany
| | - Alexey N Boyko
- Moscow City Center of Multiple Sclerosis, Russian Federation, Russia
| |
Collapse
|
126
|
Lee F, Luevano M, Veys P, Yong K, Madrigal A, Shaw BE, Saudemont A. The effects of CAMPATH-1H on cell viability do not correlate to the CD52 density on the cell surface. PLoS One 2014; 9:e103254. [PMID: 25050704 PMCID: PMC4106894 DOI: 10.1371/journal.pone.0103254] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 06/30/2014] [Indexed: 11/23/2022] Open
Abstract
Graft versus host disease (GvHD) is one of the main complications after hematological stem cell transplantation (HSCT). CAMPATH-1H is used in the pre-transplant conditioning regimen to effectively reduce GvHD by targeting CD52 antigens on T cells resulting in their depletion. Information regarding CD52 expression and the effects of CAMPATH-1H on immune cells is scant and limited to peripheral blood (PB) T and B cells. To date, the effects of CAMPATH-1H on cord blood (CB) cells has not been studied. Here we aimed to analyze CD52 expression and the effects of CAMPATH-1H on fresh or frozen, resting or activated, PB mononuclear cells (PBMC) and CB mononuclear cells (CBMC). In resting state, CD52 expression was higher in CB than PB T cell subsets (653.66±26.68 vs 453.32±19.2) and B cells (622.2±20.65 vs 612.0±9.101) except for natural killer (NK) cells where CD52 levels were higher in PB (421.0±9.857) than CB (334.3±9.559). In contrast, CD52 levels were comparable across all cell types after activation. CAMPATH-1H depleted resting cells more effectively than activated cells with approximately 80–95% of apoptosis observed with low levels of necrosis. There was no direct correlation between cell surface CD52 density and depleting effects of CAMPATH-1H. In addition, no difference in cell viability was noted when different concentrations of CAMPATH-1H were used. CD52 was not expressed on HSC but began to be expressed as the cells differentiate, implying that CAMPATH-1H could potentially affect HSC differentiation and proliferation. Our study provides insightful information, which contributes to the better understanding in the use of CAMPATH-1H as part of the conditioning regime in HSCT.
Collapse
MESH Headings
- Alemtuzumab
- Antibodies, Monoclonal, Humanized/pharmacology
- Antigens, CD/analysis
- Antigens, CD/immunology
- Antigens, Neoplasm/analysis
- Antigens, Neoplasm/immunology
- Antineoplastic Agents/pharmacology
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- CD52 Antigen
- Cell Separation
- Cell Survival/drug effects
- Glycoproteins/analysis
- Glycoproteins/immunology
- Humans
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
Collapse
Affiliation(s)
- Fuiyee Lee
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, London, United Kingdom
| | - Martha Luevano
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, London, United Kingdom
| | - Paul Veys
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, London, United Kingdom
- Great Ormond Street Hospital for Children NHS, London, United Kingdom
| | - Kwee Yong
- University College London, Cancer Institute, London, United Kingdom
| | - Alejandro Madrigal
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, London, United Kingdom
| | - Bronwen E. Shaw
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, London, United Kingdom
- Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Aurore Saudemont
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, London, United Kingdom
- * E-mail:
| |
Collapse
|
127
|
Abstract
INTRODUCTION Alemtuzumab is a humanised anti-CD52 mAb which has recently been licensed for the treatment of relapsing multiple sclerosis in Europe. AREAS COVERED The efficacy and safety of alemtuzumab from open label, Phase II and Phase III trials is reported. EXPERT OPINION Alemtuzumab causes rapid and profound complement mediated lysis of circulating lymphocytes and allows beneficial modulation of the immune system during a subsequent reconstitution phase. Clinical trials have demonstrated superior efficacy against an active comparator, with reduction in annualised relapse rates and sustained accumulation of disability at 3 years and sustained efficacy at 5 years. The main adverse effects are mild to moderate infusion reactions, an increased incidence of mild to moderate infections and autoimmune adverse events. Thyroid disorders are the most common form of autoimmune adverse events, occurring in approximately one third of patients. Overt Graves' hyperthyroidism represents approximately half of these cases. Careful patient selection and structured monitoring programs allow for effective patient management resulting in a favourable risk benefit profile.
Collapse
Affiliation(s)
- Mark Willis
- Institute of Psychological Medicine and Clinical Neuroscience, Cardiff University School of Medicine , Cardiff , UK
| | | |
Collapse
|
128
|
Hoermann G, Blatt K, Greiner G, Putz EM, Berger A, Herrmann H, Cerny-Reiterer S, Gleixner KV, Walz C, Hoetzenecker K, Müllauer L, Reiter A, Sotlar K, Sexl V, Valent P, Mayerhofer M. CD52 is a molecular target in advanced systemic mastocytosis. FASEB J 2014; 28:3540-51. [PMID: 24760752 DOI: 10.1096/fj.14-250894] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Advanced systemic mastocytosis (SM) is an aggressive hematopoietic neoplasm with poor prognosis and short survival times. So far, no curative therapy is available for affected patients. We have identified the cell surface antigen CD52 (CAMPATH-1) as a molecular target expressed abundantly on the surface of primary neoplastic mast cells (MCs) in patients with advanced SM. In contrast, neoplastic MCs of patients with indolent SM and normal MCs expressed only low levels or did not express CD52. To study the mechanisms of CD52 expression and the value of this antigen as a potential therapeutic target, we generated a human MC cell line, designated MCPV-1, by lentiviral immortalization of cord blood-derived MC progenitor cells. Functional studies revealed that activated RAS profoundly promotes surface expression of CD52. The CD52-targeting antibody alemtuzumab induced cell death in CD52(+) primary neoplastic MCs obtained from patients with SM as well as in MCPV-1 cells. NSG mice xenotransplanted with MCPV-1 cells survived significantly longer after treatment with alemtuzumab (median survival: 31 d untreated vs. 46 d treated; P=0.0012). We conclude that CD52 is a novel marker and potential therapeutic target in neoplastic MCs in patients with advanced SM.
Collapse
Affiliation(s)
| | - Katharina Blatt
- Department of Internal Medicine I, Division of Hematology and Hemostaseology
| | | | - Eva Maria Putz
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Angelika Berger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Sabine Cerny-Reiterer
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Ludwig Boltzmann Cluster Oncology, Vienna, Austria
| | - Karoline V Gleixner
- Department of Internal Medicine I, Division of Hematology and Hemostaseology
| | - Christoph Walz
- Institute of Pathology, Ludwig Maximilians University Munich, Munich, Germany
| | | | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Andreas Reiter
- Medizinische Universitätsklinik, Universitätsmedizin Mannheim, Mannheim, Germany; and
| | - Karl Sotlar
- Institute of Pathology, Ludwig Maximilians University Munich, Munich, Germany
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Ludwig Boltzmann Cluster Oncology, Vienna, Austria
| | | |
Collapse
|
129
|
Havari E, Turner MJ, Campos-Rivera J, Shankara S, Nguyen TH, Roberts B, Siders W, Kaplan JM. Impact of alemtuzumab treatment on the survival and function of human regulatory T cells in vitro. Immunology 2014; 141:123-31. [PMID: 24116901 DOI: 10.1111/imm.12178] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 09/27/2013] [Accepted: 09/30/2013] [Indexed: 12/20/2022] Open
Abstract
Alemtuzumab is a humanized monoclonal antibody specific for the CD52 protein present at high levels on the surface of B and T lymphocytes. In clinical trials, alemtuzumab has shown a clinical benefit superior to that of interferon-β in relapsing-remitting multiple sclerosis patients. Treatment with alemtuzumab leads to the depletion of circulating lymphocytes followed by a repopulation process characterized by alterations in the number, proportions and properties of lymphocyte subsets. Of particular interest, an increase in the percentage of T cells with a regulatory phenotype (Treg cells) has been observed in multiple sclerosis patients after alemtuzumab. Since Treg cells play an important role in the control of autoimmune responses, the effect of alemtuzumab on Treg cells was further studied in vitro. Alemtuzumab effectively mediated complement-dependent cytolysis of human T lymphocytes and the remaining population was enriched in T cells with a regulatory phenotype. The alemtuzumab-exposed T cells displayed functional regulatory characteristics including anergy to stimulation with allogeneic dendritic cells and ability to suppress the allogeneic response of autologous T cells. Consistent with the observed increase in Treg cell frequency, the CD25(hi) T-cell population was necessary for the suppressive activity of alemtuzumab-exposed T cells. The mechanism of this suppression was found to be dependent on both cell-cell contact and interleukin-2 consumption. These findings suggest that an alemtuzumab-mediated increase in the proportion of Treg cells may play a role in promoting the long-term efficacy of alemtuzumab in patients with multiple sclerosis.
Collapse
Affiliation(s)
- Evis Havari
- Neuroimmunology Research, Genzyme, a Sanofi Company, Framingham, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
130
|
|
131
|
Novitzky N, Davison G, Abdulla R, Mowla S. Definition of the Variables Affecting Efficacy of Immunodepletion Ex Vivo of Peripheral Blood Progenitor Cell Grafts by Alemtuzumab (Campath in the Bag). Biol Blood Marrow Transplant 2013; 19:1753-9. [DOI: 10.1016/j.bbmt.2013.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 10/01/2013] [Indexed: 11/25/2022]
|
132
|
Abstract
Alemtuzumab is a humanized anti-CD52 monoclonal antibody. Treatment in humans results in a rapid, profound, and prolonged B- and T-cell lymphopenia. Subsequently, lymphocyte reconstitution by homeostatic mechanisms alters the composition, phenotype, and function of T-cell subsets, thus allowing the immune system to be 'reset'. One phase II and two phase III randomized, multicenter, single-blinded (outcomes assessor) clinical trials of alemtuzumab in relapsing-remitting multiple sclerosis have now been completed. Against an active comparator and the current first-line therapy for relapsing-remitting multiple sclerosis (interferon-beta), alemtuzumab showed a significant reduction in annualized relapse rate as well as a significant reduction in the accumulation of disability. These outcomes are sustained over at least 5 years following treatment. The most common adverse effects are mild infusion reactions, an increased incidence of mild-to-moderate severity infections and secondary autoimmunity. The latter is observed in a third of treated patients, commonly thyroid disease but other target cells have been described including cytopenias. Marketing authorization applications have been submitted for the use of alemtuzumab in multiple sclerosis to the Food and Drug Administration and the European Medicines Agency, with licensing expected in 2013. Here, we discuss the outlook for alemtuzumab in multiple sclerosis in light of the currently available therapies, outcomes of and lessons learnt from clinical trials, and the overall position of monoclonal antibodies in modern treatment strategies.
Collapse
|
133
|
Zhang X, Tao Y, Chopra M, Ahn M, Marcus KL, Choudhary N, Zhu H, Markovic-Plese S. Differential reconstitution of T cell subsets following immunodepleting treatment with alemtuzumab (anti-CD52 monoclonal antibody) in patients with relapsing-remitting multiple sclerosis. THE JOURNAL OF IMMUNOLOGY 2013; 191:5867-74. [PMID: 24198283 DOI: 10.4049/jimmunol.1301926] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alemtuzumab (anti-CD52 mAb) provides long-lasting disease activity suppression in relapsing-remitting multiple sclerosis (RRMS). The objective of this study was to characterize the immunological reconstitution of T cell subsets and its contribution to the prolonged RRMS suppression following alemtuzumab-induced lymphocyte depletion. The study was performed on blood samples from RRMS patients enrolled in the CARE-MS II clinical trial, which was recently completed and led to the submission of alemtuzumab for U.S. Food and Drug Administration approval as a treatment for RRMS. Alemtuzumab-treated patients exhibited a nearly complete depletion of circulating CD4(+) lymphocytes at day 7. During the immunological reconstitution, CD4(+)CD25(+)CD127(low) regulatory T cells preferentially expanded within the CD4(+) lymphocytes, reaching their peak expansion at month 1. The increase in the percentage of TGF-β1-, IL-10-, and IL-4-producing CD4(+) cells reached a maximum at month 3, whereas a significant decrease in the percentages of Th1 and Th17 cells was detected at months 12 and 24 in comparison with the baseline. A gradual increase in serum IL-7 and IL-4 and a decrease in IL-17A, IL-17F, IL-21, IL-22, and IFN-γ levels were detected following treatment. In vitro studies have demonstrated that IL-7 induced an expansion of CD4(+)CD25(+)CD127(low) regulatory T cells and a decrease in the percentages of Th17 and Th1 cells. In conclusion, our results indicate that differential reconstitution of T cell subsets and selectively delayed CD4(+) T cell repopulation following alemtuzumab-induced lymphopenia may contribute to its long-lasting suppression of disease activity.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | | | | | | | | | | | | | | |
Collapse
|
134
|
Jin Q, Huang F, Sun S, Zhou Y, Xu X, Xi W. An improved method on isolation and serial passage of Chlamydia pneumoniae from human peripheral blood mononuclear cells. J Clin Lab Anal 2013; 27:471-6. [PMID: 24218129 PMCID: PMC6807342 DOI: 10.1002/jcla.21629] [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: 03/26/2013] [Accepted: 04/30/2013] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND Conventional method for Chlamydia pneumoniae (Cpn) isolation and propagation is technically challenging and time-consuming. Here, we developed a method to improve the isolation and passage of Cpn collected from human peripheral blood mononuclear cells (PBMCs). METHODS PBMCs positive with Cpn antigen (Cpn-Ag) were isolated, then centrifuged and cultured with Hep-2 cells after being broken. Cells were broken again and put into new Hep-2 cells to finish totally four passages with isolated and imported Cpn. Microimmunofluorescence method was used to detect Cpn. Inclusion forming unit (IFU) number was counted for each passage. Polymerase chain reaction (PCR) method was used to detect Cpn DNA. Efficiency of different centrifugation modes was compared. RESULTS Hep-2 cells of the first and second passages were strong positive with Cpn-Ag, the third passage was positive, and the fourth negative. Degeneration appeared in the fourth passage for isolated Cpn and third passage for imported strain. Centrifugation mode of 1,000 rpm for 2 h was the most efficient for Cpn propagation and passage. CONCLUSION This simplified method achieved efficient isolation, propagation, and passage of Cpn from PBMCs, and isolated strain was superior to imported strain on propagating ability.
Collapse
Affiliation(s)
- Qian Jin
- Department of Respiratory MedicineTongde Hospital of Zhejiang ProvinceHangzhouZhejiangChina
| | - Feihua Huang
- Department of Respiratory MedicineTongde Hospital of Zhejiang ProvinceHangzhouZhejiangChina
| | - Shuming Sun
- Department of Respiratory MedicineJinshan HospitalFudan UniversityShanghaiChina
| | - Ying Zhou
- Department of Respiratory MedicineTongde Hospital of Zhejiang ProvinceHangzhouZhejiangChina
| | - Xianrong Xu
- Department of Respiratory MedicineTongde Hospital of Zhejiang ProvinceHangzhouZhejiangChina
| | - Weixing Xi
- Department of Clinical LaboratoryTongde Hospital of Zhejiang ProvinceHangzhou, ZhejiangChina
| |
Collapse
|
135
|
Anti-mouse CD52 monoclonal antibody ameliorates iron-deficient anaemia in IL-10 knockout mice. Br J Nutr 2013; 111:987-95. [DOI: 10.1017/s0007114513003413] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Approximately 50 % of patients with inflammatory bowel disease (IBD) suffer from anaemia, with Fe deficiency being the most common cause. CD52 monoclonal antibody (mAb) targets the cell surface CD52 and is effective in depleting lymphocytes through cytolytic effects in vivo. The aim of the present study was to investigate the therapeutic effect of anti-mouse CD52 mAb on Fe-deficient anaemia in IBD. IL-10 knockout mice (IL-10− / −) of 12 weeks with established colitis were treated with anti-mouse CD52 mAb once per week for 2 weeks. Severity of colitis, blood T lymphocytes, blood Hb, haematocrit, plasma erythropoietin (EPO), serum Fe concentration, transferrin saturation, splenic Fe stores, expression of liver hepcidin mRNA, Western blotting of the phosphorylated form of Smad1/5/8 and total Smad1 were measured at the end of the experiment. IL-10− / − mice treated with CD52 mAb showed a reduction in the percentage of CD4+ and CD4+CD45+ T cells in blood and weight loss typically associated with colonic inflammation, serum levels of EPO, the expression of liver hepcidin mRNA and total Smad1 protein, while they showed an increase in Hb concentrations, haematocrit, levels of serum Fe, transferrin saturation and splenic Fe stores. The present results indicated that anti-CD52 therapy may ameliorate Fe-deficient anaemia by reducing colonic inflammation. These findings may open novel horizons in the treatment of patients with IBD by resetting of immunological homeostasis in the gut by depleting the activated T cells in the gut mucosa.
Collapse
|
136
|
Marco MRL, Dons EM, van der Windt DJ, Bhama JK, Lu LT, Zahorchak AF, Lakkis FG, Cooper DKC, Ezzelarab MB, Thomson AW. Post-transplant repopulation of naïve and memory T cells in blood and lymphoid tissue after alemtuzumab-mediated depletion in heart-transplanted cynomolgus monkeys. Transpl Immunol 2013; 29:88-98. [PMID: 24120957 DOI: 10.1016/j.trim.2013.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 10/02/2013] [Accepted: 10/02/2013] [Indexed: 11/19/2022]
Abstract
Repopulation of memory T cells (Tmem) in allograft recipients after lymphodepletion is a major barrier to transplant tolerance induction. Ineffective depletion of naïve T cells (Tn) and Tmem may predispose to repopulation of Tmem after transplantation. Cynomolgus macaque monkeys given heart allografts were lymphodepleted using Alemtuzumab (Campath-1H; anti-CD52). Peripheral blood (PB) and lymph nodes (LN) were analyzed for CD95(-) (Tn) and CD95(+) cells (Tmem), one day, one month and up to three months after Alemtuzumab infusion. CD52 expression, susceptibility to Alemtuzumab cytotoxicity and pro-apoptotic caspase-3 were evaluated in Tn and Tmem. In vivo, Alemtuzumab induction profoundly depleted lymphocytes in PB (99% reduction) but exerted a lesser effect in LN (70% reduction), with similar depletion of Tn and Tmem subsets. After transplantation, Tmem comprised the majority of lymphocytes in PB and LN. In vitro, LN T cells were more resistant to Alemtuzumab-mediated cytotoxicity than PB lymphocytes. CD4(+) Tn and Tmem were equally susceptible to Alemtuzumab-mediated cytotoxicity, whereas CD8(+) Tn were more resistant than CD8(+) Tmem. However, no significant differences in CD52 expression between lymphocyte subsets in PB and LN were observed. Caspase-3 expression was higher in PB than LN T cells. CD4(+) and CD8(+) Tn expressed lower levels of Caspase-3 than Tmem, in both PB and LN. Thus, after Alemtuzumab infusion, residual Tn in secondary lymphoid tissue may predispose to rapid recovery of Tmem in allograft recipients.
Collapse
Affiliation(s)
- M R L Marco
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Freedman MS, Kaplan JM, Markovic-Plese S. Insights into the Mechanisms of the Therapeutic Efficacy of Alemtuzumab in Multiple Sclerosis. JOURNAL OF CLINICAL & CELLULAR IMMUNOLOGY 2013; 4:1000152. [PMID: 24363961 PMCID: PMC3869402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The pathogenesis of multiple sclerosis (MS) is thought to involve peripheral activation of immune cells against central nervous system (CNS) antigens and their migration across the blood-brain barrier, leading to CNS inflammation and neurodegeneration. Alemtuzumab, a humanized anti-CD52 monoclonal antibody that rapidly depletes CD52-expressing cells from the circulation, is being investigated as a new treatment option in relapsing-remitting MS (RRMS). Clinical and radiologic results indicate robust suppression of inflammation related to the depletion of T and B lymphocytes during each treatment course of alemtuzumab. Furthermore, several lines of evidence suggest that the long-term clinical effects of alemtuzumab are attributable to qualitative changes in repopulating lymphocyte subsets potentially leading to a rebalancing of the immune system. Here, we review the contribution of data from animal models, ex vivo human studies, and clinical trials to the understanding of the mechanisms underlying the therapeutic effect of alemtuzumab in patients with RRMS.
Collapse
Affiliation(s)
- Mark S Freedman
- University of Ottawa, Multiple Sclerosis Research Clinic, the Ottawa Hospital-General Campus, Ottawa, Ontario K1H 8L6, Canada
| | | | - Silva Markovic-Plese
- Department of Neurology, Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, NC 27599-7025, USA
| |
Collapse
|
138
|
Turner MJ, Lamorte MJ, Chretien N, Havari E, Roberts BL, Kaplan JM, Siders WM. Immune status following alemtuzumab treatment in human CD52 transgenic mice. J Neuroimmunol 2013; 261:29-36. [PMID: 23759318 DOI: 10.1016/j.jneuroim.2013.04.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 04/16/2013] [Accepted: 04/22/2013] [Indexed: 10/26/2022]
Abstract
Alemtuzumab is a monoclonal antibody against the CD52 antigen present at high levels on the surface of lymphocytes. While treatment of multiple sclerosis patients with alemtuzumab results in marked depletion of lymphocytes from the circulation, it has not been associated with a high incidence of serious infections. In a human CD52 transgenic mouse, alemtuzumab treatment showed minimal impact on the number and function of innate immune cells. A transient decrease in primary adaptive immune responses was observed post-alemtuzumab but there was little effect on memory responses. These results potentially help explain the level of immunocompetence observed in alemtuzumab-treated MS patients.
Collapse
Affiliation(s)
- Michael J Turner
- Neuroimmunology Research, Genzyme, a Sanofi Company, 49 New York Avenue, Framingham, MA 01701, United States.
| | | | | | | | | | | | | |
Collapse
|
139
|
Kousin-Ezewu O, Coles A. Alemtuzumab in multiple sclerosis: latest evidence and clinical prospects. Ther Adv Chronic Dis 2013; 4:97-103. [PMID: 23634277 DOI: 10.1177/2040622313479137] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Alemtuzumab was first used in multiple sclerosis in 1991. It is a monoclonal antibody which is directed against CD52, a protein of unknown function on lymphocytes. Alemtuzumab causes a lymphopenia, following which homeostatic reconstitution leads to prolonged alteration of the immune repertoire. This reduces the risk of relapse and disability accumulation in multiple sclerosis; it is the only drug to show superiority over interferon β-1a in disability outcomes in a monotherapy phase III trial. It should be used with a parallel risk management programme to identify the principal adverse effects of alemtuzumab, especially secondary autoimmunity months or years later, mainly against the thyroid but also immune thrombocytopenia. This review charts the development of alemtuzumab as a drug for multiple sclerosis and summarizes the latest clinical trial data.
Collapse
Affiliation(s)
- Onajite Kousin-Ezewu
- Department of Clinical Neurosciences, University of Cambridge, Level 6, Block A, Box 165, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | | |
Collapse
|
140
|
Robak T. Emerging monoclonal antibodies and related agents for the treatment of chronic lymphocytic leukemia. Future Oncol 2013; 9:69-91. [DOI: 10.2217/fon.12.157] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Monoclonal antibodies (mAbs) – rituximab, ofatumumab and alemtuzumab – have been approved for use in the therapy of chronic lymphocytic leukemia (CLL). Recently, a new generation of anti-CD20 mAbs has become available for preclinical studies and clinical trials. These antibodies were engineered to have augmented antitumor activity by increasing complement-dependent cytotoxicity, antibody-dependent cellular cytotoxicity and Fc-binding affinity for the low-affinity variants of the Fcγ receptor IIIa. The most promising mAb directed against CD20 is obinutuzumab (GA-101). mAbs directed against CD22, CD37 and CD40 have also shown some activity in CLL. In addition, small modular immunopharmaceuticals – TRU-015 (anti-CD20) and TRU-016 (anti-CD37) – that retain Fc-mediated effector functions have been developed and investigated in preclinical studies and clinical trials. Antibody–drug conjugates and recombinant immunotoxins are also being evaluated in lymphoid malignancies. Further studies will elucidate the role of these agents in the treatment of CLL.
Collapse
Affiliation(s)
- Tadeusz Robak
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Ul. Ciolkowskiego 2, Poland and Copernicus Memorial Hospital, 93-510 Lodz, Ul. Ciolkowskiego 2, Poland
| |
Collapse
|