1
|
Cuesta-Mateos C, Fuentes P, Schrader A, Juárez-Sánchez R, Loscertales J, Mateu-Albero T, Vega-Piris L, Espartero-Santos M, Marcos-Jimenez A, Sánchez-López BA, Pérez-García Y, Jungherz D, Oberbeck S, Wahnschaffe L, Kreutzman A, Andersson EI, Mustjoki S, Faber E, Urzainqui A, Fresno M, Stamatakis K, Alfranca A, Terrón F, Herling M, Toribio ML, Muñoz-Calleja C. CCR7 as a novel therapeutic target in t-cell PROLYMPHOCYTIC leukemia. Biomark Res 2020; 8:54. [PMID: 33110606 PMCID: PMC7585232 DOI: 10.1186/s40364-020-00234-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 10/12/2020] [Indexed: 12/20/2022] Open
Abstract
T-cell prolymphocytic leukemia (T-PLL) is a poor prognostic disease with very limited options of efficient therapies. Most patients are refractory to chemotherapies and despite high response rates after alemtuzumab, virtually all patients relapse. Therefore, there is an unmet medical need for novel therapies in T-PLL. As the chemokine receptor CCR7 is a molecule expressed in a wide range of malignancies and relevant in many tumor processes, the present study addressed the biologic role of this receptor in T-PLL. Furthermore, we elucidated the mechanisms of action mediated by an anti-CCR7 monoclonal antibody (mAb) and evaluated whether its anti-tumor activity would warrant development towards clinical applications in T-PLL. Our results demonstrate that CCR7 is a prognostic biomarker for overall survival in T-PLL patients and a functional receptor involved in the migration, invasion, and survival of leukemic cells. Targeting CCR7 with a mAb inhibited ligand-mediated signaling pathways and induced tumor cell killing in primary samples. In addition, directing antibodies against CCR7 was highly effective in T-cell leukemia xenograft models. Together, these findings make CCR7 an attractive molecule for novel mAb-based therapeutic applications in T-PLL, a disease where recent drug screen efforts and studies addressing new compounds have focused on chemotherapy or small molecules.
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, C/ Diego de León 62, 28006 Madrid, Spain.,IMMED S.L., Immunological and Medicinal Products, Madrid, Spain
| | - Patricia Fuentes
- Immune System Development and Function Unit, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
| | - Alexandra Schrader
- Department I of Internal Medicine, Center for Integrated Oncology (CIO) Aachen-Bonn-Cologne-Duesseldorf (ABCD), Cologne Cluster of Excellence in Cellular Stress Response and Aging-Associated Diseases (CECAD), and Center of Molecular Medicine Cologne (CMMC), The University of Cologne, Cologne, Germany
| | - Raquel Juárez-Sánchez
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, C/ Diego de León 62, 28006 Madrid, Spain.,IMMED S.L., Immunological and Medicinal Products, Madrid, Spain
| | - Javier Loscertales
- Hematology Department, Hospital Universitario de La Princesa, IIS-IP, Madrid, Spain
| | - Tamara Mateu-Albero
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, C/ Diego de León 62, 28006 Madrid, Spain
| | - Lorena Vega-Piris
- Methodology Unit, Hospital Universitario de La Princesa, IIS-IP, Madrid, Spain
| | - Marina Espartero-Santos
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, C/ Diego de León 62, 28006 Madrid, Spain
| | - Ana Marcos-Jimenez
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, C/ Diego de León 62, 28006 Madrid, Spain
| | - Blanca Andrea Sánchez-López
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, C/ Diego de León 62, 28006 Madrid, Spain
| | - Yaiza Pérez-García
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, C/ Diego de León 62, 28006 Madrid, Spain
| | - Dennis Jungherz
- Department I of Internal Medicine, Center for Integrated Oncology (CIO) Aachen-Bonn-Cologne-Duesseldorf (ABCD), Cologne Cluster of Excellence in Cellular Stress Response and Aging-Associated Diseases (CECAD), and Center of Molecular Medicine Cologne (CMMC), The University of Cologne, Cologne, Germany
| | - Sebastian Oberbeck
- Department I of Internal Medicine, Center for Integrated Oncology (CIO) Aachen-Bonn-Cologne-Duesseldorf (ABCD), Cologne Cluster of Excellence in Cellular Stress Response and Aging-Associated Diseases (CECAD), and Center of Molecular Medicine Cologne (CMMC), The University of Cologne, Cologne, Germany
| | - Linus Wahnschaffe
- Department I of Internal Medicine, Center for Integrated Oncology (CIO) Aachen-Bonn-Cologne-Duesseldorf (ABCD), Cologne Cluster of Excellence in Cellular Stress Response and Aging-Associated Diseases (CECAD), and Center of Molecular Medicine Cologne (CMMC), The University of Cologne, Cologne, Germany
| | - Anna Kreutzman
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, C/ Diego de León 62, 28006 Madrid, Spain
| | - Emma I Andersson
- Department of Hematology, Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Satu Mustjoki
- Department of Hematology, Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Translational Immunology Research Program and Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| | - Edgar Faber
- Department of Hemato-Oncology, Faculty Hospital Olomouc, Faculty of Medicine and Dentistry Palacky University, Olomouc, Czech Republic
| | - Ana Urzainqui
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, C/ Diego de León 62, 28006 Madrid, Spain
| | - Manuel Fresno
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
| | - Kostantino Stamatakis
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
| | - Arantzazu Alfranca
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, C/ Diego de León 62, 28006 Madrid, Spain
| | - Fernando Terrón
- IMMED S.L., Immunological and Medicinal Products, Madrid, Spain
| | - Marco Herling
- Department I of Internal Medicine, Center for Integrated Oncology (CIO) Aachen-Bonn-Cologne-Duesseldorf (ABCD), Cologne Cluster of Excellence in Cellular Stress Response and Aging-Associated Diseases (CECAD), and Center of Molecular Medicine Cologne (CMMC), The University of Cologne, Cologne, Germany
| | - María Luisa Toribio
- Immune System Development and Function Unit, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, C/ Diego de León 62, 28006 Madrid, Spain.,Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
2
|
Johansson P, Klein-Hitpass L, Röth A, Möllmann M, Reinhardt HC, Dührsen U, Dürig J. Mutations in PIGA cause a CD52-/GPI-anchor-deficient phenotype complicating alemtuzumab treatment in T-cell prolymphocytic leukemia. Eur J Haematol 2020; 105:786-796. [PMID: 32875608 DOI: 10.1111/ejh.13511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Infusional alemtuzumab followed by consolidating allogeneic hematopoietic stem cell transplantation in eligible patients is considered a standard of care in T-cell prolymphocytic leukemia (T-PLL). Antibody selection against CD52 has been associated with the development of CD52-negative leukemic T cells at time of relapse. Clinical implications and molecular mechanisms underlying this phenotypic switch are unknown. METHODS We performed flow cytometry and real-time-PCR for CD52-expression and next generation sequencing for PIGA mutational analyses. RESULTS We identified loss of CD52 expression after alemtuzumab treatment in two of 21 T-PLL patients resulting from loss of GPI-anchor expression caused by inactivating mutations of the PIGA gene. One patient with relapsed T-PLL exhibited a single PIGA mutation, causing a CD52-negative escape variant of the initial leukemic cell clone, preventing alemtuzumab-retreatment. The second patient with continued complete remission after alemtuzumab treatment harbored three different PIGA mutations that affected either the non-neoplastic T cell or the mononuclear cell compartment and resulted in symptomatic paroxysmal nocturnal hemoglobinuria. Next generation sequencing of T-PLL cells collected before the initiation of treatment revealed PIGA wild-type sequence reads in all 16 patients with samples available for testing. CONCLUSION These data indicate that PIGA mutations were acquired during or after completion of alemtuzumab treatment.
Collapse
Affiliation(s)
- Patricia Johansson
- Department of Hematology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ludger Klein-Hitpass
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Alexander Röth
- Department of Hematology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael Möllmann
- Department of Hematology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hans Christian Reinhardt
- Department of Hematology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Ulrich Dührsen
- Department of Hematology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jan Dürig
- Department of Hematology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,Department of General Internal Medicine, St. Josef-Krankenhaus, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
3
|
Immunophenotype Expression Change From CD52+ to CD52- on Erythrodermic Peripheral T-cell Lymphoma, Not Otherwise Specified After Treatment With Alemtuzumab. Am J Dermatopathol 2017; 40:547-550. [PMID: 28937435 DOI: 10.1097/dad.0000000000001000] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
4
|
Abstract
T-cell depletion strategies are an efficient therapy for the treatment of acute rejection after organ transplantation and have been successfully used as induction regimens. Although eliminating whole T cells blocks alloreactivity, this therapy challenges the development of regulatory mechanisms because it depletes regulatory cells and modifies the profile of T cells after homeostatic repopulation. Targeting T-cell subpopulations or selectively activated T cells, without modifying Treg cells, could constitute a pro-tolerogenic approach. However, the perfect molecular target that would be totally specific probably still needs to be identified. In this study, we have reviewed the biological activities of broad or specific T-cell depletion strategies as these contribute to the induction of regulatory cells and tolerance in organ transplantation.
Collapse
Affiliation(s)
- Thomas Haudebourg
- INSERM, U643, CHU Nantes, Institut de Transplantation et de Recherche en Transplantation, ITERT, Université de Nantes, Faculté de Médecine, Nantes, France
| | | | | |
Collapse
|
5
|
Davis BH, Holden JT, Bene MC, Borowitz MJ, Braylan RC, Cornfield D, Gorczyca W, Lee R, Maiese R, Orfao A, Wells D, Wood BL, Stetler-Stevenson M. 2006 Bethesda International Consensus recommendations on the flow cytometric immunophenotypic analysis of hematolymphoid neoplasia: medical indications. CYTOMETRY PART B-CLINICAL CYTOMETRY 2008; 72 Suppl 1:S5-13. [PMID: 17803188 DOI: 10.1002/cyto.b.20365] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The clinical indications for diagnostic flow cytometry studies are an evolving consensus, as the knowledge of antigenic definition of hematolymphoid malignancies and the prognostic significance of antigen expression evolves. Additionally the standard of care is not routinely communicated to practicing clinicians and diagnostic services, especially as may relate to new technologies. Accordingly there is often uncertainty on the part of clinicians, payers of medical services, diagnostic physicians and scientists as to the appropriate use of diagnostic flow cytometry. In an attempt to communicate contemporary diagnostic utility of immunophenotypic flow cytometry in the diagnosis and follow-up of patients with hematolymphoid malignancies, the Clinical Cytometry Society organized a two day meeting of international experts in this area to reach a consensus as to this diagnostic tool. This report summarizes the appropriate use of diagnostic flow cytometry as determined by unanimous approval of these experienced practitioners.
Collapse
Affiliation(s)
- B H Davis
- Trillium Diagnostics, Brewer, Maine, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Ortonne N, Huet D, Gaudez C, Marie-Cardine A, Schiavon V, Bagot M, Musette P, Bensussan A. Significance of circulating T-cell clones in Sezary syndrome. Blood 2006; 107:4030-8. [PMID: 16418328 DOI: 10.1182/blood-2005-10-4239] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Identification of malignant Sézary cells by T-cell receptor (TCR) clonality studies is routinely used for the diagnosis of Sézary syndrome, but T-cell clones expressed in a single patient have never been accurately characterized. We previously reported that CD158k expression delineates Sézary syndrome malignant cells, and, more recently, we identified vimentin at the surface membranes of Sézary cells and normal activated lymphocytes. In the present study, T-cell clones from 13 patients with Sézary syndrome were identified by immunoscopy and further characterized in the blood according to their TCR Vbeta, CD158k, and vimentin cell-surface expression. We found in most patients a unique malignant T-cell clone that coexpressed CD158k and vimentin and that, when patients were tested, was also present in the skin. However, in some patients we detected the presence of a nonmalignant circulating clone expressing high amounts of vimentin and lacking CD158k. These results indicate that clonal expansion may originate from circulating malignant and nonmalignant CD4(+) T cell populations in patients with Sézary syndrome. Identification of the malignant cells in Sézary syndrome cannot be achieved by T-cell clonality studies or by TCR Vbeta monoclonal antibody (mAb) analysis alone; it also relies on CD158k phenotyping.
Collapse
Affiliation(s)
- Nicolas Ortonne
- INSERM U659, Faculté de Médecine de Créteil, 8 rue du Général Sarrail, F-94010 Créteil Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Garland RJ, Groves SJ, Diamanti P, West SE, Winship KL, Virgo PF, Robinson SP, Oakhill A, Cornish JM, Pamphilon DH, Marks DI, Goulden NJ, Steward CG. Early emergence of PNH-like T cells after allogeneic stem cell transplants utilising CAMPATH-1H for T cell depletion. Bone Marrow Transplant 2005; 36:237-44. [PMID: 15968291 DOI: 10.1038/sj.bmt.1705049] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
CAMPATH-1H (C-1H) is widely used in vivo and / or in vitro for T cell depletion in hematopoietic SCT. This humanised monoclonal antibody is specific for CD52, a marker coexpressed on the majority of human lymphocytes with CD48 and other glycosylphosphatidyl-inositol (GPI) anchored proteins. We detected CD52 / CD48 dual expression on >99% of CD3(+) lymphocytes from normal individuals and all 15 post-SCT patients whose transplants did not utilise C-1H. By contrast, 23 / 26 patients with transplants involving C-1H (in vivo, in vitro or both) exhibited populations lacking CD52 expression that accounted for 49.7% (4.2-86.2%) of the CD3+ lymphocytes (median and range) in samples evaluated at a median of 2 months post-SCT. Most CD52- cells also lacked CD48 expression. These GPI- T cells were of either donor or mixed donor / recipient origin. They were predominant in the early months after SCT at times of profound lymphopenia and inversely correlated with the recovery of the absolute lymphocyte count (r= - 0.663, P<0.0001). The presence of CD52- cells has been correlated previously with clinical outcome after CAMPATH therapy for both malignant and nonmalignant diseases.
Collapse
MESH Headings
- Adolescent
- Adult
- Alemtuzumab
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal, Humanized
- Antibodies, Neoplasm/chemistry
- Antigens, CD/biosynthesis
- Antigens, CD/chemistry
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/chemistry
- Antineoplastic Agents/pharmacology
- CD3 Complex/biosynthesis
- CD48 Antigen
- CD52 Antigen
- Cell Separation
- Child
- Child, Preschool
- Cohort Studies
- Female
- Flow Cytometry
- Glycoproteins/biosynthesis
- Glycoproteins/chemistry
- Glycosylphosphatidylinositols/metabolism
- Hemoglobinuria, Paroxysmal/metabolism
- Humans
- Immunomagnetic Separation
- Male
- Middle Aged
- Stem Cell Transplantation
- T-Lymphocytes/cytology
- T-Lymphocytes/metabolism
- Time Factors
- Transplantation Chimera
- Transplantation, Homologous/methods
- Treatment Outcome
Collapse
Affiliation(s)
- R J Garland
- Department of Pathology and Microbiology, University of Bristol, University Walk, Bristol, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Bremer E, Samplonius D, Kroesen BJ, van Genne L, de Leij L, Helfrich W. Exceptionally potent anti-tumor bystander activity of an scFv:sTRAIL fusion protein with specificity for EGP2 toward target antigen-negative tumor cells. Neoplasia 2005; 6:636-45. [PMID: 15548373 PMCID: PMC1531668 DOI: 10.1593/neo.04229] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Previously, we reported on the target cell-restricted fratricide apoptotic activity of scFvC54:sTRAIL, a fusion protein comprising human-soluble tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) genetically linked to the antibody fragment scFvC54 specific for the cell surface target antigen EGP2. In the present study, we report that the selective binding of scFvC54:sTRAIL to EGP2-positive target cells conveys an exceptionally potent pro-apoptotic effect toward neighboring tumor cells that are devoid of EGP2 expression (bystander cells). The anti-tumor bystander activity of scFvC54:sTRAIL was detectable at target-to-bystander cell ratios as low as 1:100. Treatment in the presence of EGP2-blocking or TRAIL-neutralizing antibody strongly inhibited apoptosis in both target and bystander tumor cells. In the absence of target cells, bystander cell apoptosis induction was abrogated. The bystander apoptosis activity of scFvC54:sTRAIL did not require internalization, enzymatic conversion, diffusion, or communication (gap junctional intracellular communication) between target and bystander cells. Furthermore, scFvC54:sTRAIL showed no detectable signs of innocent bystander activity toward freshly isolated blood cells. Further development of this new principle is warranted for approaches where cancer cells can escape from antibody-based therapy due to partial loss of target antigen expression.
Collapse
Affiliation(s)
- Edwin Bremer
- Groningen University Institute for Drug Exploration (GUIDE), Laboratory for Tumor Immunology, Section Medical Biology, Department of Pathology and Laboratory Medicine, University Hospital Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| | | | | | | | | | | |
Collapse
|
9
|
Bremer E, Samplonius DF, Peipp M, van Genne L, Kroesen BJ, Fey GH, Gramatzki M, de Leij LFMH, Helfrich W. Target Cell–Restricted Apoptosis Induction of Acute Leukemic T Cells by a Recombinant Tumor Necrosis Factor–Related Apoptosis-Inducing Ligand Fusion Protein with Specificity for Human CD7. Cancer Res 2005; 65:3380-8. [PMID: 15833872 DOI: 10.1158/0008-5472.can-04-2756] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Current treatment of human T-cell leukemia and lymphoma is predominantly limited to conventional cytotoxic therapy and is associated with limited therapeutic response and significant morbidity. Therefore, more potent and leukemia-specific therapies with favorable toxicity profiles are urgently needed. Here, we report on the construction of a novel therapeutic fusion protein, scFvCD7:sTRAIL, designed to induce target antigen-restricted apoptosis in human T-cell tumors. ScFvCD7:sTRAIL consists of the death-inducing tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) genetically linked to an scFv antibody fragment specific for the T-cell surface antigen CD7. Treatment with scFvCD7:sTRAIL induced potent CD7-restricted apoptosis in a series of malignant T-cell lines, whereas normal resting leukocytes, activated T cells, and vascular endothelial cells (human umbilical vein endothelial cells) showed no detectable apoptosis. The apoptosis-inducing activity of scFvCD7:sTRAIL was stronger than that of the immunotoxin scFvCD7:ETA. In mixed culture experiments with CD7-positive and CD7-negative tumor cells, scFvCD7:sTRAIL induced very potent bystander apoptosis of CD7-negative tumor cells. In vitro treatment of blood cells freshly derived from T-acute lymphoblastic leukemia patients resulted in marked apoptosis of the malignant T cells that was strongly augmented by vincristin. In conclusion, scFvCD7:sTRAIL is a novel recombinant protein causing restricted apoptosis in human leukemic T cells with low toxicity for normal human blood and endothelial cells.
Collapse
Affiliation(s)
- Edwin Bremer
- Laboratory for Tumor Immunology, Department of Pathology and Laboratory Medicine, Section Medical Biology, University Hospital Groningen, Groningen University Institute for Drug Exploration, 9713 GZ Groningen, the Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Lundin J, Osterborg A. Advances in the use of monoclonal antibodies in the therapy of chronic lymphocytic leukemia. Semin Hematol 2004; 41:234-45. [PMID: 15269883 DOI: 10.1053/j.seminhematol.2004.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Monoclonal antibody (moAb)-based therapies are evolving as an integrated component in the treatment of chronic lymphocytic leukemia (CLL). Advantages such as different mechanisms of action (compared with those of chemotherapy), no or minimal stem cell toxicity, as well as the absence of hair loss and delayed nausea may result in a rapidly increasing usage of these agents in different phases of the disease. The combination of moAbs with chemotheraputic agents has shown promising results in early studies as well as their role in the eradication of minimal residual disease (MRD). The availability of an increasing number of new moAbs together with a better understanding of their effector function will hopefully lead to improved therapeutic outcomes for patients with CLL and related disorders.
Collapse
Affiliation(s)
- Jeanette Lundin
- Departments of Hematology/Oncology, Karolinska University Hospital, Stockholm, Sweden.
| | | |
Collapse
|
11
|
Abstract
Alemtuzumab is an unconjugated, humanised, monoclonal antibody directed against the cell surface antigen CD52 on lymphocytes and monocytes. In noncomparative phase I/II studies in patients with B-cell chronic lymphocytic leukaemia (B-CLL) relapsed after or refractory to alkylating agents and fludarabine, intravenous (IV) administration of alemtuzumab 30 mg/day three times weekly for up to 12 weeks was associated with overall objective response (OR) rates of 21-59%. Combining alemtuzumab with fludarabine resulted in ORs >80%. In noncomparative studies in patients with previously untreated B-CLL, subcutaneous (SC) administration of alemtuzumab alone, or IV in combination with fludarabine, was highly effective, achieving OR rates of around 90%. IV alemtuzumab was active in patients with chemotherapy-resistant/relapsed T-cell prolymphocytic leukaemia, with reported OR rates of 24-76%. Alemtuzumab has been incorporated in novel conditioning regimens designed to facilitate stem cell transplantation in haematological malignancies. Adverse events with alemtuzumab are predictable and manageable. 'First-dose' flulike symptoms, frequently seen after IV infusion, can be managed by (pre)medication and minimised by dose escalation (or SC injection). Anti-infective prophylaxis is mandatory. Cytopenias are transient, although red blood cell and platelet support may be required.
Collapse
MESH Headings
- Alemtuzumab
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antibodies, Neoplasm/adverse effects
- Antibodies, Neoplasm/therapeutic use
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/therapeutic use
- Bone Marrow Transplantation
- Half-Life
- Humans
- Infusions, Intravenous
- Leukemia, B-Cell/classification
- Leukemia, B-Cell/drug therapy
- Leukemia, B-Cell/therapy
- Metabolic Clearance Rate
Collapse
|
12
|
Abstract
Treatment of malignant non-Hodgkin lymphomas (NHL) in the 21st century has been revolutionized by novel biological agents offering targeted approaches in addition to radio-chemotherapy. Monoclonal antibodies (MoAbs) against lymphatic surface antigens have been effective as monotherapeutic agents, and have already shown their superiority to conventional strategies when combined with chemotherapy. Radioimmunoconjugates are more effective than unlabelled antibodies. Specific inhibitors of neoangiogenesis as well as intracellular signal transduction pathways and antiapoptotic mechanisms have shown their efficacy in phase II studies. Long-term improvement in the setting of minimal residual disease has been observed after vaccination against surface antigens and non-myeloablative allogeneic stem cell transplantation is effective in controlling lymphoma growth. Novel specific antigens are currently identified using expression profiling of lymphomas. In the near future, combinations of biological agents will challenge conventional therapy. These exciting new strategies will improve the success rate in aggressive NHLs and may even challenge the paradigm of incurability of indolent lymphomas.
Collapse
MESH Headings
- Alemtuzumab
- Angiogenesis Inhibitors/therapeutic use
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antibodies, Monoclonal, Murine-Derived
- Antibodies, Neoplasm/therapeutic use
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Boronic Acids/therapeutic use
- Bortezomib
- Cancer Vaccines/administration & dosage
- Clinical Trials as Topic
- Clinical Trials, Phase I as Topic
- Clinical Trials, Phase II as Topic
- Clinical Trials, Phase III as Topic
- Cyclophosphamide/therapeutic use
- Disease-Free Survival
- Doxorubicin/therapeutic use
- Forecasting
- Genetic Therapy
- Humans
- Immunoconjugates
- Immunotherapy
- Lymphoma, B-Cell/drug therapy
- Lymphoma, Non-Hodgkin/drug therapy
- Lymphoma, Non-Hodgkin/mortality
- Lymphoma, Non-Hodgkin/radiotherapy
- Lymphoma, Non-Hodgkin/therapy
- Prednisone/therapeutic use
- Protease Inhibitors/therapeutic use
- Proteomics
- Pyrazines/therapeutic use
- Radioimmunotherapy
- Randomized Controlled Trials as Topic
- Rituximab
- Signal Transduction/drug effects
- Survival Analysis
- Thalidomide/therapeutic use
- Time Factors
- Vincristine/therapeutic use
Collapse
Affiliation(s)
- Ulrich Jäger
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, University of Vienna, Vienna, Austria.
| |
Collapse
|
13
|
Abstract
Monoclonal antibodies (MoAbs) are increasingly used in the treatment of patients with hematological malignancies and autoimmune diseases. The most commonly employed humanized and chimeric MoAbs are rituximab, alemtuzumab (Campath-1H, Ilex Pharmaceuticals, San Antonio, TX), and gemtuzumab-ozogamicin (Mylotarg, Wyeth-Ayerst Laboratories, St Davids, PA). The mechanism of action of these antibodies, and host and cellular factors influencing the response, are not completely known. Induction of apoptosis, antibody-dependent cell cytotoxicity (ADCC), and complement-mediated cell death (CDC) is the proposed mechanism of action of these antibodies. We review the current understanding of the mechanism of action of and resistance to these MoAbs.
Collapse
Affiliation(s)
- Neus Villamor
- Unitat d'Hematopatologia, Hospital Clínic, Barcelona, Spain
| | | | | |
Collapse
|
14
|
D'Arena G, Vigliotti ML, Matera R, Musto C, Iodice G, Tartarone A, Di Renzo N. Quantitative evaluation of CD52 expression in B-cell chronic lymphocytic leukemia. Leuk Lymphoma 2003; 44:1255-7. [PMID: 12916884 DOI: 10.1080/1042819031000076981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal
- Antigens, CD/metabolism
- Antigens, Neoplasm/metabolism
- B-Lymphocytes/metabolism
- CD52 Antigen
- Case-Control Studies
- Female
- Glycoproteins/metabolism
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Middle Aged
- Prognosis
- T-Lymphocytes/metabolism
Collapse
|
15
|
|