1
|
Ladikou EE, Sharp K, Simoes FA, Jones JR, Burley T, Stott L, Vareli A, Kennedy E, Vause S, Chevassut T, Devi A, Ashworth I, Ross DM, Hartmann TN, Mitchell SA, Pepper CJ, Best G, Pepper AGS. A Novel In Vitro Model of the Bone Marrow Microenvironment in Acute Myeloid Leukemia Identifies CD44 and Focal Adhesion Kinase as Therapeutic Targets to Reverse Cell Adhesion-Mediated Drug Resistance. Cancers (Basel) 2025; 17:135. [PMID: 39796762 PMCID: PMC11719579 DOI: 10.3390/cancers17010135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Acute myeloid leukemia (AML) is an aggressive neoplasm. Although most patients respond to induction therapy, they commonly relapse due to recurrent disease in the bone marrow microenvironment (BMME). So, the disruption of the BMME, releasing tumor cells into the peripheral circulation, has therapeutic potential. METHODS Using both primary donor AML cells and cell lines, we developed an in vitro co-culture model of the AML BMME. We used this model to identify the most effective agent(s) to block AML cell adherence and reverse adhesion-mediated treatment resistance. RESULTS We identified that anti-CD44 treatment significantly increased the efficacy of cytarabine. However, some AML cells remained adhered, and transcriptional analysis identified focal adhesion kinase (FAK) signaling as a contributing factor; the adhered cells showed elevated FAK phosphorylation that was reduced by the FAK inhibitor, defactinib. Importantly, we demonstrated that anti-CD44 and defactinib were highly synergistic at diminishing the adhesion of the most primitive CD34high AML cells in primary autologous co-cultures. CONCLUSIONS Taken together, we identified anti-CD44 and defactinib as a promising therapeutic combination to release AML cells from the chemoprotective AML BMME. As anti-CD44 is already available as a recombinant humanized monoclonal antibody, the combination of this agent with defactinib could be rapidly tested in AML clinical trials.
Collapse
Affiliation(s)
- Eleni E. Ladikou
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Kim Sharp
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Fabio A. Simoes
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - John R. Jones
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Thomas Burley
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Lauren Stott
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Aimilia Vareli
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Emma Kennedy
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Sophie Vause
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Timothy Chevassut
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
- Department of Haematology, Brighton and Sussex University Hospital Trust, Brighton BN2 5BE, UK;
| | - Amarpreet Devi
- Department of Haematology, Brighton and Sussex University Hospital Trust, Brighton BN2 5BE, UK;
| | - Iona Ashworth
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - David M. Ross
- Department of Haematology, Flinders Medical Centre, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (D.M.R.); (G.B.)
| | - Tanja Nicole Hartmann
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79085 Freiburg, Germany;
| | - Simon A. Mitchell
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Chris J. Pepper
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Giles Best
- Department of Haematology, Flinders Medical Centre, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (D.M.R.); (G.B.)
| | - Andrea G. S. Pepper
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| |
Collapse
|
2
|
Ladikou EE, Chevassut T, Pepper CJ, Pepper AG. Dissecting the role of the CXCL12/CXCR4 axis in acute myeloid leukaemia. Br J Haematol 2020; 189:815-825. [PMID: 32135579 DOI: 10.1111/bjh.16456] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023]
Abstract
Acute myeloid leukaemia (AML) is the most common adult acute leukaemia with the lowest survival rate. It is characterised by a build-up of immature myeloid cells anchored in the protective niche of the bone marrow (BM) microenvironment. The CXCL12/CXCR4 axis is central to the pathogenesis of AML as it has fundamental control over AML cell adhesion into the protective BM niche, adaptation to the hypoxic environment, cellular migration and survival. High levels of CXCR4 expression are associated with poor relapse-free and overall survival. The CXCR4 ligand, CXCL12 (SDF-1), is expressed by multiple cells types in the BM, facilitating the adhesion and survival of the malignant clone. Blocking the CXCL12/CXCR4 axis is an attractive therapeutic strategy providing a 'multi-hit' therapy that both prevents essential survival signals and releases the AML cells from the BM into the circulation. Once out of the protective niche of the BM they would be more susceptible to destruction by conventional chemotherapeutic drugs. In this review, we disentangle the diverse roles of the CXCL12/CXCR4 axis in AML. We then describe multiple CXCR4 inhibitors, including small molecules, peptides, or monoclonal antibodies, which have been developed to date and their progress in pre-clinical and clinical trials. Finally, the review leads us to the conclusion that there is a need for further investigation into the development of a 'multi-hit' therapy that targets several signalling pathways related to AML cell adhesion and maintenance in the BM.
Collapse
Affiliation(s)
- Eleni E Ladikou
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK.,Royal Sussex County Hospital, Brighton, UK
| | - Timothy Chevassut
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK.,Royal Sussex County Hospital, Brighton, UK
| | - Chris J Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Andrea Gs Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| |
Collapse
|
3
|
Klammer M, Roddie PH. Current progress in the development of a cell-based vaccine for the immunotherapy of acute myeloid leukemia. Expert Rev Vaccines 2014; 5:211-22. [PMID: 16608421 DOI: 10.1586/14760584.5.2.211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Evidence that immunological control contributes to the elimination of residual leukemia has emerged from allogeneic hematopoietic stem cell transplantation. This review assesses the current understanding of immunobiology of acute myeloid leukemia and how dendritic cells and T cells may be harnessed using in vitro and in vivo priming techniques. Preclinical and clinical dendritic cell vaccine trials reported to date are considered and the prospects for immunotherapy with dendritic cell-based vaccine constructs evaluated.
Collapse
Affiliation(s)
- Matthias Klammer
- Western General Hospital, University of Edinburgh-Leukaemia Research Fund, John Hughes Bennett Laboratory and Department of Haematology, Western General Hospital, Edinburgh, UK.
| | | |
Collapse
|
4
|
Parker JJ, Jones JC, Strober S, Knox SJ. Characterization of direct radiation-induced immune function and molecular signaling changes in an antigen presenting cell line. Clin Immunol 2013; 148:44-55. [PMID: 23649044 DOI: 10.1016/j.clim.2013.03.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 02/06/2013] [Accepted: 03/11/2013] [Indexed: 12/13/2022]
Abstract
Radiation therapy is a widely used cancer treatment and pre-transplantation conditioning regimen that has the potential to influence anti-tumor and post-transplantation immune responses. Although conventionally fractionated radiation doses can suppress immune responses by depleting lymphocytes, single high doses of local tumor radiation can enhance immune responses. Using phospho-flow cytometry analysis of a human monocytic cell line, we identified novel radiation-induced changes in the phosphorylation state of NFκB family members known in other cell types to maintain and regulate immune function. These phosphorylation changes were p53 independent, but were strongly dependent upon ATM activation due to DNA damage. We found that radiation promotes the activation and APC functional maturation through phosphorylation of NFκB Essential Modulator (NEMO). Our results and the analytic methods are especially well suited to the study of functional changes in APC when radiation is used for immune modulation in clinical protocols.
Collapse
Affiliation(s)
- Jennifer J Parker
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | | | | | | |
Collapse
|
5
|
Dolen Y, Esendagli G. Myeloid leukemia cells with a B7-2+subpopulation provoke Th-cell responses and become immuno-suppressive through the modulation of B7 ligands. Eur J Immunol 2013; 43:747-57. [DOI: 10.1002/eji.201242814] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 10/28/2012] [Accepted: 11/19/2012] [Indexed: 12/31/2022]
Affiliation(s)
- Yusuf Dolen
- Department of Basic Oncology; Hacettepe University Cancer Institute; Ankara; Turkey
| | - Gunes Esendagli
- Department of Basic Oncology; Hacettepe University Cancer Institute; Ankara; Turkey
| |
Collapse
|
6
|
Berthon C, Driss V, Liu J, Kuranda K, Leleu X, Jouy N, Hetuin D, Quesnel B. In acute myeloid leukemia, B7-H1 (PD-L1) protection of blasts from cytotoxic T cells is induced by TLR ligands and interferon-gamma and can be reversed using MEK inhibitors. Cancer Immunol Immunother 2010; 59:1839-49. [PMID: 20814675 PMCID: PMC2945474 DOI: 10.1007/s00262-010-0909-y] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Accepted: 08/16/2010] [Indexed: 12/15/2022]
Abstract
B7-H1 (PD-L1) is a B7-related protein that inhibits T-cell responses. B7-H1 participates in the immunoescape of cancer cells and is also involved in the long-term persistence of leukemic cells in a mouse model of leukemia. B7-H1 can be constitutively expressed by cancer cells, but is also induced by various stimuli. Therefore, we examined the constitutive and inducible expression of B7-H1 and the consequences of this expression in human acute myeloid leukemia (AML). We analyzed B7-H1 expression in a cohort of 79 patients with AML. In addition, we studied blast cells after incubation with interferon-gamma or toll-like receptors (TLR) ligands. Finally, we evaluated functionality of cytotoxic T-cell activity against blast cells. Expression of B7-H1 upon diagnosis was high in 18% of patients. Expression of TLR2, 4 and 9 was detected in one-third of AML samples. Expression of TLR2 and TLR4 ligands or IFN-γ induced by B7-H1 was found to protect AML cells from CTL-mediated lysis. Spontaneous B7-H1 expression was also found to be enhanced upon relapse in some patients. MEK inhibitors, including UO126 and AZD6244, reduced B7-H1 expression and restored CTL-mediated lysis of blast cells. In AML, B7-H1 expression by blasts represents a possible immune escape mechanism. The inducibility of B7-H1 expression by IFN-γ or TLR ligands suggests that various stimuli, either produced during the immune response against leukemia cells or released by infectious microorganisms, could protect leukemic cells from T cells. The efficacy of MEK inhibitors against B7-H1-mediated inhibition of CTLs suggests a possible cancer immunotherapy strategy using targeted drugs.
Collapse
Affiliation(s)
- Céline Berthon
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
- Service des Maladies du Sang, Centre Hospitalier et Universitaire de Lille, Rue Polonovski, 59037 Lille, France
| | - Virginie Driss
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
| | - Jizhong Liu
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
| | - Klaudia Kuranda
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
| | - Xavier Leleu
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
- Service des Maladies du Sang, Centre Hospitalier et Universitaire de Lille, Rue Polonovski, 59037 Lille, France
| | - Nathalie Jouy
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
| | - Dominique Hetuin
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
| | - Bruno Quesnel
- INSERM, unit 837, Institut de Recherche sur le Cancer de Lille, Lille, France
- Université Nord de France, Institut Fédératif de Recherche 114, Lille, France
- Service des Maladies du Sang, Centre Hospitalier et Universitaire de Lille, Rue Polonovski, 59037 Lille, France
| |
Collapse
|
7
|
Hardwick N, Chan L, Ingram W, Mufti G, Farzaneh F. Lytic activity against primary AML cells is stimulated in vitro by an autologous whole cell vaccine expressing IL-2 and CD80. Cancer Immunol Immunother 2010; 59:379-88. [PMID: 19711075 PMCID: PMC11030635 DOI: 10.1007/s00262-009-0756-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Accepted: 08/08/2009] [Indexed: 11/30/2022]
Abstract
Despite being of the myeloid lineage, acute myeloid leukaemia (AML) blasts are of low immunogenicity, probably because they lack the costimulatory molecule CD80 and secrete immunosuppressive factors. We have previously shown that in vitro stimulation of autologous peripheral blood mononuclear cells (PBMCs) with primary AML cells modified to express CD80 and IL-2 promotes proliferation, secretion of Th1 cytokines and expansion of activated CD8(+) T cells. In this study, we show that allogeneic effector cells (from a healthy donor or AML patients) when stimulated with IL-2/CD80 modified AML blasts were able to induce the lysis of unmodified AML blasts. Effector cells stimulated with IL-2/CD80AML blasts had higher lytic activity than cells stimulated with AML cells expressing CD80 or IL-2 alone. Similarly, AML patient PBMCs primed with autologous IL-2/CD80 AML cells had a higher frequency of IFN-gamma secreting cells and show cytotoxicity against autologous, unmodified blasts. Crucially, the response appears to be leukaemia specific, since stimulated patient PBMCs show higher frequencies of IFN-gamma secreting effector cells in response to AML blasts than to remission bone marrow cells from the same patients. Although studied in a small number of heterogeneous patient samples, the data are encouraging and support the continuing development of vaccination for poor prognosis AML patients with autologous cells genetically modified to express IL-2/CD80.
Collapse
Affiliation(s)
- Nicola Hardwick
- Department of Haematological Medicine, King’s College London, The Rayne Institute, 123 Coldharbour Lane, London, SE5 9NU UK
| | - Lucas Chan
- Department of Haematological Medicine, King’s College London, The Rayne Institute, 123 Coldharbour Lane, London, SE5 9NU UK
| | - Wendy Ingram
- Department of Haematological Medicine, King’s College London, The Rayne Institute, 123 Coldharbour Lane, London, SE5 9NU UK
| | - Ghulam Mufti
- Department of Haematological Medicine, King’s College London, The Rayne Institute, 123 Coldharbour Lane, London, SE5 9NU UK
| | - Farzin Farzaneh
- Department of Haematological Medicine, King’s College London, The Rayne Institute, 123 Coldharbour Lane, London, SE5 9NU UK
| |
Collapse
|
8
|
Dai ZS, Chen QF, Lu HZ, Xie Y. Defective expression and modulation of B7-2/CD86 on B cells in B cell chronic lymphocytic leukemia. Int J Hematol 2009; 89:656-63. [PMID: 19430862 DOI: 10.1007/s12185-009-0320-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 03/26/2009] [Accepted: 04/07/2009] [Indexed: 12/01/2022]
Abstract
Malignant monoclonal B cells of chronic B cell lymphocytic leukemia (B-CLL) usually fail to be cleared, which indicates important costimulatory molecules may be lacking. Among those costimulatory signals, B7-1/CD80 and B7-2/CD86 caused utmost attention. In this study, B7-1 and B7-2 expression on B cells in chronic B cell lymphocytic leukemia patients were detected. Data showed that B7-2 expression in chronic B cell lymphocytic leukemia patients is significantly lower than in normal people, which suggests defective B7-2 expression may be one of the pathogenic mechanisms of chronic B cell lymphocytic leukemia. Further, we confirmed interferon-gamma could induce B7-2 expression slightly and promote T-cell response against chronic B cell lymphocytic leukemia cells, indicating interferon-gamma has clinical value in chronic leukemia immunotherapy based on modulating B7-2 expression.
Collapse
Affiliation(s)
- Zhen-sheng Dai
- Department of Infectious Disease, Shanghai Public Health Clinical Center affiliated to Fudan University, Jinshan District, Shanghai, People's Republic of China.
| | | | | | | |
Collapse
|
9
|
Smits ELJM, Berneman ZN, Van Tendeloo VFI. Immunotherapy of acute myeloid leukemia: current approaches. Oncologist 2009; 14:240-52. [PMID: 19289488 DOI: 10.1634/theoncologist.2008-0165] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Following standard therapy that consists of chemotherapy with or without stem cell transplantation, both relapsed and refractory disease shorten the survival of acute myeloid leukemia (AML) patients. Therefore, additional treatment options are urgently needed, especially to fight residual AML cells. The identification of leukemia-associated antigens and the observation that administration of allogeneic T cells can mediate a graft-versus-leukemia effect paved the way to the development of active and passive immunotherapy strategies, respectively. The aim of these strategies is the eradication of AML cells by the immune system. In this review, an overview is provided of both active and passive immunotherapy strategies that are under investigation or in use for the treatment of AML. For each strategy, a critical view on the state of the art is given and future perspectives are discussed.
Collapse
Affiliation(s)
- Evelien L J M Smits
- Vaccine & Infectious Disease Institute (VIDI), Laboratory of Experimental Hematology, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | | | | |
Collapse
|
10
|
Abstract
The applications of chemotherapy for the treatment of AML have been unchanged over the past three decades, with only 30% of patients demonstrating disease-free survival (DFS) [118]. Despite achieving CR following induction chemotherapy, the majority of patients relapse and succumb to their disease [6]. In view of the limitations encountered by cytarabine/anthracycline based regimes, attention has shifted to immunotherapy as a means to treat AML and provide significant long-term DFS. This chapter will discuss the role of the immune system and recent advances in immunotherapy for the treatment of AML, focusing on cellular and non-cellular approaches.
Collapse
|
11
|
van de Loosdrecht AA, van den Ancker W, Houtenbos I, Ossenkoppele GJ, Westers TM. Dendritic cell-based immunotherapy in myeloid leukaemia: translating fundamental mechanisms into clinical applications. Handb Exp Pharmacol 2009:319-348. [PMID: 19031033 DOI: 10.1007/978-3-540-71029-5_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Immunotherapy for leukaemia patients, aiming at the generation of anti-leukaemic T cell responses, could provide a new therapeutic approach to eliminate minimal residual disease (MRD) cells in acute myeloid leukaemia (AML). Leukaemic blasts harbour several ways to escape the immune system including deficient MHC class II expression, low levels of co-stimulatory molecules and suppressive cytokines. Therapeutic vaccination with dendritic cells (DC) is now recognized as an important investigational therapy. Due to their unique antigen presenting capacity, immunosuppressive features of the leukaemic blasts can be circumvented. DC can be successfully cultured from leukaemic blasts in 60-70% of patients and show functional potential in vivo. Alternatively, monocyte derived DC obtained at time of complete remission loaded with leukaemia-specific antigens can be used as vaccine. Several sources of leukaemia-associated antigen and different methods of loading antigen onto DC have been used in an attempt to optimize antitumour responses including apoptotic cells, necrotic cell lysates and tumour-associated pep-tides. Currently, the AML-derived cell line MUTZ-3, an immortalized equivalent of CD34(+) DC precursor cells, is under investigation for vaccination purposes. For effective DC vaccination the intrinsic tolerant state of the patient must be overcome. Therefore, the development of efficient and safe adjuvants in antigen specific immunotherapeutic programs should be encouraged.
Collapse
Affiliation(s)
- A A van de Loosdrecht
- Department of Haematology, VU-Institute of Cancer and Immunology, VU University Medical Center, Amsterdam, De Boelelaan 1117, 1081 HV, The Netherlands.
| | | | | | | | | |
Collapse
|
12
|
The role of indoleamine 2,3-dioxygenase in the induction of immune tolerance: focus on hematology. Blood 2008; 113:2394-401. [PMID: 19023117 DOI: 10.1182/blood-2008-07-144485] [Citation(s) in RCA: 188] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The regulation of the interaction between the immune system and antigens, which may lead to the induction of immune tolerance, is critical both under physiologic conditions and in different pathological settings. In the past few years, major strides have been made in our understanding of the molecular and cellular bases of this process. Novel pathways have been identified and several novel therapeutic agents are currently under clinical investigation for those diseases in which the normal balance between activation and suppression of the immune response is altered. The tryptophan catabolic enzyme, indoleamine 2,3-dioxygenase (IDO), is one of the key players involved in the inhibition of cell proliferation, including that of activated T cells. Recent works have demonstrated a crucial role for IDO in the induction of immune tolerance during infection, pregnancy, transplantation, autoimmunity, and neoplasias, including hematologic malignancies. In this review, the role of IDO in the induction of immunologic tolerance is addressed with a specific focus on its recently discovered effect on hematologic malignancies.
Collapse
|
13
|
Abstract
There is a need for novel treatment for acute leukaemia as relapse rates remain unacceptably high. Immunotherapy aims to stimulate the patient's immune responses to recognize and destroy leukaemia cells whilst activating immune memory. The qualities of the most potent professional antigen-presenting cell, the dendritic cell (DC), can be used to stimulate leukaemia-specific cytotoxic T cells. DCs can be loaded with leukaemia antigens, or leukaemia blasts can be modified to express DC-like properties for use in vaccine therapy. This chapter will review the rationale for DC vaccine therapy, the preclinical and clinical trials to date, the barriers to successful DC vaccine therapies and the role of immune adjuncts to improve outcomes.
Collapse
Affiliation(s)
- Caroline Duncan
- Department of Haematology, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK.
| | | |
Collapse
|
14
|
Lee JJ, Choi BH, Kang HK, Kim SK, Nam JH, Yang DH, Kim YK, Kim HJ, Chung IJ. Monocyte-derived dendritic cells from HLA-matched allogeneic donors showed a greater ability to induce leukemic cell-specific T cells in comparison to leukemic cell-derived dendritic cells or monocyte-derived dendritic cells from AML patients. Leuk Res 2008; 32:1653-60. [PMID: 18501426 DOI: 10.1016/j.leukres.2008.03.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 03/24/2008] [Accepted: 03/24/2008] [Indexed: 12/01/2022]
Abstract
We investigated the usefulness of allogeneic monocyte-derived dendritic cells (allogeneic mDCs) pulsed with leukemic lysates in acute myeloid leukemia (AML). Allogeneic mDCs showed higher expressions of several molecules (HLA-DR, CD80, CD83 or CD86), higher production of IL-12 and higher capacity to stimulate allogeneic T cells compared to both leukemic DCs and autologous mDCs. Autologous T cells primed by allogeneic mDCs displayed a larger number of interferon-gamma-secreting cells against leukemic cells than those primed by either leukemic DCs or autologous mDCs. These results suggest that monocyte-derived DCs from HLA-matched allogeneic donors can be used as an alternative to generate leukemia-specific cytotoxic T cells and to overcome the limitation of leukemic DCs or autologous mDCs.
Collapse
Affiliation(s)
- Je-Jung Lee
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeonnam, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Nguyen Pham TN, Choi BH, Kang HK, Jin CC, Minh NHT, Kim SK, Nam JH, Yang DH, Kim YK, Kim HJ, Chung IJ, Lee JJ. Optimization and limitation of calcium ionophore to generate DCs from acute myeloid leukemic cells. Cancer Res Treat 2007; 39:175-80. [PMID: 19746185 DOI: 10.4143/crt.2007.39.4.175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Accepted: 11/12/2007] [Indexed: 11/21/2022] Open
Abstract
PURPOSE Calcium ionophore (CI) is used to generate dendritic cells (DCs) from progenitor cells, monocytes, or leukemic cells. The aim of this study was to determine the optimal dose of CI and the appropriate length of cell culture required for acute myeloid leukemia (AML) cells and to evaluate the limitations associated with CI. MATERIALS AND METHODS To generate leukemic DCs, leukemic cells (4x10(6) cells) from six AML patients were cultured with various concentrations of CI and/or IL-4 for 1, 2 or 3 days. RESULTS Potent leukemic DCs were successfully generated from all AML patients, with an average number of 1.2x10(6) cells produced in the presence of CI (270 ng/ml) for 2 days. Several surface molecules were clearly upregulated in AML cells supplemented with CI and IL-4, but not CD11c. Leukemic DCs cultured with CI had a higher allogeneic T cell stimulatory capacity than untreated AML cells, but the addition of IL-4 did not augment the MLR activity of these cells. AML cells cultured with CI in the presence or absence of IL-4 showed increased levels of apoptosis in comparison to primary cultures of AML cells. CONCLUSION Although CI appears to be advantageous in terms of time and cost effectiveness, the results of the present study suggest that the marked induction of apoptosis by CI limits its application to the generation of DCs from AML cells.
Collapse
|
16
|
Curti A, Pandolfi S, Valzasina B, Aluigi M, Isidori A, Ferri E, Salvestrini V, Bonanno G, Rutella S, Durelli I, Horenstein AL, Fiore F, Massaia M, Colombo MP, Baccarani M, Lemoli RM. Modulation of tryptophan catabolism by human leukemic cells results in the conversion of CD25- into CD25+ T regulatory cells. Blood 2007; 109:2871-7. [PMID: 17164341 DOI: 10.1182/blood-2006-07-036863] [Citation(s) in RCA: 293] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) is a novel immunosuppressive agent expressed in some subsets of normal and neoplastic cells, including acute myeloid leukemia (AML) cells. Here, we show that IDO expression correlates with increased circulating CD4+CD25+FOXP3+ T cells in patients with AML at diagnosis. In vitro, IDO+ AML cells increase the number of CD4+ CD25+ T cells expressing surface CTLA-4 and FOXP3 mRNA, and this effect is completely abrogated by the IDO inhibitor, 1-methyl tryptophan (1-MT). Purified CD4+CD25+ T cells obtained from coculture with IDO+ AML cells act as T regulatory (T(reg)) cells because they do not proliferate, do not produce interleukin (IL)-2, and inhibit naive T-cell proliferation. Coculture with IDO+AML cells results in the conversion of CD4+CD25- into CD4+CD25+ T cells, which is completely abrogated by 1-MT. Moreover, in mice, intrasplenic injection of IDO+ leukemia/ lymphoma A20 cells induces the expansion of bona fide T(reg) cells by conversion of CD4+CD25- T cells; this effect is counteracted by 1-MT treatment. These data indicate that AML cells induce T-cell tolerance by directly converting CD4+CD25- T cells into CD4+CD25+ T(reg) cells through an IDO-dependent mechanism.
Collapse
MESH Headings
- Adult
- Animals
- Base Sequence
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Coculture Techniques
- DNA Primers/genetics
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Gene Expression
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Interleukin-2 Receptor alpha Subunit/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Mice
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tryptophan/metabolism
Collapse
Affiliation(s)
- Antonio Curti
- Institute of Hematology and Medical Oncology "L. & A. Seràgnoli," University of Bologna and Stem Cell Center, S. Orsola-Malpighi Hospital, Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Jedema I, Meij P, Steeneveld E, Hoogendoorn M, Nijmeijer BA, van de Meent M, van Luxemburg-Heijs SAP, Willemze R, Falkenburg JHF. Early detection and rapid isolation of leukemia-reactive donor T cells for adoptive transfer using the IFN-gamma secretion assay. Clin Cancer Res 2007; 13:636-43. [PMID: 17255287 DOI: 10.1158/1078-0432.ccr-06-2093] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The poor immunogenicity of most leukemias and the lack of specificity of the donor T cells limit the in vivo effectiveness of conventional donor lymphocyte infusions in many patients suffering from persistent or recurrent leukemia after allogeneic stem cell transplantation. These limitations may be overcome by the adoptive transfer of in vitro generated leukemia-reactive T cells. Although the potential clinical efficacy of this approach has been shown previously, lack of reproducibility of the procedure and the inability to show persistence and survival of the transferred T cells hampered further clinical application. The purpose of this study was to develop a new, broadly applicable strategy for the efficient generation and isolation of leukemia-reactive T cells with a better probability to survive and expand in vivo. EXPERIMENTAL DESIGN Myeloid and B-cell leukemias were modified into professional immunogenic antigen-presenting cells, and used to stimulate HLA-matched donor T cells. After two stimulations, responding donor T cells were isolated based on their secretion of IFN-gamma and tested for their capacity to recognize and kill the primary leukemia. RESULTS Using one universal stimulation and isolation protocol for various forms of leukemia, T-cell populations containing high frequencies of leukemia-reactive T cells could reproducibly be generated and early isolated under mild stimulatory conditions. Isolated T cells still had high proliferative potential and their reactivity seemed to be restricted to cells of the patient's hematopoiesis. CONCLUSION We here show a new robust procedure for the generation and isolation of leukemia-reactive T cells for adoptive transfer.
Collapse
Affiliation(s)
- Inge Jedema
- Laboratory of Experimental Hematology, Department of Hematology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Haematopoietic stem cell transplantation (HSCT) remains the only curative option for patients with myelodysplastic syndrome (MDS). Developing conditioning regimens with low toxicity, at the same time as preserving an effective graft versus tumour response, is pivotal to expanding the scope for allogeneic transplantation in older patients with MDS. With the introduction of reduced intensity conditioned regimens, transplant centres worldwide are able to offer allogeneic HSCT to a much larger cohort of patients. Graft versus host disease (GvHD) remains a significant cause of morbidity and mortality, however with the use of T-cell depletion, centres have been able to utilise volunteer unrelated donors with an increasing degree of HLA disparity. The graft versus dysplasia effect resulting from allogeneic HSCT and the infusion of donor leukocytes has led to a greater understanding of the immunological mechanisms that govern outcome following transplantation in MDS.
Collapse
Affiliation(s)
- Wendy Ingram
- Department of Haematological Medicine, Kings College Hospital and Kings College London, Denmark Hill, London, SE5 9RS, UK.
| | | | | |
Collapse
|
19
|
Yadav D, Sarvetnick N. Immunomodulation of the Anti-Islet CD8 T Cell Response by B7-2. J Clin Immunol 2007; 27:221-6. [PMID: 17243007 DOI: 10.1007/s10875-006-9067-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2006] [Accepted: 12/15/2006] [Indexed: 11/29/2022]
Abstract
The absence of diabetes in NOD mice devoid of B7-2 signifies a critical role played by B7-2 in promoting autoimmunity. We asked whether the CD8 T cell compartment is impacted by the absence of B7-2. We found significantly lower expansion of anti-islet CD8 T cells in B7-2KO mice, although their survival and activation states remained unchanged in the pancreatic lymph nodes (PLNs). CD8 T cells from B7-2KO mice exhibited significantly diminished effector function compared to NOD mice. Adoptive transfer experiments using in vitro activated anti-islet CD8 T cells showed that B7-2 does not control the effector phase of the autoreactive CD8 T cell response. Our data indicate that B7-2 promotes pancreatic autoimmunity by controlling CD8 T cell expansion and effector function, but is dispensable for CD8 T cell activation, survival, and the effector phase of anti-islet CD8 T cell response.
Collapse
Affiliation(s)
- Deepak Yadav
- Department of Immunology, Scripps Research Institute, 10550 North Torrey Pines Road, Mail Drop IMM-23, La Jolla, California 92037, USA
| | | |
Collapse
|
20
|
Kang HK, Park JS, Kim SK, Choi BH, Pham TNN, Zhu XW, Cho D, Nam JH, Kim YJ, Rhee JH, Chung IJ, Kim HJ, Lee JJ. Down-regulation of cellular vascular endothelial growth factor levels induces differentiation of leukemic cells to functional leukemic-dendritic cells in acute myeloid leukemia. Leuk Lymphoma 2007; 47:2224-33. [PMID: 17071499 DOI: 10.1080/10428190600732236] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We examined the effect of cellular vascular endothelial growth factor (VEGF) levels on the generation of leukemic dendritic cells (DCs). Leukemic DCs were successfully generated in vitro from bone marrow cells of 16 of 21 acute myeloid leukemia (AML) patients, and the cellular VEGF concentrations in the leukemic cells and the neutralization of VEGF with anti-VEGF antibody were determined. AML cells that failed to generate leukemic DCs showed significantly higher cellular VEGF levels compared with generated leukemic DCs, and down-regulation of cellular VEGF levels induced the generation of leukemic DCs from AML cells. Inhibition of cellular VEGF levels increased interleukin (IL)-12 production and the allostimulatory capacity of leukemic DCs. These results suggest that the generation of leukemic DCs from AML cells is inversely related to the VEGF production of the cells and that the down-regulation of cellular VEGF levels can induce potential differentiation of leukemic cells to functional leukemic DCs in patients with AML.
Collapse
Affiliation(s)
- Hyun-Kyu Kang
- Department of Hematology-Oncology, Chonnam National University Medical School, Gwangju, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kowolik CM, Topp MS, Gonzalez S, Pfeiffer T, Olivares S, Gonzalez N, Smith DD, Forman SJ, Jensen MC, Cooper LJN. CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Res 2006; 66:10995-1004. [PMID: 17108138 DOI: 10.1158/0008-5472.can-06-0160] [Citation(s) in RCA: 390] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Chimeric antigen receptors (CAR) combine an antigen-binding domain with a CD3-zeta signaling motif to redirect T-cell specificity to clinically important targets. First-generation CAR, such as the CD19-specific CAR (designated CD19R), may fail to fully engage genetically modified T cells because activation is initiated by antigen-dependent signaling through chimeric CD3-zeta, independent of costimulation through accessory molecules. We show that enforced expression of the full-length costimulatory molecule CD28 in CD8(+)CD19R(+)CD28(-) T cells can restore fully competent antigen-dependent T-cell activation upon binding CD19(+) targets expressing CD80/CD86. Thus, to provide costimulation to T cells through a CD19-specific CAR, independent of binding to CD80/CD86, we developed a second-generation CAR (designated CD19RCD28), which includes a modified chimeric CD28 signaling domain fused to chimeric CD3-zeta. CD19R(+) and CD19RCD28(+) CD8(+) T cells specifically lyse CD19(+) tumor cells. However, the CD19RCD28(+) CD8(+) T cells proliferate in absence of exogenous recombinant human interleukin-2, produce interleukin-2, propagate, and up-regulate antiapoptotic Bcl-X(L) after stimulation by CD19(+) tumor cells. For the first time, we show in vivo that adoptively transferred CD19RCD28(+) T cells show an improved persistence and antitumor effect compared with CD19R(+) T cells. These data imply that modifications to the CAR can result in improved therapeutic potential of CD19-specific T cells expressing this second-generation CAR.
Collapse
Affiliation(s)
- Claudia M Kowolik
- Divisions of Molecular Medicine, Beckman Research Institute and City of Hope National Medical Center, Duarte, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Choi BH, Kang HK, Park JS, Kim SK, Pham TNN, Zhu XW, Cho D, Nam JH, Chung IJ, Kim YJ, Rhee JH, Kim HJ, Lee JJ. Optimization of the concentration of autologous serum for generation of leukemic dendritic cells from acute myeloid leukemic cells for clinical immunotherapy. J Clin Apher 2006; 21:233-40. [PMID: 17120232 DOI: 10.1002/jca.20105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Clinical application of immunotherapy for acute myeloid leukemia (AML) requires the efficient induction of dendritic cells (DCs) from AML blast cells using in vitro culture. We examined the effect of autologous serum on the properties of leukemic DCs derived from leukemic cells of AML patients by culture in AIM-V medium with GM-CSF, IL-4, TNF-alpha, and 0, 2, 5, or 10% human autologous serum. The expressions of CD80, CD83, CD86, and HLA-DR were upregulated under all culture conditions; however, 10% autologous serum induced the highest expression levels of several molecules. The capacity of leukemic DCs to stimulate allogeneic T cells increased with increasing serum concentration. Stimulation of autologous CD3(+) T cells with leukemic DCs grown in the presence of various concentrations of autologous serum resulted in induction of more IFN-gamma-secreting cells than was the case for unprimed CD3(+) T cells. Leukemic DCs cultured with 10% autologous serum induced the highest numbers of IFN-gamma-secreting cells and CD8(+)CD56(+) T cells from autologous T cells. These results suggest that culture of AML blast cells in the presence of autologous serum could be used to generate leukemic DCs for immunotherapy against AML. The highest serum concentration appeared optimal for generating the most potent leukemic DCs.
Collapse
Affiliation(s)
- Bo-Hwa Choi
- Department of Hematology-Oncology, Chonnam National University Medical School, Gwangju, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lee JJ, Park MS, Park JS, Kang HK, Kim SK, Nguyen Pham TN, Zhu XW, Cho D, Nam JH, Kim YJ, Rhee JH, Chung IJ, Kim HJ. Induction of leukemic-cell-specific cytotoxic T lymphocytes by autologous monocyte-derived dendritic cells presenting leukemic cell antigens. J Clin Apher 2006; 21:188-94. [PMID: 16570260 DOI: 10.1002/jca.20085] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Leukemic-dendritic cells (leukemic-DCs) have certain limitations, which include difficult generation in 30-40% of patients, and low levels of expression of several key molecules. Therefore, an alternative approach using monocyte-derived DCs pulsed with tumor antigens is required. We investigated the possibility of immunotherapy for AML using leukemic-cell-specific cytotoxic T lymphocytes that were stimulated in vitro by autologous DCs pulsed with tumor antigens. To generate DCs, CD14(+) cells were isolated from peripheral blood mononuclear cells using magnetic-activated cell sorting, and cultured in the presence of GM-CSF and IL-4. On day 6, maturation of DCs was induced by addition of cytokine cocktail (TNF-alpha, IL-1beta, IL-6, and prostaglandin E(2)) for 2 days, and then the mature DCs were pulsed with whole leukemic cell lysates or apoptotic leukemic cells. There were no differences in the phenotypic expressions of mature DCs generated by pulsing with or without leukemic antigens. The mature DCs pulsed with tumor cell lysates or apoptotic leukemic cells showed a higher allostimulatory capacity for allogeneic CD3(+) T cells as compared with mature non-pulsed DCs. Autologous CD3(+) T cells stimulated by the mature pulsed DCs showed more potent cytotoxic activities against autologous leukemic cells than those stimulated by mature non-pulsed DCs. These results suggest that use of DCs pulsed with leukemic cell lysates or apoptotic leukemic cells is a feasible alternative immunotherapeutic approach to overcome the limitations of leukemic-DCs for the treatment of AML patients.
Collapse
Affiliation(s)
- J-J Lee
- Department of Hematology-Oncology, Chonnam National University Medical School, Gwangju, Korea.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Chan L, Hardwick NR, Guinn BA, Darling D, Gäken J, Galea-Lauri J, Ho AY, Mufti GJ, Farzaneh F. An immune edited tumour versus a tumour edited immune system: Prospects for immune therapy of acute myeloid leukaemia. Cancer Immunol Immunother 2006; 55:1017-24. [PMID: 16450142 PMCID: PMC11030980 DOI: 10.1007/s00262-006-0129-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2005] [Accepted: 01/04/2006] [Indexed: 10/25/2022]
Abstract
Cell based therapies for acute myeloid leukaemia (AML) have made significant progress in the last decade benefiting the prognosis and survival of patients with this aggressive form of leukaemia. Due to advances in haematopoietic stem cell transplantation (HSCT) and particularly the advent of reduced intensity conditioning (RIC), the scope of transplantation has now extended to those patients previously ineligible due to age and health restrictions and has been associated with a decrease in transplant related mortality. The apparent graft versus leukaemia (GvL) effect observed following HSCT demonstrates the potential of the immune system to target and eradicate AML cells. Building on previously published pre-clinical studies by ourselves and others, we are now initiating a Phase I clinical study in which lentiviral vectors are used to genetically modify AML cells to express B7.1 (CD80) and IL-2. By combining allogeneic HSCT with immunisation, using the autologous AML cells expressing B7.1 and IL-2, we hope to stimulate immune eradication of residual AML cells in poor prognosis patients that have achieved donor chimerism. In this report we describe the background to cell therapy based approaches for AML, and discuss difficulties associated with the deployment of a chronically stimulated, hence exhausted/depleted immune system to eradicate tumour cells that have already escaped immune surveillance.
Collapse
Affiliation(s)
- Lucas Chan
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Nicola R. Hardwick
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Barbara-ann Guinn
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Dave Darling
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Joop Gäken
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Joanna Galea-Lauri
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Aloysius Y. Ho
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Ghulam J. Mufti
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| | - Farzin Farzaneh
- King’s College London, Department of Haematological Molecular Medicine, The Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK
| |
Collapse
|
25
|
Curti A, Pandolfi S, Aluigi M, Isidori A, Alessandrini I, Chiodoni C, Testoni N, Colombo MP, Baccarani M, Lemoli RM. Interleukin-12 production by leukemia-derived dendritic cells counteracts the inhibitory effect of leukemic microenvironment on T cells. Exp Hematol 2006; 33:1521-30. [PMID: 16338495 DOI: 10.1016/j.exphem.2005.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2005] [Revised: 07/14/2005] [Accepted: 08/15/2005] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Acute myeloid leukemia (AML) cells are poorly immunogenic and inhibit T-cell function. AML-derived dendritic cells (AML-DCs) have better antigen-presentation capacity than undifferentiated leukemic blasts, but may not be fully competent to stimulate T cells previously inhibited by leukemic cells. MATERIALS AND METHODS AML-DCs were generated from AML cells and used to stimulate proliferation and cytokine production by T cells previously inhibited by AML cells. AML-DCs were also transfected with interleukin (IL)-12 gene by the nonviral method, nucleofection. RESULTS Mature AML-DCs stimulated naive and, to a lesser extent, leukemic cell (LC)-cultured T cells more efficiently than their immature counterparts and their activity was mediated by IL-12. AML-DCs generated from CD14(-) AML samples (which represent 80% of total AML patients) were defective in IL-12 production and T-cell activation. Addition of exogenous IL-12 to LC-cultured T cells stimulated by CD14(-)-derived AML-DCs restored optimal interferon-gamma (IFN-gamma) production and Th1 skewing. IL-12 gene-nucleofected AML-DCs derived from CD14(-) cells produced significant amounts of IL-12, maintained leukemia-specific karyotype, DC-like phenotype, and function. When stimulated by IL-12-gene transduced CD14(-)-derived AML-DCs, LC-cultured T cells produced higher concentrations of IFN-gamma, thus maintaining a Th1 cytokine profile. CONCLUSION IL-12 produced by AML-DCs plays a critical role in counteracting the inhibitory activity of LCs on T-cell function. IL-12 gene can be successfully expressed into AML-DCs defective in endogenous IL-12 production by using a novel nonviral method that does not modify their phenotypical, cytogenetic, and functional features. Genetically modified AML-DCs restore a near normal T-cell function.
Collapse
Affiliation(s)
- Antonio Curti
- Institute of Hematology and Medical Oncology L. & A. Seràgnoli, University of Bologna and Stem Cell Center, S. Orsola-Malpighi Hospital, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Xing D, Decker WK, Li S, Robinson SN, Yang H, Segal H, O'Connor S, Yao X, Komanduri KV, McMannis JD, Jones RB, de Lima M, Champlin RE, Shpall EJ. AML-loaded DC generate Th1-type cellular immune responses in vitro. Cytotherapy 2006; 8:95-104. [PMID: 16698683 DOI: 10.1080/14653240600620093] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND The generation of AML-specific T-lymphocyte responses by leukemia-derived DC has been documented by multiple investigators and is being pursued clinically. An obstacle to widespread use of this strategy is that it has not been possible to generate leukemic DC from all patients, and an alternative approach is needed if the majority of leukemia patients are to receive therapeutic vaccination in conjunction with other treatment protocols. METHODS In the present study, we generated DC from CD14-selected monocytes isolated from healthy donor PBPC and loaded them with a total cell lysate from AML patient blasts. RESULTS Immature in vitro-derived DC exhibited robust phagocytic activity, and mature DC demonstrated high expression of CD80, CD83, CD86 and the chemokine receptor CCR7, important for DC migration to local lymph nodes. Mature, Ag-loaded DC were used as APC for leukemia-specific cytotoxic T-lymphocyte (CTL) induction and demonstrated cytotoxic activity against leukemic targets. CTL lysis was Ag-specific, with killing of both allogeneic leukemic blasts and autologous DC loaded with allogeneic AML lysate. HLA-matched controls were not lysed in our system. DISCUSSION These data support further research into the use of this strategy as an alternative approach to leukemia-derived DC vaccination.
Collapse
Affiliation(s)
- D Xing
- The University of Texas MD Anderson Cancer Center, Department of Blood and Marrow Transplantation, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Cheuk ATC, Chan L, Czepulkowski B, Berger SA, Yagita H, Okumura K, Farzaneh F, Mufti GJ, Guinn BA. Development of a whole cell vaccine for acute myeloid leukaemia. Cancer Immunol Immunother 2006; 55:68-75. [PMID: 15891884 PMCID: PMC11030895 DOI: 10.1007/s00262-005-0674-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2004] [Accepted: 01/06/2005] [Indexed: 01/24/2023]
Abstract
We describe the modification of tumour cells to enhance their capacity to act as antigen presenting cells with particular focus on the use of costimulatory molecules to do so. We have been involved in the genetic modification of tumour cells to prepare a whole cell vaccine for nearly a decade and we have a particular interest in acute myeloid leukaemia (AML). AML is an aggressive and difficult to treat disease, especially, for patients for whom haematopoietic stem cell (HSC) transplant is not an option. AML patients who have a suitable donor and meet HSC transplant fitness requirements, have a 5-year survival of 50%; however, for patients with no suitable donor or for who age is a factor, the prognosis is much worse. It is particularly poor prognosis patients, who are not eligible for HSC transplant, who are likely to benefit most from immunotherapy. It would be hoped that immunotherapy would be used to clear residual tumour cells in these patients in the first remission following standard chemotherapy treatments and this will extend the remission and reduce the risk of a second relapse associated with disease progression and poor mortality rates. In this symposia report, we will focus on whole cell vaccines as an immunotherapeutic option with particular reference to their use in the treatment of AML. We will aim to provide a brief overview of the latest data from our group and considerations for the use of this treatment modality in clinical trials for AML.
Collapse
Affiliation(s)
- Adam T. C. Cheuk
- Department of Haematological Medicine, Guy’s, King’s & St. Thomas’ School of Medicine, The Rayne Institute, King’s College London, 123 Coldharbour Lane, London, SE5 9NU UK
| | - Lucas Chan
- Department of Haematological Medicine, Guy’s, King’s & St. Thomas’ School of Medicine, The Rayne Institute, King’s College London, 123 Coldharbour Lane, London, SE5 9NU UK
| | - Barbara Czepulkowski
- Department of Haematological Medicine, Guy’s, King’s & St. Thomas’ School of Medicine, The Rayne Institute, King’s College London, 123 Coldharbour Lane, London, SE5 9NU UK
| | - Stuart A. Berger
- Arthritis and Immune Disorder Research Centre, University Health Network and the Department of Immunology, University of Toronto, Toronto, Canada
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ko Okumura
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Farzin Farzaneh
- Department of Haematological Medicine, Guy’s, King’s & St. Thomas’ School of Medicine, The Rayne Institute, King’s College London, 123 Coldharbour Lane, London, SE5 9NU UK
| | - Ghulam J. Mufti
- Department of Haematological Medicine, Guy’s, King’s & St. Thomas’ School of Medicine, The Rayne Institute, King’s College London, 123 Coldharbour Lane, London, SE5 9NU UK
| | - Barbara-ann Guinn
- Department of Haematological Medicine, Guy’s, King’s & St. Thomas’ School of Medicine, The Rayne Institute, King’s College London, 123 Coldharbour Lane, London, SE5 9NU UK
| |
Collapse
|
28
|
Wang X, Zheng J, Liu J, Yao J, He Y, Li X, Yu J, Yang J, Liu Z, Huang S. Increased population of CD4+CD25high regulatory T cells with their higher apoptotic and proliferating status in peripheral blood of acute myeloid leukemia patients. Eur J Haematol 2005; 75:468-76. [PMID: 16313258 DOI: 10.1111/j.1600-0609.2005.00537.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
PURPOSE Regulatory T cells (T-reg) that control harmful autoimmune T cells in the periphery may also suppress the immune response against cancer. In this study we investigated the possible involvement of CD4(+)CD25(high) T-reg in the immune impairment of patients with acute myeloid leukemia (AML). EXPERIMENTAL DESIGN The frequencies and phenotypes of CD4(+)CD25(high) T cells in the peripheral blood of AML patients were determined by flow cytometry. To assess the functional activity of CD4(+)CD25(high) T cells, CD4(+)CD25(high), and CD4(+)CD25(-) T cells were sorted from peripheral blood mononuclear cells with FACS Vantage. The immunoregulatory properties of CD4(+)CD25(high) and CD4(+)CD25(-) T cells were characterized by proliferation assays and cytokine production assays. In addition, the frequency of apoptotic and proliferating cells in CD4(+)CD25(high) T cells were respectively evaluated by 7AAD and ki67 binding cells using flow cytometry. RESULTS Compared with healthy controls, AML patients had a higher proportion of CD4(+)CD25(high) T cells in peripheral blood. These cells were CD45-RA(-), CD69(-), CD45-RO(+), CD95(+), and intercellular CTLA-4(+), and secreted low levels of TNF-alpha and IL-10, but no IL-2, IL-4, IL-5, and IFN-gamma. They inhibited the proliferation and cytokine production (IL-2, IFN-gamma) of CD4(+)CD25(-) T cells, but improved IL-10 production under the co-culture of both subsets with stimulation, thus behaving as T-reg. Notably, CD4(+)CD25(high) T cells in AML patients presented significantly higher apoptosis and proliferation than that of healthy individuals. CONCLUSIONS The frequency of CD4(+)CD25(high) T-reg in peripheral blood in AML patients is significantly higher when compared with healthy individuals, likely due to the increasing proliferation of CD4(+)CD25(high) T cells.
Collapse
Affiliation(s)
- Xingbing Wang
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Houtenbos I, Westers TM, Ossenkoppele GJ, van de Loosdrecht AA. Employing the immunological synapse in AML: Development of leukemic dendritic cells for active specific immunization. Immunobiology 2005; 210:249-57. [PMID: 16164032 DOI: 10.1016/j.imbio.2005.05.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Cytotoxic T cells directed against leukemic blasts have been observed in patients with acute myeloid leukemia (AML). However, generation of efficient T-cell responses is hampered due to several factors that enable AML blasts to protect themselves from the patients immune system. Improved immune responses can be established by the differentiation of AML blasts into AML-derived dendritic cells (DC) thereby conserving their intrinsic leukemia specific antigens and obtaining full capacity to present these antigens to naive T cells. This review discusses increased immunogenicity of AML blasts by differentiation into AML-DC and describes ways to augment the AML-DC vaccination approach.
Collapse
Affiliation(s)
- Ilse Houtenbos
- Department of Hematology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
30
|
Klammer M, Waterfall M, Samuel K, Turner ML, Roddie PH. Fusion hybrids of dendritic cells and autologous myeloid blasts as a potential cellular vaccine for acute myeloid leukaemia. Br J Haematol 2005; 129:340-9. [PMID: 15842657 DOI: 10.1111/j.1365-2141.2005.05477.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
We assessed the potential of tumour cell/dendritic cell fusion hybrids to generate in vitro anti-leukaemic T-cell responses following co-culture with autologous remission lymphocytes in six patients with acute myeloid leukaemia (AML). Comparison was made to anti-leukaemic responses induced by mature dendritic cells (mDC) co-cultured with autologous, irradiated myeloid blasts. Fusion hybrids induced anti-leukaemic T-cell immune responses in three of six patients. Tumour-pulsed mDC induced T-cellular responses in two other patients. Only one of six patients remission lymphocytes failed to develop leukaemia-directed immune responses following stimulation with either construct. Anti-proliferative properties of fusion hybrids against allogeneic lymphocytes were observed in mixed lymphocyte-leukaemia reactions and were found not to be specific to the cell fusion partners and did not prevent the ability of AML-mDC heterokaryons to induce autologous anti-leukaemic cytotoxicity. We conclude that tumour cell/dendritic cell fusion hybrids hold promise as a cellular vaccine for AML.
Collapse
Affiliation(s)
- M Klammer
- University of Edinburgh-Leukaemia Research Fund, John Hughes Bennett Laboratory, Western General Hospital, Edinburgh EH4 2XU, UK.
| | | | | | | | | |
Collapse
|
31
|
Zheng SY, Li DC, Zhang ZD, Zhao J, Ge JF. Anti-gastric cancer active immunity induced by FasL/B7-1 gene-modified tumor cells. World J Gastroenterol 2005; 11:3204-11. [PMID: 15929168 PMCID: PMC4316049 DOI: 10.3748/wjg.v11.i21.3204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the activation of cytotoxic T lymphocytes (CTLs) against gastric cancer cells induced by FasL/B7-1 (FB-11) gene-modified tumor cells, and to explore whether co-expression of FasL and B7-1 in SGC-7901 tumor cells could initiate synergistic antitumor effect.
METHODS: FasL and B7-1 genes were transfected into human SGC-7901 gastric cancer cells with adenovirus vectors. The positive clones were selected by G418. FasL and B7-1 genes were detected by flow cytometry and RT-PCR. Abdominal infiltrating lymphocytes and sensitized spleen cells were obtained from mice that were immunized with SGC-7901/FB-11 or wild type SGC-7901 cells intraperitoneally, and cytotoxicity of these CTLs against tumor cells was determined by MTT assay.
RESULTS: Flow cytometry and RT-PCR showed that FasL and B7-1 genes were highly expressed. FasL and B7-1 transfected cancer cells had a high apoptosis index. DNA laddering suggested that FasL and B7-1 genes induced gastric cancer cell apoptosis. FasL+/B7-1+SGC-7901 cells (SGC-7901/FB-11) were inoculated subcutaneously in the dorsal skin of C57BL/6 mice and then decreased their tumorigenicity greatly (z = 2.15-46.10, P<0.01). SGC-7901/FB-11 cell-sensitized mice obtained protective immune activity against the rechallenge of wild type SGC-7901 cells (z = 2.06-44.30, P<0.05). The cytotoxicity of CTLs induced by SGC-7901/FB-11 cells against SGC-7901 was significantly higher than that of CTLs activated by wild-type SGC-7901 cells (84.1±2.4% vs 30.5±2.3%, P<0.05).
CONCLUSION: FasL and B7-1 genes can effectively promote the activity of CTLs against gastric cancer cells. FasL/B7-1 molecules play an important role in CTL cytotoxicity.
Collapse
Affiliation(s)
- Shi-Ying Zheng
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Suzhou University, Suzhou 215006, Jiangsu Province, China. syzheng88@ sina.com
| | | | | | | | | |
Collapse
|
32
|
Lee JJ, Choi BH, Nam JH, Park MS, Song WH, Yang DH, Kim YK, Cho SH, Chung IJ, Park KS, Lee IK, Kim HJ. The generation of leukemic dendritic cells from acute myeloid leukemia cells is potentiated by the addition of CD40L at the terminal maturation stage. J Clin Apher 2004; 19:130-6. [PMID: 15493054 DOI: 10.1002/jca.20015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Leukemic dendritic cells (DCs) that are derived from acute myeloid leukemia (AML) cells display low-level expression of several key molecules. We investigated the optimal combination of cytokines needed to generate potent leukemic DCs from AML cells in vitro. AML cells were cultured in the presence of the following combinations of cytokines: Group A, granulocyte-macrophage colony-stimulating factor (GM-CSF) + interleukin-4 (IL-4) + tumor necrosis factor-alpha (TNF-alpha); Group B, GM-CSF + IL-4 + CD40L; and Group C, CD40L addition at the terminal maturation point of cells that were grown as for Group A. The AML cells showed clear upregulation of CD80, CD83, CD86, CD40, and HLA-DR expression under all culture conditions, without significant differences between these groups. However, the addition of CD40L (as in Group C) showed a slight upregulation in the expression of CD83 and CD86 on leukemic DCs. The leukemic DCs in Groups A and B had higher allogeneic T-cell stimulatory capacities than untreated AML cells, and the addition of CD40L (Group C) enhanced this effect. The function of the cytotoxicity-stimulating autologous T cells was also augmented by the addition of CD40L (Group C). These results suggest that AML cells may be used to generate leukemic DCs using various cytokine combinations, and that the most potent, mature leukemic DCs are generated by the addition of CD40L to terminal-stage AML cultures that are grown in the presence of conventional cytokine combinations.
Collapse
Affiliation(s)
- Je-Jung Lee
- Department of Hematology-Oncology, Chonnam National University Medical School, Gwangju, Korea.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hicks C, Cheung C, Lindeman R. Restimulation of tumour-specific immunity in a patient with AML following injection with B7-1 positive autologous blasts. Leuk Res 2003; 27:1051-61. [PMID: 12859998 DOI: 10.1016/s0145-2126(03)00058-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Leukaemic blast cells lack co-stimulatory molecules such as B7, necessary for T-lymphocyte activation. We have used modified CD80+ (B7-1+) tumour cells, with autologous, IL-2 producing, stromal marrow cells in a series of subcutaneous vaccinations to provide a localised environment for the enhancement of cytotoxic T-lymphocytes (CTL) in a patient with acute myeloid leukaemia (AML). Localised inflammation was evident after the fifth and sixth injections with a reduction in the number of circulating blasts in the following 2 weeks. Peripheral blood in vitro CTL activity increased 36-47% after five injections.CD4 T-lymphocytes (5.7%) expanded from post-injection skin biopsies, expressed intracellular IFNgamma and perforin when exposed to autologous B7-1+ blasts and when co-cultured with either B7-1+ or unmanipulated autologous blast cells showed proliferative responses. In this patient, co-injection of B7-1+ tumour cells, together with a local source of sustained IL-2, resulted in an autologous anti-leukaemic in vitro immune response.
Collapse
Affiliation(s)
- Christine Hicks
- Department of Haematology, Prince of Wales Hospital, Randwick, Sydney, NSW 2031, Australia.
| | | | | |
Collapse
|
34
|
Ge NL, Ye SL, Zheng N, Sun RX, Liu YK, Tang ZY. Prevention of hepatocellular carcinoma in mice by IL-2 and B7-1 genes co-transfected liver cancer cell vaccines. World J Gastroenterol 2003; 9:2182-5. [PMID: 14562374 PMCID: PMC4656459 DOI: 10.3748/wjg.v9.i10.2182] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the immunoprotective effect of liver cancer vaccine with co-transfected IL-2 and B7-1 genes on hepatocarcinogenesis in mice.
METHODS: The murine liver cancer cell line Hepal-6 was transfected with IL-2 and/or B7-1 gene via recombinant adenoviral vectors and the liver cancer vaccines were prepared. C57BL/6 mice were immunized with these vaccines and challenged with the parental Hepal-6 cells afterwards. The immunoprotection was investigated and the reactive T cell line was assayed.
RESULTS: The immunoprotection of the tumor vaccine was demonstrated. The effect of IL-2 and B7-1 genes co-transfected Hepal-6 liver cancer vaccine (Hep6-IL2/B7 vaccine) on the onset of tumor formation was the strongest. When attacked with wild Hepal-6 cells, the median survival period of the mice immunized with Hep6-IL2/B7 vaccine was the longest (68 d, χ2 = 7.70-11.69, P < 0.05) and the implanted tumor was the smallest (z = 3.20-44.10, P < 0.05). The effect of single IL-2 or B7-1 gene-transfected vaccine was next to the IL2/B7 gene co-transfected group, and the mean survival periods were 59 and 54 d, respectively. The mean survival periods of wild or enhanced green fluorescence protein gene modified vaccine immunized group were 51 and 48 d, respectively. The mice in control group all died within 38 d and the implanted tumor was the largest (z = 3.20-40.21, P < 0.05). The cellular immunofunction test and cytotoxicity study showed that the natural killer (NK) cell, lymphokine activated killer (LAK) cell and cytotoxic T lymphocyte (CTL) activities were significantly increased in mice immunized with the Hep6-IL2/B7 vaccine, (29.5% ± 2.5%, 65.0% ± 2.9%, 83.1% ± 1.5% respectively, compared with other groups, P < 0.05).
CONCLUSION: The Hep6-IL2/B7 liver cancer vaccines can induce the mice to produce activated and specific CTL against the parental tumor cells, and demonstrate stronger effect on the hepatocarcinogenesis than single gene modified or the regular tumor vaccine. Therefore, the vaccines may become a novel potential therapy for recurrence and metastasis of HCC.
Collapse
Affiliation(s)
- Ning-Ling Ge
- Liver Cancer Institute of Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
| | | | | | | | | | | |
Collapse
|
35
|
Erben U, Thiel E, Bittroff-Leben A, Schoch C, Fichtner I, Dürkop H, Notter M. CS-1, a novel c-kithi+ acute myeloid leukemia cell line with dendritic cell differentiation capacity and absent immunogenicity. Int J Cancer 2003; 105:232-40. [PMID: 12673685 DOI: 10.1002/ijc.11053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Complex cytogenetic abnormalities confer dismal prognoses in myeloid malignancies. Even bone marrow transplantation from siblings or matched unrelated donors offer minimal chances for cure, suggesting that these cases are not only refractory to chemotherapy but also resist the graft-vs.-leukemia effect. We herein describe the first permanent, factor-independent c-kit(hi+) cell line CS-1 derived from an unrelated donor stem cell transplanted patient with relapsed acute myeloid leukemia (AML)-M5a of high-risk karyotype [monosomy 7, t(2;11)(q31;p13), t(10;12)(q24;q24)]. Having the same karyotype, CS-1 exhibits an autonomous growth pattern and responds to stem cell factor (SCF). CS-1 did not induce T cell activation in mixed-lymphocyte-tumor-cultures (MLTCs) and, when used as third party stimulators, decreased T cell proliferation in mixed-lymphocyte reactions (MLRs). Cytokines added exogenously or secreted from bystander T cells caused CS-1 to differentiate into dendritic cells (DCs). CS-1-derived DCs, in contrast to DCs originating from non-malignant CD34(+) progenitor cells, had virtually no T cell stimulatory effect, indicating that CS-1 is both immunosuppressive and poorly immunogenic. These properties may partially be due to the detected downregulation of costimulatory molecules and appear to involve a soluble factor. CS-1 cells injected subcutaneously (s.c.) to non-obese diabetes/severe combined immunodeficient (NOD/SCID) mice produced solid tumors, disseminating into bone marrow and spleen. The data show that transforming AML blasts with high-risk karyotype into DCs is insufficient to restore their immunogenicity and that the CS-1 cell line is useful to identify tumor-related immunosuppressive mechanisms in vitro and in vivo.
Collapse
MESH Headings
- Animals
- Antigens, CD34/immunology
- Antigens, CD34/metabolism
- Bone Marrow/pathology
- Cell Differentiation
- Cell Division
- Child
- Cytokines/metabolism
- Cytokines/pharmacology
- Dendritic Cells/immunology
- Down-Regulation
- Female
- Humans
- Immunophenotyping
- In Vitro Techniques
- Injections, Subcutaneous
- Karyotyping
- Leukemia, Monocytic, Acute/immunology
- Leukemia, Monocytic, Acute/pathology
- Lymphocyte Activation
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Neoplastic Stem Cells/immunology
- Proto-Oncogene Proteins c-kit/metabolism
- Spleen/pathology
- Stem Cell Factor/pharmacology
- T-Lymphocytes/immunology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Ulrike Erben
- Department of Hematology, Oncology and Transfusion Medicine, University Hospital Benjamin Franklin, Free University, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
36
|
Holloway PA, Kaldenhoven N, Kok-Schoemaker HM, Van Kessel B, Van Blokland WTM, Bloem AC, Lokhorst HM. Antigens shared by malignant plasma cells and normal B cells may be involved in graft versus myeloma. Clin Exp Immunol 2003; 131:340-6. [PMID: 12562398 PMCID: PMC1808635 DOI: 10.1046/j.1365-2249.2003.02067.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cytotoxic T cells play an important role in graft-versus-host-disease (GvHD) and graft-versus-leukaemia/myeloma, which may occur in patients treated with an allogeneic stem cell transplantation (ASCT). Here, we describe the selection of a myeloma reactive CD4+ cytotoxic T cell-line (CTL) and two CD4+ clones from this CTL. The CTL was generated from the blood from a patient with multiple myeloma (MM) with graft versus myeloma/GvHD, following an ASCT. The CTL was stimulated using irradiated peripheral blood mononuclear cells and EBV transformed B cells from the myeloma patient (EBVp), both of which were obtained prior to ASCT. Both the CTL and the two T cell clones specifically lysed EBVp and secreted IFN-gamma after coculture with EBVp and autologous myeloma tumour cells in a class II restricted fashion. These results show that myeloma tumour cells and autologous B cells present a common polymorphic peptide that functions as a target for graft derived cytotoxic T cells. Identification of these proteins will give insight into the relationship between graft versus myeloma (GvM) and GvHD and may provide immunotherapeutical targets in the treatment of MM.
Collapse
Affiliation(s)
- P A Holloway
- Department of Haematology, UMCU, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
37
|
Barrett AJ, Rezvani K, Solomon S, Dickinson AM, Wang XN, Stark G, Cullup H, Jarvis M, Middleton PG, Chao N. New Developments in Allotransplant Immunology. Hematology 2003:350-71. [PMID: 14633790 DOI: 10.1182/asheducation-2003.1.350] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Abstract
After allogeneic stem cell transplantation, the establishment of the donor’s immune system in an antigenically distinct recipient confers a therapeutic graft-versus-malignancy effect, but also causes graft-versus-host disease (GVHD) and protracted immune dysfunction. In the last decade, a molecular-level description of alloimmune interactions and the process of immune recovery leading to tolerance has emerged. Here, new developments in understanding alloresponses, genetic factors that modify them, and strategies to control immune reconstitution are described.
In Section I, Dr. John Barrett and colleagues describe the cellular and molecular basis of the alloresponse and the mechanisms underlying the three major outcomes of engraftment, GVHD and the graft-versus-leukemia (GVL) effect. Increasing knowledge of leukemia-restricted antigens suggests ways to separate GVHD and GVL. Recent findings highlight a central role of hematopoietic-derived antigen-presenting cells in the initiation of GVHD and distinct properties of natural killer (NK) cell alloreactivity in engraftment and GVL that are of therapeutic importance. Finally, a detailed map of cellular immune recovery post-transplant is emerging which highlights the importance of post-thymic lymphocytes in determining outcome in the critical first few months following stem cell transplantation. Factors that modify immune reconstitution include immunosuppression, GVHD, the cytokine milieu and poorly-defined homeostatic mechanisms which encourage irregular T cell expansions driven by immunodominant T cell–antigen interactions.
In Section II, Prof. Anne Dickinson and colleagues describe genetic polymorphisms outside the human leukocyte antigen (HLA) system that determine the nature of immune reconstitution after allogeneic stem cell transplantation (SCT) and thereby affect transplant outcomethrough GVHD, GVL, and transplant-related mortality. Polymorphisms in cytokine gene promotors and other less characterized genes affect the cytokine milieu of the recipient and the immune reactivity of the donor. Some cytokine gene polymorphisms are significantly associated with transplant outcome. Other non-HLA genes strongly affecting alloresponses code for minor histocompatibility antigens (mHA). Differences between donor and recipient mHA cause GVHD or GVL reactions or graft rejection. Both cytokine gene polymorphisms (CGP) and mHA differences resulting on donor-recipient incompatibilities can be jointly assessed in the skin explant assay as a functional way to select the most suitable donor or the best transplant approach for the recipient.
In Section III, Dr. Nelson Chao describes non-pharmaceutical techniques to control immune reconstitution post-transplant. T cells stimulated by host alloantigens can be distinguished from resting T cells by the expression of a variety of activation markers (IL-2 receptor, FAS, CD69, CD71) and by an increased photosensitivity to rhodamine dyes. These differences form the basis for eliminating GVHD-reactive T cells in vitro while conserving GVL and anti-viral immunity. Other attempts to control immune reactions post-transplant include the insertion of suicide genes into the transplanted T cells for effective termination of GVHD reactions, the removal of CD62 ligand expressing cells, and the modulation of T cell reactivity by favoring Th2, Tc2 lymphocyte subset expansion. These technologies could eliminate GVHD while preserving T cell responses to leukemia and reactivating viruses.
Collapse
Affiliation(s)
- A John Barrett
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892-0003, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Mohty M, Olive D, Gaugler B. Leukemic dendritic cells: potential for therapy and insights towards immune escape by leukemic blasts. Leukemia 2002; 16:2197-204. [PMID: 12399962 DOI: 10.1038/sj.leu.2402710] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2002] [Accepted: 05/30/2002] [Indexed: 11/08/2022]
Abstract
Dendritic cells (DCs) are a system of potent antigen-presenting cells (APCs) specialized to initiate primary immune responses. DCs are considered important elements in the induction of specific antitumor cytotoxic effectors. At present, because of potential therapeutic implications, the critical role of DCs in cancer patients is under intensive investigation. Interactions between DCs and acute myeloid leukemia cells represent an attractive model for the study of DC physiology. Moreover, DCs can be a valuable therapeutic tool for the adjuvant treatment of leukemic patients. However, DC subsets in vivo may also be affected by leukemogenesis and may contribute to the escape of leukemia from immune control. The aim of this review is to shed further light on this paradoxical picture where the line between immune tolerance and immune defense is narrow.
Collapse
Affiliation(s)
- M Mohty
- Laboratoire d'Immunologie des Tumeurs, Institut Paoli-Calmettes, Université de la Méditerranée, Marseille, France
| | | | | |
Collapse
|
39
|
Romi F, Bø L, Skeie GO, Myking A, Aarli JA, Gilhus NE. Titin and ryanodine receptor epitopes are expressed in cortical thymoma along with costimulatory molecules. J Neuroimmunol 2002; 128:82-9. [PMID: 12098514 DOI: 10.1016/s0165-5728(02)00145-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cortical-type thymomas are associated with myasthenia gravis (MG) in 50% of the cases. MG is caused by antibodies against the acetylcholine receptors (AChR), but additional non-AChR muscle autoantibodies such as those against titin and ryanodine receptor (RyR) are found in up to 95% of MG patients with thymoma. To elucidate the induction of non-AChR autoantibodies in thymoma-associated MG, we studied cortical-type thymomas from seven thymoma MG patients, and sera from six of them. All six had titin antibodies, and four had RyR antibodies. Titin and RyR epitopes were co-expressed along with LFA3 and B7 (BB1) costimulatory molecules on thymoma antigen-presenting cells (APC) in all thymomas. In normal thymus, the staining by anti-titin, anti-RyR, anti-LFA3, and anti-BB1 antibodies was weak and occurred exclusively in the medulla and perivascularly. Our results indicate a primary autosensitization against titin and RyR antigens inside the thymoma. In MG-associated thymoma, the mechanisms involved in the initial autosensitization against titin and RyR are probably similar to those implicated in the autosensitization against AChR. In all cases, there is an overexpression of muscle-like epitopes and costimulatory molecules indicating that the T-cell autoimmunization is actively promoted by the pathogenic microenvironment inside the thymoma.
Collapse
Affiliation(s)
- Fredrik Romi
- Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway.
| | | | | | | | | | | |
Collapse
|
40
|
Hock BD, Patton WN, Budhia S, Mannari D, Roberts P, McKenzie JL. Human plasma contains a soluble form of CD86 which is present at elevated levels in some leukaemia patients. Leukemia 2002; 16:865-73. [PMID: 11986949 DOI: 10.1038/sj.leu.2402466] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2001] [Accepted: 01/17/2002] [Indexed: 11/09/2022]
Abstract
Cell surface expression of CD86 (mCD86) provides an important co-stimulatory signal which profoundly influences immune responses. In this report, we investigated the potential presence of a circulating soluble form of CD86 (sCD86) in normal individuals and patients with acute myeloid leukaemia (AML) or B cell chronic lymphocytic leukaemia (B-CLL). Circulating sCD86 was detected in the plasma of all normal individuals (1.04 +/- 0.33 ng/ml, n = 51) and patients analysed. Plasma collected from AML patients in remission (n = 6) contained only low levels of sCD86 but significantly elevated levels (> or =2.65 ng/ml, P < 0.0001) were detected in 10/24 AML patients analysed at the time of presentation or relapse. Significantly elevated levels of sCD86 were also detected in 2/17 B-CLL patients. There was no correlation between sCD86 levels and other clinical parameters. RT-PCR analysis demonstrated that normal monocytes and dendritic cells, as well as isolated AML (n = 2) and B-CLL (n = 4) cells, expressed an alternatively spliced transcript of CD86 which encoded a soluble form absent in normal T, B and NK cells. The finding that a proportion of leukaemia patients contain elevated levels of sCD86 and that at least some leukaemic cells express sCD86 transcript suggests a potential role for sCD86 in modulating mCD86 signalling during the malignant process.
Collapse
MESH Headings
- Acute Disease
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Alternative Splicing
- Antigens, CD/blood
- Antigens, CD/genetics
- B7-2 Antigen
- Case-Control Studies
- Dendritic Cells/metabolism
- Disease Progression
- Enzyme-Linked Immunosorbent Assay/standards
- Female
- Humans
- Leukemia/blood
- Leukemia/metabolism
- Leukemia/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/blood
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Myeloid/blood
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid/pathology
- Male
- Membrane Glycoproteins/blood
- Membrane Glycoproteins/genetics
- Middle Aged
- Monocytes/metabolism
- RNA, Messenger/analysis
- Solubility
- Up-Regulation
Collapse
Affiliation(s)
- B D Hock
- Haematology Research Group, Christchurch Hospital, PO Box 4345, Christchurch, New Zealand
| | | | | | | | | | | |
Collapse
|
41
|
Orleans-Lindsay JK, Barber LD, Prentice HG, Lowdell MW. Acute myeloid leukaemia cells secrete a soluble factor that inhibits T and NK cell proliferation but not cytolytic function--implications for the adoptive immunotherapy of leukaemia. Clin Exp Immunol 2001; 126:403-11. [PMID: 11737054 PMCID: PMC1906225 DOI: 10.1046/j.1365-2249.2001.01692.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2001] [Indexed: 01/07/2023] Open
Abstract
Evidence of an immune mediated graft-versus-leukaemia effect has led to the belief that T and NK cell based adoptive immunotherapy can constitute effective treatment for relapsed leukaemias. However, work on solid tumours has shown this strategy may be hampered, by an immune escape mechanism in which tumour secreted immunosuppressive factors compromise T and NK cell function. Indeed, acute myeloid leukaemia (AML) cells secrete immunosuppressive factors that block the synthesis of Th1 type cytokines in T cells. We demonstrate here that this immunosuppression, mediated by both HL60 AML cell line and primary AML blasts, inhibits T and NK cell proliferation but not cytolytic activity. Supernatants from HL60 cell line and primary AML blasts inhibited T cell proliferation to mitogenic and alloantigen stimulation but had no effect on cytolytic function. Similarly, the proliferation of NK cells to IL-2 and IL-15 stimulation was inhibited whilst their cytolytic function, shown by lysis of AML blasts, K562 and Daudi cells remained unaffected. The failure of T and NK cells to proliferate was not due to effector cell apoptosis. Indeed, removal of lymphocytes from the immunosuppressive environment partially restored their capacity to respond to mitogenic stimulation. T cells exposed to immunosuppressive supernatants did not increase expression of mitotic inhibitory proteins that arrest cell division, thereby ruling this out as a mechanism of operation for this immunosuppression. T cell expansion requires antigen stimulation, usually provided in the form of AML blasts, therefore our data suggest that NK cells may be more practical for the immunotherapy of AML.
Collapse
MESH Headings
- Cell Division
- Cytotoxicity, Immunologic
- Graft vs Leukemia Effect/immunology
- HL-60 Cells
- Humans
- Immunotherapy, Adoptive
- Interleukin-15/pharmacology
- Interleukin-2/pharmacology
- Isoantigens/administration & dosage
- K562 Cells
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Lymphocyte Activation
- Mitogens/pharmacology
- Suppressor Factors, Immunologic/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- J K Orleans-Lindsay
- Department of Haematology, Royal Free University College Medical School, London, UK
| | | | | | | |
Collapse
|
42
|
Buggins AG, Milojkovic D, Arno MJ, Lea NC, Mufti GJ, Thomas NS, Hirst WJ. Microenvironment produced by acute myeloid leukemia cells prevents T cell activation and proliferation by inhibition of NF-kappaB, c-Myc, and pRb pathways. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:6021-30. [PMID: 11698483 DOI: 10.4049/jimmunol.167.10.6021] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Tumors produce a variety of immunosuppressive factors which can prevent the proliferation and maturation of a number of normal hemopoietic cell types. We have investigated whether primary acute myeloid leukemia (AML) cells have an effect on normal T cell function and signaling. Tumor cell supernatant (TSN) from AML cells inhibited T cell activation and Th1 cytokine production and also prevented activated T cells from entering the cell cycle. These effects occurred in the absence of AML cell-T cell contact. We have demonstrated that AML TSN contained none of the immunosuppressors described to date, namely gangliosides, nitric oxide, TGF-beta, IL-10, vascular endothelial growth factor, or PGs. Furthermore, IL-2 did not overcome the block, despite normal IL-2R expression. However, the effect was overcome by preincubation with inhibitors of protein secretion and abolished by trypsinization, indicating that the active substance includes one or more proteins. To determine the mechanism of inhibition, we have studied many of the major pathways involved in T cell activation and proliferation. We show that nuclear translocation of NFATc and NF-kappaB are markedly reduced in T cells activated in the presence of primary AML cells. In contrast, calcium mobilization and activation of other signal transduction pathways, namely extracellular signal-regulated kinase1/2, p38, and STAT5 were unaffected, but activation of c-Jun N-terminal kinase 1/2 was delayed. Phosphorylation of pRb by cyclin-dependent kinase 6/4-cyclin D and of p130 did not occur and c-Myc, cyclin D3, and p107 were not induced, consistent with cell cycle inhibition early during the transition from G(0) to G(1). Our data indicate that TSN generated by AML cells induces T cell immunosuppression and provides a mechanism by which the leukemic clone could evade T cell-mediated killing.
Collapse
Affiliation(s)
- A G Buggins
- Department of Haematological Medicine, Leukaemia Sciences, Guy's, King's and St. Thomas' School of Medicine, Rayne Institute, London, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
43
|
Narita M, Takahashi M, Liu A, Nikkuni K, Furukawa T, Toba K, Koyama S, Takai K, Sanada M, Aizawa Y. Leukemia blast-induced T-cell anergy demonstrated by leukemia-derived dendritic cells in acute myelogenous leukemia. Exp Hematol 2001; 29:709-19. [PMID: 11378266 DOI: 10.1016/s0301-472x(01)00636-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVE To elucidate the mechanism of immunologic escape of leukemia cells and establish an effective anti-leukemia immunotherapy, we attempted to generate dendritic cells from leukemia cells in patients with acute myelogenous leukemia (AML). Using these leukemia-derived dendritic cells, we investigated leukemia cell-associated T-cell anergy. MATERIALS AND METHODS Leukemia cells of 30 patients with AML were cultured with granulocyte-macrophage colony-stimulating factor, interleukin-4, and tumor necrosis factor-alpha. Cultured leukemia cells were evaluated for antigen-presenting ability by mixed leukocyte culture (MLC). Normal lymphocytes, which were cocultured with leukemia blasts in the first MLC, were cultured with leukemia-derived dendritic cells in the second MLC. RESULTS In cultures of leukemia cells from 21 of 30 patients examined, cells with stellate morphology and cell fractions with CD1a(+) and/or CD83(+) were present. Autologous MLC using lymphocytes obtained in remission phase as responders as well as allogeneic MLC demonstrated antigen-presenting ability in leukemia-derived dendritic cells. Leukemia cells of FAB-M0, M1, M2, M3, or M6 morphology/phenotype gave rise to dendritic cells as well as leukemia cells of M5. The leukemic origin of dendritic cells was suggested by in situ hybridization. By coculture with CD80(-) leukemia blasts, the response of normal lymphocytes to leukemia-derived dendritic cells cultured from the same individual as that of leukemia blasts was markedly reduced, compared with the lymphocytes cultured with leukemia blasts from a different individual as leukemia blasts. CONCLUSIONS Escape of leukemia cells from anti-leukemia immunity may be associated with T-cell anergy caused by leukemia blasts. The results of the present study suggest that leukemia-derived dendritic cells can be applied efficiently in anti-leukemia immunotherapy.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Blast Crisis/immunology
- Clonal Anergy/immunology
- Coculture Techniques
- Colony-Stimulating Factors/pharmacology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Female
- Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology
- Humans
- Interleukin-4/pharmacology
- Karyotyping
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukocyte Count
- Lymphocyte Culture Test, Mixed
- Male
- Middle Aged
- T-Lymphocytes/immunology
- Tumor Cells, Cultured
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- M Narita
- School of Health Sciences, Faculty of Medicine, Niigata University, Niigata, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Affiliation(s)
- D C Reid
- Department of Haematology, Northwick Park Hospital, Watford Road, Harrow HA1 3UJ, UK.
| |
Collapse
|