101
|
Evolution of peripheral blood T lymphocyte subsets after allogenic or autologous hematopoietic stem cell transplantation. Immunobiology 2014; 219:611-8. [PMID: 24721705 DOI: 10.1016/j.imbio.2014.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 03/13/2014] [Accepted: 03/14/2014] [Indexed: 01/10/2023]
Abstract
With the aim to search for differences in T cell reconstitution after allogenic or autologous hematopoietic stem cell transplantation (HSCT), we characterized peripheral blood T-cell subsets by means of flow cytometry, in adult patients who had undergone either allogenic (n=23) or autologous (n=29) HSCT for the treatment of hematological malignancies. The patients were followed every 3 months for 21 months after HSCT. Compared to healthy controls (n=20 blood donors), the two transplanted groups displayed (i) a CD4 lymphopenia, (ii) a low percentage of naive T cells, (iii) high percentages of memory T cells and of activated T cells (HLA-DR+, CD25+) and high percentages of CD4 T cells with a high expression of CD25. The levels of TRECs (TCR rearrangement excision circles) were not significantly different between the two groups. In total, the differences of the nature and the speed of T lymphocyte reconstitution observed between the two patient groups were minor. This leads us to conclude that in allografted patients, lymphocyte activation as well as many other disturbances of subpopulations of peripheral blood lymphocytes are probably not related to the allogenicity of the graft, but are due to the expansion of T cells transfused with HSC and slow differentiation of T lymphocytes in the thymus progressively colonized by bone marrow-derived T-cell precursors.
Collapse
|
102
|
Bastidas S, Graw F, Smith MZ, Kuster H, Günthard HF, Oxenius A. CD8+T Cells Are Activated in an Antigen-Independent Manner in HIV-Infected Individuals. THE JOURNAL OF IMMUNOLOGY 2014; 192:1732-44. [DOI: 10.4049/jimmunol.1302027] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
103
|
MacVittie TJ, Bennett AW, V Cohen M, Farese AM, Higgins A, Hankey KG. Immune cell reconstitution after exposure to potentially lethal doses of radiation in the nonhuman primate. HEALTH PHYSICS 2014; 106:84-96. [PMID: 24276552 DOI: 10.1097/hp.0b013e3182a2a9b2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Delayed immune reconstitution remains a major cause of morbidity associated with myelosuppression induced by cytotoxic therapy or myeloablative conditioning for stem cell transplant, as well as potentially lethal doses of total- or partial-body irradiation. Restoration of a functional immune cell repertoire requires hematopoietic stem cell reconstitution for all immune cells and effective thymopoiesis for T cell recovery. There are no medical countermeasures available to mitigate damage consequent to high-dose, potentially lethal irradiation, and there are no well characterized large animal models of prolonged immunosuppression to assess efficacy of potential countermeasures. Herein, the authors describe a model of T and B cell reconstitution following lethal doses of partial-body irradiation with 5% bone marrow sparing that includes full exposure of the thymus. Rhesus macaques (n = 31 male, 5.5-11.3 kg body weight) were exposed to midline tissue doses of 9.0-12.0 Gy using 6 MV LINAC-derived photons at a dose rate of 0.80 Gy min, sparing approximately 5% of bone marrow (tibiae, ankles, and feet). All animals received medical management and were monitored for myeloid and lymphoid suppression and recovery through 180 d post-exposure. Myeloid recovery was assessed by neutrophil and platelet-related hematological parameters. Reconstitution of B and T cell subsets was assessed by flow cytometric immunophenotyping, and recent thymic emigrants were identified by RT-PCR of T cell receptor excision circles. Mortality was recorded through 180 d post-exposure. Acute myelo-suppression was characterized by severe neutropenia and thrombocytopenia, followed by recovery 30-60 d post-exposure. Total T (CD3+) and B (CD20+) cells were reduced significantly following exposure and exhibited differential recovery patterns post-exposure. Both CD4+ and CD8+ subsets of naïve T cells and total CD4+ T cell counts remained significantly lower than baseline through 180 d post-exposure. The failure of recent thymic emigrants and naïve T cell subsets to recover to normal baseline values reflects the severe radiation effects on the recovery of marrow-derived stem and early thymic progenitor cells, their mobilization and seeding of receptive thymic niches, and slow endogenous thymic regeneration.
Collapse
Affiliation(s)
- Thomas J MacVittie
- *University of Maryland, School of Medicine, Dept. of Radiation Oncology, Baltimore, MD; †Integrated Research Facility, Frederick, MD; ‡Naval Medical Research Center, Silver Spring, MD
| | | | | | | | | | | |
Collapse
|
104
|
Blanche S, Scott-Algara D, Le Chenadec J, Didier C, Montange T, Avettand-Fenoel V, Rouzioux C, Melard A, Viard JP, Dollfus C, Bouallag N, Warszawski J, Buseyne F. Naive T Lymphocytes and Recent Thymic Emigrants Are Associated With HIV-1 Disease History in French Adolescents and Young Adults Infected in the Perinatal Period: The ANRS-EP38-IMMIP Study. Clin Infect Dis 2013; 58:573-87. [DOI: 10.1093/cid/cit729] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
105
|
Human proT-cells generated in vitro facilitate hematopoietic stem cell-derived T-lymphopoiesis in vivo and restore thymic architecture. Blood 2013; 122:4210-9. [PMID: 24215033 DOI: 10.1182/blood-2012-12-472803] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is followed by a period of immune deficiency due to a paucity in T-cell reconstitution. Underlying causes are a severely dysfunctional thymus and an impaired production of thymus-seeding progenitors in the host. Here, we addressed whether in vitro-derived human progenitor T (proT)-cells could not only represent a source of thymus-seeding progenitors, but also able to influence the recovery of the thymic microenvironment. We examined whether co-transplantation of in vitro-derived human proT-cells with hematopoietic stem cells (HSCs) was able to facilitate HSC-derived T-lymphopoiesis posttransplant. A competitive transfer approach was used to define the optimal proT subset capable of reconstituting immunodeficient mice. Although the 2 subsets tested (proT1, CD34(+)CD7(+)CD5(-); proT2, CD34(+)CD7(+)CD5(+)) showed thymus engrafting function, proT2-cells exhibited superior engrafting capacity. Based on this, when proT2-cells were coinjected with HSCs, a significantly improved and accelerated HSC-derived T-lymphopoiesis was observed. Furthermore, we uncovered a potential mechanism by which receptor activator of nuclear factor κb (RANK) ligand-expressing proT2-cells induce changes in both the function and architecture of the thymus microenvironment, which favors the recruitment of bone marrow-derived lymphoid progenitors. Our findings provide further support for the use of Notch-expanded progenitors in cell-based therapies to aid in the recovery of T-cells in patients undergoing HSCT.
Collapse
|
106
|
Gress RE, Miller JS, Battiwalla M, Bishop MR, Giralt SA, Hardy NM, Kröger N, Wayne AS, Landau DA, Wu CJ. Proceedings from the National Cancer Institute's Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation: Part I. Biology of relapse after transplantation. Biol Blood Marrow Transplant 2013; 19:1537-45. [PMID: 24018395 PMCID: PMC3922045 DOI: 10.1016/j.bbmt.2013.08.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 08/30/2013] [Indexed: 12/01/2022]
Abstract
In the National Cancer Institute's Second Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation, the Scientific/Educational Session on the Biology of Relapse discussed recent advances in understanding some of the host-, disease-, and transplantation-related contributions to relapse, emphasizing concepts with potential therapeutic implications. Relapse after hematopoietic stem cell transplantation (HSCT) represents tumor escape, from the cytotoxic effects of the conditioning regimen and from immunologic control mediated by reconstituted lymphocyte populations. Factors influencing the biology of the therapeutic graft-versus-malignancy (GVM) effect-and relapse-include conditioning regimen effects on lymphocyte populations and homeostasis, immunologic niches, and the tumor microenvironment; reconstitution of lymphocyte populations and establishment of functional immune competence; and genetic heterogeneity within the malignancy defining potential for clonal escape. Recent developments in T cell and natural killer cell homeostasis and reconstitution are reviewed, with implications for prevention and treatment of relapse, as is the application of modern genome sequencing to defining the biologic basis of GVM, clonal escape, and relapse after HSCT.
Collapse
Affiliation(s)
- Ronald E Gress
- Experimental Transplantation Immunology Branch, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Ersvaer E, Hampson P, Wendelbo Ø, Lord JM, Gjertsen BT, Bruserud Ø. Circulating T cells in patients with untreated acute myelogenous leukemia are heterogeneous and can be activated through the CD3/TCR complex. Hematology 2013; 12:199-207. [PMID: 17558695 DOI: 10.1080/10245330701255163] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
T lymphocyte defects may contribute to the immune insufficiency seen in acute myelogenous leukemia (AML). We therefore characterized the T cell system for untreated AML patients. T lymphocyte subsets were analyzed by flow cytometry for 45 AML patients. The in vitro interferon-gamma (IFNgamma) release in response to stimulation with anti-CD3 plus anti-CD28 in the presence of autologous AML cells was examined for 31 patients. The majority of circulating lymphocytes were CD3(+)T cells, and CD19(+)B cells usually constituted < 10% of the lymphocytes. Most T cells expressed the alphabeta T cell receptor (TCRalphabeta(+)), and only a minority of the cells was TCRgammadelta(+). Both CD4(+) and CD8(+)T cells were detected, the CD4:CD8 ratio showed a wide variation but was generally >1.0. The majority of CD4(+) and CD8(+)T cells were CD45RA(+) cells. The T cells could be stimulated to release IFNgamma in response to anti-CD3 plus anti-CD28 ligation even in the presence of excess autologous AML blasts, and for a subset of patients (6 of 27) these IFNgamma levels could be further increased by the novel protein kinase C (PKC) agonist PEP005. In conclusion, circulating T cells in patients with untreated AML are mainly CD4(+) or CD8(+) TCRalphabeta(+); both CD45RA(+) and CD45R0(+) can be detected, and these cells can be activated through the CD3/TCR complex even in the presence of excess AML cells. For a subset of patients T cell responsiveness can be further increased by targeting PKC and these data therefore suggest that T cell function is not inhibited in AML patients.
Collapse
Affiliation(s)
- Elisabeth Ersvaer
- University of Bergen and Haukeland University Hospital, Institute of Medicine, Bergen, Norway.
| | | | | | | | | | | |
Collapse
|
108
|
Ribeiro D, Melão A, Barata JT. IL-7R-mediated signaling in T-cell acute lymphoblastic leukemia. Adv Biol Regul 2013; 53:211-222. [PMID: 23234870 DOI: 10.1016/j.jbior.2012.10.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 10/09/2012] [Indexed: 06/01/2023]
Abstract
Interleukin-7 (IL-7), a cytokine produced in the bone marrow, thymus and other organs, is mandatory for normal human T-cell development and peripheral homeostasis. Different studies, including phase I clinical trials, have indicated the potential therapeutic value of recombinant IL-7 in the context of anti-cancer immunotherapy and as a booster of immune reconstitution. However, the two main pathways activated by IL-7, JAK/STAT5 and PI3K/Akt/mTOR, have both been implicated in cancer and there is considerable evidence that IL-7 and its receptor (IL-7R), formed by IL-7Rα (encoded by IL7R) and γc, may partake in T-cell acute lymphoblastic leukemia (T-ALL) development. In this context, the most compelling data comes from recent studies demonstrating that around 10% of T-ALL patients display IL7R gain-of-function mutations leading, in most cases, to disulfide bond-dependent homodimerization of two mutant receptors and consequent constitutive activation of downstream signaling, with ensuing cell transformation in vitro and tumorigenic ability in vivo. Here, we review the data on the involvement of IL-7 and IL-7R in T-ALL, further discussing the peculiarities of IL-7R-mediated signaling in human leukemia T-cells that may be of therapeutic value, namely regarding the potential use of PI3K and mTOR pharmacological inhibitors.
Collapse
Affiliation(s)
- Daniel Ribeiro
- Instituto de Medicina Molecular, Faculdade de Medicina, Unversidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | | | | |
Collapse
|
109
|
Xu X, Yu Y, Wang Z, Zhu T, Wang Y, Zhu J, Chen Z, He Y, Ju L, Li Y. Mass spectrometry based phospholipidomics of mammalian thymus and leukemia patients: implication for function of iNKT cells. Anal Bioanal Chem 2013; 405:5267-78. [PMID: 23595640 DOI: 10.1007/s00216-013-6923-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 03/06/2013] [Accepted: 03/15/2013] [Indexed: 10/27/2022]
Abstract
In previous studies phospholipids have been proved to be involved in biochemical, physiological, and pathological processes. As a special class of phospholipids, peroxisome-derived lipids (PDLs) have been proved to be potential ligands of invariant natural killer T (iNKT) cells in recent studies. Here, on the basis of phospholipidomics, we focused on the relative quantity of PDLs extracted from mammalian thymus or bone marrow using electrospray ionization mass spectrometry (MS). In phospholipid analysis, we identified 12 classes of phospholipids and accounted for their relative quantities by comparing their relative abundances in the MS(1) map. Our results show that PDLs are present in mammalian thymus as well as mouse spleen and liver. Interestingly, the relative quantity of PDLs extracted from human acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) bone marrows is higher than that extracted from bone marrow of healthy donors. Our results may help to explain the close correlation between PDLs and iNKT cell function in thymus, spleen, liver, and especially in leukemia patients. We think that our phospholipidomics work may reveal a function of iNKT cells.
Collapse
Affiliation(s)
- Xiukun Xu
- Laboratory of Cellular and Molecular Tumor Immunology, Institutes of Biology and Medical Sciences, Jiangsu Laboratory of Infection Immunity, Soochow University, 199 Ren-Ai Road, Suzhou 215123, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Nars MS, Kaneno R. Immunomodulatory effects of low dose chemotherapy and perspectives of its combination with immunotherapy. Int J Cancer 2012; 132:2471-8. [PMID: 22927096 DOI: 10.1002/ijc.27801] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/29/2012] [Accepted: 08/16/2012] [Indexed: 02/06/2023]
Abstract
Given that cancer is one of the main causes of death worldwide, many efforts have been directed toward discovering new treatments and approaches to cure or control this group of diseases. Chemotherapy is the main treatment for cancer; however, a conventional schedule based on maximum tolerated dose (MTD) shows several side effects and frequently allows the development of drug resistance. On the other side, low dose chemotherapy involves antiangiogenic and immunomodulatory processes that help host to fight against tumor cells, with lower grade of side effects. In this review, we present evidence that metronomic chemotherapy, based on the frequent administration of low or intermediate doses of chemotherapeutics, can be better than or as efficient as MTD. Finally, we present some data indicating that noncytotoxic concentrations of antineoplastic agents are able to both up-regulate the immune system and increase the susceptibility of tumor cells to cytotoxic T lymphocytes. Taken together, data from the literature provides us with sufficient evidence that low concentrations of selected chemotherapeutic agents, rather than conventional high doses, should be evaluated in combination with immunotherapy.
Collapse
Affiliation(s)
- Mariana S Nars
- Department of Microbiology and Immunology, Institute of Biosciences, UNESP-Univ Estadual Paulista, Botucatu, São Paulo, Brazil
| | | |
Collapse
|
111
|
Le Saout C, Lane HC, Catalfamo M. The role of cytokines in the pathogenesis and treatment of HIV infection. Cytokine Growth Factor Rev 2012; 23:207-14. [PMID: 22738931 PMCID: PMC3726258 DOI: 10.1016/j.cytogfr.2012.05.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
HIV immune activation plays an important role in the immunopathogenesis of the disease. The mechanisms driving this immune activation are partially defined and likely are the result of multiple factors. The introduction of combination antiretroviral therapy (cART) has improved the life expectancy of HIV infected individuals, however there is evidence that in the setting of "undetectable" HIV-RNA plasma levels, there is some level of persistent immune activation in these patients. A better understanding of the immune activation pathways should be of value in developing complementary therapies to restore the immune systems of patients with HIV infection. This review discusses the cytokine mediated pathways of immune activation of the CD4 and CD8 T cell pools during HIV infection.
Collapse
Affiliation(s)
- Cecile Le Saout
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Drive, Bldg 10 Room 11B07, Bethesda, MD 20892-1360, Phone: 301-443-8313, FAX: 301-402-4097
| | - H Clifford Lane
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Drive, Rm. 4-1479, MSC 1460, Bethesda, MD 20892-1360, Office: 301-496-6572 I, Fax: 301-480-5560
| | - Marta Catalfamo
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Drive, Bldg 10 Room 11B07, Bethesda, MD 20892-1360, Phone: 301-496-5309, FAX: 301-402-4097
| |
Collapse
|
112
|
Abstract
Control of the alloimmune response requires elimination and/or suppression of alloreactive immune cells. Lymphodepleting induction therapies are increasingly used to accomplish this goal, both as part of tolerance induction protocols or to reduce the requirements for maintenance immunosuppression in the peritransplant setting. However, it is well recognized that lymphopenia induces compensatory proliferation of immune cells, generally termed ``homeostatic proliferation,'' which favors the emergence of memory T cells. Paradoxically therefore, the result may be a situation that favors graft rejection and/or makes tolerance difficult to achieve or sustain. Yet all depletion is not alike, particularly with respect to the timing of reconstitution and the types of cells that repopulate the host. Thus, to design more effective induction strategies it is important to understand the homeostatic mechanisms, which exist to maintain a balanced repertoire of naïve and memory T and B cells in the periphery and how they respond to lymphodepletion. Here we will review the biology of homeostatic proliferation stimulated by lymphopenia, the effects of specific depleting agents on reconstitution of the T- and B-cell immune repertoire, drawing from both from animal models and human experience, and potential strategies to enhance allodepletion while minimizing the adverse effects of homeostatic proliferation.
Collapse
Affiliation(s)
- N K Tchao
- Immune Tolerance Network, San Francisco, CA, USA
| | | |
Collapse
|
113
|
Morre M, Beq S. Interleukin-7 and immune reconstitution in cancer patients: a new paradigm for dramatically increasing overall survival. Target Oncol 2012; 7:55-68. [PMID: 22383042 PMCID: PMC3304058 DOI: 10.1007/s11523-012-0210-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 01/16/2012] [Indexed: 12/27/2022]
Abstract
Although great effort is being expended in the development of cancer immunotherapies, it is surprising that global lymphopenia and its various dimensions are not being systematically assessed in cancer patients. The incident pathologies associated with various immunosuppressed conditions such as those found in HIV infection have taught us that measuring various T cell populations including CD4 provides the clinician with a reliable measure for gauging the risk of cancer and opportunistic infections. Importantly, recent data emphasize the key link between lymphocyte T cell counts and overall survival in cancer patients receiving chemotherapy. Treatment of immunocompromised patients with interleukin-7 (IL-7), a critical growth and homeostatic factor for T cells, has been shown to produce a compelling profile of T cell reconstitution. The clinical results of this investigational therapy confirm data obtained from numerous preclinical studies and demonstrate the long-term stability of this immune reconstitution, not only on CD4 but also on CD8 T cells, involving recent thymic emigrants as well as naive, memory, and central memory T cells. Furthermore, IL-7 therapy also contributes to restoration of a broadened diversity of the T cell repertoire as well as to migration of these cells to lymph nodes and tissues. All these properties support the initiation of new clinical studies aimed at reconstituting the immune system of cancer patients before or immediately after chemotherapy in order to demonstrate a potentially profound increase in overall survival.
Collapse
Affiliation(s)
- Michel Morre
- Cytheris S.A., 175 rue Jean Jacques Rousseau, 92130, Issy-Les-Moulineaux, France.
| | | |
Collapse
|
114
|
Abstract
In summary, immunizations in special populations require understanding the underlying disease and how it might affect the immune system's ability to mount an antibody response to vaccines or predispose certain patient populations to developing certain serious infections. There is still a great need for research on the optimal timing of vaccines after transplants, how to assess protection and development of a protective antibody response after immunization, and whether certain groups (eg, HIV) need to be revaccinated after a certain amount of time if their antibody levels decline. In addition, there are limited data on efficacy of the newer vaccines in these special patient populations, which also requires further investigation.
Collapse
Affiliation(s)
- Michael A Miller
- Department of Pediatric Infectious Diseases and Immunology, University of Florida, Jacksonville, 32209, USA
| | | |
Collapse
|
115
|
Ruggiero A, Battista A, Coccia P, Attinà G, Riccardi R. How to manage vaccinations in children with cancer. Pediatr Blood Cancer 2011; 57:1104-8. [PMID: 21953691 DOI: 10.1002/pbc.23333] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 08/08/2011] [Indexed: 11/07/2022]
Abstract
The optimal use of routine childhood immunizations in children with malignancy is still a matter of debate. Despite their higher risk of contracting vaccine preventable diseases and of suffering important complications, there is little understanding of the magnitude of the possible benefit of administering active immunization in this population due to a paucity of clinical trial data. Our review focuses on the management of children with cancer and offers some suggestions regarding their vaccination schedules.
Collapse
Affiliation(s)
- Antonio Ruggiero
- Pediatric Oncology Division, A Gemelli Hospital, Catholic University, Rome, Italy.
| | | | | | | | | |
Collapse
|
116
|
Generation of CD19-chimeric antigen receptor modified CD8+ T cells derived from virus-specific central memory T cells. Blood 2011; 119:72-82. [PMID: 22031866 DOI: 10.1182/blood-2011-07-366419] [Citation(s) in RCA: 162] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The adoptive transfer of donor T cells that have been genetically modified to recognize leukemia could prevent or treat leukemia relapse after allogeneic HSCT (allo-HSCT). However, adoptive therapy after allo-HSCT should be performed with T cells that have a defined endogenous TCR specificity to avoid GVHD. Ideally, T cells selected for genetic modification would also have the capacity to persist in vivo to ensure leukemia eradication. Here, we provide a strategy for deriving virus-specific T cells from CD45RA(-)CD62L(+)CD8(+) central memory T (T(CM)) cells purified from donor blood with clinical grade reagents, and redirect their specificity to the B-cell lineage marker CD19 through lentiviral transfer of a gene encoding a CD19-chimeric Ag receptor (CAR). Virus-specific T(CM) were selectively transduced by exposure to the CD19 CAR lentivirus after peptide stimulation, and bi-specific cells were subsequently enriched to high purity using MHC streptamers. Activation of bi-specific T cells through the CAR or the virus-specific TCR elicited phosphorylation of downstream signaling molecules with similar kinetics, and induced comparable cytokine secretion, proliferation, and lytic activity. These studies identify a strategy for tumor-specific therapy with CAR-modified T cells after allo-HSCT, and for comparative studies of CAR and TCR signaling.
Collapse
|
117
|
Dudakov JA, van den Brink MRM. Greater than the sum of their parts: combination strategies for immune regeneration following allogeneic hematopoietic stem cell transplantation. Best Pract Res Clin Haematol 2011; 24:467-76. [PMID: 21925100 DOI: 10.1016/j.beha.2011.05.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cytoreductive conditioning regimes designed to allow for successful allogeneic hematopoietic stem cell transplantation (allo-HSCT) paradoxically are also detrimental to recovery of the immune system in general but lymphopoiesis in particular. Post-transplant immune depletion is particularly striking within the T cell compartment which is exquisitely sensitive to negative regulation, evidenced by the profound decline in thymic function with age. As a consequence, regeneration of the immune system remains a significant unmet clinical need. Over the past decade studies have revealed several promising therapeutic strategies to address ineffective lymphopoiesis and post-transplant immune deficiency. These include the use of cytokines such as IL-7, IL-12 and IL-15; growth factors and hormones like keratinocyte growth factor (KGF), insulin-like growth factor (IGF)-1 and growth hormone (GH); adoptive transfer of ex vivo-generated precursor T cells (pre-T) and sex steroid ablation (SSA). Moreover, recently several novel approaches have been proposed to generate whole thymii ex vivo using stem cell technologies and bioscaffolds. Increasingly, however, when transferred to the clinic, these strategies alone are not sufficient to restore thymopoiesis in all patients leading to the potential of combination strategies as a way to reign in non-responders. Synergistic enhancement in combination may be due to differential targets may therefore be effective in improving clinical outcomes in the transplant settings as well as in other lymphopenic states induced by high dose chemotherapy/radiation therapy or HIV, and may also be useful in improving responses to vaccination and augmenting anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Jarrod A Dudakov
- Department of Immunology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | | |
Collapse
|
118
|
Mackall CL, Fry TJ, Gress RE. Harnessing the biology of IL-7 for therapeutic application. Nat Rev Immunol 2011; 11:330-42. [PMID: 21508983 DOI: 10.1038/nri2970] [Citation(s) in RCA: 432] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Interleukin-7 (IL-7) is required for T cell development and for maintaining and restoring homeostasis of mature T cells. IL-7 is a limiting resource under normal conditions, but it accumulates during lymphopaenia, leading to increased T cell proliferation. The administration of recombinant human IL-7 to normal or lymphopenic mice, non-human primates and humans results in widespread T cell proliferation, increased T cell numbers, modulation of peripheral T cell subsets and increased T cell receptor repertoire diversity. These effects raise the prospect that IL-7 could mediate therapeutic benefits in several clinical settings. This Review summarizes the biology of IL-7 and the results of its clinical use that are available so far to provide a perspective on the opportunities for clinical application of this cytokine.
Collapse
Affiliation(s)
- Crystal L Mackall
- Immunology Section, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
119
|
Michieli M, Mazzucato M, Tirelli U, De Paoli P. Stem Cell Transplantation for Lymphoma Patients with HIV Infection. Cell Transplant 2011; 20:351-70. [DOI: 10.3727/096368910x528076] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The advent of Highly Active Antiretroviral Therapy (HAART) has radically changed incidence characteristics and prognosis of HIV-positive patients affected by lymphomas. At this time there is consensus in the literature that, in first line, HIV-positive patients should always be treated with curative intent preferentially following the same approach used in the HIV-negative counterpart. On the contrary, an approach of salvage therapy in HIV-positive lymphomas is still a matter of debate given that for a wide range of relapsed or resistant HIV-negative Hodgkin's disease (HD) and non-Hodgkin lymphoma (NHL) patients, autologous peripheral or allogeneic stem cell transplantation are among the established options. In the pre-HAART era, therapeutic options derived from pioneering experiences gave only anecdotal success, either when transplantation was used to cure lymphomas or to improve HIV infection itself. Concerns relating to the entity, quality, and kinetics of early and late immune reconstitutions and the possible worsening of underlying viroimmunological conditions were additional obstacles. Currently, around 100 relapsed or resistant HIV-positive lymphomas have been treated with an autologous peripheral stem cell transplantation (APSCT) in the HAART era. Published data compared favorably with any previous salvage attempt showing a percentage of complete remission ranging from 48% to 90%, and overall survival ranging from 36% to 85% at median follow-up approaching 3 years. However, experiences are still limited and have given somewhat confounding indications, especially concerning timing and patients' selection for APSCT and feasibility and outcome for allogeneic stem cell transplant. Moreover, little data exist on the kinetics of immunological reconstitution after APSCT or relevant to the outcome of HIV infection. The aim of this review is to discuss current knowledge of the role of allogeneic and autologous stem cell transplantation as a modality in the cure of HIV and hemopoietic cancer patients. Several topics dealing with practical aspects concerning the management of APSCT in HIV-positive patients, including patient selection, timing of transplant, conditioning regimen, and relapse or nonrelapse mortality, are discussed. Data relating to the effects of mobilization and transplantation on virological parameters and pre- and posttransplant immune reconstitution are reviewed. Finally, in this review, we examine several ethical and legal issues relative to banking infected or potentially infected peripheral blood stem cells and we describe our experience and strategies to protect positive and negative donors/recipients and the health of caretakers.
Collapse
Affiliation(s)
- Mariagrazia Michieli
- Cell Therapy and High Dose Chemotherapy Unit, Centro di Riferimento Oncologico, CRO IRCCS, Aviano, Italy
| | - Mario Mazzucato
- Stem Cell Collection and Processing Unit, Centro di Riferimento Oncologico, CRO IRCCS, Aviano, Italy
| | - Umberto Tirelli
- Medical Oncology A, Centro di Riferimento Oncologico, CRO IRCCS, Aviano, Italy
| | - Paolo De Paoli
- Scientific Directorate, Centro di Riferimento Oncologico, CRO IRCCS, Aviano, Italy
| |
Collapse
|
120
|
Lymphocyte subpopulation and dendritic cell phenotyping during antineoplastic therapy in human solid tumors. Clin Exp Med 2010; 11:199-210. [PMID: 21161672 DOI: 10.1007/s10238-010-0120-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Accepted: 10/29/2010] [Indexed: 12/21/2022]
Abstract
Patients with cancer show variable levels of immunosuppression at the time of the presentation, and cytotoxic antineoplastic therapy is the primary contributor to the clinical immunodeficiency often observed during the course of the disease. In both hematological and solid tumors, this phenomenon is primarily related to the T-cell depletion associated with inhibition of dendritic cell ability to induce both primary and secondary T- and B-cell responses. Complete restoration of immunocompetence following antineoplastic therapy implicates the progressive recovery of various cell subpopulations, and it is a complex process that also depends on the type, the dose, the scheduling, and the associations of the employed drugs. In the era of target therapies, several antiangiogenic drugs are increasingly used in combination with standard chemotherapy in the treatment of advanced solid tumors. Their clinical efficacy has been recently related not only to the specific antiangiogenic properties but also to an indirect hypothetical effect on the host immune system. In the present work, we have reviewed the most recent information regarding (1) the capacity of standard antineoplastic therapy to induce and maintain an immunodeficiency in patients with solid tumors and (2) the influence of the antiangiogenic treatment in association with standard chemotherapy on lymphocyte and dendritic cell subsets and the possible resulting additional antitumor mechanism.
Collapse
|
121
|
van Tilburg CM, van Gent R, Bierings MB, Otto SA, Sanders EAM, Nibbelke EE, Gaiser JF, Janssens-Korpela PL, Wolfs TFW, Bloem AC, Borghans JAM, Tesselaar K. Immune reconstitution in children following chemotherapy for haematological malignancies: a long-term follow-up. Br J Haematol 2010; 152:201-10. [PMID: 21114483 DOI: 10.1111/j.1365-2141.2010.08478.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Modern intensive chemotherapy for childhood haematological malignancies has led to high cure rates, but has detrimental effects on the immune system. There is little knowledge concerning long-term recovery of the adaptive immune system. Here we studied the long-term reconstitution of the adaptive immune system in 31 children treated for haematological malignancies between July 2000 and October 2006. We performed detailed phenotypical and functional analyses of the various B and T cell subpopulations until 5 years after chemotherapy. We show that recovery of newly-developed transitional B cells and naive B and T cells occurred rapidly, within months, whereas recovery of the different memory B and T cell subpopulations was slower and incomplete, even after 5 years post-chemotherapy. The speed of B and T cell recovery was age-independent, despite a significant contribution of the thymus to T cell recovery. Plasmablast B cell levels remained above normal and immunoglobulin levels normalised within 1 week. Functional T cell responses were normal, even within the first year post-chemotherapy. This study shows that after intensive chemotherapy for haematological malignancies in children, numbers of several memory B and T cell subpopulations were decreased on the long term, while functional T cell responses were not compromised.
Collapse
Affiliation(s)
- Cornelis M van Tilburg
- Department of Paediatric Haematology/Oncology, University Medical Center Utrecht, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Abstract
PURPOSE OF REVIEW The thymus provides a unique and essential microenvironment for T-cell precursors to develop into mature functionally competent T lymphocytes. Ageing causes architectural changes in the thymus resulting in a loss of thymic epithelial space required for thymopoiesis - a process known as thymic involution. Additionally, cytoablative regimens used to treat malignancies also destroy thymic architecture. The net result of both processes is diminished thymic output and function that may lead to impaired immunity. Thus, immunocompromised individuals would benefit from strategies aimed at enhancing T-cell reconstitution. RECENT FINDINGS Here we discuss strategies such as the use of sex steroid ablation, keratinocyte growth factor, interleukin-7, and in-vitro-generated progenitor T cells as candidates for restoring T-cell immunity. Using various animal models of ageing or hematopoietic stem cell transplantation, these strategies have been shown to restore thymic architecture and cellularity, resulting in increased output and T-cell function in the periphery. SUMMARY These candidate approaches are currently being tested in clinical trials, with preliminary evidence showing encouraging effects on T-cell reconstitution. Nevertheless, although these strategies show clear promise in animal models, and in early human trials, further data are needed to determine their efficacy in patients.
Collapse
|
123
|
Combination of intensive chemotherapy and anticancer vaccines in the treatment of human malignancies: the hematological experience. J Biomed Biotechnol 2010; 2010:692097. [PMID: 20625438 PMCID: PMC2896720 DOI: 10.1155/2010/692097] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 03/20/2010] [Indexed: 12/21/2022] Open
Abstract
In vitro studies have demonstrated that cancer-specific T cell cytotoxicity can be induced both ex vivo and in vivo, but this therapeutic strategy should probably be used as an integrated part of a cancer treatment regimen. Initial chemotherapy should be administered to reduce the cancer cell burden and disease-induced immune defects. This could be followed by autologous stem cell transplantation that is a safe procedure including both high-dose disease-directed chemotherapy and the possibility for ex vivo enrichment of the immunocompetent graft cells. The most intensive conventional chemotherapy and stem cell transplantation are used especially in the treatment of aggressive hematologic malignancies; both strategies induce T cell defects that may last for several months but cancer-specific T cell reactivity is maintained after both procedures. Enhancement of anticancer T cell cytotoxicity is possible but posttransplant vaccination therapy should probably be combined with optimalisation of immunoregulatory networks. Such combinatory regimens should be suitable for patients with aggressive hematological malignancies and probably also for other cancer patients.
Collapse
|
124
|
Immunosénescence et infections, mythe ou réalité ? Med Mal Infect 2010; 40:307-18. [DOI: 10.1016/j.medmal.2009.12.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Accepted: 12/10/2009] [Indexed: 01/06/2023]
|
125
|
Lineage-specific T-cell reconstitution following in vivo CD4+ and CD8+ lymphocyte depletion in nonhuman primates. Blood 2010; 116:748-58. [PMID: 20484087 DOI: 10.1182/blood-2010-01-263814] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Many features of T-cell homeostasis in primates are still unclear, thus limiting our understanding of AIDS pathogenesis, in which T-cell homeostasis is lost. Here, we performed experiments of in vivo CD4(+) or CD8(+) lymphocyte depletion in 2 nonhuman primate species, rhesus macaques (RMs) and sooty mangabeys (SMs). Whereas RMs develop AIDS after infection with simian immunodeficiency virus (SIV), SIV-infected SMs are typically AIDS-resistant. We found that, in both species, most CD4(+) or CD8(+) T cells in blood and lymph nodes were depleted after treatment with their respective antibodies. These CD4(+) and CD8(+) lymphocyte depletions were followed by a largely lineage-specific CD4(+) and CD8(+) T-cell proliferation, involving mainly memory T cells, which correlated with interleukin-7 plasma levels. Interestingly, SMs showed a faster repopulation of naive CD4(+) T cells than RMs. In addition, in both species CD8(+) T-cell repopulation was faster than that of CD4(+) T cells, with CD8(+) T cells reconstituting a normal pool within 60 days and CD4(+) T cells remaining below baseline levels up to day 180 after depletion. While this study revealed subtle differences in CD4(+) T-cell repopulation in an AIDS-sensitive versus an AIDS-resistant species, such differences may have particular relevance in the presence of active SIV repli cation, where CD4(+) T-cell destruction is chronic.
Collapse
|
126
|
Goldberg GL, Dudakov JA, Reiseger JJ, Seach N, Ueno T, Vlahos K, Hammett MV, Young LF, Heng TSP, Boyd RL, Chidgey AP. Sex steroid ablation enhances immune reconstitution following cytotoxic antineoplastic therapy in young mice. THE JOURNAL OF IMMUNOLOGY 2010; 184:6014-24. [PMID: 20483779 DOI: 10.4049/jimmunol.0802445] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cytotoxic antineoplastic therapy is used to treat malignant disease but results in long-term immunosuppression in postpubertal and adult individuals, leading to increased incidence and severity of opportunistic infections. We have previously shown that sex steroid ablation (SSA) reverses immunodeficiencies associated with age and hematopoietic stem cell transplantation in both autologous and allogeneic settings. In this study, we have assessed the effects of SSA by surgical castration on T cell recovery of young male mice following cyclophosphamide treatment as a model for the impact of chemotherapy. SSA increased thymic cellularity, involving all of the thymocyte subsets and early T lineage progenitors. It also induced early repair of damage to the thymic stromal microenvironment, which is crucial to the recovery of a fully functional T cell-based immune system. These functional changes in thymic stromal subsets included enhanced production of growth factors and chemokines important for thymopoiesis, which preceded increases in both thymocyte and stromal cellularity. These effects collectively translated to an increase in peripheral and splenic naive T cells. In conclusion, SSA enhances T cell recovery following cyclophosphamide treatment of mice, at the level of the thymocytes and their stromal niches. This provides a new approach to immune reconstitution following antineoplastic therapy.
Collapse
Affiliation(s)
- Gabrielle L Goldberg
- Immune Regeneration Laboratory, Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
|
128
|
Gutiérrez A, Mestre F, Pérez-Manga G, Rodríguez J. Diffuse large B-cell lymphoma in the older. Crit Rev Oncol Hematol 2010; 78:59-72. [PMID: 20303778 DOI: 10.1016/j.critrevonc.2010.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 02/15/2010] [Accepted: 02/18/2010] [Indexed: 10/19/2022] Open
Abstract
The incidence of diffuse large B-cell lymphoma (DLCL) in the older is growing to the point of becoming a health priority in the next decades. Prognostic factors and the biology of the tumor are not very different between younger and older populations. Furthermore, it seems that the response rate is basically similar in both populations, provided an appropriate dose of chemotherapy is administered. However, there seem to be differences with regard to a lower tolerance to treatment and a higher relapse rate in responsive older patients. To analyze these problems we review the most important differences between young and older DLCL patients in terms of immunologic status, treatment toxicity and the presence of other concomitant diseases or organ dysfunctions. We also consider the most relevant clinical studies that may allow us to make the appropriate decisions regarding DLCL therapy in this older population.
Collapse
Affiliation(s)
- A Gutiérrez
- Service of Hematology, University Hospital Son Dureta, Palma de Mallorca, Spain
| | | | | | | |
Collapse
|
129
|
Vaccinations in children with cancer. Vaccine 2010; 28:3278-84. [PMID: 20226246 DOI: 10.1016/j.vaccine.2010.02.096] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2010] [Revised: 02/22/2010] [Accepted: 02/23/2010] [Indexed: 11/20/2022]
Abstract
Children with cancer may be immunocompromised as a result of their primary underlying disease and/or the use of prolonged and intensive chemotherapy administered with or without irradiation. The damage to the immune system varies with the age of the patient, the type of cancer, and the intensity of the chemotherapy used to treat it. This review analyses the data regarding the immunogenicity, efficacy, safety and tolerability of the vaccines usually recommended in the first years of life in order to help pediatricians choose the best immunisation programme against vaccine-preventable disease in children with cancer receiving standard-dose chemotherapy. Areas for future research are highlighted because new data are required to be able to draw up evidence-based recommendations that will ensure adequate protection against infectious diseases in such high-risk children.
Collapse
|
130
|
Sportès C, Gress RE, Mackall CL. Perspective on potential clinical applications of recombinant human interleukin-7. Ann N Y Acad Sci 2010; 1182:28-38. [PMID: 20074272 DOI: 10.1111/j.1749-6632.2009.05075.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Interleukin-7 has critical and nonredundant roles in T cell development, hematopoiesis, and postdevelopmental immune functions as a prototypic homeostatic cytokine. Based on a large body of preclinical evidence, it may have multiple therapeutic applications in immunodeficiency states, either physiologic (immuno-senescence), pathologic (HIV) or iatrogenic (postchemotherapy and posthematopoietic stem cell transplant) and may have roles in immune reconstitution or enhancement of immunotherapy. Early clinical development trials in humans show that, within a short time, rhIL-7 administration results in a marked preferential expansion of both naive and memory CD4 and CD8 T cell pools with a tendency toward enhanced CD8 expansion. As a result, lymphopenic or normal older hosts develop an expanded circulating T cell pool with a profile that resembles that seen earlier in life with increased T cell repertoire diversity. These results, along with a favorable toxicity profile, open a wide perspective of potential future clinical applications.
Collapse
Affiliation(s)
- Claude Sportès
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, Maryland 20892-1104, USA
| | | | | |
Collapse
|
131
|
Abstract
Cell-based therapies with various lymphocytes and antigen-presenting cells are promising approaches for cancer immunotherapy. The transfusion of T lymphocytes, also called adoptive cell therapy (ACT), is an effective treatment for viral infections, has induced regression of cancer in early stage clinical trials, and may be a particularly important and efficacious modality in the period following hematopoietic stem cell transplantation (HSCT). Immune reconstitution post-SCT is often slow and incomplete, which in turn leads to an increased risk of infection and may impact relapse risk in patients with malignant disease. Immunization post-HSCT is frequently unsuccessful, due to the prolonged lymphopenia, especially of CD4 T cells, seen following transplant. ACT has the potential to enhance antitumor and overall immunity, and augment vaccine efficacy in the post-transplant setting. The ability to genetically engineer lymphocyte subsets has the further potential to improve the natural immune response, correct impaired immunity, and redirect T cells to an antitumor effector response. This chapter focuses on various applications of ACT for cancer immunotherapy, and we discuss some of the latest progress and hurdles in translating these technologies to the clinic.
Collapse
Affiliation(s)
- Stephan A Grupp
- Division of Oncology and Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | |
Collapse
|
132
|
Abstract
Standard therapies for many common cancers remain toxic and are often ineffective. Cellular immunotherapy has the potential to be a highly targeted alternative, with low toxicity to normal tissues but a high capacity to eradicate tumor. In this chapter we describe approaches that generate cellular therapies using active immunization with cells, proteins, peptides, or nucleic acids, as well as efforts that use adoptive transfer of effector cells that directly target antigens on malignant cells. Many of these approaches are proving successful in hematologic malignancy and in melanoma. In this chapter we discuss the advantages and limitations of each and how over the next decade investigators will attempt to broaden their reach, increase their efficacy, and simplify their application.
Collapse
Affiliation(s)
- Fatma V Okur
- Baylor College of Medicine, Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | | |
Collapse
|
133
|
Condomines M, Veyrune JL, Larroque M, Quittet P, Latry P, Lugagne C, Hertogh C, Kanouni T, Rossi JF, Klein B. Increased plasma-immune cytokines throughout the high-dose melphalan-induced lymphodepletion in patients with multiple myeloma: a window for adoptive immunotherapy. THE JOURNAL OF IMMUNOLOGY 2009; 184:1079-84. [PMID: 19966210 DOI: 10.4049/jimmunol.0804159] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
High-dose melphalan (HDM) followed by autologous stem cell transplantation (ASCT) is a standard treatment for patients with multiple myeloma. However, lymphocyte reconstitution is impaired after HDM. Recent work has suggested that the lymphopenia period occurring after various immunosuppressive or chemotherapy treatments may provide an interesting opportunity for adoptive antitumor immunotherapy. The objective of this study was to determine an immunotherapy window after HDM and ASCT, evaluating T cell lymphopenia, and measuring circulating immune cytokine concentrations in patients with multiple myeloma. The counts of T cell subpopulations reached a nadir at day 8 post-ASCT (day 10 post-HDM) and recovered by day 30. IL-6, IL-7, and IL-15 plasma levels increased on a median day 8 post-ASCT, respectively, 35-fold, 8-fold, and 10-fold compared with pre-HDM levels (p < or = 0.05). The increases in IL-7 and IL-15 levels were inversely correlated to the absolute lymphocyte count, unlike monocyte or myeloid counts. Furthermore, we have shown that CD3 T cells present in the ASC graft are activated, die rapidly when they are cultured without cytokine in vitro, and that addition of IL-7 or IL-15 could induce their survival and proliferation. In conclusion, the early lymphodepletion period, occurring 4-11 d post-HDM and ASCT, is associated with an increase of circulating immune cytokines and could be an optimal window to enhance the survival and proliferation of polyclonal T cells present in the ASC autograft and also of specific antimyeloma T cells previously expanded in vitro.
Collapse
Affiliation(s)
- Maud Condomines
- Centre Hospitalier Universitaire Montpellier, Institute of Research in Biotherapy, Montpellier, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Dudakov JA, Goldberg GL, Reiseger JJ, Vlahos K, Chidgey AP, Boyd RL. Sex steroid ablation enhances hematopoietic recovery following cytotoxic antineoplastic therapy in aged mice. THE JOURNAL OF IMMUNOLOGY 2009; 183:7084-94. [PMID: 19890044 DOI: 10.4049/jimmunol.0900196] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cytotoxic antineoplastic therapy is widely used in the clinic as a treatment for malignant diseases. The treatment itself, however, leads to long-term depletion of the adaptive immune system, which is more pronounced in older patients, predominantly due to thymic atrophy. We and others have previously shown that withdrawal of sex steroids is able to regenerate the aged thymus and enhance recovery from autologous and allogeneic hematopoietic stem cell transplant. In this study we have examined the effects of sex steroid ablation (SSA) on the recovery of lymphopoiesis in the bone marrow (BM) and thymus following treatment with the chemotherapeutic agent cyclophosphamide (Cy) in middle-aged and old mice. Furthermore, we have also examined the impact of this regeneration on peripheral immunity. SSA enhanced the recovery of BM resident hematopoietic stem cells and lymphoid progenitors and promoted lymphopoiesis. Interestingly, Cy alone caused a profound increase in the recently described common lymphoid progenitor 2 (CLP-2) population in the BM. In the thymus, SSA caused a profound increase in cellularity as well as all intrathymic T-lineage progenitors including early T-lineage progenitors (ETPs) and non-canonical T cell progenitors such as the CLP-2. We also found that these transferred into numerical increases in the periphery with enhanced B and T cell numbers. Furthermore, these lymphocytes were found to have an enhanced functional capacity with no perturbation of the TCR repertoire. Taken together, these results provide the basis for the use of SSA in the clinic to enhance treatment outcomes from cytotoxic antineoplastic therapy.
Collapse
Affiliation(s)
- Jarrod A Dudakov
- Immune Regeneration Laboratory, Monash Immunology and Stem Cell Laboratories, Monash University, Wellington Road, Clayton VIC 3800, Australia.
| | | | | | | | | | | |
Collapse
|
135
|
Turtle CJ, Swanson HM, Fujii N, Estey EH, Riddell SR. A distinct subset of self-renewing human memory CD8+ T cells survives cytotoxic chemotherapy. Immunity 2009; 31:834-44. [PMID: 19879163 DOI: 10.1016/j.immuni.2009.09.015] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 08/27/2009] [Accepted: 09/11/2009] [Indexed: 12/30/2022]
Abstract
The mechanisms that maintain human T cell memory during normal and perturbed homeostasis are not fully understood. The repeated induction of profound lymphocytopenia in patients undergoing multiple cycles of cytotoxic chemotherapy infrequently results in severe infections with viruses controlled by memory T cells, suggesting that some memory T cells survive chemotherapy and restore immunity. Here, we identified a distinct subpopulation of memory CD8(+) T cells with the ability to rapidly efflux and survive exposure to chemotherapy drugs in vitro and in vivo. T cells with high efflux capacity shared expression of molecules with hematopoietic stem cells, were quiescent in nonlymphocytopenic individuals, and were induced to proliferate in patients rendered lymphocytopenic after chemotherapy. Effluxing T cells differentiated into noneffluxing subsets in response to antigen stimulation and inflammatory signals, thereby contributing to repopulation of memory cells after chemotherapy.
Collapse
Affiliation(s)
- Cameron J Turtle
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | | | | | | | | |
Collapse
|
136
|
Tomblyn M, Chiller T, Einsele H, Gress R, Sepkowitz K, Storek J, Wingard JR, Young JAH, Boeckh MJ, Boeckh MA. Guidelines for preventing infectious complications among hematopoietic cell transplantation recipients: a global perspective. Biol Blood Marrow Transplant 2009; 15:1143-238. [PMID: 19747629 PMCID: PMC3103296 DOI: 10.1016/j.bbmt.2009.06.019] [Citation(s) in RCA: 1179] [Impact Index Per Article: 78.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Accepted: 06/23/2009] [Indexed: 02/07/2023]
|
137
|
|
138
|
Mueller YM, Do DH, Boyer JD, Kader M, Mattapallil JJ, Lewis MG, Weiner DB, Katsikis PD. CD8+ cell depletion of SHIV89.6P-infected macaques induces CD4+ T cell proliferation that contributes to increased viral loads. THE JOURNAL OF IMMUNOLOGY 2009; 183:5006-12. [PMID: 19786539 DOI: 10.4049/jimmunol.0900141] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previous studies have shown that depletion of CD8(+) cells during acute and chronic simian immunodeficiency virus (SIV) infection leads to increased viral replication, morbidity, and mortality, which have been attributed to loss of CD8(+) T cell-mediated control of SIV. However, these studies did not exclude that CD8(+) cell depletion increased homeostatic proliferation of CD4(+) T cells, resulting in increased viral targets and, therefore, viral rebound. Chronically SHIV89.6P-infected cynomolgus macaques were CD8(+) cell-depleted, and the frequency, cell number, and phenotype of CD4(+) T cells and viral infection were examined using flow cytometry and quantitative real-time PCR. The frequency and number of Ki-67-expressing CD4(+) T cells were increased with CD8(+) cell depletion. This proliferation of CD4(+) T cells occurred even in animals with no rebound of viral loads. Most of the proliferating cells were effector memory CD4(+) T cells. Plasma simian HIV (SHIV) RNA copies positively correlated with proliferating CD4(+) T cells and SHIV DNA copies in Ki-67(+) CD4(+) T cells. Although this study does not exclude an important role for virus-specific CD8(+) T cells in SIV and SHIV infection, our data suggest that homeostatic proliferation is an important contributor to increases in plasma viremia that follow CD8(+) cell depletion.
Collapse
Affiliation(s)
- Yvonne M Mueller
- Department of Microbiology and Immunology, and Center for Immunology and Vaccine Sciences, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | | | | | | | | | | | | | | |
Collapse
|
139
|
Abstract
Interleukin-7 (IL-7) is required for the development and survival of T cells and plays a critical role in modulating T-cell homeostasis. This review will address current understanding of IL-7 biology, review recent clinical experiences and discuss potential future clinical applications of IL-7, or IL-7 blockade, in the setting of disease.
Collapse
Affiliation(s)
- C M Capitini
- Immunology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | | | | |
Collapse
|
140
|
A phase 1/2 study of autologous neuroblastoma tumor cells genetically modified to secrete IL-2 in patients with high-risk neuroblastoma. J Immunother 2009; 31:812-9. [PMID: 18833006 DOI: 10.1097/cji.0b013e3181869893] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Autologous neuroblastoma (NB) tumor cells modified to secrete interleukin (IL)-2 (auto-IL-2) can be safely given to patients with advanced neuroblastoma and generate antitumor immune responses. As the benefits of tumor immunization may be greater in patients with minimal residual disease and thus rely on surrogate markers such as immune responses to measure effect, we studied the frequency of immune changes associated with vaccination. Thirteen patients (8 in first remission and 5 after treatment for recurrent NB) received 5 to 8 subcutaneous injections of auto-IL-2 at 0.3 x 10 cells/kg. The vaccine was well tolerated. Injection site biopsies revealed increased cellularity caused by infiltration of CD4 and CD8 lymphocytes, eosinophils, and dendritic cells. Enzyme-linked immunosorbent spot assays for interferon-gamma and IL-5 demonstrated that vaccination produced a rise in circulating CD4 and CD8 T cells responsive to stimulation by autologous tumor cells. Median event-free survival was 22 months for patients in first remission and 3 months for all others. Four patients treated in first remission remain alive and 3 without disease recurrence.
Collapse
|
141
|
Abrahamsson S, Muraro PA. Immune re-education following autologous hematopoietic stem cell transplantation. Autoimmunity 2009; 41:577-84. [DOI: 10.1080/08916930802197081] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
142
|
Vedel SJ, Tholstrup D, Kolte L, Gaardbo J, Ryder LP, Ersbøll A, Albrecht-Beste E, Jurlander J, Nielsen JO, Nielsen SD. Limited impact of the thymus on immunological recovery during and after chemotherapy in patients with diffuse large B-cell lymphoma. Scand J Immunol 2009; 69:547-54. [PMID: 19439016 DOI: 10.1111/j.1365-3083.2009.02252.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To investigate the impact of thymus on immunological recovery after dose-dense chemotherapy a prospective study of 17 patients diagnosed with diffuse large B-cell lymphoma (DLBCL) was conducted. Patients were monitored before, during and until 3 months after chemotherapy. The thymus was visualized using computer tomographic scans. Patients were divided into two groups according to thymic size, one group comprising of patients without detectable thymus and one group of patients with detectable thymus. Naïve CD4 and CD8 counts were measured by flow cytometry, and to measure thymic output determination of CD4+ cells containing T-cell receptor excision circles (TREC) was done. During chemotherapy, the naïve CD4 count decreased significantly as did the CD4-TREC%. Significant difference in recovery of naïve CD4 counts between patients with detectable and undetectable thymic tissue during treatment with chemotherapy was not found. CD4-TREC% was associated with lower age. It was not possible to demonstrate an association between thymic size and recovery of the naïve CD4+ cells. The study terminated 3 months after the last cycle of chemotherapy, and at that time point the naïve CD4 counts and the CD4-TREC% had not returned to pretreatment levels. However, patients with detectable thymic tissue had higher naïve CD4 counts after the first cycles of chemotherapy, suggesting that these patients may be less susceptible to infectious complications related to chemotherapy.
Collapse
Affiliation(s)
- S J Vedel
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Yakoub-Agha I, Saule P, Magro L, Cracco P, Duhamel A, Coiteux V, Bruno B, Dufossé F, Jouet JP, Dessaint JP, Labalette M. Immune Reconstitution following Myeloablative Allogeneic Hematopoietic Stem Cell Transplantation: The Impact of Expanding CD28negative CD8+ T Cells on Relapse. Biol Blood Marrow Transplant 2009; 15:496-504. [DOI: 10.1016/j.bbmt.2008.11.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Accepted: 11/27/2008] [Indexed: 12/28/2022]
|
144
|
Mir MA, Battiwalla M. Immune deficits in allogeneic hematopoietic stem cell transplant (HSCT) recipients. Mycopathologia 2009; 168:271-82. [PMID: 19156534 DOI: 10.1007/s11046-009-9181-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Accepted: 12/31/2008] [Indexed: 01/08/2023]
Abstract
Immune deficits account for the high frequency of life threatening bacterial, viral, and fungal opportunistic infections seen in allogeneic HSCT recipients. Despite advances in infectious disease management, the integrity of host defenses remains the mainstay of defense. The intensity of the preparative regimen, degree of HLA matching, source of stem cells (marrow, blood, or cord), extent of T-cell depletion, and immunosuppressive therapy are some of the factors that impact the kinetics, characteristics, and quality of immune reconstitution. Graft-versus-host disease and its prophylaxis or treatment produce a host environment that is particularly vulnerable to infections. Mucosal disruption and prolonged severe neutropenia usually confine their impact to the early course of transplant. After initial engraftment, HSCT recipients remain at great risk for opportunistic infections and this is related to prolonged and severe T-lymphocyte dysfunction of a complex multifactorial nature. B cell dysfunction is less problematic clinically, but includes deficiencies of immunoglobulin subclasses and impaired ability to mount a vaccine response. Advances in understanding of these immune deficits have resulted in successful strategies including revaccination, growth factors, thymic protection, and adoptive cellular therapy with antigen-specific cells.
Collapse
Affiliation(s)
- Muhammad A Mir
- Division of Hematology, University at Buffalo, Buffalo, NY, USA
| | | |
Collapse
|
145
|
|
146
|
Guimond M, Veenstra RG, Grindler DJ, Zhang H, Cui Y, Murphy RD, Kim SY, Na R, Hennighausen L, Kurtulus S, Erman B, Matzinger P, Merchant MS, Mackall CL. Interleukin 7 signaling in dendritic cells regulates the homeostatic proliferation and niche size of CD4+ T cells. Nat Immunol 2009; 10:149-57. [PMID: 19136960 PMCID: PMC2713006 DOI: 10.1038/ni.1695] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Accepted: 12/03/2008] [Indexed: 11/12/2022]
Abstract
Interleukin 7 (IL-7) and T cell receptor (TCR) signals have been proposed to be the primary drivers of homeostatic T cell proliferation. However, it is not known why CD4+ T cells undergo less efficient homeostatic proliferation than CD8+ T cells. Here we showed that systemic IL-7 concentrations rise during lymphopenia due to diminished IL-7 utilization, but that IL-7 signaling on IL-7Rα+ dendritic cells (DCs) in lymphopenic settings paradoxically diminishes CD4+ T cell homeostatic proliferation. This effect is mediated, at least in part, by IL-7-mediated downregulation of MHC class II expression on IL-7Rα+ DCs. These results implicate IL-7Rα+ DCs as regulators of the peripheral CD4+ T cell niche, and indicate that IL-7 signals in DCs prevent uncontrolled CD4+ T cell expansion in vivo.
Collapse
Affiliation(s)
- Martin Guimond
- Pediatric Oncology Branch, National Cancer Institute, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Westerterp M, Boermeester MA, Omloo JMT, Hulshof MCCM, Vervenne WL, Lutter R, Out TA, van Lanschot JJB. Differential responses of cellular immunity in patients undergoing neoadjuvant therapy followed by surgery for carcinoma of the oesophagus. Cancer Immunol Immunother 2008; 57:1837-47. [PMID: 18398607 PMCID: PMC11030738 DOI: 10.1007/s00262-008-0511-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Accepted: 03/25/2008] [Indexed: 11/25/2022]
Abstract
BACKGROUND To compare immune responses following neoadjuvant chemoradiation therapy in combination with hyperthermia plus surgery to those induced by surgery alone in patients with oesophageal cancer. METHODS Thirty-two patients with histopathologically proven oesophageal cancer, scheduled for potentially curative transhiatal or transthoracic oesophagectomy with (neo, n = 20) or without (control, n = 12) neoadjuvant thermochemoradiation therapy (ThCR) were included. Peripheral blood samples were obtained before ThCR, after 2 weeks of ThCR, 1 day before surgery, on postoperative days 1, 3, 7, and 6 weeks after surgery, for white blood cell counts, lymphocyte subsets and T helper type 1 (Th1) and type 2 (Th2) lymphocyte responses. RESULTS Neo patients showed a significant decrease in granulocytes and lymphocyte subsets, and T cell cytokines after 2 weeks of ThCR. Only CD8+ (cytotoxic) T cells recovered after ThCR to reach normal levels prior to surgery. In contrast, CD4+ T (helper) cells, and NK- and B cells in neo patients did not recover prior to surgery (all P < 0.05). Oesophagectomy induced a significant increase in granulocytes and a decrease in lymphocytes (and subsets). Only those subsets that had not recovered after ThCR (CD4+ T cells, NK and B cells but not CD8+ T cells), were significantly lower (all P < 0.05) during the entire postoperative study period. Postoperatively, the stimulated cytokine production capacity of Th1 and Th2 cells, corrected for number of T cells, was not significantly different between the groups. CONCLUSION Neoadjuvant thermochemoradiation for oesophageal cancer caused significant disturbances of host cellular immunity with reduced T, NK and B cell counts, and differential recovery of cytotoxic and helper T cells leading to prolonged T cell imbalance that extends beyond the time of surgery. The functional and anti-tumour consequences of this immunodisturbance need further investigation, as recovery of T helper cytokine production towards surgery was less impaired than T helper cell counts.
Collapse
Affiliation(s)
- Marinke Westerterp
- Department of Surgery, Academic Medical Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
148
|
Immune reconstitution and implications for immunotherapy following haematopoietic stem cell transplantation. Best Pract Res Clin Haematol 2008; 21:579-96. [PMID: 18790456 DOI: 10.1016/j.beha.2008.06.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recovery of a fully functional immune system is a slow and often incomplete process following allogeneic stem cell transplantation. While innate immunity reconstitutes quickly, adaptive B- and especially T-cell lymphopoeisis may be compromised for years following transplantation. In large part, these immune system deficits are due to the decrease, or even absence, of thymopoiesis following transplantation. Thereby, T-cell reconstitution initially relies upon expansion of mature donor T cells; a proliferation driven by high cytokine levels and the presence of allo-reactive antigens. This peripheral mechanism of T-cell generation may have important clinical consequences. By expanding tumouricidal T cells, it may provide a venue to enhance T-cellular immunotherapy following transplantation. Alternatively, decreased thymic function may impair long-term anti-tumour immunity and increase the likelihood of graft-versus-host disease.
Collapse
|
149
|
Abstract
Immune reconstitution following haematopoietic stem cell transplantation (SCT) is an often slow and incomplete process that leads to increased risk of infection and malignant disease. Immunization in SCT is frequently unsuccessful due to the prolonged lymphopenia, especially of CD4 T cells, seen following transplant. The transfusion of T cells, also called 'adoptive T-cell therapy', has the potential to enhance anti-tumour and overall immunity, and augment vaccine efficacy in the post-transplant setting. Recent advances in tissue culture, cellular immunology and tumour biology are guiding new approaches to adoptive T-cell therapy. This chapter will discuss the challenges that face the field before adoptive T-cell therapy can be translated into routine clinical practice.
Collapse
Affiliation(s)
- Nicole A Aqui
- Abramson Family Cancer Research Institute and the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-1416, USA.
| | | |
Collapse
|
150
|
Strategies for reconstituting and boosting T cell-based immunity following haematopoietic stem cell transplantation: pre-clinical and clinical approaches. Semin Immunopathol 2008; 30:457-77. [PMID: 18982327 DOI: 10.1007/s00281-008-0140-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2008] [Accepted: 10/14/2008] [Indexed: 12/14/2022]
Abstract
Poor immune recovery is characteristic of bone marrow transplantation and leads to high levels of morbidity and mortality. The primary underlying cause is a compromised thymic function, resulting from age-induced atrophy and further compounded by the damaging effects of cytoablative conditioning regimes on thymic epithelial cells (TEC). Several strategies have been proposed to enhance T cell reconstitution. Some, such as the use of single biological agents, are currently being tested in clinical trials. However, a more rational approach to immune restoration will be to leverage the evolving repertoire of new technologies. Specifically, the combined targeting of TEC, thymocytes and peripheral T cells, together with the bone marrow niches, promises a more strategic clinical therapeutic platform.
Collapse
|