Marrow Transplantation (2015) 50, S51–S54 © 2015 Macmillan Publishers Limited All rights reserved 0268-3369/15 www.nature.com/bmt

ORIGINAL ARTICLE for viral and fungal infections

H Einsele, J Löffler, M Kapp, L Rasche, S Mielke and UG Grigoleit

Allogenic stem cell transplantation (allo-SCT) represents the only curative option for several hematological malignancies. Due to a delayed and dysfunctional immunological recovery infectious complications and residual tumor cells following allo-SCT are still major causes of failure of this procedure. Here we discuss the most common infectious complications of allo-SCT and describe current and future strategies to prophylaxe or treat these complications using novel immunotherapeutic strategies.

Bone Marrow Transplantation (2015) 50, S51–S54; doi:10.1038/bmt.2015.96

INTRODUCTION Suitable combination of /T-cell will allow for Allogeneic transplantation (allo-SCT) optimal stimulation and selection of multiple pathogen-specific represents the only curative option for several hematological lines and a characterization of pathogen specificity and malignancies. Although several improvements in conditioning alloreactivity for safety and quality control of the generated regimens and SCT procedures (especially in vivo/ex vivo T-cell T cell lines. depletion) allow to achieve a fast and long-lasting regeneration of Furthermore, for the high-risk patients who are seropositive and hematopoiesis, patients are still suffering from major complica- receive a transplant from a seronegative donor, T-cell priming or tions due to a delayed and dysfunctional immunological recovery. redirection of donor T cells have to be used as well as the transfer of -specific cells from seropositive third party donors. Disease relapse and life-threatening infections with multiple pathogens (for example, CMV, EBV, adenovirus (ADV), Aspergillus spp., Candida spp. and so on) severely affect the ultimate outcome VIRAL INFECTIONS POST TRANSPLANT of SCT. Disease status, previous treatment, patient age and the degree of donor/patient mismatches are critical variables influen- Allo-SCT can cure patients with several hematological malignan- cing the relative risks of relapse and infections. A major strategy to cies but exposes patients to periods of marked immunosuppres- deal with viral and in the future also fungal infection is the clinical sion, during which infections, especially viral and fungal infections, application of T-cell subsets directed against multiple pathogens can cause morbidity and mortality. CMV reactivation/disease has a 1,2 in a patient-tailored approach, able to boost and shape the novel major negative impact on the outcome of SCT. Inspite of novel donor-derived toward proper antigens and anti-CMV drugs (CMX001 and letermovir) with promising activity fi 3 ultimately improve the clinical outcome. and good safety pro le, CMV infection remains a life-threatening Clinical application needs to include central memory T cells, complication and a major cause of infection-related mortality in recipients of an allo-SCT, especially in patients undergoing cord THELPER cells and probably also effector memory T cells. This goal, which represents a major progress for patients with hematological or haploidentical SCT as well as patients with severe acute malignancies, will be pursued through the development of or extensive chronic GvHD. innovative strategies of isolation, expansion and manipulation of In addition, ADV infections have been increasingly described in these patients with an incidence up to 29%, again with patients donor T- subpopulations. Adoptive transfer of T cells undergoing a stem cell transplant from an alternative donor being directed against single viral pathogens has been effective in small fi the patient cohort at a highest risk for ADV infection and disease. phase I/II studies but often therapeutic ef cacy was limited not EBV reactivations post SCT can induce life-threatening post- only owing to the lack of persistence and migration of the transplant lymphoproliferative disease. Current data reveal that transferred donor T cells to the site of infection, but also by serotherapy with anti- globulin or Campath as well as additional infections not targeted by the transferred T cells. the occurrence of severe acute GvHD and also SCT from an Therefore the aim of adoptive T-cell therapy is the generation of alternative donor or a mismatched related or unrelated donor is polyclonal T-cell populations against multiple pathogens, not only associated with an increased risk for an EBV-related lymphopro- by adding T cells directed against different infectious pathogens, liferative disorder.4 but also by the controlled coordination of a variety of T-cell It is now appreciated that T-cell reconstitution is required for populations and T-cell responses. To favor long-lasting clinical the sustained control of CMV, EBV and ADV infections5,6 and that responses new T-cell selection and manipulation procedures that drug therapy limits the infection, but cannot prevent concurrent favor the generation of antigen-specific central memory and reactivations and may induce major toxicity (hematotoxicity/ memory stem T cells, a recently discovered T-cell subset nephrotoxicity/secondary infections). characterized by a high degree of plasticity, expansion potential An urgent clinical demand for enriched virus, fungal, multivirus and self-renewal capacities has to be pursued. and multipathogen-specific T cells with reduced alloreactivity has

Department of Internal Medicine II, University Hospital of Wuerzburg, Wuerzburg, Germany. Correspondence: Professor H Einsele, Department of Internal Medicine II, University Hospital Wuerzburg, Oberduerrbacher Strasse 6, 97080 Wuerzburg, Germany. E-mail: [email protected] This article was published as part of a supplement, supported by WIS-CSP Foundation, in collaboration with Gilead, Milteny Biotec, Gamida cell, Adienne Pharma and Biotech, Medac hematology, Kiadis Pharma, Almog Diagnostic. Immunotherapy for viral/fungal infections H Einsele et al S52 been expressed.6 Several groups have reported potential with the aim of adoptive transfer directed against single antigens protocols,7–10 but only very limited clinical trial activity has been in viral infection, which can lead to reconstitution of antiviral reported so far. after SCT.21–24 So far, logistics have limited the application of cellular therapies and short-term in vivo persistence clearly interferes with the FUNGAL INFECTIONS antiviral efficacy of transferred T cells. Recently, a long-lived Invasive fungal infections are one of the major threats to patients human memory T-cell population has been described with undergoing allo-SCT. The incidence of invasive fungal infection relevance for adoptive T-cell transfer approaches. This subpopula- depending on the geographic localization of the transplantation tion has an enhanced capacity for self-renewal and a multipotent center as well as the percentage of patients belonging to high-risk ability to derive central memory, effector memory and effector group for invasive fungal infection can go beyond 15%. T cells. These cells, specific to multiple viral and self-tumor Yet diagnostic techniques are rather insensitive and not very antigens, were found within a CD45RO-, CCR7+, CD45RA+, CD62L+, reliable—leading to a vast overtreatment of patients—often as a CD27+, CD28+ and IL-7Rα+ T-cell compartment characteristic of prophylaxis—with drugs that are potentially toxic but definitely naive T cells. However, they expressed large amounts of CD95, very expensive. Thus, large transplant centers spend easily 41 IL-2Rβ, CXCR3 and LFA-1, and showed numerous functional Mio € for diagnostic and therapeutic strategies to prevent and attributes distinctive of memory cells. Compared with known treat invasive fungal infection. In spite of these high costs, the memory populations, these had increased prolifera- outcome of invasive fungal infection, especially when lung and tive capacity and more efficiently reconstituted immunodeficient even more important the central nervous system is involved, is hosts and were recently termed memory stem T cells.25 As T-cell rather poor (mortality ranging from 30 to 80% depending on the responses can reconstitute even from a single T cell, repopulating underlying disease and the transplant modality as well as the capacities are of clinical significance for adoptive T-cell transfer occurrence of severe acute and chronic GvHD). approaches.26 However, the starting population/phenotype for so- Thus, new diagnostic and treatment strategies for invasive called ‘giant T-cell clones’27 is still not clear in humans. fungal infection are highly warranted. Adoptive T-cell transfer for CMV reactivation after SCT has been performed over decades with T cell lines generated by repetitive stimulation in vitro.21,28 IMMUNOTHERAPY FOR FUNGAL INFECTIONS New strategies of T-cell selection using antigen-induced T-cell Studies in mice and humans suggest the importance of the stimulation and isolation by capture assay, streptamer T-helper type 1 (Th1) response in conferring protection against technology29 or via increased expression of activation markers has invasive aspergillosis. Th1 interferon-gamma-secreting cells are been developed.9,10,30 In a recent study, this approach has been believed to increase innate immune responses9 against aspergillus successfully applied to patients with chemorefractory CMV by activating and , and enhance infection and disease.31 Adoptive transfer of antigen-specific antimicrobial activity of respiratory epithelial cells. Mice given T cells for EBV, ADV and CMV is now established at several centers neutralizing against Th2 to indirectly in a clinical protocol with the required clinical, immunological, enhance Th1 cytokine production have increased protection virological, technical and regulatory preconditions.6,30 against aspergillosis and Th1 cells obtained from mice previously Selective allodepletion may serve as an improved platform immunized with aspergillus antigens were shown to protect from technology where GvHD-causing T cells are depleted from the aspergillosis following adoptive transfer to otherwise susceptible stem cell allograft enabling the remaining T-cell repertoire to mice. Patients undergoing allo-SCT are also at a high risk to facilitate anti-infectious antimalignant properties in the presence develop invasive aspergillosis and we have shown that a low rate of low or no immunosuppression. Various methods of allodeple- of T-cell proliferation when exposed to aspergillus antigens as well tion have been successfully established, some of which including as a predominant aspergillus-specific Th2 response are associated CD25-immunotoxin-based allodepletion and TH9402-based with a worse outcome of invasive aspergillosis in patients who photodepletion have been already transferred into the clinical have undergone allo-SCT.11 setting.32–36 International multicenter trials are recruiting. Until Several groups have established methods to select and expand now 4100 patients after allo-SCT have been treated against aspergillus-specific T cells and a small clinical trial has shown single pathogens all over Europe with Ag-specific T cells.37,38 promising activity and good safety of the transfer of in vitro However, in vivo persistence of transferred T cells in the recipient stimulated and selected aspergillus-specific T-cell clones to remains a problem of T-cell transfer either due to inherent patients with uncontrolled invasive aspergillosis following allo- properties of the T-cell product or due to immunosuppressive SCT.12 drugs in the recipient. Especially, third-party T-cell transfer from In addition, we and others have shown that also activated HLA-partially matched donors has been shown safe and natural killer cells—which can be easily selected and enriched feasible38,39 but in vivo persistence of T cells is limited. In addition, 13–15 + for—can mediate anti-aspergillus activity. the role of CD4 THELPER cells, which have been emphasized over Data from our group have also demonstrated that isolated many years for the efficacy of adoptive T-cell transfer,9,21,24,40,41 gamma-delta T cells can mediate anti-aspergillus activity.11 still remains to be investigated. Gamma-delta T lymphocytes can be highly enriched for under good manufacturing practice conditions using the Clinimacs device16,17 and also easily be activated and expanded in vivo by SELECTION/GENERATION AND TRANSFER OF stimulation with aminobisphosphonates.18 MULTIPATHOGEN-SPECIFIC T CELLS Thus, this cell population that also mediates antiviral and anti- In the past few years novel screening techniques have revealed tumor activity,19,20 but does not mediate alloreactivity will be a multiple viral and fungal pathogens to emerge as major very attractive option to be used for adoptive immunotherapy for contributors to post-transplant morbidity and mortality, expand- infection and tumor control post transplant. ing from CMV, EBV, ADV) to HSV, BK virus and VZV, also including HHV-6 and several fungal pathogens, for example, Aspergillus spp., Candida spp., especially non-albicans Candida spp. and as an IMMUNOTHERAPY FOR VIRAL INFECTIONS emergent problem also Mucor spp. Antigen-specific T cells against single pathogens (ADV, CMV and Thus, an urgent clinical demand for enriched multivirus and also EBV) have been and are currently investigated by several groups multipathogen-specific T cells with reduced alloreactivity has

Bone Marrow Transplantation (2015) S51 – S54 © 2015 Macmillan Publishers Limited Immunotherapy for viral/fungal infections H Einsele et al S53 been expressed.6 Several groups have reported potential Several hundreds of patients were treated successfully and with protocols,7,8,10,42 but only very limited clinical trial activity has little toxicity with CMV-, EBV- and ADV-specific T cells for refractory been reported so far. virus infection following allo-SCT. In addition, isolation of Broader implementation of multipathogen-specific T-cell multipathogen-specific T cells can be successfully achieved from therapy is restricted by the limited spectrum of immunodominant the seropositive donors using novel selection devices. In some fi viral and fungal proteins and de ned and available that patients multipathogen-specific T cells also were successfully can be targeted in a single T cell line and the logistics of administered and showed antiviral activity and successful control manufacture of these lines. One major problem anticipated in the of CMV, EBV and ADV infection. generation of multipathogen-specific cell lines is the antigenic competition between high- and low-frequency T cells as well as T cells of high and low avidity. Competition for shared APCs may CONFLICT OF INTEREST favor the generation of T cell lines dominated by responses to a single or only a few pathogens, thus limiting the coverage of HE received consulting and lecture fees from Celgene, Janssen, Amgen, Onyx and fi pathogens provided by the T-cell product. In addition, often the Novartis. JL received grant support from P zer Europe. SM has received lecture fees manufacturing process used to generate these multipathogen- from Deutschen Rentenversicherung, Cellex GmbH, Novartis Pharma, Shire, and grant fi support from Wilhelm-Sander-Stiftunq. Germany, Carreras-Leukamie-Stiftunq. The speci c T-cell products is very complex and extremely labor and fl time consuming. Thus, we have established two new strategies to remaining authors declare no con ict of interest. isolate mutlipathogen-specific T cell lines for the treatment of patients at risk for multiple viral and fungal infections post ACKNOWLEDGEMENTS transplant. We are evaluating the use of MHC multimers that reversibly Research Funds were received for this project by EU FP7 T-Control and the Deutsche bind to the TCR of antiviral and antifungal CD8+ and CD4+ T cells. Forschungsgemeinschaft (DFG) CRC/TR 124. Using these streptamers and pools covering several of the immunodominat viral antigenic epitopes, we have been able to generate T cell lines at high purity.29,43 The technology can also be REFERENCES used to generate multivirus-specific T cell lines at high purity and 1 Seggewiss R, Einsele H. Immune reconstitution after allogeneic transplantation with a good yield (unpublished results). An alternative technology and expanding options for immunomodulation: an update. Blood 2010; 115: developed in our labs is the selection of T cells directed against 3861–3868. viral and fungal antigenic epitopes by stimulation with peptide 2 Einsele H, Hebart H, Kauffmann-Schneider C, Sinzger C, Jahn G, Bader P et al. Risk mixes covering several relevant CMV-, EBV-, ADV-, Aspergillus factors for treatment failures in patients receiving PCR-based preemptive therapy 25 – fumigatus- and Candida albicans-specific T-cell epitopes followed for CMV infection. Bone Marrow Transplant 2000; :757 763. 3 Marty FM, Winston DJ, Rowley SD, Vance E, Papanicolaou GA, Mullane KM et al. by selection via the upregulated activation markers (for example, 9 CMX001-201 Clinical Study Group. CMX001 to prevent disease in CD137, CD154 and so on). hematopoietic-cell transplantation. N Engl J Med 2013; 369: 1227–1236. For generation from multipathogen-specific T cells from a 4 Rasche L, Kapp M, Einsele H, Mielke S. EBV-induced post transplant lymphopro- seronegative donor in vitro priming using professional APCs liferative disorders: a persisting challenge in allogeneic hematopoetic SCT. Bone pulsed with the relevant peptides from immunodominant viral Marrow Transplant 2014; 49:163–167. antigens in the presence of exogenous cytokines, especially IL7 5 Marty FM, Ljungman P, Papanicolaou GA, Winston DJ, Chemaly RF, Strasfeld L and IL15 can be used. In addition, the depletion of regulatory et al. Maribavir prophylaxis for prevention of cytomegalovirus disease in T cells and the selection of the primed pathogen-specific T cells recipients of allogeneic stem-cell transplants: a phase 3, double-blind, placebo- via surface expression of activation markers plus/without in vitro controlled, randomised trial. Lancet. 2011Infect Dis 11: 284–292. 6 Feuchtinger T, Lücke J, Hamprecht K, Richard C, Handgretinger R, Schumm M expansion have been shown to further increase the yield of fi multipathogen-specific T cells.44 This approach has been applied et al. Detection of adenovirus-speci c T cells in children with adenovirus infection after allogeneic stem cell transplantation. Br J Haematol 2005; 128:503–509. with dendritic cells out of a minimum of 100 mL whole blood, but 7 Gerdemann U, Vera JF, Rooney CM, Leen AM. Generation of multivirus-specific will require extensive T-cell expansion including CD3/28 beads for T cells to prevent/treat viral infections after allogeneic hematopoietic stem cell sufficient cell numbers (for clinical application plus T-cell assays). transplant. J Vis Exp 2011; 51: 2736–2742. Another strategy to generate multipathogen-specific T cells in 8 Zandvliet ML, van Liempt E, Jedema I, Kruithof S, Kester MG, Guchelaar HJ et al. case of a seronegative stem cell donor is to isolate these cells from Simultaneous isolation of CD8(+) and CD4(+) T cells specific for multiple viruses a not completely HLA-matched third party donor. This strategy for broad antiviral immune reconstitution after allogeneic stem cell transplanta- requires high purity and a predominant selection of T-cell tion. J Immunother 2011; 34:307–319. populations with capacity of ‘self-renewal’. 9 Stühler C, Khanna N, Bozza S, Zelante T, Moretti S, Kruhm M et al. Cross-protective These approaches will have to be tested extensively in vitro for TH1 immunity against Aspergillus fumigatus and Candida albicans. Blood 2011; 117 – safety and efficacy. : 5881 5891. 10 Khanna N, Stühler C, Conrad B, Lurati S, Krappmann S, Einsele H et al. Generation One multicenter study has used banked third party virus- fi fi of a multiple pathogen-speci c T-cell product for adoptive immunotherapy based speci c T cells to treat severe viral infections after hematopoietic on activation-dependent expression of CD154. Blood 2011; 118:1121–1131. 39 fi SCT. After preparation of a bank of 32 virus-speci c T cell lines 11 Hebart H, Bollinger C, Fisch P, Sarfati J, Meisner C, Baur M et al. Analysis of T-cell from individuals with common HLA polymorphisms with detect- responses to Aspergillus fumigatus antigens in healthy individuals and patients able T-cell responses to EBV, CMV or ADV, 50 patients with severe, with hematologic malignancies. Blood 2002; 100: 4521–4528. therapy-refractory CMV, EBV or ADV infection after allo-SCT were 12 Perruccio K, Tosti A, Burchielli E, Topini F, Ruggeri L, Carotti A et al. Transferring treated with one of these T cell lines. More than 70% of patients functional immune responses to pathogens after haploidentical hematopoietic who received multivirus-specific T cell lines from the bank third transplantation. Blood 2005; 106: 4397–4406. party donor T cells responded—independent of the treated viral 13 Bouzani M, Ok M, McCormick A, Ebel F, Kurzai O, Morton CO et al. Human NK cells infection (CMV, EBV and ADV). Only four responders had a display important antifungal activity against Aspergillus fumigatus, which is fi directly mediated by IFN-γ release. J Immunol. 2011; 187:1369–1376. recurrence or progression. The frequency of virus-speci c T cells fl fi 14 Bouzani M, Einsele H, Loef er J. Functional analysis is a paramount prerequisite that circulated in the peripheral blood increased signi cantly post for understanding the in vitro interaction of human natural killer cells with infusion, coincident with the decrease in viral load. Aspergillus fumigatus. J Infect Dis 2012; 205: 1025–1026. In patients who could be monitored for circulating T cells from 15 Lehrnbecher T, Tramsen L, Koehl U, Schmidt S, Bochennek K, Klingebiel T. the third party, donor persistence of the transferred T cells could Immunotherapy against invasive fungal diseases in stem cell transplant be demonstrated up to 12 weeks. recipients. Immunol Invest 2011; 40: 839–852.

© 2015 Macmillan Publishers Limited Bone Marrow Transplantation (2015) S51 – S54 Immunotherapy for viral/fungal infections H Einsele et al S54 16 Smetak M, Kimmel B, Birkmann J, Schaefer-Eckart K, Einsele H, Wilhelm M et al. 32 Mielke S, McIver ZA, Shenoy A, Fellowes V, Khuu H, Stroncek DF et al. Selectively Clinical-scale single-step CD4(+) and CD8(+) cell depletion for donor innate T cell-depleted allografts from HLA-matched sibling donors followed by low-dose lymphocyte infusion (DILI). Bone Marrow Transplant 2008; 41:643–650. posttransplantation immunosuppression to improve transplantation outcome in 17 Wilhelm M, Smetak M, Schaefer-Eckart K, Kimmel B, Birkmann J, Einsele H et al. patients with hematologic malignancies. Biol Blood Marrow Transplant 2011; 17: Successful adoptive transfer and in vivo expansion of haploidentical γδ T cells. 1855–1861. J Transl Med 2014; 12:45. 33 Stühler C, Mielke S, Chatterjee M, Duell J, Lurati S, Rueckert F et al. Selective 18 Kunzmann V, Kimmel B, Herrmann T, Einsele H, Wilhelm M. Inhibition of depletion of alloreactive T cells by targeted therapy of 90: a phosphoantigen-mediated gammadelta T-cell proliferation by CD4+ CD25+ novel strategy for control of graft-versus-host disease. Blood 2009; 114: FoxP3+ regulatory T cells. 2009; 126: 256–267. 2829–2836. 19 Kunzmann V, Bauer E, Wilhelm M. Gamma/delta T-cell stimulation by pami- 34 Mielke S, Nunes R, Rezvani K, Fellowes VS, Venne A, Solomon SR et al. A clinical- dronate. N Engl J Med. 1999; 340: 737–738. scale selective allodepletion approach for the treatment of HLA-mismatched and 20 Kunzmann V, Bauer E, Feurle J, Weissinger F, Tony HP, Wilhelm M. Stimulation of matched donor-recipient pairs using expanded T lymphocytes as antigen- gammadelta T cells by aminobisphosphonates and induction of antiplasma cell presenting cells and a TH9402-based photodepletion technique. Blood 2008; 111: 96 – activity in multiple myeloma. Blood 2000; :384 392. 4392–4402. fl 21 Einsele H, Roosnek E, Rufer N, Sinzger C, Riegler S, Löf er J et al. Infusion of 35 Mielke S, Rezvani K, Savani BN, Nunes R, Yong AS, Schindler J et al. Reconstitution fi cytomegalovirus (CMV)-speci c T cells for the treatment of CMV infection not of FOXP3+ regulatory T cells (Tregs) after CD25-depleted allotransplantation in 99 – responding to antiviral chemotherapy. Blood 2002; :3916 3922. elderly patients and association with acute graft-versus-host disease. Blood 2007; 22 Feuchtinger T, Matthes-Martin S, Richard C, Lion T, Fuhrer M, Hamprecht K et al. 110:1689–1697. fi Safe adoptive transfer of virus-speci c T-cell immunity for the treatment of 36 Mielke S, Solomon SR, Barrett AJ. Selective depletion strategies in allogeneic stem systemic adenovirus infection after allogeneic stem cell transplantation. Br J cell transplantation. Cytotherapy 2005; 7:109–115 Review. 134 – Haematol 2006; :64 76. 37 Feuchtinger T, Richard C, Joachim S, Scheible MH, Schumm M, Hamprecht K et al. 23 Leen AM, Myers GD, Sili U, Huls MH, Weiss H, Leung KS et al. Monoculture-derived Clinical grade generation of hexon-specific T cells for adoptive T-cell Transfer as a fi T lymphocytes speci c for multiple viruses expand and produce clinically relevant treatment of adenovirus infection after allogeneic stem cell transplantation. effects in immunocompromised individuals. Nat Med 2006; 12: 1160–1166. J Immunother 2008; 31:199–206. 24 Icheva V, Kayser S, Wolff D, Tuve S, Kyzirakos C, Bethge W et al. Adoptive transfer 38 Schottker B, Feuchtinger T, Schumm M, Klinker E, Handgretinger R, Einsele H et al. of EBNA1-specific T cells as a treatment of Epstein-Barr-Virus reactivation and Five donors-one recipient: modeling a mosaic of granulocytes, natural killer and lymphoproliferative disorders following allogeneic stem cell transplantation. J Clin T cells from cord-blood and third-party donors. Nat Clin Pract Oncol 2008; 5: Oncol 2013; 31:39–48. 291–295. 25 Gattinoni L, Lugli E, Ji Y, Pos Z, Paulos CM, Quigley MF et al. A human memory 39 Leen AM, Bollard CM, Mendizabal AM, Shpall EJ, Szabolcs P, Antin JH et al. T cell subset with stem cell-like properties. Nat Med 2011; 17: 1290–1297. Multicenter study of banked third-party virus-specific T cells to treat severe viral 26 Stemberger C, Huster KM, Koffler M, Anderl F, Schiemann M, Wagner H et al. infections after hematopoietic stem cell transplantation. Blood 2013; 121: A single naive CD8+ T cell precursor can develop into diverse effector and 5113–5123. memory subsets. Immunity 2007; 27: 985–997. 40 Walter EA, Greenberg PD, Gilbert MJ, Finch RJ, Watanabe KS, Thomas ED et al. 27 Buchholz VR, Flossdorf M, Hensel I, Kretschmer L, Weissbrich B, Graf P et al. Reconstitution of cellular immunity against cytomegalovirus in recipients of Disparate individual fates compose robust CD8+ T cell immunity. Science 2013; allogeneic bone marrow by transfer of T-cell clones from the donor. N Engl J Med. 340: 630–635. 1995; 333:1038–1044. 28 Riddell SR, Watanabe KS, Goodrich JM, Li CR, Agha ME, Greenberg PD. Restoration 41 Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD et al. Antitumor activity of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science 1991; 257:238–241. and long-term fate of chimeric antigen receptor-positive T cells in patients with 118 – 29 Schmitt A, Tonn T, Busch DH, Grigoleit GU, Einsele H, Odendahl M et al. Adoptive neuroblastoma. Blood 2011; : 6050 6056. transfer and selective reconstitution of streptamer-selected cytomegalovirus- 42 Gerdemann U, Keirnan JM, Katari UL, Yanagisawa R, Christin AS, Huye LE et al. fi specific CD8+ T cells leads to virus clearance in patients after allogeneic Rapidly generated multivirus-speci c cytotoxic T lymphocytes for the prophylaxis 20 – peripheral blood stem cell transplantation. Transfusion 2011; 51:591–599. and treatment of viral infections. Mol Ther 2012; : 1622 1632. 30 Rauser G, Einsele H, Sinzger C, Wernet D, Kuntz G, Assenmacher M et al. Rapid 43 Stemberger C, Graef P, Odendahl M, Albrecht J, Dössinger G, Anderl F et al. generation of combined CMV-specific CD4+ and CD8+ T-cell lines for adoptive Lowest numbers of primary CD8+ T cells can reconstitute protective immunity 124 – transfer into recipients of allogeneic stem cell transplants. Blood 2004; 103: upon adoptive immunotherapy. Blood 2014; : 628 637. 3565–3572. 44 Jedema I, van de Meent M, Pots J, Kester MG, van der Beek MT, Falkenburg JH. fi 31 Feuchtinger T, Opherk K, Bethge WA, Topp MS, Schuster FR, Weissinger EM et al. Successful generation of primary virus-speci c and anti-tumor T-cell responses Adoptive transfer of pp-65 specific T-cells for the treatment of chemorefractory from the naive donor T-cell repertoire is determined by the balance between cytomegalovirus disease or reactivation after haploidentical and matched antigen-specific precursor T cells and regulatory T cells. Haematologica 2011; 96: unrelated stem cell transplantation. Blood 2010; 116: 4360–4367. 1204–1212.

Bone Marrow Transplantation (2015) S51 – S54 © 2015 Macmillan Publishers Limited