Abstract
Hematopoietic stem cell transplantation has revolutionized the treatment of hematologic malignancies, but infection, graft-versus-host disease and relapse are still important problems. Calcineurin inhibitors, T-cell depletion strategies, and immunomodulators have helped to prevent graft-versus-host disease, but have a negative impact on the graft-versus-leukemia effect. T cells and natural killer cells are both thought to be important in the graft-versus-leukemia effect, and both cell types are amenable to ex vivo manipulation and clinical manufacture, making them versatile immunotherapeutics. We provide an overview of these immunotherapeutic strategies following hematopoietic stem cell transplantation, with discussions centered on natural killer and T-cell biology. We discuss the contributions of each cell type to graft-versus-leukemia effects, as well as the current research directions in the field as related to adoptive cell therapy after hematopoietic stem cell transplantation.Introduction
Hematopoietic stem cell transplantation (HSCT) has revolutionized the treatment of hematologic malignancies, bringing substantial improvements to survival outcomes for many patients.1 However, infection,2 graft-versus-host disease (GVHD),3 and relapse4 are still the most challenging sequelae to address to improve the outcomes of all patients after allogeneic transplantation.5 Over the last several decades, the introduction of calcineurin inhibitors, T-cell depletion strategies, and immunomodulators has helped to prevent GVHD, but at a cost - with inhibition of the donor-specific immune response including the graft-versus-tumor/leukemia (GVL) effect.3 Efforts have been made alone or in combination to increase GVL without increasing GVHD by: (i) optimizing conditioning regimens; (ii) selecting better matched donors; and (iii) administering GVHD prophylaxis.6 Perhaps the most direct method to restore the GVL effect would be to administer T cells (manipulated or unmanipulated), which mediate the GVL effect,7 and donor-derived natural killer (NK) cells.8 Both T and NK cells are thought to be the principal effector cells mediating the GVL effect (Figure 1):6 directly killing tumor cells through the Fas and perforin pathways, but also indirectly contributing to tumor lysis through the secretion of cytokines.9 From a therapeutic perspective, both cell types are amenable to ex vivo manipulation and clinical manufacture, thus making them versatile immunotherapeutics. We provide an overview of these two immunotherapeutic strategies following HSCT, with discussions centered on NK and T-cell biology and contributions to the GVL effect as well as the current research directions in the field as related to adoptive cell therapy after HSCT.
T-cell therapy after hematopoietic stem cell transplantation
T cells, along with B cells, comprise the major cellular components of the adaptive immune system (Figure 1). By rearranging gene segments during T-cell development, a large number of T cells with different T-cell receptors (TCR) are made that can potentially recognize an unlimited number of peptides in the context of MHC molecules. These T cells are primed to recognize foreign proteins expressed on malignant and non-self cells. Following recognition, T cells either directly lyse their targets by secreting powerful perforins and granzymes, or orchestrate a more potent immune response by secreting inflammatory cytokines and chemokines.10
The role of T cells in the GVL effect has long been established. An analysis of 2254 patients receiving bone marrow transplants for acute myeloid leukemia (AML), acute lymphoblastic leukemia, and chronic myeloid leukemia showed lower rates of relapse in patients with non-T-cell-depleted allografts with GVHD, compared to those receiving T-cell-depleted allografts without GVHD.11 This evidence was further supported by studies using donor lymphocyte infusions (especially in the setting of chronic myeloid leukemia).12 However, GVHD remains a problem with donor lymphocyte infusions, thereby necessitating the use of more specific populations of T cells to enhance the GVL effect, such as T cells targeting minor histocompatibility antigens, or leukemia-specific antigens.12
The two general strategies to manufacture T cells to exploit a GVL effect in the setting of HSCT are: (i) ex vivo expansion and (ii) genetic modification.
- Ex vivo expansion (Figure 2). This involves the selective proliferation of T cells expressing endogenous TCR that recognize tumor cells. This approach exploits repeated stimulation with antigens to expand large numbers of T cells.13 Ex vivo expansion of T cells has the advantage of decreasing alloreactivity in vitro,14 as a result of cell death or outgrowth by the antigen-specific T cells.15 Two important parameters involved in ex vivo expansion are the target antigens and the culture conditions. Target antigens include minor histocompatibility antigens and leukemia-specific antigens, which include, in some settings, viral antigens.16 Minor histocompatibility antigens are proteins that are expressed differently across individuals as a result of genetic polymorphisms.17 Leukemia-specific antigens, on the other hand, are proteins that are either mutated (e.g. bcr-abl), lineage-restricted (e.g. CD19), or overexpressed in malignancies while concomitantly absent or minimally expressed in healthy tissues.18 Culture conditions are optimized to present the best priming environment for T cells to encounter antigen, involving different antigen-presenting cells,19 stimulatory cytokines,20 and selection of sub-populations.21
- Genetic modification (Figure 3). Using various gene therapy vectors (retroviruses,22 lentiviruses,23 transposons24), investigators have been able to introduce new specificities onto T cells to allow for HLA-independent targeting of hematologic malignancies. Chimeric antigen receptor-modified T cells, in particular, have been used as both a bridge to transplant and as adjuvant therapies after HSCT. These modified cells are discussed in more detail below.
The first ex vivo-expanded T cells for therapy in HSCT began with studies of antiviral T cells and T cells recognizing minor histocompatibility antigens. The use of donor-derived Epstein Barr virus-specific T cells successfully prevented and treated Epstein Barr virus-associated post-transplant lymphoproliferative disease.2825 Furthermore, gene marking of the infused cells demonstrated that these tumor-specific T cells persist in the long term.25 Outside of the context of targeting viral tumor-associated antigens, investigators from Leiden University Medical Center and Seattle pioneered the use of T cells targeting minor histocompatibility antigens, demonstrating that T-cell clones recognizing these antigens can kill myeloid leukemic cells and inhibit leukemia growth in vitro.29 Moreover, T cells specific for minor histocompatibility antigens were expanded in vitro and infused into a patient with accelerated phase chronic myeloid leukemia after allogeneic transplant. Complete eradication of leukemic cells was achieved after three infusions of leukemia-reactive T cells.30 A more recent trial used ex vivo-expanded donor derived CD8 T cells against unknown minor histocompatibility antigens to treat leukemia relapse following allogeneic transplant. Donor T cells stimulated with recipient peripheral blood mononuclear cells, depleted of CD4 T cells, and selected for reactive clones capable of lysing transformed cells but not fibroblasts were administered to seven patients; four of whom had transient complete remissions.31
While the approaches described above show promise, very few hematologic malignancies have viral epitopes as targets, and often, identification of minor histocompatibility antigens that will not elicit GVHD is difficult. For these reasons, attempts have shifted towards generating leukemia-associated antigen-specific T cells. Studies have suggested that expansion of leukemia-associated antigen-specific T cells in the peripheral blood of patients after allogeneic HSCT contribute to the GVL effect.3332
Leukemia-associated antigens include an eclectic mix of proteins seen in hematologic malignancies,18 including cancer/testis antigens such as MAGE A3, developmental proteins such as WT1, and prosurvival/antiapoptotic proteins such as survivin. Several studies have shown that T cells targeting leukemia- and lymphoma-associated antigens can be generated from donors’ and patients’ peripheral blood mononuclear cells and show cytolytic activity against lymphoma cell lines and primary tumor cells in vitro.3734 For example, preliminary findings of WT1-specific T-cell immunotherapy from a phase I clinical trial show that these T cells are well tolerated and effective. Infusions of WT1-specific cytotoxic T lymphocytes into patients with AML, acute lymphoblastic leukemia, or myelodysplastic syndrome following allogeneic HSCT were able to transiently reduce or eliminate cells expressing WT1, without mediating toxicities or GVHD. These cells were generated from healthy donors of hematopoietic stem cells, using dendritic cells pulsed with a pool of overlapping peptides spanning the WT1 protein as antigen-presenting cells to stimulate and expand WT1-specific T cells.38 In a more recent study, T cells directed against the leukemia-associated antigens BCR-ABL, PR1, and WT1 infused after transplantation resulted in 7/14 patients with chronic myeloid leukemia remaining in molecular remission a median of 45 months after prophylactic infusion of cells.39
While the use of ex vivo-expanded T cells has advantages, especially given the ability of this approach to generate a product which targets multiple tumor antigens, they are still limited by the low affinity of their antigens as well as the HLA dependence of tumors which can downregulate their MHC. To overcome these limitations, T cells can be genetically modified to redirect their specificity. Two promising gene transfer technologies include: (i) optimized high affinity T-cell receptors and (ii) chimeric antigen receptors (CAR).40
- High affinity T-cell receptors. Certain clones from patients or healthy donors develop high affinity TCR purely by chance, and such TCR can confer high specificity onto their T cells. Cloning this TCR complex onto other cells is the basis of TCR gene transfer, and the technology has shown encouraging results against hematologic malignancies.41 However, a significant concern with TCR gene transfer is the potential mispairing that may occur between endogenous TCR and the introduced high affinity TCR. At best, the mispairing decreases effectiveness of the therapy,42 but at worst, the new TCR may recognize self-proteins43 and potentially cause harmful GVHD. Several methods have been proposed to address this, one approach being the use of short interfering RNA to silence endogenous TCR genes, which has shown promise in the setting of leukemia immunotherapy. A TCR recognizing WT1 was transduced into T cells along with short interfering RNA for endogenous TCR genes. The resultant TCR-transduced T cells derived from leukemia patients were cytolytic against autologous leukemia cells but not normal hematopoietic progenitor cells. Furthermore, these T cells were capable of eliciting anti-leukemia activity in mouse xenograft models without impairing hematopoiesis.44
- Chimeric antigen receptors. Another highly promising technology is the CAR approach, which circumvents some of the limitations of both TCR gene transfer T cells and ex vivo-expanded T cells since HLA restriction is not required. CAR-modified T cells can theoretically recognize any target (not just proteins) in an HLA-independent manner with significantly enhanced potency.45 CAR are composed of an extracellular recognition domain (usually derived from the variable regions of an antibody) coupled to intracellular signaling domains that combine both signal 1 (TCR complex) and signal 2 (co-stimulatory molecule signaling) from the T cells.46 CAR were first designed by Eshhar et al. who evaluated whether the antibody complex can confer new specificity onto T cells.47 The best experience to date with CAR-modified T cells in HSCT involve CAR that recognize CD19, present on B-cell malignancies.48 Although CD19 is also present in healthy B cells, clinical experience with patients with common variable immune deficiency and patients treated with rituximab suggest that B-cell depletion is manageable.49
The earliest experiences with CD19 CAR T cells were disappointing due to a general lack of persistence (less than a week) that coincided with poor clinical responses. These cells incorporated so-called “first generation” CAR, which only used the TCR zeta chain as the sole signaling domain.50 Subsequent studies suggested that the addition of co-stimulatory molecules in the construct (i.e. “second generation” CAR) could provide improved persistence and antitumor activity in murine models and in patients.51 The group at the Baylor College of Medicine infused two populations of T cells: (i) T cells expressing a first-generation CD19 CAR and (ii) T cells expressing a second-generation CD19 CAR (including the CD28 co-stimulatory domain). Second-generation CAR T cells persisted longer than their first-generation counterparts, demonstrating the ability of co-stimulation to enhance T-cell proliferation and persistence. However, the efficacy of the second-generation CAR T cells in this study was limited.52 Several key improvements then paved the way for the remarkable antitumor efficacies reported in literature. One improvement involved utilizing a lymphodepletion regimen to enhance persistence of CAR T cells by eliminating competing endogenous cells.53 Using a chemotherapy regimen comprising cyclophosphamide and fludarabine followed by infusion of CD19 CAR T cells, Rosenberg’s group at the National Cancer Institute observed remissions from progressive B-cell malignancies in six of eight patients.53 Another change introduced at the University of Pennsylvania was a lentivirus vector expressing 41BBL instead of CD28 as the co-stimulatory signaling domain. Infusion of these lentivirus-modified T cells following customized chemotherapy regimens initially produced impressive responses in two of three patients with chronic lymphocytic leukemia.5423 Estimates suggested that each infused CAR-expressing T cell eradicated more than a 1,000 chronic lymphocytic leukemia cells.54 Subsequently, even more dramatic successes have been observed using second-generation CAR-CD19 transduced T cells for patients with acute lymphoblastic leukemia.55 Several groups have demonstrated response rates ranging from 70–100% in some patients with poor prognosis ALL.5755 It is, however, still difficult to determine the optimal CAR approach since each protocol varies in terms of CAR design, T-cell production, prior conditioning chemotherapy, and tumor burden.58
Several lines of inquiry are being explored in our efforts to improve the use of antitumor T cells after HSCT, including: (i) improving safety; (ii) improving activity and persistence in vivo; (iii) decreasing manufacturing time; (iv) increasing the number of antigens being targeted; and (v) conferring protection against immune suppression.
- Improving safety. One concern with T-cell immunotherapies is toxicity. This is especially a concern when gene-modified T-cell approaches are utilized (e.g. using CAR with potent co-stimulatory signaling domains). Severe adverse events including cytokine storms have been observed in patients who have antitumor responses following CAR T-cell therapies.53,55 In an attempt to address such “predictable” adverse events, investigators have developed management plans aimed at curtailing the effects of cytokines. For example, it was known that interleukin (IL)-6 mediates the cytokine release syndrome and immunosuppression with an IL-6 receptor antibody was shown to be capable of reversing the syndrome, so treatment algorithms have been proposed.59 Safety switches have also been incorporated into T cells. One recent suicide gene approach is the use of the inducible caspase 9 system, in which the administration of an inert dimerizing agent that brings together two halves of a protease involved in initiating and perpetuating the apoptotic cascade results in rapid elimination of the gene-modified T cells and rapid reversal of clinical symptoms.60,61
- Improving activity and persistence. Currently, efforts are underway to determine the efficacy of CD19-CAR T cells in phase II/III studies. Attempts to develop CAR targeting other tumor targets are now a substantial focus of this field.62 Additionally, to further improve the efficacy of CAR-modified T cells some groups are exploring the incorporation of two or more co-stimulatory domains (so-called third-generation CAR)62,63 or combining them with other antibody recognition domains (so-called tandem CAR).64 Addition of cytokine signals such as IL-2165 and IL-1566 has also been explored. Other attempts to improve in vivo persistence involve utilizing the endogenous signaling provided by latent viruses67,68 which have been explored clinically in neuroblastoma and B-cell leukemias/lymphomas.69,70 Most recently, improvements within the construct itself have also allowed improvements in CAR T-cell persistence and efficacy.71
- Decreasing production time. One active avenue of research involves improvements in the ex vivo expansion of both antigen-specific T cells and CAR T cells. Prolonged culture times cause anergy in cell populations, hence efforts are underway to decrease the amount of time cells remain in culture by using novel bioreactors that allow improved gas exchange and surface area (and consequently more rapid expansion)72 or by using different cell populations such as central memory-derived,73 naïve-derived,74 or stem-cell memory-derived T cells.74,75
- Increasing target antigens. One of the advantages of ex vivo expansion using peptide mixes is the ability to increase the number of target antigens used to stimulate T cells. T cells targeting multiple proteins have been generated using this method,20 and further increases in antigen can be accomplished in vivo using epigenetic modifying drugs.76,77
- Engineering resistance to immune suppression. Finally, efforts are also underway to confer greater resistance to cells against the immunosuppressive microenvironment mediated by tumors. To counteract the suppressive cytokine transforming growth factor-beta (TGFβ), for example, T cells can be genetically modified with a mutated dominant negative TGFβ type II receptor (DNR) that prevents the formation of the functional tetrameric TGFβ.78 Modification of T cells expressing this DNR allows them to negate signals from the inhibitory cytokine. DNR-transduced cytotoxic T lymphocytes were resistant to the anti-proliferative effects of TGFβ and adoptive transfer of TGFβ-DNR transduced T cells resulted in eradication of tumor cells in vivo.79 Other current limitations of T-cell therapies are summarized in Table 1.
Natural killer cell therapy after hematopoietic stem cell transplantation
NK cells, in contrast to T cells and B cells, are lymphocytes of the innate immune response (Figure 1). These cells have receptors that have been predetermined by the germline (i.e., no further rearrangements/increased diversity occur during development).10 NK cells have diverse activating and inhibitory receptors (another unique feature, in contrast to T cells) – with the corresponding ligands providing signals for activation or inhibition, respectively, of NK cell activity. A balance of these opposing signals determines whether NK cells exert their powerful activities or remain tolerant (Figure 4).80 The absence of a corresponding matched inhibitory ligand, for example, is often enough signal for the NK cell to eliminate the defective target (the “missing self” hypothesis of NK cell activation).81 But in a remarkable evolutionary design, only NK cells with inhibitory receptors that have previously encountered self ligands (from interacting with their own hematopoietic cells) are “licensed” to kill non-self-expressing target cells.8 As components of the innate immune response, most NK cells are already licensed for activity, and can directly mediate cytotoxicity or cytokine secretion upon recognizing their allogeneic targets.10
It is now understood that killer cell immunoglobulin-like receptor (KIR)-ligand mismatches (with the “missing” ligand present on donor NK cells and absent on recipient cells, including malignant cells) account for the GVL effect exhibited by NK cells.6 An added benefit to the GVL effect elicited by NK cells is the proposed concomitant protection against GVHD. NK cells also target the recipient’s antigen-presenting cells, thereby decreasing T-cell mediated GVHD,82 while “ignoring” healthy cells which do not express ligands for the NK cells’ activating receptors.83 Evidence for a NK-cell-mediated GVL effect was seen in both murine models of leukemia, in which allogeneic NK cells eradicated disease and caused myeloablation without causing GVHD,82 as well as in clinical experience with haploidentical donor transplants, where patients with AML achieved higher survival rates in the presence of alloreactivity than in the absence of alloreactive mismatches.8482 NK cells are, therefore, believed to be the potent effector cells responsible for the antitumor efficacy of haploidentical stem cell transplants against myeloid leukemias,8584 and most “immunotherapies” utilizing NK cells are being developed in the context of a haplotype-mismatched HSCT.8 Nevertheless, not all patients benefit from an NK-mediated GVL effect following engraftment, possibly because of delayed reconstitution of functional NK cells.86 As a result, groups are exploring the direct expansion and subsequent infusion of NK cells after HSCT. This has been made possible by advances in purification and selection methods of these innate effectors.
The most common method of purifying NK cells from donor sources involves the selective depletion of T-cell populations (e.g. using magnetic depletion of CD3 populations), followed by a further purification step for CD56 populations8887 and/or subsequent enrichment with cytokines (Figure 5). Mononuclear cells are subjected to immunomagnetic selection to remove unwanted CD3 T-cell populations. Although subsequent purification steps yielded higher purity, a substantially increased processing time (and consequent lower recovery) was needed.87 Enrichment steps involve the cytokines IL-2, IL-15, or various feeder cells (K562 cells of myeloid lineage modified to express IL-15,89 irradiated autologous peripheral blood mononuclear cells in the presence of IL-2,90 or autologous mesenchymal stromal cells91). Both IL-2 and IL-15 appear equally efficacious cytokines for expanding NK cells ex vivo and no added benefit was seen when both were combined or combined with other cytokines (e.g. IL-7).92 The use of mitogen-activated feeder cells has previously been shown to increase proliferation, lytic activity, and purity of expanded NK cells.93 Some groups have also investigated the ex vivo differentiation of NK cells from stem cell progenitors by first expanding CD34 cells using static culture bags, and subsequently differentiating the stem cells using a cytokine cocktail comprising stem cell factor, IL-7, IL-15, and IL-2.94
Numerous groups have evaluated the safety and feasibility of infusing allogeneic NK cells in the HSCT setting, particularly against AML. A group in Korea infused ex vivo-expanded NK cells derived from CD34 progenitor cells into patients with acute leukemia and myelodysplastic syndrome after HLA-mismatched HSCT. Although no acute toxicity was observed, six patients developed acute or chronic GVHD after NK cell infusion.95 A group in Switzerland administered purified allogeneic NK cells on days 3, 40, and 100 after haploidentical T-cell-depleted HSCT. The NK infusions did not, however, seem to have an effect on relapse rates (compared to those in historical controls).96 A group in France reported in vivo expansion of infused alloreactive NK cells in a patient who received NK cells and IL-2 for relapsed AML following haploidentical HSCT and salvage chemotherapy.97 While the NK cells detected in vivo retained their activity as measured in functional in vitro assays, the patient still relapsed after NK cell infusion and subsequently died of his disease.97 A group from the University of Minnesota used a regulatory T-cell-depleting protein, the IL-2 diphtheria fusion protein, to suppress regulatory T-cell activity that limits NK cell function. Patients treated with the toxin and NK infusion had higher IL-15 levels and improved efficacy of haploidentical NK cell therapy for AML.98 Finally, a group from the University of Arkansas reported the use of KIR mismatched haploidentical NK cells in the setting of an autologous HSCT for multiple myeloma. The donors’ NK cells persisted transiently after infusion, but five patients still relapsed early, two patients had progressive disease, one patient had stable disease, and two patients subsequently relapsed.99 As evidenced from these studies, more research is still needed to identify the optimal NK cell product and the optimal setting for the successful use of these cells after HSCT.
Efforts to further enhance NK cells have led investigators to explore genetically modified NK cells to both redefine their specificity and/or enhance their potency. To extend NK cell activity against lymphocytic leukemias, for example, one group genetically modified the NK cell line NK-92 with CAR recognizing CD19 or CD20. This redirection of specificity enabled these cells to eliminate previously NK cell-resistant tumors in murine models.100 Similarly, another group targeted multiple myeloma cells by transfecting NK cells with an anti-CD138 CAR.101 Investigators have also modified NK cells to express granzyme B following activation by their ligands to augment NK-mediated killing of tumor cells.102 Finally, to better protect NK cells from the NK cell-inhibitory cytokine TGFβ, NK cells (NK-92) were genetically modified to express a dominant negative TGFβ receptor designed to neutralize TGFβ signaling. These cells were rendered resistant to the suppressive effects of the cytokine and were able to mediate antitumor activity.103
Several lines of inquiry are being explored in efforts to expand use of NK cells clinically, including: (i) optimizing the choice of mismatch when selecting donor NK cells; (ii) inclusion of conditioning regimens before NK cell infusion; (iii) exploring co-administration with T cells; and (iv) exploring the use of NK cells outside the HSCT setting.
- Choice of mismatch. Current mismatch algorithms for NK cell infusions involve three KIR ligands: HLAC1 alleles, HLAC2 alleles, and the Bw4 epitope found in HLA-B alleles. Removal of inhibitory ligands appears to be the primary method of activating NK cells. While mismatches involving inhibitory receptors have normally been used to guide selection of donor NK cells some settings appear to be dominated by activating KIR. Furthermore, some alloreactive clones become functional upon engagement via activating KIR. Consequently, transplants utilizing KIR-B haplotype NK cells have been observed to mediate lower relapse rates and improved survival because they contain more activating than inhibitory receptors.104
- Conditioning regimens. Similar to the experience with T cells, lymphodepleting regimens seem to provide a better hematopoietic niche for NK cell expansion and persistence in vivo, and several investigators have incorporated such conditioning regimens into NK infusion protocols. The importance of a lymphodepleted state was demonstrated in a study comparing a low intensity regimen and a high intensity regimen. Patients given the low intensity regimen followed by NK cell administration only showed transient NK cell persistence with poor expansion in vivo. In contrast, marked in vivo expansion of infused allogeneic NK cells with a concomitant induction of hematologic remission was observed in patients with poor prognosis disease who received the high intensity regimen.105 Other studies, however, suggest that lower intensity regimens may be sufficient, with results depending on the population of patients being treated.106 Combination with immunomodulatory drugs may also be an option. For example, lenalidomide has been used in patients after HSCT both to activate and increase NK cells,107 and co-administration with NK cell infusion may lead to better outcomes as the immunomodulation increases CD107 expression, interferon production and degranulation of NK cells.108
- Co-administration with T cells. The combination of NK cells and T cells as a single immunotherapeutic strategy is appealing. While NK cells provide rapid, innate activity against tumors, T cells will provide long-lasting adaptive immune activity against the disease. Because of the ability of NK cells to target recipient antigen-presenting cells, it has been suggested that T-cell infusions can be better tolerated with NK infusions with less probability of causing GVHD.82 However, some studies suggest that memory T cells impair the development and activity of NK cells in vivo,109 and more naïve T-cell populations may be better suited as “partners” for NK cells.8
- Use outside the hematopoietic stem cell transplant setting. Finally, several researchers have attempted to extend the benefits of haploidentical transplants in situations in which the procedure is too toxic for potential recipients – by directly substituting allogeneic NK cells in lieu of HSCT. A group from the University of Minnesota induced remissions in patients with poor prognosis AML treated with high-dose cyclophosphamide and fludarabine followed by NK cell infusions and subcutaneous IL-2.105 A group from Bologna, Italy, infused purified CD56+CD3− NK cells derived from KIR-ligand mismatched donors following fludarabine/cyclophosphamide lymphodepletion into elderly patients with AML not otherwise eligible for allogeneic transplants. Infused cells demonstrated alloreactivity against leukemia blasts, in poor prognosis populations.110 Finally, the group from St. Jude Children’s Research Hospital infused KIR-mismatched NK cells into pediatric patients with AML as prophylaxis following chemotherapy. These cell infusions were well tolerated and the NK cells successfully engrafted and patients remained in remission for a median of >2.5 years.106 Other limitations of NK cell therapies are summarized in Table 1.
Overall summary
Improved methods of generating T cells and NK cells have facilitated the development of novel immunotherapeutic approaches that can augment and potentially even supplant allogeneic HSCT for hematologic malignancies. Advances in genetic modification technologies will only serve to improve the antitumor properties of these cells in vivo. As indications are broadened, manufacturing protocols optimized, and safety issues addressed, cellular therapies may yet become the standard of care for the treatment of hematologic malignancies as adjunct to, bridge before, or replacement for allogeneic HSCT.
Footnotes
- Authorship and Disclosures Information on authorship, contributions, and financial & other disclosures was provided by the authors and is available with the online version of this article at www.haematologica.org.
- Received January 7, 2015.
- Accepted May 6, 2015.
References
- Spitzer TR, Dey BR, Chen YB, Attar E, Ballen KK. The expanding frontier of hematopoietic cell transplantation. Cytometry B Clin Cytom. 2012; 82(5):271-279. PubMedGoogle Scholar
- Fuji S, Kapp M, Einsele H. Monitoring of pathogen-specific T-cell immune reconstitution after allogeneic hematopoietic stem cell transplantation. Front Immunol. 2013; 4:276. PubMedGoogle Scholar
- Choi SW, Reddy P. Current and emerging strategies for the prevention of graft-versus-host disease. Nat Rev Clin Oncol. 2014; 11(9):536-547. PubMedhttps://doi.org/10.1038/nrclinonc.2014.102Google Scholar
- Rambaldi A, Biagi E, Bonini C, Biondi A, Introna M. Cell-based strategies to manage leukemia relapse: efficacy and feasibility of immunotherapy approaches. Leukemia. 2015; 29(1):1-10. PubMedhttps://doi.org/10.1038/leu.2014.189Google Scholar
- Arnaout K, Patel N, Jain M, El-Amm J, Amro F, Tabbara IA. Complications of allogeneic hematopoietic stem cell transplantation. Cancer Invest. 2014; 32(7):349-362. PubMedhttps://doi.org/10.3109/07357907.2014.919301Google Scholar
- Barrett AJ. Understanding and harnessing the graft-versus-leukaemia effect. Br J Haematol. 2008; 142(6):877-888. PubMedhttps://doi.org/10.1111/j.1365-2141.2008.07260.xGoogle Scholar
- Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008; 112(12):4371-4383. PubMedhttps://doi.org/10.1182/blood-2008-03-077974Google Scholar
- Velardi A, Ruggeri L, Mancusi A, Aversa F, Christiansen FT. Natural killer cell allorecognition of missing self in allogeneic hematopoietic transplantation: a tool for immunotherapy of leukemia. Curr Opin Immunol. 2009; 21(5):525-530. PubMedhttps://doi.org/10.1016/j.coi.2009.07.015Google Scholar
- Ringden O. Immunotherapy by allogeneic stem cell transplantation. Adv Cancer Res. 2007; 97:25-60. PubMedGoogle Scholar
- Janeway C. Immunobiology: the Immune System in Health and Disease. Garland Science: New York; 2005. Google Scholar
- Horowitz MM, Gale RP, Sondel PM. Graft-versus-leukemia reactions after bone marrow transplantation. Blood. 1990; 75(3):555-562. PubMedGoogle Scholar
- Kolb HJ, Schmid C, Barrett AJ, Schendel DJ. Graft-versus-leukemia reactions in allogeneic chimeras. Blood. 2004; 103(3):767-776. PubMedhttps://doi.org/10.1182/blood-2003-02-0342Google Scholar
- Rooney CM, Leen AM, Vera JF, Heslop HE. T lymphocytes targeting native receptors. Immunol Rev. 2014; 257(1):39-55. PubMedhttps://doi.org/10.1111/imr.12133Google Scholar
- Bollard CM, Rooney CM, Heslop HE. T-cell therapy in the treatment of post-transplant lymphoproliferative disease. Nat Rev Clin Oncol. 2012; 9(9):510-519. PubMedhttps://doi.org/10.1038/nrclinonc.2012.111Google Scholar
- Melenhorst JJ, Leen AM, Bollard CM. Allogeneic virus-specific T cells with HLA alloreactivity do not produce GVHD in human subjects. Blood. 2010; 116(22):4700-4702. PubMedhttps://doi.org/10.1182/blood-2010-06-289991Google Scholar
- Zilberberg J, Feinman R, Korngold R. Strategies for the identification of T cell-recognized tumor antigens in hematological malignancies for improved graft-versus-tumor responses after allogeneic blood and marrow transplantation. Biol Blood Marrow Transplant. 2015; 21(6):1000-1007. PubMedhttps://doi.org/10.1016/j.bbmt.2014.11.001Google Scholar
- Bleakley M, Riddell SR. Exploiting T cells specific for human minor histocompatibility antigens for therapy of leukemia. Immunol Cell Biol. 2011; 89(3):396-407. PubMedhttps://doi.org/10.1038/icb.2010.124Google Scholar
- Anguille S, Van Tendeloo VF, Berneman ZN. Leukemia-associated antigens and their relevance to the immunotherapy of acute myeloid leukemia. Leukemia. 2012; 26(10):2186-2196. PubMedhttps://doi.org/10.1038/leu.2012.145Google Scholar
- Ngo MC, Ando J, Leen AM. Complementation of antigen-presenting cells to generate T lymphocytes with broad target specificity. J Immunother. 2014; 37(4):193-203. https://doi.org/10.1097/CJI.0000000000000014Google Scholar
- Gerdemann U, Katari U, Christin AS. Cytotoxic T lymphocytes simultaneously targeting multiple tumor-associated antigens to treat EBV negative lymphoma. Mol Ther. 2011; 19(12):2258-2268. PubMedhttps://doi.org/10.1038/mt.2011.167Google Scholar
- Barrett DM, Singh N, Liu X. Relation of clinical culture method to T-cell memory status and efficacy in xenograft models of adoptive immunotherapy. Cytotherapy. 2014; 16(5):619-630. PubMedhttps://doi.org/10.1016/j.jcyt.2013.10.013Google Scholar
- Hollyman D, Stefanski J, Przybylowski M. Manufacturing validation of biologically functional T cells targeted to CD19 antigen for autologous adoptive cell therapy. J Immunother. 2009; 32(2):169-180. https://doi.org/10.1097/CJI.0b013e318194a6e8Google Scholar
- Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011; 365(8):725-733. PubMedhttps://doi.org/10.1056/NEJMoa1103849Google Scholar
- Huang X, Guo H, Kang J. Sleeping Beauty transposon-mediated engineering of human primary T cells for therapy of CD19+ lymphoid malignancies. Mol Ther. 2008; 16(3):580-589. PubMedhttps://doi.org/10.1038/sj.mt.6300404Google Scholar
- Heslop HE, Slobod KS, Pule MA. Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood. 2010; 115(5):925-935. PubMedhttps://doi.org/10.1182/blood-2009-08-239186Google Scholar
- Heslop HE, Ng CY, Li C. Long-term restoration of immunity against Epstein-Barr virus infection by adoptive transfer of gene-modified virus-specific T lymphocytes. Nat Med. 1996; 2(5):551-555. PubMedhttps://doi.org/10.1038/nm0596-551Google Scholar
- Leen AM, Myers GD, Sili U. Monoculture-derived T lymphocytes specific for multiple viruses expand and produce clinically relevant effects in immunocompromised individuals. Nat Med. 2006; 12(10):1160-1166. PubMedhttps://doi.org/10.1038/nm1475Google Scholar
- Rooney CM, Smith CA, Ng CY. Use of gene-modified virus-specific T lymphocytes to control Epstein-Barr-virus-related lymphoproliferation. Lancet. 1995; 345(8941):9-13. PubMedhttps://doi.org/10.1016/S0140-6736(95)91150-2Google Scholar
- Falkenburg JH, Goselink HM, van der Harst D. Growth inhibition of clonogenic leukemic precursor cells by minor histocompatibility antigen-specific cytotoxic T lymphocytes. J Exp Med. 1991; 174(1):27-33. PubMedhttps://doi.org/10.1084/jem.174.1.27Google Scholar
- Falkenburg JH, Wafelman AR, Joosten P. Complete remission of accelerated phase chronic myeloid leukemia by treatment with leukemia-reactive cytotoxic T lymphocytes. Blood. 1999; 94(4):1201-1208. PubMedGoogle Scholar
- Warren EH, Fujii N, Akatsuka Y. Therapy of relapsed leukemia after allogeneic hematopoietic cell transplantation with T cells specific for minor histocompatibility antigens. Blood. 2010; 115(19):3869-3878. PubMedhttps://doi.org/10.1182/blood-2009-10-248997Google Scholar
- Rezvani K, Yong AS, Mielke S. Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies. Blood. 2008; 111(1):236-242. PubMedhttps://doi.org/10.1182/blood-2007-08-108241Google Scholar
- Barrett AJ, Rezvani K, Solomon S. New developments in allotransplant immunology. Hematology Am Soc Hematol Educ Program. 2003;350-371. Google Scholar
- Weber G, Caruana I, Rouce RH. Generation of tumor antigen-specific T cell lines from pediatric patients with acute lymphoblastic leukemia–implications for immunotherapy. Clin Cancer Res. 2013; 19(18):5079-5091. PubMedhttps://doi.org/10.1158/1078-0432.CCR-13-0955Google Scholar
- Weber G, Gerdemann U, Caruana I. Generation of multi-leukemia antigen-specific T cells to enhance the graft-versus-leukemia effect after allogeneic stem cell transplant. Leukemia. 2013; 27(7):1538-1547. PubMedhttps://doi.org/10.1038/leu.2013.66Google Scholar
- Quintarelli C, Dotti G, De Angelis B. Cytotoxic T lymphocytes directed to the preferentially expressed antigen of melanoma (PRAME) target chronic myeloid leukemia. Blood. 2008; 112(5):1876-1885. PubMedhttps://doi.org/10.1182/blood-2008-04-150045Google Scholar
- Quintarelli C, Dotti G, Hasan ST. High-avidity cytotoxic T lymphocytes specific for a new PRAME-derived peptide can target leukemic and leukemic-precursor cells. Blood. 2011; 117(12):3353-3362. PubMedhttps://doi.org/10.1182/blood-2010-08-300376Google Scholar
- O’Reilly RJ, Dao T, Koehne G, Scheinberg D, Doubrovina E. Adoptive transfer of unselected or leukemia-reactive T-cells in the treatment of relapse following allogeneic hematopoietic cell transplantation. Semin Immunol. 2010; 22(3):162-172. PubMedhttps://doi.org/10.1016/j.smim.2010.02.003Google Scholar
- Bornhauser M, Thiede C, Platzbecker U. Prophylactic transfer of BCR-ABL-, PR1-, and WT1-reactive donor T cells after T cell-depleted allogeneic hematopoietic cell transplantation in patients with chronic myeloid leukemia. Blood. 2011; 117(26):7174-7184. PubMedhttps://doi.org/10.1182/blood-2010-09-308569Google Scholar
- Fujiwara H. Adoptive immunotherapy for hematological malignancies using T cells gene-modified to express tumor antigen-specific receptors. Pharmaceuticals (Basel). 2014; 7(12):1049-1068. PubMedhttps://doi.org/10.3390/ph7121049Google Scholar
- Nicholson E, Ghorashian S, Stauss H. Improving TCR gene therapy for treatment of haematological malignancies. Adv Hematol. 2012; 2012:404081. PubMedGoogle Scholar
- Morgan RA, Dudley ME, Wunderlich JR. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006; 314(5796):126-129. PubMedhttps://doi.org/10.1126/science.1129003Google Scholar
- Linette GP, Stadtmauer EA, Maus MV. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood. 2013; 122(6):863-871. PubMedhttps://doi.org/10.1182/blood-2013-03-490565Google Scholar
- Ochi T, Fujiwara H, Okamoto S. Novel adoptive T-cell immunotherapy using a WT1-specific TCR vector encoding silencers for endogenous TCRs shows marked antileukemia reactivity and safety. Blood. 2011; 118(6):1495-1503. PubMedhttps://doi.org/10.1182/blood-2011-02-337089Google Scholar
- Eshhar Z. Adoptive cancer immunotherapy using genetically engineered designer T-cells: first steps into the clinic. Curr Opin Mol Ther. 2010; 12(1):55-63. PubMedGoogle Scholar
- Eshhar Z. Tumor-specific T-bodies: towards clinical application. Cancer Immunol Immunother. 1997; 45(3–4):131-136. PubMedhttps://doi.org/10.1007/s002620050415Google Scholar
- Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci USA. 1993; 90(2):720-724. PubMedhttps://doi.org/10.1073/pnas.90.2.720Google Scholar
- Ramos CA, Savoldo B, Dotti G. CD19-CAR trials. Cancer J. 2014; 20(2):112-118. PubMedhttps://doi.org/10.1097/PPO.0000000000000031Google Scholar
- Grillo-Lopez AJ, White CA, Dallaire BK. Rituximab: the first monoclonal antibody approved for the treatment of lymphoma. Curr Pharm Biotechnol. 2000; 1(1):1-9. PubMedhttps://doi.org/10.2174/1389201003379059Google Scholar
- Jensen MC, Popplewell L, Cooper LJ. Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specific chimeric antigen receptor redirected T cells in humans. Biol Blood Marrow Transplant. 2010; 16(9):1245-1256. PubMedhttps://doi.org/10.1016/j.bbmt.2010.03.014Google Scholar
- Kowolik CM, Topp MS, Gonzalez S. CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Res. 2006; 66(22):10995-11004. PubMedhttps://doi.org/10.1158/0008-5472.CAN-06-0160Google Scholar
- Savoldo B, Ramos CA, Liu E. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest. 2011; 121(5):1822-1826. PubMedhttps://doi.org/10.1172/JCI46110Google Scholar
- Kochenderfer JN, Dudley ME, Feldman SA. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood. 2012; 119(12):2709-2720. PubMedhttps://doi.org/10.1182/blood-2011-10-384388Google Scholar
- Kalos M, Levine BL, Porter DL. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011; 3(95):95ra73. PubMedhttps://doi.org/10.1126/scitranslmed.3002842Google Scholar
- Grupp SA, Kalos M, Barrett D. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013; 368(16):1509-1518. PubMedhttps://doi.org/10.1056/NEJMoa1215134Google Scholar
- Davila ML, Riviere I, Wang X. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014; 6(224):224ra225. Google Scholar
- Brentjens RJ, Davila ML, Riviere I. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013; 5(177):177ra138. Google Scholar
- Brentjens RJ, Curran KJ. Novel cellular therapies for leukemia: CAR-modified T cells targeted to the CD19 antigen. Hematology Am Soc Hematol Educ Program. 2012; 2012:143-151. PubMedhttps://doi.org/10.1182/asheducation-2012.1.143Google Scholar
- Lee DW, Gardner R, Porter DL. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014; 124(2):188-195. PubMedhttps://doi.org/10.1182/blood-2014-05-552729Google Scholar
- Zhou X, Di Stasi A, Tey SK. Long-term outcome after haploidentical stem cell transplant and infusion of T cells expressing the inducible caspase 9 safety transgene. Blood. 2014; 123(25):3895-3905. PubMedhttps://doi.org/10.1182/blood-2014-01-551671Google Scholar
- Di Stasi A, Tey SK, Dotti G. Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med. 2011; 365(18):1673-1683. PubMedhttps://doi.org/10.1056/NEJMoa1106152Google Scholar
- Curran KJ, Pegram HJ, Brentjens RJ. Chimeric antigen receptors for T cell immunotherapy: current understanding and future directions. J Gene Med. 2012; 14(6):405-415. PubMedGoogle Scholar
- Jena B, Dotti G, Cooper LJ. Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor. Blood. 2010; 116(7):1035-1044. PubMedhttps://doi.org/10.1182/blood-2010-01-043737Google Scholar
- Grada Z, Hegde M, Byrd T. TanCAR: a novel bispecific chimeric antigen receptor for cancer immunotherapy. Mol Ther Nucleic Acids. 2013; 2:e105. https://doi.org/10.1038/mtna.2013.32Google Scholar
- Singh H, Figliola MJ, Dawson MJ. Reprogramming CD19-specific T cells with IL-21 signaling can improve adoptive immunotherapy of B-lineage malignancies. Cancer Res. 2011; 71(10):3516-3527. PubMedhttps://doi.org/10.1158/0008-5472.CAN-10-3843Google Scholar
- Hoyos V, Savoldo B, Quintarelli C. Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety. Leukemia. 2010; 24(6):1160-1170. PubMedhttps://doi.org/10.1038/leu.2010.75Google Scholar
- Terakura S, Yamamoto TN, Gardner RA, Turtle CJ, Jensen MC, Riddell SR. Generation of CD19-chimeric antigen receptor modified CD8+ T cells derived from virus-specific central memory T cells. Blood. 2012; 119(1):72-82. PubMedhttps://doi.org/10.1182/blood-2011-07-366419Google Scholar
- Micklethwaite KP, Savoldo B, Hanley PJ. Derivation of human T lymphocytes from cord blood and peripheral blood with antiviral and antileukemic specificity from a single culture as protection against infection and relapse after stem cell transplantation. Blood. 2010; 115(13):2695-2703. PubMedhttps://doi.org/10.1182/blood-2009-09-242263Google Scholar
- Pule MA, Savoldo B, Myers GD. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008; 14(11):1264-1270. PubMedhttps://doi.org/10.1038/nm.1882Google Scholar
- Cruz CR, Micklethwaite KP, Savoldo B. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood. 2013; 122(17):2965-2973. PubMedhttps://doi.org/10.1182/blood-2013-06-506741Google Scholar
- Jonnalagadda M, Mardiros A, Urak R. Chimeric antigen receptors with mutated IgG4 Fc spacer avoid Fc receptor binding and improve T cell persistence and antitumor efficacy. Mol Ther. 2015; 23(4):757-768. PubMedhttps://doi.org/10.1038/mt.2014.208Google Scholar
- Vera JF, Brenner LJ, Gerdemann U. Accelerated production of antigen-specific T cells for preclinical and clinical applications using gas-permeable rapid expansion cultureware (G-Rex). J Immunother. 2010; 33(3):305-315. https://doi.org/10.1097/CJI.0b013e3181c0c3cbGoogle Scholar
- Wang X, Naranjo A, Brown CE. Phenotypic and functional attributes of lentivirus-modified CD19-specific human CD8+ central memory T cells manufactured at clinical scale. J Immunother. 2012; 35(9):689-701. https://doi.org/10.1097/CJI.0b013e318270dec7Google Scholar
- Albrecht J, Frey M, Teschner D. IL-21-treated naive CD45RA+ CD8+ T cells represent a reliable source for producing leukemia-reactive cytotoxic T lymphocytes with high proliferative potential and early differentiation phenotype. Cancer Immunol Immunother. 2011; 60(2):235-248. PubMedhttps://doi.org/10.1007/s00262-010-0936-8Google Scholar
- Gattinoni L, Lugli E, Ji Y. A human memory T cell subset with stem cell-like properties. Nat Med. 2011; 17(10):1290-1297. PubMedhttps://doi.org/10.1038/nm.2446Google Scholar
- Goodyear O, Agathanggelou A, Novitzky-Basso I. Induction of a CD8+ T-cell response to the MAGE cancer testis antigen by combined treatment with azacitidine and sodium valproate in patients with acute myeloid leukemia and myelodysplasia. Blood. 2010; 116(11):1908-1918. PubMedhttps://doi.org/10.1182/blood-2009-11-249474Google Scholar
- Cruz CR, Gerdemann U, Leen AM. Improving T-cell therapy for relapsed EBV-negative Hodgkin lymphoma by targeting upregulated MAGE-A4. Clin Cancer Res. 2011; 17(22):7058-7066. PubMedhttps://doi.org/10.1158/1078-0432.CCR-11-1873Google Scholar
- Bollard CM, Rossig C, Calonge MJ. Adapting a transforming growth factor beta-related tumor protection strategy to enhance antitumor immunity. Blood. 2002; 99(9):3179-3187. PubMedhttps://doi.org/10.1182/blood.V99.9.3179Google Scholar
- Foster AE, Dotti G, Lu A. Antitumor activity of EBV-specific T lymphocytes transduced with a dominant negative TGF-beta receptor. J Immunother. 2008; 31(5):500-505. https://doi.org/10.1097/CJI.0b013e318177092bGoogle Scholar
- Pegram HJ, Andrews DM, Smyth MJ, Darcy PK, Kershaw MH. Activating and inhibitory receptors of natural killer cells. Immunol Cell Biol. 2011; 89(2):216-224. PubMedhttps://doi.org/10.1038/icb.2010.78Google Scholar
- Karre K. NK cells, MHC class I molecules and the missing self. Scand J Immunol. 2002; 55(3):221-228. PubMedhttps://doi.org/10.1046/j.1365-3083.2002.01053.xGoogle Scholar
- Ruggeri L, Capanni M, Urbani E. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002; 295(5562):2097-2100. PubMedhttps://doi.org/10.1126/science.1068440Google Scholar
- Locatelli F, Merli P, Rutella S. At the bedside: innate immunity as an immunotherapy tool for hematological malignancies. J Leukoc Biol. 2013; 94(6):1141-1157. PubMedhttps://doi.org/10.1189/jlb.0613343Google Scholar
- Cooley S, Weisdorf DJ, Guethlein LA. Donor selection for natural killer cell receptor genes leads to superior survival after unrelated transplantation for acute myelogenous leukemia. Blood. 2010; 116(14):2411-2419. PubMedhttps://doi.org/10.1182/blood-2010-05-283051Google Scholar
- Pende D, Marcenaro S, Falco M. Anti-leukemia activity of alloreactive NK cells in KIR ligand-mismatched haploidentical HSCT for pediatric patients: evaluation of the functional role of activating KIR and redefinition of inhibitory KIR specificity. Blood. 2009; 113(13):3119-3129. PubMedhttps://doi.org/10.1182/blood-2008-06-164103Google Scholar
- Zhao XY, Chang YJ, Huang XJ. Conflicting impact of alloreactive NK cells on transplantation outcomes after haploidentical transplantation: do the reconstitution kinetics of natural killer cells create these differences?. Biol Blood Marrow Transplant. 2011; 17(10):1436-1442. PubMedhttps://doi.org/10.1016/j.bbmt.2011.05.020Google Scholar
- McKenna DH, Sumstad D, Bostrom N. Good manufacturing practices production of natural killer cells for immunotherapy: a six-year single-institution experience. Transfusion. 2007; 47(3):520-528. https://doi.org/10.1111/j.1537-2995.2006.01145.xGoogle Scholar
- Koehl U, Brehm C, Huenecke S. Clinical grade purification and expansion of NK cell products for an optimized manufacturing protocol. Front Oncol. 2013; 3:118. PubMedGoogle Scholar
- Lapteva N, Durett AG, Sun J. Large-scale ex vivo expansion and characterization of natural killer cells for clinical applications. Cytotherapy. 2012; 14(9):1131-1143. PubMedhttps://doi.org/10.3109/14653249.2012.700767Google Scholar
- Lim O, Lee Y, Chung H. GMP-compliant, large-scale expanded allogeneic natural killer cells have potent cytolytic activity against cancer cells in vitro and in vivo. PloS One. 2013; 8(1):e53611. PubMedhttps://doi.org/10.1371/journal.pone.0053611Google Scholar
- Boissel L, Tuncer HH, Betancur M, Wolfberg A, Klingemann H. Umbilical cord mesenchymal stem cells increase expansion of cord blood natural killer cells. Biol Blood Marrow Transplant. 2008; 14(9):1031-1038. PubMedhttps://doi.org/10.1016/j.bbmt.2008.06.016Google Scholar
- Decot V, Voillard L, Latger-Cannard V. Natural-killer cell amplification for adoptive leukemia relapse immunotherapy: comparison of three cytokines, IL-2, IL-15, or IL-7 and impact on NKG2D, KIR2DL1, and KIR2DL2 expression. Exp Hematol. 2010; 38(5):351-362. PubMedhttps://doi.org/10.1016/j.exphem.2010.02.006Google Scholar
- Rabinowich H, Sedlmayr P, Herberman RB, Whiteside TL. Increased proliferation, lytic activity, and purity of human natural killer cells cocultured with mitogen-activated feeder cells. Cell Immunol. 1991; 135(2):454-470. PubMedhttps://doi.org/10.1016/0008-8749(91)90290-RGoogle Scholar
- Spanholtz J, Preijers F, Tordoir M. Clinical-grade generation of active NK cells from cord blood hematopoietic progenitor cells for immunotherapy using a closed-system culture process. PloS One. 2011; 6(6):e20740. PubMedhttps://doi.org/10.1371/journal.pone.0020740Google Scholar
- Yoon SR, Lee YS, Yang SH. Generation of donor natural killer cells from CD34(+) progenitor cells and subsequent infusion after HLA-mismatched allogeneic hematopoietic cell transplantation: a feasibility study. Bone Marrow Transplant. 2010; 45(6):1038-1046. PubMedhttps://doi.org/10.1038/bmt.2009.304Google Scholar
- Stern M, Passweg JR, Meyer-Monard S. Pre-emptive immunotherapy with purified natural killer cells after haploidentical SCT: a prospective phase II study in two centers. Bone Marrow Transplant. 2013; 48(3):433-438. PubMedhttps://doi.org/10.1038/bmt.2012.162Google Scholar
- Nguyen S, Beziat V, Norol F. Infusion of allogeneic natural killer cells in a patient with acute myeloid leukemia in relapse after haploidentical hematopoietic stem cell transplantation. Transfusion. 2011; 51(8):1769-1778. Google Scholar
- Bachanova V, Cooley S, Defor TE. Clearance of acute myeloid leukemia by haploidentical natural killer cells is improved using IL-2 diphtheria toxin fusion protein. Blood. 2014; 123(25):3855-3863. PubMedhttps://doi.org/10.1182/blood-2013-10-532531Google Scholar
- Shi J, Tricot G, Szmania S. Infusion of haploidentical killer immunoglobulin-like receptor ligand mismatched NK cells for relapsed myeloma in the setting of autologous stem cell transplantation. Br J Haematol. 2008; 143(5):641-653. PubMedhttps://doi.org/10.1111/j.1365-2141.2008.07340.xGoogle Scholar
- Boissel L, Betancur-Boissel M, Lu W. Retargeting NK-92 cells by means of CD19-and CD20-specific chimeric antigen receptors compares favorably with antibody-dependent cellular cytotoxicity. Oncoimmunology. 2013; 2(10):e26527. PubMedhttps://doi.org/10.4161/onci.26527Google Scholar
- Jiang H, Zhang W, Shang P. Transfection of chimeric anti-CD138 gene enhances natural killer cell activation and killing of multiple myeloma cells. Mol Oncol. 2014; 8(2):297-310. PubMedhttps://doi.org/10.1016/j.molonc.2013.12.001Google Scholar
- Oberoi P, Jabulowsky RA, Bahr-Mahmud H, Wels WS. EGFR-targeted granzyme B expressed in NK cells enhances natural cytotoxicity and mediates specific killing of tumor cells. PloS One. 2013; 8(4):e61267. PubMedhttps://doi.org/10.1371/journal.pone.0061267Google Scholar
- Yang B, Liu H, Shi W. Blocking transforming growth factor-beta signaling pathway augments antitumor effect of adoptive NK-92 cell therapy. Int Immunopharmacol. 2013; 17(2):198-204. PubMedhttps://doi.org/10.1016/j.intimp.2013.06.003Google Scholar
- Miller JS. Therapeutic applications: natural killer cells in the clinic. Hematology Am Soc Hematol Educ Program. 2013; 2013:247-253. PubMedhttps://doi.org/10.1182/asheducation-2013.1.247Google Scholar
- Miller JS, Soignier Y, Panoskaltsis-Mortari A. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005; 105(8):3051-3057. PubMedhttps://doi.org/10.1182/blood-2004-07-2974Google Scholar
- Rubnitz JE, Inaba H, Ribeiro RC. NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J Clin Oncol. 2010; 28(6):955-959. PubMedhttps://doi.org/10.1200/JCO.2009.24.4590Google Scholar
- Lioznov M, El-Cheikh J, Hoffmann F. Lenalidomide as salvage therapy after allo-SCT for multiple myeloma is effective and leads to an increase of activated NK (NKp44(+)) and T (HLA-DR(+)) cells. Bone Marrow Transplant. 2010; 45(2):349-353. PubMedhttps://doi.org/10.1038/bmt.2009.155Google Scholar
- Jungkunz-Stier I, Zekl M, Stuhmer T, Einsele H, Seggewiss-Bernhardt R. Modulation of natural killer cell effector functions through lenalidomide/dasatinib and their combined effects against multiple myeloma cells. Leuk Lymphoma. 2014; 55(1):168-176. PubMedhttps://doi.org/10.3109/10428194.2013.794270Google Scholar
- Vago L, Forno B, Sormani MP. Temporal, quantitative, and functional characteristics of single-KIR-positive alloreactive natural killer cell recovery account for impaired graft-versus-leukemia activity after haploidentical hematopoietic stem cell transplantation. Blood. 2008; 112(8):3488-3499. PubMedhttps://doi.org/10.1182/blood-2007-07-103325Google Scholar
- Curti A, Ruggeri L, D’Addio A. Successful transfer of alloreactive haploidentical KIR ligand-mismatched natural killer cells after infusion in elderly high risk acute myeloid leukemia patients. Blood. 2011; 118(12):3273-3279. PubMedhttps://doi.org/10.1182/blood-2011-01-329508Google Scholar
- Haso W, Lee DW, Shah NN. Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood. 2013; 121(7):1165-1174. PubMedhttps://doi.org/10.1182/blood-2012-06-438002Google Scholar
- Bonini C, Ferrari G, Verzeletti S. HSV-TK gene transfer into donor lymphocytes for control of allogeneic graft-versus-leukemia. Science. 1997; 276(5319):1719-1724. PubMedhttps://doi.org/10.1126/science.276.5319.1719Google Scholar
- Cruz CR, Micklethwaite KP, Savoldo B. Infusion of donor-derived CD19-redirected-virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase I study. Blood. 2013; 122(17):2965-2973. PubMedhttps://doi.org/10.1182/blood-2013-06-506741Google Scholar
- Ouseph S, Tappitake D, Armant M. Cellular therapies clinical research roadmap: lessons learned on how to move a cellular therapy into a clinical trial. Cytotherapy. 2015; 17(4):339-343. PubMedhttps://doi.org/10.1016/j.jcyt.2014.10.008Google Scholar
- Leen AM, Bollard CM, Mendizabal AM. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood. 2013; 121(26):5113-5123. PubMedhttps://doi.org/10.1182/blood-2013-02-486324Google Scholar
- Lee DW, Kochenderfer JN, Stetler-Stevenson M. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015; 385(9967):517-528. PubMedhttps://doi.org/10.1016/S0140-6736(14)61403-3Google Scholar
- Finn OJ. Immunooncology: understanding the function and dysfunction of the immune system in cancer. Ann Oncol. 2012; 23(Suppl 8):viii6-9. PubMedhttps://doi.org/10.1093/annonc/mds256Google Scholar
- Ruggeri L, Mancusi A, Capanni M. Donor natural killer cell allorecognition of missing self in haploidentical hematopoietic transplantation for acute myeloid leukemia: challenging its predictive value. Blood. 2007; 110(1):433-440. PubMedhttps://doi.org/10.1182/blood-2006-07-038687Google Scholar
- Gardiner CM, Guethlein LA, Shilling HG. Different NK cell surface phenotypes defined by the DX9 antibody are due to KIR3DL1 gene polymorphism. J Immunol. 2001; 166(5):2992-3001. PubMedhttps://doi.org/10.4049/jimmunol.166.5.2992Google Scholar
- Chan HW, Kurago ZB, Stewart CA. DNA methylation maintains allele-specific KIR gene expression in human natural killer cells. J Exp Med. 2003; 197(2):245-255. PubMedhttps://doi.org/10.1084/jem.20021127Google Scholar
- Ljunggren HG, Malmberg KJ. Prospects for the use of NK cells in immunotherapy of human cancer. Nat Rev Immunol. 2007; 7(5):329-339. PubMedhttps://doi.org/10.1038/nri2073Google Scholar
- Szmania S, Lapteva N, Garg T. Ex vivo-expanded natural killer cells demonstrate robust proliferation in vivo in high-risk relapsed multiple myeloma patients. J Immunother. 2015; 38(1):24-36. https://doi.org/10.1097/CJI.0000000000000059Google Scholar
- Baragano Raneros A, Martin-Palanco V, Fernandez AF. Methylation of NKG2D ligands contributes to immune system evasion in acute myeloid leukemia. Genes Immun. 2015; 16(1):71-82. PubMedhttps://doi.org/10.1038/gene.2014.58Google Scholar
- Reiners KS, Topolar D, Henke A. Soluble ligands for NK cell receptors promote evasion of chronic lymphocytic leukemia cells from NK cell antitumor activity. Blood. 2013; 121(18):3658-3665. PubMedhttps://doi.org/10.1182/blood-2013-01-476606Google Scholar