Summary
As outcomes of hematopoietic stem cell transplantation have improved over time, disease relapse has emerged as the most important causes of treatment failure. Cellular therapy represents a promising therapeutic approach, not only to treat or prevent disease relapse after hematopoietic stem cell transplantation, but also conceivably to help patients achieve remission prior to transplantation or as consolidation therapy for high risk hematologic malignancies. Of the many cellular therapies available, NK cells infusion may be the most promising approach due to strong innate activity of NK cells in vitro and in vivo against malignant or virally infected cells. Published clinical data consists only of small feasibility studies of inadequate size to show clinical benefit. Here we review the current status of clinical investigation using NK cell therapy for patients with hematologic malignancies undergoing hematopoietic stem cell transplantation.
Keywords: NK cells, NK cell expansion, cellular therapy, haploidentical stem cell transplantation
Introduction
Human Natural Killer (NK) cells play a critical role in host defense against infections and malignant transformations [1]. They comprise of approximately 10–15% of all blood lymphocytes and are identified by the lack of CD3 and the presence of CD56 and/or CD16 expression on cell surface. There are two subsets of NK cells that differ phenotypically and functionally. The majority of circulating NK cells (90%) have CD16 and low CD56 surface expression, termed CD56dim, and are important in direct and Antibody Dependent target Cell Cytotoxicity (ADCC) [2]. ADCC is mediated by CD16, the low-affinity Fc receptor which binds the Fc portion of IgG antibody [3]. These NK cells can release granules containing containing perforins, which form a pore in the cell membrane, and granzymes that enter target cells to mediate apoptosis [4]. They can also express death receptor ligands such as Fas ligand (FasL) and Tumor Necrosis Factor (TNF) Apoptosis-Inducing Ligand (TRAIL) [5].
A minority of peripheral NK cells and 100% of NK cells in secondary lymphoid tissue (SLT) express high CD56 and low CD16, which are less cytotoxic but secrete more cytokines [6]. These CD56bright NK cells have high levels of CD94/NKG2 family receptors with only very small fractions of Killer cell Immunoglobulin-like Receptors (KIRs) [7]. They express the high and intermediate affinity Interleukin 2 (IL-2) receptors and expand in vivo and in vitro in response to low doses of IL-2 [8]. Upon stimulation, they produce high levels of cytokines while cytotoxic activity and ADCC are minimal. However, cytotoxic activity can still be induced using IL-2 [9]. They also express the receptor tyrosine kinase ckit whose ligand augments IL-2 induced proliferation [8]. Since CD56bright NK cells are dominant in SLT, it has been postulated that CD56bright cells are less mature than CD56dim cells. Activation of SLT CD56bright NK cells increases expression of NK receptors that are characteristic of the CD56dim such as CD16 and KIRs [10]. Recently, a novel subset of CD34dim hematopoietic progenitor cells that reside in lymph nodes was identified and is capable of differentiating into CD56bright NK cells through a four-stage model described by Freud and Caligiuri [11].
NK cell cytotoxicity is triggered when the balance of activating and inhibiting receptor signals is tipped towards activation. Inhibitory receptors primarily recognize Major Histocompatibility Complex(MHC) class I or class-I like molecules, preventing NK-mediated cell lysis of normal MHC-expressing cells. The main group of NK receptors, KIRs, have cytoplasmic domains comprised of a short immunoreceptor tyrosine-based activating motif (ITAM) or a long immunoreceptor tyrosine-based inhibition motif (ITIM) [12]. Fifteen KIR genes and two pseudogenes have been identified on chromosome 19 and are extremely polymorphic [101]. Other receptors include C-type Lectin Receptors and Natural Cytotoxic receptors (NCR). The NCR (NKp30, NKp44, NKp46 and NKp80) are activating receptors mostly restricted to expression on NK cells [13]. The C-type Lectin receptors in the NKG2 family are expressed as heterodimers with CD94 except NKG2D which is expressed as homodimer[12].
The “missing-self hypothesis”, which postulates that NK cells recognize and destroy autologous cells with lost or altered self-MHC class I molecules [14], is an oversimplification of NK cell regulation, since MHC class I is not always required to protect from NK cytotoxicity nor is it always sufficient to prevent NK cytotoxicity[15]. Activation of NK cells with cytokines including IL-2 or IL-15 may be required to enhance the cytotoxicity and function of NK cells [15]. There have been several models used for predicting NK cell alloreactivity. Ruggeri et al. initially described the ligand-ligand mismatch model when there is incompatibility between the donor KIR ligand and recipient KIR ligand (i.e., ligand-ligand mismatch if the donor has a ligand that is absent in the recipient) [16]. However, we now recognize that KIR genotype has a profound effect on the KIR repertoire and that the expression of KIR genes is not entirely dependent on expression of their HLA ligands, but is controlled by promoter methylation [17]. In addition, it is important to note that possession of a KIR gene does not imply its expression on NK cells [18]. Thus, Leung et al. defined mismatch in the receptor-ligand model as incompatibility between the donor KIR and recipient KIR ligand on the basis of expression (i.e., receptor-ligand mismatch if the donor has an inhibitory receptor for which the cognate ligand is absent in the recipient) [19]. A third model was described by Gagne et al. in which incompatibility was defined as a mismatch between the donor KIR and recipient KIR receptors (i.e., receptor-receptor mismatch if the donor has a receptor that is absent in the recipient) [20]. The last model can be considered the prototype for the subsequent ‘KIR haplotype model’ proposed by the Stanford group, which is founded on the notion that the more activating KIR genes the donor carries, the higher the potential for alloreactivity. The ‘A’ KIR haplotype has only one activating gene (KIR2DS4), whereas the ‘B’ KIR haplotype has more than one activating KIR gene, including KIR2DS1 and KIR2DS2. Because KIR ‘B’ haplotype carries more activating genes than the ‘A’ haplotype, transplants from donors with the ‘B’ haplotype are predicted to have more prominent NK cell-mediated anti-tumor effects [21].
NK cell expansion
Even with leukapheresis, a relatively small number of NK cells can be obtained from the peripheral blood of healthy donors, generally limiting adoptive immunotherapy to median cell doses of approximately 2 × 107 NK cells/kg body weight of the recipient [22]. Studies have shown that IL-2 increases the number of circulating NK cells; however giving high doses of IL-2 can is associated with significant side effects such as fever, hypotension, pulmonary edema, hepatic and renal toxicities, among others. Giving a low dose of IL-2 was found to have more effect on specific expansion of the CD56bright NK cell subset, which may have a regulatory rather than cytotoxic role because of expressing the higher affinity IL2Rα [8]. IL-2 may also affect T regulatory (T reg) cells which have the potential to counteract the antitumor effect of other immune effector cells including NK cells [23]. Increased levels of endogenous IL-15 are associated with in vivo expansion in human trials of haploidentical NK cell therapy [24, 25], but induction of sufficient IL-15 levels requires aggressive lymphodepleting chemotherapy. With this approach, persistence of NK cells adoptively transferred after lymphodepleting chemotherapy is detectable for 2–4 weeks in both the autologous [25], and haploidentical setting [24], with donor NK cells detectable at low levels in selected patients for up to 6 months.
Thus ex vivo expansion of NK cells is likely needed to maximize the therapeutic potential of this approach. Several expansion methods have been described for this purpose including cytokine support such as IL-2 [26] or anti-CD3 antibodies [27]. Data from these trials showed that ex-vivo purification and expansion of donor NK cells from leukapheresis products is technically feasible and safe. However, cytokines alone are not sufficient for optimal cellular proliferation. Additional signals from other cells are required, and support from mesenchymal stroma or genetically-modified artificial Antigen Presenting Cells (aAPCs) can also induce expansion [28]. Contact with K562 cells, a cell line derived from patient with myeloid blast crisis of chronic myeloid leukemia (CML) is known to induce modest proliferation of NK cells [29]. K562-based aAPC transduced with 4-1BBL (CD137L) and membrane-bound IL-15 (mIL-15) was used to achieve a mean expansion of 277-fold in 21 days; however, proliferation is limited by senescence attributed to telomere shortening [30]. We have found that membrane bound IL-21 (mIL-21) resulted in greater proliferation, longer telomere length, and less senescence than NK cells expanded with mIL-15, achieving a median 21,000-fold expansion in 21 days [31]. These expanded NK cells express high levels of NKG2D, NCRs and CD16. The high proliferation and activation was enabled in this system by IL-21R-mediated phosphorylation of Signal Transducer and Activator of Transcription 3 (STAT3) which upregulates NKG2D and telomerase expression (manuscript under review). If stimulatory cells are used, irradiation prior to co-culture is important to prevent their overgrowth and to ensure that no viable cells are infused with the cultured NK cells [32].
Infusion of NK cells after transplant
Several lines of evidence suggest that increased numbers of alloreactive NK cells would be of benefit in the transplant setting [33], despite the fact that retrospective results vary depending on patient population, underlying diseases, conditioning regimens, graft composition and cell dose, degree of T-cell depletion, post-transplant immunosuppressive regimens and differing methods of determining alloreactivity. Ruggeri et al. proposed the infusion of NK cells after transplant to target disease relapse based on a retrospective evaluation of NK alloreactivity in transplanted patients [15] and supportive NK models [16]. This group analyzed 92 patients with high risk leukemia who received a haplotype mismatched hematopoietic stem cell transplant (HSCT) and reported that NK cell alloreactivity improved engraftment, protected from graft-versus-host disease (GVHD) and reduced the rate of relapse. Using mouse xenograft models they demonstrated the role of alloreactive NK cells in eradicating human leukemia, improving engraftment by targeting host T lymphocytes and reducing GVHD by eliminating recipient-type dendritic cells[16]. In addition, the NK cell dose of the infusion product has been associated with better outcomes following matched sibling transplants [34].
There is little data on tissue migration of adoptively-transferred NK cells in humans. Meller et al reported migration of Indium-labeled NK cells to lung, liver, spleen, and bone marrow [35]. There are reports of successful PET-based imaging of NK cell lines in murine tumor models [36], suggesting that this modality may be applicable to human studies in the future. There is no definitive data regarding whether NK cells cross the blood brain barrier (BBB), although it is known that naive lymphocytes do not cross the BBB, while activated lymphocytes patrol the CNS freely and are actively recruited to sites of inflammation [37]. Whether this is also true for NK cells it is unknown.
A number of studies, listed in Table 1, have reported on the safety of NK cell infusion. While no dose-limiting toxicities have been described, none of the studies in the post-transplant setting were powered to assess the therapeutic effect of the NK cells. In a study by Koehl et. al, two pediatric patients with multiply-relapsed acute lymphoblastic leukemia (ALL) and one patient with acute myeloid leukemia (AML), age 6–16 years, were treated post haploidentical HSCT with repeated infusions of haploidentical KIR mismatched NK cells starting at day 1 post transplant then every 4–6 weeks after HSCT. Single doses of 3–34×106 CD56+CD3− cells/kg were infused. The AML patient received two infusions, while the first ALL patient received three infusions and the second ALL patient received one infusion. NK cell infusions were well tolerated without significant adverse events or GVHD [26]. Although all patients had active disease with blasts of 37–97% at the time of transplant, they achieved remission and complete donor chimerism by 4th week post transplant. The AML patient died of early relapse on day +80, while the ALL patients died of thrombotic-thrombocytopenic purpura and atypical viral pneumonia on days +45 and +152, respectively [26]. In a second study by the same authors, seven pediatric patients (1 AML, 4 ALL, 1 Hodgkin’s disease (HD), 1 rhabdomyosarcoma were treated with repeated infusions of NK cells at days +3, +40, +100 after haploidentical HSCT [38]. The patients received 6.6–32.3×106 CD56+CD3− cells/kg and 0.5–53.3×103 CD3+ cells/kg from the parental donors. No immediate adverse reactions were observed after the infusion. Three patients developed GVHD, one had grade III GVHD because of a high CD3+ cell dose associated with the NK cell infusion of 53.3×103 CD3+ cells/kg, and the other two patients because of a T-cell add back of 1 and 1.5×105 CD3+ cells/kg, respectively. The remaining 4 patients who received <20×103 CD3+ cells/kg had no signs of GVHD. Six of the 7 patients who received a median of 11.5×106 (7–29.6×106) CD34+ cells/kg showed moderate or fast immune reconstitution with full donor chimerism, while one patient transplanted with 13.6×106 CD34+ cells/kg had graft rejection. Five patients were alive in complete remission with a short follow up (4–13 months) post haploidentical HSCT [38]. Passweg et al. infused NK cells up to 14 × 106 cells/kg 3–12 months following T-cell depleted haploidentical HSCT to five patients (4 AML, 1 CML), ages 3–25 years. Indications were incomplete engraftment, early relapse, and graft failure. NK alloreactivity was present in 3 of 5 patients. All patients except one received at least 2 infusions. No GVHD or infections were observed. Two patients had an increase in donor chimerism, (one of these two received an additional stem cell dose after NK), one had stable mixed chimerism, one had decreasing chimerism and another patient experienced relapsed AML. Four of the five patients are alive and in continuous remission 8–18 months after the first NK infusion (median follow-up 12 months) [39]. In an update, Passweg reported that 3 of 5 treated patients were alive and in complete remission 18–36 months after NK infusion [40]. Slavin et al. used IL-2-activated NK cells in 8 patients with median age of 25 years (range 4–63), with relapsed hematological malignancies after HSCT. Patients received positively selected CD56+ cells prepared from rIL-2 activated donor lymphocytes. Donors were haploidentically mismatched (3 patients), matched siblings (4 patients) and matched unrelated (1 patient). Disease categories included acute leukemia and myelodysplastic syndrome (MDS) (n=5) patients and lymphoma (n=3) patients. Positively selected cells were median 39% CD56+ cells (range 30–71%), with a median of 3% contaminating CD3+ cells (range 2–21%). The median number of CD56+ cells infused was 120 × 106 (range 10–600 × 106). Cell infusion was uneventful and no GVHD was observed in all 8 treated patients [41]. One patient with relapsed ALL achieved remission after receiving 25 × 106/kg, but died 8 months later due to pulmonary aspergillosis, present on admission for HSCT. A second recipient of haploidentical HSCT from KIR non-alloreactive mother treated for refractory MDS was in complete remission more than 8 months post-transplant with no GVHD, after infusion of 2.5 × 106 NK cells 3 months post SCT. Five patients are alive, two with disease 18 and 20 months post transplantation and three with no evidence of disease after 8, 12 and 20 months post transplant [41]. Barkholt et al. evaluated the infusion of long-term ex vivo expanded allogeneic NK and NK-like T-cells following allogeneic HSCT in five patients ages 48–67 years (4 with solid cancers, 1 with chronic lymphocytic leukemia) after disease progression [27]. These patients received NK cell infusions at a median of 15 months post transplant. No major adverse effects or signs of acute GVHD were observed [27]. The CLL patient in this study received a stem cell boost for disease relapse post HSCT 11 months before NK infusion followed by three doses of non-KIR alloreactive NK cells (1, 9.6 and 17.3 ×106/kg). IL-2 in dose of 6 × 106 IU/m2/day was given for three consecutive days starting on the day of the first cell infusion. Unfortunately the patient relapsed and remained alive six months after the last infusion. Uharek et al. reported on 25 patients (16 AML, 5 ALL, 2 CML, 1 Hodgkin’s disease and 1 MDS patient) who received a haploidentical HSCT followed by infusion of purified CD56+CD3−NK cells on day +2. Preparatory regimen consisted of total body irradiation, thiotepa, fludarabine, and OKT3 [42]. NK cells were isolated from the CD34− fraction using an automated two-step procedure of CD3+ depletion and subsequent CD56+ selection. No other immunosuppression was routinely given. Patients received a mean of 9.8 × 106 CD56+CD3− NK cells/kg (range 1.61–32.2) with 2.89 × 104/kg (0.95–7.4) CD3+ cells. Most of the patients developed early GVHD of the skin (median onset 12 days), which promptly resolved after a short-term treatment with steroids and cyclosporine. The main cause of death consisted of infections (n=10), chronic GVHD (n=2), and relapse (n=4). After a median follow-up of 1442 days (3.9 years) (range 0.1 to 7.1 years) 9 of 25 patients are alive and in complete remission resulting in a 2-year overall survival of 29%. Out of 16 high-risk patients with AML, 10 had refractory or active disease prior to transplantation. In a matched pair analysis with EBMT database patients, the 16 AML patients treated with haploidentical HSCT plus NK cell infusions had superior 2-year overall survival (40 vs. 11%, p=0.02) [42]. Rizzieri et al. recently reported on a total of 51 NK cell-enriched donor lymphocyte infusions (DLIs) to 30 patients following a 3–6/6 HLA matched T-cell-depleted nonmyeloablative allogeneic transplant with alemtuzumab [43]. Eight weeks following transplantation, donor NK cell-enriched DLIs were processed using a CD56+ selecting column with up to 3 fresh infusions allowed. Fourteen matched and 16 mismatched transplanted patients received a total of 51 NK cell-enriched DLIs. The median number of CD3− CD56+ NK cells infused was 10.6 × 106 cells/kg and 9.21 × 106 cells/kg for matched and mismatched, respectively. The median number of contaminating CD3+CD56− T-cells infused was 0.53 × 106 and 0.27 × 106 in the matched and mismatched setting, respectively. All but 2 subjects had donor engraftment accounting for >80% of their hematopoiesis at the time of first infusion. After a median follow-up of 12 months for matched sibling donor transplants and 27 months for the haploidentical transplants, 1 year overall survival was 43% and 42%, respectively. Evaluating outcomes by disease, 1-year survival was 50% for the 19 patients with myeloid diseases and 29% for patients with lymphoid diseases. Only one patient each in the matched (n = 14) or mismatched (n = 16) groups experienced severe aGVHD with little other toxicity attributable to the infusions. Long-term responders had improved T-cell phenotypic recovery and improved duration of responses and overall survival after multiple NK cell-enriched infusions [43]. All of these trials are small pilot studies in diverse populations of lymphoid and myeloid malignancies. However, these studies have shown that infusion of NK cells is safe and feasible with potential for future larger studies. In a few of these studies some patients developed GVHD; however, since the infusions were given early after HSCT it is impossible to ascertain whether GVHD occurring in these patients was due to the transplant or the NK cell infusion. In some instances, the development of GVHD has been associated with less efficient T-cell depletion. For clinical application most of these trials used a large scale purification methods allowing automated, efficient and relatively rapid isolation of human NK cells.
Table 1.
Clinical studies using NK cell infusion post hematopoietic stem cell transplantation.
| Author | Disease and # of treated patients |
Age of patients |
Type of SCT |
Time of NK infusion post SCT |
Nk selection/ preparation |
dose | Phase of clinical trial |
aGVHD Y/N |
Response/outcome | Ref # |
|---|---|---|---|---|---|---|---|---|---|---|
| Koehl et al | ALL (2), AML(1) | 6–16 years | Haplo | D+1, wk4-6, wk8-12 | unstimulated leukaphereses, CD3 depletion, CD56 selection then NK were IL2 activated | Repeated infusions. Single dose 3–34×106 CD56+CD3−/Kg. Total per patient ranged 20–47.4/kg |
I | N | Although all patients showed blast at time of SCT, 3/3 CR and complete donor chimerism within 1 month postSCT. | 26 |
| Koehl et al. | AML (1), ALL (4), HD (1), RMS (1) | unkown | Haplo | D+3,+40, +100 | unstimulated leukaphereses CD3 depletion, CD56 selection |
Single dose: 6.6–32.3×106 CD56+CD3− cells/kg | I | Y (3 pts developed grade III GVHD, 1 who had NK with 53.3×10^3 CD3+/Kg and 2 post T cell add back. | 5/7 patients were alive in CR 4–13 months post SCT | 38 |
| Passweg et al. | AML (4), CML (1) With mixed chimerism or impending relapse |
3–25 (median 16) years | Haplo | Several wks to months post sct | unstimulated leukaphereses CD3 depletion, CD56 selection |
0.69–1.4×107/kg (med 0.21) | I | N | 3/5 pts were alive and CR 18–36 months after NK infusion 2/5 had increased donor chimerism |
39, 40 |
| Slavin et al. | MDS (2), AML (1), ALL (1), biphenotypic (1), HD (1); NHL (2). | 4–63 (median 25) years | 3 Haplo 4 MRD 1 MUD |
unknown | Positively selected CD56+ prepared from rIL2 activated donor lymphocytes | 10–600×106 (median 120× 106) | I | N | One pt with relapsed ALL and one with MDS had CR. 4 pts alive, 3 disease free 9–22 month post SCT. | 41 |
| Barkholt et al. | Solid tumors (4), CLL (1) | 48–67 years | MRD | Time from last DLI 2–26 months | expanded NK and NK-like T cells | Escalating doses of at 1-month intervals: 1 × 106, 10 × 106 and a median of 13.2 (range from 8.1 to 40.3) × 106 NK and NK-like T cells/kg. the last 3/6 got subcutaneous IL-2 injections. | I | N | CLL patient progressed | 27 |
| Uharek et al. | AML (16), ALL (5), CML (2), HD (1), MDS (1) | unknown | Haplo | D+2 | NK cells isolated from the CD34- fraction. CD3 depletion, CD56 selection |
of 9.8 × 106 CD56+CD3− NK cells/kg (range 1.61–32.2) | I/II | Most developed early GVHD of the skin which promptly resolved after short term treatment with steroids and CSA. | 9/25 patients alive and in CR, with 2-year OS of 29% | 42 |
| Rizzieri et al. | 30 patients with myeloid and lymphoid malignancies | unknown | 14 matched 16 mis-matched SCT | 1.5–3 months | unstimulated leukaphereses were enriched for NK cells using a CD56 antibody | median infused dose of 10.60 × 106 cells/kg | I/II | Y (14 pts, 6 /14 had grade I skin aGVHD) | Total of 16 patients in CR with 1 year OS of 43% and 42% of matched and mismatched respectively | 43 |
Figure legend: AML – acute myeloid leukemia, ALL – acute lymphoid leukmemia, MDS myelodysplastic syndrome, HD – Hodgkin’s disease, NHL – non-Hodgkin’s lymphoma, CLL – chronic lymphocytic leukemia, CML – chronic myeloid leukemia, OS – overall survival, RMS – rhabdomyosarcoma, CR – complete remission, SCT – stem cell transplantation, pts – patients, MRD – Matched Related Donor, MUD – Matched Unrelated Donor
We currently have an ongoing study to assess the safety of infusing alloreactive NK cells from a haploidentical relative and to determine the maximum tolerated dose of these cells given in combination with busulfan, fludarabine, antithymocyte globulin and allogeneic transplantation from a separate HLA matched donor for treatment of AML/MDS and to asses the rate of engraftment, GVHD, immune reconstitution and survival after infusion of alloreactive haploidentical NK cells effects.
Conclusions
Relapse remains the major cause of treatment failure after allogeneic stem cell transplantation. Alloreactive NK cells mediate a potent antileukemic effect and may enhance engraftment and reduce GVHD. Aforementioned studies demonstrated that the infusion of haploidentical alloreactive NK cells is safe and does not increase the incidence of GVHD. There are still many unanswered questions regarding the type of NK cells infused, optimal dose of NK cells and timing of infusions. Future studies will assess the effectiveness of increased doses of NK cells, made possible by ex vivo expansion, to treat or prevent disease relapse post-transplant and to explore the infusion of NK cells in non-transplant disease-specific trials,
Acknowledgement
This study was supported in part by a MD Anderson Cancer Center Institutional Research Grant to S.O.C.
References
- 1.Raulet DH, Guerra N. Oncogenic stress sensed by the immune system: role of natural killer cell receptors. Nat Rev Immunol. 2009;9(8):568–580. doi: 10.1038/nri2604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Cooper MA, Fehniger TA, Caligiuri MA. The biology of human natural killer-cell subsets. Trends Immunol. 2001;22(11):633–640. doi: 10.1016/s1471-4906(01)02060-9. [DOI] [PubMed] [Google Scholar]
- 3.Trinchieri G. Biology of natural killer cells. Adv Immunol. 1989;47:187–376. doi: 10.1016/S0065-2776(08)60664-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Browne KA, Blink E, Sutton VR, Froelich CJ, Jans DA, Trapani JA. Cytosolic delivery of granzyme B by bacterial toxins: evidence that endosomal disruption, in addition to transmembrane pore formation, is an important function of perforin. Mol Cell Biol. 1999;19(12):8604–8615. doi: 10.1128/mcb.19.12.8604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zamai L, Ahmad M, Bennett IM, Azzoni L, Alnemri ES, Perussia B. Natural killer (NK) cell-mediated cytotoxicity: differential use of TRAIL and Fas ligand by immature and mature primary human NK cells. J Exp Med. 1998;188(12):2375–2380. doi: 10.1084/jem.188.12.2375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Fehniger TA, Cooper MA, Nuovo GJ, et al. CD56bright natural killer cells are present in human lymph nodes and are activated by T cell-derived IL-2: a potential new link between adaptive and innate immunity. Blood. 2003;101(8):3052–3057. doi: 10.1182/blood-2002-09-2876. [DOI] [PubMed] [Google Scholar]
- 7.Nagler A, Lanier LL, Cwirla S, Phillips JH. Comparative studies of human FcRIII-positive and negative natural killer cells. J Immunol. 1989;143(10):3183–3191. [PubMed] [Google Scholar]
- 8.Nagler A, Lanier LL, Phillips JH. Constitutive expression of high affinity interleukin 2 receptors on human CD16-natural killer cells in vivo. J Exp Med. 1990;171(5):1527–1533. doi: 10.1084/jem.171.5.1527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Cooper MA, Fehniger TA, Turner SC, et al. Human natural killer cells: a unique innate immunoregulatory role for the CD56(bright) subset. Blood. 2001;97(10):3146–3151. doi: 10.1182/blood.v97.10.3146. [DOI] [PubMed] [Google Scholar]
- 10.Chan A, Hong DL, Atzberger A, et al. CD56bright human NK cells differentiate into CD56dim cells: role of contact with peripheral fibroblasts. J Immunol. 2007;179(1):89–94. doi: 10.4049/jimmunol.179.1.89. [DOI] [PubMed] [Google Scholar]
- 11.Freud AG, Becknell B, Roychowdhury S, et al. A human CD34(+) subset resides in lymph nodes and differentiates into CD56bright natural killer cells. Immunity. 2005;22(3):295–304. doi: 10.1016/j.immuni.2005.01.013. [DOI] [PubMed] [Google Scholar]
- 12.Lanier LL. NK cell receptors. Annu Rev Immunol. 1998;16:359–393. doi: 10.1146/annurev.immunol.16.1.359. [DOI] [PubMed] [Google Scholar]
- 13.Moretta A, Biassoni R, Bottino C, Mingari MC, Moretta L. Natural cytotoxicity receptors that trigger human NK-cell-mediated cytolysis. Immunol Today. 2000;21(5):228–234. doi: 10.1016/s0167-5699(00)01596-6. [DOI] [PubMed] [Google Scholar]
- 14.Ljunggren HG, Karre K. In search of the 'missing self': MHC molecules and NK cell recognition. Immunol Today. 1990;11(7):237–244. doi: 10.1016/0167-5699(90)90097-s. [DOI] [PubMed] [Google Scholar]
- 15.Farag SS, Caligiuri MA. Human natural killer cell development and biology. Blood Rev. 2006;20(3):123–137. doi: 10.1016/j.blre.2005.10.001. [DOI] [PubMed] [Google Scholar]
- 16.Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295(5562):2097–2100. doi: 10.1126/science.1068440. [DOI] [PubMed] [Google Scholar]
- 17.Santourlidis S, Trompeter HI, Weinhold S, et al. Crucial role of DNA methylation in determination of clonally distributed killer cell Ig-like receptor expression patterns in NK cells. J Immunol. 2002;169(8):4253–4261. doi: 10.4049/jimmunol.169.8.4253. [DOI] [PubMed] [Google Scholar]
- 18.Torkar M, Norgate Z, Colonna M, Trowsdale J, Wilson MJ. Isotypic variation of novel immunoglobulin-like transcript/killer cell inhibitory receptor loci in the leukocyte receptor complex. Eur J Immunol. 1998;28(12):3959–3967. doi: 10.1002/(SICI)1521-4141(199812)28:12<3959::AID-IMMU3959>3.0.CO;2-2. [DOI] [PubMed] [Google Scholar]
- 19.Leung W, Iyengar R, Turner V, et al. Determinants of antileukemia effects of allogeneic NK cells. J Immunol. 2004;172(1):644–650. doi: 10.4049/jimmunol.172.1.644. [DOI] [PubMed] [Google Scholar]
- 20.Gagne K, Brizard G, Gueglio B, et al. Relevance of KIR gene polymorphisms in bone marrow transplantation outcome. Hum Immunol. 2002;63(4):271–280. doi: 10.1016/s0198-8859(02)00373-7. [DOI] [PubMed] [Google Scholar]
- 21.Mcqueen KL, Dorighi KM, Guethlein LA, Wong R, Sanjanwala B, Parham P. Donor-recipient combinations of group A and B KIR haplotypes and HLA class I ligand affect the outcome of HLA-matched, sibling donor hematopoietic cell transplantation. Hum Immunol. 2007;68(5):309–323. doi: 10.1016/j.humimm.2007.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sutlu T, Alici E. Natural killer cell-based immunotherapy in cancer: current insights and future prospects. J Intern Med. 2009;266(2):154–181. doi: 10.1111/j.1365-2796.2009.02121.x. [DOI] [PubMed] [Google Scholar]
- 23.Shimizu M, Fontana A, Takeda Y, Yagita H, Yoshimoto T, Matsuzawa A. Induction of antitumor immunity with Fas/APO-1 ligand (CD95L)-transfected neuroblastoma neuro-2a cells. J Immunol. 1999;162(12):7350–7357. [PubMed] [Google Scholar]
- 24.Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105(8):3051–3057. doi: 10.1182/blood-2004-07-2974. [DOI] [PubMed] [Google Scholar]
- 25.Parkhurst MR, Riley JP, Dudley ME, Rosenberg SA. Adoptive transfer of autologous natural killer cells leads to high levels of circulating natural killer cells but does not mediate tumor regression. Clin Cancer Res. 2011;17(19):6287–6297. doi: 10.1158/1078-0432.CCR-11-1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Koehl U, Sorensen J, Esser R, et al. IL-2 activated NK cell immunotherapy of three children after haploidentical stem cell transplantation. Blood Cells Mol Dis. 2004;33(3):261–266. doi: 10.1016/j.bcmd.2004.08.013. [DOI] [PubMed] [Google Scholar]
- 27.Barkholt L, Alici E, Conrad R, et al. Safety analysis of ex vivo-expanded NK and NK-like T cells administered to cancer patients: a phase I clinical study. Immunotherapy. 2009;1(5):753–764. doi: 10.2217/imt.09.47. [DOI] [PubMed] [Google Scholar]
- 28.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. doi: 10.1016/j.bbmt.2008.06.016. [DOI] [PubMed] [Google Scholar]
- 29.Harada H, Saijo K, Watanabe S, et al. Selective expansion of human natural killer cells from peripheral blood mononuclear cells by the cell line, HFWT. Jpn J Cancer Res. 2002;93(3):313–319. doi: 10.1111/j.1349-7006.2002.tb02174.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Fujisaki H, Kakuda H, Imai C, Mullighan CG, Campana D. Replicative potential of human natural killer cells. Br J Haematol. 2009;145(5):606–613. doi: 10.1111/j.1365-2141.2009.07667.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Somanchi SS, Senyukov VV, Denman CJ, Lee DA. Expansion, purification, and functional assessment of human peripheral blood NK cells. J Vis Exp. 2011;(48) doi: 10.3791/2540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Cho D, Campana D. Expansion and activation of natural killer cells for cancer immunotherapy. Korean J Lab Med. 2009;29(2):89–96. doi: 10.3343/kjlm.2009.29.2.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Gill S, Olson JA, Negrin RS. Natural killer cells in allogeneic transplantation: effect on engraftment, graft- versus-tumor, and graft-versus-host responses. Biol Blood Marrow Transplant. 2009;15(7):765–776. doi: 10.1016/j.bbmt.2009.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kim DH, Sohn SK, Lee NY, et al. Transplantation with higher dose of natural killer cells associated with better outcomes in terms of non-relapse mortality and infectious events after allogeneic peripheral blood stem cell transplantation from HLA-matched sibling donors. Eur J Haematol. 2005;75(4):299–308. doi: 10.1111/j.1600-0609.2005.00514.x. [DOI] [PubMed] [Google Scholar]
- 35.Meller B, Frohn C, Brand JM, et al. Monitoring of a new approach of immunotherapy with allogenic (111)In-labelled NK cells in patients with renal cell carcinoma. Eur J Nucl Med Mol Imaging. 2004;31(3):403–407. doi: 10.1007/s00259-003-1398-4. [DOI] [PubMed] [Google Scholar]
- 36.Meier R, Piert M, Piontek G, et al. Tracking of [18F]FDG-labeled natural killer cells to HER2/neu-positive tumors. Nucl Med Biol. 2008;35(5):579–588. doi: 10.1016/j.nucmedbio.2008.02.006. [DOI] [PubMed] [Google Scholar]
- 37.Mitchell DA, Fecci PE, Sampson JH. Immunotherapy of malignant brain tumors. Immunol Rev. 2008;222:70–100. doi: 10.1111/j.1600-065X.2008.00603.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Koehl U, Sörensen J, Esser R, et al. Immunotherapy with highly purified CD56+CD3− natural killer cells for paediatric patients after haploidentical stem cell transplantation. 32nd Annual Meeting of the European Group for Blood an Marrow Transplantation.2006. [Google Scholar]
- 39.Passweg JR, Tichelli A, Meyer-Monard S, et al. Purified donor NK-lymphocyte infusion to consolidate engraftment after haploidentical stem cell transplantation. Leukemia. 2004;18(11):1835–1838. doi: 10.1038/sj.leu.2403524. [DOI] [PubMed] [Google Scholar]
- 40.Passweg JR, Koehl U, Uharek L, Meyer-Monard S, Tichelli A. Natural-killer-cell-based treatment in haematopoietic stem-cell transplantation. Best Pract Res Clin Haematol. 2006;19(4):811–824. doi: 10.1016/j.beha.2006.06.004. [DOI] [PubMed] [Google Scholar]
- 41.Slavin S, Morecki S, Shapira MY, et al. Use of matched or mismatched rIL-2 activated donor lymphocytes positively selected for CD56+ for immunotherapy of resistant leukemia after allogeneic stem cell transplantation. Journal of Clinical Oncology, 2004 ASCO Annual Meeting Proceedings. 2004;22(14S) [Google Scholar]
- 42.Uharek L, Friedrichs B, Nogai A, et al. Successful Treatment of High-Risk and Refractory Acute Myeloid Leukemia with haploidentical Stem Cell Transplantation Plus NK cell therapy. Blood (ASH Annual Meeting Abstracts) 2010;116(21):2370. [Google Scholar]
- 43.Rizzieri DA, Storms R, Chen DF, et al. Natural killer cell-enriched donor lymphocyte infusions from A 3–6/6 HLA matched family member following nonmyeloablative allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2010;16(8):1107–1114. doi: 10.1016/j.bbmt.2010.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
Website References
- 101. [Accessed 9/20/2011]; http://www.ebi.ac.uk/ipd/kir/genes.html.
