throbber
ReprintsOesk I 21612023 2:02:03 PM
`
`•
`
`Biology and clinical effects of natural killer cells in
`allogeneic transplantation
`Jonathan E. Benjamin, Saar Gill and Robert S. Negrin
`
`Division of Blood and Marrow Transplantation,
`Stanford University School of Medicine, Stanford,
`California, USA
`
`Correspondence to Robert S. Negrin, Division of Blood
`and Marrow Transplantation, Stanford University
`School ot Medicine, 300 Pasteur Drive H3249,
`Stanford, CA 94305-5627, USA
`Tel: + 1 650 723 0822; tax: + 1 650 725 8950;
`e-mail: negrs@stanford.edu
`
`Current Opinion in Oncology 2010, 22:130- 137
`
`Purpose of review
`Following allogeneic hematopoietic cell transplantation, donor-derived natural killer
`(NK) cells target recipient hematopoietic cells, result ing in an antileukemia effect and a
`lower incidence of graft rejection. NK cells do not mediate and may diminish graft versus
`host disease. Here we review the determinants of NK cell alloreactivity and their
`implications for adoptive NK cell therapy.
`Recent findings
`NK cell alloreactivity has been defined by the absence of recipient M HC class I ligands
`for donor inhibitory killer immunoglobulin-like receptor (KIR) receptors, as predicted by a
`number of algorithms. Recently, the role of activating NK receptors and their cognate
`ligands has received more attention. The beneficial clinical effect of NK-cell alloreactivity
`has not been uniformly demonstrated, likely reflecting differences in condit ioning
`regimens, graft components and posttransplant immune suppression. Investigations of
`NK cell phenotype and function after transplantation have helped demonstrate which
`NK cell subsets mediate the graft versus leukemia effect. These advances have
`proceeded in parallel with increasing facility in GMP-grade bulk purification and
`administration of NK cell preparations.
`Summary
`NK cells are a heterogeneous population of lymphocytes with diverse patterns of target(cid:173)
`cell recognition and effector function. Further clinical and functional correlations will
`help maximize 1heir potential for clinical benefit.
`
`Keywords
`adoptive immunotherapy, hematopoietic transplantation, natural killer cells
`
`Curr 0pin 0ncol 22:130-137
`0 2010 Wolters Kluwer Health I Lippincott Williams & Wilkins
`1040-8746
`
`Introduction
`Natural ki ller ( K) cells differentiate self from nonself
`by gauging the expression of M HC class I molecules on
`potential target cells. In the context of allogeneic he ma(cid:173)
`topoietic cell transplantation (HGT), particularly with
`HLA mismatched transplants, donor-derived K cells
`have been shown in some studies to influence the out(cid:173)
`come by a direct anti tumor effect as well as by mitigating
`graft versus host d isease (GVH D) and reducing the
`incidence of graft rejection. The optimal way to harness
`NK cell alloreactivity remains the subject of vigorous
`debate. In this article, we review the basic biology of NK
`cells in the context of recent clinical trials of H CT.
`
`sion and the presence or absence of the low affinity lgG
`receptor CD16. 'T'he CD56hrighcCD16" subset is enriched
`in lymphoid organs, secretes cytokines
`to help co(cid:173)
`ordinate adaptive immunity and is the major subtype
`recruited to sites of inflammation (including malignancy)
`[1,2]. In contrast, the CD56dimCD 16+ subset circulates in
`the peripheral blood and shows potent cytotoxicity [3].
`Several lines of evidence suggest that C D56hrighc K
`cells may differentiate into CD56°im K cells under
`some conditions [4,5]. K cells are reported to comprise
`5 - 25% of peripheral b lood lymphocytes, or approxi(cid:173)
`mately 100- 600cells/µl [6].
`
`Effector functions : lytic machinery
`K cell cytotoxicity requires ta rget-cell recognition and
`Human natural killer immunophenotype
`the receipt of an activating signal in the absence of an
`inhibitory signal. Killing is mediated by several pathways;
`NK cells do not express a rearranged germline-antigen
`receptor and they are identified by expression ofCD56 in
`whereas immature N K cells rely on tumor necrosis(cid:173)
`the absence of GD3. Two subsets of mature K cells are
`related apoptosis-inducing ligand (TRAIL)-mediated
`recognized, defined by the brightness of CD56 expres-
`killing [7], mature K cells preferentially utilize the
`1040-8746 «, 2010 Wolters Kluwer Health I Lippincott W illiams & Wilkins
`
`DOI: 10.1097 /CCO.0b013e328335a559
`
`Copyright© Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`UPenn Ex. 2056
`Miltenyi v. UPenn
`IPR2022-00855
`Page 130
`
`

`

`•
`
`ReprintsOesk I 21612023 2:02:03 PM
`
`granule-exocytosis pathway (requiring perforin and gran(cid:173)
`zymes) and the Fas- Fas ligand pathway (8]. In order to
`ach ieve the ir maximal cytotoxic potential in vivo, K
`cells must be activated, with evidence in mice that this
`step requires the rranspresentation ofIL-15 by dendritic
`cells [9]. The observed enhanced cytotoxic potential of
`activated cells is due in part to the translation of pre(cid:173)
`exist ing pools of granzyme B and perforin m RNA [10].
`
`Effector functions : cytokines
`Cytokine-producing NK cells link the innate and adap(cid:173)
`tive immune responses. l nterferon--y has protean effect~
`incl ud ing Th 1 polarization (11, 12], dendritic-cell matu(cid:173)
`ration and activation f 13], d irect antiviral effect f 14, 15], as
`well as various antiproliferative effects on transformed
`cells. TNFa en hances dendritic cell maturation [16] and
`also leads to increased IFN-y production f 17]. GM-CSF
`can stimulate phagocytosis by monocytes and contributes
`to dendritic-cell maturation (18].
`
`Natural killer receptors
`MHC class I molecules are critical determinants of K
`cell activity; NK cells effectively lyse cells lacking
`expression of some or all MHC molecules. Karre et al.
`f 19] recognized chat NK cytotoxiciry was abrogated if the
`effectors and targets shared MHC class I molecules. The
`'missing-self hypothesis correctly p redicted the exist(cid:173)
`ence of receptors for self-MHC that, when e ngaged,
`would in hibit cytotoxicity. In humans, inhibitory recep(cid:173)
`tors that recognize H L A-A, B, and C molecules belong to
`the killer immunoglobulin-like receptor (KIR) family
`[20,21]. Another inhibitory receptor, the C-type lectin
`NKG2A pairs with CD94 and recognizes the nonclassical
`MHC molecule H LA-E [22-24]. T he expression pat(cid:173)
`terns of inh ibitory receptors create a repertoire of K
`cells with nonoverlapping specificities.
`
`The human KIR gene cluster is located on chromosome
`19q13.4 and contains 14 KTR genes and 2 pseudogenes.
`Inhibitory receptors possess long cytoplasmic tails with
`immunoreceptor tyrosine-based inhibitory motifs (ITIM)
`that allow docking of tyrosine phosphatase molecules,
`whereas activating KIRs have short cytoplasmic tails chat
`enable pairing to adapter molecules with immunoreceptor
`tyrosine-based activating motifa (ITAM) [25]. The extra(cid:173)
`cell ular domains of the activating KIR share sequence
`similarity with the corresponding inhibitory KIR and
`may share HLA-binding specificities, though the ligand~
`for most activating KIRs remain unknown [26,27]. It is
`thought that inhibitory KIRs bind class I molecules with
`greater affinity than the correspondi ng activating KIR.
`
`KIR proteins recog nize allorypic motifs in the class I
`alpha helix, as described in Table 1. Two KIR haplotypes
`
`Effects of natural killer cells in transplantation Benjamin et al. 131
`
`Table 1 Inhibitory and activating receptors for HLA class I
`molecules
`
`Receptor
`
`Inhibitory
`KIR2DL2/3
`KIR2DL1
`KIR3DL1
`
`CD94/NKG2NE
`KIR3DL2
`KIR2DL4
`Activating
`KIR2DS4
`KIR2DS1
`KIR2DS2
`KIR2DS3
`KIR2DS5
`KIR3DS1
`KIR Haplotype
`A
`B
`
`Ligand
`
`HLA-C group1 (Cw1, Cw3)
`HLA-C group 2 (Cw2, Cw4)
`HLA-A and B with Bw4 motifs
`at position 77- 83
`HLA-E
`HLA-A3/A1 1
`HLA-G
`
`HLA-C2
`
`Inhibitory KIR and KIR2DS4
`Inhibitory KIR and combinations
`of activating KIR
`
`KIR genes are organized into diverse haplotypes, which have simplified
`into Groups A and B.
`
`have been defined, A and B [28] . The A haplotype has
`five inhibitory genes (2D L 1, 2D L3, 3DL1, 3DL2, 3DL3)
`and one activating gene (KIR2DS4). The B haplotypes
`are less uniform in gene content and have a greater
`number of activating receptors. H LA genes on chromo(cid:173)
`some 6 and KIR genes on chromosome 19 assort inde(cid:173)
`pendently, meaning that KIR genotype, rather than HLA
`type, is the major determ inant of KI R expression pattern.
`KIR expression is stochastic, generating a n K cell
`com partment with individual members expressing one
`or more inhibitory receptors. NK cells that express only
`inhibitory KlRs for absent class I molecules are detect(cid:173)
`able bu t rather tha n being autoreactive, as would be
`predicted by the missing-self model, such cells are hy por(cid:173)
`esponsive. T he licensing theory of K cell maturation
`suggests that engagement of an inhibitory KIR by the
`cognate class I molecule is necessary to acquire effector
`function [29,30]. Another model proposes that the K
`cells become functionally anergic without self-inhibitory
`signals f3 t].
`
`Licensing notwithstanding, failure to engage an inhibi(cid:173)
`tory KI R is insufficient to elicit an K at.tack. Rather, a
`cell-surface derived activating sig nal is necessary. In
`addition to activating KI R molecules, a nu mber of acti(cid:173)
`vating K cell receptors have now been characterized
`(see Table 2).
`
`Predictors of alloreactivity
`According to the missing-self model, mismatch between
`donor KI R and recipient. MHC class I molecule expres(cid:173)
`s ion predicts the existence of a subset of donor NK cells
`that are not inhibited and are thought to be alloreactive.
`
`Copyright© Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`UPenn Ex. 2056
`Miltenyi v. UPenn
`IPR2022-00855
`Page 131
`
`

`

`132 Transplantation
`
`ReprintsOesk I 21612023 2:02:03 PM
`
`•
`
`Table 2 Some activating NK cell receptors
`
`Receptor
`
`Receptor expression
`
`Ligands
`
`Ligand expression
`
`NKG2D
`
`All NK cells, some T cells
`
`MICA, MICB, ULBP1-5
`
`Leukemia, carcinoma cells; induced by viral infection and ionizing
`radiation (DNA damage response)
`
`NKp30
`NKp44
`NKp46
`DNAM1
`
`All NK cells
`Activated NK cells only
`All NK cells
`All NK cells
`
`B7-H6, BAT3
`?
`Viral hemagglutin n
`CD112, CD155
`
`NK, natural killer.
`
`The Perugia group brought renewed atcention to K cell
`alloreacriviry in their pioneering stud ies of haploidentical
`transplantation. Recipients received myeloablative che(cid:173)
`moradiotherapy and ATG. In order to avoid GVHD ,
`donor grafts were T -cell depleted and
`'megadose'
`GD34+ (e.g. 107 /kg) cells were infused in order to over(cid:173)
`come the resistance co engraftment observed with T -cell
`depleted transplants [32].
`o posttransplant immune
`suppression was used. Under these conditions, KIR
`ligand mismatching in the GVH d irection (observed in
`30% of unselected haploidentical related donors) bene(cid:173)
`fited patients with myeloid leukemias [33,34]. Patients
`receiving KIR ligand mismatched transplants had better
`overall survival, improved rates of engraftment and a
`reduced incidence of GVHD. Protection against GVHD
`has been explored in a mouse model, where it was found
`that donor NK cell alloreactivity was linked to the
`depletion of recipient antigen-presenting cells [34].
`
`Receptor-ligand
`The Ruggeri model assumes that K cells expressing
`inhibitory KIR for ligands absent on both donor and
`recipient are nonalloreactive. T he licensing model pre(cid:173)
`d icts that developing K cells that fail to receive a signal
`th rough an inhibitory receptor are hyporesponsive to
`activating stimuli. However, licensing can he bypassed
`in a p roinflammatory e nvironment [29], as is seen follow(cid:173)
`ing myeloablative conditioning. Le u ng et al. [35] argues
`that alloreactiviry may he predicted by considering only
`the expression of donor-inhibitory KI R (as determined by
`flow cytomerry) and recipient ligand, thus eliminating
`donor ligand from the equation. In a n analysis of 36
`pediatric patients receiving T -cell depleted haploiden(cid:173)
`tical transplants, the receptor-ligand model was a better
`predictor of leukemia relapse than the ligand-ligand
`mode l.
`
`Missing killer immunoglobulin receptor ligand
`The missing ligand model is a n extensio n and sim plifica(cid:173)
`tion of the receptor-ligand model. Regardless of HLA
`expression, most individuals express the full complement
`of inhibitory KIR. An individual who is homozygous for
`KTR ligand epitopes (e.g. H LA-Ct or H LA-Bw4) would
`he predicted to have a subset of NK cells expressing an
`inhibitory KI R for the absent ligand (KI R- L). Hsu et al.
`[36] analyzed the impact of missing ligand in 1770
`
`Melanoma, carcinoma, neuroblastoma
`
`myeloahlative, T -cell replete transplants from unrelated
`donors. Recipients were grouped accord ing to homozyg(cid:173)
`osity of HLA-C group t, HLA-C group 2, or HLA-Bw6.
`Patients homozygous for KTR epitopes who received
`transplants from HLA mismatched donors had a lower
`rate of relapse than heterozygotes. T his effect was greater
`among patients with AM L than those with CM L or ALL.
`However, among pa tients receiving HLA-matched unre(cid:173)
`lated donor transplants, homozygosity of these epitopes
`was associated with a sligh tly higher risk of relapse. In a
`similar analysis Miller et al. [37] analyzed 2062 unrelated
`donor transplants with myeloid malignancies. Of these
`patients, 70% were m issing one or more KIR-L. For
`patients with early stage myeloid malignancies, that is
`AML in C Rl , early stage MDS, or early chronic phase
`CML, absence of KIR- L was protective against relapse,
`hut this effect was lost in more advanced stage disease.
`Onlike the data of H su, this effect was seen in H L A
`matched and mismatched transplants alike. In a further
`analysis of patients receiving haploidentical transplants
`for myeloid malignancies, Ruggeri et al. [38] compared
`the predictive value of the missing ligand model with the
`original mismatched ligand model, finding the latter co he
`more informative.
`
`Activating killer immunoglobulin receptor
`Increased numbers of activating KTR molecules have
`correlated with increased propensity to autoimmune d is(cid:173)
`orders as well as to resisrance to CMV reactivat.ion in
`kidney-transplant recipie n ts [39] . In the setting of allo(cid:173)
`HCT, higher n umbers of activati ng Kl R genes has been
`correlated with increased frequency of acute and chronic
`GVHD. In a multivariate analysis of 448 unrelated trans(cid:173)
`plants for AM L, Cooley et al. [40•] found that presence of
`a donor, KIR-B haplotype, wh ich contains a higher nu m(cid:173)
`ber of activating KTR genes than the A haplotype, pre(cid:173)
`dicted improved relapse free and overall survival in which
`there was no KIR ligand mismatch. This benefit was not
`observed in patients with KIR ligand mismatched trans(cid:173)
`plants. An interesting recent study showed that in the
`setting of TCD haploidentical
`transplantation with
`KTR-L mismatch, K cells co-expressing the activating
`KTRZDSl with inhihicory KTR2 D L2/3 or KG2A were
`able to kill recipient leukemia blasts, highlighting that in
`some settings, recognition by activat ing KIR is able to
`overcome inhibitory signals [41 ].
`
`Copyright© Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`UPenn Ex. 2056
`Miltenyi v. UPenn
`IPR2022-00855
`Page 132
`
`

`

`•
`
`ReprintsOesk I 21612023 2:02:03 PM
`
`Clinical outcomes
`The porential benefits of K cell alloreactivity have been
`explored in cohorts of patienrn receiving mismatched
`unrelated donor transplants, often with d iscordant con(cid:173)
`clusions. These results have been reviewed p reviously
`f42 - 4Sl.
`
`Umbilical cord transplantation
`As with haploidentical transplantation, umbilical cord
`blood (llCB) transplants are frequently c haracterized
`by a high degree of HLA m ismatching. UCB has as it~
`advantage the relatively low risk of acute GVHD due to a
`lower number of mature donor T cells and thus an
`increased ability to use HLA mismatched units. Will(cid:173)
`emze et al. [4(, ] reviewed the impact of KIR ligand
`m ismatching in lJCB transplants for acute leukemia
`(lymphoid or myeloid)
`in first complete remission
`(C Rl ). Of 218 transplants, 10% were HLA mismatched
`and 47% had greater than one mismatch. Thirty-two
`percent of donor recipient pairs were KI R-ligand incom(cid:173)
`patible in the graft versus host direction. Patients with
`KIR-ligand incompatible donors had improved overall
`survival (57 vs. 40%) and decreased relapse (20 vs. 37%)
`when compared with chose without these incompatibil(cid:173)
`ities. As was seen in the Perugia studies, benefits of KIR
`ligand incompatibility were most striking among patients
`with AM L although lJCB recipients with ALL also had a
`trend toward improved leukemia-free survival. However,
`Brunstein et al. (47] failed to observe any benefit of KIR(cid:173)
`L mismatch in lSS recipients ofOCB after myeloablative
`conditioning. In fact, in 102 patients who had received
`UCB after nonmyeloablative cond itioning, KI R-L mis(cid:173)
`match was associated with an increased rate of acute
`GVHD and higher treatment-related mortality.
`
`Nonmyeloablative transplantation
`Nonmyeloablative conditioning is marked by a period of
`mixed chimerism in which recipient NK cells may exert
`antidonor effects. KIR-L mismatch in the host versus
`graft direction in 31 patients conditioned with fludarab ine
`and 2Gy TBI was fou nd to predict for increased risk of
`graft rejectio n and lower incidence of complete do nor
`chimerism (48•].
`
`The impact of NK cell a lloreactivi cy in the graft versus host
`d irection was studied in a series of 282 donor- recipient
`pairs treated with 2Gy of'TBI with or without fludarabine,
`where the majority (88%) received H L A-matched graft~
`(49•]. High donor K cell chimerism before day 100 wa~
`associated with low relapse rates. The risk of relapse wa~
`lower for patients expressing ligands for all donor KIR,
`though this did not reach scaristical significance. Of note, a
`lower risk of relapse was nor significantly associated with
`high donor T -cell chimerism. In contrast, the risk of acute
`GVH D (grade II-IV) was associated with high levels of
`
`Effects of natural killer cells in transplantation Benjamin et al. 133
`
`do nor T -cell chimerism, whereas K cell chimerism did
`not correlate with GVHD.
`
`Natural killer reconstitution posttransplant
`During the first few mo nths post HCT, K cells are the
`predominant circulating lymphoid cell subset with the
`potential to control disease relapse. NK cells are the first
`ly mphoid cells to repopulate, reaching normal numbers
`within 1 month regardless of donor type or patient age in
`adults [S0,S 1] and children [S2]. Similar findings are
`shown after
`lJCB
`transplantation
`in children
`fS3] .
`Although some studies have highlighted the ability of
`K cells to exert a potent an tileukemic effect and
`reduced relapse in high-risk hematologic malignancy,
`the question remains whether K cells reconstituting
`after HCT manifest an actual antileukemic effect. This
`issue has been investigated by evaluating the phenotype
`and cytotoxic potential of K cells re -isolated at early
`time-points after transpla nt.
`
`Phenotypic changes
`According to the Perugia KIR-mismatch model, poren(cid:173)
`tially a lloreactive K cells posc-HCT are cells chat
`express KIR for missing KIR-L molecules in the absence
`of the inhibitory receptor GD94/NKG2A. The Perugia
`data, however, show that the majority of NK cells in the
`first S months post-HCT do not belong to this subset [38].
`Similarly, other groups have found that early after both
`HLA-matched and haploidentical HCT (with or without
`T cell depletion), NK cells are predominantly CDS6hrighc,
`KG2A+ and KIR- , a find ing which has been described
`as consistent with an ' immature phenotype' fS4,ss •,
`S6,S7] and has been related to increased post-HCT levels
`of the homeostatic cytokine IL-15 [S6]. The remaining
`CDS6tlim cells, which normally express low levels of
`KG2A, u pregulate expression of this receptor post(cid:173)
`HCT while expression of CD16 as well as of KIR is
`lower than in healthy controls fSS",57]. A mature donor(cid:173)
`type KI R repertoire appears within 3 months- 3 years
`[3s,ss· ,s8J.
`
`Expression of NKp46 as well as of the activating receptor
`KG2D appear to be increased during rhe first year pose
`HLA-marched non-TCD HC'T, whereas NKp44 a nd
`Kp30 do not appear to be u pregulated [S6].
`
`With regard to the expression of effector molecules,
`production of both perforin and IFN-y is increased among
`the C DS6hright cells in patients post HLA-matched HCT
`when compared with the same subset in normal donors
`[S6].
`
`Functional changes
`inconsistent results,
`Several grou ps, with somewhat
`have investigated the fu nctional consequences of the
`
`Copyright© Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`UPenn Ex. 2056
`Miltenyi v. UPenn
`IPR2022-00855
`Page 133
`
`

`

`134 Transplantation
`
`ReprintsOesk I 21612023 2:02:03 PM
`
`•
`
`phenotypic changes described earlier. The Perugia group
`found that N K alloreactive cells (defined as N K cells of
`donor origin capable of k ill ing cryopreserve<l host PHA
`b lasts) are detectable in about half of patie nts 1 month
`afrer haploi<len tical HCT and are no longer detectable
`afrer 12 months [38]. Afrer haplo i<lentical HCT K cells
`have poor effector function against primary leukemia
`cells, a finding which has been related to the increased
`frequency of NKG2A-bearing NK cells, althoug h result.s
`of blocking studies of this inhibitory receptor have
`yielded conflicting results [54,55•]. Evaluation of cells
`that are N KG2A- and single-KI R+ (thus predicted to
`have alloreactive potential under the models described
`earlier) after haploi<lentical HGT has demonstrated that
`these cells are first detectable approximately 75 <lays after
`HCT, their frequency is highly variable and shows no
`correlation with predicted donor K cell alloreactivity
`and that their functional capacity was reduced [55•]. T his
`finding is somewhat out of keeping with the conclusions
`from the Perugia group and may be related to the lack of
`licensing of these cells, as they are repopulating in the
`absence of a n H LA ligand for their single KI R.
`
`Furthermore, a small study in 'TCD HLA-matche<l
`HCT has shown that K cells exhibiting KI R for nonself
`class I ligands (predicted to be alloreactive according to
`the ' missing ligand' model but nonlicense<l according
`to the licensing hypothesis) are capable of mounting
`a robust cytotoxic and cytokine response in the first
`months posttransplant when incubated with MHC class
`I deficient cell lines [59•]. The a uthors explain this
`finding by postulati ng that N K cell tolerance to host
`cells takes some time to develop, <luring which time
`patients may benefit from NK cell alloreactivity, although
`it may be that licensing requirements are bypassed in the
`inflammatory posttransplant environment.
`
`The p resence of T cells in the graft has been found to
`impair K-cell reconstitution and function posttrans(cid:173)
`plant [43,60]. This find ing may in part explain the
`importance of T -cell depletion for ensuring an opti mal
`NK-cell effect. In addition, peritransplant immunosup(cid:173)
`pression could impact K cell function, as exposure ofin(cid:173)
`viuo differentiated K cells to cyclosporine A has been
`shown to lead to preferential expansion of C D56+ KIR(cid:173)
`NK cells [61].
`
`Conclusions drawn from these studies must be tempered
`by the recognition that different NK cell functional
`assays may y ield d iscordant results. Most studies evaluate
`NK cells by their capacity to <legranulate (using the
`GD107a mobilization assay) or produce IF
`'Y after
`exposure to tumor cell lines known to be susceptible
`to K cell killing, a somewhat artificial experimental. In
`addition, studies using as targets patient PHA blasts
`rather than cryopreserve<l p rimary patient leukemia cells
`
`likewise may not be representative of the actual anti(cid:173)
`tumor potential.
`
`Natural killer adoptive therapy
`As follows from the earlier d iscussion, K cells repopu(cid:173)
`lating the host after transplantat ion may be functionally
`defective as a result of immaturity, immunosuppression,
`or other u nidentified factors. Thus, it has been proposed
`that the adoptive transfer of mature NK cells may be
`beneficial by providing both a cytolytic effect and an
`immunomo<lulatory effect, the latter by utilizing NK(cid:173)
`<len<lritic cell cross talk to promote K-cell activation and
`subsequent Thl p olarization [14].
`
`GMP grade production of N K cells from apheresis pro(cid:173)
`ducts for adoptive therapy is feasible, with reported
`y ields of up to 10 x 108/kg with recovery of approximately
`40- 50% [62- 65].
`
`In the most important study of adoptive K-cell therapy
`to <late, haploi<lentical K cells were given to 43 patients
`with advanced malignancies (AML, metastatic mela(cid:173)
`noma and metastatic renal carcinoma) together with
`IL-2. There was minimal hematologic and nonhemato(cid:173)
`logic toxicity, the latter mostly attributable to IL-2.
`Results from this study revealed the following conditions
`for persistence in the host of donor NK cells: a high-dose
`conditioning regimen is g iven to achieve sig nificant lym(cid:173)
`pho<lepletion; high levels ofIL- 15 levels are present; and
`the addition of nonmyeloablative <loses of TB I, cyclopho(cid:173)
`sphami<le and flu<larabine and infusion of CD34+ p ro(cid:173)
`genitor cells leads to better K cell expansion and higher
`AML remission rates [66,67]. There was no GVHD, but
`the protocol was complicated by fatal infections and
`EBV-PTLD; interestingly, no correlation was found
`between efficacy and predicted K alloreactivity accord(cid:173)
`ing to KIR ligand mismatch.
`
`Q uestio ns remain regarding the optimal purity of the
`product, as there is evidence from early studies that
`monocytes may be required for optimal NK-cell prolifer(cid:173)
`ation [68], and more recent studies show that monocyte(cid:173)
`<lerive<l <len<lritic cells promote N K cell effector function
`[69]. Similarly, whether the optimal source of NK cells
`should be from nonmobilize<l or G-CSF mobilized per(cid:173)
`ip heral blood collections needs to be addressed, due to
`data suggesting that N K cells expanded from G-CSF
`mobilized PBPC collections are functiona lly abnormal
`and possess reduced n umbers of K progenitors [70- 72].
`
`Facilitation of NK cell eng raftment will likely depend on
`suppression of the host immune system, eradication of
`'Tregs (wh ich may inhibit transferred N K cells) [73]
`presence of 'space' for homeostatic proliferation follow(cid:173)
`ing lympho<lepletion, and the presence of high levels of
`
`Copyright© Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`UPenn Ex. 2056
`Miltenyi v. UPenn
`IPR2022-00855
`Page 134
`
`

`

`ReprintsOesk I 21612023 2:02:03 PM
`
`Effects of natural killer cells in transplantation Benjamin et al. 135
`
`•
`
`cytokines especially I L-1:5 [74,75]. The assertion that
`I L- 15 is critically required for in-vivo K cell expansion
`has heen recently challenged in murine models of infec(cid:173)
`tion [76].
`
`Receptor and ligand modulation
`Modulation of NK cell recognition of tumors may he
`ach ieved hy hlockade of inhihitory KIR as was recently
`demonstrated using a humanized antihody to KI RZDL-1,
`KIRZD L-2, and KI RZD L-3 as well as the activating
`receptors KIRZDS-1 and KIRZDS-2 [77]. A phase I trial
`of this agent is ongoing.
`
`Additional approaches to augment K cell act1v1ty
`incl ude the use of hispecific antihodies (such as for
`HE RZ/neu and CD16) [78), genetic modification of
`NK cells to express receptors for tumor-associated anti(cid:173)
`gens not normally part of the K-receptor ligand reper(cid:173)
`toire or enforced expression of signaling receptors [79,80].
`Other approaches include the indirect pharmacologic
`modulation of K receptor ligands on tumor cells hy
`compounds such as horcezomih [81 l or hisrone deacety(cid:173)
`lase inhihitors [82••1. It may he that several of these
`techniques may be required in order to realize the full
`potential of K cell adoptive therapy.
`
`Conclusion
`Successful utilization of adoptive K therapy may
`require a n availahle niche as defined either by lympho(cid:173)
`pen ia, selective host NK cell lymphopenia, high IL-15 or
`other homeostatic cytokine levels, or a comhination of
`these. Other open questions include the appropriate
`method for collection and activation, optimal cytokine
`type and schedule, and in-vivo or ex-vivo stimulation, and
`it will he important to ensu re that cells which have been
`activated for several days in vitro do not lose the ir pro(cid:173)
`liferative potential in v ivo, as has heen found for adop(cid:173)
`tively transferred T cells [83]. Although it is likely that
`apheresis products will represent the most easily acces(cid:173)
`sihle source of mature NK cells, it is important to keep in
`m ind that K cells enriched from G-CSF mohilized
`hlood exhibit reduced functional capacity compared with
`those from unstimulated peripheral hlood [70]. It may he
`that a particular suhset of K cells should he targeted for
`expansion for adoptive infusion, althoug h studies addres(cid:173)
`sing this particular point are presently lacking. Finally,
`the optimal methods to e nsure persistent expansion and
`recognition of target cells still await development, and
`perhaps will require a com hi nation of cytokines and K
`receptor ligand modulating agents.
`
`Acknowledgements
`This work was supported by the National Institutes of Health (R01-CA-
`125276) and the Hematology Society of Australia and New Zealand.
`
`We apologize to those whose studies were not cited due to space
`considerations.
`
`References and recommended reading
`Papers of particular interest, published within the annual period of review, have
`been highlighted as:
`of special interest
`•
`•• of outstanding interest
`Additional references related to this topic can also be found in the Current
`World Literature section in this issue (pp . 159-160).
`
`Dalbeth N, Gundle R, Davies RJ, et al. CD56bright NK cells are enriched at
`inflammatory sites and can engage with monocytes in a reciprocal program of
`activation. J lmmunol 2004; 173:6418-6426.
`
`2 Carrega P, Morandi B, Costa R, et al. Natural killer cells infiltrating human
`nonsmall-cell lung cancer are enriched in CD56 bright CD16(- ) cells and
`d isplay an impaired capability to kill tumor cells. Cancer 2008; 112:863-875.
`
`3 Gill S, O lson JA, Negrin RS. Natural killer cells in allogeneic transplantation:
`effect on engraftmen~ graft-versus-tumor, and graft-versus-host responses.
`Biol Blood Marrow Transplant 2009; 15:765-776.
`
`4
`
`Chan A, Hong DL, Atzberger A, et al. CD56bright human NK cells differentiate
`into C D56dim cells: role of contact with peripheral fibroblasts. J lmmunol
`2007; 179:89-94.
`5 Romagnani C, Juelke K, Falco M, et al. C D56brightCD16- killer lg-like
`receptor- NK cells display longer telomeres and acquire features of CD56dim
`NK cells upon activation. J lmmunol 2007; 178:4947-4955.
`
`6
`
`7
`
`8
`
`Jentsch-Ullrich K, Koenigsmann M, Mohren M, Franke A. Lymphocyte subsets'
`reference ranges in an age- and gender-balanced population of 1 00 healthy
`adults: a monocentric German study. Clin lmmunol 2005; 116:192-197.
`Zamai L, Ahmad M, Bennett IM, et al. Natural killer (NK) cell-mediated
`cytotosicity: differential use of TRAIL and Fas ligand by immature and mature
`primary human NK cells. J Exp Med 1998; 188:2375-2380.
`
`Freud AG, Yokohama A, Becknell B, et al. Evidence for discrete stages of
`human natural killer cell differentiation in vivo. J Exp Med 2006; 203:1033-
`1043.
`
`9
`
`Lucas M, Schachterle W , Oberle K, et al. Dendritic cells prime natural killer
`cells by trans-presenting interleukin 15. Immunity 2007; 26:503-517.
`1 0 Fehniger TA, Cai SF, Cao X, et al. Acquisition of murine NK cell cytoto,icrty
`requires the translation of a preexisting pool of granzyme B and perforin
`mRNAs. Immunity 2007; 26:798-81 1.
`
`11 Martin-Fontecha A, Thomsen LL, Brett S, et al. Induced recruitment of NK cells
`to lymph nodes provides IFN-gamma for T(H)1 priming. Nat lmmunol 2004;
`5:1260- 1265.
`
`12 Morandi B, Bougras G, Muller WA, et al. NK cells of human secondary
`lymphoid tissues enhance T ce

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket