throbber
Review |
`
`Natural killer cell-based immunotherapy in cancer: current
`insights and future prospects
`
`doi: 10.1111/j.1365-2796.2009.02121.x
`
`T. Sutlu & E. Alici
`
`From the Division of Haematology, Department of Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet,
`Stockholm, Sweden
`
`Abstract. Sutlu T, Alici E (Karolinska Institutet, Stock-
`holm, Sweden). Natural killer cell-based immunother-
`apy in cancer:
`current insights and future prospects
`(Review). J Intern Med 2009; 266: 154–181.
`
`As our understanding of the molecular mechanisms
`governing natural killer (NK) cell activity increases,
`their potential
`in cancer immunotherapy is growing
`increasingly prominent. This
`review analyses
`the
`currently available preclinical
`and clinical data
`regarding
`NK
`cell-based
`immunotherapeutic
`approaches
`in cancer
`starting from a historical
`background
`and
`an
`overview of molecular
`mechanisms taking part
`in NK cell responses. The
`status of NK cells
`in cancer patients, currently
`in vivo
`investigated clinical applications
`such as
`modulation of NK cell activity, ex vivo purifica-
`tion ⁄ expansion and adoptive transfer as well as
`future possibilities such as genetic modifications are
`discussed in detail.
`
`Keywords: cancer immunotherapy, clinical trials, cyto-
`kines, ex vivo expansion, gene therapy, natural killer
`cells.
`
`Abbreviations:
`ADCC, antibody-dependent cellular
`cytotoxicity; ALL, acute lymphoblastic leukaemia;
`AML, acute myeloid leukaemia; BM, bone marrow;
`BMT, bone marrow transplantation; CML, chronic
`myelogenous
`leukaemia; CR, complete remission;
`CRC, colorectal carcinoma; DC, dendritic cell; DLI,
`donor lymphocyte infusion; G-CSF, granulocyte colony
`stimulating factor; GM-CSF, granulocyte-macrophage
`colony stimulating factor; GMP, good manufacturing
`practice; GvHD, graft-versus-host disease; HCC,
`hepatocellular carcinoma; HLA, human leukocyte anti-
`gen; HSCT, haematopoietic stem cell transplantation;
`IFN,
`interferon;
`IL,
`interleukin; KIR, killer-cell
`immunoglobulin-like receptor; LAK cells, lymphokine-
`activated killer cells; LGL, large granular lymphocyte;
`MHC, major histocompatibility complex; MM, multiple
`myeloma; NB, neuroblastoma; NCR, natural cytotoxic-
`ity receptor; PBMC, peripheral blood mononuclear cell;
`PBSC, peripheral blood stem cell; PHA, phytohaemag-
`glutinin; PR, partial remission; RCC, renal cell carci-
`noma; ROS, reactive oxygen species; SCID, severe
`combined immunodeficiency; SCT, stem cell transplan-
`tation; TCR, T-cell receptor; TNF,
`tumour necrosis
`factor; TRAIL, TNF-related apoptosis inducing ligand;
`Treg, regulatory T cell; WBC, white blood cell.
`
`Natural killer cells: a historical background
`
`in
`Initially regarded as an ‘experimental artifact’
`T-cell cytotoxicity assays, natural killer (NK) cells
`were first discovered in mice more than 30 years ago
`by Kiessling et al., who also named them natural
`killer cells [1, 2] and in parallel by Herberman et al.
`[3, 4]. Human NK cells were initially described as
`
`nonadherent, nonphagocytic, FccR+,
`large granular
`lymphocytes (LGL) [5]. Later it was, however, appre-
`ciated that NK cells not only shared the LGL pheno-
`type and some NK cells also displayed normal small
`lymphocyte morphology, depending on their activa-
`tion status [6]. This made it difficult to detect the NK
`cell population just by the size and morphology. The
`identification of the NKR-Pl [7] and NK1.1 [8] made
`
`154 ª 2009 Blackwell Publishing Ltd
`
` 13652796, 2009, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1365-2796.2009.02121.x, Wiley Online Library on [08/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
`
`UPenn Ex. 2064
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`T. Sutlu & E. Alici |
`
`Review: Natural killer cell-based immunotherapy in cancer
`
`it possible to define the murine NK cells roughly as
`)
`)
`NK1.1+ TCR
`CD16+. Today, human NK cells
`sIg
`)
`CD56+ lymphocytes. They com-
`are defined as CD3
`prise 10–15% of all circulating lymphocytes and are
`also found in peripheral tissues, including the liver,
`peritoneal cavity and placenta. Resting NK cells cir-
`culate in the blood, but following activation by cyto-
`kines,
`they
`are
`capable
`of
`extravasation
`and
`infiltration into most
`tissues that contain pathogen-
`infected or malignant cells [9–11].
`
`The discovery of NK cells suggested a possible
`effector mechanism behind the phenomenon of
`‘hybrid resistance’. Skin and organ transplantations
`had shown that allogeneic grafts were rejected whilst
`syngeneic grafts were tolerated,
`i.e.
`rejection only
`took place when the grafts had MHC molecules dif-
`fering from the host. This rejection was mediated by
`T cells, which could induce either a graft-versus-host
`or a host-versus-graft
`reaction.
`Irradiated (AxB)F1
`mice rejected BM transplants from either parent,
`despite the fact
`that
`the transplant did not express
`any foreign MHC molecules. This was not in accor-
`dance with the reigning dogmas of T-cell-mediated
`rejection. The BM rejection could still be observed
`in severe combined immunodeficient (SCID) mice,
`which have no T and B cells but have functional
`NK cells [12].
`
`Initially, it was not clear how NK cells distinguished
`the target cells they should kill from those that they
`should spare. When Ka¨rre summarized his and other
`people’s work for his doctoral
`thesis, he found a
`common denominator not about what was commonly
`expressed on target cells but about what was com-
`monly missing. This lead him to formulate the miss-
`ing-self hypothesis, where he suggested that NK
`cells kill target cells lacking expression of self MHC
`class-I molecules
`although the mechanism was
`unclear [13, 14] (see Fig. 1). This model was later
`confirmed by the discovery of inhibitory receptors
`on NK cells. Missing-self could also explain the
`hybrid resistance phenomenon;
`the (AxB)F1 host
`killed cells from either parent A or B because these
`cells lacked complete self MHC expression (A+B).
`the missing-self hypothesis, a MHC
`To further test
`
`class I-deficient version of the tumour cell line RMA
`was established and named RMA-S. C57BL ⁄ 6 mice
`inoculated with RMA-S cells rejected the tumours,
`whilst mice inoculated with RMA developed the
`tumour. By treating the mice with NK depleting
`anti-asialo GM1 antibody,
`the difference in tumour
`outgrowth disappeared [15]. This confirmed that NK
`cells-mediated the selective rejection of MHC lack-
`ing tumour growth.
`
`Natural killer cells are separated into two subsets
`based on their CD56 antigen expression. Yet,
`this
`separation is not
`just phenotypic but
`rather has
`many functional outcomes. The majority (90%) of
`human NK cells
`have
`low-density
`expression
`of CD56 (CD56dim), whereas 10% of NK cells
`are CD56bright. Early functional
`studies of
`these
`the CD56dim cells are more
`subsets revealed that
`cytotoxic [16]. However,
`there are a number of
`other
`cell-surface markers
`that
`confer
`unique
`phenotypic and functional properties to CD56bright
`and CD56dim NK cell subsets. CD56bright subset
`is
`shown to exclusively express IL-2 receptor a chain
`(IL-2Ra ⁄ CD25), whilst
`they lack or express only
`at very low levels the FCcRIII
`(CD16). On the
`other hand, the CD56dim subset has high expression
`of CD16
`and
`lacks CD25
`expression. These
`properties set very different
`roles to the different
`subsets with regards to antibody dependent cellu-
`lar cytotoxicity (ADCC) and response to IL-2 stim-
`ulation.
`In
`addition
`to
`distinct
`expression
`of
`adhesion molecules
`and cytokine
`receptors,
`the
`CD56bright NK cell has
`the capacity to produce
`high levels of immunoregulatory cytokines, but has
`low-level expression of killer-cell
`immunoglobulin-
`like receptors (KIRs) and is poorly cytotoxic. By
`the CD56dim NK cell appears to produce
`contrast,
`low levels
`of
`cytokines
`but
`has
`high-level
`expression of KIRs
`and is
`a potent
`cytotoxic
`effector
`cell. Such evidence
`suggests
`that
`the
`CD56bright and CD56dim subsets are distinct lympho-
`cytes with unique roles
`in the immune system.
`Thus, studies of the biology of human NK cells are
`eventually
`approaching NK cells
`as
`separate
`CD56bright
`and CD56dim subsets
`rather
`than a
`homogenous population.
`
`ª 2009 Blackwell Publishing Ltd Journal of Internal Medicine 266; 154–181 155
`
` 13652796, 2009, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1365-2796.2009.02121.x, Wiley Online Library on [08/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
`
`UPenn Ex. 2064
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`T. Sutlu & E. Alici |
`
`NK
`cell
`
`Tumour
`cell
`
`Review: Natural killer cell-based immunotherapy in cancer
`
`(a)
`
`(b)
`
`(c)
`
`(d)
`
`Inhibitory
`receptor
`
`Inhibitory
`ligand
`
`Activating
`receptor
`
`Activating
`ligand
`
`As the name implies, NK cells can kill certain cells
`without prior sensitization, but
`they are also potent
`producers of various cytokines, such as IFN-c, TNF-a,
`GM-CSF and IL-3 [17]. Therefore, NK cells are also
`
`Fig. 1 The recognition of tumour cells by NK cells. The
`figure presents four hypothetical scenarios for the encounter
`of an NK cell and a tumour cell. (a) Although the tumour
`cell does not express any inhibitory ligands, it cannot be
`killed by the NK cell because it also lacks the expression of
`any activating ligands. This target is practically invisible to
`the NK cell and no recognition takes place. (b) The tumour
`cell expresses ligands for inhibitory receptors, whereas it
`lacks ligands for activating receptors. The NK cell recog-
`nizes the inhibitory ligands and, therefore, no killing takes
`place. (c) The tumour cell has significantly downregulated
`or absent expression of inhibitory ligands along with suffi-
`cient expression of activating ligands. Missing-self recogni-
`tion takes place and the target is killed. (d) The tumour cell
`expresses significant levels of both inhibitory and activating
`ligands. The NK cells recognize both types of ligands and
`the outcome of this interaction is determined by the balance
`of inhibitory and activating signals.
`
`believed to function as regulatory cells in the immune
`system, influencing other cells and responses and act-
`ing as a link between the adaptive and innate immune
`responses. For example, NK cells seem to participate
`in the development of the autoimmune disease, myas-
`thenia gravis, by regulating both the autoreactive T and
`B cells through IFN-c production [18]. Moreover, it
`has been observed that depletion of NK cells in
`C57Bl ⁄ 6 mice leads to increased engraftment of neuro-
`blastoma (NB) xenografts mainly because of dysregu-
`lation of Th1-oriented B-cell responses [19]. These
`data prove the significant impact of NK cells on adap-
`tive immune responses. Other studies have also shown
`a close interaction between NK cells and dendritic cells
`(DC) [20]. In addition to their role as the initiators of
`antigen specific responses, DCs have been shown to
`support the activity of NK cells [21], whilst recipro-
`cally, cytokine-preactivated NK cells have been shown
`to activate DCs and induce their maturation and cyto-
`kine production [22–24]. In vivo activation of NK cells
`by a DC vaccine consisting of autologous DCs loaded
`with a tumour-associated antigen has also been shown
`[25]. NK cells are also involved in the defence against
`virus infections and autoimmunity both of which have
`been elegantly reviewed elsewhere [26, 27].
`
`Today, we know that NK cell cytotoxicity is the result
`of a complex balance between the inhibitory and acti-
`vating receptors [28]. Table 1 provides a list of
`human NK cell activating and inhibitory receptors
`identified to our knowledge. Upon recognition of the
`
`156 ª 2009 Blackwell Publishing Ltd Journal of Internal Medicine 266; 154–181
`
` 13652796, 2009, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1365-2796.2009.02121.x, Wiley Online Library on [08/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
`
`UPenn Ex. 2064
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`T. Sutlu & E. Alici |
`
`Review: Natural killer cell-based immunotherapy in cancer
`
`ligands on the target cell surface by activating NK
`cell
`receptors, various intracellular signalling path-
`ways drive NK cells towards cytotoxic action and this
`results in target cell cytolysis [29].
`
`However, these processes are tightly controlled by a
`group of inhibitory receptors. These receptors act as
`negative regulators of NK cytotoxicity and inhibit the
`action of NK cells against ‘self’ targets. A main group
`of this type of receptors is KIRs, which are mainly spe-
`cific for self MHC Class-I molecules. If the target cell
`is recognized by inhibitory KIRs, which means, it has
`sufficient amount of self MHC Class-I molecules on
`the cell surface, an inhibitory signal from KIRs stops
`the action of cytotoxic pathways triggered by activating
`receptors [30, 31]. The KIRs are type I (extracellular
`amino terminus) membrane proteins that contain either
`two or three extracellular Ig-like domains [32] and they
`are designated as KIR2D or KIR3D respectively. The
`cytoplasmic domains of the KIRs can be either short
`(S) or long (L), corresponding to their function as either
`activating or inhibitory receptors respectively. Members
`of the KIR family recognize HLA-A, HLA-B and
`HLA-C alleles and KIR2DL4 recognizes HLA-G [33].
`The KIR receptors are clonally distributed on NK cells,
`which provides that even the loss of a single HLA allele
`(a common event in tumourigenesis and viral infec-
`tions) can be detected by a pool of NK cells [33, 34].
`
`The activating side of the balance also includes a ser-
`ies of different receptors (see Table 1). The main acti-
`vating receptor group is called natural cytotoxicity
`receptors (NCRs) [29] and it is believed that the main
`control over the NK cell activating pathways is regu-
`lated by these receptors. Currently, there are three dif-
`ferent NCRs identified: NKp30 [35], NKp44 [36] and
`NKp46 [37]. NKp30 and NKp46 are expressed both
`in activated and in nonactivated NK cells, whereas
`NKp44 expression is restricted to activated NK cells.
`Most activating receptors do not directly signal
`through their cytoplasmic tail, but
`instead associate
`noncovalently with other molecules containing immu-
`noreceptor
`tyrosine-based activation motifs (ITAM)
`that serve as the signal transducing proteins. NKp30
`and NKp46 are associated with CD3f, whereas
`NKp44 is associated with DAP12. NK cell activation
`
`has been studied extensively in recent years and is
`discussed elsewhere [38, 39].
`
`Natural killer cells have been described as ‘large gran-
`ular lymphocytes’ and their granularity is their means
`for target cell killing (see Fig. 2). These granules con-
`tain perforin and granzyme B [40] and it is postulated
`that granzymes and perforin both bind to the target
`surface as part of a single macromolecular complex
`[41]. When an NK cell encounters a target cell, perfo-
`rin and granzyme B are released; granzyme enters the
`target cell and mediates apoptosis, whilst perforin dis-
`rupts endosomal
`trafficking [42, 43]. NK cells can
`also express FasL and TNF-related apoptosis-inducing
`ligand (TRAIL), which are both members of the TNF
`family and are shown to induce target cell apoptosis
`when they bind their receptors on target cells [44,
`45]. TNF-a has also been suggested to mediate acti-
`vation-induced cell death by NK cells [46].
`
`NK cells in cancer
`
`The development of any malignancy is under close
`surveillance by NK cells as well as other members of
`the immune system. Nevertheless, malignant cells
`obtain means to escape from the immune system and
`proliferate. General mechanisms include saturation of
`the immune system by the rapid growth of
`the
`tumour,
`inaccessibility of
`the
`tumour owing to
`defective vascularization,
`its large dimension or its
`localization in immune-privileged sites and resistance
`to the Fas- or perforin-mediated apoptosis. The
`expression of FasL by tumour cells as a counterattack
`strategy against immune effector such as T cells and
`NK cells is also common [47–49]. Additionally, the
`defective expression activation receptors and various
`intracellular signalling molecules by T cells and NK
`cells in cancer patients was observed and reported to
`correlate with disease progression [50].
`It has
`also been shown that malignant cells secrete immuno-
`suppressive factors
`that
`inhibit T and NK cell
`proliferation [51]. As a result of all
`these events,
`defective immunity secondary to tumour development
`has been a well-established phenomenon [52]. Table 2
`presents a selection of previously defined NK cell
`abnormalities in cancer patients.
`
`ª 2009 Blackwell Publishing Ltd Journal of Internal Medicine 266; 154–181 157
`
` 13652796, 2009, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1365-2796.2009.02121.x, Wiley Online Library on [08/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
`
`UPenn Ex. 2064
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`T. Sutlu & E. Alici |
`
`Review: Natural killer cell-based immunotherapy in cancer
`
`Table 1 Activating and inhibitory receptors on human NK cells
`
`Alternative name
`
`Type of signal
`
`Ligand
`
`Distribution on NK cells
`
`CD
`
`CD2
`
`CD7
`
`CD11a
`
`CD11b
`
`CD16
`
`CD27
`
`CD44
`
`CD59
`
`CD69
`
`CD85j
`CD94 ⁄ CD159a
`CD94 ⁄ CD159c
`CD96
`
`CD160
`
`CD161
`
`BY55
`
`NKR-P1
`
`LFA-2
`
`LEU-9
`
`LFA-1
`
`Mac-1
`FccRIII
`
`Activation
`
`Activation
`
`Activation
`
`Activation
`
`Activation
`
`TNFRSF7
`
`?
`
`Hyalunorate receptor
`
`Protectin
`
`CLEC2C
`
`ILT-2
`CD94 ⁄ NKG2A
`CD94 ⁄ NKG2C
`TACTILE
`
`Activation
`
`Activation
`
`Activation
`
`Inhibition
`
`Inhibition
`
`Activation
`
`Activation
`
`Activation
`Activation ⁄
`Inhibition
`
`CD58 (LFA-3)
`
`SECTM1, Galectin
`
`ICAM-1,-2,-3,-4,-5
`
`ICAM-1, Fibrinogen
`
`IgG
`
`CD70
`
`Hyalouronan
`
`C8, C9
`
`Unkown
`
`HLA-A, -B, -G
`
`HLA-E
`
`HLA-E
`
`CD155
`
`HLA-C
`
`LLT1
`
`All
`
`All
`
`All
`
`All
`Mainly CD56dim
`Negative ⁄ dim on CD56bright
`Mainly on CD56bright
`Negative ⁄ dim on CD56dim
`All
`
`All
`
`Activated
`
`Subset
`
`Most
`
`Most
`
`Activated low expression
`
`on resting
`
`All
`
`Subset
`
`Activated
`
`CD223
`
`CD226
`
`CD244
`
`CD314
`
`CD319
`
`CD328
`
`CD329
`
`CD335
`
`CD336
`
`CD337
`
`Various
`
`Various
`
`–
`
`–
`
`Lag3
`
`DNAM-1
`
`2B4
`
`NKG2D
`
`CRACC
`
`Siglec-7
`
`Siglec-9
`
`NKp46
`
`NKp44
`
`NKp30
`
`KIR2DS, KIR3DS
`
`KIR2DL, KIR3DL
`
`NTB-A
`
`KLRG1
`
`Activation
`
`Activation
`Activation ⁄
`Inhibition
`
`Activation
`
`Activation
`
`Inhibition
`
`Inhibition
`
`Activation
`
`Activation
`
`Activation
`
`Activation
`
`Inhibition
`
`Activation
`
`Inhibition
`
`HLA Class II
`
`CD112, CD155
`
`CD48
`
`MICA, MICB,
`
`ULB-1,-2,-3,-4
`
`CRACC
`
`Sialic acid
`
`Sialic acid
`
`Viral haemagglutinin (?)
`
`All
`
`All
`
`All
`
`Mature NK cells
`
`Subset
`
`Subset
`
`All
`
`Viral haemagglutinin (?)
`
`Activated
`
`Viral haemagglutinin (?)
`
`HLA Class I
`
`HLA Class I
`
`NTB-A
`
`E-,N-,P-cadherin
`
`All
`
`Subsets
`
`Subsets
`
`All
`
`All
`
`Potential of NK cells in cancer immunotherapy
`
`Modulation of NK cell activity
`
`IL-2 alone. The cDNA encoding for the human IL-2
`gene was cloned in 1983 [53] after a long search start-
`ing in 1965 for the soluble factors in lymphocyte con-
`
`ditioned media that could sustain the proliferation of T
`cells in culture [54, 55]. It is now well known that IL-
`2 effects many types of cells in the immune system
`including cytotoxic T cells, helper T cells, regulatory T
`cells, B cells and NK cells. Currently, there are three
`distinct chains of the IL-2 receptor identified; the a
`
`158 ª 2009 Blackwell Publishing Ltd Journal of Internal Medicine 266; 154–181
`
` 13652796, 2009, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1365-2796.2009.02121.x, Wiley Online Library on [08/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
`
`UPenn Ex. 2064
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`T. Sutlu & E. Alici |
`
`Review: Natural killer cell-based immunotherapy in cancer
`
`Granule-dependent
`killing
`
`Killing by
`death ligands
`
`(b)
`
`(e)
`
`(d)
`
`(c)
`
`(a)
`
`Activating
`receptor
`
`Activating
`ligand
`
`CD16
`
`Antibody
`
`Cell surface
`antigen
`
`Cytotoxic
`granules
`
`Fas
`
`FasL
`
`TRAIL
`receptor
`
`TRAIL
`
`TNF-α
`
`TNF-αα
`receptor
`
`Fig. 2 Mechanisms of NK cell cytotoxicity. The cytotoxicity of NK cells are carried out by two main mechanisms. The first
`mechanism is granule-dependent cytotoxicity where upon triggering by (a) activating receptors or (b) the Fc receptor (CD16),
`the cytotoxic granules in the cytosol of the NK cell are polarized towards the immunological synapse and the contents (mainly
`perforin and granzyme B) are unleashed upon the target cell by exocytosis. The second mechanism is the triggering of apopto-
`sis pathways in the target cell via stimulation of death receptors on by (c) TRAIL or (d) Fas ligand expressed on the NK cell
`surface as well as (e) secretion of TNF-a.
`
`(CD25), b (CD122) and c (CD132) chains. The c
`chain is shared amongst various interleukin receptors
`(IL-4, IL-7, IL-9, IL-15, IL-21), thus named the com-
`mon c chain and it is essential for lymphoid develop-
`ment [56]. The b chain is shared between IL-2 and
`IL-15 receptors [57, 58]. The b and~c chains come
`to form the intermediate affinity IL-2 ⁄ 15
`together
`receptor. The distinction between the high affinity
`receptors for IL-2 and IL-15 comes with the a chains.
`The IL-2Ra chain alone is regarded as the low affinity
`receptor and is believed to lack the capacity to deliver
`intracellular signals because of its short intracellular
`tail [59]. However, when the a chain forms a complex
`with the b and c chains, the high affinity IL-2 receptor
`is formed. The co-expression of all three chains is con-
`fined to regulatory T cells, CD56bright NK cells as well
`as activated conventional CD4+ and CD8+ T cells [60].
`Thus, these cells are expected to give a better response
`to the presence of low dose IL-2 as they have the
`capacity to form a high affinity IL-2 receptor complex.
`
`It has been well defined that IL-2 activation of NK
`cells can result in cytotoxic activity against targets that
`were previously NK-resistant [61]. Observations on
`
`the interaction of autologous and allogeneic NK cells
`with fresh tumour cells have also proved that IL-2 acti-
`vation in vitro enhances the tumour killing potential of
`NK cells [62, 63]. Early reports of IL-2-based treat-
`ment on animal models have established a solid basis
`for efficiency of this approach for cancer immunother-
`apy in many different settings [64–72]. Although cyto-
`toxic T cells have been the primary point of interest,
`especially during the early phases of IL-2 use, the an-
`titumour response triggered by IL-2 were frequently
`attributable to NK cells [73–77]. Whiteside et al. [75]
`have demonstrated in xenograft models of head and
`neck squamous cell carcinoma, that IL-2 activated NK
`cells are as potent as tumour specific CD8+ T cells in
`vivo. Egilmez et al. [76] have shown that sustained
`delivery of IL-2 using biodegradable microspheres can
`promote the NK cell-mediated rejection of
`lung
`tumour xenografts in SCID mice. Likewise, our group
`has demonstrated in a syngeneic murine model of mul-
`tiple myeloma (MM) that NK cells are the primary
`mediators of IL-2 induced tumour rejection [77].
`
`In the clinical setting, the pioneering work of Rosen-
`berg et al.
`[78, 79], which has demonstrated the
`
`ª 2009 Blackwell Publishing Ltd Journal of Internal Medicine 266; 154–181 159
`
` 13652796, 2009, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1365-2796.2009.02121.x, Wiley Online Library on [08/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
`
`UPenn Ex. 2064
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`T. Sutlu & E. Alici |
`
`Review: Natural killer cell-based immunotherapy in cancer
`
`Table 2 NK cell abnormalities in cancer patients
`
`Abnormality
`
`Disease
`
`Decreased cytotoxic
`
`Nonsmall cell lung cancer [215]
`
`activity of NK cells
`
`Hepatocellular carcinoma [216, 217]
`
`Stage IV rectal cancer [218]
`
`Head and neck cancer [219]
`
`Breast cancer [219]
`
`Cervical carcinoma [220]
`
`Squamous cell carcinoma of the
`
`penis [221]
`
`Bronchogenic carcinoma [222]
`
`Ovarian cancer [223]
`
`AML [224]
`
`ALL [224, 225]
`
`B-CLL [226]
`
`CML [227]
`
`MM [228]
`
`Defective expression
`
`Hepatocellular carcinoma [216]
`
`of activating receptors
`
`Metastatic melanoma [229]
`
`AML [230]
`
`MM [95, 231]
`
`Defective NK cell
`
`Metastatic renal cell carcinoma [232]
`
`proliferation
`
`Nasopharyngeal cancer [233]
`
`Increased number of
`CD56bright NK cells
`Defective expression
`
`CML [234]
`
`Head and neck cancer [219]
`
`Breast cancer [219]
`
`Cervical cancer [235]
`
`of intracellular
`
`Colorectal cancer [236]
`
`signalling molecules
`
`Ovarian cancer [237]
`
`Prostate cancer [238]
`
`AML [239]
`
`CML [239]
`
`Decreased NK cell
`
`Nasopharyngeal cancer [233]
`
`counts
`
`Increased NK cell
`
`counts
`
`CML [234]
`
`MM [228]
`
`Defective cytokine
`
`AML [224]
`
`production
`
`ALL [224, 225]
`
`CML [240]
`
`potent immunostimulatory effect of IL-2 in advanced
`cancer patients, resulted in a great
`interest for the
`use of cytokines and immune effector cells for the
`treatment of cancer. Further reports have shown that
`
`IL-2 treatment results in in vivo activation of NK cell
`cytotoxicity [80] and this effect is dependent on the
`dose and schedule of IL-2 administration [81]. Since
`then, such an approach of stimulating endogenous NK
`cells with cytokines in an attempt to promote in vivo
`killing of
`tumour cells has been used by many
`investigators.
`
`It has been observed that IL-2 treatment of some can-
`cer patients receiving a T-cell depleted allogeneic
`BMT was well tolerated, decreased relapse risk and
`increased survival compared to those not receiving
`IL-2 [82]. Such observations have drawn more and
`more interest for the use of NK cells in cancer immu-
`notherapy. Other investigators have shown that IL-2
`administration stimulates the ERK signalling pathway
`in especially CD56bright NK cells and CD14+ mono-
`cytes, but not CD3+ T cells, which suggests that it
`has a distinct way of acting on this lymphocyte
`subpopulation [83].
`
`Interleukin-2 has received FDA approval for the treat-
`ment of metastatic renal cell carcinoma (RCC) in
`1992 based on its ability to induce an objective
`response rate of 15–20% [84]. It has also been dem-
`onstrated that in RCC patients undergoing IL-2-based
`therapy and nephrectomy, a higher percentage of
`circulating NK cells is a predictor of response [85].
`
`Natural killer cells have also been shown to play an
`important role in the effective treatment or preven-
`tion of AIDS-associated lymphoma using low-dose
`IL-2 infusions
`[86]. Treatment of patients with
`AIDS-associated malignancies results in an increase
`in absolute NK cell numbers, whilst no significant
`change in T-cell subsets or plasma HIV RNA level
`is seen [87].
`
`The use of IL-2 alone has been attempted in many
`other tumour types, mostly as an adjuvant to chemo-
`therapy or stem cell transplantation (SCT). Treatment
`of patients with breast cancer and lymphoma using
`IL-2 was shown to increase the number of circulating
`NK cells and their cytotoxicity against NK resistant
`breast cancer and lymphoma cell
`lines significantly
`[88]. Burns et al. have treated 23 lymphoma ⁄ breast
`
`160 ª 2009 Blackwell Publishing Ltd Journal of Internal Medicine 266; 154–181
`
` 13652796, 2009, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1365-2796.2009.02121.x, Wiley Online Library on [08/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
`
`UPenn Ex. 2064
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`T. Sutlu & E. Alici |
`
`Review: Natural killer cell-based immunotherapy in cancer
`
`cancer patients with daily s.c. IL-2 infusions supple-
`mented with two i.v. bolus IL-2 infusions. As antici-
`pated, the therapy resulted in a significant increase in
`total WBC count along with a more than 10-fold
`increase in circulating CD56bright NK cells. Yet, no
`improvement
`in survival or
`relapse was observed
`when compared with matched controls [89].
`
`Kalwak et al. administered intermediate doses of i.v.
`IL-2 to 11 children with poor prognosis solid tumours
`following autologous SCT (ASCT). A significant
`increase in T- and NK-cell counts and an elevated
`level of NK cell cytotoxic activity was observed.
`Again, mainly CD56bright NK cells expanded in vivo
`and most of them expressed the inhibitory receptor
`CD94. Despite the increased NK cell activity, relapses
`occurred frequently [90]. In another study, IL-2 treat-
`ment of NB patients after intensive chemotherapy and
`autologous BMT resulted in a drastic increase of NK
`cell numbers and cytotoxicity during 1-year treatment.
`One of seven patients relapsed and that was the
`patient that showed only a slight increase in the NK
`cell subset in response to IL-2 treatment [91].
`
`The use of IL-2 for inducing NK cell-mediated killing
`of tumour cells has also been a popular approach in
`haematological malignancies. IL-2 has been shown to
`provide stimulation of PBMCs for killing of multiple
`myeloma (MM) cells [92]. Later studies have proved
`that NK cells have an effective cytotoxic activity
`against MM cell
`lines and tumour cells from MM
`patients [93]. Our group has recently demonstrated
`that NK cells from MM patients can be expanded
`ex vivo using GMP-compliant components, and they
`show high cytotoxic activity against autologous MM
`cells whilst retaining their tolerance against normal
`cells of the patient [94]. Other researchers have also
`shown that HLA Class
`I molecules, NCRs and
`NKG2D take part in the recognition of myeloma cells
`by autologous and allogeneic NK cells [95, 96]. Like-
`wise, NK cells from AML patients at remission have
`also been expanded ex vivo and showed cytotoxic
`activity against allogeneic
`and autologous AML
`blasts, which could be further enhanced by IL-2 [97].
`In a clinical AML study, where IL-2 was used alone,
`patients older than 60 years in first complete remis-
`
`sion after induction and consolidation chemotherapy
`were
`randomly assigned to no further
`therapy
`(n = 82) or a 90-day regimen (n = 81) of 14-day
`cycles of low-dose rIL-2, aimed at expanding NK
`cells, followed by 3 day higher doses aimed to induce
`cytotoxicity of expanded NK cells. No prolongation
`of disease-free or overall survival was seen and the
`authors concluded that
`low-dose IL-2 maintenance
`immunotherapy alone is not a successful strategy to
`treat older AML patients [98]. Other researchers have
`observed that IL-2 activated autologous NK cells from
`CML patients can suppress primitive CML progeni-
`tors in long-term culture [99].
`
`Overall, data from the reports reviewed above clearly
`demonstrates that although promising outcomes have
`been observed, low-dose IL-2 treatment is not an opti-
`mal strategy for most indications. In most cases of
`low-dose IL-2 administration (picomolar serum con-
`centrations), a specific expansion of the CD56bright
`NK cell subset, that is known to have a regulatory
`rather than cytotoxic activity, is observed [59]. Within
`the NK cell population, IL-2Ra that confers high
`affinity for IL-2 is uniquely expressed by CD56bright
`cells [100], which could explain their selective expan-
`sion in response to low-dose IL-2. Although such
`treatment has proven to be safe, there is yet no con-
`vincing proof of efficacy. Likewise, the in vivo expan-
`IL-2Ra expressing regulatory cell
`sion of another
`subset; Treg cells could also overwhelm and ⁄ or sup-
`press the antitumour activity that is to be carried out
`by immune effector cells. The potential of Treg cells
`to dampen NK cell-mediated antitumour
`responses
`has primarily been suggested in a murine leukaemia
`model [101]. The effect of Treg cells in cancer immu-
`notherapy has now been well recognized [102, 103]
`and attempts to circumvent such suppression are
`underway [104].
`
`IL-2 in combination with other factors. Studies
`have shown that IL-2, IL-12 and IL-15 stimulate NK
`cell cytotoxicity in vitro and show synergy when used
`in combination [105, 106]. Such cytokines have been
`widely used for in vitro studies to define requirements
`of NK cell activation that could potentially be used in
`cancer immunotherapy. As the b and c chains of IL-2
`
`ª 2009 Blackwell Publishing Ltd Journal of Internal Medicine 266; 154–181 161
`
` 13652796, 2009, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1365-2796.2009.02121.x, Wiley Online Library on [08/02/2023]. See the Term

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