throbber
11 Feb 2004 18:54
`
`AR
`
`AR210-IY22-11.tex AR210-IY22-11.sgm
`
`LaTeX2e(2002/01/18)
`P1: IKH
`10.1146/annurev.immunol.22.012703.104803
`
`Annu. Rev. Immunol. 2004. 22:329–60
`doi: 10.1146/annurev.immunol.22.012703.104803
`Copyright c 2004 by Annual Reviews. All rights reserved
`First published online as a Review in Advance on October 15, 2003
`THE THREE ESOF CANCER IMMUNOEDITING
`Gavin P. Dunn,1Lloyd J. Old,2 and Robert D. Schreiber1
`1Department of Pathology and Immunology, Center for Immunology, Washington
`University School of Medicine, St. Louis, Missouri 63110;
`email: schreiber@immunology.wustl.edu
`2Ludwig Institute for Cancer Research, New York Branch at Memorial Sloan-Kettering
`Cancer Center, New York, NY 10021; email: lold@licr.org
`
`Key Words
`immunosurveillance, tumor, lymphocytes, interferon, tumor sculpting
`■ Abstract After a century of controversy, the notion that the immune system regu-
`lates cancer development is experiencing a new resurgence. An overwhelming amount
`of data from animal models—together with compelling data from human patients—
`indicate that a functional cancer immunosurveillance process indeed exists that acts
`as an extrinsic tumor suppressor. However, it has also become clear that the immune
`system can facilitate tumor progression, at least in part, by sculpting the immunogenic
`phenotype of tumors as they develop. The recognition that immunity plays a dual role
`in the complex interactions between tumors and the host prompted a refinement of the
`cancer immunosurveillance hypothesis into one termed “cancer immunoediting.” In
`this review, we summarize the history of the cancer immunosurveillance controversy
`and discuss its resolution and evolution into the three Es of cancer immunoediting—
`elimination, equilibrium, and escape.
`
`INTRODUCTION
`The concept that the immune system can recognize and eliminate primary develop-
`ing tumors in the absence of external therapeutic intervention has existed for nearly
`100 years. However, the validity of this concept has, in the past, been difficult to
`establish. When first proposed in 1909 (1), the hypothesis could not be experimen-
`tally tested because so little was known at the time about the molecular and cellular
`basis of immunity. Later on, as the field of immunology developed and the concept
`acquired its name—cancer immunosurveillance (2, 3)—experimental testing be-
`came possible but failed to provide evidence for the process, using mice with spon-
`taneous mutations that rendered them immunocompromised but not completely
`immunodeficient (4). Only recently, with the development of gene targeting and
`transgenicmousetechnologiesandthecapacitytoproducehighlyspecificblocking
`monoclonal antibodies (mAb) to particular immune components, has the cancer
`immunosurveillance hypothesis become testable in unequivocal, molecularly de-
`fined murine models of immunodeficiency. Over the past ten years, the use of these
`329
`0732-0582/04/0423-0329$14.00
`
` Access provided by Cedars-Sinai Medical Center on 09/23/19. For personal use only.
`Annu. Rev. Immunol. 2004.22:329-360. Downloaded from www.annualreviews.org
`
`Genome Ex. 1046
`Page 1 of 37
`
`

`

`11 Feb 2004 18:54
`
`AR
`
`AR210-IY22-11.tex AR210-IY22-11.sgm
`
`LaTeX2e(2002/01/18)
`
`P1: IKH
`
`330
`
`DUNN ⌅ OLD ⌅ SCHREIBER
`
`improved in vivo cancer models has provided strong and convincing data that have
`rekindled interest in the cancer immunosurveillance hypothesis. Most recently,
`this conundrum has been further clarified by the demonstration that the immune
`system not only can protect the host against tumor development but also, by se-
`lecting for tumors of lower immunogenicity, has the capacity to promote tumor
`growth. These dual effects of the immune system on developing tumors prompted
`us to refine the cancer immunosurveillance hypothesis into one we termed cancer
`immunoediting (5, 6). We envisage that this process is comprised of three phases
`that are collectively denoted the three Es of cancer immunoediting: elimination,
`equilibrium, and escape. In this review, we first present data supporting the ex-
`istence of the elimination phase (i.e., cancer immunosurveillance) as it occurs in
`mice and humans and propose a model for the molecular and cellular events that
`underlie this process. Second, we provide evidence for a tumor-sculpting role of
`immunity and discuss the relationship between this function and the equilibrium
`and escape phases of cancer immunoediting. Third, we outline the implications of
`this concept for the understanding and treatment of human cancer.
`
`CANCER IMMUNOSURVEILLANCEIN MICE
`Historical Perspective
`The validity of the cancer immunosurveillance hypothesis has emerged only re-
`cently from a long history of heated debate (reviewed in 6). The notion that the
`immune system could protect the host from neoplastic disease was initially pro-
`posed by Ehrlich (1) and formally introduced as the cancer immunosurveillance
`hypothesis nearly 50 years later by Burnet and Thomas (2, 3, 7–9). Based on an
`emerging understanding of the cellular basis of transplantation and tumor immu-
`nity (10–15), Burnet and Thomas predicted that lymphocytes were responsible
`for eliminating continuously arising, nascent transformed cells. However, when
`this prediction was put to the experimental test using nude mice, which were the
`most congenitally immunodeficient mice available at the time (16, 17), no con-
`vincing evidence for such a process was obtained. Specifically, CBA/H strain nude
`mice neither developed increased incidences of carcinogen [methylcholanthrene
`(MCA)]-induced or spontaneous tumors nor did they show shortened periods of
`tumor latency compared with wild-type controls (4, 18–22).
`However, in retrospect, there are several important caveats to these experiments
`that could not have been appreciated at the time. First, the nude mouse is now
`recognized to be an imperfect model of immunodeficiency. These mice produce
`low but detectable numbers of functional populations of ↵ T cells (23–25) and
`therefore can manifest at least some degree of adaptive immunity. Second, the
`existence of natural killer (NK) cells (which are present and function normally
`in nude mice) was not well established at the time (26) and thus very little was
`known about their origins, actions, or roles in promoting innate immunity. In
`addition, the profound influence of innate immunity on adaptive immunity was
`
` Access provided by Cedars-Sinai Medical Center on 09/23/19. For personal use only.
`Annu. Rev. Immunol. 2004.22:329-360. Downloaded from www.annualreviews.org
`
`Genome Ex. 1046
`Page 2 of 37
`
`

`

`11 Feb 2004 18:54
`
`AR
`
`AR210-IY22-11.tex AR210-IY22-11.sgm
`
`LaTeX2e(2002/01/18)
`
`P1: IKH
`
`CANCER IMMUNOEDITING
`
`331
`
`not recognized (27). Thus, the residual adaptive immune system in the presence
`of a fully functional innate immune system may provide the nude mouse with
`at least some cancer immunosurveillance capacity. Third, the CBA/H strain mice
`used in Stutman’s MCA carcinogenesis experiments express the highly active
`isoform of the aryl hydroxylase enzyme that is required to metabolize MCA into its
`carcinogenic form (28, 29). Therefore, it is conceivable that MCA-induced cellular
`transformation in CBA/H strain mice occurred so efficiently that it masked any
`protective effect that immunity could provide. Nevertheless, since these caveats
`can only be appreciated in hindsight, the Stutman experiments were considered to
`be so convincing that by the end of the 1970s, the death knell had sounded for the
`cancer immunosurveillance hypothesis.
`
`THERENAISSANCEOF CANCER IMMUNOSURVEILLANCE
`IFN-, Perforin, and Lymphocytes in Tumor Immunity
`In the 1990s, two sets of studies incited renewed interest in cancer immunosurveil-
`lance. First, endogenously produced interferon- (IFN-) was shown to protect
`the host against the growth of transplanted tumors and the formation of primary
`chemically induced and spontaneous tumors (30–33). The injection of neutral-
`izing monoclonal antibodies specific for IFN- into mice bearing transplanted,
`established Meth A tumors blocked LPS-induced tumor rejection (30). In addi-
`tion, transplanted fibrosarcomas grew faster and more efficiently in mice treated
`with IFN--specific mAb. These observations were then extended to models of
`primary tumor formation. IFN--insensitive 129/SvEv mice lacking either the
`IFNGR1 ligand-binding subunit of the IFN- receptor or STAT1, the transcrip-
`tion factor responsible for mediating much of IFN-’s biologic effects on cells
`(34), were found to be 10–20 times more sensitive than wild-type mice to tumor
`induction by methylcholanthrene (31). Specifically, these mice developed more
`tumors, more rapidly, and at lower MCA doses than did wild-type controls. These
`results were subsequently confirmed by independent experiments using C57BL/6
`strain mice lacking the gene encoding IFN- itself (32). Similarly, in models of
`genetically driven tumorigenesis, mice lacking the p53 tumor suppressor gene
`and either IFNGR1 or STAT1 formed a wider spectrum of tumors compared with
`IFN--sensitive mice lacking only p53 (31). In addition, compared to their IFN--
`sufficient counterparts, IFN- / C57BL/6 mice showed an increased incidence
`of disseminated lymphomas, and IFN- / BALB/c mice displayed an increased
`incidence of spontaneous lung adenocarcinomas (33).
`Second, mice lacking perforin (pfp/) were found to be more susceptible
`to MCA-induced and spontaneous tumor formation compared with their wild-
`type counterparts (32, 33, 35–37). Perforin is a component of the cytolytic gran-
`ules of cytotoxic T cells and NK cells that plays an important role in mediating
`lymphocyte-dependent killing (38). Following challenge with MCA, pfp/ mice
`developed significantly more tumors compared with wild-type mice treated in the
`
` Access provided by Cedars-Sinai Medical Center on 09/23/19. For personal use only.
`Annu. Rev. Immunol. 2004.22:329-360. Downloaded from www.annualreviews.org
`
`Genome Ex. 1046
`Page 3 of 37
`
`

`

`11 Feb 2004 18:54
`
`AR
`
`AR210-IY22-11.tex AR210-IY22-11.sgm
`
`LaTeX2e(2002/01/18)
`
`P1: IKH
`
`332
`
`DUNN ⌅ OLD ⌅ SCHREIBER
`
`same manner (32, 35, 36). Untreated pfp/ mice also showed a high incidence of
`spontaneous disseminated lymphomas, which was accelerated on a p53/ back-
`ground (37). BALB/c mice lacking perforin also displayed a low incidence of
`spontaneous lung adenocarcinomas, which was not observed in wild-type mice
`(33). Taken together, these observations demonstrated that tumor development
`in mice was controlled by components of the immune system and stimulated a
`considerable amount of work aimed at better defining this process (Table 1).
`The definitive work demonstrating the existence of an IFN-- and lymphocyte-
`dependent cancer immunosurveillance process was based on experiments employ-
`ing gene-targeted mice that lack the recombinase activating gene (RAG)-2 (5).
`Mice lacking RAG-2 (or its obligate partner RAG-1) cannot rearrange lymphocyte
`antigen receptors and thus lack T, B, and NKT cells (39). Since RAG-2 expression
`is limited to cells of the immune system, the use of RAG-2/ mice provided an
`appropriate model to study the effects of host immunodeficiency on tumor devel-
`opment because, unlike other genetic models of immunodeficiency (such as SCID
`mice), the absence of RAG-2 would not result in impaired DNA repair in nonlym-
`phoid cells undergoing transformation. Following challenge with MCA, 129/SvEv
`RAG-2/ mice developed sarcomas more rapidly and with greater frequency than
`genetically matched wild-type controls (5) (Figure 1A). After 160 days, 30/52
`RAG-2/ mice formed tumors, compared with 11/57 wild-type mice. Similar
`findings were obtained in MCA tumorigenesis experiments that used RAG-1/
`C57BL/6 mice (40). Moreover, Helicobacter-free RAG-2/ 129/SvEv mice aged
`in a specific pathogen-free mouse facility and maintained on broad-spectrum an-
`tibiotics formed far more spontaneous epithelial tumors than did wild-type mice
`housed in the same room (5; A.T. Bruce & R.D. Schreiber, unpublished observa-
`tions) (Figure 1B). Specifically, 26/26 RAG-2/ mice ranging in age from 13–
`24 months developed spontaneous neoplasia, predominantly of the intestine; 8 of
`these mice had premalignant intestinal adenomas, 17 had intestinal adenocarcino-
`mas, and 1 had both an intestinal adenoma and a lung adenocarcinoma. In contrast,
`only 5/20 wild-type mice aged 13–24 months developed spontaneous neoplasia,
`which was predominantly benign. Three wild-type mice developed adenomas of
`the Harderian gland, lung, and intestine, respectively; one developed a Harderian
`gland adenocarcinoma; and one developed an endometrial stromal carcinoma.
`Thus, lymphocytes protect mice against the formation of both chemically induced
`and spontaneous tumors.
`The overlap between the IFN-- and lymphocyte-dependent tumor suppressor
`pathways was explored by comparing tumor formation in 129/SvEv mice lack-
`ing either IFN- responsiveness (IFNGR1/ or STAT1/ mice), lymphocytes
`(RAG-2/ mice), or both [RAG-2/ X STAT1/ (RkSk) mice] (5). Each of
`the four lines of gene-targeted mice formed three times more chemically induced
`tumors than syngeneic wild-type mice when injected with a single 100 µg dose
`of MCA (Figure 1A). Since no significant differences were detected between any
`of the gene-targeted mice, the conclusion was reached that the IFN-/STAT1
`and lymphocyte-dependent extrinsic tumor suppressor mechanisms were heavily
`
` Access provided by Cedars-Sinai Medical Center on 09/23/19. For personal use only.
`Annu. Rev. Immunol. 2004.22:329-360. Downloaded from www.annualreviews.org
`
`Genome Ex. 1046
`Page 4 of 37
`
`

`

`11 Feb 2004 18:54
`
`AR
`
`AR210-IY22-11.tex AR210-IY22-11.sgm
`
`LaTeX2e(2002/01/18)
`
`P1: IKH
`
`CANCER IMMUNOEDITING
`
`333
`
`TABLE 1 Enhanced susceptibility of immunodeficient mice to chemically induced and
`spontaneous tumors
`
`Technology
`
`RAG-2/
`
`Immune status
`
`Lacks T, B, NKT cells
`
`RAG-2/⇥STAT1/
`(RkSk)
`
`Lacks T, B, NKT cells;
`IFN-, ↵/-insensitive
`
`RAG-1/
`BALB/c SCID
`TCR/
`TCR/
`
`J↵281/
`LMP2/
`Anti-asialo-GM1
`
`Anti-NK1.1
`Anti-Thy1
`STAT1/
`
`IFNGR1/
`
`IFN- /
`
`Lacks T, B, NKT cells
`Lacks T, B, NKT cells
`Lacks ↵ T cells
`Lacks T cells
`
`Lacks NKT cell subset
`Lacks LMP2 subunit
`Lacks NK cells,
`mono-cytes/macrophages
`Lacks NK, NKT cells
`Lacks T cells
`IFN--, ↵/-insensitive
`
`IFN--insensitive
`
`Lacks IFN-
`
`GM-CSF/IFN- /
`
`Lacks GM-CSF, IFN-
`
`Pfp/⇥IFN- /
`
`Lacks Perforin, IFN-
`
`Pfp/
`
`TRAIL/
`Anti-TRAIL
`
`IL-12p40/
`Wt + IL-12
`Wt + ↵-GalCer
`
`Lacks Perforin
`
`Lacks TRAIL
`Blockade of TRAIL
`function
`
`Lacks IL-12
`Exogenous IL-12
`Exogenous NKT cell
`activation
`
`Tumor susceptibility
`relative to wild type
`" MCA-induced sarcomas;
`" spontaneous intestinal neoplasia
`" MCA-induced sarcomas;
`" spontaneous intestinal and
`mammary neoplasia
`" MCA-induced sarcomas
`" MCA-induced sarcomas
`" MCA-induced sarcomas
`" MCA-induced sarcomas;
`" DMBA/TPA-induced skin
`tumors
`" MCA-induced sarcomas
`" Spontaneous uterine neoplasms
`" MCA-induced sarcomas
`" MCA-induced sarcomas
`" MCA-induced sarcomas
`" MCA-induced sarcomas;
`wider tumor spectrum
`in STAT1/⇥p53/
`" MCA-induced sarcomas;
`wider tumor spectrum in
`IFNGR1/⇥p53/
`" MCA-induced sarcomas;
`B6: " spontaneous disseminated
`lymphomas;
`BALB/c: " spontaneous lung
`adenocarcinomas
`" Spontaneous lymphomas;
`" nonlymphoid solid cancers
`" MCA-induced sarcomas;
`" spontaneous disseminated
`lymphomas
`" MCA-induced sarcomas;
`" spontaneous disseminated
`lymphomas
`" MCA-induced sarcomas
`" MCA-induced sarcomas;
`" spontaneous sarcomas,
`disseminated lymphomas
`" MCA-induced sarcomas
`# MCA-induced sarcomas
`# MCA-induced sarcomas
`
`References
`
`(5)
`
`(5)
`
`(40)
`(40)
`(58)
`(58)
`
`(32, 36, 40)
`(169)
`(40)
`
`(36, 40)
`(36)
`(5, 31)
`
`(5, 31)
`
`(32, 33)
`
`(55)
`
`(32, 33)
`
`(32, 33, 35–37)
`
`(61)
`(60)
`
`(36)
`(62)
`(63)
`
` Access provided by Cedars-Sinai Medical Center on 09/23/19. For personal use only.
`Annu. Rev. Immunol. 2004.22:329-360. Downloaded from www.annualreviews.org
`
`Genome Ex. 1046
`Page 5 of 37
`
`

`

`11 Feb 2004 18:54
`
`AR
`
`AR210-IY22-11.tex AR210-IY22-11.sgm
`
`LaTeX2e(2002/01/18)
`
`P1: IKH
`
`334
`
`DUNN ⌅ OLD ⌅ SCHREIBER
`
`Figure 1 Increased incidence of chemically induced and spontaneous tumors in im-
`munodeficient mice. (A) Age- and sex-matched mice were inoculated with 100 µg
`MCA and monitored for tumor development for 160 days. (B) Mice housed in a
`specific pathogen-free facility were monitored for spontaneous tumor development
`between 13–24 months. Adapted from Shankaran et al. (5).
`
`overlapping. However, RkSk mice developed spontaneous breast tumors that were
`not observed in wild-type or RAG-2/ mice, therefore demonstrating that the
`overlap between the two pathways was incomplete (Figure 1B). Similar findings
`were made in carcinogenesis experiments employing mice that lacked either per-
`forin, IFN-, or both, where a small increase was observed in tumor induction
`in the doubly deficient mice compared with mice lacking only one of the two
`components (32).
`Identification of the Components of the Immmunosurveillance
`Network
`TUMOR CELLS AS KEY TARGETS OF IFN- The finding that endogenously pro-
`duced IFN- played a critical role in protecting mice against tumor develop-
`ment stimulated a search for the physiologically important cellular targets of this
`cytokine. Two approaches demonstrated that the tumor cell itself is an impor-
`tant IFN- target in tumor rejection. In the first, the effects of ablating IFN-
`sensitivity on the immunogenicity of IFN--sensitive tumor cells was assessed us-
`ing models of tumor cell
`transplantation (30). Meth A tumor cells, when
`
` Access provided by Cedars-Sinai Medical Center on 09/23/19. For personal use only.
`Annu. Rev. Immunol. 2004.22:329-360. Downloaded from www.annualreviews.org
`
`Genome Ex. 1046
`Page 6 of 37
`
`

`

`11 Feb 2004 18:54
`
`AR
`
`AR210-IY22-11.tex AR210-IY22-11.sgm
`
`LaTeX2e(2002/01/18)
`
`P1: IKH
`
`CANCER IMMUNOEDITING
`
`335
`
`engineered to be unresponsive to IFN- by overexpression of a dominant-negative
`IFNGR1 mutant (mgR.1IC) (41), grew more aggressively than mock-transfected
`controls when transplanted into na¨ıve syngeneic wild-type hosts and were resistant
`to LPS-induced tumor rejection (30, 41). Unlike their IFN--sensitive counter-
`parts, IFN--insensitive Meth A.mgR.1IC cells failed to prime na¨ıve recipients
`for development of Meth A immunity and were poorly recognized when injected
`into mice with pre-established immunity to the parental wild-type tumor cell line.
`Similar results were obtained with a second fibrosarcoma derived from a C57BL/6
`mouse(MCA-207).Thesecondapproachemployedanoppositestrategywherethe
`effects on in vivo tumor growth were assessed following restoration of IFN- sen-
`sitivity to tumor cells generated in IFN--insensitive IFNGR1/ mice (31). When
`transplanted into wild-type mice, IFNGR1-deficient RAD.gR.28 tumor cells were
`highly tumorigenic and formed progressively growing tumors even when injected
`at very low cell number (10–100 cells/mouse). In contrast, when RAD.gR.28 cells
`were rendered responsive to IFN- by complementation with wild-type IFNGR1,
`theresultingtumorcellline(RAD.gR.28.mgR)washighlyimmunogenicandfailed
`toformprogressivelygrowingtumorsinwild-typerecipientsevenwheninjectedat
`high cell number (5 ⇥ 106 cells/mouse). Demonstration that RAD.gR.28.mgR re-
`jection occurred via an IFN--dependent immunologic mechanism was evidenced
`by the observations that (a) rejection of RAD.gR.28.mgR cells in wild-type mice
`was inhibited by administration of IFN- mAb (31), (b) rejection was inhibited
`if wild-type mice were depleted of either CD4+ or CD8+ T cells (A.T. Bruce &
`R.D. Schreiber, unpublished observations), and (c) RAD.gR.28.mgR cells formed
`progressively growing tumors when injected into RAG-2/ mice (31). Thus, the
`effects of using IFN--insensitive tumor cells are the same as blocking IFN-
`availability in the intact mouse: Immune rejection of the tumor is inhibited. To-
`gether, these results formed the basis for the conclusion that the tumor cell is a
`physiologically relevant target of IFN- in the tumor rejection process.
`Subsequent studies have pointed to several effects of IFN- on tumor cells
`that could promote tumor elimination. IFN-’s capacity to enhance tumor cell
`immunogenicity by upregulating components of the MHC class I antigen process-
`ing and presentation pathway has been shown to be sufficient for tumor rejection.
`IFN--insensitive RAD.gR.28 tumor cells engineered for enforced expression of
`either TAP-1 (5) or H-2Db (A.T. Bruce & R.D. Schreiber, unpublished obser-
`vations) were rejected when transplanted into na¨ıve syngeneic recipients in an
`immunologic manner that was indistinguishable from that of IFN--responsive
`RAD.gR.28.mgR cells. In contrast, RAD.gR.28 cells engineered for expression of
`H-2Kbwerenotrejected(5).ThefindingthatenforcedexpressionofH-2Db,butnot
`H-2Kb, caused rejection of RAD.gR.28 corresponds to the H-2Db MHC restriction
`displayed by protective CD8+ T cells that arise naturally in mice immunized with
`RAD.gR.28.mgR (A.T. Bruce & R.D. Schreiber, unpublished observations). Thus,
`the capacity of IFN- to regulate tumor cell immunogenicity via enhancement of
`MHC class I pathway function is a physiologically relevant action that promotes
`tumor rejection.
`
` Access provided by Cedars-Sinai Medical Center on 09/23/19. For personal use only.
`Annu. Rev. Immunol. 2004.22:329-360. Downloaded from www.annualreviews.org
`
`Genome Ex. 1046
`Page 7 of 37
`
`

`

`11 Feb 2004 18:54
`
`AR
`
`AR210-IY22-11.tex AR210-IY22-11.sgm
`
`LaTeX2e(2002/01/18)
`
`P1: IKH
`
`336
`
`DUNN ⌅ OLD ⌅ SCHREIBER
`
`Other known IFN--dependent effects on developing tumors may also con-
`tribute to the rejection process; however, their physiologic relevance to the process
`has not yet been established. IFN- has profound antiproliferative and/or proapop-
`toticeffectsoncertaintumorcells.Intheformercase,IFN- caninduceexpression
`of cell cycle inhibitors such as p21WAF1/CIP1 or p27KIP1 that bind to and inhibit the
`cyclin-dependentkinaseCDK-2(42)orCDK-4(43),respectively.Inthelattercase,
`IFN- can induce expression of gene products such as caspase-1 (44, 45), Fas, and
`Fasligand(46)that,undertheproperconditions,canpromotetumorcellapoptosis.
`IFN- can also stimulate tumor cells to produce the chemokines CXCL-9 (Mig)
`and CXCL-10 (IP-10), which, in addition to having potent chemoattractant activity
`for CXCR3-expressing leukocyte populations, also function as powerful inhibitors
`of angiogenesis (47–52). Although all the aforementioned processes likely con-
`tribute in some way to the antitumor response, the relative importance and interre-
`lationships between the immunologic and nonimmunologic actions of IFN- on
`developing tumors in promoting tumor rejection requires further analysis.
`
`HOST CELLS AS POTENTIAL ADDITIONAL TARGETS OF IFN- Evidence has also
`been obtained supporting a role for IFN- and/or STAT1 at the level of the host
`immune system in the tumor rejection process. IFN--unresponsive mice lacking
`STAT1 failed to reject highly immunogenic P198 tumor cells that were completely
`eliminated in wild-type mice (53). Similar findings have also been made using the
`highly immunogenic RAD.gR.28.mgR fibrosarcoma cell line that was rejected in
`wild-type mice but grew progressively in STAT1/ mice (V. Shankaran & R.D.
`Schreiber, unpublished observations). In addition, T cells derived from STAT1/
`mice immunized with poorly immunogenic P1.HTR tumor cells in the presence
`of IL-12 failed to express cytolytic activity against the tumor. In contrast, T cells
`derived from similarly immunized wild-type mice developed potent cytocidal ca-
`pacity.MicelackingSTAT6,whichtendtopolarizetheirCD4+Tcellcompartment
`more easily into Th1 cells, spontaneously rejected poorly immunogenic P1.HTR
`tumor cells that grew progressively in wild-type mice (54). Thus, these stud-
`ies suggest that IFN-’s well-recognized STAT1-dependent promotion of CD4+
`T cell polarization into Th1 cells facilitates development of the appropriate type
`of cellular immune response needed for tumor rejection.
`Another study revealed a more indirect immunological action of IFN- at the
`level of the host in preventing tumor development (55). Both GM-CSF/IFN- /
`doubly deficient and GM-CSF/IL-3/IFN- / triply deficient mice were found to
`be highly susceptible to bacterial infection, displayed acute and chronic inflamma-
`tion in a variety of different organs, and developed high incidences of spontaneous
`lymphoma and nonlymphoid solid cancers. The incidences of infection, inflamma-
`tion, and neoplasia were much reduced in mice lacking GM-CSF alone, IL-3 and
`GM-CSF only, or IFN- alone. Tumor development in the IL-3/GM-CSF/IFN-
`triply gene-targeted mice was prevented or delayed by maintaining the mice on
`broad-spectrum antibiotics from birth. These results suggest a role for IFN-,
`in combination with GM-CSF, in controlling chronic infections that can lead to
`
` Access provided by Cedars-Sinai Medical Center on 09/23/19. For personal use only.
`Annu. Rev. Immunol. 2004.22:329-360. Downloaded from www.annualreviews.org
`
`Genome Ex. 1046
`Page 8 of 37
`
`

`

`11 Feb 2004 18:54
`
`AR
`
`AR210-IY22-11.tex AR210-IY22-11.sgm
`
`LaTeX2e(2002/01/18)
`
`P1: IKH
`
`CANCER IMMUNOEDITING
`
`337
`
`a chronic inflammatory state that ultimately may result in cancer development.
`Clearly,therelationshipbetweenbacterial/microbialimmunosurveillanceandcan-
`cer immunosurveillance warrants further analysis but must await the development
`of in vivo models that can unequivocally differentiate between the two processes.
`Finally, other studies have suggested that host cells of nonimmunologic origin
`may also be important targets of IFN- in the antitumor response (56, 57). These
`studies report that IFN- can induce angiostatic effects in tumors by targeting
`nontransformed host cells that are in close proximity to the tumor. It is possible
`that the underlying mechanism of this effect is similar to the one that has already
`been discussed in the context of the tumor cells themselves—the IFN--dependent
`induction in host stromal cells of the angiostatic chemokines IP-10 and Mig.
`
`THE CELLULAR EFFECTORS OF CANCER IMMUNOSURVEILLANCE Otherstudieshave
`begun to shed light on the specific lymphocyte subsets that are involved in can-
`cer immunosurveillance. Together, these studies have shown that components
`of both the adaptive and innate immune systems participate in the process.
`Girardi et al. (58) examined the relative contributions of different T-cell subsets in
`blocking primary tumor formation in mice lacking ↵ T cells (TCR/) and/or
` T cells (TCR/). MCA treatment of either type of TCR/ mouse led to
`an increased incidence of fibrosarcomas and spindle cell carcinomas compared
`with wild-type controls, thereby showing that both ↵ and T-cell subsets play
`critical and nonredundant host-protective roles in this particular model of tumor
`development. However, in an initiation/promotion model of DMBA- and TPA-
`induced skin tumorigenesis, TCR/ mice showed an increased susceptibility to
`tumor formation and a higher incidence of papilloma-to-carcinoma progression
`than wild-type mice, whereas TCR/ mice did not. This result suggests that im-
`munosurveillance may be a multivariable process requiring the actions of different
`immune effectors in a manner dependent on the tumor’s cell type of origin, mech-
`anism of transformation, anatomic localization, and mechanism of immunologic
`recognition.
`NK and NKT cells represent cellular populations of the innate immune com-
`partment that were shown to protect the host from tumor formation. C57BL/6
`mice depleted of both NK and NKT cells using the NK1.1 mAb were two to three
`times more susceptible to MCA-induced tumor formation than wild-type controls
`(40). In the same study, C57BL/6 mice depleted of NK cells following anti-asialo-
`GM1 treatment were two to three times more prone to developing MCA-induced
`tumors than control counterparts. Although anti-asialo-GM1 can also deplete ac-
`tivated macrophages, this study nevertheless supports the involvement of cells of
`innate immunity in blocking primary tumor development. A role for NKT cells in
`this process was implicated when J↵281/ mice, which lack a large population of
`V↵14J↵281-expressing invariant NKT cells, were found to develop MCA-induced
`sarcomas at a higher incidence than their wild-type counterparts (36).
`Additional evidence pointing to cells of innate immunity as critical effec-
`tors of cancer immunosurveillance comes from studies of the TNF-related
`
` Access provided by Cedars-Sinai Medical Center on 09/23/19. For personal use only.
`Annu. Rev. Immunol. 2004.22:329-360. Downloaded from www.annualreviews.org
`
`Genome Ex. 1046
`Page 9 of 37
`
`

`

`11 Feb 2004 18:54
`
`AR
`
`AR210-IY22-11.tex AR210-IY22-11.sgm
`
`LaTeX2e(2002/01/18)
`
`P1: IKH
`
`338
`
`DUNN ⌅ OLD ⌅ SCHREIBER
`
`apoptosis-inducing ligand (TRAIL). A member of the TNF superfamily that in-
`duces apoptosis through engagement of the TRAIL-R2 (DR5) receptor in mice,
`TRAIL is expressed constitutively on a subset of liver NK cells and is induced by
`either IFN- or IFN-↵/ in monocytes, NK cells, and dendritic cells (59). When
`injected with low doses of MCA, C57BL/6 strain mice treated with neutralizing
`antibodies to TRAIL (60) or lacking the TRAIL gene (61) developed fibrosar-
`comas at a higher incidence than wild-type controls. Moreover, C57BL/6 strain
`p53+/ mice treated with the same neutralizing TRAIL antibody exhibited a higher
`incidence of spontaneous sarcoma and disseminated lymphoma formation over a
`two-year span than control IgG-treated mice (60). Further study will be required
`to identify the specific innate cell subsets that manifest the TRAIL-dependent
`antitumor effects.
`Finally, evidence also exists showing that enhancing immune system activity
`leads to reduced primary tumor formation in models of MCA tumorigene-
`sis. Mice treated with either IL-12 (62) or the prototypic NKT cell activator ↵-
`galactosylceramide (↵-GalCer) (63) throughout the MCA carcinogenesis process
`had a reduced incidence of tumors after longer latency periods than control mice.
`In summary, using a variety of well-characterized gene-targeted mice, specific
`immune system activators, and blocking monoclonal antibodies highly specific
`for distinct immunologic components, a large body of work has now accumulated
`to support the statement that the immune system indeed functions to protect the
`murine host against development of both chemically induced and spontaneous
`tumors (Table 1).
`
`CANCER IMMUNOSURVEILLANCEIN HUMANS
`Given that there is significant evidence supporting the existence of a cancer im-
`munosurveillanceprocessinmice,doesasimilarprocessexistinhumans?Analysis
`ofindividualswithcongenitaloracquiredimmunodeficienciesorpatientsundergo-
`ing immunosuppressive therapy has documented a highly elevated incidence of vi-
`rally induced malignancies such as Kaposi’s sarcoma, non-Hodgkin’s lymphoma,
`and cancers of the anal and urogenital tracts compared with immunocompetent
`individuals (64–66). However, the study of the incidence of cancers of nonviral
`origins that may take many years to develop is confounded by the variety of viral
`and bacterial infections to which these immunodeficient/immunosuppressed pa-
`tients are susceptible and by the more rapid appearance of virally induced tumors.
`Nevertheless, one can draw upon three lines of evidence to suggest that cancer
`immunosurveillance indeed occurs in humans: (a) immunosuppressed transplant
`recipients display higher incidences of nonviral cancers than age-matched im-
`munocompetent control populations; (b) cancer patients can develop spontaneous
`adaptive and innate immune responses to the tumors that they bear, and (c) the
`presence of lymphocytes within the tumor can be a positive prognostic indicator
`of patient survival.
`
` Access provided by Cedars-Sinai Medical Center on 09/23/19. For personal use only.
`Annu. Rev. Immunol. 2004.22:329-360. Downloaded from www.annualreviews.org
`
`Genome Ex. 1046
`Page 10 of 37
`
`

`

`11 Feb 2004 18:54
`
`AR
`
`AR210-IY22-11.tex AR210-IY22-11.sgm
`
`LaTeX2e(2002/01/18)
`
`P1: IKH
`
`CANCER IMMUNOEDITING
`
`339
`
`Transplant Recipients Display Increased Incidences
`of Malignancies
`Increased relative risk ratios have indeed been observed in immunosuppressed
`transplant recipients for a broad subset of tumors that have no apparent viral ori-
`gin. Assessment of 5692 renal transplant patients from 1964–1982 in Finland,
`Denmark, Norway, and Sweden showed increased standardized cancer incidence
`ratios for colon, lung, bladder, kidney, ureter, and endocrine tumors compared to
`the general population (67). For example, the relative risks for colon cancer were
`3.2 for men and 3.9 for women. In addition, analysis of 925 patients who received
`cadaveric renal transplants from 1965 to

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