throbber
Cell, Vol. 100, 57–70, January 7, 2000, Copyright ª 2000 by Cell Press
`
`The Hallmarks of Cancer
`
`Review
`
`Douglas Hanahan* and Robert A. Weinberg†
`* Department of Biochemistry and Biophysics and
`Hormone Research Institute
`University of California at San Francisco
`San Francisco, California 94143
`† Whitehead Institute for Biomedical Research and
`Department of Biology
`Massachusetts Institute of Technology
`Cambridge, Massachusetts 02142
`
`After a quarter century of rapid advances, cancer re-
`search has generated a rich and complex body of knowl-
`edge, revealing cancer to be a disease involving dy-
`namic changes in the genome. The foundation has been
`set in the discovery of mutations that produce onco-
`genes with dominant gain of function and tumor sup-
`pressor genes with recessive loss of function; both
`classes of cancer genes have been identified through
`their alteration in human and animal cancer cells and
`by their elicitation of cancer phenotypes in experimental
`models (Bishop and Weinberg, 1996).
`Some would argue that the search for the origin and
`treatment of this disease will continue over the next
`quarter century in much the same manner as it has in
`the recent past, by adding further layers of complexity
`to a scientific literature that is already complex almost
`beyond measure. But we anticipate otherwise: those
`researching the cancer problem will be practicing a dra-
`matically different type of science than we have experi-
`enced over the past 25 years. Surely much of this change
`will be apparent at the technical level. But ultimately,
`the more fundamental change will be conceptual.
`We foresee cancer research developing into a logical
`science, where the complexities of the disease, de-
`scribed in the laboratory and clinic, will become under-
`standable in terms of a small number of underlying prin-
`ciples. Some of these principles are even now in the
`midst of being codified. We discuss one set of them in
`the present essay: rules that govern the transformation
`of normal human cells into malignant cancers. We sug-
`gest that research over the past decades has revealed
`a small number of molecular, biochemical, and cellular
`traits—acquired capabilities—shared by most and per-
`haps all types of human cancer. Our faith in such simplifi-
`cation derives directly from the teachings of cell biology
`that virtually all mammalian cells carry a similar molecu-
`lar machinery regulating their proliferation, differentia-
`tion, and death.
`Several lines of evidence indicate that tumorigenesis
`in humans is a multistep process and that these steps
`reflect genetic alterations that drive the progressive
`transformation of normal human cells into highly malig-
`nant derivatives. Many types of cancers are diagnosed
`in the human population with an age-dependent inci-
`dence implicating four to seven rate-limiting, stochastic
`events (Renan, 1993). Pathological analyses of a number
`of organ sites reveal lesions that appear to represent
`the intermediate steps in a process through which cells
`
`evolve progressively from normalcy via a series of pre-
`malignant states into invasive cancers (Foulds, 1954).
`These observations have been rendered more con-
`crete by a large body of work indicating that the ge-
`nomes of tumor cells are invariably altered at multiple
`sites, having suffered disruption through lesions as sub-
`tle as point mutations and as obvious as changes in
`chromosome complement (e.g., Kinzler and Vogelstein,
`1996). Transformation of cultured cells is itself a
`multistep process: rodent cells require at least two intro-
`duced genetic changes before they acquire tumorigenic
`competence, while their human counterparts are more
`difficult to transform (Hahn et al., 1999). Transgenic
`models of tumorigenesis have repeatedly supported the
`conclusion that tumorigenesis in mice involves multiple
`rate-limiting steps (Bergers et al., 1998; see Oncogene,
`1999, R. DePinho and T. E. Jacks, volume 18[38], pp.
`5248–5362). Taken together, observations of human
`cancers and animal models argue that tumor develop-
`ment proceeds via a process formally analogous to Dar-
`winian evolution,
`in which a succession of genetic
`changes, each conferring one or another type of growth
`advantage, leads to the progressive conversion of nor-
`mal human cells into cancer cells (Foulds, 1954; Nowell,
`1976).
`
`An Enumeration of the Traits
`The barriers to development of cancer are embodied
`in a teleology: cancer cells have defects in regulatory
`circuits that govern normal cell proliferation and homeo-
`stasis. There are more than 100 distinct types of cancer,
`and subtypes of tumors can be found within specific
`organs. This complexity provokes a number of ques-
`tions. How many distinct regulatory circuits within each
`type of target cell must be disrupted in order for such
`a cell to become cancerous? Does the same set of
`cellular regulatory circuits suffer disruption in the cells
`of the disparate neoplasms arising in the human body?
`Which of these circuits operate on a cell-autonomous
`basis, and which are coupled to the signals that cells
`receive from their surrounding microenvironment within
`a tissue? Can the large and diverse collection of cancer-
`associated genes be tied to the operations of a small
`group of regulatory circuits?
`We suggest that the vast catalog of cancer cell geno-
`types is a manifestation of six essential alterations in cell
`physiology that collectively dictate malignant growth
`(Figure 1): self-sufficiency in growth signals, insensitivity
`to growth-inhibitory (antigrowth) signals, evasion of pro-
`grammed cell death (apoptosis),
`limitless replicative
`potential, sustained angiogenesis, and tissue invasion
`and metastasis. Each of these physiologic changes—
`novel capabilities acquired during tumor development—
`represents the successful breaching of an anticancer
`defense mechanism hardwired into cells and tissues.
`We propose that these six capabilities are shared in
`common by most and perhaps all types of human tu-
`mors. This multiplicity of defenses may explain why can-
`cer is relatively rare during an average human lifetime.
`
`NOVARTIS EXHIBIT 2029
`Breckenridge v. Novartis, IPR 2017-01592
`Page 1 of 14
`
`

`

`Cell
`58
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Figure 1. Acquired Capabilities of Cancer
`We suggest that most if not all cancers have acquired the same set
`of functional capabilities during their development, albeit through
`various mechanistic strategies.
`
`We describe each capability in turn below, illustrate with
`a few examples its functional importance, and indicate
`strategies by which it is acquired in human cancers.
`
`Acquired Capability: Self-Sufficiency
`in Growth Signals
`Normal cells require mitogenic growth signals (GS) be-
`fore they can move from a quiescent state into an active
`proliferative state. These signals are transmitted into the
`cell by transmembrane receptors that bind distinctive
`classes of signaling molecules: diffusible growth fac-
`tors, extracellular matrix components, and cell-to-cell
`adhesion/interaction molecules. To our knowledge, no
`type of normal cell can proliferate in the absence of
`such stimulatory signals. Many of the oncogenes in the
`cancer catalog act by mimicking normal growth signal-
`ing in one way or another.
`Dependence on growth signaling is apparent when
`propagating normal cells in culture, which typically pro-
`liferate only when supplied with appropriate diffusible
`mitogenic factors and a proper substratum for their inte-
`grins. Such behavior contrasts strongly with that of tu-
`mor cells, which invariably show a greatly reduced
`dependence on exogenous growth stimulation. The con-
`clusion is that tumor cells generate many of their own
`growth signals, thereby reducing their dependence on
`stimulation from their normal tissue microenvironment.
`This liberation from dependence on exogenously de-
`rived signals disrupts a critically important homeostatic
`mechanism that normally operates to ensure a proper
`behavior of the various cell types within a tissue.
`
`Acquired GS autonomy was the first of the six capabili-
`ties to be clearly defined by cancer researchers, in large
`part because of the prevalence of dominant oncogenes
`that have been found to modulate it. Three common
`molecular strategies for achieving autonomy are evi-
`dent, involving alteration of extracellular growth signals,
`of transcellular transducers of those signals, or of intra-
`cellular circuits that translate those signals into action.
`While most soluble mitogenic growth factors (GFs) are
`made by one cell type in order to stimulate proliferation
`of another—the process of heterotypic signaling—many
`cancer cells acquire the ability to synthesize GFs to
`which they are responsive, creating a positive feedback
`signaling loop often termed autocrine stimulation (Fedi
`et al., 1997). Clearly, the manufacture of a GF by a cancer
`cell obviates dependence on GFs from other cells within
`the tissue. The production of PDGF (platelet-derived
`growth factor) and TGFa (tumor growth factor a) by
`glioblastomas and sarcomas, respectively, are two illus-
`trative examples (Fedi et al., 1997).
`The cell surface receptors that transduce growth-
`stimulatory signals into the cell interior are themselves
`targets of deregulation during tumor pathogenesis. GF
`receptors, often carrying tyrosine kinase activities in
`their cytoplasmic domains, are overexpressed in many
`cancers. Receptor overexpression may enable the can-
`cer cell to become hyperresponsive to ambient levels
`of GF that normally would not trigger proliferation (Fedi
`et al., 1997). For example, the epidermal GF receptor
`(EGF-R/erbB)
`is upregulated in stomach, brain, and
`breast tumors, while the HER2/neu receptor is overex-
`pressed in stomach and mammary carcinomas (Slamon
`et al., 1987; Yarden and Ullrich, 1988). Additionally, gross
`overexpression of GF receptors can elicit ligand-inde-
`pendent signaling (DiFiore et al., 1987). Ligand-indepen-
`dent signaling can also be achieved through structural
`alteration of receptors; for example, truncated versions
`of the EGF receptor lacking much of its cytoplasmic
`domain fire constitutively (Fedi et al., 1997).
`Cancer cells can also switch the types of extracellular
`matrix receptors (integrins) they express, favoring ones
`that transmit progrowth signals (Lukashev and Werb,
`1998; Giancotti and Ruoslahti, 1999). These bifunctional,
`heterodimeric cell surface receptors physically link cells
`to extracellular superstructures known as the extracellu-
`lar matrix (ECM). Successful binding to specific moieties
`of the ECM enables the integrin receptors to transduce
`signals into the cytoplasm that influence cell behavior,
`ranging from quiescence in normal tissue to motility,
`resistance to apoptosis, and entrance into the active
`cell cycle. Conversely, the failure of integrins to forge
`these extracellular links can impair cell motility, induce
`apoptosis, or cause cell cycle arrest (Giancotti and Ru-
`oslahti, 1999). Both ligand-activated GF receptors and
`progrowth integrins engaged to extracellular matrix
`components can activate the SOS-Ras-Raf-MAP kinase
`pathway (Aplin et al., 1998; Giancotti and Ruoslahti,
`1999).
`The most complex mechanisms of acquired GS auton-
`omy derive from alterations in components of the down-
`stream cytoplasmic circuitry that receives and pro-
`cesses the signals emitted by ligand-activated GF
`receptors and integrins. The SOS-Ras-Raf-MAPK cas-
`cade plays a central role here. In about 25% of human
`
`NOVARTIS EXHIBIT 2029
`Breckenridge v. Novartis, IPR 2017-01592
`Page 2 of 14
`
`

`

`Review
`59
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Figure 2. The Emergent Integrated Circuit of the Cell
`Progress in dissecting signaling pathways has begun to lay out a circuitry that will likely mimic electronic integrated circuits in complexity
`and finesse, where transistors are replaced by proteins (e.g., kinases and phosphatases) and the electrons by phosphates and lipids, among
`others. In addition to the prototypical growth signaling circuit centered around Ras and coupled to a spectrum of extracellular cues, other
`component circuits transmit antigrowth and differentiation signals or mediate commands to live or die by apoptosis. As for the genetic
`reprogramming of this integrated circuit in cancer cells, some of the genes known to be functionally altered are highlighted in red.
`
`tumors, Ras proteins are present in structurally altered
`forms that enable them to release a flux of mitogenic
`signals into cells, without ongoing stimulation by their
`normal upstream regulators (Medema and Bos, 1993).
`We suspect that growth signaling pathways suffer
`deregulation in all human tumors. Although this point
`is hard to prove rigorously at present, the clues are
`abundant (Hunter, 1997). For example, in the best stud-
`ied of tumors—human colon carcinomas—about half
`of the tumors bear mutant ras oncogenes (Kinzler and
`Vogelstein, 1996). We suggest that the remaining colonic
`tumors carry defects in other components of the growth
`signaling pathways that phenocopy ras oncogene acti-
`vation. The nature of these alternative, growth-stimulat-
`ing mechanisms remains elusive.
`Under intensive study for two decades, the wiring
`diagram of the growth signaling circuitry of the mamma-
`lian cell is coming into focus (Figure 2). New downstream
`effector pathways that radiate from the central SOS-
`Ras-Raf-MAP kinase mitogenic cascade are being dis-
`covered with some regularity (Hunter, 1997; Rommel
`and Hafen, 1998). This cascade is also linked via a variety
`of cross-talking connections with other pathways; these
`cross connections enable extracellular signals to elicit
`
`multiple cell biological effects. For example, the direct
`interaction of the Ras protein with the survival-promot-
`ing PI3 kinase enables growth signals to concurrently
`evoke survival signals within the cell (Downward, 1998).
`While acquisition of growth signaling autonomy by
`cancer cells is conceptually satisfying, it is also too
`simplistic. We have traditionally explored tumor growth
`by focusing our experimental attentions on the geneti-
`cally deranged cancer cells (Figure 3, left panel). It is,
`however, increasingly apparent that the growth deregu-
`lation within a tumor can only be explained once we
`understand the contributions of the ancillary cells pres-
`ent in a tumor—the apparently normal bystanders such
`as fibroblasts and endothelial cells—which must play
`key roles in driving tumor cell proliferation (Figure 3,
`right panel). Within normal tissue, cells are largely in-
`structed to grow by their neighbors (paracrine signals)
`or via systemic (endocrine) signals. Cell-to-cell growth
`signaling is likely to operate in the vast majority of human
`tumors as well; virtually all are composed of several
`distinct cell types that appear to communicate via het-
`erotypic signaling.
`Heterotypic signaling between the diverse cell types
`within a tumor may ultimately prove to be as important
`
`NOVARTIS EXHIBIT 2029
`Breckenridge v. Novartis, IPR 2017-01592
`Page 3 of 14
`
`

`

`Cell
`60
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Figure 3. Tumors as Complex Tissues
`The field of cancer research has largely been
`guided by a reductionist focus on cancer cells
`and the genes within them (left panel)—a fo-
`cus that has produced an extraordinary body
`of knowledge. Looking forward in time, we
`believe that important new inroads will come
`from regarding tumors as complex tissues in
`which mutant cancer cells have conscripted
`and subverted normal cell types to serve as
`active collaborators in their neoplastic agenda
`(right panel). The interactions between the
`genetically altered malignant cells and these
`supporting coconspirators will prove critical
`to understanding cancer pathogenesis and to
`the development of novel, effective therapies.
`
`in explaining tumor cell proliferation as the cancer cell-
`autonomous mechanisms enumerated above. For ex-
`ample, we suspect that many of the growth signals driv-
`ing the proliferation of carcinoma cells originate from
`the stromal cell components of the tumor mass. While
`difficult to validate at present, such thinking recasts the
`logic of acquired GS autonomy: successful tumor cells
`are those that have acquired the ability to co-opt their
`normal neighbors by inducing them to release abundant
`fluxes of growth-stimulating signals (Skobe and Fu-
`senig, 1998). Indeed, in some tumors, these cooperating
`cells may eventually depart from normalcy, coevolving
`with their malignant neighbors in order to sustain the
`growth of the latter (Kinzler and Vogelstein, 1998; Olumi
`et al., 1999). Further, inflammatory cells attracted to sites
`of neoplasia may promote (rather than eliminate) cancer
`cells (Cordon-Cardo and Prives, 1999; Coussens et al.,
`1999; Hudson et al., 1999), another example of normal
`cells conscripted to enhance tumor growth potential,
`another means to acquire necessary capabilities.
`
`Acquired Capability: Insensitivity
`to Antigrowth Signals
`Within a normal tissue, multiple antiproliferative signals
`operate to maintain cellular quiescence and tissue ho-
`meostasis; these signals include both soluble growth
`inhibitors and immobilized inhibitors embedded in the
`extracellular matrix and on the surfaces of nearby cells.
`These growth-inhibitory signals, like their positively act-
`ing counterparts, are received by transmembrane cell
`surface receptors coupled to intracellular signaling cir-
`cuits.
`Antigrowth signals can block proliferation by two dis-
`tinct mechanisms. Cells may be forced out of the active
`proliferative cycle into the quiescent (G0) state from
`which they may reemerge on some future occasion
`when extracellular signals permit. Alternatively, cells
`may be induced to permanently relinquish their prolifera-
`tive potential by being induced to enter into postmitotic
`states, usually associated with acquisition of specific
`differentiation-associated traits.
`Incipient cancer cells must evade these antiprolifera-
`tive signals if they are to prosper. Much of the circuitry
`that enables normal cells to respond to antigrowth sig-
`nals is associated with the cell cycle clock, specifically
`
`the components governing the transit of the cell through
`the G1 phase of its growth cycle. Cells monitor their
`external environment during this period and, on the ba-
`sis of sensed signals, decide whether to proliferate, to
`be quiescent, or to enter into a postmitotic state. At the
`molecular level, many and perhaps all antiproliferative
`signals are funneled through the retinoblastoma protein
`(pRb) and its two relatives, p107 and p130. When in a
`hypophosphorylated state, pRb blocks proliferation by
`sequestering and altering the function of E2F transcrip-
`tion factors that control the expression of banks of genes
`essential for progression from G1 into S phase (Wein-
`berg, 1995).
`Disruption of the pRb pathway liberates E2Fs and
`thus allows cell proliferation, rendering cells insensitive
`to antigrowth factors that normally operate along this
`pathway to block advance through the G1 phase of the
`cell cycle. The effects of the soluble signaling molecule
`TGFb are the best documented, but we envision other
`antigrowth factors will be found to signal through this
`pathway as well. TGFb acts in a number of ways, most
`still elusive, to prevent the phosphorylation that inacti-
`vates pRb; in this fashion, TGFb blocks advance through
`G1. In some cell types, TGFb suppresses expression
`of the c-myc gene, which regulates the G1 cell cycle
`machinery in still unknown ways (Moses et al., 1990).
`More directly, TGFb causes synthesis of the p15INK4B and
`p21 proteins, which block the cyclin:CDK complexes
`responsible for pRb phosphorylation (Hannon and
`Beach, 1994; Datto et al., 1997).
`The pRb signaling circuit, as governed by TGFb and
`other extrinsic factors, can be disrupted in a variety of
`ways in different types of human tumors (Fynan and
`Reiss, 1993). Some lose TGFb responsiveness through
`downregulation of their TGFb receptors, while others
`display mutant, dysfunctional receptors (Fynan and
`Reiss, 1993; Markowitz et al., 1995). The cytoplasmic
`Smad4 protein, which transduces signals from ligand-
`activated TGFb receptors to downstream targets, may
`be eliminated through mutation of its encoding gene
`(Schutte et al., 1996). The locus encoding p15INK4B may be
`deleted (Chin et al., 1998). Alternatively, the immediate
`downstream target of its actions, CDK4, may become
`unresponsive to the inhibitory actions of p15INK4B be-
`cause of mutations that create amino acid substitutions
`
`NOVARTIS EXHIBIT 2029
`Breckenridge v. Novartis, IPR 2017-01592
`Page 4 of 14
`
`

`

`Review
`61
`
`in its INK4A/B-interacting domain; the resulting cyclin
`D:CDK4 complexes are then given a free hand to inacti-
`vate pRb by hyperphosphorylation (Zuo et al., 1996).
`Finally, functional pRb, the end target of this pathway,
`may be lost through mutation of its gene. Alternatively,
`in certain DNA virus-induced tumors, notably cervical
`carcinomas, pRb function is eliminated through seques-
`tration by viral oncoproteins, such as the E7 oncoprotein
`of human papillomavirus (Dyson et al., 1989). In addition,
`cancer cells can also turn off expression of integrins and
`other cell adhesion molecules that send antigrowth sig-
`nals, favoring instead those that convey progrowth sig-
`nals; these adherence-based antigrowth signals likely
`impinge on the pRb circuit as well. The bottom line is
`that the antigrowth circuit converging onto Rb and the
`cell division cycle is, one way or another, disrupted in
`a majority of human cancers, defining the concept and
`a purpose of tumor suppressor loss in cancer.
`Cell proliferation depends on more than an avoidance
`of cytostatic antigrowth signals. Our tissues also con-
`strain cell multiplication by instructing cells to enter irre-
`versibly into postmitotic, differentiated states, using di-
`verse mechanisms that are incompletely understood; it
`is apparent that tumor cells use various strategies to
`avoid this terminal differentiation. One strategy for
`avoiding differentiation directly involves the c-myc on-
`cogene, which encodes a transcription factor. During
`normal development, the growth-stimulating action of
`Myc, in association with another factor, Max, can be
`supplanted by alternative complexes of Max with a
`group of Mad transcription factors; the Mad–Max com-
`plexes elicit differentiation-inducing signals (Foley and
`Eisenman, 1999). However, overexpression of the c-Myc
`oncoprotein, as is seen in many tumors, can reverse this
`process, shifting the balance back to favor Myc–Max
`complexes, thereby impairing differentiation and pro-
`moting growth. During human colon carcinogenesis, in-
`activation of the APC/b-catenin pathway serves to block
`the egress of enterocytes in the colonic crypts into a
`differentiated, postmitotic state (Kinzler and Vogelstein,
`1996). Analogously, during the generation of avian eryth-
`roblastosis, the erbA oncogene acts to prevent irrevers-
`ible erythrocyte differentiation (Kahn et al., 1986).
`While the components and interconnections between
`the various antigrowth and differentiation-inducing sig-
`nals and the core cell cycle machinery are still being
`delineated, the existence of an antigrowth signaling cir-
`cuitry is clear (Figure 2), as is the necessity for its circum-
`vention by developing cancers.
`
`Acquired Capability: Evading Apoptosis
`The ability of tumor cell populations to expand in number
`is determined not only by the rate of cell proliferation
`but also by the rate of cell attrition. Programmed cell
`death—apoptosis—represents a major source of this
`attrition. The evidence is mounting, principally from
`studies in mouse models and cultured cells, as well as
`from descriptive analyses of biopsied stages in human
`carcinogenesis, that acquired resistance toward apo-
`ptosis is a hallmark of most and perhaps all types of
`cancer.
`Observations accumulated over the past decade indi-
`cate that the apoptotic program is present in latent form
`
`in virtually all cell types throughout the body. Once trig-
`gered by a variety of physiologic signals, this program
`unfolds in a precisely choreographed series of steps.
`Cellular membranes are disrupted, the cytoplasmic and
`nuclear skeletons are broken down, the cytosol is ex-
`truded, the chromosomes are degraded, and the nu-
`cleus is fragmented, all in a span of 30–120 min. In the
`end, the shriveled cell corpse is engulfed by nearby cells
`in a tissue and disappears, typically within 24 hr (Wyllie
`et al., 1980).
`The apoptotic machinery can be broadly divided into
`two classes of components—sensors and effectors. The
`sensors are responsible for monitoring the extracellular
`and intracellular environment for conditions of normality
`or abnormality that influence whether a cell should live
`or die. These signals regulate the second class of com-
`ponents, which function as effectors of apoptotic death.
`The sentinels include cell surface receptors that bind
`survival or death factors. Examples of these ligand/
`receptor pairs include survival signals conveyed by IGF-
`1/IGF-2 through their receptor, IGF-1R, and by IL-3 and
`its cognate receptor, IL-3R (Lotem and Sachs, 1996;
`Butt et al., 1999). Death signals are conveyed by the
`FAS ligand binding the FAS receptor and by TNFa bind-
`ing TNF-R1 (Ashkenazi and Dixit, 1999). Intracellular
`sensors monitor the cell’s well-being and activate the
`death pathway in response to detecting abnormalities,
`including DNA damage, signaling imbalance provoked
`by oncogene action, survival factor insufficiency, or hyp-
`oxia (Evan and Littlewood, 1998). Further, the life of most
`cells is in part maintained by cell–matrix and cell–cell
`adherence-based survival signals whose abrogation
`elicits apoptosis (Ishizaki et al., 1995; Giancotti and Ru-
`oslahti, 1999). Both soluble and immobilized apoptotic
`regulatory signals likely reflect the needs of tissues to
`maintain their constituent cells in appropriate architec-
`tural configurations.
`Many of the signals that elicit apoptosis converge
`on the mitochondria, which respond to proapoptotic
`signals by releasing cytochrome C, a potent catalyst of
`apoptosis (Green and Reed, 1998). Members of the Bcl-2
`family of proteins, whose members have either pro-
`apoptotic (Bax, Bak, Bid, Bim) or antiapoptotic (Bcl-2,
`Bcl-XL, Bcl-W) function, act in part by governing mito-
`chondrial death signaling through cytochrome C re-
`lease. The p53 tumor suppressor protein can elicit apo-
`ptosis by upregulating expression of proapoptotic Bax
`in response to sensing DNA damage; Bax in turn stimu-
`lates mitochondria to release cytochrome C.
`The ultimate effectors of apoptosis include an array
`of intracellular proteases termed caspases (Thornberry
`and Lazebnik, 1998). Two “gatekeeper” caspases, 28
`and 29, are activated by death receptors such as FAS
`or by the cytochrome C released from mitochondria,
`respectively. These proximal caspases trigger the acti-
`vation of a dozen or more effector caspases that execute
`the death program, through selective destruction of sub-
`cellular structures and organelles, and of the genome.
`The possibility that apoptosis serves as a barrier to
`cancer was first raised in 1972, when Kerr, Wyllie, and
`Currie described massive apoptosis in the cells populat-
`ing rapidly growing, hormone-dependent tumors follow-
`ing hormone withdrawal (Kerr et al., 1972). The discovery
`
`NOVARTIS EXHIBIT 2029
`Breckenridge v. Novartis, IPR 2017-01592
`Page 5 of 14
`
`

`

`Cell
`62
`
`of the bcl-2 oncogene by its upregulation via chromo-
`somal translocation in follicular lymphoma (reviewed in
`Korsmeyer, 1992) and its recognition as having anti-
`apoptotic activity (Vaux et al., 1988) opened up the in-
`vestigation of apoptosis in cancer at the molecular level.
`When coexpressed with a myc oncogene in transgenic
`mice, the bcl-2 gene was able to promote formation of
`B cell lymphomas by enhancing lymphocyte survival, not
`by further stimulating their myc-induced proliferation
`(Strasser et al., 1990); further, 50% of the infrequent
`lymphomas arising in bcl-2 single transgenic transgenic
`mice had somatic translocations activating c-myc, con-
`firming a selective pressure during lymphomagenesis
`to upregulate both Bcl-2 and c-Myc (McDonnell and
`Korsmeyer, 1991).
`Further insight into the myc-bcl-2 interaction emerged
`later from studying the effects of a myc oncogene on
`fibroblasts cultured in low serum. Widespread apoptosis
`was induced in myc-expressing cells lacking serum; the
`consequent apoptosis could be abrogated by exoge-
`nous survival factors (e.g., IGF-1), by forced overexpres-
`sion of Bcl-2 or the related Bcl-XL protein, or by disrup-
`tion of the FAS death signaling circuit (Hueber et al.,
`1997). Collectively, the data indicate that a cell’s apo-
`ptotic program can be triggered by an overexpressed
`oncogene. Indeed, elimination of cells bearing activated
`oncogenes by apoptosis may represent the primary
`means by which such mutant cells are continually culled
`from the body’s tissues.
`Other examples strengthen the consensus that apo-
`ptosis is a major barrier to cancer that must be circum-
`vented. Thus, in transgenic mice where the pRb tumor
`suppressor was functionally inactivated in the choroid
`plexus, slowly growing microscopic tumors arose, ex-
`hibiting high apoptotic rates; the additional inactivation
`of the p53 tumor suppressor protein, a component of
`the apoptotic signaling circuitry, led to rapidly growing
`tumors containing low numbers of apoptotic cells (Sy-
`monds et al., 1994). The role of extracellular survival
`factors is illustrated by disease progression in trans-
`genic mice prone to pancreatic islet tumors. If IGF-2
`gene expression, which is activated in this tumorigene-
`sis pathway, was abrogated using gene knockout mice,
`tumor growth and progression were impaired, as evi-
`denced by the appearance of comparatively small, be-
`nign tumors showing high rates of apoptosis (Christofori
`et al., 1994). In these cells, the absence of IGF-2 did not
`affect cell proliferation rates, clearly identifying it as an
`antiapoptotic survival factor. Collectively, these obser-
`vations argue that altering components of the apoptotic
`machinery can dramatically affect the dynamics of tu-
`mor progression, providing a rationale for the inactiva-
`tion of this machinery during tumor development.
`Resistance to apoptosis can be acquired by cancer
`cells through a variety of strategies. Surely, the most
`commonly occurring loss of a proapoptotic regulator
`through mutation involves the p53 tumor suppressor
`gene. The resulting functional inactivation of its product,
`the p53 protein, is seen in greater than 50% of human
`cancers and results in the removal of a key component
`of the DNA damage sensor that can induce the apoptotic
`effector cascade (Harris, 1996). Signals evoked by other
`
`abnormalities, including hypoxia and oncogene hyper-
`expression, are also funneled in part via p53 to the apo-
`ptotic machinery; these too are impaired at eliciting
`apoptosis when p53 function is lost (Levine, 1997). Addi-
`tionally, the PI3 kinase–AKT/PKB pathway, which trans-
`mits antiapoptotic survival signals, is likely involved in
`mitigating apoptosis in a substantial fraction of human
`tumors. This survival signaling circuit can be activated
`by extracellular factors such as IGF-1/2 or IL-3 (Evan
`and Littlewood, 1998), by intracellular signals emanating
`from Ras (Downward, 1998), or by loss of the pTEN
`tumor suppressor, a phospholipid phosphatase that
`normally attenuates the AKT survival signal (Cantley and
`Neel, 1999). Recently, a mechanism for abrogating the
`FAS death signal has been revealed in a high fraction
`of lung and colon carcinoma cell lines: a nonsignaling
`decoy receptor for FAS ligand is upregulated, titrating
`the death-inducing signal away from the FAS death re-
`ceptor (Pitti et al., 1998). We expect that virtually all
`cancer cells harbor alterations that enable evasion of
`apoptosis.
`It is now possible to lay out a provisional apoptotic
`signaling circuitry (Figure 2); while incomplete, it is evi-
`dent that most regulatory and effector components are
`present in redundant form. This redundancy holds im-
`portant implications for the development of novel types
`of antitumor therapy, since tumor cells that have lost
`proapoptotic components are likely to retain other simi-
`lar ones. We anticipate that new technologies will be
`able to display the apop

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