<<

[ RESEARCH (SUPPL.) 59, 1693s-1700s, April 1, 1999] BCL-2 Family and the Regulation of Programmed 1

Stanley J. Korsmeyer 2'3

Division of Molecular Oncology, Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, Missouri 63110

I hope he doesn't mind me saying this, but Dr. Stan Korsmeyer has achieved an enormous amount in a relatively short scientific career. You can take that whichever way you want to. Good, now I have landed myself in it. Stun Korsmeyer did his undergraduate degree in the University of Illinois and then did an M.D. at the same university and graduated in '76. Then, after doing an internship in UCSF Medical School, he came to the NIH where he was a clinical associate working in collaboration with Phil Leder and Tom Waldmann. There are some interesting parallels with Suzanne's career at this stage. Stan worked quite a lot on the structure of the immunoglobulin , their rearrangements in normal B cell development, and also studied aberrant rearrangements in leukemias. He set up his own lab in, I believe it was, '83 at NIH and then moved to Washington University School of Medicine at St. Louis where he became an associate professor in about 1986. The first pivotal contribution in relation to this prize, was his isolation, at the same time as others, of the bcl-2 at the breakpoint of a translocation between 14 and 18 in B cell follicular . Very shortly after Suzanne and colleagues published their seminal study showing that bcl-2 promotes and extends B cell survival, Stan showed essentially the same thing in transgenic mice where he overexpressed the bcl-2 oncogene in B cells and was able to show that B cells had extended survival and went on to develop . So, a wonderful convergence of findings in cell culture from Suzanne's lab and Stan's finding in mice. Stan has continued over the years to make some very important contributions to our understanding of bcl-2 function and also of bcl-2 relatives, as you have already heard from Suzanne. One of the first things he showed was that bcl-2 may have a function in mitochondria, and this notion received a great deal of support from findings published last year by several groups. Using a very elegant set of biochemical approaches, he was able to show that bcl-2 is a member of a larger family and that these family members can interact with each other. As you have heard, some of them act to promote and some to inhibit . So, we have a very interesting network of here. By knocking out different members of these families, Stan was also able to show that these genes really do play vital roles in cell survival in whole , obviously, a very important thing to do. He continues to work on the function of the proteins, and it is remarkable to think that, while doing this, he also was able to show the important roles of Hox-11 and Mll both in normal development and in leukemias. Since 1992 Stan has been the Chief of the Division of Molecular Oncology at Washington University School of Medicine, St Louis and since 1993 has been an investigator of the Howard Hughes Medical Institute. In recognition of his pioneering work, Stan has already received a number of awards including the Ciba Drew Award in 1996, the GHA Clowes Award, American Association for Cancer Research in 1997 and the Bristol-Myers Squibb Award for distinguished achievements in cancer research, 1997. He was elected to the National Academy of Sciences in 1995. So, we look forward very much to hearing Stan's talk on the bcl-2 family and the regulation of . Nicholas D. Hastie Medical Research Council Genetics Unit Western General Hospital Edinburgh, United

Abstract clustered within four conserved regions termed BCL-2 homology 1 through 4 (BHI-4). These novel domains control the ability of these proteins The BCL-2 gene was identified at the chromosomal breakpoint of t(14; to dimerize and function. An amphipathic a helix, BH3, is of particular 18)-bearing human follicular B cell lymphomas. BCL-2 proved to block importance for the proapoptotic family members. BID and BAD represent an programmed cell death rather than promote proliferation. Transgenic mice evolving set of proapoptotic molecules, which bear only at that overexpress Bcl-2 in the B cell lineage demonstrate extended cell survival BH3. They appear to reside more proximal in the pathway serving as death and progress to high-grade lymphomas. Thus, BCL-2 initiated a new cate- ligands. BAD connects upstream paths with the BCL-2 gory of , regulators of cell death. Bcl-2-deficient mice demonstrate family, modulating this checkpoint for apoptosis. In the presence of survival fnlminant apoptosis of lymphocytes, profound renal cell death and loss of factor interleukin-3, cells phosphorylate BAD on two residues. This melanocytes. BCL-2 duels with its counteracting twin, a partner inactivated BAD is held by the 14-3-3 protein, freeing BCL-XL and BCL-2 to known as BAX. When BAX is in excess, cells execute a death command; but, promote survival. Activation of BAX results in the initiation of apoptosis. when BCL-2 dominates, the program is inhibited and cells survive. Bax- Downstream events in this program include mitochondrial dysfunction, as deficient mice display cellular , confirming its role as a pro- well as activation. The pro- and antiapoptotic BCL-2 family mem- apoptotic molecule. An expanded family of BCL-2-related proteins shares bers represent central regulators in an evolutionarily conserved pathway of cell death. Aberrations in the BCL-2 family result in disordered homeostasis, Received 9/18/98; accepted 1/27/99. a pathogenic event in , including cancer. 1 Presented at the "General Motors Cancer Research Foundation Twentieth Annual Scientific Conference: Developmental Biology and Cancer," June 9-10, 1998, Bethesda, MD. Introduction 2 To whom requests for reprints should be addressed, at Dana-Farber Cancer Institute, Smith 758, Harvard Medical School, One Jimmy Fund Way, Boston, MA 02115. 3 Co-recipient of the Mott Prize along with Suzanne Cory, whose article can be found Programmed cell death plays an indispensable role in the develop- on pages 1685s-1692s of this Supplement. ment and maintenance of homeostasis within all multicellulax organ- 1693s

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 1999 American Association for Cancer Research. BCL-2 FAMILY REGULATES APOPTOSIS isms. Genetic and molecular analysis from to has chromosomal breakpoint. Aberrant immunoglobulin heavy chain indicated that the pathway of cellular suicide is highly conserved. rearrangements in t(14;18) lymphomas proved to be the chromo- Although the capacity to carry out apoptosis seems to be inherent in somal breakpoint and delivered a candidate oncogene, BCL-2, at all cells, the susceptibility to apoptosis varies markedly and is influ- 18q21 (1-3). BCL-2 is normally located on segment enced by external and cell-autonomous events. Considerable progress 18q21.3 in a telomere to centromere orientation. A molecular has been made in identifying the molecules that regulate the apoptotic consequence of the translocation is the movement of the BCL-2 pathway at each level. Of note, both positive and negative regulators, gene to the der (14) chromosome (Fig. 1). This places BCL-2 in the often encoded within the same family of proteins, characterize the same transcriptional orientation as the immunoglobulin heavy extracellular, cell surface, and intracellular steps. chain locus, giving rise to chimeric RNAs. However, translocation The t(14;18) Breakpoint of Human Pro- does not interrupt the protein-encoding region so that normal and vides the BCL-2 Gene. The t(14;18):(q32;q21) constitutes the most translocated alleles produce the same-sized, 25-kDa protein, t(14; common chromosomal translocation in human lymphoid malignan- 18)-bearing B cells have inappropriately elevated levels of the cies, being present in 85% of follicular and 20% of diffuse B-cell BCL-2-immunoglobulin fusion RNA and BCL-2 protein (4, 5). lymphomas. As a , follicular lymphoma provided many BCL-2-immunoglobulin Transgenic Mice Progress from Follic- clues concerning the ultimate function of the BCL-2 molecule. ular Hyperplasia to High-Grade Lymphoma. A stringent test of a Follicular lymphoma often presents as a low-grade malignancy gene's oncogenic capacity is to place it into the germline of mice composed of small resting igM/IgD B cells. Over time, conversion to observe its effects during the development of an entire organism. to an aggressive high-grade lymphoma with a diffuse large-cell Transgenic mice bearing a BCL-2-immunoglobulin minigene initially architecture frequently occurs in these patients. The location of the displayed a polyclonal follicular lymphoproliferation that selectively ex- immunoglobulin heavy chain locus at 14q32 and the B-cell phe- panded a small resting IgM/IgD B-cell population (Refs. 6, 7; Fig. 1). notype of this lymphoma provided the rationale for cloning the analysis confirmed that ~97% of the expanded B cells reside

Transgenic Construct Chromosome Chromosome I 18 I 14 J I II III P, P, J, EH Sj, Sv Cv, pA pA Translocated i ! P~2~: ~ ~,r ~ ~:~,~ .... ~;~im~:'ltI~tul~llml~;mwJ~ Bcl-2-Tg Genomic cDNA' ' Genomic 1 H23vs ,.~, Normal Sequence lOS_ Splenocytes - 0 - 0 Bcl-2 Transgenic - 9 - 9 Control Littermate

10 "~ mIm > =m > L-

r ~3 10 4-

! I V 10; 2 4 6 8 Days in Culture Fig. 1. Bcl-2-immunoglobulintransgenic mice. The Bcl-2-immunoglobulinminigene recapitulates the t(14;18) chrolnosomaltranslocation of follicular B-cell lymphoma.Gain of function Bcl-2 leads to B-cell expansion and an enlargedwhite pulp in the transgenic (left) versus control (right) spleen. B cells accumulate due to extendedcell survival (graph). 1694s

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 1999 American Association for Cancer Research. BCL-2 FAMILY REGULATES APOPTOSIS

Table 10ncogene categories membrane blebbing, volume contraction, nuclear condensation, and Category 1: Growth and proliferation genes endonucleolytic of DNA, (15) termed apoptosis by Wyllie Transcription factors or signal transduction (17). By preventing the apoptotic demise of activated lymphocytes, , Ras, Abl Gain of function BCL-2 enables the acquisition of additional genetic aberrations and Category II: tumor suppressor genes the emergence of monoclonal . By repressing normal ap- Inhibit growth and proliferation optotic pathways, BCL-2 allows cells that sustain DNA damage to Rb, Loss of function avoid a suicide response. Category 11I: programmed cell death genes Oncogenic events had concentrated on mechanisms of increased Antiapoptotic genes growth and proliferation. However, increased cell number, which Bcl-2 Gain of function violates normal homeostasis, could arise from either increased Proapoptotic genes proliferation or decreased death. BCL-2 provided the first example p53, Bax Loss of function of oncogenesis mediated by decreased cell death. Table 1 depicts two classes of oncogenes that regulate growth and proliferation (18). The first oncogenes discovered, category I, promote and proliferation. Most of these can be classified as tran- scription factors or molecules involved in signal transduction. In general, these genes contribute to cancer after an alteration result- ing in a gain of function. They usually display an autosomal dominant mechanism, in which a single altered allele is sufficient to confer the effect. Category II represents the classic tumor suppressor genes that, in their wild-type form, inhibit growth and proliferation (19). A unique aspect of two experimentally proven ipO members, Rb and p53, is their frequent contribution to unchecked growth by loss of function. The effects of deregulated BCL-2 argue that it does not qualify as either a category I or II oncogene. Instead, BCL-2 seems to be the cardinal member of a new category Fig. 2. Susceptibility to PCD. The relative ratio of BCL2 to BAX determines the of oncogenes: regulators of PCD (Table 1). susceptibility to PCD. BAX, a Proapoptotic Member and Expansion of the Family. The first proapoptotic homologue, BAX, was identified by coim- in Go/Gv These recirculating B cells accumulate because of an extended survival rather than increased proliferation. Despite a 4-fold increment in resting B cells, BCL-2-immunoglobulin mice are initially quite healthy. The BCL-2 Family However, over time, these transgenics progress from indolent follicular hyperplasia to diffuse large-cell immunoblastic lymphoma. A long la- Anti-apoptotic tency period and progression from polyclonal hyperplasia to monoclonal high-grade malignancy is an indictment of secondary genetic abnormal- Mammalian BH4 BH3 BH1 BH2 TM BCL-2 ities. Approximately half of the high-grade tumors possess a c-myc translocation involving an immunoglobulin heavy-chain locus (8). These BCL-X L tumor cells have complemented an inherent survival advantage (bcl-2) BCL-W with a gene that promotes proliferation (myc). When bcl-2 transgenic MCL-1 mice were mated with myc transgenic mice, rapidly emerging undiffer- A1 entiated hematopoietic leukemia occurred, providing further testimony for the potent synergy of this particular oncogene combination (9). In NR-13 [Rxx-q--t~N-s addition to promoting cell cycle progression, myc has been shown to C. elegans CED-9 promote cell death (10). Thus, the overexpression of myc specifically benefits from the ability of bcl-2 to block cell death. The BCL-2-imInu- Pro-apoptotic noglobulin mice document the prospective importance of the t(14;18) in Mammalian setting the stage for lymphomagenesis and tumor progression. T-cell BAX lymphomas were also found in mice where bcl-2 was overexpressed by BAK the lck (11). In this case, thymocytes and peripheral T cells also BOK exhibited enhanced cell survival (12, 13). Thus, prolonged cell survival can be a primary oncogenic event in both B and T cells. BCL-X S -~ ~ E} BCL-2 Blocks Apoptosis Initiating a New Category of Onco- BH3-only genes: Regulators of PCD. BCL-2 blocks cell death after a wide BID variety of stimuli. BCL-2 conferred a death-sparing effect to hema- BAD F'77"~~ topoietic cell lines after withdrawal, including the BIK --~ ...... E]- IL-3a-dependent early hematopoietic cell lines FDCP1, FL5.12, and BLK ~1~7"/~ {~- 32D (14-16). A detailed examination of the interference of BCL-2 with cell death revealed it does not alter the dose response to limiting HRK concentrations of IL-3 (16). Instead, BCL-2 blocked the plasma BIN F~ Fig. 3. Summary of antiapoptotic and proapoptotic BCL-2 family members. BCL-2 4 The abbreviations used are: IL-3, interleukin-3; PCD, programmed cell death. homology regions are indicated with similar shaded boxes (BH1-BH4). 1695s

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 1999 American Association for Cancer Research. BCL-2 FAMILY REGULATES APOPTOSIS

Table 2 Bcl-2 -/- BCL-2 proteins and proved essential for proapoptotic function (31, B + T cell apoptosis: immunodeficiency 32, 33). The multidimensional nuclear magnetic resonance and X-ray Renal mesangial apoptosis: polycystic kidney crystallographic structure of a BCL-X L monomer indicated that Melanocyte death: hypopigmentation $ Primordial follicles: 9 infertility BH1-4 domains corresponded to o~-helices 1-7. The a-helices of BH1-3 are closely juxtaposed to form a hydrophobic pocket (34). Nuclear magnetic resonance analysis of a BAK peptide/BCL-XL interaction and a detailed mutational analysis of the BH3 amphipathic Table 3 Bax -/- c~2-helix of BAX indicates that BH3 forms critical interactions with Mild lymphoidhyperplasia BCL-2/BCL-XL (35). Moreover, the hydrophobic face of this am- Spermatogonia hyperptasia: ~ sterility Neuronal hyperplasia: trophic factor (NGFa) independence;protection from stroke phipathic helix is critical for its death-effector function (Ref. (33); Fig. 4). 1' Primordialfollicles: oocyte chemoresistance;no menopause Loss of Function Models Reveal Critical Roles for the BCL-2 Accelerated onset of tumors in the presenceof an oncogene:Bax as a tumor suppressor Family in Homeostasis. The normal developmental roles of BCL-2 "NGF, . family members have been addressed by gene disruption. These loss of function models have proven most instructive in revealing the cell type specificity and singular roles for the BCL-2 members. munoprecipitation with the BCL-2 protein (20). BAX is a 21-kDa Newborn Bcl-2-/- knockout mice are viable, but the majority die protein that shares homology with BCL-2 clustered in conserved at a few weeks of age (Ref. (36); Table 2). They develop polycystic regions entitled BCL-2 homology domains, BH1-3. BAX het- kidney disease with marked dilation of proximal and distal tubules erodimerizes with BCL-2 and homodimerizes with itself. When and collecting ducts, resulting in renal failure (36-38). In the BAX was overexpressed in cells, apoptotic death in response to a normal fetal kidney, BCL-2 maintains cell survival during induc- death signal was enhanced, earning its designation as a death tive interactions between epithelium and mesenchyme. BCL-2-/- agonist. When BCL-2 was overexpressed, it heterodimerized with kidneys contain many fewer nephrons and greatly increased apop- BAX and death was repressed. Thus, the ratio of BCL-2 to BAX tosis within metanephric blastemas of metanephroi at embryonic serves as a rheostat to determine the susceptibility to apoptosis day 12 (39). Bcl-2-/- mice turn gray at 5-6 weeks of age, at the (Fig. 2). time of the second hair follicle cycle. The hypopigmentation of The family has further expanded to include the death antagonists Bcl-2-/- mice reflects decreased melanocyte survival. The BCL-2, BCL-X L (21), MCL-1 (22), and A1 (23), as well as the lymphoid organs, thymus, and spleen are initially normal in Bcl- proapoptotic molecules BAX, BCL-X s (21), and BAK (24-26). Con- 2-/- mice. Thymocyte development is normal, and B and T cells served domains BH1, BH2, and BH3 participate in the formation of undergo selection successfully. However, at 4-8 weeks of age, the various dimer pairs, as well as the regulation of cell death (Refs. (27 lymphoid organs undergo massive cell death and involution, show- and 28); Fig. 3). ing a failure to maintain homeostasis in both the B- and T-cell Mutational analysis of BCL-2 and BCL-X L identified key residues populations in the absence of BCL-2. within BH1 and BH2 domains required for both heterodimerization Bax-deficient mice represent the first knockout of a death- with BAX and repression of cell death (29). However, other BCL-XL promoting family member (Ref. (40); Table 3). Bax-/- mice mutants lost heterodimerization with BAX, but still retained some appear healthy, indicating that BAX is not essential for develop- death-repressor activity, suggesting that these two functions were ment of a viable organism. However, male Bax-/- mice are separable (30). An additional domain, BH3, had also been noted in infertile, and Bax-/- seminiferous tubules are markedly disorga-

Fig. 4. Three-dimensional model of the BH3 domain of BAX. Model of the molecular surface of BAX BH3. The surface is colored deep blue (23kBT) in the most positive regions and deep red (-21kBT) in the most negative, with linear inter- polation for values in between. Left, view of the hydrophobic surface of the amphipathic BH3 helix of BAX. Right, view of the polar surface of the amphipathic BH3 a-2 helix of BAX. Residues forming polar and hydrophobic surfaces are indi- cated.

1696s

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 1999 American Association for Cancer Research. BCL-2 FAMILY REGULATES APOPTOSIS

Normal Tissue autoimmune disease, whereas accelerated cell death can manifest as neurodegenerative disease, immunodeficiency syndromes, or infertil- (In) 1 ity (Fig. 5). Proapoptotic BAX as a Tumor Suppressor. Prolonged cell sur- J Homeostasis vival with resistance to apoptosis can be a primary oncogenic (Out) 1 event. This predicted that proapoptotic genes in the death pathway might contribute to oncogenesis through loss of function (Table 1). Indeed, a principal contribution from the loss of p53 Diseases of Disordered Cell Death function is the elimination of a death pathway (43, 44). In select settings, stimuli that induce p53 result in the expression of Bax Proliferation 1 (45). Bax-deficient mice indicate that approximately half of certain p53-dependent cell require BAX. Elimination of BAX Neurodegenerative blocks approximately half of chemotherapy-induced cell death Immunodeficiency Cancer (46). A transgenic model, TGT121 expresses a truncated T antigen Infertility Autoimmunity that inhibits Rb but leaves p53 intact, resulting in cell proliferation with counterbalancing cell death. The elimination of Bax in this 1 Death l model markedly decreased the apoptosis. Moreover, the TGT121 transgenic mice displayed a markedly accelerated progression to Fig. 5. Schematicrepresentation of normal tissue homeostasiswith balancedinput and output reactions. Primary aberrations in the cell death pathway that result in diminished malignancy on a Bax-deficient background (Ref. (47); Table 3). death can manifest as cancer or autoimmunity, whereas increased death can result in These experimental models argue that Bax can also be considered neurodegenerativedisease, immunodeficiency,or infertility. a tumor suppressor. Recently, mutations in BAX were noted in ~20% of human hema- nized with multinucleated giant cells, clusters of apoptotic germ topoietic malignancies studied (48). Approximately half were frame- cells, and lack of mature sperm. This follows a failure of normal shifts confined to a single mononucleotide (G) s tract (nt 114-121) postnatal death of spermatogonia, leading to an expansion of the that results in a stop codon and the loss of BAX protein. The others premeiotic 2N cell population. Thus, the failure of BAX-dependent represent point mutations within conserved BH1 and BH3 domains developmental deaths leads to organ failure and later paradoxic that alter dimerization and eliminate proapoptotic activity. Rampino et cell death during attempted meiosis (40). Bax-/- display al. (49) described the presence of frameshift mutations in the identical an accumulation of atrophic granulosa cells and excess primordial (G)8 tract of BAX in about 50% of human colon adenocarcinomas with follicles that failed to undergo apoptosis (41). Bax-/--sympa- the microsatellite mutator . Mutations of the proapoptotic thetic neurons do not reqmre nerve growth factor for survival, and BAX molecule within human malignancies support the role of BAX motor neurons survive disconnection from their targets after axo- as a tumor suppressor. tomy (42). The Bax-deficient mice separated the survival from BH3 Only Molecules as Death Ligands That Connect the anabolic action of . Despite the presence of BCL-2 Checkpoint to Signal Transduction. To extend the cell multiple BCL-2 family members within these neurons, their death death pathway, BCL-2 and BAX were used as bait in interactive by trophic factor deprivation was singularly dependent on BAX. cloning strategies. This provided a series of interacting molecules that Transgenic and knockout models of the Bcl-2 family indicate that bear sequence homology to BCL-2 principally within only the BH3 primary aberrations in the cell death pathway result in disordered domain. These now include BIK (50, 51), BID (52), BAD (53, 54), homeostasis and disease. Decreased cell death can result in cancer or BIM (55), HRK (56), and BLK (Ref. (57); Fig. 3). Mutagenesis of

IL-3

~c N

Fig. 6. Dual impact of lL-3-induced phospho- rylation of BAD on two serine sites by distinct pathways. The phosphorylated BAD is se- N Inactive questered by 14-3-3 in the cytosol as the inactive form, which releases BCL-X L to promote survival. Only nonphosphorylated BAD can heterodimerize with membrane-bound BCL-X L, which seems to be B the active form that inhibits BCL-X L. Act,ve /\ A $112 $136 cytosol BAD Kinase(s) membrane ( Phosphatase(s) !!~ i~i~84184 !~ ~ ~i~iiil~ ~ili~ membrane

Death Survival 1697s

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 1999 American Association for Cancer Research. BCL-2 FAMILY REGULATES APOPTOSIS

6 Two of these molecules, BID and BAD, lack a COOH-terminal I II hydrophobic segment (Fig. 3). These molecules can translocate Ii II Death Signal between cytosol and classic membrane based family partners such Ii Ii as BCL-2 or BCL-X L. Moreover, mutants of BID and BAD that il II fail to bind their partner proteins were functionally inert. Thus, [] BID and BAD can be envisioned as "death ligands" that bind to A.o;. downstream membrane-based BCL-2 full members, "receptors." These characteristics make BID and BAD candidates for an up- stream link between the BCL-2 checkpoint and proximal signal transduction. Extraeellular Survival Factors Reset the BCL-2 Checkpoint by Phosphorylation of BAD. Support for the above thesis comes from the recognition that BAD is rapidly phosphorylated on two serine residues in response to a survival factor, IL-3 (58). These represent canonical 14-3-3 binding sites. Phosphorylated BAD seems to be the inactive form incapable of binding BCL-X L and sequestered in the cytosol bound to 14-3-3 (Fig. 6). This would release BCL-X L Fig. 7. Schematic model of BAX activation cell death. A death signal activates BAX, resulting in the translocation of a BAX monomer in the cytosol to homodimerized,integral at membrane sites to promote cell survival. Consistent with this mitochondrial membrane BAX. Downstream effects include the activation of model, substitution of the serine phosphorylation sites eliminated and a program of mitochondrial dysfunction. binding to 14-3-3 and further enhanced the death-promoting activity of BAD. Despite the similarity of the phosphorylation motifs sur- BID and BAD revealed that their amphipathic BH3 c~ helical domain rounding Serll2 and Ser136 of BAD, distinct BAD seem was critical for both binding to the pocket of antagonists BCL-2 and responsible for the differential phosphorylation of each site after BCL-X L and their death agonist activity (52, 54). These molecules engagement of a single (58, 59). This illustrates the com- provide further support for BH3 as the critical and minimal death plexity of the posttranslational modifications that will regulate this domain. pathway.

Genotoxic i O.,ok, no l .,.an. I Oe~ Receptor J

_ Cell Death Si_gn_a_l ~

Pro-apoptotic Death Antic - - O y Checkpoint ~(ced-9) Fig. 8. Schematic model of mammalian cell death pathway. A major checkpoint in the common portion of the pathway is the ratio Caspase (ced-3) of proapoptotic (BAX) to antiapoptotic (BCL-2) members. Cyt C > Apaf,1 (ced-4) < PTP A ~ m ROS q, Death Substrates (PARP) /

1698s

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 1999 American Association for Cancer Research. BCL-2 FAMILY REGULATES APOPTOSIS

BAX Activation Induces Mitochondrial Dysfunction and Apo- recent identification of egl-1, a BH3-only gene in C. elegans (70), ptosis. Although the ratio of BCL-2:BAX determines the response indicates that the evolving set of BH3-only proteins are a conserved to an apoptotic signal, a question remained as to whether BAX component of the central apoptotic pathway. Cloning of C. elegans ced-3 itself could initiate death and, if so, how. Transient transfection revealed homology to the mammalian enzyme ICE (71). There are more and inducible expression systems for BAX or BAK proved suffi- than 10 ICE-like , now termed caspases (cysteine proteases that cient to induce apoptosis without an additional stimulus (60, 61). cleave after aspartic acid; Ref. (72). Caspases use a cysteinyl residue as BAX-induced death activated caspases which cleaved endogenous the active nucleophile and recognize a cleavage site with an aspartic acid substrates in the nucleus (PARP) and cytosol (D4-GDI; Fig. 7). residue at the P1 position of multiple death substrates. A single ced-4 However, although caspase inhibitors successfully blocked prote- homologue, Apaf-l, which interacts with cytochrome C when released ase activity and could prevent a FAS-induced death, it did not from mitochondria to activate caspases, has also been identified (73). block BAX-induced death. The localization of BCL-2 (15), and Thus, the basic tenets of the cell death pathway seem to be markedly subsequently other family members, to the mitochondria prompted conserved, likely common to all multicellular organisms. Moreover, an assessment of mitochondrial function. Although the cleavage of DNA viruses choose to interfere at these same critical steps to prevent the substrates and the final degradation of DNA was prevented, mito- apoptotic death of their cells (Fig. 8). Thus, BCL-2, which was chondrial dysfunction still occurred. BAX-induced alterations in provided by follicular lymphoma, like so many genes that are responsible mitochondrial membrane potential, production of reactive oxygen for a human disease, revealed a major genetic and biochemical pathway; species, cytoplasmic vacuolation, and plasma membrane perme- in this case, one that regulates cell survival. The BCL-2 family genes in ability apparently occurred despite the inhibition of measurable this pathway have proven critical for the successful crafting of multicel- caspase activity (60). lular lineages during development, as well as the maintenance of home- BAX has been shown to have both cytosolic and membrane- ostasis in the adult. associated locations. Death signals such as IL-3 withdrawal, stau- rosporine, dexamethasone, and ?/-irradiation induce the transloca- References tion of BAX protein from cytosol to mitochondrial membrane (62, 63). Although BAX forms both homo- and heterodimers, questions 1. Tsujimoto, Y., Gorham, J., Cossman, J., Jaffe, E., and Croce, C. M. The t(14;18) chromosome translocations involved in B-cell neoplasms result from mistakes in VDJ remained concerning its native conformation in vivo and which joining. Science (Washington DC), 229: 1390-1393, 1985. moiety was functionally active. Physiological death stimuli result 2. B',tkhshi, A., Jensen, J. P., Goldman, P., Wright, J. J., McBride, O. W., Epstein, A. L., in the translocation of monomeric BAX from the cytosol to the and Korsmeyer, S. J. Cloning the chromosomal breakpoint of t(14;18) human lym- phomas: clustering around Jn on chromosome 14 and near a transcriptional unit on mitochondria, where it can be cross-linked as a BAX homodimer 18. Cell, 41: 889-906, 1985. (62-64). In contrast, cells protected by BCL-2 demonstrate a block 3. Cleary, M. L., and Sklar, J. Nucleotide sequence of a t(14;18) chromosomal break- point in follicular lymphoma and demonstration of a breakpoint cluster region near a in this process in that BAX does not redistribute or homodimerize transcriptionally active locus on chromosome 18. Proc. Natl. Acad. Sci. USA, 82: in response to a death signal, Moreover, enforced dimerization of 7439-7443, 1985. FKBP-BAX by the bivalent FK1012 resulted in its translo- 4. Cleary, M. L., Smith, S. D., mad Sklar, J. Cloning and structural analysis of cDNAs for bcl-2 and a bcl-2/immunoglobulin transcript resulting from the t(14;18) cation to mitochondria and induced apoptosis (63). Enforced translocation. Cell, 47: 19-28, 1986. dimerization of BAX overrode the protection of BCL-X L and IL-3 5. Seto, M., Jaeger, U., Hockett, R. D., Graninger, W., Bennett, S., Goldman, P., and to kill cells. Korsmeyer, S. J. Alternativepromoters and exons, somatic and transcriptional deregulation of the Bcl-2-1g fusion gene in lympholna. EMBO J., 7: 123-131, 1988. The ability of BAX dimers to kill cells is consistent with genetic 6. McDonnell, T. J., Dean, N., Platt, F. M., Nuncz, G., Jaeger, U., McKearn, J. P., and evidence that BAX can function independently of BCL-2 (65). Korsmeyer, S. J. Bcl-2-immunoglobulin transgenic mice demonstrate extended B cell survival and follicular lymphoproliferation. Cell, 57." 79-88, 1989. Moreover, a point mutant of BAX that fails to form homodimers or 7. McDonnell, T. J., Nunez, G., Platt, F. M., Hockenbery, D., London, L., McKearn, heterodimers in classic binding assays still inserted into mitochon- J. P., and Korsmeyer, S. J. Deregulated Bcl-2-immunoglobulin transgene expands a drial membranes as a cross-linkable pair and induced apoptosis resting but responsive immunoglobulin M and D-expression B-cell population. Mol. Cell. Biol., 10: 1901-1907, 1990. (33). How might integral membrane BAX kill? One possibility 8. McDonnell, T. J., and Korsmeyer, S. J. Progression from lymphoid hyperplasia to may relate to the capacity of BAX to form distinct ion-conductive high-grade malignant lymphoma in mice transgenic for the t(14;18). (Lond.), pores (66, 67). This may prove to regulate an electrochemical 349: 254-257, 1991. 9. Strasser, A., Harris, A. W., Bath, M. L., and Cory, S. Novel primitive lymphoid gradient, alter mitochondrial volume homeostasis, or redistribute tumors induced in transgenic mice by cooperation between myc and bcl-2. Nature critical substrates or products residing in the intermembrane space. (Lond.), 348: 331-333, 1990. 10. Evan, G. l., Wyllie, A. H., Gilbert, C. S., Littlewood, T. D., Land, H., Brooks, M., Overall, these data favor a model in which a death signal results in Waters, C. M., Penn, L. Z., and Hancock, D. C. Induction of apoptosis in the activation of BAX. This conformational change in BAX man- by c-myc protein. Cell, 69: 119-128, 1992. ifests in its translocation, mitochondrial membrane insertion, ho- 11. Linette, G. P., Hess, J. L., Sentman, C. L., and Korsmeyer, S. J. Peripheral T-cell lymphoma in lckPr-bcl-2 transgenic mice. Blood, 86: 1255-1260, 1995. modimerization, and a program of mitochondrial dysfunction that 12. Sentmml, C. L., Shutter, J. R., Hockenbery, D., Kanagawa, O., and Korsmeyer, S. J. results in cell death (Fig. 7). bcl-2 inhibits multiple forms of apoptosis but not negative selection in thymocytes. BCL-2 and BAX as an Active Checkpoint Upstream of Cell, 67: 879-888, 1991. 13. Strasser, A., Harris, A. W., and Cory, S. Bcl-2 transgene inhibits T cell death and Caspases and Mitochondrial Dysfunction. Just as BAX can acti- perturbs thymic self-censorship. Cell, 67: 889-899, 1991. vate caspases and induce mitochondrial dysfunction, the expression of 14. Vaux, D. L., Cory, S., and Adams, J. M. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature (Lond.), 335: BCL-2 can block both events (Fig. 8). Thus, the BCL-2 family seems 440-442, 1988. to constitute a critical go/no go checkpoint. This decisional step 15. Hockenbery, D., Nunez, G., Milliman, C., Schreiber, R. D., and Korsmeyer, S. J. resides downstream of a myriad of cell death signals, but proximal to Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature (Lond.), 348: 334-336, 1990. irreversible cellular damage. Important contributions to the under- 16. Nunez, G., London, L., Hockenbery, D., Alexander, M., McKearn, J., and Korsmeyer, standing of PCD have come from the genetic studies of the S. J. Deregulated Bcl-2 selectively prolongs survival of ga'owth factor-deprived hematopoietic cell lines. J. lmmunol., 144: 3602-3610, 1990. Caenorhabditis elegans. In the development of that worm, 131 of the 17. Wyllie, A. H. Apoptosis. Cell death in tissue regulation. J. Pathol., 153:313-316, 1987. 1090 somatic cells undergo PCD using a common genetic pathway. 18. Bishop, J. M. The molecular genetics of cancer. Science (Washington DC), 235: Two genes, ced-3 and ced-4, are required for cell death to occur. A 305-31 l, 1987. 19. Knudson, A. G., Jr. Heredity cancer, oncogenes antioncogenes. Cancer Res., 45: third gene, ced-9, represses the death pathway and protects cells (68). 1437-1443, 1985. Ced-9 is the structural and functional homologue of BCL-2 (69). The 2o. Oltvai, Z. N., Milliman, C. L., and Korsmeyer, S. J. Bcl-2 heterodimerizes in vivo 1699s

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 1999 American Association for Cancer Research. BCL-2 FAMILY REGULATES APOPTOSIS

with a conserved homologue, Bax, that accelerates programmed cell death. Cell, 74: tumorigenesis and stimulates apoptosis in vivo. Nature (Lond.), 385: 637-640, 609-619, 1993. 1997. 21. Boise, L. H., Gonzalez-Garcia, M., Postema, C. E., Ding, L., Lindsten, T., Turka, 48. Meijerink, J. P. P., Mensink, E. J. B. M., Wang, K., Sedlak, T. W., Sloetjes, A. W., L. A., Mao, X., Nunez, G., and Thompson, C. B. bcl-x, a bcl-2-related gene that de Witte, T., Waksman, G., and Korsmeyer, S. J. Hematopoietic malignancies functions as a dominant regulator of apoptotic cell death. Cell, 74: 597-608, 1993. demonstrate Loss-of-function mutations of Bax. Blood, 9l: 2991-2997, 1998. 22. Kozopas, K. M., Yang, T., Buchan, H. L., Zhou, P., and Craig, R. W. MCL1, a gene 49. Rampino, N., Yamamoto, H., Ionov, Y., Li, Y, Sawai, H. Reed, J. C., and Perucho, expressed in programmed myeloid cell differentiation, has sequence similarity to M. Somatic frameshift mutations in the bax gene in colon of the microsatellite BCL2. Proc. Natl. Acad. Sci. USA, 90: 3516-3520, 1993. mutator phenotype. Science (Washington DC), 275: 967-969, 1997. 23. Lin, E. Y., Orlofsky, A., Berger, M. S., and Prystowsky, M. B. Characterization of 50. Boyd, J. M., Gallo, G. J., Elangovan, B., Houghton, A. B., Malstrom, S., Avery, B. J., A1, a novel hemopoietic-specific early-response gene with sequence similarity to Ebb, R. G., Subramanian, T., Chittenden, T., Lutz, R. J., et al. Bik, a novel bcl-2. J. lmmunol., 15I: 1979-1988, 1993. death-inducing protein shares a distinct sequence motif with Bcl-2 family proteins and 24. Farrow, S. N., White, J. H. M., Martinou, I., Raven, T., Pun, K-T., Grinham, C. J., interacts with viral and cellular survival-promoting proteins. Oncogene, 11: 1921- Martinou, J-C., and Brown, R. Cloning of a bcl-2 homologue by interaction with 1928, 1995. adenovirus E1B 19K. Nature (Lond.), 374: 731-733, 1995. 51. Han, L, Sabbatini, P., and White, E. Induction of apoptosis by human Nbk/Bik, a 25. Chittenden, T., Harrington, E. A., O'Connor, R., Flemington, C., Lutz, R. J., Evan, BH3-containing protein that interacts with E1B 19K. Mol. Cell. Biol., 16: 5857-5864, G. I., and Guild, B. C. Induction of apoptosis by the Bcl-2 homologue Bak. Nature 1996. (Lond.), 374: 733-736, 1995. 52. Wang, K., Yin, X-M., Chao, D. T., Milliman, C. L., and Korsmeyer, S. J. BID: a 26. Kiefer, M. C., Brauer, M. J., Powers, V. c., Wu, J. J., Umansky, S. R., Tomei, L. D., novel BH3 domain-only death agonist. Genes Dev., 10: 2859-2869, 1996. and Barr, P. J. Modulation of apoptosis by the widely distributed Bcl-2 homologue 53. Yang, E., Zha, J., Jockel, J., Boise, L. H., Thompson, C. B., and Korsmeyer, S. J. Bad, Bak. Nature (Lond.), 374: 736-739, 1995. a heterodimeric partner for Bcl-XL and Bcl-2, displaces Bax and promotes cell death. 27. Golemis, E., Fong, L., Wang, H. G., and Reed, J. C. Interactions among members of Cell, 80: 285-291, 1995. the BCL-2 protein family analyzed with a yeast two-hybrid system. Proc. Natl. Acad. 54. Zha, J. P., Jockel, J., and Korsmeyer, S. J. BH3 domain of BAD is required for death Sci. USA, 91: 9238-9242, 1994. promotion and heterodimerization with BCL-XL. J. Biol. Chem., 272: 24, 101-104, 28. Sedlak, T. W., Oltvai, Z. N., Yang, E., Wang, K., Boise, L. H., Thompson, C. B., and 1997. Korsmeyer, S. J. Multiple Bcl-2 family members demonstrate selective dimerizations 55. O'Connor, L., Strasser, A., O'Reilly, L. A., Hausmann, G., Adams, J. M., Cory, S., with Bax. Proc. Natl. Acad. Sci. USA, 92: 7834-7838, 1995. and Huang, D. C. Bim: a novel member of the Bcl-2 family that promotes apoptosis. 29. Yin, X-M., Oltvai, Z. N., and Korsmeyer, S. J. BH1 and BH2 domains of B1-2 are EMBO J., 17: 384-395, 1998. required for inhibition of apoptosis and heterodimerization with Bax. Nature (Lond.), 56. Onohara, N., Ding, L., Chen, S., and Nunez, G. Harakiri, a novel regulator of cell 369: 321-323. death, encodes a protein that activates apoptosis and interacts selectively with sur- 30. Cheng, E. H., Levine, B., Boise, L. H., Thompson, C. B., and Hardwick, J. M. Bax- vival-promoting proteins Bcl-2 and Bcl-X(L). EMBO J., 16: 1686-1694, 1997. independent inhibition of apoptosis by BcI-XL. Nature (Lond.), 379: 554-556, 1996. 57. Hegde, R., Srinivasula, S. M., Ahmad, M., Fernandes-Alnemri, T., and Alnemri, E. S. 31. Chinnadurai, G., mad Lutz, R. J. A conserved domain in Bak, distinct from BH1 and BH2, Blk, a BH3-containing mouse protein that interacts with Bcl-2 and Bcl-xL, is a potent mediates cell death and protein binding functions. EMBO J., 14: 5589-5596, 1996. death agonist. J. Biol. Chem., 273: 7783-7786, 1998. 32. Hunter, J. J., and Parslow, T. G. A peptide sequence from Bax that converts Bcl-2 into 58. Zha, J. P., Harada, H,, Yang, E., Jockel, J., and Korsmeyer, S. J. Serine phosphoryl- an activator of apoptosis. J. Biol. Chem., 271: 8521-8524, 1996. ation of death agonist Bad in response to survival factor results in binding to 14-3-3 33. Wang, K., Gross, A., Waksman, G., and Korsmeyer, S. J. Mutagenesis of the BH3 not Bcl-XL. Cell, 87." 619-628, 1996. domain of BAX identifies critical residues for dimerization and killing. Mol. Cell 59. Datta, S. R., Dudek, H., Tao, X., Masters, S., Fu, H., Gotoh, Y., and Greenberg, M. E. Biol., 18: 6083-6089, 1998. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death 34. Muchmore, S. W., Sattler, M., Liang, H., Meadows, R. P., Harlan, J. E., Yoon, H. S., machinery. Cell, 91: 231-241, 1997. Nettesheim, D., Chang, B. S., Thompson, C. B., Wong, S., Ng, S., and Fesik, S. W. 60. Xiang, J., Chao, D. T., and Korsmeyer, S. J. Bax-induced cell death may not require X-ray and NMR structure of human Bcl-xL, an inhibitor of programmed cell death. interleukin 1/3-converting enzyme-like proteases. Proc. Natl. Acad. Sci. USA, 93: Nature (Lond.), 381: 335-341, 1996. 14559-14563, 1996. 35. Sattler, M., Liang, H., Nettesheim, D., Meadows, R. P., Harlan, J. E., Eberstadt, M., 61. McCarthy, N. J., Whyte, M. K., Gilbert, C. S., and Evan, G. I. Inhibition of Yoon, H. S., Shuker, S. B., Chang, B. S., Minn, A. J., Thompson, C. B., and Fesik, Ced-3/ICE-related proteases does not prevent cell death induced by oncogenes, DNA S. W. Structure of Bcl-x(L)-Bak peptide complex-recognition between regulators of damage, or the Bcl-2 homologue Bak. J. Cell Bio1.,136: 215-227, 1997. apoptosis. Science (Washington DC), 275: 983-986, 1997. 62. Wolter, K. G., Hsu, Y. T., Smith, C. L, Nechushtan, A., Xi, Z. G., and Youle, R. J. 36. Veis, D. J., Sorenson, C. M., Shutter, J. R., and Korsmeyer, S. J. Bcl-2-deficient mice Movement of Bax from the cytosol to mitochondria during apoptosis. J. Cell Biol., demonstrate fulminant lymphoid apoptosis, polycystic kidneys, and hypopigmented 139: 1281-1292, 1997. hair. Cell, 75: 229-240, 1993. 63. Gross, A., Jockel, J., Wei, M. C., and Korsmeyer, S. J. Enforced dimerization of BAX 37. Nakayama, K., Negishi, 1., Kuida, K., Sawa, H., and Loh, D. Y. Targeted disruption results in its translocation, mitochondrial dysfunction and apoptosis. EMBO J., 17: of Bcl-2 ab in mice: occurrence of gray hair, polycystic kidney disease, and lympho- 3878-3885, 1998. cytopenia. Proc. Natl. Acad. Sci. USA, 91: 3700-3704, 1994. 64. Hsu, Y. T., and Youle, R. J. Bax in murine thymus is a soluble monomeric protein that 38. Kamada, S., Shimono, A., Shinto, Y., Tsujimura, T., Takahashi, T., Noda, T., displays differential detergent-induced conformations. J. Biol. Chem., 273: 10777- Kitamura, Y., Kondoh, H., and Tsujimoto, Y. Bcl-2 deficiency in mice leads to 10783, 1998. pleitrophic abnormalities: accelerated lymphoid cell death in thymus and spleen, 65. Knudson, C. M., and Korsmeyer, S. J. Bcl-2 and Bax function independently to polycystic kidney, hair hypopigmentation, and distorted small intestine. Cancer Res., regulate cell death. Nat. Genet., 16: 358-363, 1997. 55: 354-359, 1995. 66. Antonsson, B., Conti, F., Ciavatta, A., Montessuit, S., Lewis, S., Martinou, I., 39. Sorenson, C. M., Rogers, S. A., Korsmeyer, S. J., and Hammerman, M. R. Fulminant Bernasconi, L., Bernard, A., Mermod, J. J., Mazzei, G. Maundrell, K., Gambale, F., metanephric apoptosis and abnormal kidney development in bcl-2-deficient mice. Sadoul, R., and Martinou, J. C. Inhibition of Bax channel-forming activity by Bcl-2. Am. J. Physiol., 268: F73-F81, 1995. Science (Washington DC), 277: 370-372, 1997. 40. Knudson, C. M., Tung, K. S., Tourtellotte, W. G., Brown, G. A., and Korsmeyer, S. J. 67. Schlesinger, P. H., Gross, A., Yin, X-M., Yamamoto, K., Saito, M., Waksman, G., Bax-deficient mice with lymphoid hyperplasia and male germ cell death. Science and Korsmeyer, S. J. Comparison of the ion channel characteristics of proapop- (Washington DC), 270: 96-99, 1995. totic BAX and antiapoptotic BCL-2. Proc. Natl. Acad. Sci. USA, 94: 11357- 41. Perez, G. I., Knudson, C. M., Leykin, L., Korsmeyer, S. J., and Tilly, J. L. Apoptosis- 11362, 1997. associated signaling pathways are required for chemotherapy-mediated female germ 68. Hengartner, M. O., and Horvitz, H. R. Programmed cell death in Caenorhabditis cell destruction. Nat. Med., 3: 1228-1232, 1997. elegans. Curr. Opin. Genet. Dev., 4: 581-586, 1994. 42. Deckwerth, T. L., Elliot, J. L., Knudson, C. M., Johnson, E. M., Jr., Snider, W. D., 69. Hengartner, M. O., and Horvitz, H. R. C. elegans cell survival gene ced-9 encodes a and Korsmeyer, S. J. Bax is required for neuronal death after trophic factor depriva- functional homologue of the mammalian proto-oncogene bcl-2. Cell, 76: 665-676, 1994. tion and during development. Neuron, 17:401-411, 1996. 70. Conradt, B., and Horvitz, H. R. The C. elegans protein EGL-I is required for 43. Lowe, S. W., Ruley, H. E., Jacks, T., and Housman, D. E. p53-dependent apoptosis programmed cell death and interacts with the Bcl-2-1ike protein CED-9. Cell, 93: modulates the of anticancer agents. Cell, 74: 957-967, 1993. 519-529, 1998. 44. Symonds, H., Krall, L., Remington, L., Saenz-Robles, M., Lowe, S., Jack, T., and Van 71. Yuan, J., Shaham, S., Ledoux, S., Ellis, H. M., and Horvitz, H. R. The C. elegans cell Dyke, T. P53-dependent apoptosis suppresses tumor growth and progression in vivo. death gene ced-3 encodes a protein similar to mammalian interleukin-1/3-converting Cell, 78:703-711, 1994. enzyme. Cell, 75: 641-652, 1993. 45. Miyashita, T., and Reed. J. C. Tumor suppressor p53 is a direct transcriptional 72. Alnemri, E. S., Livingston, D. J., Nicholson, D. W., Salvesen, G. S., Thornberry, activator of the human bax gene. Cell, 80: 293-299, 1995. N. A., Wong, W. W., and Yuan, J. Human ICE/CED-3 nomenclature. Cell, 46. McCurrach, M. E., Connor, T. M., Knudson, C. M., Korsmeyer, S. J., and Lowe, S. W. 87: 171, 1996. Bax-deficiency promotes drug resistance and oncogenic transformation by attenuating 73. Zou, H., Henzel, W. J., Liu, X., Lutschg, A., and Wang, X. Apaf-1, a human protein p53-dependent apoptosis. Proc. Natl. Acad. Sci. USA, 94: 2345-2349, 1997. homologous to C. elegans CED-4, participates in cytochrome c-dependent activation 47. Yin, C. Y., Knudson, C. M., Korsmeyer, S. J., and VanDyke, T. Bax suppresses of caspase-3. Cell, 90: 405-413, 1997.

1700s

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 1999 American Association for Cancer Research. BCL-2 Gene Family and the Regulation of Programmed Cell Death

Stanley J. Korsmeyer

Cancer Res 1999;59:1693s-1700s.

Updated version Access the most recent version of this article at: http://cancerres.aacrjournals.org/content/59/7_Supplement/1693s.2

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/59/7_Supplement/1693s.2. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 1999 American Association for Cancer Research.