<<

Modern Pathology (2008) 21, S2–S7 & 2008 USCAP, Inc All rights reserved 0893-3952/08 $30.00 www.modernpathology.org

Targeted in

James R Downing

Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN, USA

Research conducted over the last two decades has yielded a detailed understanding of the molecular lesions that contribute to the malignant transformation of hematopoietic stem cells and committed progenitors into the various forms of acute and chronic leukemia. Although our understanding of the molecular pathology of leukemia remains incomplete, the information gained to date has had a profound impact on the way these malignancies are both diagnosed and monitored during therapy. More recently, targeted have been developed against some of the identified genetic lesions. These therapies have led to significant improvements in patient outcomes while simultaneously decreasing therapy-related toxicity. With the advent of genome-wide methods to define the total complement of genetic and epigenetic lesions involved in leukemogenesis, new targeted therapies can be anticipated. This review highlights some of the targeted therapies that are presently being used to treat hematopoietic malignancies and describes some of the recent advances that should have a significant impact on the development of future target therapies. Modern Pathology (2008) 21, S2–S7; doi:10.1038/modpathol.2008.13

Keywords: leukemia; translocations; tyrosine kinases; HDAC; monoclonal antibodies

A goal of modern research is to define the The first molecularly targeted cancer underlying molecular genetic lesions responsible for therapy: ATRA the establishment of the malignant clone. The hope is that this information will lead to the identification Acute Promyelocytic Leukemia of rational molecular targets against which novel Analysis of chromosomal abnormalities within therapeutic agents can be developed. Although leukemic blasts revealed somatically acquired chro- these hopes are being realized across the spectrum mosomal rearrangements in the majority of cases. of human , to date their direct influence The targets of these rearrangements are genes on clinical care is probably most evident in the encoding proteins that play fundamental roles in treatment of hematopoietic malignancies. Specifi- normal hematopoietic cell development, differentia- cally, molecular genetic data have resulted in tion, proliferation, and survival. The most common significant changes in our understanding of targets of these rearrangements are genes that the pathogenesis of both acute myeloid leukemia encode DNA-binding transcription factors or tran- (AML) and acute lymphoid leukemia (ALL), scriptional regulatory proteins. The prototypical and have resulted in the identification of a translocation is the t(15;17) translocation of acute number of molecular targets against which targeted promyelocytic leukemia (APL). The product en- 1,2 therapies have been developed. This article coded by this translocation not only drives the summarizes some of the recent advances made formation of the leukemic clone, but also serves as a in our understanding of the molecular pathology specific target for the highly effective therapy, all of acute leukemia, and highlights how this trans retinoic acid (ATRA).3,4 information is being used to develop targeted APL consists of a clonal population of malignant therapies. myeloid cells blocked at the promyelocyte stage of differentiation. In over 95% of cases, a t(15;17)(q22;q21) chromosomal translocation is pre- sent within the leukemic cells. This translocation generates a chimeric gene between the promyelocy- tic leukemia gene (PML) on chromosome 15 and the Correspondence: JR Downing, MD, Department of Pathology, retinoic acid receptor a gene (RARa) on chromosome St Jude Children’s Research Hospital, 332 North Lauderdale a Street, Memphis, TN 38105, USA. 17, and results in the production of a PML-RAR E-mail: [email protected] . Expression of PML-RARa as a Received 11 December 2007; accepted 31 December 2007 transgene in mice induces acute leukemia with in leukemia JR Downing S3 features of APL, confirming the central role of this Mechanism of Transformation in APL and the Role fusion protein in leukemogenesis. ATRA Played in Its Discovery RARa is a ligand-dependent transcription factor and a member of the nuclear hormone receptor Clinical trials initially performed in China revealed superfamily. In the presence of its ligand, retinoic a unique sensitivity of APL to ATRA. The most acid, RARa regulates the expression of a large remarkable aspect of these observations were that number of target genes, a subset of which are they occurred before RARa was identified as one important for normal myeloid cell differentiation. of the targets of the t(15;17) translocation. In fact, RARa is organized into distinct functional the results of these clinical studies were in part the domains including a DNA-binding domain and a driving force to assess whether the RARa gene might large ligand-binding, heterodimerization, and trans- be involved in the pathogenesis of this acute activation domain. PML is a multidomain protein leukemia subtype. With the cloning of the transloca- that contains a unique cysteine- and histidine- tion, the reasons for APL’s responsiveness to ATRA rich zinc-binding RING finger domain followed were quickly elucidated. by two cysteine/histidine-rich regions known as PML-RARa induces leukemia in part by inhibiting B-boxes and an a-helical coiled-coil domain. the normal biological activities of both RARa and In the PML-RARa chimeric product, nearly all of PML. As predicted from its structure, PML-RARa the key functional domains of each molecule continues to bind RAREs, but is unresponsive to are retained, suggesting that the fusion protein retinoic acid, and thus instead of activating tran- influences the biological function of both normal scription, it dominantly represses transcription proteins. of ligand-bound RARa. Moreover, the PML-RARa RARa normally dimerizes with a member of the fusion protein sequesters RXRs away from other retinoid-X receptor (RXR) family leading to the nuclear hormone receptors, such as those for formation of a heterodimer with high DNA-binding vitamin D and thyroid hormone, resulting in an affinity. In the absence of retinoic acid, these RARa/ inhibition of their transcriptional activity. The RXR heterodimers bind to specific retinoic acid combined effect on the transcriptional cascade response elements (RAREs) in the promoter of controlled by the nuclear hormone receptors target genes and induce transcriptional repression. contributes to the block in differentiation at This repression is mediated by the RARa/RXR the promyelocyte stage. PML-RARa also induces heterodimer interacting with transcriptional repres- disruption of normal PML NBs and the redistribu- sors including the nuclear receptor-corepressor tion of PML into a microparticulate pattern within N-CoR/SMRT, the transcriptional corepressors the nucleus. This change in nuclear localization Sin3A or Sin3B, and histone deacetylases (HDACs). impairs the normal growth-inhibitory and proapop- The latter enzymes deacetylate lysine residues in totic effects of PML. Recent studies suggest that nucleosomal histones, leading to a closed chromatin these activities of PML-RARa may result from the structure and repression. Binding of retinoic full-length fusion protein, as well as the cleaved acid to RARa induces a conformational change that product that results from proteolytic cleavage causes disassembly of the repression complex and by neutrophil elastase, a neutral serine protease the recruitment of transcriptional coactivators in- expressed at high levels in promyelocytes. cluding CBP and p300 to RARa/RXR. CBP and p300 PML-RARa, like wild-type RARa, directly binds contain intrinsic histone acetylase (HAT) activity, the nuclear corepressor complex, resulting in which acetylates the lysine residues of histones transcriptional repression. Unlike the wild-type resulting in nucleosome unfolding and an increased receptor, however, PML-RARa is unresponsive to accessibility of the DNA to transcription factors. physiological levels of retinoic acid and remains a In addition, HATs can directly acetylate transcrip- constitutive repressor in its presence. ATRA, by tion factors and other cellular proteins and alter contrast, induces a conformational change in PML- their activity. RARa resulting in the release of NcoR and its PML is expressed in all cells and exists both as a replacement by transcriptional coactivators. Thus, free form that localizes within the nucleoplasm and treatment with ATRA reverses the inhibitory activity as a conjugated form covalently linked to the of PML-RARa on RARa. Moreover, ATRA induces ubiquitin-like protein SUMO, which localizes to cleavage of PML-RARa through a proteasome- the so-called PML nuclear bodies (NBs) or PML mediated pathway reversing the effect of the fusion oncogenic domains. Approximately 10–30 PML NBs protein on normal PML functions. Together, these are present within a cell. In addition to PML, these activities induce growth arrest and terminal differ- NBs contain over 30 other proteins. Recent studies entiation of the malignant promyelocytes. have demonstrated that PML functions in several Although ATRA is highly effective at inducing the cellular processes, including proliferation, senes- terminal differentiation, it is not curative as a single cence, and . Importantly, chemical- agent. Moreover, ATRA resistance resulting from the induced tumors occur at an increased frequency in acquisition of mutations in the RARa ligand-binding PML-deficient mice, suggesting that PML is a tumor domain of the fusion protein is a frequent occur- suppressor. rence. As a result, ATRA is now used along with

Modern Pathology (2008) 21, S2–S7 Targeted therapy in leukemia JR Downing S4 other chemotherapeutic agents, a strategy that has In addition to PML-RARa, a large number of other been shown to be superior to either ATRA or translocation-encoded onco-fusion proteins func- alone. tion in part through transcriptional repression. Recently, studies have demonstrated APL cases to These include t(8;21) (AML1-ETO), inv(16) (CBFb- be sensitive to treatment with arsenic trioxide MYH11), t(1;19) (E2A-PBX1), t(8;16) (MOZ-CBP),

(As2O3). In contrast to ATRA, As2O3 does not bind and t(11;16) (MLL-CBP), as well as others. As to RARa, but instead binds to the PML portion of the predicted, HDACi have been demonstrated to have chimeric product, inducing its degradation through activity in these . The activity of the a proteasome-dependent mechanism. Arsenic triox- HDACi even within these leukemias, however, goes ide’s primary effect on promyelocytes is thought to beyond the transcriptional repression mediated by be induction of apoptosis. Protocols combining the the oncogenic transcription factors. Specifically, use of ATRA, As2O3, and standard chemotherapeu- cancer cells appear to have global modifications of tic are showing promise for relapse disease, and may their chromatin that lead to wide ranging changes in provide a survival advantage over standard ATRA gene transcription. This modified epigenome pro- plus chemotherapy for newly diagnosed patients. vides a setting where the HDACi lead to differential effects (gene activation) between normal and cancer cells. This differential in part explains the marked Histone deacetylase inhibitors difference in the sensitivity of cancer cells vs normal cells to the HDACi. The lessons learned from APL suggest that reversing Preclinical work with several HDACi has shown constitutive transcriptional repression could lead impressive antitumor activity for a variety of to terminal differentiation of leukemia cells. In APL, cancers, including acute leukemia. In phase I ATRA reverses repression by directly inhibiting the clinical trials, these agents have been well tolerated ability of the oncogenic PML-RARa fusion protein to and have shown evidence of partial responses. direct the repression complex to the transcriptional These encouraging results are being followed in regulatory region of target genes. An alternative single-agent phase II studies and in combination approach is to directly inhibit the enzymatic activity with a variety of other agents. The theoretical of the histone deacetylases within the transcrip- underpinning of this work suggests that HDACi will tional repression complex.5–7 likely become a stable part of the combinatorial The relative state of chromatin in the transcrip- . tional regulatory regions of genes plays a key role in controlling the level of gene transcription. The nucleosome serves as the basic unit of chromatin inhibitors in acute leukemia structure and consists of approximately two turns of the DNA double helix wound around a histone Protein kinases play a central role in the regulation octamer core. A diverse range of post-translational of a wide variety of intracellular signaling pathways. modifications of the amino-terminal tails of histones Through reversible post-translational modifications controls the packing of the nucleosomes, and thus of substrate proteins, kinases influence nearly the relative access of the DNA to transcriptional all cellular processes, including development, dif- regulatory proteins. Modification of the histone ferentiation, metabolism, proliferation, cell move- tails includes methylation, phosphorylation, ubiqui- ment, and apoptosis. In addition, aberrant kinase tination, sumoylation, and acetylation. Acetylation signaling is a recurrent mechanism implicated in is controlled by the balance between HATs and cellular transformation. Constitutive activation of HDACs. HATs transfer acetyl groups to the amino- protein kinases through oncogenic mutations, auto- terminal lysine group in histones, resulting in the crine or paracrine stimulation, or overexpression as elimination of the positive charge and an opening a result of genomic amplification has been docu- of the chromatin structure. HDACs catalyze the mented in a wide spectrum of human cancers. removal of the acetyl group and lead to a condensa- In normal hematopoiesis, both tyrosine and tion of the chromatin and transcriptional repression. serine/threonine kinases play essential roles in A relatively large number of small molecule histone cytokine-induced proliferation, survival, and differ- deacetylase inhibitors (HDACi) have been devel- entiation. Thus, oncogenic activation of members of oped and tested in preclinical and clinical trials.5–7 this extended family of genes would be predicted These agents have been shown to not only have to contribute to the neoplastic transformation of anticancer activity, but to demonstrate a remarkable hematopoietic stem cells (HSCs) and lineage-com- level of tumor specificity, with cancer cells showing mitted progenitors. In fact, it has been suggested a 10-fold greater sensitivity than normal cells. that constitutive activation of growth factor recep- Typically, these agents induce apoptosis of cancer tors or downstream components of their signaling cells through a combination of targeted effects on cascades may be an obligate step in leukemogenesis, the oncogenic transcription factors and through providing a proliferative and/or survival advantage. more global effects on gene transcription and non- As described in the first lecture in this course, one transcriptional targets.5–9 of the first examples of the contribution of altered

Modern Pathology (2008) 21, S2–S7 Targeted therapy in leukemia JR Downing S5 protein kinase signaling to leukemogenesis was the Stable expression of either class of FLT3 muta- identification in chronic myeloid leukemia (CML) tions renders hematopoietic cells growth factor- of the t(9;22)-encoded BCR-ABL fusion protein, a independent. Indeed, mice receiving bone marrow constitutively activated form of the ABL tyrosine cells expressing mutant FLT3 develop an oligoclonal kinase. A second example is the constitutive activa- myeloproliferative disorder characterized by sple- tion of the FMS-related tyrosine kinase-3 (FLT3), a nomegaly and leukocytosis. member of the class III The high frequency of FLT3 mutations makes this family, which also includes CSFR, c-, PDGFRa, an ideal candidate for a small molecule inhibitor. A and PDGFRb. FLT3 is comprised of an extracellular number of inhibitors have been developed with domain, a juxtamembrane (JM) domain just within varying degrees of specificity. These include CEP- the cytoplasmic membrane, and two tandem 701 (Cephalon), PKC-412 (Novartis), and SU11248 tyrosine kinase domains. FLT3 activates several (Sugen). Phase I/II clinical studies have been carried downstream signaling molecules through direct out in relapse patients with AML or MDS with phosphorylation or association, including the p85 mutant FLT3. These studies have demonstrated subunit of phosphatidyl inositol, SHC, SHP2, GRB2, significant partial responses in a proportion of the STAT 5A, and STAT 5B. Although the expression of patients, although the responses were short lived. FLT3 is normally restricted to early hematopoietic Biochemical analysis demonstrated effective inhibi- progenitors, it is also expressed in many primary tion of FLT3 kinase activity at the doses achieved. hematopoietic malignancies, including the majority The limited responses are not surprising and FLT3 of AMLs, a proportion of B-cell and T-cell lineage inhibitors in combination with other therapeutic ALLs, and CML in lymphoblast crisis. These data agents will likely be required for optimal clinical suggest that aberrant signaling through the FLT3 effects. Phase I studies of combinations of these receptor may play a role in the survival or prolifera- FLT3 inhibitors with standard AML therapies are in tion of leukemic blast. Consistent with this sugges- progress. tion, stimulation of FLT3-expressing leukemias with the FLT3 ligand (FL) induced a dose-dependent proliferation of leukemic blasts. In addition, a Monoclonal antibodies as therapeutic significant proportion of leukemic cell lines coex- agents press both FL and FLT3, suggesting that autocrine stimulation may play a role in the proliferation of Over the last two decades, detailed phenotypic leukemia blast. studies have led to the identification of leukemia- More recently, activating mutations have been specific immunophenotypic signatures consisting of identified in FLT3 in a subset of leukemias.10 lineage-specific and nonspecific antigens in the Specifically, an internal tandem duplication (ITD) majority of ALLs and AMLs. These phenotypic of the JM region of FLT3 was shown to induce signatures are now routinely used to measure the constitutive activation of the receptor’s tyrosine so-called minimal residual disease and to modify kinase activity. The length of ITD varies from 3 to therapy based on these measurements. What has Z400 base pairs, and is always in-frame. Overall, been less rewarding is the search for individual the frequency of this mutation has been estimated to leukemia-specific antigens, that is surface proteins be 15–25% in AML, but less than 1% in cases of that are expressed only on leukemic blasts and not ALL. A second less common type of FLT3 activating on normal HSCs or lineage-committed progenitors. mutation is a missense point mutation in codons An antigen that comes close to these criteria is encoding amino acids within the activation loop of CD33 in AML. CD33 is a 67 kDa surface glycoprotein the kinase domain. The activation loop is a general of unknown function that is expressed on the component of tyrosine kinase and exerts autoinhi- leukemic blasts in over 80% of cases, but is not bitory function by limited access of ATP and expressed on normal HSCs. Monoclonal antibodies substrate to the catalytic domain. FLT3 activation (mAbs) against CD33 are routinely used in the loop mutations have been identified in approxi- characterization of leukemias. To convert mAbs mately 7% of AML, but is again only rarely seen in to therapeutic agents, the antibodies need to be cases of ALL. Among all the mutations identified, humanized to reduce immunogenicity, enhance the most common is D835Y; other substitutions stability in circulation, and improve their ability include D835V, D835H, D835E, D835N, and I836M. to recruit cytotoxic cells and complement for Importantly, the presence of an FLT3 ITD, and to Ab-mediated cell killing.11,12 A humanized IgG4k a lesser degree the D835/836 point mutations, is anti-CD33 mAb has been developed. This antibody predictive of a poor prognosis. The frequency has been conjugated to the chemotherapy agent of FLT3 mutations in AML is somewhat higher through a hydrazone linker to form in adults than in children, largely due to the rarity the therapeutic agent known as gemtuzumab ozoga- of these mutations in children less than 10 years of micin (Mylotarg).13 age. Furthermore, the frequency of FLT3 mutations bind to CD33 on the varies significantly between genetic subtypes of surface of AML blasts and is rapidly internalized by AML, with the highest incidence in APL. receptor-medicated endocytosis. Upon entering the

Modern Pathology (2008) 21, S2–S7 Targeted therapy in leukemia JR Downing S6 acidic environment of the lysosome, the hydrozone have been developed to identify copy number linker is cleaved resulting in the release of calichea- alterations including fluorescence in situ hybridiza- micin. Calicheamicin is a highly potent antitumor tion, comparative genomic hybridization (CGH), antibiotic that cleaves double-stranded DNA, result- array-CGH, and single nucleotide polymorphism ing in tumor cell killing. Importantly, calicheamicin (SNP) arrays. Array-based methods involve hybri- is highly toxic and cannot be delivered systemically dizing leukemia cell DNA to arrays containing without significant toxicity. Thus, by targeting the thousands of pieces of DNA either as bacterial agents specifically to leukemic blasts, gemtuzumab artificial chromosome (BAC) or oligonucleotide ozogamicin allows the effective use of this agent for probes. The resolution is dependent upon the the treatment of AML. platform used and can vary from a mean intermarker In clinical trials, gemtuzumab ozogamicin has distance of over 1 Mb for older BAC arrays to less shown impressive results, with 30% of patients in than 5 kb for newer oligonucleotide-based platforms. first relapse achieving a complete remission, with A particularly powerful platform is the 500K SNP median relapse-free survival of 7.2 months. More arrays from Affymetrix. By comparing the SNP recent studies have suggested that gemtuzumab genotype of germline and tumor samples, these ozogamicin may yield a higher complete remission arrays allow the detection of copy-neutral loss of rate when administered with intensive chemother- heterozygosity (or uniparental disomy), in addition apy. Phase III studies are underway in which to directly inferring DNA copy number from probe gemtuzumab ozogamicin is being incorporated hybridization intensity. with standard chemotherapy in newly diagnosed As a representative example, we have recently patients. If positive results are obtained from these used Affymetrix 500K SNP arrays to detect copy studies, it would result in the first change in number alterations in pediatric ALL at a resolution standard induction therapy in AML in over 30 of approximately 5.0 kb. This analysis is very years. Treatment with gemtuzumab ozogamicin is accurate and provides a fingerprint of deletions not without side effects. Severe neutropenia and and amplifications. The methodology was able to thrombocytopenia are frequently seen and this is identify the known DNA copy number changes consistent with the expression of CD33 on maturing including the multiple whole chromosomal gains myeloid progenitors. Transient and reversible hepa- characteristic of hyperdiploid B-progenitor ALL totoxicity is also seen in a proportion of patients, with 450 chromosomes, the duplication of chromo- and occasional cases of veno-occlusive disease have some 1q in ALL with t(1;19)(q23;p13) as a result of been documented. unbalanced translocation, and deletions at 9p21.3 (harboring the CDKN2A and CDKN2B genes) and 12p13.2 (ETV6). In addition to these known lesions, Are there new targets for directed therapy? this analysis identified a number of new lesions. The most striking finding was mono-allelic dele- An important observation from modeling leukemias tions of genes that play a critical role in regulating in mice is that individual oncogenic lesions alone B-cell development and differentiation in approxi- are insufficient to generate a full leukemia. Thus, mately 40% of B lineage cases.15 lesions affecting multiple different pathways are The most frequently targeted gene within the required to lead to cellular transformation. Candi- B-cell development pathway, occurring in almost date gene approaches have been used to identify 30% of cases, was PAX5, a paired domain transcrip- contributing lesions and have led to the identi- tion factor located at 9p13. The PAX5 copy number fication of deletions and epigenetic silencing of alterations included mono-allelic loss in 53 cases, CDKN2A and mutations of NOTCH1 in a high bi-allelic loss in 3 cases, and an internal amplifica- percentage of T-lineage ALL cases. However, for tion in 1 case. Importantly, these alterations were most leukemias, the full complement of cooperating somatic in nature and were present in over 90% of lesions remains to be defined. It is anticipated that blasts. Moreover, almost half of the deletions were identifying these lesions will help to define the confined to PAX5, with most resulting in the molecular ‘Achilles heels’ against which targeted deletion of only a subset of PAX5 exons. The PAX5 therapy can be developed. lesions are predicted to result in either haploinsuf- Contributing mutations include deletions, ampli- ficiency or the generation of hypomorphic alleles fications, and point mutations, as well as epigenetic that produce proteins lacking the DNA-binding changes that influence gene expression.14 The domain or the transcriptional activation domain. In identification of the full complement of point addition, four cases were identified with cryptic mutations within a population of leukemia cells is translocations (two cases with PAX5-TEL, one case cost prohibitive with present sequencing methodol- with PAX5-FOXP1, and one case with PAX5-EVI3) ogy. However, rapid advances in re-sequencing and 14 cases had point mutations including mis- approaches are being made and it is anticipated sense, frame-shift, and splice-site mutations that full genome sequence of individual cases will that clustered in the DNA-binding paired domain be achievable at a reasonable cost within the next and transactivation domains. In addition to 5–10 years. A variety of genome-wide approaches PAX5, a number of other genes involved in B-cell

Modern Pathology (2008) 21, S2–S7 Targeted therapy in leukemia JR Downing S7 development and differentiation were found to be newly diagnosed patients with APL. The recent deleted including EBF (8 cases), Ikaros (19 cases), advances in our understanding of the molecular Aiolos (6 cases), TCF3 (1 case), and BLNK (2 cases). pathology of acute leukemia coupled with methodo- Taken together, mutations in genes regulating logical advances in genome-wide scans for mutations B-cell development were identified in over 40% of and high-throughput screens for drugs should lead to pediatric B-progenitor ALL. a significant increase in the number of targeted agents Mice null for PAX5 show a complete arrest in over the next decade. B-cell development at the pro-B-cell stage of devel- opment prior to completing immunoglobulin heavy chain rearrangement. Moreover, loss of a single EBF References allele leads to a partial arrest in differentiation at this stage of B-cell development. Thus, the simplest 1 Tallman MS, Gilliland G, Rowe JM. Drug therapy for interpretation of the data is that the identified acute myeloid leukemia. Blood 2005;4:1154–1163. mutations lead to a reduction in the functional 2 Estey E, Dohner H. Acute myeloid leukaemia. Lancet level of PAX5 and/or other key regulators of B-cell 2006;368:1894–1907. development and differentiation and contribute to 3 Chou WC, Dang CV. Acute promyelocytic leukemia: the block in differentiation at the pro-B-cell stage of recent advances in therapy and molecular basis of development. In addition to contributing to the response to arsenic therapies. Curr Opin Hematol block in differentiation, the mutations are likely to 2004;12:1–6. 4 Douer D, Tallman MS. Arsenic trioxide: new clinical also collaborate with other known genetic lesions in experience with an old medication in hematologic leukemogenesis. malignancies. J Clin Oncol 2005;23:2396–2410. The most exciting possibility raised by these data 5 Johnstone RW. Histone-deacetylase inhibitors: novel is to use the genetic defects as a way of developing drugs for the treatment of cancer. Nat Rev Drug Discov leukemia-specific therapy. For example, can a small 2002;1:287–299. molecule inducer of differentiation (agonist) bypass 6 Marks PA, Richon VM, Miller T, et al. Histone the block resulting from the identified lesions, and deacetylase inhibitors. Adv Cancer Res 2004;91: will this in turn trigger a leukemia cell-specific 137–168. apoptotic response. With recent advances in high- 7 Bolden JE, Peart MJ, Johnstone RW. Anticancer throughput screening methodology and the avail- activities of histone deacetylase inhibitors. Nat Rev Drug Discov 2006;5:769–784. ability of small molecule libraries, such screens 8 Insinga A, Monestiroli S, Ronzoni S, et al. Inhibitors of can be readily accomplished. The combination of histone deacetylases induce tumor-selective apoptosis genome-wide screening approaches to identify the through activation of the death receptor pathway. complete complement of genetic lesions within a Nat Med 2005;11:71–76. cancer, coupled with high-throughput screens for 9 Nebbioso A, Clarke N, Voltz E, et al. Tumor-selective new small molecules targeted toward these newly action of HDAC inhibitors involves TRAIL induction identified lesions, should lead to a rapid expansion in acute myeloid leukemia cells. Nat Med 2005;11: in our understanding of the molecular pathology of 77–84. leukemia and in the number of drugs at our disposal 10 Levis M, Small D. FLT3 tyrosine kinase inhibitors. to effectively treat these cancers. Int J Hematol 2005;82:100–107. 11 Harris M. Monoclonal antibodies as therapeutic agents for cancer. Lancet Oncol 2004;5:292–302. Summary 12 Wu AM, Senter PD. Arming antibodies: prospects and challenges for immunoconjugates. Nat Biotechnol Significant advances have been made in our under- 2005;23:1137–1146. standing of the molecular pathology of acute 13 Linenberger ML. CD33-directed therapy with gemtu- leukemias. These data are beginning to have a direct zumab ozogamicin in acute myeloid leukemia: pro- impact on our ability to accurately diagnose, gress in understanding cytotoxicity and potential classify, and treat these cancers so that each patient mechanisms of drug resistance. Leukemia 2005;19: has the best chance for a cure. The recent use of the 176–182. 14 Veltman JA, de Vries BB. Diagnostic genome profiling: so-called targeted therapies directed toward specific unbiased whole genome or targeted analysis? J Mol genetic lesions within leukemic cells has further Diagn 2006;8:534–537. advanced cure rates while simultaneously reducing 15 Mullighan CG, Goorha S, Radtke I, et al. Genome-wide treatment-associated toxicities. This is best exemplified analysis of genetic alterations in acute lymphoblastic by the use of ATRA as part of front-line therapy for leukaemia. Nature 2007;446:758–764.

Modern Pathology (2008) 21, S2–S7