Anaplastic large-cell lymphoma (ALCL) was initially recognized on the basis of morphologic features and the consistent expression of CD30. It then became evident that the majority of these tumors are derived from lymphoid cells of T or null immunophenotype. The subsequent finding that t(2;5)(p23;q35) occurs in 40% to 60% of ALCL patients established a distinct clinicopathologic entity. This chromosomal translocation induces the formation of the chimeric protein nucleophosmin–anaplastic lymphoma kinase (NPM-ALK), which possesses significant oncogenic potential resulting from the constitutive activation of the tyrosine kinase ALK. In addition to its specific pathophysiologic events, NPM-ALK–expressing lymphoma presents with consistent clinical manifestations. Only 13 years after the identification of NPM-ALK, tremendous progress has been made in our understanding of this molecule because of the relentless efforts of multiple investigators who have dissected its biologic roles using in vitro and in vivo experimental models. Several upstream modulators, cross-reacting oncogenes, and downstream effectors of NPM-ALK have been identified and characterized. Understanding these interacting oncogenic systems is expected to facilitate the design of new therapeutic strategies and agents. In this review, we briefly discuss ALCL and focus on NPM-ALK.

Anaplastic large-cell lymphoma (ALCL) is a relatively uncommon tumor. It was first recognized by Stein et al1  in 1985, who reported the consistent expression of the Ki-1 antigen (later designated CD30) in tumors with frequent cohesive proliferation of large pleomorphic cells. Most of these tumors were labeled “histiocytic malignancies.”1  The Ki-1 monoclonal antibody was originally described by the same group and was used to identify a novel antigen in the Hodgkin lymphoma cell line L428.2  Subsequent immunophenotyping and gene rearrangement studies showed that the vast majority of ALCL tumors are derived from lymphoid cells of T or null immunophenotype.3 

Histologically, several ALCL variants have been described. Of these variants, the common, lymphohistiocytic, and small-cell are the most frequently encountered. The “horseshoe” or “wreath” cell is considered the cytologic hallmark of this disease.4  ALCL occurs as 2 distinct clinical entities, as a widespread systemic disease, or as a localized cutaneous disease. Systemic ALCL comprises 2% to 8% of non-Hodgkin lymphomas in adults and 10% to 15% of these lymphomas in children.5  The frequency of ALCL increases to 30% to 40% of non-Hodgkin lymphomas in children when only cases with large-cell morphology are included.

The recognition of t(2;5)(p23;q35) established the molecular definition of a subset of ALCL tumors that harbors this translocation.6,8  In 1994, 2 independent groups cloned the genes involved in this translocation and illustrated the fusion of the nucleophosmin (NPM) gene on chromosome 5q35 to the previously unidentified gene anaplastic lymphoma kinase (ALK) gene on 2p23.9,10  This chromosomal translocation leads to the generation of the chimeric protein NPM-ALK. Identifying these tumors with this translocation became clinically feasible after the production of antibodies that specifically interact with chimeric NPM-ALK and full-length ALK proteins.11  Tumors harboring t(2;5)(p23;q35) and expressing ALK soon became a distinct clinicopathologic entity known as ALK-positive (ALK+) ALCL, which is included in the current World Health Organization classification system.12  Several other chromosomal aberrations involving ALK were subsequently identified, including t(1;2)(q21;p23) [chimeric protein TPM3-ALK], inv2(p23q35) [ATIC-ALK], t(2;3)(p23;q21) [TFG-ALK], t(2;17)(p23;q23) [CLTC-ALK], t(2;19)(p23;q13.1) [TPM4-ALK], and t(2;X)(p23;q11-12) [MSN-ALK].13  Immunohistochemical staining has shown that ALK expression is both cytoplasmic and nuclear in tumors with t(2;5)(p23;q35) but is strictly cytoplasmic in most of the other variants.14 

All the ALK chromosomal aberrations lead to the expression and constitutive activation of ALK. This transmembrane receptor tyrosine kinase belongs to the insulin receptor superfamily. ALK expression in humans is normally limited to cells of neural origin. In mice embryos, it is widely expressed in the nervous system, but its expression level decreases significantly at birth.15,16  In Drosophila melanogaster, ALK is widely expressed in the gut.17  These observations suggest that ALK plays an important role in the development of these systems. Full-length ALK has been detected in neuroblastoma and rhabdomyosarcoma.18,19  Full-length ALK protein expression has been described not only in nonhematologic tumors but also in rare cases of diffuse large B-cell lymphoma.20 

Whereas full-length ALK is not detected in normal hematopoietic cells, low levels of NPM-ALK and ATIC-ALK fusion gene mRNA have been identified in these cells.21,22  The expression of ALK in hematologic neoplasms is largely limited to ALCL tumors of T-cell or null-cell immunophenotype, in which 40% to 60% of these tumors express ALK.18  In 80% of these cases, ALK expression results from t(2;5)(p23;q35). Rare cases of large B-cell lymphoma have been found to have ALK rearrangements.23,24 ALK rearrangements and CLTC-ALK, TPM3-ALK, and TPM4-ALK chimeric proteins have also been detected in inflammatory myofibroblastic tumors and neuroblastoma.25,26 

How ALK is physiologically activated is not completely known. In D melanogaster, ALK is activated by the Jelly belly (Jeb) protein, and the Jeb/ALK pathway appears to play an important role in the development of the gut system.27,28  In humans, the heparin-binding growth factors pleiotrophin and midkine have been reported to be the ligands binding and activating ALK,29,30  but this idea is still controversial.31 

ALK+ ALCL tends to affect children and young adults and has a striking male predominance. The majority of ALK+ ALCL cases present as advanced stage (III or IV) systemic disease with generalized lymphadenopathy and extranodal involvement; particularly of the skin and soft tissue.32  Compared with ALK-negative ALCL, ALK+ ALCL demonstrates a significantly favorable prognosis.33  In this review, we specifically discuss ALK+ ALCL and focus on the pathobiology of NPM-ALK.

To understand the oncogenic role of NPM-ALK, we need to understand the physiologic functions of the ALK and NPM proteins. We discussed the physiologic functions of ALK in the previous section. In contrast to ALK, NPM is a ubiquitously expressed RNA-binding nucleolar phosphoprotein.34  Its physiologic functions include the shuttling of ribonucleoproteins between the nucleus and cytoplasm. NPM carries the oligomerizing motif that drives the homodimerization of NPM-ALK, which leads to the constitutive activation of ALK tyrosine kinase.35 

Numerous studies have proven that NPM-ALK is oncogenic, and its transforming ability has been repeatedly demonstrated in vitro.35,36  Transgenic mouse models have shown that the enforced expression of NPM-ALK leads to the development of malignant lymphoma. A significant number of these lymphomas were of plasmablastic or B-cell immunoblastic morphology and phenotype, even when NPM-ALK expression was driven by T-cell–associated antigen promoters.37,,,,42  Furthermore, several of these lymphomas were restricted to the mediastinum and they lacked CD30 antigen expression.

These important observations indicate that despite the universally accepted role of NPM-ALK in promoting lymphomagenesis, it is probably not the only factor that configures the known morphologic, immunophenotypic, and clinical features of NPM-ALK–expressing ALCL as we know it in humans. Most probably, NPM-ALK interacts with other biologic and molecular factors to result in the development of NPM-ALK–expressing lymphoma in humans with its characteristic features. NPM-ALK has been shown to interact with a wide range of oncogenic molecules (Figure 1), several of which are discussed in the subsequent sections.

Figure 1

Molecular network interacting with NPM-ALK. A complex network of protein kinases, protein phosphatases, transcription factors, apoptosis and cell-cycle regulators, adaptor proteins, and other molecules has been proposed to interact with NPM-ALK. The association and interaction between NPM-ALK and the majority of these molecules have been documented and briefly discussed in this review article. This model provides a rationale for designing specific and selective therapeutics that can target, individually or synergistically, members of this network and disrupt their oncogenic effects.

Figure 1

Molecular network interacting with NPM-ALK. A complex network of protein kinases, protein phosphatases, transcription factors, apoptosis and cell-cycle regulators, adaptor proteins, and other molecules has been proposed to interact with NPM-ALK. The association and interaction between NPM-ALK and the majority of these molecules have been documented and briefly discussed in this review article. This model provides a rationale for designing specific and selective therapeutics that can target, individually or synergistically, members of this network and disrupt their oncogenic effects.

Close modal

Jak/Stat

Signal transducers and activators of transcription (Stats) comprise a family of 7 latent transcription factors. Three major mechanisms contribute to the phosphorylation and subsequent activation of these proteins.43  The first is via a family of 4 cytokine receptor-associated tyrosine kinases known as Janus kinases (Jaks). The second mechanism of Stat activation is related to its interaction with receptor tyrosine kinases such as receptors for platelet-derived and epidermal growth factors. The third mechanism is related to its interaction with cytoplasmic tyrosine kinases such as Src and Abl. The Jak/Stat signaling pathway is one of the most extensively studied oncogenic mechanisms in NPM-ALK–expressing ALCL.

On cytokine/receptor engagement, Jaks undergo autophosphorylation at specific tyrosine residues. Some of these phosphotyrosine residues serve as docking sites where Stats bind to the receptor and undergo phosphorylation by Jaks. The activation of Stats is followed by their translocation to the nucleus where they regulate the transcription of a number of genes known to be directly involved in apoptosis, the cell cycle, and cell growth. This signaling pathway plays pivotal roles in normal cell differentiation and development.44  The activation status of Stats is under strict regulation, which involves the proper functioning of the activation and inhibition pathways.

Mounting evidence suggests that constitutively activated Stat3 is oncogenic and is found in a wide range of human cancers.45  In most experimental models, the net result of Stat3 activation is enhanced cell survival and proliferation. Blockade of Stat3 signaling has been shown to suppress tumorigenesis in vitro and in vivo. Constitutive activation of Stat3 is a highly consistent finding in ALK+ ALCL, and Stat3 activation significantly contributes to the pathogenesis of this lymphoma.46,48  Transfection of a Stat3 dominant-negative construct into ALK+ ALCL cells has been shown to induce cell-cycle arrest and apoptosis.49 

In parallel to the fact that Stat3 is activated by multiple mechanisms, the constitutive activation of Stat3 in ALK+ ALCL is multifactorial.50  NPM-ALK has been shown to physically bind to and phosphorylate Stat3, which results in its activation.51  In addition, Jak3, a physiologic activator of Stat3, is expressed and highly activated in ALK+ ALCL cell lines and primary tumors.51,53  Unlike the activation of other members of the Jak family of kinases, the activation of Jak3 is restricted to a relatively small number of cytokines that possess the IL-2 common cytokine receptor γ-chain in their respective receptors.54,55  In addition to IL-2, this family includes IL-4, IL-7, IL-9, IL-15, and IL-21. Because Jak3 is a major physiologic activator of Stat3, it is not surprising that Jak3 contributes to the activation of Stat3 in ALK+ ALCL cells.52  Jak3 also appears to be bound to NPM-ALK,52,56  and inhibition of Jak3 decreases the tyrosine kinase activity of NPM-ALK.52  In addition, we recently identified the autocrine release of IL-9 by ALK+ ALCL cells as an upstream modulator of the Jak3/Stat3 signaling system in these cells.57  Consistent with the selective inhibition of Jak3, an anti–IL-9 neutralizing antibody was shown to downregulate phosphorylated Jak3 and Stat3 and to decrease NPM-ALK kinase activity.57  It is possible that the interactions between Jak3 and NPM-ALK are similar to the recently described interactions between Src and each of NPM-ALK and Bcr-Abl. Cussac et al58  and Meyn et al59  demonstrated that Src kinases are capable of phosphorylating NPM-ALK and Bcr-Abl, respectively.58,59 

Another mechanism of Stat3 constitutive activation in ALK+ ALCL occurs via downregulation of SH2 domain-containing protein tyrosine phosphatase-1 (Shp1), which is a nontransmembrane tyrosine phosphatase predominantly expressed in hematopoietic cells.60  Shp1 plays important roles in regulating the growth and differentiation of hematopoietic cells. It has also been shown to be a major negative regulator of a wide range of cytokine-mediated signaling pathways.61  Shp1 modulates Jak/Stat signaling by binding to Jak via the SH2-binding motif, and dephosphorylating crucial tyrosine sites on Jak.61  Shp1 also has direct inhibitory effects on NPM-ALK via dephosphorylation of this protein.62  Loss of Shp1 expression is a relatively common defect in hematologic malignancies.63  In a series of ALK+ ALCL tumors derived from children and adult patients, Shp1 expression was lost in 50% and 86% of the tumors, respectively.62,64  Evidence of Shp1 gene methylation was detectable in a significant number of these cases.64  Loss of Shp1 is important to the pathogenesis of NPM-ALK–expressing lymphoma because restoration of Shp1 expression in NPM-ALK–expressing ALCL cell lines decreases Stat3 and Jak3 activation and enhances proteosome degradation of NPM-ALK and Jak3.65,66 

Other negative regulatory systems of Jak/Stat, including the suppressors of cytokine signaling and protein inhibitors of activated Stat, have not been extensively investigated in NPM-ALK–expressing ALCL. Few studies have demonstrated that suppressors of cytokine signaling-3 is highly expressed in NPM-ALK–expressing cell lines and tumors, which might represent a biologic attempt to counterbalance the constitutively active Jak3/Stat3 signaling.67 

Whereas the role of Jak3/Stat3 has been extensively explored in ALK+ ALCL, relatively few studies have addressed the role of other members of the Jak/Stat signaling family. These studies illustrated that in ALK+ ALCL, Jak2 and Stat5 exert mechanistic interactions and pathogenetic effects somehow reminiscent to those executed by Jak3 and Stat3. Nieborowska-Skorska et al68  found that NPM-ALK constitutively activates Stat5 and that this activation is essential to lymphomagenesis. However, the constitutive activation of Stat5 in these cells appears not to be restricted to NPM-ALK. Ruchatz et al69  demonstrated that Jak2 also contributes to the activation of Stat5 in these cells. In addition, Jak2 and NPM-ALK were shown to be physically associated and inhibition of Jak2 by a pharmacologic agent or dominant negative construct decreased NPM-ALK–mediated proliferation.69 

PI3K/Akt

The class-Ia phosphoinositide 3-kinase (PI3K) is a heterodimer comprising a catalytic subunit (p110) and a regulatory subunit (p85).70  Activation of PI3K occurs as a sequel to receptor and nonreceptor tyrosine kinase activation. PI3K phosphorylates phosphatidylinositol-4,5-bisphosphate (PIP2) at the 3′ position on its inositol ring, which leads to the production of the lipid second messenger phosphatidylinositol-3,4,5-triphosphate (PIP3). Thereafter, PIP3 contributes to the recruitment to the plasma membrane of a wide range of downstream targets, including the serine/threonine protein kinase Akt (protein kinase B), via binding to their pleckstrin-homology domains.70  At the plasma membrane, another pleckstrin-homology domain-containing serine/threonine protein kinase, named 3-phosphoinositide–dependent protein kinase-1 phosphorylates Akt on Thr308.71  However, maximal activation of Akt requires additional phosphorylation at Ser473 by 3-phosphoinositide–dependent protein kinase-2.72  The level of PIP3 in the cell is strictly regulated by several lipid phosphatases such as the phosphatase and tensin homologue, which converts PIP3 back to PIP2.

The PI3K/Akt signaling pathway regulates a wide array of cellular processes critical for tumorigenesis, including proliferation, survival, growth, and motility.73  A major antiapoptotic effect of activated Akt resides in its ability to sequester the proapoptotic protein Bad and promote its binding to the cytoplasmic 14-3-3 proteins, which leads to increased free Bcl-XL.74  In various experimental models, activated Akt has also been shown to inhibit the activity of caspase 9, prevent nuclear localization of the Forkhead family of transcription factors (FOXO1 [FKHR], FOXO3a [FKHRL1], and AFX [FOXO4]), decrease transcription of Fas ligand, increase the activity of nuclear factor κB (NF-κB) and promote the degradation of the NF-κB inhibitor, IκB, increase the production of nitric oxide by enhancing the activity of endothelial nitric oxide synthase, and induce activation of the serine/threonine protein kinase mammalian target of rapamycin.73 

The contribution of the PI3K/Akt signaling pathway to human cancer including ALK+ ALCL has been extensively investigated. Bai et al75  and Slupianek et al75  showed that NPM-ALK can activate this pathway. PI3K pharmacologic inhibitors have been shown to induce apoptosis in NPM-ALK–expressing lymphoma cells.75,76  An important finding is that dominant-negative PI3K and Akt mutants suppressed the proliferation of BaF3 cells transfected with NPM-ALK.76  In addition, transfection of NPM-ALK salvages Cos-1 cells from Bad-induced apoptotic cell death.75 

PI3K/Akt signaling appears to induce cell-cycle progression in NPM-ALK–expressing lymphoma cells. This effect is mediated, at least partially, via the downregulation of the cyclin-dependent kinase inhibitor p27.77,78  Gu et al79  showed that Akt activation due to forced expression of NPM-ALK in BaF3 cells induced the phosphorylation and nuclear exclusion of FOXO3a (FKHRL1). This effect was associated with the upregulation of cyclin D2 and the downregulation of p27 and Bim-1.79  In further support of the role of PI3K/Akt signaling in NPM-ALK–expressing lymphoma, it has been recently shown that overexpression of activated Akt by using an adenoviral vector into NPM-ALK–expressing cell lines induces activation of mammalian target of rapamycin signaling proteins.80  Nonetheless, another recent study demonstrated that NPM-ALK can induce the activation of mammalian target of rapamycin signaling pathway primarily via a mitogen-induced extracellular kinase/extracellular signal-regulated kinase–dependent pathway, and to a much lesser extent through the PI3K/Akt signaling.81 

CD30

CD30 is a member of the tumor necrosis factor receptor superfamily.82  The recognition of ALCL as a distinct clinicopathologic entity was primarily based on the consistent expression of CD30 by these tumors,1  although this protein is also detected in activated B- and T-lymphocytes, Reed-Sternberg cells, other types of malignant lymphomas, and rare solid tumors.83  Overexpression of CD30 in malignant lymphoma, including the NPM-ALK–expressing lymphoma, can be attributed to a constitutively active extracellular signal-regulated kinase/mitogen-activated protein kinase (MAPK)/JunB signaling cascade, which maintains high level of activity of the CD30 promoter.84,85  In addition, NPM-ALK has been shown to sustain the expression of CD30 and to be associated with its cytoplasmic domain.85,86  However, other studies showed that NPM-ALK can abrogate CD30 signaling.87  It is possible that the differences between these studies might be attributable to the experimental conditions as well as to the complexity of the biologic roles of CD30.

The physiologic function of CD30 is largely unknown. Depending on the cell type, activation of CD30 either enhances cell proliferation or induces apoptosis and cell growth arrest.88  Despite the similar mechanisms leading to CD30 overexpression in ALCL and Hodgkin lymphoma, the outcome of CD30 signaling differs significantly between the 2 diseases. Activation of CD30 decreases the proliferation and induces cycle arrest and apoptosis of NPM-ALK–expressing cells but not Hodgkin lymphoma cells.86,89,,92  The cell-cycle arrest has been shown to be associated with the expression of p21 and the hypophosphorylation of the retinoblastoma protein.90,91 

Contradictory findings have surrounded the role of NF-κB in CD30-mediated regulation of the cell cycle and apoptotic cell death in ALK+ ALCL. This role appears to be related, at least in part, to the mechanism of CD30 stimulation. Earlier studies demonstrated that activation of CD30 via cross-linking with an anti-CD30 antibody fails to induce NF-κB activation in these cells.86,89  In contrast, a subsequent study showed that an anti-CD30 antibody appears to indirectly activate NF-κB by decreasing IκB levels, which has been associated with upregulation of the cellular inhibitors of apoptosis (cIAP1 and cIAP2).93  A more recent study also showed that CD30 stimulation via interaction with its ligand (CD30L) induces activation of NF-κB in ALK+ ALCL cells.92  This study showed that the biologic outcome of this effect is time-dependent. Short, physiologic periods of stimulation induce apoptosis, and the surviving cells exhibit a high level of activation of both the canonical and alternative NF-κB pathways.92  The same study also demonstrated that longer durations of stimulation of NPM-ALK–expressing cells by CD30L induce upregulation of p21 and cell-cycle arrest, which is predominantly dependent on the activated canonical NF-κB pathway.92 

PLC-γ

Activation of phospholipase C-γ (PLC-γ) through its interaction with receptor tyrosine kinases leads to hydrolysis of phophatidylinositol-4,5-biphosphate (PIP2) to soluble inositol-1,4,5-triphosphate (IP3) and membrane-bound diacylglycerol (DAG). IP3 and DAG act as secondary messengers in cellular signaling transduction.94  Whereas IP3 stimulates the release of Ca2+ from the endoplasmic reticulum to the cytosol, DAG binds and activates the serine/threonine protein kinase C (PKC).95 

Bai et al36  demonstrated that NPM-ALK and PLC-γ are physically associated and identified Tyr664 residue on NPM-ALK as the binding site with PLC-γ. Replacement of Tyr664 with a phenylalanine residue abrogated the transforming potential of NPM-ALK; this finding supports an important role for PLC-γ in NPM-ALK oncogenic signaling.36  However, studies to further dissect the mechanisms by which PLC-γ transduces its mitogenic signaling in ALK+ ALCL are lacking. A recent gene array study showed that PKC is highly expressed in ALK+ ALCL.96  However, this study also showed that PKC is also overexpressed in ALK-negative ALCL cells, implying that PKC overexpression might not be directly associated with NPM-ALK.96 

Grb2/Shc/insulin receptor substrate-1/Ras

Ras is a small GTPase with significant transforming potential through the modulation of MAPK.97  Activation of Ras by upstream tyrosine kinases is mediated via a number of widely expressed SH2 and SH3 domain-containing adaptor proteins such as growth factor receptor–bound protein 2 (Grb2), Src homology and collagen (Shc), and insulin receptor substrate-1.98,99 

Simonitsch et al100  showed that NPM-ALK cooperates with Ras in inducing cellular transformation and that NPM-ALK is not, by itself, sufficient to transform rat embryonic cells without exogenously transfected Ras. In a more recent study, Turner et al101  demonstrated that NPM-ALK can induce the activation of Ras and the phosphorylation of extracellular signal-regulated kinase/MAPK. The physical association between NPM-ALK and Grb2, Shc, or insulin receptor substrate-1 has been documented, and these associations suggest a role for these molecules in transducing the oncogenic effects of NPM-ALK.35,36,102  Nonetheless, NPM-ALK mutants defective in the binding sites of Shc or insulin receptor substrate-1 (Tyr567 and Tyr156, respectively) were able to induce transforming effects. It is of note that the association of these mutants with Grb2 was still observed, which suggests that interaction of NPM-ALK with Grb2, but not Shc or insulin receptor substrate-1, is important for cell transformation.36,102  However, the exact binding site on NPM-ALK to Grb2 has yet to be identified. Regardless, few studies have investigated these signaling mechanisms in NPM-ALK–expressing cells, and thus a definitive role of these proteins has not been completely confirmed. Another caveat lies in the fact that these studies were performed using artificial models in which fibroblasts were transfected with NPM-ALK.

Myc

Myc is a transcription factor with potent oncogenic effects. It was originally identified as the cellular homolog of the product of the v-Myc oncogene of the avian myelocytomatosis virus.103  The Myc gene promoter is targeted by multiple signal transduction pathways including Jak/Stat and Ras/Raf/MAPK. Myc proteins drive the expression of a wide range of genes associated with cell proliferation and death.104  Tumor-associated biologic alterations result in the overexpression of Myc and the deregulation of its target genes. NPM-ALK induces its tumor-stimulatory effects, at least partially, via Myc. Transfection of rat fibroblasts with NPM-ALK results in elevated expression of Myc.105  Raetz et al106  used immunohistochemical staining to report that Myc expression was detectable in all 15 ALK+ ALCL tumors from pediatric patients, but in none of the ALK-negative tumors. The exact role of Myc and its interactions with upstream modifiers, including NPM-ALK, and downstream effectors is an area that needs further investigation.

Src

NPM-ALK associates with and activates the Src kinase pp60src; this association appears to be mediated via Tyr418 of NPM-ALK.58  Most probably, the interaction between NPM-ALK and pp60src is important for NPM-ALK–mediated oncogenesis, because loss of this interaction or direct inhibition of pp60src leads to inhibition of NPM-ALK–mediated proliferation.58  It is of note that pp60src has also been proposed to maintain the phosphorylation of NPM-ALK,58  similar to the recently identified mechanism for Bcr-Abl phosphorylation by Src.59  However, it is not yet clear which candidate targets are downstream of NPM-ALK-pp60src interactions. A recent study identified α-diacylglycerol kinase as a possible downstream target and discovered that its phosphorylation by NPM-ALK is dependent on pp60src.107  More studies are needed to delineate the role of Src kinases in ALK+ ALCL lymphoma. For example, Src is a major activator of Stat3, which is known to have an important role in the pathogenesis of ALK+ ALCL. It will be interesting to find out whether Src is capable of activating Stat3, independent of NPM-ALK, in this lymphoma.

Hsp90

Bonvini et al108  reported that pharmacologic blockade of heat shock protein (Hsp) 90 mediates effective apoptosis in ALK+ ALCL cell lines. This study highlighted the fact that NPM-ALK is unstable, and it undergoes proteosome degradation relatively rapidly. Most probably, Hsp90 protects NPM-ALK by slowing down its proteosome degradation. Targeting Hsp90 prevents its complex formation with NPM-ALK, probably by promoting the association of the latter with Hsp70 chaperone in an E3 ubiquitin ligase carboxyl terminus Hsp70-interacting protein (CHIP)–dependent manner.109 

SNT/FRS2

Suc1- and Suc2-associated neurotrophic factor-induced phosphorylated target/fibroblast receptor substrates (SNT-1/FRS2α and SNT-2/FRS2β) are membrane-anchored docking proteins. Evidence supports a role for SNT-1 and SNT-2 in mediating signaling from receptors such as fibroblast growth factor receptor and the nerve growth factor receptor TrkA to Ras and MAPK.110  Recently, Chikamori et al111  demonstrated by the immunoprecipitation technique that NPM-ALK is physically associated with SNT-1 and SNT-2. This association was also detected when kinase-negative NPM-ALK mutant was used. The interaction between NPM-ALK and these adaptor proteins occurred via Tyr156, Tyr567, and a 19-amino-acid sequence of NPM-ALK. When these binding sites were mutated, the transforming potential of NPM-ALK was markedly diminished. A surprising finding was that these mutant constructs were still capable of binding molecules previously shown to contribute to the oncogenic effects of NPM-ALK, including PLC-γ and PI3K. This observation indicates that the contribution of these molecules may not be as important as was previously thought or that there is redundancy in the effects of the different oncogenic signaling pathways interacting with NPM-ALK. This area still requires more investigation.

NIPA

Using a yeast 2-hybrid screen, Ouyang et al112  identified the nuclear interacting partner of anaplastic lymphoma kinase (NIPA) as a novel NPM-ALK–interacting protein. NIPA is widely expressed in human tissues and it contains a nuclear localization signal in its carboxyl terminus. The interaction between NIPA and ALK is not limited to NPM-ALK, because other ALK chimeric proteins can also induce NIPA phosphorylation.112  NPM-ALK induces the phosphorylation of NIPA at several serine and tyrosine residues, particularly Ser354. Most probably, NIPA plays a role in NPM-ALK–mediated oncogenic effects by inhibiting apoptotic cell death. Mutations of the nuclear translocation signal or the Ser354 phosphorylation site block the antiapoptotic effects of NIPA. A more recent study identified an important role of NIPA in controlling mitotic entry and the cell cycle.113  In light of these findings, further analysis of the role of NIPA in NPM-ALK–mediated oncogenic effects is needed.

p130Cas

The mechanisms by which NPM-ALK induces its transforming effects have been relatively well characterized. Less is known about its effects on the cell shape. Transfection of NPM-ALK into fibroblasts or lymphoma cells induces notable morphologic changes.87,114  Depending on the cell type, NPM-ALK induces morphologic changes that make the cells similar to ALCL cells or induces neurite outgrowth.87,114  Using mass spectrometry–based screening of proteins interacting with NPM-ALK and involved in the cytoskeleton morphology, Ambrogio et al115  identified p130 Crk–associated substrate (p130Cas) protein and demonstrated that NPM-ALK is able to bind and phosphorylate this protein. They also showed that p130Cas plays a role in NPM-ALK–mediated modification and transformation of the cell shape.115 

Despite significant progress in delineating the molecular mechanisms of ALK+ ALCL, the therapeutic approaches to this lymphoma have not changed significantly. The current treatment is based on doxorubicin-containing combination chemotherapy.33,116  This treatment induces complete remission in up to 95% of the patients, but relapse and resistance occur in more than 40% of the cases.116,117  The exact cause of the high rate of relapse is not known. It has been suggested that a high age-related International Prognostic Index is a reliable indicator for a worse clinical outcome.32,33  It is also possible that ALK+ ALCL patients with poor clinical outcome possess specific biologic factors that affect the outcome of their disease. For example, even though Bcl-2 is infrequently expressed in ALK+ ALCL cells,118  these cells still express high levels of Mcl-1.119  In addition, although ALK+ ALCL cells have a high apoptotic rate,118  some tumor cells may remain dormant and later become the origin of relapses. Furthermore, ALK+ ALCL patients with tumors expressing CD56 or survivin have been shown to have unfavorable clinical outcome.120,121  Similarly, ALK+ ALCL patients with high levels of c-Jun activation binding protein-1 and low levels of p27 have been shown to demonstrate worse clinical outcome.122  In addition, ALK+ ALCL patients with tumors expressing the retinoblastoma protein show a trend, albeit not statistically significant, for a less favorable clinical course.123  One possible explanation is that the tumors that express the retinoblastoma protein demonstrate a lower apoptotic index.123  Recently, Lamant et al124  used gene-expression profiling to show that ALK+ ALCL primary tumors demonstrate significantly up-regulated CEBPB, BCL6, PTPN12, and SERPINA1 genes compared with tumors that lack ALK. The potential significance of CEBPB in NPM-ALK–expressing lymphoma has also been stressed by other investigators.125,126  The relationship between these observations and patient's survival and response to therapy still needs to be investigated.

The fact that NPM-ALK plays a central role in the development and pathogenesis of ALK+ ALCL tumors makes this chimeric protein a legitimate therapeutic target in this disease. Because of its repeatedly documented oncogenic potential and its expression in this type of malignant lymphoma, specific or selective targeting of NPM-ALK will probably be associated with less toxic effects, as compared with targeting other signaling partners that might be involved in maintaining the biologic functions of the nonneoplastic cells. Selective targeting of Bcr-Abl, a chimeric protein with constitutive tyrosine kinase activity and biologic features similar to NPM-ALK, has been used to treat chronic myeloid leukemia and other neoplastic diseases.127  Several recent studies have demonstrated that specific targeting of NPM-ALK may represent a promising therapeutic modality for ALK+ ALCL.128,,,132  Indirect inhibition of NPM-ALK can also be achieved by inhibitors of Hsp90 such as herbimycin A and 17-AAG.108,133  Nonetheless, similar to Bcr-Abl, resistance to agents selectively targeting NPM-ALK is a real possibility, and therapeutic approaches based on combination or alternative agents will most probably be needed.

With our understanding of the pathobiology of NPM-ALK, blockade of other oncogenic systems interacting with NPM-ALK could be used separately or in conjunction with therapeutic approaches targeting NPM-ALK. Two of the more studied systems in this lymphoma are Jak/Stat and PI3K/Akt. These 2 pathways have recently been proposed to be the focus for the development of new pharmacologic agents to antagonize their effects in malignant diseases including NPM-ALK–expressing lymphoma.47,49,52,75,76,78  Another approach to targeting these systems is by blocking upstream cytokines and growth factors that induce their activation.57 

Immunotherapy is another possible approach to treating ALK+ ALCL. In vitro and in vivo studies showed that anti-CD30 antibodies induce ALK+ ALCL apoptotic cell death and tumor regression.134,135  Other studies have suggested that CD26 might represent a promising immunotherapeutic target.136  Finally, gene therapy also may represent a promising future therapeutic modality for ALK+ ALCL. Adenoviral-mediated transfer of p16, p21, p27, and p53 induces apoptosis and cell-cycle arrest in ALK+ ALCL cell lines and tumors implanted in nude mice.137,138  In addition, the transfer of Shp1 suppresses NPM-ALK, Jak3, and Stat3 in ALK+ ALCL cells.66 

We have briefly discussed the pathobiology of ALK+ ALCL and emphasized the role of NPM-ALK in this disease. Despite being relatively uncommon, ALK+ ALCL has become an excellent study model for cancer. Over the past 13 years, accumulated data have illustrated and highlighted the collective role of oncoproteins, tumor suppressors, adaptor proteins, and other molecules in the development and progression of ALK+ ALCL. Despite the central role of NPM-ALK in the pathogenesis of this lymphoma, the model emerging from the studies on ALK+ ALCL stresses the collaboration of several members of a molecular network rather than individual culprits. This molecular network transforms ALK+ ALCL into a well-defined clinicopathologic entity. The configuration of this molecular network is most probably a multistep process, which is a well-known characteristic of oncogenesis. The current model provides a rationale for designing therapeutic approaches that can specifically disrupt multiple targets to eliminate the synergistic effects of the various members of the molecular network. As our understanding of the biology of ALK+ ALCL continues to evolve, it is highly anticipated that the knowledge gained from the studies of this lymphoma will be applicable to other neoplastic diseases.

The authors are grateful to Elizabeth Hess and Kim-Anh Vu for their valuable help with the preparation of this manuscript. The authors apologize to the laboratories whose contributions to this field could not be discussed or cited because of space limitation.

This work was supported in part by CA114395 grant from the National Institutes of Health (NIH) and by the Physician Scientist Program Award and an Institutional Research Grant from M. D. Anderson Cancer Center to H.M.A.; and by grants from the National Cancer Institute of Canada, Alberta Cancer Foundation, and the Canadian Institute for Health Research awarded to R.L.

National Institutes of Health

Contribution: H.M.A. and R.L. wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Hesham M. Amin, Department of Hematopathology, Unit 72, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030; e-mail:hamin@mdanderson.org; or Raymond Lai, Cross Cancer Institute, Room 2342, 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada; e-mail: raymondl@cancerboard.ab.ca.

1
Stein
H
Mason
DY
Gerdes
J
et al
The expression of the Hodgkin's disease associated antigen Ki-1 in reactive and neoplastic lymphoid tissue: evidence that Reed-Sternberg cells and histiocytic malignancies are derived from activated lymphoid cells.
Blood
1985
66
848
858
2
Schwab
U
Stein
H
Gerdes
J
et al
Production of a monoclonal antibody specific for Hodgkin and Sternberg-Reed cells of Hodgkin's disease and a subset of normal cells.
Nature
1982
299
65
67
3
Herbst
H
Tippelmann
G
Anagnostopoulos
I
et al
Immunoglobulin and T-cell receptor gene rearrangements in Hodgkin's disease and Ki-1-positive anaplastic large cell lymphoma: dissociation between phenotype and genotype.
Leuk Res
1989
13
103
116
4
Stein
H
Foss
H-D
Durkop
H
et al
CD30+ anaplastic large cell lymphoma: a review of its histopathologic, genetic, and clinical features.
Blood
2000
96
3681
3695
5
Kadin
ME
Sako
D
Berliner
N
Childhood Ki-1 lymphoma presenting with skin lesions and peripheral lymphadenopathy.
Blood
1986
68
1042
1049
6
Morgan
R
Hecht
BK
Sandberg
AA
Hecht
F
Smith
SD
Chromosome 5q35 breakpoint in malignant histiocytosis.
New Engl J Med
1986
314
1322
7
Rimokh
R
Magaud
J-P
Berger
R
et al
A translocation involving a specific breakpoint (q35) on chromosome 5 is characteristic of anaplastic large cell lymphoma (‘Ki-1 lymphoma').
Br J Haematol
1989
71
31
36
8
Le Beau
MM
Bitter
MA
Larson
RA
et al
The t(2;5)(p23;q35): a recurring chromosomal abnormality in Ki-1-positive anaplastic large cell lymphoma.
Leukemia
1989
3
866
870
9
Morris
SW
Kirstein
MN
Valentine
MB
et al
Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma.
Science
1994
263
1281
1284
10
Shiota
M
Fujimoto
J
Semba
T
Satoh
H
Yamamoto
T
Mori
S
Hyperphosphorylation of a novel 80 kDa protein-tyrosine kinase similar to Ltk in a human Ki-1 lymphoma cell line, AMS3.
Oncogene
1994
9
1567
1574
11
Shiota
M
Fujimoto
J
Takenaga
M
et al
Diagnosis of t(2;5)(p23;q35)-associated Ki-1 lymphoma with immunohistochemistry.
Blood
1994
84
3648
3652
12
Delsol
G
Ralfkiaer
E
Stein
H
Wright
D
Jaffe E
S
Jaffe
ES
Harris
NL
Stein
H
Vardiman
JW
Anaplastic large cell lymphoma.
Pathology and genetics of tumors of haematopoietic and lymphoid tissues: World Health Organization Classification of Tumours
2001
Lyon, France
IARC Press
230
235
13
Pulford
K
Morris
SW
Turturro
F
Anaplastic lymphoma kinase proteins in growth control and cancer.
J Cell Physiol
2004
199
330
358
14
Falini
B
Pulford
K
Pucciarini
A
et al
Lymphomas expressing ALK fusion protein(s) other than NPM-ALK.
Blood
1999
94
3509
3515
15
Iwahara
T
Fujimoto
J
Wen
D
et al
Molecular characterization of ALK, a receptor tyrosine kinase expressed specifically in the nervous system.
Oncogene
1997
14
439
449
16
Morris
SW
Naeve
C
Mathew
P
et al
ALK, the chromosome 2 gene locus altered by the t(2;5) in non-Hodgkin's lymphoma, encodes a novel neural receptor tyrosine kinase that is highly related to leukocyte tyrosine kinase (LTK).
Oncogene
1997
14
2175
2188
17
Lorén
CE
Englund
C
Grabbe
C
Hallberg
B
Hunter
T
Palmer
RH
A crucial role for the Anaplastic lymphoma kinase receptor tyrosine kinase in gut development in Drosophila melanogaster.
EMBO Rep
2003
4
781
786
18
Falini
B
Bigerna
B
Fizzotti
M
et al
ALK expression defines a distinct group of T/null lymphomas (“ALK lymphomas”) with a wide morphological spectrum.
Am J Pathol
1998
153
875
886
19
Lamant
L
Pulford
K
Bischof
D
et al
Expression of the ALK tyrosine kianse gene in neuroblastoma.
Am J Pathol
2000
156
1711
1721
20
Delsol
G
Lamant
L
Mariame
B
et al
A new subtype of large B-cell lymphoma expressing the ALK kinase and lacking the 2;5 translocation.
Blood
1997
89
1483
1490
21
Trümper
L
Pfreundschuh
M
Bonin
FV
Daus
H
Detection of the t(2;5)-associated NPM/ALK fusion cDNA in peripheral blood cells of healthy individuals.
Br J Haematol
1998
103
1138
1144
22
Maes
B
Vanhentenrijk
V
Wlodarska
I
et al
The NPM-ALK and the ATIC-ALK fusion genes can be detected in non-neoplastic cells.
Am J Pathol
2001
158
2185
2193
23
Gascoyne
RD
Lamant
L
Martin-Subero
JI
et al
ALK-positive diffuse large B-cell lymphoma is associated with Clathrin-ALK rearrangements: report of 6 cases.
Blood
2003
102
2568
2573
24
Onciu
M
Behm
FG
Downing
JR
et al
ALK-positive plasmablastic B-cell lymphoma with expression of the NPM-ALK fusion transcript: report of 2 cases.
Blood
2003
102
2642
2644
25
Griffin
CA
Hawkins
AL
Dvorak
C
Henkle
C
Ellingham
T
Perlman
EJ
Recurrent involvement of 2p23 in inflammatory myofibroblastic tumors.
Cancer Res
1999
59
2776
2780
26
Lawrence
B
Perez-Atayde
A
Hibbard
MK
et al
TPM3-ALK and TPM4-ALK oncogenes in inflammatory myofibroblastic tumors.
Am J Pathol
2000
157
377
384
27
Lee
HH
Norris
A
Weiss
JB
Frasch
M
Jelly belly protein activates the receptor tyrosine kinase Alk to specify visceral muscle pioneers.
Nature
2003
425
507
512
28
Englund
C
Loren
CE
Grabbe
C
et al
Jeb signals through the Alk receptor tyrosine kinase to drive visceral muscle fusion.
Nature
2003
425
512
516
29
Stoica
GE
Kuo
A
Aigner
A
et al
Identification of anaplastic lymphoma kinase as a receptor for the growth factor pleiotrophin.
J Biol Chem
2001
276
16772
16779
30
Stoica
GE
Kuo
A
Powers
C
et al
Midkine binds to anaplastic lymphoma kinase (ALK) and acts as a growth factor for different cell types.
J Biol Chem
2002
277
35990
35999
31
Moog-Lutz
C
Degoutin
J
Gouzi
JY
et al
Activation and inhibition of anaplastic lymphoma kinase receptor by monoclonal antibodies and absence of agonist activity of pleiotrophin.
J Biol Chem
2005
280
26039
26048
32
Gascoyne
R
Aoun
P
Wu
D
et al
Prognostic significance of anaplastic lymphoma kinase (ALK) protein expression in adults with anaplastic large cell lymphoma.
Blood
1999
93
3913
3921
33
Falini
B
Pileri
S
Zinzani
PL
et al
ALK+ lymphoma: clinico-pathological findings and outcome.
Blood
1999
93
2697
2706
34
Borer
RA
Lehner
CF
Eppenberger
HM
Nigg
EA
Major nucleolar proteins shuttle between nucleus and cytoplasm.
Cell
1989
56
379
390
35
Bischof
D
Pulford
K
Mason
DY
Morris
SW
Role of the nucleophosmin (NPM) portion of the non-Hodgkin's lymphoma-associated NPM-anaplastic lymphoma kinase fusion protein in oncogenesis.
Mol Cell Biol
1997
17
2312
2325
36
Bai
R-Y
Dieter
P
Peschel
C
Morris
SW
Duyster
J
Nucelophosmin-anaplastic lymphoma kinase of large-cell anaplastic lymphoma is a constitutively active tyrosine kinase that utilizes phospholipase C-γ to mediate its mitogenicity.
Mol Cell Biol
1998
18
6951
6961
37
Kuefer
MU
Look
AT
Pulford
K
et al
Retrovirus-mediated gene transfer of NPM-ALK causes lymphoid malignancy in mice.
Blood
1997
90
2901
2910
38
Miething
C
Grundler
R
Fend
F
et al
The oncogenic fusion protein nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) induces two distinct malignant phenotypes in a murine retroviral transplantation model.
Oncogene
2003
22
4642
4647
39
Chiarle
R
Gong
JZ
Guasparri
I
et al
NPM-ALK transgenic mice spontaneously develop T-cell lymphomas and plasma cell tumors.
Blood
2003
101
1919
1927
40
Lange
K
Uckert
W
Blankenstein
T
et al
Overexpression of NPM-ALK induces different types of malignant lymphomas in IL-9 transgenic mice.
Oncogene
2003
22
517
527
41
Turner
SD
Tooze
R
Maclennan
K
Alexander
DR
Vav-promoter regulated oncogenic fusion protein NPM-ALK in transgenic mice causes B-cell lymphomas with hyperactive Jun kinase.
Oncogene
2003
22
7750
7761
42
Turner
SD
Merz
H
Yeung
D
Alexander
DR
CD2 promoter regulated nucleophosmin-anaplastic lymphoma kinase in transgenic mice causes B lymphoid malignancy.
Anticancer Res
2006
26
3275
3279
43
Levy
DE
Darnell
JE
Jr
STATs: transcriptional control and biological impact.
Nat Rev Mol Cell Biol
2002
3
651
662
44
Bromberg
J
Darnell
JE
Jr
The role of STATs in transcriptional control and their impact on cellular function.
Oncogene
2000
19
2468
2473
45
Bromberg
JF
Wrzeszczynska
MH
Devgan
G
et al
Stat3 as an oncogene.
Cell
1999
98
295
303
46
Khoury
JD
Medeiros
LJ
Rassidakis
GZ
et al
Differential expression and clinical significance of tyrosine-phosphorylated STAT3 in ALK+ and ALK− anaplastic large cell lymphoma.
Clin Cancer Res
2003
9
3692
3699
47
Chiarle
R
Simmons
WJ
Cai
H
et al
Stat3 is required for ALK-mediated lymphomagenesis and provides a possible therapeutic target.
Nat Med
2005
11
623
629
48
Kasprzycka
M
Marzec
M
Liu
X
Zhang
Q
Wasik
MA
Nucleophosmin/anaplastic lymphoma kinase (NPM/ALK) oncoprotein induces the T regulatory cell phenotype by activating STAT3.
Proc Natl Acad Sci USA
2006
103
9964
9969
49
Amin
HM
McDonnell
TJ
Ma
Y
et al
Selective inhibition of STAT3 induces apoptosis and G1 cell cycle arrest in ALK-positive anaplastic large cell lymphoma.
Oncogene
2004
23
5426
5434
50
Zhang
Q
Raghunath
PN
Xue
L
et al
Multilevel dysregulation of STAT3 activation in anaplastic lymphoma kinase-positive T/null-cell lymphoma.
J Immunol
2002
168
466
474
51
Zamo
A
Chiarle
R
Piva
R
et al
Anaplastic lymphoma kinase (ALK) activates Stat3 and protects hematopoietic cells from cell death.
Oncogene
2002
21
1038
1047
52
Amin
HM
Medeiros
LJ
Ma
Y
et al
Inhibition of JAK3 induces apoptosis and decreases anaplastic lymphoma kinase activity in anaplastic large cell lymphoma.
Oncogene
2003
22
5399
5407
53
Lai
R
Rassidakis
GZ
Lin
Q
Atwell
C
Medeiros
LJ
Amin
HM
Jak3 activation is significantly associated with ALK expression in anaplastic large cell lymphoma.
Hum Pathol
2005
36
939
944
54
Johnston
JA
Kawamura
M
Kirken
RA
et al
Phosphorylation and activation of Jak-3 Janus kinase in response to interleukin-2.
Nature
1994
370
151
153
55
Witthuhn
BA
Silvennoinen
O
Miura
O
et al
Involvement of the Jak-3 kinase in signaling by interleukins 2 and 4 in lymphoid and myeloid cells.
Nature
1994
370
153
157
56
Crockett
DK
Lin
Z
Elenitoba-Johnson
KS
Lim
MS
Identification of NPM-ALK interacting proteins by tandem mass spectrometry.
Oncogene
2004
23
2617
2629
57
Qiu
L
Lai
R
Lin
Q
et al
Autocrine release of interleukin-9 promotes Jak3-dependent survival of ALK+ anaplastic large-cell lymphoma.
Blood
2006
108
2407
2415
58
Cussac
D
Greenland
C
Roche
S
et al
Nucleophosmin-anaplastic lymphoma kinase of anaplastic large-cell lymphoma recruits, activates, and uses pp60c−src to mediate its mitogenicity.
Blood
2004
103
1464
1471
59
Meyn
MA
Wilson
MB
Abdi
FA
et al
Src family kinases phosphorylate the Bcr-Abl SH3-SH2 region and modulate Bcr-Abl transforming activity.
J Biol Chem
2006
281
30907
30916
60
Mizuno
K
Katagiri
T
Hasegawa
K
Ogimoto
M
Yakura
H
Hematopoietic cell phosphatase, SHP-1, is constitutively associated with the SH2 domain-containing leukocyte protein, SLP-76, in B cells.
J Exp Med
1996
184
457
463
61
Klingmuller
U
Lorenz
U
Cantley
LC
Neel
BG
Lodish
HF
Specific recruitment of SH-PTP1 to the erythropoietin receptor causes inactivation of JAK2 and termination of proliferative signals.
Cell
1995
80
729
738
62
Honorat
J-F
Ragab
A
Lamant
L
Delsol
G
Ragab-Thomas
J
SHP1 tyrosine phosphatase negatively regulates NPM-ALK tyrosine kinase signaling.
Blood
2006
107
4130
4138
63
Oka
T
Yoshino
T
Hayashi
K
et al
Reduction of hematopoietic cell-specific tyrosine phosphatase SHP-1 gene expression in natural killer cell lymphoma and various types of lymphomas/leukemias: combination analysis with cDNA expression array and tissue microarray.
Am J Pathol
2001
159
1495
1505
64
Khoury
JD
Rassidakis
GZ
Medeiros
LJ
Amin
HM
Lai
R
Methylation of SHP1 gene and loss of SHP1 protein expression are frequent in systemic anaplastic large cell lymphoma.
Blood
2004
104
1580
1581
65
Han
Y
Amin
HM
Frantz
C
et al
Restoration of shp1 expression by 5-AZA-2′-deoxycytidine is associated with downregulation of JAK3/STAT3 signaling in ALK-positive anaplastic large cell lymphoma.
Leukemia
2006
20
1602
1609
66
Han
Y
Amin
HM
Franko
B
Frantz
C
Shi
X
Lai
R
Loss of SHP1 enhances JAK3/STAT3 signaling and decreases proteosome degradation of JAK3 and NPM-ALK in ALK+ anaplastic large-cell lymphoma.
Blood
2006
108
2796
2803
67
Cho-Vega
JH
Rassidakis
GZ
Amin
HM
et al
Suppressor of cytokine signaling 3 expression in anaplastic large cell lymphoma.
Leukemia
2004
18
1872
1878
68
Nieborowska-Skorska
M
Slupianek
A
Xue
L
et al
Role of signal transducer and activator of transcription 5 in nucleophosmin/anaplastic lymphoma kinase-mediated malignant transformation of lymphoid cells.
Cancer Res
2001
61
6517
6523
69
Ruchatz
H
Couccia
AM
Stano
P
Marchesi
E
Gambacorti-Passerini
C
Constitutive activation of Jak2 contributes to proliferation and resistance to apoptosis in NPM/ALK-transformed cells.
Exp Hematol
2003
31
309
315
70
Cantley
LC
The phosphoinositide 3-kinase pathway.
Science
2002
296
1655
1657
71
Stokoe
D
Stephens
LR
Copeland
T
et al
Dual role of phosphatidylinositol-3,4,5-triphosphate in the activation of protein kinase B.
Science
1997
277
567
570
72
Alessi
DR
James
SR
Downes
CP
et al
Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Bα.
Curr Biol
1997
7
261
269
73
Vivanco
I
Sawyers
CL
The phosphatidylinositol 3-kinase-AKT pathway in human cancer.
Nat Rev Cancer
2002
2
489
501
74
Datta
SR
Dudek
H
Tao
X
et al
Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery.
Cell
1997
91
231
241
75
Bai
R-Y
Ouyang
T
Miething
C
Morris
SW
Peschel
C
Duyster
J
Nucleophosmin-anaplastic lymphoma kinase associated with anaplastic large-cell lymphoma activates the phosphatidylinositol 3-kinase/Akt antiapoptotic pathway.
Blood
2000
96
4319
4327
76
Slupianek
A
Nieborowska-Skorska
M
Hoser
G
et al
Role of phosphatidylinositol 3-kinase-Akt pathway in nucleophosmin/anaplastic lymphoma kinase-mediated lymphomagenesis.
Cancer Res
2001
61
2194
2199
77
Slupianek
A
Skorski
T
NPM/ALK downregulates p27Kip1 in a PI-3K-dependent manner.
Exp Hematol
2004
32
1265
1271
78
Rassidakis
GZ
Feretzaki
M
Atwell
C
et al
Inhibition of Akt increases p27Kip1 levels and induces cell cycle arrest in anaplastic large cell lymphoma.
Blood
2005
105
827
829
79
Gu
T-L
Tothova
Z
Scheijen
B
Griffin
JD
Gilliland
DG
Sternberg
DW
NPM-ALK fusion kinase of anaplastic large-cell lymphoma regulates survival and proliferative signaling through modulation of FOXO3a.
Blood
2004
103
4622
4629
80
Vega
F
Medeiros
LJ
Leventaki
V
et al
Activation of mammalian target of rapamycin signaling pathway contributes to tumor cell survival in anaplastic lymphoma kinase-positive anaplastic large cell lymphoma.
Cancer Res
2006
66
6589
6597
81
Marzec
M
Kasprzycka
M
Liu
X
et al
Oncogenic tyrosine kinase NPM/ALK induces activation of the rapamycin-sensitive mTOR signaling pathway.
Oncogene
2007
26
5606
5614
82
Smith
CA
Farrah
T
Goodwin
RG
The TNF receptor superfamily of cellular and viral proteins: Activation, costimulation, and death.
Cell
1994
76
959
962
83
Falini
B
Pileri
S
Pizzolo
G
et al
CD30 (Ki-1) molecule: a new cytokine receptor of the tumor necrosis factor receptor superfamily as a tool for diagnosis and immunotherapy.
Blood
1995
85
1
14
84
Watanabe
M
Sasaki
M
Itoh
K
et al
JunB induced by constitutive CD30-extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase signaling activates the CD30 promoter in anaplastic large cell lymphoma and Reed-Sternberg cells of Hodgkin lymphoma.
Cancer Res
2005
65
7628
7634
85
Hsu
FY-Y
Johnston
PB
Burke
KA
Zhao
Y
The expression of CD30 in anaplastic large cell lymphoma is regulated by nucelophosmin-anaplastic lymphoma kinase-mediated JunB level in a cell type-specific manner.
Cancer Res
2006
66
9002
9008
86
Hübinger
G
Scheffrahn
I
Muller
E
et al
The tyrosine kinase NPM-ALK, associated with anaplastic large cell lymphoma, binds the intracellular domain of the surface receptor CD30 but is not activated by CD30 stimulation.
Exp Hematol
1999
27
1796
1805
87
Horie
R
Watanabe
M
Ishida
T
et al
The NPM-ALK oncoprotein abrogates CD30 signaling and constitutive NF-κB activation in anaplastic large cell lymphoma.
Cancer Cell
2004
5
353
364
88
Gruss
HJ
Boiani
N
Williams
DE
Armitage
RJ
Smith
CA
Goodwin
RG
Pleiotropic effects of CD30 ligand on CD30-expressing cells and lymphoma cell lines.
Blood
1994
83
2045
2056
89
Mir
SS
Richter
BWM
Duckett
CS
Differential effects of CD30 activation in anaplastic large cell lymphoma and Hodgkin disease cells.
Blood
2000
96
4307
4312
90
Hübinger
G
Muller
E
Scheffrahn
I
et al
CD30-mediated cell cycle arrest associated with induced expression of p21CIP1/WAF1 in the anaplastic large cell lymphoma cell line Karpas 299.
Oncogene
2001
20
590
598
91
Levi
E
Pfeifer
WM
Kadin
ME
CD30-activation-mediated growth inhibition of anaplastic large-cell lymphoma cell lines: apoptosis or cell-cycle arrest?
Blood
2001
98
1630
1631
92
Wright
CW
Rumble
JM
Duckett
CS
CD30 activates both the canonical and alternative NF-κB pathways in anaplastic large cell lymphoma cells.
J Biol Chem
2007
282
10252
10262
93
Hübinger
G
Schneider
C
Stohr
D
et al
CD30-induced up-regulation of the inhibitor of apoptosis genes cIAP1 and cIAP2 in anaplastic large cell lymphoma cells.
Exp Hematol
2004
32
382
389
94
Berridge
MJ
Irvine
RF
Inositol triphosphate, a novel second messenger in cellular signal transduction.
Nature
1982
312
315
321
95
Nishizuka
Y
The role of protein kinase C in cell surface signal transduction and tumour promotion.
Nature
1984
308
693
698
96
Thompson
MA
Stumph
J
Henrickson
SE
et al
Differential gene expression in anaplastic lymphoma kinase-positive and anaplastic lymphoma kinase-negative anaplastic large cell lymphomas.
Hum Pathol
2005
36
494
504
97
Feramisco
JR
Gross
M
Kamata
T
Rosenberg
M
Sweet
RW
Microinjection of the oncogene form of the human H-ras (T-24) protein results in rapid proliferation of quiescent cells.
Cell
1984
38
109
117
98
Rozakis-Adcock
M
McGlade
J
Mbamalu
G
et al
Association of the Shc and Grb2/Sem5 SH2-containing proteins is implicated in activation of the Ras pathway by tyrosine kinases.
Nature
1992
360
689
692
99
Skolnik
EY
Lee
CH
Batzer
A
et al
The SH2/SH3 domain-containing protein GRB2 interacts with tyrosine-phosphorylated IRS1 and Shc: implications for insulin control of ras signaling.
EMBO J
1993
12
1929
1936
100
Simonitsch
I
Polgar
D
Hajek
M
et al
The cytoplasmic truncated receptor tyrosine kinase ALK homodimer immortalizes and cooperates with ras in cellular transformation.
FASEB J
2001
15
1416
1418
101
Turner
SD
Yeung
D
Hadfield
K
Cook
SJ
Alexander
DR
The NPM-ALK tyrosine kinase mimics TCR signalling pathways, inducing NFAT and AP-1 by RAS-dependent mechanisms.
Cell Signal
2007
19
740
747
102
Fujimoto
J
Shiota
M
Iwahara
T
et al
Characterization of the transforming activity of p80, a hyperphosphorylated protein in a Ki-1 lymphoma cell line with chromosomal translocation t(2;5).
Proc Natl Acad Sci USA
1996
93
4181
4186
103
Vennstrom
B
Sheiness
D
Zabielski
J
Bishop
JM
Isolation and characterization of c-myc, a cellular homolog of the oncogene (v-myc) of avian myelocytomatosis virus strain 29.
J Virol
1982
42
773
779
104
Pelengaris
S
Khan
M
Evan
G
c-MYC: more than just a matter of life and death.
Nat Rev Cancer
2002
2
764
776
105
Wellmann
A
Doseeva
V
Butscher
W
et al
The activated anaplastic lymphoma kinase increases cellular proliferation and oncogene up-regulation in rat 1a fibroblasts.
FASEB J
1997
11
965
972
106
Raetz
EA
Perkins
SL
Carlson
MA
Schooler
KP
Carroll
WL
Virshup
DM
The nucleophosmin-anaplastic lymphoma kinase fusion protein induces c-Myc expression in pediatric anaplastic large cell lymphomas.
Am J Pathol
2002
161
875
883
107
Bacchiocchi
R
Baldanzi
G
Carbonari
D
et al
Activation of α–diacylglycerol kinase is critical for the mitogenic properties of anaplastic lymphoma kinase.
Blood
2005
106
2175
2182
108
Bonvini
P
Gastaldi
T
Falini
B
Rosolen
A
Nucleophosmin-anaplastic lymphoma kinase (NPM-ALK), a novel Hsp90-client tyrosine kinase: down-regulation of NPM-ALK expression and tyrosine phosphorylation in ALK+ CD30+ lymphoma cells by Hsp90 antagonist 17-allylamino,17-demethoxygeldanamycin.
Cancer Res
2002
62
1559
1566
109
Bonvini
P
Rosa
HD
Vignes
N
Rosolen
A
Ubiquitination and proteasomal degradation of nucleophosmin-anaplastic lymphoma kinase induced by 17-allylamino-demethoxygeldanamycin: role of the co-chaperon carboxyl heat shock protein 70-interacting protein.
Cancer Res
2004
64
3256
3264
110
Ong
SH
Guy
GR
Hadari
YR
et al
FRS2 proteins recruit intracellular signaling pathways by binding to diverse targets on fibroblast growth factor and nerve growth factor receptors.
Mol Cell Biol
2000
20
979
989
111
Chikamori
M
Fujimoto
J
Tokai-Nishizumi
N
Yamamoto
T
Identification of multiple SNT-binding sites on NPM-ALK oncoprotein and their involvement in cell transformation.
Oncogene
2007
26
2950
2954
112
Ouyang
T
Bai
R-Y
Bassermann
F
et al
Identification and characterization of a nuclear interacting partner of anaplastic lymphoma kinase (NIPA).
J Biol Chem
2003
278
30028
30036
113
Bassermann
F
von Klitzing
C
Munch
S
et al
NIPA defines an SCF-type mammalian E3 ligase that regulates mitotic entry.
Cell
2005
122
45
57
114
Souttou
B
Carvalho
NB
Raulais
D
Vigny
M
Activation of anaplastic lymphoma kinase receptor tyrosine kinase induces neuronal differentiation through the mitogen-activated protein kinase pathway.
J Biol Chem
2001
276
9526
9531
115
Ambrogio
C
Voena
C
Manazza
AD
et al
p130Cas mediates the transforming properties of the anaplastic lymphoma kinase.
Blood
2005
106
3907
3916
116
Brugières
L
Le Deley
MC
Pacquement
H
et al
CD30+ anaplastic large-cell lymphoma in children: analysis of 82 patients enrolled in two consecutive studies of the French Society of Pediatric Oncology.
Blood
1998
92
3591
3598
117
Brugieres
L
Quartier
P
Le Deley
MC
et al
Relapses of childhood anaplastic large-cell lymphoma: treatment results in a series of 41 children – a report from the French Society of Pediatric Oncology.
Ann Oncol
2000
11
53
58
118
Rassidakis
GZ
Sarris
AH
Herling
M
et al
Differential expression of BCL-2 family proteins in ALK-positive and ALK-negative anaplastic large cell lymphoma of T/null-cell lineage.
Am J Pathol
2001
159
527
535
119
Rust
R
Harms
G
Blokziji
T
et al
High expression of Mcl-1 in ALK positive and negative anaplastic large cell lymphoma.
J Clin Pathol
2005
58
520
524
120
Suzuki
R
Kagami
Y
Takeuchi
K
et al
Prognostic significance of CD56 expression for ALK-positive and ALK-negative anaplastic large-cell lymphoma of T/null cell phenotype.
Blood
2000
96
2993
3000
121
Schlette
EJ
Medeiros
LJ
Goy
A
Lai
R
Rassidakis
GZ
Survivin expression predicts poorer prognosis in anaplastic large-cell lymphoma.
J Clin Oncol
2004
22
1682
1688
122
Rassidakis
GZ
Claret
FX
Lai
R
et al
Expression of p27Kip1 and c-Jun activation binding protein 1 are inversely correlated in systemic anaplastic large cell lymphoma.
Clin Cancer Res
2003
9
1121
1128
123
Rassidakis
GZ
Lai
R
Herling
M
Cromwell
C
Schmitt-Graeff
A
Medeiros
LJ
Retinoblastoma protein is frequently absent or phosphorylated in anaplastic large-cell lymphoma.
Am J Pathol
2004
164
2259
2267
124
Lamant
L
de Reynies
A
Duplaniter
M-M
et al
Gene-expression profiling of systemic anaplastic large-cell lymphoma reveals differences based on ALK status and two distinct morphologic ALK+ subtypes.
Blood
2007
109
2156
2164
125
Piva
R
Pellegrino
E
Mattioli
M
et al
Functional validation of anaplastic lymphoma kinase signature identifies CEBPB and BCL2A1 as critical target genes.
J Clin Invest
2006
116
3171
3182
126
Quintanilla-Martinez
L
Pittaluga
S
Miething
C
et al
NPM-ALK-dependent expression of the transcription factor CCAAT/enahncer binding protein β in ALK-positive anaplastic large cell lymphoma.
Blood
2006
108
2029
2036
127
Druker
BJ
Tamura
S
Buchdunger
E
et al
Effects of a selective inhibitor of Abl tyrosine kinase on the growth of Bcr-Abl positive cells.
Nat Med
1996
2
561
566
128
Ritter
U
Damm-Welk
C
Fuchs
U
Bohle
RM
Borkhardt
A
Woessmann
W
Design and evaluation of chemically synthesized siRNA targeting the NPM-ALK fusion site in anaplastic large cell lymphoma (ALCL).
Oligonucleotides
2003
13
365
373
129
Hübinger
G
Wehnes
E
Xue
L
Morris
SW
Maurer
U
Hammerhead ribozyme-mediated cleavage of the fusion transcript NPM-ALK associated with anaplastic large-cell lymphoma.
Exp Hematol
2003
31
226
233
130
Piva
R
Chiarle
R
Manazza
AD
et al
Ablation of oncogenic ALK is a viable therapeutic approach for anaplastic large-cell lymphomas.
Blood
2006
107
689
697
131
Wan
W
Albom
MS
Lu
L
et al
Anaplastic lymphoma kinase activity is essential for the proliferation and survival of anaplastic large-cell lymphoma cells.
Blood
2006
107
1617
1623
132
Galkin
AV
Melnick
JS
Kim
S
et al
Identification of NVP-TAE684, a potent, selective, and efficacious inhibitor of NPM-ALK.
Proc Natl Acad Sci USA
2007
104
270
275
133
Ergin
M
Denning
MF
Izban
KF
et al
Inhibition of tyrosine kinase activity induces caspase-dependent apoptosis in anaplastic large cell lymphoma with NPM-ALK (p80) fusion protein.
Exp Hematol
2001
29
1082
1090
134
Pasqualucci
L
Wasik
M
Teicher
BA
et al
Antitumor activity of anti-CD30 immunotoxin (Ber-H2/saporin) in vitro and in severe combined immunodeficiency disease mice xenografted with human CD30+ anaplastic large-cell lymphoma.
Blood
1995
85
2139
2146
135
Pfeifer
W
Levi
E
Petrogiannis-Haliotis
T
Lehmann
L
Wang
Z
Kadin
ME
A murine xenograft model of human CD30+ anaplastic large cell lymphoma: successful growth inhibition with an anti-CD30 antibody (HeFi-1).
Am J Pathol
1999
155
1353
1359
136
Ho
L
Aytac
U
Stephens
LC
et al
In vitro and in vivo antitumor effect of the anti-CD26 monoclonal antibody 1F7 on human CD30+ anaplastic large cell T-cell lymphoma Karpas 299.
Clin Cancer Res
2001
7
2031
3040
137
Turturro
F
Heineke
HL
Drevyanko
TF
Link
CJ
Jr
Seth
P
Adenovirus-p53-mediated gene therapy of anaplastic large cell lymphoma with t(2;5) in a nude mouse model.
Gene Ther
2000
7
930
933
138
Turturro
F
Arnold
MD
Frist
AY
Seth
P
Effects of adenovirus-mediated expression of p27Kip1, p21Waf1 and p16INK4A in cell lines derived from t(2;5) anaplastic large cell lymphoma and Hodgkin's disease.
Leuk Lymphoma
2002
43
1323
1328
Sign in via your Institution