Abstract
Context
Since the first discovery of anaplastic lymphoma kinase (ALK) in anaplastic large cell lymphoma (ALCL) by Morris et al in 1994, the number of ALK-positive neoplasms, either in the form of translocation or gain-of-function mutations, have been dramatically expanded from ALCL of T- and NK-cell origin, to diffuse large B-cell lymphoma, inflammatory myofibroblastic tumor (IMT), neuroblastoma, non-small cell lung carcinoma (NSCLC), undifferentiated anaplastic thyroid carcinoma, and rare type of sarcomas.
Objective
This review covers the major aspects of ALK-immunoreactive neoplasms with emphasis on the pathogenesis of ALK-positive neoplasms. The new advances and rapid-evolving practices using ALK inhibitors for therapy are also discussed at the end of this review.
Data Sources
ALK(+) articles published in English literature are retrieved and critically reviewed.
Conclusion
ALK(+) neoplasia is a rapidly growing field and the list of ALK(+) neoplasms is being expanded continuously. Accurate and correct diagnosis of ALK(+) neoplasms is of paramount importance in guiding the appropriate treatment in the era of personalized medicine using specific ALK inhibitor.
Keywords: Anaplastic lymphoma kinase (ALK), anaplastic large cell lymphoma (ALCL), ALK-positive neoplasms
Since the first discovery of anaplastic lymphoma kinase (ALK) in anaplastic large cell lymphoma (ALCL) by Morris et al in 1994, the number of ALK-positive neoplasms, either in the form of translocation or gain-of-function mutations, have been dramatically expanded from ALCL of T- and NK-cell origin, to diffuse large B-cell lymphoma, inflammatory myofibroblastic tumor (IMT), neuroblastoma, non-small cell lung carcinoma (NSCLC), undifferentiated anaplastic thyroid carcinoma, and rare type of sarcomas. This review covers the major aspects of ALK-immunoreactive neoplasms with emphasis on the pathogenesis of ALK-positive neoplasia. The new advances and rapid-evolving practices using ALK inhibitors for therapy are discussed at the end of this review.
ALK in physiology
ALK is a receptor tyrosine kinase, which belongs to the insulin receptor superfamily [1]. The ALK gene is highly conserved among species and is located on human chromosome 2p23 [2]. ALK is abundantly expressed in nervous system during embryogenesis but is only focally expressed in an adult brain, suggesting of a role for ALK in the development of central nervous system [3]. Murine knockout studies revealed that mice lacking ALK gene showed only subtle abnormality in their brain including hyperproliferation of basal hippocampal progenitor cells, which was associated with behavior alterations [4]. Recent studies shed light on the physiologic role of ALK by showing its ability to function as a “dependence receptor” where it creates cellular states of dependence on its ligand by inducing or favoring apoptosis when unoccupied by ligand, and inhibiting apoptosis in the presence of ligand (or as a result of ALK fusion proteins). In other words, there is an inverse correlation between the kinase activation of ALK and its proapoptotic activity [5,6]. The application of these findings in mammals is debatable as definitive ALK ligand, if any, has not yet been identified so far [5].
ALK as an oncogene
ALK was first identified within an oncogenic gene fusion product associated with anaplastic large cell lymphoma (ALCL) [1]. Subsequent studies revealed that 80-85% of ALK-positive ALCL cases harbor t(2;5)(p23;q35) translocation, resulting in fusion of intracytoplasmic portion of ALK located on 2p23 to the N-terminal portion of nucleophosmin (NPM) located on 5q35 [7]. NPM is a nuclear chaperon involved in many essential biological functions of a cell including transportation of pre-ribosomal particles across nuclear membrane, DNA repair and regulation of DNA transcription [8]. The NPM protein contains an N-terminal dimerization domain which is essential for oncogenic potentials of the fusion protein by promoting autophosphorylation and activation of the kinase domain within the chimeric protein leading to phosphorylation and activation of downstream signaling proteins [8]. In addition to NPM, numerous partner proteins were found to be fused to ALK, which result in functional chimeric proteins. These partner proteins include ALK lymphoma oligomerization partner on chromosome 17 (ALO17) [9], TRK-fused gene (TGF) [10], tropomyosin 3 and 4 (TPM3 and TPM4) [11,12], non-muscle myosin heavy chain (MYH9) [13], and clathrin heavy chain (CLTC) [14] among others [15] (Table 1). All of these fusion proteins are associated with chromosomal rearrangements including translocation or inversion. They share the same ALK breakpoint, although they slightly differ in their downstream signaling effectors. These discrepancies are most likely due to different subcellular localizations associated with structural characteristics of the partner proteins. NPM is unique in that it provides a nuclear localization domain in addition to the dimerization interface which leads to partial accumulation of NPM-ALK in the nucleus besides cytoplasm [16]. This has practical application in diagnostic practices where immunohistochemical analysis shows both cytoplasmic and nuclear ALK expression in tumors with t(2;5) (p23;q35) involving ALK and NPM, but is strictly cytoplasmic in most of the other variants [17].
Table 1.
Disease | ALK alteration | Chromosomal abnormality | References |
---|---|---|---|
ALCL | NPM-ALK | t(2;5)(p23;q35) | [2] |
TPM3-ALK | t(1;2)(q25;p23) | [11] | |
TPM4-ALK | t(2;19)(p23;p13) | [12] | |
TFG-ALK | t(2;3)(p23;q21) | [10] | |
ATIC-ALK | inv(2)(p23;q35) | [125-127] | |
CLTC-ALK | t(2;17)(p23;q23) | [14] | |
MSN-ALK | t(2;X)(p32;q11-12) | [128] | |
ALO17-ALK | t(2;17)(p23;q25) | [9] | |
MYH9-ALK | t(2;22)(p23;q11.2) | [13] | |
DLBCL | NPM-ALK | t(2;5)(p23;q35) | [129,130] |
CLTC-ALK | t(2;17)(p23;q23) | [69,131] | |
Unknown | ins(3’ALK)(4q22-24) | [132] | |
SQSTM1-ALK | t(2;5)(p23.1;q35.3) | [66] | |
SEC31A-ALK | ins(4)(2;4)(?;q21) t(2;4)(p24;q21) | [133,134] | |
Unknown | t(X;2)(q21;p23) and t(2;12)(p23;q24) | [68] | |
Plasmacytoma | CLTC-ALK | t(2;17)(p23;q23) | [71] |
IMT | TPM3-ALK | t(1;2)(q25;p23) | [135] |
TPM4-ALK | t(1;19)(p23;p13) | [135] | |
CLTC-ALK | t(2;17)(p23;q23) | [136] | |
CARS-ALK | t(2;11;2)(p23;p15;q31) | [9,137] | |
ATIC-ALK | inv(2)(p23;q35) | [138] | |
RANBP2-ALK | t(2;2)(p23;q13) inv(2)(p23;p15;q31) | [139] | |
SEC31L1-ALK | t(2;4)(p23;q21) | [140] | |
NSCLC | EML4-ALK | inv(2)(p21;p23) | [85,97] |
TFG-ALK | t(2;3)(p23;q21) | [97] | |
KIF5B-ALK | t(2;10)(p23;p11) | [95,96] | |
Esophageal cancer | TPM4-ALK | t(2;19)(p23;p13) | [109,110] |
Renal cell carcinoma | VCL-ALK | t(2;10)(p23;q22) | [111] |
Renal medullary carcinoma | VCL-ALK | t(2;10)(p23;q22) | [112] |
Breast cancer | EML4-ALK | inv(2)(p21;p23) | [92] |
Colon cancer | EML4-ALK | inv(2)(p21;p23) | [92] |
Neuroblastoma | Point mutations or amplification | [21-25] | |
Thyroid carcinoma | Point mutations | [26] |
Aberrant kinase activity of ALK is not always the result of ALK gene rearrangements. Amplification or increased gene copy numbers is another mechanism of ALK-mediated tumorigenesis found mainly in neuroblastoma and some of NSCLC, respectively [18-21]. Point mutations in the ALK kinase domain can also lead to constitutive activation of ALK kinase, representing an alternative mechanism for oncogenesis. These mutations have been found in some malignancies such as neuroblastoma [21-25] and anaplastic thyroid cancer [26]. Several studies in the past have documented the oncogenic potentials of ALK fusions in vivo and in vitro [27,28]. However, recent reports have shown high incidence of ALK fusion proteins including NPM-ALK and ATIC (5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase)-ALK present in the peripheral blood cells of apparently healthy individuals [29,30]. This finding indicates that the presence of ALK in its oncogenic form is required but not sufficient to induce cell transformation. In fact, aberrant ALK tyrosine kinase activity has been shown to result in cell cycle arrest and senescence induced by p16, P53 and Rb, suggesting that inactivation of these tumor suppressor genes are among those additional molecular events required for cell transformation [31,32].
ALK-mediated signaling events in cancer
Multiple signaling pathways are triggered by ALK not only to enhance cell proliferation and survival but also to induce cytoskeletal rearrangement and cell migration [16]. These include Ras/ERK (Ras/Extracellular signal Regulated Kinase), JAK/STAT (Janus Kinase/Signal Transducer and Activator of Transcription), PI3K/Akt (Phosphatidyloinositol-3 Kinase/Akt) and PLCγ (phospholipase C-γ) pathways [7,33-35]. Overall, proliferative effect of ALK chimeric proteins is mainly attributable to Ras/ERK pathway whereas JAK/STAT and PI3K/AKT pathways are mediators of cell survival and phenotypic changes [16]. Several adaptor proteins are involved to transmit the ALK-induced mitogenic signals by direct attachment to specific tyrosine residues within the intracytoplasmic segment of ALK fusion proteins. These include IRS-1 (insulin receptor substrate-1), SHC (SH2 domain-containing transforming protein), GRB2 (growth factor receptor-bound protein 2) [16,35]. It has been postulated that PLCγ also contributes to transmit the mitogenic signal downstream of ALK fusion proteins by direct binding to ALK [36]. In fact, mutational studies showed that removal of PLCγ binding site on ALK chimera (Tyr664) blocked transforming potentials of NPM-ALK [36], supporting the importance of PLCγ in oncogenic signal.
Activation of PI3K pathway generates an antiapoptotic signal via activation of Akt and subsequent downstream effectors. These include phosphorylation and activation of FOXO3A, a member of the forkhead family of transcription factors, leading to sequestration of this molecule in the cytoplasm and therefore inhibition of its transcription activity. This results in induction of cell survival as well as cell cycle progression via upregulation of cycline D2 and downregulation of Bim-1 and p27 (Kip1) [37]. The survival effects of ALK-NPM fusion is also mediated by activation of mammalian target of rapamycin (mTOR), which occurs downstream of both PI3K and Ras/ERK pathways [38,39].
The role of JAK/STAT pathway to mediate the oncogenic signals of NPM-ALK has been extensively studied. These studies have shown that among the family of STAT proteins, STAT3 is the key modulator of ALK-induced growth and survival effects [40-43]. Phosphorylation and activation of STAT3 is achieved directly by ALK kinase or alternatively through activated JAK3 [44]. Interestingly, a contrary signaling role has been suggested for STAT5, another member of the STAT family. STAT5 has emerged as a tumor suppressor gene in NPM-ALK induced ALCL cells evidenced by reciprocal inhibition of NPM-ALK expression. In fact, NPM-ALK protects its expression by epigenetic silencing of STAT5, and re-expression of STAT5, by inhibition of methylation, results in decreased expression of NPM-ALK [45]. SHP-1 (SH2 domain-containing protein tyrosine phosphatase-1), a potent negative regulator of JAK3/STAT3 signaling, is another tumor suppressor gene that can block NPM-ALK expression and is epigenetically silenced in NPM-ALK induced ALCL cells [46-48].
ALK(+) anaplastic large cell lymphoma (ALK+ ALCL)
ALCL was first described by Stein et al in 1985 as a morphologically distinct lymphoma with consistent expression of Ki-1 antigen (now known as CD30) [49]. Subsequent studies revealed that most of ALCL tumor cells have a T-cell lineage origin, although in rare cases ALCL may show NK immunophenotype (see below) [50]. Subsequently, a non-random recurrent balanced chromosomal translocation between chromosome 2 and chromosome 5 [t(2;5)] was identified [51,52]. The gene located on chromosome 2p23, namely ALK, was finally cloned in 1994 [2]. ALK-positive ALCL has been defined by WHO classification as a T-cell lymphoma consisting of lymphoid cells that are usually large with abundant cytoplasm and pleomorphic, often horseshoe-shaped nuclei, with a translocation involving the ALK gene and expression of ALK and CD30 [53].
ALK+ ALCL is most commonly diagnosed in the first decades of life with a small male predominance [54,55]. Most patients present clinically with lymphadenopathy as well as involvement of extranodal sites including bone marrow [55,56]. Although several morphologic variants have been described, most of the cases contain characteristic large cells with eccentric horseshoe-or kidney-shaped nuclei referred to as “hallmark” cells. These tumor cells have abundant cytoplasm and often have denser focal staining of CD30 in the perinuclear, Golgi region of the cytoplasm. Five morphological variants have been described in the recent edition of WHO classification including common pattern (60%), lymphohistiocytic pattern (10%), small cell pattern (5-10%), Hodgkin-like pattern (3%) and composite pattern (15%) [53]. Diagnosis of the uncommon variants may be difficult without knowledge of the ALK and CD30 expression due to small numbers of large cells. A useful diagnostic clue in these cases is the tendency of large cells to cluster around blood vessels. As mentioned before, ALCL tumor cells are CD30 positive by definition and show characteristic membrane and paranuclear (Golgi) staining. This pattern of CD30 expression is not pathognomonic for ALCL; however, it helps to distinct ALCL from the broad spectrum of CD30 expressive entities including reactive immunoblasts, Hodgkin and non Hodgkin lymphomas and nonlymphoid neoplasms such as embryonal carcinoma [57]. Depend upon the nature of ALK fusion protein, ALK expression and sub-cellular localization can be different and helpful in predicting the fusion partner gene(s). For example, nuclear and diffuse cytoplasmic staining of ALK is seen in ALK+ ALCL with t(2;5) involving ALK and NPM [2]; on the other hand, exclusive granular cytoplasmic staining pattern is observed in ALK+ ALCL with t(2;17) involving ALK and clathrin heavy polypeptide gene [14]. In the majority of the ALK+ ALCL, diffuse cytoplasmic staining is observed [17]. ALK is promiscuous, and many fusion partner genes have been identified, these genes include the following: NPM at 5q35, TPM3 at 1q25, TFG at 3q12, CLTC at 17q23, MSN a 5 xq11-12, TPM4 at 19p13.1, MYH9 at 22q11.2 and ALO17 at 17q25 [57] (Table 1).
Immunohistochemically, the majority of ALK+ ALCL tumor cells express EMA and one or more T-cell antigens [57,58]. CD3, CD5, CD7 and T-cell receptors (TCRs), are not commonly expressed but CD2 and CD4 are more often expressed [57]. Due to the loss of some T-cell markers, some of ALCL cases may present as a null immunophenotype although NK/null phenotype are now considered a single rare entity [53]. Tumor cells from ALCL frequently express cytotoxic related antigens including TIA-1, perforin and granzyme B [50]. ALCL cells strongly express IL-2 receptor (CD25) and lack evidence of Epstein-Barr virus infection [58-60].
Patients with ALK+ ALCL show an overall better prognosis than patients with ALK(-) ALCL [61,62]. ALK+ ALCL cases with small cell morphologic pattern or those with aberrant expression of CD56 have adverse prognosis [7]. ALCL cases expressing ALK fusion proteins other than NPM-ALK have a good prognosis, similar to that of cases with t(2;5) translocation [17].
ALK(+) large B-cell lymphoma (ALK+ LBCL)
ALK+ LBCL is a rare subtype of diffuse large B-cell lymphoma (DLBCL) characterized by sinusoidal growth of large anaplastic, immunoblastic or plasmablastic B-cells with very aggressive clinical course [63]. Since the first reported ALK+ LBCL by Delsol G et al in 1997 [63], there have been approximately more than 50 cases reported in the English literature. ALK+ LBCL cases have been seen from all age groups (9-70) with male:female ratio of 3-5:1 [63,64]. Although NPM-ALK fusion protein, as a result of t(2;5) translocation, can be found in a minority of the cases, the most common ALK rearrangement ALK+ LBCL involves CLTC at 17q23 within a t(2:17) translocation [65]. SQSTM1, encoding a ubiquitin binding protein and SEC31A, encoding a ER-Golgi transporter, are newly discovered but rare partners of ALK in ALK+ LBCLs [66,67]. Very recently, new translocations involved in Xq21 and 12q24 in partner with ALK in DLBCL were discovered by Shi M et al. [68].
Clinically, ALK+ LBCL usually presents as a peripheral lymphadenopathy or a mediastinal mass and commonly shows advanced stage (III-VI) at the time of diagnosis. ALK staining is cytoplasmic in almost all of the cases. Tumor cells are also positive for CD138, EMA and cytoplasmic Ig (especially IgA) but negative for most, if not all, of the B-cell associated markers including CD19, CD20, CD22, CD23 and CD79a. In fact, in reviewing of 32 cases of ALK+ LBCL, Reichard KK et al have shown that CD138 and EMA were expressed in all cases, while CD20 was expressed in only 3% of cases [69]. Of interest, in contrast to ALK+ ALCL of T-cell origin, in which CD30 and CD45 were almost always positive, ALK+ LBCL is rarely positive for CD30 [69]. In addition, CD4 and CD57 are aberrantly expressed in higher percentage of ALK+ LBCL cases with 64% and 40%, respectively [69]. The prognosis is poor with median survival of 11 to 12 months in advance stages and poor response to treatment [64,69,70].
Extramedullary plasmacytoma
Recently, a case of extramedullary plasmacytoma with expression of CLTC-ALK transcript has been reported by Wang WY, et al. [71]. Interestingly, this is in line with the previous study showing that NPM-ALK transgenic mice developed plasma cell tumors [72].
Inflammatory myofibroblastic tumor (IMT)
IMT, a poorly understood mesenchymal tumor with various names in the past, has emerged as a distinct entity from the broad category of inflammatory pseudotumor with characteristic clinical, pathological and molecular features [73]. Inflammatory pseudotumor was first described in lung but subsequently was reported in virtually every organ/system in the body [74]. Originally, inflammatory pseudotumor was considered a post-inflammatory/reactive process but further studies suggested a neoplastic nature in some cases as evidenced by clonal cytogenetic abnormalities and more aggressive clinical behavior such as local recurrence, and even distant metastatases [75]. IMT represents those lesions with a neoplastic nature although these two terms is being used in the literature interchangeably. IMTs are more common in children and young adults but it can occur in any age and the most common sites are lung, mesentry and omentum [76,77].
IMT consists of spindle to fuseform tumor cells in the background of inflammatory cells comprising of plasma cells, small mature-appearing lymphocytes, neutrophils, and eosinophils with myxoid or hyaline stroma [76]. Immunophenotypically, tumor cells in IMT are positive for smooth muscle actin (SMA), muscle specific actin (MSA) and desmin [76].
More common in younger patients, ALK immunostain is positive in approximately 50% of the cases and is associated with clonal rearrangement involving ALK oncogene [78,79]. Interestingly, NPM-ALK has not been identified in IMTs, but other fusion proteins found in ALCL such as ALK-TPM-3, ALK-TPM4, ALK-ATIC and ALK-CLTC have also been reported in IMT [73]. There is no clear correlation between ALK expression and prognosis or recurrence, although some studies reported a better prognosis in ALK positive cases [80].
Neuroblastoma
Neuroblastoma, the most common malignancy in infancy in the first year of life is derived from the neuroblasts in neural crest that give rise to simpatico-adrenal nervous system [81]. Most neuroblastomas are sporadic but small subset of cases is considered familial with autosomal dominant inheritance [33,82]. Neuroblastoma is a heterogeneous clinical entity. While some of the confirmed cases regress spontaneously, most of neuroblastomas show progression and relapse despite intense chemoradiation regimens [81]. These tumors also exhibit variable grades of histologic differentiation associating with clinical outcome. MYCN amplification, deletion of 1p and 11q, unbalanced gain (translocation) of 17q are well established genetic abnormalities in neuroblastomas and are associated with poor outcome [82]. The role of ALK gene in pathogenesis of neuroblastoma was first suggested by Lamant and colleagues where they found expression of ALK protein in 22 out of 24 human-derived neuroblastoma cell lines [83]. In 2008, several studies identified multiple germline and somatic mutations within the ALK gene in familial and sporadic neuroblastoma cases [21-25]. These mutations were found to be associated with increased ALK kinase activity and those cases harboring these mutations had worse prognosis. ALK gene amplification represents another potential mechanism for tumorigenesis in neuroblastoma [18,21,84]. Berthier and colleagues showed that ALK immunoreactivity increased with ALK gene copy number gain and this was associated with poor outcome [84]. The critical role of ALK gene mutation or amplification in pathogenesis of neuroblastoma was further supported by using ALK inhibitors and/or targeted knockdown of ALK mRNA in neuroblastoma derived cell lines carrying mutated or amplified ALK alleles [23-25]. ALK and MYCN are in close proximity on the short arm of chromosome 2 and one study showed concordant aberrancy in ALK and MYCN copy numbers in 50% of neuroblastoma cases [20].
Non-small cell lung cancer (NSCLC)
The role of ALK gene in pathogenesis of NSCLCs was first reported in 2007 when a small inversion within chromosome 2p was shown to result in the formation of a fusion gene comprising portions of the EML4 (echinoderm microtubule-associated protein-like 4) gene and the ALK gene [85]. The chromosomal inversion does not always occur in the same location and multiple EML4-ALK variants have been identified [86]. The oncogenic potential of EML4-ALK fusion was further confirmed using transgenic mouse model that express EML4-ALK specifically in lung alveolar epithelial cells. All of the transgenic mice examined developed hundreds of adenocarcinoma nodules in both lungs within a few weeks after birth and also they responded dramatically to an ALK kinase inhibitor [87]. The EML4-ALK fusion transcript was detected in 6.7% of NSCLC patients in the original report [85]. Subsequent studies have reported that between 1.6%-13% of lung tumors harbor EML4-ALK fusions [86,88-92]. In contrast to EGFR mutations, the frequency of EML4-ALK fusion gene variants is not influenced by ethnicity [93]. ALK rearrangement is associated with younger age, negative smoking history [88,89]. None of the ALK-rearranged tumors harbored coexisting EGFR mutations [88,89,91,94]. EML4 does not appear to be the exclusive fusion partner with ALK, two other fusion proteins ALK-TFG and ALK-KIF5B, have been described as well [95-97].
NSCLC are divided into three histologic subtypes including adenocarcinoma, squamous cell carcinoma and large cell carcinoma. Among these, adenocarcinomas seem to be the major NSCLC cell type harboring EML4-ALK fusions [93,98]. It has been suggested that some histologic findings may help to select cases for ALK testing. These include a solid signet-ring cell pattern and a mucinous cribriform pattern [88,89,99]. In one study, ALK amplification and increased ALK copy number were detected in 10% and 63% of the NSCLC cases [19]. There was an association between ALK amplification and EGFR FISH positivity but not with prognosis [19].
Using RT-PCR detection method, a recent study on European population reported EML4-ALK fusions in 7.5% of the NSCLC cases. However, the same oncogenic fusion was detected in non-cancerous lung tissue samples from the cases that had EML4-ALK negative NSCLC [100]. These findings turned into a matter of debate and interpreted by others as potential false positive results or technical error [101,102].
Similar to other ALK fusions, EML4-ALK rearrangement leading to ALK transcriptional upregulation and subsequent ALK protein expression, can be detected by immunohistochemistry (IHC). While some studies reported 100% concordance between the results of IHC and molecular studies (including FISH and RT-PCR) in detecting rearranged ALK fusions [103,104], other reports showed relative inability of the standard IHC method to detect ALK rearrangement in NSCLCs [89,101]. This might be due to the weak transcriptional activity of EML4 promoter to drive EML4-ALK expression compared to other ALK-rearranged fusions in ALCLs. In a more recent study, a highly sensitive antibody (D5F3) was developed that reliably detected all positive ALK fusions with high specificity [105]. These date suggest that IHC for ALK expression can be readily used in routine daily pathology practice to screen for ALK rearrangements in NSCLCs. However, it is empirical to confirm the cases with weak ALK staining using molecular studies such as FISH [101,105].
ALK+ sarcomas
ALK expression has been observed in other soft tissue tumors besides IMT. These include rhabdomyosarcomas, various lipogenic tumors, Ewing's sarcoma/primitive neuroectodermal tumors (PNETs), and leiomyosarcomas [106-108]. ALK overexpression in these cases was independent of fusion status. Amplification or increased copy number of ALK may cause ALK protein overexpression. In rhabdomyosarcomas, ALK overexpression is more associated with alveolar subtype [107].
Other rare ALK(+) carcinomas
ALK(+) esophageal squamous carcinomas
ALK rearrangements have been reported in esophageal squamous cell carcinomas from Iran and China [109,110]. In both studies TPM4-ALK rearrangement was the only identified fusion. The frequency of ALK fusions in esophageal squamous cell carcinoma remains to be determined.
ALK(+) carcinoma of breast
ELM4-ALK is found in 2.4% of breast carcinomas using exon array profiling technique [92].
ALK(+) carcinoma of colon
By using the exon array profiling, Lin E at el also demonstrated that 2.5% of colorectal cancers harbor EML4-ALK rearrangement [92]. Whether ALK rearrangement in these cases is associated with specific histologic features is yet to be determined.
ALK(+) thyroid cancer
Two point mutations in exon 23 of the ALK gene have been recently reported in anaplastic thyroid cancers both reside in ALK tyrosine kinase domain and associated with gain of function. The frequency of these mutations was 11% [26]. ALK rearrangement has not been reported in thyroid cancers.
ALK(+) renal cell carcinoma
In a recent study on six pediatric renal cell carcinomas, two cases showed chromosomal rearrangements involving the ALK locus with a resultant novel VCL-ALK fusion expressed in one of the cases [111]. ALK rearrangement in this case could also be detected by IHC [111]. The same ALK rearrangement has been reported in sickle cell trait-associated renal medullary carcinoma [112].
Therapeutic advances
Targeted therapy has been emerged in the recent past as a new approach in the treatment of different cancers with promising outcomes. Selective kinase inhibitors are one of the most important class of anticancer medications, and the use of which is already well established in the current clinical practice [113]. ALK is an appealing oncogene to target for personalized cancer therapy for two main reasons: first, limited ALK expression in adult tissues and second, discovery of a growing number of ALK-driven cancers. Several studies have been conducted to target ALK by means of RNA interference [114], monoclonal antibodies and interfere with ALK protein stability through inhibition of heat shock proteins 90 (HSP90) [115]. However, the most promising results were achieved using small molecule inhibitors to block the kinase domain of ALK protein. The first ALK inhibitor that entered into clinical trials was Crizotinib [116]. Crizotinib is an ATP competitive, orally bioavailable small molecule inhibitor of receptor tyrosine kinase c-MET (mesenchymal-epithelial transition growth factor also known as hepatocyte growth factor receptor) that was found to have similar potent inhibitory effect against ALK kinase domain [117]. Recently, the results of the early phase clinical trials using Crizotinib in 82 patients with EML4-ALK positive NSCLC were published and showed an impressive response rate up to 57% at a mean treatment duration of 6 months [118]. This result is very promising given that only 10% response can be achieved using traditional chemotherapy. Furthermore, Crizotinib has been clinically tested on patients with IMT and showed impressive response [119]. ALK inhibitors have shown remarkable effects against ALK-driven ALCL, DLBCL and neuroblastoma in vitro [120-122] and they are currently being tested clinically. However, despite dramatic initial responses, as has been observed with other tyrosine kinase inhibitors (e.g. imatinib and EGFR inhibitors), prolonged treatment with ALK inhibitors will eventually lead to drug-resistance point mutations within the ALK kinase domain [123]. Fortunately, the mechanism of drug resistance has been modeled which enables clinicians to screen and select resistant patients who most likely will benefit from second generation ALK tyrosine kinase inhibitors or HSP90 blockers or both [124]. While the physiologic role of ALK is still ambiguous, patients have already started getting benefit from targeting ALK in the personalized cancer therapy. It is expected that many ALK targeting compounds will arrive in clinics in near future.
References
- 1.Morris SW, Naeve C, Mathew P, James PL, Kirstein MN, Cui X, Witte DP. 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;14:2175–88. doi: 10.1038/sj.onc.1201062. [DOI] [PubMed] [Google Scholar]
- 2.Morris SW, Kirstein MN, Valentine MB, Dittmer KG, Shapiro DN, Saltman DL, Look AT. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma. Science. 1994;263:1281–4. doi: 10.1126/science.8122112. [DOI] [PubMed] [Google Scholar]
- 3.Iwahara T, Fujimoto J, Wen D, Cupples R, Bucay N, Arakawa T, Mori S, Ratzkin B, Yamamoto T. Molecular characterization of ALK, a receptor tyrosine kinase expressed specifically in the nervous system. Oncogene. 1997;14:439–49. doi: 10.1038/sj.onc.1200849. [DOI] [PubMed] [Google Scholar]
- 4.Bilsland JG, Wheeldon A, Mead A, Znamenskiy P, Almond S, Waters KA, Thakur M, Beaumont V, Bonnert TP, Heavens R, Whiting P, McAllister G, Munoz-Sanjuan I. Behavioral and neurochemical alterations in mice deficient in anaplastic lymphoma kinase suggest therapeutic potential for psychiatric indications. Neuropsychopharmacology. 2008;33:685–700. doi: 10.1038/sj.npp.1301446. [DOI] [PubMed] [Google Scholar]
- 5.Allouche M. ALK is a novel dependence receptor: potential implications in development and cancer. Cell Cycle. 2007;6:1533–1538. doi: 10.4161/cc.6.13.4433. [DOI] [PubMed] [Google Scholar]
- 6.Mourali J, Benard A, Lourenco FC, Monnet C, Greenland C, Moog-Lutz C, Racaud-Sultan C, Gonzalez-Dunia D, Vigny M, Mehlen P, Delsol G, Allouche M. Anaplastic lymphoma kinase is a dependence receptor whose proapoptotic functions are activated by caspase cleavage. Mol Cell Biol. 2006;26:6209–6222. doi: 10.1128/MCB.01515-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kinney MC, Higgins RA, Medina EA. Anaplastic large cell lymphoma: twenty-five years of discovery. Arch Pathol Lab Med. 2011;135:19–43. doi: 10.5858/2010-0507-RAR.1. [DOI] [PubMed] [Google Scholar]
- 8.Grisendi S, Mecucci C, Falini B, Pandolfi PP. Nucleophosmin and cancer. Nat Rev Cancer. 2006;6:493–505. doi: 10.1038/nrc1885. [DOI] [PubMed] [Google Scholar]
- 9.Cools J, Wlodarska I, Somers R, Mentens N, Pedeutour F, Maes B, De Wolf-Peeters C, Pauwels P, Hagemeijer A, Marynen P. Identification of novel fusion partners of ALK, the anaplastic lymphoma kinase, in anaplastic large-cell lymphoma and inflammatory myofibroblastic tumor. Genes Chromosomes Cancer. 2002;34:354–362. doi: 10.1002/gcc.10033. [DOI] [PubMed] [Google Scholar]
- 10.Hernandez L, Bea S, Bellosillo B, Pinyol M, Falini B, Carbone A, Ott G, Rosenwald A, Fernández A, Pulford K, Mason D, Morris SW, Santos E, Campo E. Diversity of genomic breakpoints in TFG-ALK translocations in anaplastic large cell lymphomas: identification of a new TFG-ALK(XL) chimeric gene with transforming activity. Am J Pathol. 2002;160:1487–1494. doi: 10.1016/S0002-9440(10)62574-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lamant L, Dastugue N, Pulford K, Delsol G, Mariame B. A new fusion gene TPM3-ALK in anaplastic large cell lymphoma created by a (1;2)(q25;p23) translocation. Blood. 1999;93:3088–3095. [PubMed] [Google Scholar]
- 12.Meech SJ, McGavran L, Odom LF, Liang X, Meltesen L, Gump J, Wei Q, Carlsen S, Hunger SP. Unusual childhood extramedullary hematologic malignancy with natural killer cell properties that contains tropomyosin 4--anaplastic lymphoma kinase gene fusion. Blood. 2001;98:1209–1216. doi: 10.1182/blood.v98.4.1209. [DOI] [PubMed] [Google Scholar]
- 13.Lamant L, Gascoyne RD, Duplantier MM, Armstrong F, Raghab A, Chhanabhai M, Rajcan-Separovic E, Raghab J, Delsol G, Espinos E. Non-muscle myosin heavy chain (MYH9): a new partner fused to ALK in anaplastic large cell lymphoma. Genes Chromosomes Cancer. 2003;37:427–432. doi: 10.1002/gcc.10232. [DOI] [PubMed] [Google Scholar]
- 14.Touriol C, Greenland C, Lamant L, Pulford K, Bernard F, Rousset T, Mason DY, Delsol G. Further demonstration of the diversity of chromosomal changes involving 2p23 in ALK-positive lymphoma: 2 cases expressing ALK kinase fused to CLTCL (clathrin chain polypeptide-like) Blood. 2000;95:3204–3207. [PubMed] [Google Scholar]
- 15.Barreca A, Lasorsa E, Riera L, Machiorlatti R, Piva R, Ponzoni M, Kwee I, Bertoni F, Piccaluga PP, Pileri SA, Inghirami G European T-Cell Lymphoma Study Group. Anaplastic lymphoma kinase in human cancer. J Mol Endocrinol. 2011;47:R11–23. doi: 10.1530/JME-11-0004. [DOI] [PubMed] [Google Scholar]
- 16.Chiarle R, Voena C, Ambrogio C, Piva R, Inghirami G. The anaplastic lymphoma kinase in the pathogenesis of cancer. Nat Rev Cancer. 2008;8:11–23. doi: 10.1038/nrc2291. [DOI] [PubMed] [Google Scholar]
- 17.Falini B, Pulford K, Pucciarini A, Carbone A, De Wolf-Peeters C, Cordell J, Fizzotti M, Santucci A, Pelicci PG, Pileri S, Campo E, Ott G, Delsol G, Mason DY. Lymphomas expressing ALK fusion protein(s) other than NPM-ALK. Blood. 1999;94:3509–3515. [PubMed] [Google Scholar]
- 18.Osajima-Hakomori Y, Miyake I, Ohira M, Nakagawara A, Nakagawa A, Sakai R. Biological role of anaplastic lymphoma kinase in neuroblastoma. Am J Pathol. 2005;167:213–222. doi: 10.1016/S0002-9440(10)62966-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Salido M, Pijuan L, Martinez-Aviles L, Galván AB, Cañadas I, Rovira A, Zanui M, Martínez A, Longarón R, Sole F, Serrano S, Bellosillo B, Wynes MW, Albanell J, Hirsch FR, Arriola E. Increased ALK gene copy number and amplification are frequent in non-small cell lung cancer. J Thorac Oncol. 2011;6:21–27. doi: 10.1097/JTO.0b013e3181fb7cd6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Subramaniam MM, Piqueras M, Navarro S, Noguera R. Aberrant copy numbers of ALK gene is a frequent genetic alteration in neuroblastomas. Hum Pathol. 2009;40:1638–1642. doi: 10.1016/j.humpath.2009.05.002. [DOI] [PubMed] [Google Scholar]
- 21.Caren H, Abel F, Kogner P, Martinsson T. High incidence of DNA mutations and gene amplifications of the ALK gene in advanced sporadic neuroblastoma tumours. Biochem J. 2008;416:153–159. doi: 10.1042/bj20081834. [DOI] [PubMed] [Google Scholar]
- 22.Chen Y, Takita J, Choi YL, Kato M, Ohira M, Sanada M, Wang L, Soda M, Kikuchi A, Igarashi T, Nakagawara A, Hayashi Y, Mano H, Ogawa S. Oncogenic mutations of ALK kinase in neuroblastoma. Nature. 2008;455:971–974. doi: 10.1038/nature07399. [DOI] [PubMed] [Google Scholar]
- 23.Janoueix-Lerosey I, Lequin D, Brugieres L, Ribeiro A, de Pontual L, Combaret V, Raynal V, Puisieux A, Schleiermacher G, Pierron G, Valteau-Couanet D, Frebourg T, Michon J, Lyonnet S, Amiel J, Delattre O. Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature. 2008;455:967–970. doi: 10.1038/nature07398. [DOI] [PubMed] [Google Scholar]
- 24.George RE, Sanda T, Hanna M, Fröhling S, Luther W 2nd, Zhang J, Ahn Y, Zhou W, London WB, McGrady P, Xue L, Zozulya S, Gregor VE, Webb TR, Gray NS, Gilliland DG, Diller L, Greulich H, Morris SW, Meyerson M, Look AT. Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature. 2008;455:975–978. doi: 10.1038/nature07397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Mosse YP, Laudenslager M, Longo L, Cole KA, Wood A, Attiyeh EF, Laquaglia MJ, Sennett R, Lynch JE, Perri P, Laureys G, Speleman F, Kim C, Hou C, Hakonarson H, Torkamani A, Schork NJ, Brodeur GM, Tonini GP, Rappaport E, Devoto M, Maris JM. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature. 2008;455:930–935. doi: 10.1038/nature07261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Murugan AK, Xing M. Anaplastic thyroid cancers harbor novel oncogenic mutations of the ALK gene. Cancer Res. 2011;71:4403–4411. doi: 10.1158/0008-5472.CAN-10-4041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Turner SD, Alexander DR. What have we learnt from mouse models of NPM-ALK-induced lymphomagenesis? Leukemia. 2005;19:1128–1134. doi: 10.1038/sj.leu.2403797. [DOI] [PubMed] [Google Scholar]
- 28.Kuefer MU, Look AT, Pulford K, Behm FG, Pattengale PK, Mason DY, Morris SW. Retrovirus-mediated gene transfer of NPM-ALK causes lymphoid malignancy in mice. Blood. 1997;90:2901–2910. [PubMed] [Google Scholar]
- 29.Maes B, Vanhentenrijk V, Wlodarska I, Cools J, Peeters B, Marynen P, de Wolf-Peeters C. The NPM-ALK and the ATIC-ALK fusion genes can be detected in non-neoplastic cells. Am J Pathol. 2001;158:2185–2193. doi: 10.1016/S0002-9440(10)64690-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Trumper 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. doi: 10.1046/j.1365-2141.1998.01097.x. [DOI] [PubMed] [Google Scholar]
- 31.McDuff FK, Turner SD. Aberrant anaplastic lymphoma kinase activity induces a p53 and Rb-dependent senescence-like arrest in the absence of detectable p53 stabilization. PLoS One. 2011;6:e17854. doi: 10.1371/journal.pone.0017854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Martinelli P, Bonetti P, Sironi C, Pruneri G, Fumagalli C, Raviele PR, Volorio S, Pileri S, Chiarle R, McDuff FK, Tusi BK, Turner SD, Inghirami G, Pelicci PG, Colombo E. The lymphoma-associated NPM-ALK oncogene elicits a p16INK4a/pRb-dependent tumor-suppressive pathway. Blood. 2011;117:6617–6626. doi: 10.1182/blood-2010-08-301135. [DOI] [PubMed] [Google Scholar]
- 33.Palmer RH, Vernersson E, Grabbe C, Hallberg B. Anaplastic lymphoma kinase: signalling in development and disease. Biochem J. 2009;420:345–361. doi: 10.1042/BJ20090387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wasik MA, Zhang Q, Marzec M, Kasprzycka M, Wang HY, Liu X. Anaplastic lymphoma kinase (ALK)-induced malignancies: novel mechanisms of cell transformation and potential therapeutic approaches. Semin Oncol. 2009;36:S27–35. doi: 10.1053/j.seminoncol.2009.02.007. [DOI] [PubMed] [Google Scholar]
- 35.Amin HM, Lai R. Pathobiology of ALK+ anaplastic large-cell lymphoma. Blood. 2007;110:2259–2267. doi: 10.1182/blood-2007-04-060715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bai RY, Dieter P, Peschel C, Morris SW, Duyster J. Nucleophosmin-anaplastic lymphoma kinase of large-cell anaplastic lymphoma is a constitutively active tyrosine kinase that utilizes phospholipase C-gamma to mediate its mitogenicity. Mol Cell Biol. 1998;18:6951–6961. doi: 10.1128/mcb.18.12.6951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Gu TL, 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. doi: 10.1182/blood-2003-03-0820. [DOI] [PubMed] [Google Scholar]
- 38.Marzec M, Kasprzycka M, Liu X, El-Salem M, Halasa K, Raghunath PN, Bucki R, Wlodarski P, Wasik MA. Oncogenic tyrosine kinase NPM/ALK induces activation of the rapamycin-sensitive mTOR signaling pathway. Oncogene. 2007;26:5606–5614. doi: 10.1038/sj.onc.1210346. [DOI] [PubMed] [Google Scholar]
- 39.Vega F, Medeiros LJ, Leventaki V, Atwell C, Cho-Vega JH, Tian L, Claret FX, Rassidakis GZ. 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. doi: 10.1158/0008-5472.CAN-05-3018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Chiarle R, Simmons WJ, Cai H, Dhall G, Zamo A, Raz R, Karras JG, Levy DE, Inghirami G. Stat3 is required for ALK-mediated lymphomagenesis and provides a possible therapeutic target. Nat Med. 2005;11:623–629. doi: 10.1038/nm1249. [DOI] [PubMed] [Google Scholar]
- 41.Zamo A, Chiarle R, Piva R, Howes J, Fan Y, Chilosi M, Levy DE, Inghirami G. Anaplastic lymphoma kinase (ALK) activates Stat3 and protects hematopoietic cells from cell death. Oncogene. 2002;21:1038–1047. doi: 10.1038/sj.onc.1205152. [DOI] [PubMed] [Google Scholar]
- 42.Khoury JD, Medeiros LJ, Rassidakis GZ, Yared MA, Tsioli P, Leventaki V, Schmitt-Graeff A, Herling M, Amin HM, Lai R. Differential expression and clinical significance of tyrosine-phosphorylated STAT3 in ALK+ and ALK-anaplastic large cell lymphoma. Clin Cancer Res. 2003;9:3692–3699. [PubMed] [Google Scholar]
- 43.Amin HM, McDonnell TJ, Ma Y, Lin Q, Fujio Y, Kunisada K, Leventaki V, Das P, Rassidakis GZ, Cutler C, Medeiros LJ, Lai R. Selective inhibition of STAT3 induces apoptosis and G(1) cell cycle arrest in ALK-positive anaplastic large cell lymphoma. Oncogene. 2004;23:5426–5434. doi: 10.1038/sj.onc.1207703. [DOI] [PubMed] [Google Scholar]
- 44.Amin HM, Medeiros LJ, Ma Y, Feretzaki M, Das P, Leventaki V, Rassidakis GZ, O'Connor SL, McDonnell TJ, Lai R. Inhibition of JAK3 induces apoptosis and decreases anaplastic lymphoma kinase activity in anaplastic large cell lymphoma. Oncogene. 2003;22:5399–5407. doi: 10.1038/sj.onc.1206849. [DOI] [PubMed] [Google Scholar]
- 45.Zhang Q, Wang HY, Liu X, Wasik MA. STAT5A is epigenetically silenced by the tyrosine kinase NPM1-ALK and acts as a tumor suppressor by reciprocally inhibiting NPM1-ALK expression. Nat Med. 2007;13:1341–1348. doi: 10.1038/nm1659. [DOI] [PubMed] [Google Scholar]
- 46.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. doi: 10.1182/blood-2004-03-1151. [DOI] [PubMed] [Google Scholar]
- 47.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. doi: 10.1182/blood-2006-04-017434. [DOI] [PubMed] [Google Scholar]
- 48.Han Y, Amin HM, Frantz C, Franko B, Lee J, Lin Q, Lai R. 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. doi: 10.1038/sj.leu.2404323. [DOI] [PubMed] [Google Scholar]
- 49.Stein H, Mason DY, Gerdes J, O'Connor N, Wainscoat J, Pallesen G, Gatter K, Falini B, Delsol G, Lemke H, Schwarting R, Lennert K. 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. [PubMed] [Google Scholar]
- 50.Foss HD, Anagnostopoulos I, Araujo I, Assaf C, Demel G, Kummer JA, Hummel M, Stein H. Anaplastic large-cell lymphomas of T-cell and null-cell phenotype express cytotoxic molecules. Blood. 1996;88:4005–4011. [PubMed] [Google Scholar]
- 51.Kaneko Y, Frizzera G, Edamura S, Maseki N, Sakurai M, Komada Y, Sakurai M, Tanaka H, Sasaki M, Suchi T. A novel translocation, t(2;5) (p23;q35), in childhood phagocytic large T-cell lymphoma mimicking malignant histiocytosis. Blood. 1989;73:806–813. [PubMed] [Google Scholar]
- 52.Le Beau MM, Bitter MA, Larson RA, Doane LA, Ellis ED, Franklin WA, Rubin CM, Kadin ME, Vardiman JW. The t(2;5)(p23;q35): a recurring chromosomal abnormality in Ki-1-positive anaplastic large cell lymphoma. Leukemia. 1989;3:866–870. [PubMed] [Google Scholar]
- 53.Swerdlow SH, Campo E, Harris NL, et al. WHO classification of tumors of hematopoietic and lymphoid tissues. 2008. [Google Scholar]
- 54.Greer JP, Kinney MC, Collins RD, Salhany KE, Wolff SN, Hainsworth JD, Flexner JM, Stein RS. Clinical features of 31 patients with Ki-1 anaplastic large-cell lymphoma. J. Clin. Oncol. 1991;9:539–547. doi: 10.1200/JCO.1991.9.4.539. [DOI] [PubMed] [Google Scholar]
- 55.Falini B, Pileri S, Zinzani PL, Carbone A, Zagonel V, Wolf-Peeters C, Verhoef G, Menestrina F, Todeschini G, Paulli M, Lazzarino M, Giardini R, Aiello A, Foss HD, Araujo I, Fizzotti M, Pelicci PG, Flenghi L, Martelli MF, Santucci A. ALK+ lymphoma: clinico-pathological findings and outcome. Blood. 1999;93:2697–2706. [PubMed] [Google Scholar]
- 56.Stein H, Foss HD, Durkop H, Marafioti T, Delsol G, Pulford K, Pileri S, Falini B. CD30(+) anaplastic large cell lymphoma: a review of its histopathologic, genetic, and clinical features. Blood. 2000;96:3681–3695. [PubMed] [Google Scholar]
- 57.Medeiros LJ, Elenitoba-Johnson KS. Anaplastic Large Cell Lymphoma. Am J Clin Pathol. 2007;127:707–722. doi: 10.1309/r2q9ccuvtlrycf3h. [DOI] [PubMed] [Google Scholar]
- 58.Delsol G, Al Saati T, Gatter KC, Gerdes J, Schwarting R, Caveriviere P, Rigal-Huguet F, Robert A, Stein H, Mason DY. Coexpression of epithelial membrane antigen (EMA), Ki-1, and interleukin-2 receptor by anaplastic large cell lymphomas. Diagnostic value in so-called malignant histiocytosis. Am J Pathol. 1988;130:59–70. [PMC free article] [PubMed] [Google Scholar]
- 59.Brousset P, Rochaix P, Chittal S, Rubie H, Robert A, Delsol G. High incidence of Epstein-Barr virus detection in Hodgkin's disease and absence of detection in anaplastic large-cell lymphoma in children. Histopathology. 1993;23:189–191. doi: 10.1111/j.1365-2559.1993.tb00480.x. [DOI] [PubMed] [Google Scholar]
- 60.Herling M, Rassidakis GZ, Jones D, Schmitt-Graeff A, Sarris AH, Medeiros LJ. Absence of Epstein-Barr virus in anaplastic large cell lymphoma: a study of 64 cases classified according to World Health Organization criteria. Hum Pathol. 2004;35:455–459. doi: 10.1016/j.humpath.2003.10.013. [DOI] [PubMed] [Google Scholar]
- 61.Gascoyne RD, Aoun P, Wu D, Chhanabhai M, Skinnider BF, Greiner TC, Morris SW, Connors JM, Vose JM, Viswanatha DS, Coldman A, Weisenburger DD. Prognostic significance of anaplastic lymphoma kinase (ALK) protein expression in adults with anaplastic large cell lymphoma. Blood. 1999;93:3913–3921. [PubMed] [Google Scholar]
- 62.Savage KJ, Harris NL, Vose JM, Ullrich F, Jaffe ES, Connors JM, Rimsza L, Pileri SA, Chhanabhai M, Gascoyne RD, Armitage JO, Weisenburger DD. International Peripheral T-Cell Lymphoma Project. ALK- anaplastic large-cell lymphoma is clinically and immunophenotypically different from both ALK+ ALCL and peripheral T-cell lymphoma, not otherwise specified: report from the International Peripheral T-Cell Lymphoma Project. Blood. 2008;111:5496–5504. doi: 10.1182/blood-2008-01-134270. [DOI] [PubMed] [Google Scholar]
- 63.Delsol G, Lamant L, Mariame B, Pulford K, Dastugue N, Brousset P, Rigal-Huguet F, al Saati T, Cerretti DP, Morris SW, Mason DY. A new subtype of large B-cell lymphoma expressing the ALK kinase and lacking the 2; 5 translocation. Blood. 1997;89:1483–1490. [PubMed] [Google Scholar]
- 64.Laurent C, Do C, Gascoyne RD, Lamant L, Ysebaert L, Laurent G, Delsol G, Brousset P. Anaplastic lymphoma kinase-positive diffuse large B-cell lymphoma: a rare clinicopathologic entity with poor prognosis. J. Clin. Oncol. 2009;27:4211–4216. doi: 10.1200/JCO.2008.21.5020. [DOI] [PubMed] [Google Scholar]
- 65.Gascoyne RD, Lamant L, Martin-Subero JI, Lestou VS, Harris NL, Müller-Hermelink HK, Seymour JF, Campbell LJ, Horsman DE, Auvigne I, Espinos E, Siebert R, Delsol G. ALK-positive diffuse large B-cell lymphoma is associated with Clathrin-ALK rearrangements: report of 6 cases. Blood. 2003;102:2568–2573. doi: 10.1182/blood-2003-03-0786. [DOI] [PubMed] [Google Scholar]
- 66.Takeuchi K, Soda M, Togashi Y, Ota Y, Sekiguchi Y, Hatano S, Asaka R, Noguchi M, Mano H. Identification of a novel fusion, SQSTM1-ALK, in ALK-positive large B-cell lymphoma. Haematologica. 2010;96:464–467. doi: 10.3324/haematol.2010.033514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Van Roosbroeck K, Cools J, Dierickx D, Thomas J, Vandenberghe P, Stul M, Delabie J, De Wolf-Peeters C, Marynen P, Wlodarska I. ALK-positive large B-cell lymphomas with cryptic SEC31A-ALK and NPM1-ALK fusions. Haematologica. 2010;95:509–513. doi: 10.3324/haematol.2009.014761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Shi M, Minehart Miron P, Hutchinson L, Woda BA, Nath R, Cerny J, Yu H. Anaplastic lymphoma kinase-positive large B-cell lymphoma with complex karyotype and novel ALK gene rearrangements. Hum Pathol. 2011;42:1562–1567. doi: 10.1016/j.humpath.2011.01.012. [DOI] [PubMed] [Google Scholar]
- 69.Reichard KK, McKenna RW, Kroft SH. ALK-positive diffuse large B-cell lymphoma: report of four cases and review of the literature. Mod Pathol. 2007;20:310–319. doi: 10.1038/modpathol.3800742. [DOI] [PubMed] [Google Scholar]
- 70.Lee HW, Kim K, Kim W, Ko YH. ALK-positive diffuse large B-cell lymphoma: report of three cases. Hematol Oncol. 2008;26:108–113. doi: 10.1002/hon.841. [DOI] [PubMed] [Google Scholar]
- 71.Wang WY, Gu L, Liu WP, Li GD, Liu HJ, Ma ZG. ALK-positive extramedullary plasmacytoma with expression of the CLTC-ALK fusion transcript. Pathol Res Pract. 2011;207:587–591. doi: 10.1016/j.prp.2011.07.001. [DOI] [PubMed] [Google Scholar]
- 72.Chiarle R, Gong JZ, Guasparri I, Pesci A, Cai J, Liu J, Simmons WJ, Dhall G, Howes J, Piva R, Inghirami G. NPM-ALK transgenic mice spontaneously develop T-cell lymphomas and plasma cell tumors. Blood. 2003;101:1919–1927. doi: 10.1182/blood-2002-05-1343. [DOI] [PubMed] [Google Scholar]
- 73.Gleason BC, Hornick JL. Inflammatory myofibroblastic tumours: where are we now? J Clin Pathol. 2008;61:428–437. doi: 10.1136/jcp.2007.049387. [DOI] [PubMed] [Google Scholar]
- 74.Yi E, Aubry MC. Pulmonary pseudoneoplasms. Arch Pathol Lab Med. 2010;134:417–426. doi: 10.5858/134.3.417. [DOI] [PubMed] [Google Scholar]
- 75.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. [PubMed] [Google Scholar]
- 76.Coffin CM, Watterson J, Priest JR, Dehner LP. Extrapulmonary inflammatory myofibroblastic tumor (inflammatory pseudotumor). A clinicopathologic and immunohistochemical study of 84 cases. Am J Surg Pathol. 1995;19:859–872. doi: 10.1097/00000478-199508000-00001. [DOI] [PubMed] [Google Scholar]
- 77.Janik JS, Janik JP, Lovell MA, Hendrickson RJ, Bensard DD, Greffe BS. Recurrent inflammatory pseudotumors in children. J Pediatr Surg. 2003;38:1491–1495. doi: 10.1016/s0022-3468(03)00501-3. [DOI] [PubMed] [Google Scholar]
- 78.Cook JR, Dehner LP, Collins MH, Ma Z, Morris SW, Coffin CM, Hill DA. Anaplastic lymphoma kinase (ALK) expression in the inflammatory myofibroblastic tumor: a comparative immunohistochemical study. Am J Surg Pathol. 2001;25:1364–1371. doi: 10.1097/00000478-200111000-00003. [DOI] [PubMed] [Google Scholar]
- 79.Coffin CM, Patel A, Perkins S, Elenitoba-Johnson KS, Perlman E, Griffin CA. ALK1 and p80 expression and chromosomal rearrangements involving 2p23 in inflammatory myofibroblastic tumor. Mod Pathol. 2001;14:569–576. doi: 10.1038/modpathol.3880352. [DOI] [PubMed] [Google Scholar]
- 80.Chun YS, Wang L, Nascimento AG, Moir CR, Rodeberg DA. Pediatric inflammatory myofibroblastic tumor: anaplastic lymphoma kinase (ALK) expression and prognosis. Pediatr Blood Cancer. 2005;45:796–801. doi: 10.1002/pbc.20294. [DOI] [PubMed] [Google Scholar]
- 81.Maris JM, Hogarty MD, Bagatell R, Cohn SL. Neuroblastoma. Lancet. 2007;369:2106–2120. doi: 10.1016/S0140-6736(07)60983-0. [DOI] [PubMed] [Google Scholar]
- 82.Ogawa S, Takita J, Sanada M, Hayashi Y. Oncogenic mutations of ALK in neuroblastoma. Cancer Sci. 2011;102:302–308. doi: 10.1111/j.1349-7006.2010.01825.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Lamant L, Pulford K, Bischof D, Morris SW, Mason DY, Delsol G, Mariamé B. Expression of the ALK tyrosine kinase gene in neuroblastoma. Am J Pathol. 2000;156:1711–1721. doi: 10.1016/S0002-9440(10)65042-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Berthier A, Piqueras M, Villamon E, Berbegall A, Tadeo I, Castel V, Navarro S, Noguera R. Anaplastic lymphoma kinase expression in neuroblastomas and its relationship with genetic, prognostic, and predictive factors. Hum Pathol. 2011;42:301–302. doi: 10.1016/j.humpath.2010.07.024. [DOI] [PubMed] [Google Scholar]
- 85.Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Fujiwara S, Watanabe H, Kurashina K, Hatanaka H, Bando M, Ohno S, Ishikawa Y, Aburatani H, Niki T, Sohara Y, Sugiyama Y, Mano H. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448:561–566. doi: 10.1038/nature05945. [DOI] [PubMed] [Google Scholar]
- 86.Takahashi T, Sonobe M, Kobayashi M, Yoshizawa A, Menju T, Nakayama E, Mino N, Iwakiri S, Sato K, Miyahara R, Okubo K, Manabe T, Date H. Clinicopathologic features of non-small-cell lung cancer with EML4-ALK fusion gene. Ann Surg Oncol. 2010;17:889–897. doi: 10.1245/s10434-009-0808-7. [DOI] [PubMed] [Google Scholar]
- 87.Soda M, Takada S, Takeuchi K, Choi YL, Enomoto M, Ueno T, Haruta H, Hamada T, Yamashita Y, Ishikawa Y, Sugiyama Y, Mano H. A mouse model for EML4-ALK-positive lung cancer. Proc Natl Acad Sci USA. 2008;105:19893–19897. doi: 10.1073/pnas.0805381105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR, Costa DB, Heist RS, Solomon B, Stubbs H, Admane S, McDermott U, Settleman J, Kobayashi S, Mark EJ, Rodig SJ, Chirieac LR, Kwak EL, Lynch TJ, Iafrate AJ. Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK. J. Clin. Oncol. 2009;27:4247–4253. doi: 10.1200/JCO.2009.22.6993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Rodig SJ, Mino-Kenudson M, Dacic S, Yeap BY, Shaw A, Barletta JA, Stubbs H, Law K, Lindeman N, Mark E, Janne PA, Lynch T, Johnson BE, Iafrate AJ, Chirieac LR. Unique clinicopathologic features characterize ALK-rearranged lung adenocarcinoma in the western population. Clin Cancer Res. 2009;15:5216–5223. doi: 10.1158/1078-0432.CCR-09-0802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Koivunen JP, Mermel C, Zejnullahu K, Murphy C, Lifshits E, Holmes AJ, Choi HG, Kim J, Chiang D, Thomas R, Lee J, Richards WG, Sugarbaker DJ, Ducko C, Lindeman N, Marcoux JP, Engelman JA, Gray NS, Lee C, Meyerson M, Jänne PA. EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin Cancer Res. 2008;14:4275–4283. doi: 10.1158/1078-0432.CCR-08-0168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wong DW, Leung EL, So KK, Tam IY, Sihoe AD, Cheng LC, Ho KK, Au JS, Chung LP, Pik Wong M University of Hong Kong Lung Cancer Study Group. The EML4-ALK fusion gene is involved in various histologic types of lung cancers from nonsmokers with wild-type EGFR and KRAS. Cancer. 2009;115:1723–1733. doi: 10.1002/cncr.24181. [DOI] [PubMed] [Google Scholar]
- 92.Lin E, Li L, Guan Y, Soriano R, Rivers CS, Mohan S, Pandita A, Tang J, Modrusan Z. Exon array profiling detects EML4-ALK fusion in breast, colorectal, and non-small cell lung cancers. Mol Cancer Res. 2009;7:1466–1476. doi: 10.1158/1541-7786.MCR-08-0522. [DOI] [PubMed] [Google Scholar]
- 93.Kelleher FC, McDermott R. The emerging pathogenic and therapeutic importance of the anaplastic lymphoma kinase gene. Eur J Cancer. 2010;46:2357–2368. doi: 10.1016/j.ejca.2010.04.006. [DOI] [PubMed] [Google Scholar]
- 94.Inamura K, Takeuchi K, Togashi Y, Hatano S, Ninomiya H, Motoi N, Mun MY, Sakao Y, Okumura S, Nakagawa K, Soda M, Choi YL, Mano H, Ishikawa Y. EML4-ALK lung cancers are characterized by rare other mutations, a TTF-1 cell lineage, an acinar histology, and young onset. Mod Pathol. 2009;22:508–515. doi: 10.1038/modpathol.2009.2. [DOI] [PubMed] [Google Scholar]
- 95.Takeuchi K, Choi YL, Togashi Y, Soda M, Hatano S, Inamura K, Takada S, Ueno T, Yamashita Y, Satoh Y, Okumura S, Nakagawa K, Ishikawa Y, Mano H. KIF5B-ALK, a novel fusion oncokinase identified by an immunohistochemistry-based diagnostic system for ALK-positive lung cancer. Clin Cancer Res. 2009;15:3143–3149. doi: 10.1158/1078-0432.CCR-08-3248. [DOI] [PubMed] [Google Scholar]
- 96.Wong DW, Leung EL, Wong SK, Tin VP, Sihoe AD, Cheng LC, Au JS, Chung LP, Wong MP. A novel KIF5B-ALK variant in nonsmall cell lung cancer. Cancer. 2011;117:2709–2718. doi: 10.1002/cncr.25843. [DOI] [PubMed] [Google Scholar]
- 97.Rikova K, Guo A, Zeng Q, Possemato A, Yu J, Haack H, Nardone J, Lee K, Reeves C, Li Y, Hu Y, Tan Z, Stokes M, Sullivan L, Mitchell J, Wetzel R, Macneill J, Ren JM, Yuan J, Bakalarski CE, Villen J, Kornhauser JM, Smith B, Li D, Zhou X, Gygi SP, Gu TL, Polakiewicz RD, Rush J, Comb MJ. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell. 2007;131:1190–1203. doi: 10.1016/j.cell.2007.11.025. [DOI] [PubMed] [Google Scholar]
- 98.Horn L, Pao W. EML4-ALK: honing in on a new target in non-small-cell lung cancer. J. Clin. Oncol. 2009;27:4232–4235. doi: 10.1200/JCO.2009.23.6661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Yoshida A, Tsuta K, Nakamura H, Kohno T, Takahashi F, Asamura H, Sekine I, Fukayama M, Shibata T, Furuta K, Tsuda H. Comprehensive histologic analysis of ALK-rearranged lung carcinomas. Am J Surg Pathol. 2011;35:1226–1234. doi: 10.1097/PAS.0b013e3182233e06. [DOI] [PubMed] [Google Scholar]
- 100.Martelli MP, Sozzi G, Hernandez L, Pettirossi V, Navarro A, Conte D, Gasparini P, Perrone F, Modena P, Pastorino U, Carbone A, Fabbri A, Sidoni A, Nakamura S, Gambacorta M, Fernández PL, Ramirez J, Chan JK, Grigioni WF, Campo E, Pileri SA, Falini B. EML4-ALK rearrangement in non-small cell lung cancer and non-tumor lung tissues. Am J Pathol. 2009;174:661–670. doi: 10.2353/ajpath.2009.080755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Sasaki T, Rodig SJ, Chirieac LR, Janne PA. The biology and treatment of EML4-ALK non-small cell lung cancer. Eur J Cancer. 2010;46:1773–1780. doi: 10.1016/j.ejca.2010.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Mano H, Takeuchi K. EML4-ALK fusion in lung. Am J Pathol. 2010;176:1552–1553. doi: 10.2353/ajpath.2010.091057. author reply 1553-1554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Boland JM, Erdogan S, Vasmatzis G, Yang P, Tillmans LS, Johnson MR, Wang X, Peterson LM, Halling KC, Oliveira AM, Aubry MC, Yi ES. Anaplastic lymphoma kinase immunoreactivity correlates with ALK gene rearrangement and transcriptional up-regulation in non-small cell lung carcinomas. Hum Pathol. 2009;40:1152–1158. doi: 10.1016/j.humpath.2009.01.012. [DOI] [PubMed] [Google Scholar]
- 104.Inamura K, Takeuchi K, Togashi Y, Nomura K, Ninomiya H, Okui M, Satoh Y, Okumura S, Nakagawa K, Soda M, Choi YL, Niki T, Mano H, Ishikawa Y. EML4-ALK fusion is linked to histological characteristics in a subset of lung cancers. J Thorac Oncol. 2008;3:13–17. doi: 10.1097/JTO.0b013e31815e8b60. [DOI] [PubMed] [Google Scholar]
- 105.Mino-Kenudson M, Chirieac LR, Law K, Hornick JL, Lindeman N, Mark EJ, Cohen DW, Johnson BE, Jänne PA, Iafrate AJ, Rodig SJ. A novel, highly sensitive antibody allows for the routine detection of ALK-rearranged lung adenocarcinomas by standard immunohistochemistry. Clin Cancer Res. 2010;16:1561–1571. doi: 10.1158/1078-0432.CCR-09-2845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Pillay K, Govender D, Chetty R. ALK protein expression in rhabdomyosarcomas. Histopathology. 2002;41:461–467. doi: 10.1046/j.1365-2559.2002.01534.x. [DOI] [PubMed] [Google Scholar]
- 107.Corao DA, Biegel JA, Coffin CM, Barr FG, Wainwright LM, Ernst LM, Choi JK, Zhang PJ, Pawel BR. ALK expression in rhabdomyosarcomas: correlation with histologic subtype and fusion status. Pediatr Dev Pathol. 2009;12:275–283. doi: 10.2350/08-03-0434.1. [DOI] [PubMed] [Google Scholar]
- 108.Li XQ, Hisaoka M, Shi DR, Zhu XZ, Hashimoto H. Expression of anaplastic lymphoma kinase in soft tissue tumors: an immunohistochemical and molecular study of 249 cases. Hum Pathol. 2004;35:711–721. doi: 10.1016/j.humpath.2003.12.004. [DOI] [PubMed] [Google Scholar]
- 109.Jazii FR, Najafi Z, Malekzadeh R, Conrads TP, Ziaee AA, Abnet C, Yazdznbod M, Karkhane AA, Salekdeh GH. Identification of squamous cell carcinoma associated proteins by proteomics and loss of beta tropomyosin expression in esophageal cancer. World J Gastroenterol. 2006;12:7104–7112. doi: 10.3748/wjg.v12.i44.7104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Du XL, Hu H, Lin DC, Xia SH, Shen XM, Zhang Y, Luo ML, Feng YB, Cai Y, Xu X, Han YL, Zhan QM, Wang MR. Proteomic profiling of proteins dysregulted in Chinese esophageal squamous cell carcinoma. J Mol Med (Berl) 2007;85:863–875. doi: 10.1007/s00109-007-0159-4. [DOI] [PubMed] [Google Scholar]
- 111.Debelenko LV, Raimondi SC, Daw N, Shivakumar BR, Huang D, Nelson M, Bridge JA. Renal cell carcinoma with novel VCL-ALK fusion: new representative of ALK-associated tumor spectrum. Mod Pathol. 2011;24:430–442. doi: 10.1038/modpathol.2010.213. [DOI] [PubMed] [Google Scholar]
- 112.Marino-Enriquez A, Ou WB, Weldon CB, Fletcher JA, Perez-Atayde AR. ALK rearrangement in sickle cell trait-associated renal medullary carcinoma. Genes Chromosomes Cancer. 2011;50:146–153. doi: 10.1002/gcc.20839. [DOI] [PubMed] [Google Scholar]
- 113.McDermott U, Settleman J. Personalized cancer therapy with selective kinase inhibitors: an emerging paradigm in medical oncology. J. Clin. Oncol. 2009;27:5650–5659. doi: 10.1200/JCO.2009.22.9054. [DOI] [PubMed] [Google Scholar]
- 114.Piva R, Chiarle R, Manazza AD, Taulli R, Simmons W, Ambrogio C, D'Escamard V, Pellegrino E, Ponzetto C, Palestro G, Inghirami G. Ablation of oncogenic ALK is a viable therapeutic approach for anaplastic large-cell lymphomas. Blood. 2006;107:689–697. doi: 10.1182/blood-2005-05-2125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Sequist LV, Gettinger S, Senzer NN, Martins RG, Jänne PA, Lilenbaum R, Gray JE, Iafrate AJ, Katayama R, Hafeez N, Sweeney J, Walker JR, Fritz C, Ross RW, Grayzel D, Engelman JA, Borger DR, Paez G, Natale R. Activity of IPI-504, a novel heat-shock protein 90 inhibitor, in patients with molecularly defined non-small-cell lung cancer. J. Clin. Oncol. 2010;28:4953–4960. doi: 10.1200/JCO.2010.30.8338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Hallberg B, Palmer RH. Crizotinib--latest champion in the cancer wars? N Engl J Med. 2010;363:1760–1762. doi: 10.1056/NEJMe1010404. [DOI] [PubMed] [Google Scholar]
- 117.Shaw AT, Solomon B. Targeting anaplastic lymphoma kinase in lung cancer. Clin Cancer Res. 2011;17:2081–2086. doi: 10.1158/1078-0432.CCR-10-1591. [DOI] [PubMed] [Google Scholar]
- 118.Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, Ou SH, Dezube BJ, Jänne PA, Costa DB, Varella-Garcia M, Kim WH, Lynch TJ, Fidias P, Stubbs H, Engelman JA, Sequist LV, Tan W, Gandhi L, Mino-Kenudson M, Wei GC, Shreeve SM, Ratain MJ, Settleman J, Christensen JG, Haber DA, Wilner K, Salgia R, Shapiro GI, Clark JW, Iafrate AJ. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2010;363:1693–1703. doi: 10.1056/NEJMoa1006448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Butrynski JE, D'Adamo DR, Hornick JL, Dal Cin P, Antonescu CR, Jhanwar SC, Ladanyi M, Capelletti M, Rodig SJ, Ramaiya N, Kwak EL, Clark JW, Wilner KD, Christensen JG, Jänne PA, Maki RG, Demetri GD, Shapiro GI. Crizotinib in ALK-rearranged inflammatory myofibroblastic tumor. N Engl J Med. 2010;363:1727–1733. doi: 10.1056/NEJMoa1007056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Galkin AV, Melnick JS, Kim S, Hood TL, Li N, Li L, Xia G, Steensma R, Chopiuk G, Jiang J, Wan Y, Ding P, Liu Y, Sun F, Schultz PG, Gray NS, Warmuth M. Identification of NVP-TAE684, a potent, selective, and efficacious inhibitor of NPM-ALK. Proc Natl Acad Sci USA. 2007;104:270–275. doi: 10.1073/pnas.0609412103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Azarova AM, Gautam G, George RE. Emerging importance of ALK in neuroblastoma. Semin Cancer Biol. 2011;21:267–275. doi: 10.1016/j.semcancer.2011.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Cerchietti L, Damm-Welk C, Vater I, Klapper W, Harder L, Pott C, Yang SN, Reiter A, Siebert R, Melnick A, Woessmann W. Inhibition of anaplastic lymphoma kinase (ALK) activity provides a therapeutic approach for CLTC-ALK-positive human diffuse large B cell lymphomas. PLoS One. 2011;6:e18436. doi: 10.1371/journal.pone.0018436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Gerber DE, Minna JD. ALK inhibition for non-small cell lung cancer: from discovery to therapy in record time. Cancer Cell. 2010;18:548–551. doi: 10.1016/j.ccr.2010.11.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Sasaki T, Janne PA. New Strategies for Treatment of ALK Rearranged Non-Small Cell Lung Cancers. Clin Cancer Res. 2011;17:7213–7218. doi: 10.1158/1078-0432.CCR-11-1404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Colleoni GW, Bridge JA, Garicochea B, Liu J, Filippa DA, Ladanyi M. ATIC-ALK: A novel variant ALK gene fusion in anaplastic large cell lymphoma resulting from the recurrent cryptic chromosomal inversion, inv(2)(p23q35) Am J Pathol. 2000;156:781–789. doi: 10.1016/S0002-9440(10)64945-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Ma Z, Cools J, Marynen P, Cui X, Siebert R, Gesk S, Schlegelberger B, Peeters B, De Wolf-Peeters C, Wlodarska I, Morris SW. Inv(2) (p23q35) in anaplastic large-cell lymphoma induces constitutive anaplastic lymphoma kinase (ALK) tyrosine kinase activation by fusion to ATIC, an enzyme involved in purine nucleotide biosynthesis. Blood. 2000;95:2144–2149. [PubMed] [Google Scholar]
- 127.Trinei M, Lanfrancone L, Campo E, Pulford K, Mason DY, Pelicci PG, Falini B. A new variant anaplastic lymphoma kinase (ALK)-fusion protein (ATIC-ALK) in a case of ALK-positive anaplastic large cell lymphoma. Cancer Res. 2000;60:793–798. [PubMed] [Google Scholar]
- 128.Tort F, Pinyol M, Pulford K, Roncador G, Hernandez L, Nayach I, Kluin-Nelemans HC, Kluin P, Touriol C, Delsol G, Mason D, Campo E. Molecular characterization of a new ALK translocation involving moesin (MSN-ALK) in anaplastic large cell lymphoma. Lab Invest. 2001;81:419–426. doi: 10.1038/labinvest.3780249. [DOI] [PubMed] [Google Scholar]
- 129.Adam P, Katzenberger T, Seeberger H, Gattenlöhner S, Wolf J, Steinlein C, Schmid M, Müller-Hermelink HK, Ott G. A case of a diffuse large B-cell lymphoma of plasmablastic type associated with the t(2;5)(p23;q35) chromosome translocation. Am J Surg Pathol. 2003;27:1473–1476. doi: 10.1097/00000478-200311000-00012. [DOI] [PubMed] [Google Scholar]
- 130.Onciu M, Behm FG, Downing JR, Shurtleff SA, Raimondi SC, Ma Z, Morris SW, Kennedy W, Jones SC, Sandlund JT. ALK-positive plasmablastic B-cell lymphoma with expression of the NPM-ALK fusion transcript: report of 2 cases. Blood. 2003;102:2642–2644. doi: 10.1182/blood-2003-04-1095. [DOI] [PubMed] [Google Scholar]
- 131.De Paepe P, Baens M, van Krieken H, Verhasselt B, Stul M, Simons A, Poppe B, Laureys G, Brons P, Vandenberghe P, Speleman F, Praet M, De Wolf-Peeters C, Marynen P, Wlodarska I. ALK activation by the CLTC-ALK fusion is a recurrent event in large B-cell lymphoma. Blood. 2003;102:2638–2641. doi: 10.1182/blood-2003-04-1050. [DOI] [PubMed] [Google Scholar]
- 132.Stachurski D, Miron PM, Al-Homsi S, Hutchinson L, Harris NL, Woda B, Wang SA. Anaplastic lymphoma kinase-positive diffuse large B-cell lymphoma with a complex karyotype and cryptic 3' ALK gene insertion to chromosome 4 q22-24. Hum Pathol. 2007;38:940–945. doi: 10.1016/j.humpath.2006.12.019. [DOI] [PubMed] [Google Scholar]
- 133.Bedwell C, Rowe D, Moulton D, Jones G, Bown N, Bacon CM. Cytogenetically complex SEC31A-ALK fusions are recurrent in ALK-positive large B-cell lymphomas. Haematologica. 2010;96:343–346. doi: 10.3324/haematol.2010.031484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Van Roosbroeck K, Cools J, Dierickx D, Thomas J, Vandenberghe P, Stul M, Delabie J, De Wolf-Peeters C, Marynen P, Wlodarska I. ALK-positive large B-cell lymphomas with cryptic SEC31A-ALK and NPM1-ALK fusions. Haematologica. 2010;95:509–513. doi: 10.3324/haematol.2009.014761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Lawrence B, Perez-Atayde A, Hibbard MK, Rubin BP, Dal Cin P, Pinkus JL, Pinkus GS, Xiao S, Yi ES, Fletcher CD, Fletcher JA. TPM3-ALK and TPM4-ALK oncogenes in inflammatory myofibroblastic tumors. Am J Pathol. 2000;157:377–384. doi: 10.1016/S0002-9440(10)64550-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Bridge JA, Kanamori M, Ma Z, Pickering D, Hill DA, Lydiatt W, Lui MY, Colleoni GW, Antonescu CR, Ladanyi M, Morris SW. Fusion of the ALK gene to the clathrin heavy chain gene, CLTC, in inflammatory myofibroblastic tumor. Am J Pathol. 2001;159:411–415. doi: 10.1016/S0002-9440(10)61711-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Debelenko LV, Arthur DC, Pack SD, Helman LJ, Schrump DS, Tsokos M. Identification of CARS-ALK fusion in primary and metastatic lesions of an inflammatory myofibroblastic tumor. Lab Invest. 2003;83:1255–1265. doi: 10.1097/01.lab.0000088856.49388.ea. [DOI] [PubMed] [Google Scholar]
- 138.Debiec-Rychter M, Marynen P, Hagemeijer A, Pauwels P. ALK-ATIC fusion in urinary bladder inflammatory myofibroblastic tumor. Genes Chromosomes Cancer. 2003;38:187–190. doi: 10.1002/gcc.10267. [DOI] [PubMed] [Google Scholar]
- 139.Ma Z, Hill DA, Collins MH, Morris SW, Sumegi J, Zhou M, Zuppan C, Bridge JA. Fusion of ALK to the Ran-binding protein 2 (RANBP2) gene in inflammatory myofibroblastic tumor. Genes Chromosomes Cancer. 2003;37:98–105. doi: 10.1002/gcc.10177. [DOI] [PubMed] [Google Scholar]
- 140.Panagopoulos I, Nilsson T, Domanski HA, Isaksson M, Lindblom P, Mertens F, Mandahl N. Fusion of the SEC31L1 and ALK genes in an inflammatory myofibroblastic tumor. Int J Cancer. 2006;118:1181–1186. doi: 10.1002/ijc.21490. [DOI] [PubMed] [Google Scholar]