Anaplastic lymphoma kinase fusions: Roles in cancer and therapeutic perspectives (Review)

  • Authors:
    • Zhifa Cao
    • Qian Gao
    • Meixian Fu
    • Nan Ni
    • Yuting Pei
    • Wen‑Bin Ou
  • View Affiliations

  • Published online on: December 20, 2018     https://doi.org/10.3892/ol.2018.9856
  • Pages: 2020-2030
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Receptor tyrosine kinase (RTK) anaplastic lymphoma kinase (ALK) serves a crucial role in brain development. ALK is located on the short arm of chromosome 2 (2p23) and exchange of chromosomal segments with other genes, including nucleophosmin (NPM), echinoderm microtubule‑associated protein‑like 4 (EML4) and Trk‑fused gene (TFG), readily occurs. Such chromosomal translocation results in the formation of chimeric X‑ALK fusion oncoproteins, which possess potential oncogenic functions due to constitutive activation of ALK kinase. These proteins contribute to the pathogenesis of various hematological malignancies and solid tumors, including lymphoma, lung cancer, inflammatory myofibroblastic tumors (IMTs), Spitz tumors, renal carcinoma, thyroid cancer, digestive tract cancer, breast cancer, leukemia and ovarian carcinoma. Targeting of ALK fusion oncoproteins exclusively, or in combination with ALK kinase inhibitors including crizotinib, is the most common therapeutic strategy. As is often the case for small‑molecule tyrosine kinase inhibitors (TKIs), drug resistance eventually develops via an adaptive secondary mutation in the ALK fusion oncogene, or through engagement of alternative signaling mechanisms. The updated mechanisms of a variety of ALK fusions in tumorigenesis, proliferation and metastasis, in addition to targeted therapies are discussed below.

Introduction

Located on chromosome 2p23, receptor tyrosine kinase (RTK) anaplastic lymphoma kinase (ALK) is physiologically expressed in fetal neural cells. Phosphorylated and activated ALK controls the basic mechanisms of cell proliferation, survival and differentiation during development of the nervous system (1). In 1994 ALK t(2;5) chromosomal translocation was reported in anaplastic large cell lymphoma (ALCL) (2). This translocation induced formation of the nucleophosmin (NPM)-ALK chimeric protein (3). Over the ensuing two decades, ALK fusion oncogenes have been associated with the development of diverse tumor types of different lineages, including, but not limited to, lymphoma, lung cancer, inflammatory myofibroblastic tumors (IMTs), Spitz tumors, renal carcinoma, thyroid cancer, digestive tract cancer, breast cancer, leukemia and ovarian carcinoma. During this period, the discovery of EML4-ALK in non-small cell lung cancer (NSCLC) was a major development that led to significant diagnostic and therapeutic advances (4).

In general, ALK fusions arise from fusion of the 3′ end of the ALK gene (exons 20–29) with the 5′portion of a different gene (5). To date, numerous X-ALK fusion oncoproteins have been identified in various tumor types of different lineages. Although targeting ALK fusions markedly promotes tumor shrinkage due to acquisition of activating mutations, genomic rearrangement or copy number amplification of ALK, a subset of patients inevitably acquire resistance to ALK inhibitors. The functional roles of a variety of ALK fusions in neoplasms and targeted therapy advances are summarized below.

ALK rearrangement

In the majority of cancer types, ALK is activated via chromosomal rearrangement. The breakpoint of ALK often occurs at intron 19, which results in dissociation of the 3′ end of exons 20–29 from 5′ end sequences, including the gene promoter, regulatory elements and coding sequences corresponding to the extracellular and transmembrane domains of ALK. The other breakpoint affects a diverse group of genes that contribute to the fusion oncogene, including a different gene promoter and a series of 5′ exons of variable lengths and properties, which predominantly share the ability to self-associate. Additionally, clinical data indicate that different fusion partners affect treatment responses in patients with lung cancer (6). The resulting fusion oncoproteins (X-ALK) are chimeric, self-associating polypeptides with a variety of N-terminal domains and a common, constitutively active C-terminal tyrosine kinase domain (Fig. 1) (5).

In 1994, Morris et al (2), first demonstrated NPM-ALK expression in ALCL. Subsequently, a variety of fusion partners have been found (Table I), including the following: α-2-macroglobulin (A2M); 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase (ATIC); carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (CAD); cysteinyl-tRNA synthetase (CARS); clathrin heavy chain (CLTC); dynactin (DCTN1); echinoderm microtubule-associated protein like-4 (EML4); fibronectin 1 (FN1); huntingtin-interacting protein 1 (HIP1); kinesin family member 5B (KIF5B); kinesin light chain 1 (KLC1); moesin (MSN); non-muscle myosin heavy chain 9 (MYH9); PTPRF interacting protein, binding protein 1 (PPFIBP1); RAN binding protein 2 (RANBP2); ring finger protein 213 (RNF213); SEC31 homolog A (SEC31A); spectrin beta non-erythrocytic 1 (SPTBN1); sequestosome 1 (SQSTM1); striatin (STRN); TRK-fused gene (TFG); tropomyosin 3 (TPM3); tropomyosin 4 (TPM4); translocated promoter region (TPR); TNF receptor-associated factor 1 (TRAF1); and vinculin (VCL).

Table I.

ALK fusion proteins described in diverse tumors.

Table I.

ALK fusion proteins described in diverse tumors.

Gene fusionChromosomal aberrationPartner proteinTumor typeFrequency, %(Refs.)
NPM-ALK t(2;5)(p23;q35)NucleophosminLymphoma45(3,22)
MSN-ALK t(X;2)(q11-12;p23)MoesinLymphoma<1(106)
MYH9-ALK t(2;22)(p23;q11)Non-muscle myosin heavy chain 9Lymphoma<1(107)
RNF213-ALK t(2;17)(p23;q25)Ring finger protein 213Lymphoma<1(108)
TRAF1-ALK t(2;9)(p23;q33.2)Tumor necrosis factor receptor-associated factor 1LymphomaN/A(109)
ATIC-ALKinv(2)(p23q35) 5-aminoimidazole-4-Lymphoma2(110)
carboxamideRibonucleotide formyltransferaseIMT<1(39)
CLTC-ALK t(2;17)(p23;q23)Clathrin heavy chainLymphoma<1(23,108)
IMT13(111)
SQSTM1-ALK t(2;5)(p23.1;q35.3)Sequestosome 1Lymphoma<1(26)
Lung cancer<1(112)
TFG-ALK t(2;3)(p23;q21)Tyrosine kinaseLymphoma<1(113)
receptor-fused geneLung cancer<1(39)
Thyroid cancer2(63)
TPM4-ALK t(2;19)(p23;p13)Tropomyosin 4Lymphoma3(114,115)
IMT17(67)
Digestive tract cancer2
TPM3-ALK t(1;2)(q21;p23)Tropomyosin 3Lymphoma9(115,116)
IMT21(39)
Renal carcinoma<1(53,54)
Spitz tumor6
A2M-ALK t(2;12)(p23;p13) α-2-macroglobulinLung cancer<1(117)
HIP1-ALK t(2;7)(p23;q11.23) Huntingtin-interacting protein 1Lung cancerN/A(118,119)
KIF5B-ALK t(2;10)(p23;p11)Kinesin family member 5BLung cancer<1(40)
KLC1-ALK t(2;14)(p23;q32.1)Kinesin light chain 1Lung cancerN/A(41)
TPR-ALK t(1;2)(q31.1;p23)Translocated promoter regionLung cancerN/A(120)
EML4-ALKinv(2)(p21p23)Echinoderm microtubule-Lung cancer5(4)
associated protein like-4IMT<1(50)
Thyroid cancer2(121)
Renal carcinoma<1(39)
Digestive tract cancerN/A(71)
Breast cancer<1(71)
DCTN1-ALKinv(2)(p13p23)DynactinLung cancer<1(112,122)
IMT<1(123)
Thyroid cancer<1(53,54)
Spitz tumor4
CARS-ALK t(2;11;2)(p23;p15;q31)Cysteinyl-tRNA synthetaseIMT<1(108)
PPFIBP1-ALK t(2;12)(p23;p11)Protein tyrosine phosphatase, receptor type F-interacting protein, binding protein 1IMT<1(124)
SEC31A-ALK t(2;4)(p23;q21)SEC31 homolog AIMT<1(125)
FN1-ALKinv(2)(p23q34)Fibronectin 1IMT<1(126)
Ovarian sarcoma<1(73)
RANBP2-ALK inv(2)(p23q11-13)RAN binding protein 2IMT3(127)
Leukemia<1(72)
STRN-ALK t(2)(p23;p22.2)StriatinThyroid cancer<1   (63,128)
Renal carcinomaN/A
VCL-ALK t(2;10)(p23;q22)VinculinRenal carcinoma<1(59)
CAD-ALK inv(2)(p23;p22)Carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotaseDigestive tract cancer<1(69)
SPTBN1-ALK t(2)(p16.2;p23)Spectrin β non-erythrocytic 1Digestive tract cancer<1(70)

[i] Not all ALK fusions identified worldwide are included; clear statistics are not available for several ALK fusions found in tumors. IMT, inflammatory myofibroblastic tumor; N/A, data unavailable.

The precise mechanisms of ALK gene rearrangement remain unclear. Widely considered a key source of genomic rearrangement, non-homologous end-joining may be divided into 3 steps: i) Generation of double-stranded DNA breaks; ii) ligation of DNA; and iii) gene rearrangement (7,8). Fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC) are widely used in clinical settings to detect ALK rearrangements (911). However, FISH and IHC exhibit low specificity in the recognition of fusion partners, which may be identified by reverse transcription polymerase chain reaction (RT-PCR) or rapid amplification of cDNA ends (RACE)-coupled PCR sequencing (10,12).

Roles of ALK fusion oncoproteins in cancer pathogenesis

Lymphoma

Lymphomas comprise a group of blood cancer types that develop from lymphocytes and are classified as either Hodgkin's lymphoma (HL, 10%) or non-Hodgkin's (NHL, 90%) lymphoma. Based on the normal function of lymphocytes, NHL may be further divided into three subtypes: i) B cell NHL; ii) T cell NHL; and iii) natural killer cell NHL. Compared with HL, NHL patients have a poor prognosis, and the five-year survival rate is ~69% (13,14).

According to certain studies, ALK rearrangements are common in ALCL, which is a type of T cell NHL (15). Statistically, a total of ~90% of ALCLs in children and teenagers, and 50% of ALCLs in adults are ALK-fusion-positive (1618). The most frequent ALK fusion partner is NPM, as the ALK-NPM fusion protein is observed in ~70-80% of all ALCL cases. A total of ~25% cases of ALCL exhibit the TPM3-ALK rearrangement, whereas other rearrangements, including TFG-ALK, ATIC-ALK and CLTC1-ALK, are rare (Table I). Notably, the prognoses of patients with ALK-fusion-positive ALCL are substantially improved compared with those of patients with ALK-fusion-negative ALCL (the five-year survival rate is 70–80% for ALK-fusion-positive patients compared with 15–45% for ALK-fusion-negative patients) (19,20).

Expression of X-ALK was thought to be restricted to ALK-fusion-positive ALCLs; however, in 1997, Delsol et al (21), first demonstrated aberrant expression of NPM-ALK in diffuse large B cell lymphoma (DLBCL). ALK-fusion-positive DLBCL is usually a nodal disease that affects 34~55 years old males, presents at advanced clinical stages and has a poor prognosis (22). The most common ALK rearrangement in DLBCL is t(2;17)(p23;q23), which corresponds to the CLTC-ALK fusion; a minority are NPM-ALK rearrangements (23). Rare cases that harbor SEC31A-ALK and SQSTM1-ALK fusions have also been described (2427).

Lung cancer

Lung cancer is the most prevalent type of cancer and the leading cause of mortality among all malignancies. Despite tremendous progress in the diagnosis and treatment of lung cancer, prognosis for these patients remains poor, with only 15% surviving more than 5 years after initial diagnosis (28). NSCLC accounts for ~80-85% of these cases of lung cancer, whereas the remainder involve small cell lung cancer and lung carcinoid tumors (29).

The EML4-ALK fusion was first observed in 5 out of 75 (6.7%) Japanese patients with NSCLC; notably, these patients did not harbor epidermal growth factor receptor (EGFR) or KRAS mutations (4). Multiple studies have determined the frequency of the EML4-ALK translocation in NSCLC patients, which ranges from 2 to 7% in individual studies, with an average frequency of ~5% (3037). During the past decade, over 11 different variants of EML4-ALK have been identified in a variety of tumors, including NSCLC, digestive tract and breast cancer. The most common variant among EML4-ALK fusions is variant 1 (33%), followed by variant 3 (29%) and variant 2 (10%) (12,38). Furthermore, other ALK fusion partners have been identified in NSCLC, including KLC, TFG, KLC, and KIF5B (3941). ALK-rearranged NSCLC is frequently observed in young patients, in addition to never or former light smokers. Morphologically, acinar, tubulopapillary, cribriform and solid patterns are the most common histological subtypes, and >10% of tumor cells display a distinctive signet ring morphology with abundant intracellular mucin (42). In addition, the oncogenic potential of X-ALK has been confirmed in lung cancer models, including patient-derived cell lines and transgenic mouse models. Several studies have identified the X-ALK gene in a number of NSCLC patients harboring EGFR mutations (38,4346). The majority of these patients are insensitive to the ALK inhibitor crizotinib, but exhibit a partial response to the EGFR inhibitor erlotinib. Therefore, they may not further benefit from coordinated treatment with ALK and EGFR inhibitors compared with either intervention alone.

IMTs

IMT is a type of mesenchymal neoplasm composed of a mixture of several inflammatory cells, which primarily occurs in children (47,48). IMTs are generally benign or low-grade malignant tumors, and patients usually only require surgical treatment (49,50). According to certain statistics, ~50% of IMTs are ALK-fusion-positive, and two of the most common fusion partners are TPM3 and TPM4 (51). Similar to ALCL, various ALK fusion partners have been identified in IMTs, including PPF1BP1, PCTN1, RANBP2, EML4, CLTC, CARS, ATIC, SEC31A and FN1 (Table I). Additionally, a study suggested that patients with ALK-fusion-positive IMT may exhibit a more favorable prognosis compared with those with ALK-fusion-negative IMT (52).

Spitz tumors

Spitz tumors are a type of melanocytic neoplasm that tend to occur in younger people (2–35 years old). Spitz tumors may be divided into three subtypes: i) Benign Spitz nevus; ii) atypical Spitz tumor; and iii) Spitz malignant melanoma (53). In 2014, DCTN1-ALK and TPM3-ALK were identified in Spitz tumors (53,54). Follow-up studies have demonstrated that activation of the X-ALK oncoprotein serves an important role in the pathogenesis of Spitz tumors (55).

Renal carcinoma

Renal carcinoma, a type of tumor that originates from cells in the kidney, accounts for <2% of all cancer types. Renal carcinoma may be divided into two main subtypes: i) renal cell carcinoma (RCC) with a poor prognosis; and ii) transitional cell carcinoma (accounting for 5–10% of cases) (56). Due to the difficulty of early diagnosis in renal carcinomas, their pathogenesis is not completely known. ALK fusions have been documented in a small percentage of RCCs (<1%) (57,58). Based on clinical settings, RCCs with ALK translocation are divided into two categories: i) RCCs with VCL-ALK, composed of sickle cells; and ii) other fusions, which are not associated with sickle cell composition (59,60). In addition to ALK rearrangements, up to 10% of RCC cases show a low level of ALK copy number gains (58). The therapeutic relevance of these findings in RCC is yet to be established.

Thyroid cancer

Thyroid cancer is a common type of endocrine tumor that is classified as either benign thyroid adenoma or a thyroid malignancy (61). Based on the cells that comprise these tumors, thyroid malignancies can be further divided into four subtypes: i) papillary (PTC; 80–85%); ii) follicular (10–15%); iii) medullary (3%); and iv) anaplastic thyroid cancer (ATC; 2%). Among these four types of tumor, the degree of malignance of ATC is high, and its prognosis is poor, with a median patient survival of only 5 months (6264). In 2015, translocations involving ALK were detected by Chou et al (65), in 2.2% of PTC patients. Several other ALK fusion genes have been reported in thyroid cancer, including EML4-ALK, TFG-ALK and STRN-ALK (Table I).

Digestive tract cancer

Digestive tract cancer refers to neoplasms of the digestive system, including cancer of the mouth, esophagus, stomach and intestines. Epidemiological studies have indicated that the frequency of different digestive tract cancer types differs widely in different countries. A recent study illustrated that several factors determine the prognosis of patients with digestive tract cancer, including the location of the tumor, clinical stage and the type of cancer cell (66). In 2006, the TPM4-ALK fusion was first reported in esophageal squamous cell carcinomas (67). Subsequently, other fusion partners have been described in digestive tract cancer, including EML4, CAD and SPTBN1 (6870).

Other neoplasms

Surveys in which a variety of techniques have been applied to a large series of tumors have revealed differentially convincing evidence of ALK rearrangement in rare cases of breast carcinoma (fusions in 5 out of 209 cases assessed by RT-PCR) (71), leukemia (fusions in 3 out of 1,708 cases assessed by RT-PCR) (72) and ovarian carcinoma (3 out of 69 tumors expressed ALK) (73). Although these reports are technically sound, for the most part, the relevance of these findings remains to be clarified through functional studies in pertinent models.

Therapeutic implications

ALK is a compelling therapeutic target, as it is a critical oncogenic driver in diverse tumor types of different lineages. However, its expression and functions are limited in normal tissues. Indeed, Bilsland et al (74) confirmed that ALK double-knockout mice exhibited no significant phenotypic differences, a normal life span, no structurally detectable defects and minor behavioral abnormalities, which advocates a wide non-toxic therapeutic window of ALK-specific inhibition. Various therapeutic methods for tumor treatment are currently in development, including direct targeting of activated ALK with small-molecule inhibitors or immunotherapeutic agents and modulation of downstream signaling intermediates in cancer types with ALK rearrangement. In addition, the X-ALK fusion oncoprotein predominantly activates the RAS/MAPK cell proliferation pathway, in addition to the PI3K/AKT/mTOR and JAK/STAT cell survival pathways. Therefore, an understanding of these downstream effectors has prompted the development of novel therapeutic strategies, some of which are being tested in preclinical/clinical trials.

Multiple structurally distinct ALK drugs are being developed based on a deep understanding of the structure of ALK (Table II), three of which are currently in clinical use for the treatment of ALK-fusion-positive lung cancer, including crizotinib, ceritinib and alectinib. Crizotinib, an oral ALK TKI, has been extensively studied in preclinical and clinical settings. Early phase I studies (PROFILE 1001) have indicted notable activity of crizotinib, with satisfactory tolerability in patients with ALK-fusion-positive NSCLC (75,76). Two-phase III studies further demonstrated the superiority of crizotinib to standard chemotherapy in patients with advanced NSCLC with X-ALK. One of these studies (PROFILE 1007) illustrated that crizotinib treatment significantly prolonged progression-free survival (PFS), which was the primary end point, compared with chemotherapy with either pemetrexed or docetaxel (7.7 vs. 3.0 months, respectively) (77). Another study (PROFILE 1014) compared crizotinib with carboplatin or cisplatin plus pemetrexed in 343 patients with advanced X-ALK NSCLC, and clarified the significance of crizotinib as a first-line treatment for these tumors (78). Furthermore, crizotinib displayed excellent activity in IMT and ALCL cases harboring X-ALK fusions (79).

Table II.

Novel drugs for use in therapies targeting ALK rearrangement tumors.

Table II.

Novel drugs for use in therapies targeting ALK rearrangement tumors.

DrugMolecular targetTumorPhase(Refs.)
Crizotinib NPM-ALK,lung cancerApproved by FDA(7578)
EMLA-ALK, RANBP2-ALKIMTPhase II/III ongoing(129,130)
Ceritinib EML4-ALKlung cancerApproved by FDA(90)
thyroid cancerPhase II/III ongoing(79)
Alectinib EML4-ALKlung cancerApproved by FDA(131,132)
LorlatinibNPM-ALK, EML4-ALKlung cancerPhase I/II ongoing(133,134)
lymphomaPhase I/II ongoing(135)
Entrectinib EML4-ALK,lung cancerPhase I/II ongoing(98)
CAD-ALKdigestive tract cancerPhase I/II ongoing(69)
BrigatinibNPM-ALK, EML4-ALKlung cancerPhase I/II ongoing(136,137)
CEP-28122NPM-ALKlung cancerPreclinical study(138)
lymphomaPreclinical study
TSR-011 EML4-ALKlung cancerPhase I/II ongoing(139)
X-396 EML4-ALKlung cancerPhase I/II ongoing(98)
ASP3026NPM-ALK, EML4-ALKlung cancerPhase I ended(134,140)
lymphomaPhase I ended(96)
Retaspimycin EML4-ALKlung cancerPreclinical study(88,89)
(HSP90 inhibitor)
TanespimycinNPM-ALK, EML4-ALK,lung cancerPreclinical study(141)
(HSP90 inhibitor)TPR-ALK, RANBP2-ALKlymphomaPreclinical study(100)
IMTPreclinical study(84)

[i] Only clinically available drugs are listed; the development of ASP3026 was discontinued due to strategic adjustment of the company. IMT, inflammatory myofibroblastic tumor; HSP90, heat shock protein 90; ALK, anaplastic lymphoma kinase; FDA, Food and Drug Administration.

Despite the excellent efficacy of crizotinib in the setting of NSCLC with ALK translocation, almost all patients developed resistance to crizotinib, but the exact molecular mechanism underlying this phenomenon is yet to be confirmed. The known mechanisms that confer intrinsic or acquired resistance to crizotinib are as follows: i) secondary mutations in the ALK kinase domain (L1152R, C1156Y, I1171T, F1174C/L/V, L1196M, G1202R, S1206Y, E1210K and G1269A/S); ii) ALK gene amplification; and iii) activation of alternative ALK-independent survival pathways, including the EGF signaling pathway, the IGF signaling pathway, the RAS/SRC signaling pathway, and the AKT/mammalian target of rapamycin (mTOR) signaling pathway (8087). Synergistic and/or complementary treatment strategies to overcome resistance are being investigated. Second-generation ALK TKIs, such as ceritinib and alectinib, have been demonstrated to be effective not only in crizotinib-sensitive patients, but also in those who are resistant to crizotinib. Furthermore, other therapeutic options to overcome drug resistance have been proposed, e.g., the use of heat shock protein 90 (HSP90) inhibitors, which can indirectly inhibit ALK fusion (88,89).

Currently, multiple ALK TKIs, including ceritinib, alectinib, lorlatinib, entrectinib, brigatinib, CEP-28122, TSR-011, X-396 and ASP3026, are being investigated as potential therapies for cancer types characterized by ALK rearrangement (Table II). Ceritinib, a highly potent and selective TKI, was approved by the Food and Drug Administration (FDA) as a second-line treatment for patients with X-ALK NSCLC, and following unsuccessful treatment with crizotinib. A total of 114 patients with ALK-fusion-positive NSCLC were enrolled in a global multi-institutional phase I trial, among whom 70% were crizotinib-sensitive and 30% were crizotinib-resistant. All patients received at least 400 mg of crizotinib per day, and the overall response rate (ORR) was 59% (90). Alectinib is a TKI used clinically that exhibits minimal inhibitory activity against kinases other than ALK and RET (91,92). Furthermore, in vitro and in vivo studies have demonstrated that alectinib effectively inhibits ALK with or without the gatekeeper mutation L1196M (92). A separate clinical study was conducted to investigate the safety and activity of alectinib in TKI-naive patients with X-ALK NSCLC, with an ORR of 48% (93). Lorlatinib, which is structurally similar to crizotinib, has been demonstrated to be active against identified crizotinib-resistant ALK mutations, such as the most common mutation seen clinically (G1202R) (94). In 2014, Brigatinib received breakthrough therapy designation from the FDA and a nationwide phase III clinical study in which brigatinib was compared with crizotinib in patients with X-ALK NSCLC was recently initiated (95). Furthermore, the antitumor activities of at least 5 other novel ALK inhibitors, including entrectinib, CEP-28122, TSR-011, X-396 and ASP3026, have been shown in vitro, and these agents are currently under clinical investigation (9698). In addition to targeting ALK directly, several pharmacological strategies allow its indirect targeting. Specifically, HSP90 inhibitors, including retaspimycin and tanespimycin, have displayed certain clinical efficacy in the treatment of patients with ALK rearrangements (84,99,100).

Conclusion

ALK fusions are remarkably versatile oncoproteins that may drive a variety of tumors of different lineages, including, but not limited to, lymphoma, lung cancer, IMTs, Spitz tumors, renal carcinoma, thyroid cancer, digestive tract cancer, breast cancer, leukemia and ovarian carcinoma. Furthermore, a profusion of ALK fusion partners has been consistently identified in ALK-translocated cancer types, which are unique neoplasms that can be effectively targeted by several clinically available TKIs, including crizotinib, ceritinib and alectinib. By using alternative methods of tumor detection, novel ALK translocations may be discovered in upcoming years, which may reveal novel aspects of ALK biology. Substantial efforts are focused on therapeutic considerations and novel approaches to target ALK, including rationally designed tyrosine kinase inhibitors, the study of resistance mechanisms, the design of dual-blockade therapeutic strategies that target downstream signaling intermediates, and immunotherapy against activated receptor tyrosine kinases.

In addition to disease-causing gene mutations, genome-level alterations, including chromosomal imbalances and instability, clonal chromosomal aberrations (CCAs, also known as recurrent karyotypic alterations) and non-clonal chromosome aberrations (NCCAs), also serve a significant role in carcinogenesis and the development of malignant tumors. Since cancer-specific aneuploidy catalyzes karyotypic variation, the degree of aneuploidy predicts the clinical risk of tumor progression. Increasing evidence has indicated the complexity of cancer, which cannot be explained by somatic mutation theory. To address this complexity, additional ad hoc explanations have been postulated, and carcinogenesis is thought to represent a problem of tissue organization on the basis of tissue organization field theory (101103). According to recent studies, chromosomal aberration-mediated genome evolution is responsible for all major transitions in cancer evolution, including phenotypic plasticity, metastasis and drug resistance (104,105). It is believed that the genome serves as the evolutionary platform that links gene/epigene interaction and multiple levels of omics, which can be driven by genome-level alteration rather than individual hallmarks as gene mutation or epigenetic alteration. Conclusively, ongoing research with the aim of characterizing the clinicopathological and biological consequences of ALK rearrangement may allow us to better understand the genome-mediated evolutionary mechanism of cancer.

Acknowledgements

Not applicable.

Funding

The present review was supported by the National Natural Science Foundation of China (grant no. 81728012), the Natural Science Foundation of Zhejiang Province (grant no. LY18H160065), the Zhejiang Medical and Health Science and Technology Plan Project (grant no. 2018260845), the Science Foundation of Zhejiang Sci-Tech University (grant no. 14042107-Y), the National Undergraduate Training Program for Innovation and Entrepreneurship and Graduate Research and Innovation Projects of Zhejiang Sci-Tech University, China.

Availability of data and materials

Not applicable.

Authors' contributions

ZFC and WBO drafted the manuscript. ZFC, QG, MXF, NN and YTP were responsible for the collection of the relevant literature. WBO designed the outline and revised the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Yao S, Cheng M, Zhang Q, Wasik M, Kelsh R and Winkler C: Anaplastic lymphoma kinase is required for neurogenesis in the developing central nervous system of zebrafish. PLoS One. 8:e637572013. View Article : Google Scholar : PubMed/NCBI

2 

Morris SW, Kirstein MN, Valentine MB, Dittmer KG, Shapiro DN, Saltman DL and Look AT: Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma. Science. 263:1281–1284. 1994. View Article : Google Scholar : PubMed/NCBI

3 

Morris SW, Kirstein MN, Valentine MB, Dittmer K, Shapiro DN, Look AT and Saltman DL: Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma. Science. 267:316–317. 1995. View Article : Google Scholar : PubMed/NCBI

4 

Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Fujiwara S, Watanabe H, Kurashina K, Hatanaka H, et al: Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 448:561–566. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Mariño-Enríquez A and Dal Cin P: ALK as a paradigm of oncogenic promiscuity: Different mechanisms of activation and different fusion partners drive tumors of different lineages. Cancer Genet. 206:357–373. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Ninomiya H, Kato M, Sanada M, Takeuchi K, Inamura K, Motoi N, Nagano H, Nomura K, Sakao Y, Okumura S, et al: Allelotypes of lung adenocarcinomas featuring ALK fusion demonstrate fewer onco- and suppressor gene changes. BMC Cancer. 13:82013. View Article : Google Scholar : PubMed/NCBI

7 

Bunting SF and Nussenzweig A: End-joining, translocations and cancer. Nat Rev Cancer. 13:443–454. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Shaw AT and Engelman JA: ALK in lung cancer: Past, present, and future. J Clin Oncol. 31:1105–1111. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Cui S, Zhang W, Xiong L, Pan F, Niu Y, Chu T, Wang H, Zhao Y and Jiang L: Use of capture-based next-generation sequencing to detect ALK fusion in plasma cell-free DNA of patients with non-small-cell lung cancer. Oncotarget. 8:2771–2780. 2017.PubMed/NCBI

10 

Pekar-Zlotin M, Hirsch FR, Soussan-Gutman L, Ilouze M, Dvir A, Boyle T, Wynes M, Miller VA, Lipson D, Palmer GA, et al: Fluorescence in situ hybridization, immunohistochemistry, and next-generation sequencing for detection of EML4-ALK rearrangement in lung cancer. Oncologist. 20:316–322. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Hofman P, Ilie M, Hofman V, Roux S, Valent A, Bernheim A, Alifano M, Leroy-Ladurie F, Vaylet F, Rouquette I, et al: Immunohistochemistry to identify EGFR mutations or ALK rearrangements in patients with lung adenocarcinoma. Ann Oncol. 23:1738–1743. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Li T, Maus MK, Desai SJ, Beckett LA, Stephens C, Huang E, Hsiang J, Zeger G, Danenberg KD, Astrow SH and Gandara DR: Large-scale screening and molecular characterization of EML4-ALK fusion variants in archival non-small-cell lung cancer tumor specimens using quantitative reverse transcription polymerase chain reaction assays. J Thorac Oncol. 9:18–25. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Jaffe ES, Harris NL, Stein H and Vardiman JW: Pathology and genetics of tumours of haematopoietic and lymphoid tissues. IARC Press. 2001.

14 

Cheson BD, Fisher RI, Barrington SF, Cavalli F, Schwartz LH, Zucca E, Lister TA; Alliance, Australasian Leukaemia and Lymphoma Group; Eastern Cooperative Oncology Group; European Mantle Cell Lymphoma Consortium, ; et al: Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: The Lugano classification. J Clin Oncol. 32:3059–3068. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Medeiros LJ and Elenitobajohnson KS: Anaplastic large cell lymphoma. Am J Clin Pathol. 127:707–722. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Gustafson S, Medeiros LJ, Kalhor N and Buesoramos CE: Anaplastic large cell lymphoma: Another entity in the differential diagnosis of small round blue cell tumors. Ann Diagn Pathol. 13:413–427. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Damm-Welk C, Pillon M, Woessmann W and Mussolin L: Prognostic factors in paediatric anaplastic large cell lymphoma: Role of ALK. Front Biosci (Schol Ed). 7:205–216. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Holla VR, Elamin YY, Bailey AM, Johnson AM, Litzenburger BC, Khotskaya YB, Sanchez NS, Zeng J, Shufean MA, Shaw KR, et al: ALK: A tyrosine kinase target for cancer therapy. Cold Spring Harb Mol Case Stud. 3:a0011152017. View Article : Google Scholar : PubMed/NCBI

19 

Savage KJ, Harris NL, Vose JM, Ullrich F, Jaffe ES, Connors JM, Rimsza L, Pileri SA, Chhanabhai M, Gascoyne RD, et al: 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. 111:5496–5504. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Roskoski R Jr: Anaplastic lymphoma kinase (ALK): Structure, oncogenic activation, and pharmacological inhibition. Pharmacol Res. 68:68–94. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Delsol G, Lamant L, Mariamé B, Pulford K, Dastugue N, Brousset P, Rigal-Huguet F, al Saati T, Cerretti DP, Morris SW and Mason DY: A new subtype of large B-cell lymphoma expressing the ALK kinase and lacking the 2; 5 translocation. Blood. 89:1483–1490. 1997.PubMed/NCBI

22 

Laurent C, Do C, Gascoyne RD, Lamant L, Ysebaert L, Laurent G, Delsol G and Brousset P: Anaplastic lymphoma kinase-positive diffuse large B-cell lymphoma: A rare clinicopathologic entity with poor prognosis. J Clin Oncol. 27:4211–4216. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Gascoyne RD, Lamant L, Martin-Subero JI, Lestou VS, Harris NL, Müller-Hermelink HK, Seymour JF, Campbell LJ, Horsman DE, Auvigne I, et al: ALK-positive diffuse large B-cell lymphoma is associated with Clathrin-ALK rearrangements: Report of 6 cases. Blood. 102:2568–2573. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Van Roosbroeck K, Cools J, Dierickx D, Thomas J, Vandenberghe P, Stul M, Delabie J, De Wolf-Peeters C, Marynen P and Wlodarska I: ALK-positive large B-cell lymphomas with cryptic SEC31A-ALK and NPM1-ALK fusions. Haematologica. 95:509–513. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Bedwell C, Rowe D, Moulton D, Jones G, Bown N and Bacon CM: Cytogenetically complex SEC31A-ALK fusions are recurrent in ALK-positive large B-cell lymphomas. Haematologica. 96:343–346. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Takeuchi K, Soda M, Togashi Y, Ota Y, Sekiguchi Y, Hatano S, Asaka R, Noguchi M and Mano H: Identification of a novel fusion, SQSTM1-ALK, in ALK-positive large B-cell lymphoma. Haematologica. 96:464–467. 2011. View Article : Google Scholar : PubMed/NCBI

27 

D'Amore ES, Visco C, Menin A, Famengo B, Bonvini P and Lazzari E: STAT3 pathway is activated in ALK-positive large B-cell lymphoma carrying SQSTM1-ALK rearrangement and provides a possible therapeutic target. Am J Surg Pathol. 37:780–786. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Ettinger DS, Akerley W, Borghaei H, Chang AC, Cheney RT, Chirieac LR, D'Amico TA, Demmy TL, Ganti AK, Govindan R, et al: Non-small cell lung cancer. J Natl Compr Canc Netw. 10:1236–1271. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Koivunen JP, Mermel C, Zejnullahu K, Murphy C, Lifshits E, Holmes AJ, Choi HG, Kim J, Chiang D, Thomas R, et al: EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin Cancer Res. 14:4275–4283. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Rodig SJ, Mino-Kenudson M, Dacic S, Yeap BY, Shaw A, Barletta JA, Stubbs H, Law K, Lindeman N, Mark E, et al: Unique clinicopathologic features characterize ALK-rearranged lung adenocarcinoma in the western population. Clin Cancer Res. 15:5216–5223. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR, Costa DB, Heist RS, Solomon B, Stubbs H, Admane S, McDermott U, et al: Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK. J Clin Oncol. 27:4247–4253. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Li Y, Li Y, Yang T, Wei S, Wang J, Wang M, Wang Y, Zhou Q, Liu H and Chen J: Clinical significance of EML4-ALK fusion gene and association with EGFR and KRAS gene mutations in 208 Chinese patients with non-small cell lung cancer. PLoS One. 8:e520932013. View Article : Google Scholar : PubMed/NCBI

34 

Shaozhang Z, Xiaomei L, Aiping Z, Jianbo H, Xiangqun S and Qitao Y: Detection of EML4-ALK fusion genes in non-small cell lung cancer patients with clinical features associated with EGFR mutations. Genes Chromosomes Cancer. 51:925–932. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Zhang X, Zhang S, Yang X, Yang J, Zhou Q, Yin L, An S, Lin J, Chen S, Xie Z, et al: Fusion of EML4 and ALK is associated with development of lung adenocarcinomas lacking EGFR and KRAS mutations and is correlated with ALK expression. Mol Cancer. 9:1882010. View Article : Google Scholar : PubMed/NCBI

36 

Wong DW, Leung EL, So KK, Tam IY, Sihoe AD, Cheng LC, Ho KK, Au JS, Chung LP and 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. 115:1723–1733. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Guo Y, Ma J, Lyu X, Liu H, Wei B, Zhao J, Fu S, Ding L and Zhang J: Non-small cell lung cancer with EML4-ALK translocation in Chinese male never-smokers is characterized with early-onset. BMC Cancer. 14:8342014. View Article : Google Scholar : PubMed/NCBI

38 

Ou SH, Bartlett CH, Mino-Kenudson M, Cui J and Iafrate AJ: Crizotinib for the treatment of ALK-rearranged non-small cell lung cancer: A success story to usher in the second decade of molecular targeted therapy in oncology. Oncologist. 17:1351–1375. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Rikova K, Guo A, Zeng Q, Possemato A, Yu J, Haack H, Nardone J, Lee K, Reeves C, Li Y, et al: Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell. 131:1190–1203. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Takeuchi K, Choi YL, Togashi Y, Soda M, Hatano S, Inamura K, Takada S, Ueno T, Yamashita Y, Satoh Y, et al: KIF5B-ALK, a novel fusion oncokinase identified by an immunohistochemistry-based diagnostic system for ALK-positive lung cancer. Clin Cancer Res. 15:3143–3149. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Togashi Y, Soda M, Sakata S, Sugawara E, Hatano S, Asaka R, Nakajima T, Mano H and Takeuchi K: KLC1-ALK: A novel fusion in lung cancer identified using a formalin-fixed paraffin-embedded tissue only. PLoS One. 7:e313232012. View Article : Google Scholar : PubMed/NCBI

42 

Nishino M, Klepeis VE, Yeap BY, Bergethon K, Morales-Oyarvide V, Dias-Santagata D, Yagi Y, Mark EJ, Iafrate AJ and Mino-Kenudson M: Histologic and cytomorphologic features of ALK-rearranged lung adenocarcinomas. Mod Pathol. 25:1462–1472. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Lee JK, Kim TM, Koh Y, Lee SH, Kim DW, Jeon YK, Chung DH, Yang SC, Kim YT, Kim YW, et al: Differential sensitivities to tyrosine kinase inhibitors in NSCLC harboring EGFR mutation and ALK translocation. Lung Cancer. 77:460–463. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Yang J, Zhang X, Su J, Chen H, Tian H, Huang Y, Xu C and Wu YL: Concomitant EGFR mutation and EML4-ALK gene fusion in non-small cell lung cancer. J Clin Oncol. 29 Suppl 15:S10517. 2011. View Article : Google Scholar

45 

Popat S, Vieira de Araújo A, Min T, Swansbury J, Dainton M, Wotherspoon A, Lim E, Nicholson AG and O'Brien ME: Lung adenocarcinoma with concurrent exon 19 EGFR mutation and ALK rearrangement responding to erlotinib. J Thorac Oncol. 6:1962–1963. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Kris MG, Johnson BE, Kwiatkowski DJ, Iafrate AJ, Wistuba II, Aronson SL, Engelman JA, Shyr Y, Khuri FR, Rudin CM, et al: Identification of driver mutations in tumor specimens from 1,000 patients with lung adenocarcinoma: The NCI's lung cancer mutation consortium (LCMC). J Clin Oncol. 29:CRA75062011. View Article : Google Scholar

47 

Leuschner I: Inflammatory myofibroblastic tumor. Pathologe. 31:106–108. 2010.(In German). View Article : Google Scholar : PubMed/NCBI

48 

Coffin CM, Watterson J, Priest JR and Dehner LP: Extrapulmonary inflammatory myofibroblastic tumor (inflammatory pseudotumor). A clinicopathologic and immunohistochemical study of 84 cases. Am J Surg Pathol. 19:859–872. 1995. View Article : Google Scholar : PubMed/NCBI

49 

Coffin CM, Hornick JL and Fletcher CD: Inflammatory myofibroblastic tumor: Comparison of clinicopathologic, histologic, and immunohistochemical features including ALK expression in atypical and aggressive cases. Am J Surg Pathol. 31:509–520. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Sokai A, Enaka M, Sokai R, Mori S, Mori S, Gunji M, Fujino M and Ito M: Pulmonary inflammatory myofibroblastic tumor harboring EML4-ALK fusion gene. Jpn J Clin Oncol. 44:93–96. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Griffin CA, Hawkins AL, Dvorak C, Henkle C, Ellingham T and Perlman EJ: Recurrent involvement of 2p23 in inflammatory myofibroblastic tumors. Cancer Res. 59:2776–2780. 1999.PubMed/NCBI

52 

Chun YS, Wang L, Nascimento AG, Moir CR and Rodeberg DA: Pediatric inflammatory myofibroblastic tumor: Anaplastic lymphoma kinase (ALK) expression and prognosis. Pediatr Blood Cancer. 45:796–801. 2005. View Article : Google Scholar : PubMed/NCBI

53 

Busam KJ, Kutzner H, Cerroni L and Wiesner T: Clinical and pathologic findings of Spitz nevi and atypical Spitz tumors with ALK fusions. Am J Surg Pathol. 38:925–933. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Wiesner T, He J, Yelensky R, Esteve-Puig R, Botton T, Yeh I, Lipson D, Otto G, Brennan K, Murali R, et al: Kinase fusions are frequent in Spitz tumours and spitzoid melanomas. Nat Commun. 5:31162014. View Article : Google Scholar : PubMed/NCBI

55 

Yeh I, de la Fouchardiere A, Pissaloux D, Mully TW, Garrido MC, Vemula SS, Busam KJ, LeBoit PE, McCalmont TH and Bastian BC: Clinical, histopathologic, and genomic features of Spitz tumors with ALK fusions. Am J Surg Pathol. 39:581–591. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Seo AN, Yoon G and Ro JY: Clinicopathologic and molecular pathology of collecting duct carcinoma and related renal cell carcinomas. Adv Anat Pathol. 24:65–77. 2017.PubMed/NCBI

57 

Stöhr CG, Amann K and Hartmann A: Histopathologie des Nierenzellkarzinoms. Der Urologe. 52:942–948. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Sukov WR, Hodge JC, Lohse CM, Akre MK, Leibovich BC, Thompson RH and Cheville JC: ALK alterations in adult renal cell carcinoma: Frequency, clinicopathologic features and outcome in a large series of consecutively treated patients. Mod Pathol. 25:1516–1525. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Mariño-Enríquez A, Ou WB, Weldon CB, Fletcher JA and Pérez-Atayde AR: ALK rearrangement in sickle cell trait-associated renal medullary carcinoma. Genes Chromosomes Cancer. 50:146–153. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Debelenko LV, Raimondi SC, Daw N, Shivakumar BR, Huang D, Nelson M and Bridge JA: Renal cell carcinoma with novel VCL-ALK fusion: New representative of ALK-associated tumor spectrum. Mod Pathol. 24:430–442. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Xing M: Molecular pathogenesis and mechanisms of thyroid cancer. Nat Rev Cancer. 13:184–199. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Smallridge RC and Copland JA: Anaplastic thyroid carcinoma: Pathogenesis and emerging therapies. Clin Oncol (R Coll Radiol). 22:486–497. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Kelly LM, Barila G, Liu P, Evdokimova VN, Trivedi S, Panebianco F, Gandhi M, Carty SE, Hodak SP, Luo J, et al: Identification of the transforming STRN-ALK fusion as a potential therapeutic target in the aggressive forms of thyroid cancer. Proc Natl Acad Sci USA. 111:4233–4238. 2014. View Article : Google Scholar : PubMed/NCBI

64 

Baudin E and Schlumberger M: New therapeutic approaches for metastatic thyroid carcinoma. Lancet Oncol. 8:148–156. 2007. View Article : Google Scholar : PubMed/NCBI

65 

Chou A, Fraser S, Toon CW, Clarkson A, Sioson L, Farzin M, Cussigh C, Aniss A, O'Neill C, Watson N, et al: A detailed clinicopathologic study of ALK-translocated papillary thyroid carcinoma. Am J Surg Pathol. 39:652–659. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Rassouli FB, Matin MM and Saeinasab M: Cancer stem cells in human digestive tract malignancies. Tumor Biol. 37:7–21. 2016. View Article : Google Scholar

67 

Jazii FR, Najafi Z, Malekzadeh R, Conrads TP, Ziaee AA, Abnet C, Yazdznbod M, Karkhane AA and Salekdeh GH: Identification of squamous cell carcinoma associated proteins by proteomics and loss of beta tropomyosin expression in esophageal cancer. World J Gastroenterol. 14:7104–7112. 2006. View Article : Google Scholar

68 

Aisner DL, Nguyen TT, Paskulin DD, Le AT, Haney J, Schulte N, Chionh F, Hardingham J, Mariadason J, Tebbutt N, et al: ROS1 and ALK fusions in colorectal cancer, with evidence of intratumoral heterogeneity for molecular drivers. Mol Cancer Res. 12:111–118. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Amatu A, Somaschini A, Cerea G, Bosotti R, Valtorta E, Buonandi P, Marrapese G, Veronese S, Luo D, Hornby Z, et al: Novel CAD-ALK gene rearrangement is drugable by entrectinib in colorectal cancer. Br J Cancer. 113:1730–1734. 2015. View Article : Google Scholar : PubMed/NCBI

70 

Ying J, Lin C, Wu J, Guo L, Qiu T, Ling Y, Shan L, Zhou H, Zhao D, Wang J, et al: Anaplastic lymphoma kinase rearrangement in digestive tract cancer: Implication for targeted therapy in Chinese population. PLoS One. 10:e01447312015. View Article : Google Scholar : PubMed/NCBI

71 

Lin E, Li L, Guan Y, Soriano R, Rivers CS, Mohan S, Pandita A, Tang J and Modrusan Z: Exon array profiling detects EML4-ALK fusion in breast, colorectal, and non-small cell lung cancers. Mol Cancer Res. 7:1466–1476. 2009. View Article : Google Scholar : PubMed/NCBI

72 

Röttgers S, Gombert M, Teigler-Schlegel A, Busch K, Gamerdinger U, Slany R, Harbott J and Borkhardt A: ALK fusion genes in children with atypical myeloproliferative leukemia. Leukemia. 24:1197–1200. 2010. View Article : Google Scholar : PubMed/NCBI

73 

Ren H, Tan ZP, Zhu X, Crosby K, Haack H, Ren JM, Beausoleil S, Moritz A, Innocenti G, Rush J, et al: Identification of anaplastic lymphoma kinase as a potential therapeutic target in ovarian cancer. Cancer Res. 72:3312–3323. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Bilsland JG, Wheeldon A, Mead A, Znamenskiy P, Almond S, Waters KA, Thakur M, Beaumont V, Bonnert TP, Heavens R, et al: Behavioral and neurochemical alterations in mice deficient in anaplastic lymphoma kinase suggest therapeutic potential for psychiatric indications. Neuropsychopharmacology. 33:685–700. 2007. View Article : Google Scholar : PubMed/NCBI

75 

Camidge DR, Bang YJ, Kwak EL, Iafrate AJ, Varella-Garcia M, Fox SB, Riely GJ, Solomon B, Ou SH, Kim DW, et al: Activity and safety of crizotinib in patients with ALK-positive non-small-cell lung cancer: Updated results from a phase 1 study. Lancet Oncol. 13:1011–1019. 2012. View Article : Google Scholar : PubMed/NCBI

76 

Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, Ou SH, Dezube BJ, Jänne PA, Costa DB, et al: Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 363:1693–1703. 2010. View Article : Google Scholar : PubMed/NCBI

77 

Shaw AT, Kim DW, Nakagawa K, Seto T, Crinó L, Ahn MJ, De Pas T, Besse B, Solomon BJ, Blackhall F, et al: Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. 368:2385–2394. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Solomon BJ, Mok T, Kim DW, Wu YL, Nakagawa K, Mekhail T, Felip E, Cappuzzo F, Paolini J, Usari T, et al: First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med. 371:2167–2177. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Godbert Y, Henriques de Figueiredo B, Bonichon F, Chibon F, Hostein I, Pérot G, Dupin C, Daubech A, Belleannée G, Gros A, et al: Remarkable response to crizotinib in woman with anaplastic lymphoma kinase-rearranged anaplastic thyroid carcinoma. J Clin Oncol. 33:e84–e87. 2015. View Article : Google Scholar : PubMed/NCBI

80 

Choi YL, Soda M, Yamashita Y, Ueno T, Takashima J, Nakajima T, Yatabe Y, Takeuchi K, Hamada T, Haruta H, et al: EML4-ALK mutations in lung cancer that confer resistance to ALK inhibitors. N Engl J Med. 363:1734–1739. 2010. View Article : Google Scholar : PubMed/NCBI

81 

Heuckmann JM, Hölzel M, Sos ML, Heynck S, Balke-Want H, Koker M, Peifer M, Weiss J, Lovly CM, Grütter C, et al: ALK mutations conferring differential resistance to structurally diverse ALK inhibitors. Clin Cancer Res. 17:7394–7401. 2011. View Article : Google Scholar : PubMed/NCBI

82 

Doebele RC, Pilling AB, Aisner DL, Kutateladze TG, Le AT, Weickhardt AJ, Kondo KL, Linderman DJ, Heasley LE, Franklin WA, et al: Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer. Clin Cancer Res. 18:1472–1482. 2012. View Article : Google Scholar : PubMed/NCBI

83 

Katayama R, Shaw AT, Khan TM, Mino-Kenudson M, Solomon BJ, Halmos B, Jessop NA, Wain JC, Yeo AT, Benes C, et al: Mechanisms of acquired crizotinib resistance in ALK-rearranged lung cancers. Sci Transl Med. 4:120ra1172012. View Article : Google Scholar

84 

Sasaki T, Okuda K, Zheng W, Butrynski J, Capelletti M, Wang L, Gray NS, Wilner K, Christensen JG, Demetri G, et al: The neuroblastoma associated F1174L ALK mutation causes resistance to an ALK kinase inhibitor in ALK translocated cancers. Cancer Res. 70:10038–10043. 2010. View Article : Google Scholar : PubMed/NCBI

85 

Crystal AS, Shaw AT, Sequist LV, Friboulet L, Niederst MJ, Lockerman EL, Frias RL, Gainor JF, Amzallag A, Greninger P, et al: Patient-derived models of acquired resistance can identify effective drug combinations for cancer. Science. 346:1480–1486. 2014. View Article : Google Scholar : PubMed/NCBI

86 

Ji C, Zhang L, Cheng Y, Patel R, Wu H, Zhang Y, Wang M, Ji S, Belani CP, Yang JM and Ren X: Induction of autophagy contributes to crizotinib resistance in ALK-positive lung cancer. Cancer Biol Ther. 15:570–577. 2014. View Article : Google Scholar : PubMed/NCBI

87 

Mengoli MC, Barbieri F, Bertolini F, Tiseo M and Rossi G: K-RAS mutations indicating primary resistance to crizotinib in ALK-rearranged adenocarcinomas of the lung: Report of two cases and review of the literature. Lung Cancer. 93:55–58. 2016. View Article : Google Scholar : PubMed/NCBI

88 

Sequist LV, Gettinger S, Senzer NN, Martins RG, Jänne PA, Lilenbaum R, Gray JE, Iafrate AJ, Katayama R, Hafeez N, et al: Activity of IPI-504, a novel heat-shock protein 90 inhibitor, in patients with molecularly defined non-small-cell lung cancer. J Clin Oncol. 28:4953–4960. 2010. View Article : Google Scholar : PubMed/NCBI

89 

Normant E, Paez G, West KA, Lim AR, Slocum KL, Tunkey C, McDougall J, Wylie AA, Robison K, Caliri K, et al: The Hsp90 inhibitor IPI-504 rapidly lowers EML4-ALK levels and induces tumor regression in ALK-driven NSCLC models. Oncogene. 30:2581–2586. 2011. View Article : Google Scholar : PubMed/NCBI

90 

Shaw AT, Kim DW, Mehra R, Tan DS, Felip E, Chow LQ, Camidge DR, Vansteenkiste J, Sharma S, De Pas T, et al: Ceritinib in ALK-rearranged non-small-cell lung cancer. N Engl J Med. 370:1189–1197. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Kodama T, Tsukaguchi T, Satoh Y, Yoshida M, Watanabe Y, Kondoh O and Sakamoto H: Alectinib shows potent antitumor activity against RET-rearranged non-small cell lung cancer. Mol Cancer Ther. 13:2910–2918. 2014. View Article : Google Scholar : PubMed/NCBI

92 

Sakamoto H, Tsukaguchi T, Hiroshima S, Kodama T, Kobayashi T, Fukami TA, Oikawa N, Tsukuda T, Ishii N and Aoki Y: CH5424802, a selective ALK inhibitor capable of blocking the resistant gatekeeper mutant. Cancer Cell. 19:679–690. 2011. View Article : Google Scholar : PubMed/NCBI

93 

Shaw AT, Gandhi L, Gadgeel S, Riely GJ, Cetnar J, West H, Camidge DR, Socinski MA, Chiappori A, Mekhail T, et al: Alectinib in ALK-positive, crizotinib-resistant, non-small-cell lung cancer: A single-group, multicentre, phase 2 trial. Lancet Oncol. 17:234–242. 2016. View Article : Google Scholar : PubMed/NCBI

94 

Zou HY, Friboulet L, Kodack DP, Engstrom LD, Li Q, West M, Tang RW, Wang H, Tsaparikos K, Wang J, et al: PF-06463922, an ALK/ROS1 inhibitor, overcomes resistance to first and second generation ALK inhibitors in preclinical models. Cancer Cell. 28:70–81. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Huang WS, Liu S, Zou D, Thomas M, Wang Y, Zhou T, Romero J, Kohlmann A, Li F, Qi J, et al: Discovery of Brigatinib (AP26113), a phosphine oxide-containing, potent, orally active inhibitor of anaplastic lymphoma kinase. J Med Chem. 59:4948–4964. 2016. View Article : Google Scholar : PubMed/NCBI

96 

George SK, Vishwamitra D, Manshouri R, Shi P and Amin HM: The ALK inhibitor ASP3026 eradicates NPM-ALK+ T-cell anaplastic large-cell lymphoma in vitro and in a systemic xenograft lymphoma model. Oncotarget. 5:5750–5763. 2014. View Article : Google Scholar : PubMed/NCBI

97 

Lee J, Kim HC, Hong JY, Wang K, Kim SY, Jang J, Kim ST, Park JO, Lim HY, Kang WK, et al: Detection of novel and potentially actionable anaplastic lymphoma kinase (ALK) rearrangement in colorectal adenocarcinoma by immunohistochemistry screening. Oncotarget. 6:24320–24332. 2015.PubMed/NCBI

98 

Lovly CM, Heuckmann JM, de Stanchina E, Chen H, Thomas RK, Liang C and Pao W: Insights into ALK-driven cancers revealed through development of novel ALK tyrosine kinase inhibitors. Cancer Res. 71:4920–4931. 2011. View Article : Google Scholar : PubMed/NCBI

99 

Sang J, Acquaviva J, Friedland JC, Smith DL, Sequeira M, Zhang C, Jiang Q, Xue L, Lovly CM, Jimenez JP, et al: Targeted inhibition of the molecular chaperone Hsp90 overcomes ALK inhibitor resistance in non-small cell lung cancer. Cancer Discov. 3:430–443. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Bonvini P, Gastaldi T, Falini B and 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 the Hsp90 antagonist 17-allylamino, 17-demethoxygeldanamycin. Cancer Res. 62:1559–1566. 2002.PubMed/NCBI

101 

Bloomfield M and Duesberg P: Inherent variability of cancer-specific aneuploidy generates metastases. Mol Cytogenet. 9:902016. View Article : Google Scholar : PubMed/NCBI

102 

Heng HH, Regan SM, Liu G and Ye CJ: Why it is crucial to analyze non clonal chromosome aberrations or NCCAs? Mol Cytogenet. 9:152016. View Article : Google Scholar : PubMed/NCBI

103 

Ye CJ, Regan S, Liu G, Alemara S and Heng HH: Understanding aneuploidy in cancer through the lens of system inheritance, fuzzy inheritance and emergence of new genome systems. Mol Cytogenet. 11:312018. View Article : Google Scholar : PubMed/NCBI

104 

Bloomfield M and Duesberg P: Is cancer progression caused by gradual or simultaneous acquisitions of new chromosomes? Mol Cytogenet. 11:42018. View Article : Google Scholar : PubMed/NCBI

105 

Horne SD, Pollick SA and Heng HH: Evolutionary mechanism unifies the hallmarks of cancer. Int J Cancer. 136:2012–2021. 2015. View Article : Google Scholar : PubMed/NCBI

106 

Tort F, Pinyol M, Pulford K, Roncador G, Hernandez L, Nayach I, Kluin-Nelemans HC, Kluin P, Touriol C, Delsol G, et al: Molecular characterization of a new ALK translocation involving moesin (MSN-ALK) in anaplastic large cell lymphoma. Lab Invest. 81:419–426. 2001. View Article : Google Scholar : PubMed/NCBI

107 

Lamant L, Gascoyne RD, Duplantier MM, Armstrong F, Raghab A, Chhanabhai M, Rajcan-Separovic E, Raghab J, Delsol G and Espinos E: Non-muscle myosin heavy chain (MYH9): A new partner fused to ALK in anaplastic large cell lymphoma. Genes Chromosomes Cancer. 37:427–432. 2003. View Article : Google Scholar : PubMed/NCBI

108 

Cools J, Wlodarska I, Somers R, Mentens N, Pedeutour F, Maes B, De Wolf-Peeters C, Pauwels P, Hagemeijer A and 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. 34:354–362. 2002. View Article : Google Scholar : PubMed/NCBI

109 

Feldman AL, Vasmatzis G, Asmann YW, Davila J, Middha S, Eckloff BW, Johnson SH, Porcher JC, Ansell SM and Caride A: Novel TRAF1-ALK fusion identified by deep RNA sequencing of anaplastic large cell lymphoma. Genes Chromosomes Cancer. 52:1097–1102. 2013. View Article : Google Scholar : PubMed/NCBI

110 

Trinei M, Lanfrancone L, Campo E, Pulford K, Mason DY, Pelicci PG and 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. 60:793–798. 2000.PubMed/NCBI

111 

Bridge JA, Kanamori M, Ma Z, Pickering D, Hill DA, Lydiatt W, Lui MY, Colleoni GW, Antonescu CR, Ladanyi M and Morris SW: Fusion of the ALK gene to the clathrin heavy chain gene, CLTC, in inflammatory myofibroblastic tumor. Am J Pathol. 159:411–415. 2001. View Article : Google Scholar : PubMed/NCBI

112 

Iyevleva AG, Raskin GA, Tiurin VI, Sokolenko AP, Mitiushkina NV, Aleksakhina SN, Garifullina AR, Strelkova TN, Merkulov VO, Ivantsov AO, et al: Novel ALK fusion partners in lung cancer. Cancer Lett. 362:116–121. 2015. View Article : Google Scholar : PubMed/NCBI

113 

Hernández L, Pinyol M, Hernández S, Beà S, Pulford K, Rosenwald A, Lamant L, Falini B, Ott G, Mason DY, et al: TRK-fused gene (TFG) is a new partner of ALK in anaplastic large cell lymphoma producing two structurally different TFG-ALK translocations. Blood. 94:3265–3268. 1999.PubMed/NCBI

114 

Liang X, Meech SJ, Odom LF, Bitter MA, Ryder JW, Hunger SP, Lovell MA, Meltesen L, Wei Q, Williams SA, et al: Assessment of t(2;5)(p23;q35) translocation and variants in pediatric ALK+ anaplastic large cell lymphoma. Am J Clin Pathol. 121:496–506. 2004. View Article : Google Scholar : PubMed/NCBI

115 

Lawrence B, Perez-Atayde A, Hibbard MK, Rubin BP, Dal Cin P, Pinkus JL, Pinkus GS, Xiao S, Yi ES, Fletcher CD and Fletcher JA: TPM3-ALK and TPM4-ALK oncogenes in inflammatory myofibroblastic tumors. Am J Pathol. 157:377–384. 2000. View Article : Google Scholar : PubMed/NCBI

116 

Lamant L, Dastugue N, Pulford K, Delsol G and Mariamé B: A new fusion gene TPM3-ALK in anaplastic large cell lymphoma created by a (1;2)(q25;p23) translocation. Blood. 93:3088–3095. 1999.PubMed/NCBI

117 

Onoda T, Kanno M, Sato H, Takahashi N, Izumino H, Ohta H, Emura T, Katoh H, Ohizumi H, Ohtake H, et al: Identification of novel ALK rearrangement A2M-ALK in a neonate with fetal lung interstitial tumor. Genes Chromosomes Cancer. 53:865–874. 2014. View Article : Google Scholar : PubMed/NCBI

118 

Ou SH, Klempner SJ, Greenbowe JR, Azada M, Schrock AB, Ali SM, Ross JS, Stephens PJ and Miller VA: Identification of a novel HIP1-ALK fusion variant in non-small-cell lung cancer (NSCLC) and discovery of ALK I1171 (I1171N/S) mutations in two ALK-rearranged NSCLC patients with resistance to Alectinib. J Thorac Oncol. 9:1821–1825. 2014. View Article : Google Scholar : PubMed/NCBI

119 

Fang DD, Zhang B, Gu Q, Lira M, Xu Q, Sun H, Qian M, Sheng W, Ozeck M, Wang Z, et al: HIP1-ALK, a novel ALK fusion variant that responds to crizotinib. J Thorac Oncol. 9:285–294. 2014. View Article : Google Scholar : PubMed/NCBI

120 

Choi YL, Lira ME, Hong M, Kim RN, Choi SJ, Song JY, Pandy K, Mann DL, Stahl JA, Peckham HE, et al: A novel fusion of TPR and ALK in lung adenocarcinoma. J Thorac Oncol. 9:563–566. 2014. View Article : Google Scholar : PubMed/NCBI

121 

Ji JH, Oh YL, Hong M, Yun JW, Lee HW, Kim D, Ji Y, Kim DH, Park WY, Shin HT, et al: Identification of driving ALK fusion genes and genomic landscape of medullary thyroid cancer. PLoS Genet. 11:e10054672015. View Article : Google Scholar : PubMed/NCBI

122 

Wang X, Krishnan C, Nguyen E, Meyer KJ, Oliveira JL, Yang P, Yi ES, Yaszemski MJ, Maran A, Erickson-Johnson MR and Oliveira AM: Fusion of dynactin 1 (DCTN1) to ALK in inflammatory myofibroblastic tumor. Lab Invest. 2011.

123 

Shimada Y, Kohno T, Ueno H, Ino Y, Hayashi H, Nakaoku T, Sakamoto Y, Kondo S, Morizane C, Shimada K, et al: An oncogenic ALK fusion and an RRAS mutation in KRAS mutation-negative pancreatic ductal adenocarcinoma. Oncologist. 22:158–164. 2017. View Article : Google Scholar : PubMed/NCBI

124 

Takeuchi K, Soda M, Togashi Y, Sugawara E, Hatano S, Asaka R, Okumura S, Nakagawa K, Mano H and Ishikawa Y: Pulmonary inflammatory myofibroblastic tumor expressing a novel fusion, PPFIBP1-ALK: Reappraisal of Anti-ALK immunohistochemistry as a tool for novel ALK fusion identification. Clin Cancer Res. 17:3341–3348. 2011. View Article : Google Scholar : PubMed/NCBI

125 

Panagopoulos I, Nilsson T, Domanski HA, Isaksson M, Lindblom P, Mertens F and Mandahl N: Fusion of the SEC31L1 and ALK genes in an inflammatory myofibroblastic tumor. Int J Cancer. 118:1181–1186. 2006. View Article : Google Scholar : PubMed/NCBI

126 

Ouchi K, Miyachi M, Tsuma Y, Tsuchiya K, Iehara T, Konishi E, Yanagisawa A and Hosoi H: FN1: A novel fusion partner of ALK in an inflammatory myofibroblastic tumor. Pediatric Blood Cancer. 62:909–911. 2015. View Article : Google Scholar : PubMed/NCBI

127 

Ma Z, Hill DA, Collins MH, Morris SW, Sumegi J, Zhou M, Zuppan C and Bridge JA: Fusion of ALK to the Ran-binding protein 2 (RANBP2) gene in inflammatory myofibroblastic tumor. Genes Chromosomes Cancer. 37:98–105. 2003. View Article : Google Scholar : PubMed/NCBI

128 

Kusano H, Togashi Y, Akiba J, Moriya F, Baba K, Matsuzaki N, Yuba Y, Shiraishi Y, Kanamaru H, Kuroda N, et al: Two cases of renal cell carcinoma harboring a novel STRN-ALK fusion gene. Am J Surg Pathol. 40:761–769. 2016. View Article : Google Scholar : PubMed/NCBI

129 

Lovly CM, Mcdonald NT, Chen H, Ortiz-Cuaran S, Heukamp LC, Yan Y, Florin A, Ozretić L, Lim D, Wang L, et al: Rationale for co-targeting IGF-1R and ALK inALKfusion positive lung cancer. Nat Med. 20:1027–1034. 2014. View Article : Google Scholar : PubMed/NCBI

130 

Di Paolo D, Yang D, Pastorino F, Emionite L, Cilli M, Daga A, Destafanis E, Di Fiore A, Piaggio F, Brignole C, et al: New therapeutic strategies in neuroblastoma: Combined targeting of a novel tyrosine kinase inhibitor and liposomal siRNAs against ALK. Oncotarget. 6:28774–28789. 2015. View Article : Google Scholar : PubMed/NCBI

131 

Seto T, Kiura K, Nishio M, Nakagawa K, Maemondo M, Inoue A, Hida T, Yamamoto N, Yoshioka H, Harada M, et al: CH5424802 (RO5424802) for patients with ALK-rearranged advanced non-small-cell lung cancer (AF-001JP study): A single-arm, open-label, phase 1–2 study. Lancet Oncol. 14:590–598. 2013. View Article : Google Scholar : PubMed/NCBI

132 

Gadgeel SM, Gandhi L, Riely GJ, Chiappori AA, West HL, Azada MC, Morcos PN, Lee RM, Garcia L, Yu L, et al: Safety and activity of alectinib against systemic disease and brain metastases in patients with crizotinib-resistant ALK-rearranged non-small-cell lung cancer (AF-002JG): Results from the dose-finding portion of a phase 1/2 study. Lancet Oncol. 15:1119–1128. 2014. View Article : Google Scholar : PubMed/NCBI

133 

Johnson TW, Richardson PF, Bailey S, Brooun A, Burke BJ, Collins MR, Cui JJ, Deal JG, Deng YL, Dinh D, et al: Discovery of (10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]-benzoxadiazacyclotetradecine-3-carbonitrile (PF-06463922), a macrocyclic inhibitor of anaplastic lymphoma kinase (ALK) and c-ros oncogene 1 (ROS1) with preclinical brain exposure and broad-spectrum potency against ALK-resistant mutations. J Med Chem. 57:4720–4744. 2014. View Article : Google Scholar : PubMed/NCBI

134 

Mologni L, Ceccon M, Pirola A, Chiriano G, Piazza R, Scapozza L and Gambacorti-Passerini C: NPM/ALK mutants resistant to ASP3026 display variable sensitivity to alternative ALK inhibitors but succumb to the novel compound PF-06463922. Oncotarget. 6:5720–5734. 2015. View Article : Google Scholar : PubMed/NCBI

135 

Basit S, Ashraf Z, Lee K and Latif M: First macrocyclic 3rd-generation ALK inhibitor for treatment of ALK/ROS1 cancer: Clinical and designing strategy update of lorlatinib. Eur J Med Chem. 134:348–356. 2017. View Article : Google Scholar : PubMed/NCBI

136 

Katayama R, Khan TM, Benes C, Lifshits E, Ebi H, Rivera VM, Shakespeare WC, Iafrate AJ, Engelman JA and Shaw AT: Therapeutic strategies to overcome crizotinib resistance in non-small cell lung cancers harboring the fusion oncogene EML4-ALK. Proc Natl Acad Sci USA. 108:7535–7540. 2011. View Article : Google Scholar : PubMed/NCBI

137 

Ceccon M, Mologni L, Bisson W, Scapozza L and Gambacorti-Passerini C: Crizotinib-resistant NPM-ALK mutants confer differential sensitivity to unrelated Alk inhibitors. Mol Cancer Res. 11:122–132. 2013. View Article : Google Scholar : PubMed/NCBI

138 

Cheng M, Quail MR, Gingrich DE, Ott GR, Lu L, Wan W, Albom MS, Angeles TS, Aimone LD, Cristofani F, et al: CEP-28122, a highly potent and selective orally active inhibitor of anaplastic lymphoma kinase with antitumor activity in experimental models of human cancers. Mol Cancer Ther. 11:670–679. 2012. View Article : Google Scholar : PubMed/NCBI

139 

Arkenau HT, Sachdev JC, Mita MM, Dziadziuszko R, Lin CC, Yang JC, Infante JR, Anthony SP, Voskoboynik M, Su WC, et al: Phase (Ph) 1/2a study of TSR-011, a potent inhibitor of ALK and TRK, in advanced solid tumors including crizotinib-resistant ALK positive non-small cell lung cancer. J Clin Oncol. 33:8063. 2015.

140 

Mori M, Ueno Y, Konagai S, Fushiki H, Shimada I, Kondoh Y, Saito R, Mori K, Shindou N, Soga T, et al: The selective anaplastic lymphoma receptor tyrosine kinase inhibitor ASP3026 induces tumor regression and prolongs survival in non-small cell lung cancer model mice. Mol Cancer Ther. 13:329–340. 2014. View Article : Google Scholar : PubMed/NCBI

141 

Katayama R, Friboulet L, Koike S, Lockerman EL, Khan TM, Gainor JF, Iafrate AJ, Takeuchi K, Taiji M, Okuno Y, et al: Two novel ALK mutations mediate acquired resistance to the next-generation ALK inhibitor alectinib. Clin Cancer Res. 20:5686–5696. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 17 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cao Z, Gao Q, Fu M, Ni N, Pei Y and Ou WB: Anaplastic lymphoma kinase fusions: Roles in cancer and therapeutic perspectives (Review). Oncol Lett 17: 2020-2030, 2019
APA
Cao, Z., Gao, Q., Fu, M., Ni, N., Pei, Y., & Ou, W. (2019). Anaplastic lymphoma kinase fusions: Roles in cancer and therapeutic perspectives (Review). Oncology Letters, 17, 2020-2030. https://doi.org/10.3892/ol.2018.9856
MLA
Cao, Z., Gao, Q., Fu, M., Ni, N., Pei, Y., Ou, W."Anaplastic lymphoma kinase fusions: Roles in cancer and therapeutic perspectives (Review)". Oncology Letters 17.2 (2019): 2020-2030.
Chicago
Cao, Z., Gao, Q., Fu, M., Ni, N., Pei, Y., Ou, W."Anaplastic lymphoma kinase fusions: Roles in cancer and therapeutic perspectives (Review)". Oncology Letters 17, no. 2 (2019): 2020-2030. https://doi.org/10.3892/ol.2018.9856