BLU-667

RET fusions in solid tumors
Andrew Y. Lia, Michael G. McCuskerb, Alessandro Russob,c, Katherine A. Scillab, Allison Gittensb,
Katherine Arensmeyerb, Ranee Mehrab, Vincenzo Adamoc, Christian Rolfob,⁎
a Department of Medicine, Division of General Internal Medicine, University of Maryland Medical Center, Baltimore, United States
b Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
c Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Italy

A R T I C L E I N F O

Keywords:
RET
Fusion Rearrangement Solid
LOXO-292 BLU-667 NSCLC
Selpercatinib Pralsetinib
A B S T R A C T

The RET proto-oncogene has been well-studied. RET is involved in many different physiological and develop- mental functions. When altered, RET mutations influence disease in a variety of organ systems from Hirschsprung’s disease and multiple endocrine neoplasia 2 (MEN2) to papillary thyroid carcinoma (PTC) and non-small cell lung cancer (NSCLC). Changes in RET expression have been discovered in 30–70% of invasive breast cancers and 50–60% of pancreatic ductal adenocarcinomas in addition to colorectal adenocarcinoma, melanoma, small cell lung cancer, neuroblastoma, and small intestine neuroendocrine tumors. RET mutations have been associated with tumor proliferation, invasion, and migration. RET fusions or rearrangements are somatic juxtapositions of 5′ sequences from other genes with 3′ RET sequences encoding tyrosine kinase. RET rearrangements occur in approximately 2.5–73% of sporadic PTC and 1–3% of NSCLC patients. The most common RET fusions are CDCC6-RET and NCOA4-RET in PTC and KIF5B-RET in NSCLC. Tyrosine kinase in- hibitors are drugs that target kinases such as RET in RET-driven (RET-mutation or RET-fusion-positive) disease. Multikinase inhibitors (MKI) target various kinases and other receptors. Several MKIs are FDA-approved for cancer therapy (sunitinib, sorafenib, vandetanib, cabozantinib, regorafenib, ponatinib, lenvatinib, alectinib) and non-oncologic disease (nintedanib). Selective RET inhibitor drugs LOXO-292 (selpercatinib) and BLU-667 (pralsetinib) are also undergoing phase I/II and I clinical trials, respectively, with preliminary results demon- strating partial response and low incidence of serious adverse events. RET fusions provide a viable therapeutic target for oncologic treatment, and further study is warranted into the prevalence and pathogenesis of RET fusions as well as development of current and new tyrosine kinase inhibitors.

RET Proto-Oncogene pathway

The RET (REarranged during Transfection) gene derives its name from its discovery via transfection of NIH/3T3 cells with human lym- phoma DNA. RET codes for a transmembrane receptor tyrosine kinase (RTK) with proto-oncogene properties [1]. RET binds with the ligand- co-receptor complex of glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) consisting of GDNF, neurturin (NRTN), artemin (ARTN), or persephin (PSPN) and one of four GDNF family
receptor-α (GFRα). The RET-bound complex is then incorporated into cholesterol-rich transmembrane subdomains known as lipid rafts, where adaptor and signaling proteins bind to RET intracellular tyrosine
kinase residues that have undergone dimerization and autopho- sphorylation [2–6] (Fig. 1). These signaling proteins then bind to docking sites, primarily phosphotyrosine 1062 (pY1062) and pY1096,

leading to activation of signaling pathways such as RAS/mitogen acti- vated protein kinase (MAPK), RAS/extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K)/AKT, and c-Jun N-terminal kinase (JNK) [7–12]. RET signaling is vital for renal morphogenesis, neural and neuroendocrine tissue development, and spermatogonial stem cell maintenance [8,13,14].
RET (REarranged during Transfection) forms a heterodimeric com- plex with GDNF (glial cell line-derived neurotrophic factor) family li- gands GDNF, NRTN (Neurturin), ARTN (Artemin), PSPN (Persephin)
and GDNF family co-receptors GFRα1-4. This leads to autopho- sphorylation of the intracellular tyrosine kinase domain, leading to
downstream signaling pathway activation.
Given its prominent involvement in multi-system tissue develop- ment, RET mutations have similarly been implicated in the progression of several different disorders. Germline nonsense and/or missense

⁎ Corresponding author at: Thoracic Oncology Department and Early Phase Clinical Trials Section, School of Medicine, Maryland University, Maryland, United States. 22 South Greene Street. Baltimore, MD 21201, United States.
E-mail address: [email protected] (C. Rolfo).

https://doi.org/10.1016/j.ctrv.2019.101911

Received 1 September 2019; Received in revised form 20 October 2019; Accepted 21 October 2019
0305-7372/©2019ElsevierLtd.Allrightsreserved.

Fig. 1.

mutations decrease the amount of functional RET receptors on devel- oping gut tissue, leading to the failure of neuroblast migration and enteric nervous system development seen in Hirschsprung’s disease [15–17]. Reduced or loss of function RET mutations have also been associated with congenital anomalies of the kidney and urinary tract (CAKUT) and renal agenesis [18]. Aberrant activation of the RET re- ceptor through gain of function mutations primarily by single amino acid substitution have been associated with multiple endocrine neo- plasia 2 (MEN 2), an autosomal dominant cancer predisposition syn- drome consisting of subtypes Familial MTC (FMTC), MEN2A, and MEN2B. MEN2 consists of three primary tumor types: medullary thyroid cancer, pheochromocytoma, and parathyroid hyperplasia or adenoma [19–22] RET germline mutations have also been identified in nonsyndromic pheochromocytomas [23].

RET expression in solid tumors

Variations in RET expression have been discovered in several dif- ferent solid tumor types. RET has been found to be expressed in 30–70% of invasive breast cancers, and more frequently expressed in ER+, HER2+, and a subset of ER- breast cancers [24–28]. Both RET and GFRA1 have been found to be upregulated and linked to cellular pro- liferation, survival, and scattering [25,29,30]. RET expression is asso- ciated with tamoxifen and aromatase inhibitor resistance in ER+ breast cancers [24,31]. Targeting RET with the multikinase inhibitor vande- tanib potentiated the effect of tamoxifen (selective estrogen receptor modulator), demonstrating greater reduction in tumor growth com- pared to single agent therapy in estrogen-receptor alpha (ERα) positive
breast cancer cells [31]. RET inhibitor NVP-AST487 in combination
with aromatase inhibitor letrozole was effective in inhibiting breast cell line motility and growth [32].
Frequent aberrant methylation of RET is found in colon adenomas and adenocarcinomas, and is associated with decreased RET expression, potentially leading to inhibition of RET-induced apoptosis of colon cancer cells [33]. RET G533C variant was found to promote increased cellular proliferation and migration in colon cancer. In vitro studies demonstrated that vandetanib induced a dose-dependent reduction in RET G533C mutant cell number [34].
In pancreatic cancer, RET expression was linked to lymphatic in- vasion, and found to be significantly higher in patients with lymph node
metastasis. RET was found to be expressed in 50–65% of pancreatic ductal adenocarcinomas [35,36]. In vitro studies demonstrated sig- nificantly lower neural invasion index and invasion severity score of RET knockdown tumors compared with controls, suggesting involve- ment of RET upregulation in pancreatic ductal adenocarcinoma nerve invasion [37].
Positive RET expression is associated with low Fuhrman nuclear grade in papillary renal cell carcinoma [38]. Cytoplasmic and nuclear expression of RET in renal clear cell carcinoma were demonstrated to be strong negative predictors of survival, with high expression correlating with shorter median progression-free survival (PFS) and overall sur- vival (OS). High RET nuclear expression was also found to be an in- dependent predictor of distant and postoperative metastasis in clear cell carcinoma [39]. In prostate cancer, moderately to poorly differentiated tumors (Gleason scores greater than 2) displayed overexpression of RET. Cytoplasmic RET expression was increased in all variants of pro- static intraepithelial neoplasia [40]. In vitro knockdown of RET in prostate cancer cell lines reduced cellular proliferation, invasion, and colony formation. In vivo RET knockdown was associated with de- creased tumor growth. Lenvatinib (multikinase inhibitor including RET) also inhibited invasion and soft agar colony formation in vivo. Furthermore, RET was strongly expressed in small-cell neuroendocrine prostate cancer [41].
The RET G691S polymorphism (RETp) was present in high fre- quency in desmoplastic melanoma, and was shown to interact with GDNF to promote proliferation, migration, and invasion [42,43]. In- creased RFP-RET protein, c-RET, GFRα1, and GDNF levels were noted in malignant murine melanomas compared with benign melanocytic tu- mors [44]. RET was also found to be highly expressed in neuroblastoma
and small-cell lung cancer (SCLC). One RET mutation was associated with increased ERK activation, MYC expression, and cellular pro- liferation in two SCLC cell lines [45]. Tyrosine kinase inhibitors with RET inhibitory activity cabozantinib, regorafenib, sunitinib, and sor- afenib inhibited cell growth of GOT1 metastatic small intestinal neu- roendocrine tumor cells [46]. No longer isolated to the MEN2 syndrome of malignancies, RET is becoming increasingly recognized as an onco- genic contributor to disease and potential therapeutic target.

Detection of RET fusions

RET fusions or rearrangements are a type of somatic mutation leading to formation of distinct RET oncoproteins. RET fusions occur when 5′ sequences from another gene encoding protein dimerization domains juxtapose with RET 3′ sequences that encode the intracellular tyrosine kinase domain via chromosomal inversion or translocation [22]. RET somatic mutations have been increasingly identified in multiple tumor types, and are a focus of study in regards to tumor- igenesis, progression, prognosis, therapy, and therapeutic response. Fluorescence in situ hybridization (FISH) can be used to detect gene fusions, however is limited in single gene assays and is unable to con- sistently identify fusion partners. Real-time PCR assays can detect both fusion drivers and partners, but possesses the same limitation in de- tecting a limited quantity of genes at a time and cannot identify novel fusion partners. Immunohistochemistry (IHC) was found to be limited in detecting RET rearrangements due to variable staining patterns and weak reactivity [47,48].
The introduction of high-throughput next generation sequencing (NGS) of whole genomes or transcriptomes offered simultaneous se- quencing analyses of multiple genes, resulting in high-sensitivity de- tection of multiple mutations including gene fusions and variant allele frequencies. Unfortunately, NGS of whole genomes comes at a high per- sample cost. Targeted NGS addresses this issue by sequencing a selected subset of clinically relevant genes. The gene detection sensitivity of targeted sequencing has been found to meet or exceed that of FISH [49–55]. Several different targeted NGS methodologies have been cre- ated to assess gene fusions [56–58]. The recent FDA authorizations of NGS panels provides increasing accessibility to engage in in- dividualized, precision oncology practice and research [59]. The use of RNA sequencing can allow a more comprehensive detection of action- able gene rearrangements compared with DNA sequencing only and might be considered as a complementary option in apparently driver- negative cases by DNA sequencing [60]. Some gene fusions in fact arise from rearrangements in very long introns or in introns that harbor re- petitive sequence elements also present elsewhere in the genome, making difficult their assessment with DNA sequencing. In addition, RNA sequencing can allow direct evidence that the rearrangement produces a fusion expressed at the mRNA level, overcoming the limits of DNA sequencing when rearrangements appear non-canonical at the genomic DNA level [60].
Liquid biopsy has emerged as a new powerful tool for tumor gen-
otyping to guide the clinical management of advanced solid tumors, including NSCLC [61]. Multiple NGS platforms are now widely avail- able and can allow the identification of less common, but potentially targetable alterations such as activating RET alterations [62]. The identification of gene fusions in plasma is feasible using hybrid capture- based technologies, although differences in hybrid capture techniques and bioinformatic calling may be sources of variations in sensitivity among the different assays available [63].

RET fusions in thyroid cancer

There are at least 13 different RET fusions in PTC, the two most prevalent being coiled-coil domain containing 6 (CCDC6)-RET (also known as RET/PTC1) and nuclear receptor co-activator 4 (NCOA4)-RET (also known as RET/PTC3), which account for > 90% of all re- arrangements [64–69]. Other RET fusions seen in PTC include RET/ PTC2, RET/PTC4 through RET/PTC9, ELKS-RET, PCM1-RET, RFP-RET,
and HOOK3-RET [68], (Fig. 2). Although sparse, RET fusions in MTC have been reported in the literature [70,71]. A targeted 244 cancer- related gene and 20 fusion gene paralleled sequencing assay detected RET fusions in 4.35% of papillary thyroid carcinoma (PTC) samples [72]. Integrated multiplatform analyses performed through The Cancer Genome Atlas (TCGA) network yielded RET fusions in 6.8% of PTC samples [73]. Other studies have shown somatic RET locus

Fig. 2.

rearrangements at chromosome 10q11.2 presenting in 2.5–73% of sporadic PTC and 22–87% of post-Chernobyl PTC, occurring more frequently in childhood than adulthood thyroid cancer.
RET fusions are more prevalent in radiation-exposed populations. RET rearrangements were present in 84% of PTC and 45% of follicular adenomas from patients exposed to external thyroid radiation, with RET/PTC1 being most common followed by RET/PTC3 [74]. RET/PTC rearrangement was found in 22% of PTC patients exposed to radiation from the Hiroshima or Nagasaki atomic bombings. RET fusions oc- curred in higher frequency of 50% in patients with high dose (> 0.5 Gy) exposure, and novel rearrangements have been found in this group [75,76]. RET fusion mutations were detected in 35–69% of Uk- ranian patients with PTC living in Chernobyl-contamined areas at the time of the accident [77,78]. One Chernobyl patient with spontaneous medullary thyroid carcinoma (MTC) was found to have RET re- arrangement of p.Met918Thr alteration, suggesting that radiation ex- posure is not associated with RET fusions in MTC compared to PTC [79].
There are conflicting reports regarding the clinical significance of RET rearrangements in PTC. RET/PTC fusions have been associated with advanced stage disease, more aggressive phenotype, and extra- thyroid extension at diagnosis [80–84]. In contrast, another study found no significant correlation of RET/PTC with clinical aggressive- ness [85]. RET/PTC1 was shown to be associated with a more benign clinical course while RET/PTC3 was associated with more aggressive tumor behavior [67]. Multiple genetico-clinical analyses performed on PTC samples have demonstrated no association between RET/PTC ex- pression and clinico-pathological and prognostic features such as age, sex, thyroid function, lifestyle, tumor size, pT, pN, number of tumor foci, histological subtype, high grade cancers, solid follicular or classic papillary variants [64,86–93]. The important prognostic significance of RET rearrangements remains unclear and represents an unmet area of medical study.

RET fusions in lung cancer

RET mutations, albeit rare, but not RET fusions have been found in neuroendocrine small cell lung cancers (SCLC) [94]. Chimeric RET re- arrangements have been identified in 1–3% of non-small cell lung cancers (NSCLC) in various ethnic populations, and were found to have significantly higher frequencies in younger (< 60 years of age), female, non-smoking, and patients with lung adenocarcinoma histology (LADC) [95–101]. RET fusions in LADC were also associated with poor differ- entiation, solid subtype, smaller T stage (≤3 cm) with N2 disease [102]. The most common RET fusions in lung cancer are kinesin family

[101]. RET/PTC3 has been detected in human malignant pleural me- sothelioma cell line EHMES-10 [114]. RET rearrangements CCDC6-RET, NCOA4-RET, and KIF5B-RET were discovered in 0.2–1.6% of colorectal carcinomas [115,116], defying a new subgroup of patients (RAS/BRAF wild type, right-sided, MSI-high) at poor prognosis with conventional therapeutic strategies [117]. RET rearrangements including CCDC6- RET, NCOA4-RET, and RASGEF1A-RET were detected in 0.16% of breast cancers. Similar to PTC and NSCLC studies, the NCOA4-RET fu- sion detected in this study encoded the characteristic coiled-coil do-
main and RET exons encoding the kinase domain. Expression of
NCOA4-RET and RASGEF1A-RET along with RET amplification in NIH/ 3T3 fibroblasts and MCF10A mammary cells resulted in increased growth capacity and clonogenic expansion, demonstrating constitutive kinase activation. NIH/3T3 cells transduced with NCOA4-RET formed tumors within 2 weeks [118].

Fig. 3.

member 5B (KIF5B)-RET (70–90%) and CCDC6-RET (10–25%), fol- lowed by NCOA4-RET, TRIM33-RET, ZNF477P-RET, ERCC1-RET, HTR4- RET, CLIP1-RET (18%) [23,103–105] (Fig. 3).
The RET tyrosine kinase is preserved across all fusions despite the breakpoint, promoting ligand-independent dimerization, phosphoryla- tion, and constitutive RET activation with resultant intact downstream intracellular kinase activity. Exogenous KIF5B-RET expression was found to confer interleukin-3 (IL-3) independent growth in Ba/F3 cells and induce morphological transformation and anchorage-independent growth in NIH/3T3 fibroblasts. The coiled-coil domain of KIF5B in- duces homodimerization and activates the RET tyrosine kinase domain by autophosphorylation. Drosophila models demonstrated CCDC6-RET and NCOA4-RET-directed cellular migration, delamination, and epi- thelial-mesenchymal transition ultimately leading to death when broadly expressed [106]. LADCs positive for KIF5B-RET were shown to possess two to thirty-fold higher RET expression compared with benign lung tissue, suggesting RET-driven carcinogenesis [95,97,98,103,107,108].
Clinically, both KIF5b-RET and CCDC6-RET rearrangements have been found to co-exist with activated epidermal growth factor receptor (EGFR) mutations in EGFR-mutated NSCLC patients who had pro- gressed on first or second generation EGFR TKI (tyrosine kinase in- hibitors) [109]. EGF (epidermal growth factor) decreased the sensitivity of CCDC6-RET-positive LADC cells, and transduced bypass signaling through extracellular signal-regulated kinases (ERK) and protein kinase
B (AKT) leading to development of resistance to tyrosine kinase in-
RET tyrosine kinase inhibitors

Studies involving RET-targeted therapy primarily involve thyroid cancer and NSCLC. Pyrazolopyrimidine, a RET kinase inhibitor with sedative, anxiolytic, and pesticidal properties, was shown to inhibit RET/PTC1 at IC(50) in the nanomolar range, preventing growth in RET/PTC1-fusion-positive NIH/3T3 fibroblasts and two human PTC cell lines harboring RET/PTC1 [119]. AUY922 is a heat shock protein 90 (HSP90) inhibitor that was found to inhibit downstream RET signaling targets in RET/PTC1 cell lines [120]. Several multikinase inhibitors (MKI) are FDA-approved for cancer therapy (sunitinib, sorafenib, van- detanib, cabozantinib, regorafenib, ponatinib, lenvatinib, alectinib) and non-oncologic diseases (nintedanib), and are being evaluated in clinical trials for a variety tumor types. Several drugs are undergoing clinical trials in RET-fusion-positive malignancies (Table 1). Importantly, a phase I/Ib trial is currently underway for RXDX-105, a VEGFR-sparing RET and BRAF inhibitor [121]. Selective RET inhibitor drugs LOXO-292 (selpercatinib) and BLU-667 (pralsetinib) are also in phase I/II and I clinical trials, respectively [122,123].

Table 1
Studies on Therapies directed towards RET-rearranged Cancers.

Drug Cancer Study Type

Alectinib NSCLC, Thyroid Cancer Phase I/II (NCT03131206) Alectinib NSCLC Phase II (NCT03445000)
Alectinib NSCLC Phase II (NCT02314481)
Apatinib NSCLC Phase II (NCT02540824)
AUY922 NSCLC Phase II (NCT01922583)

hibitors sunitinib, lenvatinib, vandetanib, and sorafenib [110]. CCDC6-
BLU-667 NSCLC, MTC, PTC, Colon Cancer,
and other Solid Tumors
Phase I (NCT03037385)

RET fusion expression was associated with conferring EGFR TKI re- sistance in EGFR-mutant NSCLC cell lines via continuing cellular pro- liferation as well as RET phosphorylation in CCDC6-RET cell lines when
BOS172738 NSCLC Phase I (NCT03780517)
Cabozantinib NSCLC Phase II (NCT01639508)
Cabozantinib NSCLC Phase II (NCT03468985)
Dasatinib Salivary Gland Cancer Phase II (NCT00859937)

treated with osimertinib or afatinib [111]. Some retrospective studies have evaluated the activity of conventional approved therapies in these
Dovitinib Solid and Hematological
Malignancies
Phase II (NCT01831726)

patients. RET rearranged NSCLCs show durable benefits with peme-
Lenvatinib LADC Phase II (NCT01877083)

trexed-based therapies, similarly to other rearranged lung cancers (ALK and ROS1) [112]. Furthermore, RET translocated NSCLCs have an im-
LOXO-292 NSCLC, MTC, PTC, Colon Cancer,
other Solid Tumors
Phase I/II (NCT03157128)

munophenotype “cold” that is associated with low response to immune
Ponatinib NSCLC Phase II (NCT01813734)
Regorafenib Melanoma Phase II (NCT02587650)

checkpoint inhibition (PD-1/PD-L1 +/− CTLA-4 inhibitors), due to low PD-L1 expression and low tumor mutation burden (TMB), con-
RXDX-105 NSCLC
Expanded Access (NCT03784378)

sistent with other oncogene-addicted NSCLCs [113].

RET fusions in other solid tumors
Sunitinib Solid Tumors Pilot Study
(NCT02450123)
Sunitinib Solid Tumors Pilot Study (NCT02691793)

RET fusions are not as extensively studies in solid tumors aside from
Sunitinib MTC, PTC, FTC, Hurthle Cell Carcinoma
Phase II (NCT00381641)

thyroid and lung cancer. One review notes RET fusion expression in small cohorts of lung carcinosarcoma (16.7%), ovarian epithelial car- cinoma (1.9%), salivary gland adenocarcinoma (3.2%), pancreatic ductal carcinoma (0.6%), and carcinoma of unknown primary (0.7%)
Vandetanib NSCLC Phase II (NCT01823068)

RET, rearranged during transfection, LADC, lung adenocarcinoma, NSCLC, non- small cell lung cancer, MTC, medullary thyroid carcinoma, PTC, papillary thyroid carcinoma, FTC, follicular thyroid carcinoma.

There are several preclinical studies on multikinase inhibitors in RET-fusion-positive cell lines. Ba/F3 cells harboring the KIF5B-RET fusions common in RET-fusion-positive NSCLC were found to be sen- sitive to sorafenib [124]. LADC cell line LC-2/ad harboring RET-fusions were sensitive to vandetanib, regorafenib, ponatinib and lenvatinib treatment [115,125,126]. CCDC6-RET-expressing cell lines were sensi- tive to combined therapy of BLU-667 (selective RET inhibitor) or ca- bozantinib (multikinase inhibitor with RET activity) and either afatinib or osimertinib (EGFR inhibitors), suggesting that acquired EGFR TKI resistance from CCDC6-RET fusion can be overcome with dual EGFR and RET inhibition [111]. Cabozantinib and vandetanib inhibited the colony-forming abilities of RET fusion cell lines and lead to colony re- duction, causing rapid tumor regression in mouse models with NCOA4- RET xenografts [118]. Vandetanib was shown to potently inhibit AKT and ERK phosphorylation in metastatic colon cancer patient-derived cells expressing NCOA4-RET fusion [116]. Lenvatinib inhibited autop- hosphorylation of KIF5B-RET, CCDC6-RET, and NCOA4-RET, sup- pressed CCDC6-RET-positive human lung and thyroid cancer cell lines, and suppressed tumorigenicity and anchorage-independent growth of RET-fusion harboring NIH/3T3 cells [127]. Sunitinib suppressed RET/ PTC-fusion-positive PTC cell growth in vitro and in vivo via inhibition of MEK/ERK pathway and G1 arrest [128]. Alectinib demonstrated anti- tumor activity in vivo of RET-fusion-positive mouse models [129]. RXDX-105 inhibited in vitro cell lines harboring CCDC6-RET, NCOA4- RET and PRKAR1A-RET, and also inhibited the in vivo tumor growth of patient-derived xenografts harboring CCDC6-RET, NCOA4-RET and KIF5B-RET [130].
Ongoing clinical trials elaborate on RET-directed therapy efficacy and safety. A prospective phase II trial of 25 RET-rearranged NSCLC patients treated with cabozantinib yielded overall response rate (ORR) of 28%, median progression-free survival (mPFS) of 5.5 months and median overall survival (mOS) of 9.9 months [131]. A phase II trial of 25 RET-rearranged NSCLC patients treated with levantinib resulted in ORR of 16%, mPFS of 7.3 months and non-evaluable mOS [132]. A phase II trial of 17 RET-fusion-positive NSCLC patients treated with vandetanib showed ORR of 18%, mPFS of 4.5 months, and mOS of
⦁ months [133]. A phase II trial of 19 RET-fusion-positive, EGFR- negative NSCLC patients treated with vandetanib resulted in ORR of 47%, mPFS of 6.5 months, and mOS of 13.5 months [134,135]. Of note, the ORR was lower in KIF5B-RET rearranged patients in the two aforementioned trials compared to other RET fusions, and no objective response was observed in KIF5B-RET patients in another trial. Taken together, these four clinical trials [Table 2] show an average ORR of 24,25%, average mPFS of 5.96 months, and average mOS of
⦁ months. Similar trials are currently underway, such as a phase I/II clinical trial of alectinib treatment in RET-rearranged NSCLC in Japan [136].

RET-specific inhibitors

Multikinase inhibitors target several other receptors, and thus are associated with a variety of treatment-emergent adverse events (TEAE) [121]. Resistance to MKIs has been found through development of gatekeeper position mutations that sterically hinder inhibitor binding. It is hypothesized that RET-specific antagonists may produce improved clinical outcomes with potentially less adverse events compared to MKIs [139]. Experimental selective RET inhibitors LOXO-292 (LIBRE- TTO-001 Clinical Trial, Phase I/II, NCT03157128) and BLU-667 (ARROW Clinical Trial, Phase I, NCT03037385) seek to address this issue (Table 3). BLU-667 and LOXO-292 have recently received FDA breakthrough therapy designation for RET-fusion-positive NSCLC after progressing to platinum-based chemotherapy, RET-mutant MTC, and RET-fusion-positive thyroid cancer in patients who require systemic therapy, progressed on prior treatment, or have no other acceptable alternative treatment options [122,123]. Early results show that these two selective RET antagonists are well-tolerated (Table 4). The RET
inhibitor BOS172738 is currently undergoing Phase I clinical trial (NCT03780517) with RET fusion-positive NSCLC, RET mutant MTC, and RET gene-altered NSCLC/MTC with prior specific RET gene-tar- geted therapy. GSK3352589 and GSK3179106 are RET-specific in- hibitors with recently completed Phase I clinical trials designed for ir- ritable bowel syndrome with potential applications for MEN2 and other RET-positive malignancies [140,141].
LOXO-292 was found to be 60 to 1300-fold more effective in in- hibiting KIF5B-RET in engineered cells compared to other targets in- hibited by MKIs. LOXO-292 caused significant tumor regression in RET- fusion-positive mouse models compared to cabozantinib which only caused mild regression. Additionally, intrathecal LOXO-292 caused significantly prolonged survival in intracranial CCDC6-RET-fusion-po- sitive mouse models compared to ponatinib. One patient with meta- static RET-fusion-positive MTC treated with LOXO-292 experienced dramatic decreases in serum carcinoembryonic antigen (CEA) and cal- citonin levels, symptom resolution, and RECIST tumor response of 54% after 6.9 months of treatment. Another patient with metastatic RET- fusion-positive NSCLC treated with LOXO-292 had symptom resolution, RECIST tumor response of 57%, and RANO-BM brain metastases shrinkage or resolution of 89% after 2 months of treatment. LOXO-292 was well-tolerated in both patients and was associated with Grade 1 TEAEs such as fatigue, dyspnea, joint pain, insomnia, and aspartate aminotransferase elevation [137]. Preliminary results from the LIBRE- TTO-001 trial of RET-fusion-positive NSCLC patients treated with LOXO-292 (selpercatinib) were recently presented at the 2019 Internal Association for the Study of Lung Cancer (IASLC) World Conference on Lung Cancer and the 2019 European Society for Medical Oncology (ESMO) Annual Meeting. Selpercatinib was associated with an im- pressive activity in RET fusion-positive NSCLCs, reporting an ORR of 68% in the primary analysis set (PAS), including 105 patients who had received previous platinum-based chemotherapy, and 85% in treat- ment-naïve patients (n = 39). Responses were durable (median dura- tion of response of 20.3 months in the PAS and not reached in treat- ment-naïve patients) with a median PFS of 18.4 months (95% CI, 12.9–24.9) and not reached (95% CI, 9.2-NE) for the PAS and treat- ment-naïve population, respectively [122]. LOXO-292 was overall well- tolerated with mostly Grade 1–2 TEAEs, with only a 1.7% dis- continuation rate [122]. Similar activity was also reported in the RET- mutant MTC and RET fusion-positive thyroid cancer cohorts, with an ORR of 56–59% in RET-mutant MTC pretreated with cabozantinib/ vandetanib and treatment-naïve, respectively, and 62% in RET fusion- positive thyroid cancer [145]. LOXO-292 showed activity also against RET gatekeeper mutations, such as RET V804M and RET V804L, which are responsible of acquired resistance to MKI [122,145]. Based on these impressive results, a new drug application has been submitted to the FDA and two randomized phase III trials versus platinum-peme- trexed ± pembrolizumab in the upfront setting in RET fusion-positive NSCLC and versus cabozantinib/vandetanib in TKI-naïve RET-mutant MTC have been planned.
BLU-667 was found to be 8 to 28-fold more potent against wild-type RET compared with vandetanib, cabozantinib, and RXDX-105, and also displayed greater potency against RET V804L/M, RET M918T, and CCDC6-RET compared with MKIs. BLU-667 also possessed potency against RETV804L, RETV804M, and RETV804E, gatekeeper mutations that can confer MKI resistance, suppressing proliferation in vitro. BLU-667 was found to be at least 100-fold more selective for RET over 96% of tested MKIs, and demonstrated at least 10 times more potency than vandetanib, cabozantinib, and RXDX-105 in inhibiting RET autopho- sphorylation in Ba/F3 cells harboring KIF5B-RET fusion. BLU-667 re- tained this antitumor activity and RET selectivity in vivo. Two patients with MTC treated with BLU-667 showed over 90% reduction in serum calcitonin, and 57–75% decrease in CEA with 19% reduction in target lesion size after 8 weeks, a confirmed partial response of 47% maximal reduction at 10 months in one patient and 35% radiographic response by RECIST after 8 weeks in another. Two patients with NSCLC treated

Table 2
Activity of RET inhibitors in RET fusion positive NSCLCs in phase I/II studies.
Study n Drug RET detection method (s) RET Fusion partners ORR PFS OS
Drilon, 2016 [131] 26 Cabozantinib FISH, hybrid-capture KIF5B-RET 62% 28% (95% CI 5.5 months (95% CI 9.9 months (95% CI
NGS CCDC6-RET 4% 12–49). 3.8–8.4). 8.1–NR)
Unknown 23%
Yoh, 2017 & 2018 19 Vandetanib RT-PCR and FISH KIF5B-RET 53% 47% (95% CI 6.5 months (95% CI 13.5 months (95% CI
[134,135] CCDC6-RET 24–71) 2.8–8.5) 9.8–28.1)
31%
Unknown 16%
Lee, 2017 [133] 18 Vandetanib FISH (10/18 confirmed KIF5B-RET 28% 18% 4.54 months 11.63 months
with other methods) CCDC6-RET
11%
Unknown 55%
Velcheti, 2016 [132] 25* Lenvatinib Not reported KIF5B-RET 52% 16% 7.3 (95% CI NR (5.8–NR)

Oxnard, 2018 [122]
38**
LOXO-292
Not reported Others 48%
KIF5B-RET
68% (95% CI 3.6–10.2)
Not reported
Not reported
60.5% 51–83)
CCDC6-RET
26.3%
Unknown 7.9%
Subbiah, 2018 [137] 19 BLU-667 Not reported Not reported 50% Not reported Not reported
Drilon, 2017 [138] 40*** (22 treatment- RXDX-105 Not reported KIF5B-RET 50% 27% (treatment- Not reported Not reported
naïve, evaluable) CCDC6-RET naïve)
15% 75% (non-KIF5B
Unknown 5% fusions)
Legend. RN, not reached; FISH, fluorescent in situ hybridization; NGS, next generation sequencing; RT-PCR, real-time polymerase chain reaction; IHC, im- munohistochemistry.
* 28% had previously received a RET-targeted therapy.
** 55% had previously received ≥ 1 multi-target inhibitors.
*** 22.5% had previously received a RET-targeted therapy.

with BLU-667 developed 25–34% tumor reduction followed by partial response. All four patients tolerated the treatment well with Grade 1 TEAEs of nausea, constipation, dry skin, rash, leukopenia, and hyper- phosphatemia [137]. Two patients in a separate study with EGFR-mu- tant NSCLC and acquired CCDC6-RET fusion developed RECIST tumor shrinkage of 78% after concurrent osimertinib and BLU-667 treatment [111]. Preliminary results from the ARROW trial of BLU-667 treatment of RET-driven (RET mutation or RET fusion) cancers were presented at the 2018 American Association for Cancer Research (AACR) Annual Meeting (Abstract CT043) and at the 2019 ASCO annual meeting. Over 90% of PTC and MTC patients with measurable target lesions achieved radiographic tumor reduction. The preliminary ORR 60% (DCR 100%) in RET fusion-positive NSCLC previously treated with platinum-based chemotherapy, 63% (94% DCR) in RET-mutated MTC pretreated with cabozantinib or vandetanib, and 83% in RET fusion-positive papillary thyroid cancer. The majority of TEAEs were Grade 1–2 and Grade 3 TEAEs of anemia, hypertension, diarrhea, leukopenia, neutropenia, and increased alanine aminotransferase [123,142,143].
Preliminary data were also reported with the selective RET inhibitor RXDX-105. Based on analysis of safety, efficacy and pharmacokinetics across multiple doses in a phase 1/1b study, 275 mg fed, administered orally once daily, was selected as the RP2D. An expansion cohort of the phase 1b portion of the study enrolled 47 RET fusion positive patients,
including 40 NSCLCs. In treatment-naïve patients (22 patients evalu- able), receiving RXDX-105 at 275 mg or 350 mg/daily, an ORR of 27% was reported, with a differential activity between KIF5B-RET fusions (ORR 0%, albeit 21.4% experienced a SD lasting ≥ 6 months) and non- KIF5B-RET fusions (ORR 75%) [138] (Table 2), further confirming the lower sensitivity of KIF5B-RET fusions to RET-targeted inhibition. RXDX-105 demonstrated a manageable safety profile, with the majority of TEAEs of grade ≤2 and the most frequent AEs of grade ≥3 were rash (10%), hypophosphatemia (7%), increase of ALT/AST (7% and 4%,
respectively), and diarrhea (4%) [138].

Discussion

From its discovery of RET rearrangement over two decades ago to the development of selective RET inhibitors, the RET gene has under- gone extensive study in the field of oncology [1,22]. No longer known solely as the source of germline mutations involved in Hirschsprung’s disease and MEN2, RET has become increasingly recognized as a key driver of tumorigenesis and prognostic indicator or treatment response. RET mutations and RET fusions occur in an abundance of tumor types from papillary thyroid carcinoma to non-small cell lung cancer. RET fusions such as KIF5B-RET, CDCC6-RET, and NCOA4-RET are now being studied in the laboratory and clinical trials in a variety of different

Table 3
Selective RET Inhibitors.
Drug Clinical Trial FDA-Approved Indications

LOXO-292 Phase I/II (NCT03157128) Breakthrough therapy designation for RET-fusion-positive NSCLC, RET-mutant MTC, RET-fusion-positive thyroid cancer
requiring systemic therapy, progressed on prior treatment, or without acceptable alternative treatment options
BLU-667 Phase I (NCT03037385) N/A
BOS172738 Phase I (NCT03780517) N/A
GSK3352589 Phase I (NCT03154086) N/A

GSK3179106 Phase I (NCT02798991,
NCT02727283)
N/A

RET, rearranged during transfection, NSCLC, non-small cell lung cancer, MTC, medullary thyroid carcinoma.

Table 4
Treatment-Emergent Adverse Events of Selective and non-selective RET Inhibitors.
Drug Grade 1 Grade 2 Grade 3 Grade 4 Grade 5 Ref.
LOXO-292 Diarrhea (21%), Fatigue (15%), Dry Mouth (29%), Diarrhea (8%), Fatigue (9%), Constipation (3%), Diarrhea (2%), Fatigue (1%), Constipation Hypertension (< 1%), None [122,145]
(selpercatinib) Constipation (19%), Headache (15%), Nausea Headache (4%), Nausea (4%), Dry mouth (4%), (< 1%), Headache (1%), Nausea (< 1%), Increased AST (1%), Increased
(15%), Hypertension (4%), Increased AST (17%), Hypertension (11%), Increased AST (5%), Hypertension (14%), Increased AST (6%), ALT (1%), Increased
Increased ALT (13%), Peripheral edema (16%), Increased ALT (4%), Peripheral edema (4%), Increased ALT (7%), Peripheral edema (< 1%) creatinine (< 1%)
Increased creatinine (14%) Increased creatinine (4%)
BLU-667 (pralsetinib) Decreased WBC (10%), Neutropenia (7%), Anemia Decreased WBC (15%), Neutropenia (7%), Anemia Decreased WBC (4%), Neutropenia (9%), Anemia Neutropenia (4%) None [143]
(12%), Increased blood Creatinine (28%), (10%), Increased blood Creatinine (1%), Increased (9%), Increased ALT (1%), Hypertension (16%),
Increased ALT (23%), Increased AST (29%), AST (4%), Hypertension (7%), Constipation (3%), Diarrhea (7%), Fatigue (1%), Headache (1%)
Hypertension (7%), Constipation (32%), Diarrhea Diarrhea (4%), Fatigue (4%), Headache (3%)
(16%), Fatigue (13%), Headache (13%)
Vandetanib Diarrhea (26–33%), Rash acneiform (16–39%), Dry skin (21–22%), Prolonged QT corrected interval (0–11%), Anorexia (5–21%), Increased
creatinine (21%), Vomiting (5–21%), Paronychia
(16–17%), Oral mucositis (21%), Nausea (5–11%),
Liver dysfunction (16%), Hypoalbuminemia (5%),
Photosensitivity (11%), Pruritus (16%) Hypertension (26–39%), Diarrhea (5–42%), Rash
acneiform (11–32%), Dry skin (0–16%), Prolonged
QT corrected interval (21%), Anorexia (5%),
Increased creatinine (11%), Vomiting (5%),
Paronychia (11%), Proteinuria (21%), Nausea
(0–5%), Liver dysfunction (5%),
Hypoalbuminemia (16%), Photosensitivity (5%) Hypertension (17–58%), Diarrhea (0–11%), Rash
acneiform (0–16%), Dry skin (0–5%), Prolonged
QT corrected interval (5–11%), Anorexia (0–5%),
Proteinuria (5%), Nausea (0–5%), Photosensitivity
(5%) Prolonged QT corrected interval (5%), None [133,134]
Cabozantinib ALT increased (81%), AST increased (62%), ALT increased (8%), AST increased (4%), ALT increased (8%), AST increased (8%), Palmar- None [131]
Hypothyroidism (15%), Diarrhea (46%), Palmar- Hypothyroidism (54%), Diarrhea (15%), Palmar- plantar erythrodysaesthesia (4%),
plantar erythrodysaesthesia (35%), plantar erythrodysaesthesia (23%), Skin Thrombocytopenia (8%), Fatigue (4%), Oral
Thrombocytopenia (31%), Fatigue (15%), Oral hypopigmentation (50%), Thrombocytopenia mucositis (4%), Lipase increased (15%),
mucositis (42%), Lipase increased (12%), Nausea (12%), Fatigue (27%), Lipase increased (8%), Hypertension (4%)
(19%), Dysgeusia (23%), Amylase increased Nausea (12%), Dysgeusia (8%), Amylase increased
(19%), Anorexia (4%), Dry skin (19%) (8%), Anorexia (15%), Hypertension (15%)

Lenvatinib Hypertension (12%), Nausea (48%), Anorexia (52%), Diarrhea (44%), Proteinuria (32%), Vomiting
(36%), Headache (40%), Fatigue (28%), Thrombocytopenia (24%), ALT increased (20%), AST increased
(24%), Constipation (24%), Cough (24%), Peripheral edema (20%), Hyponatremia (4%)
Hypertension (56%), Nausea (12%), Diarrhea
(8%), Proteinuria (16%), Vomiting (8%), Fatigue
(8%), Thrombocytopenia (4%), Hyponatremia
(12%)
Hyponatremia (8%) None [132]

CancerTreatmentReviews81(2019)101911
A.Y. Li, et al.
7

Treatment-Emergent Adverse Events were assessed via CTCAE, Common Terminology Criteria for Adverse Events. Grade 1, mild, Grade 2, moderate, Grade 3, severe, Grade 4, life-threatening, Grade 5, death, ALT, alanine aminotransferase, AST, aspartate aminotransferase, WBC, white blood cell.

cancers as potential therapeutic targets. Early clinical trial results in NSCLC show encouraging results in treating RET-fusion-positive ma- lignancy with MKIs [131–135]. The majority of TKIs are already FDA- approved for a diverse array of cancers such as GIST, CRC, RCC, HCC, pNET, DTC, MTC, NSCLC, CML and ALL and are well-tolerated, while others such as apatinib and LOXO-292 have received orphan drug and breakthrough therapy designations, respectively by the FDA. Selective TKIs LOXO-292 and BLU-667 have been shown to be efficacious and well tolerated due to their selectivity compared to MKIs in preliminary Phase I clinical trial data and early studies [111,122,123,137,142]. Moreover, the excellent intracranial activity of LOXO-292 and BLU-667 seen in these preliminary clinical trials provides a further advantage compared with MKIs that were associated with low CNS activity in RET fusion positive NSCLCs, but are associated with a high cumulative in- cidence of brain metastases during the course of their disease (46% lifetime prevalence in stage IV disease) [121]. The widespread use and accessibility of NGS allow for increasingly targeted therapies tailored to each individual patient, holding promise for the field of precision on- cology [59].
There are vast opportunities for further research into RET fusions from bench to bedside. Novel RET rearrangements continue to be dis- covered in solid tumors. More studies are necessary into the patho- genesis of RET fusions and clinic-pathological correlations. One ex- periment induced the KIF5B-RET fusion in 201T human lung cells using 1 Gy of γ radiation [144]. Further study is warranted to investigate the relationship between RET fusions and radiation exposure, especially
with the frequent use of gamma radiation via computed tomography and nuclear imaging in modern medicine [144]. Multikinase inhibitors have been FDA-approved for over a decade, yet much more work re- mains to be done in terms of efficacy and safety in current Phase IV trials as well as breaking new ground in TKI therapy of RET fusions in rarely studied and unstudied tumors such as pancreatic cancer, mela- nomatous and non-melanomatous skin cancers, head and neck malig- nancies, soft tissue tumors, and bone malignancies. More research into selective RET inhibitors LOXO-292 and BLU-667 will be conducted, eventually progressing to Phase II-IV clinical trials and potential FDA approval. Studies into RET fusions at every level from risk factors and genetic understanding to clinical presentation and therapeutic response have great potential to impact patients across all spectrum of cancers.

Declaration of Competing Interest

Dr. Rolfo reports personal fees from Novartis, personal fees from MSD, non-financial support from OncoDNA, personal fees and non-fi- nancial support from GuardantHealth, institutional grant from Biomark inc., outside the submitted work. All other authors have no conflicts of interest to report.

References

⦁ Takahashi M, Ritz J, Cooper GM. Activation of a novel human transforming gene, ret, by DNA rearrangement. Cell 1985;42:581–8. ⦁ https://doi.org/10.1016/0092- ⦁ 8674(85)90115-⦁ 1⦁ .
⦁ Amoresano A, Incoronato M, Monti G, Pucci P, de Franciscis V, Cerchia L. Direct interactions among Ret, GDNF and GFRalpha1 molecules reveal new insights into the assembly of a functional three-protein complex. Cell Signal 2005;17:717–27. https://doi.org/10.1016/j.cellsig.2004.10.012⦁ .
⦁ Wang X. Structural studies of GDNF family ligands with their receptors-Insights into ligand recognition and activation of receptor tyrosine kinase RET. Biochim Biophys Acta 2013;1834:2205–12. ⦁ https://doi.org/10.1016/j.bbapap.2012.10. ⦁ 008⦁ .
⦁ Arighi E, Borrello MG, Sariola H. RET tyrosine kinase signaling in development and cancer. Cytokine Growth Factor Rev 2005;16:441–67. ⦁ https://doi.org/10.1016/j. ⦁ cytogfr.2005.05.010⦁ .
⦁ Tansey MG, Baloh RH, Milbrandt J, Johnson EMJ. GFRalpha-mediated localization of RET to lipid rafts is required for effective downstream signaling, differentiation, and neuronal survival. Neuron 2000;25:611–23. ⦁ https://doi.org/10.1016/s0896- ⦁ 6273(00)81064-8⦁ .
⦁ Ibáñez CF. Structure and physiology of the RET receptor tyrosine kinase. Cold Spring Harb Perspect Biol 2013;5. https://doi.org/10.1101/cshperspect.a009134⦁ .
⦁ Besset V, Scott RP, Ibanez CF. Signaling complexes and protein-protein interactions involved in the activation of the Ras and phosphatidylinositol 3-kinase pathways by the c-Ret receptor tyrosine kinase. J Biol Chem 2000;275:39159–66. ⦁ https:// ⦁ doi.org/10.1074/jbc.M00690820⦁ 0⦁ .
⦁ Coulpier M, Anders J, Ibanez CF. Coordinated activation of autophosphorylation sites in the RET receptor tyrosine kinase: importance of tyrosine 1062 for GDNF mediated neuronal differentiation and survival. J Biol Chem 2002;277:1991–9. https://doi.org/10.1074/jbc.M107992200⦁ .
De⦁ ⦁ Vita⦁ ⦁ G,⦁ ⦁ Melillo⦁ ⦁ RM,⦁ ⦁ Carlomagno⦁ ⦁ F,⦁ ⦁ Visconti⦁ ⦁ R,⦁ ⦁ Castellone⦁ ⦁ MD,⦁ ⦁ Bellacosa⦁ ⦁ A,⦁ ⦁ et⦁ ⦁ al. ⦁ Tyrosine 1062 of RET-MEN2A mediates activation of Akt (protein kinase B) and ⦁ mitogen-activated protein kinase pathways leading to PC12 cell survival. Cancer ⦁ Res⦁ ⦁ 2000;60:3727⦁ –⦁ 31⦁ .
⦁ Hayashi H, Ichihara M, Iwashita T, Murakami H, Shimono Y, Kawai K, et al. Characterization of intracellular signals via tyrosine 1062 in RET activated by glial cell line-derived neurotrophic factor. Oncogene 2000;19:4469–75. ⦁ https://doi. ⦁ org/10.1038/sj.onc.1203799⦁ .
⦁ Ichihara M, Murakumo Y, Takahashi M. RET and neuroendocrine tumors. Cancer Lett 2004;204:197–211. https://doi.org/10.1016/S0304-3835(03)00456-7⦁ .
⦁ Segouffin-Cariou C, Billaud M. Transforming ability of MEN2A-RET requires ac- tivation of the phosphatidylinositol. J Biol Chem 2000;275:3568–76. ⦁ https://doi. ⦁ org/10.1074/jbc.275.5.3568⦁ .
⦁ Jain S, Encinas M, Johnson EMJ, Milbrandt J. Critical and distinct roles for key RET tyrosine docking sites in renal development. Genes Dev 2006;20:321–33. https://doi.org/10.1101/gad.1387206⦁ .
⦁ Jijiwa M, Kawai K, Fukihara J, Nakamura A, Hasegawa M, Suzuki C, et al. GDNF- mediated signaling via RET tyrosine 1062 is essential for maintenance of sper- matogonial stem cells. Genes Cells 2008;13:365–74. ⦁ https://doi.org/10.1111/j. ⦁ 1365-2443.2008.01171.⦁ x⦁ .
⦁ Amiel J, Sproat-Emison E, Garcia-Barcelo M, Lantieri F, Burzynski G, Borrego S, et al. Hirschsprung disease, associated syndromes and genetics: a review. J Med Genet 2008;45:1–14. https://doi.org/10.1136/jmg.2007.053959⦁ .
⦁ Edery P, Lyonnet S, Mulligan LM, Pelet A, Dow E, Abel L, et al. Mutations of the RET proto-oncogene in Hirschsprung’s disease. Nature 1994;367:378–80. ⦁ https:// ⦁ doi.org/10.1038/367378a0⦁ .
⦁ Romeo G, Ronchetto P, Luo Y, Barone V, Seri M, Ceccherini I, et al. Point muta- tions affecting the tyrosine kinase domain of the RET proto-oncogene in Hirschsprung’s disease. Nature 1994;367:377–8. ⦁ https://doi.org/10.1038/ ⦁ 367377a0⦁ .
⦁ Davis TK, Hoshi M, Jain S. To bud or not to bud: the RET perspective in CAKUT. Pediatr Nephrol 2014;29:597–608. https://doi.org/10.1007/s00467-013-2606-5⦁ .
⦁ Donis-Keller H, Dou S, Chi D, Carlson KM, Toshima K, Lairmore TC, et al. Mutations in the RET proto-oncogene are associated with MEN 2A and FMTC. Hum Mol Genet 1993;2:851–6. https://doi.org/10.1093/hmg/2.7.851⦁ .
⦁ Mulligan LM, Kwok JB, Healey CS, Elsdon MJ, Eng C, Gardner E, et al. Germ-line mutations of the RET proto-oncogene in multiple endocrine neoplasia type 2A. Nature 1993;363:458–60. https://doi.org/10.1038/363458a0⦁ .
⦁ Hofstra RM, Landsvater RM, Ceccherini I, Stulp RP, Stelwagen T, Luo Y, et al. A mutation in the RET proto-oncogene associated with multiple endocrine neoplasia type 2B and sporadic medullary thyroid carcinoma. Nature 1994;367:375–6. ⦁ https://doi.org/10.1038/367375a⦁ 0⦁ .
⦁ Mulligan LM. RET revisited: expanding the oncogenic portfolio. Nat Rev Cancer 2014;14:173–86. https://doi.org/10.1038/nrc3680⦁ .
⦁ Neumann HPH, Bausch B, McWhinney SR, Bender BU, Gimm O, Franke G, et al. Germ-line mutations in nonsyndromic pheochromocytoma. N Engl J Med 2002;346:1459–66. https://doi.org/10.1056/NEJMoa020152⦁ .
⦁ Morandi A, Martin L-A, Gao Q, Pancholi S, Mackay A, Robertson D, et al. GDNF- RET signaling in ER-positive breast cancers is a key determinant of response and resistance to aromatase inhibitors. Cancer Res 2013;73:3783–95. ⦁ https://doi.org/ ⦁ 10.1158/0008-5472.CAN-12-426⦁ 5⦁ .
⦁ Esseghir S, Todd SK, Hunt T, Poulsom R, Plaza-Menacho I, Reis-Filho JS, et al. A role for glial cell derived neurotrophic factor induced expression by inflammatory cytokines and RET/GFR alpha 1 receptor up-regulation in breast cancer. Cancer Res 2007;67:11732–41. https://doi.org/10.1158/0008-5472.CAN-07-2343⦁ .
⦁ Plaza-Menacho I, Morandi A, Robertson D, Pancholi S, Drury S, Dowsett M, et al. Targeting the receptor tyrosine kinase RET sensitizes breast cancer cells to ta- moxifen treatment and reveals a role for RET in endocrine resistance. Oncogene 2010;29:4648–57. https://doi.org/10.1038/onc.2010.209⦁ .
⦁ Gattelli A, Nalvarte I, Boulay A, Roloff TC, Schreiber M, Carragher N, et al. Ret inhibition decreases growth and metastatic potential of estrogen receptor positive breast cancer cells. EMBO Mol Med 2013;5:1335–50. ⦁ https://doi.org/10.1002/ ⦁ emmm.201302625⦁ .
⦁ Hatem R, Labiod D, Chateau-Joubert S, de Plater L, El Botty R, Vacher S, et al. Vandetanib as a potential new treatment for estrogen receptor-negative breast cancers. Int J Cancer 2016;138:2510–21. https://doi.org/10.1002/ijc.29974⦁ .
⦁ Boulay A, Breuleux M, Stephan C, Fux C, Brisken C, Fiche M, et al. The Ret receptor tyrosine kinase pathway functionally interacts with the ERalpha pathway in breast cancer. Cancer Res 2008;68:3743–51. ⦁ https://doi.org/10.1158/0008-5472.CAN- ⦁ 07-5100⦁ .
⦁ Tozlu S, Girault I, Vacher S, Vendrell J, Andrieu C, Spyratos F, et al. Identification of novel genes that co-cluster with estrogen receptor alpha in breast tumor biopsy specimens, using a large-scale real-time reverse transcription-PCR approach. Endocr Relat Cancer 2006;13:1109–20. https://doi.org/10.1677/erc.1.01120⦁ .
⦁ Spanheimer PM, Park J-M, Askeland RW, Kulak MV, Woodfield GW, De Andrade JP, et al. Inhibition of RET increases the efficacy of antiestrogen and is a novel treatment strategy for luminal breast cancer. Clin Cancer Res 2014;20:2115–25. https://doi.org/10.1158/1078-0432.CCR-13-2221⦁ .

⦁ Andreucci E, Francica P, Fearns A, Martin L-A, Chiarugi P, Isacke CM, et al. Targeting the receptor tyrosine kinase RET in combination with aromatase in- hibitors in ER positive breast cancer xenografts. Oncotarget 2016;7:80543–53. ⦁ https://doi.org/10.18632/oncotarget.1182⦁ 6⦁ .
⦁ Luo Y, Tsuchiya KD, Il Park D, Fausel R, Kanngurn S, Welcsh P, et al. RET is a potential tumor suppressor gene in colorectal cancer. Oncogene 2013;32:2037–47. ⦁ https://doi.org/10.1038/onc.2012.22⦁ 5⦁ .
⦁ Mendes Oliveira D, Grillone K, Mignogna C, De Falco V, Laudanna C, Biamonte F, et al. Next-generation sequencing analysis of receptor-type tyrosine kinase genes in surgically resected colon cancer: identification of gain-of-function mutations in the RET proto-oncogene. J Exp Clin Cancer Res 2018;37:84. ⦁ https://doi.org/10.1186/ ⦁ s13046-018-0746-⦁ y⦁ .
⦁ Ito Y, Okada Y, Sato M, Sawai H, Funahashi H, Murase T, et al. Expression of glial cell line-derived neurotrophic factor family members and their receptors in pan- creatic cancers. Surgery 2005;138:788–94. ⦁ https://doi.org/10.1016/j.surg.2005. ⦁ 07.007⦁ .
⦁ Zeng Q, Cheng Y, Zhu Q, Yu Z, Wu X, Huang K, et al. The relationship between overexpression of glial cell-derived neurotrophic factor and its RET receptor with progression and prognosis of human pancreatic cancer. J Int Med Res 2008;36:656–64. https://doi.org/10.1177/147323000803600406⦁ .
⦁ Amit M, Na’ara S, Leider-Trejo L, Binenbaum Y, Kulish N, Fridman E, et al. Upregulation of RET induces perineurial invasion of pancreatic adenocarcinoma. Oncogene 2017;36:3232–9. https://doi.org/10.1038/onc.2016.483⦁ .
⦁ Flavin R, Finn SP, Choueiri TK, Ingoldsby H, Ring M, Barrett C, et al. RET protein expression in papillary renal cell carcinoma. Urol Oncol 2012;30:900–5. ⦁ https:// ⦁ doi.org/10.1016/j.urolonc.2010.08.02⦁ 5⦁ .
⦁ Wang L, Zhang Y, Gao Y, Fan Y, Chen L, Liu K, et al. Prognostic and predictive values of subcellular localisation of RET in renal clear-cell carcinoma. Dis Markers 2016;2016:6870470. https://doi.org/10.1155/2016/6870470⦁ .
⦁ Dawson DM, Lawrence EG, MacLennan GT, Amini SB, Kung HJ, Robinson D, et al. Altered expression of RET proto-oncogene product in prostatic intraepithelial neoplasia and prostate cancer. J Natl Cancer Inst 1998;90:519–23. ⦁ https://doi. ⦁ org/10.1093/jnci/90.7.51⦁ 9⦁ .
Ban K, Feng S, Shao L, Ittmann M. RET signaling in prostate cancer. Clin Cancer ⦁ Res⦁ ⦁ 2017;23:4885⦁ –⦁ 96⦁ .
⦁ Narita N, Tanemura A, Murali R, Scolyer RA, Huang S, Arigami T, et al. Functional RET G691S polymorphism in cutaneous malignant melanoma. Oncogene 2009;28:3058–68. https://doi.org/10.1038/onc.2009.164⦁ .
⦁ Lawrence NF, Hammond MR, Frederick DT, Su Y, Dias-Santagata D, Deng A, et al. Ki-67, p53, and p16 expression, and G691S RET polymorphism in desmoplastic melanoma (DM): A clinicopathologic analysis of predictors of outcome. J Am Acad Dermatol 2016;75:595–602. https://doi.org/10.1016/j.jaad.2016.04.059⦁ .
⦁ Ohshima Y, Yajima I, Takeda K, Iida M, Kumasaka M, Matsumoto Y, et al. c-RET molecule in malignant melanoma from oncogenic RET-carrying transgenic mice and human cell lines. PLoS ONE 2010;5:e10279. ⦁ https://doi.org/10.1371/journal. ⦁ pone.001027⦁ 9⦁ .
⦁ Dabir S, Babakoohi S, Kluge A, Morrow JJ, Kresak A, Yang M, et al. RET mutation and expression in small-cell lung cancer. J Thorac Oncol 2014;9:1316–23. ⦁ https:// ⦁ doi.org/10.1097/JTO.000000000000023⦁ 4⦁ .
⦁ Andersson E, Arvidsson Y, Sward C, Hofving T, Wangberg B, Kristiansson E, et al. Expression profiling of small intestinal neuroendocrine tumors identifies sub- groups with clinical relevance, prognostic markers and therapeutic targets. Mod Pathol 2016;29:616–29. https://doi.org/10.1038/modpathol.2016.48⦁ .
⦁ Go H, Jung YJ, Kang HW, Park I-K, Kang C-H, Lee JW, et al. Diagnostic method for the detection of KIF5B-RET transformation in lung adenocarcinoma. Lung Cancer 2013;82:44–50. https://doi.org/10.1016/j.lungcan.2013.07.009⦁ .
⦁ Wong L-JC. Next Generation Sequencing 2013. ⦁ https://doi.org/10.1007/978-1- ⦁ 4614-7001-⦁ 4⦁ .
⦁ Abel HJ, Al-Kateb H, Cottrell CE, Bredemeyer AJ, Pritchard CC, Grossmann AH, et al. Detection of gene rearrangements in targeted clinical next-generation se- quencing. J Mol Diagn 2014;16:405–17. ⦁ https://doi.org/10.1016/j.jmoldx.2014. ⦁ 03.006⦁ .
⦁ Ali SM, Ou S-HI, He J, Peled N, Chmielecki J, Pinder MC, et al. Identifying ALK rearrangements that are not detected by FISH with targeted next-generation se- quencing of lung carcinoma. J Clin Oncol 2014;32:8049. ⦁ https://doi.org/10.1200/ ⦁ jco.2014.32.15_suppl.8049⦁ .
⦁ Drilon A, Wang L, Arcila ME, Balasubramanian S, Greenbowe JR, Ross JS, et al. Broad, hybrid capture-based next-generation sequencing identifies actionable genomic alterations in lung adenocarcinomas otherwise negative for such altera- tions by other genomic testing approaches. Clin Cancer Res 2015;21:3631–9. https://doi.org/10.1158/1078-0432.CCR-14-2683⦁ .
⦁ Frampton GM, Fichtenholtz A, Otto GA, Wang K, Downing SR, He J, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol 2013;31:1023–31. ⦁ https://doi. ⦁ org/10.1038/nbt.2696⦁ .
⦁ Moskalev EA, Frohnauer J, Merkelbach-Bruse S, Schildhaus H-U, Dimmler A, Schubert T, et al. Sensitive and specific detection of EML4-ALK rearrangements in non-small cell lung cancer (NSCLC) specimens by multiplex amplicon RNA massive parallel sequencing. Lung Cancer 2014;84:215–21. ⦁ https://doi.org/10.1016/j. ⦁ lungcan.2014.03.002⦁ .
⦁ Pekar-Zlotin M, Hirsch FR, Soussan-Gutman L, Ilouze M, Dvir A, Boyle T, et al. Fluorescence in situ hybridization, immunohistochemistry, and next-generation sequencing for detection of EML4-ALK rearrangement in lung cancer. Oncologist 2015;20:316–22. https://doi.org/10.1634/theoncologist.2014-0389⦁ .
⦁ Zheng Z, Liebers M, Zhelyazkova B, Cao Y, Panditi D, Lynch KD, et al. Anchored multiplex PCR for targeted next-generation sequencing. Nat Med
2014;20:1479–84. https://doi.org/10.1038/nm.3729.
⦁ Beadling C, Wald AI, Warrick A, Neff TL, Zhong S, Nikiforov YE, et al. A Multiplexed Amplicon Approach for Detecting Gene Fusions by Next-Generation Sequencing. J Mol Diagn 2016;18:165–75. ⦁ https://doi.org/10.1016/j.jmoldx. ⦁ 2015.10.002⦁ .
⦁ Levin JZ, Berger MF, Adiconis X, Rogov P, Melnikov A, Fennell T, et al. Targeted next-generation sequencing of a cancer transcriptome enhances detection of se- quence variants and novel fusion transcripts. Genome Biol 2009;10:R115. ⦁ https:// ⦁ doi.org/10.1186/gb-2009-10-10-r115⦁ .
⦁ Reeser JW, Martin D, Miya J, Kautto EA, Lyon E, Zhu E, et al. Validation of a targeted RNA sequencing assay for kinase fusion detection in solid tumors. J Mol Diagn 2017;19:682–96. https://doi.org/10.1016/j.jmoldx.2017.05.006⦁ .
⦁ Cheng ML, Berger MF, Hyman DM, Solit DB. Clinical tumour sequencing for pre- cision oncology: time for a universal strategy. Nat Rev Cancer 2018;18:527–8. ⦁ https://doi.org/10.1038/s41568-018-0043-⦁ 2⦁ .
⦁ Benayed R, Offin M, Mullaney K, Sukhadia P, Rios K, Desmeules P, et al. High yield of RNA sequencing for targetable kinase fusions in lung adenocarcinomas with no mitogenic driver alteration detected by DNA sequencing and low tumor mutation burden. Clin Cancer Res 2019. https://doi.org/10.1158/1078-0432.CCR-19-0225⦁ .
⦁ Rolfo C, Mack PC, Scagliotti GV, Baas P, Barlesi F, Bivona TG, et al. Liquid biopsy for advanced non-small cell lung cancer (NSCLC): a statement paper from the IASLC. J Thorac Oncol 2018;13:1248–68. ⦁ https://doi.org/10.1016/j.jtho.2018.05. ⦁ 030⦁ .
⦁ Rich TA, Reckamp KL, Chae YK, Doebele RC, Iams WT, Oh MS, et al. Analysis of cell-free DNA from 32,989 advanced cancers reveals novel co-occurring activating RET alterations and oncogenic signaling pathway aberrations. Clin Cancer Res 2019. https://doi.org/10.1158/1078-0432.CCR-18-4049⦁ .
⦁ Supplee JG, Milan MSD, Lim LP, Potts KT, Sholl LM, Oxnard GR, et al. Sensitivity of next-generation sequencing assays detecting oncogenic fusions in plasma cell- free DNA. Lung Cancer 2019;134:96–9. ⦁ https://doi.org/10.1016/j.lungcan.2019. ⦁ 06.004⦁ .
⦁ Fenton CL, Lukes Y, Nicholson D, Dinauer CA, Francis GL, Tuttle RM. The ret/PTC mutations are common in sporadic papillary thyroid carcinoma of children and young adults. J Clin Endocrinol Metab 2000;85:1170–5. ⦁ https://doi.org/10.1210/ ⦁ jcem.85.3.6472⦁ .
⦁ Elisei R, Romei C, Vorontsova T, Cosci B, Veremeychik V, Kuchinskaya E, et al. RET/PTC rearrangements in thyroid nodules: studies in irradiated and not irra- diated, malignant and benign thyroid lesions in children and adults. J Clin Endocrinol Metab 2001;86:3211–6. https://doi.org/10.1210/jcem.86.7.7678⦁ .
⦁ Cheung CC, Carydis B, Ezzat S, Bedard YC, Asa SL. Analysis of ret/PTC gene re- arrangements refines the fine needle aspiration diagnosis of thyroid cancer. J Clin Endocrinol Metab 2001;86:2187–90. https://doi.org/10.1210/jcem.86.5.7504⦁ .
Nikiforov YE. RET/PTC rearrangement in thyroid tumors. Endocr Pathol ⦁ 2002;13:3⦁ –⦁ 16⦁ .
⦁ Romei C, Elisei R. RET/PTC translocations and clinico-pathological features in human papillary thyroid carcinoma. Front Endocrinol (Lausanne) 2012;3:54. https://doi.org/10.3389/fendo.2012.00054⦁ .
⦁ Romei C, Fugazzola L, Puxeddu E, Frasca F, Viola D, Muzza M, et al. Modifications in the papillary thyroid cancer gene profile over the last 15 years. J Clin Endocrinol Metab 2012;97:E1758–65. https://doi.org/10.1210/jc.2012-1269⦁ .
⦁ Grubbs EG, Ng PK-S, Bui J, Busaidy NL, Chen K, Lee JE, et al. RET fusion as a novel driver of medullary thyroid carcinoma. J Clin Endocrinol Metab 2015;100:788–93. https://doi.org/10.1210/jc.2014-4153⦁ .
⦁ Romei C, Ciampi R, Elisei R. A comprehensive overview of the role of the RET proto-oncogene in thyroid carcinoma. Nat Rev Endocrinol 2016;12:192–202. ⦁ https://doi.org/10.1038/nrendo.2016.1⦁ 1⦁ .
⦁ Lu Z, Zhang Y, Feng D, Sheng J, Yang W, Liu B. Targeted next generation se- quencing identifies somatic mutations and gene fusions in papillary thyroid car- cinoma. Oncotarget 2017;8:45784–92. ⦁ https://doi.org/10.18632/oncotarget. ⦁ 17412⦁ .
⦁ Integrated genomic characterization of papillary thyroid carcinoma. Cell 2014; 159: 676–90. https://doi.org/10.1016/j.cell.2014.09.050.
⦁ Bounacer A, Wicker R, Caillou B, Cailleux AF, Sarasin A, Schlumberger M, et al. High prevalence of activating ret proto-oncogene rearrangements, in thyroid tu- mors from patients who had received external radiation. Oncogene 1997;15:1263–73. https://doi.org/10.1038/sj.onc.1200206⦁ .
⦁ Hamatani K, Eguchi H, Ito R, Mukai M, Takahashi K, Taga M, et al. RET/PTC rearrangements preferentially occurred in papillary thyroid cancer among atomic bomb survivors exposed to high radiation dose. Cancer Res 2008;68:7176–82. https://doi.org/10.1158/0008-5472.CAN-08-0293⦁ .
⦁ Hamatani K, Eguchi H, Koyama K, Mukai M, Nakachi K. Kusunoki Y. A novel RET rearrangement (ACBD5/RET) by pericentric inversion, inv(10)(p12.1;q11.2), in papillary thyroid cancer from an atomic bomb survivor exposed to high-dose ra- diation. Oncol Rep 2014;32:1809–14. https://doi.org/10.3892/or.2014.3449⦁ .
⦁ Leeman-Neill RJ, Brenner AV, Little MP, Bogdanova TI, Hatch M, Zurnadzy LY, et al. RET/PTC and PAX8/PPARgamma chromosomal rearrangements in post- Chernobyl thyroid cancer and their association with iodine-131 radiation dose and other characteristics. Cancer 2013;119:1792–9. ⦁ https://doi.org/10.1002/cncr. ⦁ 27893⦁ .
⦁ Ricarte-Filho JC, Li S, Garcia-Rendueles MER, Montero-Conde C, Voza F, Knauf JA, et al. Identification of kinase fusion oncogenes in post-Chernobyl radiation-in- duced thyroid cancers. J Clin Invest 2013;123:4935–44. ⦁ https://doi.org/10.1172/ ⦁ JCI6976⦁ 6⦁ .
Chung JH, Kim KW, Kim BJ, et al. ret/PTC-1, -2, and -3 Incogene Rearrangements ⦁ of Papillary Thyroid Carcinomas in Korea and Its Relevance to Clinical ⦁ Aggressiveness. Endocrinol Metabol 1999;14(1):53⦁ .

⦁ Fisher SB, Cote GJ, Bui-Griffith JH, Lu W, Tang X, Hai T, et al. Genetic char- acterization of medullary thyroid cancer in childhood survivors of the Chernobyl accident. Surgery 2019;165:58–63. https://doi.org/10.1016/j.surg.2018.08.029⦁ .
Nikiforov YE, Rowland JM, Bove KE, Monforte-Munoz H, Fagin JA. Distinct ⦁ pat- ⦁ tern of ret oncogene rearrangements in morphological variants of radiation-in- ⦁ duced and sporadic thyroid papillary carcinomas in children. Cancer Res ⦁ 1997;57:1690⦁ –⦁ 4⦁ .
Powell⦁ ⦁ DJJ,⦁ ⦁ Russell⦁ ⦁ J,⦁ ⦁ Nibu⦁ ⦁ K,⦁ ⦁ Li⦁ ⦁ G,⦁ ⦁ Rhee⦁ ⦁ E,⦁ ⦁ Liao⦁ ⦁ M,⦁ ⦁ et⦁ ⦁ al.⦁ ⦁ The⦁ ⦁ RET/PTC3⦁ ⦁ oncogene: ⦁ metastatic solid-type papillary carcinomas in murine thyroids. Cancer Res ⦁ 1998;58:5523⦁ –⦁ 8⦁ .
Sugg SL, Ezzat S, Zheng L, Freeman JL, Rosen IB, Asa SL. Oncogene profi⦁ le of ⦁ papillary thyroid carcinoma. Surgery⦁ ⦁ 1999;125:46⦁ –⦁ 52⦁ .
⦁ Zafon C, Obiols G, Castellvi J, Tallada N, Baena JA, Simo R, et al. Clinical sig- nificance of RET/PTC and p53 protein expression in sporadic papillary thyroid carcinoma. Histopathology 2007;50:225–31. ⦁ https://doi.org/10.1111/j.1365- ⦁ 2559.2006.02555.⦁ x⦁ .
⦁ Mochizuki K, Kondo T, Nakazawa T, Iwashina M, Kawasaki T, Nakamura N, et al. RET rearrangements and BRAF mutation in undifferentiated thyroid carcinomas having papillary carcinoma components. Histopathology 2010;57:444–50. ⦁ https://doi.org/10.1111/j.1365-2559.2010.03646.⦁ x⦁ .
Tallini G, Santoro M, Helie M, Carlomagno F, Salvatore G, Chiappetta G, et al. ⦁ RET/PTC oncogene activation de⦁ fi⦁ nes a subset of papillary thyroid carcinomas ⦁ lacking evidence of progression to poorly diff⦁ erentiated or undiff⦁ erentiated ⦁ tumor ⦁ phenotypes. Clin Cancer Res⦁ ⦁ 1998;4:287⦁ –⦁ 94⦁ .
⦁ Musholt TJ, Musholt PB, Khaladj N, Schulz D, Scheumann GF, Klempnauer J. Prognostic significance of RET and NTRK1 rearrangements in sporadic papillary thyroid carcinoma. Surgery 2000;128:984–93. ⦁ https://doi.org/10.1067/msy. ⦁ 2000.110845⦁ .
Basolo F, Molinaro E, Agate L, Pinchera A, Pollina L, Chiappetta G, et al. RET ⦁ protein⦁ ⦁ expression⦁ ⦁ has⦁ ⦁ no⦁ ⦁ prognostic⦁ ⦁ impact⦁ ⦁ on⦁ ⦁ the⦁ ⦁ long-term⦁ ⦁ outcome⦁ ⦁ of⦁ ⦁ papillary ⦁ thyroid carcinoma. Eur J Endocrinol⦁ ⦁ 2001;145:599⦁ –⦁ 604⦁ .
Pu⦁ x⦁ eddu⦁ ⦁ E,⦁ ⦁ Moretti⦁ ⦁ S,⦁ ⦁ Giannico⦁ ⦁ A,⦁ ⦁ Martinelli⦁ ⦁ M,⦁ ⦁ Marino⦁ ⦁ C,⦁ ⦁ Avenia⦁ ⦁ N,⦁ ⦁ et⦁ ⦁ al.⦁ ⦁ Ret/ ⦁ PTC⦁ ⦁ activation⦁ ⦁ does⦁ ⦁ not⦁ ⦁ infl⦁ uence⦁ ⦁ clinical⦁ ⦁ and⦁ ⦁ pathological⦁ ⦁ features⦁ ⦁ of⦁ ⦁ adult⦁ ⦁ pa- ⦁ pillary thyroid carcinomas. Eur J Endocrinol⦁ ⦁ 2003;148:505⦁ –⦁ 13⦁ .
Zhu X, Zhou X, Zhang T, Zhu X. [E⦁ x⦁ pressions of wildtype-RET and RET/PTC re- ⦁ arrangements in sporadic adult papillary thyroid carcinoma and their clin- ⦁ icopathologic correlation]. Zhonghua Bing Li Xue Za Zhi⦁ ⦁ 2006;35:87⦁ –⦁ 91⦁ .
⦁ Romei C, Ciampi R, Faviana P, Agate L, Molinaro E, Bottici V, et al. BRAFV600E mutation, but not RET/PTC rearrangements, is correlated with a lower expression of both thyroperoxidase and sodium iodide symporter genes in papillary thyroid cancer. Endocr Relat Cancer 2008;15:511–20. ⦁ https://doi.org/10.1677/ERC-07- ⦁ 0130⦁ .
⦁ Mishra A, Agrawal V, Krishnani N, Mishra SK. Prevalence of RET/PTC expression in papillary thyroid carcinoma and its correlation with prognostic factors in a north Indian population. J Postgrad Med 2009;55:171–5. ⦁ https://doi.org/10. ⦁ 4103/0022-3859.57390⦁ .
⦁ Zhang X, Su X, Chen WC, Li Y, Yang ZY, Deng WZ, et al. RET/PTC rearrangement affects multifocal formation of papillary thyroid carcinoma. Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 2017;52:435–9. ⦁ https://doi.org/10.3760/cma.j.issn. ⦁ 1673-0860.2017.06.00⦁ 8⦁ .
⦁ Rudin CM, Drilon A, Poirier JT. RET mutations in neuroendocrine tumors: in- cluding small-cell lung cancer. J Thorac Oncol 2014;9:1240–2. ⦁ https://doi.org/10. ⦁ 1097/JTO.000000000000030⦁ 1⦁ .
⦁ Kohno T, Ichikawa H, Totoki Y, Yasuda K, Hiramoto M, Nammo T, et al. KIF5B- RET fusions in lung adenocarcinoma. Nat Med 2012;18:375–7. ⦁ https://doi.org/10. ⦁ 1038/nm.2644⦁ .
⦁ Li F, Feng Y, Fang R, Fang Z, Xia J, Han X, et al. Identification of RET gene fusion by exon array analyses in “pan-negative” lung cancer from never smokers. Cell Res 2012;22:928–31. https://doi.org/10.1038/cr.2012.27⦁ .
⦁ Lipson D, Capelletti M, Yelensky R, Otto G, Parker A, Jarosz M, et al. Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies. Nat Med 2012;18:382–4. https://doi.org/10.1038/nm.2673⦁ .
⦁ Takeuchi K, Soda M, Togashi Y, Suzuki R, Sakata S, Hatano S, et al. RET, ROS1 and ALK fusions in lung cancer. Nat Med 2012;18:378–81. ⦁ https://doi.org/10.1038/ ⦁ nm.2658⦁ .
⦁ Yoo SS, Jin G, Jung HJ, Hong MJ, Choi JE, Jeon H-S, et al. RET fusion genes in Korean non-small cell lung cancer. J Korean Med Sci 2013;28:1555–8. ⦁ https://doi. ⦁ org/10.3346/jkms.2013.28.10.1555⦁ .
⦁ Lin C, Wang S, Xie W, Chang J, Gan Y. The RET fusion gene and its correlation with demographic and clinicopathological features of non-small cell lung cancer: a meta-analysis. Cancer Biol Ther 2015;16:1019–28. ⦁ https://doi.org/10.1080/ ⦁ 15384047.2015.104664⦁ 9⦁ .
⦁ Kato S, Subbiah V, Marchlik E, Elkin SK, Carter JL, Kurzrock R. RET aberrations in diverse cancers: next-generation sequencing of 4871 patients. Clin Cancer Res 2017;23:1988–97. https://doi.org/10.1158/1078-0432.CCR-16-1679⦁ .
⦁ Wang R, Hu H, Pan Y, Li Y, Ye T, Li C, et al. RET fusions define a unique molecular and clinicopathologic subtype of non-small-cell lung cancer. J Clin Oncol 2012;30:4352–9. https://doi.org/10.1200/JCO.2012.44.1477⦁ .
⦁ Ju YS, Lee W-C, Shin J-Y, Lee S, Bleazard T, Won J-K, et al. A transforming KIF5B and RET gene fusion in lung adenocarcinoma revealed from whole-genome and transcriptome sequencing. Genome Res 2012;22:436–45. ⦁ https://doi.org/10. ⦁ 1101/gr.133645.111⦁ .
⦁ Sarfaty M, Moore A, Neiman V, Dudnik E, Ilouze M, Gottfried M, et al. RET fusion lung carcinoma: response to therapy and clinical features in a case series of 14 patients. Clin Lung Cancer 2017;18:e223–32. ⦁ https://doi.org/10.1016/j.cllc.2016. ⦁ 09.003⦁ .
⦁ Gautschi O, Milia J, Filleron T, Wolf J, Carbone DP, Owen D, et al. Targeting RET in patients with RET-rearranged lung cancers: results from the global. Multicenter RET Registry. J Clin Oncol 2017;35:1403–10. ⦁ https://doi.org/10.1200/JCO.2016. ⦁ 70.935⦁ 2⦁ .
⦁ Levinson S, Cagan RL. Drosophila cancer models identify functional differences between ret fusions. Cell Rep 2016;16:3052–61. ⦁ https://doi.org/10.1016/j.celrep. ⦁ 2016.08.019⦁ .
⦁ Score J, Curtis C, Waghorn K, Stalder M, Jotterand M, Grand FH, et al. Identification of a novel imatinib responsive KIF5B-PDGFRA fusion gene following screening for PDGFRA overexpression in patients with hypereosinophilia. Leukemia 2006;20:827–32. https://doi.org/10.1038/sj.leu.2404154⦁ .
⦁ Takeuchi K, Choi YL, Togashi Y, Soda M, Hatano S, Inamura K, et al. KIF5B-ALK, a novel fusion oncokinase identified by an immunohistochemistry-based diagnostic system for ALK-positive lung cancer. Clin Cancer Res 2009;15:3143–9. ⦁ https://doi. ⦁ org/10.1158/1078-0432.CCR-08-324⦁ 8⦁ .
⦁ Klempner SJ, Bazhenova LA, Braiteh FS, Nikolinakos PG, Gowen K, Cervantes CM, et al. Emergence of RET rearrangement co-existing with activated EGFR mutation in. Lung Cancer 2015;89:357–9. https://doi.org/10.1016/j.lungcan.2015.06.021⦁ .
⦁ Chang H, Sung JH, Moon SU, Kim HS, Kim JW, Lee JS. EGF induced RET inhibitor resistance in CCDC6-RET lung cancer cells. Yonsei Med J 2017;58:9–18. ⦁ https:// ⦁ doi.org/10.3349/ymj.2017.58.1.⦁ 9⦁ .
⦁ Piotrowska Z, Isozaki H, Lennerz JK, Gainor JF, Lennes IT, Zhu VW, et al. Landscape of acquired resistance to osimertinib in EGFR-mutant NSCLC and clinical validation of combined EGFR and RET inhibition with osimertinib and BLU-667 for acquired RET fusion. Cancer Discov 2018;8:1529–39. ⦁ https://doi. ⦁ org/10.1158/2159-8290.CD-18-102⦁ 2⦁ .
⦁ Drilon A, Bergagnini I, Delasos L, Sabari J, Woo KM, Plodkowski A, et al. Clinical outcomes with pemetrexed-based systemic therapies in RET-rearranged lung cancers. Ann Oncol 2016;27:1286–91. https://doi.org/10.1093/annonc/mdw163⦁ .
⦁ Offin M, Guo R, Wu SL, Sabari J, Land JD, Ni A, et al. Immunophenotype and response to immunotherapy of RET-rearranged lung cancers. JCO Precis Oncol 2019;3. https://doi.org/10.1200/PO.18.00386⦁ .
⦁ Ogino H, Yano S, Kakiuchi S, Yamada T, Ikuta K, Nakataki E, et al. Novel dual targeting strategy with vandetanib induces tumor cell apoptosis and inhibits an- giogenesis in malignant pleural mesothelioma cells expressing RET oncogenic rearrangement. Cancer Lett 2008;265:55–66. ⦁ https://doi.org/10.1016/j.canlet. ⦁ 2008.02.01⦁ 8⦁ .
⦁ Le Rolle A-F, Klempner SJ, Garrett CR, Seery T, Sanford EM, Balasubramanian S, et al. Identification and characterization of RET fusions in advanced colorectal cancer. Oncotarget 2015;6:28929–37. https://doi.org/10.18632/oncotarget.4325⦁ .
⦁ Kim SY, Oh SO, Kim K, Lee J, Kang S, Kim K-M, et al. NCOA4-RET fusion in colorectal cancer: therapeutic challenge using patient-derived tumor cell lines. J Cancer 2018;9:3032–7. https://doi.org/10.7150/jca.26256⦁ .
⦁ Pietrantonio F, Di Nicolantonio F, Schrock AB, Lee J, Morano F, Fuca G, et al. RET fusions in a small subset of advanced colorectal cancers at risk of being neglected. Ann Oncol 2018;29:1394–401. https://doi.org/10.1093/annonc/mdy090⦁ .
⦁ Paratala BS, Chung JH, Williams CB, Yilmazel B, Petrosky W, Williams K, et al. RET rearrangements are actionable alterations in breast cancer. Nat Commun 2018;9:4821. https://doi.org/10.1038/s41467-018-07341-4⦁ .
⦁ Carlomagno F, Vitagliano D, Guida T, Basolo F, Castellone MD, Melillo RM, et al. Efficient inhibition of RET/papillary thyroid carcinoma oncogenic kinases by. J Clin Endocrinol Metab 2003;88:1897–902. ⦁ https://doi.org/10.1210/jc.2002- ⦁ 021278⦁ .
⦁ Gild ML, Bullock M, Pon CK, Robinson BG, Clifton-Bligh RJ. Destabilizing RET in targeted treatment of thyroid cancers. Endocr Connect 2016;5:10–9. ⦁ https://doi. ⦁ org/10.1530/EC-15-009⦁ 8⦁ .
⦁ Drilon A, Fu S, Patel MR, Fakih M, Wang D, Olszanski AJ, et al. A Phase I/Ib Trial of the VEGFR-Sparing Multikinase RET Inhibitor RXDX-105. Cancer Discov 2019;9:384–95. https://doi.org/10.1158/2159-8290.CD-18-0839⦁ .
⦁ Drilon A, Oxnard GR, Wirth L, Besse B, Gautchi O, Tan DS, et al. PL02.08 Registrational results of LIBRETTO-001: A phase 1/2 trial of LOXO-292 in patients with RET fusion-positive lung cancers. Presented at: IASLC 2019 World Conference on Lung Cancer hosted by the International Association for the Study of Lung Cancer; September 7-10, 2019; Barcelona, Spain. Abstract PL02.08.
⦁ Gainor JF, Lee DH, Curigliano G, Doebele RC, Kim D-W, Baik CS, et al. Clinical activity and tolerability of BLU-667, a highly potent and selective RET inhibitor, in patients (pts) with advanced RET-fusion+ non-small cell lung cancer (NSCLC). J Clin Oncol. 2019;37:9008. https://doi.org/10.1200/JCO.2019.37.15_suppl.9008⦁ .
⦁ Horiike A, Takeuchi K, Uenami T, Kawano Y, Tanimoto A, Kaburaki K, et al. Sorafenib treatment for patients with RET fusion-positive non-small cell lung cancer. Lung Cancer 2016;93:43–6. ⦁ https://doi.org/10.1016/j.lungcan.2015.12. ⦁ 011⦁ .
⦁ Matsubara D, Kanai Y, Ishikawa S, Ohara S, Yoshimoto T, Sakatani T, et al. Identification of CCDC6-RET fusion in the human lung adenocarcinoma cell line. J Thorac Oncol 2012;7:1872–6. https://doi.org/10.1097/JTO.0b013e3182721ed1⦁ .
⦁ Nelson-Taylor SK, Le AT, Yoo M, Schubert L, Mishall KM, Doak A, et al. Resistance to RET-inhibition in RET-rearranged NSCLC Is Mediated By reactivation of RAS/ MAPK signaling. Mol Cancer Ther 2017;16:1623–33. ⦁ https://doi.org/10.1158/ ⦁ 1535-7163.MCT-17-000⦁ 8⦁ .
⦁ Okamoto K, Kodama K, Takase K, Sugi NH, Yamamoto Y, Iwata M, et al. Antitumor activities of the targeted multi-tyrosine kinase inhibitor lenvatinib (E7080) against RET gene fusion-driven tumor models. Cancer Lett 2013;340:97–103. ⦁ https://doi. ⦁ org/10.1016/j.canlet.2013.07.00⦁ 7⦁ .
⦁ Jeong W-J, Mo J-H, Park MW, Choi IJ, An S-Y, Jeon E-H, et al. Sunitinib inhibits papillary thyroid carcinoma with RET/PTC rearrangement but not BRAF muta- tion. Cancer Biol Ther 2011;12:458–65. https://doi.org/10.4161/cbt.12.5.16303⦁ .

⦁ Kodama T, Tsukaguchi T, Satoh Y, Yoshida M, Watanabe Y, Kondoh O, et al. Alectinib shows potent antitumor activity against RET-rearranged non-small cell lung cancer. Mol Cancer Ther 2014;13:2910–8. ⦁ https://doi.org/10.1158/1535- ⦁ 7163.MCT-14-027⦁ 4⦁ .
⦁ Li GG, Somwar R, Joseph J, Smith RS, Hayashi T, Martin L, et al. Antitumor ac- tivity of RXDX-105 in multiple cancer types with RET rearrangements or muta- tions. Clin Cancer Res 2017;23:2981–90. ⦁ https://doi.org/10.1158/1078-0432. ⦁ CCR-16-188⦁ 7⦁ .
⦁ Drilon A, Rekhtman N, Arcila M, Wang L, Ni A, Albano M, et al. Cabozantinib in patients with advanced RET-rearranged non-small-cell lung cancer: an open-label, single-centre, phase 2, single-arm trial. Lancet Oncol 2016;17:1653–60. ⦁ https:// ⦁ doi.org/10.1016/S1470-2045(16)30562-⦁ 9⦁ .
⦁ Velcheti V, Hida T, Reckamp KL, Yang JC, Nokihara H, Sachdev P, et al. Phase 2 study of lenvatinib (LN) in patients (Pts) with RET fusion-positive adenocarcinoma of the lung. Ann Oncol. 2016;27(suppl_6):1204PD. ⦁ https://doi.org/10.1093/ ⦁ annonc/mdw383.0⦁ 5⦁ .
⦁ Lee S-H, Lee J-K, Ahn M-J, Kim D-W, Sun J-M, Keam B, et al. Vandetanib in pretreated patients with advanced non-small cell lung cancer-harboring RET re- arrangement: a phase II clinical trial. Ann Oncol 2017;28:292–7. ⦁ https://doi.org/ ⦁ 10.1093/annonc/mdw55⦁ 9⦁ .
⦁ Yoh K, Seto T, Satouchi M, Nishio M, Yamamoto N, Murakami H, et al. Vandetanib in patients with previously treated RET-rearranged advanced non-small-cell lung cancer (LURET): an open-label, multicentre phase 2 trial. Lancet Respir Med 2017;5:42–50. https://doi.org/10.1016/S2213-2600(16)30322-8⦁ .
⦁ Yoh K, Seto T, Satouchi M, Nishio M, Yamamoto N, Murakami H, et al. 1487P LURET: Final survival results of the phase II trial of vandetanib in patients with advanced RET-rearranged non-small cell lung cancer. Annals Oncol 2018;29. https://doi.org/10.1093/annonc/mdy292.108⦁ .
⦁ Takeuchi S, Murayama T, Yoshimura K, Kawakami T, Takahara S, Imai Y, et al. Phase I/II study of alectinib in lung cancer with RET fusion gene: study protocol. J Med Invest 2017;64:317–20. https://doi.org/10.2152/jmi.64.317⦁ .
⦁ Subbiah V, Gainor JF, Rahal R, Brubaker JD, Kim JL, Maynard M, et al. Precision targeted therapy with BLU-667 for RET-driven cancers. Cancer Discov 2018;8:836–49. https://doi.org/10.1158/2159-8290.CD-18-0338⦁ .
Drilon⦁ ⦁ A,⦁ ⦁ Liu⦁ ⦁ S,⦁ ⦁ Doebele⦁ ⦁ R,⦁ ⦁ Rodriguez⦁ ⦁ C,⦁ ⦁ Fakih⦁ ⦁ M,⦁ ⦁ Reckamp⦁ ⦁ K,⦁ ⦁ et⦁ ⦁ al.⦁ ⦁ LBA19A⦁ ⦁ phase ⦁ 1b⦁ ⦁ study⦁ ⦁ of⦁ ⦁ RXDX-105,⦁ ⦁ a⦁ ⦁ VEGFR-sparing⦁ ⦁ potent⦁ ⦁ RET⦁ ⦁ inhibitor,⦁ ⦁ in⦁ ⦁ RETi-naïve⦁ ⦁ pa- ⦁ tients⦁ ⦁ with⦁ ⦁ RET⦁ ⦁ fusion-positive⦁ ⦁ NSCLC.⦁ ⦁ Ann⦁ ⦁ Oncol.⦁ ⦁ 2017;28(suppl_5):v605⦁ –⦁ 49⦁ .
⦁ Roskoski RJ, Sadeghi-Nejad A. Role of RET protein-tyrosine kinase inhibitors in the treatment RET-driven thyroid and lung cancers. Pharmacol Res 2018;128:1–17. https://doi.org/10.1016/j.phrs.2017.12.021⦁ .
⦁ Abdel-Magid AF. RET kinase inhibitors may treat cancer and gastrointestinal disorders. ACS Med Chem Lett 2014;6:13–4. https://doi.org/10.1021/ml500402t⦁ .
⦁ Redaelli S, Plaza-Menacho I, Mologni L. Novel targeted therapeutics for MEN2. Endocr Relat Cancer 2018;25:T53–68. https://doi.org/10.1530/ERC-17-0297⦁ .
Starr P. Selective RET inhibitor, BLU-667, hits the target in patients with lung or ⦁ thyroid⦁ cancers. Value-Based Cancer⦁ ⦁ Care 2018;9(3):22⦁ .
⦁ Taylor MH, Gainor JF, Hu MI-N, Zhu VW, Lopes G, Leboulleux S, et al. Activity and tolerability of BLU-667, a highly potent and selective RET inhibitor, in patients with advanced RET-altered thyroid cancers. J Clin Oncol. 2019;37:6018. ⦁ https:// ⦁ doi.org/10.1200/JCO.2019.37.15_suppl.601⦁ 8⦁ .
⦁ Dacic S, Luvison A, Evdokimova V, Kelly L, Siegfried JM, Villaruz LC, et al. RET rearrangements in lung adenocarcinoma and radiation. J Thorac Oncol 2014;9:118–20. https://doi.org/10.1097/JTO.0000000000000015⦁ .
⦁ Wirth L, Sherman E, Drilon A, Solomon B, Robinson B, Lorch J, et al. LBA93Registrational results of LOXO-292 in patients with RET-altered thyroid cancers. Ann Oncol 2019;30. https://doi.org/10.1093/annonc/mdz394.093⦁ .