RET alterations have been characterized as oncogenic drivers in multiple cancers. The clinical validation of highly selective RET inhibitors demonstrates the utility of specific targeting of aberrantly activated RET in patients with cancers such as medullary thyroid cancer or non–small cell lung cancer. The remarkable responses observed have opened the field of RET-targeted inhibitors. In this review, we seek to focus on the impact of therapeutic RET targeting in cancers.

Significance:

Successful clinical translation of selective RET inhibitors is poised to alter the therapeutic landscape of altered cancers. Questions that clearly need to be addressed relate to the ability to maintain long-term inhibition of tumor cell growth, how to prepare for the potential mechanisms of acquired resistance, and the development of next-generation selective RET inhibitors.

Genome-driven precision oncology has altered the therapeutic landscape of multiple kinase-driven hematologic and solid malignancies. The use of imatinib in BCR–ABL fusion–positive chronic myelogenous leukemia (1) and the use of crizotinib in ALK fusion– and ROS1 fusion–positive non–small cell lung cancer (NSCLC) are prime examples of transformative first-generation kinase inhibitor therapies (2, 3). Twenty-five years ago, inherited mutations in RET mutations were identified as the cause of multiple endocrine neoplasia type 2 (MEN2; refs. 4–7), which was soon followed by the discovery of somatic activating RET aberrations, genetic fusions, or mutations in diverse malignancies (8–10) warranting their choice as prospective therapeutic targets (Fig. 1). RET fusions are seen in NSCLC (2%) and papillary thyroid cancers (PTC; 10%–20%), whereas somatic (60%–90%) or germline (100%) RET mutations are seen in medullary thyroid cancer (MTC; refs. 11, 12).

Figure 1.

Frequency and distribution of RET fusions and RET mutations across malignancies. Visual art © 2019 The University of Texas MD Anderson Cancer Center. Red text indicates the most prevalent RET-dependent malignancies. CMML, chronic myelomonocytic leukemia.

Figure 1.

Frequency and distribution of RET fusions and RET mutations across malignancies. Visual art © 2019 The University of Texas MD Anderson Cancer Center. Red text indicates the most prevalent RET-dependent malignancies. CMML, chronic myelomonocytic leukemia.

Close modal

Although RET was one of the earliest genes to be cloned, and several multikinase inhibitors (MKI) have RET inhibitory activity, patients with RET-driven malignancies, especially patients with RET fusion–positive NSCLC, have derived only limited benefit from MKIs with secondary RET activity. MKIs such as vandetanib and cabozantinib are FDA-approved in the treatment of advanced MTC and have demonstrated activity in patients with RET fusion–positive NSCLC, but their response rates and duration of response are lower when compared with other selective kinase inhibitors for ALK or ROS1 fusion–driven NSCLC (2, 3, 11, 13, 14). Development of selective RET inhibitors is poised to change this paradigm. In this review, we focus on the impact of RET therapeutic targeting in cancers, within the context of a relatively short-term treatment experience. Questions that need to be further addressed include the ability to maintain long-term inhibition of tumor cell growth, and how to prepare for the potential mechanisms of acquired resistance. We also consider the need for development of second-generation selective RET inhibitors, and finally the potential side effects associated with reduced RET activity in tissues reliant on its expression.

By 1985, the search for human oncogenes was rapidly advancing. Approximately a dozen or so transforming genes, most notably the RAS family members, had already been identified using a simple assay of transfecting human tumor DNA into NIH 3T3 cells. Serial passaging of the transformed NIH 3T3 cells allowed for these human oncogenes to be cloned through their association with human repetitive DNA sequences. Interestingly, the coincidence of using DNA isolated from a patient with T-cell lymphoma, 3215, led to the discovery of the RET oncogene (15). A single transformed colony was ultimately expanded through secondary and tertiary transfections, providing both confirmation of a transforming oncogene and a DNA source for characterization (15). However, when the 3215 transforming DNA was compared with normal human DNA, a discontinuity was discovered, leading to the hypothesis that the oncogene was derived from recombination of two unlinked segments. The authors proposed a mechanism of REarrangement during Transfection that ultimately led to the naming of the RET oncogene. Molecular analysis of the RET transforming gene determined the fusion partner to be an upstream ring finger domain (originally unrelated to genes identified at the time) and a downstream transmembrane linked to a tyrosine kinase domain (16). As other tyrosine kinases had previously been linked to oncogenic transformation, it was ultimately the gene encoding this domain that was given the name “RET proto-oncogene.” Despite the belief that the RET oncogene was created through an experimental artifact, the same NIH 3T3 transformation assay was able to subsequently demonstrate the frequent occurrence of RET gene fusions in papillary thyroid cancers (17) and to confirm the transforming ability of MEN2-associated RET-activating mutations (18).

In the decades that have passed since the discovery of RET, much has been uncovered related to its role in cancer. To date, three general mechanisms of aberrant RET activation have been reported in cancer: in-frame RET gene fusions (15, 16), targeted mutation of the RET gene itself (4–6), and finally aberrant overexpression of the RET gene (19, 20). What the three mechanisms appear to share in common is the inappropriate activation of the tyrosine kinase, most commonly in the complete absence of ligand. The multifunctional docking sites at phosphotyrosine 1062 (pY1062) and pY1096 serve as the primary RET signaling hubs (reviewed in refs. 20, 21). Activation of RAS–MAPK and PI3K–AKT signaling pathways results from adaptor protein binding to these docking sites (Fig. 2; refs. 22, 23).

Figure 2.

Oncogenic RET signaling: RET is the signaling receptor for the glial cell–derived neurotrophic factor (GDNF) family of ligands. These ligands play a key role in organ development and tissue homeostasis. Targeted mutation of RET (e.g., RETM918T and RETV804M/L) results in aberrant activation through three broad mechanisms—dimerization through the formation of intermolecular cysteine disulfide bonds, impacting of the ATP-binding domain, and finally enhancement of the kinase domain activity. Chromosomal rearrangements (e.g., KIF5BRET and CCDC6RET) produce hybrid proteins that fuse a partner protein that often contains a dimerization domain with the RET kinase domain. Visual art © 2019 The University of Texas MD Anderson Cancer Center.

Figure 2.

Oncogenic RET signaling: RET is the signaling receptor for the glial cell–derived neurotrophic factor (GDNF) family of ligands. These ligands play a key role in organ development and tissue homeostasis. Targeted mutation of RET (e.g., RETM918T and RETV804M/L) results in aberrant activation through three broad mechanisms—dimerization through the formation of intermolecular cysteine disulfide bonds, impacting of the ATP-binding domain, and finally enhancement of the kinase domain activity. Chromosomal rearrangements (e.g., KIF5BRET and CCDC6RET) produce hybrid proteins that fuse a partner protein that often contains a dimerization domain with the RET kinase domain. Visual art © 2019 The University of Texas MD Anderson Cancer Center.

Close modal

Targeted mutation of RET results in aberrant activation through three broad mechanisms—dimerization through the formation of intermolecular cysteine disulfide bonds, impacting of the ATP-binding domain, and finally enhancement of the kinase domain activity (Fig. 2). Because germline RET activating mutations occur in patients with hereditary MEN2, we know that tumorigenesis is primarily limited to a subpopulation of cells of neuroendocrine origin, most notably thyroid C cells and cells within the adrenal medulla. Although it has been argued that the specificity of tumor formation derives from the high level of RET expression in these cell types, this is clearly an oversimplification. RET is known to be more broadly expressed and has clear roles in mediating progenitor stem-cell function.

To date, germline RET fusions have not been observed, only cell-specific somatic fusions. They were initially thought to be limited to thyroid follicular cell tumors, but have more recently been identified in 1% to 2% of NSCLCs, and at a <1% frequency in a range of tumor types, including colorectal cancer, breast cancer, and pancreatic cancer (11, 12). There are numerous RET fusion partners, all of which appear to provide dimerization domains (Fig. 1). A survey of datasets for cancer types within cBioPortal finds that approximately 10% of reported RET mutations are fusions, with approximately 10% of those comprising tumor types other than thyroid or lung. RET fusions are the most commonly observed aberration in tumors that are not of neuroendocrine origin. RET fusions are thought to be oncogenic for two reasons. First, fusion provides a mechanism to aberrantly express RET in a cell type where it is normally transcriptionally silent. Second, in all cases the extracellular domain is replaced with a protein dimerization domain. The outcome is the production of an intracellular RET tyrosine kinase domain capable of ligand-independent activation. Interestingly, despite the absence of a transmembrane domain, the RET fusion proteins remain capable of MAPK pathway and PI3K–AKT activation. However, beyond the examination of proliferative signaling, the cellular functions of RET fusion have not been extensively studied. It is also important to point out that key regulatory mechanisms of RET inactivation, such as endocytosis and recruitment of membrane-associated ubiquitin ligases, do not appear to affect the fusion proteins, which additionally may enhance their oncogenicity (24, 25). Furthermore, when localized to the cytoplasm or nucleus, monomeric RET has additional functions, such as regulation of ATF4, that could certainly be affected (26). These circumstances seemingly predict a greater oncogenic potential for RET fusions compared with activating mutations that remains to be fully addressed. Direct comparisons of RET fusions with full-length RET containing activating mutations are limited, but it is important to note that experimental inclusion of RET-activating mutations into fusion constructs does not increase tumorigenicity in xenograft models (27, 28). Given these differences, we are largely left to speculate on the precise impact of targeted inhibition on oncogenesis driven by these two mechanisms of aberrant RET activation. As a result, our understanding of targeting RET-driven cancer continues to evolve largely through clinical trials (27, 28).

The original concept of therapeutically targeting RET largely stems from MTC studies. Activating germline mutations cause MTC, somatic activating mutations are found in sporadic MTC, and these same mutations are capable of inducing cellular transformation in the NIH 3T3 cell assay. These facts ultimately led to the hypothesis that MTC cells were addicted to oncogenic RET and therefore sensitive to targeted inhibition. The first proof-of-concept came when it was demonstrated that ribozyme-mediated cleavage of mutant RET mRNA inhibited MTC tumor cell growth in vitro (29). Subsequently, it was demonstrated that overexpression of a dominant-negative RET was capable of inhibiting human MTC cell line growth in vitro (30) and in xenograft models (31). Neither approach was clinically viable, however, thereby opening the door for small-molecule inhibitors.

Vandetanib/ZD6474 was among the first drugs demonstrated to inhibit the activity of both oncogenic RET fusions as well as RET activated through the M918T mutation (32). However, as vandetanib is a repurposed VEGFR inhibitor, it has never been definitively established whether its antitumor actions (and those of similar multikinase-targeting drugs like cabozantinib, lenvatinib, and sorafenib) function primarily through inhibition of RET, particularly as MKIs without RET activity (axitinib) have been shown to be effective in RET-altered thyroid cancers (33).

Vandetanib and cabozantinib are MKIs with nonselective RET activity that have been approved by the FDA for MTC. They were originally designed to target other kinases, such as VEGFR2 and MET, but were repurposed because of the discovery of their inhibitory actions on RET. This MKI activity leads to significant and sometimes prohibitive “off-target” clinical side effects such as nausea, diarrhea, rash, and hypertension that limit use in some patients or limit the dose that patients can tolerate, leading to drug discontinuation or dose reduction (Table 1). Together with nonselectivity for RET and inferior pharmacokinetic properties, these MKIs prevented potent RET inhibition.

Table 1.

ORRs, dose-reduction rates, and drug-discontinuation rates of MKIs versus selective RET inhibitors

ORRsDose-reduction rate and drug-discontinuation ratesb
DrugThyroidNSCLCThyroidNSCLC
Vandetanib (Caprelsa) 45% (MTC) 18%, 53% (Japan) DRR = 81/231 (35%) DRR = 10/19 (53%) 
   DDR = 28/231 (12%) DDR = 4/19 (21%) 
Cabozantinib (Cabometyx/Cometriq) 28% (MTC) 28% DRR = 169/214 (79%) DRR = 19/26 (73%) 
   DDR = 35/214 (16%) DDR = 2/26 (8%) 
Lenvatinib (Lenvima) 36% 16% DRR = 35/59 (59%) DRR = 16/25 (64%) 
   DDR = 14/59 (24%) DDR = 5/25 (20%) 
Selpercatiniba (LOXO-292) 78% (RET+ thyroid cancer) 68% DRR = N/A  
  59% (MTC) DDR = 9/531 (1.7%)  
   DRR = N/A  
   9/531 (1.7%)  
Pralsetiniba (BLU-667) 56% (MTC) 58% DRR = N/A  
   DDR = 4% (276 patients)  
ORRsDose-reduction rate and drug-discontinuation ratesb
DrugThyroidNSCLCThyroidNSCLC
Vandetanib (Caprelsa) 45% (MTC) 18%, 53% (Japan) DRR = 81/231 (35%) DRR = 10/19 (53%) 
   DDR = 28/231 (12%) DDR = 4/19 (21%) 
Cabozantinib (Cabometyx/Cometriq) 28% (MTC) 28% DRR = 169/214 (79%) DRR = 19/26 (73%) 
   DDR = 35/214 (16%) DDR = 2/26 (8%) 
Lenvatinib (Lenvima) 36% 16% DRR = 35/59 (59%) DRR = 16/25 (64%) 
   DDR = 14/59 (24%) DDR = 5/25 (20%) 
Selpercatiniba (LOXO-292) 78% (RET+ thyroid cancer) 68% DRR = N/A  
  59% (MTC) DDR = 9/531 (1.7%)  
   DRR = N/A  
   9/531 (1.7%)  
Pralsetiniba (BLU-667) 56% (MTC) 58% DRR = N/A  
   DDR = 4% (276 patients)  

Abbreviations: DDR, drug-discontinuation rate; DRR, dose-reduction rate; ORR, objective response rate.

aClinical outcomes data on overall survival and progression-free survival emerging as clinical trials are ongoing.

bRelatively high dose reduction and discontinuation rates in MKIs due to drug-related adverse events like hypertension, diarrhea, rash, fatigue, hand-foot syndrome, proteinuria, hypopigmentation, QT prolongation, thrombosis, and hemorrhage preclude effective long-term use of MKIs in thyroid cancer.

BLU-667 (pralsetinib) and LOXO-292 (selpercatinib) are two highly potent and selective RET inhibitors designed to offset the weaknesses of MKIs (refs. 11, 27, 28; Table 1). High potency and RET selectivity were confirmed in robust preclinical models using multiple in vitro and in vivo RET-dependent tumor models (27, 28). In addition, favorable pharmacokinetic properties, including high bioavailability, predictable exposure, and minimal potential for drug–drug interactions, were confirmed as well. On-target acquired resistance with tyrosine kinase inhibitors (TKI) is always an issue in developmental therapeutics, specifically a mutation at the gatekeeper position that sterically hinders inhibitor binding, as seen in chronic myeloid leukemia (BCR–ABLT315I) or EGFR-mutant NSCLC (EGFRT790M). Similarly, the RET gatekeeper mutations V804L and V804M have been described and have been shown as an acquired resistance mechanism to MKIs (28, 34). Interestingly, these gatekeeper aberrations have been seen as germline mutations in patients presenting with MTC, and although cabozantinib and vandetanib do not cover these mutations adequately, the selective RET inhibitors BLU-667 and LOXO-292 were specifically designed to inhibit these mutations in subnanomolar concentrations (27, 28, 34).

Following preclinical validation, early clinical studies with BLU-667 showed remarkable responses in MKI-naïve and MKI-refractory patients with RET-rearranged NSCLC and patients with RET-mutant advanced MTC (27). Contemporaneously, utilizing rapid dose titration guided by real-time pharmacokinetic assessments to achieve meaningful clinical exposures safely and rapidly, a patient with RETM918T-mutant MTC and an acquired RETV804M gatekeeper resistance mutation, previously treated with six MKI regimens, was treated with LOXO-292 and achieved a rapid tumor response (28). A second patient with symptomatic brain metastases who was treated similarly experienced rapid tumor regression in the brain metastases. The mature clinical data that we currently have for selective RET inhibitors is mainly from RET fusion–positive NSCLC and RET-mutant MTC. Patients harboring RET-fusion NSCLC or PTC respond regardless of fusion partner, and patients with RET-mutant MTC respond regardless of germline or somatic mutation, including patients harboring germline V804M. Below we discuss the emerging clinical data for the selective RET inhibitors.

ARROW, a phase I clinical trial designed to evaluate the safety, tolerability, and efficacy of BLU-667 in multiple ascending doses in adults with RET-altered NSCLC, MTC, and other advanced solid tumors showed that the recommended phase II dose was 400 mg every day. As per the data presented at the American Society of Clinical Oncology 2019 annual meeting, 48 patients with RET-fusion NSCLC were evaluable for response assessment, including 35 patients previously treated with platinum-based chemotherapy (35). Nearly all patients (90%) had radiographic tumor reductions. The objective response rate (ORR) was 60% [one complete response and 20 partial responses (PR); all responses were confirmed], and the disease control rate (DCR) was 100% in the patients previously treated with platinum-based chemotherapy. Remarkably, pralsetinib was highly active regardless of RET fusion partner, including RET–KIF5B and RET–CCDC6. Thirty-two patients with RET-mutant MTC were evaluable for response assessment, including 16 patients previously treated with the MKIs cabozantinib or vandetanib (36). The ORR was 63% (nine confirmed PRs, one PR pending confirmation) and the DCR was 94%. On the basis of these data, pralsetinib has received two FDA breakthrough designations for NSCLC and MTC. In addition to NSCLC and MTC, in patients with RET fusion–positive PTC the ORR was 83% (5/6 patients) and PRs were noted in RET fusion–positive gastrointestinal malignancies (pancreatic cancer and cholangiocarcinoma) as well.

LIBRETTO-001 is a global phase I/II trial of selpercatinib (LOXO-292) in RET-altered cancers. On the basis of early promising data, selpercatinib received FDA breakthrough designations for RET fusion–positive NSCLC, RET mutation–positive MTC, and RET fusion–positive PTC. In the registration dataset consisting of the first 105 enrolled patients with RET fusion–positive NSCLC with prior platinum-based chemotherapy, selpercatinib treatment resulted in a 68% ORR [95% confidence interval (CI), 58%–76%; ref. 37]. These patients received a median of three prior systemic regimens (55% previously treated with an anti–PD-1/PD-L1 antibody and 48% previously treated with at least one MKI) and ORR was similar regardless of prior therapy. Up to 50% of RET fusion–positive NSCLCs can metastasize to the brain, and in the subset of patients with brain metastases in the registrational dataset, selpercatinib treatment demonstrated a central nervous system ORR of 91% (95% CI, 59%–100%). As of the data cutoff date of June 17, 2019, median duration of response (DOR) was 20.3 months (95% CI, 13.8–24.0) and median progression-free survival (PFS) was 18.4 months (95% CI, 12.9–24.9; ref. 37). In a safety analysis of all 531 patients enrolled to LIBRETTO-001, selpercatinib was well tolerated, with only 9 patients (1.7%) discontinuing therapy due to treatment-related toxicity.

In the RET-mutant MTC registration dataset (38) consisting of the first 55 enrolled patients with prior cabozantinib and/or vandetanib therapy, selpercatinib treatment resulted in a 56% ORR (95% CI, 42%–70%). ORR was similar regardless of prior MKI therapy, and 53% of patients were previously treated with ≥2 prior MKI. As of the data cutoff date of June 17, 2019, median DOR was not reached [95% CI, 11.1–not estimable (NE)] and median PFS was not reached (95% CI, 11.3–NE). Interestingly, in 76 cabozantinib/vandetanib (MKI)–naïve patients with RET-mutant MTC, selpercatinib treatment resulted in a 59% ORR (95% CI, 47%–70%; ref. 38). In 26 patients with RET fusion–positive thyroid cancer (PTC, Hürthle cell, poorly differentiated, and anaplastic) 62% ORR (95% CI, 41%–80%) was reached. Median DOR and PFS were not reached in the MKI-naïve or RET fusion–positive thyroid cancer cohorts, as the vast majority of patients continue to show tumor regression or remain progression-free (38).

The treatment of MTC with vandetanib represented the first attempt to target oncogenic RET. Despite a high DCR (73%), resistance developed with a median PFS of 27.9 months (39). With nearly a decade of clinical use, the mechanisms of vandetanib resistance remain largely unknown (40). This has largely been complicated by two factors. First, vandetanib is an MKI with antiangiogenic and anti-RET activity. As such, both pathways could be subject to resistance mechanisms. Second, the tools to examine resistance mechanisms have only recently begun to be applied to study RET-driven cancer. From clinical trial data, it is clear that both primary and acquired resistance mechanisms exist. Furthermore, similar to other oncogenic receptor tyrosine kinases, acquired resistance is expected to occur primarily through either target modification or bypass signaling (41, 42). Indeed, the earliest preclinical studies demonstrated that the RETV804M/L mutation functioned as a vandetanib gatekeeper, severely limiting its efficacy (43). More recent studies have demonstrated that V804M/L provides a gatekeeper function in oncogenic RET fusions, as well as limiting efficacy of other MKIs such as cabozantinib (44, 45). For this reason, the selective RET inhibitors have been screened and designed for activity against gatekeeper mutations.

It is only recently that an acquired RETV804 mutation has been demonstrated following resistance to multi-TKI treatment (28). The gatekeeper was detected in the circulating cell-free DNA (cfDNA) of a patient with MTC following treatment with consecutive TKIs over a 5-year time frame. In follow-up studies examining a small cohort of patients with MTC with RETM918T somatic driver mutations, 75% of patients had detectable circulating RETV804M cfDNA after development of resistance to vandetanib or cabozantinib (46), suggesting that acquisition of a RET gatekeeper mutation is a common mechanism of resistance in MTC. Acquired RET gatekeeper mutations on treatment with MKIs and successful treatment with the selective RET inhibitor selpercatinib has been reported for NSCLC as well (34). In the LURET patient cohort of vandetanib-treated patients, only a single case was observed to acquire a RET mutation after development of resistance (47, 48). This patient, with a CCDC6RET fusion, developed a novel RETS904F activation loop mutation shown to reduce drug binding. Additional studies will be needed to clarify the frequency of acquired RET mutation in these two cancer types and whether target modification is a less-favored mechanism of resistance for oncogenic fusions.

Evidence also exists for bypass mechanisms of resistance to RET inhibition. Notably, resistance to cabozantinib has been associated with acquisition of METD1228V in NSCLC (49) and MET amplification in colorectal cancer (50). In support of these findings, preclinical models have demonstrated that RET inhibition can be overcome through activation of MET or EGFR family members, as well as through activating RAS mutations (51–53).

In the near future, studies of acquired resistance in patients treated with selective RET inhibitors are expected, but as of now, unlike with EGFR- and ALK-driven cancers, we do not yet have vast patient experience to draw upon (54). However, what we have learned is that many of the discoveries of resistance mechanisms initially made in preclinical studies found themselves duplicated in patient studies, and later addressed by new clinical approaches. If we apply a similar approach to treatment of RET-driven cancer, then identification of the mechanism(s) of resistance becomes a critical step in providing an effective treatment. First, it is clear that the current generation of RET inhibitors provides the ability to use a higher targeting dose and is largely insensitive to acquisition of gatekeeper mutations. Thus, these inhibitors provide a logical treatment choice for TKI resistance in MTC, where acquired gatekeeper mutation frequency is high, or as a first-line therapy where the use of higher pharmacologic dosing has proven more effective. Addressing resistance mediated through bypass mechanisms presents a more difficult challenge, one that will become more common with next-generation TKIs. Here the specific mechanism of resistance needs to be identified, and a targeted drug combination therapy is needed. There is considerable evidence demonstrating cross-talk between RET and EGFR, with the activation of EGF known to trigger resistance to multikinase RET inhibitors. Clinically, targeting both receptors has proven effective (55) and should be considered. A more generalized approach is the use of an mTOR inhibitor to target resistance mediated through PI3K–AKT (56, 57). Phase I studies have demonstrated that the addition of everolimus is associated with a greater response rate and longer PFS than vandetanib alone in RET-driven tumors (58, 59). Whether a similar approach targeting MAPK pathway activation using MEK inhibitors would be effective remains to be addressed, although at least preclinically the combination has been shown to be effective in MTC cell lines (60). Development of an acquired NRASQ61K mutation was also seen with prolonged treatment of NSCLC cells with ponatinib (53).

Multiple resistance mutations have been reported from preclinical studies, including the gatekeeper mutation V804L to cabozantinib and solvent front mutations (SFM) G810A/S to vandetanib (61). Although the first-generation selective RET inhibitors are designed to cover the gatekeeper mutation, they do not adequately cover the SFMs. Emergence of SFMs has been reported preclinically (62). Recently, RET G810R, S, and C SFMs as acquired resistance mechanisms have been demonstrated in RET-aberrant patients who progressed on selective RET inhibitors (63). Second-generation RET inhibitors are in development that may have different properties. TPX-0046, a next-generation RET inhibitor that is structurally differentiated and potent against a broad range of mutations, has currently entered first-in-human studies (ClinicalTrials.gov Identifier: NCT04161391) after investigational new drug–enabling preclinical studies. Other selective RET inhibitors in development include BOS-172738 (NCT03780517) and TAS0953/HM06 (in preclinical development).

The considerable enthusiasm in the precision oncology of RET-dependent cancers including RET fusion–positive NSCLC, RET fusion–positive PTC, and RET-mutant MTC stems from the successful clinical trial results of selective RET inhibitors and offers a tantalizing array of opportunities in RET-dependent cancer research. The fewer off-target side effects, more effective control of growth, and sustained antitumor activity compared with MKIs opens up a new era of precision oncology in RET-driven cancers. Approval of these selective RET inhibitors will further the use of the drugs in the community. Next steps include clinical trials with these drugs earlier in the disease course, and combination therapy trials. Current RET inhibitors are a proof of principle for selective RET inhibition, but we need to know when we willget the most benefit from this specific targeting, and we clearly need to be prepared for addressing acquired resistance and escape from RET targeting.

V. Subbiah is a consultant/advisory board member at LOXO Oncology/Eli Lilly, Helsinn, R-Pharma US, Incyte, QED, Novartis, and Medimmune, and reports receiving commercial research grants from LOXO Oncology/Eli Lilly, Blueprint Medicines, Fujifilm, Pharmamar, D3, Pfizer, Multivir, Amgen, AbbVie, Agensys, Boston Biomedical, Idera, Turning Point Therapeutics, Exelixis, Inhibrx, Altum, Medimmune, Dragonfly Therapeutics, Takeda, Roche/Genentech, Novartis, Bayer, GSK, Nanocarrier, Berghealth, Incyte, and Northwest Biotherapeutics. No potential conflicts of interest were disclosed by the other author.

This work was supported in part by The Cancer Prevention and Research Institute of Texas (RP1100584), the Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, 1U01 CA180964, NCATS Grant UL1 TR000371 (Center for Clinical and Translational Sciences), and The MD Anderson Cancer Center Support Grant (P30 CA016672). The funders had no role in the design of the study; the collection, analysis, and interpretation of the data; the writing of the manuscript; and the decision to submit the manuscript for publication.

1.
Goldman
JM
,
Melo
JV
. 
Targeting the BCR-ABL tyrosine kinase in chronic myeloid leukemia
.
N Engl J Med
2001
;
344
:
1084
6
.
2.
Shaw
AT
,
Kim
D-W
,
Nakagawa
K
,
Seto
T
,
Crinó
L
,
Ahn
M-J
, et al
Crizotinib versus chemotherapy in advanced ALK-positive lung cancer
.
N Engl J Med
2013
;
368
:
2385
94
.
3.
Shaw
AT
,
Ou
S-HI
,
Bang
Y-J
,
Camidge
DR
,
Solomon
BJ
,
Salgia
R
, et al
Crizotinib in ROS1-rearranged non–small-cell lung cancer
.
N Engl J Med
2014
;
371
:
1963
71
.
4.
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
.
5.
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
.
6.
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
.
7.
Arighi
E
,
Borrello
MG
,
Sariola
H
. 
RET tyrosine kinase signaling in development and cancer
.
Cytokine Growth Factor Rev
2005
;
16
:
441
67
.
8.
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
.
9.
Stransky
N
,
Cerami
E
,
Schalm
S
,
Kim
JL
,
Lengauer
C
. 
The landscape of kinase fusions in cancer
.
Nat Commun
2014
;
5
:
4846
.
10.
Soares
P
,
Trovisco
V
,
Rocha
AS
,
Lima
J
,
Castro
P
,
Preto
A
, et al
BRAF mutations and RET/PTC rearrangements are alternative events in the etiopathogenesis of PTC
.
Oncogene
2003
;
22
:
4578
80
.
11.
Drilon
A
,
Hu
ZI
,
Lai
GGY
,
Tan
DSW
. 
Targeting RET-driven cancers: lessons from evolving preclinical and clinical landscapes
.
Nat Rev Clin Oncol
2018
;
15
:
150
.
12.
Kato
S
,
Subbiah
V
,
Marchlik
E
,
Elkin
SK
,
Carter
JL
,
Kurzrock
R
. 
RET aberrations in diverse cancers: next-generation sequencing of 4,871 patients
.
Clin Cancer Res
2017
;
23
:
1988
97
.
13.
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
.
14.
Lee
SH
,
Lee
JK
,
Ahn
MJ
,
Kim
DW
,
Sun
JM
,
Keam
B
, et al
Vandetanib in pretreated patients with advanced non-small cell lung cancer-harboring RET rearrangement: a phase II clinical trial
.
Ann Oncol
2017
;
28
:
292
7
.
15.
Takahashi
M
,
Ritz
J
,
Cooper
GM
. 
Activation of a novel human transforming gene, ret, by DNA rearrangement
.
Cell
1985
;
42
:
581
8
.
16.
Takahashi
M
,
Cooper
GM
. 
ret transforming gene encodes a fusion protein homologous to tyrosine kinases
.
Mol Cell Biol
1987
;
7
:
1378
85
.
17.
Grieco
M
,
Santoro
M
,
Berlingieri
MT
,
Melillo
RM
,
Donghi
R
,
Bongarzone
I
, et al
PTC is a novel rearranged form of the ret proto-oncogene and is frequently detected in vivo in human thyroid papillary carcinomas
.
Cell
1990
;
60
:
557
63
.
18.
Santoro
M
,
Carlomagno
F
,
Romano
A
,
Bottaro
DP
,
Dathan
NA
,
Grieco
M
, et al
Activation of RET as a dominant transforming gene by germline mutations of MEN2A and MEN2B
.
Science
1995
;
267
:
381
3
.
19.
Horibata
S
,
Rice
EJ
,
Mukai
C
,
Marks
BA
,
Sams
K
,
Zheng
H
, et al
ER-positive breast cancer cells are poised for RET-mediated endocrine resistance
.
PLoS One
2018
;
13
:
e0194023
.
20.
Mulligan
LM
. 
GDNF and the RET receptor in cancer: new insights and therapeutic potential
.
Front Physiol
2018
;
9
:
1873
.
21.
Mulligan
LM
. 
RET revisited: expanding the oncogenic portfolio
.
Nat Rev Cancer
2014
;
14
:
173
86
.
22.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
23.
Hanahan
D
,
Weinberg
RA
. 
The hallmarks of cancer
.
Cell
2000
;
100
:
57
70
.
24.
Richardson
DS
,
Gujral
TS
,
Peng
S
,
Asa
SL
,
Mulligan
LM
. 
Transcript level modulates the inherent oncogenicity of RET/PTC oncoproteins
.
Cancer Res
2009
;
69
:
4861
9
.
25.
Hyndman
BD
,
Crupi
MJF
,
Peng
S
,
Bone
LN
,
Rekab
AN
,
Lian
EY
, et al
Differential recruitment of E3 ubiquitin ligase complexes regulates RET isoform internalization
.
J Cell Sci
2017
;
130
:
3282
96
.
26.
Bagheri-Yarmand
R
,
Williams
MD
,
Grubbs
EG
,
Gagel
RF
. 
ATF4 targets RET for degradation and is a candidate tumor suppressor gene in medullary thyroid cancer
.
J Clin Endocrinol Metab
2017
;
102
:
933
41
.
27.
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
.
28.
Subbiah
V
,
Velcheti
V
,
Tuch
BB
,
Ebata
K
,
Busaidy
NL
,
Cabanillas
ME
, et al
Selective RET kinase inhibition for patients with RET-altered cancers
.
Ann Oncol
2018
;
29
:
1869
76
.
29.
Kikumori
T
,
Cote
GJ
,
RF
G
. 
A ribozyme directed against RET proto-oncogene inhibits medullary thyroid carcinoma cell growth
.
In
proceedings of the 92th Annual Meeting of the American Association of Cancer Research; 2001 Mar 24–28
;
New Orleans, LA:
AACR
; 
2001
.
30.
Drosten
M
,
Frilling
A
,
Stiewe
T
,
Putzer
BM
. 
A new therapeutic approach in medullary thyroid cancer treatment: inhibition of oncogenic RET signaling by adenoviral vector-mediated expression of a dominant-negative RET mutant
.
Surgery
2002
;
132
:
991
7
.
31.
Drosten
M
,
Stiewe
T
,
Putzer
BM
. 
Antitumor capacity of a dominant-negative RET proto-oncogene mutant in a medullary thyroid carcinoma model
.
Hum Gene Ther
2003
;
14
:
971
82
.
32.
Carlomagno
F
,
Vitagliano
D
,
Guida
T
,
Ciardiello
F
,
Tortora
G
,
Vecchio
G
, et al
ZD6474, an orally available inhibitor of KDR tyrosine kinase activity, efficiently blocks oncogenic RET kinases
.
Cancer Res
2002
;
62
:
7284
90
.
33.
Cohen
EE
,
Rosen
LS
,
Vokes
EE
,
Kies
MS
,
Forastiere
AA
,
Worden
FP
, et al
Axitinib is an active treatment for all histologic subtypes of advanced thyroid cancer: results from a phase II study
.
J Clin Oncol
2008
;
26
:
4708
13
.
34.
Wirth
LJ
,
Kohno
T
,
Udagawa
H
,
Matsumoto
S
,
Ishii
G
,
Ebata
K
, et al
Emergence and targeting of acquired and hereditary resistance to multikinase RET inhibition in patients with RET-altered cancer
.
JCO Precision Oncology
2019
Sep 11
.
35.
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
.
36.
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
.
37.
Drilon
A
,
Oxnard
G
,
Wirth
L
,
Besse
B
,
Gautschi
O
,
Tan
SWD
, 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
.
J Thorac Oncol
2019
;
14
:
S6
S7
.
38.
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
.
39.
Wells
SA
 Jr
,
Gosnell
JE
,
Gagel
RF
,
Moley
J
,
Pfister
D
,
Sosa
JA
, et al
Vandetanib for the treatment of patients with locally advanced or metastatic hereditary medullary thyroid cancer
.
J Clin Oncol
2010
;
28
:
767
72
.
40.
Sim
MW
,
Cohen
MS
. 
The discovery and development of vandetanib for the treatment of thyroid cancer
.
Expert Opin Drug Discov
2014
;
9
:
105
14
.
41.
Lovly
CM
,
Shaw
AT
. 
Molecular pathways: resistance to kinase inhibitors and implications for therapeutic strategies
.
Clin Cancer Res
2014
;
20
:
2249
56
.
42.
Pagliarini
R
,
Shao
W
,
Sellers
WR
. 
Oncogene addiction: pathways of therapeutic response, resistance, and road maps toward a cure
.
EMBO Rep
2015
;
16
:
280
96
.
43.
Carlomagno
F
,
Guida
T
,
Anaganti
S
,
Vecchio
G
,
Fusco
A
,
Ryan
AJ
, et al
Disease associated mutations at valine 804 in the RET receptor tyrosine kinase confer resistance to selective kinase inhibitors
.
Oncogene
2004
;
23
:
6056
63
.
44.
Huang
Q
,
Schneeberger
VE
,
Luetteke
N
,
Jin
C
,
Afzal
R
,
Budzevich
MM
, et al
Preclinical modeling of KIF5B-RET fusion lung adenocarcinoma
.
Mol Cancer Ther
2016
;
15
:
2521
9
.
45.
Liu
X
,
Shen
T
,
Mooers
BHM
,
Hilberg
F
,
Wu
J
. 
Drug resistance profiles of mutations in the RET kinase domain
.
Br J Pharmacol
2018
;
175
:
3504
15
.
46.
Busaidy
NL
,
Cabanillas
ME
,
Sherman
SI
,
Habra
M
,
Dadu
R
,
Hu
MI
, et al
Emergence of V804M resistance gatekeeper mutation in sporadic medullary thyroid carcinoma patients treated with TKI tyrosine kinase inhibitors
.
Thyroid
2017
;
2017
:
A-166
A-88
.
47.
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
.
48.
Nakaoku
T
,
Kohno
T
,
Araki
M
,
Niho
S
,
Chauhan
R
,
Knowles
PP
, et al
A secondary RET mutation in the activation loop conferring resistance to vandetanib
.
Nat Commun
2018
;
9
:
625
.
49.
Bahcall
M
,
Sim
T
,
Paweletz
CP
,
Patel
JD
,
Alden
RS
,
Kuang
Y
, et al
Acquired METD1228V mutation and resistance to MET inhibition in lung cancer
.
Cancer Discov
2016
;
6
:
1334
41
.
50.
Jia
J
,
Morse
MA
,
Nagy
RJ
,
Lanman
RB
,
Strickler
JH
. 
Cell-free DNA profiling to discover mechanisms of exceptional response to cabozantinib plus panitumumab in a patient with treatment refractory metastatic colorectal cancer
.
Front Oncol
2018
;
8
:
305
.
51.
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
.
52.
Plenker
D
,
Riedel
M
,
Bragelmann
J
,
Dammert
MA
,
Chauhan
R
,
Knowles
PP
, et al
Drugging the catalytically inactive state of RET kinase in RET-rearranged tumors
.
Sci Transl Med
2017
;
9
:
pii
:
eaah6144
.
53.
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
.
54.
Camidge
DR
,
Pao
W
,
Sequist
LV
. 
Acquired resistance to TKIs in solid tumours: learning from lung cancer
.
Nat Rev Clin Oncol
2014
;
11
:
473
81
.
55.
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
.
56.
Heilmann
AM
,
Subbiah
V
,
Wang
K
,
Sun
JX
,
Elvin
JA
,
Chmielecki
J
, et al
Comprehensive genomic profiling of clinically advanced medullary thyroid carcinoma
.
Oncology
2016
;
90
:
339
46
.
57.
Gild
ML
,
Landa
I
,
Ryder
M
,
Ghossein
RA
,
Knauf
JA
,
Fagin
JA
. 
Targeting mTOR in RET mutant medullary and differentiated thyroid cancer cells
.
Endocr Relat Cancer
2013
;
20
:
659
67
.
58.
Subbiah
V
,
Berry
J
,
Roxas
M
,
Guha-Thakurta
N
,
Subbiah
IM
,
Ali
SM
, et al
Systemic and CNS activity of the RET inhibitor vandetanib combined with the mTOR inhibitor everolimus in KIF5B-RET re-arranged non-small cell lung cancer with brain metastases
.
Lung Cancer
2015
;
89
:
76
9
.
59.
Subbiah
V
,
Cascone
T
,
Hess
KR
,
Subbiah
IM
,
Nelson
S
,
Morikawa
N
, et al
Multi-kinase RET inhibitor vandetanib combined with mTOR inhibitor everolimus in patients with RET rearranged non-small cell lung cancer
.
J Clin Oncol
2018
;
36
:
9035
.
60.
Koh
YW
,
Shah
MH
,
Agarwal
K
,
McCarty
SK
,
Koo
BS
,
Brendel
VJ
, et al
Sorafenib and Mek inhibition is synergistic in medullary thyroid carcinoma in vitro
.
Endocr Relat Cancer
2012
;
19
:
29
38
.
61.
Terzyan
SS
,
Shen
T
,
Liu
X
,
Huang
Q
,
Teng
P
,
Zhou
M
, et al
Structural basis of resistance of mutant RET protein-tyrosine kinase to its inhibitors nintedanib and vandetanib
.
J Biol Chem
2019
;
294
:
10428
37
.
62.
Drilon
A
,
Rogers
E
,
Zhai
D
,
Deng
W
,
Zhang
X
,
Lee
D
, et al
506P TPX-0046 is a novel and potent RET/SRC inhibitor for RET-driven cancers
.
Ann Oncol
2019
;
30
:
mdz244.068
.
63.
Solomon
BJ
,
Tan
L
,
Lin
JJ
,
Wong
SQ
,
Hollizeck
S
,
Ebata
K
, et al
RET solvent front mutations mediate acquired resistance to selective RET inhibition in RET-driven malignancies
.
J Thorac Oncol
2020
Jan 24 [Epub ahead of print]
.