Skip to main content
ACS Medicinal Chemistry Letters logoLink to ACS Medicinal Chemistry Letters
. 2021 Nov 6;12(12):1912–1919. doi: 10.1021/acsmedchemlett.1c00450

Antitarget Selectivity and Tolerability of Novel Pyrrolo[2,3-d]pyrimidine RET Inhibitors

Casey J N Mathison †,*, Yang Yang , John Nelson , Zhihong Huang , Jiqing Jiang , Donatella Chianelli , Paul V Rucker , Jason Roland , Yun Feng Xie , Robert Epple , Badry Bursulaya , Christian Lee , Mu-Yun Gao , Jennifer Shaffer , Sergio Briones , Yelena Sarkisova , Anna Galkin , Lintong Li , Nanxin Li , Chun Li , Su Hua , Shailaja Kasibhatla , Jacqueline Kinyamu-Akunda , Rie Kikkawa , Valentina Molteni , John E Tellew
PMCID: PMC8667305  PMID: 34917254

Abstract

graphic file with name ml1c00450_0011.jpg

The selective inhibition of RET kinase as a treatment for relevant cancer types including lung adenocarcinoma has garnered considerable interest in recent years and prompted a variety of efforts toward the discovery of small-molecule therapeutics. Hits uncovered via the analysis of archival kinase data ultimately led to the identification of a promising pyrrolo[2,3-d]pyrimidine scaffold. The optimization of this pyrrolo[2,3-d]pyrimidine core resulted in compound 1, which demonstrated potent in vitro RET kinase inhibition and robust in vivo efficacy in RET-driven tumor xenografts upon multiday dosing in mice. The administration of 1 was well-tolerated at established efficacious doses (10 and 30 mg/kg, po, qd), and plasma exposure levels indicated a minimal risk of KDR or hERG inhibition in vivo, as evaluated by Miles assay and free plasma concentrations, respectively.

Keywords: RET, receptor tyrosine kinase, KDR, pyrrolo[2, 3-d]pyrimidine, Miles assay


The RET (REarranged during Transfection) gene, first described by Takahashi,1 encodes an RTK (transmembrane Receptor Tyrosine Kinase) that plays a key role in neuronal signaling as the canonical receptor for ligands of the GDNF (Glial cell-line Derived Neurotrophic Factor) family. Consequently, downstream signaling via RET activation has been demonstrated to be critical for early-stage peripheral nervous system development.2,3 Gain-of-function mutations of the proto-oncogene, however, have long been recognized as potential drivers in a range of malignancies, including a relatively small subset (1–2%) of non-small cell lung cancer (NSCLC) cases.4 As such, the therapeutic deployment of small-molecule inhibitors to address RET oncogene-addiction in the context of lung adenocarcinoma has garnered considerable interest.58 Early efforts in this area have focused on the assessment of multikinase RET inhibitors in the clinic, but this approach has suffered from instances of dose-limiting toxicity attributed to the inhibition of non-RET kinases, most notably KDR (Kinase insert Domain Receptor).9 Current efforts in this field have therefore focused on the discovery of novel RET inhibitors with broad kinase selectivity to minimize the risk of off-target toxicities.

Late-stage clinical reports and subsequent approval of the treatment of RET-driven cancers with the selective inhibitors pralsetinib (BLU-667)10,11 and selpercatinib (LOXO-292)12,13 are promising breakthroughs and have reinforced the notion that the potent and selective inhibition of acquired RET kinase fusions can effect improved clinical outcomes in relevant oncogenic cohorts.14,15 Our preliminary efforts to devise selective RET inhibitors resulted in the discovery of a highly potent pyrazolo[1,5-a]pyrimidine compound with considerable selectivity against antitarget KDR, brain penetration, and robust efficacy in vivo. Ultimately, though, poor tolerability proximal to the efficacious dose precluded further preclinical evaluation, as has been outlined in a previous disclosure.16 Our subsequent investigation of alternative scaffolds, in the hope of improving upon in vivo compound tolerability thresholds, resulted in focused efforts on a pyrrolo[2,3-d]pyrimidine core substructure, which is described herein.

The decision to pursue a pyrrolo[2,3-d]pyrimidine scaffold was initially informed by Gini coefficient analysis17 performed on historical enzymatic and Ba/F318 kinase data, which resulted in several intriguing hits, including 2 and 3 (Table 1).19 Additional factors, including favorable ligand efficiency and LipE of these pyrrolo[2,3-d]pyrimidine hits, reinforced our interest in this scaffold and substantiated the assessment of additional archival analogs. During the course of this evaluation, our internal collection revealed several additional pyrrolo[2,3-d]pyrimidine RET kinase inhibitors including piperazine-containing compound 4. We found 4 to be particularly appealing because it demonstrated both broad selectivity across a panel of tyrosine kinases (Table 1) and favorable physicochemical properties. Although 4 demonstrated only moderate RET potency (Table 1), the activity of structurally similar hit compounds 2 and 3 gave us confidence that further cell-based RET inhibitory activity could be subsequently introduced during the course of wet chemistry efforts.

Table 1. Cellular Activity and Tyrosine Kinase Selectivity of Archival Hit Compoundsa.

graphic file with name ml1c00450_0007.jpg

compound 2 3 4
WTb,c 1.3 1.6 >10
KIF5B-RETc 0.011 0.007 0.24
KIF5B-RETV804Mc 2.1 1.1 >10
RET 0.051 0.0040 0.92
BCR-ABL 0.043 0.058 4.2
NPM-ALK 0.37 0.87 >8.5
BLK 0.57 0.14 3.6
BMX 6.9 10 >11
FGFR1 2.1 0.99 >11
FGFR2 0.027 0.19 >2.9
FGFR3 0.49 1.1 >9.4
FGFR4 1.1 2.4 >11
FGR2 0.37 0.022 2.6
FLT1 0.11 0.14 >6.7
FLT3 4.4 0.97 >11
FMS 0.96 0.48 >11
IGF1R 2.8 2.7 >11
INSR 1.6 1.4 >8.2
JAK2 5.1 6.6 >11
KDR 0.15 0.058 >7.8
KIT 0.042 0.043 3.4
LCK 0.13 0.062 3.5
LYN 0.13 0.17 5.0
MER 5.5 4.9 >11
MET 5.8 6.0 >11
PDGFRα 0.34 0.18 >10
PDGFRβ 0.42 0.27 >11
RON 6.0 7.0 >11
ROS NTd 0.042 >11
SRC 0.063 0.12 2.4
SYK 2.4 4.9 >11
TIE1 4.4 2.5 >11
TRKA 1.5 1.2 9.3
TRKB 1.3 1.4 8.5
TRKC 0.92 0.51 >8.1
TYRO3 2.2 3.1 >11
ZAP70 2.1 2.9 >11
a

Proliferation assays run in 1536 well format, employing Ba/F3 cells transfected with Tel fusions of the indicated kinase, and IC50 values reported in μM, unless otherwise noted.

b

Parental Ba/F3 cells utilized.

c

Assay run in 384 well format.

d

Not tested.

Using 4 as a basis template for structure–activity relationship (SAR) investigations, we first examined substitution at R1. Our initial objectives focused on both improving RET inhibition in cellular assays and finding an appropriate phenol isostere due to the well-recognized reactive and phase II metabolism concerns for the moiety. A library of R1 derivatives was synthesized via Suzuki coupling of the appropriate aryl boronic acid/ester with the intermediate (cis)-1-(4-(−3-(4-amino-5-bromo-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclobutyl)piperazin-1-yl)ethanone (Scheme 1), and the key findings are summarized in Table 2. A cursory exploration of R1 phenol trajectory found meta substitution to be ideal for both enzymatic and cell-based RET activity, akin to the phenolic compounds that surfaced from screening efforts (Table 1). A survey of halogen substitution of the phenol ring found chloro-substitution at the four-position to be particularly advantageous, and gratifyingly, this boost in potency was also translated to the corresponding aniline compound 10 (Table 2). Furthermore, the transition from phenol to aniline also led to increased selectivity versus KDR, as evidenced by the matched pair 8/10. These initial findings at R1 were found to be consistent with an X-ray cocrystal structure of 10 with RET kinase, which indicated H bonds between the R1 aniline and kinase residues E775 (αC-helix) and S891 (adjacent to the DFG motif). Additional hydrophobic interaction of the R1 4-chloro moiety (10) with the nonpolar carbon chain of amphipathic residue K758 (<4 Å, Figure 1) also was supportive of our preliminary SAR (Table 2).

Scheme 1. Synthetic Route for R1 Analogs.

Scheme 1

NaBH(OAc)3, DCM.

TFA, DCM.

2-(4,6-Dichloropyrimidin-5-yl)acetaldehyde, iPr2NEt, EtOH.

Br2, AcOH.

NH4OH, THF.

R1B(OH)2 or R1B(pin), Na2CO3, K3PO4, SiliaCat DPP-Pd, dioxane.

Table 2. Impact of R1 Modification on RET Potencya.

graphic file with name ml1c00450_0009.jpg

a

IC50 values reported in μM.

b

Biochemical (aa 658–1072) HTRF-based assay.

c

Cell-based proliferation assays.

Figure 1.

Figure 1

X-ray cocrystal structure of 10 and RET kinase reveals interaction of R1 NH2 with kinase residues E775 (3.3 Å) and S891 (2.8 Å).

The increased cellular potency achieved through replacement of the phenol moiety at R1 also advantageously coincided with an improvement in the pharmacokinetics (PK) of the scaffold. When comparing compounds 4 and 10 in PK studies, the latter was found to possess much improved oral plasma exposure, clearance, and bioavailability (Table 3). The suitable PK profile of 10 subsequently allowed for its rapid evaluation in mouse efficacy models, where it was hoped that sufficient in vivo RET-inhibitory exposures could be achieved. We also desired to quickly benchmark the scaffold in terms of tolerability, as poor tolerability ultimately doomed our previous efforts on a pyrazolo[1,5-a]pyrimidine series.16 As such, mice were dosed orally with 10 in a transgenic RIE KIF5B-RET xenograft tumor model at 10, 30, and 60 mg/kg once daily for 14 days. Gratifyingly, robust tumor regression was observed in the 60 mg/kg cohort, with corresponding inhibition/stasis demonstrated at lower doses. All doses were well-tolerated, with no adverse events or body weight loss observed (Supporting Information). Subsequent western blot analysis of intratumoral tissue collected on day 14 of the study confirmed target engagement, where a significant inhibition of RET phosphorylation was observed at a 5 h postdose time point for the 60 mg/kg study group, although the activity substantially declined by 24 h (Supporting Information). Ultimately, the promising results of this study gave us confidence to move forward and screen additional pyrrolo[2,3-d]pyrimidine analogs for efficacy in relevant murine xenograft models.

Table 3. PK Parameters of Key Compoundsa.

graphic file with name ml1c00450_0010.jpg

a

PK studies run in CD-1 mice with formulation of 1 mg/mL in PEG300/D5W, 3:1 for iv and in 0.5% MC/Tween80 for po, unless otherwise noted.

b

Formulation of 2.5 mg/mL in PEG300/D5W, 3:1 for both iv and po in Balb/C mice.

c

Balb/C mice used.

d

20 mg/kg of compound HCl salt dosed at 2 mg/mL in Wistar mice.

e

Formulated in 10% Eudragit, 0.5% Tween80 in 50 mM acetate buffer in Balb/C mice.

f

2 mg/mL used.

Resulting efforts to improve upon compound 10 primarily focused on lowering the in vivo efficacious dose. The first manner in which we sought to accomplish this was through strategic efforts to further increase the RET potency. Satisfied, for the time being, with prior modification at R1 (vide supra), we turned our attention to the evaluation of R2. The 10/RET kinase cocrystal structure (Figure 1) informed a detailed understanding of the R2-region topography and suggested that more wide-ranging changes may be tolerated to improve the potency. Molecular docking served to guide these synthetic efforts, where four-, five-, and six-membered aliphatic rings flanked by terminal hydrophilicity abutting the solvent front were thought to be well-suited for RET inhibitory activity. More tailored attempts to create additional discrete polar contacts seemed to offer only a slim chance of success, however, as RET-specific residues in this region appeared to be poorly accessible. Second, we sought to lower the in vivo efficacious dose of this scaffold through the identification and elimination of R2 metabolic soft spots. Metabolic identification (MetID) analysis of selected analogs streamlined the prosecution of the R2-region SAR, with the objective of achieving reduced clearance and increased oral exposures. Subsequent testing of these R2 analogs in vitro and in vivo validated our efforts, as cell-based RET potency was successfully increased by two- to three-fold (Table 4), and CL reduced from 15 to <2 mL/min/kg (Table 3), respectively.

Table 4. Optimization of in Vivo Efficacy and Antitarget Windowsa.

  Ba/F3
LC2/ad hERG
  in vivo mouse efficacyb
Cmpd KIF5B-RET Tel-KDR WT CCDC6-RET bindingc MPCd PPBe Eff Dosef Eff AUCg Eff Cmaxg Eff AUCfreeh Eff Cmax,freeh
10 0.044 0.84 4.6 0.046 19 7.9 64.4 60 115 19 41 6.8
11 0.015 0.66 2.6 0.035 25 5.3 96.9 10 50 8.3 1.6 0.26
12 0.020 0.44 4.1 0.11 9.9 3.0 99.58 3 106 7.4 0.45 0.031
1 0.022 0.80 8.1 0.051 >30 6.0 99.34 10 136 8.4 0.90 0.055
a

IC50 values reported in μM for cell-based proliferation and hERG assays.

b

Evaluated in multiday RIE KIF5B-RET xenograft model.

c

Assessed with 3H-dofetilide.

d

Manual patch clamp.

e

Mouse plasma protein binding percentage. For cases >99%, 20% diluted plasma and rapid equilibrium dialysis utilized with calculation by Kalvass equation.

f

Oral dose (mg/kg, qd) at which tumor regression was observed.

g

Area under curve (AUC) (h*μM) and Cmax (μM) observed at regression dose after days 14, 14, 4, and 13 for compounds 10, 11, 12, and 1, respectively.

h

Unbound fraction of AUC (h*μM) and Cmax (μM) at regression dose.

As a result of the improved RET cellular potency and oral exposures achieved, we felt that several of our advanced R2 analogs were well-suited for further evaluation in murine xenograft models (Table 4). When compound 11 was assessed for efficacy and tolerability in the aforementioned 14-day KIF5B-RET xenograft model, its significant RET potency and low clearance translated to significant tumor regression at 10 mg/kg, with growth inhibition observed at 1 and 3 mg/kg. With respect to tolerability, mice dosed at 30 mg/kg required a drug holiday following significant body weight loss (BWL); however, once dosing resumed on day 9, no further adverse events were noted during in-life observation or upon necropsy. Similarly, the in vivo analysis of compound 12, studied in mice for 12 days with oral dosing qd at 1, 3, and 10 mg/kg, revealed significant tumor regression at 3 mg/kg. Although, at 10 mg/kg, body weights initially dropped <10% but gradually stabilized over time with ongoing dosing. Whereas the origin of the documented BWL remained unclear at this point, its manageability and the robust low-dose efficacy observed with 11 and 12 made us cautiously optimistic that further widening of the therapeutic index could be achieved with other scaffold analogs.

During the course of our optimization efforts, we discovered compound 1, a derivative of 12 where R1 fluorination served to modestly reduce activity against antitargets KDR and hERG while retaining RET potency (Table 4). The PK properties were similar to those of 12, where a prolonged half-life, low clearance, and good bioavailability (Table 3) translated to high levels of compound exposure in multiday in vivo mouse xenograft models (Table 4, Supporting Information). For instance, when compound 1 was examined in a 16 day RIE KIF5B-RET mechanistic mouse xenograft model at 1, 3, 10, 30, and 60 mg/kg, exposures were generally dose proportional, and analysis on day 13 versus day 1 suggested that only minor compound accumulation occurred (Supporting Information). With regard to compound efficacy in this study, tumor inhibition, stasis, and regression were observed in the 1, 3, and 10 mg/kg cohorts, respectively (Figure 2). The tolerability for compound 1 was found to be significantly improved relative to that for other scaffold analogs (vide supra), as oral dosing of 30 mg/kg also demonstrated robust efficacy with no adverse effects. However, in the 60 mg/kg arm of the study, significant BWL among mice was observed, where subsequent necropsy revealed substantial gastrointestinal toxicity. The slightly lower exposures on day 1 for the 60 mg/kg group (AUC: 369 h*μM, Cmax: 19 μM) than on day 13 for the well-tolerated 30 mg/kg dose cohort (AUC: 409 h*μM, Cmax: 25 μM) seemed to be indicative of a relatively steep and acutely sensitive dose-toxicity paradigm, akin to compounds 11 and 12 (vide supra). Nevertheless, this widened therapeutic window marked a key achievement for the project and warranted the advancement of 1 to further profiling and more advanced mouse xenograft models.

Figure 2.

Figure 2

In a 16 day study, the administration of compound 1 (po, qd, MC/Tween80, suspension) resulted in significant tumor regression at well-tolerated doses of 10 and 30 mg/kg in RIE KIF5B-RET xenograft mice.

To gain an understanding of the PK/PD (pharmacodynamics) relationship for compound 1, we conducted a time-course analysis in RIE KIF5B-RET tumor-bearing mice with a single administration of the efficacious dose (10 mg/kg). As depicted in Figure 3, a clear relationship between the in vivo exposure of 1 and the intratumoral pRET inhibition, as measured by western blot, was found over the course of 48 h. In addition, 1 was analyzed in the context of a 21-day disease-relevant CCDC6-RET LC-2 tumor tissue model, which resulted in tumor regression at 10 and 30 mg/kg, with the compound being well tolerated (Figure 4). Following the cessation of treatment, mice were monitored for several weeks, and tumor regrowth was found to be heterogeneous but generally weak in the case of fully regressed tumors (Supporting Information).

Figure 3.

Figure 3

Intratumoral RET target engagement relative to plasma exposure in RIE KIF5B-RET xenograft mice at tumor regression dose (10 mg/kg, po, MC/Tween80, suspension) following a single dose of compound 1.

Figure 4.

Figure 4

Multiday treatment of LC-2/ad tumor-bearing mice with 1 (21 day, po, qd, MC/Tween80, suspension) is well tolerated and leads to significant regression at 10 and 30 mg/kg.

Now confident that well-tolerated robust tumor regression could be achieved with 1, we sought to examine how safety windows against specific antitargets might be impacted by efficacious levels of in vivo compound exposure. With regard to KDR, a well-recognized side effect of its inhibition in vivo is vascular toxicity mediated by disruption of the VEGF signaling pathway. Therefore, we hoped to determine if efficacious exposures of 1 would only minimally engage KDR in vivo and not result in the significant interruption of VEGF-mediated vascular effects. As such, a Miles assay20 was performed at the maximum tolerated dose of 30 mg/kg to establish if in vitro cell-based RET/KDR selectivity for 1 (36-fold, Table 4) would be translated in vivo. As can be seen in Figure 5, compound 1 does not significantly impact the vascular permeability in non-tumor-bearing mice as compared with the multi-RTK inhibitor cabozantinib (at 14 day RIE KIF5B-RET model tumor stasis dose of 60 mg/kg, Supporting Information).

Figure 5.

Figure 5

Representative images of non-tumor-bearing mice (four or five per group) demonstrate that 1 does not significantly impact VEGF-driven Evan’s Blue Dye vascular permeability in the Miles assay over the course of 24 h.

Furthermore, despite hERG manual patch clamp data being <8 μM across the late-stage lead compounds in Table 4, plasma exposure data gleaned from efficacy studies gave us confidence that we could effectively engage RET in vivo without leading to significant hERG inhibition. The calculation of maximum free plasma concentrations for compounds at their efficacious dose (where tumor regression was observed in mouse RIE KIF5B-RET efficacy studies) established that the modification of R2 led to progressively lower free concentrations, which were ultimately orders of magnitude below hERG IC50 values from both binding and manual patch clamp assays in the case of 1 (Table 4). Proportionally wide hERG safety margins for 1 were also found at the tolerated dose of 30 mg/kg (AUCfree: 2.7 h*μM, Cmax,free: 0.16 μM), stemming from its reasonably linear PK profile and minimal compound accumulation.

To summarize, efforts to optimize a pyrrolo[2,3-d]pyrimidine scaffold successfully demonstrated a correlation between in vitro cell-based kinase inhibition and in vivo murine model efficacy, as evidenced by robust RET fusion tumor regression and VEGF/KDR-driven vascular permeability. Furthermore, multiday studies with compound 1 proved that good tolerability could be achieved, with substantial tumor regression being observed at both 10 and 30 mg/kg in both KIF5B-RET and LC-2/ad xenograft mouse models. Calculations involving the free plasma exposure of compound 1 in these murine efficacy studies also suggested that other known off-target toxicity risks, such as hERG inhibition, should be minimal. The balanced composite profile of compound 1 ultimately warranted its advancement to a range of preclinical studies, including, but not limited to, dose formulation, rat toxicology, and pharmacokinetics in nonrodent species.

Acknowledgments

We thank Thomas Hollenbeck for LCMS-ES data and David Jones for NMR data. We also thank the staff at BL5.0.3 Advanced Light Source (Berkeley, CA), a DOE Office of Science User Facility under contract no. DE-AC02-05CH11231, supported in part by the ALS-ENABLE program funded by the National Institutes of Health, National Institute of General Medical Sciences, grant P30 GM124169-01.

Glossary

Abbreviations

RET

rearranged during transfection

RTK

receptor tyrosine kinase

GDNF

glial cell-line derived neurotrophic factor

NSCLC

non-small cell lung cancer

KDR

kinase insert domain receptor

SAR

structure–activity relationship

RIE

rat intestinal epithelial

BWL

body weight loss

VEGF

vascular endothelial growth factor

Supporting Information Available

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsmedchemlett.1c00450.

  • Experimental procedures and characterization of all compounds; in vitro assay descriptions; cellular kinase panel activity for compounds 4, 10, 11, 12, and 1; PK data for compounds 4, 10, 11, 12, and 1; in vivo efficacy/tolerability study design and results for compounds 10, 11, 12, and 1; PK/PD study design and results for compound 1; Miles assay study design and representative images for compound 1 (PDF)

Accession Codes

PDB accession code: 7RUN.

The authors declare no competing financial interest.

Supplementary Material

ml1c00450_si_001.pdf (1.5MB, pdf)

References

  1. Takahashi M.; Ritz J.; Cooper G. M. Activation of a novel human transforming gene, ret, by DNA rearrangement. Cell 1985, 42, 581–588. 10.1016/0092-8674(85)90115-1. [DOI] [PubMed] [Google Scholar]
  2. Delalande J.-M.; Barlow A. J.; Thomas A. J.; Wallace A. S.; Thapar N.; Erickson C. A.; Burns A. J. The receptor tyrosine kinase RET regulates hindgut colonization by sacral neural crest cells. Dev. Biol. 2008, 313, 279–292. 10.1016/j.ydbio.2007.10.028. [DOI] [PubMed] [Google Scholar]
  3. Mulligan L. M. GDNF and the RET receptor in cancer: new insights and therapeutic potential. Front. Physiol. 2019, 9, 1873. 10.3389/fphys.2018.01873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Shaw A. T.; Hsu P. P.; Awad M. M.; Engelman J. A. Tyrosine kinase gene rearrangements in epithelial malignancies. Nat. Rev. Cancer 2013, 13, 772–787. 10.1038/nrc3612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Mologni L.; Gambacorti-Passerini C.; Goekjian P.; Scapozza L. RET kinase inhibitors: a review of recent patents (2012–2015). Expert Opin. Ther. Pat. 2017, 27, 91–99. 10.1080/13543776.2017.1238073. [DOI] [PubMed] [Google Scholar]
  6. Roskoski R. Jr.; 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. 10.1016/j.phrs.2017.12.021. [DOI] [PubMed] [Google Scholar]
  7. O’Leary C.; Xu W.; Pavlakis N.; Richard D.; O’Byrne K. Rearranged during transfection fusions in non-small cell lung cancer. Cancers 2019, 11, 620–625. 10.3390/cancers11050620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Li A. Y.; McCusker M. G.; Russo A.; Scilla K. A.; Gittens A.; Arensmeyer K.; Mehra R.; Adamo V.; Rolfo C. RET fusions in solid tumors. Cancer Treat. Rev. 2019, 81, 101911–101925. 10.1016/j.ctrv.2019.101911. [DOI] [PubMed] [Google Scholar]
  9. Song M. Progress in discovery of KIF5B-RET kinase inhibitors for the treatment of non-small-cell lung cancer. J. Med. Chem. 2015, 58, 3672–3681. 10.1021/jm501464c. [DOI] [PubMed] [Google Scholar]
  10. Subbiah V.; Hu M. I.; Gainor J. F.; Mansfield A. S.; Alonso G.; Taylor M. H.; Zhu V. W.; Garrido P.; Amatu A.; Doebele R. C.; Cassier P.; Keam B.; Schuler M. H.; Zhang H.; Clifford C.; Palmer M.; Green J.; Turner C. D.; Curigliano G. Clinical activity of the RET inhibitor pralsetinib (BLU-667) in patients with RET fusion-positive solid tumors. J. Clin. Oncol. 2021, 39, 467. 10.1200/JCO.2021.39.3_suppl.467.33434059 [DOI] [Google Scholar]
  11. Subbiah V.; Gainor J. F.; Rahal R.; Brubaker J. D.; Kim J. L.; Maynard M.; Hu W.; Cao Q.; Sheets M. P.; Wilson D.; Wilson K. J.; DiPietro L.; Fleming P.; Palmer M.; Hu M. I.; Wirth L.; Brose M. S.; Ou S.-H. I.; Taylor M.; Garralda E.; Miller S.; Wolf B.; Lengauer C.; Guzi T.; Evans E. K. Precision targeted therapy with BLU-667 for RET-driven cancers. Cancer Discovery 2018, 8, 836–849. 10.1158/2159-8290.CD-18-0338. [DOI] [PubMed] [Google Scholar]
  12. Markham A. Selpercatinib: first approval. Drugs 2020, 80, 1119–1124. 10.1007/s40265-020-01343-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Velcheti V.; Bauer T.; Subbiah V.; Cabanillas M.; Lakhani N.; Wirth L.; Oxnard G.; Shah M.; Sherman E.; Smith S.; Eary T.; Cruickshank S.; Tuch B.; Ebata K.; Nguyen M.; Corsi-Travali S.; Rothenberg S.; Drilon A. LOXO-292, a potent, highly selective RET inhibitor, MKI-resistant RET fusion-positive lung cancer patients with and without brain metastases. J. Thorac. Oncol. 2017, 12, S1778. 10.1016/j.jtho.2017.09.399. [DOI] [Google Scholar]
  14. Falchook G. S.; Ordóñez N. G.; Bastida C. C.; Stephens P. J.; Miller V. A.; Gaido L.; Jackson T.; Karp D. D. Effect of the RET inhibitor vandetanib in a patient with RET fusion-positive metastatic non-small-cell lung cancer. J. Clin. Oncol. 2016, 34, e141–e144. 10.1200/JCO.2013.50.5016. [DOI] [PubMed] [Google Scholar]
  15. Bronte G.; Ulivi P.; Verlicchi A.; Cravero P.; Delmonte A.; Crinò L. Targeting RET-rearranged non-small-cell lung cancer: future prospects. Lung Cancer: Targets Ther. 2019, 10, 27–36. 10.2147/LCTT.S192830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Mathison C. J. N.; Chianelli D.; Rucker P. V.; Nelson J.; Roland J.; Huang Z.; Yang Y.; Jiang J.; Xie Y. F.; Epple R.; Bursulaya B.; Lee C.; Gao M.-Y.; Shaffer J.; Briones S.; Sarkisova Y.; Galkin A.; Li L.; Li N.; Li C.; Hua S.; Kasibhatla S.; Kinyamu-Akunda J.; Kikkawa R.; Molteni V.; Tellew J. E. Efficacy and tolerability of pyrazolo[1,5-a]pyrimidine RET kinase inhibitors for the treatment of lung adenocarcinoma. ACS Med. Chem. Lett. 2020, 11, 558–565. 10.1021/acsmedchemlett.0c00015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Graczyk P. P. Gini coefficient: a new way to express selectivity of kinase inhibitors against a family of kinases. J. Med. Chem. 2007, 50, 5773–5779. 10.1021/jm070562u. [DOI] [PubMed] [Google Scholar]
  18. Warmuth M.; Kim S.; Gu X.; Xia G.; Adrian F. Ba/F3 cells and their use in kinase drug discovery. Curr. Opin. Oncol. 2007, 19, 55–60. 10.1097/CCO.0b013e328011a25f. [DOI] [PubMed] [Google Scholar]
  19. Widler L.; Green J.; Missbach M.; Susa M.; Altmann E. 7-Alkyl- and 7-cycloalkyl-5-aryl-pyrrolo[2,3-d]pyrimidines-potent inhibitors of the tyrosine kinase c-Src. Bioorg. Med. Chem. Lett. 2001, 11, 849. 10.1016/S0960-894X(01)00079-8. [DOI] [PubMed] [Google Scholar]
  20. Miles A. A.; Miles E. M. Vascular reactions to histamine, histamine-liberator and leukotaxine in the skin of guinea-pigs. J. Physiol. 1952, 118, 228–257. 10.1113/jphysiol.1952.sp004789. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

ml1c00450_si_001.pdf (1.5MB, pdf)

Articles from ACS Medicinal Chemistry Letters are provided here courtesy of American Chemical Society

RESOURCES