Skip to main content
MedComm logoLink to MedComm
. 2021 Jun 17;2(3):341–350. doi: 10.1002/mco2.42

Two novel strategies to overcome the resistance to ALK tyrosine kinase inhibitor drugs: Macrocyclic inhibitors and proteolysis‐targeting chimeras

Xiaoling Song 1,#,, Hui Zhong 1,2,#, Xiaojuan Qu 1,#, Linsong Yang 2,, Biao Jiang 1,3,
PMCID: PMC8554663  PMID: 34766150

Abstract

Lung cancer is the most malignant tumor in the worldwide. About 3%‐5% non‐small cell lung cancer (NSCLC) patients carry anaplastic lymphoma kinase (ALK) gene fusions and receive great benefits from ALK‐targeted therapy. However, drug resistance inevitably occurs even with the most potent inhibitor drug lorlatinib. About half of the resistance are caused by alteration in ALK proteins for earlier ALK TKI drugs and near one‐third of loratinib resistant cases are caused by compound mutations without current effective treatment strategy in clinic. Novel strategies are in great need to overcome drug resistance. Lately, two novel strategies have been developed and attracted great attentions for their potentials to overcome drug resistance problems: (1) developed small compact macrocyclic ALK kinase inhibitors and (2) developed ALK targeted proteolysis‐targeting chimera (PROTAC) drugs. The macrocyclic molecules are small and compact in size, brain barrier permeable, and highly potent against lorlatinib‐resistant compound mutations. Developed ALK targeted PROTAC molecules could degrade oncogenic ALK driver proteins. Some showed superiority in killing ALK positive cancer cells and inhibiting the growth of cells expressing G1202R resistant ALK proteins comparing to inhibitor drugs. The update on these two treatment strategies was reviewed.

Keywords: ALK PROTACs, ALK‐TKIs, EML4‐ALK, macrocyclic inhibitors, non‐small cell lung cancer, resistance mechanisms


Acquired resistance mutations including G1202R and L1196M, alone or in compound form, in ALK proteins lead to drug resistance to ALK‐targeted therapy. Two novel strategies showed great potential in overcoming such resistance: (1) More effective inhibition by small and compact macrocyclic inhibitors; (2) Degradation of oncogenic ALK proteins including those with resistant mutations by ALK targeted proteolysis‐targeting chimeras.

graphic file with name MCO2-2-341-g002.jpg


Abbreviations

ALK

anaplastic lymphoma kinase

CDX

cell line‐derived xenograft

CRBN

cereblon

FDA

food and drug administration

IC50

half maximal inhibitory concentration

IND

investigational new drug

NSCLC

non‐small cell lung cancer

NTRK

neurotrophin receptor kinase

PDX

patient‐derived xenograft

PROTAC

proteolysis targeting chimera

ROS1

ROS proto‐oncogene 1

TBK1

TANK binding kinase 1

TKIs

tyrosine kinase inhibitors

VHL

von Hippel‐Lindau

1. INTRODUCTION

Lung cancer is the most common malignant cancer in the world, and it is also the leading cause of cancer‐related death. The latest data published by the national cancer center in 2020 showed that in 2015 lung cancer accounted for 20% of new cases in China and led to 27% of cancer deaths, ranking first in both morbidity and mortality among malignant tumors in China. 1 A latest report released that lung cancer has the highest fatality rate of malignant tumor in the United States, and the 5‐year lung Cancer survival rate is 18.4%. 2 Treating ALK mutant cancer patients with targeted therapy greatly reduces toxic adverse effect and improves patients’ quality of life comparing to traditional chemotherapy. 3

At present, five different ALK‐Tyrosine kinase inhibitors (ALK‐TKIs) have been approved by food and drug administration (FDA) to treat ALK‐positive non‐small cell lung cancer (NSCLC) patients (Figure 1). These drugs include the first‐generation ALK‐TKIs crizotinib, the second‐generation ALK‐TKIs ceritinib, alectinib and brigatinib, and the third generation ALK‐TKIs lorlatinib. Although later generation ALK TKI inhibitors have better kinase selectivity and enhanced ability to overcome drug resistance, it has been proved that drug resistance inevitably occurs within a certain period of time after the initial drug treatment. It is urgent to develop novel and effective treatment strategies to overcome drug resistance. This article was focused on the update on two novel strategies to overcome drug resistance.

FIGURE 1.

FIGURE 1

FDA approved ALK inhibitor drugs and novel macrocyclic ALK inhibitors. The structure and molecular weight (MW) of five FDA approved ALK inhibitor drugs (1st to 3rd generations) and newly developed macrocyclic ALK kinase inhibitors were presented. When there are no resistant mutations in ALK tyrosine kinase domain, ALK kinase activities can be effectively inhibited by all currently approved ALK tyrosine kinase inhibitor drugs (TKIs). The 2nd generation ALK TKI drugs can effectively inhibit the activity of gate keeper mutation L1196M, but they are not so effective on solvent front mutation G1202R and many compound mutations. The 3rd generation ALK TKI drug lorlatinib is a small and compact macrocyclic inhibitor. It can effectively inhibit the activity of G1202R mutation but not for those compound mutations. Latest developed macrocyclic ALK TKI drugs are even smaller and more compact than lorlatinib. They are highly potent against G1202R mutation and many lorlatinib resistant compound mutations. They were proposed to be the next generation ALK TKI drugs (references 14, 19)

Abbreviations: MW, molecular weight.

2. RESISTANCE MECHANISMS OF ALK KINASE INHIBITORS

About half of the acquired resistance mechanisms to ALK TKIs are ALK kinase protein‐dependent. The acquired resistance mechanisms of first‐generation and second‐ generation ALK‐TKIs include: secondary mutations in ALK kinase domain, amplification of ALK fusion gene, epithelial mesenchymal transformation, activation of ERK, SRC, IGF‐1R and other bypass signaling pathway, and transformation into small‐cell lung cancer. 4 Firstly, within above resistant mechanisms, ALK‐dependent resistant mechanisms including ALK gene amplification and acquired secondary mutations in ALK kinase domains make up about half of the incidence for resistance to crizotinib (1st generation ALK TKI). 5 , 6 These mutations include L1196M, L1152R, and G1202R, etc. Because drug‐resistant mutations transform ALK protein into an active conformation and introduce new steric hindrance between ALK kinase and ATP, they increase the binding affinity between the kinase and ATP resulting in drug resistance. Secondly, secondary mutations in the ALK kinase domain are more common in patients with acquired resistance to later generation ALK inhibitor drugs, reaching about 50‐70% of resistant cases. 7 The mutation spectrum was also different from that of crizotinib. Solvent front mutation G1202R is the most dominant acquired mutation among mutations that confer the resistance to second generations ALK TKI drugs. It makes up about 42% of resistant cases for patients on brigatinib treatment, 21% for patients treated with ceritinib, and 29% for patients on alectinib. 7 These results indicate that acquired aberrant ALK protein alterations are the major source of drug resistance. Developing strategies to target these aberrant ALK alterations are critical to overcome such drug resistance.

The third generation ALK TKI drug lorlatinib is a potent, brain permeable, small macrocyclic molecule (Figure 1). It can inhibit most of the drug‐resistant mutations of crizotinib and second‐generation ALK‐TKIs especially G1202R mutation. 8 Clinical data showed that lorlatinib led to an objective response rate of 45% and durable responses in ALK‐positive patients who already developed resistance to previous generation of ALK TKI drugs. 9 , 10 Lorlatinib was approved by FDA in 2018 for second‐line treatment of advanced ALK‐positive NSCLC patients. A full user guide to lorlatinib has been recently reviewed. 11

However, the development of drug resistance to lorlatinib is also inevitable in clinic. Studies showed that about 35% of the acquired resistance mechanisms to lorlatinib are compound mutations at two or three sites, such as I1171N + L1198F, G1202R + L1204V + G1269A (Table 1). 12 In addition, other complex mutations conferring drug resistance to lorlatinib were also reported. 13 The sensitivities of 14 different ALK compound mutations that conferring resistance to lorlatinib have been studied using current FAD approved ALK inhibitor drugs and were summarized in Table 1. Results showed that some of the compound mutations (6/14) were sensitive to 1st or 2nd generation of ALK TKI inhibitors, but about half of compound mutations were resistant to all current approved ALK TKI drugs. Such resistant mutations include ALK G1202R+L1196M, G1202R+F1174C, and G1202R+ F1174L etc. Therefore, novel treatment strategies are in great need to be developed to solve these treatment difficulties in clinic.

TABLE 1.

Reported mutations resistant to lorlatinib and their sensitivities to other ALK kinase inhibitors

graphic file with name MCO2-2-341-g003.jpg lorlatinib 3rd crizotinib 1st ceritinib 2nd alectinib 2nd brigatinib 2nd repotrectinib TPX‐0131 Reference
Lorlatinib sensitive variants
WT 0.7‐1.9 22‐74.8 2.1‐10 10‐18.9 2.4‐11.8 17.8 <5 13,15,19
G1202R 69‐80 230‐560 120‐388 507‐640 56‐399 20.5 <2 13,15,19
L1196M 18 713 6 131 13 50 13
I1171N 81 220 34‐36 600 27‐28 13
Lorlatinib resistant variants
C1156Y + L1198F 67‐140 0.6‐22 1123‐1493 24‐776 12‐118 1.1 12,13,15,35
L1196M + L1198F 1169 350.7 1794 1249 341 34.8 <2 15,19
G1202R + L1198F 132‐370 47 477‐1100 >1000 290‐2040 0.2 <2 12,13,15,19
G1202R + C1156Y >1000 >1000 >1000 >1000 >1000 <2 13,19
G1202R + G1269A 700 390 120 960 75 12,13
G1202R + L1196M >1000 370‐800 690‐1260 >1000 320‐1100 <2 12,13,19
G1202R + F1174C 280‐300 320‐370 280‐420 700‐1400 180‐220 13
G1202R + F1174L 110‐190 200‐370 260‐420 560‐2300 160‐190 13
G1202R + L1204V + G1269A 673 634 345 6740 176 12,13
I1171N + L1198F 240 19 660 750 43 12,13
I1171N + L1196M 360 460 21 460 31 12,13
I1171N + L1256F 6000 560 400 73 42 12,13
I1171N + G1269A 470 670 14 1100 7.1 12,13
I1171N + L1198H 670 160 540 1600 140 13
I1171N + F1174I 320 350 140 1300 110 13
I1171N + F1174L 220 440 160 1600 100 13
I1171N + D1203N Resistant 12
E1210K + D1203N + G1269A Resistant 12

Half maximal inhibitory concentration (IC50s) in Ba/F3 cells for different ALK TKI inhibitors were summarized. Lorlatinib resistant mutations reported in clinics or identified in lab were labeled in bold color.

– : means data are not available.

3. NOVEL TREATMENT STRATEGIES

Lots of efforts have been put to develop newer treatment strategies to fight against drug resistance. Two latest developed treatment strategies are as follows: (1) developing smaller highly compact macrocyclic ALK‐TKIs and (2) developing ALK‐targeting proteolysis targeting chimeria (PROTAC) molecules. The updates on these treatment strategies were reviewed in the following.

3.1. Develop smaller and more compact macrocyclic ALK‐TKIs

Most FDA approved ALK‐TKIs are ATP‐competitive type I TKIs, which are large in size and have some motifs close to or across the hydrophobic posterior capsule, which makes them susceptible to various drug‐resistant mutations 14 (Figure 1). Further development of similar inhibitors is difficult to block the activities of such evolutionary mutations. Based on this idea, new inhibitors were designed smaller and more compact to avoid previous resistance problems. For example, the third generation lorlatinib was designed as a small macrocyclic molecule (Figure 1). It is smaller than all earlier generation of FDA approved ALK inhibitor drugs; and it is more potent in inhibiting the activities of mutant ALK kinase proteins than all previous approved ALK TKI drugs. Therefore, newer ALK TKIs were designed even smaller and more compact to evade the intrinsic resistance to previous reported mutations. Two new small and compact macrocyclic molecules were designed by Turning Point Therapeutics, Inc. lately, which are repotrectinib and TPX‐0131. The molecular weight of these two molecules was smaller than all current FDA approved ALK TKI drugs (Figure 1). Because these two molecules are very potent against many lorlatinib resistant compound mutations, they are proposed to be the 4th generation ALK TKI drugs.

Repotrectinib (TPX‐0005) is one of the two small compact macrocyclic molecules (Figure 1). It is smaller than lorlatinib with a molecular weight about 355 Kilo‐Dalton and having a high activity in central nervous system. 14 It can accurately anchor molecules on adenine binding sites on ALK protein, target kinase resistant active conformations, effectively avoid mutations especially G1202R precursor solvent mutation caused by space interference. 14 Data showed that repotrectinib could effectively inhibit previously reported tough resistant mutations including G1202R and L1196M, and it is also effective in some lorlatinib resistant ALK compound mutations such as L1198F+C1156Y and G1202R+L1198F (Table 1). 15 Additionally, It effectively induced G1202R resistant tumor regression in clinical relevant mouse models including both cell line derived xenograft (CDX) and patient‐derived (PDX) tumor models. 16 Furthermore, repotrectinib can also sensitize H2228 cells that are intrinsically resistant to ALK TKI by inhibiting SRC activities. 17 , 18 Lastly, it is a multi‐function inhibitor that can inhibit the activity of NTRK (neurotrophin receptor kinase), ROS1 (ROS proto‐oncogene 1), and ALK. 14 In general, repotrectinib is a very promising treatment to overcome various mechanisms of ALK resistance, including secondary mutations and bypass signal activation. The efficacy of repotrectinib in patients with advanced solid tumor harboring ALK gene rearrangement is currently being evaluated in a phase I/II clinical trial (www.clinicaltrials.gov: NCT03093116).

TPX‐0131 is another small compact macrocyclic ALK inhibitor developed by Turning Point Therapeutics. It was firstly reported in 2020 AACR annual meeting. Released data showed that TPX‐0131 is not only sensitive to most single resistant mutations including solvent front G1202R resistant mutation, but also is very effective for many compound mutations such as G1202R+L1196M, G1202R+L1198F, G1202R+C1156F, and L1196M+ L1198F. 19 Currently, this compound is in a process of investigational new drug enabling study. With its strong activity in overcoming many lorlatinib resistant compound mutations, TPX‐0131 has the great potential to be the candidate of the next‐generation ALK inhibitor drug.

3.2. Proteolysis targeting chimeras of ALK

PROTAC technology is a technology utilizing cell endogenous proteasome degradation system to degrade protein of interest. The compounds designed based on PROTAC technology are hetero‐functional molecules (Figure 2A), which include three parts: (1) ligand for targeted protein, (2) ligand to recruit an E3 ubiquitin ligase, and (3) a linker for connection of the two ligands. The most frequently used E3 ubiquitin ligase ligands include cereblon (CRBN) ligands and VHL (von‐Hippel‐Lindau) ligands. CRBN ligands include thalidomide, lenalidomide, and pomalidomide. Both CRBN protein and VHL protein are substrate recognition subunits of two widely expressed and functionally important cullin ring E3 ubiquitin ligases complexes. By connecting the target protein and the E3 ligase together, PROTAC molecules recruit the target protein to the proximity of the E3 ubiquitin ligase, which leads to ubiquitination of the target protein, and subsequently degradation of target protein by proteasome. Because aberrant expression of many biologically important proteins are highly involved in the progress of human diseases, targeted degradation of these proteins have been considered as a critical way to treat such diseases. Because of its great potential to degrade previously undrugable protein and overcome drug resistance in precision medicine, it has attracted a lot of attentions recently to be used in the development of new drugs for human diseases. 20 At present, PROTAC technique has been successfully applied to selective degradation of various protein targets, such as BCR‐ABL, 21 TBK1, 22 epidermal growth factor receptor (EGFR), 23 HER2, 23 and c‐MET. 23

FIGURE 2.

FIGURE 2

ALK‐targeted proteolysis targeting chimeras. A, The working principles of ALK PROTAC molecules. An ALK‐PROTAC molecule includes three parts: (1) an ALK protein binder, (2) an E3 ubiquitin ligase binder, and (3) a connection linker. When an ALK PROTAC molecule enters cells, it binds to ALK protein and in the meanwhile, it recruits the E3 ubiquitin ligase to the proximity of ALK protein. Followed by labeled with ubiquitin, ALK protein was degraded by proteasome. The degradation of ALK protein by ALK PROTACs will lead to the death of ALK positive cancer cells, including those baring TKI resistant mutations, provide a promising strategy to treat cancer patients and overcome drug resistance in clinic in the future. B, Six published ALK PROTAC molecules. Three different ALK binders were used: TAE684, ceritinib, and brigatinib (as shown in left rectangular part); and two different E3 binders were used: pomalidomide and VHL ligand (as shown in brown colored right part)

Because oncogenic ALK protein is the driver force of ALK mutant positive cancer, and about half of the resistant mechanisms are related to either ALK mutations or ALK amplification. Targeted degradation of ALK protein theoretically can improve drug treatment efficacy and evade drug resistance. In addition, although ALK is expressed in many types of cancer, it is barely expressed in adult animals. 24 Get rid of ALK protein in patients will not affect normal function of human health and only affect ALK dependent tumors. Furthermore, mouse models have showed that knock out ALK only had a slight impact on the behavioral phenotype of mice without affecting their survival. 25 Therefore, pharmacological degradation of ALK by PROTAC technology can effectively kill cancer cells, reduce the off‐target side toxicity effect and provide a new and potential treatment strategy for ALK‐positive cancer patients.

Currently, there are six different ALK PROTACs having been developed (Figure 2 and Table 2). All published ALK PROTACs were designed based on the second generation ALK‐TKIs. Based on the ALK binder, these ALK targeting PROTACs can be categorized into three classes: (1) ceritinib‐based ALK PROTAC 26 , 27 , 28 , 29 , (2) TAE684‐based ALK PROTAC 27 , and (3) brigtinib‐based ALK PROTAC. 30

TABLE 2.

Bio‐activities of reported ALK PROTAC molecules

ALK PROTACs TL13‐12 TL13‐112 MS4077 MS4078 TD‐004 SIAIS117
PROTAC elements
ALK binders TAE684 Ceritinib Ceritinib Ceritinib Ceritinib Brigatinib
E3 type CRBN CRBN CRBN CRBN VHL VHL
E3 binders Pomalidomide Pomalidomide Pomalidomide Pomalidomide VHL ligand VHL ligand
In vitro DC50 (nM)
EML4‐ALK (NSCLC cell line) 10 (H3122) 10 (H3122) 34 (H2228) 59 (H2228) 300 (H3122) ∼100 (H2228)
NPM‐ALK (ALCL cell line) 180 (Karpas299) 40 (Karpas299) 3 (SUDHL1) 11 (SUDHL1) 300 (SUDHL1) 7 (SR)
ALK F1174L (NB cell line) 50 (Kelly) 50 (Kelly)
EML4‐ALKG1202R 189
In vitro IC50 (nM)
EML4‐ALK 25 50 30 45 180 46
NPM‐ALK 5 5‐15 46 33 58 1.7
EML4‐ALKG1202R 166
Activity (comparing to inhibitor) Slightly stronger Slightly stronger Weaker Weaker Weaker Slightly stronger
Effect on bypass pathway Inhibit SCLC
Degrade Aurora A Yes Yes No
In vivo activities
Serum levels >500 nM (IP, 50 mg/kg)
Anti‐tumor activity Inhibit H3122 CDX growth (IP)
Reference 27 27 28 28 26 30

Abbreviations: CDX, cell line‐derived xenograft; DC50, half maximal degradation concentration; IC50, half maximal inhibitory concentration; IP, intraperitoneal drug administration.

– : No data available.

Ceritinib‐based ALK PROTACs exhibited a moderate ALK protein degradation ability. In these studies, ligands for two different E3 ubiquitin ligases, CRBN (cereblon), and VHL (von Hippel‐Lindau), were linked to ceritinib to generate ALK PROTAC molecules (Figure 2B and Table 2). ALK PROTACs using polidomide as CRBN E3 ligase ligand exhibited stronger degradation ability comparing to those using VHL ligand (Figure 2B and Table 2). Kang et al 26 used VHL as E3 ligands to generate ALK degrader TD‐004. At the concentration of 300 nM, TD‐004 could degrade NPM‐ALK fusion protein in ALCL cell line SU‐DHL‐1 cells and degrade EML4‐ALK protein (Version 1) in non‐small cell lung cancer cell line H3122. Powell et al 27 developed ALK degraders with CRBN ligand pomalidomide. ALK degrader TL13‐112 degraded NPM‐ALK protein at the concentration of 40 nM in ALCL cell line Karpas299 and degraded EM4‐ALK at the concentration of 10 nM in H3122 cell lines. Zhang et al also utilized CRBN ligand pomalidomide to design their ALK degrader, which could degrade NPM‐ALK (SU‐DHL‐1) and EML4‐ALK (version3, in non‐small cell lung cancer cell line H2228) at the concentration of 3‐60 nM. 28 Besides using ceritinib in ALK PROTAC, Powell et al also used TAE684 and pomalidomide to generate ALK PROTAC molecules, but the degradation capability of TAE684 PROTAC was not better than that of ceritinib‐pomalidomide PROTAC. 27

However, there was no dramatic improvement for ceritinib‐based ALK degraders comparing to ceritinib in terms of anti‐proliferation effect against ALK mutant lung cancer cells (Figure 2B and Table 2). In NSCLC and ALCL cell lines, the anti‐proliferation activities of the degraders and parental kinase inhibitors are roughly the same. VHL‐based ceritinib ALK degrader TD‐004 could inhibit the proliferation of SU‐DHL‐1 but with the ability reduced about 2‐20 fold (Table 2). The ceritinib‐pomalidomid ALK degraders MS4077 and MS4078 designed by Zhang exhibited an anti‐proliferation ability (IC50s) of 33 nM and 46 nM in SU‐DHL‐1 cells, and which was about 2‐3 fold weaker than that of ceritinib (IC50: 15 nM). Ceritinib‐ and pomalidomide‐based degraders TL13‐12 and TL13‐112 designed by Powell could inhibit the proliferation of H3122, Karpas299, and SU‐DHL‐1 cells, but the IC50 was about similar to or weaker than ceritinib. 27 They also tried to study the effect on the growth inhibition of Ba/F3 cells expressing EML4‐ALK resistant mutations. However, no ALK degrader showed better growth inhibition effect than ceritinib did. 27

Brigatinib‐based ALK degrader displayed a good potential to overcome drug resistance (Figure 2B and Table 2). Currently, only one brigatinib‐based ALK degrader, SIAIS117, was reported, which used ligand of VHL as the E3 ubiquitinase ligand. 30 It could not only effectively degrade NPM‐ALK proteins in ALCL cell line SR and EML4‐ALK proteins in NSCLC cell line H2228, but also could degrade exogenously expressed ALK fusion proteins with G1202R resistant mutations in 293T cells. Additionally, it also exhibited a better growth inhibition effect in several ALK mutation positive cancer cells including ALCL cell line SR, non‐small cell lung cancer cell line H2228, and ALK TKI resistant cells that exogenously expressed G1202R mutant EML4‐ALK proteins. Furthermore, SIAIS117 displayed an advantage in inhibiting the growth of two different small cell lung cancer cell lines at a physiological achievable concentration for most approved drugs, but brigatinib did not have such function at the same concentration. Cell type transformation from lung adenocarcinoma cancer cells to small cell lung cancer cells is one resistance mechanism to crizotinib. 31 , 32 , 33 Thus, SIAIS117 is potentially effective in overcoming such resistance by possessing a more potent growth inhibition effect on small cell lung cancer cells.

Two different data supported that ALK PROTAC molecules could also have good bioavailabilities in vivo (Table 2). When administered ALK PROTAC molecules MS4078 in mice via intraperitoneal injection, it was pharmacokinetically stable. It could achieve a relative stable serum exposure level more than 500 nM within a 12‐hour observing window 28 (Table 2). ALK degrader TD‐004 could inhibit the growth of H3122 xenograft tumor in mouse model after intraperitoneal administration 26 (Table 2). A big obstacle for PROTAC molecules to become successful drug candidates in clinic is that most generated PROTAC molecules barely had good exposure in vivo. However, the data from MS4078 and TD‐004 indicate that current PROTAC developing technologies have broken the obstacle and are able to generate ALK PROTAC candidates with good pharmacokinetics in vivo. This will greatly expedite the speed of developing a successful ALK PROTAC drug to be used in clinic in the future.

Different ALK degraders have different side effects on cell activities. Ceritinib‐based ALK degraders and TAE684‐based ALK degrader showed some non‐specific degradation of cellular proteins such as Aurora A. 27 However, brigatinib‐based ALK PROTAC SIAIS117 did not degrade Aurora A. It indicates a different ALK binder may bring in differential specificity to the final cellular action. Because brigatinib also provides patients with a longer progression‐free survival rate comparing to other second generation ALK inhibitor drugs, brigatinib‐based ALK degraders may be more beneficial than ceritinib‐based ALK degraders in treating cancer patients in the future.

4. CONCLUSIONS

In summary, two different strategies have been developed to overcome current drug resistance issues. Designing smaller and more compact macrocyclic ALK inhibitors seems to be a very good way to develop effective ALK targeted drug candidates. Small and compact macrocyclic can bind completely inside the ATP pocket even in the presence of solvent front mutation and gatekeeper mutation. This empower them the ability to evade the resistance to previous generation ALK TKI drugs. ALK TKI drug repotrectinib (TPX‐0005) showed great potency in overcoming ALK drug resistant mutation such as L1196M and G1202R mutations, and it is currently being tested in phase I clinical trials to treat ALK TKI resistant patients. Another small compact macrocyclic molecule TPX‐0131 has also been showed with very potent activities against most of compound mutations conferring resistance to lorlatinib and currently is in an IND‐enabling study. These macrocyclic inhibitors were proposed to be the next‐generation ALK TKI drugs14, 19 and they will provide great opportunities to solve some current resistance problems in ALK‐positive cancer patients in the future.

The strategy of treating ALK positive cancer with ALK‐specific PROTAC molecules provides another good opportunity to overcome drug resistance problems. In order to achieve sustained ALK inhibition, inhibitors must bind to the ALK protein for a long period of time to reach binding saturation. During this process off‐target effects and kinase mutations may occur, resulting in drug resistance, becoming a major clinical obstacle. PROTAC technology provides an ideal way to degrade ALK driver proteins to kill ALK mutant cancer cells and evade drug resistance. Currently, the PROTAC technology is still in development. Most ALK degraders have not shown in vivo anti‐tumor efficacy yet. Even brigatinib‐based ALK degrader is very promising for its ability to overcome ALK drug resistance in vitro, the pharmokinetics and dynamics of this compound remain to be studied. Results from MS4078 and TD004 showed that ALK PORTACs are able to have good exposure in vivo. It indicates that current technologies are able to break the obstacle of poor exposure for PROTAC molecules in vivo, and ALK PROTAC molecules have great opportunities to become real therapeutic drugs in clinic in the future. The latest news from ASCO2020 showed that the pioneer PROTAC drug ARV110 developed by Arvinas could induce tumor regression in cancer patients in clinical trials (NCT03888612). 34 It proved that PROTAC molecules could have good biological effect in clinic in real patients. With the fast developing PORTAC field, more and more PROTAC drugs are anticipated to be shown in clinical trials, and the time for a successful ALK PROTAC drug to be used in clinic is about only 5‐10 years away.

CONFLICT OF INTEREST

The authors declare no conflict of interest for the manuscript.

AUTHOR CONTRIBUTIONS

Xiaoling Song, Hui Zhong, and Xiaojuan Qu wrote the manuscript; Xiaoling Song designed and revised the manuscript; Xiaoling Song, Biao Jiang, and Linsong Yang supervised the work.

ETHICS STATEMENT

Not applicable.

ACKNOWLEDGMENT

Xiaoling Song received financial support from National Natural Science Foundation of China (Grand No.: 82072592 and 81602602) and Science and Technology Commission of Shanghai Municipality (Grant No.: 20S11903100); Biao Jiang received financial support from ShanghaiTech University.

Song X, Zhong H, Qu X, Yang L, Jiang B. Two novel strategies to overcome the resistance to ALK tyrosine kinase inhibitor drugs: Macrocyclic inhibitors and proteolysis‐targeting chimeras. MedComm. 2021;2:341–350. 10.1002/mco2.42

Contributor Information

Xiaoling Song, Email: songxl@shanghaitech.edu.cn.

Linsong Yang, Email: linsongyang@cczu.edu.cn.

Biao Jiang, Email: jiangbiao@shanghaitech.edu.cn.

REFERENCES

  • 1. Gao S, Li N, Wang S, et al. Lung cancer in People's Republic of China. J Thorac Oncol. 2020;15(10):1567‐1576. [DOI] [PubMed] [Google Scholar]
  • 2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA‐Cancer J Clin. 2019;69(1):7‐34. [DOI] [PubMed] [Google Scholar]
  • 3. Solomon BJ, Mok T, Kim DW, et al. First‐line crizotinib versus chemotherapy in ALK‐ positive lung cancer. N Engl J Med. 2014;371(23):2167‐2177. [DOI] [PubMed] [Google Scholar]
  • 4. Qian M, Zhu B, Wang X, Liebman M. Drug resistance in ALK‐positive non‐small cell lung cancer patients. Semin Cell Dev Biol. 2017;64:150‐157. [DOI] [PubMed] [Google Scholar]
  • 5. Doebele RC, Pilling AB, Aisner DL, et al. Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non‐small cell lung cancer. Clin Cancer Res. 2012;18(5):1472‐1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Katayama R, Shaw AT, Khan TM, et al. Mechanisms of acquired crizotinib resistance in ALK‐rearranged lung cancers. Sci Transl Med. 2012;4(120):120ra17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Redaelli S, Ceccon M, Zappa M, et al. Lorlatinib treatment elicits multiple on‐ and off‐target mechanisms of resistance in ALK‐driven cancer. Cancer Res. 2018;78(24):6866‐6880. [DOI] [PubMed] [Google Scholar]
  • 8. Zou HY, Friboulet L, Kodack DP, et al. PF‐06463922, an ALK/ROS1 inhibitor, overcomes resistance to first and second generation ALK inhibitors in preclinical models. Cancer Cell. 2015;28(1):70‐81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Shaw AT, Felip E, Bauer TM, et al. Lorlatinib in non‐small‐cell lung cancer with ALK or ROS1 rearrangement: an international, multicentre, open‐label, single‐arm first‐in‐man phase 1 trial. Lancet Oncol. 2017;18(12):1590‐1599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Solomon BJ, Besse B, Bauer TM, et al. Lorlatinib in patients with ALK‐positive non‐small‐cell lung cancer: results from a global phase 2 study. Lancet Oncol. 2018;19(12):1654‐1667. [DOI] [PubMed] [Google Scholar]
  • 11. Nagasaka M, Ge Y, Sukari A, Kukreja G, Ou SI. A user's guide to lorlatinib. Crit Rev Oncol Hematol. 2020;151:102969. [DOI] [PubMed] [Google Scholar]
  • 12. Yoda S, Lin JJ, Lawrence MS, et al. Sequential ALK inhibitors can select for lorlatinib‐resistant compound ALK mutations in ALK‐positive lung cancer. Cancer Discov. 2018;8(6):714‐729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Okada K, Araki M, Sakashita T, et al. Prediction of ALK mutations mediating ALK‐TKIs resistance and drug re‐purposing to overcome the resistance. EBioMedicine. 2019;41:105‐119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Drilon A, Ou SI, Cho BC, et al. Repotrectinib (TPX‐0005) is a next‐generation ROS1/TRK/ALK inhibitor that potently inhibits ROS1/TRK/ALK solvent‐ front mutations. Cancer Discov. 2018;8(10):1227‐1236. [DOI] [PubMed] [Google Scholar]
  • 15. Zhai D, Deng W, Rogers E, et al. Abstract B186: TPX‐0005, a polypharmacology inhibitor, overcomes ALK treatment resistance from acquired mutations, bypass signaling, and EMT. Mol Cancer Ther. 2018;17(1):B186. [Google Scholar]
  • 16. Satoshi Yoda, Leila Dardaei, Kylie Prutisto‐Chang, Cui J, Shaw AT, Hata AN. Abstract 4795: potency of a new ALK/ROS1 inhibitor TPX‐0005 to ALK G1202R mutation and ROS1 G2032R mutation. Cancer Res. 2018;78(13):4795. [Google Scholar]
  • 17. Cui JJ, Zhai D, Deng W, et al. P3.02a‐009 TPX‐0005: a multi‐faceted approach to overcoming clinical resistances from current ALK or ROS1 inhibitor treatment in lung cancer. J Thorac Oncol. 2017;12(1):S1164‐S1165. [Google Scholar]
  • 18. Zhai D, Deng W, Huang Z, Rogers E, Cui JJ. Abstract 2132: the novel, ratio‐nally‐designed, ALK/SRC inhibitor TPX‐0005 overcomes multiple acquired resistance mechanisms to current ALK inhibitors. Cancer Res. 2016;76(14):2132. [Google Scholar]
  • 19. Cui JJ, Rogers E, Zhai D, et al. TPX‐0131: a next generation macrocyclic ALK inhibitor that overcomes ALK resistant mutations refractory to currently approved ALK inhibitors. Cancer Res. 2020;80(16 Suppl): 5226. [Google Scholar]
  • 20. Lai AC, Crews CM. Induced protein degradation: an emerging drug discovery paradigm. Nat Rev Drug Discov. 2017;16(2):101‐114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Lai AC, Toure M, Hellerschmied D, et al. Modular PROTAC design for the degradation of oncogenic BCR‐ABL. Angew Chem Int Ed Engl. 2016;55(2):807‐810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Crew AP, Raina K, Dong H, et al. Identification and characterization of von hippel‐lindau‐recruiting proteolysis targeting chimeras (PROTACs) of TANK‐binding kinase 1. J Med Chem. 2018;61(2):583‐598. [DOI] [PubMed] [Google Scholar]
  • 23. Burslem GM, Smith BE, Lai AC, et al. The advantages of targeted protein degradation over inhibition: an RTK case study. Cell Chem Biol. 2018;25(1):67‐77 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Kargbo RB. PROTACs and targeted protein degradation for treating ALK‐mediated cancers. ACS Med Chem Lett. 2019;10(8):1102‐1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Weiss JB, Xue C, Benice T, Xue L, Morris SW, Raber J. Anaplastic lymphoma kinase and leukocyte tyrosine kinase: functions and genetic interactions in learning, memory and adult neurogenesis. Pharmacol Biochem Be. 2012;100(3):566‐574. [DOI] [PubMed] [Google Scholar]
  • 26. Kang CH, Lee DH, Lee CO, Du Ha J, Park CH, Hwang JY. Induced protein degradation of anaplastic lymphoma kinase (ALK) by proteolysis targeting chimera (PROTAC). Biochem Bioph Res Co. 2018;505(2):542‐547. [DOI] [PubMed] [Google Scholar]
  • 27. Powell CE, Gao Y, Tan L, et al. Chemically induced degradation of anaplastic lymphoma kinase (ALK). J Med Chem. 2018;61(9):4249‐4255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Zhang C, Han XR, Yang X, et al. Proteolysis targeting chimeras (PROTACs) of anaplastic lymphoma kinase (ALK). Eur J Med Chem. 2018;151:304‐314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Wang Y, Han L, Liu F, et al. Targeted degradation of anaplastic lymphoma kinase by gold nanoparticle‐based multi‐headed proteolysis targeting chimeras. Colloids Surf B. 2020;188:110795. [DOI] [PubMed] [Google Scholar]
  • 30. Sun N, Ren C, Kong Y, et al. Development of a brigatinib degrader (SIAIS117) as a potential treatment for ALK positive cancer resistance. Eur J Med Chem. 2020;193:112190. [DOI] [PubMed] [Google Scholar]
  • 31. Cha YJ, Cho BC, Kim HR, Lee HJ, Shim HS. A case of ALK‐rearranged adenocarcinoma with small cell carcinoma‐like transformation and resistance to crizotinib. J J Thorac Oncol. 2016;11(5):e55‐e58. [DOI] [PubMed] [Google Scholar]
  • 32. Fujita S, Masago K, Katakami N, Yatabe Y. Transformation to SCLC after treatment with the ALK inhibitor alectinib. J Thorac Oncol. 2016;11(6):e67‐e72. [DOI] [PubMed] [Google Scholar]
  • 33. Takegawa N, Hayashi H, Iizuka N, et al. Transformation of ALK rearrangement‐ positive adenocarcinoma to small‐cell lung cancer in association with acquired resistance to alectinib. Ann Oncol. 2016;27(5):953‐955. [DOI] [PubMed] [Google Scholar]
  • 34. Petrylak DP. First‐in‐human phase I study of ARV‐110, an androgen receptor (AR) PROTAC degrader in patients (pts) with metastatic castrate‐resistant prostate cancer (mCRPC) following enzalutamide (ENZ) and/or abiraterone (ABI). 2020 ASCO annual meeting proceedings. J Clin Oncol. 2020;38(15s):165s. [Google Scholar]
  • 35. Shaw AT, Friboulet L, Leshchiner I, et al . Resensitization to crizotinib by the lorlatinib ALK resistance mutation L1198F. N Engl J Med. 2016;374(1): 54–61. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from MedComm are provided here courtesy of Wiley

RESOURCES