ABSTRACT
Dysentery caused by Entamoeba histolytica affects millions of people annually. Current treatment regimens are based on metronidazole to treat invasive parasites combined with paromomycin for luminal parasites. Issues with treatment include significant side effects, inability to easily treat breastfeeding and pregnant women, the use of two sequential agents, and concern that all therapy is based on nitroimidazole agents, with no alternatives if clinical resistance emerges. Thus, the need for new drugs against amebiasis is urgent. To identify new therapeutic candidates, we screened 11,948 compounds assembled for the ReFRAME (Repurposing, Focused Rescue, and Accelerated Medchem) library against E. histolytica trophozoites. We identified 159 hits in the primary screen at 10 μM, and 46 compounds were confirmed in secondary assays. Overall, 26 were selected as priority molecules for further investigation, including 6 FDA approved, 5 orphan designations, and 15 that are currently in clinical trials (3 phase III, 7 phase II, and 5 phase I). We found that all 26 compounds are active against metronidazole-resistant E. histolytica, and 24 are able to block parasite recrudescence after drug removal. Additionally, 14 are able to inhibit encystation and 2 (lestaurtinib and LY-2874455) are active against mature cysts. Two classes of compounds are most interesting for further investigations: (i) the Bcr-Abl tyrosine kinase (TK) inhibitors, with ponatinib (50% effective concentration [EC50], 0.39) as the most potent; and (ii) mTOR or phosphatidylinositol 3-kinase (PI3K) inhibitors, with 8 compounds in clinical development, of which 4 have nanomolar potency. Overall, these are promising candidates and represent a significant advance for development of drugs against E. histolytica.
KEYWORDS: ReFRAME library, amebiasis, drug discovery, repurposing
INTRODUCTION
Entamoeba histolytica is an anaerobic protozoan and a waterborne pathogen generally spread in situations associated with contaminated food and water and lack of access to sanitation, as occurs in developing countries. The parasite is estimated to infect 500 million people annually (1, 2), with a prevalence of infection as high as 40% in some populations in regions of endemicity (3) and is estimated to cause 55,000 deaths each year (4). Amebiasis is also a major cause of traveler’s diarrhea (5). Among returning travelers reporting morbidity at GeoSentinel sites (mostly in Europe and the United States), the estimated incidence of E. histolytica infection is 12.5% of all microbiologically confirmed cases of infectious gastrointestinal diseases (6). Traveler’s diarrhea is also a problem among military personnel returning from regions of endemicity, with an incidence as high as 36.3 cases per 100 person-months (7). Sexual transmission and immigration are also among the reasons for amebiasis reemergence in developed countries (8).
Infection occurs when E. histolytica cysts are ingested in contaminated food or water and undergo excystation to produce the replicative and invasive trophozoite form. Cysts are resistant in the environment and responsible for spreading the disease to new patients. Disease begins with the adherence of the trophozoite form to colonic mucins and epithelial cells: the most common manifestation is amebic colitis, but infection may spread to other organs, particularly the liver, lung, and brain (9). Amebic colitis can be misdiagnosed as inflammatory bowel disease, which may lead to fulminant colitis after corticosteroid treatment (10).
Metronidazole (MNZ) is the most commonly prescribed treatment for invasive amebiasis. It is a low-cost nitroimidazole compound that is oral, rapidly absorbed, and highly effective against invasive trophozoites (9, 11). However, it has mild to severe side effects and is ineffective against luminal parasites and cysts: 40% to 60% of patients have parasites persist in the colonic lumen after treatment. Therefore, after metronidazole treatment, a second round of treatment with luminal agents such as paromomycin is required (12). The complexity of this regimen and the lack of symptoms during the second phase, when the second agent is needed, may reduce patient compliance and result in recurrent disease (13, 14). Other treatments for amebiasis are available; however, all rely on other nitroimidazole compounds with similar issues (11). Additionally, metronidazole resistance is easily induced under laboratory conditions (15). In Giardia and Trichomonas, other protozoan parasites that are treated with metronidazole, clinical metronidazole resistance is an increasing problem, indicating that similar problems may also arise with Entamoeba (16, 17). Thus, there is a need for development of new treatments for amebiasis, preferably with different modes of action to serve as optional therapy if parasite drug resistance emerges.
Drug development is costly and risky, with an average of $1.6 billion of investment for each drug launched (18). For this reason, diseases with lower predicted returns, including neglected diseases such as amebiasis, receive less attention from traditional routes of drug development. Repurposing drugs with known human safety profiles is an alternative that offers fewer risks, lower costs, and shorter timelines (19, 20). Auranofin is a successful example of repurposing, having been identified against amebiasis in a screening of ∼700 FDA-approved drugs (21); it has subsequently passed phase I clinical trials (22). Another recent screen of ∼3,400 unique compounds identified anisomycin, obatoclax, and prodigiosin as potent inhibitors of E. histolytica with activity against cysts and metronidazole-resistant parasites (23). Disulfiram, an antabuse FDA-approved drug, has also being highlighted as an interesting option for repurposing against amebiasis as its metabolite, zinc-ditiocarb complex, was shown to be efficient against E. histolytica in a mouse model (24, 25).
In this effort we screened the ReFRAME (Repurposing, Focused Rescue, and Accelerated Medchem) library, a set of ∼12,000 drugs that were assembled with a majority of compounds tested for clinical safety to accelerate discovery of drugs for new indications, including neglected and rare diseases (26). The ReFRAME library has been screened for many infectious disease indications, and the results are compiled in the a publicly available platform (https://reframedb.org). In our final analysis, we identified 26 priority compounds, including compounds with relevant features such as activity against metronidazole-resistant parasites, blockage of recrudescence, and inhibition of cysts. This effort demonstrates the value of screening the carefully assembled ReFRAME library against E. histolytica and represents a significant advance for drug development against amebiasis.
RESULTS
Screening the ReFRAME library identifies 86 hits against E. histolytica.
New treatments are needed for amebiasis, a neglected disease that is largely prevalent in areas of the developing world. To identify drug development candidates, we screened the ReFRAME library, a collection with 11,948 compounds assembled with the focus on accelerating drug development for neglected diseases (26). The workflow from screen to the list of final selected compounds is presented in Fig. 1. We identified 159 active compounds using a cutoff of 75% of viability inhibition (see Table S1 in the supplemental material). Primary screen hits were tested in 7-point dose-response curves (from 9 nM to 20 μM) in duplicate, using luciferase-expressing parasites. Setting a cutoff of 50% effective concentrations (EC50s) lower than 20 μM, 86 hits were considered active (data available at the ReFRAME online portal at https://reframedb.org/assays/A00203 and in Table S2 in the supplemental material).
Both primary and secondary screenings were performed using a transgenic cell line. Although G418 is removed from the medium for the assay, residues of the antibiotic could impact compound activity. This concern is resolved in subsequent confirmatory and phenotypic assays, in which we used the nontransgenic E. histolytica HM-1:IMSS strain. Other limitations should also be considered in drug discovery using a high-throughput library in vitro. For instance, in the case of therapies designed with prodrugs, the molecules we tested may not be the active form, which may lead to false-negative results. Moreover, in our screening we used a laboratory strain of E. histolytica. Under this condition, no host factor is considered, no pharmacology is addressed, and in the adaptation to the laboratory culture, the parasite may lose important aspects of its biology in vivo. Nevertheless, the significant findings emerging from high-throughput screenings with E. histolytica (21–23) are evidence that important drug discovery knowledge can be accessed with this methodology. Additionally, a number of hits which had been previously shown to inhibit E. histolytica viability were identified in this screen such as obatoclax, nithiamide, auranofin, and etofamine, further validating our approach (21, 23, 27).
Confirming compound activity.
With the list of 86 hits after the secondary screen, we selected 51 compounds based on potency, advanced clinical stages, and commercial availability for further testing. These compounds, plus an additional 20 related compounds, were purchased and tested in 20-point dose-response curves (0.09 nM to 50 μM). Assays were performed in triplicate, using E. histolytica HM-1:IMSS trophozoites in a fluorescent dye (fluorescein diacetate) viability assay to evaluate parasite survival. We chose to perform the confirmatory assay with a different readout to identify compounds that truly inhibit parasite viability and remove those that may have been identified in the first screen simply because they interfere with luciferase. All of the data obtained in this step, including EC50, EC90, R2 values, and efficacy are shown in Table S3 in the supplemental material. After this assay, a total of 46 compounds were considered active with a confirmed EC50 lower than 20 μM. We further selected compounds for ongoing testing using a variable threshold for potency depending on the clinical development stage of the compound. We applied the following criteria: EC50 of <10 μM for drugs that received FDA approval (6 molecules) or orphan designation (5 molecules), EC50 of <2 μM for drugs in phase III clinical trials (3 molecules), and EC50 of <1 μM for drugs in phase II and I clinical trials (7 and 5 molecules). With these criteria, we selected 26 compounds that are more potent than metronidazole (EC50, 11.85 μM), with 16 drugs presenting submicromolar potencies (Table 1). The targets in humans for each molecule are listed; a majority of kinases and some repeated antineoplastic targets such as mTOR/phosphatidylinositol 3-kinase (PI3K) and Bcr-Abl tyrosine kinase (TK) can be observed. Overall, these results identify a number of new druggable compounds that can be explored as therapeutic options against Entamoeba.
TABLE 1.
Highest phase | Compound name | EC50 (μM) for E. histolytica | Known target(s) in human cells |
---|---|---|---|
Positive control | Metronidazole | 11.85 | |
FDA approved | Plicamycin | 0.26 | DNA |
Bortezomib | 0.61 | Proteasome | |
Ponatinib | 0.39 | Bcr-Abl TK | |
Dasatinib | 1.73 | Bcr-Abl TK | |
Bosutinib | 2.44 | Bcr-Abl TK | |
Telotristat etiprate | 6.70 | TPH1 | |
FDA orphan clinical trial | |||
Phase III | Vosaroxin | 2.85 | DNA topoisomerase |
Lestaurtinib | 4.77 | FLK, TrK, JAK | |
Bardoxolone methyl | 6.31 | Multiple | |
Phase II | Zotiraciclib (TG-02) | 2.32 | CDKs, JAK2, FLT3 |
Milciclib maleate | 7.60 | CDK, TrKA | |
Clinical trial | |||
Phase III | Beloranib | 0.02 | MetAP2 |
Taselisib | 1.39 | PIK3 | |
Dactolisib | 1.53 | mTOR/PI3K | |
Phase II | CC-115 | 0.03 | mTOR, DNA-PK |
NVP-BGT226 | 0.06 | mTOR/PI3K | |
TNP-470 | 0.07 | MetAP2 | |
Bruceantin | 0.11 | Myc | |
CUDC-907 | 0.44 | HDAC, PI3K | |
PF-04691502 | 0.47 | mTOR | |
Sapanisertib | 0.93 | mTOR | |
Phase I | Omipalisib | 0.03 | mTOR/PI3K |
LY-2874455 | 0.31 | FGFR | |
Latrunculin B | 0.52 | Actin | |
R-547 | 0.53 | CDK 1/2 | |
PF-03814735 | 0.82 | Aurora kinase |
Specific target inhibitors.
Among the 26 selected compounds in Table 1, we noted that 8 are mTOR or PI3K inhibitors. To understand if this activity against the parasite could be related to the potency of these compounds against the targeted enzymes, we expanded this panel by screening an additional 13 compounds, including 3 ReFRAME compounds that were active but not included on our priority compound list, and 10 new compounds selected from commercially available with specific activities that were organized in the following categories: selective to mTOR, selective to PI3K, selective to both mTOR and PI3K (dual inhibitors), and rapalogs, which contain rapamycin and derivatives.
Of the 13 new inhibitors included, 6 were active against E. histolytica, in addition to the 8 previously identified mTOR/PI3K inhibitors (Table 2). Further data for these compounds, such as EC90, R2 values, and efficacy, are shown in Table S3. Neither mTOR nor PI3K ATP binding pocket inhibitors seem to be preferred, since no relationship was found when the inhibition of E. histolytica viability and the target-specific inhibition potencies of the compounds tested were compared (Table 2). Interestingly, none of the rapalog compounds showed significant activity, which is notable since rapalogs have a different mechanism than PI3K or mTOR selective inhibitors. The former class binds to FKBP12, and the complex inhibits mTOR by blocking substrate access (28), while the latter class competes with ATP for the binding pocket of these kinases (29). Hence, the lack of activity of rapamycin and rapalogs compared to variable activity in the other three categories indicates that the inhibition in parasite viability may be related to the compounds’ affinity to the ATP binding site of mTOR and PI3K.
TABLE 2.
Selectivity | Compound name | Potency against: |
Other target(s) | EC50 (μM) for E. histolytica | Highest clinical trial phase | |
---|---|---|---|---|---|---|
mTOR or Abl | PI3K or Bcr-Abl | |||||
mTOR | PI3K | |||||
mTOR | Sapanisertib | ++++ | + | 0.93 | Phase II | |
Gedatolisib* | ++++ | + | Not active | Phase II | ||
Vistusertib* | ++++ | P-Akt, PS6 | Not active | Phase II | ||
CC-115 | ++ | + | DNA-PK | 0.03 | Phase II | |
PI3K | Taselisib | +++++ | C2β, hVps34 | 1.39 | Phase III | |
GDC-0084* | + | +++ | 19 | Phase II | ||
NVP-BGT226 | ++ | 0.06 | Phase I/II | |||
AZD-8835 | + | 5.71 | Phase I | |||
Wortmannin* | +++ | DNA-PK, ATM, MLCK | 2.47 | Preclinical | ||
GSK-2636771 | +++ | 18.48 | Phase I/II | |||
XL765* | + | ++ | DNA-PK | Not active | Phase II | |
CUDC-907 | ++ | HDAC | 0.44 | Phase II | ||
Dual | Omipalisib | +++++ | +++++ | 0.03 | Phase I | |
SB-2343 | ++++ | +++ | 8.18 | Phase I | ||
Dactolisib | +++ | ++ | ATR | 1.53 | Phase III | |
Bimiralisib* | ++ | +++ | Not active | Phase II | ||
PF-04691502 | ++ | ++++ | P-Akt | 0.47 | Phase II | |
Apitolisib* | ++ | +++ | 15.53 | Phase II | ||
Rapalogs | Rapamycin* | ++++ | FKBP12 | Not active | FDA approved | |
Everolimus* | ++++ | FKBP12 | Not active | FDA approved | ||
Temsirolimus* | ++++ | FKBP12 | Not active | FDA approved | ||
Abl | Bcr-Abl | |||||
Bcr-bl | Ponatinib | +++++ | PDGFRα, VEGFR2, FGFR1 | 0.3927 | FDA approved | |
Dasatinib | +++++ | Src, c-Kit(D816V), c-Kit (WT) | 1.728 | FDA approved | ||
Bosutinib | +++++ | STAT3, ERK, S6 kinase | 2.44 | FDA approved | ||
Rebastinib | ++++ | FLT3, KDR, Tie-2 | 13.4 | FDA orphan (phase II) | ||
Imatinib | + | c-Kit, PDGFR | Not active | FDA approved | ||
Nilotinib | ++ | Not active | FDA approved | |||
Bafetinib | +++ | Lyn | Not active | Phase II | ||
Danusertib | ++ | Aurora A, TrkA, RET | Not active | Phase II | ||
Tozasertib | ++ | Aurora A, Aurora C, Aurora B | Not active | Phase I |
In the top part of the table, inhibitors of mTOR and PI3K with distinct selectivity were tested in 8- or 16-point drug response curves against trophozoites of E. histolytica. The top part of the table is divided into categories of selectivity to mTOR, PI3K, or dual mTOR/PI3K inhibitors, as well as rapalogs. The potency of each molecule against the purified human protein is represented in the following scale: +++++, <1 nM; ++++, 1 nM to <5 nM; +++, 5 nM to <10 nM; ++, 10 nM to <50 nM; +, >50 nM (50, 62–82). In the bottom part of the table, inhibitors of Bcr-Abl TK are listed, and the potency of each molecule against the purified human protein is represented using the scale defined above (83–90). Activity is shown according to the EC50 (μM). A compound name followed by an asterisk (*) indicates additional compounds not identified in the primary screen. All other molecules were identified from the ReFRAME library. The highest phase of clinical studies achieved with each compound and the targets in humans of each compound are listed.
ReFRAME screens against other intestinal parasites revealed less activity of mTOR/PI3K inhibitors than what we observed. SB-2343 is the only compound active against Giardia lamblia and NVP-BGT226, and sapanisertib, dactolisib, and gedatolisib are active against Cryptosporidium parvum (30). The activities of mTOR/PI3K inhibitors have been found enriched against amebic parasites that can infect the brain, including Naegleria, Balamuthia, and Acanthamoeba (31), suggesting this class of compounds may have greater activity against amebic parasites. Interfering with mTOR pathway in the host has also been studied as a possible therapeutic route for parasitic diseases, such as toxoplasmosis, leishmaniasis, and malaria (32).
Another target that is repeated among the selected compounds is Bcr-Abl TK, with ponatinib, dasatinib, and bosutinib as potent FDA-approved compounds against E. histolytica. We expanded this list by testing rebastinib, bafetinib, nilotinib, imatinib, danusertib, and tozasertib. We observed increasing activity against E. histolytica corresponding to increasing potency of these compounds against human ABL1 (Table 2). Although the parasite target has not been determined for these molecules, our results indicate that Bcr-Abl TK inhibitors may be an interesting class of compounds for further investigations of their activity against Entamoeba.
Activity against metronidazole-resistant parasites.
In order to identify compounds that are active against metronidazole-resistant parasites, we tested the 26 selected compounds against a metronidazole-resistant cell line that we previously generated (23). We performed parallel experiments in 6-point dose-response curves and in both wild-type (WT) and MNZ-resistant parasites.
The EC50 calculated for metronidazole against the MNZ resistant parasites was almost 4× greater than that for the WT strain, validating the MNZ resistance of this cell line (Table 3). Of the 26 drugs tested against wild-type and metronidazole-resistant parasites, 24 had comparable EC50s for WT and MNZ-resistant parasites, with a fold change lower than 2 indicating similar sensitivity to the drug of interest in the MNZ-resistant line. This may indicate that the drugs tested have mechanisms of action in E. histolytica that are different from the one of metronidazole, which will be useful in case of emergence of clinical resistance. A few compounds had higher EC50s (fold change of >2) for the metronidazole-resistant parasites, such as bosutinib (fold change of 2.1) and NVP-BGT226 (fold change of 2.05). However, even for these drugs, the EC50s against MNZ-resistant parasites were still low (5.2 and 0.13 μM, respectively). The slight increase in EC50 for bosutinib is probably not related to its mechanism of action, since the effect was not observed for other Bcr-Abl TK inhibitors, ponatinib and dasatinib. Anisomycin and prodigiosin were previously identified as active against metronidazole-resistant parasites (23), and lestaurtinib and bardoxolone methyl have also been found active against Giardia (30), which makes these compounds promising for treating both intestinal parasitic diseases. The ReFRAME library has not been tested against Trichomonas vaginalis, but this represents another anaerobic pathogen typically treated with MNZ, but in which resistance has been demonstrated (17). These results show that all 26 compounds represent viable alternatives to nitroimidazole therapies as they are active against MNZ-resistant E. histolytica parasites, and some may also be interesting leads against other parasites that develop MNZ resistance. Of note, in this assay the absolute EC50 value for metronidazole against the HM-1:IMSS strain is somewhat lower than the one found at the 20-point dose-response curve that we have previously demonstrated; this could be due to having lower resolution, with a 6-point dose-response curve versus a 20-point dose response curve, or to the parasite growth differences in 96-well plates versus the experiment’s 384-well plates.
TABLE 3.
Highest phase | Compound name | EC50 (μM) from 6-pt dose-response curves |
Fold change | |
---|---|---|---|---|
WT (HM-1:iMSS) | MNZ resistant | |||
Positive control | Metronidazole | 4.93 | 18.77 | 3.8 |
FDA approved | Plicamycin | 0.26 | 0.05 | 0.2 |
Bortezomib | 0.42 | 0.18 | 0.4 | |
Ponatinib | 0.39 | 0.73 | 1.9 | |
Dasatinib | 1.73 | 1.82 | 1.1 | |
Bosutinib | 2.44 | 5.20 | 2.1 | |
Telotristat etiprate | 6.70 | 5.15 | 0.8 | |
FDA orphan clinical trial | ||||
Phase III | Vosaroxin | 2.14 | 2.95 | 1.4 |
Lestaurtinib | 2.64 | 1.94 | 0.7 | |
Bardoxolone methyl | 2.81 | 4.93 | 1.8 | |
Phase II | Zotiraciclib (TG-02) | 2.32 | 1.57 | 0.7 |
Milciclib maleate | 4.89 | 4.27 | 0.9 | |
Clinical trial | ||||
Phase III | Beloranib | 0.02 | 0.02 | 1.1 |
Taselisib | 0.83 | 1.39 | 1.7 | |
Dactolisib | 0.17 | 0.04 | 0.3 | |
Phase II | CC-115 | 0.03 | 0.04 | 1.4 |
NVP-BGT226 | 0.06 | 0.13 | 2.1 | |
TNP-470 | 0.03 | 0.02 | 0.9 | |
Bruceantin | 0.05 | 0.03 | 0.6 | |
CUDC-907 | 0.18 | 0.12 | 0.6 | |
PF-04691502 | 0.47 | 0.48 | 1.0 | |
Sapanisertib | 0.41 | 0.30 | 0.7 | |
Phase I | Omipalisib | 0.01 | 0.02 | 1.4 |
LY-2874455 | 0.28 | 0.48 | 1.7 | |
Latrunculin B | 0.23 | 0.42 | 1.8 | |
R-547 | 0.55 | 0.50 | 0.9 | |
PF-03814735 | 0.57 | 0.66 | 1.2 |
The 26 compounds were tested in 6-point dose-response curves against MNZ resistant in parallel with the wild-type parasite (HM1:IMSS) E. histolytica. Results are shown as EC50 (μM) of both parasite lines, and the fold change (MNZ EC50 versus WT EC50) for each compound tested is shown in the last column.
Activity in preventing parasite recrudescence.
Recrudescence was evaluated by testing the parasite’s viability when treated with the compounds’ EC90 before and after a 72-h period of drug removal. (Fig. 2; see Table S4 in the supplemental material). Metronidazole was included at its EC90 for comparison with the standard of care and at 50 μM in attempt to promote complete inhibition of the parasite growth. Vosaroxin (33%) and bruceantin (61%) exhibited higher recrudescence than metronidazole (18%). All other 24 compounds were effective in preventing recrudescence, suggesting a cytotoxic effect, which is ideal for treating parasitic diseases and would help prevent recurrent disease by dormant parasites. These data are useful in helping to prioritize compounds for further development against Entamoeba. A similar approach in another amebic parasite, Balamuthia mandrillaris, identified plicamycin, zotiraciclib (TG-02), and auranofin as drugs that inhibit recrudescence (31).
Drug activity against encystation and mature cysts.
Current treatment of amebiasis involves metronidazole to treat trophozoites, followed by paromomycin to target luminal parasites’ trophozoites and cysts (11, 33). Compounds that target trophozoites and can also inhibit encystation or kill mature cysts can drastically reduce amebiasis transmission and could lead to a single-drug treatment for Entamoeba. To identify compounds with this feature, we tested the selected drugs against encystation and mature cysts. E. histolytica can be cultivated as trophozoites, but the encystation process has not yet been developed to high efficiency in the lab for the purposes of a drug screen. The currently accepted model for high-level, reliable in vitro encystation uses Entamoeba invadens, a species that infects reptiles (34). Thus, we defined the EC50 for the 26 selected compounds in E. invadens (Table 4). R2 values and efficacy are shown in Table S5 in the supplemental material. Of the 26 compounds, plicamycin is the only drug that did not have any effect on E. invadens viability. Bosutinib, telotristat etiprate, CUDC-907, and R-547 showed low efficacy (EC50 higher than 20 μM) (Table S5). Drug susceptibility differences between E. histolytica and E. invadens have previously been noted, and so these results are not surprising (23).
TABLE 4.
Highest phase | Compound name | EC50 (μM) for E. invadens | % Encystation after treatment at 1× EC50 | Significance vs DMSO |
---|---|---|---|---|
Positive control | A23187 (at 5 μM) | NA | 26 ± 3.4 | **** |
FDA approved | Plicamycin | Not calculated | Not tested | |
Bortezomib | 3.93 | 4 ± 0.9 | **** | |
Ponatinib | 6.97 | 76 ± 5.3 | * | |
Dasatinib | 4.42 | 87 ± 8.8 | NS | |
Bosutinib | 22.96 | 11 ± 1.6 | **** | |
Telotristat etiprate | 26.24 | 41 ± 7.5 | **** | |
FDA orphan clinical trial | ||||
Phase III | Vosaroxin | 3.03 | 11 ± 2.2 | **** |
Lestaurtinib | 1.27 | 5 ± 0.9 | **** | |
Bardoxolone methyl | 2.38 | 2 ± 0.3 | **** | |
Phase II | Zotiraciclib (TG-02) | 15.62 | 4 ± 1.2 | **** |
Milciclib maleate | 13.34 | 27 ± 4.2 | **** | |
Clinical trial | ||||
Phase III | Beloranib | 0.15 | 11 ± 2.2 | **** |
Taselisib | 1.43 | 55 ± 19.8 | **** | |
Dactolisib | 1.05 | 68 ± 9.0 | ** | |
Phase II | CC-115 | 0.31 | 68 ± 14.7 | ** |
NVP-BGT226 | 0.67 | 64 ± 11.4 | ** | |
TNP-470 | 0.06 | 11 ± 2.6 | **** | |
Bruceantin | 0.06 | 79 ± 19.9 | NS | |
CUDC-907 | 27.96 | 3 ± 0.9 | **** | |
PF-04691502 | 0.13 | 36 ± 8.4 | **** | |
Sapanisertib | 3.01 | 70 ± 14.3 | ** | |
Phase I | Omipalisib | 0.01 | 113 ± 16.6 | NS |
LY-2874455 | 1.40 | 9 ± 2.2 | **** | |
Latrunculin B | 0.10 | 24 ± 7.0 | **** | |
R-547 | 5.01 | 14 ± 1.5 | **** | |
PF-03814735 | 1.60 | 22 ± 9.7 | **** |
Trophozoites of E. invadens were incubated in encystation medium for 48 h with the selected compounds at a concentration equal to the EC50 obtained in E. invadens. Cysts were counted using high-content imaging. The results are presented as a percentage of the encystation value ± standard error. Treatment with DMSO is considered 100% encystation. NA, not applicable. Significance was calculated with a t test against DMSO treatment values. Asterisks: *, adjusted P values between 0.05 and 0.01; **, adjusted P values between 0.01 and 0.001; ****, adjusted P values of <0.0001. Nonsignificant differences are indicated (NS). Boldface indicates compounds that were considered active, reducing encystation efficiency (<30% encystation).
After defining the EC50 in E. invadens trophozoites, the activity against encystation was tested (23). The results are expressed as a percentage of the control number of cysts after 48 h of treatment (Table 4). In this in vitro model, we have determined that the calcium ionophore A23187 (35) blocks encystation when added at the initiation of encystation (see Fig. S1 in the supplemental material). We adapted this compound for use as a positive control in our encystation assay and saw significant reduction of encystation. Treatment with dimethyl sulfoxide (DMSO) is considered to show 100% encystation efficiency, and the calcium ionophore A23187 is the positive control for blocking encystation. Metronidazole had been shown to be ineffective against encystation efficiency (23). Of the 25 molecules tested in this manner, 14 were considered active by using the criterion that treatment with 1× EC50 resulted in >70% encystation inhibition. Encystation inhibitors have been previously identified from several repurposing drug libraries (23). These compounds may serve as a useful tool to block an important biological pathway of the parasite and would be of high priority as they may serve as a one-drug approach to treat both trophozoites and cysts.
The assay described above tests the encystation process, but additional inhibition of mature cysts is high priority as it is the shedding of mature cysts into the environment that propagates disease. We previously developed an assay to test this feature and have demonstrated that metronidazole does not inhibit mature cysts (23). We now evaluated the ability of 10 compounds that were efficient encystation inhibitors to target mature cysts. Data are expressed as percentage of inhibition compared to treatment with DMSO (Fig. 3). Obatoclax, previously identified as targeting mature cysts, was used as a positive control (23). In our assay, lestaurtinib was the most active compound, with 75% and 50% mature cyst inhibition at 20 and 10 μM, respectively, and no activity observed at 5 μM. LY-2874455 was also active at 20 μM, with 65% inhibition, and with a slight but not significant inhibition at 10 μM. Milciclib maleate, bardoxolone methyl, and vosaroxin presented slight inhibition in cyst viability, although without significant difference. R-547, PF-03814735, bortezomib, and zotiraciclib (TG-02) did not inhibit cyst viability. These results highlight lestaurtinib and LY-2874455 as interesting compounds for further development against amebic cysts; both are oral drugs (36, 37) and potent inhibitors of both E. histolytica life cycle stages, trophozoites and cysts.
DISCUSSION
Screening of the ReFRAME library opened several drug development opportunities for amebiasis, with identification of 26 molecules in different stages of clinical trials and various target classes to be explored. Among this set of drugs, we identified compounds with advantages for clinical use, such as activity against MNZ-resistant parasites, blockage of parasite recrudescence, and efficacy against encystation or mature cysts. We compiled the results in a heat map to give an overview of the drugs, their regulatory status, and their features of interest in specific areas of parasite biology (Fig. 4).
We identified six FDA-approved compounds highly active against E. histolytica, of which plicamycin (EC50, 0.26 μM), ponatinib (EC50, 0.39 μM), and bortezomib (EC50, 0.61 μM) have submicromolar potency (Table 1). This highlights one advantage of the ReFRAME library, as it was assembled using compounds with significant clinical use. For a repurposing strategy, ponatinib is the most interesting among the three cited drugs as plicamycin has been discontinued and bortezomib is tolerated in mice in very low doses, which would make it challenging to test in animal models of amebiasis for in vivo efficacy (38, 39).
Ponatinib is a multikinase inhibitor with primary affinity to Bcr-Abl TK and with oral bioavailability (40). It is the most potent among the compounds in this class (EC50, 0.39 μM, compared to dasatinib and bosutinib, with EC50s of 1.73 and 2.44 μM, respectively) against E. histolytica and is 30× more potent than metronidazole. All three Bcr-Abl TK inhibitors are active against MNZ-resistant parasites and are able to block recrudescence, but are not able to inhibit encystation. These drugs, as well as other antineoplastic kinase inhibitors, were previously described to be active against E. histolytica with similar potencies (41). In addition, a cross talk was found between the Bcr-abl protein and the Trx system (42), a known target of amebiasis therapies with nitroimidazole molecules and auranofin (43, 44), highlighting the Bcr-Abl TK inhibitors as potential drug development candidates against the parasite.
All five drugs that have received FDA orphan designation were able to inhibit MNZ-resistant parasite viability: most blocked parasite recrudescence, were able to inhibit encystation, and, therefore, were tested against mature cysts. Lestaurtinib, a multitargeted tyrosine kinase inhibitor, was the most efficient compound, with 76% cyst inhibition at 20 μM and 50% at 10 μM. This molecule was found active against Naegleria (a free-living ameba) (31, 45), Trypanosoma cruzi, Leishmania major, Mycobacterium tuberculosis, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (30). Most interestingly, lestaurtinib was also found to inhibit G. lamblia, with similar potency found against E. histolytica (EC50 of 4.25 μM) (https://reframedb.org/assays/A00229). Giardiasis is also an intestinal disease spread by infectious cysts that is treated with metronidazole, with significant clinical resistance (16). Thus, lestaurtinib should be considered for further studies as a treatment for multiple intestinal parasitic pathogens.
Beloranib and TNP470 are among the most potent compounds found in this study (Table 1). Both are MetAP2 inhibitors and chemically similar to fumagillin, previously shown be active against Entamoeba (46). Both are very potent against metronidazole-resistant parasites, block recrudescence at very low concentrations, and are also able to inhibit encystation. However, concerns about venous thromboembolism in beloranib-treated patients motivated the development of alternative MetAP2 inhibitors with better safety profiles (47).
The majority of the other compounds are mTOR/PI3K inhibitors; most are in clinical trials in phase II and III, with omipalisib in phase I. All mTOR/PI3K inhibitors among the selected compounds (dactolisib, CC-115, NVP-BGT226, CUDC-907, PF-04691502, sapanisertib, and omipalisib) are able to inhibit the metronidazole-resistant parasites and block recrudescence, but most are not efficient encystation inhibitors. These compounds are all orally bioavailable, and several have been reported in phase I clinical trials to be well tolerated (48–54). Even though no relationship was observed between enzymatic specificity or potency and amoebicidal activity (Table 2), the behaviors of mTOR and PI3K inhibitors were similar across the phenotypic assays. Considering the lack of activity of rapalogs, compounds with higher affinity to the mTOR and PI3K ATP binding sites are of interest for development of therapy against amebiasis and further target validation studies.
As a neglected disease endemic in communities with poor sanitation conditions, a good candidate for drug development against amebiasis is an inexpensive, orally bioavailable molecule. It should ideally be safe for children and elderly people, as well as pregnant and breastfeeding women, with no severe or intolerable side effects. Slow absorption would be an interesting feature to ensure good luminal concentration. In addition to good cytotoxic potency against trophozoites and cysts of E. histolytica, the preferred compounds would also have good activity against metronidazole-resistant parasites.
Given these considerations, ponatinib would be a good candidate for a repurposing strategy as an FDA-approved oral drug potent against E. histolytica. It has been noted to cause severe side effects (e.g., pancreatitis, arrhythmia, and hypertension), which is a concern; however, those events were identified over several months of treatment, and lower doses of the drug are associated with fewer side effects (55). Thus, lower drug doses and a shorter treatment of duration may be able to overcome these hurdles and make this a viable option for amebiasis therapy. Lestaurtinib, another orally bioavailable drug with orphan status, is generally well tolerated (36, 56), has activity against Entamoeba cysts and Giardia, and is thus a very interesting option for further development. Molecules that are in earlier stages of development, such as LY-2874455 and the mTOR/PI3K inhibitors, also have relevant activity against Entamoeba, are orally bioavailable, and should be considered for further studies as more robust data on clinical safety profile and pharmacology parameters become available.
In summary, we identified several molecules and potential targets with promising profiles for the development of therapies against amebiasis, a devastating disease that affects millions of people worldwide. All of our selected compounds are more potent than the current standard of care, metronidazole, and several performed better in our phenotypic assays, representing clinical advantages, such as the abilities to inhibit encystation or mature cysts, block recrudescence, and inhibit MNZ-resistant parasites. As we move to the next steps, it will be important to design animal studies considering the particularities of selected drugs, such as possible immunosuppression and other side effects, to obtain animal pharmacology profiles and to determine in vivo efficacy in an animal model of colitis.
MATERIALS AND METHODS
Parasite culture and strains.
Trophozoites of Entamoeba histolytica strain HM-1:IMSS were grown and maintained at 37°C in TYI medium under standard conditions (57). For primary and secondary screen assays, we used a stable transgenic heterogeneous cell line expressing luciferase (Eh-CS-luc) maintained in 6 μg/mL G418 (58). Trophozoites of Entamoeba invadens (strain IP-1) were grown and maintained at 25°C in LYI medium under standard conditions (34). For the cyst assay, a stable transgenic heterogeneous E. invadens cell line expressing luciferase (Ei-CK-luc) was established and maintained in 40 μg/mL G418 in LYI medium (59). Both E. histolytica and E. invadens parasites were maintained in glass tubes and expanded for 2 to 3 days before the assays into plastic T25 flasks.
Compound library and screening.
For the primary and secondary screen assays, the compounds from the ReFRAME library were pinned for a total of 0.1% of volume in white 384-well Greiner Bio-One plates, which were heat-sealed, shipped frozen, and kept at −20°C until the day before testing. Our screening was performed with the support of the Stanford High Throughput Biosciences Center (http://htbc.stanford.edu). The plates were thawed at room temperature overnight. E. histolytica cells expressing luciferase (Eh-CS-luc) grown in T25 flasks were iced for 10 min, centrifuged for 10 min at 230 relative centrifugal force (RCF), resuspended in fresh medium, and seeded into the library plates at 3,000 Eh-CS-luc cells/well in 100 μl of fresh TYI medium using a Multidrop DW (Thermo Fisher, USA). Plates were covered with a lid and incubated at 37°C for 48 h in an anaerobic chamber using a BD GasPak System with two CO2 generator sachets (Becton Dickinson and Company, USA). BrightGlo (Promega, USA) reagent was prepared and kept at −80°C until the day before reading, when it was transferred to 4°C to thaw overnight. At 2 h before reading, the BrightGlo reagent and assay plates were placed on the bench to equilibrate at room temperature. After 40 min, plates were centrifuged for 10 min at 230 RCF, the liquid was aspirated with Bio-Tek ELx405 (BioTek, USA) to leave 10 μL per well, and then 20 μL per well of BrightGlo diluted in phosphate-buffered saline (PBS [1:1]) was added. Luminescence signal was read on the Tecan Infinite M1000 Pro (Molecular Devices, US). DMSO at 0.1% and metronidazole at 50 μM were used as negative and positive controls, respectively.
The primary screening was performed with the whole ReFRAME library at a single dose of 10 μM and a single replicate. Compounds that inhibited E. histolytica viability at least 75% compared to the DMSO control were selected as primary hits (Table S1). These were used for the secondary screen, in which compounds were pinned for 7-point dose-response curves (from 9 nM to 20 μM) in two biological replicates. Raw data were normalized based on percentage of blank control, and the EC50 was calculated using Genedata software. The dose-response curves of the 86 compounds considered active were used to calculate EC50, efficacy and R2 values (Table S2). All data are available at https://reframedb.org/assays/A00203.
Confirmatory assays.
For the confirmatory assays, we purchased 71 molecules (51 identified from the ReFRAME library and 20 related compounds) from commercial sources and tested them in 20-point dose-response curves in Greiner-Bio Cellstar 384-well plates with black walls. For the assay, 10 μL of medium was added to all wells, and another 10 μL of medium containing 2 μL of DMSO stock solution of each compound at 5 mM was added to the well with the highest concentration of the dose-response curve. Serial dilution (1:2) was performed by transferring 10 μL of the diluted solution to the following 19 wells, to result in a curve ranging from 0.09 nM to 50 μM when completed to a final volume of 100 μL. Cells of wild-type E. histolytica strain HM-1:IMSS growing in T25 flasks were iced for 10 min, centrifuged for 10 min at 230 RCF, resuspended in fresh medium, and primed to the multidrop pipettor. Parasites were plated (3,000 cells per well in 90 μL), and plates were covered with a lid and incubated in anaerobic chamber at 37°C for 72 h. Two columns were left with medium only, to calculate the background value. Plates were then centrifuged for 10 min at 230 RCF, and the medium was aspirated with Bio-Tek ELx405 (BioTek, USA), leaving 10 μL. Then, 30 μL of fluorescein diacetate diluted in fresh medium at a concentration of 20 μg/mL was added to all wells. The plates were incubated for 20 min at 37°C. To wash the medium with fluorescein diacetate and avoid high fluorescence background, 70 μL of 1× PBS was added per well, the plates were centrifuged for 10 min at 230 RCF, the liquid was again aspirated with Bio-Tek ELx405 (BioTek, USA), leaving 10 μL, and another 90 μL of 1× PBS was added to each well. Fluorescence was read using the Tecan Infinite M1000 Pro. Metronidazole dose curves were used as a positive control in all biological replicates, and 1% DMSO was used as a negative control. EC50s were calculated as described below. Each compound was tested in at least three biological replicates.
EC50 calculation.
In order to calculate the EC50, the percentage of inhibition relative to maximum and minimum reference signal controls was calculated using the formula % inhibition = [(mean of maximum signal reference control − experimental value)/(mean of maximum signal reference control − mean of minimum signal reference control)] × 100.
The relative dose-response data in triplicate were exported to GraphPad Prism software 8.0 for EC50 calculations using 4PL sigmoidal nonlinear regression. R2 values and efficacy were used for statistical analysis.
Evaluating E. histolytica recrudescence after drug treatment.
To investigate if any viable E. histolytica were remaining after compound treatment, we evaluated the ability of parasites to recover growth after treatment with the selected drugs for 72 h. For that, 200,000 parasites were treated in 8-mL glass tubes with each of the 26 compounds at a concentration able to inhibit 90% of E. histolytica growth in 72 h of treatment at 37°C (EC90). After treatment, the tubes were iced and centrifuged for 6 min at 230 RCF. The pellets were resuspended in 8 mL of fresh medium, and 300 μL of the suspension was transferred to multiple wells of two 96-well black plates with clear bottoms. One of the plates was incubated with fluorescein diacetate dye immediately, and the viability was evaluated as described previously. The second plate was incubated in an anaerobic chamber at 37°C for 72 h to allow any remaining viable parasites to grow. After this 72-h recovery period, parasite viability was also evaluated with fluorescein diacetate dye. Treatment with metronidazole at 50 μM was used as a positive control and 0.5% DMSO as a negative control. Each compound was tested in three biological replicates. Percent values were exported to GraphPad Prism software 8.0. For statistical analysis, values obtained before and after the 72-h period of drug removal were compared using a t test. Recrudescence was considered positive when the value after drug removal was above 10% and significantly different (P < 0.05) from the value previously obtained for 72-h treatment with the same compound.
Drug effect on metronidazole-resistant E. histolytica.
Parasites resistant to metronidazole (MNZ) were generated by continuous growth of E. histolytica HM-1:IMSS in MNZ, with steadily increasing concentrations from 1 μM to 15 μM (23). Parasites growing well and able to be sequentially passaged multiple times at 15 μM MNZ were defined as MNZ resistant and used for further experiments. A total of 15,000 amebae from the MNZ-resistant strain were seeded into a 96-well plate with medium and dose-response curves or the DMSO control. The plates were covered with a lid and incubated in an anaerobic chamber for 72 h at 37°C. Dose-response curves were done with 6 points in a 1:3 dilution, and the maximum concentrations were 45 μM for drugs with an EC50 of >1 μM, 5 μM for drugs with an EC50 between 0.1 μM and 1 μM, and 1.6 μM for drugs with an EC50 of <0.1 μM. Viability was assayed by fluorescein diacetate fluorescence, as detailed previously. Parallel dose-response curves were performed using the MNZ-sensitive E. histolytica HM-1:IMSS strain as a control, seeded at 6,000 parasites per well. Each compound was tested in three biological replicates. The compounds’ EC50s against the WT (HM-1:IMSS) and MNZ resistance cell lines were calculated and analyzed as described above. Fold change (EC50 in the MNZ-resistant strain versus EC50 in the WT HM-1:IMSS strain) was used to evaluate if any change of activity is found when the parasite is resistant to metronidazole.
EC50 definition in E. invadens.
E. invadens EC50s were determined by seeding E. invadens IP-1 parasites in logarithmic growth in 96-well black plates with clear bottoms, which were covered with a lid in an anaerobic chamber at a density of 40,000 parasites per well, with the compounds in dose-response curves done with 9 points ranging from 7 nM to 45 μM. Potent drugs were further tested in dose-response curves ranging from 0.8 nM to 5 μM. After 72 h at 25°C, plates were centrifuged at 230 RCF, liquid was aspirated, 50 μL fluorescein diacetate dye was added at a concentration of 20 μg/mL, and the plates were incubated for 20 min. Once again, plates were centrifuged at 230 RCF, liquid was aspirated, and 300 μL of 1× PBS was added to each well. Fluorescence was read using the Tecan Infinite M1000 Pro. Each compound was tested in three biological replicates. Metronidazole dose curves were used as a positive control in all biological replicates, and 1% DMSO was used as a negative control. EC50s were calculated as described above.
Evaluating drug effect on encystation efficiency.
Encystation assays were performed as previously described (23). Briefly, E. invadens IP-1 trophozoites were harvested in mid-log phase, washed once, resuspended in encystation medium (47% LG) (34), and plated in 96-well black plates with clear bottoms, at a density of 50,000 parasites per well. Drugs were added at a concentration that was 1× the E. invadens EC50, as determined above. Plates were covered with a lid and incubated for 48 h at 25°C in an anaerobic chamber and then spun, and the medium was removed. Cysts were stained by addition of 50 μL of 50 μM calcofluor white (Sigma) and 4% paraformaldehyde (PFA) in PBS, incubated at room temperature for 20 min, and evaluated by high-content imaging in an ImageExpress Micro (Molecular Devices). The number of cysts per well was quantified using MetaExpress software. Each compound was tested in three biological replicates. Parasites treated with DMSO only were used as a positive control, and media without parasites were used to establish background signal. A23187 was used as a positive control. A23187 is a calcium ionophore that blocks encystation when incubated at 5 μM with trophozoites in encystation medium for 48 h, and this was evaluated by counting cysts after a 30-min incubation with 0.1% Sarkosyl (Fig. S1). We adapted this compound for use as a positive control in our encystation assay and saw significant reduction of encystation. Percent values were exported to GraphPad Prism software 8.0 for statistical analysis. The percentage of encystation was calculated as the mean ± standard error. Significance was calculated based on t test using percentage of encystation obtained with DMSO treatment as a reference for comparison.
Evaluating effect on mature cyst viability.
Compounds identified as encystation inhibitors were tested for the ability to reduce mature cyst viability, as previously described (23). E. invadens cells expressing luciferase (Ei-CK-luc) were induced to encyst by incubation in encystation medium (47% LG). After 72 h, cells were harvested, washed once in distilled water, resuspended in water, and incubated at 25°C for 5 h to lyse trophozoites. Purified cysts were pelleted, counted, resuspended in encystation medium, and distributed in 8-mL glass tubes to have 4 × 105 cells per tube. Drug or DMSO was added, and tubes were incubated at 25°C for 72 h. On the day of the assay, cysts were pelleted, washed, and treated once more with distilled water (5 h) to lyse any trophozoites that had emerged during treatment. Purified cysts were then resuspended in 75 μL cell lysis buffer (Promega) and sonicated for 2 × 10 s to break the cyst wall. The luciferase assay was performed using the Promega luciferase assay kit according to the manufacturer's instructions, using 100 μL of luciferase substrate and 35 μL of cyst lysate, keeping equal volumes, and not normalizing by protein content. Obatoclax was used as a positive control (23), and 0.2% DMSO was used as a negative control. Each compound was tested in at least two biological replicates. The effect of the drug was calculated by comparison to the DMSO control, after subtraction of background signal. Percent values of replicates were exported to GraphPad Prism software 8.0 and calculated as the mean ± standard error. Significance was calculated based on t test using values obtained with DMSO treatment as a reference for comparison.
ACKNOWLEDGMENTS
We thank David E. Solow-Cordero and Jason Wu from the Stanford High-Throughput Biosciences Center and Kevin Grimes, Steve Schow, Toni Kline, and other advisers and staff members from the Stanford SPARK. This study was mentored and financially supported by Stanford’s SPARK Translational Research Program. We are indebted to all members of the Singh lab for input and especially to Gretchen Ehrenkaufer for detailed protocols and in-depth discussions. We also thank Case McNamara and Mitchell Hull from Calibr at Scripps Research for compound supply from the ReFRAME library, data processing assistance, and data interpretation.
The work was supported by grants from the Stanford Department of Medicine, including a TRAM grant to M.M., a grant from Stanford SPARK to U.S., a Weston Havens Foundation grant for U.S., and a grant from the Stanford Maternal and Child Health Research Institute (MCHRI) to U.S., as well as an MCHRI Postdoctoral Support grant to M.M. and NIAID grant R21-AI146651 to U.S.
Footnotes
Supplemental material is available online only.
REFERENCES
- 1.Walsh JA. 1986. Problems in recognition and diagnosis of amebiasis: estimation of the global magnitude of morbidity and mortality. Rev Infect Dis 8:228–238. 10.1093/clinids/8.2.228. [DOI] [PubMed] [Google Scholar]
- 2.WHO. 1997. Entamoeba taxonomy. Bull World Health Organ 75:291–294. [PMC free article] [PubMed] [Google Scholar]
- 3.Shirley DT, Farr L, Watanabe K, Moonah S. 2018. A review of the global burden, new diagnostics, and current therapeutics for amebiasis. Open Forum Infect Dis 5:ofy161. 10.1093/ofid/ofy161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, Adair T, Aggarwal R, Ahn SY, Alvarado M, Anderson HR, Anderson LM, Andrews KG, Atkinson C, Baddour LM, Barker-Collo S, Bartels DH, Bell ML, Benjamin EJ, Bennett D, Bhalla K, Bikbov B, Bin Abdulhak A, Birbeck G, Blyth F, Bolliger I, Boufous S, Bucello C, Burch M, Burney P, Carapetis J, Chen H, Chou D, Chugh SS, Coffeng LE, Colan SD, Colquhoun S, Colson KE, Condon J, Connor MD, Cooper LT, Corriere M, Cortinovis M, de Vaccaro KC, Couser W, Cowie BC, Criqui MH, Cross M, Dabhadkar KC, et al. 2012. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380:2095–2128. 10.1016/S0140-6736(12)61728-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Steffen R, Hill DR, DuPont HL. 2015. Traveler's diarrhea: a clinical review. JAMA 313:71–80. 10.1001/jama.2014.17006. [DOI] [PubMed] [Google Scholar]
- 6.Swaminathan A, Torresi J, Schlagenhauf P, Thursky K, Wilder-Smith A, Connor BA, Schwartz E, Vonsonnenberg F, Keystone J, O'Brien DP, GeoSentinel Network. 2009. A global study of pathogens and host risk factors associated with infectious gastrointestinal disease in returned international travellers. J Infect 59:19–27. 10.1016/j.jinf.2009.05.008. [DOI] [PubMed] [Google Scholar]
- 7.Olson S, Hall A, Riddle MS, Porter CK. 2019. Travelers' diarrhea: update on the incidence, etiology and risk in military and similar populations—1990–2005 versus 2005–2015, does a decade make a difference? Trop Dis Travel Med Vaccines 5:1. 10.1186/s40794-018-0077-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Shirley DT, Watanabe K, Moonah S. 2019. Significance of amebiasis: 10 reasons why neglecting amebiasis might come back to bite us in the gut. PLoS Negl Trop Dis 13:e0007744. 10.1371/journal.pntd.0007744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Haque R, Huston CD, Hughes M, Houpt E, Petri WA, Jr.. 2003. Amebiasis. N Engl J Med 348:1565–1573. 10.1056/NEJMra022710. [DOI] [PubMed] [Google Scholar]
- 10.Shirley DA, Moonah S. 2016. Fulminant amebic colitis after corticosteroid therapy: a systematic review. PLoS Negl Trop Dis 10:e0004879. 10.1371/journal.pntd.0004879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Li J, Cui Z, Li X, Zhang L. 2021. Review of zoonotic amebiasis: epidemiology, clinical signs, diagnosis, treatment, prevention and control. Res Vet Sci 136:174–181. 10.1016/j.rvsc.2021.02.021. [DOI] [PubMed] [Google Scholar]
- 12.Petri WA, Haque R. 2013. Entamoeba histolytica brain abscess. Handb Clin Neurol 114:147–152. 10.1016/B978-0-444-53490-3.00009-1. [DOI] [PubMed] [Google Scholar]
- 13.Guyon C, Greve E, Hag B, Cuilleron M, Jospe R, Nourrisson C, Raberin H, Tran Manh Sung R, Balique JG, Flori P. 2013. Amebic liver abscess and late recurrence with no travel in an endemic area. Med Sante Trop 23:344–346. (In French.) 10.1684/mst.2013.0189. [DOI] [PubMed] [Google Scholar]
- 14.Escola-Verge L, Arando M, Vall M, Rovira R, Espasa M, Sulleiro E, Armengol P, Zarzuela F, Barbera MJ. 2017. Outbreak of intestinal amoebiasis among men who have sex with men, Barcelona (Spain), October 2016 and January. Euro Surveill 22:2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wassmann C, Hellberg A, Tannich E, Bruchhaus I. 1999. Metronidazole resistance in the protozoan parasite Entamoeba histolytica is associated with increased expression of iron-containing superoxide dismutase and peroxiredoxin and decreased expression of ferredoxin 1 and flavin reductase. J Biol Chem 274:26051–26056. 10.1074/jbc.274.37.26051. [DOI] [PubMed] [Google Scholar]
- 16.Morch K, Hanevik K. 2020. Giardiasis treatment: an update with a focus on refractory disease. Curr Opin Infect Dis 33:355–364. 10.1097/QCO.0000000000000668. [DOI] [PubMed] [Google Scholar]
- 17.Alessio C, Nyirjesy P. 2019. Management of resistant trichomoniasis. Curr Infect Dis Rep 21:31. 10.1007/s11908-019-0687-4. [DOI] [PubMed] [Google Scholar]
- 18.DiMasi JA, Grabowski HG, Hansen RW. 2016. Innovation in the pharmaceutical industry: new estimates of R&D costs. J Health Econ 47:20–33. 10.1016/j.jhealeco.2016.01.012. [DOI] [PubMed] [Google Scholar]
- 19.Parvathaneni V, Kulkarni NS, Muth A, Gupta V. 2019. Drug repurposing: a promising tool to accelerate the drug discovery process. Drug Discov Today 24:2076–2085. 10.1016/j.drudis.2019.06.014. [DOI] [PubMed] [Google Scholar]
- 20.Trouiller P, Olliaro P, Torreele E, Orbinski J, Laing R, Ford N. 2002. Drug development for neglected diseases: a deficient market and a public-health policy failure. Lancet 359:2188–2194. 10.1016/S0140-6736(02)09096-7. [DOI] [PubMed] [Google Scholar]
- 21.Debnath A, Parsonage D, Andrade RM, He C, Cobo ER, Hirata K, Chen S, Garcia-Rivera G, Orozco E, Martinez MB, Gunatilleke SS, Barrios AM, Arkin MR, Poole LB, McKerrow JH, Reed SL. 2012. A high-throughput drug screen for Entamoeba histolytica identifies a new lead and target. Nat Med 18:956–960. 10.1038/nm.2758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Capparelli EV, Bricker-Ford R, Rogers MJ, McKerrow JH, Reed SL. 2017. Phase I clinical trial results of auranofin, a novel antiparasitic agent. Antimicrob Agents Chemother 61:e01947-16. 10.1128/AAC.01947-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ehrenkaufer GM, Suresh S, Solow-Cordero D, Singh U. 2018. High-throughput screening of Entamoeba identifies compounds which target both life cycle stages and which are effective against metronidazole resistant parasites. Front Cell Infect Microbiol 8:276. 10.3389/fcimb.2018.00276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ghosh S, Farr L, Singh A, Leaton LA, Padalia J, Shirley DA, Sullivan D, Moonah S. 2020. COP9 signalosome is an essential and druggable parasite target that regulates protein degradation. PLoS Pathog 16:e1008952. 10.1371/journal.ppat.1008952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Shirley DA, Sharma I, Warren CA, Moonah S. 2021. Drug repurposing of the alcohol abuse medication disulfiram as an anti-parasitic agent. Front Cell Infect Microbiol 11:633194. 10.3389/fcimb.2021.633194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Janes J, Young ME, Chen E, Rogers NH, Burgstaller-Muehlbacher S, Hughes LD, Love MS, Hull MV, Kuhen KL, Woods AK, Joseph SB, Petrassi HM, McNamara CW, Tremblay MS, Su AI, Schultz PG, Chatterjee AK. 2018. The ReFRAME library as a comprehensive drug repurposing library and its application to the treatment of cryptosporidiosis. Proc Natl Acad Sci USA 115:10750–10755. 10.1073/pnas.1810137115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Slighter RG, Yarinsky A, Drobeck HP, Bailey DM. 1980. Activity of quinfamide against natural infections of Entamoeba criceti in hamsters: a new potent agent for intestinal amoebiasis. Parasitology 81:157–168. 10.1017/s0031182000055128. [DOI] [PubMed] [Google Scholar]
- 28.Yang H, Rudge DG, Koos JD, Vaidialingam B, Yang HJ, Pavletich NP. 2013. mTOR kinase structure, mechanism and regulation. Nature 497:217–223. 10.1038/nature12122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zheng Y, Jiang Y. 2015. mTOR inhibitors at a glance. Mol Cell Pharmacol 7:15–20. [PMC free article] [PubMed] [Google Scholar]
- 30.reframedb.org. 2021. reframedb: a screening library of 12,000 molecules assembled by combining three databases (Clarivate Integrity, GVK Excelra GoStar and Citeline Pharmaprojects) to facilitate drug repurposing. https://reframedb.org/#/. Accessed 15 May 2021.
- 31.Kangussu-Marcolino MM, Ehrenkaufer GM, Chen E, Debnath A, Singh U. 2019. Identification of plicamycin, TG02, panobinostat, lestaurtinib, and GDC-0084 as promising compounds for the treatment of central nervous system infections caused by the free-living amebae Naegleria, Acanthamoeba and Balamuthia. Int J Parasitol Drugs Drug Resist 11:80–94. 10.1016/j.ijpddr.2019.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Rashidi S, Mansouri R, Ali-Hassanzadeh M, Mojtahedi Z, Shafiei R, Savardashtaki A, Hamidizadeh N, Karimazar M, Nguewa P, Manzano-Roman R. 2021. The host mTOR pathway and parasitic diseases pathogenesis. Parasitol Res 120:1151–1166. 10.1007/s00436-021-07070-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kikuchi T, Koga M, Shimizu S, Miura T, Maruyama H, Kimura M. 2013. Efficacy and safety of paromomycin for treating amebiasis in Japan. Parasitol Int 62:497–501. 10.1016/j.parint.2013.07.004. [DOI] [PubMed] [Google Scholar]
- 34.Ehrenkaufer GM, Weedall GD, Williams D, Lorenzi HA, Caler E, Hall N, Singh U. 2013. The genome and transcriptome of the enteric parasite Entamoeba invadens, a model for encystation. Genome Biol 14:R77. 10.1186/gb-2013-14-7-r77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Erdahl WL, Chapman CJ, Taylor RW, Pfeiffer DR. 1994. Ca2+ transport properties of ionophores A23187, ionomycin, and 4-BrA23187 in a well defined model system. Biophys J 66:1678–1693. 10.1016/S0006-3495(94)80959-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Knapper S, Burnett AK, Littlewood T, Kell WJ, Agrawal S, Chopra R, Clark R, Levis MJ, Small D. 2006. A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy. Blood 108:3262–3270. 10.1182/blood-2006-04-015560. [DOI] [PubMed] [Google Scholar]
- 37.Michael M, Bang YJ, Park YS, Kang YK, Kim TM, Hamid O, Thornton D, Tate SC, Raddad E, Tie J. 2017. A phase 1 study of LY2874455, an oral selective pan-FGFR inhibitor, in patients with advanced cancer. Target Oncol 12:463–474. 10.1007/s11523-017-0502-9. [DOI] [PubMed] [Google Scholar]
- 38.Boccadoro M, Morgan G, Cavenagh J. 2005. Preclinical evaluation of the proteasome inhibitor bortezomib in cancer therapy. Cancer Cell Int 5:18. 10.1186/1475-2867-5-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.FDA. May 2021. US Food and Drug Administration. Orange Book: approved drug products with therapeutic equivalence evaluations. https://www.accessdata.fda.gov/Scripts/cder/ob/index.cfm. Accessed 15 May 2021.
- 40.Jain P, Kantarjian H, Jabbour E, Gonzalez GN, Borthakur G, Pemmaraju N, Daver N, Gachimova E, Ferrajoli A, Kornblau S, Ravandi F, O'Brien S, Cortes J. 2015. Ponatinib as first-line treatment for patients with chronic myeloid leukaemia in chronic phase: a phase 2 study. Lancet Haematol 2:e376-83. 10.1016/S2352-3026(15)00127-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Sauvey C, Ehrenkaufer G, Shi D, Debnath A, Abagyan R. 2021. Antineoplastic kinase inhibitors: a new class of potent anti-amoebic compounds. PLoS Negl Trop Dis 15:e0008425. 10.1371/journal.pntd.0008425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Clapper E, Wang S, Raninga PV, Di Trapani G, Tonissen KF. 2020. Cross-talk between Bcr-abl and the thioredoxin system in chronic myeloid leukaemia: implications for CML treatment. Antioxidants 9:207. 10.3390/antiox9030207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Leitsch D, Kolarich D, Wilson IB, Altmann F, Duchene M. 2007. Nitroimidazole action in Entamoeba histolytica: a central role for thioredoxin reductase. PLoS Biol 5:e211. 10.1371/journal.pbio.0050211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Shaulov Y, Nagaraja S, Sarid L, Trebicz-Geffen M, Ankri S. 2020. Formation of oxidised (OX) proteins in Entamoeba histolytica exposed to auranofin and consequences on the parasite virulence. Cell Microbiol 22:e13174. 10.1111/cmi.13174. [DOI] [PubMed] [Google Scholar]
- 45.Rice CA, Colon BL, Chen E, Hull MV, Kyle DE. 2020. Discovery of repurposing drug candidates for the treatment of diseases caused by pathogenic free-living amoebae. PLoS Negl Trop Dis 14:e0008353. 10.1371/journal.pntd.0008353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.McCowen MC, Callender ME, Lawlis JF, Jr.. 1951. Fumagillin (H-3), a new antibiotic with amebicidal properties. Science 113:202–203. 10.1126/science.113.2930.202. [DOI] [PubMed] [Google Scholar]
- 47.Burkey BF, Hoglen NC, Inskeep P, Wyman M, Hughes TE, Vath JE. 2018. Preclinical efficacy and safety of the novel antidiabetic, antiobesity MetAP2 inhibitor ZGN-1061. J Pharmacol Exp Ther 365:301–313. 10.1124/jpet.117.246272. [DOI] [PubMed] [Google Scholar]
- 48.Wise-Draper TM, Moorthy G, Salkeni MA, Karim NA, Thomas HE, Mercer CA, Beg MS, O'Gara S, Olowokure O, Fathallah H, Kozma SC, Thomas G, Rixe O, Desai P, Morris JC. 2017. A phase Ib study of the dual PI3K/mTOR inhibitor dactolisib (BEZ235) combined with everolimus in patients with advanced solid malignancies. Target Oncol 12:323–332. 10.1007/s11523-017-0482-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Munster P, Mita M, Mahipal A, Nemunaitis J, Massard C, Mikkelsen T, Cruz C, Paz-Ares L, Hidalgo M, Rathkopf D, Blumenschein G, Jr, Smith DC, Eichhorst B, Cloughesy T, Filvaroff EH, Li S, Raymon H, de Haan H, Hege K, Bendell JC. 2019. First-in-human phase I study of a dual mTOR kinase and DNA-PK inhibitor (CC-115) in advanced malignancy. Cancer Manag Res 11:10463–10476. 10.2147/CMAR.S208720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Markman B, Tabernero J, Krop I, Shapiro GI, Siu L, Chen LC, Mita M, Melendez Cuero M, Stutvoet S, Birle D, Anak O, Hackl W, Baselga J. 2012. Phase I safety, pharmacokinetic, and pharmacodynamic study of the oral phosphatidylinositol-3-kinase and mTOR inhibitor BGT226 in patients with advanced solid tumors. Ann Oncol 23:2399–2408. 10.1093/annonc/mds011. [DOI] [PubMed] [Google Scholar]
- 51.Younes A, Berdeja JG, Patel MR, Flinn I, Gerecitano JF, Neelapu SS, Kelly KR, Copeland AR, Akins A, Clancy MS, Gong L, Wang J, Ma A, Viner JL, Oki Y. 2016. Safety, tolerability, and preliminary activity of CUDC-907, a first-in-class, oral, dual inhibitor of HDAC and PI3K, in patients with relapsed or refractory lymphoma or multiple myeloma: an open-label, dose-escalation, phase 1 trial. Lancet Oncol 17:622–631. 10.1016/S1470-2045(15)00584-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Britten CD, Adjei AA, Millham R, Houk BE, Borzillo G, Pierce K, Wainberg ZA, LoRusso PM. 2014. Phase I study of PF-04691502, a small-molecule, oral, dual inhibitor of PI3K and mTOR, in patients with advanced cancer. Invest New Drugs 32:510–517. 10.1007/s10637-013-0062-5. [DOI] [PubMed] [Google Scholar]
- 53.Moore KN, Bauer TM, Falchook GS, Chowdhury S, Patel C, Neuwirth R, Enke A, Zohren F, Patel MR. 2018. Phase I study of the investigational oral mTORC1/2 inhibitor sapanisertib (TAK-228): tolerability and food effects of a milled formulation in patients with advanced solid tumours. ESMO Open 3:e000291. 10.1136/esmoopen-2017-000291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lukey PT, Harrison SA, Yang S, Man Y, Holman BF, Rashidnasab A, Azzopardi G, Grayer M, Simpson JK, Bareille P, Paul L, Woodcock HV, Toshner R, Saunders P, Molyneaux PL, Thielemans K, Wilson FJ, Mercer PF, Chambers RC, Groves AM, Fahy WA, Marshall RP, Maher TM. 2019. A randomised, placebo-controlled study of omipalisib (PI3K/mTOR) in idiopathic pulmonary fibrosis. Eur Respir J 53:1801992. 10.1183/13993003.01992-2018. [DOI] [PubMed] [Google Scholar]
- 55.Chan O, Talati C, Isenalumhe L, Shams S, Nodzon L, Fradley M, Sweet K, Pinilla-Ibarz J. 2020. Side-effects profile and outcomes of ponatinib in the treatment of chronic myeloid leukemia. Blood Adv 4:530–538. 10.1182/bloodadvances.2019000268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Hexner EO, Mascarenhas J, Prchal J, Roboz GJ, Baer MR, Ritchie EK, Leibowitz D, Demakos EP, Miller C, Siuty J, Kleczko J, Price L, Jeschke G, Weinberg R, Basu T, Pahl HL, Orazi A, Najfeld V, Marchioli R, Goldberg JD, Silverman LR, Hoffman R. 2015. Phase I dose escalation study of lestaurtinib in patients with myelofibrosis. Leuk Lymphoma 56:2543–2551. 10.3109/10428194.2014.1001986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Diamond LS, Harlow DR, Cunnick CC. 1978. A new medium for the axenic cultivation of Entamoeba histolytica and other Entamoeba. Trans R Soc Trop Med Hyg 72:431–432. 10.1016/0035-9203(78)90144-x. [DOI] [PubMed] [Google Scholar]
- 58.Morf L, Pearson RJ, Wang AS, Singh U. 2013. Robust gene silencing mediated by antisense small RNAs in the pathogenic protist Entamoeba histolytica. Nucleic Acids Res 41:9424–9437. 10.1093/nar/gkt717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ehrenkaufer GM, Singh U. 2012. Transient and stable transfection in the protozoan parasite Entamoeba invadens. Mol Biochem Parasitol 184:59–62. 10.1016/j.molbiopara.2012.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.ClinicalTrials.gov. 2019. ClinicalTrials.gov is a database of privately and publicly funded clinical studies conducted around the world. https://clinicaltrials.gov. Accessed 22 March 2019.
- 61.FDA. 2019. Orphan drug listing US Food and Drug Administration. Search Orphan Drug Designations and Approvals. https://www.accessdata.fda.gov/scripts/opdlisting/oopd/. Accessed 22 March 2019.
- 62.Barlaam B, Cosulich S, Delouvrie B, Ellston R, Fitzek M, Germain H, Green S, Hancox U, Harris CS, Hudson K, Lambert-van der Brempt C, Lebraud H, Magnien F, Lamorlette M, Le Griffon A, Morgentin R, Ouvry G, Page K, Pasquet G, Polanska U, Ruston L, Saleh T, Vautier M, Ward L. 2015. Discovery of 1-(4-(5–(5-amino-6-(5-tert-butyl-1,3,4-oxadiazol-2-yl)pyrazin-2-yl)-1-ethyl-1,2,4-triazol-3-yl)piperidin-1-yl)-3-hydroxypropan-1-one (AZD8835): a potent and selective inhibitor of PI3Kalpha and PI3Kdelta for the treatment of cancers. Bioorg Med Chem Lett 25:5155–5162. 10.1016/j.bmcl.2015.10.002. [DOI] [PubMed] [Google Scholar]
- 63.Beaufils F, Cmiljanovic N, Cmiljanovic V, Bohnacker T, Melone A, Marone R, Jackson E, Zhang X, Sele A, Borsari C, Mestan J, Hebeisen P, Hillmann P, Giese B, Zvelebil M, Fabbro D, Williams RL, Rageot D, Wymann MP. 2017. 5-(4,6-Dimorpholino-1,3,5-triazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (PQR309), a potent, brain-penetrant, orally bioavailable, pan-class I PI3K/mTOR inhibitor as clinical candidate in oncology. J Med Chem 60:7524–7538. 10.1021/acs.jmedchem.7b00930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Edwards SR, Wandless TJ. 2007. The rapamycin-binding domain of the protein kinase mammalian target of rapamycin is a destabilizing domain. J Biol Chem 282:13395–13401. 10.1074/jbc.M700498200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Garcia-Echeverria C, Sellers WR. 2008. Drug discovery approaches targeting the PI3K/Akt pathway in cancer. Oncogene 27:5511–5526. 10.1038/onc.2008.246. [DOI] [PubMed] [Google Scholar]
- 66.Burris HA, III, Kurkjian CD, Hart L, Pant S, Murphy PB, Jones SF, Neuwirth R, Patel CG, Zohren F, Infante JR. 2017. TAK-228 (formerly MLN0128), an investigational dual TORC1/2 inhibitor plus paclitaxel, with/without trastuzumab, in patients with advanced solid malignancies. Cancer Chemother Pharmacol 80:261–273. 10.1007/s00280-017-3343-4. [DOI] [PubMed] [Google Scholar]
- 67.Heffron TP, Ndubaku CO, Salphati L, Alicke B, Cheong J, Drobnick J, Edgar K, Gould SE, Lee LB, Lesnick JD, Lewis C, Nonomiya J, Pang J, Plise EG, Sideris S, Wallin J, Wang L, Zhang X, Olivero AG. 2016. Discovery of clinical development candidate GDC-0084, a brain penetrant inhibitor of PI3K and mTOR. ACS Med Chem Lett 7:351–356. 10.1021/acsmedchemlett.6b00005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hsieh AC, Liu Y, Edlind MP, Ingolia NT, Janes MR, Sher A, Shi EY, Stumpf CR, Christensen C, Bonham MJ, Wang S, Ren P, Martin M, Jessen K, Feldman ME, Weissman JS, Shokat KM, Rommel C, Ruggero D. 2012. The translational landscape of mTOR signalling steers cancer initiation and metastasis. Nature 485:55–61. 10.1038/nature10912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Knight SD, Adams ND, Burgess JL, Chaudhari AM, Darcy MG, Donatelli CA, Luengo JI, Newlander KA, Parrish CA, Ridgers LH, Sarpong MA, Schmidt SJ, Van Aller GS, Carson JD, Diamond MA, Elkins PA, Gardiner CM, Garver E, Gilbert SA, Gontarek RR, Jackson JR, Kershner KL, Luo L, Raha K, Sherk CS, Sung CM, Sutton D, Tummino PJ, Wegrzyn RJ, Auger KR, Dhanak D. 2010. Discovery of GSK2126458, a highly potent inhibitor of PI3K and the mammalian target of rapamycin. ACS Med Chem Lett 1:39–43. 10.1021/ml900028r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Maira SM, Stauffer F, Brueggen J, Furet P, Schnell C, Fritsch C, Brachmann S, Chene P, De Pover A, Schoemaker K, Fabbro D, Gabriel D, Simonen M, Murphy L, Finan P, Sellers W, Garcia-Echeverria C. 2008. Identification and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity. Mol Cancer Ther 7:1851–1863. 10.1158/1535-7163.MCT-08-0017. [DOI] [PubMed] [Google Scholar]
- 71.Mateo J, Ganji G, Lemech C, Burris HA, Han SW, Swales K, Decordova S, DeYoung MP, Smith DA, Kalyana-Sundaram S, Wu J, Motwani M, Kumar R, Tolson JM, Rha SY, Chung HC, Eder JP, Sharma S, Bang YJ, Infante JR, Yan L, de Bono JS, Arkenau HT. 2017. A first-time-in-human study of GSK2636771, a phosphoinositide 3 kinase beta-selective inhibitor, in patients with advanced solid tumors. Clin Cancer Res 23:5981–5992. 10.1158/1078-0432.CCR-17-0725. [DOI] [PubMed] [Google Scholar]
- 72.Mortensen DS, Perrin-Ninkovic SM, Shevlin G, Elsner J, Zhao J, Whitefield B, Tehrani L, Sapienza J, Riggs JR, Parnes JS, Papa P, Packard G, Lee BG, Harris R, Correa M, Bahmanyar S, Richardson SJ, Peng SX, Leisten J, Khambatta G, Hickman M, Gamez JC, Bisonette RR, Apuy J, Cathers BE, Canan SS, Moghaddam MF, Raymon HK, Worland P, Narla RK, Fultz KE, Sankar S. 2015. Optimization of a series of triazole containing mammalian target of rapamycin (mTOR) kinase inhibitors and the discovery of CC-115. J Med Chem 58:5599–5608. 10.1021/acs.jmedchem.5b00627. [DOI] [PubMed] [Google Scholar]
- 73.Ndubaku CO, Heffron TP, Staben ST, Baumgardner M, Blaquiere N, Bradley E, Bull R, Do S, Dotson J, Dudley D, Edgar KA, Friedman LS, Goldsmith R, Heald RA, Kolesnikov A, Lee L, Lewis C, Nannini M, Nonomiya J, Pang J, Price S, Prior WW, Salphati L, Sideris S, Wallin JJ, Wang L, Wei B, Sampath D, Olivero AG. 2013. Discovery of 2-{3-[2-(1-isopropyl-3-methyl-1H-1,2–4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1, 2-d][1,4]oxazepin-9-yl]-1H-pyrazol-1-yl}-2-methylpropanamide (GDC-0032): a beta-sparing phosphoinositide 3-kinase inhibitor with high unbound exposure and robust in vivo antitumor activity. J Med Chem 56:4597–4610. 10.1021/jm4003632. [DOI] [PubMed] [Google Scholar]
- 74.Qian C, Lai CJ, Bao R, Wang DG, Wang J, Xu GX, Atoyan R, Qu H, Yin L, Samson M, Zifcak B, Ma AW, DellaRocca S, Borek M, Zhai HX, Cai X, Voi M. 2012. Cancer network disruption by a single molecule inhibitor targeting both histone deacetylase activity and phosphatidylinositol 3-kinase signaling. Clin Cancer Res 18:4104–4113. 10.1158/1078-0432.CCR-12-0055. [DOI] [PubMed] [Google Scholar]
- 75.Sarkaria JN, Tibbetts RS, Busby EC, Kennedy AP, Hill DE, Abraham RT. 1998. Inhibition of phosphoinositide 3-kinase related kinases by the radiosensitizing agent wortmannin. Cancer Res 58:4375–4382. [PubMed] [Google Scholar]
- 76.Sedrani R, Cottens S, Kallen J, Schuler W. 1998. Chemical modification of rapamycin: the discovery of SDZ RAD. Transplant Proc 30:2192–2194. 10.1016/s0041-1345(98)00587-9. [DOI] [PubMed] [Google Scholar]
- 77.Shor B, Zhang WG, Toral-Barza L, Lucas J, Abraham RT, Gibbons JJ, Yu K. 2008. A new pharmacologic action of CCI-779 involves FKBP12-independent inhibition of mTOR kinase activity and profound repression of global protein synthesis. Cancer Res 68:2934–2943. 10.1158/0008-5472.CAN-07-6487. [DOI] [PubMed] [Google Scholar]
- 78.Sun SY. 2013. mTOR kinase inhibitors as potential cancer therapeutic drugs. Cancer Lett 340:1–8. 10.1016/j.canlet.2013.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Sutherlin DP, Bao L, Berry M, Castanedo G, Chuckowree I, Dotson J, Folks A, Friedman L, Goldsmith R, Gunzner J, Heffron T, Lesnick J, Lewis C, Mathieu S, Murray J, Nonomiya J, Pang J, Pegg N, Prior WW, Rouge L, Salphati L, Sampath D, Tian Q, Tsui V, Wan NC, Wang S, Wei B, Wiesmann C, Wu P, Zhu BY, Olivero A. 2011. Discovery of a potent, selective, and orally available class I phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) kinase inhibitor (GDC-0980) for the treatment of cancer. J Med Chem 54:7579–7587. 10.1021/jm2009327. [DOI] [PubMed] [Google Scholar]
- 80.Venkatesan AM, Dehnhardt CM, Delos Santos E, Chen Z, Dos Santos O, Ayral-Kaloustian S, Khafizova G, Brooijmans N, Mallon R, Hollander I, Feldberg L, Lucas J, Yu K, Gibbons J, Abraham RT, Chaudhary I, Mansour TS. 2010. Bis(morpholino-1,3,5-triazine) derivatives: potent adenosine 5'-triphosphate competitive phosphatidylinositol-3-kinase/mammalian target of rapamycin inhibitors: discovery of compound 26 (PKI-587), a highly efficacious dual inhibitor. J Med Chem 53:2636–2645. 10.1021/jm901830p. [DOI] [PubMed] [Google Scholar]
- 81.Yano H, Nakanishi S, Kimura K, Hanai N, Saitoh Y, Fukui Y, Nonomura Y, Matsuda Y. 1993. Inhibition of histamine secretion by wortmannin through the blockade of phosphatidylinositol 3-kinase in RBL-2H3 cells. J Biol Chem 268:25846–25856. 10.1016/S0021-9258(19)74466-4. [DOI] [PubMed] [Google Scholar]
- 82.Yuan J, Mehta PP, Yin MJ, Sun S, Zou A, Chen J, Rafidi K, Feng Z, Nickel J, Engebretsen J, Hallin J, Blasina A, Zhang E, Nguyen L, Sun M, Vogt PK, McHarg A, Cheng H, Christensen JG, Kan JL, Bagrodia S. 2011. PF-04691502, a potent and selective oral inhibitor of PI3K and mTOR kinases with antitumor activity. Mol Cancer Ther 10:2189–2199. 10.1158/1535-7163.MCT-11-0185. [DOI] [PubMed] [Google Scholar]
- 83.O'Hare T, Shakespeare WC, Zhu X, Eide CA, Rivera VM, Wang F, Adrian LT, Zhou T, Huang WS, Xu Q, Metcalf CA, III, Tyner JW, Loriaux MM, Corbin AS, Wardwell S, Ning Y, Keats JA, Wang Y, Sundaramoorthi R, Thomas M, Zhou D, Snodgrass J, Commodore L, Sawyer TK, Dalgarno DC, Deininger MW, Druker BJ, Clackson T. 2009. AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell 16:401–412. 10.1016/j.ccr.2009.09.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.O'Hare T, Walters DK, Stoffregen EP, Jia T, Manley PW, Mestan J, Cowan-Jacob SW, Lee FY, Heinrich MC, Deininger MW, Druker BJ. 2005. In vitro activity of Bcr-Abl inhibitors AMN107 and BMS-354825 against clinically relevant imatinib-resistant Abl kinase domain mutants. Cancer Res 65:4500–4505. 10.1158/0008-5472.CAN-05-0259. [DOI] [PubMed] [Google Scholar]
- 85.Cheetham GM, Charlton PA, Golec JM, Pollard JR. 2007. Structural basis for potent inhibition of the Aurora kinases and a T315I multi-drug resistant mutant form of Abl kinase by VX-680. Cancer Lett 251:323–329. 10.1016/j.canlet.2006.12.004. [DOI] [PubMed] [Google Scholar]
- 86.Chan WW, Wise SC, Kaufman MD, Ahn YM, Ensinger CL, Haack T, Hood MM, Jones J, Lord JW, Lu WP, Miller D, Patt WC, Smith BD, Petillo PA, Rutkoski TJ, Telikepalli H, Vogeti L, Yao T, Chun L, Clark R, Evangelista P, Gavrilescu LC, Lazarides K, Zaleskas VM, Stewart LJ, Van Etten RA, Flynn DL. 2011. Conformational control inhibition of the BCR-ABL1 tyrosine kinase, including the gatekeeper T315I mutant, by the switch-control inhibitor DCC-2036. Cancer Cell 19:556–568. 10.1016/j.ccr.2011.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Buchdunger E, Zimmermann J, Mett H, Meyer T, Muller M, Regenass U, Lydon NB. 1995. Selective inhibition of the platelet-derived growth factor signal transduction pathway by a protein-tyrosine kinase inhibitor of the 2-phenylaminopyrimidine class. Proc Natl Acad Sci USA 92:2558–2562. 10.1073/pnas.92.7.2558. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 88.Weisberg E, Manley PW, Breitenstein W, Bruggen J, Cowan-Jacob SW, Ray A, Huntly B, Fabbro D, Fendrich G, Hall-Meyers E, Kung AL, Mestan J, Daley GQ, Callahan L, Catley L, Cavazza C, Azam M, Neuberg D, Wright RD, Gilliland DG, Griffin JD. 2005. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell 7:129–141. 10.1016/j.ccr.2005.01.007. [DOI] [PubMed] [Google Scholar]
- 89.Kimura S, Naito H, Segawa H, Kuroda J, Yuasa T, Sato K, Yokota A, Kamitsuji Y, Kawata E, Ashihara E, Nakaya Y, Naruoka H, Wakayama T, Nasu K, Asaki T, Niwa T, Hirabayashi K, Maekawa T. 2005. NS-187, a potent and selective dual Bcr-Abl/Lyn tyrosine kinase inhibitor, is a novel agent for imatinib-resistant leukemia. Blood 106:3948–3954. 10.1182/blood-2005-06-2209. [DOI] [PubMed] [Google Scholar]
- 90.Carpinelli P, Ceruti R, Giorgini ML, Cappella P, Gianellini L, Croci V, Degrassi A, Texido G, Rocchetti M, Vianello P, Rusconi L, Storici P, Zugnoni P, Arrigoni C, Soncini C, Alli C, Patton V, Marsiglio A, Ballinari D, Pesenti E, Fancelli D, Moll J. 2007. PHA-739358, a potent inhibitor of Aurora kinases with a selective target inhibition profile relevant to cancer. Mol Cancer Ther 6:3158–3168. 10.1158/1535-7163.MCT-07-0444. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.