Abstract
Histone acetylation plays an important role in regulating gene transcription and silencing in Plasmodium falciparum. Histone deacetylase (HDAC) inhibitors, particularly those of the hydroxamate class, have been shown to have potent in vitro activity against drug-resistant and -sensitive laboratory strains of P. falciparum, raising their potential as a new class of antimalarial compounds. In the current study, stage-specific ex vivo susceptibility profiles of representative hydroxamate-based HDAC inhibitors suberoylanilide hydroxamic acid (SAHA), 2-ASA-9, and 2-ASA-14 (2-ASA-9 and 2-ASA-14 are 2-aminosuberic acid-based HDAC inhibitors) were assessed in multidrug-resistant clinical isolates of P. falciparum (n = 24) and P. vivax (n = 25) from Papua, Indonesia, using a modified schizont maturation assay. Submicromolar concentrations of SAHA, 2-ASA-9, and 2-ASA-14 inhibited the growth of both P. falciparum (median 50% inhibitory concentrations [IC50s] of 310, 533, and 266 nM) and P. vivax (median IC50s of 170, 503, and 278 nM). Inverse correlation patterns between HDAC inhibitors and chloroquine for P. falciparum and mefloquine for P. vivax indicate species-specific susceptibility profiles for HDAC inhibitors. These HDAC inhibitors were also found to be potent ex vivo against P. vivax schizont maturation, comparable to that in P. falciparum, suggesting that HDAC inhibitors may be promising candidates for antimalarial therapy in geographical locations where both species are endemic. Further studies optimizing the selectivity and in vivo efficacy of HDAC inhibitors in Plasmodium spp. and defining drug interaction with common antimalarial compounds are warranted to investigate the role of HDAC inhibitors in antimalarial therapy.
Malaria is a major public health issue in more than 100 countries with 40% of the world's population at risk of acquiring the disease (21). Early diagnosis, followed by prompt and effective treatment, remains a cornerstone of malaria control programs (47). However, the effectiveness of this strategy is compromised by the emergence and spread of drug-resistant malaria. Most antimalarial drug research has focused on the treatment of Plasmodium falciparum, the dominant species in sub-Saharan Africa. However, outside Africa, P. falciparum and Plasmodium vivax are sympatric, with the latter species often accounting for half of malaria episodes. In Papua, Indonesia, P. vivax has become highly resistant to chloroquine and sulfadoxine-pyrimethamine (36, 41), with evidence now emerging of declining chloroquine efficacy throughout most areas where vivax malaria is endemic (11). Reports from the island of New Guinea, where chloroquine resistance is highly prevalent, have demonstrated an association between vivax malaria and severe disease and death (5, 17, 35, 45). In this and other regions where drug resistance has emerged, new treatment strategies that focus on different plasmodial targets are urgently needed for both P. falciparum and P. vivax.
In eukaryotes, histone deacetylase (HDAC) and histone acetyltransferase (HAT) enzymes control the acetylation of both histones and nonhistone proteins and are thus important for a variety of vital cellular functions, such as transcription, DNA replication and repair, cell signaling, as well as cell cycle regulation and differentiation (49, 50). HDACs have been recognized as therapeutic targets in cancer cells for over a decade, with one hydroxamate-based HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) (Vorinostat), approved for the treatment of persistent or refractory T-cell lymphoma (18). More recently, HDACs have been identified as important regulators of transcription in P. falciparum (7, 14, 16, 22). HDAC inhibitors have been shown to have activity against apicomplexan parasites, raising their potential as a new class of antimalarial compounds (3, 23, 33).
The principal aims of this study were to investigate the ex vivo susceptibility profiles of antimalarial HDAC inhibitors against multidrug-resistant clinical isolates of P. falciparum and P. vivax in Papua, Indonesia, and to examine the stage-specific actions of these compounds. The three HDAC inhibitors compared in this study were the clinically approved antitumor compound SAHA, which has in vitro 50% inhibitory concentrations (IC50s) against P. falciparum laboratory lines of ∼100 to 300 nM (12), and two other structurally related hydroxamate-based compounds with greater in vitro activity against laboratory lines (2-ASA-9 with an IC50 of 15 to 39 nM and 2-ASA-14 with an IC50 of 13 to 33 nM [2-ASA-9 and 2-ASA-14 are 2-aminosuberic acid-based HDAC inhibitors]; Fig. 1). These compounds contain a hydroxamic acid zinc binding group and are currently the most promising class of HDAC inhibitors under investigation for use against malaria.
FIG. 1.
Chemical structures of suberoylanilide hydroxamic acid (SAHA) (WR308364) and 2-aminosuberic acid-based HDAC inhibitors, 2-ASA-9 and 2-ASA-14.
MATERIALS AND METHODS
Compounds.
The synthesis, purification, and characterization of 2-aminosuberic acid-based HDAC inhibitors (2-ASA-9 and 2-ASA-14; Fig. 1) have been previously described (24). Suberoylanilide hydroxamic acid (SAHA) was purchased from Sigma. All compounds were prepared as 10 mM stock solutions in dimethyl sulfoxide (DMSO). Drug plates were then predosed by diluting the compounds in 50% ethanol followed by lyophilization and storage at 4°C.
Field location and sample collection.
Between April and September 2008, Plasmodium isolates were collected from patients attending malaria clinics in Timika, Papua province, Indonesia, a region where multidrug-resistant strains of both P. vivax and P. falciparum are endemic (25, 36, 38). Patients with symptomatic malaria presenting to an outpatient facility were recruited into the study if they presented with single-species infections of either P. falciparum or P. vivax. Parasite counts were determined on Giemsa-stained thick films as the number of parasites per 200 white blood cells (WBC), and peripheral parasitemia was calculated assuming a white cell count of 7,300 μl−1. Only isolates with a parasitemia of between 2,000 μl−1 and 80,000 μl−1 were selected for ex vivo analysis. Since previous studies have highlighted a major difference in the stage-specific activities of chloroquine and amodiaquine, isolates of P. vivax were processed only if the majority of asexual forms were at the ring stage (38). Patients treated with antimalarial drugs in the month prior to sampling were excluded from the study. Venous blood (5 ml) was collected by venipuncture, and after removal of host white blood cells using CF 11 cellulose (37), packed infected red blood cells (IRBC) were used for the ex vivo drug susceptibility assay.
Ex vivo drug susceptibility assay.
Drug susceptibility of P. vivax and P. falciparum isolates was measured using a protocol modified from the WHO microtest as described previously (38, 39). Two hundred microliters of a 2% hematocrit blood medium mixture (BMM), consisting of RPMI 1640 medium plus 10% AB+ human serum (P. falciparum) or McCoy's 5A medium plus 20% AB+ human serum (P. vivax) was added to each well of predosed drug plates containing 11 serial concentrations (2-fold dilutions) of the following antimalarial drugs (maximum concentration shown in parentheses): chloroquine (2,992 nM), amodiaquine (80 nM), piperaquine (769 nM), mefloquine (338 nM), and artesunate (25 nM) and the three HDAC inhibitors SAHA (2,000 nM), 2-ASA-9 (1,000 nM), and 2-ASA-14 (1,000 nM). A candle jar was used to mature the parasites at 37.5°C for 24 to 56 h. Incubation was stopped when >40% of ring-stage parasites had reached the mature schizont stage in the drug-free control.
Thick blood films made from each well were stained with 5% Giemsa solution for 30 min and examined microscopically. Differential counts of 200 asexual parasites in both the preincubation and test slides were classified into ring stage, mature trophozoites, and schizonts. To ensure optimal maturity, ease of parasite identification, and reduction of parasite classification error between microscopists, only schizonts with at least five well-defined chromatin dots were classified as schizonts at harvest. Free merozoites and gametocytes were not included in the count.
The number of schizonts per 200 asexual-stage parasites was determined for each drug concentration and normalized to that of the control well. The dose-response data were analyzed using nonlinear regression analysis (WinNonLin 4.1; Pharsight Corporation), and the IC50s were derived using an inhibitory sigmoid Emax model. IC50 ex vivo data were used only from predicted curves where the maximum effect attributable to the drug (Emax) and the basal effect, corresponding to the response when the dose of the drug is zero (E0) were within 15% of 100 or 0, respectively.
Data analysis.
Data analysis was performed using STATA (version 10.1; Stata Corp., College Station, TX) and GraphPad Prism (version 5) software. The Mann-Whitney U test, Wilcoxon signed-rank test, and Spearman rank correlation were used for nonparametric comparisons and correlations. In order to examine the robustness of our observations, we used restricted and stratified analysis to control for three major confounding factors: (i) the percentage of ring stages at the start of the assay, (ii) the duration of the assay, and (iii) the initial parasitemia. Robust regression analysis was performed on log-transformed data.
Ethical approval.
Ethical approval for this study was obtained from the ethics committees of the National Institute of Health Research and Development, Ministry of Health, Indonesia, and the Menzies School of Health Research, Casuarina, Darwin, Australia.
RESULTS
Between April and September 2008, ex vivo susceptibility of the HDAC inhibitors SAHA, 2-ASA-9, and 2-ASA-14 was tested in clinical isolates from 49 patients presenting with single-species infections of either P. vivax (n = 25) or P. falciparum (n = 24). Susceptibility profiles on the same isolates were also tested against chloroquine, amodiaquine, piperaquine, mefloquine, and artesunate. Adequate growth for harvest (i.e., maturation from the ring or trophozoite stage to >40% at the schizont stage) was successfully achieved in 83% (20/24) of P. falciparum isolates and 80% (20/25) of P. vivax isolates. Baseline characteristics of the isolates processed are presented in Table 1, and the geometric mean IC50s for the two species, in comparison with published IC50s for laboratory lines, are shown in Table 2 and Fig. 2. A difference in drug susceptibility in the two species was observed for amodiaquine, which showed a higher median IC50 (9.1 versus 5.0 nM) for P. vivax (P = 0.005), and SAHA, which had a higher median IC50 for P. falciparum (301 versus 170 nM; P < 0.001).
TABLE 1.
Baseline characteristics of isolates for which ex vivo assay was accomplished
Baseline characteristic | Value for species: |
|
---|---|---|
P. falciparum | P. vivax | |
Total no. of isolates reaching harvest | 20 | 20 |
Median (range) delay from venipuncture to start of culture (h) | 2.4 (1.6-4.9) | 2.5 (1.6-3.7) |
Median (range) duration of assay (h) | 46 (41-50) | 48 (43-53) |
Geometric mean (95% CI)a parasitemia (no. of asexual parasites/μl) | 22,622 (11,900-43,001) | 11,675 (7,527-18,110) |
Median initial % (range) of parasites at ring stage | 100b | 94 (54-100) |
Mean (95% CI) schizont count at harvest | 46 (39-52) | 40 (35-44) |
95% CI, 95% confidence interval.
No range given (all values were 100%).
TABLE 2.
Overall ex vivo drug susceptibility for each drug according to the species tested
Antimalarial | Mean IC50 (nM) for P. falciparum lab linesa |
P. falciparum |
P. vivax |
|||
---|---|---|---|---|---|---|
CQs | CQr | n (%)b | Median IC50(nM) (range) | n (%) | Median IC50(nM) (range) | |
Chloroquine | 12 | 240 | 20 (100) | 30.2 (6.4-97.1) | 20 (100) | 40.4 (8.8-81.4) |
Amodiaquine | 2 | 8 | 20 (100) | 5.0 (1.0-28.6) | 19 (95) | 9.1 (2.8-16.0) |
Piperaquine | 3 | 11 | 20 (100) | 10.8 (4.4-46.9) | 20 (100) | 13.2 (3.4-38.4) |
Mefloquine | 11 | 8 | 19 (95) | 6.3 (1.7-21.3) | 18 (90) | 7.7 (1.5-23.2) |
Artesunate | 2 | 1 | 20 (100) | 0.92 (0.31-4.1) | 20 (100) | 0.74 (0.09-1.42) |
SAHA | 247 | 161 | 19 (95) | 301 (120-484) | 20 (100) | 170 (67-281) |
2-ASA-9 | 15 | 39 | 15 (75)c | 533 (199-964) | 19 (95) | 503 (203-766) |
2-ASA-14 | 13 | 33 | 18 (90)d | 266 (87-704) | 20 (100) | 278 (121-781) |
Mean IC50s (in vitro growth quantified by [3H]hypoxanthine incorporation) as previously published for chloroquine (8, 9), amodiaquine (1, 19), piperaquine (1, 30), mefloquine (1, 10), artesunate (1, 10) (the chloroquine-sensitive and -resistant lines were 3D7 and K1, respectively), SAHA (12), 2-ASA-9 (2), and 2-ASA-14 (2) (the chloroquine-sensitive and -resistant laboratory lines, respectively, were D6 and W2 for SAHA and 3D7 and Dd2 for 2-ASA-9 and 2-ASA-14). CQs, chloroquine-sensitive laboratory strain; CQr, chloroquine-resistant laboratory strain.
n, total number of assays with acceptable IC50; %, total number of assays with acceptable IC50/number of assays with adequate growth harvested.
No IC50 estimates of 2-ASA-9 for four P. falciparum isolates and one P. vivax isolate (MIC > 1,000 nM).
No IC50 estimate of 2-ASA-14 for one P. falciparum isolate (MIC > 1,000 nM).
FIG. 2.
Ex vivo sensitivity (IC50) of clinical field isolates of P. falciparum (closed circles) and P. vivax (open circles) from Papua, Indonesia, to SAHA, 2-ASA-9, and 2-ASA-14. Each symbol represents the IC50 for one patient. Each horizontal bar represents the median IC50 for the group of patients.
Stage-specific antimalarial activity.
To investigate the stage-specific drug susceptibility, isolates with greater than 90% rings were set up in culture in the presence of drug directly and again after ex vivo growth to 90% trophozoites in the absence of drug. For P. falciparum, isolates added to the assay at the trophozoite stage had significantly higher median IC50s for SAHA (520 nM versus 329 nM) and 2-ASA-14 (467 nM versus 238 nM) than the same isolates exposed to drug at the ring stage (P = 0.036 and P = 0.028, respectively; Table 3). The derived IC50s were also significantly higher for chloroquine, amodiaquine, piperaquine, and mefloquine, but not for artesunate. For P. vivax, a stage-specific difference in drug response was observed only for chloroquine (median IC50 of 841 nM versus 53 nM; P = 0.008), and a less-significant difference was observed for amodiaquine (median IC50 of 21.2 nM versus 10.1 nM; P = 0.036; Table 3).
TABLE 3.
Ex vivo drug susceptibility for paired isolates tested at the ring (>90% before culture) and trophozoite (>90% after culture) stage
Antimalarial |
P. falciparumassay |
P. vivaxassay |
||||||
---|---|---|---|---|---|---|---|---|
na | Median IC50(range) (nM) for: |
Pb | n | Median IC50(range) (nM) for: |
P | |||
Rings | Trophozoites | Rings | Trophozoites | |||||
Chloroquine | 8 | 30.2 (14.0-97.1) | 63.2 (21.0-673.3) | 0.036 | 9 | 53.0 (8.8-81.4) | 841.4 (133-1930) | 0.008 |
Amodiaquine | 8 | 5.7 (1.0-28.6) | 9.6 (4.5-40.5) | 0.025 | 8 | 10.1 (3.9-15.5) | 21.1 (4.0-36.3) | 0.036 |
Piperaquine | 8 | 11.5 (4.4-46.9) | 87.6 (30.4-692.7) | 0.012 | 9 | 15.5 (3.7-34.8) | 15.9 (1.32-42.5) | 0.515 |
Mefloquine | 6 | 6.4 (2.7-12.7) | 19.9 (6.4-22.5) | 0.046 | 7 | 9.2 (1.5-23.2) | 10.2 (2.3-27.2) | 0.612 |
Artesunate | 8 | 0.88 (0.31-4.11) | 1.19 (0.59-4.18) | 0.400 | 8 | 0.75 (0.09-1.03) | 0.39 (0.2-1.09) | 0.484 |
SAHA | 8 | 329 (232-482) | 520 (346-1016) | 0.036 | 9 | 159 (67-223) | 147 (40-229) | 0.767 |
2-ASA-9 | 4 | 492 (463-533) | 519 (362-988) | 1.000 | 9 | 545 (203-710) | 438 (149-2491) | 0.767 |
2-ASA-14 | 6 | 238 (209-704) | 467 (273-1043) | 0.028 | 9 | 327 (121-502) | 214 (145-588) | 0.110 |
n is the number of paired isolates.
The Pvalues compare the median IC50 values for rings and trophozoites by the Wilcoxon rank sum test. The statistically significant values are shown in boldface type.
Correlation in antimalarial susceptibility.
The drug responses to the different HDAC inhibitors were positively correlated in both P. falciparum and P. vivax isolates (Spearman rank correlation coefficients [rs] of 0.515 to 0.818; Table 4). In P. falciparum isolates, inhibition by mefloquine (IC50) correlated positively with SAHA (rs = 0.635 and P = 0.004), but not 2-ASA-9 or 2-ASA-14. Conversely, in P. vivax, 2-ASA-9 and 2-ASA-14 (IC50), but not SAHA, positively correlated with mefloquine (rs = 0.527 and P = 0.030 and rs = 0.610 and P = 0.007, respectively). In addition, P. vivax susceptibility to 2-ASA-14 was positively correlated with chloroquine (rs = 0.490 and P = 0.028).
TABLE 4.
Correlation coefficients (rs) for ex vivo antimalarial susceptibilities in P. falciparum and P. vivax
Antimalarial combination | All P. falciparumisolates |
All P. vivaxisolates |
||||
---|---|---|---|---|---|---|
Correlationa | Pb | dfc | Correlation | P | df | |
SAHA-chloroquine | 0.367 | 0.123 | 18 | −0.012 | 0.960 | 19 |
SAHA-amodiaquine | 0.167 | 0.495 | 18 | 0.363 | 0.127 | 18 |
SAHA-piperaquine | 0.158 | 0.519 | 18 | 0.239 | 0.310 | 19 |
SAHA-mefloquine | 0.635 | 0.004 | 18 | 0.191 | 0.448 | 17 |
SAHA-artesunate | 0.156 | 0.523 | 18 | 0.300 | 0.212 | 18 |
SAHA-2-ASA-9 | 0.818 | <0.001 | 14 | 0.519 | 0.023 | 18 |
SAHA-2-ASA-14 | 0.515 | 0.029 | 17 | 0.523 | 0.018 | 18 |
2-ASA-9-chloroquine | 0.418 | 0.121 | 14 | 0.307 | 0.201 | 18 |
2-ASA-9-amodiaquine | 0.104 | 0.713 | 14 | 0.401 | 0.099 | 19 |
2-ASA-9-piperaquine | 0.282 | 0.308 | 14 | 0.209 | 0.391 | 18 |
2-ASA-9-mefloquine | 0.450 | 0.092 | 14 | 0.527 | 0.030 | 16 |
2-ASA-9-artesunate | −0.057 | 0.840 | 14 | 0.174 | 0.489 | 17 |
2-ASA-9-2-ASA-14 | 0.662 | 0.010 | 13 | 0.542 | 0.017 | 18 |
2-ASA-14-chloroquine | 0.123 | 0.627 | 17 | 0.490 | 0.028 | 19 |
2-ASA-14-amodiaquine | −0.108 | 0.669 | 17 | 0.321 | 0.180 | 18 |
2-ASA-14-piperaquine | 0.389 | 0.111 | 17 | 0.087 | 0.715 | 19 |
2-ASA-14-mefloquine | 0.123 | 0.627 | 17 | 0.610 | 0.007 | 17 |
2-ASA-14-artesunate | −0.113 | 0.657 | 17 | 0.233 | 0.336 | 18 |
Spearman rank correlation coefficients (rs) are shown. The statistically significant values are shown in boldface type.
The P values show the significance of the correlation between each pair of drugs, with those reaching statistical significance shown in boldface type.
df, degrees of freedom.
DISCUSSION
Inhibitors of histone deacetylase (HDAC) enzymes represent a promising new class of antimalarial compounds (3). Suberoylanilide hydroxamic acid (SAHA) (Vorinostat) was licensed for use in humans for the treatment of advanced cutaneous lymphoma in 2006, and knowledge about the safety and pharmacokinetics of HDAC inhibitors is accruing (6, 44). In vitro drug susceptibility testing has demonstrated that several hydroxamate-based HDAC inhibitors are potent inhibitors (IC50s in low nanomolar concentrations) of laboratory strains of P. falciparum and, importantly, display promising selectivity for malaria parasites versus mammalian cell lines (>100 fold), depending on the inhibitor and cell lines used (2, 12).
In the present study, we compared the ex vivo activities of three hydroxamate-based HDAC inhibitors against multidrug-resistant clinical isolates of P. falciparum and P. vivax. This is the first report of antimalarial activity of this class of compounds directly against clinical isolates and against P. vivax parasites. The derived potencies (IC50) ranged from 87 to 964 nM against P. falciparum and 67 to 766 nM for P. vivax. This level of drug activity was within the range of well-tolerated plasma drug concentrations observed in phase 1 clinical drug trials in oncology patients (26, 27). However, the ex vivo susceptibility of clinical isolates of P. falciparum to HDAC inhibitors was significantly lower than previously reported in culture-adapted P. falciparum laboratory strains (Table 2). Although some of this difference may be explained by the level of multidrug resistance in Papua, Indonesia, the IC50s for chloroquine were not excessive (median of 30 nM) and significantly lower than that reported for the chloroquine-resistant laboratory strain W2 (400 nM; Table 2). An alternative explanation is the difference in assay methodology. Previous studies employed proliferation-based growth inhibition assays of culture-adapted laboratory strains with in vitro growth quantified by [3H]hypoxanthine incorporation (4), whereas in the present study, inhibitory effects were determined in isolates fresh from the patient with quantification of ex vivo schizont maturation assessed by microscopy (38). This phenomenon (i.e., microscopic assessment of schizont maturation producing higher IC50s than those derived from radioisotopic incorporation) has been described previously (46). In addition, even by applying the same methodology (i.e., [3H]hypoxanthine incorporation) but using different assay durations, IC50 estimates derived by a one-cycle assay (42 h) can be 2- to 6-fold higher than the estimates derived from the standard assay (72 h) (29). In our study, IC50s in chloroquine-sensitive and -resistant laboratory strains (K1 and 3D7, respectively) assessed by using the schizont maturation assay were indeed higher (700 to 1,000 nM) than the reported values derived by the isotopic assay (15 to 300 nM).
Our study also presents data for the first time on the ex vivo activity of HDAC inhibitors against the second most significant malaria parasite, P. vivax. On the basis of sequence homology, three class I/II HDAC homologues have been identified in P. falciparum—the class I HDAC homologue (P. falciparum HDAC 1 [PfHDAC1]) and two less-conserved putative HDACs (PfHDAC2 and PfHDAC3) about which little is known. Homologues for all three of these genes are present in other Plasmodium species, including P. vivax. The other HDAC homologues cannot be ruled out as possible targets of HDAC inhibitors in P. falciparum, and furthermore, a correlation between HDAC inhibitors with selective antimalarial action on parasite HDACs versus mammalian cell HDACs has not yet been demonstrated. However, recombinant PfHDAC1 has recently been generated, and the activity of this enzyme has been shown to be inhibited by hydroxamate-based HDAC inhibitors (32). PfHDAC1 and the P. vivax homologue (PlasmoDB gene identification [ID] PVX_099700) share ∼95% sequence identity at the amino acid level, and preliminary homology modeling using the same alignment as for an in silico PfHDAC1 homology model previously generated by us (2) revealed no substituted residues near the active site that were different (D. P. Fairlie and A. J. Lucke, unpublished results). There is currently no homology model available for the other PfHDACs due to limited sequence similarity with other eukaryotic HDAC enzymes.
In view of the confounding effects of stage specificity and duration of assay for drug susceptibility testing in P. vivax (38, 40), we have adopted more-stringent criteria for our P. vivax assay. In the present study, only isolates with more than 70% of the parasites at the ring stage were selected for testing and with culture maintained for a minimum of 30 h prior to harvest. Our results for P. vivax isolates tested at both the ring and trophozoite stages confirm the marked stage specificity of action of chloroquine, with trophozoites being almost completely resistant to the effect of the drug (38, 40). In contrast, the stage-specific action in P. falciparum was modest, with IC50s less than 2-fold higher for trophozoite stages compared to ring stages. The results with HDAC inhibitors are consistent with a previous study in which the effect of continual exposure of P. falciparum line Dd2 to 2-ASA-9 and 2-ASA-14 was examined at the ring stage (∼18 h after invasion), trophozoite stage (∼26 h after invasion), and schizont stage (∼38 h after invasion). Microscopic examination showed that all stages were inhibited, with very small numbers of invasive merozoites being released from schizonts (2).
The ex vivo activity between the HDAC inhibitors was strongly correlated (rs = 0.515 to 0.818; Table 4). Whereas the drug susceptibility of SAHA was correlated with mefloquine in P. falciparum (rs = 0.635), this did not reach statistical significance in P. vivax. Conversely, the activities of 2-ASA-9 and 2-ASA-14 were highly correlated with mefloquine in P. vivax, but not in P. falciparum. The statistical significance of our observations may simply reflect the small sample size of the study and intrinsic variance of our assay. Alternatively, ex vivo drug efficacy may not be determined solely by structural differences in the HDAC inhibitors and their plasmodial targets. While cross-susceptibility between HDAC inhibitors and chloroquine or mefloquine has not been observed in sensitive and resistant laboratory strains (2, 12), the differences in correlation profiles for P. falciparum and P. vivax may indicate different susceptibility profiles in these species. Molecular mechanisms underlying chloroquine resistance are likely to differ significantly between P. vivax and P. falciparum (31, 43). However, this is not the case for mefloquine susceptibility, which has been correlated with increased copy number of the mdr1 gene (P-glycoprotein) in both species (34, 42). There is limited data on reduced susceptibility to HDAC inhibitors in eukaryotic cells, but mdr1-mediated tolerance has been reported for the non-hydroxamate-based compounds apicidin and depsipeptide (FK-228) in several cancer cell lines (28, 48). However, there is also evidence for mdr1-independent mechanisms (15), and a more recent study has shown that HDAC inhibitors were able to promote cell line-specific and reversible induction of multiple drug transporter genes (20). Antimalarial resistance is postulated to be a multigenic process (13), and it is possible that reduced Plasmodium susceptibility to HDAC inhibitors will occur. However, since HDAC inhibitors have an effect on multiple vital eukaryotic cell functions (49) and potentially act on both histone and non-histone-related pathways, it is difficult to predict exactly how Plasmodium will develop resistance to this drug class.
In our study, hydroxamate-based HDAC inhibitors SAHA, 2-ASA-9, and 2-ASA-14 showed potent ex vivo efficacy against multidrug-resistant Plasmodium field isolates. Importantly, ex vivo activity of HDAC inhibitors against P. vivax was comparable to that in P. falciparum, suggesting that HDAC inhibitors may be promising candidates for antimalarial therapy in geographical areas where both species are endemic. Elucidation of the species-specific structure of HDAC analogues in Plasmodium should facilitate molecular design aimed at optimizing the selectivity and efficacy of HDAC inhibitors. Studies assessing the comparative clinical efficacy in Plasmodium species and defining drug interaction with common antimalarial compounds will reveal further the potential of HDAC inhibitors in antimalarial therapy.
Acknowledgments
We are grateful to Lembaga Pengembangan Masyarakat Amungme dan Kamoro, the staff of the Rumah Sakit Mitra Masyarakat Hospital, and Paulus Sugiarto for their support in conducting this study. We thank the Australian Red Cross blood transfusion service for supplying human sera.
This study was funded by the Wellcome Trust (ICRG GR071614MA), the Australian National Health and Medical Research Council (ICRG ID 283321, program 496600) and AusAID (infrastructure support for the Timika Translational Research Facility). J.M. is funded by a Swiss National Science Foundation (SNSF) fellowship, D.P.F. is funded by an Australian Research Council fellowship, K.T.A. is funded by an ARC Future Fellowship, N.M.A. is supported by an NHMRC Practitioner Fellowship, and R.N.P. is a Wellcome Trust Senior Research Fellow in Clinical Science.
Footnotes
Published ahead of print on 6 December 2010.
The authors have paid a fee to allow immediate free access to this article.
REFERENCES
- 1.Akoachere, M., et al. 2005. In vitro assessment of methylene blue on chloroquine-sensitive and -resistant Plasmodium falciparum strains reveals synergistic action with artemisinins. Antimicrob. Agents Chemother. 49:4592-4597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Andrews, K. T., et al. 2008. Potent antimalarial activity of histone deacetylase inhibitor analogues. Antimicrob. Agents Chemother. 52:1454-1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Andrews, K. T., T. N. Tran, N. C. Wheatley, and D. P. Fairlie. 2009. Targeting histone deacetylase inhibitors for anti-malarial therapy. Curr. Top. Med. Chem. 9:292-308. [DOI] [PubMed] [Google Scholar]
- 4.Andrews, K. T., et al. 2000. Anti-malarial effect of histone deacetylation inhibitors and mammalian tumour cytodifferentiating agents. Int. J. Parasitol. 30:761-768. [DOI] [PubMed] [Google Scholar]
- 5.Barcus, M. J., et al. 2007. Demographic risk factors for severe and fatal vivax and falciparum malaria among hospital admissions in northeastern Indonesian Papua. Am. J. Trop. Med. Hyg. 77:984-991. [PubMed] [Google Scholar]
- 6.Cang, S., Y. Ma, and D. Liu. 2009. New clinical developments in histone deacetylase inhibitors for epigenetic therapy of cancer. J. Hematol. Oncol. 2:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chaal, B. K., A. P. Gupta, B. D. Wastuwidyaningtyas, Y. H. Luah, and Z. Bozdech. 2010. Histone deacetylases play a major role in the transcriptional regulation of the Plasmodium falciparum life cycle. PLoS Pathog. 6:e1000737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Daher, W., et al. 2006. In vitro metabolism of ferroquine (SSR97193) in animal and human hepatic models and antimalarial activity of major metabolites on Plasmodium falciparum. Drug Metab. Dispos. 34:667-682. [DOI] [PubMed] [Google Scholar]
- 9.Dar, O., M. S. Khan, and I. Adagu. 2008. The potential use of methotrexate in the treatment of falciparum malaria: in vitro assays against sensitive and multidrug-resistant falciparum strains. Jpn. J. Infect. Dis. 61:210-211. [PubMed] [Google Scholar]
- 10.de Pilla Varotti, F., et al. 2008. Synthesis, antimalarial activity, and intracellular targets of MEFAS, a new hybrid compound derived from mefloquine and artesunate. Antimicrob. Agents Chemother. 52:3868-3874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Douglas, N. M., N. M. Anstey, B. J. Angus, F. Nosten, and R. N. Price. 2010. Artemisinin combination therapy for vivax malaria. Lancet Infect. Dis. 10:405-416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Dow, G. S., et al. 2008. Antimalarial activity of phenylthiazolyl-bearing hydroxamate-based histone deacetylase inhibitors. Antimicrob. Agents Chemother. 52:3467-3477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Duraisingh, M. T., and P. Refour. 2005. Multiple drug resistance genes in malaria-from epistasis to epidemiology. Mol. Microbiol. 57:874-877. [DOI] [PubMed] [Google Scholar]
- 14.Duraisingh, M. T., et al. 2005. Heterochromatin silencing and locus repositioning linked to regulation of virulence genes in Plasmodium falciparum. Cell 121:13-24. [DOI] [PubMed] [Google Scholar]
- 15.Fedier, A., K. J. Dedes, P. Imesch, A. O. Von Bueren, and D. Fink. 2007. The histone deacetylase inhibitors suberoylanilide hydroxamic (Vorinostat) and valproic acid induce irreversible and MDR1-independent resistance in human colon cancer cells. Int. J. Oncol. 31:633-641. [PubMed] [Google Scholar]
- 16.Freitas-Junior, L. H., et al. 2005. Telomeric heterochromatin propagation and histone acetylation control mutually exclusive expression of antigenic variation genes in malaria parasites. Cell 121:25-36. [DOI] [PubMed] [Google Scholar]
- 17.Genton, B., et al. 2008. Plasmodium vivax and mixed infections are associated with severe malaria in children: a prospective cohort study from Papua New Guinea. PLoS Med. 5:e127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Grant, S., C. Easley, and P. Kirkpatrick. 2007. Vorinostat. Nat. Rev. Drug Discov. 6:21-22. [DOI] [PubMed] [Google Scholar]
- 19.Guglielmo, S., et al. 2009. A new series of amodiaquine analogues modified in the basic side chain with in vitro antileishmanial and antiplasmodial activity. Eur. J. Med. Chem. 44:5071-5079. [DOI] [PubMed] [Google Scholar]
- 20.Hauswald, S., et al. 2009. Histone deacetylase inhibitors induce a very broad, pleiotropic anticancer drug resistance phenotype in acute myeloid leukemia cells by modulation of multiple ABC transporter genes. Clin. Cancer Res. 15:3705-3715. [DOI] [PubMed] [Google Scholar]
- 21.Hay, S. I., C. A. Guerra, A. J. Tatem, A. M. Noor, and R. W. Snow. 2004. The global distribution and population at risk of malaria: past, present, and future. Lancet Infect. Dis. 4:327-336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hu, G., et al. 2010. Transcriptional profiling of growth perturbations of the human malaria parasite Plasmodium falciparum. Nat. Biotechnol. 28:91-98. [DOI] [PubMed] [Google Scholar]
- 23.Joshi, M. B., et al. 1999. Molecular cloning and nuclear localization of a histone deacetylase homologue in Plasmodium falciparum. Mol. Biochem. Parasitol. 99:11-19. [DOI] [PubMed] [Google Scholar]
- 24.Kahnberg, P., et al. 2006. Design, synthesis, potency, and cytoselectivity of anticancer agents derived by parallel synthesis from alpha-aminosuberic acid. J. Med. Chem. 49:7611-7622. [DOI] [PubMed] [Google Scholar]
- 25.Karyana, M., et al. 2008. Malaria morbidity in Papua Indonesia, an area with multidrug resistant Plasmodium vivax and Plasmodium falciparum. Malar. J. 7:148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kelly, W. K., et al. 2005. Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J. Clin. Oncol. 23:3923-3931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kelly, W. K., et al. 2003. Phase I clinical trial of histone deacetylase inhibitor: suberoylanilide hydroxamic acid administered intravenously. Clin. Cancer Res. 9:3578-3588. [PubMed] [Google Scholar]
- 28.Kim, Y. K., et al. 2008. Histone deacetylase inhibitor apicidin-mediated drug resistance: involvement of P-glycoprotein. Biochem. Biophys. Res. Commun. 368:959-964. [DOI] [PubMed] [Google Scholar]
- 29.Kocken, C. H., et al. 2006. Plasmodium vivax: in vitro susceptibility of blood stages to synthetic trioxolane compounds and the diamidine DB75. Exp. Parasitol. 113:197-200. [DOI] [PubMed] [Google Scholar]
- 30.Muangnoicharoen, S., D. J. Johnson, S. Looareesuwan, S. Krudsood, and S. A. Ward. 2009. Role of known molecular markers of resistance in the antimalarial potency of piperaquine and dihydroartemisinin in vitro. Antimicrob. Agents Chemother. 53:1362-1366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Nomura, T., et al. 2001. Evidence for different mechanisms of chloroquine resistance in 2 Plasmodium species that cause human malaria. J. Infect. Dis. 183:1653-1661. [DOI] [PubMed] [Google Scholar]
- 32.Patel, V., et al. 2009. Identification and characterization of small molecule inhibitors of a class I histone deacetylase from Plasmodium falciparum. J. Med. Chem. 52:2185-2187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Patil, V., et al. 2010. Antimalarial and antileishmanial activities of histone deacetylase inhibitors with triazole-linked cap group. Bioorg. Med. Chem. 18:415-425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Price, R. N., et al. 1999. The pfmdr1 gene is associated with a multidrug-resistant phenotype in Plasmodium falciparum from the western border of Thailand. Antimicrob. Agents Chemother. 43:2943-2949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Price, R. N., N. M. Douglas, and N. M. Anstey. 2009. New developments in Plasmodium vivax malaria: severe disease and the rise of chloroquine resistance. Curr. Opin. Infect. Dis. 22:430-435. [DOI] [PubMed] [Google Scholar]
- 36.Ratcliff, A., et al. 2007. Therapeutic response of multidrug-resistant Plasmodium falciparum and P. vivax to chloroquine and sulfadoxine-pyrimethamine in southern Papua, Indonesia. Trans. R. Soc. Trop. Med. Hyg. 101:351-359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Richards, W. H., and S. G. Williams. 1973. The removal of leucocytes from malaria infected blood. Ann. Trop. Med. Parasitol. 67:249-250. [DOI] [PubMed] [Google Scholar]
- 38.Russell, B., et al. 2008. Determinants of in vitro drug susceptibility testing of Plasmodium vivax. Antimicrob. Agents Chemother. 52:1040-1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Russell, B. M., et al. 2003. Simple in vitro assay for determining the sensitivity of Plasmodium vivax isolates from fresh human blood to antimalarials in areas where P. vivax is endemic. Antimicrob. Agents Chemother. 47:170-173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Sharrock, W. W., et al. 2008. Plasmodium vivax trophozoites insensitive to chloroquine. Malar. J. 7:94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Sumawinata, I. W., et al. 2003. Very high risk of therapeutic failure with chloroquine for uncomplicated Plasmodium falciparum and P. vivax malaria in Indonesian Papua. Am. J. Trop. Med. Hyg. 68:416-420. [PubMed] [Google Scholar]
- 42.Suwanarusk, R., et al. 2008. Amplification of pvmdr1 associated with multidrug-resistant Plasmodium vivax. J. Infect. Dis. 198:1558-1564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Suwanarusk, R., et al. 2007. Chloroquine resistant Plasmodium vivax: in vitro characterisation and association with molecular polymorphisms. PLoS One 2:e1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Tan, J., S. Cang, Y. Ma, R. L. Petrillo, and D. Liu. 2010. Novel histone deacetylase inhibitors in clinical trials as anti-cancer agents. J. Hematol. Oncol. 3:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Tjitra, E., et al. 2008. Multidrug-resistant Plasmodium vivax associated with severe and fatal malaria: a prospective study in Papua, Indonesia. PLoS Med. 5:e128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wernsdorfer, W. H., and I. McGregor. 1989. Malaria: principles and practice of malariology. Churchill Livingstone, Edinburgh, Scotland, United Kingdom.
- 47.World Health Organization. 2008. World malaria report 2008. World Health Organization publication no. WHO/HTM/GMP/2008.1. World Health Organization, Geneva, Switzerland.
- 48.Xiao, J. J., et al. 2005. Chemoresistance to depsipeptide FK228 [(E)-(1S,4S,10S,21R)-7-[(Z)-ethylidene]-4,21-diisopropyl-2-oxa-12,13-dithia-5,8,20,23-tetraazabicyclo[8,7,6]-tricos-16-ene-3,6,9,22-pentanone] is mediated by reversible MDR1 induction in human cancer cell lines. J. Pharmacol. Exp. Ther. 314:467-475. [DOI] [PubMed] [Google Scholar]
- 49.Xu, W. S., R. B. Parmigiani, and P. A. Marks. 2007. Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene 26:5541-5552. [DOI] [PubMed] [Google Scholar]
- 50.Yang, X. J., and E. Seto. 2007. HATs and HDACs: from structure, function and regulation to novel strategies for therapy and prevention. Oncogene 26:5310-5318. [DOI] [PubMed] [Google Scholar]