Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

J Biol Inorg Chem (2012) 17:275–283

DOI 10.1007/s00775-011-0848-x

ORIGINAL PAPER

Isoniazid metal complex reactivity and insights


for a novel anti-tuberculosis drug design
Eduardo Henrique Silva Sousa • Luiz Augusto Basso • Diógenes S. Santos •

Izaura Cirino Nogueira Diógenes • Elisane Longhinotti •


Luiz Gonzaga de França Lopes • Ícaro de Sousa Moreira

Received: 27 May 2011 / Accepted: 10 September 2011 / Published online: 28 September 2011
Ó SBIC 2011

Abstract For over a decade, tuberculosis (TB) has been the KatG enzyme was used. However, upon metal coordination,
leading cause of death among infectious diseases. Since a significant enhancement in the formation of isonicotinic
the 1950s, isoniazid has been used as a front-line drug in the acid was observed compared with that of isonicotinamide.
treatment of TB; however, resistant TB strains have limited These results suggest that the pathway of a carbonyl-cen-
its use. The major route of isoniazid resistance relies on KatG tered radical might be favored upon coordination to the
enzyme disruption, which does not promote an electron Fe(II) owing to the p-back-bonding effect promoted by this
transfer reaction. Here, we investigated the reactivity of metal center; therefore, the isoniazid metal complex could
isoniazid metal complexes as prototypes for novel self- serve as a potential metallodrug. Enzymatic inhibition assays
activating metallodrugs against TB with the aim to overcome conducted with InhA showed that the cyanoferrate moiety is
resistance. Reactivity studies were conducted with hydrogen not the major player involved in this inhibition but the pres-
peroxide, hexacyanoferrate(III), and aquopentacyanofer- ence of isoniazid is required in this process. Other isoniazid
rate(III). The latter species showed a preference for the inner- metal complexes, [Ru(CN)5(izd)]3- and [Ru(NH3)5(izd)]2?
sphere electron transfer reaction pathway. Additionally, (where izd is isoniazid), were also unable to inhibit InhA,
electron transfer reaction performed with either free isonia- supporting our proposed self-activating mechanism of
zid or (isoniazid)pentacyanoferrate(II) complex resulted in action. We propose that isoniazid reactivity can be rationally
similar oxidized isoniazid derivatives as observed when the modulated by metal coordination chemistry, leading to the
development of novel anti-TB metallodrugs.

In memory of Ícaro de Sousa Moreira. Keywords Tuberculosis  Isoniazid  Metallodrugs 


Enoyl reductase  Cyanoferrate
E. H. S. Sousa (&)  I. C. N. Diógenes  E. Longhinotti 
L. G. de França Lopes  Í. de Sousa Moreira
Departamento de Quı́mica Orgânica e Inorgânica, Abbreviations
Laboratório de Bioinorgânica, DMSO Dimethyl sulfoxide
Universidade Federal do Ceará, HPLC High-performance liquid chromatography
P.O. Box 6021,
IQG607 Sodium (isoniazid)pentacyanoferrate(II)
Fortaleza 60455-970, Brazil
e-mail: eduardohss@dqoi.ufc.br TB Tuberculosis

L. A. Basso  D. S. Santos
Centro de Pesquisas em Biologia Molecular e Funcional,
Pontifı́cia Universidade Católica do Rio Grande do Sul, Av.
Ipiranga, 6681, Tecnopuc, Prédio 92A Partenon,
Introduction
Porto Alegre, RS 90619-900, Brazil
Mycobacterium tuberculosis infection has affected mankind
L. A. Basso  D. S. Santos  L. G. de França Lopes for centuries, leading to the death of billions of people.
Instituto Nacional de Ciência e Tecnologia em Tuberculose,
Pontifı́cia Universidade Católica do Rio Grande do Sul,
Extremely efficient therapeutic compounds were discovered
Porto Alegre, Brazil in the mid-1900, leading to great expectation of eradicating

123
276 J Biol Inorg Chem (2012) 17:275–283

tuberculosis (TB) [1]. Unfortunately, this goal has not been works as an efficient inhibitor for InhA enzyme
attained and a worse-case scenario has emerged in which TB (Kd = 0.4 nM) in a slow-onset inhibition step [17]. This
has become the leading cause of death worldwide compared protein is an enoyl reductase enzyme involved in one of the
with other known infectious diseases. It latently infects about steps for mycolic acid biosynthesis and it is a very efficient
two billion people, one third of the world’s population, therapeutic target.
creating a huge reservoir of this bacterium [2]. Each year, Approximately 50% of isolated isoniazid-resistant
there are about eight million new TB cases and two million strains have either a deletion or mutations in the katG gene
deaths [3]. The growing number of multiple-drug-resistant [18, 19]. It appears that the enzymatic activation of isoni-
M. tuberculosis strains and the recently discovered extre- azid through an electron transfer reaction is a major step in
mely drug resistant strains are a cause for alarm among health TB resistance, as described elsewhere [20]. These reports
agencies around the world. According to the WHO, multiple- along with our previous chemical and biochemical data
drug-resistant M. tuberculosis strains are found in at least 72 prompted us to design novel ‘‘self-activating metallodrugs’’
countries, accounting for 3–41% of TB cases [4]. In 2005, it for TB treatment, which would potentially overcome iso-
was reported that there were 27,000 cases of extremely drug- niazid resistance. Isoniazid is an acylhydrazine pyridine
resistant TB in developed and underdeveloped countries [5]. derivative and, from an inorganic chemistry view point, has
Despite the fact that TB can be cured using therapy appealing properties. For decades, pyridine derivatives
involving multiple drugs, the minimum treatment time of have been shown to form stable and well-characterized
6 months is too long and the drugs are highly toxic. These complexes with cyanoferrates [21–24]. Moreover, there
factors make patient compliance with therapy very have been a number of reports on electron transfer reac-
difficult, and as a result, this noncompliance leads to drug- tions involving cyanoferrate complexes [21, 25–27]. Oxi-
resistant M. tuberculosis. Since the most recently devel- dation of many organic ligands exhibits large kinetic
oped TB drug is about 40 years old, there is a strong barriers, which are usually dramatically lowered by
requirement for the development of new drugs that would metalloenzymes. Intramolecular electron transfer reactions
shorten the treatment time, lower the toxicity levels, and promoted by metals could be used as a strategy to mimic
yet be effective against the various resistant strains [3, 6]. biocatalysts. This strategy has just recently gained appeal
Isoniazid is the front-line and most effective anti-TB drug in metallodrug design. Isoniazid requires enzymatic catal-
discovered so far, along with rifampin. The discovery of ysis to promote an electron transfer reaction leading to drug
isoniazid was a major milestone in chemotherapy for TB activation; a coordinated metal complex could produce a
because it was highly active, inexpensive, and relatively less faster alternative route for intramolecular-assisted ligand
toxic [7]. Its molecular mechanism of action was unknown oxidation. On the basis of this, we have asked the question
for decades long after its first clinical use, but a more detailed whether the cyanoferrate moiety could indeed favor a faster
picture has emerged during recent decades [8]. At least five intramolecular electron transfer reaction of isoniazid in
different genes (katG, inhA, ahpC, kasA, and ndh) have been support of this strategy. Additionally, we evaluated how
found to correlate with isoniazid resistance [6]. However, the similar these oxidized species were to enzymatically and
primary target of isoniazid has been shown to be the product nonenzymatically generated active species and what their
of the inhA structural gene [9]. A proteomic approach using implications are for the design of a novel anti-TB drug. It
isoniazid adducts was conducted, which led to the identifi- was shown that sodium (isoniazid)pentacyanoferrate(II)
cation of a panel of other promising potential isoniazid tar- (IQG607) could efficiently inhibit the TB target enzyme
gets in M. tuberculosis [10]; hence, this suggests that there (InhA) in vitro and kill M. tuberculosis, including isoni-
may be a broader spectrum of isoniazid anti-TB action. In azid-resistant strains [28–30]. A mechanism of action has
addition, there is a search for novel molecular anti-TB targets been proposed that is based on the chemical reactivity of
for a generation of new drugs, including those potentially this complex along with biochemical and biological data
involved in M. tuberculosis latency [11–13]. supporting this compound as a prototype for metallodrugs
It has been shown that isoniazid is a prodrug that is against M. tuberculosis. However, there was no clear evi-
activated by an electron transfer reaction catalyzed by the dence that supported the mechanism mentioned above.
catalase–peroxidase KatG [14]. This reaction can generate
a range of reactive oxygen species and reactive organic
radicals, which can attack multiple targets in M. tubercu- Materials and methods
losis [3]. Several reports have suggested that the main
mechanism for isoniazid is based on the formation of a Materials
radical intermediate species [15, 16]. This isonicotinic acyl
radical would promptly react with NAD(H) and produced a All experiments were performed under an inert atmosphere
NAD–isoniazid adduct [15]. It has been shown this adduct (N2 or Ar) using conventional techniques. Milli-Q grade or

123
J Biol Inorg Chem (2012) 17:275–283 277

doubly distilled water was used throughout. Pyridine meter in the UV and visible range. A 1.0-cm quartz cell
derivatives (isonicotinamide, isonicotinic acid, 4-pyri- was used and sample concentrations were kept between
dinemethanol, 4-pyridinecarboxaldehyde) used as stan- 1.0 9 10-3 and 1.0 9 10-2 M for most of the experiments
dards in high-performance liquid chromatography (HPLC) unless otherwise stated. IR spectra were taken with a Shi-
along with NAD? and NADH were purchased from Sigma. madzu FTIR-8300 spectrophotometer, using solid samples
Isoniazid, from Aldrich, was recrystallized in methanol dispersed in KBr pellets. Electrochemical experiments
solution. Potassium hexacyanoferrate(III) was purchased were conducted with a BAS 100BW electrochemical ana-
from Mallinckrodt. Na3[FeII(CN)5(NH3)]3H2O and lyzer (Bioanalytical Systems, West Lafayette, IN, USA).
Na2[FeIII(CN)5(NH3)]H2O complexes were synthesized as A conventional three-electrode glass configuration cell with
previously described [29, 30] with minor modifications a glassy carbon electrode of 0.126-cm2 geometrical area, and
[20]. All other chemicals were of analytical reagent grade. a platinum foil were used as working and auxiliary elec-
trodes, respectively. A 0.1 M sodium trifluoroacetate solu-
Synthesis of Na3[Fe(CN)5(izd)]4H2O (IQG607) tion, pH 5.0, was used as the electrolyte, at 25 °C. Potential
values were measured versus Ag/AgCl (3.5 M KCl, Bioan-
A 100.0 mg sample of Na3[Fe(CN)5(NH3)]3H2O was alytical Systems) as a reference electrode. All potentials
dissolved in 5 mL of water under argon flow. The isoniazid were converted to the normal hydrogen electrode and are
ligand (90.0 mg), dissolved in 2 mL of water, was added to cited accordingly throughout the text. The Mössbauer
the complex solution. Upon mixing, the resulting solution spectra were obtained with a conventional constant accel-
immediately developed an orange color and was allowed to eration spectrometer operating in a triangular wave mode
react for 2 h under argon, with stirring, and in the with a 57Co source in a rhodium matrix. All spectra were
absence of light. The reaction vessel was then cooled in an taken with the source and absorber at room temperature, and
ice bath, and cold NaI-saturated solution in ethanol was sodium nitroprusside was used as a reference.
added dropwise until precipitation was complete. Chromatography measurements were conducted with a
Na3[Fe(CN)5(izd)]4H2O (IQG607; where izd is isoniazid) Shimadzu liquid chromatograph (HPLC) equipped with a
as a red solid was collected by filtration, washed with model LC-10AD pump and an SPD-M10A UV–vis pho-
ethanol and diethyl ether, dried, and stored under a vacuum todiode-array detector with a CBM-10AD interface. An
in the absence of light. Anal. Calc.: C: 28.4; H: 3.2; N: 24; octadecyl silica column (250 mm 9 4.6-mm inner diame-
Found: C: 28.0; H: 3.5; N: 23.7. 1H NMR: d(D2O, 500 ter, 5-lm particles, from Alltech) was employed and
MHz) 9.11 (d, 2H, H-2,20 ), 7.41 (d, 2H, H-3,30 ). The yield injection volumes of 5 lL were used for most of the
was greater than 85%. analyses unless otherwise stated.

Reaction of isoniazid and Na2[FeIII(CN)5(NH3)]H2O Kinetic measurements

A 100.0-mg sample of Na2[Fe(CN)5(NH3)]H2O was dissolved Outer-sphere electron transfer reaction rates were deter-
in 5 mL of water. The isoniazid ligand (100.0 mg), dissolved in mined using a Hewlett-Packard 8453 spectrophotometer at
2 mL of water, was added to the complex solution. Upon 25 °C. Solutions of the reactants were freshly prepared
mixing, bubbling of gas and a color change occurred immedi- under an argon atmosphere for each run. Buffer solutions
ately. The reaction was allowed to proceed for 2 h under argon, consisting of a mixture of K2HPO4 and KH2PO4 were used.
with stirring, and in the absence of light. The reaction vessel The ionic strength of the solutions (0.5 M) was adjusted by
was then cooled in an ice bath, and ice-cold 1:2 diethyl ether/ the addition of KCl. The pH measurements were performed
ethanol was added dropwise until precipitation was complete. with a Corning 440 pH meter equipped with a Cole-Parmer
The orange solid was collected by filtration, washed with eth- glass electrode. Values of kobs were calculated from a least-
anol and diethyl ether, dried, and stored under a vacuum in the squares fitting to the equation ln(At - A?) = ln(Ao -
absence of light. The yield was better than 85%. Formula A?) - kobst, where At, Ao, and A? are absorbances at time
proposed: Na3[FeII(CN)5(isn)]3H2O (where isn is isonicoti- t, at time zero (first point), and at longer times (four or
namide) Anal. Calc.: C: 30.6; H: 2.8; N: 22.7; Found: C: 30.0; more half lives), respectively. The calculations were done
H: 2.6; N: 23.0. 1H NMR: d(D2O, 500 MHz) 9.18 (d, 2H, using the Hewlett-Packard 8453 spectrophotometer
H-2,20 ), 7.57 (d, 2H, H-3,30 ). kinetic software. The reaction progress was followed at
420 nm, the absorbance maximum of [Fe(CN)6]3-, under
Apparatus pseudo-first order conditions. Kinetic measurements for
the intramolecular reaction were performed with an
The electronic absorption spectra were acquired with a Aminco-Morrow 126550-3 stopped-flow spectrophoto-
Hewlett-Packard 8453 diode-array scanning spectrophoto- meter.

123
278 J Biol Inorg Chem (2012) 17:275–283

Reactions with hexacyanoferrate(III) from the rates of decrease in absorbance at 340 nm using
2-trans-dodecenoyl-CoA (100.0 lM) and NADH (200 lM).
The reactions of isoniazid with hexacyanoferrate(III) in The substrate 2-trans-dodecenoyl-CoA was synthesized
excess were performed in buffered solution, pH 7.0, fol- from 2-trans-dodecenoic acid and CoA by the mixed anhy-
lowed by electronic spectroscopy (k = 420 nm) until dride method [28, 31] and was purified by reverse-phase
completion. The products of this reaction were also char- HPLC using a 19 mm 9 300 mm C18 lBondapak column
acterized using HPLC. We injected 5 lL of the product (Waters Associates, Milford, MA, USA) as previously
reaction mixture into the HPLC system and separated the described [31]. The ratio of the absorbance of purified
mixture by isocratic elution. Standards were injected to 2-trans-dodecenoyl-CoA at 232 and 260 nm was 0.62, a
identify the products formed. value that meets the established criterion for pure thioesters
The reaction of 2.0 mM NAD? and isoniazid, in buf- (A232 nm/A260 nm C 0.52). Wild-type InhA and I21V mutant
fered phosphate solution pH 7.0, was started by addition of enzymes were expressed and purified to homogeneity as
4 mM hexacyanoferrate(III) and the reaction was allowed described elsewhere [28, 29]. The specific activities of wild-
to proceed for 1 h. This mixture was loaded on a C18 type InhA and I21V mutant with 2-trans-dodecenoyl-CoA
reverse-phase column and separated by a gradient elution (100 lM) and NADH (200.0 lM) were 13.5 ± 0.4 and
of 5–60% acetonitrile/water over a period of 40 min. The 8.1 ± 0.3 U mg-1, respectively. Activity measurements for
adduct products were identified by the appearance of two both wild-type and I21V InhA proteins were performed for
new peaks with typical electronic spectra. Standard mix- up to 1 h in 100 mM Na2HPO4, pH 7.5 at 25 °C, but no loss
tures of only two of the three components were prepared to of enzyme activity was observed in the time range studied.
characterize and validate the appearance of the peak.
After mixing, we loaded three control mixtures: (1)
NAD? and hexacyanoferrate(III), (2) NAD? and isoniazid, Results and discussion
and (3) isoniazid and hexacyanoferrate(III), and their peaks
were identified and excluded from the reaction conducted A product of the reaction of isoniazid
with the three components [NAD?, isoniazid, and hexa- with [FeIII(CN)5(NH3)]2-
cyanoferrate(III)].
On the basis of previous observations of electron transfer
Reactions with hydrogen peroxide reactions involving isoniazid-like compounds, we conducted
a direct reaction of isoniazid with [FeIII(CN)5(NH3)]2-. The
Free and coordinated isoniazid (1.0 mM) were oxidized final product was isolated and further analyzed. This isolated
using 66.0 mM hydrogen peroxide solution, in phosphate metal complex showed the presence of a peak profile com-
buffer pH 7.0, at 25 °C and the reaction was allowed to patible with diamagnetic iron(II) with predominant signals at
proceed for 6 h. Their oxidized products were identified on d 9.18 and d 7.57, which, respectively, correspond to the
the basis of HPLC injections of standards. The H2O2-oxi- H-2,20 and H-3,30 of a pyridine derivative such as isonico-
dized isoniazid iron complex was subjected to a 100-fold tinamide. Additionally, this result indicates that the coordi-
concentration excess of dimethyl sulfoxide (DMSO) to nation occurs through the nitrogen atom of the pyridine ring.
ensure complete ligand displacement, and to prevent any The IR spectroscopy measurements were also consistent
further oxidation reaction, ascorbic acid was added to the with a reduced pentacyanoferrate(II) complex as assigned by
mixture. This mixture was loaded on the HPLC column and mCN and mFe–C at 2,046 and 569 cm-1, respectively [20, 21,
the organic products released from the complex were 32]. The electronic spectrum showed an intense solvent-
promptly characterized by the injection of standards. dependent band with a maximum at 438 nm (e = 4.8 9
103 M-1 cm-1) assigned to a metal-to-ligand charge trans-
Enzyme inhibition assays fer transition, which matches a band found in the (isonico-
tinamide)pentacyanoferrate(II) complex [22]. Furthermore,
Wild-type InhA and I21V InhA were expressed, purified, the cyclic voltammogram with E1/2 at 578 mV was consis-
and characterized as described in the literature [28]. Inhi- tent with an isonicotinamide ligand bound to a pentacyano-
bition assays were performed as described elsewhere ferrate(II) complex.
[28, 31]. Briefly, experiments on time-dependent inacti- HPLC was used to further identify and quantify the
vation of the enoyl reductases were conducted in production of pyridine derivatives. It is well known that
100.0 mM NaH2PO4/Na2HPO4, pH 7.5 at 25 °C, with pentacyanoferrate complexes are very hydrophilic and their
either 3.0 lM wild-type InhA or 3.0 lM I21V, in the presence separation on reverse-phase columns is poor and close to
of 100.0 lM complexes. Aliquots were taken at specified the void time; hence, a new strategy was developed to take
times, and steady-state enzyme activities were determined full advantage of this technique. Since organic ligands

123
J Biol Inorg Chem (2012) 17:275–283 279

usually interact well with C18 reverse-phase columns, the and 8.0, respectively). Therefore, the reaction with
strategy was to dislodge these ligands from the pentacy- [FeIII(CN)5(H2O)]2- is at least 1,000 times faster than the
anoferrate moiety for this study. Taking into account the outer-sphere electron transfer reaction using [FeIII(CN)6]3-
well-known use of DMSO in substitution reactions of under identical conditions. Thus, it supports an inner-
pentacyanoferrate(II) complexes [33, 34], we made the sphere mechanism where isoniazid is oxidized by
isolated complex react with DMSO in a pseudo-first-order [FeIII(CN)5(H2O)]2- producing [FeII(CN)5(izd*)]3-, where
condition and investigated the products by HPLC. The izd* are products of oxidation, such as isonicotinamide and
DMSO molecule displaces the sixth ligand, thus forming isonicotinic acid. Another relevant point in this study is a
[Fe(CN)5(dmso)]3- in a reaction in which the aquation is significant pH dependence for outer-sphere oxidation. As
the rate-determining step. Two peaks assigned to isonico- far as we know, no connection between mycobacterial
tinamide (major, over 90%) and isonicotinic acid (minor) enzymatic oxidation and acid–base assistance has been
were detected, meaning that the reaction of pentacyano- reported. Previously, we reported that pyridine thioamide
ferrate(III) with isoniazid resulted in the formation of compounds showed a similar pH-dependent behavior, but
mainly [Fe(CN)5(isn)]3- (where isn is isonicotinamide) at higher pH [20]. Thus, it is evident that a base assistance
and impurities of [Fe(CN)5(isa)]3- (where isa is isonicot- can accelerate catalytic oxidation of these compounds,
inic acid). These results are consistent with the mechanism which might also take place in the active sites of the
suggested for acylhydrazine decomposition [35] and are activating enzymes KatG and EthA. Isoniazid-activating
in agreement with isoniazid oxidation investigated using enzyme KatG, a heme catalase–peroxidase, has a con-
mycobacterial enzymes and manganese compounds served amino acid triad of arginine–tryptophan–histidine in
[35, 36]. the active site, which could work well in acid–base catal-
To further provide solid evidence for the iron redox state ysis. The X-ray structures available for three KatG
in this isolated complex, Mössbauer spectroscopy was enzymes from M. tuberculosis [Protein Data Bank (PDB)
employed. The Mössbauer spectra of iron pentacyano ID 1SJ2)], Haloarcula marismortui (PDB ID 1ITK), and
complexes are shown as doublets owing to the presence of Burkholderia pseudomallei (PDB ID 1MWV) showed that
an electronic field gradient with noncubic symmetry in the these residues are close to the heme group. In addition,
57
Fe nucleus [37]. We observed a doublet with an isomer Wengenack et al. [38] showed by NMR spectroscopy that
shift (d) and quadrupolar splitting (D) of 0.229 ± 0.003 the acylhydrazine group of isoniazid is close to the heme
and 0.893 ± 0.004 mm/s, respectively, which is a typical center of KatG, which would support a hypothesis of
profile for low-spin iron(II) cyano complexes. Our results acid–base assistance during catalysis.
are consistent with NMR profiles, which indicate charac-
teristics of diamagnetic species. Isoniazid and metal complex oxidations

Kinetics of the electron transfer reaction The reaction of isoniazid with an excess of [FeIII(CN)6]3-
was monitored by HPLC using a UV–vis diode array.
As mentioned earlier, an electron transfer process is Figure 1 shows the chromatogram of the final products,
involved in the reaction of isoniazid with where four peaks with retention times of 2.3, 3.2, 5.2, and
[FeIII(CN)5(H2O)]2- (note ammonia is quickly released 10 min were observed and assigned to [FeIII(CN)6]3-,
upon dissolution in water, resulting in the aquo compound). isonicotinic acid, isonicotinamide, and 4-pyridinecarbo-
This reaction can occur through an inner-sphere or an xaldehyde, respectively, on the basis of the electronic
outer-sphere mechanism, although a mixture of both is also spectra and injection of standards. These pyridine deriva-
possible. To study this mechanism, stopped-flow mea- tives matched those observed for in vivo and in vitro
surements were performed under pseudo-first-order reac- enzymatic oxidation of isoniazid [35, 36, 39, 40]. Inter-
tion conditions for isoniazid at 438 nm (pH 7.0). kobs of estingly, the relative proportion of isonicotinamide and
12 s-1 was determined and was independent of the con- isonicotinic acid switched on altering the isoniazid
centration, indicating an inner-sphere mechanism. To fur- concentration and this indicates that isonicotinic acid pro-
ther validate the data, the contribution of an outer-sphere duction strongly decreased at a concentration above
mechanism using hexacyanoferrate(III) ([FeIII(CN)6]3-) 20 mM, whereas isonicotinamide production increased.
was examined. This compound, with E1/2 = 450 mV, This behavior has also been observed for enzymatic reac-
should react with isoniazid through the outer-sphere path- tion of KatG, which highlights a suitable use of this
way faster than [FeIII(CN)5(H2O)]2- (E1/2 = 390 mV) on oxidizing reactant for modeling and reactivity comparison
the basis of Marcus theory. The kinetic data for the hexa- [35, 40].
cyanoferrate reaction showed a second-order rate with kobs Manganese complexes have been used as a model
dependent on the pH (k = 0.5 and 41 M-1 s-1 for pH 7.0 reactant for the isoniazid activation through oxidation

123
280 J Biol Inorg Chem (2012) 17:275–283

Fig. 1 Chromatogram for the reaction of isoniazid (2 mM) with


hexacyanoferrate(II), in molar excess, pH 7.0. Isocratic elution at
10% acetonitrile pH 3.5, flow rate of 1.0 mL min-1, monitored at
260 nm, after 1 h of reaction at 25 °C

[35, 36]. This reaction conducted in the presence of NAD?


leads to the formation of NAD–isoniazid adducts. To
evaluate the potential use of [FeIII(CN)6]3- instead of
manganese complexes, isoniazid was mixed with NAD?,
which triggered a reaction upon addition of [FeIII(CN)6]3-
at pH 7.0. The chromatogram showed two new peaks at 7.1
Fig. 2 Chromatogram for the reaction of hydrogen peroxide
and 8.2 min, which were not seen in the standards or (66 mM) with isoniazid (1 mM) (a) and (isoniazid)pentacyanofer-
control mixtures (isoniazid plus [FeIII(CN)6]3-, NAD? plus rate(II) (b). Isocratic elution at 10% acetonitrile pH 3.5, flow rate of
isoniazid, or NAD? plus [FeIII(CN)6]3-) examined for 1.0 mL min-1, monitored at 260 nm, after 6 h of reaction at 25 °C
identical incubation time and medium. Those peaks were
found in the electronic spectra, which showed typical bands chromatograms obtained for both reactions, indicating
at 260 and 330 nm consistent with dihydropyridine com- general similarities (Fig. 2). However, remarkable changes
pounds. In addition, A260 nm/A330 nm absorbance ratios of in the ratio of isonicotinic acid to isonicotinamide might
about 3.2 were obtained, which is consistent with NAD– reflect a preferential reaction pathway, suggesting that the
isoniazid adducts described in the literature [15, 41]. metal bonding could be influencing the isoniazid reactivity.
Optimization of product formation and the conditions to For free isoniazid, the relative area of the peak assigned to
obtain better yields is clearly necessary and is under way. isonicotinamide is more intense than that of isonicotinic
However, this result suggests the possibility of employing a acid. However, the opposite profile was seen for the reaction
very common and clean outer-sphere oxidizing compound, containing isoniazid coordinated to pentacyanoferrate(II)
[FeIII(CN)6]3-, to prepare these important adducts. ([FeII(CN)5(izd)]3-). Also, after 5 h of reaction more iso-
Oxidative reaction of isoniazid (1 mM) promoted by niazid was oxidized in the reaction with the complex than in
hydrogen peroxide (66 mM) exhibited kinetics slower than the reaction with the free ligand (Fig. 2). Two pathways
initially expected in comparison with [FeIII(CN)6]3- oxi- have been proposed for the formation of isonicotinamide
dation (Fig. 2). This was also found in previous reports, and isonicotinic acid and are based on the origin of the
where it was reported that a slow reaction with hydrogen radical produced as well as the initial concentration of
peroxide was indeed accelerated by the catalytic effect of isoniazid [35]. Isonicotinamide is produced by a nitrogen-
the catalase–peroxidase KatG enzyme [14]. In the work based radical that decomposes by a bimolecular reaction,
reported in the literature, 200 lM isoniazid and 25 mM whereas isonicotinic acid is produced by an acyl-based
H2O2 in the presence of KatG was used and yet still radical in a reaction with water. When this latter radical is
resulted in incomplete reaction even after 6 h [14]. The engaged in resonance, the p-back-bonding effect is a rea-
reaction performed in our studies produced isonicotin- sonable explanation for the observed result. This effect may
amide as the major product despite it being used at rela- favor the stabilization of p-resonant radicals, thus enhanc-
tively low isoniazid concentration. To evaluate and ing the formation of the acyl radical. Hence, the p-back-
compare the reactivity of free isoniazid and isoniazid bonding phenomenon manifested in pentacyanoferrate(II)
coordinated to pentacyanoferrate(II), the oxidation of this complexes can be used as a way to adjust isoniazid
metal complex with hydrogen peroxide was conducted. reactivity. One interesting example is 4-pyridine-
The peaks assigned to isonicotinic acid, isonicotinamide, carboxaldehyde, which in solution is found to react with
and 4-pyridinecarboxaldehyde were observed in the water and reaches an equilibrium between the hydrated

123
J Biol Inorg Chem (2012) 17:275–283 281

form (alcohol) and the aldehyde. The free ligand produces the InhA enzyme either in the presence or in the absence of
an equilibrium constant of 0.84, but once the ligand is NAD(H) or even in its reduced form, as illustrated in
coordinated to pentacyanoferrate(II), equilibrium constant Fig. 3. The data suggest the isoniazid moiety is still a major
shifts to of 0.48. What is particularly interesting is that and relevant pharmacophore in this compound. However, it
aldehyde formation is favored. This new equilibrium is due is evident that cyanoferrate has altered the mechanism of
to the extended electronic density delocalization of the p action of isoniazid. This new metal compound inhibits
orbitals in the back-bonding, which favors the aldehyde InhA enzyme without KatG, bypassing the enzymatic
instead of the hydrated form (alcohol). In our studies, the activation. This behavior could explain the action of
favored product formed was isonicotinic acid and upon IQG607 against an isoniazid-resistant strain as was recently
oxidation it might have important functional implications observed [30]. The mechanism we predicted was based on
since it has been suggested that the acyl radical is the an electron transfer reaction scheme (Fig. 4) in which an
‘‘preactive’’ form of isoniazid in vivo [3]. Therefore having inner-sphere reaction would be used to promote faster
a higher production level or a longer lifetime for this radical isoniazid oxidation, bypassing the enzymatic activation as
could improve the action and performance of isoniazid as a discussed elsewhere [20]. The metal atom, which is
reactivity strategy. Further investigations of radical forma- kinetically easier to oxidize, triggers the oxidation of the
tion employing EPR spectroscopy and suitable spin traps coordinated isoniazid, thus forming activated species. To
are under way. investigate our mechanistic hypothesis, we prepared two
other isoniazid metal complexes, [Ru(NH3)5(izd)]2? and
InhA inhibition assays [Ru(CN)5(izd)]3-. The former has a lower electrochemical
potential (E1/2 = 356 mV) than IQG607, whereas the latter
As previously reported [28] [FeII(CN)5(izd)]3- (IQG607) has a much higher value (E1/2 * 1,000 mV). Remarkably,
showed very efficient enzymatic inhibition of wild-type both compounds were unable to inhibit InhA (see Fig. 4)
InhA and its isoniazid-resistant mutant InhA I21V. Its even though [Ru(CN)5(izd)]3- exhibited high similarity to
ability to inhibit the enzyme even in the absence of KatG [FeII(CN)5(izd)]3- (IQG607), including its overall size and
and NAD(H) was also remarkable and suggested its charge. The electrochemical potentials of both compounds,
potential use as a metallodrug against KatG- and InhA- however, are very different, thus reinforcing the potential
resistant strains. More recently, it was shown that this role of an intramolecular electron transfer reaction in InhA
compound is indeed effective against isoniazid-sensitive inhibition. Furthermore, evaluation of toxicity has shown
and isoniazid-resistant strains [30]. A comparative inhibi-
tion study was done with [FeII(CN)5(izd)]3- and
[FeIII(CN)6]3- to evaluate the role of the inorganic moiety.
It was observed that [FeIII(CN)6]3- had no ability to inhibit

100
% Activity Remaining

80

60

40

20

Fig. 4 Self-activating anti-tuberculosis metallodrug scheme. Two


0 routes to generate activated drug are shown. In the first route, as
0 20 40 60 proposed in the literature, free isoniazid is enzymatically converted to
TIME (minutes) an active species, but this has suffered from resistance, particularly
deletion or mutation in key activating enzymes, which has blocked
Fig. 3 Assay of inactivation of wild-type InhA (3 lM) by 100 lM this pathway. The alternative route proposed here relies on the
[Fe(CN)6]3- (circles), 100 lM [Ru(NH3)5(izd)]2? (where izd is generation of the active species using an intramolecular electron
isoniazid) (triangles), 100 lM [Ru(CN)5(izd)]3- (squares), and transfer reaction pathway promoted by a metal complex which would
100 lM [Fe(CN)5(izd)]3- (IQG607) (diamonds), at 25 °C, as bypass the enzymatic system of activation, thus leading to isoniazid
described in ‘‘Materials and methods’’ function even against isoniazid-resistant strains

123
282 J Biol Inorg Chem (2012) 17:275–283

that IQG607 shows promise as a viable candidate for fur- overcoming the necessity for KatG. The inhibition data for
ther development as an anti-TB agent on the basis of tox- InhA show KatG is not required, thus supporting our pro-
icological profile assays and the ability to mass produce it posal. The InhA inhibition experiment conducted with
on a large scale [28]. ruthenium(II) (isoniazid)pentamine shows that it does not
inhibit InhA (Fig. 3); however, it has a much lower elec-
trochemical potential than the cyanoferrate complex. The
Conclusions (isoniazid)pentacyanoruthenate(II) complex was also unable
to inhibit InhA, likely because of its very high electro-
As reported in the literature, isoniazid must be activated by chemical potential which was harder to reach. Further studies
electron transfer reactions in vivo, which depends on KatG are under way to provide evidence for this mechanism as well
enzyme. It is also known that the highest percentage of as to examine other analogous metal complexes to support
isoniazid-resistant strains is based on katG gene deletion or this hypothesis. Our data reinforce the idea that electro-
mutation in approximately 50% of strains. These data chemical potential might be a key to design these novel
indicate that there is an obvious need for the development inhibitors. Interestingly, a somehow related process was
of drugs that could overcome TB resistance and yet are still reported that was based on the enhanced activation of iso-
keep the isoniazid efficacy against M. tuberculosis. To niazid activation by KatG in the presence of Mn2?. It was
accomplish this goal we proposed an isoniazid metal proposed that transient Mn3? would be produced by KatG,
complex that could be self-activated in vivo by endogenous which would oxidize isoniazid and reduce the metal back to
oxidizing species found in macrophages. We were able to Mn2?, which is reminiscent of our proposal [43]. However,
find the mechanism of oxidation of isoniazid with this proposed mechanism is still unclear and it is unknown
[FeIII(CN)5(H2O)]2-, followed by a preferential inner- whether it is fully viable in vivo. We are currently trying to
sphere electron transfer process. This concept supports our answer this question with the complexes used in our own
earlier proposal to design ‘‘self-activating’’ metallodrugs studies.
against TB [20]. On the basis of our data, isoniazid is Another important point in our anti-TB metallodrug
intramolecularly oxidized by pentacyanoferrate(III), which design is the use of an iron compound as a deadly bait.
gives us hope to generate an activated drug. Another Following this similar strategy, we believe it might facili-
interesting observation was that the coordinated pentacy- tate drug uptake by taking advantage of the iron trans-
anoferrate(II) has likely favored a pathway for isonicotinic porters, which could serve as a shuttle for this metallodrug.
acyl radical formation resulting in the major production of This could overcome one of the drawbacks in anti-TB drug
isonicotinic acid. Despite current debate regarding the design and therapy, namely, improving the bioavailability
isoniazid activation, several reports suggest that an acyl of the drug in M. tuberculosis, but clearly this idea remains
radical is the actual active isoniazid intermediate [35, 42]. to be proven. However, this kind of strategy can potentially
This species would be directly involved in the reaction with serve as a platform for other pathogenic systems such as
NAD(H) to produce NAD–isoniazid adducts in vivo as malaria.
well as isonicotinic acid in in vitro experiments.
Our proposal for how this complex could work in vivo as Acknowledgments The authors thank the Brazilian agencies CNPq
and CAPES for financial support. This work was also supported by
an anti-TB metallodrug is quite straightforward. It is known the National Institute of Science and Technology on Tuberculosis
that M. tuberculosis faces a strong oxidizing environment (Decit/SCTIE/MS-MCT-CNPq-FNDCT-CAPES) and the Millennium
inside the macrophage, where it is able to supply endogenous Initiative Program (CNPq), Brazil, to D.S.S. and L.A.B.. D.S.S. also
activating agents such as hydrogen peroxide and superoxide. acknowledges a grant awarded by BNDES. L.G.F.L. (301478/2008-2),
I.C.N.D. (303530/2008-1), D.S.S. (304051/1975-06), and L.A.B.
Therefore, we came up with the idea to have a metal complex (520182/99-5) are research career awardees of the National Council for
of isoniazid where the metal unit can be quickly oxidized Scientific and Technological Development of Brazil (CNPq). We are
then subsequently trigger isoniazid activation intramolecu- also thankful to CENAUREMN/UFC and Douglas W. Franco for the
larly (Fig. 4); hence, it would no longer require the presence use of NMR and stopped-flow equipment.
of activating enzymes such as KatG. Now instead, this metal
compound would serve as a way to kill isoniazid-resistant
strains. Our data obtained using the pentacyanoferrate References
complex support this hypothesis. Other pyridine thioamide
and isoniazid drugs have also been reported to support this 1. Blanchard JS (1996) Annu Rev Biochem 65:215–239
concept [20, 28]. Our proposed mechanism, tested with these 2. World Health Organization (2003) The World Health Organiza-
tion Global Tuberculosis Program. http://www.who.int/gtb/
metallodrugs, as shown in Fig. 4 indicates that an
3. Zhang Y (2005) Annu Rev Pharmacol Toxicol 45:529–564
inner-sphere electron transfer reaction occurs between the 4. World Health Organization (2000) Anti-tuberculosis drug resis-
metal and isoniazid, thereby activating this prodrug and tance in the world. World Health Organization, Geneva

123
J Biol Inorg Chem (2012) 17:275–283 283

5. Goldman RC, Plumley KV, Laughon BE (2007) Infect Disord 24. Toma HE, Batista AA, Gray HB (1982) J Am Chem Soc
Drug Targets 7(2):73–91 104(26):7509–7515
6. Schroeder EK, de Souza N, Santos DS, Blanchard JS, Basso LA 25. Leal JM, Garcia B, Domingo PL (1998) Coord Chem Rev
(2002) Curr Pharm Biotechnol 3(3):197–225 173:79–131
7. Zhang Y (2003) In: Rom W, Garay S (eds) Tuberculosis. Lip- 26. Olabe JA (2004) Adv Inorg Chem 55:61–126
pincott, New York, pp 739–758 27. Chen MH, Lee S, Liu S, Yeh A (1996) Inorg Chem
8. Ducati RG, Ruffino-Netto A, Basso LA, Santos DS (2006) Mem 35(9):2627–2629
Inst Oswaldo Cruz 101(7):697–714 28. Oliveira JS, Sousa EHS, Basso LA, Palaci M, Dietze R, Santos
9. Banerjee A, Dubnau E, Quemard A, Balasubramanian V, Um KS, DS, Moreira IS (2004) Chem Commun 312–313
Wilson T, Collins D, de Lisle G, Jacobs WR Jr (1994) Science 29. Oliveira JS, de Sousa EHS, de Souza ON, Moreira IS, Santos DS,
263(5144):227–230 Basso LA (2006) Curr Pharm Des 12(19):2409–2424
10. Argyrou A, Jin L, Siconilfi-Baez L, Angeletti RH, Blanchard JS 30. Basso LA, Schneider CZ, dos Santos AJAB, dos Santos AA,
(2006) Biochemistry 45(47):13947–13953 Campos MM, Souto AA, Santos DS (2010) J Braz Chem Soc
11. Gupta RK, Thakur TS, Desiraju GR, Tyagi JSJ (2009) Med Chem 21(7):1384–1389
52(20):6324–6334 31. Basso LA, Zheng RJ, Blanchard JS (1996) J Am Chem Soc
12. Murphy DJ, Brown JR (2008) Curr Opin Microbiol 118(45):11301–11302
11(5):422–427 32. Nakamoto K (1997) Infrared and Raman spectra of inorganic and
13. Sousa EHS, Tuckerman JR, Gonzalez G, Gilles-Gonzalez MA coordination compounds. Wiley, New York
(2007) Protein Sci 16(8):1708–1719 33. Toma HE (1978) J Coord Chem 7(4):231–238
14. Magliozzo RS, Marcinkeviciene JA (1996) J Am Chem Soc 34. Toma HE, Martins JM, Giesbrecht E (1978) J Chem Soc Dalton
118(45):11303–11304 Trans 1610–1617
15. Nguyen M, Claparols C, Bernadou J, Meunier B (2001) Chem- 35. Bodiguel J, Nagy JM, Brown KA, Jamart-Gregoire B (2001)
biochem 2(12):877–883 J Am Chem Soc 123(16):3832–3833
16. Wilming M, Johnsson K (1999) Angew Chem Int Ed 36. Nguyen M, Claparols C, Bernadou J, Meunier B (2002) Cr Chim
38(17):2588–2590 5(4):325–330
17. Rawat R, Whitty A, Tonge PJ (2003) Proc Natl Acad Sci USA 37. Dickson DPE, Berry FJ (1986) Mossbauer spectroscopy. Cam-
100(24):13881–13886 bridge University Press, London
18. Yu SW, Girotto S, Lee C, Magliozzo RS (2003) J Biol Chem 38. Wengenack NL, Todorovic S, Yu L, Rusnak F (1998) Bio-
278(17):14769–14775 chemistry 37(45):15825–15834
19. Ramaswamy SV, Reich R, Dou SJ, Jasperse L, Pan X, Wanger A, 39. Wang F, Langley R, Gulten C, Dover LG, Besra GS, Jacobs WR
Quitugua T, Graviss EA (2003) Antimicrob Agents Chemother Jr, Sacchettini JC (2007) J Exp Med 204(1):73–78
47(4):1241–1250 40. Johnsson K, Schultz PG (1994) J Am Chem Soc 116(16):
20. Sousa EHS, Pontes DL, Diogenes IC, Lopes LG, Oliveira JS, 7425–7426
Basso LA, Santos DS, Moreira IS (2005) J Inorg Biochem 41. Lei B, Wei CJ, Tu SC (2000) J Biol Chem 275(4):2520–2526
99(2):368–375 42. Amos RIJ, Gourlay BS, Schiesser CH, Smith JA, Yates BF
21. Moreira ID, Franco DW (1994) Inorg Chem 33(8):1607–1613 (2008) Chem Commun 1695–1697
22. Toma HE, Malin JM (1973) Inorg Chem 12(5):1039–1045 43. Magliozzo RS, Marcinkeviciene JA (1997) J Biol Chem
23. Moreira IS (1990) Sı́ntese e Caracterização de Compostos Mono 272(14):8867–8870
e Binucleares de Ru e Fe: Estudos das Reações de Transferência
de Elétrons‘‘, PhD Thesis, In: Departamento de Quı́mica, Uni-
versidade de São Paulo, São Carlos

123
Copyright of Journal of Biological Inorganic Chemistry is the property of Springer Science & Business Media
B.V. and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright
holder's express written permission. However, users may print, download, or email articles for individual use.

You might also like