Abstract
Pyrrolobenzodiazepines (PBDs) are sequence selective DNA alkylating agents with remarkable antineoplastic activity. They are either naturally produced by actinomycetes or synthetically produced. The remarkable broad spectrum of activities of the naturally produced PBDs encouraged the synthesis of several PBDs, including dimeric and hybrid PBDs yielding to an improvement in the DNA binding sequence specificity and in the potency of this class of compounds. However, limitation in the chemical synthesis prevented the testing of one of the most potent PBDs, sibiromycin, a naturally produced glycosylated PBDs. Only recently the biosynthetic gene clusters for PBDs have been identified opening the doors to the production of glycosylated PBDs by mutasynthesis and biosynthetic engineering. The present review describes the recent studies on the biosynthesis of naturally produced pyrrolobenzodiazepines. In addition, it provides an overview on the isolation and characterization of naturally produced PBDs, on the chemical synthesis of PBDs, on the mechanism of DNA alkylation, and on the DNA binding affinity and cytotoxic properties of both naturally produced and synthetic pyrrolobenzodiazepines.
Keywords: pyrrolobenzodiazepines, DNA alkylation, anticancer, biosynthesis, anthramycin, sibiromycin, tomaymycin, SJG-136, lincomycin, DSB-120
1. INTRODUCTION
The 1,4-benzodiazepine ring system characterizes a class of compounds with diverse chemical structures and biological activities (Fig. 1). Four pharmacophore groups compose this class, the 1,4-benzodiazepine-2-ones, 1,4-benzodiazepine-2,5-diones, the dibenzodiazepinones and the pyrrolobenzodiazepines (PBDs). The most widely used members of the 1,4- benzodiazepines are the 1,4-benzodiazepine-2-ones, whose most prominent member is diazepam (valium®, Hoffmann-La Roche). At least 50 members of this group are clinically approved psychoactive drugs used for the treatment of insomnia, alcohol withdrawal, epileptic seizures, muscles spasms, and anxiety as anxyiolitic1. Some members of the second group, the 1,4- benzodiazepine-2,5-ones, have been tested as panxiolytic2 and anticonvulsant3 as well as peptidomimetic inhibitors4,5. In addition to their medicinal properties they also possess herbicidal activity6. Cyclopenin, the only naturally produced 1,4-benzodiazepine-2,5-one isolated, is particularly interesting as the biosynthetic intermediate of viridicatin7, an inhibitor of replication of HIV8. The naturally produced TLN-4601 (formerly ECO-4601) is the sole member of the dibenzodiazepinones’ group9 with a broad spectrum of antitumor activity in cancer cells and human xenografts10. Phase I/II trials (Thallion Pharma company web site: http://www.thallion.com/en/drug-development/tln-4601.php) are underway. The last group contains several naturally and synthetically produced PBDs. These compounds are minor groove sequence specific DNA alkylating agents with remarkable potency against cancer cells11–13 and are the subject of this review. Excellent reviews have recently covered advances in the synthesis of PBDs12,14–16. Hence, we will report only on significantly bioactive synthetic PBDs as well as recently synthesized PBDs not covered by the above-mentioned review. The main focus of this review will be on recent discoveries in the biosynthesis of naturally produced PBDs and on the biological activities of these compounds.
Figure 1.
The 1,4-benzodiazepine ring system and representative members of the 1,4- benzodiazepine class.
2. ISOLATION AND CHARACTERIZATION OF NATURALLY PRODUCED PYRROLOBENZODIAZEPINES
Since the discovery of anthramycin in 196317 18 others PBDs produced by micrococci and streptomyces have been isolated (Table I, Fig. 2). Sixteen of these possess moderate antimicrobial activity (Fig. 2.A). The three C11-oxo-PBDs (Fig. 2.B) do not display any biological activity and, as discussed in section 3, are believed to be the product of a resistance mechanism present in the RK1441A, tomaymycin and limazepines producers. More recently 10 new pentacyclic pyrrolobenzodiazepines, the circumdatins, have been isolated from terrestrial and marine Aspergillus fungi18–22 (Fig. 3). The chemical structure of the circumdatins have been revisited based on the crystal structures of circumdatins A and B21. Only the isolation, spectroscopic and crystallographic characterization, and chemical synthesis23–25 but not the biosynthetic gene clusters have been reported on the circumdatins. Therefore, this review will not cover the pentacyclic pyrrolobenzodiazepines but it will focus on the bacterial PBDs, hereafter referred to as PBDs.
Table I.
Producers and antimicrobial activity of bacterial PBDs
PBDs | Producer | Antimicrobial activity, two best MICs or inhibition diameter |
---|---|---|
| ||
Anthramycin153,171 |
Streptomyces refuineus thermotolerans NRRL 3143 Strept. spadicogriseus ATCC 31179 |
Staph. aureus: 0.1–5 μg/mL B. subtilis: 0.1–5 μg/mL |
Tomaymycin31,172 | Streptomyces achromogenes ATCC 3143 |
Staph. aureus: 6. μg/mL B. subtilis ATCC-6633: 12.5 μg/mL |
Oxotomaymycin57 | Streptomyces achromogenes ATCC 3143 | No activity |
Sibiromycin173 | Streptosporangium sibiricum |
Staph. aureus: 1 μg/mL B. subtilis: 0.3 μg/mL |
Sibanomicin37 | Micromonospora sp. SF2364 |
Staph. aureus S-424: 12.5 μg/mL B. anthracis 119: 25 μg/mL |
Chicamycin A33 | Streptomyces sp. J576-99 |
Micrococcus luteus PCI 1001: 50 μg/mL Streptococccus pyogenes A20201: 50 μg/mL |
Mazethramycin35 | Streptomyces thioluteus ME561-L4 |
B. subtilis PCI 219: 1.6 μg/mL Xanthomonas oryzae: 1.6 μg/mL |
Prothracarcin174 | Streptomyces umbrosus subsp. raffinophilus DO-62 |
B. subtilis 10707: 50 μg/mL E. coli ATCC 26: 50 μg/mL |
Abbeymycin32 | Streptomyces sp. AB-99F-52 |
B. loeschii ATCC 15930: 16 μg/mL Bacteroides fragilis ATCC 25285: 16 μg/mL |
Porothramycin34 | Streptomyces albus K731-113 |
B. subtilis PCI 219: 1.6 μg/mL Bacteroides fragilis A 22053: 0.8 μg/mL E. coli ATCC 26: 50 μg/mL |
DC-81175,176 | Streptomyces sp. | Not reported |
RK-1441A177,178 | Streptomyces sp. RK-1441 | E. coli BE 1186: 100 μg/mL |
RK-1441B177,178 | Streptomyces sp. RK-1441 | No activity |
Neothramycin A38,179 | Streptomyces sp. MC916-C4 |
Aeromonas salmonecida ATCC14174: 25 μg/mL Xanthomonas oryzae: 50 μg/mL |
Neothramycin B38,179 | Streptomyces sp. MC916-C4 |
Aeromonas salmonecida ATCC14174: 50 μg/mL Xanthomonas oryzae: 100 μg/mL |
Limazepine A180 | Micrococcus sp. ICBB8177 | No activity |
Limazepine C180 | Micrococcus sp. ICBB8177 |
E. coli 10 mm (40 μg/disk) Staph. aureus 18 mm (40 μg/disk) |
Limazepine E180 | Micrococcus sp. ICBB8177 |
E. coli 10 mm (40 μg/disk) Staph. aureus 12 mm (40 μg/disk) |
Limazepine F180 | Micrococcus sp. ICBB8177 | No activity |
Figure 2.
Naturally produced pyrrolobenzodiazepines: (A) bioactive and (B) not bioactive PBDs.
Figure 3.
Revised structures of circumdatins A and B.
All PBDs contain the tricyclic ring system formed by an anthranilate (A), a diazepine (B) and a hydropyrrole (C) moieties shown in figure 4. Different degrees and types of substituents at the A- and C-rings provide chemical diversity among PBDs. For example, PBDs such as sibiromycin and sibanomicin are glycosylated at C7 of the A-ring (Fig. 2). In addition, the ring C can be fully saturated, unsaturated at the C2-C3 bond or exocyclically unsaturated at C2 as in neothramycin, anthramycin and tomaymycin, respectively. The PBDs can be in three difference forms depending on the storage conditions (Fig. 4). In aqueous solution the imine form is in equilibrium with the carbinolamine form and it is stabilized by the presence of electron-withdrawing substituents at C826–28. The carbinolamine ether form is formed in protic solvent such as alcohols and it is in equilibrium with the carbinolamine form29,30. The PBDs are most stable in the carbinolamine ether forms and as such they are usually stored in methanol at −20 °C28. The crystalline imine and the carbinolamine methyl ether forms can be respectively obtained from boiling acetone and methanol-water28. The literature, especially on the naturally produced PBDs as on tomaymycin31 and abbemycin32, has lead to significant confusion by naming the methoxy form and referring to the carbinolamine and imine forms, respectively, as the 11-hydroxy and anhydro derivatives. Particularly indicative are the cases of chicamycin and porothramycin. The methoxy derivative of chicamycin was named chicamycin A, while the imine form of chicamycin was named chicamycin B33. The methoxy derivative of porothramycin was named porothramycin B, while the carbinolamine form was named porothramycin A34. In most cases the carbinolamine form is consider the parent compound such as in mazethramycin35, sibiromycin36 and anthramycin28. However, adding to this confusion, in other cases the imine form is considered such as in sibanomicin37 and neothramycin38. Equilibrium between the three forms, imine, carbinolamine, and methoxy of PBDs not only depends on the solvent used but also on the conditions of the analysis. For example, LC-ESI spectra of PBDs shows three peaks corresponding to the three forms and the ratio of the peaks can be changed by changing the acquisition parameters and the solvent composition39,40. I am proposing to revisit the nomenclature by naming the imine form of PBDs as the parent form considering, as we will see later, that it is the biologically active form of this class of compounds. Figure 2 was drawn to follow this convention.
Figure 4.
Epimerization of C11 in the presence of protic solvent such as water and methanol.
The literature on many naturally produced PBDs is limited to the initial isolation and characterization referenced in table I. Sibiromycin, tomaymycin, anthramycin and, to some extent, neothramycin are the only exceptions with several studies devoted to these compounds. Isolation of naturally produced PBDs entails a general protocol that includes an initial extraction using chloroform or ethyl acetate, followed by silica and gel filtration chromatography. The absorbance spectrum of PBDs has a characteristic peak at ~320 nm. 1D 1H- and 13C-NMR have mainly been used for structural determination of PBDs. I refer to the references of table I for detailed conditions and structural characterization of the naturally produced PBDs. The crystal structure of anthramycin41,42 and tomaymycin43 were pivotal not only to confirm the structure of these compounds but, more importantly, to assign the S-stereochemistry of C11a, which gives the characteristic right-handed twist of the PBD scaffold. The assignment of the structure of sibiromycin was not as straightforward. Spectroscopic characterization of the degradation products of sibiromycin obtained by harsh acidic hydrolysis and, subsequently, by catalytic hydrogenation and base hydrolysis lead to the wrong assignment of the aglycone of sibiromycin with an aromatic pyrrole ring36 further confirmed by synthesis of the degradation products44. Facile oxidation of the dihydropyrrole under the harsh acidic degradation conditions45 used resulted in the incorrect assignment. 1D and 2D NMR studies of sibiromycin lead to the final assignment of the aglycone of sibiromycin46 with a hydropyrrole moiety (Fig. 2). Similarly, the original structure36 of sibirosamine moiety was revisited by detailed spectroscopic47 analysis and further confirmed by synthesis of L-sibirosamine48.
3. BIOSYNTHESIS OF THE PYRROLOBENZODIAZEPINES
Excellent reviews on precursors feeding experiments of the biosynthesis of anthramycin, sibiromycin and tomaymycin are available49,50. In summary, the following conclusions were drawn:
L-Methionine, L-tyrosine and L-tryptophan are the immediate biosynthetic precursors of anthramycin, sibiromycin and tomaymycin51,52 (Fig. 5).
The indole nitrogen of tryptophan and the α-amino nitrogen of tyrosine are conserved in PBDs (Fig. 5)53.
The biosynthesis of the hydropyrrole moiety of anthramycin, sibiromycin and tomaymycin proceed through a common pathway shared also by the biosynthesis of lincomycin, a natural product produced by Streptomyces lincolnensis54,55 (Fig. 6.A and Fig. 7). The pathway includes DOPA formation from L-tyrosine followed by extradiol cleavage to a 4-alanyl-2-hydroxy-muconate-6-semialdehyde, which undergoes cyclization to yield the cyclic α-ketoacid shown in figure 6.A. Removal of two carbon atoms from the side chain results in 4-vinyl-2,3-dihydropyrrole-2-carboxylic acid, considered the branch point of all these biosynthetic pathways53.
Transformation of L-tryptophan to kynurenine by a common biosynthetic pathway for the anthranilate moiety of anthramycin, sibiromycin, and tomaymycin is proposed. For sibiromycin, kynurenine undergoes hydroxylation at C3, methylation at C4 and then transformation to the 3-hydroxy-4-methylanthranilic acid (Fig. 6.B)52. A similar pathway was proposed for anthramycin due to the similarity of the anthranilate moiety between these compounds but it could not be verified based on the impermeability of S. refuineus to anthranilate analogs52. A very speculative pathway in which kynurenine is first hydroxylated at C4, then C5 and eventually converted to the 4,5-dihydroxyanthranilic acid was proposed for tomaymycin (Fig. 6.B)56.
The tomaymycin to oxotomaymycin transformation is attributed to a constitutively enzyme. It is likely not co-regulated with the biosynthetic enzymes because this reaction occurs independently from the time of growth and in a strain incapable of producing tomaymycin57. This can be a resistance mechanism of some PBDs producers.
Resistance to PBDs in producing strain is mainly achieved by changes in cell permeability to the antibiotic and it is regulated with the biosynthesis58.
Figure 5.
Labeling patterns in tomaymycin, sibiromycin and anthramycin. Atoms from Ltryptophan, L-tyrosine, and L-methionine are in grey, bold black and italic/shadow black, respectively.
Figure 6.
Proposed biosynthetic pathways based on the feeding experiments for the hydropyrrole (A) and anthranilate (B) moieties.
Figure 7.
The biosynthesis of the tetrahydropyrrole moiety in lincomycin.
A quarter of a century has passed before another study on the biosynthesis of PBDs was published39,40,59. In the mean time progress on the lincomycin biosynthesis was laying the foundation for future assignments of the PBDs biosynthetic pathways. Characterization of a nonproducing S. lincolnensis, yielded to the isolation from the growth media of 4-propylidene- 3,4-dihydropyrrole-2-carboxylic acid, proposed as an intermediate in the lincomycin pathway (Fig. 7)60. Accumulation of this intermediate is due to the inability in this strain to synthesize the F420 cofactor. Thus, this compound is the substrate of an F420-reductase whose gene was identified when the biosynthetic gene cluster was sequenced61. Limited functional assignment of the enzymes coded by the other genes of the cluster was possible due to the low or no similarity to characterize proteins in Protein Data Bank. Currently, the activities of only two enzymes, LmbB1 and LmbB2, involved in the tyrosine to hydropyrrole transformation in lincomycin biosynthesis have been assigned (Fig. 7). LmbB1 contains the catechol-2,3-dioxygenase signature sequence Hx7FYx2DPxGx3E62 and shares very low sequence similarity to hypothetical glyoxylases. The reaction of purified LmbB1 with DOPA yields a yellow product with absorbance at 414 nm63, which was later assigned to 4-(3-carboxy-3-oxopropenyl)2,3- dihydropyrrole-2-carboxylic acid by capillary electrophoresis coupled to ESI-MS64. The formation of a transient product with absorbance at 378 nm followed by the 414 nm product has been observed spectrophotometrically65. 1H-NMR study of the reaction and spectral comparison with chemical standards support the enzymatic formation of the semialdehyde followed by non-enzymatic cyclization to the cyclic α-hydroxyacid65 (Fig. 7). The activity of LmbB2 was indirectly assigned in vivo by observing conversion of tyrosine to the yellow colored compound only in cells transformed with a plasmid containing lmbB1 and lmbB2 63.
The gene clusters for anthramycin59, sibiromycin39 and tomaymycin40 have been recently identified and contain 25, 26 and 17 open reading frames, respectively (Tables II, III and IV). Functional assignments of the enzymes coded by the gene clusters were made possible by homology analysis, gene inactivation, chemical complementation, and comparison of the sibiromycin, tomaymycin, anthramycin and lincomycin gene clusters. The presence of genes in the sibiromycin and anthramycin gene clusters coding for proteins homologous to characterized enzymes involved in degradation of L-tryptophan to kynurenine confirms the tryptophan origin of the anthranilate moiety in these PBDs proposed by Hurley51,52,66. Chemical complementation experiments support the intermediacy of L-3-hydroxykynurenine, 3-hydroxyanthranilic and 3- hydroxy-4-methylanthranilic acid (Table V). The failure of 4-methyl-3-hydroxyanthranilic acid to rescue production of anthramycin is simply due to the known impermeability of S. refuineus towards this compound52 and, therefore, it is not inconsistent with the intermediacy of this compound in the biosynthetic pathway. Adenylation by the ORF21 A-T didomain of 4-methyl-3- hydroxyanthranilic acid confirms the intermediacy of this compound67. ORF24 based on inactivation and feeding experiment was proposed to be involved in the biosynthesis of the anthranilate moiety of anthramycin59. Hu et al. speculated that ORF24 could be involved in the formation of 3-hydroxykynurenine and L-kynurenine59. However, the absence of a gene encoding a homologous protein in the sibiromycin gene cluster39 raises question on this assignment, especially considering that all the substituents present on the anthranilate moiety of anthramycin are present on sibiromycin. The precise role of this protein in the anthramycin biosynthesis remains unclear. The anthranilate moieties of sibiromycin is C-methylated at C8 and hydroxylated at C9 as in anthramycin, and is hydroxylated at C7 as in tomaymycin. Hydroxylation at C9 is likely to occur prior to the diazepine ring formation due to the similarity of SibC to kynurenine-3-monoxygenases and based on the chemical complementation experiments, which also support C8 methylation to occur after C9 hydroxylation and before diazepine ring formation39 (Fig. 8.A). Tomaymycin contains the same C7 hydroxyl substitution at the anthranilate moiety found in sibiromycin while anthramycin does not. Gene inactivation and chemical complementation experiments confirm the assignment of SibG and TomO as the C7 hydroxylase40 (Table V, Fig. 8.B).
Table II.
Deduced functions of ORFs in the anthramycin biosynthetic gene cluster59
gene | sizea | putative function | NCBI accession number of protein homolog | %identity/s imilarity |
---|---|---|---|---|
ORF1 | 624 | amidotransferase | BAB12569 | 57/68 |
ORF2 | 500 | aldehyde dehydrogenase | CAD30313 | ------- |
ORF3 | 354 | alcohol dehydrogenase | EAO60654 | 50/65 |
ORF4 | 410 | cytochrome P-450 hydroxylase | CAJ23858 | 49/63 |
ORF5 | 352 | methyltransferase | ABW71852 (ORF21) | 47/58 |
TomA | 40/53 | |||
ORF6 | 621 | γ-glutamyltransferase | LmbBA | 79/87 |
SibY | 56/67 | |||
TomL | 50/59 | |||
ORF7 | 487 | FAD oxidoreductase | SibW | 41/48 |
ORF8 | 764 | UvrA-drug resistance pump | SibF | 61/75 |
TomM | 66/78 | |||
ORF9 | 256 | putative hydroxylase/glyoxylase | CAB55527 | 27/40 |
ORF10 | 377 | transporter | EAL16816 | 41/66 |
ORF11 | 89 | none | ---- | ---- |
ORF12 | 169 | extradiol dioxygenase | SibV | 45/59 |
TomH | 44/52 | |||
ORF13 | 302 | tyrosine hydroxylase | SibU | 36/46 |
TomI | 44/52 | |||
ORF14 | 297 | F420-dependent reductase | LmbY | 50/63 |
SibT | 57/70 | |||
TomJ | 47/59 | |||
ORF15 | 276 | unknown | LmbX | 34/41 |
SibS | 32/43 | |||
TomK | 34/42 | |||
ORF16 | 413 | kynureninase | SibQ | 45/57 |
ORF17 | 261 | tryptophan 2,3-dioxygenase | SibP | 58/68 |
ORF18 | 58 | None | ---- | ---- |
ORF19 | 348 | aromatic C-methylltransferase | SibL | 45/54 |
ORF20 | 296 | aryl formamidase | SibK | 45/54 |
ORF21 | 600 | NRPS | SibE | 47/58 |
TomA | 39/52 | |||
ORF22 | 1146 | NRPS | SibD | 41/53 |
TomB | 37/49 | |||
ORF23 | 500 | kynurenine 3-monoxygenase | SibC | 44/58 |
ORF24 | 475 | FAD-oxidoreductase | ABF87356 | 36/50 |
ORF25 | 273 | repressor response regulator | SibB | 33/46 |
Numbers are in amino acids.
Table III.
Deduced functions of ORFs in the sibiromycin biosynthetic gene cluster39
gene | sizea | putative function | NCBI accession number of protein homolog | %identity/s imilarity |
---|---|---|---|---|
sibA | 299 | putative regulator | BAC79018 | 31/43 |
ORF25 | 33/46 | |||
sibB | 94 | none | ----- | ------- |
sibC | 477 | kynurenine 3-monoxygenase | XP_001514157 | 31/48 |
ORF23 | 44/58 | |||
sibD | 1504 | nonribosomal peptide synthetase | ORF22 | 41/53 |
TomB | 42/54 | |||
sibE | 597 | nonribosomal peptide synthetase | ORF21 | 47/58 |
TomA | 40/53 | |||
sibF | 771 | UvrA-drug resistance pump | YP_001822519 | 55/70 |
ORF8 | 61/75 | |||
TomM | 61/75 | |||
sibG | 352 | NADH-dependent flavin oxidoreductase | YP_001823080 | 20/25 |
TomO | 40/49 | |||
sibH | 393 | glycosyltransferase | ABO28818 | 48/57 |
sibI | 332 | dTDP-glucose synthase | AAS79450 | 57/69 |
sibJ | 582 | dTDP-4-keto-6-deoxyglucose 3,5-epimerase | BAC55217 | 48/61 |
sibK | 822 | esterase/aryl formamidase | YP_001676677 | 38/53 |
ORF20 | 45/54 | |||
sibL | 345 | C-methyltransferase | AAL33761 | 38/53 |
ORF19 | 45/54 | |||
sibM | 417 | Sugar C-methyltransferase | AAF01816 | 54/66 |
sibN | 390 | dTDP-4-keto-6-deoxyglucose transaminase | ZP_02842724 | 51/68 |
sibO | 246 | N-methyltransferase | CAA63163 | 38/49 |
sibP | 262 | tryptophan 2,3-dioxygenase | YP_001812683 | 37/57 |
ORF17 | 58/68 | |||
sibQ | 394 | kynurenine hydrolase | CAJ89348 | 50/60 |
ORF16 | 45/57 | |||
sibR | 251 | unknown | AAK59995 | 33/39 |
sibS | 300 | unknown | LmbX | 41/52 |
ORF15 | 32/43 | |||
TomK | 43/52 | |||
sibT | 293 | F420-dependent reductase | LmbY | 54/67 |
ORF14 | 57/70 | |||
TomJ | 53/64 | |||
sibU | 318 | tyrosine hydroxylase | LmbB2 | 39/49 |
ORF13 | 36/46 | |||
TomI | 39/48 | |||
sibV | 152 | L-DOPA 2,3-dioxygenase | LmbB1 | 56/67 |
ORF12 | 45/59 | |||
TomH | 51/58 | |||
sibW | 492 | FAD-dependent oxidoreductase | ORF7 | 41/48 |
sibX | 391 | Transcriptional regulator | YP_001823517 | 21/33 |
sibY | 598 | γ-glutamyltransferase | LmbA | 57/68 |
ORF6 | 56/67 | |||
TomL | 52/62 | |||
sibZ | 337 | methyltransferase | LmbW | 56/65 |
ORF5 | 60/74 |
Numbers are in amino acids.
Table IV.
Deduced functions of ORFs in the tomaymycin biosynthetic gene cluster40
gene | sizea | putative function | NCBI accession number of protein homolog | %identity/similarity |
---|---|---|---|---|
tomA | 614 | nonribosomal peptide synthetase | ORF21 | 39/52 |
SibE | 41/54 | |||
tomB | 1542 | nonribosomal peptide synthetase | ORF22 | 37/49 |
SibD | 42/54 | |||
tomC | 406 | putative 3-deoxy-D_arabinose-heptulosonic 7-phosphate (DHAP) synthase | CAL34108 | 47/58 |
tomD | 677 | phenazine biosynthesis protein PhzE | YP_001348741 | 44/59 |
tomE | 206 | Phenol-2-monoxygenase reductase component | YP_707281 | 36/46 |
tomF | 533 | Phenol-2-monoxygenase oxygenase component | YP_702343 | 63/74 |
NP_927609 | 66/79 | |||
tomG | 234 | O-methyltransferase | YP_001644688 | 47/60 |
tomH | 145 | L-DOPA-2,3-dioxygenase | LmbB1 | 54/59 |
ORF12 | 44/52 | |||
SibV | 51/58 | |||
tomI | 318 | tyrosine hydroxylase | LmbB2 | 48/61 |
ORF13 | 44/52 | |||
SibU | 39/48 | |||
tomJ | 319 | F-420 dependent reductase | LmbY | 50/63 |
ORF14 | 47/59 | |||
SibT | 53/64 | |||
tomK | 287 | unknown | LmbX | 45/56 |
ORF15 | 34/42 | |||
SibS | 43/52 | |||
tomL | 577 | γ-glutamyltransferase | LmbA | 50/59 |
ORF6 | 49/58 | |||
SibY | 52/62 | |||
tomM | 783 | UvrA-drug resistance pump | YP_001822519 | 60/72 |
ORF8 | 66/78 | |||
SibF | 61/75 | |||
tomN | 66 | 4-oxalocrotonate tautomerase | ZP_01514863 | 44/63 |
tomO | 394 | salicylylCoA hydroxylase (NADH-dependent flavin oxidoreductase) | YP_117454 | 25/32 |
SibG | 40/49 | |||
tomP | 637 | anthranilate synthase | T03800 | 42/51 |
TomD | 35/45 | |||
tomQ | 482 | flavin containing amine oxidase | YP_633086 | 32/47 |
ORF24 | 57/71 |
Numbers are in amino acids.
Table V.
Abolishment and rescue of PBD production by gene inactivation and plasmid/chemical complementation39,40,59
Inactivated Gene | Chemical/plasmid complementation | Analog production |
---|---|---|
ORF1 | Not reported | Not reported |
ORF12 | Not reported | Not reported |
ORF19 | Rescued with 3-hydrokynurenine and 3-hydroxyanthranilic acid. Not rescued with kynurenine and 3-hydroxy-4-mehyl-anthranilic acid. |
Not reported |
ORF21-22 | Not reported | Not reported |
ORF23 | Rescued with 3-hydrokynurenine and 3- hydroxyanthranilic acid. Not rescued with kynurenine and 3-hydroxy-4- mehyl-anthranilic acid. |
Not reported |
ORF24 | Rescued with 3-hydrokynurenine and 3- hydroxyanthranilic acid. Not rescued with kynurenine and 3-hydroxy-4- mehyl-anthranilic acid. |
Not reported |
tomA | Rescued by plasmid complementation | Not reported |
tomO | Rescued with 5-hydroxylanthranilic acid | ΔtomO strain produces prothracarcin (Table 1) |
sibA | Not reported | Not reported |
sibC | Rescued with 3-hydrokynurenine, 3-hydroxyanthranilic acid, and 3-hydroxy-4-mehylanthranilic acid. Not rescued with kynurenine. |
Not reported |
sibE | Not reported | Not reported |
sibG | Not reported | ΔsibG strain produces 7-deoxyaglycone of sibiromcyin |
Figure 8.
Biosynthetic pathways for the anthranilate moiety of sibiromycin and anthramycin (A), and of tomaymycin (B).
Tryptophan was proposed as the precursor for the anthranilate moiety of tomaymycin based on the feeding experiments52, even though higher level of incorporations of radiolabeled were observed in tomaymycin when the cells were fed with labeled anthranilic acid compared to labeled L-tryptophan. In addition, in feeding experiment with tryptophan-[5-3H, 7-14C]56 minimal 3H retention (16%) in tomaymycin was measured compared to anthramycin (100%) and sibiromycin (91%). Notably, only in tomaymycin, and not in sibiromycin and anthramycin, incorporation of radiolabeled anthranilic acid was detected, which indicates a possible different biosynthetic pathway for the anthranilate moiety. The sequenced tomaymycin gene cluster is consistent with anthranilic acid as the precursor for the anthranilate moiety of tomaymycin. The presence of genes coding for proteins (TomD, TomP and TomC) with high similarity to enzymes involved in the transformation of chorismate to anthranilic acid and to enzymes involved in the shikimate pathway (Fig. 8.B) confirm the chorismate/anthranilic acid origin of the anthranilate moiety in tomaymycin. The biosynthesis of L-tryptophan in plants and microbes proceeds through chorismate, the product of the shikimate pathway, which is then transformed into anthranilic acid by anthranilate synthetase68. Gene inactivation of tomO and rescue of tomaymycin production with 5-hydroxyanthranilic acid support C7 hydroxylation occurring prior to C8 hydroxylation, assigned by homology search to TomE and TomF. Both reactions occur prior to diazepine ring formation40 (Fig. 8.B).
The biosynthetic pathways proposed for the hydropyrrole moiety of tomaymycin, sibiromycin, and anthramycin is mainly based on homology search, gene clusters comparison and the experimental data mentioned above on the lincomycin biosynthesis. We were able to confirm the tyrosine hydroxylase activity of SibU, ORF13 and TomI and of the extradiol dioxygenase activity of SibV, ORF12 and TomH (Fig. 9, unpublished data). 4-Vinyl-2,3- dihydropyrrole-2-carboxylic acid (Fig. 9) represents the branch point of the three biosynthetic pathways. Tomaymycin contains an ethylidene substituent at C2, while anthramycin and sibiromycin have a propylidene substituent at C2 (Fig. 2). As expected, the methyltransferase homologous to SibZ, ORF5 and LmbW necessary to elongate the C2 chain of the 4-vinyl-2,3- dihydropyrrole-2-carboxylic acid intermediate is not found in the tomaymycin gene cluster40. The methyltransferase is proposed to catalyze also a methyl transfer coupled to a tautomerizaton reaction. Accordingly, the tomaymycin gene cluster contains a gene coding for TomN, a putative tautomerase40, likely involved in the tautomerizaton of the 4-vinyl-2,3-dihydropyrrole-2- carboxylic acid intermediate. The presence of homologous proteins SibS, ORF15, TomK and LmbX in all PBDs and lincomycin gene clusters strongly supports a role for these enzymes in the formation of 4-vinyl-2,3-dihydropyrrole-2-carboxylic acid39. Blast search of the Protein Data Bank fails to identify similar characterized proteins. However, a recent gene inactivation experiment showed the requirement of LmbX for production of lincomycin69. We have proposed that these enzymes catalyze an unusual C-C hydrolysis of 4-(3-carboxy-3-oxo-propenyl)-2,3- dihydropyrrole-2-carboxylic acid similarly to BphD, a C-C bond hydrolase39,70.
Figure 9.
Biosynthetic pathways for the hydropyrrole moiety of sibiromycin, anthramycin, lincomycin and tomaymycin.
4-propylidene-tetrahydropyrrole-2-carboxylic acid is the last intermediate in common in the anthramycin, sibiromycin and lincomycin A biosyntheses (Fig. 9), which is the product of F420- dependent reduction of 4-propylidene-3,4-dihydropyrrole-2-carboxylic acid and accordingly it accumulates in lincomycin strain unable to produce F420 cofactor60. LmbA, ORF6 and SibY contain conserved sequence motifs of F420-dependent reductases. F420 are flavin like cofactors involved in redox reaction substituted with side chain with different number of glutamate residues. Proteins homologous to γ-glutamyltransferases are found in all gene clusters considered consistent with the mechanism proposed (Fig. 9; Tables II, III and IV). The biosynthesis of lincomycin requires an additional reduction of 4-propylidene-tetrahydropyrrole-2-carboxylic acid to yield the propyl side chain. Instead, the biosyntheses of anthramycin and sibiromycin diverge with the oxidation of 4-propylidine-tetrahydropyrrole-2-carboxylic acid to 4-propenyl- 2,3-dihydropyrrole-2-carboxylic acid (Fig. 9). Enzymes involved in this transformation were assigned based on homology search and gene cluster comparison (Tables II and III)39,59. Enzymes involved in the oxidation and amidation of the propenyl side chain should only be present in the anthramaycin and, therefore, were assigned by a process of elimination (Table II)59. The biosynthetic pathway for the sibirosamine moiety in sibiromycin is mainly based on sequence homology to characterized enzymes involved in sugar biosynthesis (Table III, Fig. 10)39. Sibirosamine loading occurs after the PBD scaffold is formed as shown by the formation of the aglycone in the ΔsibG strain (Table V, Fig. 10)39. A clear biosynthetic strategy emerges from these studies in which the anthranilate and dihydropyrrole moieties are fully synthesized before formation of the diazepine ring (Fig. 11). This modular and assembly approach significantly eases future combinatorial biosynthetic studies.
Figure 10.
Biosynthetic pathways for the sibirosamine moiety of sibiromycin and for its transfer to the PBD aglycone.
Figure 11.
Biosynthetic strategy for PBDs.
4. SYNTHETICALLY PRODUCED PYRROLOBENZODIAZEPINES
Monomeric PBDs
Chemical syntheses of naturally occurring PBDs such as anthramycin71–73, DC-8174–76, tomaymycin77 and chicaymycin78 have been reported. Over 60 not naturally occurring monomeric PBDs have been synthesized76,78–90 and are shown in figures 12 and 13. Chemical synthesis of naturally occurring PBDs and their analogs is hampered by the stability of the N10-C11 imine bond, the retention of stereochemistry at C11a, the loss of unsaturation in the C-ring under reductive cyclization and dependency on ring A substituents for the diazepine ring closure (reviewed in 12,16). The predominance of DC-81 analogs (Fig. 12) reflects the relatively ease of these synthetic procedures (9 steps and 18% overall yield75) compared to those for more chemically diverse PBDs (Fig. 13)87,88, and especially for PBDs with C2-endo-exo unsaturation (Fig. 13.B), which are laborious and often plague by modest yields. A common synthetic strategy has been employed for almost all these compounds (Fig. 14) in which the C5-N4 amide bond is formed first then followed by intramolecular cyclization through aza-Witting reaction of azidocarbonyl compounds80, catalytic hydrogenation of nitrocarbonyl compounds38,89, or Swern oxidation of aminohydroxyl compounds82–84,91. The first reaction results in direct formation of the PBD. Reduction of the amide bond with NaBH4 or of the protected carbinolamine with Pd yields the final PBD in the second and third reactions, respectively. C2-exo unsaturation is introduced before the formation of the diazepine ring by Wittig oleifination of the C2 ketone84. The C2-endo unsaturation is introduced after formation of the diazepine ring by reacting a C2 ketone compound with trifluoromethanesulphonic anhydride forming the C2-C3 enoltriflate91. A Stille reaction on the enoltriflate PBD allows introduction of the C2-endo-exo unsaturation88. The C2-C3 enoltriflate was recently used in the synthesis of a 66-member library of C2-aryl PBDs87 (Fig. 15). The novelties in this PBD synthesis are the use of solid-supported palladium catalyst and diethanolamine in the Suzuki coupling and in the subsequent work-up, respectively, and the microwave conditions during the Suzuki coupling. Both modifications improved the time of the reaction and the ease of purification. Some members of this family (55 and 56, Fig. 13) display interesting biological properties (see section 5 and 6)92. The general appeal of the synthetic procedures of PBDs is diminished by the need of highly toxic reagents and by the tedious and lengthy syntheses of PBDs with different substituted anthranilate and C2-endo-exo pyrrole moieties75 such in the synthesis of the sibiromycin aglycone44. In addition, synthetic access to sibiromycin, the PBD with highest affinity to DNA among all synthetically or naturally produced monomeric PBDs (see section 5 and Table VI), and to glycosylated PBDs is prevented by difficulties associated with the synthesis of the sibirosamine sugar48 and efficient conjugation to the core.
Figure 12.
Chemically synthesized analogs of DC-81.
Figure 13.
Chemically synthesized C2-exo (A) and C2-endo-exo (B) unsaturated PBDs, and C2- endo unsaturated aryl-substituted PBDs (C).
Figure 14.
The common retrosynthetic approach for monomeric PBDs.
Figure 15.
Synthetic procedure for a 66-member C2-endo aryl-PBD library.
Table VI.
Compound | 0 h | 4 h | 18 h |
---|---|---|---|
Sibiromycin13 | 15.7 | 15.9 | 16.3 |
Anthramycin13 | 9.4 | 11.2 | 13.0 |
Tomaymycin13 | 1.0 | 2.4 | 2.6 |
DC-8113 | 0.3 | 0.5 | 0.7 |
3484 | 0.9 | 1.6 | 2.3 |
3583 | 3.4 | 4.6 | 5.9 |
3683 | 3.1 | 4.0 | 4.7 |
3783 | 1.8 | 2.3 | 3.5 |
3883 | 1.6 | 2.1 | 2.5 |
3983 | 4.0 | 4.6 | 4.6 |
4184 | 1.5 | 2.0 | 2.4 |
5592 | n/a | n/a | 15.8 |
| |||
DSB-12081 | 10.2 | 13.1 | 15.1 |
SJG-136148 | 25.7 | 31.9 | 33.6 |
61 n=1110 | 37.0 | n/a | n/a |
61 n=2110 | 38 | n/a | n/a |
61 n=3110 | 37.0 | n/a | n/a |
62 n=1110 | 11.0 | n/a | n/a |
62 n=2110 | 14 | n/a | n/a |
62 n=3110 | 13 | n/a | n/a |
63 n=3110 | 28.9 | n/a | n/a |
67 n=2, m=2110 | 21.9 | n/a | 22.7 |
67 n=2, m=3110 | 25.9 | n/a | 26.5 |
67 n=2, m=4110 | 23.1 | n/a | 23.9 |
67 n=3, m=3110 | 12.0 | n/a | 12.9 |
67 n=4, m=4110 | 19.9 | n/a | 20.8 |
69 n=1, m=190 | 9.0 | n/a | 9.0 |
69 n=1, m=290 | 9.0 | n/a | 9.0 |
69 n=2, m=290 | 9.0 | n/a | 9.9 |
69 n=3, m=290 | 6.2 | n/a | 7.1 |
69 n=3, m=290 | 6.1 | n/a | 7.2 |
70 n=5112 | 2.1 | 2.4 | 2.6 |
75117 | 2.9 | 3.4 | 3.6 |
76117 | 1.1 | 1.4 | 2.0 |
A 5:1 = DNA: PBD molar ratio was used at pH 7.00.
Only compounds with a ΔTm higher than 2.0 °C are listed.
Dimeric and hybrid PBDs
A DNA alkylating compound should be specific for a 10 bp sequence to specifically modify a target double-stranded DNA. Most common DNA alkylating agents, including monomeric PBDs, fell short of this requirement. In order to increase the DNA base specificity of PBDs (see section 5), Thurston’s group designed a PBD dimer with two DC- 81 unit linked together by 1,3-propanediyldioxy ether linkage named DSB-12093 (Fig. 16). The length of the diether linker was optimized to 3 methylenes by synthesis of DSB-120 analog with 3, 4, 5 and 6 methylenes94,95 and by comparison of their biological activities96,97 to DSB-120. The synthesized C7-linked dimers have not proven as biologically active as C8-linked98,99. The most noteworthy PBD dimer is SJG-136 (Figure 16). The remarkable biological properties of this compound are discussed in section 6. The increased base sequence specificity and DNA affinity, the interstrand-crosslinking and the good cytotoxicity properties of DSB-120 and SJG-136 encouraged the synthesis of a variety of PBD dimers. Synthesized analogs of DSB-120 and SJG- 136 are fluoride substituted dimers (as an example 57, Fig. 16) 86,100,101, mixed imine-amide and imine-amine dimers (as an example 58, Fig. 16)102,103, and dimers with one of the methylenes of the linker substitutes with a piperazine or antraquinone moieties (as an example 59 and 60, Fig. 16)104,105. These compounds and all the other dimeric PBDs synthesized before 2008 are reported in the excellent review by Cipolla et al.14. A new generation of PBD dimers includes C2-fluoro substituted analogs of DSB-120 and SJG-136 with a piperazine linker86 (61–63, Fig. 16). The biological properties of the C2-aryl monomeric PBDs have recently inspired the synthesis of the dimeric C2/C2′-aryl compound 64 (Fig. 16)106. Because of the insolubility of 61, which prevented in vivo studies, compound 65 (Fig. 16) was synthesized106. The slow reactivity of 65 towards DNA suggests that it is acting as a prodrug. Kamal and coworkers have recently synthesized the monomeric and dimeric β-glucuronide prodrugs (Fig. 17) encouraged by the efficient activation by E. coli β-galactosidase of a previously produced monomeric PBD β-galactoside prodrug107 and on the high level of expression of β-glucuronidase in solid tumors compared to healthy tissues108. Both monomeric and dimeric glucuronide prodrugs are efficiently cleaved in vitro by E. coli β-glucuronidase (Fig. 17) as determined by HPLC-activity assays108. Two main strategies have been adopted for the synthesis of dimeric PBDs (Fig. 18). The first synthesis (Fig. 18.A) includes formation of a 2-nitrobenzoic acid dimer core followed by coupling of the pyrrolidine fragment, Swern mediated cyclization and deprotection of the carbinolamine or reduction yielding the imine106,109. The introduction of C2-exo and endo-exo unsaturation is accomplished through the same synthetic reactions used for the monomers (Fig. 15). The second synthesis (Fig. 18.B) includes coupling of a nitrobenzoic acid compound to a pyrrolidine-2-carboxylate, esterification with dibromoalkanes yielding the dimeric intermediate, which undergoes cyclization after reduction of the nitro groups86,100. The syntheses of 64 and of the difluoro-DSB-120 (Fig. 18) are examples of the first and second strategies, respectively. The synthesis of the glucuronide prodrug is similar to the one adopted for the difluro-DSB-120 (Fig. 18.B) with the additional steps required for the addition of the glucuronide promoiety that is added using triphosgene and dibutyltin dilaurate before formation of the diazepine ring and after reduction of the nitro group108.
Figure 16.
PBD dimers.
Figure 17.
Activation of the dimeric β-glucuronide PBD prodrug.
Figure 18.
Syntheses of the compound 64 (A), a C2-aryl dimeric PBD, and difluro-DSB-120 (B).
The ability to add a second monomeric PBD through a linker to form a dimeric PBD inspired the design of hybrid PBDs in which instead of a second monomeric PBD a different pharmacore is added. The main rational for making these conjugates is to produce a compound with two pharmacophoric heads with different DNA sequence specificity. Classical example is the distamycin-DC-81 hybrid82 designed to exploit the AT and GC rich sequence specificity of distamycin and DC-81, respectively. This review will describe literature on hybrid PBDs published since 2008 (Fig. 19). For coverage of hybrid PBDs reported prior to 2008 I refer to the excellent review of Cipolla et al. 14. Hybrid PBDs 66, 67 and 68 (Fig. 19) with a naphthalimide and phenyl benzimidazole moieties tethered to DC-81 were produced to exploit their DNA intercalating properties and minor groove binding specificity at AT rich sequences, respectively110,111. Additionally, the chromophoric properties of these moieties allow a detailed study of their binding mode to DNA discussed in section 5. Diethylphosphonate (69)90, triazolebenzothiadiazine (70)112, indole (71)113,114, and enediyne (72, 73)115 conjugates were synthesized to increase cytotoxicity and in some cases solubility. Two types of hybrid PBDs synthesized by Thurston et al. are of particular interest116,117. The first type is a dimeric PBD, 74, with a tripyrrole linker with a TATAA base pairs specificity116. The length of the linker allows interstrand cross-linking of DNA that covers roughly one turn of the helix. The second type is the fluorescent 7-diethylaminocoumarin PBD conjugates 75, 76, and 77 117 (Fig. 19). The fluorescent moiety can be utilized as a fluorescent marker in cellular localization studies allowing for the first time to characterize the cellular distribution of PBDs and to correlate it to the cytotoxicity.
Figure 19.
PBD conjugates.
5. DNA ALKYLATING PROPERTIES OF PYRROLOBENZODIAZEPINES
Monomeric PBDs
The inhibition of replication and transcription in cells by anthramycin as well as the high wavelength shift from 333 to 343 nm of the spectrum of anthramycin in the presence of DNA provided the initial clues on the DNA alkylating properties of monomeric PBDs118–121. The reaction of anthramycin with RNA and DNA molecules in vitro show a lack of activity with RNA and a preference for double-stranded over single-stranded DNA122. The binding of anthramycin with DNA stabilizes the DNA as evident by an increase of the Tm of the complex compared to the unbound DNA118,122. Copurification by gel filtration and coprecipitation in alcohol of PBD and DNA provided the first indication that a covalent bond is formed between PBD and DNA118,121. Although the covalent bond formed is generally stable, the reverse reaction can occur albeit very slowly123 but it can significantly speed up at pH below 3122. The reaction of only polydG single stranded DNA molecules, among the four single-stranded DNA molecules polydG, polydC, polydA and polydT tested, pointed to guanine as the reactive site on DNA124. CPK modeling125, footprinting123,126, NMR127–130, and crystalloghraphic131 studies identified as the chemical functionalities involved in the covalent bond the C11 on the PBD scaffold and the exocyclic amino group of guanine base on the DNA (Fig. 20). Alkylation of DNA by anthramycin, sibiromycin and tomaymycin is sequence specific with the following trend for binding preference 5′-Pu-G-Pu>5′-Py-G-Pu>5′-Pu-G-Py>5′-Py-G-Py126,132.
Figure 20.
Mechanism of the formation of the PBD-DNA complex.
The presence of the imine and carbinolamine forms in aqueous solution prompted the proposal of three different mechanisms133,134. The first mechanism entails a direct SN2 attack from the guanine nitrogen on the carbinolamine carbon with water elimination. The second mechanism includes an intramolecular cyclization of the Schiff base formed between the guanine nitrogen and the aldehyde resulting from the opening of the diazepine ring. The third mechanism entails a direct reaction between the imine form and the guanine nitrogen (Fig. 20). Lack of adduct formation with aldehyde analogs ruled out the Schiff base mechanism133–135. The third mechanism is strongly supported over the carbinolamine mechanism by the formation of two diastereomeric tomaymycin-DNA adducts (11S, 11aS and 11R, 11aS) in amount different to the free diastereomers130 (Fig. 21). Chirality is lost in the imine reaction intermediate also in the formation of the anthramycin-DNA adduct131. Further proof of the imine mechanism is the increased reactivity of PBDs with electron-donating substituents at the aromatic ring, which is consistent with a facile protonation of the imine nitrogen and a favored nucleophilic attack26. Recent quantum mechanics/dynamics simulations of the binding of anthramycin to DNA shows that in the imine bond is polarized facilitating nucleophilic attack from the guanine136.
Figure 21.
Four possible modes of binding of monomeric PBD to DNA, the example of tomaymycin-DNA.
The imine group at N10-C11 and the S configuration at C11a are the two chemical features always conserved among naturally occurring PBDs and intrinsically linked to the biological properties of these antitumor natural products. Because of the 11aS stereochemistry the PBD scaffold adopts a right handed twist from the anthranilate to the hydropyrrole ring that is a perfect fit for the minor groove of B-DNA molecule42 (Fig. 22). The right-handed twists of free anthramycin and tomaymycin are 35.4º 41,137 and 9.1º 43, respectively. The right-handed twist increases by few degrees once anthramycin is bound to the DNA131. Significantly, the 11aR PBD with a left-handed twist does not bind to DNA26. Two diastereoisomers can result from the reaction of the imine form of a PBD with DNA, the 11S, 11aS and the 11R, 11aS. The substituents on the PBD scaffold and the local sequence of DNA dictate which diastereoisomer is formed and, thus, the mode of binding. There are four possible modes of binding of a monomeric PBD to DNA (Fig. 21). In the first two, the 11R, 11aS and 11S, 11aS are bound with the anthranilate moiety pointing towards the 5′ end of the modified strand. In the other two, the hydropyrrole moiety is pointing towards the 5′ end. Anthramycin mainly binds to DNA with the hydropyrrole moiety ring on the 5′ end of the modified strand resulting in the 11S, 11aS adduct as proposed first by molecular mechanics calculations30,138,139 and then shown by 2D-NMR studies128,140 and crystallography131. The formation of one adduct of anthrmaycin-DNA is likely due to the significant 35° twist of the anthramycin scaffold and to the steric clashes resulting from the 11R, whose axial bond would move the molecule away from the minor groove131. Hydrogen bonding among the anthranilate substituents and the acrylamide tail to the DNA bases seems to contribute to the stability of the 11S, 11aS adduct131,140. The C9 hydroxyl group, that in solution has a pKa of 8.7124, is protonated in the DNA adduct125.
Figure 22.
Crystal structure of the anthramycin (in black)-DNA (CCACGTTGG decamer, in grey) at 2.3 Å resolution (PDB code 274D). The duplex is represented in CPK in (A) and in sticks in (B).
All four possible orientations have been observed in tomaymycin-DNA complexes as shown by NMR and fluorescence studies using the same DNA oligomer used in the anthramycin studies141 (Fig. 21). Fluorescence studies are possible because of the different fluorescent spectra of the 11S, 11aS and 11R, 11aS diastereoisomers130. Fluorescence of free tomaymycin is lost at basic pHs when the phenolate ion is formed (pKa of hydroxyl group at C8 is 8.0)142. However, at basic pH a quenching of fluorescence was not observed in the tomaymycin-DNA adduct indicating that also in this adduct the pKa is perturbed and the C8 hydroxyl group is protonated142. The equilibrium distribution between the different possible adducts seems to be dependent not only on the chemical structure of the PBD such as the modest right-handed twist of tomaymycin but also on the sequence of DNA adjacent to the guanine base, site of the covalent bond132,141. For example, with the d(CICGAATTCICG) duplex tomaymycin mainly forms only the 11S, 11aS with the hydropyrrole at the 5′ end of the modified strand129. The 11R stereoisomer is partly destabilized by an unfavorable interaction between O4 of tomaymycin and the phosphate oxygen of the DNA backbone129 not present in the dATGCAT duplex.
Binding of anthramycin and tomaymycin to DNA causes very little distortion of the double helix129,131,136. Although, formation of the PBD-DNA adduct induces a modest bending of the DNA around the covalent bond 129,131,132. The degree of bending is more pronounced in the tomaymycin-DNA (8.2–14.5º) than in the anthramycin-DNA (5–8.9º) adduct as judged by gel mobility assay132. Some degree of correlation between the structural flexibility of DNA and the sequence specificity has been observed132 with increased flexibility yielding to an increase in sequence specificity and in reactivity. The pseudo-first order rate constant for the formation of PBD-adducts is directly related to the sequence specificity132. Pseudo-first order rate constants measured for anthramycin varied from 0.1 to 0.005 min−1 at pH 7.6132. The conformational flexibility of the drug and the degree of the right–handed twist, 9.1º for tomaymycin, contribute to explain the lower reactivity of tomaymycin vs. anthramycin and the higher bending of DNA in the tomaymycin covalent adduct131,132. The DNA affinity of anthramycin, sibiromycin, tomaymycin, DC-81 and neothramycin shows the following trend from the highest affinity to the least sibiromycin>anthramycin>tomaymycin>DC-81>neothramycin13,143 (Table VI). Table VI lists all naturally and synthetically produced PBD compounds with a ΔTm above 2.0 °C in complex with calf thymus DNA with the exception of DC-81. Compounds not listed either show a ΔTm lower than 2.0 °C when bound to DNA or their ΔTm has not been reported. The high affinity of sibiromycin for DNA is likely due to the additional interactions between the amino sugar and the DNA backbone27 as inferred from the structure of the anthramycin-DNA adduct. Inhibition of transcription in vitro correlates well with the DNA binding reactivity of these monomeric PBDs1. The molecular basis for the 3 bp sequence selectivity of PBDs and the different reactivity of PBDs is still unclear. Among the synthetic monomeric PBDs only 55 (Fig. 13) has a DNA binding affinity comparable to sibiromycin as judged by thermal denaturation experiments with calf thymus DNA92 (Table VI). High resolution NMR studies shows that the C11S,C11aS diastereoisomer of 55 is bound with the hydropyrrole ring towards 5′ end of DNA of the covalently modified strand (Fig. 23, PBD code 2K4L). The C2-haphthyl ring buried in the minor groove of the DNA establishing van der Waals interactions that significantly contributes to the high binding affinity of 55 92.
Figure 23.
NMR structure of 55 (in sticks)-DNA (in CPK) complex (PDB code 2K4L).
Dimeric and hybrid PBDs
Characterization of a DNA duplex with two molecules of tomaymycin bound to guanine bases separated by six base pairs on opposite strand129,144 provided the structural basis for the design of the first PBD dimer, DSB-12093 (Fig. 16). DSB- 120 forms interstrand cross-links at 6 base pairs specific sequences 5′-PuGATCPy-3′ or 5′-PyGATCPu-3′. When bound to DNA, DSB-120 assumes the 11S, 11aS stereochemistry93,145. DSB-120, the dimeric equivalent of two DC-81, is much more reactive towards DNA than many monomeric PBDs (Table VI). The linker position at C8 is important for activity as shown by the negligible inhibition of transcription measured for the C7-linked homolog of DSB-120146. However, as a result of the enhanced biological properties of monomeric PBDs with endocyclic/exocyclic unsaturation and substitution at C2147 a new PBD dimer, SJG-136, was pursued148. Footprinting experiments indicate that SJG-136 forms interstrand cross-links with a similar base pairs selectivity as per DSB-120 by preferentially binding to 5′-GGATCC-3′ but with higher affinity (Table VI)146. Specificity for the AT bp is possibly due to the hydrogen bonding interaction of the N10 hydrogen and the adenine base148. Recent HPLC-MS studies149 of the cross-links products of SJG-136 to DNA show that dimeric PBDs such as SJG-136 in addition to the interstrand crosslinks form an intrastrand cross-links (Fig. 24) and monoalkylated adducts in the absence of a appropriately spaced guanine base. These results are apparently confirmed by high-field NMR studies149 but at the time of writing this report they have yet to be published. Nonetheless, the above-mentioned data signify that the product distribution of these dimeric PBDs is not as simple as previously believed150 and it can have serious implication for their biological activities.
Figure 24.
Interstrand (A) and intrastrand (B) cross-links of SJG-136 to DNA.
Among the recently synthesized hybrid PBDs, the binding mode and specificity to DNA of 68 and 74 have been studies. Consistent to the majority of PBDs, the C11S,C11aS diastereoisomer of 68 is bound to the DNA. However, it assumes an opposite orientation in the minor groove with the anthranilate moiety, instead of the hydropyrrole, towards the 5′ end of the covalently modified strand151. Structural distortions are apparent both by CD and NMR studies around the covalent bond in the complex111,151. The binding of the PBD also causes a slight unwinding of the double helix as well as a narrowing of the minor groove. The major contribution to the binding affinity of 68 to DNA comes from van der Waals interactions. The N-methylpiperazine moiety is extremely disordered in the complex and fails to establish interactions with the DNA. As mentioned in section 4, 74 is a very remarkable hybrid dimeric PBD with a high affinity towards 5′GCTTATAATGG-3′ (in bold are the sites of the covalent bond with the PBD moieties) with a XL50 of 0.06 μM compared to the XL50 of 0.23 μM of SJG-136116. Energy calculations of the binding of 74 to DNA with differently spaced covalently linked guanines indicate that a 7 bp spacer stabilizes interstrand over intrastrand cross-links. Molecular dynamic simulations suggests that very little distortion of the double helix upon binding occurs and that 74 binding site is 11 bp long116 unprecedented among any PBDs.
6. ANTINEOPLASTIC PROPERTIES OF PYRROLOBENZODIAZEPINES
Monomeric PBDs
The antimicrobial properties of monomeric PBDs are not particularly potent to warrant interest in this class of natural products. However, their antitumor properties are remarkable. The first animal152 and human studies on PBDs have been reported for anthamycin153. Among the 219 patients treated some received crystalline pure anthramycin and others a crude extract. All had unresectable or unresponsive tumors such as lymphomas and sarcomas as well as tumors in the gastrointestinal tract and breast. Almost half of the patients were responsive and some showed a significant decrease in tumor size. No side effects were reported besides ulceration at the injection site153. Using higher daily doses (0.5–1.0 mg/Kg) than those used in the clinical trials (0.02 mg/kg) intestinal necrosis was observed but it was reversed 36 h after drug administration154. However, the identification of cardiotoxic side effects in rats at doses between 0.1–0.5 mg/Kg were most discouraging and suggested a disruption of the electron transport chain in mitochondria155. The diminished lethality of anthramycin in vivo in the presence of CoQ points to inhibition of CoQ-dependent enzymes as the cause of the cardiotoxicity156. The inhibitory form of anthramycin is the quinolinic resonance form of the C9 hydroxyl group. Neothramycin displays broad antitumor activities without bone marrow and neurotoxicity and, more importantly, without any cardiotoxic effects157. Significantly, neothramycin does not have a C9 hydroxyl substituent (Fig. 2). The lack of side effects of neothramycin resulted in Phase I clinical trials that were not successful due to the lack of potency of this compound158. However, neothramycin seems particularly effective in the topical treatment of bladder cancer with complete disappearance of the tumor in 36 % of patients and with more than 50% regression of the tumor in 55 % of patients159. Because the twist of the PBD structure matches very well the twist in the minor groove, it is believed that PBD modification is particularly effective at evading the DNA error correction machinery that would otherwise strip modified nucleobases at damaged positions123. Another attractive property of PBDs is the absence of bone marrow suppression in model organisms, a serious side effect of many chemotherapeutics160.
Structural-activity relationships studies
The availability of naturally produced PBDs and of synthetic PBDs has allowed the determination of their structure-activity relationships (Tables VI and 7). Change in the stereochemistry of C11a from S to R results in PBD compounds devoid of the biological properties of the corresponding S stereoisomers27. Substituting the anthranilate ring with a heterocycle ring such as pyridine, pyrazine, pyrimidine or pyrazole decreases the binding affinity and potency13. The presence of unsubstituted anthranilate moiety in PBDs diminishes their potency13. Bulky substituents at C9 of the anthranilate moiety inhibit DNA binding and diminish the potency of PBDs13. C9 hydroxyl group has been proposed to be the source of the cardiotoxic properties of anthramycin27,155,156. C9 hydroxyl group is likely not significantly contributing to the potency of anthramycin. Compounds 48–50, analogs of anthramycin missing the C9-hydroxyl groups, have similar average GI50 in the NCI cancer cells screen (Table VII)91. Therefore, an unsubstituted C9 is preferred for PBDs with chemotherapeutic properties. The presence of electro-donating groups such as methoxy substituents at C7 and C8 shifts the equilibrium from the carbinolamine to the imine form, increasing the biological activities of PBDs13. As previously mentioned, O-glycosylation at C7 further increases the DNA binding affinity and cytotoxic properties of PBDs as in sibiromycin13. All PBDs with high antitumor activities, such as anthramycin, sibiromycin and tomaymycin, share a common endocyclic or exocyclic C2 unsaturation83,84,91. C2-endo or exo unsaturation is likely to enhance the properties of PBDs by flattening the ring and, thus, improving the fit to the minor groove13. Therefore, C2-endo or exo unsaturation is a prerequisite for optimization of PBDs’s properties. Full saturation at the ring C significantly inhibits DNA binding, while complete unsaturation at the ring C prevents any DNA binding161. The presence of an additional unsaturation at the C2 side chain characterizes the most potent PBDs as in the propenyl side chain of sibiromycin and was the basis of the design and synthesis of the C2-aryl PBDs (Fig. 13). A screening of C2-aryl PBDs identified DHR-417 (52) with mean IC50 values of 3 nM against melanoma, breast and renal cells carcinoma162. The maximum tolerated dose by intraperitoneal administration for this compound is 0.5 mg kg−1. DHR-417 shows promising potency only in renal xenografts studies162.
Table VII.
Average GI50, TGI (dose that inhibits 100% cell growth) and LC50 values of C2-endo-exo PBDs across the 60 cell lines of the NCI screen.
Compound | GI50 (μM) | TGI (μM) | LC50 (μM) |
---|---|---|---|
Anthramycin91 | 0.029 | 0.61 | 12.7 |
SJG-136164 | 0.0074 | 0.00083 for sensitive cells |
0.0071 for sensitive cells |
4288 | 0.023 | n/a | n/a |
4388 | <0.01 | n/a | n/a |
4488 | 0.123 | n/a | n/a |
4688 | <0.01 | n/a | n/a |
4891 | 0.053 | 3.72 | 69.2 |
4991 | 0.013 | 1.32 | 55.0 |
5091 | 0.012 | 0.096 | 0.65 |
52162 | <0.01 | 0.065 | 10.5 |
5489 | <0.01 | n/a | n/a |
C2-F2 DSB86 | 0.42 | 4.2 | 39.8 |
6186 n=1 | 0.014 | 0.76 | 22.4 |
6286 n=1 | 3.9 | 16.2 | 63.1 |
Dimeric and hybrid PBDs
The modest potency of DSB-120 in in vivo studies and its high reactivity with thiols163 lead to the development of SJG-136. SJG-136 is a very potent antiproliferating compound in vitro with GI50 values between 0.14–320 nM and significant cell selectivity164. Interestingly, the activity profile of SJG-136 is different from other known alkylating compounds indicative of a novel mode of action164. Four hours after administration SJG-136 can still be detected in plasma. The half-time for removal of SJG-136 in mice is 0.98 h. Most significantly the remarkable activity of SJG-136 is persistent in vivo showing significant tumor regression especially in melanoma, leukemia and lung xenografts tumor161. An LD50 of 9.06 nM for SJG-136 was determined in efficacy studies on tumor cells taken from lymphocytic leukemia (B-CLL) affected patient11. The high potency of SJG-136 is likely due to the failure of recognition of the PBD-DNA complex by repair proteins as for other PBDs165. The potency, the unique specificity and the low toxicity in animals of SJG-136 prompted the start of phase I clinical trials to evaluate the maximum tolerated dose through intravenous administration and the optimal dosage to be used in phase II clinical trials. The MTD tested for SJG-136 had to be reduced significantly to 45 μg m-2 due to unforeseen side effects such as edema and ascite in surrounding tissue to the injection site (vascular leak syndrome, VLS) and some degree of hepatotoxicity166. The VLS symptoms can be decreased by treatment with steroids and dieteutics167. Based on the company web-site (http://www.spirogen.com/sg2000.php) at the time of writing this review phase II trials of SJG-136 (SG2000) will begin soon.
Among the most recently synthesized PBD compounds, the coumarin PBD conjugates are of particular interest117. The fluorescence properties of these compounds allowed evaluating their cellular distribution and, specifically, whether they can successfully localized in the nucleus. Compound 75 with the ideal three-carbon linker is the most potent and it is the only one among the three tested that predominantly localizes in the nucleus. The less potency of compound 76 can be attributed to a less efficient localization in the nucleus. Consistent with this analysis compound 77, which is only found in the cytoplasm, is the least potent of the three. It is apparent that the cellular distribution of this compounds is a function of the chemical structure of the linker117. Lack of uptake in the nucleus could explain the relatively modest cytotoxic activity of 67 compared to its DNA binding affinity110.
GI50 and TGI50 values support localization of 74 in the nucleus despite its MW of 984116. This compound showed significant cytotoxic activity in the NCI 60 cell lines screen. Other noteworthy compounds recently made are 72 and 73 with GI50 values from 0.28 to 0.92 μM for leukemia, colon, ovarian, renal and breast cancer cells115. The most noteworthy PBDs recently synthesized is the dimer of DHR-417 (64) and its produrg 65 106 that show cytotoxic activity in the picomolar range (Table VIII). Preliminary data indicates that this compound is similarly active in vivo 106. The poor DNA binding affinity of 70 is reflected in modest potency. Weak cytotoxic activity characterizes the difluoro substituted DC-81 dimer with a piperazine linker86. The piperazine linker decreases the cytotoxic activity as evident by comparison of 62 to C2- difluoro-DSB-120 (Table VII). However, the C2-difluoro modification has an even more deleterious effect as shown by the very good GI50 average value for 61 compared to those of 62 (Table VII). Among the phosphonate PBD conjugates, only compound 69 with n=3 and m=1 displays significant cytotoxic activity especially for myeloma90.
Table VIII.
In vitro cytotoxicity of PBDs with IC50 values lower than 0.1 μM with the exception of DC-81.
Compound | IC50 μM [cells type] | ||
---|---|---|---|
Sibiromycin13 | 0.0029 [L1210, leukemia] | 0.000017 [ADJ/PC6, plasmacytoma] | 0.04 [CH1, ovarian] |
Anthramycin13 | 0.022 [L1210, leukemia] | 0.0028 [ADJ/PC6, plasmacytoma] | 0.32 [CH1, ovarian] |
Tomaymycin13 | 0.0037 [L1210, leukemia] | 0.0018 [ADJ/PC6, plasmacytoma] | 0.00013 [CH1, ovarian] |
DC-8113 | 0.38 [L1210, leukemia] | 0.33 [ADJ/PC6, plasmacytoma] | 0.10 [CH1, ovarian] |
DSB-12093 | 0.01 [L1210, leukemia] | 0.0005 [ADJ/PC6, plasmacytoma] | 0.003 [CH1, ovarian] |
| |||
181 | 0.045 [A2780, ovarian] | 0.047 [CH1cisR, ovarian] | 0.059 [CH1, ovarian] |
3284 | n/a | 0.082 [CH1cisR, ovarian] | 0.017 [CH1, ovarian] |
3384 | n/a | 0.056 [CH1cisR, ovarian] | 0.035 [CH1, ovarian] |
3484 | n/a | 0.031 [CH1cisR, ovarian] | 0.031 [CH1, ovarian] |
3683 | 0.0145 [A2780, ovarian] | 0.04 [CH1cisR, ovarian] | 0.016 [CH1, ovarian] |
3983 | 0.07 [A2780, ovarian] | 0.037 [CH1cisR, ovarian] | 0.09 [CH1, ovarian] |
4184 | n/a | 0.084 [CH1cisR, ovarian] | 0.066 [CH1, ovarian] |
DSB-12081 | 0.0072 [A2780, ovarian] | 0.022 [CH1cisR, ovarian] | 0.033 [CH1, ovarian] |
SJG-136148 | 0.000022 [A2780, ovarian] | 0.00012 [CH1cisR, ovarian] | 0.0006 [CH1, ovarian] |
| |||
47181 | 0.005 [KB, epidermal] | 0.020 [HCT116, colon] | 0.050 [K562, leukemia] |
51181 | 0.086 [KB, epidermal] | 0.050 [HCT116, colon] | 0.085 [K562, leukemia] |
74116 | n/a | n/a | 0.037 [K562, leukemia] |
| |||
67110 n=2, m=2 | 0.5 [A2780, ovarian] | 0.8 [PC3, prostate] | 1.6 [MCF7, breast] |
67110 n=2, m=3 | 0.5 [A2780, ovarian] | 0.5 [PC3, prostate] | 1.6 [MCF7, breast] |
67110 n=2, m=4 | 0.5 [A2780, ovarian] | 0.5 [PC3, prostate] | 1.6 [MCF7, breast] |
| |||
6990 n=3, m=1 | 0.17 [A2780, ovarian] | 0.17 [GURAV, oral] | 0.0051 [RPMI8226, myeloma] |
| |||
64106 | 0.0024 [A2780, ovarian] | 0.0014 [CCRF-CEM, all] | 0.028 [RPMI8226, myeloma] |
65106 | 0.0005 [A2780, ovarian] | 0.0001 [CCRF-CEM, all] | 0.21 [SiHa, cervix] |
64106 | 0.033 [A549, NSCL] | 0.015 [DU145, prostate] | 0.0027 [LNCaP-FGC, prostate] |
65106 | 0.019 [A549, NSCL] | 0.0064 [DU145, prostate] | 0.0007 [LNCaP-FGC, prostate] |
| |||
64106 | 0.018 [MCF7, breast] | 0.022 [LOXIMI, melanoma] | 0.0007 [LS174T, colon] |
65106 | 0.031 [MCF7, breast] | 0.053 [LOXIMI, melanoma] | 0.0043 [LS174T, colon] |
Mode of action of PBDs
It is only recently that researchers have started to address the mechanism of PBD-induced cell death. Inhibition of DNA binding by the Sp-1, NF-Y, NF-κB and AP-1 transcription factors occurs in the presence of PBD-distamycin168, of the polyamide GWL-78169 and of the indole PBD 71 170, respectively. Recent genetic profiling of sensitive tumors treated with DHR-417 identified many transcriptional factors in the first 150 up-regulated genes but Sp-1 is not included. DHR-417 was proposed to induce cell apoptosis through an insulin-like growth factor signaling pathway162. Clearly, research aimed at elucidation the mode of action of PBD and, specifically, the identification of the target genes is at its infancy but it is necessary for a successful use of the PBDs as antineoplastic drugs.
Biography
Barbara Gerratana received her degree in Chemistry from the Universita’ degli Studi di Pavia in Italy. Soon after she pursued a Ph.D. in Biochemistry with Profs. W. W. Cleland and P.A. Frey at the University of Wisconsin-Madison. She worked as a post-doc with Prof. C.A. Townsend in the Chemistry Department at The Johns Hopkins University. She joined the faculty of the Department of Chemistry and Biochemistry at the University of Maryland, College Park, where she is now an Assistant Professor. Her current research focuses on the biosynthesis of pyrrolobenzodiazepines and on the structural and mechanistic characterization of NAD+ synthetase, a multifunctional enzyme essential for M. tuberculosis.
References
- 1.Puvvada MS, Forrow SA, Hartley JA, Stephenson P, Gibson I, Jenkins TC, Thurston DE. Inhibition of bacteriophage T7 RNA polymerase in vitro transcription by DNA-binding pyrrolo[2,1-c][1,4]benzodiazepines. Biochemistry. 1997;36(9):2478–2484. doi: 10.1021/bi952490r. [DOI] [PubMed] [Google Scholar]
- 2.Wright WB, Jr, Brabander HJ, Greenblatt EN, Day IP, Hardy RA., Jr Derivatives of 1,2,3,11a-tetrahydro-5H-pyrrolo[2,1-c][1,4]benzodiazepine-5,11(10H)-dione as anxiolytic agents. J Med Chem. 1978;21(10):1087–1089. doi: 10.1021/jm00208a017. [DOI] [PubMed] [Google Scholar]
- 3.Demartino G, Massa S, Corelli F, Pantaleoni G, Fanini D, Palumbo G. Cns Agents - Neuropsychopharmacological Effects of 5h-Pyrrolo[2,1-C] [1,4]Benzodiazepine Derivatives. European Journal of Medicinal Chemistry. 1983;18(4):347–350. [Google Scholar]
- 4.Joseph CG, Wilson KR, Wood MS, Sorenson NB, Phan DV, Xiang Z, Witek RM, Haskell-Luevano C. The 1,4-benzodiazepine-2,5-dione small molecule template results in melanocortin receptor agonists with nanomolar potencies. J Med Chem. 2008;51(5):1423–1431. doi: 10.1021/jm701303z. [DOI] [PubMed] [Google Scholar]
- 5.Cummings MD, Schubert C, Parks DJ, Calvo RR, LaFrance LV, Lattanze J, Milkiewicz KL, Lu T. Substituted 1,4-benzodiazepine-2,5-diones as alpha-helix mimetic antagonists of the HDM2-p53 protein-protein interaction. Chem Biol Drug Des. 2006;67(3):201–205. doi: 10.1111/j.1747-0285.2006.00365.x. [DOI] [PubMed] [Google Scholar]
- 6.Karp GM, Manfredi MC, Guaciaro MA, Ortlip CL, Marc P, Szamosi IT. Synthesis and herbicidal activity of 1H-1,4-benzodiazepine-2,5-diones. Journal of Agricultural and Food Chemistry. 1997;45(2):493–500. [Google Scholar]
- 7.Ross W. Benzodiazepine Alkaloids. In: Brossi A, editor. The Alkaloids. San Diego, California: Academic Press; 1990. pp. 73–79. [Google Scholar]
- 8.Heguy A, Cai P, Meyn P, Houck D, Russo S, Michitsch R, Pearce C, Katz B, Bringmann G, Feineis D, Taylor DL, Tyms AS. Isolation and characterization of the fungal metabolite 3-O-methylviridicatin as an inhibitor of tumour necrosis factor alpha-induced human immunodeficiency virus replication. Antivir Chem Chemother. 1998;9(2):149–155. doi: 10.1177/095632029800900206. [DOI] [PubMed] [Google Scholar]
- 9.McAlpine JB, Banskota AH, Charan RD, Schlingmann G, Zazopoulos E, Piraee M, Janso J, Bernan VS, Aouidate M, Farnet CM, Feng X, Zhao Z, Carter GT. Biosynthesis of diazepinomicin/ECO-4601, a Micromonospora secondary metabolite with a novel ring system. J Nat Prod. 2008;71(9):1585–1590. doi: 10.1021/np800376n. [DOI] [PubMed] [Google Scholar]
- 10.Gourdeau H, McAlpine JB, Ranger M, Simard B, Berger F, Beaudry F, Farnet CM, Falardeau P. Identification, characterization and potent antitumor activity of ECO-4601, a novel peripheral benzodiazepine receptor ligand. Cancer Chemother Pharmacol. 2008;61(6):911–921. doi: 10.1007/s00280-007-0544-2. [DOI] [PubMed] [Google Scholar]
- 11.Pepper CJ, Hambly RM, Fegan CD, Delavault P, Thurston DE. The novel sequence-specific DNA cross-linking agent SJG-136 (NSC 694501) has potent and selective in vitro cytotoxicity in human B-cell chronic lymphocytic leukemia cells with evidence of a p53-independent mechanism of cell kill. Cancer Res. 2004;64(18):6750–6755. doi: 10.1158/0008-5472.CAN-04-1713. [DOI] [PubMed] [Google Scholar]
- 12.Kumar R, Lown JW. Recent developments in novel pyrrolo[2,1-c][1,4]benzodiazepine conjugates: synthesis and biological evaluation. Mini Rev Med Chem. 2003;3(4):323–339. doi: 10.2174/1389557033488097. [DOI] [PubMed] [Google Scholar]
- 13.Thurston DE, Bose DS, Howard PW, Jenkins TC, Leoni A, Baraldi PG, Guiotto A, Cacciari B, Kelland LR, Foloppe MP, Rault S. Effect of A-ring modifications on the DNA-binding behavior and cytotoxicity of pyrrolo[2,1-c][1,4]benzodiazepines. J Med Chem. 1999;42(11):1951–1964. doi: 10.1021/jm981117p. [DOI] [PubMed] [Google Scholar]
- 14.Cipolla L, Araujo AC, Airoldi C, Bini D. Pyrrolo[2,1-c][1,4]benzodiazepine as a scaffold for the design and synthesis of anti-tumour drugs. Anticancer Agents Med Chem. 2009;9(1):1–31. doi: 10.2174/187152009787047743. [DOI] [PubMed] [Google Scholar]
- 15.Kamal A, Reddy KL, Devaiah V, Shankaraiah N, Reddy DR. Recent advances in the solid-phase combinatorial synthetic strategies for the benzodiazepine based privileged structures. Mini Rev Med Chem. 2006;6(1):53–69. doi: 10.2174/138955706775197875. [DOI] [PubMed] [Google Scholar]
- 16.Kamal A, Rao MV, Laxman N, Ramesh G, Reddy GS. Recent developments in the design, synthesis and structure-activity relationship studies of pyrrolo[2,1- c][1,4]benzodiazepines as DNA-interactive antitumour antibiotics. Curr Med Chem Anticancer Agents. 2002;2(2):215–254. doi: 10.2174/1568011023354119. [DOI] [PubMed] [Google Scholar]
- 17.Tendler MD, Korman S. ‘Refuin’: A Non-Cytotoxic Carcinostatic Compound Proliferated by a Thermophilic Actinomycete. Nature. 1963;199:501. doi: 10.1038/199501a0. [DOI] [PubMed] [Google Scholar]
- 18.Rahbaek L, Breinholt J, Frisvad JC, Christophersen C. Circumdatin A, B, and C: Three New Benzodiazepine Alkaloids Isolated from a Culture of the Fungus Aspergillus ochraceus. J Org Chem. 1999;64(5):1689–1692. doi: 10.1021/jo981536u. [DOI] [PubMed] [Google Scholar]
- 19.Rahbaek L, Breinholt J. Circumdatins D, E, and F: further fungal benzodiazepine analogues from aspergillus ochraceus. J Nat Prod. 1999;62(6):904–905. doi: 10.1021/np980495u. [DOI] [PubMed] [Google Scholar]
- 20.Dai J, Carte BK, Sidebottom PJ, Sek Yew AL, Ng S, Huang Y, Butler MS. Circumdatin G, a new alkaloid from the fungus Aspergillus ochraceus. J Nat Prod. 2001;64(1):125–126. doi: 10.1021/np000381u. [DOI] [PubMed] [Google Scholar]
- 21.Ookura R, Kito K, Ooi T, Namikoshi M, Kusumi T. Structure revision of circumdatins A and B, benzodiazepine alkaloids produced by marine fungus Aspergillus ostianus, by Xray crystallography. J Org Chem. 2008;73(11):4245–4247. doi: 10.1021/jo800348d. [DOI] [PubMed] [Google Scholar]
- 22.Zhang D, Yang X, Kang JS, Choi HD, Son BW. Circumdatin I, a new ultraviolet-A protecting benzodiazepine alkaloid from a marine isolate of the fungus Exophiala. J Antibiot (Tokyo) 2008;61(1):40–42. doi: 10.1038/ja.2008.108. [DOI] [PubMed] [Google Scholar]
- 23.Tseng MC, Lai CY, Chu YW, Chu YH. Tin triflate-mediated total synthesis of circumdatin F, sclerotigenin, asperlicin C, and other quinazolino[3,2- a][1,4]benzodiazepines. Chem Commun (Camb) 2009;(4):445–447. doi: 10.1039/b813880j. [DOI] [PubMed] [Google Scholar]
- 24.Liu JF, Kaselj M, Isome Y, Chapnick J, Zhang B, Bi G, Yohannes D, Yu L, Baldino CM. Microwave-assisted concise total syntheses of quinazolinobenzodiazepine alkaloids. J Org Chem. 2005;70(25):10488–10493. doi: 10.1021/jo051876x. [DOI] [PubMed] [Google Scholar]
- 25.Witt A, Bergman J. Total syntheses of the benzodiazepine alkaloids circumdatin F and circumdatin C. J Org Chem. 2001;66(8):2784–2788. doi: 10.1021/jo001696h. [DOI] [PubMed] [Google Scholar]
- 26.Hurley LH, Reck T, Thurston DE, Langley DR, Holden KG, Hertzberg RP, Hoover JR, Gallagher G, Jr, Faucette LF, Mong SM, et al. Pyrrolo[1,4]benzodiazepine antitumor antibiotics: relationship of DNA alkylation and sequence specificity to the biological activity of natural and synthetic compounds. Chem Res Toxicol. 1988;1(5):258–268. doi: 10.1021/tx00005a002. [DOI] [PubMed] [Google Scholar]
- 27.Petrusek RL, Anderson GL, Garner TF, Fannin QL, Kaplan DJ, Zimmer SG, Hurley LH. Pyrrolo[1,4]Benzodiazepine Antibiotics - Proposed Structures and Characteristics of the Invitro Deoxyribonucleic-Acid Adducts of Anthramycin, Tomaymycin, Sibiromycin, and Neothramycin-a and Neothramycin-B. Biochemistry. 1981;20(5):1111–1119. doi: 10.1021/bi00508a011. [DOI] [PubMed] [Google Scholar]
- 28.Leimgrub W, Stefanov V, Schenker F, Karr A, Berger J. Isolation and Characterization of Anthramycin a New Antitumor Antibiotic. J Am Chem Soc. 1965;87(24):5791. doi: 10.1021/ja00952a050. [DOI] [PubMed] [Google Scholar]
- 29.Barkley MD, Cheatham S, Thurston DE, Hurley LH. Pyrrolo[1,4]Benzodiazepine Antitumor Antibiotics - Evidence for 2 Forms of Tomaymycin Bound to DNA. Biochemistry. 1986;25(10):3021–3031. doi: 10.1021/bi00358a043. [DOI] [PubMed] [Google Scholar]
- 30.Remers WA, Mabilia M, Hopfinger AJ. Conformations of complexes between pyrrolo[1,4]benzodiazepines and DNA segments. J Med Chem. 1986;29(12):2492–2503. doi: 10.1021/jm00162a012. [DOI] [PubMed] [Google Scholar]
- 31.Tozuka Z, Takaya T. Studies on Tomaymycin .1. The Structure Determination of Tomaymycin on the Basis of Nmr-Spectra. J Antibiot. 1983;36(2):142–146. doi: 10.7164/antibiotics.36.142. [DOI] [PubMed] [Google Scholar]
- 32.Hochlowski JE, Andres WW, Theriault RJ, Jackson M, McAlpine JB. Abbeymycin, a new anthramycin-type antibiotic produced by a streptomycete. J Antibiot (Tokyo) 1987;40(2):145–148. doi: 10.7164/antibiotics.40.145. [DOI] [PubMed] [Google Scholar]
- 33.Konishi M, Hatori M, Tomita K, Sugawara M, Ikeda C, Nishiyama Y, Imanishi H, Miyaki T, Kawaguchi H. Chicamycin, a new antitumor antibiotic. I. Production, isolation and properties. J Antibiot (Tokyo) 1984;37(3):191–199. doi: 10.7164/antibiotics.37.191. [DOI] [PubMed] [Google Scholar]
- 34.Tsunakawa M, Kamei H, Konishi M, Miyaki T, Oki T, Kawaguchi H. Porothramycin, a new antibiotic of the anthramycin group: production, isolation, structure and biological activity. J Antibiot (Tokyo) 1988;41(10):1366–1373. doi: 10.7164/antibiotics.41.1366. [DOI] [PubMed] [Google Scholar]
- 35.Kunimoto S, Masuda T, Kanbayashi N, Hamada M, Naganawa H, Miyamoto M, Takeuchi T, Umezawa H. Mazethramycin, a new member of anthramycin group antibiotics. J Antibiot (Tokyo) 1980;33(6):665–667. doi: 10.7164/antibiotics.33.665. [DOI] [PubMed] [Google Scholar]
- 36.Mesentsev AS, Kuljaeva VV, Rubasheva LM. Structure of sibiromycin. J Antibiot (Tokyo) 1974;27(11):866–873. doi: 10.7164/antibiotics.27.866. [DOI] [PubMed] [Google Scholar]
- 37.Itoh J, Watabe H, Ishii S, Gomi S, Nagasawa M, Yamamoto H, Shomura T, Sezaki M, Kondo S. Sibanomicin, a new pyrrolo[1,4]benzodiazepine antitumor antibiotic produced by a Micromonospora sp. J Antibiot (Tokyo) 1988;41(9):1281–1284. doi: 10.7164/antibiotics.41.1281. [DOI] [PubMed] [Google Scholar]
- 38.Miyamoto M, Kondo S, Naganawa H, Maeda K, Ohno M. Structure and synthesis of neothramycin. J Antibiot (Tokyo) 1977;30(4):340–343. doi: 10.7164/antibiotics.30.340. [DOI] [PubMed] [Google Scholar]
- 39.Li W, Khullar A, Chou S, Sacramo A, Gerratana B. Biosynthesis of sibiromycin, a potent antitumor antibiotic. Appl Environ Microbiol. 2009;75(9):2869–2878. doi: 10.1128/AEM.02326-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Li W, Chou S, Khullar A, Gerratana B. Cloning and characterization of the biosynthetic gene cluster for tomaymycin, an SJG-136 monomeric analog. Appl Environ Microbiol. 2009;75(9):2958–2963. doi: 10.1128/AEM.02325-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Mostad A, Romming C, Storm B. Structure of the DNA complexing agent anthramycin. Acta Chemica Scandinavica B. 1978;32:639–645. [Google Scholar]
- 42.Kopka ML, Goodsell DS, Baikalov I, Grzeskowiak K, Cascio D, Dickerson RE. Crystal- Structure of a Covalent DNA Drug Adduct - Anthramycin Bound to C-C-a-a-C-G-T-TG- G and a Molecular Explanation of Specificity. Biochemistry. 1994;33(46):13593–13610. doi: 10.1021/bi00250a011. [DOI] [PubMed] [Google Scholar]
- 43.Arora SK. Structure of tomaymycin, a DNA binding antitumor antibiotic. J Antibiot (Tokyo) 1981;34(4):462–464. doi: 10.7164/antibiotics.34.462. [DOI] [PubMed] [Google Scholar]
- 44.Parker KA, Fedynyshyn TH. Synthesis of Anhydrosibiromycinone - New Method for the Direct Synthesis of Pyrrolo-1,4-Benzodiazepin-5-Ones. Tetrahedron Letters. 1979;(19):1657–1660. [Google Scholar]
- 45.Malhotra RK, Ostrander JM, Hurley LH, McInnes AG, Smith DG, Walter JA, Wright JL. Chemical conversion of anthramycin 11-methyl ether to didehydroanhydroanthramycin and its utilization in studies of the biosynthesis and mechanism of action of anthramycin. J Nat Prod. 1981;44(1):38–44. doi: 10.1021/np50013a007. [DOI] [PubMed] [Google Scholar]
- 46.Leber JD, Hoover JRE, Holden KG, Johnson RK, Hecht SM. A Revised Structure for Sibiromycin. Journal of the American Chemical Society. 1988;110(9):2992–2993. [Google Scholar]
- 47.Parker KA, Babine RE. Revision of the Assignment of Relative Stereochemistry in Sibirosamine - Synthesis of Methyl N-Tosyl Alpha-D-Sibirosaminopyranoside. Tetrahedron Letters. 1982;23(17):1763–1766. [Google Scholar]
- 48.Maurer PJ, Knudsen CG, Palkowitz AD, Rapoport H. Alpha-Amino-Acids as Chiral Educts for Asymmetric Products - Chirospecific Syntheses of Methyl L-Sibirosaminide and Its C-3 Epimer from L-Allothreonine. J Org Chem. 1985;50(3):325–332. [Google Scholar]
- 49.Hurley LH, Speedie MK, editors. Pyrrolo(1,4)benzodiazepines Antibiotics: Anthramycin, Tomaymycin and Sibiromycin. 4. Berlin: Springer-Verlag; 1981. pp. 261–294. [Google Scholar]
- 50.Hurley LH. Elucidation and Formulation of Novel Biosynthetic Pathways Leading to the Pyrrolo[1,4]Benzodiazepine Antibiotics Anthramycin, Tomaymycin, and Sibiromycin. Acc Chem Res. 1980;13(8):263–269. [Google Scholar]
- 51.Hurley LH, Zmijewski M, Chang CJ. Biosynthesis of Anthramycin - Determination of Labeling Pattern by Use of Radioactive and Stable Isotope Techniques. Journal of the American Chemical Society. 1975;97(15):4372–4378. doi: 10.1021/ja00848a040. [DOI] [PubMed] [Google Scholar]
- 52.Hurley LH, Gairola C. Pyrrolo (1,4) benzodiazepine antitumor antibiotics: Biosynthetic studies on the conversion of tryptophan to the anthranilic acid moieties of sibiromycin and tomaymycin. Antimicrob Agents Chemother. 1979;15(1):42–45. doi: 10.1128/aac.15.1.42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Hurley LH, Lasswell WL, Malhotra RK, Das NV. Pyrrolo[1,4]Benzodiazepine Antibiotics - Biosynthesis of the Anti-Tumor Antibiotic Sibiromycin by Streptosporangium-Sibiricum. Biochemistry. 1979;18(19):4225–4229. doi: 10.1021/bi00586a029. [DOI] [PubMed] [Google Scholar]
- 54.Witz DF, Hessler EJ, Miller TL. Bioconversion of tyrosine into the propylhygric acid moity of lincomycin. Biochemistry. 1971;10(7):1128–1133. doi: 10.1021/bi00783a005. [DOI] [PubMed] [Google Scholar]
- 55.Brahme NM, Gonzalez JE, Rolls JP, Hessler EJ, Mizsak S, Hurley LH. Biosynthesis of the Lincomycins .1. Studies Using Stable Isotopes on the Biosynthesis of the Propyl-LHygric and Ethyl-L-Hygric Acid Moieties of Lincomycin-a and Lincomycin-B. Journal of the American Chemical Society. 1984;106(25):7873–7878. [Google Scholar]
- 56.Hurley L, Das N, Gairola C, Zmijewski M. Biosynthetic Incorporation of Dl-Tryptophan- (5-H-3) into Anthramycin, Sibiromycin and Tomaymycin - Nih Shift Produced by Actinomycetes. Tetrahedron Letters. 1976;18:1419–1422. [Google Scholar]
- 57.Hurley LH, Gairola C, Das NV. Pyrrolo[1,4]benzodiazepine antibiotics. Biosynthesis of the antitumor antibiotic 11-demethyltomaymycin and its biologically inactive metabolite oxotomaymycin by Streptomyces achromogenes. Biochemistry. 1976;15(17):3760–3769. doi: 10.1021/bi00662a019. [DOI] [PubMed] [Google Scholar]
- 58.Rokem JS, Hurley LH. Sensitivity and permeability of the anthramycin producing organism Streptomyces refuineus to anthramycin and structurally related antibiotics. J Antibiot (Tokyo) 1981;34(9):1171–1174. doi: 10.7164/antibiotics.34.1171. [DOI] [PubMed] [Google Scholar]
- 59.Hu Y, Phelan V, Ntai I, Farnet CM, Zazopoulos E, Bachmann BO. Benzodiazepine Biosynthesis in Streptomyces refuineus. Chem Biol. 2007;14(6):691–701. doi: 10.1016/j.chembiol.2007.05.009. [DOI] [PubMed] [Google Scholar]
- 60.Kuo MS, Yurek DA, Coats JH, Chung ST, Li GP. Isolation and identification of 3- propylidene-delta 1-pyrroline-5-carboxylic acid, a biosynthetic precursor of lincomycin. J Antibiot (Tokyo) 1992;45(11):1773–1777. doi: 10.7164/antibiotics.45.1773. [DOI] [PubMed] [Google Scholar]
- 61.Peschke U, Schmidt H, Zhang HZ, Piepersberg W. Molecular characterization of the lincomycin-production gene cluster of Streptomyces lincolnensis 78-11. Mol Microbiol. 1995;16(6):1137–1156. doi: 10.1111/j.1365-2958.1995.tb02338.x. [DOI] [PubMed] [Google Scholar]
- 62.Harayama S, Rekik M. Bacterial aromatic ring-cleavage enzymes are classified into two different gene families. J Biol Chem. 1989;264(26):15328–15333. [PubMed] [Google Scholar]
- 63.Neusser D, Schmidt H, Spizek J, Novotna J, Peschke U, Kaschabeck S, Tichy P, Piepersberg W. The genes lmbB1 and lmbB2 of Streptomyces lincolnensis encode enzymes involved in the conversion of L-tyrosine to propylproline during the biosynthesis of the antibiotic lincomycin A. Arch Microbiol. 1998;169(4):322–332. doi: 10.1007/s002030050578. [DOI] [PubMed] [Google Scholar]
- 64.Novotna J, Honzatko A, Bednar P, Kopecky J, Janata J, Spizek J. l-3,4-Dihydroxyphenyl alanine-extradiol cleavage is followed by intramolecular cyclization in lincomycin biosynthesis. Eur J Biochem. 2004;271(18):3678–3683. doi: 10.1111/j.1432-1033.2004.04308.x. [DOI] [PubMed] [Google Scholar]
- 65.Colabroy KL, Hackett WT, Markham AJ, Rosenberg J, Cohen DE, Jacobson A. Biochemical characterization of L-DOPA 2,3-dioxygenase, a single-domain type I extradiol dioxygenase from lincomycin biosynthesis. Arch Biochem Biophys. 2008;479(2):131–138. doi: 10.1016/j.abb.2008.08.022. [DOI] [PubMed] [Google Scholar]
- 66.Hurley LH, Zmijewski M, Chang CJ. Biosynthesis of Anthramycin - Determination of Labeling Pattern by Use of Radioactive and Stable Isotope Techniques. J Am Chem Soc. 1975;97(15):4372–4378. doi: 10.1021/ja00848a040. [DOI] [PubMed] [Google Scholar]
- 67.Phelan VV, Du Y, McLean JA, Bachmann BO. Adenylation enzyme characterization using gamma -(18)O(4)-ATP pyrophosphate exchange. Chem Biol. 2009;16(5):473–478. doi: 10.1016/j.chembiol.2009.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Voet D, Voet JG. John Wiley & Sons, editor. Biochemistry. 2005;Ch 26 doi: 10.1002/bmb.20. [DOI] [PubMed] [Google Scholar]
- 69.Ulanova D, Novotna J, Smutna Y, Kamenik Z, Gazak R, Sulc M, Sedmera P, Kadlcik S, Plhackova K, Janata J. Mutasynthesis of lincomycin derivatives with activity against drug-resistant staphylococci. Antimicrob Agents Chemother. 54(2):927–930. doi: 10.1128/AAC.00918-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Horsman GP, Bhowmik S, Seah SYK, Kumar P, Bolin JT, Eltis LD. The tautomeric half-reaction of BphD, a C-C bond hydrolase - Kinetic and structural evidence supporting a key role for histidine 265 of the catalytic triad. Journal of Biological Chemistry. 2007;282(27):19894–19904. doi: 10.1074/jbc.M702237200. [DOI] [PubMed] [Google Scholar]
- 71.Pena MR, Stille JK. A Total Synthesis of Anthramycin - Application of Palladium- Catalyzed Coupling Reactions for the Attachment of the Acrylic Side-Chain. Journal of the American Chemical Society. 1989;111(14):5417–5424. [Google Scholar]
- 72.Leimgrub W, Batcho AD, Czajkows Rc. Total Synthesis of Anthramycin. Journal of the American Chemical Society. 1968;90(20):5641. doi: 10.1021/ja01022a078. [DOI] [PubMed] [Google Scholar]
- 73.Kitamura T, Sato Y, Mori M. Synthetic study of (+)-anthramycin using ring-closing enyne metathesis and cross-metathesis. Tetrahedron. 2004;60(43):9649–9657. [Google Scholar]
- 74.Hu WP, Wang JJ, Lin FL, Lin YC, Lin SR, Hsu MH. An efficient synthesis of pyrrolo[2,1-c][1,4]benzodiazepine. Synthesis of the antibiotic DC-81. J Org Chem. 2001;66(8):2881–2883. doi: 10.1021/jo010043d. [DOI] [PubMed] [Google Scholar]
- 75.Correa A, Tellitu I, Dominguez E, Moreno I, SanMartin R. An efficient, PIFA mediated approach to benzo-, naphtho-, and heterocycle-fused pyrrolo[2,1-c][1,4]diazepines. An advantageous access to the antitumor antibiotic DC-81. J Org Chem. 2005;70(6):2256–2264. doi: 10.1021/jo047872u. [DOI] [PubMed] [Google Scholar]
- 76.Kamal A, Laxman E, Laxman N, Rao NV. Synthesis of pyrrolo[2,1- c][1,4]benzodiazepines via reductive cyclization of omega-azido carbonyl compounds by TMSI: An efficient preparation of antibiotic DC-81 and its dimers. Bioorganic & Medicinal Chemistry Letters. 2000;10(20):2311–2313. doi: 10.1016/s0960-894x(00)00468-6. [DOI] [PubMed] [Google Scholar]
- 77.Tozuka Z, Takasugi H, Takaya T. Studies on tomaymycin. II. Total syntheses of the antitumor antibiotics, E-and Z-tomaymycins. J Antibiot (Tokyo) 1983;36(3):276–282. doi: 10.7164/antibiotics.36.276. [DOI] [PubMed] [Google Scholar]
- 78.Kaneko T, Wong H, Doyle TW, Rose WC, Bradner WT. Bicyclic and Tricyclic Analogs of Anthramycin. Journal of Medicinal Chemistry. 1985;28(3):388–392. doi: 10.1021/jm00381a020. [DOI] [PubMed] [Google Scholar]
- 79.Langlois N, Andriamialisoa RZ. Synthesis of Sulfonamide Analogs of the Pyrrolo[1,4]Benzodiazepine Antibiotic Abbeymycin. Heterocycles. 1989;29(8):1529–1536. [Google Scholar]
- 80.Oneil IA, Murray CL, Hunter RC, Kalindjian SB, Jenkins TC. The synthesis of functionalized pyrrolo-[2,1-c][1,4]-benzodiazepines. Synlett. 1997;1:75. [Google Scholar]
- 81.Wilson SC, Howard PW, Forrow SM, Hartley JA, Adams LJ, Jenkins TC, Kelland LR, Thurston DE. Design, synthesis, and evaluation of a novel sequence-selective epoxide-containing DNA cross-linking agent based on the pyrrolo[2,1-c][1,4]benzodiazepine system. Journal of Medicinal Chemistry. 1999;42(20):4028–4041. doi: 10.1021/jm981124d. [DOI] [PubMed] [Google Scholar]
- 82.Baraldi PG, Balboni G, Cacciari B, Guiotto A, Manfredini S, Romagnoli R, Spalluto G, Thurston DE, Howard PW, Bianchi N, Rutigliano C, Mischiati C, Gambari R. Synthesis, in vitro antiproliferative activity, and DNA-binding properties of hybrid molecules containing pyrrolo[2,1-c][1, 4]benzodiazepine and minor-groove-binding oligopyrrole carriers. J Med Chem. 1999;42(25):5131–5141. doi: 10.1021/jm991033w. [DOI] [PubMed] [Google Scholar]
- 83.Gregson SJ, Howard PW, Barcella S, Nakamya A, Jenkins TC, Kelland LR, Thurston DE. Effect of C2/C3-endo unsaturation on the cytotoxicity and DNA-binding reactivity of pyrrolo[2,1-c][1,4]benzodiazepines. Bioorg Med Chem Lett. 2000;10(16):1849–1851. doi: 10.1016/s0960-894x(00)00350-4. [DOI] [PubMed] [Google Scholar]
- 84.Gregson SJ, Howard PW, Corcoran KE, Barcella S, Yasin MM, Hurst AA, Jenkins TC, Kelland LR, Thurston DE. Effect of C2-exo unsaturation on the cytotoxicity and DNA-binding reactivity of pyrrolo[2,1-c][1,4]benzodiazepines. Bioorg Med Chem Lett. 2000;10(16):1845–1847. doi: 10.1016/s0960-894x(00)00351-6. [DOI] [PubMed] [Google Scholar]
- 85.Thurston DE, Bose DS, Howard PW, Jenkins TC, Leoni A, Baraldi PG, Guiotto A, Cacciari B, Kelland LR, Foloppe MP, Rault S. Effect of A-ring modifications on the DNA-binding behavior and cytotoxicity of pyrrolo[2,1-c] [1,4]benzodiazepines. Journal of Medicinal Chemistry. 1999;42(11):1951–1964. doi: 10.1021/jm981117p. [DOI] [PubMed] [Google Scholar]
- 86.Kamal A, Rajender, Reddy DR, Reddy MK, Balakishan G, Shaik TB, Chourasia M, Sastry GN. Remarkable enhancement in the DNA-binding ability of C2-fluoro substituted pyrrolo[2,1-c][1,4] benzodiazepines and their anticancer potential. Bioorganic & Medicinal Chemistry. 2009;17(4):1557–1572. doi: 10.1016/j.bmc.2008.12.068. [DOI] [PubMed] [Google Scholar]
- 87.Antonow D, Cooper N, Howard PW, Thurston DE. Parallel Synthesis of a Novel C2-Aryl Pyrrolo[2,1-c][1,4]benzodiazepine (PBD) Library. J Comb Chem. 2007 doi: 10.1021/cc0601332. [DOI] [PubMed] [Google Scholar]
- 88.Tiberghien AC, Hagan D, Howard PW, Thurston DE. Application of the Stille coupling reaction to the synthesis of C2-substituted endo-exo unsaturated pyrrolo[2,1- c][1,4]benzodiazepines (PBDs) Bioorg Med Chem Lett. 2004;14(20):5041–5044. doi: 10.1016/j.bmcl.2004.08.002. [DOI] [PubMed] [Google Scholar]
- 89.Cooper N, Hagan DR, Tiberghien A, Ademefun T, Matthews CS, Howard PW, Thurston DE. Synthesis of novel C2-aryl pyrrolobenzodiazepines (PBDs) as potential antitumour agents. Chemical Communications. 2002;16:1764–1765. doi: 10.1039/b205136b. [DOI] [PubMed] [Google Scholar]
- 90.Kamal A, Kumar PP, Seshadri BN, Srinivas O, Kumar MS, Sen S, Kurian N, Juvekar AS, Zingde SM. Phosphonate-linked pyrrolo[2,1-c][1,4]benzodiazepine conjugates: Synthesis, DNA-binding affinity and cytotoxicity. Bioorganic & Medicinal Chemistry. 2008;16(7):3895–3906. doi: 10.1016/j.bmc.2008.01.040. [DOI] [PubMed] [Google Scholar]
- 91.Chen ZZ, Gregson SJ, Howard PW, Thurston DE. A novel approach to the synthesis of cytotoxic C2-C3 unsaturated pyrrolo[2,1-c][1,4]benzodiazepines (PBDs) with conjugated acrylyl C2-substituents. Bioorganic & Medicinal Chemistry Letters. 2004;14(6):1547–1549. doi: 10.1016/j.bmcl.2003.12.094. [DOI] [PubMed] [Google Scholar]
- 92.Antonow D, Barata T, Jenkins TC, Parkinson GN, Howard PW, Thurston DE, Zloh M. Solution structure of a 2:1 C2-(2-naphthyl) pyrrolo[2,1-c][1,4]benzodiazepine DNA adduct: molecular basis for unexpectedly high DNA helix stabilization. Biochemistry. 2008;47(45):11818–11829. doi: 10.1021/bi801225q. [DOI] [PubMed] [Google Scholar]
- 93.Bose DS, Thompson AS, Ching JS, Hartley JA, Berardini MD, Jenkins TC, Neidle S, Hurley LH, Thurston DE. Rational Design of a Highly Efficient Irreversible DNA Interstrand Cross-Linking Agent Based on the Pyrrolobenzodiazepine Ring-System. Journal of the American Chemical Society. 1992;114(12):4939–4941. [Google Scholar]
- 94.Thurston DE, Bose DS, Thompson AS, Howard PW, Leoni A, Croker SJ, Jenkins TC, Neidle S, Hartley JA, Hurley LH. Synthesis of sequence-selective C8-linked pyrrolo[2,1- c][1,4]benzodiazepine DNA interstrand cross-linking agents. J Org Chem. 1996;61(23):8141–8147. doi: 10.1021/jo951631s. [DOI] [PubMed] [Google Scholar]
- 95.Kamal A, Rao NV. Facile and Efficient Synthesis of the Dimers of Dc-81 Antitumor Antibiotics. Tetrahedron Letters. 1995;36(24):4299–4302. [Google Scholar]
- 96.Smellie M, Kelland LR, Thurston DE, Souhami RL, Hartley JA. Cellular Pharmacology of Novel C8-Linked Anthramycin-Based Sequence-Selective DNA Minor-Groove Cross- Linking Agents. British Journal of Cancer. 1994;70(1):48–53. doi: 10.1038/bjc.1994.248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Bose DS, Thompson AS, Smellie M, Berardini MD, Hartley JA, Jenkins TC, Neidle S, Thurston DE. Effect of Linker Length on DNA-Binding Affinity, Cross-Linking Efficiency and Cytotoxicity of C8-Linked Pyrrolobenzodiazepine Dimers. Journal of the Chemical Society-Chemical Communications. 1992;(20):1518–1520. [Google Scholar]
- 98.Farmer JD, Rudnicki SM, Suggs JW. Synthesis and DNA Crosslinking Ability of a Dimeric Anthramycin Analog. Tetrahedron Letters. 1988;29(40):5105–5108. [Google Scholar]
- 99.Farmer JD, Jr, Gustafson GR, Conti A, Zimmt MB, Suggs JW. DNA binding properties of a new class of linked anthramycin analogs. Nucleic Acids Res. 1991;19(4):899–903. doi: 10.1093/nar/19.4.899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kamal A, Reddy PSMM, Reddy DR, Laxman E, Murthy YLN. Synthesis of fluorinated analogues of SJG-136 and their DNA-binding potential. Bioorganic & Medicinal Chemistry Letters. 2004;14(22):5699–5702. doi: 10.1016/j.bmcl.2004.08.050. [DOI] [PubMed] [Google Scholar]
- 101.Kamal A, Reddy PSMM, Reddy DR. The effect of C2-fluoro group on the biological activity of DC-81 and its ditners. Bioorganic & Medicinal Chemistry Letters. 2004;14(10):2669–2672. doi: 10.1016/j.bmcl.2004.02.063. [DOI] [PubMed] [Google Scholar]
- 102.Kamal A, Ramesh G, Srinivas O, Ramulu P, Laxman N, Rehana T, Deepak M, Achary MS, Nagarajaram HA. Design, synthesis, and evaluation of mixed imine-amine pyrrolobenzodiazepine dimers with efficient DNA binding affinity and potent cytotoxicity. Bioorg Med Chem. 2004;12(20):5427–5436. doi: 10.1016/j.bmc.2004.07.045. [DOI] [PubMed] [Google Scholar]
- 103.Kamal A, Ramesh G, Laxman N, Ramulu P, Srinivas O, Neelima K, Kondapi AK, Sreenu VB, Nagarajaram HA. Design, synthesis, and evaluation of new noncross-linking pyrrolobenzodiazepine dimers with efficient DNA binding ability and potent antitumor activity. J Med Chem. 2002;45(21):4679–4688. doi: 10.1021/jm020124h. [DOI] [PubMed] [Google Scholar]
- 104.Kamal A, Ramu R, Tekumalla V, Khanna GB, Barkume MS, Juvekar AS, Zingde SM. Synthesis, DNA binding, and cytotoxicity studies of pyrrolo[2,1-c][1,4]benzodiazepine-anthraquinone conjugates. Bioorg Med Chem. 2007;15(22):6868–6875. doi: 10.1016/j.bmc.2007.08.026. [DOI] [PubMed] [Google Scholar]
- 105.Kamal A, Reddy PS, Reddy DR, Laxman E. DNA binding potential and cytotoxicity of newly designed pyrrolobenzodiazepine dimers linked through a piperazine side-armed-alkane spacer. Bioorg Med Chem. 2006;14(2):385–394. doi: 10.1016/j.bmc.2005.08.020. [DOI] [PubMed] [Google Scholar]
- 106.Howard PW, Chen Z, Gregson SJ, Masterson LA, Tiberghien AC, Cooper N, Fang M, Coffils MJ, Klee S, Hartley JA, Thurston DE. Synthesis of a novel C2/C2′-arylsubstituted pyrrolo[2,1-c][1,4]benzodiazepine dimer prodrug with improved water solubility and reduced DNA reaction rate. Bioorg Med Chem Lett. 2009;19(22):6463–6466. doi: 10.1016/j.bmcl.2009.09.012. [DOI] [PubMed] [Google Scholar]
- 107.Kamal A, Tekumalla V, Krishnan A, Pal-Bhadra M, Bhadra U. Development of pyrrolo[2,1-c][1,4]benzodiazepine beta-galactoside prodrugs for selective therapy of cancer by ADEPT and PMT. ChemMedChem. 2008;3(5):794–802. doi: 10.1002/cmdc.200700328. [DOI] [PubMed] [Google Scholar]
- 108.Kamal A, Tekumalla V, Raju P, Naidu VG, Diwan PV, Sistla R. Pyrrolo[2,1- c][1,4]benzodiazepine-beta-glucuronide prodrugs with a potential for selective therapy of solid tumors by PMT and ADEPT strategies. Bioorg Med Chem Lett. 2008;18(13):3769–3773. doi: 10.1016/j.bmcl.2008.05.038. [DOI] [PubMed] [Google Scholar]
- 109.Gregson SJ, Howard PW, Gullick DR, Hamaguchi A, Corcoran KE, Brooks NA, Hartley JA, Jenkins TC, Patel S, Guille MJ, Thurston DE. Linker length modulates DNA crosslinking reactivity and cytotoxic potency of C8/C8′ ether-linked C2-exo-unsaturated pyrrolo[2,1-c][1,4]benzodiazepine (PBD) dimers. J Med Chem. 2004;47(5):1161–1174. doi: 10.1021/jm030897l. [DOI] [PubMed] [Google Scholar]
- 110.Kamal A, Ramu R, Tekumalla V, Khanna GB, Barkume MS, Juvekar AS, Zingde SM. Remarkable DNA binding affinity and potential anticancer activity of pyrrolo[2,1- c][1,4]benzodiazepine-naphthalimide conjugates linked through piperazine side-armed alkane spacers. Bioorg Med Chem. 2008;16(15):7218–7224. doi: 10.1016/j.bmc.2008.06.034. [DOI] [PubMed] [Google Scholar]
- 111.Rettig M, Kamal A, Ramu R, Mikolajczak J, Weisz K. Spectroscopic and calorimetric studies on the DNA recognition of pyrrolo[2,1-c][1,4]benzodiazepine hybrids. Bioorg Med Chem. 2009;17(2):919–928. doi: 10.1016/j.bmc.2008.11.033. [DOI] [PubMed] [Google Scholar]
- 112.Kamal A, Khan MN, Srikanth YV, Reddy KS, Juvekar A, Sen S, Kurian N, Zingde S. Synthesis, DNA-binding ability and evaluation of antitumour activity of triazolo[1,2,4]benzothiadiazine linked pyrrolo[2,1-c][1,4]benzodiazepine conjugates. Bioorg Med Chem. 2008;16(16):7804–7810. doi: 10.1016/j.bmc.2008.06.056. [DOI] [PubMed] [Google Scholar]
- 113.Wang JJ, Shen YK, Hu WP, Hsieh MC, Lin FL, Hsu MK, Hsu MH. Design, synthesis, and biological evaluation of pyrrolo[2,1-c][1,4]benzodiazepine and indole conjugates as anticancer agents. J Med Chem. 2006;49(4):1442–1449. doi: 10.1021/jm050956q. [DOI] [PubMed] [Google Scholar]
- 114.Lee CH, Hu WP, Hong CH, Yu HS, Liao WT, Chen CY, Chen YL, Chen BH, Chen GS, Wang JJ. Pyrrolo[2,1-c][1,4]benzodiazepine and indole conjugate (IN6CPBD) has better efficacy and superior safety than the mother compound DC-81 in suppressing the growth of established melanoma in vivo. Chem Biol Interact. 2009;180(3):360–367. doi: 10.1016/j.cbi.2009.05.001. [DOI] [PubMed] [Google Scholar]
- 115.Hu WP, Liang JJ, Kao CL, Chen YC, Chen CY, Tsai FY, Wu MJ, Chang LS, Chen YL, Wang JJ. Synthesis and antitumor activity of novel enediyne-linked pyrrolo[2,1- c][1,4]benzodiazepine hybrids. Bioorg Med Chem. 2009;17(3):1172–1180. doi: 10.1016/j.bmc.2008.12.036. [DOI] [PubMed] [Google Scholar]
- 116.Tiberghien AC, Evans DA, Kiakos K, Martin CR, Hartley JA, Thurston DE, Howard PW. An asymmetric C8/C8′-tripyrrole-linked sequence-selective pyrrolo[2,1- c][1,4]benzodiazepine (PBD) dimer DNA interstrand cross-linking agent spanning 11 DNA base pairs. Bioorg Med Chem Lett. 2008;18(6):2073–2077. doi: 10.1016/j.bmcl.2008.01.096. [DOI] [PubMed] [Google Scholar]
- 117.Wells G, Suggitt M, Coffils M, Baig MA, Howard PW, Loadman PM, Hartley JA, Jenkins TC, Thurston DE. Fluorescent 7-diethylaminocoumarin pyrrolobenzodiazepine conjugates: synthesis, DNA interaction, cytotoxicity and differential cellular localization. Bioorg Med Chem Lett. 2008;18(6):2147–2151. doi: 10.1016/j.bmcl.2008.01.083. [DOI] [PubMed] [Google Scholar]
- 118.Kohn KW, Bono VH, Jr, Kann HE., Jr Anthramycin, a new type of DNA-inhibiting antibiotic: reaction with DNA and effect on nucleic acid synthesis in mouse leukemia cells. Biochim Biophys Acta. 1968;155(1):121–129. doi: 10.1016/0005-2787(68)90342-0. [DOI] [PubMed] [Google Scholar]
- 119.Stefanovic V. Spectrophotometric studies of the interaction of anthramycin with deoxyribonucleic acid. Biochemical Pharmacology. 1968;17(2):315–323. [Google Scholar]
- 120.Horwitz SB, Chang SC, Grollman AP, Borkovec AB. Chemosterilant action of anthramycin: a proposed mechanism. Science. 1971;174(5):159–161. doi: 10.1126/science.174.4005.159. [DOI] [PubMed] [Google Scholar]
- 121.Nishioka Y, Beppu T, Kosaka M, Arima K. Mode of action of tomaymycin. J Antibiot (Tokyo) 1972;25(11):660–667. doi: 10.7164/antibiotics.25.660. [DOI] [PubMed] [Google Scholar]
- 122.Kohn KW, Spears CL. Reaction of anthramycin with deoxyribonucleic acid. J Mol Biol. 1970;51(3):551–572. doi: 10.1016/0022-2836(70)90008-2. [DOI] [PubMed] [Google Scholar]
- 123.Petrusek RL, Uhlenhopp EL, Duteau N, Hurley LH. Reaction of anthramycin with DNA. Biological consequences of DNA damage in normal and xeroderma pigmentosum cell. J Biol Chem. 1982;257(11):6207–6216. [PubMed] [Google Scholar]
- 124.Kohn KW, Glaubiger D, Spears CL. The reaction of anthramycin with DNA. II. Studies of kinetics and mechanism. Biochim Biophys Acta. 1974;361(3):288–302. doi: 10.1016/0005-2787(74)90372-4. [DOI] [PubMed] [Google Scholar]
- 125.Hurley LH, Petrusek R. Proposed structure of the anthramycin-DNA adduct. Nature. 1979;282(5738):529–531. doi: 10.1038/282529a0. [DOI] [PubMed] [Google Scholar]
- 126.Hertzberg RP, Hecht SM, Reynolds VL, Molineux IJ, Hurley LH. DNA sequence specificity of the pyrrolo[1,4]benzodiazepine antitumor antibiotics. Methidiumpropyl- EDTA-iron(II) footprinting analysis of DNA binding sites for anthramycin and related drugs. Biochemistry. 1986;25(6):1249–1258. doi: 10.1021/bi00354a009. [DOI] [PubMed] [Google Scholar]
- 127.Graves DE, Pattaroni C, Krishnan BS, Ostrander JM, Hurley LH, Krugh TR. The reaction of anthramycin with DNA. Proton and carbon nuclear magnetic resonance studies on the structure of the anthramycin-DNA adduct. J Biol Chem. 1984;259(13):8202–8209. [PubMed] [Google Scholar]
- 128.Krugh TR, Graves DE, Stone MP. Two-dimensional NMR studies on the anthramycin-d( ATGCAT)2 adduct. Biochemistry. 1989;28(26):9988–9994. doi: 10.1021/bi00452a017. [DOI] [PubMed] [Google Scholar]
- 129.Boyd FL, Stewart D, Remers WA, Barkley MD, Hurley LH. Characterization of a unique tomaymycin-d(CICGAATTCICG)2 adduct containing two drug molecules per duplex by NMR, fluorescence, and molecular modeling studies. Biochemistry. 1990;29(9):2387–2403. doi: 10.1021/bi00461a024. [DOI] [PubMed] [Google Scholar]
- 130.Barkley MD, Cheatham S, Thurston DE, Hurley LH. Pyrrolo[1,4]benzodiazepine antitumor antibiotics: evidence for two forms of tomaymycin bound to DNA. Biochemistry. 1986;25(10):3021–3031. doi: 10.1021/bi00358a043. [DOI] [PubMed] [Google Scholar]
- 131.Kopka ML, Goodsell DS, Baikalov I, Grzeskowiak K, Cascio D, Dickerson RE. Crystal structure of a covalent DNA-drug adduct: anthramycin bound to C-C-A-A-C-G-T-T-G-G and a molecular explanation of specificity. Biochemistry. 1994;33(46):13593–13610. doi: 10.1021/bi00250a011. [DOI] [PubMed] [Google Scholar]
- 132.Kizu R, Draves PH, Hurley LH. Correlation of DNA sequence specificity of anthramycin and tomaymycin with reaction kinetics and bending of DNA. Biochemistry. 1993;32(33):8712–8722. doi: 10.1021/bi00084a043. [DOI] [PubMed] [Google Scholar]
- 133.Hurley LH. Pyrrolo(1,4)benzodiazepine antitumor antibiotics. Comparative aspects of anthramycin, tomaymycin and sibiromycin. J Antibiot (Tokyo) 1977;30(5):349–370. doi: 10.7164/antibiotics.30.349. [DOI] [PubMed] [Google Scholar]
- 134.Hurley LH, Gairola C, Zmijewski M. Pyrrolo(1,4)benzodiazepine antitumor antibiotics. In vitro interaction of anthramycin, sibiromycin and tomaymycin with DNA using specifically radiolabelled molecules. Biochim Biophys Acta. 1977;475(3):521–535. doi: 10.1016/0005-2787(77)90067-3. [DOI] [PubMed] [Google Scholar]
- 135.Lown JW, Joshua AV. Molecular mechanism of binding of pyrrolo(1,4)benzodiazepine antitumour agents to deoxyribonucleic acid--anthramycin and tomaymycin. Biochem Pharmacol. 1979;28(13):2017–2026. doi: 10.1016/0006-2952(79)90218-1. [DOI] [PubMed] [Google Scholar]
- 136.Vargiu AV, Ruggerone P, Magistrato A, Carloni P. Anthramycin-DNA binding explored by molecular simulations. J Phys Chem B. 2006;110(48):24687–24695. doi: 10.1021/jp063155n. [DOI] [PubMed] [Google Scholar]
- 137.Arora SK. Structural investigations of mode of action of drugs. I. Molecular structure of mitomycin C. Life Sci. 1979;24(16):1519–1526. doi: 10.1016/0024-3205(79)90036-5. [DOI] [PubMed] [Google Scholar]
- 138.Zakrzewska K, Pullman B. A theoretical investigation of the sequence specificity in the binding of the antitumor drug anthramycin to DNA. J Biomol Struct Dyn. 1986;4(1):127–136. doi: 10.1080/07391102.1986.10507650. [DOI] [PubMed] [Google Scholar]
- 139.Rao SN, Singh UC, Kollman PA. Molecular mechanics simulations on covalent complexes between anthramycin and B DNA. J Med Chem. 1986;29(12):2484–2492. doi: 10.1021/jm00162a011. [DOI] [PubMed] [Google Scholar]
- 140.Boyd FL, Cheatham SF, Remers W, Hill GC, Hurley LH. Characterization of the Structure of the Anthramycin-D(Atgcat)2 Adduct by Nmr and Molecular Modeling Studies - Determination of the Stereochemistry of the Covalent Linkage Site, Orientation in the Minor Groove of DNA, and Effect on Local DNA-Structure. Journal of the American Chemical Society. 1990;112(9):3279–3289. [Google Scholar]
- 141.Cheatham S, Kook A, Hurley LH, Barkley MD, Remers W. One- and two-dimensional 1H NMR, fluorescence, and molecular modeling studies on the tomaymycind( ATGCAT)2 adduct. Evidence for two covalent adducts with opposite orientations and stereochemistries at the covalent linkage site. J Med Chem. 1988;31(3):583–590. doi: 10.1021/jm00398a016. [DOI] [PubMed] [Google Scholar]
- 142.Barkley MD, Thomas TJ, Maskos K, Remers WA. Steady-state fluorescence and molecular-modeling studies of tomaymycin-DNA adducts. Biochemistry. 1991;30(18):4421–4431. doi: 10.1021/bi00232a008. [DOI] [PubMed] [Google Scholar]
- 143.Puvvada MS, Hartley JA, Jenkins TC, Thurston DE. A Quantitative Assay to Measure the Relative DNA-Binding Affinity of Pyrrolo[2,1-C][1,4]Benzodiazepine (Pbd) Antitumor Antibiotics Based on the Inhibition of Restriction-Endonuclease Bamhi. Nucleic Acids Res. 1993;21(16):3671–3675. doi: 10.1093/nar/21.16.3671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Wang JJ, Hill GC, Hurley LH. Template-Directed Design of a DNA-DNA Cross-Linker Based Upon a Bis-Tomaymycin Duplex Adduct. Journal of Medicinal Chemistry. 1992;35(16):2995–3002. doi: 10.1021/jm00094a010. [DOI] [PubMed] [Google Scholar]
- 145.Jenkins TC, Hurley LH, Neidle S, Thurston DE. Structure of a covalent DNA minor groove adduct with a pyrrolobenzodiazepine dimer: evidence for sequence-specific interstrand cross-linking. J Med Chem. 1994;37(26):4529–4537. doi: 10.1021/jm00052a012. [DOI] [PubMed] [Google Scholar]
- 146.Martin C, Ellis T, McGurk CJ, Jenkins TC, Hartley JA, Waring MJ, Thurston DE. Sequence-selective interaction of the minor-groove interstrand cross-linking agent SJG- 136 with naked and cellular DNA: footprinting and enzyme inhibition studies. Biochemistry. 2005;44(11):4135–4147. doi: 10.1021/bi0479813. [DOI] [PubMed] [Google Scholar]
- 147.Gregson SJ, Howard PW, Corcoran KE, Jenkins TC, Kelland LR, Thurston DE. Synthesis of the first example of a C2-C3/C2 ′-C3 ′-endo unsaturated pyrrolo[2,1- c][1,4]benzodiazepine dimer. Bioorganic & Medicinal Chemistry Letters. 2001;11(21):2859–2862. doi: 10.1016/s0960-894x(01)00560-1. [DOI] [PubMed] [Google Scholar]
- 148.Gregson SJ, Howard PW, Hartley JA, Brooks NA, Adams LJ, Jenkins TC, Kelland LR, Tedhurston DE. Design, synthesis, and evaluation of a novel pyrrolobenzodiazepine DNA-interactive agent with highly efficient cross-linking ability and potent cytotoxicity. J Med Chem. 2001;44(5):737–748. doi: 10.1021/jm001064n. [DOI] [PubMed] [Google Scholar]
- 149.Rahman KM, Thompson AS, James CH, Narayanaswamy M, Thurston DE. The Pyrrolobenzodiazepine Dimer SJG-136 Forms Sequence-Dependent Intrastrand DNA Cross-Links and Monoalkylated Adducts in Addition to Interstrand Cross-Links. Journal of the American Chemical Society. 2009;131(38):13756–13766. doi: 10.1021/ja902986x. [DOI] [PubMed] [Google Scholar]
- 150.Arnould S, Spanswick VJ, Macpherson JS, Hartley JA, Thurston DE, Jodrell DI, Guichard SM. Time-dependent cytotoxicity induced by SJG-136 (NSC 694501): influence of the rate of interstrand cross-link formation on DNA damage signaling. Mol Cancer Ther. 2006;5(6):1602–1609. doi: 10.1158/1535-7163.MCT-06-0018. [DOI] [PubMed] [Google Scholar]
- 151.Rettig M, Weingarth M, Langel W, Kamal A, Kumar PP, Weisz K. Solution structure of a covalently bound pyrrolo[2,1-c][1,4]benzodiazepine-benzimidazole hybrid to a 10mer DNA duplex. Biochemistry. 2009;48(51):12223–12232. doi: 10.1021/bi901655t. [DOI] [PubMed] [Google Scholar]
- 152.Grunberg E, Prince HN, Titsworth E, Beskid G, Tendler MD. Chemotherpeutic properties of anthramycin. Chemotherapia. 1960;11:249–260. doi: 10.1159/000220459. [DOI] [PubMed] [Google Scholar]
- 153.Korman S, Tendler MD. Clinical investigation of cancer chemotherapeutic agents for neoplastic disease. J New Drugs. 1965;5(5):275–285. doi: 10.1002/j.1552-4604.1965.tb00247.x. [DOI] [PubMed] [Google Scholar]
- 154.Sigdesta Cp, Hagemann RF, Lesher S. Biological Effects of Anthramycin Methyl Ether .1. Acute Intestinal Necrosis. Chemotherapy. 1970;15(5):286. doi: 10.1159/000220693. [DOI] [PubMed] [Google Scholar]
- 155.Cargill C, Bachmann E, Zbinden G. Effects of daunomycin and anthramycin on electrocardiogram and mitochondrial metabolism of the rat heart. J Natl Cancer Inst. 1974;53(2):481–486. doi: 10.1093/jnci/53.2.481. [DOI] [PubMed] [Google Scholar]
- 156.Lubawy WC, Dallam RA, Hurley LH. Protection against anthramycin-induced toxicity in mice by coenzyme Q10. J Natl Cancer Inst. 1980;64(1):105–109. [PubMed] [Google Scholar]
- 157.Hisamatsu T, Uchida S, Takeuchi T, Ishizuka M, Umezawa H. Anti-Tumor Effect of a New Antibiotic, Neothramycin. Gann. 1980;71(3):308–312. [PubMed] [Google Scholar]
- 158.Kimura K, Ogawa M. Phase I trial of a new antitumor antibiotic, Neothramycin: A cooperative study group of Neothramycin Study. Cancer Treament Symposia. 1983;1:75–78. [Google Scholar]
- 159.Tsugaya M, Washida H, Hirao N, Hachisuka Y, Sakagami H, Iwase Y. The treatment of bladder cancer by neothramycin. Hinyokika Kiyo. 1986;32(10):1443–1448. [PubMed] [Google Scholar]
- 160.Gairola C, Thomas H, Szeinbach SL, Lubawy WC. The genetic activity of anthramycin, tomaymycin and sibiromycin in bacterial forward- and reverse-mutation assays and in the mouse bone-marrow micronucleus test. J Appl Toxicol. 1983;3(6):317–320. doi: 10.1002/jat.2550030609. [DOI] [PubMed] [Google Scholar]
- 161.Alley MC, Hollingshead MG, Pacula-Cox CM, Waud WR, Hartley JA, Howard PW, Gregson SJ, Thurston DE, Sausville EA. SJG-136 (NSC 694501), a novel rationally designed DNA minor groove interstrand cross-linking agent with potent and broad spectrum antitumor activity: part 2: efficacy evaluations. Cancer Res. 2004;64(18):6700–6706. doi: 10.1158/0008-5472.CAN-03-2942. [DOI] [PubMed] [Google Scholar]
- 162.Burger AM, Loadman PM, Thurston DE, Schultz R, Fiebig HH, Bibby MC. Preclinical pharmacology of the pyrrolobenzodiazepine (PBD) monomer DRH-417 (NSC 709119) J Chemother. 2007;19(1):66–78. doi: 10.1179/joc.2007.19.1.66. [DOI] [PubMed] [Google Scholar]
- 163.Walton MI, Goddard P, Kelland LR, Thurston DE, Harrap KR. Preclinical pharmacology and antitumour activity of the novel sequence-selective DNA minor-groove cross-linking agent DSB-120. Cancer Chemother Pharmacol. 1996;38(5):431–438. doi: 10.1007/s002800050507. [DOI] [PubMed] [Google Scholar]
- 164.Hartley JA, Spanswick VJ, Brooks N, Clingen PH, McHugh PJ, Hochhauser D, Pedley RB, Kelland LR, Alley MC, Schultz R, Hollingshead MG, Schweikart KM, Tomaszewski JE, Sausville EA, Gregson SJ, Howard PW, Thurston DE. SJG-136 (NSC 694501), a novel rationally designed DNA minor groove interstrand cross-linking agent with potent and broad spectrum antitumor activity: part 1: cellular pharmacology, in vitro and initial in vivo antitumor activity. Cancer Res. 2004;64(18):6693–6699. doi: 10.1158/0008-5472.CAN-03-2941. [DOI] [PubMed] [Google Scholar]
- 165.Clingen PH, De Silva IU, McHugh PJ, Ghadessy FJ, Tilby MJ, Thurston DE, Hartley JA. The XPF-ERCC1 endonuclease and homologous recombination contribute to the repair of minor groove DNA interstrand crosslinks in mammalian cells produced by the pyrrolo[2,1-c][1,4]benzodiazepine dimer SJG-136. Nucleic Acids Res. 2005;33(10):3283–3291. doi: 10.1093/nar/gki639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Hochhauser D, Meyer T, Spanswick VJ, Wu J, Clingen PH, Loadman P, Cobb M, Gumbrell L, Begent RH, Hartley JA, Jodrell D. Phase I study of sequence-selective minor groove DNA binding agent SJG-136 in patients with advanced solid tumors. Clin Cancer Res. 2009;15(6):2140–2147. doi: 10.1158/1078-0432.CCR-08-1315. [DOI] [PubMed] [Google Scholar]
- 167.Puzanov I, Lee W, Berlin JD, Calcutt MW, Hacehy DL, Vermeulen WL, Spanswick VJ, Haartley JA, Chen AP, Rothenberg ML. Final results of phase I and phamacokinetic trial of SJG-136 administeres on a daily x 3 schedule. J Clinical Oncology. 2008;26(May 20 suppl) [Google Scholar]
- 168.Baraldi PG, Cacciari B, Guiotto A, Romagnoli R, Spalluto G, Leoni A, Bianchi N, Feriotto G, Rutigliano C, Mischiati C, Gambari R. [2,1-c][1,4]benzodiazepine (PBD)- distamycin hybrid inhibits DNA binding to transcription factor Sp1. Nucleosides Nucleotides Nucleic Acids. 2000;19(8):1219–1229. doi: 10.1080/15257770008033045. [DOI] [PubMed] [Google Scholar]
- 169.Kotecha M, Kluza J, Wells G, O’Hare CC, Forni C, Mantovani R, Howard PW, Morris P, Thurston DE, Hartley JA, Hochhauser D. Inhibition of DNA binding of the NF-Y transcription factor by the pyrrolobenzodiazepine-polyamide conjugate GWL-78. Mol Cancer Ther. 2008;7(5):1319–1328. doi: 10.1158/1535-7163.MCT-07-0475. [DOI] [PubMed] [Google Scholar]
- 170.Hu WP, Tsai FY, Yu HS, Sung PJ, Chang LS, Wang JJ. Induction of apoptosis by DC- 81-indole conjugate agent through NF-kappaB and JNK/AP-1 pathway. Chem Res Toxicol. 2008;21(7):1330–1336. doi: 10.1021/tx700394h. [DOI] [PubMed] [Google Scholar]
- 171.Leimgruber W, Batcho AD, Schenker F. The structure of anthramycin. J Am Chem Soc. 1965;87(24):5793–5795. doi: 10.1021/ja00952a051. [DOI] [PubMed] [Google Scholar]
- 172.Arima K, Kohsaka M, Tamura G, Sakai H, Imanaka H. Studies on Tomaymycin, a New Antibiotic. 1. Isolation and Properties of Tomaymycin. J Antibiot (Tokyo) 1972;25(8):437. doi: 10.7164/antibiotics.25.437. [DOI] [PubMed] [Google Scholar]
- 173.Brazhnikova MG, Kovsharova IN, Konstantinova NV, Mezentsev AS, Proshliakova VV. Chemical study of the antineoplastic abtibiotic sibiromycin. Antibiotiki. 1970;15(4):297–300. [PubMed] [Google Scholar]
- 174.Shimizu K, Kawamoto I, Tomita F, Morimoto M, Fujimoto K. Prothracarcin, a novel antitumor antibiotic. J Antibiot (Tokyo) 1982;35(8):972–978. doi: 10.7164/antibiotics.35.972. [DOI] [PubMed] [Google Scholar]
- 175.Ltd. KHKC. Japanese Patent Japan. 1983:58–180. 487.
- 176.Hu WP, Yu HS, Chen YC, Wang JJ. Biological evaluation of an antibiotic DC-81-indole conjugate agent in human melanoma cell lines. Kaohsiung J Med Sci. 2003;19(1):6–12. doi: 10.1016/S1607-551X(09)70441-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Osada H, Ishinabe K, Yano T, Kajikawa K, Isono K. New pyrrolobenzodiazepine antibiotics, RK-1441A and B. I. Biological properties. Agric Biol Chem. 1990;54(11):2875–2881. [PubMed] [Google Scholar]
- 178.Osada H, Uramoto M, Uzawa J, Kajikawa K, Isono K. New pyrrolobenzodiazepine antibiotics, RK-1441A and B. II. Isolation and structure. Agric Biol Chem. 1990;54(11):2883–2887. [PubMed] [Google Scholar]
- 179.Takeuchi T, Miyamoto T, Ishizuka M, Naganawa H, Kondo S. Neothramycins A and B, new antitumor antibiotics. J Antibiot (Tokyo) 1976;29(1):93–96. doi: 10.7164/antibiotics.29.93. [DOI] [PubMed] [Google Scholar]
- 180.Fotso S, Zabriskie TM, Proteau PJ, Flatt PM, Santosa DA, Mahmud T. Limazepines A-F, pyrrolo[1,4]benzodiazepine Antibiotics from an Indonesian Micrococcus sp. J Nat Prod. 2009;72(4):690–695. doi: 10.1021/np800827w. [DOI] [PubMed] [Google Scholar]
- 181.Langlois N, Rojas-Rousseau A, Gaspard C, Werner GH, Darro F, Kiss R. Synthesis and cytotoxicity on sensitive and doxorubicin-resistant cell lines of new pyrrolo[2,1- c][1,4]benzodiazepines related to anthramycin. J Med Chem. 2001;44(22):3754–3757. doi: 10.1021/jm010937q. [DOI] [PubMed] [Google Scholar]