1
|
Morcos A, Jung Y, Galvan Bustillos J, Fuller RN, Caba Molina D, Bertucci A, Boyle KE, Vazquez ME, Wall NR. A Comprehensive Review of the Antitumor Properties and Mechanistic Insights of Duocarmycin Analogs. Cancers (Basel) 2024; 16:3293. [PMID: 39409913 PMCID: PMC11475672 DOI: 10.3390/cancers16193293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
The duocarmycin family is a group of potent cytotoxic agents originally isolated from the bacterium Streptomyces. This discovery has spurred significant interest due to duocarmycins' unique chemical structures and powerful mechanism of action. This review comprehensively details the history of the duocarmycin family, the current understanding of their therapeutic potential, and the major clinical trials that have been conducted. Chemically, the duocarmycin family is characterized by a DNA-binding unit that confers specificity, a subunit-linking amide that positions the molecule within the DNA helix, and an alkylating unit that interacts with the DNA. This configuration allows them to bind selectively to the minor groove of DNA and alkylate adenine bases, a notable deviation from the more common guanine targeting performed by other alkylating agents. Duocarmycin's mechanism of action involves the formation of covalent adducts with DNA, leading to the disruption of the DNA architecture and subsequent inhibition of replication and transcription. Recent advancements in drug delivery systems, such as antibody-drug conjugates (ADCs), have further elevated the therapeutic prospects of duocarmycin analogs by providing a promising mechanism for enhancing intracellular concentrations and selective tumor delivery. Preclinical studies have highlighted the efficacy of duocarmycin derivatives in various in vitro models, providing a strong foundation for translational research. However, further biological research is required to fully understand the toxicology of duocarmycin family members before it can be clinically relevant. The major focus of this review is to cache the major biologically relevant findings of different duocarmycin analogs as well as their biological shortcomings to propose next steps in the field of cancer therapy with these potent therapeutics.
Collapse
Affiliation(s)
- Ann Morcos
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA
- Division of Biochemistry, Department of Basic Science, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Yeonkyu Jung
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA
- Division of Biochemistry, Department of Basic Science, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Joab Galvan Bustillos
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA
- Division of Biochemistry, Department of Basic Science, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Division of Surgical Oncology, Department of Surgery, Loma Linda University Health, Loma Linda, CA 92350, USA;
| | - Ryan N. Fuller
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA
- Division of Biochemistry, Department of Basic Science, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - David Caba Molina
- Division of Surgical Oncology, Department of Surgery, Loma Linda University Health, Loma Linda, CA 92350, USA;
| | - Antonella Bertucci
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA
- Nuclear Response & Analysis, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | | | - Marcelo E. Vazquez
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA
- Radiobiology & Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | - Nathan R. Wall
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA
- Division of Biochemistry, Department of Basic Science, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
2
|
Benham CJ. DNA superhelicity. Nucleic Acids Res 2024; 52:22-48. [PMID: 37994702 PMCID: PMC10783518 DOI: 10.1093/nar/gkad1092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 10/20/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023] Open
Abstract
Closing each strand of a DNA duplex upon itself fixes its linking number L. This topological condition couples together the secondary and tertiary structures of the resulting ccDNA topoisomer, a constraint that is not present in otherwise identical nicked or linear DNAs. Fixing L has a range of structural, energetic and functional consequences. Here we consider how L having different integer values (that is, different superhelicities) affects ccDNA molecules. The approaches used are primarily theoretical, and are developed from a historical perspective. In brief, processes that either relax or increase superhelicity, or repartition what is there, may either release or require free energy. The energies involved can be substantial, sufficient to influence many events, directly or indirectly. Here two examples are developed. The changes of unconstrained superhelicity that occur during nucleosome attachment and release are examined. And a simple theoretical model of superhelically driven DNA structural transitions is described that calculates equilibrium distributions for populations of identical topoisomers. This model is used to examine how these distributions change with superhelicity and other factors, and applied to analyze several situations of biological interest.
Collapse
Affiliation(s)
- Craig J Benham
- UC Davis Genome Center, University of California, One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
3
|
Huo YF, Zhu LN, Liu KK, Zhang LN, Zhang R, Kong DM. Water-Soluble Cationic Metalloporphyrins: Specific G-Quadruplex-Stabilizing Ability and Reversible Chirality of Aggregates Induced by AT-Rich DNA. Inorg Chem 2017; 56:6330-6342. [DOI: 10.1021/acs.inorgchem.7b00426] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Yan-Fang Huo
- Department of Chemistry, Tianjin University, Tianjin 300072, People’s Republic of China
- State Key
Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of
Biosensing and Molecular Recognition, Nankai University, Tianjin 300071, People’s Republic of China
| | - Li-Na Zhu
- Department of Chemistry, Tianjin University, Tianjin 300072, People’s Republic of China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300071, People’s Republic of China
| | - Ke-Ke Liu
- Department of Chemistry, Tianjin University, Tianjin 300072, People’s Republic of China
| | - Li-Na Zhang
- Department of Chemistry, Tianjin University, Tianjin 300072, People’s Republic of China
| | - Ran Zhang
- Department of Chemistry, Tianjin University, Tianjin 300072, People’s Republic of China
| | - De-Ming Kong
- State Key
Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of
Biosensing and Molecular Recognition, Nankai University, Tianjin 300071, People’s Republic of China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300071, People’s Republic of China
| |
Collapse
|
4
|
Nikolova EN, Zhou H, Gottardo FL, Alvey HS, Kimsey IJ, Al-Hashimi HM. A historical account of Hoogsteen base-pairs in duplex DNA. Biopolymers 2016; 99:955-68. [PMID: 23818176 DOI: 10.1002/bip.22334] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 06/17/2013] [Indexed: 11/05/2022]
Abstract
In 1957, a unique pattern of hydrogen bonding between N3 and O4 on uracil and N7 and N6 on adenine was proposed to explain how poly(rU) strands can associate with poly(rA)-poly(rU) duplexes to form triplexes. Two years later, Karst Hoogsteen visualized such a noncanonical A-T base-pair through X-ray analysis of co-crystals containing 9-methyladenine and 1-methylthymine. Subsequent X-ray analyses of guanine and cytosine derivatives yielded the expected Watson-Crick base-pairing, but those of adenine and thymine (or uridine) did not yield Watson-Crick base-pairs, instead favoring "Hoogsteen" base-pairing. More than two decades ensued without experimental "proof" for A-T Watson-Crick base-pairs, while Hoogsteen base-pairs continued to surface in AT-rich sequences, closing base-pairs of apical loops, in structures of DNA bound to antibiotics and proteins, damaged and chemically modified DNA, and in polymerases that replicate DNA via Hoogsteen pairing. Recently, NMR studies have shown that base-pairs in duplex DNA exist as a dynamic equilibrium between Watson-Crick and Hoogsteen forms. There is now little doubt that Hoogsteen base-pairs exist in significant abundance in genomic DNA, where they can expand the structural and functional versatility of duplex DNA beyond that which can be achieved based only on Watson-Crick base-pairing. Here, we provide a historical account of the discovery and characterization of Hoogsteen base-pairs, hoping that this will inform future studies exploring the occurrence and functional importance of these alternative base-pairs.
Collapse
Affiliation(s)
- Evgenia N Nikolova
- Department of Chemistry & Biophysics, The University of Michigan, 930 North University Avenue, Ann Arbor, MI, 48109-1055; Integrative Structural & Computational Biology Department, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037
| | | | | | | | | | | |
Collapse
|
5
|
Sershen CL, Mell JC, Madden SM, Benham CJ. Superhelical duplex destabilization and the recombination position effect. PLoS One 2011; 6:e20798. [PMID: 21695263 PMCID: PMC3111454 DOI: 10.1371/journal.pone.0020798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 05/12/2011] [Indexed: 11/19/2022] Open
Abstract
The susceptibility to recombination of a plasmid inserted into a chromosome varies with its genomic position. This recombination position effect is known to correlate with the average G+C content of the flanking sequences. Here we propose that this effect could be mediated by changes in the susceptibility to superhelical duplex destabilization that would occur. We use standard nonparametric statistical tests, regression analysis and principal component analysis to identify statistically significant differences in the destabilization profiles calculated for the plasmid in different contexts, and correlate the results with their measured recombination rates. We show that the flanking sequences significantly affect the free energy of denaturation at specific sites interior to the plasmid. These changes correlate well with experimentally measured variations of the recombination rates within the plasmid. This correlation of recombination rate with superhelical destabilization properties of the inserted plasmid DNA is stronger than that with average G+C content of the flanking sequences. This model suggests a possible mechanism by which flanking sequence base composition, which is not itself a context-dependent attribute, can affect recombination rates at positions within the plasmid.
Collapse
Affiliation(s)
- Cheryl L Sershen
- Baylor College of Medicine, Houston, Texas, United States of America.
| | | | | | | |
Collapse
|
6
|
Rao L, West TK, Saluta G, Kucera GL, Bierbach U. Probing platinum-adenine-n3 adduct formation with DNA minor-groove binding agents. Chem Res Toxicol 2010; 23:1148-50. [PMID: 20578739 PMCID: PMC2930193 DOI: 10.1021/tx100170p] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Me-lex(py/py), an adenine-N3-selective alkylating agent, and the reversible minor-groove binder netropsin were used to probe the formation of unusual minor-groove adducts by the cytotoxic hybrid agent PT-ACRAMTU ([PtCl(en)(ACRAMTU)](NO(3))(2); en = ethane-1,2-diamine, ACRAMTU = 1-[2-(acridin-9-ylamino)ethyl]-1,3-dimethylthiourea). PT-ACRAMTU was found by chemical footprinting to inhibit specific Me-lex-mediated DNA cleavage at several adenine sites but not at nonspecific guanine, which is consistent with the platination of adenine-N3. In a cell proliferation assay, a significant decrease in cytotoxicity was observed for PT-ACRAMTU, when cancer cells were pretreated with netropsin, suggesting that minor-groove adducts in cellular DNA contribute to the biological activity of the hybrid agent.
Collapse
Affiliation(s)
| | | | | | | | - Ulrich Bierbach
- To whom correspondence should be addressed. Te: 336-758-3507. Fax: 336-758-4656.
| |
Collapse
|
7
|
Prajapati RK, Kumar J, Verma S. Silver-catalyzed intramolecular cyclization of 9-propargyladenine via N3 alkylation. Chem Commun (Camb) 2010; 46:3312-4. [DOI: 10.1039/c001947j] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
8
|
Rahimian M, Miao Y, Wilson WD. Influence of DNA structure on adjacent site cooperative binding. J Phys Chem B 2008; 112:8770-8. [PMID: 18582108 DOI: 10.1021/jp801997v] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Previous NMR studies of Hoechst 33258 with the d(CTTTTGCAAAAG)2 sequence have shown very strong (K2 >> K1) cooperativity between two adjacent binding sites (Searle, M. S.; Embrey, K. J. Nucleic Acids Res. 1990, 18 (13), 3753- 3762). In contrast, surface plasmon resonance (SPR) results with the hairpin analog of the same sequence show significantly reduced cooperativity. In an effort to explain the difference, two-dimensional (2-D) NMR experiments were done on both duplex and hairpin. Hoechst 33258 and an amidine analog, DB183, show very strong cooperativity with the duplex DNA but much weaker cooperativity with the hairpin. The significantly lower thermal melting temperature (Tm) of the duplex (34.8 degrees C) in comparison to its hairpin analog (62.3 degrees C) supports the idea of a dynamic difference between the two DNA structures that can influence cooperativity in binding. These results confirm the role of conformational entropy in positive cooperativity in some DNA interactions.
Collapse
Affiliation(s)
- Maryam Rahimian
- Department of Chemistry, Georgia State University, P.O. Box 4098, Atlanta, Georgia 30303, USA
| | | | | |
Collapse
|
9
|
Gluch A, Vidakovic M, Bode J. Scaffold/matrix attachment regions (S/MARs): relevance for disease and therapy. Handb Exp Pharmacol 2008:67-103. [PMID: 18491049 DOI: 10.1007/978-3-540-72843-6_4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
There is increasing awareness that processes, such as development, aging and cancer, are governed, to a considerable extent, by epigenetic processes, such as DNA and histone modifications. The sites of these modifications in turn reflect their position and role in the nuclear architecture. Since epigenetic changes are easier to reverse than mutations, drugs that remove or add the chemical tags are at the forefront of research for the treatment of cancerous and inflammatory diseases. This review will use selected examples to develop a unified view that might assist the systematic development of novel therapeutic regimens.
Collapse
Affiliation(s)
- A Gluch
- Helmholtz-Zentrum für Infektionsforschung MBIO/Epigenetic Regulation, Inhoffenstrasse 7, Braunschweig, Germany
| | | | | |
Collapse
|
10
|
Woynarowski JM, Krugliak M, Ginsburg H. Pharmacogenomic analyses of targeting the AT-rich malaria parasite genome with AT-specific alkylating drugs. Mol Biochem Parasitol 2007; 154:70-81. [PMID: 17524501 DOI: 10.1016/j.molbiopara.2007.04.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 04/01/2007] [Accepted: 04/13/2007] [Indexed: 10/23/2022]
Abstract
UNLABELLED Human malaria parasites, including the most lethal Plasmodium falciparum, are increasingly resistant to existing antimalarial drugs. One remarkable opportunity to selectively target P. falciparum stems from the unique AT-richness of its genome (80% A/T, relative to 60% in human DNA). To rationally explore this opportunity, we used drugs (adozelesin and bizelesin) which distinctly target AT-rich minisatellites and an in silico approach for genome-wide analysis previously experimentally validated in human cells [Woynarowski JM, Trevino AV, Rodriguez KA, Hardies SC, Benham CJ. AT-rich islands in genomic DNA as a novel target for AT-specific DNA-reactive antitumor drugs. J Biol Chem 2001;276:40555-66]. Both drugs demonstrate a potent, rapid and irreversible inhibition of the cultured P. falciparum (50% inhibition at 110 and 10+/-2.3 pM, respectively). This antiparasital activity reflects most likely drug binding to specific super-AT-rich regions. Relative to the human genome, the P. falciparum genome shows 3.9- and 7-fold higher frequency of binding sites for adozelesin and bizelesin, respectively. The distribution of these sites is non-random with the most prominent clusters found in large unique minisatellites [median size 3.5 kbp of nearly pure A/T, with multiple converging repeats but no shared consensus other than (A/T)(n)]. Each of the fourteen P. falciparum chromosomes contains only one such "super-AT island" located within approximately 3-7.5 kbp of gene-free and nucleosome-free loci. Important functions of super-AT islands are suggested by their exceptional predicted potential to serve as matrix attachment regions (MARs) and a precise co-localization with the putative centromeres. CONCLUSION Super-AT islands, identified as unique domains in the P. falciparum genome with presumably crucial functions, offer therapeutically exploitable opportunity for new antimalarial strategies.
Collapse
Affiliation(s)
- Jan M Woynarowski
- Department of Radiation Oncology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78245, USA.
| | | | | |
Collapse
|
11
|
Stojković MR, Piantanida I, Kralj M, Marjanović M, Zinić M, Pawlica D, Eilmes J. The dicationic derivatives of DBTAA: Interactions with DNA/RNA and antiproliferative effects on human cell lines. Bioorg Med Chem 2007; 15:1795-801. [PMID: 17161951 DOI: 10.1016/j.bmc.2006.11.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2006] [Revised: 11/15/2006] [Accepted: 11/21/2006] [Indexed: 10/23/2022]
Abstract
Three dibenzotetraaza[14]annulenes non-covalently interacted with double-stranded DNA and RNA by mixed minor groove and/or intercalative binding mode. Observed interactions were strongly dependent on the steric exposure of positive charges and the length of the linkers of studied compounds as well as on the secondary structure and basepair composition of DNA/RNA. Compound 2 showed pronounced selectivity toward dA-dT-rich sequences and binding mode switch from dominant minor groove binding to ds-DNA to dominant intercalation into ds-RNA. Antiproliferative effect of studied compounds on human tumor and normal cell lines was in good agreement with the strength of observed interactions with DNA/RNA.
Collapse
Affiliation(s)
- Marijana Radić Stojković
- Laboratory for Supramolecular and Nucleoside Chemistry, Division of Organic Chemistry and Biochemistry, Ruder Bosković Institute, PO Box 180, HR-10002 Zagreb, Croatia
| | | | | | | | | | | | | |
Collapse
|
12
|
Guddneppanavar R, Saluta G, Kucera GL, Bierbach U. Synthesis, biological activity, and DNA-damage profile of platinum-threading intercalator conjugates designed to target adenine. J Med Chem 2006; 49:3204-14. [PMID: 16722638 DOI: 10.1021/jm060035v] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
PT-ACRAMTU {[PtCl(en)(ACRAMTU)](NO3)2, 2; ACRAMTU = 1-[2-(acridin-9-ylamino)ethyl]-1,3-dimethylthiourea, 1, en = ethane-1,2-diamine} is the prototype of a series of DNA-targeted adenine-affinic dual intercalating/platinating agents. Several novel 4,9-disubstituted acridines and the corresponding platinum-acridine conjugates were synthesized. The newly introduced 4-carboxamide side chains contain H-bond donor/acceptor functions designed to promote groove- and sequence-specific platinum binding. In HL-60 (leukemia) and H460 (lung) cancer cells, IC50 values in the micromolar to millimolar range were observed. Several of the intercalators show enhanced cytotoxicity compared to prototype 1, but conjugate 2 appears to be the most potent hybrid agent. Enzymatic digestion assays in conjunction with liquid chromatography-electrospray mass spectrometry analysis indicate that the new conjugates produce PT-ACRAMTU-type DNA damage. Platinum-modified 2'-deoxyguanosine, dG, and several dinucleotide fragments, d(NpN)*, were detected. One of the conjugates showed significantly higher levels of binding to A-containing sites than conjugate 2 (35 +/- 3% vs 24 +/- 3%). Possible structure-activity relationships are discussed.
Collapse
|
13
|
Barry CG, Day CS, Bierbach U. Duplex-promoted platination of adenine-N3 in the minor groove of DNA: challenging a longstanding bioinorganic paradigm. J Am Chem Soc 2005; 127:1160-9. [PMID: 15669855 DOI: 10.1021/ja0451620] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The interactions of [Pt(en)Cl(ACRAMTU-S)](NO3)2 (PT-ACRAMTU, en = ethane-1,2-diamine, ACRAMTU = 1-[2-(acridin-9-ylamino)ethyl]-1,3-dimethylthiourea) with adenine in DNA have been studied using a combination of analytical and high-resolution structural methods. For the first time, a cytotoxic platinum(II) complex has been demonstrated to form adducts in the minor groove of DNA through platination of the adenine-N3 endocyclic nitrogen. An acidic depurination assay was developed that allowed the controlled and selective (pH 2, 60 degrees C, 12 h) release of platinum-modified adenine from drug-treated nucleic acid samples. From the digested mixtures, three adducts were isolated by semipreparative reverse phase high-performance liquid chromatography and studied by electrospray ionization mass spectrometry (in-line LC-MS), variable-pH 1H NMR spectroscopy, and, where applicable, X-ray crystallography. The three species were identified as the N7 (A-I), N3 (A-II), and N1 (A-III) linkage isomers of [Pt(en)(ACRAMTU-S)(adenine)]3+ (A). Incubations carried out with the single- and double-stranded model sequences, d(TA)5 and d(TA)15, as well as native DNA indicate that the adduct profiles (A-I:A-II:A-IIIratios) are sensitive to the nature of the nucleic acid template. A-II was found to be a double-strand specific adduct. The crystal structure of this adduct has been determined, providing ultimate evidence for the N3 connectivity of platinum. A-II crystallizes in the triclinic space group P in the form of centrosymmetric dimers, {[Pt(en)(ACRAMTU-S)(adenine-N3)]2}6+. The cations are stabilized by a combination of adenine-adenine base pairing (N6...N1 2.945(5) A) and mutual acridine-adenine base stacking. Tandem mass spectra and 1H chemical shift anomalies indicate that this type of self-association is not merely a crystal packing effect but persists in solution. The monofunctional platination of adenine at its N7, N3, and N1 positions in a significant fraction of adducts breaks a longstanding paradigm in platinum-DNA chemistry, the requirement for nucleophilic attack of guanine-N7 as the principal step in cross-link formation. The biological consequences and potential therapeutic applications of the unique base and groove recognition of PT-ACRAMTU are discussed.
Collapse
Affiliation(s)
- Colin G Barry
- Department of Chemistry, Wake Forest University, PO Box 7486, Reynolda Station, Winston-Salem, North Carolina 27109, USA
| | | | | |
Collapse
|
14
|
Nelson SM, Ferguson LR, Denny WA. Demonstration by Real-Time Polymerase Chain Reaction that Cellular DNA Alkylation by Novel Aminoindoline Compounds Affects Expression of the Protooncogene c-myc. Chem Res Toxicol 2005; 18:239-48. [PMID: 15720128 DOI: 10.1021/tx049852t] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aminoindolines, analogues of the potent DNA alkylating agent seco-CBI-TMI, bind to and alkylate in the minor groove of AT-rich DNA in vitro. Here we extend the in vitro mechanism of action studies by treating cells in culture and examining the DNA binding patterns within AT-rich regions of the protooncogene locus c-myc, using a real-time polymerase chain reaction (PCR) stop assay. In addition, real-time reverse transcriptase (RT) PCR is used to examine the immediate effects of drug treatment on c-myc expression. These analyses demonstrate a concentration and time dependence for DNA alkylation at the chosen sites within the c-myc locus, as well as a prompt and significant downregulation of c-myc expression. While downregulation of this important growth regulator is likely not the only consequence of aminoindoline treatment, these studies begin to address the cellular pathways that are involved in the potent cytotoxic effects observed and provide insights for the future development of anticancer drugs of this class.
Collapse
Affiliation(s)
- Stephanie M Nelson
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 10000, New Zealand.
| | | | | |
Collapse
|
15
|
Benham CJ, Bi C. The analysis of stress-induced duplex destabilization in long genomic DNA sequences. J Comput Biol 2005; 11:519-43. [PMID: 15579230 DOI: 10.1089/cmb.2004.11.519] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We present a method for calculating predicted locations and extents of stress-induced DNA duplex destabilization (SIDD) as functions of base sequence and stress level in long DNA molecules. The base pair denaturation energies are assigned individually, so the influences of near neighbors, methylated bases, adducts, or lesions can be included. Sample calculations indicate that copolymeric energetics give results that are close to those derived when full near-neighbor energetics are used; small but potentially informative differences occur only in the calculated SIDD properties of moderately destabilized regions. The method presented here for analyzing long sequences calculates the destabilization properties within windows of fixed length N, with successive windows displaced by an offset distance d(o). The final values of the relevant destabilization parameters for each base pair are calculated as weighted averages of the values computed for each window in which that base pair appears. This approach implicitly assumes that the strength of the direct coupling between remote base pairs that is induced by the imposed stress attenuates with their separation distance. This strategy enables calculations of the destabilization properties of DNA sequences of any length, up to and including complete chromosomes. We illustrate its utility by calculating the destabilization properties of the entire E. coli genomic DNA sequence. A preliminary analysis of the results shows that promoters are associated with SIDD regions in a highly statistically significant manner, suggesting that SIDD attributes may prove useful in the computational prediction of promoter locations in prokaryotes.
Collapse
Affiliation(s)
- Craig J Benham
- UC Davis Genome Center, University of California, One Shields Avenue, Davis, CA 95616, USA.
| | | |
Collapse
|
16
|
Kamal A, Srinivas O, Ramulu P, Ramesh G, Kumar PP, Kumar MS. Synthesis and DNA binding affinity of novel A-C8/C-C2-exo unsaturated alkoxyamido-linked pyrrolo[2,1-c][1,4]benzodiazepine dimers. Bioorg Med Chem 2004; 12:4337-50. [PMID: 15265486 DOI: 10.1016/j.bmc.2004.06.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2004] [Revised: 06/10/2004] [Accepted: 06/11/2004] [Indexed: 11/22/2022]
Abstract
The synthesis of novel A-C8/C-C2-exo unsaturated alkoxyamido-linked pyrrolo[2,1-c][1,4]benzodiazepine dimers is reported and these dimers show significant DNA binding affinity and they also exhibit moderate anticancer activity.
Collapse
Affiliation(s)
- Ahmed Kamal
- Biotransformation Laboratory, Division of Organic Chemistry, Indian Institute of Chemical Technology, Hyderabad 500007, India.
| | | | | | | | | | | |
Collapse
|
17
|
Nelson SM, Ferguson LR, Denny WA. DNA and the chromosome - varied targets for chemotherapy. CELL & CHROMOSOME 2004; 3:2. [PMID: 15157277 PMCID: PMC421739 DOI: 10.1186/1475-9268-3-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Accepted: 05/24/2004] [Indexed: 12/29/2022]
Abstract
The nucleus of the cell serves to maintain, regulate, and replicate the critical genetic information encoded by the genome. Genomic DNA is highly associated with proteins that enable simple nuclear structures such as nucleosomes to form higher-order organisation such as chromatin fibres. The temporal association of regulatory proteins with DNA creates a dynamic environment capable of quickly responding to cellular requirements and distress. The response is often mediated through alterations in the chromatin structure, resulting in changed accessibility of specific DNA sequences that are then recognized by specific proteins. Anti-cancer drugs that target cellular DNA have been used clinically for over four decades, but it is only recently that nuclease specific drugs have been developed to not only target the DNA but also other components of the nuclear structure and its regulation. In this review, we discuss some of the new drugs aimed at primary DNA sequences, DNA secondary structures, and associated proteins, keeping in mind that these agents are not only important from a clinical perspective but also as tools for understanding the nuclear environment in normal and cancer cells.
Collapse
Affiliation(s)
- Stephanie M Nelson
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 10000, New Zealand
| | - Lynnette R Ferguson
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 10000, New Zealand
| | - William A Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 10000, New Zealand
| |
Collapse
|
18
|
Lockhart AC, Howard M, Hande KR, Roth BJ, Berlin JD, Vreeland F, Campbell A, Fontana E, Fiorentini F, Fowst C, Paty VA, Lankford O, Rothenberg ML. A Phase I Dose-Escalation and Pharmacokinetic Study of Brostallicin (PNU-166196A), a Novel DNA Minor Groove Binder, in Adult Patients with Advanced Solid Tumors. Clin Cancer Res 2004; 10:468-75. [PMID: 14760067 DOI: 10.1158/1078-0432.ccr-0658-03] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE This study was performed to determine the maximum tolerated dose, dose-limiting toxicities, and pharmacokinetics of brostallicin, a nonalkylating DNA minor groove binder and a synthetic derivative of distamycin A, given as a weekly i.v. infusion. EXPERIMENTAL DESIGN Using an accelerated dose escalation design, patients with advanced solid tumor malignancies were treated with brostallicin administered as a 10-min i.v. infusion on days 1, 8, and 15 of a 28-day cycle. The starting dose of brostallicin was 0.3 mg/m(2)/week. To study the pharmacokinetic behavior of brostallicin, serial blood samples were obtained before and after the first and last infusions during cycle 1, and in cycles 2 and 4 in a limited number of patients. RESULTS Fourteen patients received 32 complete cycles of brostallicin. Dose-limiting toxicity was febrile neutropenia and was observed in 3 of 5 patients treated at 4.8 mg/m(2)/week. The maximum tolerated dose and recommended Phase II dose was 2.4 mg/m(2)/week. The mean +/- SD terminal half-life at the maximum tolerated dose was 4.6 +/- 4.1 h. There was moderate distribution of brostallicin into tissues, and the clearance was approximately 20% of the hepatic blood flow. The area under the concentration time curve(0- infinity ) of brostallicin increased in a dose-linear fashion. No significant relationship was observed between any plasma pharmacokinetic parameter and clinical toxicities. There were no objective responses during the trial, but 5 patients had stable disease after two cycles of treatment. CONCLUSIONS The dose-limiting toxicity of weekly brostallicin was neutropenia. Systemic exposure increases linearly with dose. The recommended dose for Phase II studies is 2.4 mg/m(2) on days 1, 8, and 15 of a 28-day cycle.
Collapse
|
19
|
Jackson JA, Trevino AV, Herzig MC, Herman TS, Woynarowski JM. Matrix attachment region (MAR) properties and abnormal expansion of AT island minisatellites in FRA16B fragile sites in leukemic CEM cells. Nucleic Acids Res 2003; 31:6354-64. [PMID: 14576323 PMCID: PMC275474 DOI: 10.1093/nar/gkg832] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AT-rich minisatellites (AT islands) are sites of genomic instability in cancer cells and targets for extremely lethal AT-specific drugs, such as bizelesin. Here we investigated the AT islands in the FRA16B fragile site region for their possible roles in the organization of DNA on the nuclear matrix. The FRA16B AT island nominally spans approximately 3 kb of mostly >90% A/T DNA. In silico analysis indicates that this domain exhibits characteristics of nuclear matrix attachment regions (MARs): an exceptionally intense computed 'MAR potential' and profound duplex destabilization and flexibility. FRA16B repeats specifically bind to isolated nuclear matrices, which indicates their in vitro MAR function. This binding is several-fold greater than that of a known MAR in the c-myc gene. AT islands in fragile sites FRA16B and FRA16D are significantly more abundant in CEM cells that are hypersensitive to bizelesin compared to normal WI-38 cells. FRA16B overabundance in CEM is due to an approximately 10-fold expansion of FRA16B repeats. The expanded FRA16B minisatellites in CEM cells preferentially localize to the nuclear matrix-associated DNA indicating their in vivo MAR function. The unexpanded repeats in WI-38 cells localize to the loop DNA. The c-myc MAR is also matrix-associated in CEM cells while localizing to loop DNA in WI-38 cells. These results are the first to demonstrate that AT islands in fragile sites can function as MARs both in vitro and in vivo. The ability of FRA16B-mediated MAR sites to rearrange depending on the repeat expansion status could be relevant to both genomic instability of cancer cells and their sensitivity to AT-island targeting drugs.
Collapse
Affiliation(s)
- Jennifer A Jackson
- Department of Radiation Oncology, University of Texas Health Science Center, 14960 Omicron Drive, San Antonio, TX 78245, USA
| | | | | | | | | |
Collapse
|
20
|
Tercel M, Stribbling SM, Sheppard H, Siim BG, Wu K, Pullen SM, Botting KJ, Wilson WR, Denny WA. Unsymmetrical DNA cross-linking agents: combination of the CBI and PBD pharmacophores. J Med Chem 2003; 46:2132-51. [PMID: 12747786 DOI: 10.1021/jm020526p] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A set of 10 compounds, each combining the seco-1,2,9,9a-tetrahydrocyclopropa[c]benz[e]indol-4-one (seco-CBI) and pyrrolo[2,1-c][1,4]benzodiazepine (PBD) pharmacophores, was designed and prepared. These compounds were anticipated to cross-link between N3 of adenine and N2 of guanine in the minor groove of DNA. The compounds, which differ in the chain length separating the two alkylation subunits, and the configuration of the CBI portion, showed great variation in cellular toxicity (over 4 orders of magnitude in a cell line panel) with the most potent example exhibiting IC50s in the pM range. Cytotoxicity correlated with the ability of the compounds to cross-link naked DNA. Cross-linking was also observed in living cells, at much lower concentrations than for a related symmetrical PBD dimer. A thermal cleavage assay was used to assess sequence selectivity, demonstrating that the CBI portion controlled the alkylation sites, while the PBD substituent increased the overall efficiency of alkylation. Several compounds were tested for in vivo activity using a tumor growth delay assay against WiDr human colon carcinoma xenografts, with one compound (the most cytotoxic and most efficient cross-linker) showing a statistically significant increase in survival time following a single iv dose.
Collapse
Affiliation(s)
- Moana Tercel
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Schwartz GH, Patnaik A, Hammond LA, Rizzo J, Berg K, Von Hoff DD, Rowinsky EK. A phase I study of bizelesin, a highly potent and selective DNA-interactive agent, in patients with advanced solid malignancies. Ann Oncol 2003; 14:775-82. [PMID: 12702533 DOI: 10.1093/annonc/mdg215] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The aim of this study was to assess the feasibility of administering bizelesin, a cyclopropylpyrroloindole with extraordinarily high potency as a bifunctional DNA-damaging agent and selectivity for specific AT-rich DNA sequences, as a single i.v. bolus injection every 4 weeks in patients with advanced solid malignancies. The study also sought to determine the maximum tolerated dose (MTD) of bizelesin, characterize its pharmacokinetic behavior, and seek preliminary evidence of anticancer activity. PATIENTS AND METHODS Patients with advanced solid malignancies were treated with escalating doses of bizelesin as an i.v. bolus injection every 4 weeks. The selection of the specific starting dose, 0.1 micro g/m(2), which was equivalent to one-tenth the toxic dose low in dogs, factored in large interspecies differences in myelotoxicity as gauged using an ex vivo hematopoietic colony-forming assay. Due to concerns about the high potency of bizelesin and the large interspecies differences in toxicity, a conservative dose-escalation scheme was used for dose-level assignment to determine the MTD levels for both minimally pretreated (MP) and heavily pretreated (HP) patients. A variety of analytical assays were assessed to reliably measure bizelesin concentrations in plasma. RESULTS Sixty-two patients were treated with 185 courses of bizelesin at eight dose levels ranging from 0.1 to 1.5 micro g/m(2). Myelosuppression, principally neutropenia that was always brief, was the most common toxicity observed. Thrombocytopenia and anemia were uncommon and severe non-hematological effects were not observed. Severe neutropenia alone and/or associated with fever was consistently experienced by HP and MP patients at doses exceeding 0.71 and 1.26 micro g/m(2), respectively. These doses also resulted in functionally non-cumulative myelosuppression as repetitive treatment was well-tolerated. A 40% reduction in measurable disease lasting 24 months was noted in a patient with advanced ovarian carcinoma. Various analytical methods were evaluated but none demonstrated the requisite sensitivity to reliably quantify the minute plasma concentrations of bizelesin and metabolites resulting from administering microgram quantities of drug. CONCLUSIONS The highly potent and unique cytotoxic agent, bizelesin can be feasibly administered to patients with advanced solid malignancies. The recommended doses for phase II studies of bizelesin as a bolus i.v. injection every 4 weeks are 0.71 and 1.26 micro g/m(2) in HP and MP patients, respectively. The characteristics of the myelosuppression, the paucity of severe toxicities with repetitive treatment, the preliminary antitumor activity noted, and, above all, its unique mechanism of action as a selective DNA-damaging agent and high potency, warrant disease-directed evaluations of bizelesin in solid and hematopoietic malignancies and consideration of its use as a cytotoxic in targeted conjugated therapeutics.
Collapse
Affiliation(s)
- G H Schwartz
- Brooke Army Medical Center, San Antonio, TX 78229, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Woynarowski JM. Targeting critical regions in genomic DNA with AT-specific anticancer drugs. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1587:300-8. [PMID: 12084472 DOI: 10.1016/s0925-4439(02)00093-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Cellular DNA is not a uniform target for DNA-reactive drugs. At the nucleotide level, drugs recognize and bind short motifs of a few base pairs. The location of drug adducts at the genomic level depends on how these short motifs are distributed in larger domains. This aspect, referred to as region specificity, may be critical for the biological outcome of drug action. Recent studies demonstrated that certain minor groove binding (MGB) drugs, such as bizelesin, produce region-specific lesions in cellular DNA. Bizelesin binds mainly T(A/T)(4)A sites, which are on average scarce, but occasionally cluster in distinct minisatellite regions (200-1000 bp of approximately 85-100% AT), herein referred to as AT islands. Bizelesin-targeted AT islands are likely to function as strong matrix attachment regions (MARs), domains that organize DNA loops on the nuclear matrix. Distortion of MAR-like AT islands may be a basis for the observed inhibition of new replicon initiation and the extreme lethality of bizelesin adducts (<10 adducts/cell for cell growth inhibition). Hence, long AT-islands represent a novel class of critical targets for anticancer drugs. The AT island paradigm illustrates the potential of the concept of regional targeting as an essential component of the rational design of new sequence-specific DNA-reactive drugs.
Collapse
Affiliation(s)
- Jan M Woynarowski
- Cancer Therapy and Research Center, Institute for Drug Development, University of Texas Health Science Center at San Antonio, 14960 Omicron Drive, San Antonio, TX 78245, USA.
| |
Collapse
|
23
|
Abstract
DNA is the molecular target for many of the drugs that are used in cancer therapeutics, and is viewed as a non-specific target of cytotoxic agents. Although this is true for traditional chemotherapeutics, other agents that were discovered more recently have shown enhanced efficacy. Furthermore, a new generation of agents that target DNA-associated processes are anticipated to be far more specific and effective. How have these agents evolved, and what are their molecular targets?
Collapse
|
24
|
Herzig MCS, Rodriguez KA, Trevino AV, Dziegielewski J, Arnett B, Hurley L, Woynarowski JM. The genome factor in region-specific DNA damage: the DNA-reactive drug U-78779 prefers mixed A/T-G/C sequences at the nucleotide level but is region-specific for long pure AT islands at the genomic level. Biochemistry 2002; 41:1545-55. [PMID: 11814348 DOI: 10.1021/bi011907s] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bizelesin is the first anticancer drug capable of damaging specific regions of the genome with clusters of its binding sites T(A/T)(4)A. This study characterized the sequence- and region-specificity of a bizelesin analogue, U-78779, designed to interact with mixed A/T-G/C motifs. At the nucleotide level, U-78779 was found to prefer runs of A/Ts interspersed with 1 or 2 G/C pairs, although 25% of the identified sites corresponded to pure AT motifs similar to bizelesin sites. The in silico computational analysis showed that the preferred mixed A/T-G/C motifs distribute uniformly at the genomic level. In contrast, the secondary, pure AT motifs (A/T)(6)A were found densely clustered in the same long islands of AT-rich DNA that bizelesin targets. Mapping the sites and quantitating the frequencies of U-78779 adducts in model AT island and non-AT island naked DNAs demonstrated that clusters of pure AT motifs outcompete isolated mixed A/T-G/C sites in attracting drug binding. Regional preference of U-78779 for AT island domains was verified also in DNA from drug-treated cells. Thus, while the primary sequence preference gives rise to non-region-specific scattered lesions, the clustering of the minor pure AT binding motifs seems to determine region-specificity of U-78779 in the human genome. The closely correlated cytotoxic activities of U-78779 and bizelesin in several cell lines further imply that both drugs may share common cellular targets. This study underscores the significance of the genome factor in a drug's potential for region-specific DNA damage, by showing that it can take precedence over drug binding preferences at the nucleotide level.
Collapse
MESH Headings
- Adenine Nucleotides/isolation & purification
- Adenine Nucleotides/metabolism
- Antineoplastic Agents, Alkylating/chemistry
- Antineoplastic Agents, Alkylating/metabolism
- Antineoplastic Agents, Alkylating/toxicity
- Binding Sites/drug effects
- Binding Sites/genetics
- Cell Nucleus/chemistry
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cross-Linking Reagents/chemistry
- Cross-Linking Reagents/metabolism
- Cytosine Nucleotides/metabolism
- DNA Adducts/chemistry
- DNA Adducts/metabolism
- DNA Damage
- DNA, Neoplasm/chemistry
- DNA, Neoplasm/metabolism
- DNA, Superhelical/chemistry
- DNA, Superhelical/metabolism
- DNA, Viral/chemistry
- DNA, Viral/metabolism
- Dinucleotide Repeats/drug effects
- Duocarmycins
- Genome, Human
- Guanine Nucleotides/metabolism
- Humans
- Indoles/chemistry
- Indoles/metabolism
- Indoles/toxicity
- Intracellular Fluid/chemistry
- Intracellular Fluid/metabolism
- Reproducibility of Results
- Sequence Analysis, DNA
- Simian virus 40/drug effects
- Simian virus 40/genetics
- Thymine Nucleotides/isolation & purification
- Thymine Nucleotides/metabolism
- Tumor Cells, Cultured
- Urea/analogs & derivatives
- Urea/chemistry
- Urea/metabolism
- Urea/toxicity
Collapse
Affiliation(s)
- Maryanne C S Herzig
- Cancer Therapy and Research Center, 14960 Omicron Drive, San Antonio, Texas 78245, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Preferential damage to defined regions of genomic DNA by AT-specific anticancer drugs. ACTA ACUST UNITED AC 2002. [DOI: 10.1016/s1067-568x(02)80003-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
26
|
Lichanska AM. AT targets for anti-tumor drugs. Genome Biol 2001. [DOI: 10.1186/gb-2001-3-1-reports0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|