Antitumor drugs based on indolocarbazol derivatives

Cover Page

Cite item

Abstract

The aim of the work is to generalize the literature data on indolocarbazole derivatives with an antitumor activity.

Materials and methods. The objects of the study were the preparations based on indolocarbazole derivatives with the antitumor activity. To search for materials on the problem under study, the following search and information as well as library databases were used: Ebibrary, PubMed, CyberLeninka, ResearchGate, the State Register of Medicines, clinical trials registries clinline.ru and clinicaltrials.gov. The search for the following words / phrases was performed: indolocarbazoles, indolocarbazole derivatives, staurosporine, rebeccamycin, staurosporine derivatives. The search was conducted from January 11 until March 1, 2021. The compounds with a biological activity which were undergoing or had undergone preclinical and clinical trials, were taken into account. All the materials from 1977 to January 1, 2021, were taken into account.

Results. The materials obtained indicate that indolocarbazole derivatives are promising compounds for the creation of anticancer medicinal preparations due to their properties and peculiarities of the action mechanism. These drugs have a selective action due to the targeted interaction with specific molecular targets: kinases (especially protein kinase C and its isozymes), DNA and DNA topoisomerase. To date, many compounds from the class of indolocarbazoles have been synthesized and investigated. They have shown a high antitumor activity in the treatment of systemic and solid tumors. However, despite this, only one MP based on a staurosporine derivative, registered by the TN of Rydapt® (in the USA and EU countries) and Miticaid® (in the Russian Federation), is approved for use in the clinical practice.

Conclusion. Thus, the basic data from scientific publications on promising anticancer medicinal preparations based on compounds from the class of indolocarbazoles, have been summarized. The information is provided, in particular, on their molecular structure, the origin, classification, the main representatives of the class, which are at various stages of the research and are approved for use in the clinic.

Full Text

Abbreviations: P – pharmaceutical; MP – medicinal preparation; DF – dosage form; PKC – protein kinase C; DNA – deoxyribonucleic acid; TN – trade name

 

INTRODUCTION

Cancer is often referred to as “the pathology of the century” in the context of an endemic disease spreading throughout the world. Cancer has also been identified as “a true disease of modernity” (Roy Porter) or even “an important product of modernity” (Siddhartha Mukherjee). These two definitions are generally accepted and justified by a sharp increase in morbidity and mortality, which has been observed since the end of the 18th century [1]. In 2020, cancer continued to be one of the leading causes of death and an important obstacle to increasing life expectancy in all countries of the world. In 2019, The World Health Organization estimates cancer as the first or second leading cause of people’s deaths under 70 in 112 out of 183 countries, and is ranked as the third or fourth in 23 more countries [2].

Over the past two decades, cancer treatment with the use of pharmacological approaches has changed dramatically. Long years of fundamental and clinical research have led to the transition from classical anticancer therapy, characterized by a low selectivity of a drug action and accompanied by severe intoxication of the body, to more targeted antitumor “snipers” that effectively destroy populations of tumor cells with fewer side effects [3].

Among a wide range of anticancer drugs, compounds from the group of indolocarbazole derivatives are of particular interest. Indolocarbazoles are a unique class of indole alkaloids of a natural or synthetic kinds of origin, which have a number of therapeutic properties – antitumor, antibacterial, antiparasitic, antiviral, and an immunomodulatory activity [4–7].

The most significant biological profile of compounds from the group of indolocarbazole derivatives is their potential antitumor effect [8]. A distinctive feature of the action mechanism of these drugs is their ability to interact with several targets and induce various pathways of tumor cell death [9]. For them, such targets are DNA, topoisomerase and protein kinase C enzymes, which are responsible for regulating the main aspects of cell metabolism, including the progression of the cell cycle [10, 11].

Protein kinases C are a family of protein kinases, enzymes that phosphorylate proteins and thus participate in cell signaling cascades. The term “protein kinase C” refers to all described isoenzymes [12]. PKC inhibitors can reduce the expression of P-glycoprotein in tumor cells and thereby increase their sensitivity to chemotherapy [13]. PKC activation is also required for tumor angiogenesis [14].

Topoisomerases affect the topology of DNA and are able to relax their supercoiled molecules by introducing single- or double-stranded breaks followed by their DSB repair, as well as negative supercoils, or catenans. Inhibitors of these enzymes are widely used to suppress the activity of type I and / or type II tumor topoisomerases, blocking cells in the G2 phase and delaying their entry into mitosis [15].

Inhibitors of topoisomerases are ones of the most effective inducers of apoptosis, i. e., a programmed death of tumor cells [10]. In addition, a number of indolocarbazole derivatives with antiangiogenic activity have been synthesized. They are able to block vasculogenic mimicry in a tumor and restore the sensitivity of resistant cells to chemotherapeutic drugs [16, 17]. These features of the action mechanism determine a wide range of cytotoxic and antitumor activities of indolocarbazole derivatives.

THE AIM of the work is to generalize the literature data on indolocarbazole derivatives with an antitumor activity.

MATERIALS AND METHODS

The objects of the study were the preparations based on indolocarbazole derivatives with the antitumor activity. To search for materials on the problem under study, the following search and information as well as library databases were used: Ebibrary, PubMed, CyberLeninka, ResearchGate, the State Register of Medicines, clinical trials registries clinline.ru and clinicaltrials.gov. The search for the following words / phrases was performed: indolocarbazoles, indolocarbazole derivatives, staurosporine, rebeccamycin, staurosporine derivatives. The search was conducted from January 11 until March 1, 2021. The compounds with a biological activity that were undergoing or had undergone preclinical and clinical trials were taken into account. All the materials from 1977 until January 1, 2021, were taken into account.

The article is a review of the publications devoted to indolocarbazole derivatives, i.e., to the information on their structure, origin, classification, the main representatives of the class, which are at various stages of research and are approved for use in the clinic.

RESULTS AND DISCUSSION

General characteristics of indolocarbazole group compounds

The first indolocarbazoles were found out in streptomycetes and subsequently isolated from numerous representatives of flora and fauna. To date, this class of compounds has also been supplemented by a wide variety of synthetic compounds [18]1.

Indolocarbazoles are a class of heterocyclic compounds that include a planar ring consisting of indole and carbazole elements (Fig. 1, 2) [18]. Indole, carbazole and their derivatives are colorless solid crystalline substances that do not dissolve in water. Under standard conditions, the melting point of indole is 52°C, of carbazole, it is 247–248°C, and their boiling points are 253°C and 354–355°C, respectively2,3. The carbazole fragment serves as a ligand for many receptors and has the property of reverse coupling to enzymes, in particular, to DNA topoisomerase I [19], and the indole element is responsible for the interaction with DNA [20, 21].

 

Figure 1 – Structural formula of indole

 

Figure 2 – Structural formula of carbazole

 

The class of indolocarbazoles includes 5 subclasses of compounds differing in the structure of a planar aromatic ring. In this case, 5 isomers of the polycyclic system – indolo[2, 3-a]carbazole (1), indolo[2, 3-b]carbazole (2), indolo[2,3-c]carbazole (3), indolo[3, 2-a]carbazole (4) and indolo[3, 2-b]carbazole (5) (Fig. 3) [18] – are meant.

 

Figure 3 – Structural formulas of indolocarbazole isomers

 

The most extensive, biologically significant and studied in detail is the subclass of 11,12-dihydroindolo[2, 3-a]carbazole derivatives, including mainly compounds having indolo[2, 3-a]pyrrolo[3,4-c]carbazole ring, in which 2 indole fragments are linked through a benzene ring with an amide or imide group. The indole moieties are linked through 1 or 2 bonds to the carbohydrate moiety. At the same time, this subclass also includes a small group of compounds that do not include an additional pyrrole ring in their composition [18].

Based on the number of glycosidic bonds that bind the carbohydrate moiety to the isoindole backbone, indolocarbazole derivatives can be divided into 2 subclasses – compounds of the staurosporin group (a) and rebeccamycin (c). In staurosporin and its derivatives, for example, K252a (b), glycoside is bound to 2 indole groups through nitrogen atoms, in contrast to the representatives of the rebeccamycin group, for example, cholyrin A (d), in which the carbohydrate residue is attached to only one indole. The staurosporin heterocycle is connected to the lactone ring, the rebeccamycin heterocycle – to the imide ring (Fig. 4) [25]. Both monosaccharides [23, 24] and disaccharides [25–28] can be included in the structure of indolocarbazoles as carbohydrate residues.

 

Figure 4 – Structural formulas of indolocarbazole derivatives

 

However, the literature also describes indolocarbazole derivatives with a different structure, for example, Go 6976 (Fig. 5) and AEB071 (sotrastaurin) (Fig. 6) – selective inhibitors of protein kinase C α, β1, δ, υ and ζ isozymes. Unlike staurosporine, Go 6976 is methyl- and cyanoalkyl-substituted non-glycoside indolocarbazole [29]; sotrastaurin contains a piperazine ring, in which the nitrogen atom in the ring carries an aryl group [30].

 

Figure 5 – Structural formula of Go 6976

 

Figure 6 – Structural formula of sotrastaurine

 

Various modifications of natural and synthetic derivatives of indolocarbazoles lead to changes in their physicochemical properties and biological activity. These factors are important for the development of potential antitumor agents. It is obvious that the antitumor effect of the target compound can be influenced by both substituents in the aglycone and the nature of the glycosidic residue, changing the pharmacodynamic and pharmacokinetic properties. [31].

Representatives of indolocarbazoles class

Staurosporin, the first of the discovered compounds of indolocarbazole derivatives, was isolated in 1977 at the Kitasato Institute (Japan) from the cultures of Streptomyces staurosporeus and Streptomyces actuosus. Staurosporin has antifungal and hypotensive effects, inhibits platelet aggregation, and is a potent inhibitor of various protein kinases. These factors lead to its use as an antitumor drug [32, 33].

In 1983, the parent of the second group of indolocarbazole derivatives, rebeccamycin (NSC 655649) (Bristol-Myers Co., USA) [34], which is structurally similar to staurosporin, but has a weaker inhibitory activity against protein kinases, was isolated from the strain of actinobacteria C-38383 [35]. The mechanism of the antitumor action of rebeccamycin is associated with the inhibition of topoisomerase I, which is due to its ability to interact with DNA [36].

In order to increase the solubility and the biological activity, the hydrophobic indolocarbazoles staurosporin and rebeccamycin underwent various modifications: a) the addition of substituents to the upper heterocycle, replacement of atoms in the upper heterocycle or removal of a heterocycle, b) modification of flat chromophore, c) modification of replacement or removal of the carbohydrate moiety [37–39].

Staurosporine derivatives

Midostaurine (CGP 41251, PKC 412, NVP-PKC412) (Fig. 7) is an N-benzoyl4 derivative of staurosporin; it is a synthetic inhibitor of many kinases, including FLT3 and KIT, with antiangiogenic and antitumor activities [40]. It is approved by the FDA5 and EMA6 by the TN of Rydapt® (Novartis Pharmaceuticals, Switzerland)7, in Russia this drug is registered by the TN of Miticaid® (LP-005927)8. This MP is a liquid capsule for the oral administration, each capsule contains 25 mg of midostaurin9.

 

Figure 7 – Structural formula of midostaurine

 

Enzastaurine (LY-317615, LY317615) (Eli Lilly and Company, USA) (Fig. 8) is an acyclic bisindolylmaleimide derived from staurosporin that selectively inhibits protein kinase-β. The mechanism of the antitumor action of enzastaurin is due to several effects. First, the drug has anti-angiogenic properties associated with a decrease in the level of vascular endothelial growth factor. Second, enzastaurine directly induces the death of tumor cells by reducing the phosphorylation of protein kinase [41]. Numerous studies have been carried out in mono- and combined therapy of oncological diseases of various nosologies, for example, tumors of the nervous system [42–44], colon [45], lymphoma [46–49], Waldenstrom’s myeloma and macroglobulinemia [50], non-small-cell lung cancer [51], prostate [52], ovaries [53], etc.

 

Figure 8 – Structural formula of enzastaurin

 

Sotrastaurine (AEB071) (Novartis Pharmaceuticals, Switzerland) is a selective inhibitor of PKC β [57], which prevents the activation of T cells, has a piperazine ring; therefore, this compound can be attributed to the class of organic compounds known as n-arylpiperazines (Fig. 6) [55]. The use of sotrastaurine in the treatment of diffuse large B-cell lymphoma, stomach cancer [56], ulveal melanoma [57], psoriasis [58], as well as in kidney transplantation, has been investigated [59, 60].

Lestaurtinib (A-154475, A-154475.0, CEP-701, KT-555, KT-5555, KT5555, SP-924, SP924, SPM-924) (Cephalon, Inc., USA) (Fig. 9)10 has been studied in the treatment of infections of the central nervous system caused by free living amoebae [61], myeloid leukemia [62–64], polycythemia and essential thrombocythemia [65], myelofibrosis [66], prostate cancer [67, 68], neuroblastoma [69, 70] psoriasis [71].

 

Figure 9 – Structural formula of Lestaurtinib

 

Among the staurosporin derivatives, the antibiotic K-252a (Kyowa Hakko Kogyo Co., Ltd., Japan) (Fig. 10)11, isolated from the culture of Nocardiopsis sp. K-252a, is a unique in its structure indolocarbazole glycoside, and exhibits a powerful neuroprotective antitumor activity. K-252a consists of K-252c and an extraordinary dihydrostreptose fragment linked together by two C-N bonds [72]. Its semi-synthetic derivative KT5720 inhibits cAMP-dependent protein kinase. The activity of KT5720 has been confirmed on granulosa cells of animal ovaries [73, 74].

 

Figure 10 – Structural formula of K-252a

 

A promising semi-synthetic staurosporine derivative is stauprimide (The Scripps Research Institute, USA), which inhibits the transcription of the MYC NME2 oncogene and also increases the efficiency of directed differentiation of embryonic stem cells [75, 76].

CEP-11981 (Cephalon, Inc., USA) (Fig. 11) is a targeted drug for oral administration, exhibiting a high inhibitory activity against several targets – receptors for vascular endothelial growth factor 1 and 2, tyrosine kinase 2, and a fibroblast growth factor-1, protooncogene c-SRC, and Aurora A. The studies of the pharmacological activity in animal and human tumor models have shown sustained dose-dependent antiangiogenic and antitumor effects. In addition, CEP-11981 has shown an excellent bioavailability, a metabolic stability, and other pharmacokinetic properties. Phase I clinical trials to evaluate the pharmacokinetics and pharmacodynamics of CEP-11981 in patients with advanced, recurrent / refractory solid tumors, have been completed [77, 78].

 

Figure 11 – Structural formula of CEP-11981

 

Go 6976 (Godecke AG, Germany) (Fig. 5) is an indolocarbazole derivative containing a propane nitrile radical instead of a glycosidic residue12. Go 6976 is a selective inhibitor of PKC α and β, it moderately inhibits the activation of protein kinase regulated by extracellular signals [79]. In addition, this indolocarbazole is a potential anticancer drug due to its ability to stimulate the formation of cellular compounds (the formation of an increased number of desmosomes and adhesions), to suppress migration and invasion of tumor cells [80].

In the literature, there are also data on many other compounds of staurosporin derivatives: ZHD-0501 [81]; BMY-41950 (RK 1409) [82]; UCN-01 and UCN-02 [83]; CEP-7055 and CEP-5214 [84]; CEP-701; CEP-2563 and CEP-751 (KT-6587) [85]; KT5926 [86]; Ro 318220 and GF 109203X [87]; CEP-134713, and others.

Among the domestic compounds of staurosporine derivatives, the most famous are the N-glycosides of indolo [2, 3-a] pyrrolo [3,4-c] carbazole-5,7-diones of indolocarbazoles: LHS-976, LHS-983, LHS-985, LHS-999, LHS-1006, LHS-1007, LHS-1040, LHS-1054, LHS-1098, LHS-1208, LHS-1269, etc. [88-93]. Today, compounds LHS-1208 and LHS-1269 are the most studied among them as antitumor agents.

An indolocarbazole derivative LHS-1208 exhibits a strong inhibitory activity against kinases14 – cyclin-dependent kinase, protein kinase C and tyrosine kinase; the second target is DNA and the DNA topoisomerase complex. To date, preclinical trials of an injectable dosage form LHS-1208 containing dimethyl sulfoxide and a solubilizer Kollidon 17PF as a co-solvent of the hydrophobic active substance, have been completed [94]. For this compound, a liposome-based DF was also developed in the form of a lyophilisate for the preparation of an injection emulsion [95].

LHS-1269 is an indolocarbazole derivative with a carbohydrate residue xylose, which has cytotoxic and antiangiogenic effects and has shown a high antitumor activity against a number of transplanted ascites and solid tumor models [96, 97]. To date, a composition and technology for producing an injectable liposomal dosage form have been developed for LHS-1269 [98].

Rebeccamycin derivatives

On the basis of rebeccamycin, a glycosyl-dichloroindolocarbazole analogue with the improved water solubility denoted as becatecarin (BMS-181176, BMY-27557, NSC-655649, XL 119, XL-119, XL119) (National Cancer Institute, USA) (Fig. 12)15 was obtained [99]. Becatecarin is an antitumor antibiotic with an inhibitory activity against topoisomerase I and topoisomerase II, as well as the ability to intercalate DNA [100, 104]. It has been studied in the treatment of lung cancer [101, 104], blood cancer [102], tumors of the nervous system [99] and solid tumors [103].

 

Figure 12 – Structural formula of becatecarin

 

NB-506 (Banyu Co., Japan) (Fig. 13) is a glycoside derivative of rebeccamycin, the antitumor activity of which is due to its ability to interact with DNA and inhibit topoisomerase I. The glucose residue attached to the planar chromophore of indolocarbazole, plays a significant role in the interaction of drugs with nucleic acids; it promotes the stabilization of covalent complexes of topoisomerase I – DNA [105]. It has been reported that NB-506 is in clinical trials [106].

 

Figure 13 – Structural formula of NB-506

 

Edothecarin (J-107088, J-107088, PF-804950, PHA-782615) (Banyu Co., Japan) (Fig. 14) is a NB-506 derivative with a broad spectrum of antitumor activity, it is a topoisomerase I inhibitor that induces cleavage of single-stranded DNA more effectively than original indolocarbazole or camptothecin. In contrast to other inhibitors of topoisomerase I, the antitumor activity is less dependent on the cell cycle. Despite the fact that J-107088 has a structure similar to staurosporin, this drug does not possess the properties of a protein kinase inhibitor [107]. It has been actively studied in mono- and combined therapy of oncological diseases [108–115].

 

Figure 14 – Structural formula of edothecarin

 

It was also found out that when grown in a specific medium containing 0.05% potassium bromide, Saccharothrix aerocolonigenes ATCC 39243 produces a rebeccamycin analog which has been indicated as brombeccamycin. It has the same structure as rebeccamycin, except the replacement of two chlorine atoms with bromine atoms in the molecule. The authors of the study suggest that the compound has an activity against mouse P-388 leukemia [116].

Rebeccamycin-based compounds have also been obtained. They are: BMS-250749, BMS-210287, BMS-251873, SA315F, AT2433-A1, AT2433-A2, AT2433-B1, AT2433-B2, etc. [117].

CONCLUSION

An important issue in medical science is the creation of new MPs for the treatment of cancer. Indolocarbazole derivatives are a promising class of anticancer drugs characterized by a directed mechanism of the action on targets such as kinases (especially PKC and its isozymes), DNA and DNA topoisomerases I and II. These compounds, along with the antitumor effect, have a wide spectrum of a biological activity, which also makes it possible to use them in the therapy of other nosologies, including transplantology.

To date, a fairly large number of compounds that are at various stages of preclinical and clinical studies, have been synthesized. They belong to two subclasses – derivatives of staurosporin and rebeccamycin.

However, for clinical practice, only one drug based on a staurosporine derivative, midostaurine, registered abroad by the TN of Rydapt® (in the Russian Federation its TN is Miticaid) has been approved for use. Therefore, to expand the arsenal of targeted anticancer drugs, it is necessary to study the known synthesized indolocarbazole derivatives, as well as to search for new compounds with improved characteristics, further.

FUNDING

This study did not receive any support from outside organizations.

CONFLICT OF INTERESTS

The authors declare no conflict of interest.

AUTHORS’ CONTRIBUTIONS

Alexander P. Kolpaksidi – searching for materials, writing, planning and editing the review; Maria V. Dmitrieva – searching for materials, planning and editing the review; Ilya V. Yarosh – searching for materials; Ivan I. Krasnyuk – planning and searching for materials.

 

1 A few examples of indolocarbazoles found in the ClassyFire database. The Metabolomics Innovation Centre (TMIC) [Electronic resource]. Available from: http://classyfire.wishartlab.com/tax_nodes/C0001866

2 PubChem Compound Summary for CID 798, Indole. PubChem. Bethesda (MD): National Library of Medicine (US), National Center for Biotechnology Information [Electronic resource]. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/6854

3 PubChem Compound Summary for CID 6854, Carbazole. PubChem. Bethesda (MD): National Library of Medicine (US), National Center for Biotechnology Information [Electronic resource]. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/6854

4 PubChem. Compound Summary for CID 9829523, Midostaurin. PubChem. Bethesda (MD): National Library of Medicine (US), National Center for Biotechnology Information [Electronic resource]. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/Midostaurin.

5 Highlights of prescribing information. Rydapt. U.S. Food and Drug Administration [Electronic resource]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/207997s000lbl.pdf

6 Rydapt. European Medicines Agency [Electronic resource]. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/rydapt

7 RYDAPT® (midostaurin) Capsules. AML & ASM Treatment Novartis AG [Electronic resource]. Available from: https://www.rydapt.com.

8 Instructions for the use of a medicinal product for medical application of Mitikaid®. State Registermedicines. Available from: http://grls.rosminzdrav.ru

9 Highlights of prescribing information. Rydapt. U.S. Food and Drug Administration [Electronic resource]. Available from: https://www. accessdata.fda.gov/drugsatfda_docs/label/2017/207997s000lbl.pdf

10 Lestaurtinib. DrugBank. Available from: https://www.drugbank.ca/drugs/DB06469

11 PubChem. Compound Summary for CID 3035817, Antibiotic K 252a. PubChem. Bethesda (MD): National Library of Medicine (US), National Center for Biotechnology Information [Электронный ресурс]. URL: https://pubchem.ncbi.nlm.nih.gov/compound/Antibiotic-K-252a

12 PubChem Compound Summary for CID 3501, Go-6976. PubChem. Bethesda (MD): National Library of Medicine (US), National Center for Biotechnology Information [Electronic resource]. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/Go-6976

13 CEP-1347. DrugBank [Electronic resource]. Available from: https://go.drugbank.com/drugs/DB05403

14 Isoenmers are not specified.

15 PubChem Compound Summary for CID 101524, Becatecarin. PubChem. Bethesda (MD): National Library of Medicine (US), National Center for Biotechnology Information [Электронный ресурс] Available from: https://pubchem.ncbi.nlm.nih.gov/compound/Becatecarin

×

About the authors

Alexander P. Kolpaksidi

I.M. Sechenov First Moscow State Medical University (Sechenov University); N.N. Blokhin National Medical Research Center of Oncology

Author for correspondence.
Email: aleksandrkolpaksidi@gmail.com
ORCID iD: 0000-0001-9388-1019

post-graduate student of the Department of Pharmaceutical Technology of the Institute of Pharmacy; laboratory assistant-researcher Laboratory for the Development of Dosage Forms Research Institute for Experimental Diagnostics and Therapy of Tumors

Russian Federation, Bldg. 2, 8, Trubetskaya St., Moscow, Russia, 119991; 24, Kashirskoe sh., Moscow, Russia, 115478

Maria V. Dmitrieva

N.N. Blokhin National Medical Research Center of Oncology

Email: dmitrieva.m@ronc.ru
ORCID iD: 0000-0001-6740-5692

Candidate of Sciences (Pharmacy), Senior Researcher Laboratory for the Development of Dosage Forms Research Institute for Experimental Diagnostics and Therapy of Tumors

Russian Federation, 24, Kashirskoe sh., Moscow, Russia, 115478

Ilya V. Yarosh

I.M. Sechenov First Moscow State Medical University (Sechenov University)

Email: ilya96yarosh@gmail.com
ORCID iD: 0000-0002-3347-9674

resident of the Department of Pharmaceutical Technology of the Institute of Pharmacy

Russian Federation, Bldg. 2, 8, Trubetskaya St., Moscow, Russia, 119991

Ivan I. Krasnyuk

I.M. Sechenov First Moscow State Medical University (Sechenov University)

Email: krasnyuki@mail.ru
ORCID iD: 0000-0003-4382-7377

Doctor of Sciences (Pharmacy), Professor, head of the Department of Pharmaceutical Technology of the Institute of Pharmacy

Russian Federation, Bldg. 2, 8, Trubetskaya St., Moscow, Russia, 119991

References

  1. Falzone L, Salomone S, Libra M. Evolution of Cancer Pharmacological Treatments at the Turn of the Third Millennium. Front Pharmacol. 2018 Nov 13;9:1300. doi: 10.3389/fphar.2018.01300.
  2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021 May;71(3):209–49. doi: 10.3322/caac.21660.
  3. Kroschinsky F, Stölzel F, von Bonin S, Beutel G, Kochanek M, Kiehl M, Schellongowski P; Intensive Care in Hematological and Oncological Patients (iCHOP) Collaborative Group. New drugs, new toxicities: severe side effects of modern targeted and immunotherapy of cancer and their management. Crit Care. 2017 Apr 14;21(1):89. doi: 10.1186/s13054-017-1678-1.
  4. Olano C, Méndez C, Salas JA. Antitumor compounds from marine actinomycetes. Mar Drugs. 2009 Jun 11;7(2):210–48. doi: 10.3390/md7020210.
  5. Bashir M, Bano A, Ijaz AS, Chaudhary BA. Recent Developments and Biological Activities of N-Substituted Carbazole Derivatives: A Review. Molecules. 2015 Jul 23;20(8):13496–517. doi: 10.3390/molecules200813496.
  6. Cartuche L, Sifaoui I, López-Arencibia A, Bethencourt-Estrella CJ, San Nicolás-Hernández D, Lorenzo-Morales J, Piñero JE, Díaz-Marrero AR, Fernández JJ. Antikinetoplastid Activity of Indolocarbazoles from Streptomyces sanyensis. Biomolecules. 2020 Apr 24;10(4):657. doi: 10.3390/biom10040657.
  7. Knübel G, Larsen LK, Moore RE, Levine IA, Patterson GM. Cytotoxic, antiviral indolocarbazoles from a blue-green alga belonging to the Nostocaceae. J Antibiot (Tokyo). 1990 Oct;43(10):1236–9. doi: 10.7164/antibiotics.43.1236.
  8. Wang W, Lv M, Zhao X, Zhang J. Developing a Novel Indolocarbazole as Histone Deacetylases Inhibitor against Leukemia Cell Lines. J Anal Methods Chem. 2015;2015:675053. doi: 10.1155/2015/675053.
  9. Kiseleva MP, Pokrovsky VS, Tataskiy VV, Borisova LM, Golubeva IS, L. Ektova1 LV. indolocarbazole derivatives – a promising class of anticancer drugs. Russian Biotherapeutic Journal. 2018;17(4):20–6. doi: 10.17650/1726-9784-2018-17-4-20-26. Russian
  10. Dezhenkova LG., Tsvetkov VB., Shtil AA. Topoisomerase I and II inhibitors: chemical structure, mechanisms of action and role in cancer chemotherapy. (Russian) Russian Chemical Reviews. 2014;83(1):82–94.
  11. Cartuche L, Reyes-Batlle M, Sifaoui I, Arberas-Jiménez I, Piñero JE, Fernández JJ, Lorenzo-Morales J, Díaz-Marrero AR. Antiamoebic Activities of Indolocarbazole Metabolites Isolated from Streptomyces sanyensis Cultures. Mar Drugs. 2019 Oct 17;17(10):588. doi: 10.3390/md17100588.
  12. Mellor H, Parker PJ. The extended protein kinase C superfamily. Biochem J. 1998 Jun 1;332 (Pt 2)(Pt 2):281–92. doi: 10.1042/bj3320281.
  13. List AF. Non-P-glycoprotein drug export mechanisms of multidrug resistance. Semin Hematol. 1997 Oct;34(4 Suppl 5):20–4.
  14. Blokhin DY, Chmutin EF, Ivanov PK. Molecular targets for anticancer therapy: growth factors, angiogenesis and apoptosis. Russian Biotherapeutic Journal. 2011;10(3);17–24. Russian
  15. Treshalin MI, Neborak EV. topoisomerases: features of the action, classification, cell functions, inhibition, anthrafurandion. Russian Journal of Oncology. 2018;23(2):60–70. (Russian) DOI: 0.18821/1028-9984-2018-23-2-60-7
  16. Vartanjan AA, Baryshnikova MA, Eremina VA, Miniker TD, Tikhonova NI, Kuz’mina NE, Ehktova LV. indolocarbazole derivative blocking tumour vasculogenic mimicry. Patent No. RU 2557554 C1. 27 July 2015. Russian
  17. Acero N, Braña MF, Añorbe L, Domínguez G, Muñoz-Mingarro D, Mitjans F, Piulats J. Synthesis and biological evaluation of novel indolocarbazoles with anti-angiogenic activity. Eur J Med Chem. 2012 Feb;48:108-13. doi: 10.1016/j.ejmech.2011.11.040.
  18. Zenkov R.G., Ektova L.V., Vlasova O.A., Belitsky G.A., Yakubovskaya M.G., Kirsanov K.I. Indolo[2,3-a]carbazoles: diversity, biological properties, application in antitumor therapy. Chemistry of Heterocyclic Compounds. 2020;56(6):644–58. (Russian) doi: 10.17650/1726-9784-2019-18-2-32-39.
  19. Parisi OI, Morelli C, Puoci F, Saturnino C, Caruso A, Sisci D, Trombino GE, Picci N, Sinicropi MS. Magnetic molecularly imprinted polymers (MMIPs) for carbazole derivative release in targeted cancer therapy. J Mater Chem B. 2014 Oct 14;2(38):6619–6625. doi: 10.1039/c4tb00607k.
  20. Caruso A, Ceramella J, Iacopetta D, Saturnino C, Mauro MV, Bruno R, Aquaro S, Sinicropi MS. Carbazole Derivatives as Antiviral Agents: An Overview. Molecules. 2019 May 17;24(10):1912. doi: 10.3390/molecules24101912.
  21. Lafayette EA, de Almeida SMV, Cavalcanti Santos RV, de Oliveira JF, Amorim CADC, da Silva RMF, Pitta MGDR, Pitta IDR, de Moura RO, de Carvalho Júnior LB, de Melo Rêgo MJB, de Lima MDCA. Synthesis of novel indole derivatives as promising DNA-binding agents and evaluation of antitumor and antitopoisomerase I activities. Eur J Med Chem. 2017 Aug 18;136:511–22. doi: 10.1016/j.ejmech.2017.05.012.
  22. Speck K, Magauer T. The chemistry of isoindole natural products. Beilstein J Org Chem. 2013 Oct 10;9:2048-78. doi: 10.3762/bjoc.9.243.
  23. Borisova LM, Golubeva IS, Goryunova OV, Eremina VA, Zhukova OS, Kiseleva MP, Markova NP, Medvedeva LA, Melnik SY, Miniker TD, Smirnova ZS, Tikhonova NI, Fetisova LV, Ektova LV, Yartseva IV. N-glycosides of indolo[2,3-a]pyrrolo[3,4-c]carbazoles with antitumor activity. Patent No. 2548045. 27 Feb 2014. Russian
  24. Ohkubo M, Nishimura T, Kawamoto H, Nakano M, Honma T, Yoshinari T, Arakawa H, Suda H, Morishima H, Nishimura S. Synthesis and biological activities of NB-506 analogues modified at the glucose group. Bioorg Med Chem Lett. 2000 Mar 6;10(5):419–22. doi: 10.1016/s0960-894x(00)00004-4.
  25. Carrasco C, Facompré M, Chisholm JD, Van Vranken DL, Wilson WD, Bailly C. DNA sequence recognition by the indolocarbazole antitumor antibiotic AT2433-B1 and its diastereoisomer. Nucleic Acids Res. 2002 Apr 15;30(8):1774–81. doi: 10.1093/nar/30.8.1774.
  26. Animati F, Berettoni M, Bigioni M, Binaschi M, Felicetti P, Gontrani L, Incani O, Madami A, Monteagudo E, Olivieri L, Resta S, Rossi C, Cipollone A. Synthesis, biological evaluation, and molecular modeling studies of rebeccamycin analogues modified in the carbohydrate moiety. ChemMedChem. 2008 Feb;3(2):266–79. doi: 10.1002/cmdc.200700232.
  27. Singh S, Kim Y, Wang F, Bigelow L, Endres M, Kharel MK, Babnigg G, Bingman CA, Joachimiak A, Thorson JS, Phillips GN Jr. Structural characterization of AtmS13, a putative sugar aminotransferase involved in indolocarbazole AT2433 aminopentose biosynthesis. Proteins. 2015 Aug;83(8):1547-54. doi: 10.1002/prot.24844.
  28. Shaaban KA, Elshahawi SI, Wang X, Horn J, Kharel MK, Leggas M, Thorson JS. Cytotoxic Indolocarbazoles from Actinomadura melliaura ATCC 39691. J Nat Prod. 2015 Jul 24;78(7):1723–9. doi: 10.1021/acs.jnatprod.5b00429.
  29. Martiny-Baron G, Kazanietz MG, Mischak H, Blumberg PM, Kochs G, Hug H, Marmé D, Schächtele C. Selective inhibition of protein kinase C isozymes by the indolocarbazole Gö 6976. J Biol Chem. 1993 May 5;268(13):9194–7.
  30. Wagner J, von Matt P, Sedrani R, Albert R, Cooke N, Ehrhardt C, Geiser M, Rummel G, Stark W, Strauss A, Cowan-Jacob SW, Beerli C, Weckbecker G, Evenou JP, Zenke G, Cottens S. Discovery of 3-(1H-indol-3-yl)-4-[2-(4-methylpiperazin-1-yl) quinazolin-4-yl] pyrrole-2, 5-dione (AEB071), a potent and selective inhibitor of protein kinase C isotypes. Journal of medicinal chemistry. 2009 Sept; 52(20):6193–6. doi: 10.1021/jm901108b.
  31. Kiseleva MP, Pokrovsky VS, Borisova LM, Golubeva IS, Ektova LV. N-glycosidesindolo[2,3,-a]pyrrolo[3,4,-c]carbazole derivatives chemical structure influence on antitumor activity. Russian Biotherapeutic Journal. 2019;18(2). (Russian) DOI: 10.17650 / 1726-9784-2019-18-2-32-39.
  32. Omura S, Sasaki Y, Iwai Y, Takeshima H. Staurosporine, a potentially important gift from a microorganism. J Antibiot (Tokyo). 1995 Jul;48(7):535–48. doi: 10.7164/antibiotics.48.535.
  33. Salas AP, Zhu L, Sánchez C, Braña AF, Rohr J, Méndez C, Salas JA. Deciphering the late steps in the biosynthesis of the anti-tumour indolocarbazole staurosporine: sugar donor substrate flexibility of the StaG glycosyltransferase. Mol Microbiol. 2005 Oct;58(1):17–27. doi: 10.1111/j.1365-2958.2005.04777.x.
  34. Bush JA, Long BH, Catino JJ, Bradner WT, Tomita K. Production and biological activity of rebeccamycin, a novel antitumor agent. J Antibiot (Tokyo). 1987 May;40(5):668–78. doi: 10.7164/antibiotics.40.668.
  35. Nettleton D.E.,.Doyle T.W., Krishnan B., Matsumoto G.K., Clardy J. Isolation and structure of rebeccamycin – a new antitumor antibiotic from Nocardia aerocoligenes. Tetrahedron. Letters. 1985;26:4011–4014. doi: 10.1016/S0040-4039(00)89280-1.
  36. Bailly C, Riou JF, Colson P, Houssier C, Rodrigues-Pereira E, Prudhomme M. DNA cleavage by topoisomerase I in the presence of indolocarbazole derivatives of rebeccamycin. Biochemistry. 1997 Apr 1;36(13):3917–29. doi: 10.1021/bi9624898.
  37. Wada Y., Nagasaki H., Tokuda M., Orito K. Synthesis of N-protected staurosporinones. J. Org. Chem. 2007;72:2008–14. doi: 10.1021/jo062184r.
  38. Prudhomme M. Biological targets of antitumor indolocarbazoles bearing a sugar moiety. Curr Med Chem Anticancer Agents. 2004 Nov;4(6):509–21. doi: 10.2174/1568011043352650.
  39. Gescher A. Analogs of staurosporine: potential anticancer drugs? Gen Pharmacol. 1998 Nov;31(5):721–8. doi: 10.1016/s0306-3623(98)00069-x.
  40. Kim ES. Midostaurin: First Global Approval. Drugs. 2017 Jul;77(11):1251–9. doi: 10.1007/s40265-017-0779-0.
  41. Jane EP, Pollack IF. Enzastaurin induces H2AX phosphorylation to regulate apoptosis via MAPK signalling in malignant glioma cells. Eur J Cancer. 2010 Jan;46(2):412–9. doi: 10.1016/j.ejca.2009.10.014.
  42. Kilburn LB, Kocak M, Decker RL, Wetmore C, Chintagumpala M, Su J, Goldman S, Banerjee A, Gilbertson R, Fouladi M, Kun L, Boyett JM, Blaney SM. A phase 1 and pharmacokinetic study of enzastaurin in pediatric patients with refractory primary central nervous system tumors: a pediatric brain tumor consortium study. Neuro Oncol. 2015 Feb;17(2):303–11. doi: 10.1093/neuonc/nou114.
  43. Butowski N, Chang SM, Lamborn KR, Polley MY, Pieper R, Costello JF, Vandenberg S, Parvataneni R, Nicole A, Sneed PK, Clarke J, Hsieh E, Costa BM, Reis RM, Hristova-Kazmierski M, Nicol SJ, Thornton DE, Prados MD. Phase II and pharmacogenomics study of enzastaurin plus temozolomide during and following radiation therapy in patients with newly diagnosed glioblastoma multiforme and gliosarcoma. Neuro Oncol. 2011 Dec;13(12):1331–8. doi: 10.1093/neuonc/nor130.
  44. Wick W, Puduvalli VK, Chamberlain MC, van den Bent MJ, Carpentier AF, Cher LM, Mason W, Weller M, Hong S, Musib L, Liepa AM, Thornton DE, Fine HA. Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J Clin Oncol. 2010 Mar 1;28(7):1168–74. doi: 10.1200/JCO.2009.23.2595.
  45. Glimelius B, Lahn M, Gawande S, Cleverly A, Darstein C, Musib L, Liu Y, Spindler KL, Frödin JE, Berglund A, Byström P, Qvortrup C, Jakobsen A, Pfeiffer P. A window of opportunity phase II study of enzastaurin in chemonaive patients with asymptomatic metastatic colorectal cancer. Ann Oncol. 2010 May;21(5):1020–6. doi: 10.1093/annonc/mdp521.
  46. He Y, Li J, Ding N, Wang X, Deng L, Xie Y, Ying Z, Liu W, Ping L, Zhang C, Song Y, Zhu J. Combination of Enzastaurin and Ibrutinib synergistically induces anti-tumor effects in diffuse large B cell lymphoma. J Exp Clin Cancer Res. 2019 Feb 18;38(1):86. doi: 10.1186/s13046-019-1076-4.
  47. Li X, Fang X, Li S, Zhang W, Yang N, Cui Y, Huang H, Cai R, Lin X, Fu X, Hong H, Lin T. A pharmacokinetic and safety study of a fixed oral dose of enzastaurin HCl in native Chinese patients with refractory solid tumors and lymphoma. Oncotarget. 2016 Apr 5;7(14):18585–93. doi: 10.18632/oncotarget.7875.
  48. Morschhauser F, Seymour JF, Kluin-Nelemans HC, Grigg A, Wolf M, Pfreundschuh M, Tilly H, Raemaekers J, van ‘t Veer MB, Milpied N, Cartron G, Pezzutto A, Spencer A, Reyes F, Dreyling M. A phase II study of enzastaurin, a protein kinase C beta inhibitor, in patients with relapsed or refractory mantle cell lymphoma. Ann Oncol. 2008 Feb;19(2):247–53. doi: 10.1093/annonc/mdm463.
  49. Querfeld C, Kuzel TM, Kim YH, Porcu P, Duvic M, Musiek A, Rook AH, Mark LA, Pinter-Brown L, Hamid O, Lin B, Bian Y, Boye M, Day JM, Rosen ST. Multicenter phase II trial of enzastaurin in patients with relapsed or refractory advanced cutaneous T-cell lymphoma. Leuk Lymphoma. 2011 Aug;52(8):1474–80. doi: 10.3109/10428194.2011.572265.
  50. Ghobrial IM, Vij R, Siegel D, Badros A, Kaufman J, Raje N, Jakubowiak A, Savona MR, Obreja M, Berdeja JG. A Phase Ib/II Study of Oprozomib in Patients with Advanced Multiple Myeloma and Waldenström Macroglobulinemia. Clin Cancer Res. 2019 Aug 15;25(16):4907–16. doi: 10.1158/1078-0432.CCR-18-3728.
  51. Chiappori A, Bepler G, Barlesi F, Soria JC, Reck M, Bearz A, Barata F, Scagliotti G, Park K, Wagle A, Liepa AM, Zhao YD, Chouaki N, Iscoe N, von Pawel J. Phase II, double-blinded, randomized study of enzastaurin plus pemetrexed as second-line therapy in patients with advanced non-small cell lung cancer. J Thorac Oncol. 2010 Mar;5(3):369–75. doi: 10.1097/JTO.0b013e3181cee24f.
  52. Dreicer R, Garcia J, Hussain M, Rini B, Vogelzang N, Srinivas S, Somer B, Zhao YD, Kania M, Raghavan D. Oral enzastaurin in prostate cancer: a two-cohort phase II trial in patients with PSA progression in the non-metastatic castrate state and following docetaxel-based chemotherapy for castrate metastatic disease. Invest New Drugs. 2011 Dec;29(6):1441–8. doi: 10.1007/s10637-010-9428-0.
  53. Usha L, Sill MW, Darcy KM, Benbrook DM, Hurteau JA, Michelin DP, Mannel RS, Hanjani P, De Geest K, Godwin AK. A Gynecologic Oncology Group phase II trial of the protein kinase C-beta inhibitor, enzastaurin and evaluation of markers with potential predictive and prognostic value in persistent or recurrent epithelial ovarian and primary peritoneal malignancies. Gynecol Oncol. 2011 Jun 1;121(3):455–61. doi: 10.1016/j.ygyno.2011.02.013.
  54. Naylor TL, Tang H, Ratsch BA, Enns A, Loo A, Chen L, Lenz P, Waters NJ, Schuler W, Dörken B, Yao YM, Warmuth M, Lenz G, Stegmeier F. Protein kinase C inhibitor sotrastaurin selectively inhibits the growth of CD79 mutant diffuse large B-cell lymphomas. Cancer Res. 2011 Apr 1;71(7):2643–53. doi: 10.1158/0008-5472.CAN-10-2525.
  55. Fang YH, Joo DJ, Lim BJ, Huh KH, Kim MS, Suh H, Kim YS. The effects of AEB071 (sotrastaurin) with tacrolimus on rat heterotopic cardiac allograft rejection and survival. J Surg Res. 2011 Nov;171(1):e133–7. doi: 10.1016/j.jss.2011.06.039.
  56. Yuan Y, Yangmei Z, Rongrong S, Xiaowu L, Youwei Z, Sun S. Sotrastaurin attenuates the stemness of gastric cancer cells by targeting PKCδ. Biomed Pharmacother. 2019 Sep;117:109165. doi: 10.1016/j.biopha.2019.109165З.
  57. Piperno-Neumann S, Larkin J, Carvajal RD, Luke JJ, Schwartz GK, Hodi FS, Sablin MP, Shoushtari AN, Szpakowski S, Chowdhury NR, Brannon AR, Ramkumar T, de Koning L, Derti A, Emery C, Yerramilli-Rao P, Kapiteijn E. Genomic Profiling of Metastatic Uveal Melanoma and Clinical Results of a Phase I Study of the Protein Kinase C Inhibitor AEB071. Mol Cancer Ther. 2020 Apr;19(4):1031–9. doi: 10.1158/1535-7163.MCT-19-0098.
  58. Skvara H, Dawid M, Kleyn E, Wolff B, Meingassner JG, Knight H, Dumortier T, Kopp T, Fallahi N, Stary G, Burkhart C, Grenet O, Wagner J, Hijazi Y, Morris RE, McGeown C, Rordorf C, Griffiths CE, Stingl G, Jung T. The PKC inhibitor AEB071 may be a therapeutic option for psoriasis. J Clin Invest. 2008 Sep;118(9):3151–9. doi: 10.1172/JCI35636.
  59. Kovarik JM, Steiger JU, Grinyo JM, Rostaing L, Arns W, Dantal J, Proot P, Budde K. Sotrastaurin Renal Transplant Study Group. Pharmacokinetics of sotrastaurin combined with tacrolimus or mycophenolic acid in de novo kidney transplant recipients. Transplantation. 2011 Feb;91(3),317–22. doi: 10.1097/TP.0b013e318203860d.
  60. Matz M, Naik M, Mashreghi MF, Glander P, Neumayer HH, Budde K. Evaluation of the novel protein kinase C inhibitor sotrastaurin as immunosuppressive therapy after renal transplantation. Expert Opin Drug Metab Toxicol. 2011 Jan;7(1):103–13.
  61. Kangussu-Marcolino MM, Ehrenkaufer GM, Chen E, Debnath A, Singh U. Identification of plicamycin, TG02, panobinostat, lestaurtinib, and GDC-0084 as promising compounds for the treatment of central nervous system infections caused by the free-living amebae Naegleria, Acanthamoeba and Balamuthia. Int J Parasitol Drugs Drug Resist. 2019 Dec;11:80–94. doi: 10.1016/j.ijpddr.2019.10.003.
  62. Ramos NR, Mo CC, Karp JE, Hourigan CS. Current Approaches in the Treatment of Relapsed and Refractory Acute Myeloid Leukemia. J Clin Med. 2015 Apr;4(4):665–95. doi: 10.3390/jcm4040665.
  63. Knapper S, Burnett AK, Littlewood T, Kell WJ, Agrawal S, Chopra R, Clark R, Levis MJ, Small D. A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy. Blood. 2006 Nov 15;108(10):3262–70. doi: 10.1182/blood-2006-04-015560.
  64. Sutamtewagul G, Vigil CE. Clinical use of FLT3 inhibitors in acute myeloid leukemia. Onco Targets Ther. 2018 Oct 16;11:7041–52. doi: 10.2147/OTT.S171640.
  65. Hexner E, Roboz G, Hoffman R, Luger S, Mascarenhas J, Carroll M, Clementi R, Bensen-Kennedy D, Moliterno A. Open-label study of oral CEP-701 (lestaurtinib) in patients with polycythaemia vera or essential thrombocythaemia with JAK2-V617F mutation. Br J Haematol. 2014 Jan;164(1):83–93. doi: 10.1111/bjh.12607.
  66. Mascarenhas J, Baer MR, Kessler C, Hexner E, Tremblay D, Price L, Sandy L, Weinberg R, Pahl H, Silverman LR, Goldberg JD, Kosiorek H, Dueck AC, Hoffman R. Phase II trial of Lestaurtinib, a JAK2 inhibitor, in patients with myelofibrosis. Leuk Lymphoma. 2019 May;60(5):1343–5. doi: 10.1080/10428194.2018.1532509.
  67. Festuccia C, Muzi P, Gravina GL, Millimaggi D, Speca S, Dolo V, Ricevuto E, Vicentini C, Bologna M. Tyrosine kinase inhibitor CEP-701 blocks the NTRK1/NGF receptor and limits the invasive capability of prostate cancer cells in vitro. Int J Oncol. 2007 Jan;30(1):193–200.
  68. Collins C, Carducci MA, Eisenberger MA, Isaacs JT, Partin AW, Pili R, Sinibaldi VJ, Walczak JS, Denmeade SR. Preclinical and clinical studies with the multi-kinase inhibitor CEP-701 as treatment for prostate cancer demonstrate the inadequacy of PSA response as a primary endpoint. Cancer Biol Ther. 2007 Sep;6(9):1360–7. doi: 10.4161/cbt.6.9.4541.
  69. Iyer R, Evans AE, Qi X, Ho R, Minturn JE, Zhao H, Balamuth N, Maris JM, Brodeur GM. Lestaurtinib enhances the antitumor efficacy of chemotherapy in murine xenograft models of neuroblastoma. Clin Cancer Res. 2010 Mar 1;16(5):1478–85. doi: 10.1158/1078-0432.CCR-09-1531.
  70. Minturn JE, Evans AE, Villablanca JG, Yanik GA, Park JR, Shusterman S, Groshen S, Hellriegel ET, Bensen-Kennedy D, Matthay KK, Brodeur GM, Maris JM. Phase I trial of lestaurtinib for children with refractory neuroblastoma: a new approaches to neuroblastoma therapy consortium study. Cancer Chemother Pharmacol. 2011 Oct;68(4):1057–65. doi: 10.1007/s00280-011-1581-4.
  71. Volc S, Ghoreschi K. Pathophysiological basis of systemic treatments in psoriasis. J Dtsch Dermatol Ges. 2016 Jun;14(6):557–72. doi: 10.1111/ddg.13050.
  72. Chiu HT, Chen YL, Chen CY, Jin C, Lee MN, Lin YC. Molecular cloning, sequence analysis and functional characterization of the gene cluster for biosynthesis of K-252a and its analogs. Mol Biosyst. 2009 Oct;5(10):1180–91. doi: 10.1039/b905293c.
  73. Gadbois DM, Crissman HA, Tobey RA, Bradbury EM. Multiple kinase arrest points in the G1 phase of nontransformed mammalian cells are absent in transformed cells. Proc Natl Acad Sci USA. 1992 Sep 15;89(18):8626–30. doi: 10.1073/pnas.89.18.8626.
  74. Makarevich AV, Sirotkin AV, Rafay J. Comparison of effects of protein kinase A, mitogen-activated protein kinase, and cyclin-dependent kinase blockers on rabbit ovarian granulosa cell functions. Horm Metab Res. 2010 Dec;42(13):936–43. doi: 10.1055/s-0030-1267226.
  75. Zhu S, Wurdak H, Wang J, Lyssiotis CA, Peters EC, Cho CY, Wu X, Schultz PG. A small molecule primes embryonic stem cells for differentiation. Cell Stem Cell. 2009 May 8;4(5):416–26. doi: 10.1016/j.stem.2009.04.001.
  76. Bouvard C, Lim SM, Ludka J, Yazdani N, Woods AK, Chatterjee AK, Schultz PG, Zhu S. Small molecule selectively suppresses MYC transcription in cancer cells. Proc Natl Acad Sci USA. 2017 Mar 28;114(13):3497–502. doi: 10.1073/pnas.1702663114.
  77. Hudkins RL, Becknell NC, Zulli AL, Underiner TL, Angeles TS, Aimone LD, Albom MS, Chang H, Miknyoczki SJ, Hunter K, Jones-Bolin S, Zhao H, Bacon ER, Mallamo JP, Ator MA, Ruggeri BA. Synthesis and biological profile of the pan-vascular endothelial growth factor receptor/tyrosine kinase with immunoglobulin and epidermal growth factor-like homology domains 2 (VEGF-R/TIE-2) inhibitor 11-(2-methylpropyl)-12,13-dihydro-2-methyl-8-(pyrimidin-2-ylamino)-4H-indazolo[5,4-a]pyrrolo[3,4-c]carbazol-4-one (CEP-11981): a novel oncology therapeutic agent. J Med Chem. 2012 Jan 26;55(2):903–13. doi: 10.1021/jm201449n.
  78. Pili R, Carducci M, Brown P, Hurwitz H. An open-label study to determine the maximum tolerated dose of the multitargeted tyrosine kinase inhibitor CEP-11981 in patients with advanced cancer. Invest New Drugs. 2014 Dec;32(6):1258–68. doi: 10.1007/s10637-014-0147-9.
  79. Higa-Nakamine S, Maeda N, Toku S, Yamamoto H. Involvement of Protein Kinase D1 in Signal Transduction from the Protein Kinase C Pathway to the Tyrosine Kinase Pathway in Response to Gonadotropin-releasing Hormone. J Biol Chem. 2015 Oct 23;290(43):25974–85. doi: 10.1074/jbc.M115.681700.
  80. Koivunen J, Aaltonen V, Koskela S, Lehenkari P, Laato M, Peltonen J. Protein kinase C alpha/beta inhibitor Go6976 promotes formation of cell junctions and inhibits invasion of urinary bladder carcinoma cells. Cancer Res. 2004 Aug 15;64(16):5693–701. doi: 10.1158/0008-5472.CAN-03-3511.
  81. Xiao-Xian Han, Cheng-Bin Cui, Qian-Qun Gu, Wei-Ming Zhu, Hong-Bing Liu, Jing-Yan Gu, Hiroyuki Osada. “ZHD-0501, a novel naturally occurring staurosporine analog from Actinomadura sp. 007.” Tetrahedron letters 46.36(2005):6137–40. doi: 10.1016/j.tetlet.2005.06.154.
  82. Schroeder D., Lam K.S., Mattei J., Hesler G.A. BMY-41950 antitumor antibiotic. U.S. Patent No. 5,073,633. 17 Dec. 1991.
  83. Takahashi I, Saitoh Y, Yoshida M, Sano H, Nakano H, Morimoto M, Tamaoki T. UCN-01 and UCN-02, new selective inhibitors of protein kinase C. II. Purification, physico-chemical properties, structural determination and biological activities. J Antibiot (Tokyo). 1989 Apr;42(4):571–6. doi: 10.7164/antibiotics.42.571.
  84. Ruggeri B, Singh J, Gingrich D, Angeles T, Albom M, Yang S, Chang H, Robinson C, Hunter K, Dobrzanski P, Jones-Bolin S, Pritchard S, Aimone L, Klein-Szanto A, Herbert JM, Bono F, Schaeffer P, Casellas P, Bourie B, Pili R, Isaacs J, Ator M, Hudkins R, Vaught J, Mallamo J, Dionne C. CEP-7055: a novel, orally active pan inhibitor of vascular endothelial growth factor receptor tyrosine kinases with potent antiangiogenic activity and antitumor efficacy in preclinical models. Cancer Res. 2003 Sep 15;63(18):5978–9.
  85. Strock CJ, Park JI, Rosen M, Dionne C, Ruggeri B, Jones-Bolin S, Denmeade SR, Ball DW, Nelkin BD. CEP-701 and CEP-751 inhibit constitutively activated RET tyrosine kinase activity and block medullary thyroid carcinoma cell growth. Cancer Res. 2003 Sep 1;63(17):5559–63.
  86. Nakanishi S, Yamada K, Iwahashi K, Kuroda K, Kase H. KT5926, a potent and selective inhibitor of myosin light chain kinase. Mol Pharmacol. 1990 Apr;37(4):482–8.
  87. Alessi DR. The protein kinase C inhibitors Ro 318220 and GF 109203X are equally potent inhibitors of MAPKAP kinase-1beta (Rsk-2) and p70 S6 kinase. FEBS Lett. 1997 Feb 3;402(2–3):121–3. doi: 10.1016/S0014-5793(96)01510-4.
  88. Melnik SY., Vlasenkova NK, Garaeva LD, Golubeva IS, Goryunova OV, Eremina VA, Markova NP, Miniker TD, Plikhtyak IL, Tikhonova NI, Ektova LV, Yartseva IV. A method for obtaining N-glycosides of indolo[2,3-a]pyrrolo[3,4-c]carbazole-5,7-diones with cytotoxic and antitumor activity. Patent No. №2427585. 10 Dec.2009. Russian
  89. Kiseleva MP, Smirnova ZS, Borisova LM, Kubasova IYu, Ektova LV, Miniker TD, Plikhtiak IL, Medvedeva LA, Eremina VA, Tikhonova NI. Search for new antitumor compounds among n-glycoside indolo[2,3-a]carbazole derivatives. Russian Journal of Oncology. 2015;20(1):33–7. Russian
  90. Golubeva IS, Goryunova OV, Yavorskaya NP. Comparative in vivo studying of potential antineoplastic properties among of amino-acid derivative glycosides of the indolocarbazole (detailed report). Russian Biotherapeutic Journal. 2018;17(2). (Russian) doi: 10.17650/1726-9784-2018-17-2-71-77.
  91. Golubeva IS, Yavorskaya NP, Eremina VA, Tikhonova NI, Miniker TD, Ektova LV, Dmitrieva MV. Antitumor activity of indolocarbazole glycosides. Russian Biotherapeutic Journal. 2016;15(1):23–4. Russian
  92. Ektova LV, Eremina VA, Tikhonova NI, Plikhtyak IL, Medvedeva LA, Yartseva IV, Moiseeva NI, Golubeva IS, Yavorskaya NP, Budko AP, Tarasova OI, Pugacheva RB. Synthesis and cytotoxic activity of N-glycosides of indolo[2,3-a]pyrrolo[3,4-c]carbazole-5,7-diones substituted by a maleimide nitrogen atom. Pharmaceutical Chemistry Journal. 2020;54(5):26–9. DOI: 0023-1134-2020-54-5-26-29. Russian
  93. Kiseleva MP, Borisova LM, Ektova LV, Eremina VA, Tikhonova NI, Dmitrieva MV, Mironova SE, Medvedeva LA. Investigation of antitumor activity of new compounds in a number of indolocarbazole glycoside derivatives. Russian Biotherapeutic Journal. 2018;14, S1:35. Russian
  94. Gulyakin ID, Nikolaeva LL, Dmitrieva MV, Orlova OL, Polozkova AP, Oborotova NA, Ignatieva EV, Dmitricheva NA, Yartseva IV, Shprakh ZS. Preparation and analysis of lyophilized dosage form lhs-1208 by thin chromatography and a spectrophotometery. Development and registration of medicines. 2016;4(17):62–7.
  95. Gulyakin ID., Hashem A, Nikolaeva LL, Dmitrieva MV, Afanasyeva DA, Baryshnikova MA, Oborotova NA, Lantsova AV. The development of new technology of the dosage form for intravenous administration indolocarbazole derivative LHS-1208. Russian Biotherapeutic Journal. 2016;15(2):55–60. doi: 10.17650/1726-9784-2016-15-2-55-60. Russian
  96. Yavorskaya NP, Golubeva IS, Ektova LV, Eremina VA, Tikhonova NI, Miniker TD, Dmitrieva MV. Antitumor activity of indolocarbazole LHS-1269. Russian Biotherapeutic Journal. 2016;15(1):125–6. Russian
  97. Vartanyan A.A., Baryshnikova M.A., Burova O.S., Ektova L.V., Smirnova L.I., Shprach Z.S. The blocker of vasculogenic mimicry restores the sensitivity of resistant melanoma cells to DNA-damaging agents. Russian Biotherapeutic Journal. 2016;15(1):19–20. (Russian)
  98. Lugen B, Dmitrieva MV, Orlova OL, Krasnyuk II, Krasnyuk Jr II, Bokov DO, Stepanova OI, Belyatskaya AV. Development of the composition of a liposomal dosage form of a hydrophobic derivative of indolocarbazole // Development and registration of medicines. 2020;9(3):21–6. doi: 10.33380/2305-2066-2020-9-3-21-26. Russian
  99. Sherer C, Snape TJ. Heterocyclic scaffolds as promising anticancer agents against tumours of the central nervous system: Exploring the scope of indole and carbazole derivatives. Eur J Med Chem. 2015 Jun 5;97:552–60. doi: 10.1016/j.ejmech.2014.11.007.
  100. Rewcastle GW. Becatecarin (Helsinn Healthcare). IDrugs. 2005 Oct;8(10):838–47.
  101. Robey RW, Obrzut T, Shukla S, Polgar O, Macalou S, Bahr JC, Di Pietro A, Ambudkar SV, Bates SE. Becatecarin (rebeccamycin analog, NSC 655649) is a transport substrate and induces expression of the ATP-binding cassette transporter, ABCG2, in lung carcinoma cells. Cancer Chemother Pharmacol. 2009 Aug;64(3):575–83. doi: 10.1007/s00280-008-0908-2.
  102. Borthakur G, Alvarado Y, Ravandi-Kashani F, Cortes J, Estrov Z, Faderl S, Ivy P, Bueso-Ramos C, Nebiyou Bekele B, Giles F. Phase 1 study of XL119, a rebeccamycin analog, in patients with refractory hematologic malignancies. Cancer. 2008 Jul 15;113(2):360–6. doi: 10.1002/cncr.23559.
  103. Pommerehne K, Walisko J, Ebersbach A. Krull R. Phase I trial of combination becatecarin and oxaliplatin in patients with advanced solid tumors. Journal of Clinical Oncology. 2007 June 20; 25(18_suppl), 2561–2561. doi: 10.1007/s00253-019-09741-y.
  104. Schwandt A, Mekhail T, Halmos B, O’Brien T, Ma PC, Fu P, Ivy P, Dowlati A. Phase-II trial of rebeccamycin analog, a dual topoisomerase-I and -II inhibitor, in relapsed “sensitive” small cell lung cancer. J Thorac Oncol. 2012 Apr;7(4):751–4. doi: 10.1097/JTO.0b013e31824abca2.
  105. Qu X, Chaires JB, Ohkubo M, Yoshinari T, Nishimura S, Bailly C. A DNA binding indolocarbazole disaccharide derivative remains highly cytotoxic without inhibiting topoisomerase I. Anticancer Drug Des. 1999 Oct;14(5):433–42.
  106. Saijo N. Preclinical and clinical trials of topoisomerase inhibitors. Ann N Y Acad Sci. 2000;922:92–9. doi: 10.1111/j.1749-6632.2000.tb07028.x.
  107. Saif MW, Diasio RB. Edotecarin: a novel topoisomerase I inhibitor. Clin Colorectal Cancer. 2005 May;5(1):27–36. doi: 10.3816/ccc.2005.n.014.
  108. Yamada Y, Tamura T, Yamamoto N, Shimoyama T, Ueda Y, Murakami H, Kusaba H, Kamiya Y, Saka H, Tanigawara Y, McGovren JP, Natsumeda Y. Phase I and pharmacokinetic study of edotecarin, a novel topoisomerase I inhibitor, administered once every 3 weeks in patients with solid tumors. Cancer Chemother Pharmacol. 2006 Aug;58(2):173–82. doi: 10.1007/s00280-005-0149-6.
  109. Saif MW, Sellers S, Diasio RB, Douillard JY. A phase I dose-escalation study of edotecarin (J-107088) combined with infusional 5-fluorouracil and leucovorin in patients with advanced/metastatic solid tumors. Anticancer Drugs. 2010 Aug;21(7):716–23. doi: 10.1097/CAD.0b013e32833cb658.
  110. Ciomei M, Croci V, Ciavolella A, Ballinari D, Pesenti E. Antitumor efficacy of edotecarin as a single agent and in combination with chemotherapy agents in a xenograft model. Clin Cancer Res. 2006 May 1;12(9):2856–61. doi: 10.1158/1078-0432.CCR-05-1859.
  111. Hurwitz HI, Cohen RB, McGovren JP, Hirawat S, Petros WP, Natsumeda Y, Yoshinari T. A phase I study of the safety and pharmacokinetics of edotecarin (J-107088), a novel topoisomerase I inhibitor, in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2007 Jan;59(1):139–47. doi: 10.1007/s00280-006-0267-9.
  112. Ciomei M, Croci V, Stellari F, Amboldi N, Giavarini R, Pesenti E. Antitumor activity of edotecarin in breast carcinoma models. Cancer Chemother Pharmacol. 2007 Jul;60(2):229–35. doi: 10.1007/s00280-006-0365-8.
  113. Vrdoljak E, Boban M, Saratlija-Novaković Z, Jović J. Long-lasting partial regression of glioblastoma multiforme achieved by edotecarin: case report. Croat Med J. 2006 Apr;47(2):305–9.
  114. Yin D, Toler S, Guo F, Duncan B, Sharma A. Pharmacokinetics (PK) of edotecarin (J-107088), a topoisomerase I inhibitor, in patients with metastatic breast cancer (mBC) or glioblastoma multiforme (GBM). Journal of Clinical Oncology. 2005 June 01;23(16_suppl), 2073–2073. doi: 10.1200/jco.2005.23.16_suppl.2073.
  115. Carvajal RD, Ilson DH, Noy A. Possible role of edotecarin, a novel topoisomerase I inhibitor, in therapy-related myelodysplastic syndrome. Leuk Lymphoma. 2007 Jan;48(1):192–4. doi: 10.1080/10428190600968350.
  116. Lam KS, Schroeder DR, Veitch JM, Matson JA, Forenza S. Isolation of a bromo analog of rebeccamycin from Saccharothrix aerocolonigenes. J Antibiot (Tokyo). 1991 Sep;44(9):934–9. doi: 10.7164/antibiotics.44.934.
  117. Matson JA, Claridge C, Bush JA, Titus J, Bradner WT, Doyle TW, Horan AC, Patel M. AT2433-A1, AT2433-A2, AT2433-B1, and AT2433-B2 novel antitumor antibiotic compounds produced by Actinomadura melliaura. Taxonomy, fermentation, isolation and biological properties. J Antibiot (Tokyo). 1989 Nov;42(11):1547–55. doi: 10.7164/antibiotics.42.1547.

Supplementary files

Supplementary Files
Action
1. JATS XML
2. Figure 1 – Structural formula of indole

Download (12KB)
3. Figure 2 – Structural formula of carbazole

Download (16KB)
4. Figure 3 – Structural formulas of indolocarbazole isomers

Download (87KB)
5. Figure 4 – Structural formulas of indolocarbazole derivatives

Download (120KB)
6. Figure 5 – Structural formula of Go 6976

Download (44KB)
7. Figure 6 – Structural formula of sotrastaurine

Download (52KB)
8. Figure 7 – Structural formula of midostaurine

Download (47KB)
9. Figure 8 – Structural formula of enzastaurin

Download (38KB)
10. Figure 9 – Structural formula of Lestaurtinib

Download (52KB)
11. Figure 10 – Structural formula of K-252a

Download (52KB)
12. Figure 11 – Structural formula of CEP-11981

Download (40KB)
13. Figure 12 – Structural formula of becatecarin

Download (49KB)
14. Figure 13 – Structural formula of NB-506

Download (48KB)
15. Figure 14 – Structural formula of edothecarin

Download (57KB)

Copyright (c) 2021 Kolpaksidi I.P., Dmitrieva M.V., Yarosh I.V., Krasnyuk I.I.

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.

СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: ПИ № ФС 77 - 67428 от 13.10.2016. 

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies