Open access peer-reviewed chapter

Development of Benzimidazole Compounds for Cancer Therapy

Written By

Puranik Purushottamachar, Senthilmurugan Ramalingam and Vincent C.O. Njar

Submitted: 27 November 2018 Reviewed: 06 May 2019 Published: 10 June 2019

DOI: 10.5772/intechopen.86691

From the Edited Volume

Chemistry and Applications of Benzimidazole and its Derivatives

Edited by Maria Marinescu

Chapter metrics overview

1,299 Chapter Downloads

View Full Metrics

Abstract

A fact that is largely unknown in the lay press and even the scientific community is that today cancer kills more people worldwide than tuberculosis (TB), malaria, and human immunodeficiency virus (HIV) combined. Benzimidazole is a heterocyclic aromatic organic compound considered to be a useful pharmacophore in a variety of impactful drugs. The purpose of this review is to highlight the benzimidazole-containing agents that are currently in clinical use or in clinical development as anticancer drugs. It is hoped that this review would function as comprehensive working reference of research accomplishment in the field of discovery and development of benzimidazole-based anticancer drugs.

Keywords

  • benzimidazole derivatives
  • privileged pharmacophore
  • anticancer drugs/agents

1. Introduction

Benzimidazole (1) (Figure 1) is used as the major scaffold or as a moiety on other scaffolds for the development of a variety of drugs [1, 2, 3, 4]. The wide range of pharmacological activities of benzimidazole-containing agents are attributed to the unique fused benzene and imidazole rings, which can interact in a noncovalent manner with a range of biological targets due to the presence of an electron-rich aromatic system and the two hetero-nitrogen atoms [5, 6]. Because of the ability of benzimidazole derivative to interact with a variety of unrelated molecular targets, the term “privileged substructure/moiety” is ascribed to this unique azole agent. It is believed that the interest in benzimidazole chemistry and as a scaffold/moiety in the discovery and development of drugs arose from the discovery of the rare and most prominent benzimidazole compound in nature, N-ribosyl-dimethylbenzimidazole (2) (Figure 1), which serves as an axial ligand for cobalt in vitamin B12 [7].

Figure 1.

Chemical structures of benzimidazole (1) and N-ribosyl-dimethylbenzimidazole (2).

Although several benzimidazole derivatives have been approved for clinical use, including antiparasitic, antiulcer, antihypertensive, antihistaminic, and antiemetic drugs [1, 2, 3, 4], only one anticancer drug, bendamustine (3) (Figure 2), has received FDA approval [8, 9, 10]. Two prominent benzimidazole agents, selumetinib (4) (Figure 2) [1, 6] and galeterone (5) (Figure 2) [11], that advanced to phase III clinical trials, but are yet to be approved as anticancer drugs, will also be discussed.

Figure 2.

Chemical structures of bendamustine (3), selumetinib (4), and galeterone (5).

Advertisement

2. Benzimidazole agents in the clinic and in clinical development

2.1 Bendamustine (3)

Bendamustine (3) (Figure 2) was discovered in a structure-activity relationship (SAR) campaign directed to obtain more effective and safer water-soluble analogs of chlorambucil (6) (Figure 3), a nitrogen mustard, which is used clinically against chronic lymphatic leukemia, lymphomas, and advanced ovarian and breast carcinomas [12]. The strategy was replacement of the benzene ring in compound 6 with purine-like N-methylbenzimidazole moiety in the hope of obtaining an anticancer agent with antimetabolite and DNA-alkylating activities. Although bendamustine was first synthesized in the early 1960s [13], it was approved under the trade name Treanda® by the US Food and Drug Administration (FDA) in 2008 for the treatment of chronic lymphocytic leukemia, multiple myeloma, and non-Hodgkin’s lymphoma [10, 14, 15, 16].

Figure 3.

Replacement of aromatic benzene ring of chlorambucil (6) to produce bendamustine (3).

2.1.1 Chemistry

Bendamustine (3) 4-{5-[bis(2-chloroethyl)amino]-1-methylbenzimidazol-2-yl} butanoic acid was first synthesized via eight synthetic steps with an overall yield of 12% [13, 17]. However, Chen and colleagues have developed a new, more efficient, and cost-effective route focused on the use of sustainable chemistry for the synthesis of bendamustine hydrochloride, with the overall yield improved from 12 to 45% as outlined in Figure 4 [18]. This new synthesis is currently used for the commercial production of 3.

Figure 4.

New optimized synthesis of bendamustine hydrochloride (3).

2.1.2 Summary of bendamustine’s preclinical and clinical pharmacology

Even though bendamustine is an alkylating agent, due to its ability to cause intra-strand and inter-strand cross-links between DNA bases, it has been reported that the DNA breaks induced by bendamustine are more extensive/durable than those induced by other alkylating agents, such as chlorambucil, cyclophosphamide, or carmustine [19, 20, 21]. In addition, the drug was shown to exhibit partial cross-resistance to other alkylating agents. These data suggested that bendamustine may possess additional mechanisms of action. Indeed, a comprehensive study by Leoni and colleagues clearly demonstrated that bendamustine exhibits a distinct pattern of activities unrelated to other alkylating drugs. Using a variety of lymphoid cancer cell lines, the study concluded that mechanisms of action include induction of mitotic catastrophe, inhibition of mitotic checkpoints, and activation of DNA-damage stress response and apoptosis. Compared to other alkylating agents, bendamustine was shown to activate the base excision DNA repair pathway rather than the alkyl transferase DNA repair mechanism [20].

Although bendamustine is approved for the treatment of a variety of lymphoid cancers, its activity has also been reported in several cancers, including cancers of small cell lung, breast, hepatic, bile duct, and head and neck. The studies by Chow and colleagues using leukemic cell lines in vitro or ex vivo cells from patients with leukemic progression to clarify interactions between bendamustine and other chemotherapeutic drugs unraveled synergy with cladribine, in contrast to observed antagonism with mitoxantrone or doxorubicin. The observation of synergism between bendamustine and rituximab (an anti-CD20 antibody) in in vitro CD20-positive DOHH-2 and WSU-NHL cell lines and ex vivo B-cell chronic lymphocytic leukemia (CLL) cells [22] and in mice with Daudi xenografts [23] provided the impetus for clinical trials combining these two drugs [24, 25].

Based on the discussion above, it is obvious that bendamustine is an “old drug rediscovered.” For over 30 years, bendamustine was used in Eastern Germany as monotherapy for several cancers, including breast cancer, chronic lymphocytic leukemia (CLL), Hodgkin’s lymphoma, non-Hodgkin’s lymphoma (NHL), and multiple myeloma (MM) [26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37]. However, following the reunification of Germany, other countries initiated clinical trials of bendamustine as a single agent and in combination with other drugs. Bendamustine has achieved worldwide regulatory approval and is a standard-of-care drug for the treatment on many lymphoid malignancies. Several articles that provide comprehensive reviews of the discovery and development of this unique drug are available [8, 9, 10].

2.2 Selumetinib (4)

Selumetinib (4) (AZD6244: ARRY-142866) is an orally available, potent, selective inhibitor of mitogen-activated protein kinase (MAPK)/extracellular signal-related kinase (ERK) kinases 1 and 2 (MEK1 and MEK2) [6]. This agent has been extensively studied in many preclinical and clinical studies in several tumor types with mixed results. Here, we will summarize the chemistry, preclinical studies, and clinical studies.

2.2.1 Chemistry

Selumetinib (6-(4-bromo-2-chloroanilino)-7-fluoro-N-(2-hydroxyethoxy)-3-methyl benzimidazole-5-carboxamide) (4) is a diarylamine hydroxamide, containing mono-methylated benzimidazole subunit [38, 39]. It is a second-generation, orally active small molecule that acts as a selective and ATP-uncompetitive inhibitor of MEK1 and MEK2, binding to the allosteric binding site [38, 39]. The synthesis of selumetinib is yet to be reported in the literature.

2.2.2 Summary of selumetinib’s preclinical and clinical pharmacology

Selumetinib inhibits the enzymatic activity of purified constitutively active MEK1 with a half maximal inhibitory concentration (IC50) of 14 nM and was shown to be highly selective for inhibition of these targets compared to other related kinases [39]. Using several human cancer cell lines such as NSCLC, melanoma, and pancreatic and colorectal cell lines, it was shown that selumetinib was a potent antiproliferative agent. Analysis of the data revealed that cell lines with mutant BRAF and RAS were sensitive to selumetinib [40]. Selumetinib had little effect on the growth of Malme-3, the control cell line to the melanoma. Additional studies suggested that the growth inhibitory effects of selumetinib was not due to wide-ranging cytotoxicity [39], and it was also established that selumetinib effectively inhibits the phosphorylation of ERK 1 and ERK 2, which are substrates of MEK1 and MEK2 in the MAP kinase pathway. This mechanism of action was confirmed in tumor xenografts. Additionally, increased markers of apoptosis such as cleaved caspase 3 and decreased cell proliferation were seen in response to treatment with selumetinib in the xenograft models [39, 40].

The promising preclinical in vitro and in vivo data provided the rationale for multiple clinical trials in cancers with activated Raf-MEK-ERK signaling. In preparation for clinical evaluation of selumetinib, it was originally developed as a free base and administered as a liquid suspension, but subsequently a capsule formulation of the hydrogen sulfate salt was found to be more suitable for further development [38]. Several phase I and II clinical trials conducted against solid tumors to test the impact of selumetinib as a monotherapy were unsuccessful [41, 42, 43, 44]. This led to the conduct of several clinical trials with selumetinib in combination with other cancer drugs. A notable trial was the randomized phase II study of selumetinib in combination with docetaxel, as a second-line treatment for patients with KRAS-mutant advanced NSCLC which showed very promising results [45]. The median progression-free survival was 5.3 months with selumetinib + docetaxel and 2.1 months with docetaxel alone. The objective response rate was 37% for selumetinib + docetaxel vs. 0% for docetaxel alone (p < 0.001), and the median overall survival was 9.4 months for selumetinib + docetaxel vs. 5.2 months for docetaxel alone (HR for death, 0.80 [80% CI, 0.56–1.14]; one-sided p = 0.21). Unfortunately, in a multinational 510 randomized patients with previously treated advanced KRAS-mutant NSCLC trial, the combination of selumetinib with docetaxel did not improve progression-free survival compared with docetaxel alone [46]. Clearly, addition clinical studies are required to realize the potential impact of selumetinib alone and in combination with other drugs for the treatment of a variety of cancers [47, 48].

2.3 Galeterone (5)

Galeterone (also called VN/124-1 or TOK-001) is an orally available anticancer agent. It was rationally designed as an inhibitor of androgen biosynthesis via inhibition of 17α-hydroxylase/17,20-lyase (CYP17), the key enzyme which catalyzes the biosynthesis of androgens from the progestins. Through extensive and rigorous preclinical studies, galeterone was shown to modulate two other targets in the androgen/androgen receptor (AR) signaling pathway [11] and shown to inhibit the eukaryotic initiation factor 4E (eIF4E) protein translational machinery [49]. Galeterone advanced successfully through phases I and II clinical trials in prostate cancer patients but was unsuccessful in the pivotal phase III clinical trial in men with castration-resistant prostate cancer (CRPC), harboring AR splice variants (e.g., AR-V7). We present a summary of the chemistry, preclinical studies, and clinical studies [50].

2.3.1 Chemistry

Galeterone, 3β-hydroxy-17-(1H-benzimidazole-1-yl)androsta-5,16-diene (5), is one of a series of novel ∆16-17-azolyl steroid, which, unlike previously known 17-heteroaryl steroids, the azole moiety is attached to the steroid nucleus at C-17 via a nitrogen of the azole. The synthesis of galeterone from commercially available 3β-acetoxyandrosta-5-en-17-one (12) is presented in Figure 5 [11, 51], and a facile and large-scale preparation (commercial process) of the compound has been developed but is yet to appear in the literature.

Figure 5.

Synthesis of galeterone.

2.3.2 Summary of galeterone’s preclinical and clinical pharmacology

Using intact CYP17 expressing Escherichia coli, galeterone was shown to be a potent inhibitor of the enzyme with an IC50 value of 300 nM and was shown to be more potent than abiraterone (IC50 value of 800 nM) [52]. Additional studies by our group revealed that galeterone could disrupt androgen signaling through multiple targets [51, 52, 53, 54].

We strongly believe that the increased efficacy of galeterone in several prostate cancer models both in vitro and in vivo is due to its ability to downregulate the AR and block androgen binding to AR. Using well-established AR-competitive binding assays (against the synthetic androgen [3H]R1881), galeterone was equipotent to Casodex in LNCaP cells but had a slightly higher affinity for the wild-type receptor in PC3-AR cells. In transcriptional activation assays (utilizing a luciferase reporter), galeterone was shown to be a pure AR antagonist of the wild-type AR and the T877A mutation found in LNCaP cells [53]. In prostate cancer cell lines, galeterone inhibited the growth of CRPCs, which had increased AR and were no longer sensitive to Casodex [53] and was also shown to inhibit the growth of AR-negative prostate cancer cells [54]. In addition, galeterone demonstrated superior synergy for growth inhibition in combination with everolimus or gefitinib compared with Casodex [55].

Recent in vitro studies have shown additional activities of galeterone, including proteasomal degradation of AR and its splice variants [56, 57] and inhibition of the eukaryotic initiation factor 4E (eIF4E) protein translational machinery via induction of proteasomal degradation of mitogen-activated protein kinase-interacting kinases 1 and 2 (Mnk1 and Mnk2) [58, 59].

Because of the short half-life (t½ = ∼40 min) in mice, galeterone was administered twice daily in our antitumor efficacy studies. Galeterone (0.13 mmol/kg twice daily) caused a 93.8% reduction (p = 0.00065) in the mean final LAPC-4 xenograft volume compared with controls, and this efficacy was significantly more effective than castration or our most potent CYP17 inhibitor, VN/85-1 [51]. In another antitumor efficacy study, treatment of galeterone (0.13 mmol twice daily) was very effective in preventing the formation of LAPC4 tumors (6.94 vs. 2410.28 mm3 in the control group). Galeterone (0.13 mmol/kg twice daily) and VN/124-1 (0.13 mmol/kg twice daily) + castration induced regression of LAPC4 tumor xenografts by 26.55 and 60.67%, respectively [53]. Using castration-resistant prostate cancer (CRPC) HP-LNCaP tumor xenografts, we showed that galeterone + everolimus (m-TORC1 inhibitor) acted in concert to reduce tumor growth [60]. Utilizing the androgen-dependent LAPC-4 prostate cancer xenograft model, we have shown galeterone is more efficacious than the blockbuster prostate cancer drug abiraterone (Zytiga®) [61]. We also reported that galeterone potently inhibits the growth of CRPC CWR22Rv1 tumor xenografts [56].

Based on these impressive preclinical data, galeterone was licensed by the University of Maryland, Baltimore, to Tokai Pharmaceuticals, Inc., who initiated Androgen Receptor Modulation Optimized for Response 1 (ARMOR1) phase 1/phase 2 trials in castrate-resistant prostate cancer patients on November 5, 2009 [11]. The ARMOR phase I and phase II studies conducted with galeterone demonstrated that galeterone is well tolerated with promising clinical activity in patients with CRCP [62, 63]. To determine whether galeterone has clinical activity in patients with C-terminal loss of the androgen receptor, circulating tumor cells were retrospectively tested for C-terminal loss. Of the seven patients identified, six had PSA50 responses. These promising phases I and II studies enabled the selection of galeterone 2500 mg/day dose for the pivotal phase III trial (ARMOR3-SV, NCT02438007). The retrospective data of patients with C-terminal loss of the androgen receptor supported the design of ARMOR3-SV pivotal trial in which patients with AR-V7 were randomized to receive either galeterone or enzalutamide. Regrettably, the trial was discontinued following review by the independent Data Monitoring Committee, though no safety concerns were cited regarding this recommendation [50]. Gratifyingly, Educational and Scientific LLC (ESL), Baltimore, announced (December 17, 2018) that the University of Maryland, Baltimore (UMB), has granted ESL an exclusive license for the development of galeterone for the treatment of patients with CRPC. We eagerly await the initiation of a new phase III clinical study of galeterone in men with prostate cancer.

Advertisement

3. Concluding remarks

Despite the enormous literature on the synthesis and preclinical evaluation of numerous benzimidazole-containing compounds, it is unclear why very few of this class of compounds have entered clinical trials for evaluation as potential anticancer drugs. Given the fact that many benzimidazole-containing drugs have achieved blockbuster status for other diseases, it may be reasonable to suggest that the researchers interested in the development of benzimidazole-contained anticancer drugs should carefully study the process that have resulted in successful non-cancer benzimidazole drugs. We hope that this review will stimulate research activities that would eventually produce new anticancer benzimidazole drugs.

Advertisement

Acknowledgments

This work was supported in part by a grant from the National Institutes of Health (NIH) and the National Cancer Institute (NCI) (R01CA224696) to VCON.

Advertisement

Conflict of interest

VCON is the lead inventor of galeterone; the patents and technologies thereof are owned by the University of Maryland, Baltimore. The other authors declare no potential conflict of interest.

Advertisement

Note added in proof

During the review of this manuscript, it was reported that an analog of Selumetinib called Binimetinib (Mektovi) in combination with a BRAF inhibitor (Encorafenib, Braftovi) was approved by US Food and Drug Administration (FDA) for the treatment of unresectable or metastatic melanoma with BRAF mutations [64].

Advertisement

List of abbreviations

AME

apparent mineralocorticoid excess

AR

androgen receptor

ARMOR

androgen receptor modulation optimized for response

AR-V7

a type of androgen receptor splice variant

BRAF

a human gene that encodes a protein called B-Raf

CI

confidence interval

CLL

chronic lymphocytic leukemia

CRPC

castration-resistant prostate cancer

CWR22Rv1

a type of AR/AR-splice variants positive human prostate cancer cell line

CYP17

cytochrome P450 17α-hydroxylase/17,20-lyase

DNA

deoxyribonucleic acid

eIF4E

eukaryotic initiation factor 4E

ERK1 and ERK2

extracellular signal-regulated kinases 1 and 2

ESL

Educational and Scientific LLC

FDA

Food and Drug Administration

HIV

human immunodeficiency virus

HR

hazard ratio

IC50

is the concentration of inhibitor required to inhibit the enzyme activity by 50%

KRAS

Kirsten retrovirus-associated DNA sequences

LAPC-4

a type of AR-positive human prostate cancer cell line

LNCaP

a type of AR-positive human prostate cancer cell line

MAPK

mitogen-activated protein kinase

MEK1 and MEK2

MAPK/ERK kinase

MM

multiple myeloma

Mnk1 and Mnk2

mitogen-activated protein kinase-interacting kinases 1 and 2

NHL

non-Hodgkin’s lymphoma

NSCLC

non-small cell lung carcinoma

PC3-AR

a type of AR-negative prostate cancer cell line transfected with AR

PSA

prostate-specific antigen

RAF

rapidly accelerated fibrosarcoma

RAS

retrovirus-associated DNA sequences

SAR

structure-activity relationship

TB

tuberculosis

TOK-001

another code name of galeterone

UMB

University of Maryland, Baltimore

VN/85-1

code name for a CYP17 inhibitor/AR antagonist/AR degrader

VN/124-1

original code name of galeterone

References

  1. 1. Gaba M, Mohan C. Development of drugs based on imidazole and benzimidazole bioactive heterocycles: Recent advances and future directions. Medicinal Chemistry Research. 2016;25:173-210
  2. 2. Keri RS, Hiremathad A, Budagumpi S, Nagaraja BM. Comprehensive review in current developments of benzimidazole-based medicinal chemistry. Chemical Biology & Drug Design. 2015;86:19-65. DOI: 10.1111/cbdd.12462
  3. 3. Shrivastava N, Naim MJ, Alam MJ, Nawaz F, Ahmed S, Alam O. Benzimidazole scaffold as anticancer agent: Synthetic approaches and structure-activity relationship. Archiv der Pharmazie. 2017;350:1-80. e1700040. DOI: 10.1002/ardp.201700040
  4. 4. Yadav G, Ganguly S. Structure activity relationship (SAR) study of benzimidazole scaffold for different biological activities: A mini-review. European Journal of Medicinal Chemistry. 2015;97:419-443. DOI: 10.1016/j.ejmech.2014.11.053
  5. 5. DeSimone RW, Currie KS, Mitchell SA, Darrow JW, Pippin DA. Privileged structures: Applications in drug discovery. Combinatorial Chemistry & High Throughput Screening. 2004;7:473-494
  6. 6. Gaba M, Singh S, Mohan C. Benzimidazole: An emerging scaffold for analgesic and anti-inflammatory agents. European Journal of Medicinal Chemistry. 2014;76:494-505. DOI: 10.1016/j.ejmech.2014.01.030
  7. 7. Barker HA, Smyth RD, Weissbach H, Toohey JI, Ladd JN, Volcani BE. Isolation and properties of crystalline cobamide coenzymes containing benzimidazole or 5,6-dimethylbenzimidazole. The Journal of Biological Chemistry. 1960;235:480-488
  8. 8. Cheson BD, Brugger W, Damaj G, Dreyling M, Kahl B, Kimby E, et al. Optimal use of bendamustine in hematologic disorders: Treatment recommendations from an international consensus panel—An update. Leukemia & Lymphoma. 2016;57:766-782. DOI: 10.3109/10428194.2015.1099647
  9. 9. Cheson BD, Rummel MJ. Bendamustine: Rebirth of an old drug. Journal of Clinical Oncology. 2009;27:1492-1501. DOI: 10.1200/JCO.2008.18.7252
  10. 10. Tageja N, Nagi J. Bendamustine: Something old, something new. Cancer Chemotherapy and Pharmacology. 2010;66:413-423. DOI: 10.1007/s00280-010-1317-x
  11. 11. Njar VC, Brodie AM. Discovery and development of Galeterone (TOK-001 or VN/124-1) for the treatment of all stages of prostate cancer. Journal of Medicinal Chemistry. 2015;58:2077-2087. DOI: 10.1021/jm501239f
  12. 12. Nicolle A, Proctor SJ, Summerfield GP. High dose chlorambucil in the treatment of lymphoid malignancies. Leukemia & Lymphoma. 2004;45:271-275
  13. 13. Ozegowski W, Krebs D. IMET 3393, (−[1-methyl-5-bis-(−chloroethyl)-amino-benzimidazolyl-(2)]-butyric) acid hydrochloride, a new cytostatic agent from among the series of benzimidazole mustard compounds. Zentralblatt für die Pharmazie. 1971;110:1013-1019
  14. 14. Knauf WU, Lissichkov T, Aldaoud A, Liberati A, Loscertales J, Herbrecht R, et al. Phase III randomized study of bendamustine compared with chlorambucil in previously untreated patients with chronic lymphocytic leukemia. Journal of Clinical Oncology. 2009;27:4378-4384. DOI: 10.1200/JCO.2008.20.8389
  15. 15. Rasschaert M, Schrijvers D, Van den Brande J, Dyck J, Bosmans J, Merkle K, et al. A phase I study of bendamustine hydrochloride administered once every 3 weeks in patients with solid tumors. Anti-Cancer Drugs. 2007;18:587-595. DOI: 10.1097/CAD.0b013e3280149eb1
  16. 16. Rasschaert M, Schrijvers D, Van den Brande J, Dyck J, Bosmans J, Merkle K, et al. A phase I study of bendamustine hydrochloride administered day 1+2 every 3 weeks in patients with solid tumours. British Journal of Cancer. 2007;96:1692-1698. DOI: 10.1038/sj.bjc.6603776
  17. 17. Werner W, Letsch G, Ihn W, Sohr R, Preiss R. Synthesis of a potential metabolite of the carcinostatic bendamustin (Cytostasen). Pharmazie. 1991;46:113-114
  18. 18. Chen J, Przyuski K, Roemmele R, Bakale RP. Discovery of a novel, efficient, and scalable route to bendamustine hydrochloride: The API in Treanda. Organic Process Research and Development. 2011;15:1063-1072
  19. 19. Hartmann M, Zimmer C. Investigation of cross-link formation in DNA by the alkylating cytostatic IMET 3106, 3393 and 3943. Biochimica et Biophysica Acta. 1972;287:386-389
  20. 20. Leoni LM, Bailey B, Reifert J, Bendall HH, Zeller RW, Corbeil J, et al. Bendamustine (Treanda) displays a distinct pattern of cytotoxicity and unique mechanistic features compared with other alkylating agents. Clinical Cancer Research. 2008;14:309-317. DOI: 10.1158/1078-0432.CCR-07-1061
  21. 21. Strumberg D, Harstrick A, Doll K, Hoffmann B, Seeber S. Bendamustine hydrochloride activity against doxorubicin-resistant human breast carcinoma cell lines. Anti-Cancer Drugs. 1996;7:415-421
  22. 22. Chow KU, Sommerlad WD, Boehrer S, Schneider B, Seipelt G, Rummel MJ, et al. Anti-CD20 antibody (IDEC-C2B8, rituximab) enhances efficacy of cytotoxic drugs on neoplastic lymphocytes in vitro: Role of cytokines, complement, and caspases. Haematologica. 2002;87:33-43
  23. 23. Kanekal S, Crain B., Elliott G. SDX-105 (Trenada) enhances the tumor inhibitory effect of rituximab in Daudi lymphoma xenografts. Blood 2004, 104:229b, (abstr 4580)
  24. 24. Robinson KS, Williams ME, van der Jagt RH, Cohen P, Herst JA, Tulpule A, et al. Phase II multicenter study of bendamustine plus rituximab in patients with relapsed indolent B-cell and mantle cell non-Hodgkin’s lymphoma. Journal of Clinical Oncology. 2008;26:4473-4479. DOI: 10.1200/JCO.2008.17.0001
  25. 25. Rummel MJ, Al-Batran SE, Kim SZ, Welslau M, Hecker R, Kofahl-Krause D, et al. Bendamustine plus rituximab is effective and has a favorable toxicity profile in the treatment of mantle cell and low-grade non-Hodgkin’s lymphoma. Journal of Clinical Oncology. 2005;23:3383-3389. DOI: 10.1200/JCO.2005.08.100
  26. 26. Bottke D, Bathe K, Wiegel T, Hinkelbein W. Phase I trial of radiochemotherapy with bendamustine in patients with recurrent squamous cell carcinoma of the head and neck. Strahlentherapie und Onkologie. 2007;183:128-132. DOI: 10.1007/s00066-007-1597-1
  27. 27. Eichbaum MH, Schuetz F, Khbeis T, Lauschner I, Foerster F, Sohn C, et al. Weekly administration of bendamustine as salvage therapy in metastatic breast cancer: Final results of a phase II study. Anti-Cancer Drugs. 2007;18:963-968. DOI: 10.1097/CAD.0b013e328165d11a
  28. 28. Hartmann JT, Mayer F, Schleicher J, Horger M, Huober J, Meisinger I, et al. Bendamustine hydrochloride in patients with refractory soft tissue sarcoma: A noncomparative multicenter phase II study of the German sarcoma group (AIO-001). Cancer. 2007;110:861-866. DOI: 10.1002/cncr.22846
  29. 29. Kollmannsberger C, Gerl A, Schleucher N, Beyer J, Kuczyk M, Rick O, et al. Phase II study of bendamustine in patients with relapsed or cisplatin-refractory germ cell cancer. Anti-Cancer Drugs. 2000;11:535-539
  30. 30. Koster W, Heider A, Niederle N, Wilke H, Stamatis G, Fischer JR, et al. Phase II trial with carboplatin and bendamustine in patients with extensive stage small-cell lung cancer. Journal of Thoracic Oncology. 2007;2:312-316. DOI: 10.1097/01.JTO.0000263714.46449.4c
  31. 31. Koster W, Stamatis G, Heider A, Avramidis K, Wilke H, Koch JA, et al. Carboplatin in combination with bendamustine in previously untreated patients with extensive-stage small cell lung cancer (SCLC). Clinical Drug Investigation. 2004;24:611-618. DOI: 10.2165/00044011-200424100-00007
  32. 32. Reichmann U, Bokemeyer C, Wallwiener D, Bamberg M, Huober J. Salvage chemotherapy for metastatic breast cancer: Results of a phase II study with bendamustine. Annals of Oncology. 2007;18:1981-1984. DOI: 10.1093/annonc/mdm378
  33. 33. Schmidt-Hieber M, Schmittel A, Thiel E, Keilholz U. A phase II study of bendamustine chemotherapy as second-line treatment in metastatic uveal melanoma. Melanoma Research. 2004;14:439-442
  34. 34. Schmittel A, Knodler M, Hortig P, Schulze K, Thiel E, Keilholz U. Phase II trial of second-line bendamustine chemotherapy in relapsed small cell lung cancer patients. Lung Cancer. 2007;55:109-113. DOI: 10.1016/j.lungcan.2006.09.029
  35. 35. Schoppmeyer K, Kreth F, Wiedmann M, Mossner J, Preiss R, Caca K. A pilot study of bendamustine in advanced bile duct cancer. Anti-Cancer Drugs. 2007;18:697-702. DOI: 10.1097/CAD.0b013e32803d36e6
  36. 36. von Minckwitz G, Chernozemsky I, Sirakova L, Chilingirov P, Souchon R, Marschner N, et al. Bendamustine prolongs progression-free survival in metastatic breast cancer (MBC): A phase III prospective, randomized, multicenter trial of bendamustine hydrochloride, methotrexate and 5-fluorouracil (BMF) versus cyclophosphamide, methotrexate and 5-fluorouracil (CMF) as first-line treatment of MBC. Anti-Cancer Drugs. 2005;16:871-877
  37. 37. Zulkowski K, Kath R, Semrau R, Merkle K, Hoffken K. Regression of brain metastases from breast carcinoma after chemotherapy with bendamustine. Journal of Cancer Research and Clinical Oncology. 2002;128:111-113. DOI: 10.1007/s00432-001-0303-4
  38. 38. Uehling DE, Harris PA. Recent progress on MAP kinase pathway inhibitors. Bioorganic & Medicinal Chemistry Letters. 2015;25:4047-4056. DOI: 10.1016/j.bmcl.2015.07.093
  39. 39. Yeh TC, Marsh V, Bernat BA, Ballard J, Colwell H, Evans RJ, et al. Biological characterization of ARRY-142886 (AZD6244), a potent, highly selective mitogen-activated protein kinase kinase 1/2 inhibitor. Clinical Cancer Research. 2007;13:1576-1583. DOI: 10.1158/1078-0432.CCR-06-1150
  40. 40. Davies BR, Logie A, McKay JS, Martin P, Steele S, Jenkins R, et al. AZD6244 (ARRY-142886), a potent inhibitor of mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 kinases: Mechanism of action in vivo, pharmacokinetic/pharmacodynamic relationship, and potential for combination in preclinical models. Molecular Cancer Therapeutics. 2007;6:2209-2219. DOI: 10.1158/1535-7163.MCT-07-0231
  41. 41. Beloueche-Babari M, Jamin Y, Arunan V, Walker-Samuel S, Revill M, Smith PD, et al. Acute tumour response to the MEK1/2 inhibitor selumetinib (AZD6244, ARRY-142886) evaluated by non-invasive diffusion-weighted MRI. British Journal of Cancer. 2013;109:1562-1569. DOI: 10.1038/bjc.2013.456
  42. 42. El-Hoss J, Kolind M, Jackson MT, Deo N, Mikulec K, McDonald MM, et al. Modulation of endochondral ossification by MEK inhibitors PD0325901 and AZD6244 (selumetinib). Bone. 2014;59:151-161. DOI: 10.1016/j.bone.2013.11.013
  43. 43. Jain N, Curran E, Iyengar NM, Diaz-Flores E, Kunnavakkam R, Popplewell L, et al. Phase II study of the oral MEK inhibitor selumetinib in advanced acute myelogenous leukemia: A University of Chicago phase II consortium trial. Clinical Cancer Research. 2014;20:490-498. DOI: 10.1158/1078-0432.CCR-13-1311
  44. 44. Ma BB, Lui VW, Cheung CS, Lau CP, Ho K, Hui EP, et al. Activity of the MEK inhibitor selumetinib (AZD6244; ARRY-142886) in nasopharyngeal cancer cell lines. Investigational New Drugs. 2013;31:30-38. DOI: 10.1007/s10637-012-9828-4
  45. 45. Janne PA, Shaw AT, Pereira JR, Jeannin G, Vansteenkiste J, Barrios C, et al. Selumetinib plus docetaxel for KRAS-mutant advanced non-small-cell lung cancer: A randomised, multicentre, placebo-controlled, phase II study. The Lancet Oncology. 2013;14:38-47. DOI: 10.1016/S1470-2045(12)70489-8
  46. 46. Janne PA, van den Heuvel MM, Barlesi F, Cobo M, Mazieres J, Crino L, et al. Selumetinib plus docetaxel compared with docetaxel alone and progression-free survival in patients with KRAS-mutant advanced non-small cell lung cancer: The SELECT-1 randomized clinical trial. JAMA. 2017;317:1844-1853. DOI: 10.1001/jama.2017.3438
  47. 47. Cheng Y, Tian H. Current development status of MEK inhibitors. Molecules. 2017;22:1-20. DOI: 10.3390/molecules22101551
  48. 48. Ciombor KK, Bekaii-Saab T. Selumetinib for the treatment of cancer. Expert Opinion on Investigational Drugs. 2015;24:111-123. DOI: 10.1517/13543784.2015.982275
  49. 49. Ramamurthy VP, Ramalingam S, Kwegyir-Afful AK, Hussain A, Njar VC. Targeting of protein translation as a new treatment paradigm for prostate cancer. Current Opinion in Oncology. 2017;29:210-220. DOI: 10.1097/CCO.0000000000000367
  50. 50. McKay RR, Werner L, Fiorillo M, Roberts J, Heath EI, Bubley GJ, et al. Efficacy of therapies after galeterone in patients with castration-resistant prostate cancer. Clinical Genitourinary Cancer. 2017;15:463-471. DOI: 10.1016/j.clgc.2016.10.006
  51. 51. Handratta VD, Vasaitis TS, Njar VC, Gediya LK, Kataria R, Chopra P, et al. Novel C-17-heteroaryl steroidal CYP17 inhibitors/antiandrogens: Synthesis, in vitro biological activity, pharmacokinetics, and antitumor activity in the LAPC4 human prostate cancer xenograft model. Journal of Medicinal Chemistry. 2005;48:2972-2984. DOI: 10.1021/jm040202w
  52. 52. Clement OO, Freeman CM, Hartmann RW, Handratta VD, Vasaitis TS, Brodie AM, et al. Three dimensional pharmacophore modeling of human CYP17 inhibitors. Potential agents for prostate cancer therapy. Journal of Medicinal Chemistry. 2003;46:2345-2351. DOI: 10.1021/jm020576u
  53. 53. Vasaitis T, Belosay A, Schayowitz A, Khandelwal A, Chopra P, Gediya LK, et al. Androgen receptor inactivation contributes to antitumor efficacy of 17{alpha}-hydroxylase/17,20-lyase inhibitor 3beta-hydroxy-17-(1H-benzimidazole-1-yl)androsta-5,16-diene in prostate cancer. Molecular Cancer Therapeutics. 2008;7:2348-2357. DOI: 10.1158/1535-7163.MCT-08-0230
  54. 54. Bruno RD, Gover TD, Burger AM, Brodie AM, Njar VC. 17alpha-hydroxylase/17,20 lyase inhibitor VN/124-1 inhibits growth of androgen-independent prostate cancer cells via induction of the endoplasmic reticulum stress response. Molecular Cancer Therapeutics. 2008;7:2828-2836. DOI: 10.1158/1535-7163.MCT-08-0336.
  55. 55. Schayowitz A, Sabnis G, Njar VC, Brodie AM. Synergistic effect of a novel antiandrogen, VN/124-1, and signal transduction inhibitors in prostate cancer progression to hormone independence in vitro. Molecular Cancer Therapeutics. 2008;7:121-132. DOI: 10.1158/1535-7163.MCT-07-0581
  56. 56. Kwegyir-Afful AK, Ramalingam S, Purushottamachar P, Ramamurthy VP, Njar VC. Galeterone and VNPT55 induce proteasomal degradation of AR/AR-V7, induce significant apoptosis via cytochrome c release and suppress growth of castration resistant prostate cancer xenografts in vivo. Oncotarget. 2015;6:27440-27460. DOI: 10.18632/oncotarget.4578.
  57. 57. Purushottamachar P, Godbole AM, Gediya LK, Martin MS, Vasaitis TS, Kwegyir-Afful AK, et al. Systematic structure modifications of multitarget prostate cancer drug candidate galeterone to produce novel androgen receptor down-regulating agents as an approach to treatment of advanced prostate cancer. Journal of Medicinal Chemistry. 2013;56:4880-4898. DOI: 10.1021/jm400048v
  58. 58. Kwegyir-Afful AK, Bruno RD, Purushottamachar P, Murigi FN, Njar VC. Galeterone and VNPT55 disrupt Mnk-eIF4E to inhibit prostate cancer cell migration and invasion. The FEBS Journal. 2016;283:3898-3918. DOI: 10.1111/febs.13895
  59. 59. Kwegyir-Afful AK, Murigi FN, Purushottamachar P, Ramamurthy VP, Martin MS, Njar VCO. Galeterone and its analogs inhibit Mnk-eIF4E axis, synergize with gemcitabine, impede pancreatic cancer cell migration, invasion and proliferation and inhibit tumor growth in mice. Oncotarget. 2017;8:52381-52402. DOI: 10.18632/oncotarget.14154
  60. 60. Schayowitz A, Sabnis G, Goloubeva O, Njar VC, Brodie AM. Prolonging hormone sensitivity in prostate cancer xenografts through dual inhibition of AR and mTOR. British Journal of Cancer. 2010;103:1001-1007. DOI: 10.1038/sj.bjc.6605882
  61. 61. Bruno RD, Vasaitis TS, Gediya LK, Purushottamachar P, Godbole AM, Ates-Alagoz Z, et al. Synthesis and biological evaluations of putative metabolically stable analogs of VN/124-1 (TOK-001): Head to head anti-tumor efficacy evaluation of VN/124-1 (TOK-001) and abiraterone in LAPC-4 human prostate cancer xenograft model. Steroids. 2011;76:1268-1279. DOI: 10.1016/j.steroids.2011.06.002
  62. 62. McKay RR, Mamlouk K, Montgomery B, Taplin ME. Treatment with galeterone in an elderly man with castration-resistant prostate cancer: A case report. Clinical Genitourinary Cancer. 2015;13:e325-e328. DOI: 10.1016/j.clgc.2014.12.015
  63. 63. Montgomery B, Eisenberger MA, Rettig MB, Chu F, Pili R, Stephenson JJ, et al. Androgen receptor modulation optimized for response (ARMOR) phase I and II studies: Galeterone for the treatment of castration-resistant prostate cancer. Clinical Cancer Research. 2016;22:1356-1363. DOI: 10.1158/1078-0432.CCR-15-1432
  64. 64. Rose AAN. Drugs Today (Barc). 2019;55(4):247-264

Written By

Puranik Purushottamachar, Senthilmurugan Ramalingam and Vincent C.O. Njar

Submitted: 27 November 2018 Reviewed: 06 May 2019 Published: 10 June 2019