Next Article in Journal
In Situ Formation of a Relatively Transparent Ion-Associate Liquid Phase from an Aqueous Phase and Its Application to Microextraction/High-Performance Liquid Chromatography–Fluorescence Detection of Bisphenol A in Water
Next Article in Special Issue
Chemical Properties and Biological Activity of Bee Pollen
Previous Article in Journal
Lupin as a Source of Bioactive Antioxidant Compounds for Food Products
Previous Article in Special Issue
In Vitro and In Silico Activities of E. radiata and E. cinerea as an Enhancer of Antibacterial, Antioxidant, and Anti-Inflammatory Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Pharmacological Implications of Flavopiridol: An Updated Overview

by
Hemant Joshi
1,
Hardeep Singh Tuli
2,
Anuj Ranjan
3,
Abhishek Chauhan
4,
Shafiul Haque
5,6,7,
Seema Ramniwas
8,
Gurpreet Kaur Bhatia
9 and
Divya Kandari
1,*
1
School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
2
Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India
3
Academy of Biology and Biotechnology, Southern Federal University, Stachki 194/1, Rostov-on-Don 344090, Russia
4
Amity Institute of Environmental Toxicology Safety and Management, Amity University, Sector 125, Noida 201301, India
5
Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
6
Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut 11022801, Lebanon
7
Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 13306, United Arab Emirates
8
University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali 140413, India
9
Department of Physics, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(22), 7530; https://doi.org/10.3390/molecules28227530
Submission received: 15 October 2023 / Revised: 6 November 2023 / Accepted: 6 November 2023 / Published: 10 November 2023
(This article belongs to the Special Issue Advances in Natural Products and Their Biological Activities)

Abstract

:
Flavopiridol is a flavone synthesized from the natural product rohitukine, which is derived from an Indian medicinal plant, namely Dysoxylum binectariferum Hiern. A deeper understanding of the biological mechanisms by which such molecules act may allow scientists to develop effective therapeutic strategies against a variety of life-threatening diseases, such as cancer, viruses, fungal infections, parasites, and neurodegenerative diseases. Mechanistic insight of flavopiridol reveals its potential for kinase inhibitory activity of CDKs (cyclin-dependent kinases) and other kinases, leading to the inhibition of various processes, including cell cycle progression, apoptosis, tumor proliferation, angiogenesis, tumor metastasis, and the inflammation process. The synthetic derivatives of flavopiridol have overcome a few demerits of its parent compound. Moreover, these derivatives have much improved CDK-inhibitory activity and therapeutic abilities for treating severe human diseases. It appears that flavopiridol has potential as a candidate for the formulation of an integrated strategy to combat and alleviate human diseases. This review article aims to unravel the potential therapeutic effectiveness of flavopiridol and its possible mechanism of action.

Graphical Abstract

1. Introduction

Plants, the only producers in ecosystems, have influenced humankind and other life systems. Novel and effective therapeutic agents can be developed using bioactive products from plants that can be applicable to various dreadful illnesses, such as cancer and viral, fungal, and neurological diseases [1]. This review emphasizes the therapeutic benefits associated with a compound named flavopiridol (FP). Basically, FP is a flavone and semi-synthetic derivative of the naturally occurring product rohitukine [2]. Rohitukine is a chromone alkaloid found in four different plants: Dysoxylum binectariferum Hiern. (stem bark), Amoora rohituka (Roxb.) Wight & Arn.-(stem and leaves), Schumanniophyton problematicum, and Schumanniophyton magnificum [3,4]. Rohitukine is widely used as a medicine on the Indian subcontinent. It has been found in preclinical studies that FP is a potent anti-cancer medication for leukemia, lymphoma, prostate carcinoma, breast cancer, bladder cancer, and liver cancer [5,6,7,8,9]. Flavopiridol performs its anti-cancer function by inhibiting multiple cyclin-dependent kinases (CDKs) [10]. CDK protein kinases belong to the serine/threonine protein kinase family. They regulate cell cycle progression and transcription and are a potential target for developing chemopreventive therapies [11,12]. Several CDKs, including CDK1, 2, 4, and 6, directly participate in cell progression, whereas CDK7, 8, and 9 assist in transcription [2,13].
The enzymatic activities of CDK1, 2, and 4 are suppressed by flavopiridol, which leads to cell cycle arrests in the G1 and G2 phases, preventing them from entering the synthesis (S) and metaphase (M) phases, respectively [14,15,16]. In addition, it inhibits CDK9, a part of the positive transcription elongation factor b (P-TEFb) that leads to the suppression of phosphorylation at the C-terminus of RNA polymerase II, inhibiting transcription [17]. Among other CDK inhibitors, FP was the first to be studied in human trials [18,19,20]. In 1994, clinical studies first evaluated it as a combined therapeutic regimen for acute myeloid and chronic lymphocytic leukemia [21]. Despite the good results in preclinical studies, some unwanted toxicities (i.e., secretory diarrhea, hypotension, and pro-inflammatory syndrome) were found at higher doses of FP in clinical trials [22]. Numerous derivatives of FP were synthesized to overcome this issue by chemical modification to improve its CDK activity, biodistribution, and therapeutic potential [23,24,25].
Flavopiridol has been extensively explored for its pharmacological therapeutic implications over the last few decades. Flavopiridol modulates several signaling pathways associated with tumor proliferation and metastasis, viral replication, fungal infection, and inflammation-associated diseases [26,27,28,29,30]. Even though such bioactive semi-synthetic compounds are available, their use in treating cancer and viral, fungal, and inflammation-associated diseases is yet to be determined. Thus, understanding how a therapeutic agent such as FP acts is essential. This is imperative so that the research community can better comprehend the different signaling mechanisms underlying the onset of disease and develop innovative treatment approaches. A review is provided here, summarizing the many therapeutic applications of FP, as well as the possible molecular mechanisms of its actions.

2. Chemistry of Flavopiridol

Flavopiridol is also known as alvocidib, L86-8275, HL275, and NSC649890. The chemical name of FP is [4,31]-1-benzopyran-4-one] [32]. This flavone is synthetically produced from the natural anti-rheumatic flavonoid rohitukine. Flavopiridol is a yellow crystalline solid that can be identified through different spectroscopic and chromatographic techniques. Its molecular formula is C21H20ClNO5; its melting temperature ranges from 186 °C to 190 °C, and its molecular weight is 401.84 g mol−1. Flavopiridol has minimal solubility in water; however, it is soluble in organic solvents, including ethanol, dimethyl sulfoxide, and dimethyl formamide [33]. The synthesis of FP occurs in seven steps of chemical reactions. A detailed version of its synthesis has been reported previously [34]. The crystal structure of FP was determined in complex with the cyclin-dependent kinase2 (CDK2) enzyme, which confers the flavone nucleus of the former bound to the ATP binding site pocket of the latter [35]. It shows CDK-inhibitory activity by containing the D-ring, also known as the 3-hydroxy-1-methylpiperidinyl ring, as shown in Figure 1. In contrast, this ring is absent in two structurally relevant natural flavonoids, quercetin and genistein, that show poor CDK-inhibitory activity [34,36,37].

3. Molecular Insights into the Chemopreventive Actions of Flavopiridol

3.1. Apoptosis Activation and Cell Cycle Arrest

An important criterion that distinguishes tumor cells from healthy cells and promotes tumor formation is the loss of apoptotic functions [38]. Apoptosis is a programmed cell death (PCD) that involves condensation and disintegration of less compact chromatin, cleavage of DNA while forming ladders, and membrane blebbing associated with phosphatidylserine exposure [39]. It is a strictly organized process regulated mainly by two types of apoptotic proteins: pro-apoptotic proteins and anti-apoptotic proteins [31]. Currently, anti-cancer therapies focus on inhibiting the development and proliferation of tumor cells by targeting apoptotic pathways. In the quest to develop effective anti-cancer therapies, numerous anti-cancer compounds are being used, including FP. Flavopiridol shows anti-cancer activities mainly by directly inhibiting CDK (i.e., CDK1, 2, 4, 6, and 7) through ATP-competitive inhibitions and indirectly minimizing the levels of cyclins (i.e., cyclin D1 and cyclin D3) or CDK inhibitors (p21 and p27). Reduced levels of cyclin D (cyclin D1, cyclin A, and cyclin E) further reduce CDK enzymatic activity, leading to impaired phosphorylation of the pRb, p107, and p120 proteins (Figure 2) [17,40].
These hypophosphorylated proteins inhibit various transcription factors (MDM-2, Myc, c-Jun, E2F) mainly responsible for growth arrest and promoting apoptosis [40]. Likewise, FP decreased the levels of p21 and p27, led to the growth arrest at the G1/S phase of the cell cycle, activated caspase 9, increased the ratio of bax/bcl2, and induced apoptosis [41,42]. Smith et al., (2008) have reported that a nanomolar concentration of FP was sufficient to arrest the cell cycle in the G1 and G2 phases, and it decreased expression of cyclin D1, increased expression of p21, and promoted apoptosis by activation of caspase 3 or 7, thereby inhibiting rhabdoid tumor growth [43]. In a study using non-small-cell lung cancer cell lines (NCI-H661 and A549) and an osteosarcoma cell line (U2OS), the pro-apoptotic effect of FP was linked with cdk2/cyclin A inhibition and inactivation of the E2F-1 transcription factor during the late S-phase of the cell cycle, resulting in tumor cell apoptosis [44]. A recent study reported that FP, with CKAP2L (cytoskeleton-associated protein 2-like) depletion, led to the suppression of tumor growth and apoptosis activation in esophageal squamous cell carcinoma [45]. Several studies reported that active proliferating cancerous cells are more sensitive to FP after 24 h of treatment than resting cancerous cells [5,32,40]. Later, it was observed that FP has equal potential to activate cell death in both proliferating and non-proliferating cancer cells after 24 h of treatment [4,46]. Since all solid tumors contain resting cancer cells within hypoxic zones due to a lack of oxygen, FP is highly attractive in this context. This feature of FP gains explicit attention for treating chronic lymphocytic leukemia, where most of the lymphocytes exist in the quiescent stage [4]. In a majority of studies to date, FP-induced tumor cell death has been determined to be independent of p53, decreasing the expression of several anti-apoptotic proteins (i.e., Bcl-XL, Bcl-2, XIAP, survivin, and Mcl-1) as shown in Figure 3 [47,48,49,50,51].
Survivin is an apoptosis inhibitor in its phosphorylated form and is responsible for abnormal progression of cell cycles, as well as dodging apoptosis. However, FP suppresses the phosphorylation of survivin at threonine 34, resulting in loss of survivin activity and favoring the activation of cell death in MCF-7 (human breast cancer cells) and HeLa (human cervical cancer cells) [52]. In a study using atypical thyroid cancer cells, Pinto et al., (2020) observed that FP inhibits cell proliferation, downregulates Mcl-1, and favors induction of cell cycle arrest [53]. In multiple myeloma cells, FP was found to downregulate the levels of anti-apoptotic proteins (Mcl-1) and the induction of apoptosis. However, overexpressed Mcl-1 multiple myeloma cell lines restrict FP-induced apoptosis [49]. Further, FP mediates apoptosis in tumor cells via caspase-dependent and -independent mechanisms (Figure 3). Caspase-dependent tumor cell death in breast cancer cells (MDA-MB-435), Burkitt’s lymphoma cell line (GA-10), and human myeloid leukemic cells (U937 and HL-60) has been mediated by FP through the induction of classical mitochondrial proteins, including activation of caspases (caspase 3, 8, and 9), DNA laddering, cleavage of PARP (poly ADP-ribose polymerase) and Bid proteins, elevated levels of Bax, decreased expression of Bcl2, and the release of cytochrome C (cytC) (Figure 3) [54,55,56,57]. In lung or esophageal cancer cells, FP, combined with depsipeptide, mediates apoptosis by activating caspase 9 and the mitochondrial release of cytC [58]. Both in vitro and in vivo studies on human cholangiocarcinoma cell lines showed that FP potentiates cell cycle abortion and enhances caspase-dependent cell death [59]. Some researchers reported that FP in combination with CPT11 (a DNA topoisomerase inhibitor) promotes the cleavage of apoptosis inhibitors and activation of cell death in colon cancer cells (Hct116) [60]. In contrast, caspase- and Bcl2-independent cell death occurred in glioma and lung carcinoma tumors by FP via the secretion of mitochondrial apoptosis-inducing factor (AIF) [61,62]. In addition, a study reported that FP promotes cell death in murine glioma cells (GL261) through the induction of caspase-dependent and -independent cell death by the secretion of cytC and AIF from the mitochondria, respectively [63]. Moreover, FP may suppress the activities of the transcription factor NF-κB and make tumor cells more vulnerable to chemotherapy and radiation therapy. Through the phosphorylation of the Akt protein, the NF-κB transcription factor indirectly regulates a variety of cellular events, including cell proliferation, migration, and apoptosis. Studies have demonstrated that FP potentiates apoptosis in human leukemic cell lines in conjunction with proteasome inhibitors and PI3K (phosphatidylinositol-3-kinase) inhibitors by inhibiting the expression of NF-κB transcription factor [50,64]. Furthermore, FP could also perform synergistic effects with cytostatic drugs to induce tumor cell apoptosis throughout the cell cycle by induction of caspase-dependent and -independent mechanisms [65]. Using MKN-74 (human gastric tumor cells) and MCF-7, Motwani et al., (1999) investigated the synergistic effect of FP with paclitaxel to enhance the caspase activity and found that PARP cleavage led to the activation of cell death [66]. Interestingly, FP in synergy with gemcitabine potentiates the activation of caspase-dependent cell death in human gastric, colon, and pancreatic tumors by suppression of the transcriptional activation of the ribonucleotide reductase M2 subunit [67]. In another experimental study, it was demonstrated that FP enhances mitomycin-C-induced apoptosis in MKN-74 (gastric tumor cells) and MDA-MB-468 (breast cancer tumor cells) by inhibiting the activity of protein kinase C [68]. Likewise, irinotecan-induced apoptosis in advanced hepatocellular carcinoma is potentiated by FP [69]. As a collective, these shreds of evidence suggest that FP individually or in synergy with chemopreventive drugs enhances the induction of tumor cell apoptosis via caspase-dependent or -independent mitochondrial pathways by the downregulation of anti-apoptotic proteins, inactivation of cell cycle promoting proteins, and inhibition of tumor cell proliferation.

3.2. Anti-Metastatic Effect

Tumor metastasis is one of the fatal characteristics of tumor cells through which they disseminate beyond their original location to surrounding and distant tissues or organs [70]. It occurs in a sequential manner where tumor cells first evade anchorage-dependent cell death (anoikis) by detaching from the extracellular matrix (ECM); second, tumor cells invade across the ECM to gain access to blood circulation through the circulatory system and lymphatic system; third, tumor cells extravasate and arrest at distant sites of the human body; fourth, tumor cells colonize and form micro-metastases by warding off immune attack; and finally, the cells instigate neovascularization to promote tumor growth and establish macro-metastases [71]. An estimated 90% of cancer mortality occurs due to tumor metastasis [38]. Detecting and inhibiting malignant tumor metastasis is a significant challenge that must always be addressed in the modern practice of cancer treatment [70,72]. In order to combat this problem, scientists have found several natural and chemical compounds that inhibit tumor metastasis [73,74,75]. Anti-metastatic agents generally include angiogenesis inhibitors, integrin antagonists, VEGF inhibitors, anti-MMP agents, and receptor tyrosine kinase inhibitors [76,77]. The use of FP as an anti-metastatic agent has been documented for more than three decades in cancer research because it possesses various activities, such as decreased VEGF secretion, anti-angiogenesis activity, anti-invasion activity, and decreased secretion of MMP enzymes (Figure 4) [54,78,79]. Using ovarian carcinoma (OCa-I) as an in vivo tumor model, Mason et al., (2004) determined a significant decrease in metastatic nodules in the lungs after FP treatment [80]. Another group of researchers found that FP-treated human osteosarcoma cells showed anti-metastatic activity, as the inhibition of R-cadherin and induction of P-cadherin expression led to the reduced migration and invasion of cancerous cells [81]. Similarly, it was evidenced that FP decreased expression of galectin-3 and increased expression of E-cadherin in KRAS mutant lung adenocarcinoma cells, which reflects its anti-metastatic effects (Figure 4) [82]. Additionally, in vivo mice experiments confirmed that FP can significantly reduce the ability of human osteosarcoma cells (SJSA-1 and 143B) to metastasize into the lungs [81]. Further, several studies reported that the synergistic interaction between FP and other chemopreventive agents increased their anti-invasive and anti-metastatic activities against metastatic cancer. A recent study investigated FP in conjunction with quisinostat (a histone deacetylase inhibitor), and found that the combination is a potential regimen for treating cutaneous and uveal metastatic melanoma, irrespective of NRAS and KRAS mutations [83]. Likewise, another study reported that a synergistic combination of FP with carfilzomib led to the activation of G2/M cell cycle arrest, decreased levels of X-linked inhibitor of apoptosis (XIAP), and augmented apoptosis. This drug combination exposed to mice with human adrenocortical cancer xenografts provoked a significant decrease in cancerous growth due to increased cleaved caspase and decreased expression of XIAP [84]. Holkova et al., (2011) noted that a bortezomib (proteasome inhibitor) and FP combination is a potential therapeutic option against recurrent or refractory B-cell neoplasms [85]. Similarly, researchers have found that gemcitabine and irinotecan, in combination with FP, possess potentiated anti-metastatic effects that facilitate the treatment of metastatic cancers [86]. In an in vivo breast cancer mice model, FP in combination with sorafenib (Raf inhibitor) had anti-metastatic activity, causing a significant reduction in the lung metastatic tumor. This is possibly due to the decreased Mcl-1 expression and Rb signaling in the cancer cell [87]. In summary, it was concluded that FP itself or combined with other anti-cancerous drugs, has potent tumor metastasis inhibitory activity due to its ability to suppress the exodus and infiltration of tumor cells through numerous mechanisms.

3.3. Anti-Angiogenesis

Angiogenesis is the process of developing new blood vessels from existing ones. It occurs during menstruation, wound healing, and under various pathological conditions, including tumor growth, arthritic synovium, and proliferative retinopathy [88]. In tumor cells, angiogenesis is mainly responsible for neovascularization through the activation of endothelial cells by tumor-derived cytokines like vascular endothelial growth factor (VEGF), which encourages tumor cells to survive and expand [89]. The most potent angiogenic growth factor, VEGF, is generally released during hypoxic conditions to help tumor cells adapt to this microenvironment, driven by hypoxia-induced factor-1 (HIF-1α) [90]. Inhibition of cancer cell proliferation can be achieved by targeting angiogenic growth factors, oncogenes, membrane receptors, proteases, and signaling transduction factors involved in the neovascularization of tumor cells. Flavopiridol decreased the expression of VEGF mRNA and its consecutive protein in human monocytes under hypoxic environments by reducing VEGF mRNA stability (Figure 5) [91]. Another group of researchers found that FP inhibited the Src transcription factor and downregulated the expression of VEGF [92]. Further, it was evidenced that FP could also decrease c-fos-induced VEGFD expression by the inhibition of fos promoter activation and PKC [93]. In the studies on GI-LI-N, LAN-5, and ACN (human neuroblastoma cells), researchers demonstrated that FP inhibits the expression of VEGF, which is induced by HIF-1α, picolinic acid, and desferrioxamine [94]. Also, Newcomb et al., (2005) found that the inhibition of angiogenesis by FP in human glioma cell lines (U87MG and T98G) via downregulation of VEGF expression through a decrease in HIF-1α levels occurred even when a proteasome inhibitor was present [78]. Another vital component of the angiogenesis process is matrix metalloproteinase (MMP) enzymes, which are secreted by tumor cells to facilitate cancer cell infiltration and migration through destruction of extracellular matrix components (Figure 5). It was found that FP impaired the release of MMP2 and MMP9 enzymes in breast tumor cells, which eventually caused the suppression of tumor invasion and migration, as observed in a Matrigel invasion assay [54]. Likewise, FP-treated human glioma cells showed decreased secretion of MMP2, were unable to induce the proteolysis of the extracellular matrix, and favored the inhibition of tumor invasion [78]. Takada et al., (2004) noted that FP-mediated inhibition of MMP9 secretion occurred through impaired NF-κB enzymatic activity induced by NF-κB kinase, IκBα kinase, tumor necrosis factor (TNF) receptor 1 (also referred as TNFR1), TNF-receptor-associated factor-2 (TRAF2), and TNF receptor-associated death domain (TRADD) (Figure 5) [79]. Another study observed the anti-angiogenic function of FP in the human colon carcinoma cell line (RKO). It showed a 70% decrease in blood vessel formation using the mouse xenograft Matrigel invasion model [95]. Since human RKO cell lines are devoid of αvβ3- and αvβ5-integrins, this suggests that the FP-mediated anti-angiogenic effect is independent of integrin inhibition. In contrast, Yang et al., (2014) found a synergistic combination of FP with paclitaxel that showed anti-angiogenic effects by decreasing αvβ3 integrin expression in endothelial cells and increasing expression of αvβ3 integrin in ovarian carcinoma cell lines (SKOV3). It was assessed that both in vitro and in vivo anti-angiogenic activity was evaluated using endothelial cell tube formation and tumor microvessel density, respectively [96]. Similarly, a combination of FP with docetaxel in transgenic Gγ/T-15 mice induced apoptosis and decreased angiogenesis, leading to the inhibition of primary and metastatic prostate cancer [97]. Further, authors reported that FP is a better anti-angiogenic molecule than the known angiogenesis inhibitor TNP-470, as observed in a Matrigel invasion model of human colon carcinoma xenografts [98].
Interestingly, Mcferrin et al., (2010) reported another possible mechanism for FP-mediated anti-angiogenic effects. Researchers observed that FP inhibits the activity of CDK9 and vGPCR (Kaposi sarcoma-associated herpesvirus G-protein coupled receptor) in human umbilical vein endothelial cells (HUVECs), which further decreases the secretion of VEGF-A and VEGF-C, leading to a decrease in cell migration and new blood vessel formation [99]. All the pieces of evidence indicated that FP possesses anti-angiogenic effects through numerous mechanisms such as decreasing hypoxic-induced VEGF secretion through decreased HIF-1α expression, decreasing MMP enzyme secretion by inactivation of NF-κB, and inhibition of vGPCR that further inhibits VEGF secretion (Figure 5). Altogether, FP possesses anti-apoptotic and anti-angiogenic activities. Hence, it could be applied to the treatment of different cancers in the future.

3.4. Anti-Inflammatory Effects

Inflammatory diseases occur when the immune system fails to eliminate organisms or foreign substances, resulting in excessive and prolonged inflammation. This event contributes to the progression and pathogenesis of a broad range of severe inflammation-associated diseases such as cancer, asthma, atherosclerosis, neurological illnesses, and arthritis [100,101]. A study reported the dose-dependent inhibition of inflammatory processes in murine collagen-induced arthritis conditions by FP [102]. One of the marked characteristics of inflammation is massive leukocyte infiltration from the bloodstream into the tissue site via the ability of leukocytes to interact with endothelial cells. FP has the potential to show anti-inflammatory effects by inhibiting the leukocyte extravasation and leukocyte–endothelial cell interaction through the suppression of endothelial cell activation [103]. In support of this fact, it was observed that FP blocked neutrophil infiltration and reduced the cell adhesion molecules (E-selectin, ICAM-1, and VCAM-1) expressions in a concanavalin A-induced murine hepatitis model [103]. Additionally, it was proved that FP-mediated suppression of macrophage infiltration occurs when given through the intrathecal route [104]. Further, it was found that FP abrogates ICAM-1 expression as well as NF-κB mediated gene transcription by impairment of CDK9 activity, not by suppressing the canonical NF-κB activation pathway (i.e., abrogation of IκBα kinase and p65 subunit phosphorylation) [103]. Despite being part of P-TEFb (positive transcriptional elongation factor b), CDK9 has been reported to bind with the cytoplasmic part of glycoprotein (gp130), the receptor for IL-6, a pro-inflammatory cytokine [103]. To strengthen the observation of the previous study, it was observed that FP inhibited IL-6/STAT3 (signal transducer and activation of transcription 3) signaling in hepatocarcinoma (hepG2 cell line) via a JAK/STAT signaling pathway, which is a crucial inflammatory pathway to increase neutrophil survival [105]. Further, it was evidenced that FP suppressed IFN-γ mediated nitric oxide (NO) formation within vascular endothelial cells by reducing expression levels of inducible NO synthase (iNOS), which leads to the inactivation of STAT1 and its downstream molecule IFN-γ responsive factor (IRF1) in JAK/STAT signaling, suggesting an anti-inflammatory effect (Figure 6) [106]. It is imperative to recognize that NO and TNF-α are crucial inflammatory molecules and excess production of them is associated with the pathophysiology of many inflammatory illnesses [107]. Interestingly, Haque et al., (2011) reported that FP decreased the production of pro-inflammatory mediators, including TNF-α and NO, in lipopolysaccharide (LPS)-activated cells [108]. In addition to NF-κB and IκBα kinase, it also inhibits the series of MAPK (mitogen-activated protein kinases), including p38, extracellular signal-regulated kinases 1/2 (ERK1/2), c-Jun N-terminal kinases, and stress-induced protein kinases, in the presence of LPS [108]. Further, it was found that FP-treated inhibition of TNF-α and NO in the presence of lipopolysaccharide occurred by abrogation of MAPK and NF-κB stimulation via the MyD88-(Myeloid differentiation factor 88) dependent TLR2 (Toll-like receptor 2) ligand, which may explain the importance of FP to mitigate the inflammatory response (Figure 6) [108]. Basically, the four major signaling pathways exist, including JAK/STAT, MAPK, PI3K, and NF-κB, which regulate inflammatory signals and promote the survival of leukocytes [101]. FP can inhibit the Akt phosphorylation that suppresses the NF-κB induction and controls different biological events, including migration, proliferation, inflammation, and apoptosis (Figure 6) [57]. Dose- and time-dependent abrogation of TNF-α-induced NF-κB by FP was reported in different cell lines, including human kidney cells (A293), human myeloid leukemic cells (HL60), and human T-lymphocyte cells (Jurkat) [79]. In addition to TNF-α, FP also suppressed NF-κB induced by other molecules, such as inflammatory agents, tumor promoters, and carcinogens [79]. This inhibition occurs through the suppression of Akt phosphorylation, and a canonical NF-κB stimulation cascade results in the suppression of TNF-α-induced NF-κB regulated genes such as MMP9, cyclooxygenase-2, and cyclin D1, indicating the immunomodulatory and anti-inflammatory role of FP as illustrated in Figure 6 [79]. In another study, FP was found to have anti-arthritic effects by inhibiting the production of inflammatory mediators (i.e., iNOS) and extracellular-matrix-degrading genes (i.e., MMP1, 3, 9, and 13), protecting human cartilage explants from the adverse effects of pro-inflammatory cytokines (i.e., IL-1β, TNF-α, and LPS) while maintaining cell survival and function [109]. The in vitro and in vivo studies on the bone remodeling model indicate that FP suppresses RANKL (receptor activator of nuclear kappa B ligand) signaling pathways by suppressing the non-canonical NF-κB activation cascade [110]. Information on RANKL suggests that it is a transmembrane protein of the TNF superfamily and is closely linked to acute and chronic inflammation, which strengthens the role of FP as an anti-inflammatory agent [111,112]. Chang and his colleagues reported the abrogation of inflammatory mediators in FP alone or in combination with PLGA (poly (lactic-co-glycolic acid)) nanoparticle-treated astrocytes through significant inhibition of pro-inflammatory cytokines (i.e., IL-1β, IL-6, and TNF-α) and induction of an anti-inflammatory cytokine (i.e., IL-10) [104,113]. Similar results were obtained in a fungal keratitis mouse model, wherein FP inhibited the inflammation induced in fungal keratitis through the production of the IL-1β, IL-6, and TNF-α cytokines while promoting the production of the IL-10 cytokine [29]. All the evidence indicated herein points towards FP’s ability to inhibit inflammation through a multitude of mechanisms, as discussed above, making it a potential anti-inflammatory agent for future clinical research.

4. Efficacy of Flavopiridol as a Therapeutic Agent

The emergence of pathogens that cause life-threatening diseases remains a problem for their treatment and effective disease management and is a significant threat to global public health. Prevention of new infections and their progression to active disease is critical to reducing the burden of the diseases and death caused by them. To address this problem, research is constantly sought for better alternatives to antimicrobial agents due to drug resistance among pathogenic microbes [114]. There is potential for bioactive compounds derived from natural sources to be used to combat drug resistance issues and reduce antibiotic off-target effects [115]. In addition to the anti-cancerous, anti-metastasis, anti-angiogenic, and anti-inflammatory functions of FP, it has also been extensively used as a treatment for different diseases such as viral infections, parasitic infections, and fungal infections caused by several harmful pathogens, as shown in Table 1. It has been observed that it abrogates the replication of different viruses, such as human immunodeficiency virus (HIV), herpes simplex virus (HSV1 and 2), human cytomegalovirus (HCMV), human adenovirus (hAdV5 and hAdV53), and influenza virus A (HINI, H3N2, H7N9) by suppression of mRNA transcription through P-TEFb/CDK9 complex inhibition [26,27,28,116,117]. Similarly, it inhibits the growth of different parasites, including Leishmania mexicana, Toxoplasma gondii, and Plasmodium falciparum [29,118]. Interestingly, it has been noted that FP inhibits fungal growth, biofilm formation, and the adhesion ability of Aspergillus fumigatus, decreases inflammation, and induces autophagy [29]. Aside from these, it has several other pharmacological benefits, described in Table 1, and the underlying molecular mechanisms have been proposed. In summary, it was concluded that FP has antimicrobial and other therapeutic benefits that could be used to overcome drug resistance and unnecessary outcomes.

5. Derivatives of Flavopiridol

As already stated, FP has CDK-inhibitory activity that abrogates the continuation of the cell cycle by arresting the G1 and G2/M phases. This CDK-inhibitory activity is mainly responsible for anti-cancerous, anti-inflammatory, and other pharmacological applications. However, the significant issues hampering FP’s transition from research to the clinic are non-selective kinase inhibitory activity, low target specificity, moderate target affinity, and bioavailability in biological systems. To overcome all these problems, a wide range of FP derivatives have been synthesized by chemical modifications aiming for better biological and pharmacological functionalities. The structure–activity relationship of FP was mentioned in Figure 1. It has been observed that replacement of the chromone moiety of FP by 4-hydroxy benzofuranone improves the kinase inhibitory activity (CDK1 and CDK2) and selectivity against CDK4 and also enhances the anti-cancerous activity [130]. Thio- and oxo-FP analogs increase the selectivity of FP for CDK1 by discriminating from other kinases. In addition, these FP analogs also possess anti-proliferative activity against different human cancer cell lines [131]. Another piece of research indicates that the D-ring of FP is critical for CDK-inhibitory activity, and olefin analogs of FP have shown selective kinase inhibitory activity against CDK4, as well as tumor growth inhibition in MCF-7 cells [34]. Interestingly, P-276-00 is another derivate of FP that shows selective CDK1, CDK4, and CDK9 inhibitory activity compared to other CDKs. Additionally, it also has anti-tumor activities against 14 human cancer cell lines [132]. Further, it was found that the 2-fluorophenyl analog of FP shows 40-fold more selective inhibition to P-TEFb than other CDKs, leading to the inhibition of HIV-1 Tat transactivation and replication; therefore, it could be used as an anti-HIV1 therapeutic [133]. Voruciclib and IIIM-290 are oral bioavailable FP analogs with selective CDK9 inhibitory activity. Voruciclib has shown anti-tumor activity when combined with other anti-cancer drugs to treat different cancers [24]. Another derivative, IIIM-290, displayed caspase-dependent apoptosis in pancreatic cancer and exhibited anti-tumor activity against a wide range of cancer cell lines [23]. Surprisingly, Ibrahim et al., (2020) have reported that thioether-benzimidazoles analogs of FP by chemical modification at the C-ring led to the inhibition of CDK9 and CDK10. This FP derivative is the only compound that has shown CDK10 inhibitory activity, and it also possesses anti-cancer effects by inhibiting tumor growth in seven cancer cell lines [25]. The CDK-inhibitory activity and anti-cancerous effects of other clinically active FP derivatives are summarized in Table 2 and Table 3, respectively.

6. Adverse Effects of Flavopiridol

Flavopiridol has several therapeutic benefits, including anti-cancerous, anti-inflammatory, anti-viral, anti-fungal, and anti-parasitic functions, which have already been described in the previous sections. Despite all these advantages of FP, there is still some dirt in the picture. The significant toxicity concerns associated with using FP are secretory diarrhea and pro-inflammatory syndrome (i.e., tiredness, fever, local tumor pain, and intonation of acute phase reactants) associated with hypotension [19]. Flavopiridol’s effects on in vitro epithelial cell models were assessed to explain its unanticipated toxic effects. Flavopiridol caused chloride secretion in these models, implying that therapeutic antidiarrheal medicines should be used to prevent secretory diarrhea [2]. The maximum tolerated dose (MTD) was increased from 50 mg m−2/24 h to 78 mg m−2/24 h for three days following prophylactic treatment for diarrhea with cholestyramine and loperamide [2]. As an attempt to understand the pro-inflammatory syndrome, different cytokines were examined in the plasma prior to and following FP administration. It was observed that plasma IL-6 levels significantly increased dose-dependently [19,136]. In the phase I clinical study, it was observed that the MTD of FP was 37.5 mg m−2/24 h, and dose-limiting side effects were observed at 53 mg m−2/24 h, including fatigue, vomiting, neutropenia, diarrhea, and nausea [2,19]. In addition, non-dose-limiting toxicities, such as anorexia and local tumor pain, were observed. Similarly, in another clinical study, dose-limiting side effects were observed at 40 mg m−2/24 h MTD for three days with vomiting and nausea [2]. In a phase II dose escalation study of FP, substantial side effects were observed, including anorexia, fatigue, nausea, tumor pain, dyspnea, diarrhea, and cough [137,138]. In another toxicological study, Wirger et al., (2005) reported that at 1 mg/kg/day of FP over three weeks, different side effects were observed, including leukopenia, lesions in the spleen and bone marrow, and gastrointestinal toxicity [7]. Interestingly, in one study, it was found that FP showed drug resistance, though the mechanism is unknown. Later, it was revealed that FP mediates endoplasmic reticulum (ER) stress-induced autophagy, which is responsible for its resistance [139]. In a recent study, it was found that a majority of FP-treated human prostate cancer cells (DU145) died. However, a small sub-population survived and emerged as FP-resistant cancer cells (DU145FP) [140]. DU145FP cancer cells have shown different characteristics, including slow growth, mitochondrial depolarization, induced abundant anti-apoptotic proteins, and less sensitivity to docetaxel and cisplatin [140]. These FP-resistant cancer cells induce mitochondrial lesions that modulate cell metabolism, motility, and advanced neoplastic growth potential [140].

7. Clinical Trials: Results and Experiences

Several clinical trials have demonstrated FP’s effectiveness against many cancers, including leukemia, lymphoma, and breast, prostate, pancreatic, stomach, lung, liver, head and neck tumors [141,142]. It has been shown that FP is safe and effective in patients suffering from different cancers when administered intravenously per day for several weeks [141,142]. Two significant drawbacks to this drug being used alone as an anti-cancer agent in clinical trials are low plasma concentrations due to its binding with serum proteins and unwanted side effects due to its high doses [142]. However, in some cases, this phytochemical can exert anti-cancer activity alone or in combination with some established anti-cancer medications, including vorinostat and gemcitabine hydrochloride in adult solid tumors, cisplatin in ovarian epithelial cancer, bortezomib in B-cell neoplasms, doxorubicin hydrochloride in sarcomas, venetoclax in leukemia, paclitaxel in esophageal cancer, and docetaxel in breast and pancreatic cancer [141,142]. It has been reported that over 60 clinical trials, varying in clinical phases, have been performed with FP; these trials are listed in detail in Table 4.

8. Conclusions and Future Perspectives

The current research on FP has demonstrated its effectiveness in numerous clinical areas. It has been suggested that FP can modulate multiple molecular signaling pathways associated with the development of different cancers, and inflammatory, neurological, viral, fungal, and parasitic diseases. However, future research should explore other therapeutic functions of FP using different omics approaches such as system biology, genomics, epigenomics, transcriptomics, proteomics, and metabolomics. It would be possible to overcome the evolution of cellular resistance by synthesizing FP derivatives to improve the therapeutic efficacy of drugs for targeted diseases. It is possible to increase the effectiveness of the FP manifold by using synergistic approaches with other natural and synthetic anti-cancer medications or drug molecules. Furthermore, the application of FP with engineered nanoparticles, such as polymeric nanoparticles, liposomes, magnetic nanoparticles, exosomes, and metallic nanoparticles, enhanced the drug loading capacity, half-life in biological systems, sustained release, and selective biodistribution. Additionally, the pharmacokinetic and pharmacodynamic profiles were ameliorated over traditional formulations, preventing tumor metastasis and enhancing cancer therapy. Yang et al. (2009) reported that liposomal formulations of FP administered to mice led to reduced systemic clearance, improved plasma concentrations, and increased elimination phase half-life relative to FP alone [143,144]. Moreover, the clinical efficacy of FP with different nanoformulations against different cancers and other targeted diseases must be studied.

Author Contributions

H.J., H.S.T., A.R., A.C., S.H., S.R., G.K.B. and D.K. contributed to the Conceptualization, Formal analysis, Investigation, Methodology, and Resources. D.K. and H.S.T. provided their supervision. H.J. and D.K. wrote the original draft and performed the review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

The authors thank the Department of Biotechnology, Government of India [Fellowship DBT/2017/JNU/849]. The authors thank the Deputyship for Research & Innovation, Ministry of Education in Saudi Arabia, for funding this research work through project number ISP23-101.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

This document includes citations for all the data that were analyzed throughout the literature review.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tuli, H.S.; Sharma, A.K.; Sandhu, S.S.; Kashyap, D. Cordycepin: A bioactive metabolite with therapeutic potential. Life Sci. 2013, 93, 863–869. [Google Scholar] [CrossRef] [PubMed]
  2. Deep, A.; Marwaha, R.K.; Marwaha, M.G.; Nandal, R.; Sharma, A.K. Flavopiridol as cyclin dependent kinase (CDK) inhibitor: A review. New J. Chem. 2018, 42, 18500–18507. [Google Scholar] [CrossRef]
  3. Raju, U.; Nakata, E.; Mason, K.A.; Ang, K.K.; Milas, L. Flavopiridol, a cyclin-dependent kinase inhibitor, enhances radiosensitivity of ovarian carcinoma cells. Cancer Res. 2003, 63, 3263–3267. [Google Scholar] [PubMed]
  4. Blachly, J.S.; Byrd, J.C. Emerging drug profile: Cyclin-dependent kinase inhibitors. Leuk. Lymphoma 2013, 54, 2133–2143. [Google Scholar] [CrossRef]
  5. Carlson, B.A.; Dubay, M.M.; Sausville, E.A.; Brizuela, L.; Worland, P.J. Flavopiridol induces G1 arrest with inhibition of cyclin-dependent kinase (CDK) 2 and CDK4 in human breast carcinoma cells. Cancer Res. 1996, 56, 2973–2978. [Google Scholar]
  6. Drees, M.; A Dengler, W.; Roth, T.; LaBonte, H.; Mayo, J.; Malspeis, L.; Grever, M.; A Sausville, E.; Fiebig, H.H. Flavopiridol (L86-8275): Selective antitumor activity in vitro and activity in vivo for prostate carcinoma cells. Clin. Cancer Res. 1997, 3, 273–279. [Google Scholar]
  7. Wirger, A.; E Perabo, F.G.; Burgemeister, S.; Haase, L.; Schmidt, D.H.; Doehn, C.; Mueller, S.C.; Jocham, D. Flavopiridol, an inhibitor of cyclin-dependent kinases, induces growth inhibition and apoptosis in bladder cancer cells in vitro and in vivo. Anticancer. Res. 2005, 25, 4341–4347. [Google Scholar]
  8. Jackman, K.M.; Frye, C.B.; Hunger, S.P. Flavopiridol displays preclinical activity in acute lymphoblastic leukemia. Pediatr. Blood Cancer 2008, 50, 772–778. [Google Scholar] [CrossRef]
  9. Garcia-Cuellar, M.-P.; Füller, E.; Mäthner, E.; Breitinger, C.; Hetzner, K.; Zeitlmann, L.; Borkhardt, A.; Slany, R.K. Efficacy of cyclin-dependent-kinase 9 inhibitors in a murine model of mixed-lineage leukemia. Leukemia 2014, 28, 1427–1435. [Google Scholar] [CrossRef]
  10. Stewart, Z.A.; Westfall, M.D.; Pietenpol, J.A. Cell-cycle dysregulation and anticancer therapy. Trends Pharmacol. Sci. 2003, 24, 139–145. [Google Scholar] [CrossRef]
  11. Desai, D.; Gu, Y.; O Morgan, D. Activation of human cyclin-dependent kinases in vitro. Mol. Biol. Cell 1992, 3, 571–582. [Google Scholar] [CrossRef] [PubMed]
  12. Zhai, S.; Senderowicz, A.M.; Sausville, E.A.; Figg, W.D. Flavopiridol, a novel cyclin-dependent kinase inhibitor, in clinical development. Ann. Pharmacother. 2002, 36, 905–911. [Google Scholar] [CrossRef] [PubMed]
  13. Zaharevitz, D.W.; Gussio, R.; Leost, M.; Senderowicz, A.M.; Lahusen, T.; Kunick, C.; Meijer, L.; A Sausville, E. Discovery and initial characterization of the paullones, a novel class of small-molecule inhibitors of cyclin-dependent kinases. Cancer Res. 1999, 59, 2566–2569. [Google Scholar] [PubMed]
  14. Arguello, F.; Alexander, M.; Sterry, J.A.; Tudor, G.; Smith, E.M.; Kalavar, N.T.; Greene, J.F.; Koss, W.; Morgan, C.D.; Stinson, S.F.; et al. Flavopiridol induces apoptosis of normal lymphoid cells, causes immunosuppression, and has potent antitumor activity in vivo against human leukemia and lymphoma xenografts. Blood J. Am. Soc. Hematol. 1998, 91, 2482–2490. [Google Scholar]
  15. Schwartz, G.K.; O’Reilly, E.; Ilson, D.; Saltz, L.; Sharma, S.; Tong, W.; Maslak, P.; Stoltz, M.; Eden, L.; Perkins, P.; et al. Phase I study of the cyclin-dependent kinase inhibitor flavopiridol in combination with paclitaxel in patients with advanced solid tumors. J. Clin. Oncol. 2002, 20, 2157–2170. [Google Scholar] [CrossRef]
  16. Lapenna, S.; Giordano, A. Cell cycle kinases as therapeutic targets for cancer. Nat. Rev. Drug Discov. 2009, 8, 547–566. [Google Scholar] [CrossRef]
  17. Chen, R.; Keating, M.J.; Gandhi, V.; Plunkett, W. Transcription inhibition by flavopiridol: Mechanism of chronic lymphocytic leukemia cell death. Blood 2005, 106, 2513–2519. [Google Scholar] [CrossRef]
  18. Senderowicz, A.M.; Headlee, D.; Stinson, S.F.; Lush, R.M.; Kalil, N.; Villalba, L.; Hill, K.; Steinberg, S.M.; Figg, W.D.; Tompkins, A.; et al. Phase I trial of continuous infusion flavopiridol, a novel cyclin-dependent kinase inhibitor, in patients with refractory neoplasms. J. Clin. Oncol. 1998, 16, 2986–2999. [Google Scholar] [CrossRef]
  19. Senderowicz, A.M. Flavopiridol: The first cyclin-dependent kinase inhibitor in human clinical trials. Investig. New Drugs 1999, 17, 313–320. [Google Scholar] [CrossRef]
  20. Aklilu, M.; Kindler, H.L.; Donehower, R.C.; Mani, S.; Vokes, E.E. Phase II study of flavopiridol in patients with advanced colorectal cancer. Ann. Oncol. 2003, 14, 1270–1273. [Google Scholar] [CrossRef]
  21. Colevas, D.; Blaylock, B.; Gravell, A. Clinical trials referral resource. Flavopiridol. Oncology 2002, 16, 1204–1214. [Google Scholar] [PubMed]
  22. Jäger, W.; Zembsch, B.; Wolschann, P.; Pittenauer, E.; Senderowicz, A.M.; Sausville, E.A.; Sedlacek, H.H.; Graf, J.; Thalhammer, T. Metabolism of the anticancer drug flavopiridol, a new inhibitor of cyclin dependent kinases, in rat liver. Life Sci. 1998, 62, 1861–1873. [Google Scholar] [CrossRef] [PubMed]
  23. Bharate, S.B.; Kumar, V.; Jain, S.K.; Mintoo, M.J.; Guru, S.K.; Nuthakki, V.K.; Sharma, M.; Bharate, S.S.; Gandhi, S.G.; Mondhe, D.M.; et al. Discovery and preclinical development of IIIM-290, an orally active potent cyclin-dependent kinase inhibitor. J. Med. Chem. 2018, 61, 1664–1687. [Google Scholar] [CrossRef] [PubMed]
  24. Zhao, L.; Yuan, X.; Wang, J.; Feng, Y.; Ji, F.; Li, Z.; Bian, J. A review on flavones targeting serine/threonine protein kinases for potential anticancer drugs. Bioorg. Med. Chem. 2019, 27, 677–685. [Google Scholar] [CrossRef] [PubMed]
  25. Ibrahim, N.; Bonnet, P.; Brion, J.-D.; Peyrat, J.-F.; Bignon, J.; Levaique, H.; Josselin, B.; Robert, T.; Colas, P.; Bach, S.; et al. Identification of a new series of flavopiridol-like structures as kinase inhibitors with high cytotoxic potency. Eur. J. Med. Chem. 2020, 199, 112355. [Google Scholar] [CrossRef]
  26. Chao, S.-H.; Fujinaga, K.; Marion, J.E.; Taube, R.; Sausville, E.A.; Senderowicz, A.M.; Peterlin, B.M.; Price, D.H. Flavopiridol inhibits P-TEFb and blocks HIV-1 replication. J. Biol. Chem. 2000, 275, 28345–28348. [Google Scholar] [CrossRef]
  27. Yamamoto, M.; Onogi, H.; Kii, I.; Yoshida, S.; Iida, K.; Sakai, H.; Abe, M.; Tsubota, T.; Ito, N.; Hosoya, T.; et al. CDK9 inhibitor FIT-039 prevents replication of multiple DNA viruses. J. Clin. Investig. 2014, 124, 3479–3488. [Google Scholar] [CrossRef]
  28. Perwitasari, O.; Yan, X.; O’Donnell, J.; Johnson, S.; Tripp, R.A.; Schor, S.; Einav, S.; Bloom, B.E.; Krouse, A.J.; Gray, L.; et al. Repurposing kinase inhibitors as antiviral agents to control influenza A virus replication. Assay Drug Dev. Technol. 2015, 13, 638–649. [Google Scholar] [CrossRef]
  29. Gu, L.; Li, C.; Peng, X.; Lin, H.; Niu, Y.; Zheng, H.; Zhao, G.; Lin, J. Flavopiridol Protects against Fungal Keratitis due to Aspergillus fumigatus by Alleviating Inflammation through the Promotion of Autophagy. ACS Infect. Dis. 2022, 8, 2362–2373. [Google Scholar] [CrossRef]
  30. Srikumar, T.; Padmanabhan, J. Potential use of flavopiridol in treatment of chronic diseases. In Drug Discovery from Mother Nature; Springer: Berlin/Heidelberg, Germany, 2016; pp. 209–228. [Google Scholar]
  31. AbAbotaleb, M.; Samuel, S.M.; Varghese, E.; Varghese, S.; Kubatka, P.; Liskova, A.; Büsselberg, D. Flavonoids in cancer and apoptosis. Cancers 2018, 11, 28. [Google Scholar] [CrossRef]
  32. Kaur, G.; Stetler-Stevenson, M.; Sebers, S.; Worland, P.; Sedlacek, H.; Myers, C.; Czech, J.; Naik, R.; Sausville, E. Growth inhibition with reversible cell cycle arrest of carcinoma cells by flavone L86-8275. JNCI J. Natl. Cancer Inst. 1992, 84, 1736–1740. [Google Scholar] [CrossRef] [PubMed]
  33. Senderowicz, A.M.; Sausville, E.A. Preclinical and clinical development of cyclin-dependent kinase modulators. JNCI J. Natl. Cancer Inst. 2000, 92, 376–387. [Google Scholar] [CrossRef] [PubMed]
  34. Murthi, K.K.; Dubay, M.; McClure, C.; Brizuela, L.; Boisclair, M.D.; Worland, P.J.; Mansuri, M.M.; Pal, K. Structure–activity relationship studies of flavopiridol analogues. Bioorg. Med. Chem. Lett. 2000, 10, 1037–1041. [Google Scholar] [CrossRef] [PubMed]
  35. De Azevedo, W.F., Jr.; Mueller-Dieckmann, H.-J.; Schulze-Gahmen, U.; Worland, P.J.; Sausville, E.; Kim, S.-H. Structural basis for specificity and potency of a flavonoid inhibitor of human CDK2, a cell cycle kinase. Proc. Natl. Acad. Sci. USA 1996, 93, 2735–2740. [Google Scholar] [CrossRef]
  36. Joshi, H.; Gupta, D.S.; Abjani, N.K.; Kaur, G.; Mohan, C.D.; Kaur, J.; Aggarwal, D.; Rani, I.; Ramniwas, S.; Abdulabbas, H.S.; et al. Genistein: A promising modulator of apoptosis and survival signaling in cancer. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2023, 396, 2893–2910. [Google Scholar] [CrossRef]
  37. Joshi, H.; Gupta, D.S.; Kaur, G.; Singh, T.; Ramniwas, S.; Sak, K.; Aggarwal, D.; Chhabra, R.S.; Gupta, M.; Saini, A.K.; et al. Nanoformulations of quercetin for controlled delivery: A review of preclinical anticancer studies. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2023, 1–16. [Google Scholar] [CrossRef]
  38. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  39. Bhadra, K. A Mini Review on Molecules Inducing Caspase-Independent Cell Death: A New Route to Cancer Therapy. Molecules 2022, 27, 6401. [Google Scholar] [CrossRef]
  40. Sedlacek, H. Mechanisms of action of flavopiridol. Crit. Rev. Oncol./Hematol. 2001, 38, 139–170. [Google Scholar] [CrossRef]
  41. Javelaud, D.; Besançon, F. Inactivation of p21WAF1Sensitizes Cells to Apoptosis via an Increase of Both p14ARF and p53 Levels and an Alteration of the Bax/Bcl-2 Ratio. J. Biol. Chem. 2002, 277, 37949–37954. [Google Scholar] [CrossRef]
  42. Blagosklonny, M.V.; Darzynkiewicz, Z.; Figg, W., II. Flavopiridol inversely affects p21WAF1/CIP1 and p53 and protects p21-sensitive cells from paclitaxel. Cancer Biol. Ther. 2002, 1, 420–425. [Google Scholar] [CrossRef] [PubMed]
  43. Smith, M.E.; Cimica, V.; Chinni, S.; Challagulla, K.; Mani, S.; Kalpana, G.V.; Tan, B.K.; Tan, L.K.; Yu, K.; Tan, P.H.; et al. Rhabdoid tumor growth is inhibited by flavopiridol. Clin. Cancer Res. 2008, 14, 523–532. [Google Scholar] [CrossRef] [PubMed]
  44. Jiang, J.; Matranga, C.B.; Cai, D.; Latham, V.M., Jr.; Zhang, X.; Lowell, A.M.; Martelli, F.; Shapiro, G.I. Flavopiridol-induced apoptosis during S phase requires E2F-1 and inhibition of cyclin A-dependent kinase activity. Cancer Res. 2003, 63, 7410–7422. [Google Scholar] [PubMed]
  45. Chen, L.; Fang, B.; Qiao, L.; Zheng, Y. Discovery of Anticancer Activity of Amentoflavone on Esophageal Squamous Cell Carcinoma: Bioinformatics, Structure-Based Virtual Screening, and Biological Evaluation. J. Microbiol. Biotechnol. 2022, 32, 718–729. [Google Scholar] [CrossRef]
  46. Byrd, J.C.; Shinn, C.; Waselenko, J.K.; Fuchs, E.J.; Lehman, T.A.; Nguyen, P.L.; Flinn, I.W.; Diehl, L.F.; Sausville, E.; Grever, M.R. Flavopiridol induces apoptosis in chronic lymphocytic leukemia cells via activation of caspase-3 without evidence of bcl-2 modulation or dependence on functional p53. Blood J. Am. Soc. Hematol. 1998, 92, 3804–3816. [Google Scholar]
  47. König, A.; Schwartz, G.K.; Mohammad, R.M.; Al-Katib, A.; Gabrilove, J.L. The novel cyclin-dependent kinase inhibitor flavopiridol downregulates Bcl-2 and induces growth arrest and apoptosis in chronic B-cell leukemia lines. Blood J. Am. Soc. Hematol. 1997, 90, 4307–4312. [Google Scholar]
  48. Kitada, S.; Zapata, J.M.; Andreeff, M.; Reed, J.C. Protein kinase inhibitors flavopiridol and 7-hydroxy-staurosporine down-regulate antiapoptosis proteins in B-cell chronic lymphocytic leukemia. Blood J. Am. Soc. Hematol. 2000, 96, 393–397. [Google Scholar]
  49. Gojo, I.; Zhang, B.; Fenton, R.G. The cyclin-dependent kinase inhibitor flavopiridol induces apoptosis in multiple myeloma cells through transcriptional repression and down-regulation of Mcl-1. Clin. Cancer Res. 2002, 8, 3527–3538. [Google Scholar]
  50. Dai, Y.; Rahmani, M.; Grant, S. Proteasome inhibitors potentiate leukemic cell apoptosis induced by the cyclin-dependent kinase inhibitor flavopiridol through a SAPK/JNK-and NF-κB-dependent process. Oncogene 2003, 22, 7108–7122. [Google Scholar] [CrossRef]
  51. Wittmann, S.; Bali, P.; Donapaty, S.; Nimmanapalli, R.; Guo, F.; Yamaguchi, H.; Huang, M.; Jove, R.; Wang, H.G.; Bhalla, K. Flavopiridol down-regulates antiapoptotic proteins and sensitizes human breast cancer cells to epothilone B-induced apoptosis. Cancer Res. 2003, 63, 93–99. [Google Scholar]
  52. Wall, N.R.; O’Connor, D.S.; Plescia, J.; Pommier, Y.; Altieri, D.C. Suppression of survivin phosphorylation on Thr34 by flavopiridol enhances tumor cell apoptosis. Cancer Res. 2003, 63, 230–235. [Google Scholar] [PubMed]
  53. Pinto, N.; Prokopec, S.D.; Ghasemi, F.; Meens, J.; Ruicci, K.M.; Khan, I.M.; Mundi, N.; Patel, K.; Han, M.W.; Yoo, J.; et al. Flavopiridol causes cell cycle inhibition and demonstrates anti-cancer activity in anaplastic thyroid cancer models. PLoS ONE 2020, 15, e0239315. [Google Scholar] [CrossRef] [PubMed]
  54. Li, Y.; Bhuiyan, M.; Alhasan, S.; Senderowicz, A.M.; Sarkar, F.H. Induction of Apoptosis and Inhibition of c-erb B-2 in Breast Cancer Cells by Flavopiridol. Clin. Cancer Res. 2000, 6, 223–229. [Google Scholar] [PubMed]
  55. Rapoport, A.P.; Simons-Evelyn, M.; Chen, T.; Sidell, R.; Luhowskyj, S.; Rosell, K.; Obrig, T.; Hicks, D.; Hinkle, P.M.; Nahm, M.; et al. Flavopiridol induces apoptosis and caspase-3 activation of a newly characterized Burkitt’s lymphoma cell line containing mutant p53 genes. Blood Cells Mol. Dis. 2001, 27, 610–624. [Google Scholar] [CrossRef] [PubMed]
  56. Cartee, L.; Smith, R.; Dai, Y.; Rahmani, M.; Rosato, R.; Almenara, J.; Dent, P.; Grant, S. Synergistic induction of apoptosis in human myeloid leukemia cells by phorbol 12-myristate 13-acetate and flavopiridol proceeds via activation of both the intrinsic and tumor necrosis factor-mediated extrinsic cell death pathways. Mol. Pharmacol. 2002, 61, 1313–1321. [Google Scholar] [CrossRef] [PubMed]
  57. Newcomb, E.W. Flavopiridol: Pleiotropic biological effects enhance its anti-cancer activity. Anti-Cancer Drugs 2004, 15, 411–419. [Google Scholar] [CrossRef]
  58. Nguyen, D.M.; Schrump, W.D.; Tsai, W.S.; Chen, A.; Stewart, J.H., IV; Steiner, F.; Schrump, D.S. Enhancement of depsipeptide-mediated apoptosis of lung or esophageal cancer cells by flavopiridol: Activation of the mitochondria-dependent death-signaling pathway. J. Thorac. Cardiovasc. Surg. 2003, 125, 1132–1142. [Google Scholar] [CrossRef]
  59. Saisomboon, S.; Kariya, R.; Vaeteewoottacharn, K.; Wongkham, S.; Sawanyawisuth, K.; Okada, S. Antitumor effects of flavopiridol, a cyclin-dependent kinase inhibitor, on human cholangiocarcinoma in vitro and in an in vivo xenograft model. Heliyon 2019, 5, e01675. [Google Scholar] [CrossRef]
  60. Motwani, M.; Jung, C.; Sirotnak, F.M.; She, Y.; A Shah, M.; Gonen, M.; Schwartz, G.K. Augmentation of apoptosis and tumor regression by flavopiridol in the presence of CPT-11 in Hct116 colon cancer monolayers and xenografts. Clin. Cancer Res. 2001, 7, 4209–4219. [Google Scholar]
  61. Achenbach, T.V.; Müller, R.; Slater, E.P. Bcl-2 Independence of Flavopiridol-induced Apoptosis: Mitochondrial depolarization in the absence of cytochromec release. J. Biol. Chem. 2000, 275, 32089–32097. [Google Scholar] [CrossRef]
  62. Alonso, M.; Tamasdan, C.; Miller, D.C.; Newcomb, E.W. Flavopiridol induces apoptosis in glioma cell lines independent of retinoblastoma and p53 tumor suppressor pathway alterations by a caspase-independent pathway. Mol. Cancer Ther. 2003, 2, 139–150. [Google Scholar] [PubMed]
  63. Newcomb, E.W.; Tamasdan, C.; Entzminger, Y.; Alonso, J.; Friedlander, D.; Crisan, D.; Miller, D.C.; Zagzag, D. Flavopiridol induces mitochondrial-mediated apoptosis in murine glioma GL261 cells via release of cytochrome c and apoptosis inducing factor. Cell Cycle 2003, 2, 242–249. [Google Scholar] [CrossRef]
  64. Yu, C.; Rahmani, M.; Dai, Y.; Conrad, D.; Krystal, G.; Dent, P.; Grant, S. The lethal effects of pharmacological cyclin-dependent kinase inhibitors in human leukemia cells proceed through a phosphatidylinositol 3-kinase/Akt-dependent process. Cancer Res. 2003, 63, 1822–1833. [Google Scholar] [PubMed]
  65. Decaudin, D.; Marzo, I.; Brenner, C.; Kroemer, G. Mitochondria in chemotherapy-induced apoptosis: A prospective novel target of cancer therapy. Int. J. Oncol. 1998, 12, 141–193. [Google Scholar] [CrossRef] [PubMed]
  66. Motwani, M.; Delohery, T.M.; Schwartz, G.K. Sequential dependent enhancement of caspase activation and apoptosis by flavopiridol on paclitaxel-treated human gastric and breast cancer cells. Clin. Cancer Res. 1999, 5, 1876–1883. [Google Scholar]
  67. Jung, C.P.; Motwani, M.V.; Schwartz, G.K. Flavopiridol increases sensitization to gemcitabine in human gastrointestinal cancer cell lines and correlates with down-regulation of ribonucleotide reductase M2 subunit. Clin. Cancer Res. 2001, 7, 2527–2536. [Google Scholar]
  68. Schwartz, G.K.; Farsi, K.; Maslak, P.; Kelsen, D.P.; Spriggs, D. Potentiation of apoptosis by flavopiridol in mitomycin-C-treated gastric and breast cancer cells. Clin. Cancer Res. 1997, 3, 1467–1472. [Google Scholar]
  69. Ang, C.; O’Reilly, E.M.; Carvajal, R.D.; Capanu, M.; Gonen, M.; Doyle, L.; Ghossein, R.; Schwartz, L.; Jacobs, G.; Ma, J.; et al. A nonrandomized, phase II study of sequential irinotecan and flavopiridol in patients with advanced hepatocellular carcinoma. Gastrointest. Cancer Res. GCR 2012, 5, 185. [Google Scholar]
  70. Joshi, H.; Kumar, G.; Tuli, H.S.; Mittal, S. Inhibition of Cancer Cell Metastasis by Nanotherapeutics: Current Achievements and Future Trends. In Nanotherapeutics in Cancer; Jenny Stanford Publishing: Singapore, 2023; pp. 161–209. [Google Scholar]
  71. Banerjee, D.; Cieslar-Pobuda, A.; Zhu, G.H.; Wiechec, E.; Patra, H.K. Adding nanotechnology to the metastasis treatment arsenal. Trends Pharmacol. Sci. 2019, 40, 403–418. [Google Scholar] [CrossRef]
  72. Lu, W.; Kang, Y. Epithelial-mesenchymal plasticity in cancer progression and metastasis. Dev. Cell 2019, 49, 361–374. [Google Scholar] [CrossRef]
  73. Kumar, G.; Tuli, H.S.; Mittal, S.; Shandilya, J.K.; Tiwari, A.; Sandhu, S.S. Isothiocyanates: A class of bioactive metabolites with chemopreventive potential. Tumor Biol. 2015, 36, 4005–4016. [Google Scholar] [CrossRef] [PubMed]
  74. Kashyap, D.; Mittal, S.; Sak, K.; Singhal, P.; Tuli, H.S. Molecular mechanisms of action of quercetin in cancer: Recent advances. Tumor Biol. 2016, 37, 12927–12939. [Google Scholar] [CrossRef] [PubMed]
  75. Tuli, H.S.; Joshi, H.; Vashishth, K.; Ramniwas, S.; Varol, M.; Kumar, M.; Rani, I.; Rani, V.; Sak, K. Chemopreventive mechanisms of amentoflavone: Recent trends and advancements. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2023, 396, 865–876. [Google Scholar] [CrossRef] [PubMed]
  76. Kong, X.; Cheng, R.; Wang, J.; Fang, Y.; Hwang, K.C. Nanomedicines inhibiting tumor metastasis and recurrence and their clinical applications. Nano Today 2021, 36, 101004. [Google Scholar] [CrossRef]
  77. Tuli, H.S.; Rath, P.; Chauhan, A.; Parashar, G.; Parashar, N.C.; Joshi, H.; Rani, I.; Ramniwas, S.; Aggarwal, D.; Kumar, M.; et al. Wogonin, as a potent anticancer compound: From chemistry to cellular interactions. Exp. Biol. Med. 2023, 248, 820–828. [Google Scholar] [CrossRef]
  78. Newcomb, E.W.; Ali, M.A.; Schnee, T.; Lan, L.; Lukyanov, Y.; Fowkes, M.; Miller, D.C.; Zagzag, D. Flavopiridol downregulates hypoxia-mediated hypoxia-inducible factor-1α expression in human glioma cells by a proteasome-independent pathway: Implications for in vivo therapy. Neuro-Oncology 2005, 7, 225–235. [Google Scholar] [CrossRef]
  79. Takada, Y.; Aggarwal, B.B. Flavopiridol inhibits NF-κB activation induced by various carcinogens and inflammatory agents through inhibition of IκBα kinase and p65 phosphorylation: Abrogation of cyclin D1, cyclooxygenase-2, and matrix metalloprotease-9. J. Biol. Chem. 2004, 279, 4750–4759. [Google Scholar] [CrossRef]
  80. Mason, K.A.; Hunter, N.R.; Raju, U.; Ariga, H.; Husain, A.; Valdecanas, D.; Neal, R.; Ang, K.K.; Milas, L. Flavopiridol increases therapeutic ratio of radiotherapy by preferentially enhancing tumor radioresponse. Int. J. Radiat. Oncol. Biol. Phys. 2004, 59, 1181–1189. [Google Scholar] [CrossRef]
  81. Zocchi, L.; Wu, S.C.; Wu, J.; Hayama, K.L.; Benavente, C.A. The cyclin-dependent kinase inhibitor flavopiridol (alvocidib) inhibits metastasis of human osteosarcoma cells. Oncotarget 2018, 9, 23505. [Google Scholar] [CrossRef]
  82. Dogan Turacli, I.; Demirtas Korkmaz, F.; Candar, T.; Ekmekci, A. Flavopiridol’s effects on metastasis in KRAS mutant lung adenocarcinoma cells. J. Cell. Biochem. 2019, 120, 5628–5635. [Google Scholar] [CrossRef]
  83. Heijkants, R.; Willekens, K.; Schoonderwoerd, M.; Teunisse, A.; Nieveen, M.; Radaelli, E.; Hawinkels, L.; Marine, J.-C.; Jochemsen, A. Combined inhibition of CDK and HDAC as a promising therapeutic strategy for both cutaneous and uveal metastatic melanoma. Oncotarget 2017, 9, 6174–6187. [Google Scholar] [CrossRef] [PubMed]
  84. Nilubol, N.; Boufraqech, M.; Zhang, L.; Gaskins, K.; Shen, M.; Zhang, Y.-Q.; Gara, S.K.; Austin, C.P.; Kebebew, E. Synergistic combination of flavopiridol and carfilzomib targets commonly dysregulated pathways in adrenocortical carcinoma and has biomarkers of response. Oncotarget 2018, 9, 33030–33042. [Google Scholar] [CrossRef] [PubMed]
  85. Holkova, B.; Perkins, E.B.; Ramakrishnan, V.; Tombes, M.B.; Shrader, E.; Talreja, N.; Wellons, M.D.; Hogan, K.T.; Roodman, G.D.; Coppola, D.; et al. Phase I Trial of Bortezomib (PS-341; NSC 681239) and Alvocidib (Flavopiridol; NSC 649890) in Patients with Recurrent or Refractory B-Cell Neoplasms Phase I Trial of Bortezomib and Alvocidib. Clin. Cancer Res. 2011, 17, 3388–3397. [Google Scholar] [CrossRef] [PubMed]
  86. Fekrazad, H.M.; Verschraegen, C.F.; Royce, M.; Smith, H.O.; Lee, F.C.; Rabinowitz, I. A phase I study of flavopiridol in combination with gemcitabine and irinotecan in patients with metastatic cancer. Am. J. Clin. Oncol. 2010, 33, 393–397. [Google Scholar] [CrossRef] [PubMed]
  87. Nagaria, T.S.; Williams, J.L.; Leduc, C.; A Squire, J.; A Greer, P.; Sangrar, W. Flavopiridol synergizes with sorafenib to induce cytotoxicity and potentiate antitumorigenic activity in EGFR/HER-2 and mutant RAS/RAF breast cancer model systems. Neoplasia 2013, 15, 939–951, IN25–IN27. [Google Scholar] [CrossRef]
  88. Carmeliet, P. Angiogenesis in health and disease. Nat. Med. 2003, 9, 653–660. [Google Scholar] [CrossRef]
  89. Tammela, T.; Enholm, B.; Alitalo, K.; Paavonen, K. The biology of vascular endothelial growth factors. Cardiovasc. Res. 2005, 65, 550–563. [Google Scholar] [CrossRef]
  90. Carmeliet, P.; Dor, Y.; Herbert, J.-M.; Fukumura, D.; Brusselmans, K.; Dewerchin, M.; Neeman, M.; Bono, F.; Abramovitch, R.; Maxwell, P.; et al. Role of HIF-1α in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 1998, 394, 485–490. [Google Scholar] [CrossRef]
  91. MMelillo, G.; A Sausville, E.; Cloud, K.; Lahusen, T.; Varesio, L.; Senderowicz, A.M. Flavopiridol, a protein kinase inhibitor, down-regulates hypoxic induction of vascular endothelial growth factor expression in human monocytes. Cancer Res. 1999, 59, 5433–5437. [Google Scholar]
  92. Mukhopadhyay, D.; Tsiokas, L.; Sukhatme, V.P. Wild-type p53 and v-Src exert opposing influences on human vascular endothelial growth factor gene expression. Cancer Res. 1995, 55, 6161–6165. [Google Scholar]
  93. Marconcini, L.; Marchiò, S.; Morbidelli, L.; Cartocci, E.; Albini, A.; Ziche, M.; Bussolino, F.; Oliviero, S. c-fos-induced growth factor/vascular endothelial growth factor D induces angiogenesis in vivo and in vitro. Proc. Natl. Acad. Sci. USA 1999, 96, 9671–9676. [Google Scholar] [CrossRef] [PubMed]
  94. Rapella, A.; Negrioli, A.; Melillo, G.; Pastorino, S.; Varesio, L.; Bosco, M.C. Flavopiridol inhibits vascular endothelial growth factor production induced by hypoxia or picolinic acid in human neuroblastoma. Int. J. Cancer 2002, 99, 658–664. [Google Scholar] [CrossRef] [PubMed]
  95. Kerr, J.S.; Wexler, R.S.; A Mousa, S.; Robinson, C.S.; Wexler, E.J.; Mohamed, S.; E Voss, M.; Devenny, J.J.; Czerniak, P.M.; Gudzelak, A.; et al. Novel small molecule alpha v integrin antagonists: Comparative anti-cancer efficacy with known angiogenesis inhibitors. Anticancer Res. 1999, 19, 959–968. [Google Scholar] [PubMed]
  96. Yang, G.; Sun, H.; Kong, Y.; Hou, G.; Han, J. Diversity of RGD radiotracers in monitoring antiangiogenesis of flavopiridol and paclitaxel in ovarian cancer xenograft-bearing mice. Nucl. Med. Biol. 2014, 41, 856–862. [Google Scholar] [CrossRef]
  97. Reiner, T.; de las Pozas, A.; Perez-Stable, C. Sequential combinations of flavopiridol and docetaxel inhibit prostate tumors, induce apoptosis, and decrease angiogenesis in the Gγ/T-15 transgenic mouse model of prostate cancer. Prostate 2006, 66, 1487–1497. [Google Scholar] [CrossRef]
  98. Robinson, C.; Slee, A.; Kerr, J. Flavopiridol inhibits angiogenesis. FASEB J. 1997, 9650, 20814–23998. [Google Scholar]
  99. McFerrin, H.; Angelova, M.; Abboud, E.; Nelson, A.; Betancourt, A.; Morris, G.; Shelby, B.; Morris, C.; Sullivan, D. The angiogenic properties of Kaposi’s sarcoma-associated herpesvirus encoded G-protein coupled receptor are reduced by flavopiridol, an inhibitor of cyclin-dependent kinase 9. Infect. Agents Cancer 2010, 5, A75. [Google Scholar] [CrossRef]
  100. Han, Y.; Zhan, Y.; Hou, G.; Li, L. Cyclin-dependent kinase 9 may as a novel target in downregulating the atherosclerosis inflammation. Biomed. Rep. 2014, 2, 775–779. [Google Scholar] [CrossRef]
  101. Leitch, A.; Haslett, C.; Rossi, A. Cyclin-dependent kinase inhibitor drugs as potential novel anti-inflammatory and pro-resolution agents. Br. J. Pharmacol. 2009, 158, 1004–1016. [Google Scholar] [CrossRef]
  102. Krystof, V.; Baumli, S.; Furst, R. Perspective of cyclin-dependent kinase 9 (CDK9) as a drug target. Curr. Pharm. Des. 2012, 18, 2883–2890. [Google Scholar] [CrossRef]
  103. Schmerwitz, U.K.; Sass, G.; Khandoga, A.G.; Joore, J.; Mayer, B.A.; Berberich, N.; Totzke, F.; Krombach, F.; Tiegs, G.; Zahler, S.; et al. Flavopiridol protects against inflammation by attenuating leukocyte-endothelial interaction via inhibition of cyclin-dependent kinase 9. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 280–288. [Google Scholar] [CrossRef] [PubMed]
  104. Chang, L.; Zhang, J.; Pei-Qian, Z.; Chang-Hua, M.; Lin-Hui, Y.; Jun, L.; Zhen-Zhong, L.U.O.; Guo-Hai, X.U. Intrathecal administration of flavopiridol promotes regeneration in experimental model of spinal cord injury. Turk. Neurosurg. 2016, 26, 922–929. [Google Scholar]
  105. Hou, T.; Ray, S.; Brasier, A.R. The functional role of an interleukin 6-inducible CDK9· STAT3 complex in human γ-fibrinogen gene expression. J. Biol. Chem. 2007, 282, 37091–37102. [Google Scholar] [CrossRef] [PubMed]
  106. Terashima, T.; Haque, A.; Kajita, Y.; Takeuchi, A.; Nakagawa, T.; Yokochi, T. Flavopiridol inhibits interferon-γ-induced nitric oxide production in mouse vascular endothelial cells. Immunol. Lett. 2012, 148, 91–96. [Google Scholar] [CrossRef]
  107. Sharma, J.N.; Al-Omran, A.; Parvathy, S.S. Role of nitric oxide in inflammatory diseases. Inflammopharmacology 2007, 15, 252–259. [Google Scholar] [CrossRef]
  108. Haque, A.; Koide, N.; Iftakhar-E-Khuda, I.; Noman, A.S.M.; Odkhuu, E.; Badamtseren, B.; Naiki, Y.; Komatsu, T.; Yoshida, T.; Yokochi, T. Flavopiridol inhibits lipopolysaccharide-induced TNF-α production through inactivation of nuclear factor-κB and mitogen-activated protein kinases in the MyD88-dependent pathway. Microbiol. Immunol. 2011, 55, 160–167. [Google Scholar] [CrossRef]
  109. Yik, J.H.; Hu Za Kumari, R.; Christiansen, B.A.; Haudenschild, D.R. Cyclin-Dependent Kinase 9 Inhibition Protects Cartilage From the Catabolic Effects of Proinflammatory Cytokines. Arthritis Rheumatol. 2014, 66, 1537–1546. [Google Scholar] [CrossRef]
  110. Hu Za Chen, Y.; Song, L.; Yik, J.H.; Haudenschild, D.R.; Fan, S. Flavopiridol protects bone tissue by attenuating RANKL induced osteoclast formation. Front. Pharmacol. 2018, 9, 174. [Google Scholar]
  111. Brendan, F.; Boyce, M.; Xing, L. Functions of RANKL/RANK/OPG in bone modelling and remodelling. Arch. Biochem. Biophys. 2008, 473, 139–146. [Google Scholar]
  112. Xing, L.; Xiu, Y.; Boyce, B.F. Osteoclast fusion and regulation by RANKL-dependent and independent factors. World J. Orthop. 2012, 3, 212. [Google Scholar] [CrossRef]
  113. Ren, H.; Han, M.; Zhou, J.; Zheng, Z.-F.; Lu, P.; Wang, J.-J.; Wang, J.Q.; Mao, Q.J.; Gao, J.Q.; Ouyang, H.W. Repair of spinal cord injury by inhibition of astrocyte growth and inflammatory factor synthesis through local delivery of flavopiridol in PLGA nanoparticles. Biomaterials 2014, 35, 6585–6594. [Google Scholar] [CrossRef] [PubMed]
  114. Joshi, H.; Verma, A.; Soni, D.K. Impact of Microbial Genomics Approaches for Novel Antibiotic Target. In Microbial Genomics in Sustainable Agroecosystems; Springer: Berlin/Heidelberg, Germany, 2019; pp. 75–88. [Google Scholar]
  115. Kashyap, D.; Tuli, H.S.; Sharma, A.K. Ursolic acid (UA): A metabolite with promising therapeutic potential. Life Sci. 2016, 146, 201–213. [Google Scholar] [CrossRef] [PubMed]
  116. Nelson, P.J.; D’agati, V.D.; Gries, J.-M.; Suarez, J.-R.; Gelman, I.H. Amelioration of nephropathy in mice expressing HIV-1 genes by the cyclin-dependent kinase inhibitor flavopiridol. J. Antimicrob. Chemother. 2003, 51, 921–929. [Google Scholar] [CrossRef] [PubMed]
  117. Ou, M.; Sandri-Goldin, R.M. Inhibition of cdk9 during herpes simplex virus 1 infection impedes viral transcription. PLoS ONE 2013, 8, e79007. [Google Scholar] [CrossRef] [PubMed]
  118. Del Campo, J.S.M.; Eckshtain-Levi, M.; Sobrado, P. Identification of eukaryotic UDP-galactopyranose mutase inhibitors using the ThermoFAD assay. Biochem. Biophys. Res. Commun. 2017, 493, 58–63. [Google Scholar] [CrossRef]
  119. Leggio, G.M.; Catania, M.V.; Puzzo, D.; Spatuzza, M.; Pellitteri, R.; Gulisano, W.; Torrisi, S.A.; Giurdanella, G.; Piazza, C.; Impellizzeri, A.R.; et al. The antineoplastic drug flavopiridol reverses memory impairment induced by Amyloid-ß1-42 oligomers in mice. Pharmacol. Res. 2016, 106, 10–20. [Google Scholar] [CrossRef]
  120. Wang, F.; Corbett, D.; Osuga, H.; Osuga, S.; Ikeda, J.-E.; Slack, R.S.; Hogan, M.J.; Hakim, A.M.; Park, D.S. Inhibition of cyclin-dependent kinases improves CA1 neuronal survival and behavioral performance after global ischemia in the rat. J. Cereb. Blood Flow Metab. 2002, 22, 171–182. [Google Scholar] [CrossRef]
  121. Osuga, H.; Osuga, S.; Wang, F.; Fetni, R.; Hogan, M.J.; Slack, R.S.; Hakim, A.M.; Ikeda, J.-E.; Park, D.S. Cyclin-dependent kinases as a therapeutic target for stroke. Proc. Natl. Acad. Sci. USA 2000, 97, 10254–10259. [Google Scholar] [CrossRef]
  122. Wu, J.; Stoica, B.A.; Dinizo, M.; Pajoohesh-Ganji, A.; Piao, C.; Faden, A.I. Delayed cell cycle pathway modulation facilitates recovery after spinal cord injury. Cell Cycle 2012, 11, 1782–1795. [Google Scholar] [CrossRef]
  123. Padmanabhan, J.; Brown, K.R.; Padilla, A.; Shelanski, M.L. Functional role of RNA polymerase II and P70 S6 kinase in KCl withdrawal-induced cerebellar granule neuron apoptosis. J. Biol. Chem. 2015, 290, 5267–5279. [Google Scholar] [CrossRef]
  124. Di Giovanni, S.; Movsesyan, V.; Ahmed, F.; Cernak, I.; Schinelli, S.; Stoica, B.; Faden, A.I. Cell cycle inhibition provides neuroprotection and reduces glial proliferation and scar formation after traumatic brain injury. Proc. Natl. Acad. Sci. USA 2005, 102, 8333–8338. [Google Scholar] [CrossRef] [PubMed]
  125. Jaschke, B.; Milz, S.; Vogeser, M.; Michaelis, C.; Vorpahl, M.; Schömig, A.; Kastrati, A.; Wessely, R. Local cyclin-dependent kinase inhibition by flavopiridol inhibits coronary artery smooth muscle cell proliferation and migration: Implications for the applicability on drug-eluting stents to prevent neointima formation following vascular injury. FASEB J. 2004, 18, 1285–1287. [Google Scholar] [CrossRef] [PubMed]
  126. Hassan, P.; Fergusson, D.; Grant, K.M.; Mottram, J.C. The CRK3 protein kinase is essential for cell cycle progression of Leishmania mexicana. Mol. Biochem. Parasitol. 2001, 113, 189–198. [Google Scholar] [CrossRef]
  127. Deshmukh, A.S.; Mitra, P.; Kolagani, A.; Gurupwar, R. Cdk-related kinase 9 regulates RNA polymerase II mediated transcription in Toxoplasma gondii. Biochim. Biophys. Acta (BBA)–Gene Regul. Mech. 2018, 1861, 572–585. [Google Scholar] [CrossRef]
  128. Graeser, R.; Wernli, B.; Franklin, R.M.; Kappes, B. Plasmodium falciparum protein kinase 5 and the malarial nuclear division cycles. Mol. Biochem. Parasitol. 1996, 82, 37–49. [Google Scholar] [CrossRef] [PubMed]
  129. Oikonomakos, N.G.; Schnier, J.B.; Zographos, S.E.; Skamnaki, V.T.; Tsitsanou, K.E.; Johnson, L.N. Flavopiridol Inhibits Glycogen Phosphorylase by Binding at the Inhibitor Site. J. Biol. Chem. 2000, 275, 34566–34573. [Google Scholar] [CrossRef]
  130. Schoepfer, J.; Fretz, H.; Chaudhuri, B.; Muller, L.; Seeber, E.; Meijer, L.; Lozach, O.; Vangrevelinghe, E.; Furet, P. Structure-Based Design and Synthesis of 2-Benzylidene-benzofuran-3-ones as Flavopiridol Mimics. J. Med. Chem. 2002, 45, 1741–1747. [Google Scholar] [CrossRef]
  131. Kim, K.S.; Sack, J.S.; Tokarski, J.S.; Qian, L.; Chao, S.T.; Leith, L.; Kelly, Y.F.; Misra, R.N.; Hunt, J.T.; Kimball, S.D.; et al. Thio- and Oxoflavopiridols, Cyclin-Dependent Kinase 1-Selective Inhibitors: Synthesis and Biological Effects. J. Med. Chem. 2000, 43, 4126–4134. [Google Scholar] [CrossRef]
  132. Joshi, K.S.; Rathos, M.J.; Joshi, R.D.; Sivakumar, M.; Mascarenhas, M.; Kamble, S.; Lal, B.; Sharma, S. In vitro antitumor properties of a novel cyclin-dependent kinase inhibitor, P276-00. Mol. Cancer Ther. 2007, 6, 918–925. [Google Scholar] [CrossRef]
  133. Ali, A.; Ghosh, A.; Nathans, R.S.; Sharova, N.; O’Brien, S.; Cao, H.; Stevenson, M.; Rana, T.M. Identification of Flavopiridol Analogues that Selectively Inhibit Positive Transcription Elongation Factor (P-TEFb) and Block HIV-1 Replication. ChemBioChem 2009, 10, 2072–2080. [Google Scholar] [CrossRef]
  134. Ahn, Y.M.; Vogeti, L.; Liu, C.-J.; Santhapuram, H.K.; White, J.M.; Vasandani, V.; Mitscher, L.A.; Lushington, G.H.; Hanson, P.R.; Powell, D.R.; et al. Design, synthesis, and antiproliferative and CDK2-cyclin a inhibitory activity of novel flavopiridol analogues. Bioorg. Med. Chem. 2007, 15, 702–713. [Google Scholar] [CrossRef] [PubMed]
  135. Dey, J.; Deckwerth, T.L.; Kerwin, W.S.; Casalini, J.R.; Merrell, A.J.; Grenley, M.O.; Burns, C.; Ditzler, S.H.; Dixon, C.P.; Beirne, E.; et al. Voruciclib, a clinical stage oral CDK9 inhibitor, represses MCL-1 and sensitizes high-risk diffuse large B-cell lymphoma to BCL2 inhibition. Sci. Rep. 2017, 7, 18007. [Google Scholar] [CrossRef] [PubMed]
  136. Messmann, R.A.; Ullmann, C.D.; Lahusen, T.; Kalehua, A.; Wasfy, J.; Melillo, G.; Ding, I.; Headlee, D.; Figg, W.D.; A Sausville, E.; et al. Flavopiridol-related proinflammatory syndrome is associated with induction of interleukin-6. Clin. Cancer Res. 2003, 9, 562–570. [Google Scholar]
  137. Connors, J.M.; Kouroukis, C.; Belch, A.; Crump, M.; Imrie, K. Flavopiridol for mantle cell lymphoma: Moderate activity and frequent disease stabilization. Blood 2001, 98, 3355. [Google Scholar]
  138. Burdette-Radoux, S.; Tozer, R.; Lohmann, R.; Quirt, I.; Ernst, D.; Walsh, W.; Wainman, N.; Colevas, D.; Eisenhauer, E. A. NCIC CTG phase II study of flavopiridol in patients with previously untreated metastatic malignant melanoma. Proc. Am. Soc. Clin. Oncol. 2002, 21, 346. [Google Scholar]
  139. Mahoney, E.; Byrd, J.C.; Johnson, A.J. Autophagy and ER stress play an essential role in the mechanism of action and drug resistance of the cyclin-dependent kinase inhibitor flavopiridol. Autophagy 2013, 9, 434–435. [Google Scholar] [CrossRef]
  140. Li, X.; Lu, J.; Kan, Q.; Li, X.; Fan, Q.; Li, Y.; Huang, R.; Slipicevic, A.; Dong, H.P.; Eide, L.; et al. Metabolic reprogramming is associated with flavopiridol resistance in prostate cancer DU145 cells. Sci. Rep. 2017, 7, 5081. [Google Scholar] [CrossRef]
  141. Zeidner, J.F.; Karp, J.E. Clinical activity of alvocidib (flavopiridol) in acute myeloid leukemia. Leuk. Res. 2015, 39, 1312–1318. [Google Scholar] [CrossRef]
  142. Wiernik, P.H. Alvocidib (flavopiridol) for the treatment of chronic lymphocytic leukemia. Expert Opin. Investig. Drugs 2016, 25, 729–734. [Google Scholar] [CrossRef]
  143. Yang, X.; Zhao, X.; Phelps, M.A.; Piao, L.; Rozewski, D.M.; Liu, Q.; Lee, L.J.; Marcucci, G.; Grever, M.R.; Byrd, J.C.; et al. A novel liposomal formulation of flavopiridol. Int. J. Pharm. 2009, 365, 170–174. [Google Scholar] [CrossRef]
  144. Chen, K.T.; Militao, G.G.; Anantha, M.; Witzigmann, D.; Leung, A.W.; Bally, M.B. Development and characterization of a novel flavopiridol formulation for treatment of acute myeloid leukemia. J. Control. Release 2021, 333, 246–257. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The chemical structure of flavopiridol. A, B, C, and D represent the different aromatic rings present in the structure. The encircled regions mark the functional groups essential to flavopiridol’s functional activity.
Figure 1. The chemical structure of flavopiridol. A, B, C, and D represent the different aromatic rings present in the structure. The encircled regions mark the functional groups essential to flavopiridol’s functional activity.
Molecules 28 07530 g001
Figure 2. Flavopiridol controls cell cycle progression by targeting different cyclin and CDK proteins of the G1 and G2 phases. The red arrows represent the effect of flavopiridol on the expression, and the arrow directions indicate the increase or decrease in the expression of these proteins.
Figure 2. Flavopiridol controls cell cycle progression by targeting different cyclin and CDK proteins of the G1 and G2 phases. The red arrows represent the effect of flavopiridol on the expression, and the arrow directions indicate the increase or decrease in the expression of these proteins.
Molecules 28 07530 g002
Figure 3. An illustration of the apoptosis induced by flavopiridol. Flavopiridol promotes the apoptosis of tumor cells by targeting different proteins involved in the intrinsic and extrinsic apoptotic pathways. The red arrows represent the effect of flavopiridol on the expression, and the arrow directions indicate the increase or decrease in the expression of the respective proteins.
Figure 3. An illustration of the apoptosis induced by flavopiridol. Flavopiridol promotes the apoptosis of tumor cells by targeting different proteins involved in the intrinsic and extrinsic apoptotic pathways. The red arrows represent the effect of flavopiridol on the expression, and the arrow directions indicate the increase or decrease in the expression of the respective proteins.
Molecules 28 07530 g003
Figure 4. Schematic representation of flavopiridol targeting the regulatory molecular players of tumor metastasis. Flavopiridol mediates the inhibition of tumor metastasis by decreasing MMP proteins’ secretion via inhibition of NF-κB activity, increasing expression of E-cadherin and P-cadherin, and inhibition of wnt signaling through the inhibition of GSK3β. The red arrows represent the actions performed by flavopiridol, whereas the arrow directions indicate the increase or decrease in expression.
Figure 4. Schematic representation of flavopiridol targeting the regulatory molecular players of tumor metastasis. Flavopiridol mediates the inhibition of tumor metastasis by decreasing MMP proteins’ secretion via inhibition of NF-κB activity, increasing expression of E-cadherin and P-cadherin, and inhibition of wnt signaling through the inhibition of GSK3β. The red arrows represent the actions performed by flavopiridol, whereas the arrow directions indicate the increase or decrease in expression.
Molecules 28 07530 g004
Figure 5. Modulation of molecular targets contributing to tumor angiogenesis by flavopiridol. Flavopiridol inhibits the formation of new blood vessels by targeting different signaling pathways, including decreasing VEGF secretion, suppressing MAPK pathways by inhibiting c-Jun and c-Fos transcription factors, and decreasing the secretion of MMP proteins. The red arrows here represent the actions performed by flavopiridol, whereas the arrow directions indicate the increase or decrease in expression.
Figure 5. Modulation of molecular targets contributing to tumor angiogenesis by flavopiridol. Flavopiridol inhibits the formation of new blood vessels by targeting different signaling pathways, including decreasing VEGF secretion, suppressing MAPK pathways by inhibiting c-Jun and c-Fos transcription factors, and decreasing the secretion of MMP proteins. The red arrows here represent the actions performed by flavopiridol, whereas the arrow directions indicate the increase or decrease in expression.
Molecules 28 07530 g005
Figure 6. Schematic representation of the anti-inflammatory activity of flavopiridol expression. Flavopiridol exerts its anti-inflammatory effects by targeting proteins of different cell signaling pathways, such as NF-κB, JAK-STAT, MAPK, PI-3K, and PKC. The red arrows here represent the actions performed by flavopiridol, whereas the arrow directions indicate the increase or decrease in expression.
Figure 6. Schematic representation of the anti-inflammatory activity of flavopiridol expression. Flavopiridol exerts its anti-inflammatory effects by targeting proteins of different cell signaling pathways, such as NF-κB, JAK-STAT, MAPK, PI-3K, and PKC. The red arrows here represent the actions performed by flavopiridol, whereas the arrow directions indicate the increase or decrease in expression.
Molecules 28 07530 g006
Table 1. Description of the therapeutic effects of flavopiridol and the proposed mechanisms of action.
Table 1. Description of the therapeutic effects of flavopiridol and the proposed mechanisms of action.
S. No.TherapeuticsDiseasesMechanismsDoseRouteExperimental ModelsRefs.
1Anti-AlzheimerAlzheimerRescue from memory impairment and cell cycle reactivation caused by Aβ1-42 oligomers; Aβ-treated mice had improved long-term memory response 0.5–1 mg/kgi.p.CD1 mice[119]
2NeuroprotectiveIschaemic strokeInhibits the phosphorylation of Rb; increased levels of E2F1; inhibits CDK activation and prevents CA1 neuronal cell death500 µmol/L
500 µM
i.c.v.Wistar rats[120,121]
Spinal cord injuryReduces expression of cyclin D1, pRb, CDK4, E2F1, and PCNA; increases expression of endogenous CDK inhibitor p27; reduces levels of galactin-3 and Iba-1, leading to decreased number of Iba-1+ microglial cells; increases CC1+ oligodendrocytes and white matter myelinated area; abrogates RNA Pol II phosphorylation and induction to promote neuronal survival1 mg/kgi.p.Male Sprague-Dawley rats[122,123]
Traumatic brain injuryIncreases neuronal survival post DNA damage and inactivated astroglial proliferation, microglial activation, and scar formation by blocking cell cycle progression proteins250 µMi.c.v.Male Sprague-Dawley rats[124]
3Anti-vasoproliferativeIn-stent restenosisAnti-proliferative effects were observed in human coronary artery smooth muscle cells (HCASMC) by cell cycle arrest at G1/S and G2/M phases; increases levels of p21, p27, and p53; abrogation of Rb hyperphosphorylation; reduces apoptosis; prevents neointima formation0.1 µM, 25 mg/mL-HCASMC, human coronary artery endothelial cells, rat[125]
4Anti-hepatitisConcavalin A-induced hepatitisInhibits ConA-induced hepatitis and neutrophil infiltration; suppresses TNF-α-induced leukocyte–endothelial cell interaction; abrogates the levels of ICAM-1, VCAM-1, E-selectin, and NF-κB by inhibition of CDK9 activity44 ngi.v.C57BL/6 male mice[103]
5Anti-viralAcquired immunodeficiency syndromeInhibits the phosphorylation induced by P-TEFb at the C-terminus region of RNA Pol II large subunit; abrogates Tat transactivation and HIV-1 viral replication6–12 nM-Transfecting 293T cells with the HIV-1HXB2 provirus, HIV-1NL4–3 viral particles, Jurkat cells[26]
Human-immunodeficiency-virus-associated nephropathyInhibition of HIV-1 transcript levels in infected glomerular visceral epithelial cells; improved nephropathy in mouse model2.5 mg/kgi.p.HIV-1 NL4-3 transgenic mouse model[116]
Herpes simplex virus 1 infectionInhibition of P-TEFb/CDK9 complex by blocking the phosphorylation at serine-2 residue on the C-terminus region of RNA Pol II; suppresses the replication of HSV-1 and HSV-2 through inhibition of mRNA transcription450 nM
30 mg
-HeLa cells, Wistar rats, BALB/c mice[27,117]
Human cytomegalovirus infectionSuppresses the replication of HCMV through inhibition of mRNA transcription by CDK9 inhibition1–5 µM-A549, Vero cells[27]
Human adenovirus infectionSuppresses the replication of HAdV5 and HAdV53 through inhibition of mRNA transcription by CDK9 inhibition; decreases expression of an early gene of adenovirus E1A1–10 µM-A549, Vero cells[27]
Influenza A virus infectionSuppresses the replication of H1N1, H3N2, and H7N9 through inhibition of mRNA transcription by CDK9 inhibition0.59 µM
0.70 µM
0.24 µM
-A549 cells[28]
6Anti-fungalFungal keratitisDownregulation of IL-1β, IL-6, and TNF-α expression and induction of IL-10 expression; increases expression of LC3, Beclin-1, and Atg7 proteins; promotes the phagocytosis of RAW264.7 cells; suppresses biofilm formation, growth, and attachment of Aspergillus fumigatus; decreases inflammation in fungal keratitis disease by inducing autophagy5 µMs.c.iRAW 264.7 cells, C57BL/6 female mice[29]
AspergillosisActs as a non-competitive inhibitor of UDP-galactopyranose mutase (UGM) to treat Aspergillus fumigatus infection200 µM-AfUGM[118]
7Anti-leishmanialLeishmaniasisInhibition of CRK3 kinase; inhibition of in vitro growth of Leishmania Mexicana promastigotes; suppresses cell cycle progression at G2 and G2/M phase2.5 µM-Leishmania Mexicana promastigotes[126]
8Anti-parasiticToxoplasma gondii infectionInhibition of TgCRK9 kinase activity led to the abrogation of RNA pol II dependent transcription elongation; inhibition of parasite multiplication and proliferation4 nM, 8 nM-HFF cells, Toxoplasma gondii culture[127]
9Anti-malarialMalariaAbrogates the activity of PfPK5 kinase in Plasmodium falciparum; inhibition of DNA synthesis0.06 µM, 2 µM-Red blood cells, Plasmodium falciparum culture[128]
10Anti-diabeticDiabetesInhibition of glycogen phosphorylase a and b enzymes15.5 µM-Rabbit skeletal muscle, A549 cells[129]
11Anti-arthritisOsteoarthritisAbolishes the activation of iNOS by IL-1β; inhibits a broad range of inflammatory mediators; inhibits the induction of MMP1,3, 9, and 13; protects the cartilage from the harmful effects of pro-inflammatory cytokines300 nM-Human chondrocytes, human cartilage explants[109]
Table 2. An overview of CDK-inhibitory activity by flavopiridol-based derivatives.
Table 2. An overview of CDK-inhibitory activity by flavopiridol-based derivatives.
DerivativesIC50 (in µM)Refs.
CDKGSK3β
12467910
14,6-Dihydroxy-2-[1-[4-(4-methyl-piperazin-1-yl)-phenyl]-meth-(E)-ylidene]-7-(1-methyl-piperidin-4-yl)-benzofuran-3-one0.802.981.92----1.75[130]
24-[4,6-Dihydroxy-7-(1-methyl-piperidin-4-yl)-3-oxo-3Hbenzofuran-(2E)-ylidenemethyl] benzenesulfonamide0.0090.031.87----3.7
34,6-Dihydroxy-7-(1-methyl-piperidin-4-yl)-2-[1-(4-nitrophenyl)-meth-(E)-ylidene]-benzofuran-3-one0.060.312.21----4.8
42-[1-(2-Chloro-phenyl)-meth-(E)-ylidene]-4,6-dihydroxy-7-(1-methyl-piperidin-4-yl)-benzofuran-3-one0.603.9725.25----10
54,6-Dihydroxy-7-(1-methyl-piperidin-4-yl)-2-[1-phenylmeth-(E)-ylidene]-benzofuran-3-one0.111.284.41----4.2
R1-N-[2-(2-Chlorophenyl)-5,7-dihydroxy-4-oxo-4Hchromen-8-yl]-R2[134]
6R2 = benzamide-91------
7R2 = 4-methylbenzamide-90------
8R2 = 4-methoxybenzamide-54------
9R1 = 3,5-Dichloro; R2 = 2- hydroxyl benzene sulfonamide-94------
R-5,7-dihydroxy-8-(3-hydroxy-1-methyl-4-piperidinyl)-4H-1-benzopyran-4-one[131]
10R = (±)-(3SR,4RS)-2-(Ethylthio)0.463.932.06-----
11R = (3S,4R)-2-[(2-Chlorophenyl)thio]0.112.1016.2-----
12R = (3S,4R)-2-(2-Chlorophenoxy)0.132.116.15-----
13R = (±)-(3SR,4RS)-2-(Phenylthio)0.446.594.10-----
14R = (±)-(3SR,4RS)-2-(tert-Butylthio)0.081.072.07-----
15R = (±)-(3SR,4RS)-2-[(4,6-Dimethylpyrimidin-2-yl)thio]6.4040.482.5-----
16R = (±)-(3SR,4RS)-2-(Phenylamino)16.3>25>25-----
17R = (±)-(3SR,4RS)-2-N-Piperidyl2.509.693.70-----
cis-5,7-dihydroxy-2-(R1)-8-[R2-piperidinyl]-1-benzopyran-4-one[34]
18R1 = 2-chlorophenyl; R2 = 4-(3-one-1-methyl)10-8-----
19R1 = 2-chlorophenyl; R2 = cis-8-2-hydroxycyclohexyl12-31-----
20R1 = 2-chlorophenyl; R2 = 4-(3-en)1.1-0.8-----
21R1 = 3-chlorophenyl; R2 = 4-(3-en)1.2-2.4-----
22R1 = 4-chlorophenyl; R2 = 4-(3-en)1.7-0.55-----
23R1 = 2-florophenyl; R2 = 4-(3-en)2.3-1.8-----
24R1 = 2-bromophenyl; R2 = 4-(3-en)0.98-0.65-----
25R1 = 2-florophenyl; R2 = 4-(3-en)--2.5-----
26R1 = 2-phenyl; R2 = 4-(3-en)--1.0-----
27R1 = 2, 4-dichlorophenyl; R2 = 4-(3-en)1.7-1.2-----
28R1 = 4-pyridyl; R2 = 4-(3-en)--0.8-----
29R1 = cyclohexyl; R2 = 4-(3-en)--7-----
302-(2-chlorophenyl)-5,7-dihydroxy-8-((2R,3S)-2-(hydroxymethyl)-1-methylpyrrolidin-3-yl)-4H-chromen-4-one hydrochloride (P-276-00)0.0790.2240.0630.3962.8700.020-2.771[132]
312-(2-Chloro-4-(trifluoromethyl)phenyl)-5,7-dihydroxy-8-((2R,3S)-2-(hydroxymethyl)-1-methylpyrrolidin-3-yl)-4H-1-benzopyran-4-one (Voruciclib)0.0054-0.00390.0029-0.0017--[135]
322-(2,6-dichlorophenyl)-5,7-dihydroxy-8-[(3S,4R)-3-hydroxy-1-methylpiperidin-4-yl]chromen-4-one (IIIM-290)0.00490.01550.02250.0450.7110.0019--[23]
332-(4-((1H-benzo[d]imidazol-2-yl)thio)phenyl)-5,7-dihydroxy-8-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4H-chromen-4-one-0.162---0.008-0.117[25]
345,7-Dihydroxy-8-(1-methyl-1,2,3,6-tetrahydro pyridin-4-yl)-2-(4-((1-methyl-1H-benzo[d] imidazol-2-yl)thio)phenyl)-4Hchromen-4-one-0.349---0.014-0.160
355,7-Dihydroxy-8-(1-methyl-1,2,3,6-tetrahydro pyridin-4-yl)-2-(4-((5-phenyl-1H-imidazol-2-yl)thio) phenyl)-4H-chromen-4-one-0.469---0.009-0.356
362-(4-((1H-benzo[d]imidazol-2-yl)thio)-2-chloro phenyl)-5,7-dihydroxy-8-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4H-chromen-4-one-1.556---0.015-0.216
372-(2-Chloro-4-((1-methyl-1H-benzo[d] imidazol-2-yl)thio) phenyl)-5,7-dihydroxy-8-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4H-chromen-4-one-1.725---0.0640.1490.059
2-R-5,7-dihydroxy-8[(3S,4R)-3-hydroxy-1-methylpiperidin-4-yl]chromen-4-one[133]
38R = phenyl-0.196---0.009--
39R = 3-chlorophenyl-0.164---0.004--
40R = 4-chlorophenyl-0.287---0.012--
41R = 2-fluorophenyl0.1230.356-->100.003->1
42R = 4-fluorophenyl-0.129---0.002--
43R = 4-bromophenyl-0.223---0.005--
44R = 4-tert-butylphenyl-0.567---0.019--
45R = 4-trifluoromethylphenyl-0.302---0.019--
46R = 4-hydroxyphenyl-0.196---0.010--
47R = 2-pyridyl-0.886---0.011--
48R = 3-pyridyl-0.247---0.005--
49R = 4-pyridyl-0.208---0.006--
50R = 2-chloro-3-pyridyl-0.314---0.013--
51R = 5-methylisoxazole-0.238---0.020--
52R = 3-vinylphenyl-0.130---0.010--
53R = 4-vinylphenyl-0.206---0.010--
54R = 4-fluorophenyl-0.208---0.006--
55R = 2-bromophenyl-0.639---0.005--
56R = 3-pyridyl-1.023---0.012--
Table 3. Summary of the anti-cancerous activity of flavopiridol-based derivatives.
Table 3. Summary of the anti-cancerous activity of flavopiridol-based derivatives.
S. No.DerivativesExperimental ModelsDose (IC50) in µMRefs.
cis-5,7-dihydroxy-2-(R)-8-[4-(3-en)-piperidinyl]-1-benzopyran-4-one[34]
1R = 2-chlorophenylMCF-70.75
2R = 3-chlorophenylMCF-71
3R = 4-chlorophenylMCF-73
4R = 2-florophenylMCF-71
5R = 2-chlorophenylMCF-71
6R = 2-bromophenylMCF-71.5
7(3S,4R)-2-[(2-Chlorophenyl)thio]-5,7-dihydroxy-8-(3-hydroxy-1-methyl-4-piperidinyl)-4H-1-benzopyran-4-onePC3, Mia PaCa-2, HCT116, A27800.02, 0.03, 0.21, 0.87[131]
84,6-Dihydroxy-7-(1-methyl-piperidin-4-yl)-2-[1-phenylmeth-(E)-ylidene]-benzofuran-3-oneHCT-116>50[130]
92-[1-(2-Chloro-phenyl)-meth-(E)-ylidene]-4,6-dihydroxy-7-(1-methyl-piperidin-4-yl)-benzofuran-3-oneHCT-11620.1
104,6-Dihydroxy-7-(1-methyl-piperidin-4-yl)-2-[1-(4-nitrophenyl)-meth-(E)-ylidene]-benzofuran-3-oneHCT-11635.6
114-[4,6-Dihydroxy-7-(1-methyl-piperidin-4-yl)-3-oxo-3Hbenzofuran-(2E)-ylidenemethyl]-benzene sulfonamideHCT-116>50
124,6-Dihydroxy-2-[1-[4-(4-methyl-piperazin-1-yl)-phenyl]-meth-(E)-ylidene]-7-(1-methyl-piperidin-4-yl)-benzofuran-3-oneHCT-11624.8
R1-N-[2-(2-Chlorophenyl)-5,7-dihydroxy-4-oxo-4Hchromen-8-yl]-R2[134]
13R2 = benzamideMCF-78.5
14R2 = 4-methylbenzamideMCF-79.7
15R2 = 4-methoxybenzamideMCF-713
16R1 = 3,5-Dichloro; R2 = 2-hydroxyl benzene sulfonamideID-8, MCF-724, 17
172-(2-chlorophenyl)-5,7-dihydroxy-8-((2R,3S)-2-(hydroxymethyl)-1-methylpyrrolidin-3-yl)-4H-chromen-4-one hydrochloride (P-276-00)HCT-116, T-24, U2OS, SiHa, MCF-7, PC-3, HT-29, Colo-205, Caco-2, HL-60, SW-480, H-460, MRC-5, WI-380.31, 0.39, 0.4, 0.42, 0.52, 0.56, 0.6, 0.65, 0.65, 0.75, 0.76, 0.8, 11.5, 16.5[132]
182-(2-Chloro-4-(trifluoromethyl)phenyl)-5,7-dihydroxy-8-((2R,3S)-2-(hydroxymethyl)-1-methylpyrrolidin-3-yl)-4H-1-benzopyran-4-one (Voruciclib)RIVA56.3%[135]
192-(2,6-dichlorophenyl)-5,7-dihydroxy-8-[(3S,4R)-3-hydroxy-1-methylpiperidin-4-yl]chromen-4-one (IIIM-290)HL60, MOLT-4, MIAPaCa-2, Panc-1, PC-3, DU145, MCF-7, MDAMB-231, MDAMB-468, BT-549, T47D, Caco-2, SW630, Colo-205, HCT116, A549, NCIH322, NCIH522, HOP62, HOP92, NCIH-226, 786-O, A431, LOXIMVI, OVCAR-3, OVCAR-4, OVCAR-5, mouse adenocarcinoma, HGF, fR2, HEK2930.9, 0.5, 1, 4, 6, 5, 4, 4, 4, 5, 6, 7, 0.3, 7, 5, 4, 2, 5, 7, 3, 4, 6, 8, 4, 8, 9, 7, 1.2, 18, 19, 22[23]
202-(4-((1H-benzo[d]imidazol-2-yl)thio)phenyl)-5,7-dihydroxy-8-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4H-chromen-4-oneNCI-N87, K562, SKBR3, HCT116, SKOV3, PC3, MiaPaCa-20.183, 0.194, 0.254, 0.293, 0.595, 0.742, 0.852[25]
215,7-Dihydroxy-8-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-2-(4-((1-methyl-1H-benzo[d]imidazol-2-yl)thio)phenyl)-4Hchromen-4-oneHCT116, SKBR3, SKOV3, K562, SKBR3, MiaPaCa-2, NCI-N870.173, 0.243, 0.295, 0.300, 0.352, 0.448, 4.796
225,7-Dihydroxy-8-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-2-(4-((5-phenyl-1H-imidazol-2-yl)thio)phenyl)-4H-chromen-4-oneHCT116, SKBR3, PC3, SKOV3, K562, MiaPaCa-2, NCI-N870.219, 0.249, 0.276, 0.344, 0.345, 0.361, 0.391
232-(4-((1H-benzo[d]imidazol-2-yl)thio)-2-chlorophenyl)-5,7-dihydroxy-8-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4H-chromen-4-oneNCI-N87, K562, SKBR3, HCT116, PC3, SKOV3, MiaPaCa-20.012, 0.036, 0.066, 0.170, 0.326, 0.333, 0.361
242-(2-Chloro-4-((1-methyl-1H benzo[d]imidazol-2-yl)thio) phenyl)-5,7-dihydroxy-8-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4H-chromen-4-oneNCI-N87, K562, MiaPaCa-2, SKBR3, PC3, SKOV3, HCT1160.049, 0.051, 0.053, 0.054, 0.085, 0.094, 0.181
2-R-5,7-dihydroxy-8[(3S,4R)-3-hydroxy-1-methylpiperidin-4-yl]chromen-4-one[133]
25R = phenylHeLa0.190
26R = 3-chlorophenylHeLa0.170
27R = 4-chlorophenylHeLa0.200
28R = 2-fluorophenylHeLa0.274
29R = 4-fluorophenylHeLa0.200
30R = 4-bromophenylHeLa0.280
31R = 4-tert-butylphenylHeLa0.660
32R = 4-trifluoromethylphenylHeLa1.200
33R = 4-hydroxyphenylHeLa20.920
34R = 2-pyridylHeLa1.490
35R = 3-pyridylHeLa3.300
36R = 4-pyridylHeLa3.350
37R = 2-chloro-3-pyridylHeLa0.177
38R = 5-methylisoxazoleHeLa0.645
39R = 3-vinylphenylHeLa0.195
40R = 4-vinylphenylHeLa0.219
41R = 4-fluorophenylHeLa0.266
42R = 2-bromophenylHeLa0.331
43R = 3-pyridylHeLa1.440
Table 4. Various clinical trials are reported for the inhibition of tumor development and metastasis.
Table 4. Various clinical trials are reported for the inhibition of tumor development and metastasis.
Clinical TrialCancerPhaseStatusSample SizeTreatment
NCT00003256Recurrent prostate cancerPhase IICompleted40FP
NCT00007917Adult solid tumorsPhase ICompleted58FP and gemcitabine hydrochloride
NCT00016939Renal cell carcinomaPhase IICompleted35FP
NCT00070239Hematopoietic, adult solid tumors, and lymphoid cancerPhase ITerminated100FP and fludeoxyglucose F 18
NCT00006245Metastatic esophageal cancerPhase IICompleted37FP and paclitaxel
NCT00006485Adult solid tumorsPhase ICompleted50FP and irinotecan hydrochloride
NCT00112684Metastatic solid tumorsPhase ITerminated25FP
NCT00324480Adult solid tumorsPhase ICompleted60FP and vorinostat
NCT00020332Metastatic breast cancerPhase I/IICompleted49FP and docetaxel
NCT00331682Recurrent pancreatic cancer and pancreatic adenocarcinomaPhase IICompleted10FP and docetaxel
NCT00080990Adult solid tumorsPhase I Completed46FP, fluorouracil, oxaliplatin, and leucovorin calcium
NCT00039455Metastatic breast cancerPhase ITerminated50FP and trastuzumab
NCT00087282Advanced liver cancerPhase IICompleted32FP and irinotecan hydrochloride
NCT00072436Adult solid tumorsPhase ICompleted 58FP and gemcitabine hydrochloride
NCT00045448Advanced solid tumorsPhase ICompleted 56FP and docetaxel
NCT00046917Adult solid tumorsPhase ICompleted13FP, irinotecan hydrochloride, and cisplatin
NCT00019344Adult solid tumors, lymphoma, prostate cancer, and small intestine cancerPhase ICompleted 36FP
NCT00047307Locally advanced and unresectable pancreatic cancerPhase ICompleted46FP, gemcitabine hydrochloride, and radiation therapy
NCT00023894Recurrent or persistent endometrial cancerPhase II Completed51FP
NCT00016185Advanced solid tumorsPhase ICompleted24FP and docetaxel
NCT00003690Breast cancer, melanoma, prostate cancer, and adult solid tumorsPhase I Completed48FP
NCT00042874Metastatic solid tumorsPhase ICompleted77FP, irinotecan hydrochloride, fluorouracil, and leucovorin calcium
NCT00003004Refractory or recurrent solid tumorsPhase I Completed73FP, cisplatin, and paclitaxel
NCT00079352Metastatic solid tumorsPhase ICompleted24FP, gemcitabine hydrochloride, and irinotecan hydrochloride
NCT00020189Metastatic head and neck cancer, ThromboembolismPhase IICompleted37FP, acetylsalicylic acid, and clopidogrel bisulfate
NCT00094978Small cell carcinoma, non-small cell lung cancer, esophageal neoplasms, and mesotheliomaPhase ITerminated23FP and depsipeptide
NCT00021073Unspecified adult solid tumorsPhase ICompleted90FP, leucovorin calcium, fluorouracil, and irinotecan hydrochloride
NCT00012181Recurrent childhood solid tumors, recurrent neuroblastoma, recurrent osteosarcoma, and recurrent retinoblastomaPhase ICompleted30FP
NCT00957905Recurrent or relapsed germ cell tumorsPhase II Completed36FP, leucovorin calcium, fluorouracil, and oxaliplatin
NCT00064285LeukemiaPhase ICompleted22FP and imatinib mesylate
NCT00991952Advanced stomach and gastroesophageal junction cancerPhase II Completed19FP and irinotecan hydrochloride
NCT00083122Advanced ovarian epithelial cancer and primary peritoneal cancerPhase IICompleted45FP and cisplatin
NCT00082784Recurrent or refractory indolent B-cell neoplasmsPhase ICompleted93FP and bortezomib
NCT00098579Metastatic or recurrent sarcomaPhase ICompleted36FP and doxorubicin hydrochloride
NCT03441555Relapsed or refractory acute myeloid leukemiaPhase ICompleted36FP and venetoclax
NCT00047203Relapsed or refractory multiple myelomaPhase II Completed35FP
NCT03298984Acute myeloid leukemiaPhase I Completed32FP, cytarabine, and daunorubicin
NCT03969420Acute myeloid leukemiaPhase II Terminated11FP and cytarabine
NCT03563560Acute myeloid leukemiaPhase I Completed10FP, cytarabine, mitoxantrone, and daunorubicin
NCT00005974Recurrent and metastatic soft tissue sarcomaPhase II Completed18FP
NCT02520011Acute myeloid leukemiaPhase II Terminated104FP, cytarabine, and mitoxantrone
NCT03593915Myelodysplastic syndromesPhase I/IITerminated20FP and decitabine or azacitidine
NCT00470197Relapsed or refractory acute leukemiaPhase ICompleted35FP, cytarabine, and mitoxantrone hydrochloride
NCT00112723Relapsed or refractory lymphoma and multiple myelomaPhase I/II Terminated 46FP
NCT00445341Relapsed mantle cell lymphoma and diffuse large B-cell lymphomaPhase I/IICompleted 28FP
NCT01349972Acute myeloid leukemiaPhase IICompleted172FP, mitoxantrone hydrochloride, cytarabine, and daunorubicin hydrochloride
NCT00795002Acute myeloid leukemiaPhase IICompleted78FP, mitoxantrone hydrochloride, and cytarabine
NCT00634244Relapsed or refractory acute myeloid leukemiaPhase IICompleted92FP, mitoxantrone hydrochloride, carboplatin, cytarabine, sirolimus, etoposide, and topotecan hydrochloride
NCT00003039LymphomaPhase IICompleted40FP
NCT00058240Chronic lymphocytic leukemia and lymphocytic lymphomaPhase I/IICompleted52FP
NCT00005971Metastatic malignant melanomaPhase IICompleted17FP
NCT00101231Relapsed or refractory acute myeloid leukemia, acute lymphoblastic leukemia, and chronic myelogenous leukemiaPhase ITerminated88FP
NCT00058227Lymphoproliferative disorders or mantle cell lymphomaPhase ICompleted37FP, fludarabine phosphate, and rituximab
NCT00098371Chronic lymphocytic leukemia and prolymphocytic leukemiaPhase IITerminated64FP
NCT00003620Chronic lymphocytic leukemiaPhase IICompleted37FP
NCT00464633Chronic lymphocytic leukemiaPhase IICompleted165FP
NCT00278330Relapsed or refractory acute leukemia, chronic myelogenous leukemia, and refractory anemiaPhase ICompleted24FP and vorinostat
NCT00735930Relapsed or refractory B-cell chronic lymphocytic leukemia and small lymphocytic lymphomaPhase ICompleted39FP and lenalidomide
NCT00377104B-cell chronic lymphocytic leukemia and small lymphocytic lymphomaPhase ITerminated24FP
NCT00016016Acute leukemiaPhase I/IICompleted53FP, cytarabine, and mitoxantrone hydrochloride
NCT01076556B-cell chronic lymphocytic leukemia and small lymphocytic lymphomaPhase ITerminated9FP, cyclophosphamide, and rituximab
NCT00407966Acute myeloid leukemiaPhase IICompleted45FP, cytarabine, and mitoxantrone hydrochloride
NCT00005074Relapsed or untreated mantle cell lymphomaPhase IICompleted33FP
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Joshi, H.; Tuli, H.S.; Ranjan, A.; Chauhan, A.; Haque, S.; Ramniwas, S.; Bhatia, G.K.; Kandari, D. The Pharmacological Implications of Flavopiridol: An Updated Overview. Molecules 2023, 28, 7530. https://doi.org/10.3390/molecules28227530

AMA Style

Joshi H, Tuli HS, Ranjan A, Chauhan A, Haque S, Ramniwas S, Bhatia GK, Kandari D. The Pharmacological Implications of Flavopiridol: An Updated Overview. Molecules. 2023; 28(22):7530. https://doi.org/10.3390/molecules28227530

Chicago/Turabian Style

Joshi, Hemant, Hardeep Singh Tuli, Anuj Ranjan, Abhishek Chauhan, Shafiul Haque, Seema Ramniwas, Gurpreet Kaur Bhatia, and Divya Kandari. 2023. "The Pharmacological Implications of Flavopiridol: An Updated Overview" Molecules 28, no. 22: 7530. https://doi.org/10.3390/molecules28227530

Article Metrics

Back to TopTop