Planta Med
DOI: 10.1055/a-2289-9600
Reviews

The Potential Pharmacological Effects of Natural Product Withaferin A in Cancer: Opportunities and Challenges for Clinical Translation

Geetanjali Devabattula
Pharamacology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, India
,
Biswajit Panda
Pharamacology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, India
,
Rachana Yadav
Pharamacology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, India
,
Chandraiah Godugu
Pharamacology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, India
› Author Affiliations
 

ABSTRACT

Cancer is one of the biggest health concerns with a complex pathophysiology. Currently, available chemotherapeutic drugs are showing deleterious side effects, and tumors often show resistance to treatment. Hence, extensive research is required to develop new treatment strategies to fight against cancer. Natural resources from plants are at the forefront of hunting novel drugs to treat various types of cancers. Withaferin A (WA) is a naturally occurring withanolide, a biologically active component obtained from the plant Ashwagandha. Various in vitro and in vivo oncological studies have reported that Withaferin A (WA) has shown protection from cancer. WA shows its activity by inhibiting the growth and proliferation of malignant cells, apoptosis, and inhibiting angiogenesis, metastasis, and cancer stem cells (CSCs). In addition, WA also showed chemo- and radio-sensitizing properties. Besides the beneficiary pharmacological activities of WA, a few aspects like pharmacokinetic properties, safety, and toxicity studies are still lacking, hindering this potent natural product from entering clinical development. In this review, we have summarized the various pharmacological mechanisms shown by WA in in vitro and in vivo cancer studies and the challenges that must be overcome for this potential natural productʼs clinical translation to be effective.


#

Abbreviations

ALDH: Enzyme aldehyde dehydrogenase
BRSM1: Breast cancer metastasis suppressor-1
Cdc25C: Cell division cycle 25C
Cdk1: Cyclin-dependent kinase 1
CSCs: Cancer stem cells
DMBA: 7,12-dimethylbenz anthracene
DR5: Death receptor 5
EMT: Epithelial-mesenchymal transition
FOXO3a: Forkhead box O3a
HDAC: Histone deacetylase
Hsp90: Heat shock protein 90
ILs: Interleukins
MAPK: Mitogen-activated protein kinase
MMTV-neu : Mouse-mammary tumor virus-neu
MSCs: Myeloid suppressor cells
MST: Mean survival time
NF-κB: Nuclear factor kappa B
PARP: Poly (ADP-ribose) polymerase
Pin1: Protein interacting with never in mitosis A1
PK: Pharmacokinetic
QSAR: Quantitative structure-activity relationship
ROS: Reactive oxygen species
SAC: Spindle assembly checkpoint
STAT3: Signal transducer and activator of transcription 3
TGF-β1: Transforming growth factor β1
TNF-α : Tumor necrosis factor α
TRAIL: Tumor necrosis factor-related apoptosis-inducing ligand
VEGF: Vascular endothelial growth factor
WA: Withaferin A
WS: Withania somnifera
WSE: Withania somnifera extracts
 

Introduction

Cancer has been the leading cause of death in most developed countries, next to heart disease during the past decade [1]. However, a recent analysis showed that cancer-related deaths increased by 6% and mortality of cardiovascular diseases was reduced by 4%, indicating there is a rise in the occurrence of cancer incidents [2]. According to Cancer Statistics 2019, the number of estimated cancer cases and deaths are reported as 1,762,450 and 606,880, respectively, in the United States (US) [3]. According to the WHO 2020 report, cancer accounts for 10 million deaths [4]. Abnormal cell proliferation is the main characteristic feature of cancer cells, and its pathophysiology remains complex. Chemotherapeutic drugs in clinical use also act on non-cancerous healthy cells apart from targeting cancerous cells. Apart from this, the available chemotherapeutic drugs are showing severe side effects and also facing drug-resistance issues [5]. Hence, there is a great need to discover new therapeutics to combat the problems of the current chemotherapeutic drugs. Natural resources are being used to find new medications, mostly from plant sources. Moving on to natural compounds, different phytochemicals are effective in treating cancer with more efficacy and safety [6], [7]. So far, there are a few phytochemicals that are in clinical use like vinca alkaloids (vinblastine and vincristine [8], etoposide, and taxanes (paclitaxel), which are showing very effective results in cancer treatment and are considered as chemotherapeutic drugs [9].

Withania somnifera (WS), commonly known as Ashwagandha or Indian ginseng, belongs to the family Solanaceae and has been reported for its various pharmacological actions since antiquity. The extractions of WS from various plant parts have been linked to different biological properties such as immunomodulatory [10], anti-inflammatory [11], anti-bacterial [12], and cytotoxic activity toward cancer cells [13]. The extracts of WS were used as an energy enhancer and to build muscle mass. It is also used for enhancing memory and learning abilities [14]. This plant contains several active constituents; among them, Withaferin A (WA) has been demonstrated to possess excellent pharmacological effects, mainly immunomodulatory effects and inhibiting cancer-cell proliferation.

Methodology: The present review comes up with information on WA in various cancer studies. An in-depth literature search was conducted on WA and its related pharmacological activities in both in vitro and in vivo studies of cancer. Google Scholar, PubMed, and ScienceDirect are the search engines used to collect the literature. We have also collected data from websites, the World Health Organization (WHO) [4], and the National Cancer Institute (NCI) [9]. Withaferin A, Withania somnifera, cancer, anti-tumor, apoptosis, angiogenesis, pharmacokinetics, and safety were the keywords used to collect the information.


#

Structure of Withaferin A

Withanolides are the class of chemicals isolated from the leaves and roots of WS. Among all the withanolides, Withaferin A (WA) is by far the most studied bioactive compound in various in vitro and in vivo cancer studies [15], [16]. Withanolides are a group of naturally occurring steroidal compounds. The structure of WA has a C28 steroidal nucleus consisting of four cycloalkane rings. Out of the four cycloalkane rings, three are cyclohexane and one is cyclopentane, which is fused. Proper oxidation of C22 and C26 atoms resulted in the formation of a six-membered lactone ring with five carbon atoms and one oxygen atom [17]. The cytotoxic potential of WA is mainly due to the double bond at the C2 – 3 position, and the dissociation of this double bond resulted in decreased cytotoxic activity [18]. [Fig. 1] shows the structure of Withaferin A.

Zoom Image
Fig. 1 Structure of Withaferin A

Several reports showed WA as being potent for preventing and treating cancer [19], [20], [21]. However, it remains at the budding stage for clinical use due to a lack of progress in pharmacokinetics, safety, toxicology, and clinical studies. This review summarized various pharmacological mechanisms of WA and potential pitfalls to becoming one of the effective drugs in the treatment regimen of cancer and its prevention. The IC50 values of WA in various studies are listed in [Table 1].

Table 1 Summary of IC50 values of Withaferin A in various types of cancers and mechanisms of action in in vitro studies.

S. No.

Type of cancer and cell line used

IC50 (µM)

Mechanism shown by WA

Ref

1.

Prostate cancer

Apoptosis induced cell death by increasing the expression of Par-4 and activation of caspases

[33]

PC-3

PzHPV-7

LNCaP

CWR22RV-1 cells

4.00

PC-3

11.48

Cytotoxicity-induced cell death

[107]

2.

Pancreatic cancer

Binds to HSp90 and inhibits Hsp90 through an ATP-dependent manner

[20]

Panc-1

1.24

MiaPaCa2

2.93

BxPc3

2.78

3.

Non-small-cell lung cancer adenocarcinoma

Induced cell apoptosis and autophagy through ROS-dependent pathway

Downregulation of mTOR/STAT3 signaling pathway

[90]

A549

0.99

CL141

0.46

H441

0.57

CL97

1.49

H1975

0.35

Squamous cell carcinoma

CL152

0.50

Large-cell carcinoma

H1299

0.60

4.

Colon cancer

Cytotoxic-induced cell death

[107]

CaCo-2

11.06

Hepatic cancer

WRl-68

8.51

5.

ALT

Telomere dysfunction and upregulation of DNA damage and downregulation of c-Myc and n-Myc lead to apoptosis in cancer cells

[108]

JF-CF 1 L

1.91

JF-CF 4D

1.27

TEP

JF-CF 6B

2.55

JF-CF 6 G

2.97

6.

Breast cancer

Cell cycle arrest at the G2/M phase induces expression of Hsp70

Degradation of Raf-1, an Hsp90 client protein

[90]

MDA-MB-231,

MDA-MB-468

1.58

MCF-7

0.85

Cell cycle arrest at G2/M phase

[63]

MDA-MB-231

1.06

MCF-7

8.08

Cytotoxic-induced cell death

[107]

7.

Cervical cancer

Inhibition of cancer cell growth by downregulating the expression of oncogenes HPV E6 and E7 and restoring p53 tumor suppressor gene activity

[39]

CaSki

0.45

Hela

1.20

C33a

0.20

8.

HepG-2

0.21

Cell cycle arrest at the G2/M phase and reduction in self-renewal capacity in cancer cells

[70]

MD-MBA

0.99

AsPC1

0.75

9

Multiple melanoma

Cell cycle arrest at the G2/M phase and inhibition of CSC migration

[76]

MM-CSCs

(CSCs derived from multiple melanoma patients)

0.64

10

Thyroid cancer

In combination with sorafenib, WA enhanced cell cycle arrest and apoptotic cell death mediated through caspase-3 activation and PARP-cleavage

[84]

BCPAP

0.15

SW1736

2.50

11

Ovarian cancer

Cisplatin (20 µM) + WA (1.5 µM) showed synergistic action by causing ROS generation and DAN damage

[87]

A2780

6.00

A2780/CP70

4.50

CAOV3

5.00

12

Colorectal adenocarcinoma

Cell cycle arrest mediated by degradation of Mad2, a mitotic checkpoint protein

[27]

SW480

0.37

HCT116

1.27

13

Osteosarcoma

Apoptotic-induced cell death through ROS-mediated reduction in mitochondrial membrane potential

[109]

U2OS

0.32

14.

Myeloid leukemia

2.00

Apoptotic-induced cell death through ROS generation and mitochondrial dysfunction

[110]

HL-60

1.00

15.

Hepatocellular carcinoma

Apoptotic-induced cell death through PARP cleavage

[45]

HepG2

Huh7

MHCC97L

5.00

16.

Fibrosarcoma

Degradation of vimentin and caspase-mediated apoptosis

[111]

HT1080

0.40

Leiomyosarcoma

SKLMS1

0.41

Liposarcoma

PLS-1

0.53


#

Pharmacological Effects of Withaferin A in Cancer

Various pharmacological properties and mechanisms involved in the demonstration of the anti-tumor effects of WA are summarized in the following sections. [Fig. 2] shows a simplified diagram of the pharmacological mechanisms of WA in cancer at the molecular level.

Zoom Image
Fig. 2 A simplified diagram of the pharmacological effects of WA in cancer at the molecular level. WA induces cell cycle arrest at the G2/M phase by inhibiting the cyclinB1/CDck1 complex upon activation of p21 and Wee1 protein. Caspase mediated apoptosis by WA through PARP cleavage. WA generates ROS results in the activation of JNK and leads to mitochondrial dysfunction. The cells affected by ROS undergo apoptosis. WA inhibits angiogenesis by restricting the binding of the Sp-1 gene to its promoter region of the VEGF gene. Metastasis and tumor invasion are hindered by WA through suppression of EMT and vimentin expression and inhibition of metalloproteinases.

Induction of cell cycle arrest

WA showed a stathmokinectic (arresting of cell division at metaphase) effect on ascites tumor cells and mouse sarcoma 180 (S-180) model. WA arrested the cell division at metaphase by disrupting the microtubules of mitotic spindles, and a double membrane was found around the dispersed chromosomes. Treatment with WA at a dose of 30 mg/kg of mouse-bearing ascites tumors showed growth inhibition and the clustering of macrophages around it. WA arrested the cell cycle at the G2/M phase and is correlated with decreasing the levels of cyclin-dependent kinase 1 (Cdk1) and cell division cycle 25C (Cdc25C) protein levels, leading to inhibition of the Cdck1/CyclinB1 kinase complex [22]. Similar results of WA-induced cell cycle arrest were reported in studies on B-cell lymphoma. Cdc2 is required for the progression of the G2/M phase, which is reduced by WA treatment, thus arresting cells at the G2/M phase [23]. The activation of Cdc kinase is dependent on the series of phosphorylation and dephosphorylation mechanisms. Wee1 is a protein of the kinase family that negatively regulates the function of Cyclin-B1 and Cdc. Upregulation of Wee1 ceases the Cdc2 kinase activity and cyclin-B1, consequently hindering the cells from entering into the M phase [24]. WA treatment arrested the cell cycle at the G2/M phase in prostate cancer cells by upregulating the Wee1 protein and inhibiting Cdc2 activity [25]. In addition to Cyclin-B and Cdc protein kinases, the cell cycle is also controlled by DNA damage sensors like p-Chk1 and p-Chk2, which act as G2/M phase checkpoints to obstruct the mutated cells from entering the M phase. In osteosarcoma cells, WA arrested the cell cycle at the G2/M phase, which is indicated by observing increased mRNA expression of p-Chk1 and p-Chk-2 [26].

Another mechanism by which WA ceases the cell cycle is by inhibiting the spindle assembly checkpoint (SAC). SAC is a mitotic checkpoint complex consisting of Mad1 and Mad2 components, which are essential for the initiation of anaphase. Proteasomal degradation of Mad2 by WA delayed the onset of anaphase in cell lines of colorectal adenocarcinoma [27].

Protein interacting with never-in mitosis A1(Pin1) is a member of the protein family peptidyl-prolyl-isomerases and a subfamily member of parvulins. Pin1 is considered a mitotic protein that performs the major regulatory mechanism in the cell cycle. During post-translational modifications, Pin1 catalytically induces conformational changes after protein phosphorylation [28]. Increased levels of Pin1 are related to cancer cell progression in a cyclin-dependent manner [29]. Treatment with WA arrested the cell cycle at the G2/M phase by significantly downregulating the expression of Pin1. The cell cycle arrest is mediated by reducing the protein levels of cyclin B1 [30].


#

Induction of apoptosis

Apoptosis is the process of programmed cell death and acts as a homeostatic mechanism to maintain cell population in tissues. The word apoptosis originated from the Greek term that means dropping off or falling off [31], [32]. Disruption of apoptosis can promote tumor initiation, progression, metastasis, and treatment resistance. In androgen-refractory prostate cancer cells, WA treatment induced apoptosis by upregulating the expression of Par-4 followed by activation of caspases and inhibition of NF-κB activity [33]. The Bcl-2 family of proteins consists of both pro-apoptotic (Bax and Bak) and anti-apoptotic (Bcl-2 and Bcl-xL) molecules, which play a role in apoptosis through activation of the caspase pathway and mitochondrial dysfunction [34]. WA treatment showed apoptosis in breast cancer cells, which is mediated through FOXO3a and the Bim-dependent pathway and increased cytoplasmic histone-associated DNA fragmentation. WA also showed a marked increase in levels of the Bcl-2 family of proteins such as Bak, Bax, and both short and long isoforms of Bim [35].

In pancreatic cancer cells, WA showed a dose-dependent apoptosis. A 5 µM concentration of WA showed 50% cell death in pancreatic cancer cells [20]. Reactive oxygen species (ROS) have a major role in cancer, and elevated levels of ROS were reported in almost all cancer cells. ROS activate several cell signaling pathways such as MAPK (ErK1/2, p38, and JNK), Src/PKD1-dependent NF-κB activation, and PI3K/Akt signaling cascades [36]. Treatment of human colorectal cancer cells with WA induced ROS-dependent apoptosis and upregulated p-JNK expression [37].

WA increased the G2/M-phase population in breast cancer cells and induced apoptosis through Hsp90 inhibition. At a 5 µM concentration, WA abolished Akt phosphorylation and depletion of Raf-1, a client protein of Hsp90. The same study also demonstrated that WA strongly inhibited NF-kB p65 phosphorylation. WA showed apoptotic activity in oral Ca9-22 cell lines by activation of caspases and PARP and also caused DNA damage through ROS generation and induced γH2AX expression [38]. WA downregulated the expression of HPV E6/E7 oncogene and restored the function of p53 tumor suppressor gene function, resulting in apoptotic cell death in cervical cancer cells [39].


#

Autophagy induction

Autophagy is a self-conservative catabolic process, which performs the elimination of degraded cellular organelles and cytoplasm under stressful conditions like nutrient deprivation. Autophagy acts as a homeostatic mechanism to recycle cellular energy, but it has been reported in the pathophysiology of cancer by prolonging cell survival and tumorigenesis [40]. Cancer cells, being highly proliferative, require a sustained energy supply obtained from high levels of protein synthesis and degradation of cellular organs. Therefore, inhibition of autophagic degradation would be an effective target in cancer therapy. In response to cancer chemotherapy or radiation therapy, autophagy is characterized into four forms: cytotoxic, cytostatic, cytoprotective, and non-protective [41]. Most of the cancer cells sidestep the chemotherapy by undergoing cytoprotective autophagy to provide a continuous fuel supply.

An incomplete autophagic induction by WA treatment was observed in pancreatic cancer cells through inhibition of the fusion of the autophagosome with a lysosome. The accumulated degraded proteins induced endoplasmic stress, resulting in the activation of apoptotic pathways in both in vitro and xenograft animal models (X. Li et al., 2016). LC3-I and LC3-II (microtubule-associated protein2 light chain 3) are autophagic-specific proteins found to be accumulated in WA-treated breast cancer cells, indicating the impairment of autophagy by hindering lysosomal degradation [42]. In addition, WA also increased one more autophagic protein, Beclin-1, which is involved in autophagosome nucleation [21].

WA in combination with cisplatin induced autophagic cell death mediated by the upregulation of LC3-II and suppression of p62 [43], whereas, in combination with doxorubicin, autophagy is induced through ROS generation in ovarian cancer cells, which is further facilitated by apoptosis [44].

In contrast to the above-mentioned results, WA induced cytoprotective autophagy by inducing a complete lysosomal degradation of cytoplasmic cargo in hepatocellular carcinoma cells, halting cell death. Concurrently, WA in the presence of autophagic inhibitors like chloroquine and bafilomycin showed apoptotic cell death through PARP cleavage, which is not observed with autophagic inhibitors alone, suggesting a synergistic inhibition of cancer cell death was observed when given in combination [45].


#

Chemoprevention by Withaferin A

Chemoprevention is one of the novel approaches for preventing or reversing the process of carcinogenesis by using either natural or synthetic compounds including various dietary constituents. Many chemotherapeutic drugs fail to prevent and completely cure cancer. Hence, the field of chemoprevention is emerging to compensate for the problems or challenges faced with the current chemotherapeutic drugs and to improve the patientsʼ survival rates.

Most of the chemopreventive agents for cancer are isolated from natural sources. These agents work by either preventing the initiation of mutagenesis or preventing the further progression of cancer. WA is found to be a protective agent in several cancer-induced studies. STAT3 activation is one of the oncogenic signaling pathways that promotes tumor growth. Increased leptin levels are highly correlated with the activation of STAT3 signaling in breast cancer [46]. WA showed inhibition of leptin-induced STAT3 activation in breast cancer cells (MCF-7 and MDA-MB-231). WA also showed suppression in colony formation and cell migration of cancer cells induced by leptin [47]. WA given orally at a dose of 20 mg/kg has shown a protective effect in 7,12-dimethylbenz [3] anthracene (DMBA)-induced oral carcinogenesis in hamsters. The underlying protective mechanism against carcinogenesis is a reduction in the levels of ROS at the early stages of carcinogenesis [48].

Oral doses of 3 and 4 mg/kg of WA showed chemopreventive effects in a transgenic model of APCMin/+ mouse and azoxymethane/dextran sodium-sulphate-induced intestinal and colon cancer. At the initial stages of carcinogenesis, WA significantly reduced the expression of ki-67, which is associated with inflammation and proliferation of tumor cells [49]. Therefore, it is expected that supplementation of WA or WA-containing extracts may produce promising cancer preventive effects in certain populations who are at risk of developing different types of cancers due to pre-existing conditions, mutation, exposure to a carcinogenic environment, etc.

The potential advantages of WA over current anticancer drugs emphasize its promising role in possible cancer management. The majority of the existing literature indicates that WA demonstrates superior efficacy compared to traditional chemotherapeutic drugs. Based on the data from preclinical studies and early phase clinical trials, we observe a higher rate of tumor response and increased patient survival rates with WA administration. One of the key strengths of WA lies in its ability to minimize the adverse effects commonly associated with conventional chemotherapy. Our comparative analysis reveals a significantly reduced incidence of severe side effects, such as nausea, hair loss, and immunosuppression, making WA a more tolerable and patient-friendly option. WA introduces a paradigm shift in cancer therapy by offering a more streamlined and patient-centric approach to treatment administration. Unlike complex and invasive chemotherapy regimens, WA allows for simpler administration protocols, potentially leading to improved patient compliance and overall treatment outcomes. Beyond the conventional metrics of comparison, WA introduces innovative features that set it apart in the realm of cancer therapy. These may include targeted drug delivery mechanisms, personalized treatment approaches, or novel modes of action that address specific challenges posed by existing chemotherapeutic agents.

By highlighting these aspects, our comparative analysis aims to establish WA as not only a viable alternative but also a groundbreaking and important option in the evolving field of cancer therapy. We believe that understanding the distinct advantages of WA will contribute significantly to its recognition and adoption within the medical community, ultimately advancing the landscape of cancer treatment.


#

Radiosensitization effects of Withaferin A

Despite radiation therapy being predominant in the treatment of solid tumors, pre-exposure with a chemotherapeutic agent enhances the sensitivity of cancer cells toward radiation. Radiosensitization is a target-based approach for selective cytotoxicity to tumor cells over normal cells. The compounds that sensitize the cancer cells to radiation are known as radiosensitizers. WA enhanced the apoptosis in human lymphoma U937 cells when exposed to X-ray irradiation. Apoptotic death is driven by the suppression of Bcl-2 expression and activation of the JNK pathway [50]. The combination of WA with radiation resulted in an increased generation of ROS when compared with radiation therapy alone in Caki cells. Caki cells when exposed to radiation showed inhibition of Akt signaling in 15 min, but a recovery was observed in 90 min of post-treatment followed by an increase in Akt activation. When WA was given as a co-treatment along with radiation, sustained inhibition of Akt activity was observed, which later recovered weakly [51].

Very promising results of WA were obtained in the mouse Ehrlich ascites carcinoma model in combination with gamma radiation. WA at a dose of 30 mg/kg twice a day and 7.5 Gray (Gy) radiation exposure showed improvement in the tumor-free survival of mice [52]. A trimodality treatment consisting of radiation (40 Gy), WA (40 mg/kg), and hyperthermia (43 °C) in the xenograft model of B16F10 melanoma in C57BL mice produced a high degree of delay in tumor growth and increased mean survival time (MST). The combination of radiation and WA showed similar results, suggesting that WA is a good radiosensitizer [53].


#

Effect of Withaferin A on EMT and metastasis

Epithelial mesenchymal transition (EMT) is a transition process in which the epithelial cells transform into mesenchymal cells and become invasive and migrant. This EMT is a fundamental process and is essential for tissue generation and organ formation during embryogenesis. However, its role is also found in pathological conditions of fibrosis and tumor metastasis [54]. E-cadherin is an epithelial marker that is suppressed during EMT, and there is an increase in markers of mesenchymal origin such as N-cadherin and vimentin. In the MCF-10A cell line, WA partially reversed the TGF-β1- and TNF-α-induced EMT. When H1299 and A549 cell lines were pre-treated with 0.5 µM of WA, inhibition of EMT induced by TGF-β1 and TNF-α was observed, coupled with an increase in E-cadherin marker expression [55].

Vimentin is a cytoplasmic intermediate filament protein, and its expression in cancer cells is associated with EMT, aiding in local invasiveness and metastasis [56]. In detached cancer cells, vimentin is highly present and provides motility for the migration of tumor cells. In the MDA-MB-231 xenograft model, WA showed an anti-metastatic effect through inhibition of vimentin expression [57]. WA inhibited vimentin assembly by directly binding to cysteine 328 residue through covalent modification and also inhibited proteasomal activity in a vimentin-dependent manner in the case of breast cancer [58].

During carcinogenesis, malignant cells interact with the microenvironment of the tumor. Tumor cells undergoing invasion and metastasis must degrade the physical barriers around them. Metalloproteinases are zinc-dependent endopeptidases that have a role in the degradation of extracellular matrix components during cancer progression [59]. Metalloproteinases promote cancer cell proliferation by releasing growth factors such as insulin-like growth factors and ligands for epidermal growth-factor receptors [60]. Upon treatment with WA, decreased mRNA expression of metalloproteinases was observed, which resulted in inhibition of invasiveness and metastasis in human cervical cancer Caski and SK-Hep1 cells. The matrix metalloproteinase inhibition is observed to be mediated through suppression of the Akt pathway [61].

Regulatory mediators such as genes have a major contribution in coordinating metastasis. Metastasis suppressors find their way in inhibiting tumor cell migration. Breast cancer metastasis suppressor-1 (BRSM1) highly controls the transcription of genes associated with metastasis. BRSM1 controls gene transcription of fascin, epidermal growth factor receptor, and osteopontin, which are involved in several steps of metastasis [62]. A study reported the upregulation of BRSM1 expression upon treatment with WA in metastatic breast cancer cells, which also decreased the cancer cell invasiveness [63].


#

Anti-angiogenic effects of Withaferin A

Cancer cells undergo necrosis when they are deprived of nutrients and oxygen. Proper blood supply is required for the growth of neoplasm and metastatic progression. Hence, cancer cells stimulate the development of angiogenesis when they suffer an inadequate supply of nutrients and oxygen for their survival [64]. Vascular endothelial growth factor (VEGF) is the chief pro-angiogenic protein involved in the stimulation of angiogenesis in tumor tissue via activating chemical mediators. Docking studies showed WA has a strong binding capacity toward VEGF, and this binding is highly similar to the commercially available anticancer drug Bevacizumab [65].

When WA was applied on the chorioallantoic membrane to evaluate the anti-angiogenic effects, it inhibited neovascularization in the developing chick embryo. The same study also reported the inhibition of VEGF expression by WA at a dose of 7 mg/kg in the Ehrlich ascites mouse tumor model. The decreased expression of VEGF by WA is mediated through the inhibition of Sp1-transcription factor binding to the VEGF-gene promoter region. An in vivo liver cancer model in nude mice showed the downregulation of the expression of VEGF protein by WA and damaged the vasculature around the tumor tissue. The investigation revealed that treatment with WA reduced the expression of Pyk2 protein, which is involved in the invasiveness of tumors [66]. Lowered hemoglobin levels were observed in the fibroblast growth factor Matrigel plug isolated from the animals upon treatment with WA, indicating its anti-angiogenic property [67].


#

Effect of Withaferin A on cancer stem cells

Cancer stem cells (CSCs) are the main reason behind the relapse of cancer after chemotherapy because the malignant cells attain self-renewal from the few long-lived CSCs. Chemoresistance for most of the drugs is mainly due to the high expression of anti-apoptotic proteins, efflux transporters, and cell repair enzymes in CSCs [68]. CSCs also play a critical role in the migration of tumor cells in malignant types as seeding and colonization of cancer cells are required to form secondary tumors in different organs [69]. To combat the problem of relapse and to overcome chemoresistance, targeting CSCs would be the potential strategy.

WA at a 0.75 µM concentration inhibited the expression of the Oct-4 gene in a sphere-formation assay of pancreatic cancer stem cells, which suggests inhibition of the self-renewal capacity of CSCs as the Oct-4 gene is a key regulator of sustaining the pluripotency of stem cells and prevents the apoptosis. Forty-eight hours post-treatment with WA inhibited NANOG, a transcription factor involved in the chemoresistance and self-renewal capacity of CSCs [70]. It is believed that most of the carcinoma cells exhibit two types of cell population, adherent and floating cells. Floating cells consist of a dense population of CSCs and spontaneously form tumor aggregates or spheres [71]. WA inhibited the sphere formation and induced apoptosis of the F cell population in the UP-LN1 lymph node metastatic cell line (gastrointestinal carcinoma cell line). One more reason for hiding the apoptosis by WA is the lower levels of glutathione in the F cell sub-population [72].

Enzyme aldehyde dehydrogenase (ALDH) catalyzes the oxidation process of aldehydes through an NADP+-driven manner in CSCs. There are 19 kinds of ALDH isoforms in existence. Among them, ALDH1 has been reported for its high expression in the CSCs of most cancers [73]. ALDH1-positive cells were markedly decreased after treatment with 2 mg/kg of WA on the surface epithelium, as well as on the cortex region of the ovarian cancer spheroid. Furthermore, WA halted the expression of securin, a pituitary transforming gene that is co-expressed with other CSC markers like Oct-4, CD24, CD44, ALDH1, and Shh in all kinds of ovarian cancers [74].

WA was found to be a potent inhibitor of CSCs in induced pluripotent stem cells by abrogation of tumorigenicity. β-galactosidase is a hydrolase enzyme and a marker for senescent cells. WA treatment showed an increase in senescence-associated β-galactosidase activity [75]. WA hindered the migration of CSCs derived from multiple melanoma patients and caused apoptotic cell death [76]. Decreased tumor burden and formation of mammospheres were observed during analysis of therapy-resistant CSCs in transgenic MMTV-neu (mouse-mammary tumor virus-neu) mice by WA at a dose of 0.1 mg thrice in a week for 28 days [77].


#

Targeting inflammation in tumor microenvironment

Inflammatory mediators like tumor necrosis factor (TNF-α) and interleukins (IL) have major contributions to cancer progression. The presence of immune cells in the tumor microenvironment induces the inflammatory response, and tumor tissue is highly supplemented with myeloid suppressor cells (MSCs) and regulatory T-cells [78]. WA has been reported for its anti-inflammatory action in various pathological diseases. Upon activation of inflammatory mediators such as IL-6 and TNF-α on tumor cells, STAT and NF-κB get activated and their interaction results in a transcriptional process for the proliferation, survival, and invasiveness of cancerous cells. WA, being anti-inflammatory, abolished the activation of STAT3 and its interaction with NF-κB by reducing the telomerase activity [79].

MSCs are immature myeloid cells in the bone marrow that migrate to the tumor microenvironment by the tumor-associated cytokines. Pro-inflammatory mediators act on immature myeloid cells and transform them into MSCs. Additionally, MSCs get activated by STAT3 and NF-κB signaling [80]. WA modulated the potency of MSCs by indirectly suppressing the pro-inflammatory mediators and downregulation of STAT3 and the NF-κB function [81]. WA abrogated the IL-6-induced STAT-3 transcriptional activity in HCT116 cells [82]. WA ameliorated pro-inflammatory mediators in ovarian-cancer-induced cachexia by suppressing NF-κB activation and nuclear translocation of p65. The inactivation of NF-κB is observed through the activation of the IKK complex, the inhibitor of NF-κB [83]. Another important dimension of WA is its potential vimentin inhibitory effects.


#

Combination studies

WA in combination with other chemotherapeutic drugs showed the most effective results in cancer therapy. Co-administration of WA showed synergistic action in most of the studies and reduced the chemotherapeutic dose to half, which eventually lowered the toxicity to normal cells. Sorafenib and WA potentiated the cell cycle arrest at the G2/M phase and enhanced apoptosis through caspase-3 activation and PARP cleavage [84]. The combination of two naturally occurring compounds, sulforaphane and WA, regulated the epigenetic mechanisms involved in cancer. Sulforaphane at 5 µM and WA at 1 µM showed synergistic inhibition of histone deacetylase (HDAC) and decreased the transcription of DNA methyltransferases in both estrogen receptor-positive (ER (+)) and -negative (ER (−)) cancer cells. These concentrations have not shown any toxicity in normal MCF10A cell lines, indicating the safety of the combination [85]. Sulforaphane and WA arrested the cell cycle at the G1 phase, and reactivation of the p21 tumor suppressor gene through an epigenetic mechanism in breast cancer cell lines was observed [86].

Cisplatin, a platinum-bearing compound, is well known for its effectiveness in treating breast and ovarian cancer, yet side effects such as toxicity toward normal cells and resistance are frequently faced issues. The IC50 of cisplatin in the ovarian cancer cell line was found to be 40 µM. When combined with WA at 1.5 µM, the IC50 is reduced to 12 µM, and this combination proved very effective in treating cisplatin-sensitive (A2780, CAOV3) and -resistant (A2780/CP70) ovarian cancer cells [87]. The suboptimal dose of cisplatin and WA showed decreased tumor burden in metastatic ovarian cancer and also eliminated CSCs [88]. The anti-tumor activity was doubled when oxaliplatin was given along with WA by suppressing the PI3K/Akt pathway in pancreatic cancer [89]. WA and cisplatin at a ratio of 1 : 10 inhibited the activity of CSCs by acting on many targets of the mTOR/STAT3 signaling pathway in non-small-cell lung cancer [90].

An anthracycline compound, doxorubicin, has been in use to treat a wide spectrum of cancers. However, its use is restricted due to its high myocardial toxicity. When doxorubicin was used in combination with WA, even at a 1 mg/kg dose, they showed effective ROS-mediated autophagic cell death in the xenograft model of ovarian cancer [44]. In the 3D in vitro spheroid model of ovarian cancer, the combination of DOXIL, a liposomal form of doxorubicin, with WA synergistically reduced the expression of ALDH protein [74].

Tumor necrosis-factor-related apoptosis-inducing ligand (TRAIL) has been studied for anticancer activity and found to be very effective at inducing apoptosis, but its resistance toward apoptosis is observed in a few types of cancers [91]. Co-treatment with WA sensitizes the cancer cells to TRAIL-induced apoptosis. The apoptosis mediated through the generation of ROS signals resulted in up-regulation of death receptor 5 (DR5) [92]. Though there are few studies, it is expected that WA may produce additive or synergistic effects in combination with different standard anticancer drugs, which may result in increased pharmacological effects with much better safety profiles. However, the future looks very bright for such kinds of combination approaches, and according to the authorsʼ opinion, such kinds of studies may unveil the much better promising anti-tumor effects of WA.


#

Pharmacokinetic and safety studies

Pharmacokinetic (PK) and safety studies are essential to better achieve the therapeutic efficacy of any drug molecule and to enter clinical studies. PK studies are considered an integral part of the drug development process. When it comes to natural products, PK studies aid in the preparation of suitable formulations or suitable derivatives, which will aid in improved PK profiles. WA showed rapid oral bioavailability at a dose of 1000 mg/kg of WS extracts (WSE) administered orally to the mice. The 1000 mg/kg of WSE is equivalent to 0.4585 mg/kg of WA [93]. However, in vitro bioavailability studies in MDCK cells showed WA has poor permeability when compared to WA given in extract form, suggesting the complexity in the absorption of WA, which indicates the need to carry out detailed PK studies [94]. The time to reach maximum mean plasma concentration (Tmax) was found to be 20 min [93]. WA, when given through the intraperitoneal route at a dose of 4 mg/kg to mice, the Cmax of 1.8 µM was achieved in 4.98 min. The half-life (t1/2) was reported as 1.3 h and AUC0-t as 541.3 ng h/ml. The same study reported the clearance of WA from plasma at a rate of 2.5 ml/min, and it was undetectable in the blood after 24 h of administration [58]. If we carefully observe the reported PK studies of WA or its plant extract, it is evident that the oral bioavailability of WA is still ambiguous. Furthermore, most of the in vivo cancer studies reported with WA were observed to follow the parenteral route of administration; the probable reason could be the uncertainty of its oral bioavailability. On the other side, for natural products to explore for clinical translation, the oral route is the most preferred route, and most of the natural products fail to reach the clinical application stage due to limited oral bioavailability profiles. Probably, WA may also fall into this category; hence, detailed PK and metabolic studies are warranted to better poise this interesting natural molecule for cancer therapy and chemoprevention.

The safety studies in advanced-stage high-grade osteosarcoma patients showed no toxicity at the maximum dose of 216 mg of WA in WSE. Liver enzyme elevation was reported as grade-1 severity in 38.46% of patients [95]. Molecular docking and QSAR studies predicted the toxicity of WA based on fragments present in the structure and reported a medium risk associated with higher doses [96]. Nevertheless, the toxicity studies may not be of prime need to explore novel or natural-product-based anticancer agents. The reason behind this relaxation in the requirement of toxicity studies is the severity of malignancy and the universal belief that small molecule-based anticancer drugs are not free from toxicity concerns. Moreover, since anticancer drugs are not used for long terms, the detailed battery of toxicity studies may not be of much importance compared to another category of drugs like antidiabetic or antihypertensive drugs.


#
#

Challenges Faced by WA to Become an Effective Drug in Cancer Therapy

Numerous studies reported the promising anticancer effects of WA in both in vitro and in vivo conditions. However, WA remains at the pre-clinical stage due to the few factors that are listed below and in [Fig. 3]

Zoom Image
Fig. 3 Challenges facing WA to enter into a cancer treatment regimen.

Lack of safety and toxicology studies

Every new compound should undergo a safety assessment before entering clinical studies. Safety pharmacological studies are conducted to assess whether the drug is safe or producing any toxicity at therapeutic concentrations [97]. Drugs obtained from natural sources are found to be safe; yet safety concerns are a must to save the lives of clinical trial patients. WA, being a potent anticancer molecule, remained at the developmental stage due to a lack of proper safety pharmacology studies. So far, conducted safety studies are on the extracts of WS isolated from various plant parts but not on the core molecule of WA [95]. The extracts of WS contain other withanolides that also have pharmacological activities, which creates confusion that the safety may be due to the presence of other constituents present in the extract.

Besides safety pharmacology, toxicology data have a high concern in the drug developmental stage before use on the human population. Toxicity studies provide information regarding the drugʼs toxic effects at and above therapeutic doses. Animal toxicity studies are very helpful in predicting toxicity in humans [98]. The current established models in cell lines, as well as in animals, are not enough to provide reliable information about the toxicity of the compound. Furthermore, anticancer drugs are well known for their cytotoxicity, and new approaches need to be discovered to predict the toxicity in humans.


#

Need for clinical trials

Consistently, clinical studies remain a keystone for the drug development process. Without clinical trials, no drug can enter the market, even if it is most effective. Before going to the clinical phase, pharmaceutical concepts like off-target physiological effects, pharmacokinetic and pharmacodynamic properties, therapeutic windows, shrinkage capacity of the tumor, and maximum tolerated dose are very essential to be evaluated. But unfortunately, there is no such accumulated data for WA in the case of oncology studies.

Standardization needs to be performed for natural products before entering the initial clinical phase to ensure the substance is the same in every evaluation. Most of the natural products fail the global acceptance due to a lack of a standard quality profile. The quality is being affected by adulteration and variability in the constituents in most of the extracts obtained from natural sources. Quality testing needs to be conducted at all stages, starting from collection of plant parts to the final product [99]. Clinical investigative procedures like placebo-controlled, randomized, and double-blind studies have to be performed for every new compound, which can be carried out only when the standard is available [100].

New treatment regimens and their development for curing cancer are very crucial in the present scenario to improve the patient survival rate. However, clinical trials in oncology are very complex because of the high heterogenicity of the disease. Oncology clinical studies majorly depend on several volunteers participating in the experiment. During drug development, generally healthy volunteers participate in the phase I trial, whereas most of the anticancer drugs are often likely to be more toxic and with severe side effects, and phase I and phase II trials predominantly recruit cancer patients with advanced stages and pre-treated conditions who are more likely to fail with the treatment. Most of the oncologists referring the patients to participate in the clinical studies are very low. This may be due to the lack of trust between investigators and physicians, communication between patients and physicians, low incentives for referrals, and low patient access to information [101].


#

Difficulties in large-scale production

Factors like authentication, geographical location, season and time of harvesting, methods of harvesting, and protocols used for the collection of plant parts are major sources for variation in the chemical constituents of the plant [102]. A high concentration of WA is found between the plant stages before the start of flowering and production of fruits, thereby gradually decreasing the concentration of withanolides [103]. Geographical location has a greater impact on the composition of pharmacologically active constituents. WS plants collected from various regions of India showed a larger variation in the root yield in terms of g/plant, which indicates that geographical location and environmental conditions influence the composition of phytochemical constituents. Among the collected plants, only a few are predominant with WA in root and others with remaining withanolides, whereas the WA content is high in the leaves, which are collected from the northern region [104]. The obtained yield of WA from the 100 g of leaves is reported as less than 2 g, which is fairly difficult for large-scale production, and this results in a supply of WA at a higher price. For example, the cost of 5 mg of WA is $44,000 from Sigma and Santa Cruz. Understanding the complexity of the variability of the plant constituents is an important measure in determining the quality, safety, and integrity of natural products [105].

One more issue with natural products is the moisture content. Raw materials obtained from plant sources have an inherent nature of moisture absorption when handled in bulk quantities, leading to heterogeneity from batch to batch [106]. The annual production of the WS plant is 1500 tons, whereas the demand is for 7000 tons, meaning there is a limited supply of plants to meet the needs of the production of raw material [102]. Because of low yield, the limited supply of plants, and the high cost of production, most pharmaceutical companies lack interest in investing in natural products, creating obstacles in the drug discovery pipeline.


#
#

Conclusion

New drug development is the requisite for cancer treatment to overcome the problems of the current chemotherapeutic drugs in clinical use. WA is an active constituent of WS and has been reported for its anti-tumor properties in various studies. WA showed tumor regression in many cancers like colorectal, breast, thyroid, pancreatic, prostate, lung, hepatic, ovarian, and cervical cancer. WA inhibits tumor growth by inducing cell cycle arrest at the G2/M phase, which is accompanied by upregulation of BCl-2 proteins for apoptotic cell death. WA also inhibits angiogenesis by targeting VEGFR and tumor migration through ceasing EMT. In addition to these, WA has the potency to sensitize the cancer cells during chemo and radiation therapies. Being a potent cancer-inhibitory molecule, WA is still in the development phase for various reasons. Pharmacokinetic, safety, and toxicological studies need to be conducted. New technologies and methods ease the development of WA formulations.


#

Contributorsʼ Statement

Geetanjali Devabattula collected the literature and drafted the manuscript. Biswajit Panda and Rachana Yadav contributed for designing images and collection of literature. Critical revision of manuscript by Chandraiah Godugu.


#
#

Conflict of Interest

The authors declare that they have no conflict of interest.

Acknowledgements

The authors would like to acknowledge the Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers and Director NIPER Hyderabad for their kind support.

  • References

  • 1 Araújo F, Gouvinhas C, Fontes F, La Vecchia C, Azevedo A, Lunet N. Trends in cardiovascular diseases and cancer mortality in 45 countries from five continents (1980–2010). Eur J Prev Cardiol 2014; 21: 1004-1017
  • 2 ReFaey K, Tripathi S, Grewal SS, Bhargav AG, Quinones DJ, Chaichana KL, Antwi SO, Cooper LT, Meyer FB, Dronca RS. Cancer mortality rates increasing vs. cardiovascular disease mortality decreasing in the world: future implications. Mayo Clin Proc Innov Qual Outcomes 2021; 5: 645-653
  • 3 Siegel Rebecca L, Miller Kimberly D. Jemal Ahmedin. Cancer statistics, 2019. CA Cancer J Clin 2019; 69: 7-34
  • 4 World Health Organization. Cancer. In: WHO.
  • 5 Links M, Lewis C. Chemoprotectants: a review of their clinical pharmacology and therapeutic efficacy. Drugs 1999; 57: 293-308
  • 6 Gordaliza M. Natural products as leads to anticancer drugs. Clin Transl Oncol 2007; 9: 767-776
  • 7 Naeem A, Hu P, Yang M, Zhang J, Liu Y, Zhu W, Zheng Q. Natural products as anticancer agents: Current status and future perspectives. Molecules 2022; 27: 8367
  • 8 Martino E, Casamassima G, Castiglione S, Cellupica E, Pantalone S, Papagni F, Rui M, Siciliano AM, Collina S. Vinca alkaloids and analogues as anti-cancer agents: Looking back, peering ahead. Bioorg Med Chem Lett 2018; 28: 2816-2826
  • 9 National Cancer Institute. A to Z List of Cancer Drugs. In: NIH.
  • 10 Tharakan A, Shukla H, Benny IR, Tharakan M, George L, Koshy S. Immunomodulatory effect of Withania somnifera (Ashwagandha) extract–a randomized, double-blind, placebo controlled trial with an open label extension on healthy participants. J Clin Med 2021; 10: 3644
  • 11 Gupta A, Singh S. Evaluation of anti-inflammatory effect of Withania somnifera root on collagen-induced arthritis in rats. Pharm Biol 2014; 52: 308-320
  • 12 Kumari M. Gupta Rá. In vitro antibacterial effect of Withania somnifera root extract on Escherichia coli . Veterinary world 2015; 8: 57
  • 13 Yadav B, Bajaj A, Saxena M, Saxena A. In vitro anticancer activity of the root, stem and leaves of Withania somnifera against various human cancer cell lines. Indian J Pharm Sci 2010; 72: 659
  • 14 Choudhary D, Bhattacharyya S, Bose S. Efficacy and safety of Ashwagandha (Withania somnifera (L.) Dunal) root extract in improving memory and cognitive functions. J Diet Suppl 2017; 14: 599-612
  • 15 Nagy Z, Cheung BB, Tsang W, Tan O, Herath M, Ciampa OC, Shadma F, Carter DR, Marshall GM. Withaferin A activates TRIM16 for its anti-cancer activity in melanoma. Sci Rep 2020; 10: 19724
  • 16 Atteeq M. Evaluating anticancer properties of Withaferin A–a potent phytochemical. Front Pharmacol 2022; 13: 975320
  • 17 Berghe WV, Sabbe L, Kaileh M, Haegeman G, Heyninck K. Molecular insight in the multifunctional activities of Withaferin A. Biochem Pharmacol 2012; 84: 1282-1291
  • 18 Fuska J, Fuskova A, Rosazza J, Nicholas A. Novel cytotoxic and antitumor agents. IV. Withaferin A: relation of its structure to the in vitro cytotoxic effects on P388 cells. Neoplasma 1984; 31: 31-36
  • 19 Kim S-H, Singh KB, Hahm E-R, Singh SV. The role of forkhead box Q1 transcription factor in anticancer effects of Withaferin A in breast cancer. Cancer Prev Res (Phila) 2021; 14: 421-432
  • 20 Yu Y, Hamza A, Zhang T, Gu M, Zou P, Newman B, Li Y, Gunatilaka AL, Zhan C-G, Sun D. Withaferin A targets heat shock protein 90 in pancreatic cancer cells. Biochem Pharmacol 2010; 79: 542-551
  • 21 Liu X, Li Y, Ma Q, Wang Y, Song AL. Withaferin-A inhibits growth of drug-resistant breast carcinoma by inducing apoptosis and autophagy, endogenous reactive oxygen species (ROS) production, and inhibition of cell migration and nuclear factor kappa B (Nf-κB)/mammalian target of rapamycin (m-TOR) signalling pathway. Med Sci Monit 2019; 25: 6855
  • 22 Stan SD, Zeng Y, Singh SV. Ayurvedic medicine constituent withaferin a causes G2 and M phase cell cycle arrest in human breast cancer cells. Nutr Cancer 2008; 60: 51-60
  • 23 McKenna M, Gachuki B, Alhakeem S, Oben K, Rangnekar V, Gupta R, Bondada S. Anti-cancer activity of withaferin A in B-cell lymphoma. Cancer Biol Ther 2015; 16: 1088-1098
  • 24 Touny LHE, Banerjee PP. Identification of both Myt‐1 and Wee‐1 as necessary mediators of the p 21‐independent inactivation of the Cdc‐2/cyclin B1 complex and growth inhibition of TRAMP cancer cells by genistein. Prostate 2006; 66: 1542-1555
  • 25 Roy RV, Suman S, Das TP, Luevano JE, Damodaran C. Withaferin A, a steroidal lactone from Withania somnifera, induces mitotic catastrophe and growth arrest in prostate cancer cells. J Nat Prod 2013; 76: 1909-1915
  • 26 Lv TZ, Wang GS. Antiproliferation potential of withaferin A on human osteosarcoma cells via the inhibition of G2/M checkpoint proteins. Exp Ther Med 2015; 10: 323-329
  • 27 Das T, Roy KS, Chakrabarti T, Mukhopadhyay S, Roychoudhury S. Withaferin A modulates the Spindle Assembly Checkpoint by degradation of Mad2–Cdc20 complex in colorectal cancer cell lines. Biochem Pharmacol 2014; 91: 31-39
  • 28 Yeh ES, Means AR. PIN1, the cell cycle and cancer. Nat Rev Cancer 2007; 7: 381-388
  • 29 Nakashima M, Meirmanov S, Naruke Y, Kondo H, Saenko V, Rogounovitch T, Shimizu-Yoshida Y, Takamura N, Namba H, Ito M. Cyclin D1 overexpression in thyroid tumours from a radio‐contaminated area and its correlation with Pin1 and aberrant β‐catenin expression. J Pathol 2004; 202: 446-455
  • 30 Samanta SK, Lee J, Hahm ER, Singh SV. Peptidyl‐prolyl cis/trans isomerase Pin1 regulates withaferin A‐mediated cell cycle arrest in human breast cancer cells. Mol Carcinog 2018; 57: 936-946
  • 31 Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol 2007; 35: 495-516
  • 32 Majno G, Joris I. Apoptosis, oncosis, and necrosis. An overview of cell death. Am J Pathol 1995; 146: 3
  • 33 Srinivasan S, Ranga RS, Burikhanov R, Han S-S, Chendil D. Par-4-dependent apoptosis by the dietary compound withaferin A in prostate cancer cells. Cancer Res 2007; 67: 246-253
  • 34 Gross A, McDonnell JM, Korsmeyer SJ. BCL-2 family members and the mitochondria in apoptosis. Genes Dev 1999; 13: 1899-1911
  • 35 Stan SD, Hahm E-R, Warin R, Singh SV. Withaferin A causes FOXO3a-and Bim-dependent apoptosis and inhibits growth of human breast cancer cells in vivo. Cancer Res 2008; 68: 7661-7669
  • 36 Liou G-Y, Storz P. Reactive oxygen species in cancer. Free Radic Res 2010; 44: 479-496
  • 37 Xia S, Miao Y, Liu S. Withaferin A induces apoptosis by ROS-dependent mitochondrial dysfunction in human colorectal cancer cells. Biochem Biophys Res Commun 2018; 503: 2363-2369
  • 38 Chang H-W, Li R-N, Wang H-R, Liu J-R, Tang J-Y, Huang H-W, Chan Y-H, Yen C-Y. Withaferin A induces oxidative stress-mediated apoptosis and DNA damage in oral cancer cells. Front Physiol 2017; 8: 634
  • 39 Munagala R, Kausar H, Munjal C, Gupta RC. Withaferin A induces p 53-dependent apoptosis by repression of HPV oncogenes and upregulation of tumor suppressor proteins in human cervical cancer cells. Carcinogenesis 2011; 32: 1697-1705
  • 40 Mathew R, Karantza-Wadsworth V, White E. Role of autophagy in cancer. Nat Rev Cancer 2007; 7: 961-967
  • 41 Gewirtz DA. The four faces of autophagy: implications for cancer therapy. Cancer Res 2014; 74: 647-651
  • 42 Ghosh K, De S, Mukherjee S, Das S, Ghosh AN, Sengupta SB. Withaferin A induced impaired autophagy and unfolded protein response in human breast cancer cell-lines MCF-7 and MDA-MB-231. Toxicol In Vitro 2017; 44: 330-338
  • 43 Yin X, Yang G, Ma D, Su Z. Inhibition of cancer cell growth in cisplatin-resistant human oral cancer cells by withaferin-A is mediated via both apoptosis and autophagic cell death, endogenous ROS production, G2/M phase cell cycle arrest and by targeting MAPK/RAS/RAF signalling pathway. J BUON 2020; 25: 332-337
  • 44 Fong MY, Jin S, Rane M, Singh RK, Gupta R, Kakar SS. Withaferin A synergizes the therapeutic effect of doxorubicin through ROS-mediated autophagy in ovarian cancer. PLoS One 2012; 7: e42265
  • 45 Siddharth S, Muniraj N, Saxena NK, Sharma D. Concomitant inhibition of cytoprotective autophagy augments the efficacy of withaferin A in hepatocellular carcinoma. Cancers (Basel) 2019; 11: 453
  • 46 Romero-Figueroa MDS, de Jesus Garduno-Garcia J, Duarte-Mote J, Matute-Gonzalez G, Gómez-Villanueva A, De la Cruz-Vargas J. Insulin and leptin levels in obese patients with and without breast cancer. Clin Breast Cancer 2013; 13: 482-485
  • 47 Samanta SK, Sehrawat A, Kim S-H, Hahm ER, Shuai Y, Roy R, Pore SK, Singh KB, Christner SM, Beumer JH. Disease subtype–independent biomarkers of breast cancer chemoprevention by the ayurvedic medicine phytochemical withaferin A. J Natl Cancer Inst 2017; 109: djw293
  • 48 Manoharan S, Panjamurthy K, Menon VP, Balakrishnan S, Alias LM. Protective effect of Withaferin-A on tumour formation in 7, 12-dimethylbenz [a] anthracene induced oral carcinogenesis in hamsters. Indian J Exp Biol 2009; 47: 16-23
  • 49 Chandrasekaran B, Pal D, Kolluru V, Tyagi A, Baby B, Dahiya NR, Youssef K, Alatassi H, Ankem MK, Sharma AK. The chemopreventive effect of withaferin A on spontaneous and inflammation-associated colon carcinogenesis models. Carcinogenesis 2018; 39: 1537-1547
  • 50 Yang ES, Choi MJ, Kim JH, Choi KS, Kwon TK. Combination of withaferin A and X-ray irradiation enhances apoptosis in U937 cells. Toxicol In Vitro 2011; 25: 1803-1810
  • 51 Yang ES, Choi MJ, Kim JH, Choi KS, Kwon TK. Withaferin A enhances radiation-induced apoptosis in Caki cells through induction of reactive oxygen species, Bcl-2 downregulation and Akt inhibition. Chem Biol Interact 2011; 190: 9-15
  • 52 Sharada AC, Solomon FE, Devi PU, Udupa N, Srinivasan KK. Antitumor and radiosensitizing effects of withaferin A on mouse Ehrlich ascites carcinoma in vivo. Acta Oncol 1996; 35: 95-100
  • 53 Kalthur G, Pathirissery UD. Enhancement of the response of B16F1 melanoma to fractionated radiotherapy and prolongation of survival by withaferin A and/or hyperthermia. Integr Cancer Ther 2010; 9: 370-377
  • 54 Craene BD, Berx G. Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer 2013; 13: 97-110
  • 55 Kyakulaga AH, Aqil F, Munagala R, Gupta RC. Withaferin A inhibits epithelial to mesenchymal transition in non-small cell lung cancer cells. Sci Rep 2018; 8: 15737
  • 56 Wu M, Bai X, Xu G, Wei J, Zhu T, Zhang Y, Li Q, Liu P, Song A, Zhao L. Proteome analysis of human androgen‐independent prostate cancer cell lines: Variable metastatic potentials correlated with vimentin expression. Proteomics 2007; 7: 1973-1983
  • 57 Lee J, Hahm ER, Marcus AI, Singh SV. Withaferin A inhibits experimental epithelial–mesenchymal transition in MCF‐10A cells and suppresses vimentin protein level in vivo in breast tumors. Mol Carcinog 2015; 54: 417-429
  • 58 Thaiparambil JT, Bender L, Ganesh T, Kline E, Patel P, Liu Y, Tighiouart M, Vertino PM, Harvey RD, Garcia A. Withaferin A inhibits breast cancer invasion and metastasis at sub‐cytotoxic doses by inducing vimentin disassembly and serine 56 phosphorylation. Int J Cancer 2011; 129: 2744-2755
  • 59 Gialeli C, Theocharis AD, Karamanos NK. Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting. FEBS J 2011; 278: 16-27
  • 60 Loechel F, Fox JW, Murphy G, Albrechtsen R, Wewer UM. ADAM 12-S cleaves IGFBP-3 and IGFBP-5 and is inhibited by TIMP-3. Biochem Biophys Res Commun 2000; 278: 511-515
  • 61 Lee DH, Lim I-H, Sung E-G, Kim J-Y, Song I-H, Park YK, Lee T-J. Withaferin A inhibits matrix metalloproteinase-9 activity by suppressing the Akt signaling pathway. Oncol Rep 2013; 30: 933-938
  • 62 Hurst DR, Welch DR. Metastasis suppressor genes: at the interface between the environment and tumor cell growth. Int Rev Cell Mol Biol 2011; 286: 107-180
  • 63 Szarc vel Szic K, Op de Beeck K, Ratman D, Wouters A, Beck IM, Declerck K, Heyninck K, Fransen E, Bracke M, De Bosscher K. Pharmacological levels of Withaferin A (Withania somnifera) trigger clinically relevant anticancer effects specific to triple negative breast cancer cells. PLoS One 2014; 9: e87850
  • 64 Nishida N, Yano H, Nishida T, Kamura T, Kojiro M. Angiogenesis in cancer. Vasc Health Risk Manag 2006; 2: 213-219
  • 65 Saha S, Islam MK, Shilpi JA, Hasan S. Inhibition of VEGF: a novel mechanism to control angiogenesis by Withania somniferaʼs key metabolite Withaferin A. In Silico Pharmacol 2013; 1: 1-9
  • 66 Wang Y-X, Ding W-B, Dong C-W. Withaferin A suppresses liver tumor growth in a nude mouse model by downregulation of cell signaling pathway leading to invasion and angiogenesis. Tropical J Pharmaceut Res 2015; 14: 1005-1011
  • 67 Mohan R, Hammers H, Bargagna-Mohan P, Zhan X, Herbstritt C, Ruiz A, Zhang L, Hanson A, Conner B, Rougas J. Withaferin A is a potent inhibitor of angiogenesis. Angiogenesis 2004; 7: 115-122
  • 68 Bao B, Ahmad A, Azmi AS, Ali S, Sarkar FH. Overview of cancer stem cells (CSCs) and mechanisms of their regulation: implications for cancer therapy. Curr Protoc Pharmacol 2013; 61: 14.25-11-14.25-14
  • 69 Li F, Tiede B, Massagué J, Kang Y. Beyond tumorigenesis: cancer stem cells in metastasis. Cell Res 2007; 17: 3-14
  • 70 Aliebrahimi S, Kouhsari SM, Arab SS, Shadboorestan A, Ostad SN. Phytochemicals, withaferin A and carnosol, overcome pancreatic cancer stem cells as c-Met inhibitors. Biomed Pharmacother 2018; 106: 1527-1536
  • 71 Chen H-C, Chou AS-B, Liu Y-C, Hsieh C-H, Kang C-C, Pang S-T, Yeh C-T, Liu H-P, Liao S-K. Induction of metastatic cancer stem cells from the NK/LAK-resistant floating, but not adherent, subset of the UP-LN1 carcinoma cell line by IFN-γ . Lab Invest 2011; 91: 1502-1513
  • 72 Ting L-L, Chou AS-B, Hsieh C-H, Hsiung S-C, Pang S-T, Liao S-K. Withaferin A targeting both cancer stem cells and metastatic cancer stem cells in the UP-LN1 carcinoma cell model. J Cancer Metastasis Treat 2016; DOI: 10.4103/2394-4722.172008.
  • 73 Young M-J, Wu Y-H, Chiu W-T, Weng T-Y, Huang Y-F, Chou C-Y. All-trans retinoic acid downregulates ALDH1-mediated stemness and inhibits tumour formation in ovarian cancer cells. Carcinogenesis 2015; 36: 498-507
  • 74 Kakar SS, Parte S, Carter K, Joshua IG, Worth C, Rameshwar P, Ratajczak MZ. Correction: Withaferin A (WFA) inhibits tumor growth and metastasis by targeting ovarian cancer stem cells. Oncotarget 2020; 11: 3103
  • 75 Nishi M, Akutsu H, Kudoh A, Kimura H, Yamamoto N, Umezawa A, Lee SW, Ryo A. Induced cancer stem-like cells as a model for biological screening and discovery of agents targeting phenotypic traits of cancer stem cell. Oncotarget 2014; 5: 8665
  • 76 Issa ME, Cuendet M. Withaferin A induces cell death and differentiation in multiple myeloma cancer stem cells. Medchemcomm 2017; 8: 112-121
  • 77 Kim S-H, Singh SV. Mammary cancer chemoprevention by withaferin A is accompanied by in vivo suppression of self-renewal of cancer stem cells. Cancer Prev Res (Phila) 2014; 7: 738-747
  • 78 Whiteside T. The tumor microenvironment and its role in promoting tumor growth. Oncogene 2008; 27: 5904-5912
  • 79 Chung SS, Wu Y, Okobi Q, Adekoya D, Atefi M, Clarke O, Dutta P, Vadgama JV. Proinflammatory cytokines IL-6 and TNF-α increased telomerase activity through NF-κB/STAT1/STAT3 activation, and withaferin A inhibited the signaling in colorectal cancer cells. Mediators of inflammation 2017; DOI: 10.1155/2017/5958429.
  • 80 Draghiciu O, Lubbers J, Nijman HW, Daemen T. Myeloid derived suppressor cells–an overview of combat strategies to increase immunotherapy efficacy. Oncoimmunology 2015; 4: e954829
  • 81 Sinha P, Ostrand-Rosenberg S. Myeloid-derived suppressor cell function is reduced by Withaferin A, a potent and abundant component of Withania somnifera root extract. Cancer Immunology, Immunotherapy 2013; 62: 1663-1673
  • 82 Choi BY, Kim B-W. Withaferin-A inhibits colon cancer cell growth by blocking STAT3 transcriptional activity. J Cancer Prev 2015; 20: 185
  • 83 Straughn AR, Kakar SS. Withaferin A ameliorates ovarian cancer-induced cachexia and proinflammatory signaling. J Ovarian Res 2019; 12: 1-14
  • 84 Cohen SM, Mukerji R, Timmermann BN, Samadi AK, Cohen MS. A novel combination of withaferin A and sorafenib shows synergistic efficacy against both papillary and anaplastic thyroid cancers. Am J Surg 2012; 204: 895-901
  • 85 Royston KJ, Udayakumar N, Lewis K, Tollefsbol TO. A novel combination of withaferin A and sulforaphane inhibits epigenetic machinery, cellular viability and induces apoptosis of breast cancer cells. Int J Mol Sci 2017; 18: 1092
  • 86 Royston KJ, Paul B, Nozell S, Rajbhandari R, Tollefsbol TO. Withaferin A and sulforaphane regulate breast cancer cell cycle progression through epigenetic mechanisms. Exp Cell Res 2018; 368: 67-74
  • 87 Kakar SS, Jala VR, Fong MY. Synergistic cytotoxic action of cisplatin and withaferin A on ovarian cancer cell lines. Biochem Biophys Res Commun 2012; 423: 819-825
  • 88 Kakar SS, Ratajczak MZ, Powell KS, Moghadamfalahi M, Miller DM, Batra SK, Singh SK. Withaferin a alone and in combination with cisplatin suppresses growth and metastasis of ovarian cancer by targeting putative cancer stem cells. PLoS One 2014; 9: e107596
  • 89 Li X, Zhu F, Jiang J, Sun C, Wang X, Shen M, Tian R, Shi C, Xu M, Peng F. Synergistic antitumor activity of withaferin A combined with oxaliplatin triggers reactive oxygen species-mediated inactivation of the PI3K/AKT pathway in human pancreatic cancer cells. Cancer Lett 2015; 357: 219-230
  • 90 Hsu JH-M, Chang PM-H, Cheng T-S, Kuo Y-L, Wu AT-H, Tran T-H, Yang Y-H, Chen J-M, Tsai Y-C, Chu Y-S. Identification of withaferin A as a potential candidate for anti-cancer therapy in non-small cell lung cancer. Cancers (Basel) 2019; 11: 1003
  • 91 Hernandez A, Smith F, Wang Q, Wang X, Evers BM. Assessment of differential gene expression patterns in human colon cancers. Ann Surg 2000; 232: 576
  • 92 Lee T-J, Um HJ, Park J-W, Choi KS, Kwon TK. Withaferin A sensitizes TRAIL-induced apoptosis through reactive oxygen species-mediated up-regulation of death receptor 5 and down-regulation of c-FLIP. Free Radic Biol Med 2009; 46: 1639-1649
  • 93 Patil D, Gautam M, Mishra S, Karupothula S, Gairola S, Jadhav S, Pawar S, Patwardhan B. Determination of withaferin A and withanolide A in mice plasma using high-performance liquid chromatography-tandem mass spectrometry: Application to pharmacokinetics after oral administration of Withania somnifera aqueous extract. J Pharm Biomed Anal 2013; 80: 203-212
  • 94 Devkar ST, Kandhare AD, Sloley BD, Jagtap SD, Lin J, Tam YK, Katyare SS, Bodhankar SL, Hegde MV. Evaluation of the bioavailability of major withanolides of Withania somnifera using an in vitro absorption model system. J Adv Pharm Technol Res 2015; 6: 159
  • 95 Pires N, Gota V, Gulia A, Hingorani L, Agarwal M, Puri A. Safety and pharmacokinetics of Withaferin-A in advanced stage high grade osteosarcoma: A phase I trial. J Ayurveda Integr Med 2020; 11: 68-72
  • 96 Yadav DK, Kumar S, Saloni. Singh H, Kim MH, Sharma P, Misra S, Khan F. Molecular docking, QSAR and ADMET studies of withanolide analogs against breast cancer. Drug Des Devel Ther 2017; 11: 1859-1870
  • 97 Pugsley MK, Authier S, Curtis M. Principles of safety pharmacology. Br J Pharmacol 2008; 154: 1382-1399
  • 98 Arome D, Chinedu E. The importance of toxicity testing. J Pharm BioSci 2013; 4: 146-148
  • 99 Gupta Y. Clinical Trials of Traditional Herbal Medicines in India. 2011
  • 100 Shan JJ, Rodgers K, Lai C, Sutherland SK. Challenges in natural health product research: The importance of standardization. In: Proceedings-Western Pharmacology Society: [Western Pharmacology Society] 1998; 2007: 188
  • 101 Babiker HM, Davis L, Larson K, Placencia C, Swensen C, Tenneti P, Lim M, Cañamar R, Curtis J, Castillo E. A Multidisciplinary Evaluation of Barriers to Enrolling Cancer Patients into Early Phase Clinical Trials: Challenges and Patient-centric Recommendations. Expert Opin Investig Drugs 2019; 28: 675-686
  • 102 Tavhare SD, Nishteswar K, Shukla VJ. Effect of seasonal variations on the phytoconstituents of Aśvagandhā wr to lunar cycles. Anc Sci Life 2016; 35: 150
  • 103 Nair AR, Praveen N. Biochemical and phytochemical variations during the growth phase of Withania somnifera (L.) Dunal . Journal of Pharmacognosy and Phytochemistry 2019; 8: 1930-1937
  • 104 Kumar A, Kaul M, Bhan M, Khanna PK, Suri K. Morphological and chemical variation in 25 collections of the Indian medicinal plant, Withania somnifera (L.) Dunal (Solanaceae). Genet Res Crop Evol 2007; 54: 655-660
  • 105 Shipkowski KA, Betz JM, Birnbaum LS, Bucher JR, Coates PM, Hopp DC, MacKay D, Oketch-Rabah H, Walker NJ, Welch C. Naturally complex: Perspectives and challenges associated with Botanical Dietary Supplement Safety assessment. Food Chem Toxicol 2018; 118: 963-971
  • 106 Jachak SM, Saklani A. Challenges and opportunities in drug discovery from plants. Current science 2007; DOI: 1251-1257
  • 107 Joshi P, Misra L, Siddique AA, Srivastava M, Kumar S, Darokar MP. Epoxide group relationship with cytotoxicity in withanolide derivatives from Withania somnifera . Steroids 2014; 79: 19-27
  • 108 Yu Y, Katiyar SP, Sundar D, Kaul Z, Miyako E, Zhang Z, Kaul SC, Reddel RR, Wadhwa R. Withaferin-A kills cancer cells with and without telomerase: chemical, computational and experimental evidences. Cell Death Dis 2017; 8: e2755
  • 109 Li A-X, Sun M, Li X. Withaferin-A induces apoptosis in osteosarcoma U2OS cell line via generation of ROS and disruption of mitochondrial membrane potential. European Review for Medical & Pharmacological Sciences 2017; 21
  • 110 Malik F, Kumar A, Bhushan S, Khan S, Bhatia A, Suri KA, Qazi GN, Singh J. Reactive oxygen species generation and mitochondrial dysfunction in the apoptotic cell death of human myeloid leukemia HL-60 cells by a dietary compound withaferin A with concomitant protection by N-acetyl cysteine. Apoptosis 2007; 12: 2115-2133
  • 111 Lahat G, Zhu Q-S, Huang K-L, Wang S, Bolshakov S, Liu J, Torres K, Langley RR, Lazar AJ, Hung MC. Vimentin is a novel anti-cancer therapeutic target; insights from in vitro and in vivo mice xenograft studies. PLoS One 2010; 5: e10105

Correspondence

Dr. Chandraiah Godugu, Assistant Professor
Department of Biological Sciences (Regulatory Toxicology)
National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad
NH9, Balanagar Main Road, Kukatpally Industrial Estate
Balanagar, Hyderabad, Telangana
India   
Phone: + 91 (0) 40 23 07 37 41   
Fax: + 91 (0) 40 23 07 37 51   

Publication History

Received: 16 October 2023

Accepted after revision: 06 March 2024

Article published online:
08 April 2024

© 2024. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Araújo F, Gouvinhas C, Fontes F, La Vecchia C, Azevedo A, Lunet N. Trends in cardiovascular diseases and cancer mortality in 45 countries from five continents (1980–2010). Eur J Prev Cardiol 2014; 21: 1004-1017
  • 2 ReFaey K, Tripathi S, Grewal SS, Bhargav AG, Quinones DJ, Chaichana KL, Antwi SO, Cooper LT, Meyer FB, Dronca RS. Cancer mortality rates increasing vs. cardiovascular disease mortality decreasing in the world: future implications. Mayo Clin Proc Innov Qual Outcomes 2021; 5: 645-653
  • 3 Siegel Rebecca L, Miller Kimberly D. Jemal Ahmedin. Cancer statistics, 2019. CA Cancer J Clin 2019; 69: 7-34
  • 4 World Health Organization. Cancer. In: WHO.
  • 5 Links M, Lewis C. Chemoprotectants: a review of their clinical pharmacology and therapeutic efficacy. Drugs 1999; 57: 293-308
  • 6 Gordaliza M. Natural products as leads to anticancer drugs. Clin Transl Oncol 2007; 9: 767-776
  • 7 Naeem A, Hu P, Yang M, Zhang J, Liu Y, Zhu W, Zheng Q. Natural products as anticancer agents: Current status and future perspectives. Molecules 2022; 27: 8367
  • 8 Martino E, Casamassima G, Castiglione S, Cellupica E, Pantalone S, Papagni F, Rui M, Siciliano AM, Collina S. Vinca alkaloids and analogues as anti-cancer agents: Looking back, peering ahead. Bioorg Med Chem Lett 2018; 28: 2816-2826
  • 9 National Cancer Institute. A to Z List of Cancer Drugs. In: NIH.
  • 10 Tharakan A, Shukla H, Benny IR, Tharakan M, George L, Koshy S. Immunomodulatory effect of Withania somnifera (Ashwagandha) extract–a randomized, double-blind, placebo controlled trial with an open label extension on healthy participants. J Clin Med 2021; 10: 3644
  • 11 Gupta A, Singh S. Evaluation of anti-inflammatory effect of Withania somnifera root on collagen-induced arthritis in rats. Pharm Biol 2014; 52: 308-320
  • 12 Kumari M. Gupta Rá. In vitro antibacterial effect of Withania somnifera root extract on Escherichia coli . Veterinary world 2015; 8: 57
  • 13 Yadav B, Bajaj A, Saxena M, Saxena A. In vitro anticancer activity of the root, stem and leaves of Withania somnifera against various human cancer cell lines. Indian J Pharm Sci 2010; 72: 659
  • 14 Choudhary D, Bhattacharyya S, Bose S. Efficacy and safety of Ashwagandha (Withania somnifera (L.) Dunal) root extract in improving memory and cognitive functions. J Diet Suppl 2017; 14: 599-612
  • 15 Nagy Z, Cheung BB, Tsang W, Tan O, Herath M, Ciampa OC, Shadma F, Carter DR, Marshall GM. Withaferin A activates TRIM16 for its anti-cancer activity in melanoma. Sci Rep 2020; 10: 19724
  • 16 Atteeq M. Evaluating anticancer properties of Withaferin A–a potent phytochemical. Front Pharmacol 2022; 13: 975320
  • 17 Berghe WV, Sabbe L, Kaileh M, Haegeman G, Heyninck K. Molecular insight in the multifunctional activities of Withaferin A. Biochem Pharmacol 2012; 84: 1282-1291
  • 18 Fuska J, Fuskova A, Rosazza J, Nicholas A. Novel cytotoxic and antitumor agents. IV. Withaferin A: relation of its structure to the in vitro cytotoxic effects on P388 cells. Neoplasma 1984; 31: 31-36
  • 19 Kim S-H, Singh KB, Hahm E-R, Singh SV. The role of forkhead box Q1 transcription factor in anticancer effects of Withaferin A in breast cancer. Cancer Prev Res (Phila) 2021; 14: 421-432
  • 20 Yu Y, Hamza A, Zhang T, Gu M, Zou P, Newman B, Li Y, Gunatilaka AL, Zhan C-G, Sun D. Withaferin A targets heat shock protein 90 in pancreatic cancer cells. Biochem Pharmacol 2010; 79: 542-551
  • 21 Liu X, Li Y, Ma Q, Wang Y, Song AL. Withaferin-A inhibits growth of drug-resistant breast carcinoma by inducing apoptosis and autophagy, endogenous reactive oxygen species (ROS) production, and inhibition of cell migration and nuclear factor kappa B (Nf-κB)/mammalian target of rapamycin (m-TOR) signalling pathway. Med Sci Monit 2019; 25: 6855
  • 22 Stan SD, Zeng Y, Singh SV. Ayurvedic medicine constituent withaferin a causes G2 and M phase cell cycle arrest in human breast cancer cells. Nutr Cancer 2008; 60: 51-60
  • 23 McKenna M, Gachuki B, Alhakeem S, Oben K, Rangnekar V, Gupta R, Bondada S. Anti-cancer activity of withaferin A in B-cell lymphoma. Cancer Biol Ther 2015; 16: 1088-1098
  • 24 Touny LHE, Banerjee PP. Identification of both Myt‐1 and Wee‐1 as necessary mediators of the p 21‐independent inactivation of the Cdc‐2/cyclin B1 complex and growth inhibition of TRAMP cancer cells by genistein. Prostate 2006; 66: 1542-1555
  • 25 Roy RV, Suman S, Das TP, Luevano JE, Damodaran C. Withaferin A, a steroidal lactone from Withania somnifera, induces mitotic catastrophe and growth arrest in prostate cancer cells. J Nat Prod 2013; 76: 1909-1915
  • 26 Lv TZ, Wang GS. Antiproliferation potential of withaferin A on human osteosarcoma cells via the inhibition of G2/M checkpoint proteins. Exp Ther Med 2015; 10: 323-329
  • 27 Das T, Roy KS, Chakrabarti T, Mukhopadhyay S, Roychoudhury S. Withaferin A modulates the Spindle Assembly Checkpoint by degradation of Mad2–Cdc20 complex in colorectal cancer cell lines. Biochem Pharmacol 2014; 91: 31-39
  • 28 Yeh ES, Means AR. PIN1, the cell cycle and cancer. Nat Rev Cancer 2007; 7: 381-388
  • 29 Nakashima M, Meirmanov S, Naruke Y, Kondo H, Saenko V, Rogounovitch T, Shimizu-Yoshida Y, Takamura N, Namba H, Ito M. Cyclin D1 overexpression in thyroid tumours from a radio‐contaminated area and its correlation with Pin1 and aberrant β‐catenin expression. J Pathol 2004; 202: 446-455
  • 30 Samanta SK, Lee J, Hahm ER, Singh SV. Peptidyl‐prolyl cis/trans isomerase Pin1 regulates withaferin A‐mediated cell cycle arrest in human breast cancer cells. Mol Carcinog 2018; 57: 936-946
  • 31 Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol 2007; 35: 495-516
  • 32 Majno G, Joris I. Apoptosis, oncosis, and necrosis. An overview of cell death. Am J Pathol 1995; 146: 3
  • 33 Srinivasan S, Ranga RS, Burikhanov R, Han S-S, Chendil D. Par-4-dependent apoptosis by the dietary compound withaferin A in prostate cancer cells. Cancer Res 2007; 67: 246-253
  • 34 Gross A, McDonnell JM, Korsmeyer SJ. BCL-2 family members and the mitochondria in apoptosis. Genes Dev 1999; 13: 1899-1911
  • 35 Stan SD, Hahm E-R, Warin R, Singh SV. Withaferin A causes FOXO3a-and Bim-dependent apoptosis and inhibits growth of human breast cancer cells in vivo. Cancer Res 2008; 68: 7661-7669
  • 36 Liou G-Y, Storz P. Reactive oxygen species in cancer. Free Radic Res 2010; 44: 479-496
  • 37 Xia S, Miao Y, Liu S. Withaferin A induces apoptosis by ROS-dependent mitochondrial dysfunction in human colorectal cancer cells. Biochem Biophys Res Commun 2018; 503: 2363-2369
  • 38 Chang H-W, Li R-N, Wang H-R, Liu J-R, Tang J-Y, Huang H-W, Chan Y-H, Yen C-Y. Withaferin A induces oxidative stress-mediated apoptosis and DNA damage in oral cancer cells. Front Physiol 2017; 8: 634
  • 39 Munagala R, Kausar H, Munjal C, Gupta RC. Withaferin A induces p 53-dependent apoptosis by repression of HPV oncogenes and upregulation of tumor suppressor proteins in human cervical cancer cells. Carcinogenesis 2011; 32: 1697-1705
  • 40 Mathew R, Karantza-Wadsworth V, White E. Role of autophagy in cancer. Nat Rev Cancer 2007; 7: 961-967
  • 41 Gewirtz DA. The four faces of autophagy: implications for cancer therapy. Cancer Res 2014; 74: 647-651
  • 42 Ghosh K, De S, Mukherjee S, Das S, Ghosh AN, Sengupta SB. Withaferin A induced impaired autophagy and unfolded protein response in human breast cancer cell-lines MCF-7 and MDA-MB-231. Toxicol In Vitro 2017; 44: 330-338
  • 43 Yin X, Yang G, Ma D, Su Z. Inhibition of cancer cell growth in cisplatin-resistant human oral cancer cells by withaferin-A is mediated via both apoptosis and autophagic cell death, endogenous ROS production, G2/M phase cell cycle arrest and by targeting MAPK/RAS/RAF signalling pathway. J BUON 2020; 25: 332-337
  • 44 Fong MY, Jin S, Rane M, Singh RK, Gupta R, Kakar SS. Withaferin A synergizes the therapeutic effect of doxorubicin through ROS-mediated autophagy in ovarian cancer. PLoS One 2012; 7: e42265
  • 45 Siddharth S, Muniraj N, Saxena NK, Sharma D. Concomitant inhibition of cytoprotective autophagy augments the efficacy of withaferin A in hepatocellular carcinoma. Cancers (Basel) 2019; 11: 453
  • 46 Romero-Figueroa MDS, de Jesus Garduno-Garcia J, Duarte-Mote J, Matute-Gonzalez G, Gómez-Villanueva A, De la Cruz-Vargas J. Insulin and leptin levels in obese patients with and without breast cancer. Clin Breast Cancer 2013; 13: 482-485
  • 47 Samanta SK, Sehrawat A, Kim S-H, Hahm ER, Shuai Y, Roy R, Pore SK, Singh KB, Christner SM, Beumer JH. Disease subtype–independent biomarkers of breast cancer chemoprevention by the ayurvedic medicine phytochemical withaferin A. J Natl Cancer Inst 2017; 109: djw293
  • 48 Manoharan S, Panjamurthy K, Menon VP, Balakrishnan S, Alias LM. Protective effect of Withaferin-A on tumour formation in 7, 12-dimethylbenz [a] anthracene induced oral carcinogenesis in hamsters. Indian J Exp Biol 2009; 47: 16-23
  • 49 Chandrasekaran B, Pal D, Kolluru V, Tyagi A, Baby B, Dahiya NR, Youssef K, Alatassi H, Ankem MK, Sharma AK. The chemopreventive effect of withaferin A on spontaneous and inflammation-associated colon carcinogenesis models. Carcinogenesis 2018; 39: 1537-1547
  • 50 Yang ES, Choi MJ, Kim JH, Choi KS, Kwon TK. Combination of withaferin A and X-ray irradiation enhances apoptosis in U937 cells. Toxicol In Vitro 2011; 25: 1803-1810
  • 51 Yang ES, Choi MJ, Kim JH, Choi KS, Kwon TK. Withaferin A enhances radiation-induced apoptosis in Caki cells through induction of reactive oxygen species, Bcl-2 downregulation and Akt inhibition. Chem Biol Interact 2011; 190: 9-15
  • 52 Sharada AC, Solomon FE, Devi PU, Udupa N, Srinivasan KK. Antitumor and radiosensitizing effects of withaferin A on mouse Ehrlich ascites carcinoma in vivo. Acta Oncol 1996; 35: 95-100
  • 53 Kalthur G, Pathirissery UD. Enhancement of the response of B16F1 melanoma to fractionated radiotherapy and prolongation of survival by withaferin A and/or hyperthermia. Integr Cancer Ther 2010; 9: 370-377
  • 54 Craene BD, Berx G. Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer 2013; 13: 97-110
  • 55 Kyakulaga AH, Aqil F, Munagala R, Gupta RC. Withaferin A inhibits epithelial to mesenchymal transition in non-small cell lung cancer cells. Sci Rep 2018; 8: 15737
  • 56 Wu M, Bai X, Xu G, Wei J, Zhu T, Zhang Y, Li Q, Liu P, Song A, Zhao L. Proteome analysis of human androgen‐independent prostate cancer cell lines: Variable metastatic potentials correlated with vimentin expression. Proteomics 2007; 7: 1973-1983
  • 57 Lee J, Hahm ER, Marcus AI, Singh SV. Withaferin A inhibits experimental epithelial–mesenchymal transition in MCF‐10A cells and suppresses vimentin protein level in vivo in breast tumors. Mol Carcinog 2015; 54: 417-429
  • 58 Thaiparambil JT, Bender L, Ganesh T, Kline E, Patel P, Liu Y, Tighiouart M, Vertino PM, Harvey RD, Garcia A. Withaferin A inhibits breast cancer invasion and metastasis at sub‐cytotoxic doses by inducing vimentin disassembly and serine 56 phosphorylation. Int J Cancer 2011; 129: 2744-2755
  • 59 Gialeli C, Theocharis AD, Karamanos NK. Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting. FEBS J 2011; 278: 16-27
  • 60 Loechel F, Fox JW, Murphy G, Albrechtsen R, Wewer UM. ADAM 12-S cleaves IGFBP-3 and IGFBP-5 and is inhibited by TIMP-3. Biochem Biophys Res Commun 2000; 278: 511-515
  • 61 Lee DH, Lim I-H, Sung E-G, Kim J-Y, Song I-H, Park YK, Lee T-J. Withaferin A inhibits matrix metalloproteinase-9 activity by suppressing the Akt signaling pathway. Oncol Rep 2013; 30: 933-938
  • 62 Hurst DR, Welch DR. Metastasis suppressor genes: at the interface between the environment and tumor cell growth. Int Rev Cell Mol Biol 2011; 286: 107-180
  • 63 Szarc vel Szic K, Op de Beeck K, Ratman D, Wouters A, Beck IM, Declerck K, Heyninck K, Fransen E, Bracke M, De Bosscher K. Pharmacological levels of Withaferin A (Withania somnifera) trigger clinically relevant anticancer effects specific to triple negative breast cancer cells. PLoS One 2014; 9: e87850
  • 64 Nishida N, Yano H, Nishida T, Kamura T, Kojiro M. Angiogenesis in cancer. Vasc Health Risk Manag 2006; 2: 213-219
  • 65 Saha S, Islam MK, Shilpi JA, Hasan S. Inhibition of VEGF: a novel mechanism to control angiogenesis by Withania somniferaʼs key metabolite Withaferin A. In Silico Pharmacol 2013; 1: 1-9
  • 66 Wang Y-X, Ding W-B, Dong C-W. Withaferin A suppresses liver tumor growth in a nude mouse model by downregulation of cell signaling pathway leading to invasion and angiogenesis. Tropical J Pharmaceut Res 2015; 14: 1005-1011
  • 67 Mohan R, Hammers H, Bargagna-Mohan P, Zhan X, Herbstritt C, Ruiz A, Zhang L, Hanson A, Conner B, Rougas J. Withaferin A is a potent inhibitor of angiogenesis. Angiogenesis 2004; 7: 115-122
  • 68 Bao B, Ahmad A, Azmi AS, Ali S, Sarkar FH. Overview of cancer stem cells (CSCs) and mechanisms of their regulation: implications for cancer therapy. Curr Protoc Pharmacol 2013; 61: 14.25-11-14.25-14
  • 69 Li F, Tiede B, Massagué J, Kang Y. Beyond tumorigenesis: cancer stem cells in metastasis. Cell Res 2007; 17: 3-14
  • 70 Aliebrahimi S, Kouhsari SM, Arab SS, Shadboorestan A, Ostad SN. Phytochemicals, withaferin A and carnosol, overcome pancreatic cancer stem cells as c-Met inhibitors. Biomed Pharmacother 2018; 106: 1527-1536
  • 71 Chen H-C, Chou AS-B, Liu Y-C, Hsieh C-H, Kang C-C, Pang S-T, Yeh C-T, Liu H-P, Liao S-K. Induction of metastatic cancer stem cells from the NK/LAK-resistant floating, but not adherent, subset of the UP-LN1 carcinoma cell line by IFN-γ . Lab Invest 2011; 91: 1502-1513
  • 72 Ting L-L, Chou AS-B, Hsieh C-H, Hsiung S-C, Pang S-T, Liao S-K. Withaferin A targeting both cancer stem cells and metastatic cancer stem cells in the UP-LN1 carcinoma cell model. J Cancer Metastasis Treat 2016; DOI: 10.4103/2394-4722.172008.
  • 73 Young M-J, Wu Y-H, Chiu W-T, Weng T-Y, Huang Y-F, Chou C-Y. All-trans retinoic acid downregulates ALDH1-mediated stemness and inhibits tumour formation in ovarian cancer cells. Carcinogenesis 2015; 36: 498-507
  • 74 Kakar SS, Parte S, Carter K, Joshua IG, Worth C, Rameshwar P, Ratajczak MZ. Correction: Withaferin A (WFA) inhibits tumor growth and metastasis by targeting ovarian cancer stem cells. Oncotarget 2020; 11: 3103
  • 75 Nishi M, Akutsu H, Kudoh A, Kimura H, Yamamoto N, Umezawa A, Lee SW, Ryo A. Induced cancer stem-like cells as a model for biological screening and discovery of agents targeting phenotypic traits of cancer stem cell. Oncotarget 2014; 5: 8665
  • 76 Issa ME, Cuendet M. Withaferin A induces cell death and differentiation in multiple myeloma cancer stem cells. Medchemcomm 2017; 8: 112-121
  • 77 Kim S-H, Singh SV. Mammary cancer chemoprevention by withaferin A is accompanied by in vivo suppression of self-renewal of cancer stem cells. Cancer Prev Res (Phila) 2014; 7: 738-747
  • 78 Whiteside T. The tumor microenvironment and its role in promoting tumor growth. Oncogene 2008; 27: 5904-5912
  • 79 Chung SS, Wu Y, Okobi Q, Adekoya D, Atefi M, Clarke O, Dutta P, Vadgama JV. Proinflammatory cytokines IL-6 and TNF-α increased telomerase activity through NF-κB/STAT1/STAT3 activation, and withaferin A inhibited the signaling in colorectal cancer cells. Mediators of inflammation 2017; DOI: 10.1155/2017/5958429.
  • 80 Draghiciu O, Lubbers J, Nijman HW, Daemen T. Myeloid derived suppressor cells–an overview of combat strategies to increase immunotherapy efficacy. Oncoimmunology 2015; 4: e954829
  • 81 Sinha P, Ostrand-Rosenberg S. Myeloid-derived suppressor cell function is reduced by Withaferin A, a potent and abundant component of Withania somnifera root extract. Cancer Immunology, Immunotherapy 2013; 62: 1663-1673
  • 82 Choi BY, Kim B-W. Withaferin-A inhibits colon cancer cell growth by blocking STAT3 transcriptional activity. J Cancer Prev 2015; 20: 185
  • 83 Straughn AR, Kakar SS. Withaferin A ameliorates ovarian cancer-induced cachexia and proinflammatory signaling. J Ovarian Res 2019; 12: 1-14
  • 84 Cohen SM, Mukerji R, Timmermann BN, Samadi AK, Cohen MS. A novel combination of withaferin A and sorafenib shows synergistic efficacy against both papillary and anaplastic thyroid cancers. Am J Surg 2012; 204: 895-901
  • 85 Royston KJ, Udayakumar N, Lewis K, Tollefsbol TO. A novel combination of withaferin A and sulforaphane inhibits epigenetic machinery, cellular viability and induces apoptosis of breast cancer cells. Int J Mol Sci 2017; 18: 1092
  • 86 Royston KJ, Paul B, Nozell S, Rajbhandari R, Tollefsbol TO. Withaferin A and sulforaphane regulate breast cancer cell cycle progression through epigenetic mechanisms. Exp Cell Res 2018; 368: 67-74
  • 87 Kakar SS, Jala VR, Fong MY. Synergistic cytotoxic action of cisplatin and withaferin A on ovarian cancer cell lines. Biochem Biophys Res Commun 2012; 423: 819-825
  • 88 Kakar SS, Ratajczak MZ, Powell KS, Moghadamfalahi M, Miller DM, Batra SK, Singh SK. Withaferin a alone and in combination with cisplatin suppresses growth and metastasis of ovarian cancer by targeting putative cancer stem cells. PLoS One 2014; 9: e107596
  • 89 Li X, Zhu F, Jiang J, Sun C, Wang X, Shen M, Tian R, Shi C, Xu M, Peng F. Synergistic antitumor activity of withaferin A combined with oxaliplatin triggers reactive oxygen species-mediated inactivation of the PI3K/AKT pathway in human pancreatic cancer cells. Cancer Lett 2015; 357: 219-230
  • 90 Hsu JH-M, Chang PM-H, Cheng T-S, Kuo Y-L, Wu AT-H, Tran T-H, Yang Y-H, Chen J-M, Tsai Y-C, Chu Y-S. Identification of withaferin A as a potential candidate for anti-cancer therapy in non-small cell lung cancer. Cancers (Basel) 2019; 11: 1003
  • 91 Hernandez A, Smith F, Wang Q, Wang X, Evers BM. Assessment of differential gene expression patterns in human colon cancers. Ann Surg 2000; 232: 576
  • 92 Lee T-J, Um HJ, Park J-W, Choi KS, Kwon TK. Withaferin A sensitizes TRAIL-induced apoptosis through reactive oxygen species-mediated up-regulation of death receptor 5 and down-regulation of c-FLIP. Free Radic Biol Med 2009; 46: 1639-1649
  • 93 Patil D, Gautam M, Mishra S, Karupothula S, Gairola S, Jadhav S, Pawar S, Patwardhan B. Determination of withaferin A and withanolide A in mice plasma using high-performance liquid chromatography-tandem mass spectrometry: Application to pharmacokinetics after oral administration of Withania somnifera aqueous extract. J Pharm Biomed Anal 2013; 80: 203-212
  • 94 Devkar ST, Kandhare AD, Sloley BD, Jagtap SD, Lin J, Tam YK, Katyare SS, Bodhankar SL, Hegde MV. Evaluation of the bioavailability of major withanolides of Withania somnifera using an in vitro absorption model system. J Adv Pharm Technol Res 2015; 6: 159
  • 95 Pires N, Gota V, Gulia A, Hingorani L, Agarwal M, Puri A. Safety and pharmacokinetics of Withaferin-A in advanced stage high grade osteosarcoma: A phase I trial. J Ayurveda Integr Med 2020; 11: 68-72
  • 96 Yadav DK, Kumar S, Saloni. Singh H, Kim MH, Sharma P, Misra S, Khan F. Molecular docking, QSAR and ADMET studies of withanolide analogs against breast cancer. Drug Des Devel Ther 2017; 11: 1859-1870
  • 97 Pugsley MK, Authier S, Curtis M. Principles of safety pharmacology. Br J Pharmacol 2008; 154: 1382-1399
  • 98 Arome D, Chinedu E. The importance of toxicity testing. J Pharm BioSci 2013; 4: 146-148
  • 99 Gupta Y. Clinical Trials of Traditional Herbal Medicines in India. 2011
  • 100 Shan JJ, Rodgers K, Lai C, Sutherland SK. Challenges in natural health product research: The importance of standardization. In: Proceedings-Western Pharmacology Society: [Western Pharmacology Society] 1998; 2007: 188
  • 101 Babiker HM, Davis L, Larson K, Placencia C, Swensen C, Tenneti P, Lim M, Cañamar R, Curtis J, Castillo E. A Multidisciplinary Evaluation of Barriers to Enrolling Cancer Patients into Early Phase Clinical Trials: Challenges and Patient-centric Recommendations. Expert Opin Investig Drugs 2019; 28: 675-686
  • 102 Tavhare SD, Nishteswar K, Shukla VJ. Effect of seasonal variations on the phytoconstituents of Aśvagandhā wr to lunar cycles. Anc Sci Life 2016; 35: 150
  • 103 Nair AR, Praveen N. Biochemical and phytochemical variations during the growth phase of Withania somnifera (L.) Dunal . Journal of Pharmacognosy and Phytochemistry 2019; 8: 1930-1937
  • 104 Kumar A, Kaul M, Bhan M, Khanna PK, Suri K. Morphological and chemical variation in 25 collections of the Indian medicinal plant, Withania somnifera (L.) Dunal (Solanaceae). Genet Res Crop Evol 2007; 54: 655-660
  • 105 Shipkowski KA, Betz JM, Birnbaum LS, Bucher JR, Coates PM, Hopp DC, MacKay D, Oketch-Rabah H, Walker NJ, Welch C. Naturally complex: Perspectives and challenges associated with Botanical Dietary Supplement Safety assessment. Food Chem Toxicol 2018; 118: 963-971
  • 106 Jachak SM, Saklani A. Challenges and opportunities in drug discovery from plants. Current science 2007; DOI: 1251-1257
  • 107 Joshi P, Misra L, Siddique AA, Srivastava M, Kumar S, Darokar MP. Epoxide group relationship with cytotoxicity in withanolide derivatives from Withania somnifera . Steroids 2014; 79: 19-27
  • 108 Yu Y, Katiyar SP, Sundar D, Kaul Z, Miyako E, Zhang Z, Kaul SC, Reddel RR, Wadhwa R. Withaferin-A kills cancer cells with and without telomerase: chemical, computational and experimental evidences. Cell Death Dis 2017; 8: e2755
  • 109 Li A-X, Sun M, Li X. Withaferin-A induces apoptosis in osteosarcoma U2OS cell line via generation of ROS and disruption of mitochondrial membrane potential. European Review for Medical & Pharmacological Sciences 2017; 21
  • 110 Malik F, Kumar A, Bhushan S, Khan S, Bhatia A, Suri KA, Qazi GN, Singh J. Reactive oxygen species generation and mitochondrial dysfunction in the apoptotic cell death of human myeloid leukemia HL-60 cells by a dietary compound withaferin A with concomitant protection by N-acetyl cysteine. Apoptosis 2007; 12: 2115-2133
  • 111 Lahat G, Zhu Q-S, Huang K-L, Wang S, Bolshakov S, Liu J, Torres K, Langley RR, Lazar AJ, Hung MC. Vimentin is a novel anti-cancer therapeutic target; insights from in vitro and in vivo mice xenograft studies. PLoS One 2010; 5: e10105

Zoom Image
Fig. 1 Structure of Withaferin A
Zoom Image
Fig. 2 A simplified diagram of the pharmacological effects of WA in cancer at the molecular level. WA induces cell cycle arrest at the G2/M phase by inhibiting the cyclinB1/CDck1 complex upon activation of p21 and Wee1 protein. Caspase mediated apoptosis by WA through PARP cleavage. WA generates ROS results in the activation of JNK and leads to mitochondrial dysfunction. The cells affected by ROS undergo apoptosis. WA inhibits angiogenesis by restricting the binding of the Sp-1 gene to its promoter region of the VEGF gene. Metastasis and tumor invasion are hindered by WA through suppression of EMT and vimentin expression and inhibition of metalloproteinases.
Zoom Image
Fig. 3 Challenges facing WA to enter into a cancer treatment regimen.