Skip to main content

Paclitaxel and its semi-synthetic derivatives: comprehensive insights into chemical structure, mechanisms of action, and anticancer properties

Abstract

Cancer is a disease that can cause abnormal cell growth and can spread throughout the body. It is among the most significant causes of death worldwide, resulting in approx. 10 million deaths annually. Many synthetic anticancer drugs are available, but they often come with side effects and can interact negatively with other medications. Additionally, many chemotherapy drugs used for cancer treatment can develop resistance and harm normal cells, leading to dose-limiting side effects. As a result, finding effective cancer treatments and developing new drugs remains a significant challenge. However, plants are a potent source of natural products with the potential for cancer treatment. These biologically active compounds may be the basis for enhanced or less toxic derivatives. Herbal medicines/phytomedicines, or plant-based drugs, are becoming more popular in treating complicated diseases like cancer due to their effectiveness and are a particularly attractive option due to their affordability, availability, and lack of serious side effects. They have broad applicability and therapeutic efficacy, which has spurred scientific research into their potential as anticancer agents. This review focuses on Paclitaxel (PTX), a plant-based drug derived from Taxus sp., and its ability to treat specific tumors. PTX and its derivatives are effective against various cancer cell lines. Researchers can use this detailed information to develop effective and affordable treatments for cancer.

Introduction

Cancer is a severe public health issue, with around six million new cases yearly. Research has identified several significant causes, including exposure to certain chemicals and types of electromagnetic radiation in the diet, environment, or workplace [1]. The level of risk associated with these exposures is a topic of much debate, prompting preventative efforts such as the US national "Smoke Out" program, which aims to limit exposure to carcinogenic chemicals [2]. Many advancements have been made in developing anticancer medications due to studies on the molecular pathways involved in cancer growth. However, despite these efforts, using chemically made medications has not significantly improved overall survival rates. Treating cancer remains a challenging task with limited success. Available treatment choices comprise surgery, radiation therapy, and systemic chemotherapy. In the chemotherapy drug category, medications like methotrexate (antimetabolites), cisplatin and doxorubicin (DNA-interactive drugs), taxanes (anti-tubulin) are most widely used in addition to other hormones, and molecular targeting drugs [3]. Chemotherapy has several disadvantages, such as cancer recurrence, drug resistance, and harmful effects on healthy tissues, which can hinder the effectiveness of anticancer drugs and negatively affect a patient's well-being. Researchers are always searching for new, improved therapies with fewer side effects to overcome these obstacles and maintain the quality of life for those living with cancer [4].

Medicinal plants have numerous advantages over artificial products, as they are non-toxic to normal human cells. Conventionally, cancer is treated with radiotherapy and chemotherapy, but unfortunately, these methods have adverse side effects that can seriously harm a patient's health. These side effects include neurological, cardiac, renal, and pulmonary toxicity. Therefore, developing an alternative strategy that utilizes anticancer medications that are more effective and less hazardous than those currently available is crucial. The National Cancer Institute, Maryland, USA has examined approximately 35,000 plant species to determine their potential anti-cancer properties. As a result, they have discovered 3000 species having repeatable anticancer efficacy [5]. To develop additional medications for treating this illness, it is indispensable to research the primary anticancer agents that have arisen from natural sources. Medicinal plants contain secondary metabolites such as flavonoids, flavones, anthocyanins, lignans, coumarins, and catechins. These bioactive molecules are responsible for the high levels of antioxidants in medicinal plants [6, 7]. Research on herbal treatments has shifted nowadays due to expensive synthetic drugs and their side effects. Significantly, interest in preventing, eradicating, and treating diseases like cancer and metabolic disorders has risen because of more deaths worldwide [8, 9]. These studies have shown that Taxus sp. and its components, especially Paclitaxel (PTX), have various biological actions.

There are at least 10 diverse species of Taxus, such as T. baccata, T. cuspidata, T. wallichiana, and T. xmedia cv. Hicksii, which contain taxol [10,11,12]. The first instance of Taxol, specifically Taxus brevifolia, was created by Wani and colleagues by utilizing Nutt (Taxaceae) and a synthetic form of the plant [13]. After the finding of Taxol, more than 400 naturally occurring taxol analogs were discovered, leading to the isolation of numerous other taxoids. Typically, Taxus species contain a relatively small amount of taxol, ranging from 0.001 to 0.06% of the dried bark's weight [14]. Taxol is most prevalent in T. brevifolia: bark, needles, roots, branches, seeds, and wood. PTX was first extracted from the stem bark of the western yew, Taxus brevifolia, in 1960s; Wani and colleagues discovered its structure in 1971. Taxol® is a drug currently marketed as PTX. It was approved for clinical use in 1994, with the first medicinal application being for the treatment of ovarian cancer [15]. In 1971, researchers discovered that PTX was the active ingredient in the extract. This drug belongs to the taxane family and has a unique chemical structure (Fig. 1a) with the molecular formula C47H51NO14. Taxus brevifolia, a type of Pacific yew, was originally used to obtain this compound. However many of the species of fungus Pestalotiopsis viz, Pestalotiopsis versicolor, Pestalotiopsis neglecta (isolated from Japanese Yew tree, Taxus cuspidata), Pestalotiopsis pauciseta (isolated from Cardiospermum helicacabum) and Pestalotiopsis terminaliae was extracted from the fresh healthy leaves of Terminalia arjuna (arjun tree) and then examined for the ability to produce the anticancer medication taxol in a synthetic culture medium (Gangadevi [16,17,18]. According to [19], PTX is a vital anticancer medication to treat ovarian, breast, and lung cancer.

Fig. 1
figure 1

Chemical structure of paclitaxel and its derivatives. A Paclitaxel, B 2-debenzoyl-2-trigloyl paclitaxel, C baccatin III, D docetaxel, E cabazitaxel, and F Abraxane

To treat a single patient, approximately 2 g of PTX is requisite, and this amount can be extracted from the 3–10 tree’s bark. On average, taxol has a yield of 0.015% approximately. It takes 2000–2500 yew trees to produce just one kilogram of taxol [20,21,22]. The tree's slow growth would cause its natural stands to be depleted entirely if it were harvested for taxol. Many researchers have used the bark of Taxus spp. instead of needles to obtain docetaxel and Taxotere, rather than PTX, to protect this crucial population [23,24,25,26]. Research on taxane diterpenoids, commonly known as taxoids, has increased over the past 20 years from a small field of natural product chemistry into a nearly $1 billion business [27]. This is because the yew develops slowly, and losing its bark kills the tree. Baccatin III or 10-deacetyl baccatin III can be made widely available as a precursor without adversely hurting the trees. The production of taxol and the recently released taxotere (docetaxel) began with 10-deacetyl baccatin III. As a result, taxol's generic name was decided upon as PTX [28]. Six unique taxane diterpenoids have been found in the seeds of Taxus yunnanesis and Taxus chinensis var. mairei [29].

PTX and its derivative docetaxel prevent cell mitosis by stabilizing the microtubule polymer, which leads to cell death [30, 31]. Due to a unique method of action targeting microtubule assembly, the FDA approved PTX for managing ovarian and breast cancer [32, 33]. Currently, PTX is used to treat breast, ovarian, and non-small cell lung cancer by itself or in combination with other medications [34, 35]. It operates by obstructing microtubules' typical cell-division activity. PTX has anticancer properties, as the National Cancer Institute shows, that examined plant extracts from thousands of different species. PTX slows the advancement of the cell cycle, mitosis, and the proliferation of cancer cells by promoting tubulin assembly into microtubules and inhibiting their dissociation [36]. Contrary to previous tubulin-binding anticancer drugs, this one allows tubulin to assemble into microtubules [37, 38]. Clinical trials are being carried out for degenerative brain diseases, and it is employed in coronary heart disease, skin conditions, renal and hepatic fibrosis, inflammation, and axon regeneration [39, 40]. Beyond oncology, paclitaxel has found important applications in the field of vascular devices such as stents and balloons. The paclitaxel-coated devices are used to address the restenosis in blood vessels following procedures like angioplasty (https://www.fda.gov/medical-devices/cardiovascular-devices/paclitaxel-coated-balloons-and-stents-peripheral-arterial-disease. This strategy improves the effectiveness of treatments and subsequently improves patient outcomes, marking a substantial advancement in the fields of vascular medicine and interventional cardiology.

The fact that PTX is widely disseminated throughout the body volume demonstrates its affinity for the bound albumin protein. The liver mainly carries out PTX's metabolism and involves biliary excretion. When PTX is taken in its entirety, 6–10% of it is found in urine as an unaltered medication, and 70% of it is excreted in feces with the metabolite 6-hydroxy PTX [41]. Despite PTX's impressive anticancer activity, its lesser solubility in water and other solvents restricts its use and bioavailability. However, PTX was blended with  cremophor and dehydrated alcohol at a 50:50 v/v ratio to overcome the solubility problem, but this alteration revealed significant adverse consequences, including hypersensitivity and incompatibility with standard intravenous infusion settings [42]. P-glycoprotein (P-gp), a substance that PTX is a substrate of, expels the drug from cells, leading to the advance of drug resistance [43]. Many P-gp inhibitors were co-administered with PTX to overcome solubility issues, including verapamil [44] and valspodar [45]. However, the outcomes were unsatisfactory due to these inhibitors' toxic effects and ability to alter the pharmacokinetics and biodistribution of PTX.

In addition, this review tries to elucidate the various forms of paclitaxel, their chemical makeup, semi-synthetic derivatives, and how these substances work as anticancer medications. Scientific data supporting their classification as anticancer compounds and their current and historical uses as beneficial remedies are also discussed. The review will contribute to a better understanding of plant-based cancer therapies, including different PTX and its derivatives.

Review methodology

In this review, we analyzed and discussed recent data on PTX's chemo-preventive and chemotherapeutic effects. The most recent studies involving cellular and molecular anticancer mechanisms were reviewed based on pharmacological evidence in specialized databases such as PubMed/MedLine, SCOPUS, Google Scholar, and the TRIP database. A database study revealed that from 1965 to 2024, approximately 400 papers have been published on various aspects of paclitaxel. For the search, we used the following MeSH terms: “paclitaxel/chemistry”, “Taxol”, “Anticancer Chemotherapeutic Agents”, Anticancer therapy use”, “Tumour immunotherapy”,” Taxol/paclitaxel kills cancer cell”, Carcinoma cell line/apoptosis. We included preclinical studies highlighting the mechanisms of action, signaling pathways, and molecular mechanisms of action of andrographolide. The studies that did not have precise pharmacological mechanistic results explained or that used homeopathic remedies were excluded. Plant taxonomy was validated with World Flora Online, and chemical formulations with PubChem [46, 47].

General characterization of PTX and its semi-synthesis derivatives

One of the most important secondary metabolites known to have anticancer properties is taxol, a complex diterpene derived from Taxus spp. with a molecular weight of 853.9 Da. Its chemical name is 5, 20-epoxy-1,2,4,7,13-hexahydroxytax-11-en-9-one-4, 10-diacetate-2-benzoate 13 esters with (2R,3S)-N-benzoyl-3-phenylisoserine [48]. Taxol's structural foundation comprises the A, B, and C ring systems, each containing various functional groups such as two hydroxyl groups, one benzoyl group, two acetyl groups, and an oxetane ring. The C13 side chain, specifically (2′R,3′S)-N-benzoyl-3′-phenylisoserine, also contains hydroxyl and benzoyl functional groups and is connected to the core at C13. Although the ester group at the C2 position has been altered, 2-debenzoyl-2-tigloyl PTX (Fig. 1b), the initial natural derivative of PTX, still displays tubulin binding activity [49]. The PTX molecule is made up of an amide tail and a tetracyclic core known as baccatin III (Fig. 1c). The core rings are referred to as rings A (a cyclohexene), B (a cyclooctane), C (a cyclohexane), and D (an oxetane) in that order.

A derivative of taxane, called PTX-TTHA (PTX-triethylenetetramine hexaacetic acid conjugate), was created using PTX, triethylenetetramine hexaacetic acid, dimethylaminopyridine, and triethylamine. This semi-synthetic, water-soluble derivative showed improved PTX cosolvent toxicity, better water solubility, and efficacy against triple-negative breast cancer. PTX-TTHA caused cell apoptosis, decreased cell proliferation, and mediated TUNEL-positive apoptotic cells [51]. Additionally, docetaxel (Taxotere®), a semi-synthetic alternative to PTX, was found to be highly practical and, in some cases, more effective than PTX. The FDA has approved Docetaxel as a medication (Fig. 1d) for head and neck cancer, advanced breast cancer, and metastatic hormone-refractory prostate cancer (HRPC) treatment. Despite being significant drugs for treating various cancers, PTX and docetaxel are less effective due to drug resistance. Both drugs are vulnerable to multidrug resistance.

An increased expression in taxanes' resistance is done by the multidrug resistance gene, which encodes the P-glycoprotein gene, mostly related to increased expression. Cabazitaxel (Fig. 1e) is superior to PTX and docetaxel owing to methoxy groups' presence at C7 and C10, which results in a lower affinity for P-gp. Due to its enhanced properties, this drug effectively treats tumors resistant to docetaxel [52]. If a patient with metastatic HRPC has previously used docetaxel–prednisone therapy, it is recommended to use prednisone and cabazitaxel for treatment [52]. The clinical advantages of cabazitaxel's unique capacity to cross the blood–brain barrier (BBB) have not yet been studied [53]. Cabazitaxel significantly improved the cytotoxicity in docetaxel-sensitive cell lines, including lymphoblastic leukemia, promyelocytic leukemia (HL60), cervical adenocarcinoma (KB), and breast cancer (Calc18). The drug also proved effective in cancer cell lines that had previously been resistant to docetaxel, such as document number 1/DOX, document number 1/TXT, document number 1/VCR, HL60/TAX, Calc18/TXT, and KBV1 [54]. The resistance factor ratios of docetaxel were between 4.8 and 5.9, while those of capaztaxel ranged from 1.8 to 10.

A nanoparticle albumin-bound PTX, Abraxane (Fig. 1f), is a unique PTX version that does not contain CrEL. The particles of PTX are stabilized by human albumin 130 nm in size, making it safe for intravenous administration without the possibility of capillary occlusion [55]. To prepare Abraxane, it can be mixed with normal saline in doses ranging between 2 and 10 mg/mL. This differs from CrEL-PTX, resulting in a smaller infusion volume and duration [56, 57]. Additionally, unlike CrEL-PTX [58], Abraxane does not run the hazard of plasticizer leakage from infusion bags and can, therefore, be produced in standard plastic IV infusion bags.

PTX can be conjugated with biodegradable polymer Poly(l-glutamic acid) a water-soluble, having carboxylic acid side chains (Fig. 2a). The resulting conjugate is extremely water-soluble (> 20 mg/kg) and does not require CrEL for formulation. In chemotherapy-naive patients with advanced NSCLC, PG-PTX revealed equal effectiveness with less myelotoxicity but higher neurotoxicity than gemcitabine or vinorelbine [59]. When used as second-line therapy for NSCLC, PG-PTX exhibited survival rates that were comparable to those of docetaxel while having higher rates of neurotoxicity and lower incidences of alopecia, neutropenia, and febrile neutropenia. Utilizing the identical isoserine C13-side chain as SB-T-1214, a library of 7, 10-modified PTX, cabazitaxel, and ortataxel analogs was published [60], several of these taxanes showed outstanding to good efficacy against various cancer cell lines. Numerous "abeo-taxanes" (Fig. 2b) have been produced by altering the C7- and C9-hydroxyl groups. These taxane skeletons are derived from baccatin III via skeletal rearrangement. These abeo-taxanes showed good potency when applied to cancer cell lines resistant to PTX, vinblastine, and doxorubicin [61].

Fig. 2
figure 2

Chemical structure of poly(l-glutamic acid) paclitaxel (PG-paclitaxel) (A); conjugates abeotaxane (B); biotin-taxoid (SB-T-1214) (C)

A self-immolating disulfide linker was used to create biotin-taxoid (SB-T-1214) conjugates (Fig. 2c), including single-walled carbon nanotube (SWNT) nano-conjugates [62, 63]. These conjugates demonstrated remarkable efficiency with far lower toxicity to normal human cells and very good internalization of cancer cells via receptor-mediated endocytosis.

Many PTX and docetaxel ferrocenyl derivatives have been designed by replacing the 3′-N-benzoyl group of PTX with a ferrocenoyl moiety, resulting in enhanced anti-proliferative property of the derivative as compared to the parent compound. To synthesize PTX derivatives (2), the reaction of optically pure (3R,4S)-3-triethylsilyloxy-4-phenylazetidin-2-one (3) (shown in Fig. 4) and 10-deacetyl baccatin III (1) with trimethylsilyl chloride in pyridine was employed which was followed by selective O-acetylation of 10-OH with LiHMDS and acetyl chloride in THF at − 40 °C for 30 min (Fig. 3).

Fig. 3
figure 3

Synthesis of paclitaxel derivative (2) (reaction condition i) TESCl, pyridine, room temp., 5 min; (reaction condition 2) LiHMDS, CH3COCl, THF, − 40 °C, 30 min

Further, (3R,4S)-N-Ferrocenoyl-4-phenyl-3-triethylsilyloxyazetidin-2-one (4) was synthesized by N-acylation reaction of (3) with ferrocenoyl chloride. The N-acylation of (3) with 4-ferrocene butyric acid using diisopropyl carbodiimide functioning as a coupling agent with a catalytic amount of 4-dimethylamino pyridine in dichloromethane at room temperature resulted in conversion to N-4-ferrocenyl butyryll-3-triethylsilyloxyazetidin-2-one (5) (Fig. 4).

Fig. 4
figure 4

Synthesis of N-ferrocenyl-substituted azetidin-2-ones (4) and (5). Reaction conditions: (i) FcCOCl (Fc = ferrocenyl), Et3N, DMAP, DCM, room temperature, 2 h; (ii) Fc(CH2)3COOH, DIC, DMAP, DCM, room temperature, 24 h

Furthermore, under specific conditions with LiHMDS as a base at − 40 °C, 13-O-acylation of (4) (shown in Fig. 4) with azetidine-2-ones (6) and (3) (shown in Fig. 4) produced the corresponding PTXs (7) and (8) derivatives, and deprotection of -OH groups using a greater quantity of HFpyridine produced (9) and (10) PTX analogs (Fig. 5).

Fig. 5
figure 5

Synthesis of N-debenzoyl-N-ferrocenoylpaclitaxel derivatives, (9) and (10). (i) LiHMDS, THF, − 40 °C, 40 min; (ii) HF·Py, pyridine/MeCN, room temperature, 24 h

Substitution of ferrocenyl moiety linked to the PTX 2′-OH group led to compounds with lower toxicity than that of the PTX. Anti-proliferative activity was decreased on substitution at the 7′-OH group compared to the parent compound. The synthesized derivatives (16–19) and (25–26) possessed lower activity than the PTX parent compound (Figs. 6, 8).

Fig. 6
figure 6

Synthesis of 2′-O-ferrocene-substituted paclitaxel 13–16 and docetaxel 17–20 derivatives. Reaction conditions: (i) FcR3COOH, DIC (1.5 equiv), DMAP (0.1 equiv), DCM, room temperature, 24 h

For substitution at the 7′-OH position, (11) and (12) compounds were protected at the 2′-O-position as tert-butyldimethylsilyl ethers with tert-butyldimethylsilyl chloride in the presence of imidazole in DMF at room temperature [64]. The compound synthesized (21) was selectively 7-O-acylated with 3-ferrocenoyl propionic acid or 4-ferrocenyric acid using DIC as a coupling agent at 0 °C resulting in corresponding products 22–23 (Fig. 7).

Fig. 7
figure 7

Synthesis of 2′-O-TBS ethers of paclitaxel 11 and docetaxel 12. Reaction conditions: (i) TBSCl, imidazole, room temperature, 24 h

The 7-O-acylated-2′-O-TBS-docetaxel derivatives (23) and (24), shown in Fig. 7, were produced by reacting 2′-O-TBS-docetaxel (22) (shown in Fig. 7) using 5-ferrocenoylpentanoic and 6-ferrocenylhexanoic acids as acylating agents at 0 °C. Moreover, the deprotection of hydroxy groups was done using HF·Py, which led to the synthesis of the 7-O-ferrocenyl-substituted taxanes (25–26), respectively [65,66,67,68] (Fig. 8).

Fig. 8
figure 8

Synthesis of docetaxel derivatives (25) and (26). Reaction conditions: (ii) HF·Py/pyridine/MeCN, room temperature, 24 h

[69] prepared a water-soluble ester-linked glucoside derivative of PTX, in which diols anomers of allyl 2,3,4-tri-O-benzyl-6-O-tritylglycoside were synthesized, followed by their chromatographic separation (Fig. 9).

Fig. 9
figure 9

Synthesis of paclitaxel-2′-O-3-pentadecylhemiglutarate

7-glycolyl PTX 2′′-O-α-maltoside (Fig. 10a), an ester-linked PTX-glycoside conjugate, has been synthesized by condensing 2′-TES PTX with α-glycosyloxy acetic acid and then deprotecting the hydroxy groups. A PTX dicarboxylic acid derivative was synthesized for specific binding to ubiquitous protein, serum albumin. In the synthesis, hexadecanol was oxidized to palmitaldehyde using PCC and then Wittig olefinated. The synthesized triester was decarboxylated and saponified employing KOH, resulting in an activated anhydride of 3-pentadecyl glutaric acid. The synthesized derivative showed higher cytotoxicity, high serum stability, and efficiency than PTX [70]. Li created NucA-PTX, a water-soluble PTX-nucleolin-aptamer combination, to precisely deliver PTX to the ovarian cancer tumor spot. A more durable and inactive dipeptide bond sensitive to cathepsin B joins the tumor-targeting nucleolin aptamer to the active hydroxyl group at the 2′-position of PTX [71].

Fig. 10
figure 10

Chemical structure of 7-glycolypacilitaxel (A), DHA-paclitaxel (B), larotaxel (C), milataxel (D), ortataxel (E), tesetaxel (F)

Over the past 10 years, taxane liposomal formulations have undergone substantial research and development [72]. For instance, clinical studies employing the PTX-loaded cationic liposome EndoTAG-1 and the PTX-entrapped liposome LEP-ETU have shown promising results. New-generation taxoids carrying DHA at the C2′ position have been generated with the creation of DHA-PTX (Taxoprexin; Fig. 10b). Some of these taxoids showed promising activity against (Pgp +) DLD-1 human colon and (Pgp-) A121 human ovarian cancer xenografts in mice, as well as substantially less toxicity than the parent taxoids in terms of systemic toxicity [73].

The creation of PTX prodrugs has considerably used the C2 position to boost water solubility and anticancer effectiveness. The effects of several PTX conjugates, including docosahexaenoic acid (DHA)-PTX ("Taxoprexin") and poly (l-glutamic acid) PG-PTX ("Opaxio"), are now being researched in humans [74]. In contrast to PTX and DHA-PTX ("Taxoprexin") and PTX, DHA-SB-T-1214 had a significant antitumor impact on mouse xenografts of the DLD-1 human colon, H460 human non-small cell lung, CFPAC-1, and PANC-1 human pancreatic cancer [75].

Currently being evaluated in clinical settings are the PTX analogs larotaxel, milataxel, ortataxel, and tesetaxel (Fig. 10c–f). Larotaxel is being evaluated in clinical studies for metastatic breast cancer, advanced NSCLC, urethral bladder cancer, and advanced pancreatic cancer [76,77,78]. Larotaxel plus cisplatin did not outperform cisplatin/gemcitabine in a Phase III trial for locally advanced or metastatic bladder or urothelial tract cancer [79]. For NSCLC resistant to taxanes, recurrent glioblastoma, and metastatic breast cancer, Ortataxel is now undergoing phase II research [80]. Tesetaxel has already finished Phase I and II trials in solid tumors [81, 82]. Milataxel was promising in a distinct study of individuals with platinum-refractory NSCLC [83]. However, it was unsuccessful in a Phase II trial for advanced previously treated colorectal cancer. When given at a 60 mg/m2 dose, BMS-184476 was well tolerated and effective against NSCLC in patients with previous therapy [84].

Mechanism of antitumor action of paclitaxel

Stabilization of the microtubule

Paclitaxel (PTX) primarily targets the microtubules (microtubule targeting agent; MTA), the cytoskeletal architecture of the cells, that play critical roles in cellular processes such as cell cycle progression and division, motility, and intracellular trafficking [33, 85, 86]. The net microtubule assembly rate equals the net disassembly rate during steady-state conditions, and the length of the microtubule is unchanged [33]. Microtubules are assembled from α- and β-tubulin heterodimers in a head-to-tail pattern (rapidly growing '+ve end' at one side and slower growing '−ve end' at the other) during the G2 phase and the prophase of mitosis and its disassembly (a process called dynamic instability) requires GTP hydrolysis [33, 85]. PTX blocks depolymerization of the microtubule during cell cycle progression, specifically by binding to the N-terminus of the β-tubulin subunit, thereby stabilizing the polymerized microtubule, causing cell cycle arrest at G2/M-phase, causing non-progression of the cell cycle and subsequently leading to apoptosis [86,87,88,89] [Fig. 11-(1)].

Fig. 11
figure 11

Mechanism of anticancer action of Paclitaxel (PTX): PTX intervention leads to (1) stabilization of microtubule, cell arrest, and subsequent apoptosis, (2) inhibition of the TLR4 signaling pathway, (3) increase in the immunomodulatory effects of the drug and (4) activates ER stress-mediated cell death in different cancers. BAK: Bcl-2 homologous antagonist/killer; BAX: Bcl-2 associated X protein; Bcl-2: B cell lymphoma 2; Cyt C: cytochrome C; eIF2α: eukaryoic translation initiation factor 2 alpha; ER: endoplasmic reticulum; IRAK: interleukin 1 receptor-associated kinase; MAPK: mitogen-activated protein kinase; MEK: MAPK/extracellular signal-regulated (ERK) kinase; MyD88: myeloid differentiation primary response protein 88; NFκB: nuclear factor light chain enhancer of kappa; PERK: PRKR-like endoplasmic reticulum kinase; PTX: paclitaxel; TLR4: toll-like receptor 4; TRAF: tumor necrosis factor (TNF) receptor-associated factor; TRIF: TIR domain-containing adaptor protein. Created with Biorender.com

Interestingly, PTX stabilized microtubule formation in vitro and was resistant to low-temperature or calcium-triggered depolymerization [34, 90]. PTX intervention caused mitotic arrest in cell cultures and animal tumor models [34]. The findings suggest that PTX-arrested cells in the metaphase and the cells maintain near-normal bipolar spindles [32]. Studies have also revealed that low PTX concentrations blocked the depolymerization of microtubules. In contrast, at higher concentrations, it increased the stability of microtubules and inhibited the separation of microtubule '-ve' ends from centrosomes [91,92,93]. The efficacy of PTX as an MTA is reduced when cells confer resistance to the drug through premature mitotic exit (mitotic slippage), thus evading the PTX-induced cell cycle arrest and subsequent apoptosis [94].

Effect on the TLR4 pathway

Concentration and time-dependent anticancer and pro-apoptotic effects of PTX, independent of its effect on microtubule depolymerization, have been reported. PTX modulates the transcription of several genes, directly or indirectly involved in cell proliferation, apoptosis, and inflammation via dysregulation of the TLR4 (toll-like receptor 4) pathway, which in turn can be either via MyD88 (myeloid differentiation primary response protein 88) dependent and/or independent pathways [95]. In normal cells, the TLR4 pathway plays an integral role in cellular defense/survival mechanisms, pathogen recognition, and pattern recognition, activating innate immunity and eliciting immune responses [96,97,98]. Active TLR4 signaling, initially identified in breast cancer cells, has been implicated in the chronic inflammation-mediated development of different cancers, cancer progression, chemotherapeutic resistance, cancer cell stemness, invasion, metastasis, and disease relapse [95, 99,100,101,102]. The activation of TLR4 mediated MyD88-dependent pathway subsequently activates several pro-oncogenic and anti-apoptotic signaling mechanisms in cancer cells that include the Raf1/MEPK/MAPK pathway (associated with cell survival) and the IRAK/TRAF/NFκB pathway (associated with the synthesis and secretion of pro-inflammatory cytokines) [86, 98] [Fig. 11-(2)]. The MyD88-dependent pathway also activates the MAPK pathway in cancers that contribute to cancer cell proliferation, resistance to programmed cell death, and synthesis of pro-inflammatory cytokines (via AP-1 activation) [103, 104] [Fig. 11-(2)]. The TRIF (MyD88-independent) mediated mechanism of the TLR4 pathway results in the phosphorylation and activation of transcription factor IRF3, which then translocates to the nucleus and drives the expression of Type 1 interferons [98].

PTX intervention effectively blocks the MyD88-dependent and independent cancer-promoting TLR4 mechanisms and trigger apoptosis (via upregulation of pro-apoptotic BAX/BAK and downregulation of anti-apoptotic Bcl-2), reduce chronic inflammation, and activate several immunomodulatory tumor combating effects of PTX (Figs. 11-(3) and 12) [33, 86].

Fig. 12
figure 12

Antiangiogenic and immunomodulatory effects of Paclitaxel (PTX): PTX exerts its anticancer activity via its antiangiogenic and immunomodulatory effects. CTL: cytotoxic T-lymphocyte; DC: dendritic cells; M1: M1 macrophages; MDSC: myeloid-derived suppressor cells; NK1: natural killer type 1 cells; Th1: Type 1 T-helper cells; Treg: regulatory T cells; VEGFR2: vascular endothelial growth factor receptor 2. Adapted and modified from Kampan et al., 2015 [33]. Created with Biorender.com

However, PTX-mediated modulation of the TLR4 pathway is implicated in the activation of NF-κB and MAPK-related downstream signaling and subsequent release of pro-inflammatory molecules that enhance the progression of cancers and confer chemoresistance to drug intervention in cancers [105, 106]. PTX at lower doses induces cytokines and pro-inflammatory proteins and apoptosis at higher doses. Hence, the dose-dependent effects of PTX in different cancers must be carefully addressed to maximize the anticancer efficacy of the drug [107].

Paclitaxel-mediated activation of ER stress

Certain studies have implicated the PTX-mediated activation of the endoplasmic reticulum (ER) stress response or the unfolded protein response (UPR) via the PERK and IRE1α [Fig. 11-(4)], subsequent apoptotic cell death in different cancers [108, 109]. PTX chemotherapy triggers the activation of antitumour immunity through immunogenic cell death via TLR4 and enhances the expression of CALR mediated by CCL2 transcription and IκB kinase-2 SNARE-dependent exocytosis resulting in activation of NF-κB signaling pathway [108]. Combinational therapy of apatinib and PTX induces ER stress, autophagy and apoptosis in ECA-109 and KYSE-150 esophageal squamous cancer cells (ESCC). Further, combination of apatinib and chloroquine enhances the sensitivity in ESCC which in turn triggers PTX for apoptosis through IRE-1α-AKT-mTOR signaling pathway [109]. In conclusion, PTX in combination with other drugs mediates the ER stress response via PERK, IRE1α and NF-κB signaling pathway resulting in apoptosis and immunogenic cell death of tumor cells.

Other anticancer effects of paclitaxel

Several studies have reported the solid angiogenic inhibitory activity of PTX at low doses by modulating VEGF expression and the VEGF signaling pathway by downregulating VEGFR2 [33, 110]. One of the mechanisms of action of PTX involves tumor cell death via the generation of reactive oxygen species (ROS) [86]. The combinational therapy of PTX with glucose inhibitors, such as 2-deoxy D-glucose and hydro peroxide (L-buthionine-S, R-sulfoximine), selectively increased hydrogen peroxide mediated breast cancer cell death [111].

There are many challenges in understanding the detailed and precise action of PTX and the specific relevant concentration for using PTX in cell culture. A probable reason could be varying usable concentrations of PTX in diverse cancer types and chemotherapy-associated interventions. The changing PTX levels in plasma and excess PTX accumulation in cell lines reflect greater concentration in cells than in plasma; hence, it is not easy to measure its effective concentration [32]. Instinctively, how PTX interferes specifically at interphase without affecting prior mitosis is still unclear. Some authors hypothesized that PTX might have interfered with cell signaling and microtubule-mediated transport. The microenvironment of cell culture has a profound effect on PTX antitumor activity, e.g., in the context of drug testing, clinically relevant amounts of PTX do not cause death in cells at interphase and have not gone through mitosis [112].

Scientific studies on the antitumor effect of PTX

Non-small-cell lung cancer (NSCLC) is one of the most common cancers in the USA, accounting for 85% of lung cancer cases. In 1999, PTX was recognized as an FDA-approved drug for NSCLC. Mohiuddin et al. examined the underlying mechanisms of PTX's inhibitory effect on gefitinib-resistant NSCLC cells (PC9-MET). The results demonstrate that PTX significantly reduced the PC9-MET cell viability and apoptosis induction. The apoptotic impact was also accompanied by enhanced cleaved caspase-3, 9, and PARP levels. PTX augmented oxidative stress by enhancing ROS production, which in turn caused DNA damage in tumor cells. PTX eliminated cellular senescence related to the inactivation of p53/p21 and p16/pRb signaling pathways. The authors concluded that PTX is a hopeful antitumor drug offering a new therapeutic approach for managing gefitinib-resistant NSCLC during the COVID-19 pandemic [113].

In another study, using a time-dependent approach, PTX nanoparticles loaded with polylactic-co-glycolic acid were employed to observe their antitumor effect on NSCLC cells in vitro. The authors demonstrated that PTX nanoparticles inhibited A549 and H1650 cell activity. Although the inhibitory activity was less at 12 and 24 h with the progression of time, a potent inhibition occurred at 48 and 72 h. The nanoparticles were more effective in triggering apoptosis, blocking invasion, and migrating NSCLC cells than normal PTX. The sustained release with more efficient cellular uptake made PTX nanoparticles a hope as a promising drug carrier in lung carcinoma [114].

The PTX chemotherapy is extensively implemented to manage several tumors listed as breast, ovarian, and NSCLC. Nowadays, combinational therapy has been effective, overcoming many challenges associated with single-drug chemotherapy. Kim et al. [115] employed a combinational approach to study the antitumor activity of PTX with sorafenib and radiation in vitro and in vivo in anaplastic thyroid cancer (ATC) cells. The authors concluded that a combination of synergistically in vitro lowered the cell viability of tumor cells and increased cell apoptosis. The xenograft model reported a significant decline in tumor volume and enhanced survival rate, representing it as a potential therapy in preclinical models.

Another combinational therapy by Khing et al. [116], in which PTX was given in adjunction with fluoxetine, was evaluated for antitumor activity in gastric adenocarcinoma cells. The combination resulted in the G2/M-phase arrest and triggered early and late cell death plus necroptosis in a time and dose-dependent fashion.

[117] studied the caffeic acid and PTX (in combination) effect on NSCLC cells both in vivo and in vitro. Co-treatment showed that caffeic acid enhanced the cytotoxicity of PTX in H1299 cells at low concentrations but not in Beas-2b cells.

The cells H1299 were arrested at the sub-G1 phase and triggered caspase-3, 9 followed by apoptosis. Caffeic acid improved the phosphorylation of c-Jun NH2-terminal protein kinase1/2, Bax, and Bid, and their activation. Additionally, in vivo, study reported that PTX and caffeic acid suppressed the tumor growth in the H1299 xenograft model without any adverse effects.

The synergistic influence of silibinin and PTX on ovarian cancer has been investigated by [118] in ovarian carcinoma cell lines SKOV-3. Results revealed a considerable slowing of the SKOV-3 cells' development followed by induction of apoptosis. Tumor suppressor genes p53 and p21 upregulation is reported along with a crosswalk between PTX, silibinin, and cancer via computational analysis.

In human prostate cancer, a combination of PTX and noscapine was analyzed for antitumor properties in vitro. The tumor cells’ viability declined, improved apoptosis, decreased expression of Bcl-2, and increased Bax and Bcl/Bax ratio LNCaP and PC-3 cells. The expression of androgen receptor and prostate-specific antigens declined in LNCaP cell lines [119]. Han et al. [120] reported that a combination of PTX and ruxolitinib synergistically enhanced the antitumor property of an antineoplastic agent and suppressed tumor growth in the human ovarian mouse model. However, a recent study on ovarian cancer cell lines MES synergistically demonstrating the effect of low-dose PTX with Asparagus officinalis revealed that the combination of congested cell proliferation and cell invasion triggered apoptosis. The mechanism of action was DNA-dependent damage, suppression of microtubule dynamics and associated proteins, and AKT/mTOR pathway interference [121]. Trastuzumab, a humanized anti-human epidermal growth factor receptor 2 antibody drug, when given in combination with PTX enhanced the antitumor efficacy of trastuzumab-resistant in resistant and sensitive xenografted tumors. The combination resulted AKT–p27kip1–retinoblastoma protein pathway and apoptosis [122].

Researchers have also prepared nano-formulations of PTX to enhance the drug's bioavailability, specificity, and antitumor activity in different types of cancers. Huang et al. [123] encapsulated PTX with PEG-PLA/TPGS and found the PTX-micelles to improve the anticancer property of PTX in A549 non-small lung cancer cells. The xenograft model studies on nude mice revealed that PTX micelles could block tumor growth more efficiently than other formulations. Leiva et al. [124] formulated PTX nanoparticles with glyceryl tripalmitate (tripalmitin), including adjustments by adding hexa (ethylene glycol), β-cyclodextrin, and macelignan. All the nano-formulations reported excellent hemocompatibility and improved antitumor activity in breast and lung cancer cells. Tripalm-NPs-PC declined IC50 by 40.5 and 38.8-fold in breast and lung cancer, respectively. Moreover, the exact formulation reduced the breast volume and lung multicellular tumors. The authors concluded that Tripalm-NPs-PC enhanced the antitumor property and is an alternative and practical PTX delivery system in lung and breast cancer.

Traditional medicine versus standard clinical practice: current medical applications

The discovery of PTX was a significant breakthrough in cancer treatment. Today, PTX is commonly used as a chemotherapy drug and is available in various formulations for different administration routes, including intravenous and oral administration. PTX has played a vital role in cancer treatment and has significantly improved patient outcomes in several cancer types (Table 1 and Fig. 13). It is the principal taxane-derived antineoplastic drug used in the cancer treatment [125]. The PTX detection involved screening 35,000 medicinal plants by U.S. National Cancer Institute (1958) for cytotoxic efficacy. Later, in 1971, it was extracted from the Taxus brevifolia bark [126]. PTX is regarded as a gold standard chemotherapeutic drug for healing different cancer types, such as ovarian, breast, urothelial, head and neck, Kaposi's sarcoma, and non-small cell lung carcinoma [127, 128]. To enhance its therapeutic efficacy and overcome limitations, novel drug formulations incorporating PTX, such as polymeric micelle nanoparticles, have been developed and applied in anticancer curing regimes. PTX also exhibits radiation-sensitizing effects.

Table 1 Medical applications of paclitaxel for curing many diseases
Fig. 13
figure 13

Paclitaxel vital role in several cancer types

The discovery of PTX from plant screening led to its inclusion in clinical trials, and it became the only plant-derived drug to be enlisted in such trials [129]. A unique mechanism of PTX action, targeting microtubule assembly, was discovered in 1979 and approved by the Food and Drug Administration in 1992 and 1994 for ovarian and breast cancer [129]. At the moment, PTX is used individually or in combination with supplementary medications to treat breast, ovarian, and non-small cell lung cancer [129, 130].

PTX is a potent anticancer drug frequently utilized in ovarian cancer prevention, both in adjuvant and advanced settings, often administered in combination with additional chemotherapy drugs like carboplatin and doxorubicin or cyclophosphamide. The usage of PTX in ovarian cancer treatment has been well-established and is supported by several clinical trials and guidelines [131]. PTX-based regimens have been extensively studied and recommended as a standard option for the prevention of breast as well as metastatic cell lung cancer [132]. It is often administered in amalgamation with platinum-based chemotherapy drugs like cisplatin or carboplatin. PTX-based regimens were evaluated in clinical trials and are commonly recommended for the management of the NSCLC [133]. PTX has demonstrated activity against Kaposi sarcoma, a type of cancer often occurring in people with debilitated immune systems, like HIV/AIDS. It can also be used alone or with other medications to treat Kaposi sarcomas [134].

As a chemotherapeutic agent, it can be practiced to cure various cancers like breast, ovarian, lung, pancreatic, and solid tumors. While PTX is an effective chemotherapeutic agent, it can cause several adverse effects, including decreased red blood cells, white blood cells, and platelets (bone marrow suppression), peripheral neuropathy, myalgia (muscle pain), arthralgia (joint pain), gastrointestinal disturbances, and hair loss [135].

PTX derived from endophytes of different plant species for improved production

The challenges associated with sluggish plant growth (Taxus) and the limited PTX yield have prompted the search for alternative strategies for PTX production. Over the past 40 years, various biotechnological approaches have been developed to address these challenges. These approaches include field cultivation, chemical synthesis, cell suspension, callus, hairy root, and tissue culture [136,137,138]. While these biotechnological methods have shown promise, they have not been widely adopted for large-scale PTX production due to several limitations. These limitations include massive reaction steps, lengthy incubation times, and low yields, which make these methods impractical for meeting the increasing demand for PTX. Researchers have been working to overcome these limitations and develop more efficient and scalable strategies for PTX production [139,140,141].

In recent years, endophytes have gained the attention of researchers for their potential in PTX production. Endophytes can be sequestered from various host plant species belonging to different families. They are in diverse ecological and geographical conditions, indicating their adaptability to various environments. Endophytes possess the genetic machinery necessary for PTX biosynthesis and modulate the gene expression in secondary metabolite biosynthesis pathways. Endophytes can potentially overcome the limitations associated with traditional methods of PTX production, such as low yields and long incubation times. However, the exploration of endophytes for PTX production is still in its early stage, and vital research efforts are required to harness their potential fully [142].

Researchers have discovered over 35 species of endophytic fungi proficient in producing PTX. Some of these species include Glomerella cingulata, Pestalotiopsis terminaliae, Fusarium oxysporum, Nigrospora sphaerica, Lasiodiplodia theobromae, Colletotrichum gloesporioides, Phyllosticta tabernaemontanae, Pestalotiopsis microspora, Chaetomella raphigera, Alternaria alternata, and Cladosporium oxysporum [143, 144]. PTX can also be created by endophytes connected to a variety of other plant groups in addition to those belonging to the Taxaceae family, which are known for producing the drug, such as Rubiaceae, Rutaceae, Rhizophoraceae, Solanaceae, Sapindaceae, Plantaginaceae, Podocarpaceae, Pinaceae, Malvaceae, Magnoliaceae, Moraceae, Lamiaceae, Ginkgoaceae, Combretaceae, Cupressaceae, Acanthaceae and Araucariaceae [144].

Several species of Aspergillus, including A. terreus, A. fumigatus, A. niger, A. aculeatinus and A. oryzae, have been specified as PTX producers. Notably, A. fumigatus is confirmed as a high PTX-producing species on the S7 medium. Other PTX-producing fungi includes Beauveria sp., Mycelia sterilia, Epicoccum sp., Fusarium sp., Stemphylium sedicola, Alternaria sp., Cladosporium sp. and Paraconiothyrium variabile. These fungi have been sequestered from diverse host plants and demonstrated PTX production under specific culture conditions [145] (Table 2).

Table 2 Paclitaxel isolated from endophytes of different host plant species

To test the activity, studies by [146, 147] suggest that low concentrations of PTX derived from endophytic fungi can effectively inhibit cell proliferation during mitosis by stabilizing the spindle fibers. It promotes cell death in various cancer cell lines, encompassing lung (HL251), breast (MCF-7, BT220), intestine (Int4070), colon (H116), and leukemia (HLK210) [148] (Table 3).

Table 3 Anticancer properties of paclitaxel derived from endophytes through in vitro study

Rajendran et al. [149] extracted endophytic fungi (Pestalotiopsis microspore) from Plectranthus amboinicus, producing a better yield of PTX, which showed cytotoxic activity against the Hep G2 cell line. The PTX effects (Pestalotiopsis pauciseta) on the growth of MCF-7 breast cancer cells were examined by [150]. They observed that PTX exhibited a higher cytotoxic effect on MCF-7 cells. Ismaiel et al. [151] identified A. tenuissima and A. fumigatus isolated from Terminalia arjuna and Taxodium distichum as producers of PTX. Various human cancer cell lines (HepG-2, A-549, MCF-7, CHO-K1, and HEp-2) were subjected to the MTT assay, and this fungus-derived PTX demonstrated robust action against them. Raj et al. [152] demonstrated that Cladosporium oxysporum-derived PTX inhibited HCT-15 cell growth at higher concentrations. They hypothesized that this fungus might serve as a different source of PTX.

[153] isolated the PTX (Fusarium solani) from T. celebica. They observed that PTX-induced induced DNA fragmentation, caspase-10 activation, apoptosis, and mitochondrial membrane potential loss. Suresh et al. [154] treated NCI-H460 cells with fungal-derived PTX from A. oryzae. The treatment resulted in alterations in cell structure, with the cells becoming sphere-shaped. Fungal PTX displayed significant in vitro cytotoxic activity, inducing apoptosis. It is important to remember that this research may utilize different techniques, cell lines, and particular doses. However, taken as a whole, they show that PTX can kill different cancer cell lines. Matrix metalloproteinase 9 (MMP9) activity is decreased by PTX treatment, which is identified to play a crucial control over the tumor microenvironment and the development of cancer [155]. Inhibiting MMP9 activity can help to impede cancer progression.

Fungal-derived PTX also prompts the initiation of cytotoxic activity in mice [156]. The effects of PTX on Sprague Dawley rat breast tissue generated from Botryodiplodia theobromae were discovered by [157]. They noticed that the levels of antioxidants such as catalase (CAT), glutathione peroxidases (GPx), and superoxide dismutase (SOD) were raised by PTX. PTX also increased the levels of non-enzymatic antioxidants like glutathione (GSH), vitamin C, and vitamin E. Superoxide dismutase (SOD) is a tumor suppressor protein as well as an intracellular enzyme that guards cells from oxidative damage and breaks down superoxide radicals into hydrogen peroxide [158]. By increasing the levels of antioxidants (CAT, SOD, and GPx), fungal PTX helps to block ROS and their cell-induced effects [159]. In Sprague Dawley rats, COX-2 (cyclooxygenase-2) is recognized for stimulating cancer cell growth. PTX considerably declines the COX-2 expression [157]. By reducing COX-2 levels, fungal PTX may hinder cancer cell growth and enhance the cancer cell's susceptibility to undergo programmed cell death. These findings suggest that fungal-derived PTX exhibits various beneficial effects, including inhibition of MMP9 activity, induction of caspase 3-mediated cytotoxicity, modulation of antioxidant levels, and suppression of COX-2 expression. These effects collectively contribute to the potential anticancer properties of fungal PTX.

Conclusion

This article aims to give an overview of the molecular and pharmacological aspects of PTX's ability to fight cancer. PTX (marketed as Taxol®) is a tetracyclic diterpenoid that was initially discovered in the bark of Taxus brevifolia, a Pacific yew tree. It is the first taxane to undergo clinical trials and is an active chemotherapy drug against a wide range of cancers, typically resistant to conventional treatments. The US FDA in 1992 approved it for ovarian cancer treatment and advanced and early-stage breast cancer treatments in 1994 and 1999, respectively. PTX is administered as a second-line drug in monotherapy when combination chemotherapy fails to treat breast cancer or the disease recurs within 6 months of adjuvant chemotherapy. Based on published preclinical data, PTX affects various pathways, causing an overall clinical activity that is not solely dependent on its direct cytotoxic effects on cancer cells. As a result, PTX (including its new formulations) may provide unique and rational therapeutic approaches to managing tumor progression in patients.

Availability of data and materials

Not applicable.

Abbreviations

ABC:

ATP-binding cassette

ATC:

Anaplastic thyroid cancer

BBB:

Blood–brain barrier

CAT:

Catalase

COX-2:

Cyclooxygenase-2

DHA:

Docosahexaenoic acid

FDA:

Food and Drug Administration

GPx:

Glutathione peroxidases

HPLC/LC–MS:

High-performance liquid chromatography/mass spectrometry

HPLC:

High-performance liquid chromatography

HRPC:

Hormone-refractory prostate cancer

LC–ESI-MS:

Liquid chromatography–electrospray ionization tandem mass spectrometry

LEP-ETU:

Liposome-entrapped

MDR:

Multidrug resistance

MMP9:

Matrix metalloproteinase

NMR:

Nuclear magnetic resonance

NSCLC:

Non-small-cell lung cancer

P-gp:

P-glycoprotein

PTX-TTHA:

Paclitaxel-triethylenetetramine hexaacetic acid conjugate

SOD:

Superoxide dismutase

SWNT:

Single-walled carbon nanotubes

TLC:

Thin-layer chromatography

UV:

Ultraviolet

References

  1. Parsa N. Environmental factors inducing human cancers [2251-6085 (Print)]

  2. Klepeis NE, Hughes SC, Edwards RD, Allen T, Johnson M, Chowdhury Z, Smith KR, Boman-Davis M, Bellettiere J, Hovell MF. Promoting smoke-free homes: a novel behavioral intervention using real-time audio-visual feedback on airborne particle levels. PLoS ONE. 2013;8(8):e73251. https://doi.org/10.1371/journal.pone.0073251.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Choudhari AS, Mandave PC, Deshpande M, Ranjekar P, Prakash O. Phytochemicals in cancer treatment: from preclinical studies to clinical practice. Front Pharmacol. 2019;10:1614. https://doi.org/10.3389/fphar.2019.01614.

    Article  CAS  PubMed  Google Scholar 

  4. Anand U, Dey A, Chandel AKS, Sanyal R, Mishra A, Pandey DK, De Falco V, Upadhyay A, Kandimalla R, Chaudhary A, Dhanjal JK, Dewanjee S, Vallamkondu J, Pérez de la Lastra JM. Cancer chemotherapy and beyond: current status, drug candidates, associated risks and progress in targeted therapeutics. Genes Dis. 2023;10(4):1367–401. https://doi.org/10.1016/j.gendis.2022.02.007.

    Article  CAS  PubMed  Google Scholar 

  5. Greenwell M, Rahman PK. Medicinal plants: their use in anticancer treatment. Int J Pharm Sci Res. 2015;6(10):4103–12. https://doi.org/10.13040/IJPSR.0975-8232.6(10).4103-12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sun W, Shahrajabian MH. Therapeutic potential of phenolic compounds in medicinal plants—natural health products for human health. Molecules. 2023. https://doi.org/10.3390/molecules28041845.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Riaz M, Khalid R, Afzal M, Anjum F, Fatima H, Zia S, Rasool G, Egbuna C, Mtewa AG, Uche CZ, Aslam MA. Phytobioactive compounds as therapeutic agents for human diseases: a review. Food Sci Nutr. 2023;11(6):2500–29. https://doi.org/10.1002/fsn3.3308.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Dong S, Wang Z, Shen K, Chen X. Metabolic syndrome and breast cancer: prevalence, treatment response, and prognosis. Front Oncol. 2021. https://doi.org/10.3389/fonc.2021.629666.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Garzoli S, Alarcón-Zapata P, Seitimova G, Alarcón-Zapata B, Martorell M, Sharopov F, Fokou PVT, Dize D, Yamthe LRT, Les F, Cásedas G, López V, Iriti M, Rad JS, Gürer ES, Calina D, Pezzani R, Vitalini S. Natural essential oils as a new therapeutic tool in colorectal cancer. Cancer Cell Int. 2022;22(1):407. https://doi.org/10.1186/s12935-022-02806-5.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Sénilh V, Blechert S, Colin M, Guénard D, Picot F, Potier P, Varenne P. Mise en Évidence de Nouveaux Analogues du Taxol Extraits de Taxus baccata. J Nat Prod. 1984;47(1):131–7. https://doi.org/10.1021/np50031a019.

    Article  Google Scholar 

  11. Velde DGV, Georg GI, Gollapudi SR, Jampani HB, Liang X-Z, Mitscher LA, Ye Q-M. Wallifoliol, a taxol congener with a novel carbon Skeleton, from Himalayan Taxus wallichiana. J Nat Prod. 1994;57(6):862–7. https://doi.org/10.1021/np50108a032.

    Article  Google Scholar 

  12. Appendino G, Cravotto G, Enriù R, Gariboldi P, Barboni L, Torregiani E, Gabetta B, Zini G, Bombardelli E. Taxoids from the Roots of Taxus × media cv. Hicksii J Nat Prod. 1994;57(5):607–13. https://doi.org/10.1021/np50107a007.

    Article  CAS  Google Scholar 

  13. Wani MC, Taylor HL, Wall ME, Coggon P, McPhail AT. Plant antitumor agents. VI. The isolation and structure of taxol, a novel antileukemic and antitumor agent from Taxus brevifolia. J Am Chem Soc. 1971;93(9):2325–7. https://doi.org/10.1021/ja00738a045.

    Article  CAS  PubMed  Google Scholar 

  14. Mamadalieva NZ, Mamedov NA. Taxus brevifolia a high-value medicinal plant, as a source of taxol. In: Máthé Á (ed) Medicinal and aromatic plants of North America. Cham: Springer; 2020. p. 201–218. https://doi.org/10.1007/978-3-030-44930-8_9.

  15. Bănică D, Algeorge G, Moisoiu A, Petre A, Toancă M. The possibilities for improving the serological diagnosis of active tuberculosis by using new mycobacterial antigens and immunoblot and ELISA technics. Pneumoftiziologia. 1994;43(3–4):173–7.

    PubMed  Google Scholar 

  16. Gangadevi V, Murugan M, Johnpaul M. Taxol Determination from Pestalotiopsis pauciseta, a Fungal Endophyte of a Medicinal Plant. Sheng wu gong cheng xue bao = Chin J Biotechnol, 2008;24:1433–38. https://doi.org/10.1016/S1872-2075(08)60065-5.

  17. Gangadevi V, Muthumary J. Taxol production by Pestalotiopsis terminaliae, an endophytic fungus of Terminalia arjuna (arjun tree). Biotechnol Appl Biochem. 2009;52(1):9–15.

    Article  CAS  PubMed  Google Scholar 

  18. Kumaran RS, Kim HJ, Hur BK. Taxol-producing [corrected] fungal endophyte, Pestalotiopsis species isolated from Taxus cuspidata. J Biosci Bioeng. 2010;110(5):541–6. https://doi.org/10.1016/j.jbiosc.2010.06.007.

    Article  CAS  PubMed  Google Scholar 

  19. Kumar P, Raza K, Kaushik L, Malik R, Arora S, Katare OP. Role of colloidal drug delivery carriers in taxane-mediated chemotherapy: a review. Curr Pharm Des. 2016;22(33):5127–43. https://doi.org/10.2174/1381612822666160524144926.

    Article  CAS  PubMed  Google Scholar 

  20. Vidensek N, Lim P, Campbell A, Carlson C. Taxol content in bark, wood, root, leaf, twig, and seedling from several Taxus species. J Nat Prod. 1990;53(6):1609–10. https://doi.org/10.1021/np50072a039.

    Article  CAS  PubMed  Google Scholar 

  21. Croteau R, Ketchum RE, Long RM, Kaspera R, Wildung MR. Taxol biosynthesis and molecular genetics. Phytochem Rev. 2006;5(1):75–97. https://doi.org/10.1007/s11101-005-3748-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Nadeem M, Rikhari HC, Kumar A, Palni LM, Nandi SK. Taxol content in the bark of Himalayan Yew in relation to tree age and sex. Phytochemistry. 2002;60(6):627–31. https://doi.org/10.1016/s0031-9422(02)00115-2.

    Article  CAS  PubMed  Google Scholar 

  23. Expósito O, Moyano E, Onrubia M, Mirjalili M, Cusido R, Palazon J. Biotechnological production of taxol and related taxoids: current state and prospects. Anticancer Agents Med Chem. 2009;9:109–21. https://doi.org/10.2174/187152009787047761.

    Article  PubMed  Google Scholar 

  24. Frense D. Taxanes: perspectives for biotechnological production. Appl Microbiol Biotechnol. 2007;73:1233–40. https://doi.org/10.1007/s00253-006-0711-0.

    Article  CAS  PubMed  Google Scholar 

  25. Kingston DG. The shape of things to come: structural and synthetic studies of taxol and related compounds. Phytochemistry. 2007;68(14):1844–54. https://doi.org/10.1016/j.phytochem.2006.11.009.

    Article  CAS  PubMed  Google Scholar 

  26. Witherup KM, Look SA, Stasko MW, Ghiorzi TJ, Muschik GM, Cragg GM. Taxus spp. needles contain amounts of taxol comparable to the bark of Taxus brevifolia: analysis and isolation. J Nat Prod. 1990;53(5):1249–55. https://doi.org/10.1021/np50071a017.

    Article  CAS  PubMed  Google Scholar 

  27. Pauling L. The significance of the evidence about ascorbic acid and the common cold. Proc Natl Acad Sci USA. 1971;68(11):2678–81. https://doi.org/10.1073/pnas.68.11.2678.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Walsh V, Goodman J. The billion dollar molecule: taxol in historical and theoretical perspective. Clio Med. 2002;66:245–67. https://doi.org/10.1163/9789004333499_013.

    Article  PubMed  Google Scholar 

  29. Shi Q, Oritani T, Sugiyama T, Murakami R, Wei H. Six new taxane diterpenoids from the seeds of Taxus chinensis var. mairei and taxus yunnanensis. J Nat Prod. 1999;62(8):1114–8. https://doi.org/10.1021/np990106b.

    Article  CAS  PubMed  Google Scholar 

  30. Morris H, Price S. Langerhans’ cells, papillomaviruses and oesophageal carcinoma. A hypothesis. S Afr Med J. 1986;69(7):413–7.

    CAS  PubMed  Google Scholar 

  31. Mukhtar E, Adhami VM, Mukhtar H. Targeting microtubules by natural agents for cancer therapy. Mol Cancer Ther. 2014;13(2):275–84. https://doi.org/10.1158/1535-7163.MCT-13-0791.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Weaver BA. How taxol/paclitaxel kills cancer cells. Mol Biol Cell. 2014;25(18):2677–81. https://doi.org/10.1091/mbc.E14-04-0916.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kampan NC, Madondo MT, McNally OM, Quinn M, Plebanski M. Paclitaxel and its evolving role in the management of ovarian cancer. Biomed Res Int. 2015;2015: 413076. https://doi.org/10.1155/2015/413076.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Abu Samaan TM, Samec M, Liskova A, Kubatka P, Büsselberg D. Paclitaxel’s mechanistic and clinical effects on breast cancer. Biomolecules. 2019. https://doi.org/10.3390/biom9120789.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Monfort JC, Manus A. Schizophrenia and choline acetyltransferase. Am J Psychiatry. 1994;151(4):627. https://doi.org/10.1176/ajp.151.4.627a.

    Article  CAS  PubMed  Google Scholar 

  36. Zhu L, Chen L. Progress in research on paclitaxel and tumor immunotherapy. Cell Mol Biol Lett. 2019;24(1):40. https://doi.org/10.1186/s11658-019-0164-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Badiner GJ, Hamilton RD, Li LH, Bhuyan BK. Drug sensitivity of ten human tumor cell lines compared to mouse leukemia (L1210) cells. Invest New Drugs. 1987;5(3):219–29. https://doi.org/10.1007/BF00175291.

    Article  CAS  PubMed  Google Scholar 

  38. Khwaja S, Kumar K, Das R, Negi AS. Microtubule associated proteins as targets for anticancer drug development. Bioorg Chem. 2021;116:105320. https://doi.org/10.1016/j.bioorg.2021.105320.

    Article  CAS  PubMed  Google Scholar 

  39. Bledsoe A. The importance of touch in nursing care. Imprint. 1984;13(4):58–9.

    CAS  PubMed  Google Scholar 

  40. Zhu L, Chen L. Progress in research on paclitaxel and tumor immunotherapy. Cell Mol Biol Lett. 2019;24:40. https://doi.org/10.1186/s11658-019-0164-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Choi JS, Shin SC. Enhanced paclitaxel bioavailability after oral coadministration of paclitaxel prodrug with naringin to rats. Int J Pharm. 2005;292(1–2):149–56. https://doi.org/10.1016/j.ijpharm.2004.11.031.

    Article  CAS  PubMed  Google Scholar 

  42. Zhang JA, Anyarambhatla G, Ma L, Ugwu S, Xuan T, Sardone T, Ahmad I. Development and characterization of a novel Cremophor EL free liposome-based paclitaxel (LEP-ETU) formulation. Eur J Pharm Biopharm. 2005;59(1):177–87. https://doi.org/10.1016/j.ejpb.2004.06.009.

    Article  CAS  PubMed  Google Scholar 

  43. Gallo JM, Li S, Guo P, Reed K, Ma J. The effect of P-glycoprotein on paclitaxel brain and brain tumor distribution in mice. Cancer Res. 2003;63(16):5114–7.

    CAS  PubMed  Google Scholar 

  44. Berg SL, Tolcher A, O’Shaughnessy JA, Denicoff AM, Noone M, Ognibene FP, Cowan KH, Balis FM. Effect of R-verapamil on the pharmacokinetics of paclitaxel in women with breast cancer. J Clin Oncol. 1995;13(8):2039–42. https://doi.org/10.1200/JCO.1995.13.8.2039.

    Article  CAS  PubMed  Google Scholar 

  45. Bardelmeijer HA, Ouwehand M, Beijnen JH, Schellens JH, van Tellingen O. Efficacy of novel P-glycoprotein inhibitors to increase the oral uptake of paclitaxel in mice. Invest New Drugs. 2004;22(3):219–29. https://doi.org/10.1023/B:DRUG.0000026248.45084.21.

    Article  CAS  PubMed  Google Scholar 

  46. WFO. WFO the world flora online; 2021. http://www.worldfloraonline.org/.

  47. PubChem. PubChem; 2022. https://pubchem.ncbi.nlm.nih.gov/.

  48. Defraigne JO, Pincemail J. Local and systemic consequences of severe ischemia and reperfusion of the skeletal muscle. Physiopathol Prevent Acta Chir Belg. 1998;98(4):176–86.

    CAS  Google Scholar 

  49. Galsky MD, Dritselis A, Kirkpatrick P, Oh WK. Cabazitaxel. Nat Rev Drug Discov. 2010;9(9):677–8. https://doi.org/10.1038/nrd3254.

    Article  CAS  PubMed  Google Scholar 

  50. Liu Y, Hong G, Mao L, Su Z, Liu T, Liu H. A Novel Paclitaxel Derivative for Triple-Negative Breast Cancer Chemotherapy. Molecules. 2023;28(9):3662. https://doi.org/10.3390/molecules28093662. PMID: 37175072; PMCID:PMC10180349.

    Article  Google Scholar 

  51. Kartner N, Riordan JR, Ling V. Cell surface P-glycoprotein associated with multidrug resistance in mammalian cell lines. Science. 1983;221(4617):1285–8. https://doi.org/10.1126/science.6137059.

    Article  CAS  PubMed  Google Scholar 

  52. de Bono JS, Oudard S, Ozguroglu M, Hansen S, Machiels JP, Kocak I, Gravis G, Bodrogi I, Mackenzie MJ, Shen L, Roessner M, Gupta S, Sartor AO, Investigators T. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet. 2010;376(9747):1147–54. https://doi.org/10.1016/S0140-6736(10)61389-X.

    Article  CAS  PubMed  Google Scholar 

  53. Abidi A. Cabazitaxel: a novel taxane for metastatic castration-resistant prostate cancer-current implications and future prospects. J Pharmacol Pharmacother. 2013;4(4):230–7. https://doi.org/10.4103/0976-500X.119704.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Bissery MC. Preclinical evaluation of new taxoids. Curr Pharm Des. 2001;7(13):1251–7. https://doi.org/10.2174/1381612013397465.

    Article  CAS  PubMed  Google Scholar 

  55. Desai N, Trieu V, Yao Z, Louie L, Ci S, Yang A, Tao C, De T, Beals B, Dykes D, Noker P, Yao R, Labao E, Hawkins M, Soon-Shiong P. Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin Cancer Res. 2006;12(4):1317–24. https://doi.org/10.1158/1078-0432.CCR-05-1634.

    Article  CAS  PubMed  Google Scholar 

  56. Ibrahim NK, Desai N, Legha S, Soon-Shiong P, Theriault RL, Rivera E, Esmaeli B, Ring SE, Bedikian A, Hortobagyi GN, Ellerhorst JA. Phase I and pharmacokinetic study of ABI-007, a Cremophor-free, protein-stabilized, nanoparticle formulation of paclitaxel. Clin Cancer Res. 2002;8(5):1038–44.

    CAS  PubMed  Google Scholar 

  57. Stinchcombe TE. Nanoparticle albumin-bound paclitaxel: a novel Cremphor-EL-free formulation of paclitaxel. Nanomedicine (Lond). 2007;2(4):415–23. https://doi.org/10.2217/17435889.2.4.415.

    Article  CAS  PubMed  Google Scholar 

  58. Donyai P, Sewell GJ. Physical and chemical stability of paclitaxel infusions in different container types. J Oncol Pharm Pract. 2006;12(4):211–22. https://doi.org/10.1177/1078155206073589.

    Article  CAS  PubMed  Google Scholar 

  59. Paz-Ares L, Ross H, O’Brien M, Riviere A, Gatzemeier U, Von Pawel J, Kaukel E, Freitag L, Digel W, Bischoff H, García-Campelo R, Iannotti N, Reiterer P, Bover I, Prendiville J, Eisenfeld AJ, Oldham FB, Bandstra B, Singer JW, Bonomi P. Phase III trial comparing paclitaxel poliglumex vs. docetaxel in the second-line treatment of non-small-cell lung cancer. Br J Cancer. 2008;98(10):1608–13. https://doi.org/10.1038/sj.bjc.6604372.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Jing YR, Zhou W, Li WL, Zhao LX, Wang YF. The synthesis of novel taxoids for oral administration. Bioorg Med Chem. 2014;22(1):194–203. https://doi.org/10.1016/j.bmc.2013.11.037.

    Article  CAS  PubMed  Google Scholar 

  61. Yared JA, Tkaczuk KH. Update on taxane development: new analogs and new formulations. Drug Des Devel Ther. 2012;6:371–84. https://doi.org/10.2147/DDDT.S28997.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Petri WA, Joyce MP, Broman J, Smith RD, Murphy CF, Ravdin JI. Recognition of the galactose- or N-acetylgalactosamine-binding lectin of Entamoeba histolytica by human immune sera. Infect Immun. 1987;55(10):2327–31. https://doi.org/10.1128/iai.55.10.2327-2331.1987.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Chen J, Chen S, Zhao X, Kuznetsova LV, Wong SS, Ojima I. Functionalized single-walled carbon nanotubes as rationally designed vehicles for tumor-targeted drug delivery. J Am Chem Soc. 2008;130(49):16778–85. https://doi.org/10.1021/ja805570f.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ojima I, Habus I, Zhao M, Zucco M, Park YH, Sun CM, Brigaud T. New and efficient approaches to the semisynthesis of taxol and its C-13 side chain analogs by means of β-lactam synthon method. Tetrahedron. 1992;48(34):6985–7012. https://doi.org/10.1016/S0040-4020(01)91210-4.

    Article  CAS  Google Scholar 

  65. Magri NF, Kingston DG. Modified taxols, 4. Synthesis and biological activity of taxols modified in the side chain. J Nat Prod. 1988;51(2):298–306. https://doi.org/10.1021/np50056a017.

    Article  CAS  PubMed  Google Scholar 

  66. Turunen BJ, Ge H, Oyetunji J, Desino KE, Vasandani V, Güthe S, Himes RH, Audus KL, Seelig A, Georg GI. Paclitaxel succinate analogs: Anionic and amide introduction as a strategy to impart blood-brain barrier permeability. Bioorg Med Chem Lett. 2008;18(22):5971–4. https://doi.org/10.1016/j.bmcl.2008.09.103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Nevarez DM, Mengistu YA, Nawarathne IN, Walker KD. An N-aroyltransferase of the BAHD superfamily has broad aroyl CoA specificity in vitro with analogues of N-dearoylpaclitaxel. J Am Chem Soc. 2009;131(16):5994–6002. https://doi.org/10.1021/ja900545m.

    Article  CAS  PubMed  Google Scholar 

  68. Wieczorek A, Błauż A, Żal A, Arabshahi HJ, Reynisson J, Hartinger CG, Rychlik B, Plażuk D. Ferrocenyl paclitaxel and docetaxel derivatives: impact of an organometallic moiety on the mode of action of taxanes. Chemistry. 2016;22(32):11413–21. https://doi.org/10.1002/chem.201601809.

    Article  CAS  PubMed  Google Scholar 

  69. Hamada H, Hamada H, Shimoda K, Mandai T, Ishihara K, Kiriake Y, Kuboki A. Synthesis of ester-linked paclitaxel-glycoside conjugate as a water-soluble paclitaxel derivative—maltoside modification of paclitaxel through ester-linker (ester-spacer). Nat Prod Commun. 2021;16(9):1934578X211038788. https://doi.org/10.1177/1934578x211038788.

  70. Hackett MJ, Joolakanti S, Hartranft ME, Guley PC, Cho MJ. A dicarboxylic fatty acid derivative of paclitaxel for albumin-assisted drug delivery. J Pharm Sci. 2012;101(9):3292–304. https://doi.org/10.1002/jps.23213.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Li F, Lu J, Liu J, Liang C, Wang M, Wang L, Li D, Yao H, Zhang Q, Wen J, Zhang ZK, Li J, Lv Q, He X, Guo B, Guan D, Yu Y, Dang L, Wu X, Li Y, Chen G, Jiang F, Sun S, Zhang BT, Lu A, Zhang G. A water-soluble nucleolin aptamer-paclitaxel conjugate for tumor-specific targeting in ovarian cancer. Nat Commun. 2017;8(1):1390. https://doi.org/10.1038/s41467-017-01565-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Torchilin VP. Multifunctional, stimuli-sensitive nanoparticulate systems for drug delivery. Nat Rev Drug Discov. 2014;13(11):813–27. https://doi.org/10.1038/nrd4333.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Wendt F, Kappler C, Burckhardt K, Bohle A. Induction of a generalized Sanarelli-Shwartzman reaction in spite of extreme granulocytopenia after nitrogenmustard. Verh Dtsch Ges Inn Med. 1967;73:361–5.

    CAS  PubMed  Google Scholar 

  74. Ojima I, Lichtenthal B, Lee S, Wang C, Wang X. Taxane anticancer agents: a patent perspective. Expert Opin Ther Pat. 2016;26(1):1–20. https://doi.org/10.1517/13543776.2016.1111872.

    Article  CAS  PubMed  Google Scholar 

  75. Li JA, Xu XF, Han X, Fang Y, Shi CY, Jin DY, Lou WH. Nab-paclitaxel plus S-1 shows increased antitumor activity in patient-derived pancreatic cancer xenograft mouse models. Pancreas. 2016;45(3):425–33. https://doi.org/10.1097/MPA.0000000000000501.

    Article  CAS  PubMed  Google Scholar 

  76. Robert F, Harper K, Ackerman J, Gupta S. A phase I study of larotaxel (XRP9881) administered in combination with carboplatin in chemotherapy-naïve patients with stage IIIB or stage IV non-small cell lung cancer. Cancer Chemother Pharmacol. 2010;65(2):227–34. https://doi.org/10.1007/s00280-009-1026-5.

    Article  CAS  PubMed  Google Scholar 

  77. Metzger-Filho O, Moulin C, de Azambuja E, Ahmad A. Larotaxel: broadening the road with new taxanes. Expert Opin Investig Drugs. 2009;18(8):1183–9. https://doi.org/10.1517/13543780903119167.

    Article  CAS  PubMed  Google Scholar 

  78. Ojima I, Kamath A, Seitz JD. Taxol, taxoids, and related taxanes. In: Natural products in medicinal chemistry. Methods and principles in medicinal chemistry; 2014. P. 127–80. https://doi.org/10.1002/9783527676545.ch04.

  79. Sternberg CN, Skoneczna IA, Castellano D, Theodore C, Blais N, Voog E, Bellmunt J, Peters F, Le-Guennec S, Cerbone L, Risse ML, Machiels JP. Larotaxel with Cisplatin in the first-line treatment of locally advanced/metastatic urothelial tract or bladder cancer: a randomized, active-controlled, phase III trial (CILAB). Oncology. 2013;85(4):208–15. https://doi.org/10.1159/000354085.

    Article  CAS  PubMed  Google Scholar 

  80. Flores JP, Saif MW. Novel oral taxane therapies: recent Phase I results. Clin Investig (Lond). 2013;3(4):333–41. https://doi.org/10.4155/cli.13.18.

    Article  CAS  PubMed  Google Scholar 

  81. Gautier C, Mehtali M, Lathe R. A ubiquitous mammalian expression vector, pHMG, based on a housekeeping gene promoter. Nucl Acids Res. 1989;17(20):8389. https://doi.org/10.1093/nar/17.20.8389.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Roche M, Kyriakou H, Seiden M. Drug evaluation: tesetaxel—an oral semisynthetic taxane derivative. Curr Opin Investig Drugs. 2006;7(12):1092–9.

    CAS  PubMed  Google Scholar 

  83. Ramanathan RK, Picus J, Raftopoulos H, Bernard S, Lockhart AC, Frenette G, Macdonald J, Melin S, Berg D, Brescia F, Hochster H, Cohn A. A phase II study of milataxel: a novel taxane analogue in previously treated patients with advanced colorectal cancer. Cancer Chemother Pharmacol. 2008;61(3):453–8. https://doi.org/10.1007/s00280-007-0489-5.

    Article  CAS  PubMed  Google Scholar 

  84. Camps C, Felip E, Sanchez JM, Massuti B, Artal A, Paz-Ares L, Carrato A, Alberola V, Blasco A, Baselga J, Astier L, Voi M, Rosell R, Group SLC. Phase II trial of the novel taxane BMS-184476 as second-line in non-small-cell lung cancer. Ann Oncol. 2005;16(4):597–601. https://doi.org/10.1093/annonc/mdi120.

  85. Knossow M, Campanacci V, Khodja LA, Gigant B. The mechanism of tubulin assembly into microtubules: insights from structural studies. iScience. 2020;23(9):101511. https://doi.org/10.1016/j.isci.2020.101511.

  86. Gallego-Jara J, Lozano-Terol G, Sola-Martínez RA, Cánovas-Díaz M, de Diego Puente T. A compressive review about taxol(®): history and future challenges. Molecules. 2020. https://doi.org/10.3390/molecules25245986.

  87. Wang TH, Wang HS, Soong YK. Paclitaxel-induced cell death: where the cell cycle and apoptosis come together. Cancer. 2000;88(11):2619–28. https://doi.org/10.1002/1097-0142(20000601)88:11%3c2619::aid-cncr26%3e3.0.co;2-j.

    Article  CAS  PubMed  Google Scholar 

  88. Schiff PB, Horwitz SB. Taxol stabilizes microtubules in mouse fibroblast cells. Proc Natl Acad Sci USA. 1980;77(3):1561–5. https://doi.org/10.1073/pnas.77.3.1561.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Zhang D, Yang R, Wang S, Dong Z. Paclitaxel: new uses for an old drug. Drug Des Devel Ther. 2014;8:279–84. https://doi.org/10.2147/dddt.S56801.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Schiff PB, Fant J, Horwitz SB. Promotion of microtubule assembly in vitro by taxol. Nature. 1979;277(5698):665–7. https://doi.org/10.1038/277665a0.

    Article  CAS  PubMed  Google Scholar 

  91. Ganguly A, Yang H, Cabral F. Paclitaxel-dependent cell lines reveal a novel drug activity. Mol Cancer Ther. 2010;9(11):2914–23. https://doi.org/10.1158/1535-7163.MCT-10-0552.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. De Brabander M, Geuens G, Nuydens R, Willebrords R, De Mey J. Taxol induces the assembly of free microtubules in living cells and blocks the organizing capacity of the centrosomes and kinetochores. Proc Natl Acad Sci USA. 1981;78(9):5608–12. https://doi.org/10.1073/pnas.78.9.5608.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Jordan MA, Toso RJ, Thrower D, Wilson L. Mechanism of mitotic block and inhibition of cell proliferation by taxol at low concentrations. Proc Natl Acad Sci USA. 1993;90(20):9552–6. https://doi.org/10.1073/pnas.90.20.9552.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Luserna G, di Rorà A, Martinelli G, Simonetti G. The balance between mitotic death and mitotic slippage in acute leukemia: a new therapeutic window? J Hematol Oncol. 2019;12(1):123. https://doi.org/10.1186/s13045-019-0808-4.

    Article  Google Scholar 

  95. Rajput S, Volk-Draper LD, Ran S. TLR4 is a novel determinant of the response to paclitaxel in breast cancer. Mol Cancer Ther. 2013;12(8):1676–87. https://doi.org/10.1158/1535-7163.Mct-12-1019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Molteni M, Gemma S, Rossetti C. The role of toll-like receptor 4 in infectious and noninfectious inflammation. Mediators Inflamm. 2016;2016:6978936. https://doi.org/10.1155/2016/6978936.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Saitoh S, Miyake K. Mechanism regulating cell surface expression and activation of Toll-like receptor 4. Chem Rec. 2006;6(6):311–9. https://doi.org/10.1002/tcr.20093.

    Article  CAS  PubMed  Google Scholar 

  98. Kashani B, Zandi Z, Pourbagheri-Sigaroodi A, Bashash D, Ghaffari SH. The role of toll-like receptor 4 (TLR4) in cancer progression: A possible therapeutic target? J Cell Physiol. 2021;236(6):4121–37. https://doi.org/10.1002/jcp.30166.

    Article  CAS  PubMed  Google Scholar 

  99. Zhou S, Du R, Wang Z, Shen W, Gao R, Jiang S, Fang Y, Shi Y, Chang A, Liu L, Liu C, Li N, Xiang R. TLR4 increases the stemness and is highly expressed in relapsed human hepatocellular carcinoma. Cancer Med. 2019;8(5):2325–37. https://doi.org/10.1002/cam4.2070.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Oblak A, Jerala R. Toll-like receptor 4 activation in cancer progression and therapy. Clin Dev Immunol. 2011;2011:609579. https://doi.org/10.1155/2011/609579.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Ju H, Hu Z, Lu Y, Wu Y, Zhang L, Wei D, Guo W, Xia W, Liu S, Ren G, Hu J. TLR4 activation leads to anti-EGFR therapy resistance in head and neck squamous cell carcinoma. Am J Cancer Res. 2020;10(2):454–72.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Jiang N, Xie F, Guo Q, Li MQ, Xiao J, Sui L. Toll-like receptor 4 promotes proliferation and apoptosis resistance in human papillomavirus-related cervical cancer cells through the Toll-like receptor 4/nuclear factor-κB pathway. Tumour Biol. 2017;39(6):1010428317710586. https://doi.org/10.1177/1010428317710586.

    Article  CAS  PubMed  Google Scholar 

  103. Shetab Boushehri MA, Lamprecht A. TLR4-based immunotherapeutics in cancer: a review of the achievements and shortcomings. Mol Pharm. 2018;15(11):4777–800. https://doi.org/10.1021/acs.molpharmaceut.8b00691.

    Article  CAS  PubMed  Google Scholar 

  104. Papadakos SP, Arvanitakis K, Stergiou IE, Lekakis V, Davakis S, Christodoulou MI, Germanidis G, Theocharis S. The role of TLR4 in the immunotherapy of hepatocellular carcinoma: can we teach an old dog new tricks? Cancers (Basel). 2023. https://doi.org/10.3390/cancers15102795.

  105. Kelly MG, Alvero AB, Chen R, Silasi DA, Abrahams VM, Chan S, Visintin I, Rutherford T, Mor G. TLR-4 signaling promotes tumor growth and paclitaxel chemoresistance in ovarian cancer. Cancer Res. 2006;66(7):3859–68. https://doi.org/10.1158/0008-5472.Can-05-3948.

    Article  CAS  PubMed  Google Scholar 

  106. Szajnik M, Szczepanski MJ, Czystowska M, Elishaev E, Mandapathil M, Nowak-Markwitz E, Spaczynski M, Whiteside TL. TLR4 signaling induced by lipopolysaccharide or paclitaxel regulates tumor survival and chemoresistance in ovarian cancer. Oncogene. 2009;28(49):4353–63. https://doi.org/10.1038/onc.2009.289.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Pfannenstiel LW, Lam SS, Emens LA, Jaffee EM, Armstrong TD. Paclitaxel enhances early dendritic cell maturation and function through TLR4 signaling in mice. Cell Immunol. 2010;263(1):79–87. https://doi.org/10.1016/j.cellimm.2010.03.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Lau TS, Chan LKY, Man GCW, Wong CH, Lee JHS, Yim SF, Cheung TH, McNeish IA, Kwong J. Paclitaxel induces immunogenic cell death in ovarian cancer via TLR4/IKK2/SNARE-dependent exocytosis. Cancer Immunol Res. 2020;8(8):1099–111. https://doi.org/10.1158/2326-6066.Cir-19-0616.

    Article  CAS  PubMed  Google Scholar 

  109. Wang YM, Xu X, Tang J, Sun ZY, Fu YJ, Zhao XJ, Ma XM, Ye Q. Apatinib induces endoplasmic reticulum stress-mediated apoptosis and autophagy and potentiates cell sensitivity to paclitaxel via the IRE-1α-AKT-mTOR pathway in esophageal squamous cell carcinoma. Cell Biosci. 2021;11(1):124. https://doi.org/10.1186/s13578-021-00640-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Bocci G, Di Paolo A, Danesi R. The pharmacological bases of the antiangiogenic activity of paclitaxel. Angiogenesis. 2013;16(3):481–92. https://doi.org/10.1007/s10456-013-9334-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Hadzic T, Aykin-Burns N, Zhu Y, Coleman MC, Leick K, Jacobson GM, Spitz DR. Paclitaxel combined with inhibitors of glucose and hydroperoxide metabolism enhances breast cancer cell killing via H2O2-mediated oxidative stress. Free Radic Biol Med. 2010;48(8):1024–33. https://doi.org/10.1016/j.freeradbiomed.2010.01.018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Zasadil LM, Andersen KA, Yeum D, Rocque GB, Wilke LG, Tevaarwerk AJ, Raines RT, Burkard ME, Weaver BA. Cytotoxicity of paclitaxel in breast cancer is due to chromosome missegregation on multipolar spindles. Sci Transl Med. 2014;6(229):229ra243. https://doi.org/10.1126/scitranslmed.3007965.

  113. Mohiuddin M, Kasahara K. The mechanisms of the growth inhibitory effects of paclitaxel on Gefitinib-resistant non-small cell lung cancer cells. Cancer Genomics Proteomics. 2021;18(5):661–73. https://doi.org/10.21873/cgp.20288.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Zuo Y, Shen W, Wang L, Wang C, Pu J. Study on the mechanism of action of paclitaxel-loaded polylactic-co-glycolic acid nanoparticles in non-small-cell lung carcinoma cells. Comput Math Methods Med. 2022;2022:8524951. https://doi.org/10.1155/2022/8524951.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Kim SY, Kim SM, Chang H, Chang HS, Park CS, Lee YS. Synergistic anticancer activity of sorafenib, paclitaxel, and radiation therapy on anaplastic thyroid cancer in vitro and in vivo. Head Neck. 2020;42(12):3678–84. https://doi.org/10.1002/hed.26431.

    Article  PubMed  Google Scholar 

  116. Khing TM, Po WW, Sohn UD. Fluoxetine enhances anti-tumor activity of paclitaxel in gastric adenocarcinoma cells by triggering apoptosis and necroptosis. Anticancer Res. 2019;39(11):6155–63. https://doi.org/10.21873/anticanres.13823.

    Article  CAS  PubMed  Google Scholar 

  117. Min J, Shen H, Xi W, Wang Q, Yin L, Zhang Y, Yu Y, Yang Q, Wang ZN. Synergistic anticancer activity of combined use of caffeic acid with paclitaxel enhances apoptosis of non-small-cell lung cancer H1299 cells in vivo and in vitro. Cell Physiol Biochem. 2018;48(4):1433–42. https://doi.org/10.1159/000492253.

    Article  CAS  PubMed  Google Scholar 

  118. Pashaei-Asl F, Pashaei-Asl R, Khodadadi K, Akbarzadeh A, Ebrahimie E, Pashaiasl M. Enhancement of anticancer activity by silibinin and paclitaxel combination on the ovarian cancer. Artif Cells Nanomed Biotechnol. 2018;46(7):1483–7. https://doi.org/10.1080/21691401.2017.1374281.

    Article  CAS  PubMed  Google Scholar 

  119. Rabzia A, Khazaei M, Rashidi Z, Khazaei MR. Synergistic anticancer effect of paclitaxel and noscapine on human prostate cancer cell lines. Iran J Pharm Res. 2017;16(4):1432–42.

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Han ES, Wen W, Dellinger TH, Wu J, Lu SA, Jove R, Yim JH. Ruxolitinib synergistically enhances the anti-tumor activity of paclitaxel in human ovarian cancer. Oncotarget. 2018;9(36):24304–19. https://doi.org/10.18632/oncotarget.24368.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Zhang X, Wang J, Fan Y, Zhao Z, Paraghamian SE, Hawkins GM, Buckingham L, O’Donnell J, Hao T, Suo H, Yin Y, Sun W, Kong W, Sun D, Zhao L, Zhou C, Bae-Jump VL. Asparagus officinalis combined with paclitaxel exhibited synergistic anti-tumor activity in paclitaxel-sensitive and -resistant ovarian cancer cells. J Cancer Res Clin Oncol. 2022. https://doi.org/10.1007/s00432-022-04276-8.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Shu S, Yamashita-Kashima Y, Yanagisawa M, Nakanishi H, Kodera Y, Harada N, Yoshimura Y. Trastuzumab in combination with paclitaxel enhances antitumor activity by promoting apoptosis in human epidermal growth factor receptor 2-positive trastuzumab-resistant gastric cancer xenograft models. Anticancer Drugs. 2020;31(3):241–50. https://doi.org/10.1097/CAD.0000000000000853.

    Article  CAS  PubMed  Google Scholar 

  123. Huang G, Zang B, Wang X, Liu G, Zhao J. Encapsulated paclitaxel nanoparticles exhibit enhanced anti-tumor efficacy in A549 non-small lung cancer cells. Acta Biochim Biophys Sin (Shanghai). 2015;47(12):981–7. https://doi.org/10.1093/abbs/gmv110.

    Article  CAS  PubMed  Google Scholar 

  124. Leiva MC, Ortiz R, Contreras-Cáceres R, Perazzoli G, Mayevych I, López-Romero JM, Sarabia F, Baeyens JM, Melguizo C, Prados J. Tripalmitin nanoparticle formulations significantly enhance paclitaxel antitumor activity against breast and lung cancer cells in vitro. Sci Rep. 2017;7(1):13506. https://doi.org/10.1038/s41598-017-13816-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Jordan MA, Wilson L. Microtubules as a target for anticancer drugs. Nat Rev Cancer. 2004;4(4):253–65.

    Article  CAS  PubMed  Google Scholar 

  126. Farina V. Chemistry and pharmacology of taxol® and its derivatives, The. Pharmacochemistry Library, Volume 22. Elsevier Science & Technology; 1995.

  127. Kim SC, Kim DW, Shim YH, Bang JS, Oh HS, Kim SW, Seo MH. In vivo evaluation of polymeric micellar paclitaxel formulation: toxicity and efficacy. J Control Release. 2001;72(1–3):191–202.

    Article  CAS  PubMed  Google Scholar 

  128. Bajorin DF. Paclitaxel in the treatment of advanced urothelial cancer. Oncology (Williston Park). 2000;14(1):43–52.

    CAS  PubMed  Google Scholar 

  129. Kampan NC, Madondo MT, McNally OM, Quinn M, Plebanski M. Paclitaxel and its evolving role in the management of ovarian cancer. BioMed Re int. 2015;2015:1.

    Article  Google Scholar 

  130. Abu Samaan TM, Samec M, Liskova A, Kubatka P, Büsselberg D. Paclitaxel’s mechanistic and clinical effects on breast cancer. Biomolecules. 2019;9(12):789.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. McGuire WP, Rowinsky EK, Rosenshein NB, Grumbine FC, Ettinger DS, Armstrong DK, Donehower RC. Taxol: a unique antineoplastic agent with significant activity in advanced ovarian epithelial neoplasms. Ann Intern Med. 1989;111(4):273–9.

    Article  CAS  PubMed  Google Scholar 

  132. Sparano JA, Wang M, Martino S, Jones V, Perez EA, Saphner T, Wolff AC, Sledge GW Jr, Wood WC, Davidson NE. Weekly paclitaxel in the adjuvant treatment of breast cancer. N Engl J Med. 2008;358(16):1663–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Schiller JH, Harrington D, Belani CP, Langer C, Sandler A, Krook J, Zhu J, Johnson DH. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med. 2002;346(2):92–8.

    Article  CAS  PubMed  Google Scholar 

  134. Gill PS, Tulpule A, Espina BM, Cabriales S, Bresnahan J, Ilaw M, Louie S, Gustafson NF, Brown MA, Orcutt C. Paclitaxel is safe and effective in the treatment of advanced AIDS-related Kaposi’s sarcoma. J Clin Oncol. 1999;17(6):1876–1876.

    Article  CAS  PubMed  Google Scholar 

  135. Gradishar WJ. Taxanes for the treatment of metastatic breast cancer. Breast Cancer Basic Clin Res. 2012;6:BCBCR-S8205.

    Article  Google Scholar 

  136. Zhou X, Zhu H, Liu L, Lin J, Tang K. A review: recent advances and future prospects of taxol-producing endophytic fungi. Appl Microbiol Biotechnol. 2010;86:1707–17.

    Article  CAS  PubMed  Google Scholar 

  137. Croteau R, Ketchum REB, Long RM, Kaspera R, Wildung MR. Taxol biosynthesis and molecular genetics. Phytochem Rev. 2006;5:75–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Debbab A, Aly AH, Edrada-Ebel R, Wray V, Müller WEG, Totzke F, Zirrgiebel U, Schachtele C, Kubbutat MHG, Lin WH. Bioactive metabolites from the endophytic fungus Stemphylium globuliferum isolated from Mentha pulegium. J Nat Prod. 2009;72(4):626–31.

    Article  CAS  PubMed  Google Scholar 

  139. Masters JJ, Link JT, Snyder LB, Young WB, Danishefsky SJ. A total synthesis of taxol. Angew Chem Int Ed Engl. 1995;34(16):1723–6.

    Article  CAS  Google Scholar 

  140. Patel RN. Tour de paclitaxel: biocatalysis for semisynthesis. Annu Rev Microbiol. 1998;52(1):361–95.

    Article  CAS  PubMed  Google Scholar 

  141. Yukimune Y, Tabata H, Higashi Y, Hara Y. Methyl jasmonate-induced overproduction of paclitaxel and baccatin III in Taxus cell suspension cultures. Nat Biotechnol. 1996;14(9):1129–32.

    Article  CAS  PubMed  Google Scholar 

  142. Wang W-X, Kusari S, Spiteller M. Unraveling the chemical interactions of fungal endophytes for exploitation as microbial factories. Fungal Appl Sustain Environ Biotechnol. 2016;2016:353–70.

    Article  Google Scholar 

  143. Swamy MK, Das T, Nandy S, Mukherjee A, Pandey DK, Dey A. Endophytes for the production of anticancer drug, paclitaxel. In: Paclitaxel. London: Elsevier; 2022. p. 203–28.

  144. Mirjalili MH, Farzaneh M, Bonfill M, Rezadoost H, Ghassempour A. Isolation and characterization of Stemphylium sedicola SBU-16 as a new endophytic taxol-producing fungus from Taxus baccata grown in Iran. FEMS Microbiol Lett. 2012;328(2):122–9.

    Article  CAS  PubMed  Google Scholar 

  145. Qiao W, Ling F, Yu L, Huang Y, Wang T. Enhancing taxol production in a novel endophytic fungus, Aspergillus aculeatinus Tax-6, isolated from Taxus chinensis var. mairei. Fungal Biol. 2017;121(12):1037–44.

    Article  CAS  PubMed  Google Scholar 

  146. Senthil Kumaran R, Muthumary J, Hur BK. Production of taxol from Phyllosticta spinarum, an endophytic fungus of Cupressus sp. Eng Life Sci. 2008;8(4):438–46.

    Article  Google Scholar 

  147. Gangadevi V, Murugan M, Muthumary J. Taxol determination from Pestalotiopsis pauciseta, a fungal endophyte of a medicinal plant. Chin J Biotechnol. 2008;24(8):1433–8.

    Article  CAS  Google Scholar 

  148. Pandi M, Kumaran RS, Choi Y-K, Kim HJ, Muthumary J. Isolation and detection of taxol, an anticancer drug produced from Lasiodiplodia theobromae, an endophytic fungus of the medicinal plant Morinda citrifolia. Afr J Biotech. 2011;10(8):1428–35.

    CAS  Google Scholar 

  149. Rajendran L, Rajagopal K, Subbarayan K, Ulagappan K, Sampath A, Karthik G. Efficiency of fungal taxol on human liver carcinoma cell lines. Am J Res Commun. 2013;1:112–21.

    Google Scholar 

  150. Vennila R, Kamalraj S, Muthumary J. In vitro studies on anticancer activity of fungal taxol against human breast cancer cell line MCF-7 cells. Asian Pac J Trop Biomed. 2012;2(2):S1159–61.

    Article  Google Scholar 

  151. Ismaiel AA, Ahmed AS, Hassan IA, El-Sayed E-SR, Karam El-Din A-ZA. Production of paclitaxel with anticancer activity by two local fungal endophytes, Aspergillus fumigatus and Alternaria tenuissima. Appl Microbiol Biotechnol. 2017;101:5831–46.

    Article  CAS  PubMed  Google Scholar 

  152. Raj KG, Manikandan R, Arulvasu C, Pandi M. Anti-proliferative effect of fungal taxol extracted from Cladosporium oxysporum against human pathogenic bacteria and human colon cancer cell line HCT 15. Spectrochim Acta Part A Mol Biomol Spectrosc. 2015;138:667–74.

    Article  Google Scholar 

  153. Chakravarthi BVSK, Sujay R, Kuriakose GC, Karande AA, Jayabaskaran C. Inhibition of cancer cell proliferation and apoptosis-inducing activity of fungal taxol and its precursor baccatin III purified from endophytic Fusarium solani. Cancer Cell Int. 2013;13:1–11.

    Article  Google Scholar 

  154. Suresh G, Kokila D, Suresh TC, Kumaran S, Velmurugan P, Vedhanayakisri KA, Sivakumar S, Ravi AV. Mycosynthesis of anticancer drug taxol by Aspergillus oryzae, an endophyte of Tarenna asiatica, characterization, and its activity against a human lung cancer cell line. Biocatal Agric Biotechnol. 2020;24:101525.

    Article  Google Scholar 

  155. Abbas NF, Shabana ME-A, Habib FM, Soliman AA. Histopathological and immunohistochemical study of matrix metalloproteinase-2 and matrix metalloproteinase-9 in breast carcinoma. J Arab Soc Med Res. 2017;12(1):6.

    Google Scholar 

  156. Zein N, Aziz SW, El-Sayed AS, Sitohy B. Comparative cytotoxic and anticancer effect of Taxol derived from Aspergillus terreus and Taxus brevifolia. Biosci Res. 2019;16(2):1500–9.

    Google Scholar 

  157. Pandi M, Manikandan R, Muthumary J. Anticancer activity of fungal taxol derived from Botryodiplodia theobromae Pat., an endophytic fungus, against 7, 12 dimethyl benz (a) anthracene (DMBA)-induced mammary gland carcinogenesis in Sprague dawley rats. Biomed Pharmacother. 2010;64(1):48–53.

    Article  CAS  PubMed  Google Scholar 

  158. Weydert CJ, Waugh TA, Ritchie JM, Iyer KS, Smith JL, Li L, Spitz DR, Oberley LW. Overexpression of manganese or copper–zinc superoxide dismutase inhibits breast cancer growth. Free Radical Biol Med. 2006;41(2):226–37.

    Article  CAS  Google Scholar 

  159. Simon HU, Haj-Yehia A, Levi-Schaffer F. Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis. 2000;5:415–8.

    Article  CAS  PubMed  Google Scholar 

  160. Guo B, Wang Y, Zhou X, Hu K, Tan F, Miao Z, Tang K. An endophytic taxol-producing fungus BT2 isolated from Taxus chinensis var. mairei. Afr J Biotechnol. 2006;5(10):1.

    CAS  Google Scholar 

  161. Caruso M, Colombo AL, Fedeli L, Pavesi A, Quaroni S, Saracchi M, Ventrella G. Isolation of endophytic fungi and actinomycetes taxane producers. Ann Microbiol. 2000;50(1):3–14.

    CAS  Google Scholar 

  162. Somjaipeng S, Medina A, Kwaśna H, Ortiz JO, Magan N. Isolation, identification, and ecology of growth and taxol production by an endophytic strain of Paraconiothyrium variabile from English yew trees (Taxus baccata). Fungal Biol. 2015;119(11):1022–31.

    Article  CAS  PubMed  Google Scholar 

  163. Kasaei A, Mobini-Dehkordi M, Mahjoubi F, Saffar B. Isolation of taxol-producing endophytic fungi from Iranian yew through novel molecular approach and their effects on human breast cancer cell line. Curr Microbiol. 2017;74:702–9.

    Article  CAS  PubMed  Google Scholar 

  164. Garyali S, Kumar A, Reddy MS. Taxol production by an endophytic fungus, Fusarium redolens, isolated from Himalayan yew. J Microbiol Biotechnol. 2013;23(10):1372–80.

    Article  CAS  PubMed  Google Scholar 

  165. Stierle A, Strobel G, Stierle D. Taxol and taxane production by Taxomyces andreanae, an endophytic fungus of Pacific yew. Science. 1993;260(5105):214–6.

    Article  CAS  PubMed  Google Scholar 

  166. Chakravarthi B, Das P, Surendranath K, Karande AA, Jayabaskaran C. Production of paclitaxel by Fusarium solani isolated from Taxus celebica. J Biosci. 2008;33:259–67.

    Article  CAS  PubMed  Google Scholar 

  167. Liu K, Ding X, Deng B, Chen W. Isolation and characterization of endophytic taxol-producing fungi from Taxus chinensis. J Ind Microbiol Biotechnol. 2009;36(9):1171.

    Article  CAS  PubMed  Google Scholar 

  168. Deng BW, Liu KH, Chen WQ, Ding XW, Xie XC. Fusarium solani, Tax-3, a new endophytic taxol-producing fungus from Taxus chinensis. World J Microbiol Biotechnol. 2009;25:139–43.

    Article  CAS  Google Scholar 

  169. Miao Z, Wang Y, Yu X, Guo B, Tang K. A new endophytic taxane production fungus from Taxus chinensis. Appl Biochem Microbiol. 2009;45:81–6.

    Article  CAS  Google Scholar 

  170. Kumaran RS, Hur BK. Screening of species of the endophytic fungus Phomopsis for the production of the anticancer drug taxol. Biotechnol Appl Biochem. 2009;54(1):21–30.

    Article  CAS  PubMed  Google Scholar 

  171. Zhao K, Ping W, Li Q, Hao S, Zhao L, Gao T, Zhou D. Aspergillus niger var. taxi, a new species variant of taxol-producing fungus isolated from Taxus cuspidata in China. J Appl Microbiol. 2009;107(4):1202–7.

    Article  CAS  PubMed  Google Scholar 

  172. Chang-Tian L, Yu L, Wang Q-J, Sung C-K. Taxol production by Fusarium arthrosporioides isolated from yew, Taxus cuspidata. J Med Biochem. 2008;27(4):454–8.

    Article  Google Scholar 

  173. Xu F, Tao W, Cheng L, Guo L. Strain improvement and optimization of the media of taxol-producing fungus Fusarium maire. Biochem Eng J. 2006;31(1):67–73.

    Article  CAS  Google Scholar 

  174. Wang J, Li G, Lu H, Zheng Z, Huang Y, Su W. Taxol from Tubercularia sp. strain TF5, an endophytic fungus of Taxus mairei. FEMS Microbiol Lett. 2000;193(2):249–53.

    Article  CAS  PubMed  Google Scholar 

  175. Xiong Z-Q, Yang Y-Y, Zhao N, Wang Y. Diversity of endophytic fungi and screening of fungal paclitaxel producer from Anglojap yew, Taxus x media. BMC Microbiol. 2013;13(1):1–10.

    Article  Google Scholar 

  176. Zhang P, Zhou P-P, Yu L-J. An endophytic taxol-producing fungus from Taxus media, Cladosporium cladosporioides MD2. Curr Microbiol. 2009;59:227–32.

    Article  CAS  PubMed  Google Scholar 

  177. Soliman SSM, Tsao R, Raizada MN. Chemical inhibitors suggest endophytic fungal paclitaxel is derived from both mevalonate and non-mevalonate-like pathways. J Nat Prod. 2011;74(12):2497–504.

    Article  CAS  PubMed  Google Scholar 

  178. Strobel G, Yang X, Sears J, Kramer R, Sidhu RS, Hess WM. Taxol from Pestalotiopsis microspora, an endophytic fungus of Taxus wallachiana. Microbiology. 1996;142(2):435–40.

    Article  CAS  PubMed  Google Scholar 

  179. Shrestha K, Strobel GA, Shrivastava SP, Gewali MB. Evidence for paclitaxel from three new endophytic fungi of Himalayan yew of Nepal. Planta Med. 2001;67(04):374–6.

    Article  CAS  PubMed  Google Scholar 

  180. Gangadevi V, Muthumary J. Taxol, an anticancer drug produced by an endophytic fungus Bartalinia robillardoides Tassi, isolated from a medicinal plant, Aegle marmelos Correa ex Roxb. World J Microbiol Biotechnol. 2008;24:717–24.

    Article  CAS  Google Scholar 

  181. Zaiyou J, Li M, Xiqiao H. An endophytic fungus efficiently producing paclitaxel isolated from Taxus wallichiana var. mairei. Medicine. 2017;96(27):1.

    Article  Google Scholar 

  182. Hemamalini V, Kumar DM, Rebecca AIN, Srimathi S, Muthumary J, Kalaichelvan P. Isolation and characterization of taxol producing endophytic Phoma sp. from Calotropis gigantea and its anti-proliferative studies. J Acad Ind Res. 2015;3:645–9.

    CAS  Google Scholar 

  183. Das A, Rahman MI, Ferdous AS, Amin A, Rahman MM, Nahar N, Uddin MA, Islam MR, Khan H. An endophytic Basidiomycete, Grammothele lineata, isolated from Corchorus olitorius, produces paclitaxel that shows cytotoxicity. PLoS ONE. 2017;12(6):e0178612.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Kumaran RS, Muthumary J, Kim E-K, Hur B-K. Production of taxol from Phyllosticta dioscoreae, a leaf spot fungus isolated from Hibiscus rosa-sinensis. Biotechnol Bioprocess Eng. 2009;14:76–83.

    Article  CAS  Google Scholar 

  185. Gangadevi V, Muthumary J. Isolation of Colletotrichum gloeosporioides, a novel endophytic taxol-producing fungus from the leaves of a medicinal plant, Justicia gendarussa. Mycol Balcanica. 2008;5(1):1–4.

    Google Scholar 

  186. Rebecca AIN, Hemamalini V, Kumar DM, Srimathi S, Muthumary J, Kalaichelvan PT. Endophytic Chaetomium sp. from Michelia champaca L. and its taxol production. J Acad Ind Res. 2012;1(68):72.

    Google Scholar 

  187. Andrade HFD, Araújo LCAD, Santos BSD, Paiva PMG, Napoleão TH, Correia MTDS, Oliveira MBMD, Lima GMDS, Ximenes RM, Silva TDD. Screening of endophytic fungi stored in a culture collection for taxol production. Braz J Microbiol. 2018;49:59–63.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Roopa G, Madhusudhan MC, Sunil KCR, Lisa N, Calvin R, Poornima R, Zeinab N, Kini KR, Prakash HS, Geetha N. Identification of Taxol-producing endophytic fungi isolated from Salacia oblonga through genomic mining approach. J Genetic Eng Biotechnol. 2015;13(2):119–27.

    Article  CAS  Google Scholar 

  189. Elavarasi A, Rathna GS, Kalaiselvam M. Taxol producing mangrove endophytic fungi Fusarium oxysporum from Rhizophora annamalayana. Asian Pac J Trop Biomed. 2012;2(2):S1081–5.

    Article  Google Scholar 

  190. Gill H, Vasundhara M. Isolation of taxol producing endophytic fungus Alternaria brassicicola from non-Taxus medicinal plant Terminalia arjuna. World J Microbiol Biotechnol. 2019;35:1–8.

    Article  CAS  Google Scholar 

  191. Gangadevi V, Muthumary J. A novel endophytic taxol-producing fungus Chaetomella raphigera isolated from a medicinal plant, Terminalia arjuna. Appl Biochem Biotechnol. 2009;158:675–84.

    Article  CAS  PubMed  Google Scholar 

  192. Li JY, Sidhu RS, Ford EJ, Long DM, Hess WM, Strobel GA. The induction of taxol production in the endophytic fungus—Periconia sp from Torreya grandifolia. J Ind Microbiol Biotechnol. 1998;20:259–64.

    Article  CAS  Google Scholar 

  193. Kumaran RS, Choi Y-K, Lee S, Jeon HJ, Jung H, Kim HJ. Isolation of taxol, an anticancer drug produced by the endophytic fungus, Phoma betae. Afr J Biotechnol. 2012;11(4):950–60.

    CAS  Google Scholar 

  194. Gangadevi V, Muthumary J. Preliminary studies on cytotoxic effect of fungal taxol on cancer cell lines. Afr J Biotechnol. 2007;6(12):1.

    Google Scholar 

  195. Wang Y, Tang K. A new endophytic taxol-and baccatin III-producing fungus isolated from Taxus chinensis var. mairei. Afr J Biotechnol. 2011;10(72):16379–86.

    CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

S.M.S. was supported by a National Priorities Research Program Grant (NPRP11S-1214-170101; awarded to Professor Dr. Dietrich Büsselberg, June 2019–Current) from the Qatar National Research Fund (QNRF, a member of Qatar Foundation). The statements made herein are solely the responsibility of the authors.

Author information

Authors and Affiliations

Authors

Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis, and interpretation, or in all these areas that is, revising or critically reviewing the article; giving final approval of the version to be published; agreeing on the journal to which the article has been submitted; and confirming to be accountable for all aspects of the work. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Dietrich Büsselberg, Samson Mathews Samuel or Javad Sharifi-Rad.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

Authors wish to confirm that there are no known conflicts of interest associated with this publication and there has been no significant financial support for this work that could have influenced its outcome.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sati, P., Sharma, E., Dhyani, P. et al. Paclitaxel and its semi-synthetic derivatives: comprehensive insights into chemical structure, mechanisms of action, and anticancer properties. Eur J Med Res 29, 90 (2024). https://doi.org/10.1186/s40001-024-01657-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-024-01657-2

Keywords