Download PDF
Review  |  Open Access  |  18 Oct 2023

Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistance

Views: 822 |  Downloads: 161 |  Cited:   4
Cancer Drug Resist 2023;6:729-47.
10.20517/cdr.2023.78 |  © The Author(s) 2023.
Author Information
Article Notes
Cite This Article

Abstract

The induction of cell death is recognized as a potent strategy for cancer treatment. Apoptosis is an extensively studied form of cell death, and multiple anticancer drugs exert their therapeutic effects by inducing it. Nonetheless, apoptosis evasion is a hallmark of cancer, rendering cancer cells resistant to chemotherapy drugs. Consequently, there is a growing interest in exploring novel non-apoptotic forms of cell death, such as ferroptosis, necroptosis, pyroptosis, and paraptosis. Natural compounds with anticancer properties have garnered significant attention due to their advantages, including a reduced risk of drug resistance. Over the past two decades, numerous natural compounds have been discovered to exert anticancer and anti-resistance effects by triggering these four non-apoptotic cell death mechanisms. This review primarily focuses on these four non-apoptotic cell death mechanisms and their recent advancements in overcoming drug resistance in cancer treatment. Meanwhile, it highlights the role of natural compounds in effectively addressing cancer drug resistance through the induction of these forms of non-apoptotic cell death.

Keywords

Drug resistance, cancer therapy, natural compound, non-apoptotic cell death

INTRODUCTION

Cancer, as a major global public health concern, poses a severe threat to people’s well-being. In 2018, the World Health Organization reported that cancer ranked the second leading cause of death globally, resulting in approximately 9.6 million annual fatalities[1]. Cell death is a fundamental process crucial for human health, playing a pivotal role in regulating cell division, facilitating organ development, and upholding tissue homeostasis[2]. Nevertheless, the disruption and avoidance of cell death mechanisms facilitate malignant cell transformation and advance tumorigenesis[3]. From a therapeutic perspective, conventional cancer treatments, such as chemotherapy and radiation, achieve their anticancer effects by inducing cell death[2].

Over the span of several decades, there has been a growing comprehension of the diverse mechanisms governing cell death. This process can be classified into two primary categories: programmed cell death (PCD), which is genetically controlled, and unprogrammed cell death, which represents a passive response to both biotic and abiotic stress. Among the various forms of PCD, apoptosis has emerged as one of the earliest and most extensively explored pathways. For a significant duration, the development of anticancer drugs targeting apoptosis has been a focal point of research. Numerous drugs, such as cisplatin, oxaliplatin, and pirarubicin, have showcased their ability to elicit anticancer effects through this mechanism[4,5]. However, recent studies have increasingly validated that cancer cells possess the capability to elude apoptosis through a range of mechanisms, including the overexpression of apoptosis-inhibiting proteins, the inhibition of apoptosis-inducing factors, and the activation of survival signaling pathways[6]. This evasion of apoptosis directly contributes to chemoresistance, a phenomenon intricately associated with alterations in various aspects of cancer, encompassing angiogenesis, the tumor microenvironment, and oxidative stress[7-10]. Ultimately, this leads to the failure of cancer treatment. To improve the efficacy of cancer treatment, the exploration of non-apoptotic cell death mechanisms has gradually emerged as a prominent priority. Over the last two decades, researchers have sequentially unveiled and extensively investigated novel different cell death mechanisms, including ferroptosis, necroptosis, pyroptosis, and paraptosis. These unique mechanisms, each with its distinct regulators and pathways, hold the potential to be activated within apoptosis-resistant cancer cells, offering novel strategies for the treatment of cancer and overcoming cancer drug resistance.

Throughout history, natural compounds derived from plants, animals, microorganisms, and minerals have consistently served as a valuable source for drug discovery[11,12]. Unlike synthetic compounds, these substances are not artificially created and can be categorized into various forms, such as alkaloids, flavonoids, and terpenes, due to their diverse chemical structures[13]. Many of these natural compounds exhibit significant potential in the realm of cancer treatment. In fact, certain natural compounds, such as paclitaxel (PTX), camptothecin, and vincristine, have already gained widespread acceptance as chemotherapeutic drugs in clinical practice[14,15]. Moreover, an interesting aspect is that numerous natural compounds have been found to induce various non-apoptotic cell death pathways when administered to resistant cancer cells that evade apoptosis.

This review focuses on ferroptosis, necroptosis, pyroptosis, and paraptosis, providing a comprehensive overview of the latest research advancements in these mechanisms within the framework of cancer. We place specific emphasis on their relevance in the context of combatting cancer drug resistance. Additionally, this review compiles information on natural compounds with the capacity to induce these four modes of cell death in the context of addressing cancer resistance over the past two decades.

FERROPTOSIS

Overview of ferroptosis

Ferroptosis, a recently discovered form of PCD that is iron-dependent, was first proposed in 2012[16]. Morphologically, ferroptotic cells exhibit intact nuclei without chromatin condensation. However, their mitochondria undergo significant changes, including reduced size, increased membrane density, reduced cristae, and outer membrane rupture[17]. Interestingly, the discovery of ferroptosis inducers was earlier than its naming. Yang et al. discovered some new compounds as early as 2003 and 2008, including Erastin, RSL3, and RSL5, which induce cell death through a mechanism distinct from apoptosis[18]. Erastin reduces cysteine uptake by inhibiting cysteine/glutamate transporter receptor, known as System Xc-, resulting in a decrease in glutathione (GSH) synthesis and an increase in iron-dependent lipid peroxidation (LPO), which finally leads cells to ferroptosis[19]. Differently, RSL3 directly inhibits the activity of Glutathione Peroxidase 4 (GPX4), a GSH-utilizing enzyme that prevents the accumulation of toxic lipid hydroperoxide, thereby inducing ferroptosis[20]. Additionally, Erastin also indirectly triggers ferroptosis through the inactivation of GPX4 via the inhibition of GSH synthesis[21].

It is known that LPO and intracellular iron accumulation play pivotal roles in triggering ferroptosis. Polyunsaturated fatty acids (PUFAs) are the most susceptible lipids to peroxidation during ferroptosis. The regulation of PUFA synthesis may be influenced by enzymes such as Long-chain acyl-CoA synthetases (ACSLs) and lyso-phosphatidylcholine acyltransferase-3 (LPCAT3). Additionally, arachidonate lipoxygenases (ALOXs) and cytochrome p450s (POR) can directly or indirectly modulate PUFA peroxidation, resulting in ferroptosis[22]. Additionally, iron metabolism is crucial for ferroptosis. As Fe2+ is released from the labile iron pool (LIP) into the cytoplasm, excess Fe2+ oxidizes PUFAs to hydroxyl radicals and leads to ferroptosis.

The mitochondria are responsible for cellular metabolism and also play an important role in the regulation of ferroptosis[23]. Within the mitochondria, dihydroorotate dehydrogenase (DHODH) reduces ubiquinone (CoQ) to ubiquinol (CoQH2), serving as a radical-trapping antioxidant with anti-ferroptosis activity. DHODH plays a significant role in mediating ferroptosis defense independent of the GSH pathway[24]. Similarly, the plasma enzyme ferroptosis suppressor protein 1 (FSP1) inhibits lipid hydroperoxides by reducing ubiquinone to ubiquinol, operating in parallel with GPX4 to counteract ferroptosis[25]. Obviously, the mechanisms of ferroptosis are complex and being explored constantly [Figure 1].

Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistance

Figure 1. The mechanism of ferroptosis. Ferroptosis can be initiated by an increase in intracellular iron levels and the accumulation of iron-dependent lipid peroxidation. It can be induced through the SystemXc-/GSH/GPX4 axis, the DHODH/CoQ10 axis, and the FSP1/CoQ10 axis. ACSL: Acyl-CoA synthetase; ALOXs: arachidonate lipoxygenases; CoQ: ubiquinone; FSP1: ferroptosis suppressor protein 1; GPX4: glutathione peroxidase 4; GSH: glutathione; LPCAT3: lyso-phosphatidylcholine acyltransferase-3; LIP: labile iron pool; POR: cytochrome p450s; PUFA: polyunsaturated fatty acid.

Ferroptosis pathways for chemotherapy resistance in cancer

In recent years, ferroptosis has been a hot topic in cancer development, treatment and cancer drug resistance. Several signaling pathways have been found to participate in promoting cancer development and drug resistance through the inhibition of ferroptosis. With multiple functions for proliferation, metastasis, and differentiation of cancer cells, the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of the rapamycin (mTOR) signaling pathway is responsible for ferroptosis prevention via sterol regulatory element-binding protein 1 (SREBP1)/stearoyl-CoA desaturase-1 (SCD1)-mediated adipogenesis[26]. Hippo pathway activity is also responsible for cell growth and proliferation. Activating the Hippo pathway can suppress downstream YAP, leading cancer cells resistant to ferroptosis by downregulating acyl-CoA synthetase long-chain family member 4 (ACSL4) and TCP friendly rate control (TFRC)[27]. Since RAS may regulate some processes to escape ferroptosis, RAS-mutated cells are always susceptible to ferroptosis. High-mobility group box 1 (HMGB1), a leukemia pathogenic gene, inhibits LPO via the RAS/MAP kinase (MAPK) pathway, promoting ferroptosis resistance[28].

Additionally, several other pathways, including nuclear factor erythroid 2-related factor 2 (Nrf2), p53, and hypoxia-inducible factor (HIF), are also involved in chemotherapeutic resistance through ferroptosis regulation. Recent research highlights the significant impact of the Nrf2 signaling pathway on both organ protection and resistance to cisplatin (DDP) across various cancer types[29]. For instance, in non-small cell lung cancer, Erastin and Sorafenib, either alone or in combination, induce ferroptosis by inhibiting the Nrf2/SLC7A11 (also known as xCT) pathway, thereby overcoming DDP resistance[30]. The p53 gene, extensively studied in cancer research, influences metabolic pathways related to ferroptosis, such as enhancing ferroptosis by down-regulating xCT expression[31]. According to this mechanism, Flubendazole and 5-fluorouracil (5-FU) demonstrate synergistic effects in treating castration-resistant prostate cancer[32]. HIF serves a dual role in the regulation of ferroptosis in cancer cells. Lowering HIF-α levels has been found to increase LPO and enhance ferroptosis in clear cell renal cell carcinoma[33]. Conversely, in temozolomide-resistant glioblastoma, activating HIF-1α and HIF-2α can induce ferroptosis, with HIF-2α possibly promoting LPO as the primary mechanism[34].

Natural compounds inducing ferroptosis for cancer treatment

Many natural compounds have been found to induce ferroptosis via single or combinational therapies, offering opportunities for cancer treatment and drug resistance [Table 1]. Here, we summarize those natural compounds that reverse drug resistance by inducing ferroptosis. Most of these compounds can synergize with chemotherapeutic drugs by regulating ferroptosis-related proteins and genes. Recent studies have revealed that sorafenib can induce ferroptosis in various types of cancer[35]. However, sorafenib-induced ferroptosis can be suppressed, specifically through the activation of the PI3K/Akt signaling pathway or the NRF2/GPX4 axis[36,37]. To overcome this challenge, drug combinations have proven to be effective strategies. Dihydroartemisinin, a derivative of artemisinin, shares similar mechanisms with sorafenib regarding ferroptosis-related proteins, such as GPX4, and has a stronger effect in liver cancer cells when combined with sorafenib[38]. Ursolic acid, a pentacyclic triterpene compound, also exhibits synergistic anticancer effects with sorafenib by inhibiting xCT in many cancers[39]. While DDP is considered a front-line chemotherapy drug for various cancers, including ovarian cancer, its resistance poses a significant impediment to achieving effective treatment outcomes[40]. Inhibition of apoptosis plays a significant role in contributing to DDP resistance, whereas the induction of novel forms of cell death, such as ferroptosis, has been shown to effectively kill DDP-resistant cancer cells that evade apoptosis[41]. The combination of DDP with Shikonin, a hydroxy-1,4-naphthoquinone isolated from Lithospermum erythrorhizon Sieb. et Zucc.(Boraginaceae), promotes Fe2+ accumulation by upregulating heme oxygenase 1 (HMOX1), initiating ferroptosis in DDP-resistant ovarian cancer cells[42]. Piperlongumine, an alkaloid derived from long pepper (Piper longum L.), exhibits anticancer activity in lung cancer cells by targeting the glutathione regeneration enzyme, thioredoxin reductase 1 (TXNRD1). Although it does not induce ferroptosis, it can significantly enhance erastin-induced LPO[43].

Table 1

Natural compounds for anticancer drug resistance by inducing ferroptosis

CompoundsOriginStructureCancerAnti-drug resistant effectsRefs
ArtesunateArtemisia annua L.Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceRenal cell carcinomaIncreasing cytotoxicity in sunitinib-resistant renal cell carcinoma by triggering ferroptosis, increasing ROS generation, and decreasing metabolism.[50]
ChrysinOroxylum indicum (L.) KurzRecent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistancePancreatic cancerInhibiting CBR1 activity in gemcitabine-resistant pancreatic cancer to trigger ferroptosis through ROS accumulation.[48]
DihydroartemisininArtemisia annua L.Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceHepatocellular carcinomaSynergizing with sorafenib to induce ferroptosis by increasing the levels of L-ROS, LIP, and MDA and decreasing the level of GSH.[38,127-129]
GinkgetinGinkgo biloba L.Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceNon-small cell lung cancerSynergizing with DDP to induce ferroptosis by increasing ROS formation, decreasing the expression of xCT and GPX4, and inactivating the Nrf2/HO-1 axis.[46]
KayadiolTorreya nucifera Sieb. et Zucc. (Taxaceae)Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceExtranodal natural killer/T cell lymphomaInducing p53-mediated ferroptosis through the xCT/GPX4 axis and exhibiting synergistic effects when combined with L-asparaginase and DDP.[49]
PiperlonguminePiper longum L.Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistancePancreatic cancerEnhancing the antitumor effects of erastin by inducing ROS generation, GSH depletion and inhibiting TXNRD activity.[43,130]
ShikoninLithospermum erythrorhizon Sieb. et Zucc.Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceOvarian cancerSynergizing with DDP to induce ferroptosis through upregulation of HMOX1 and increased levels of ROS, LPO, and Fe2+.[42]
TilirosideTribulus terrestris L.Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceHepatocellular carcinomaEnhancing the antitumor effects of sorafenib by inducing ferroptosis via targeting TBK1 to promote Keap1-mediated Nrf2 ubiquitination and degradation.[45]
Ursolic acidLigustrum lucidum W. T. AitonRecent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceColon cancer, gastric cancer, prostate cancerEnhancing the antitumor effects of sorafenib by inducing xCT-dependent ferroptosis.[39]
Withaferin AWithania somnifera (L.) DunalRecent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceHepatocellular carcinomaEnhancing the antitumor effects of sorafenib in sorafenib-resistant hepatocellular carcinoma cells by regulating Keap1/Nrf2-associated ferroptosis and EMT.[44]

In addition to ferroptosis-related genes and proteins, some natural compounds induce ferroptosis via regulation of the Nrf2 signaling pathway. Withaferin A, a steroidal lactone isolated from the medicinal plant Ashwagandha [Withania somnifera (L.) Dunal], and Tiliroside, a flavonoid found in the herbs of Tribulus terrestris L., both modulate the Kelch-like ECH-associated protein 1 (Keap1)/Nrf2 pathway in hepatocellular carcinoma. Withaferin A enhances sorafenib sensitivity in sorafenib-resistant hepatocellular carcinoma cells by regulating the Keap1/Nrf2-associated epithelial-to-mesenchymal transition (EMT) and ferroptosis[44]. Additionally, tiliroside synergistically combines with sorafenib to inhibit Tank-binding kinase (TBK1) activity, prompting Keap1-mediated Nrf2 ubiquitination and degradation, leading to ferroptosis in hepatocellular carcinoma cells[45]. Ginkgetin, a natural biflavonoid isolated from the leaves of Ginkgo biloba L., has been found to induce ferroptosis in non-small cell lung cancer cells through decreased expression of xCT and GPX4, decreased GSH/glutathione disulfide (GSSG) ratio, and inactivation of the Nrf2/HMOX1 axis. Interestingly, it can promote DDP-induced anticancer activity, which is also a result of ferroptosis induction[46].

As a key regulator of iron homeostasis, ferritin plays a vital role in storing intracellular free iron and is involved in ferritinophagy, a form of autophagic ferroptosis. In ferritinophagy, the autophagic cargo receptor nuclear receptor coactivator 4 (NCOA4) binds to ferritin heavy chains (FTH1) and delivers it to autophagosomes for degradation and iron release[47]. Human carbonyl reductase 1 (CBR1) contributes to gemcitabine resistance in pancreatic cancer. The upregulation of CBR1 induced by gemcitabine inhibits the antitumor effects of the drug. Conversely, reducing CBR1 activity enhances the sensitivity of cancer cells to gemcitabine, thereby improving its therapeutic efficacy. Chrysin, a natural bioflavonoid compound, has been discovered to induce ferritinophagy, thus enhancing gemcitabine sensitivity in pancreatic cancer cells. In Chrysin-treated cells, there is a deregulation of FTH1 and an increase in intracellular free iron levels, followed by the inhibition of CBR1, which is involved in the induction of ferroptosis[48].

The tumor suppressor protein p53 also plays an essential role in ferroptosis in certain cancers. As a diterpenoid extracted from Torreya nucifera, kayadiol exhibits anticancer properties through p53-mediated ferroptosis in NK/T lymphoma cells, and it could synergistically combine with L-asparaginase and DDP[49]. Artesunate (AST), another derivative of artemisinin, was found to inhibit the growth of sunitinib-resistant renal cell carcinoma cells by both inhibiting cell cycle progression and inducing ferroptosis. Interestingly, the induction of ferroptosis was associated with its inhibitory effect only in renal cell carcinoma cells expressing p53, suggesting that AST induces p53-dependent ferroptosis[50].

NECROPTOSIS

Overview of necroptosis

Necroptosis, first described in 2005, is a form of cell death characterized by morphological features similar to necrosis, including a lack of nuclear chromatin, organelle swelling, and cell membrane disruption[51]. However, unlike necrosis, which is a passive and non-programmed form of cell death, necroptosis can be regulated by multiple signal transduction pathways[52].

The classical form of necroptotic cell death is mediated by tumor necrosis factor-α (TNF-α). Initially, TNF-α binds to its specific receptor TNF receptor 1 (TNFR1), promoting its trimerization and facilitating the recruitment of several proteins including receptor-interacting protein 1 (RIP1) kinase, TNF-α receptor-associated death domain (TRADD), cellular inhibitor of apoptosis 1 (cIAP1), cellular inhibitor of apoptosis 2 (cIAP2), TNFR-associated factor 2 (TRAF2), and TNFR-associated factor 5 (TRAF5) to form complex I. Within complex I, RIP1 can undergo polyubiquitination by TRAF2, TRAF5, cIAP1, and cIAP2. The ubiquitination status of RIP1 determines whether complex I activates the nuclear factor-κB (NF-κB) pathway to promote cell survival, or triggers cell death[53].

When the ubiquitylation of RIP is impaired, complex I can transform into complex IIa and complex IIb, leading to cell death[54]. Complex IIa can trigger caspase-8-dependent apoptosis in the absence of RIP1, whereas complex IIb depends on RIP1 for caspase-8 activation due to its deficiency in TRADD compared to complex IIa[55]. Additionally, complex IIa can transform into complex IIb. When the levels of receptor-interacting protein 3 (RIP3) and mixed lineage kinase domain-like protein (MLKL) are sufficiently high and caspase-8 is blocked, complex IIb may develop into a necrosome. Following the phosphorylation of RIP1 and RIP3, two core components of the necrosome, the activated RIP3 can further recruit and phosphorylate MLKL, triggering its oligomerization. The oligomerized MLKL is then translocated to the plasma membrane, increasing its permeability and ultimately leading to necroptotic cell death[56] [Figure 2]. In addition to TNF superfamily receptors, various other types of receptors, such as Toll-like receptors, T-cell receptors, and interferon receptors, also contribute to the activation of necroptosis[57].

Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistance

Figure 2. The classical pathway of necroptosis. The classical form of necroptosis begins with the binding of TNF-α to TNFR1, initiating the formation of complex I. If the ubiquitylation of RIP1 within complex I is inhibited, it undergoes a transition into complex IIA and complex IIB. When caspase-8 is inactive, complex IIB recruits RIP3 to form the necrosome. Once RIP3 is phosphorylated, it recruits and phosphorylates MLKL, leading to its oligomerization. The oligomerized MLKL is subsequently translocated to the plasma membrane, resulting in necroptosis. MLKL: Mixed lineage kinase domain-like protein; PIR1: receptor-interacting protein 1; RIP3: receptor-interacting protein 3; TNF: tumor necrosis factor; TNFR: TNF receptor; TRADD: TNF-α receptor-associated death domain TRAF: TNFR-associated factor.

Regulation of necroptosis for anti-drug resistance in cancer

As independent of caspase activation and involving distinct components from apoptotic pathways, necroptosis is an effective mechanism to overcome apoptosis resistance in cancer. However, some necroptotic core components are always lacking in cancer cells, resulting in the evasion of this mechanism. RIP3 is silenced in numerous cancer types, and this silencing is likely ascribed to genomic methylation near the RIP3 transcriptional start site or driven by oncogenes BRAF and AXL[58]. Current studies have revealed that demethylation treatment can activate necroptotic pathways by restoring the expression of RIP3. Moreover, upregulating RIPK3 expression can enhance the sensitivity of colon cancer cells to 5-FU and lung cancer cells to DDP through mediating necroptosis[59,60].

Furthermore, the low expression of MLKL appears to be associated with a poor patient prognosis in certain cancers, making it a potential novel potential prognostic biomarker for these cancers[61]. In addition, some compounds can induce MLKL-mediated necroptosis without the phosphorylation of RIP1 and RIP3, which presents a very promising prospect for future studies[62].

In addition to modulating necroptosis genes and proteins, increasing glycolytic metabolism may confer resistance to necroptosis in cancer cells under hypoxic conditions. The mechanism involves the suppression of RIP-dependent necroptosis through pyruvate scavenging of mitochondrial superoxide[63]. Several studies have suggested that inhibiting glycolysis may be a potential mechanism for necroptosis induction. For instance, selenite-induced necroptosis in prostate cancer resulted from the inhibition of glycolysis through adenosine triphosphate (ATP) depletion and phosphofructokinase activity reduction[64].

Natural compounds inducing necroptosis for cancer treatment

So far, many natural compounds can induce necroptosis in diverse cancer types, and some can also inhibit drug resistance [Table 2]. As mentioned above, modulating necroptosis core proteins is critical for necroptosis induction. By regulating the RIP1/reactive oxygen species (ROS)-mediated pathway, bufalin, an endogenous cardiotonic steroid, can induce necroptosis in adriamycin-resistant triple-negative breast cancer cell lines[65]. Numerous studies have indicated that when 5-FU is in combination with other anticancer agents, its therapeutic efficacy can be effectively enhanced[66]. Gambogenic acid is one of the main components of Gamboge which can be used in combination with 5-FU to upregulate necroptosis-related proteins such as RIP1 in lung cancer cells, thereby inducing necroptosis[67]. Piperlongumine can also activate RIP1 to produce excessive ROS, triggering necroptosis in DDP-resistant bladder cells[68]. Berberine can effectively enhance the antitumor effect of DDP in ovarian cancer by increasing the expression and activation of RIP3 and MLKL, thereby inducing necroptotic cell death[69]. In TNF-α induced necroptosis, MLKL is a key downstream component of RIP1 and RIP3. However, it is worth mentioning that tanshinol A, a phenolic compound extracted from Salvia miltiorrhiza Bunge, can trigger non-canonical necroptosis mediated by MLKL in lung cancer independently of RIP1 and RIP3[62].

Table 2

Natural compounds for anticancer drug resistance by inducing necroptosis

CompoundsOriginStructureCancerAnti-drug resistant effectsRefs
BerberineCoptis chinensis FranchRecent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceOvarian cancerSynergizing with DDP to induce necroptosis by activating the RIP3/MLKL pathway.[69]
BufalinBufo bufo gargarizans CantorRecent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceTriple-negative breast cancerInducing necroptosis in adriamycin-resistant triple-negative breast cancer through mediating the RIP1/ROS pathway.[65]
Gambogenic AcidGarcinia hanburyi Hook. f.Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceLung cancerSynergizing with 5-FU to induce necroptosis by increasing the expression of RIP1.[67]
Ganoderic acid TGanoderma lucidum (Leyss.ex Fr.) Karst.Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceCervical cancerIncreasing the radiosensitivity of cervical cancer by inducing necroptosis via ROS generation and increased expression of RIP and MLKL.[71]
OridoninIsodon rubescens (Hemsley) H. HaraRecent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceRenal carcinomaInducing necroptosis in renal carcinoma to enhance the antitumor effects of 5-FU via ROS generation, GSH depletion, and activation of p38, ERK, and JNK.[70]

MAPK signaling pathways also play a role in necroptosis induction. For instance, oridonin, a diterpenoid derived from Isodon rubescens (Hemsley) H. Hara, has been shown to enhance the cytotoxicity of 5-FU in renal cancer cells by inducing necroptosis. This process is associated with the activation of c-Jun N-terminal kinase (JNK), p38, and extracellular signal-regulated kinase (ERK)[70].

Moreover, certain studies have demonstrated that the inhibition of caspases may cause a switch from apoptosis to necroptosis in specific cancer types. Ganoderic acid T (GAT) is a triterpene of Garcinia hanburyi Hook. f., inducing both necroptosis and apoptosis in cervical cancer cells. Interestingly, the percentage ratio of necroptosis is increased following the increase of GAT concentration, as GAT can reduce the matrix metalloproteinase (MMP) and ATP levels and caspase-8 expression under radiation conditions[71].

Notably, glycolysis suppression has also emerged as an effective mechanism for necroptosis induction by natural compounds. Docetaxel is indeed a valuable chemotherapeutic agent utilized in the treatment of prostate cancer, primarily by inducing cell death[72]. However, similar to DDP, the effectiveness of docetaxel-induced apoptosis can also be hindered through various pathways, including the p38/p53/p21 pathway, USP33-DUSP1-JNK pathway, and PI3K/Akt/NF-κB pathway[73-75]. Therefore, several studies are currently focused on identifying alternative cell death pathways, such as necroptosis, that can be induced in docetaxel-resistant cancer cells as well. Shikonin has been extensively studied as a natural necroptosis inducer in various cancer types, and it has been demonstrated to overcome drug resistance to docetaxel in prostate cancer and DDP in bladder cancer[76,77]. Furthermore, shikonin can induce glycolysis suppression in glioma cells, which is closely associated with the accumulation of intracellular H2O2 triggered by the activation of RIP1 and RIP3[78].

PYROPTOSIS

Overview of pyroptosis

Pyroptosis, a pro-inflammatory programmed cell death, was originally termed by Cookson et al. in 2001[79]. The term derives from the Greek roots “pyro”, which relates to fire or fever, and “ptosis”, denoting a falling, reflecting its nature. Pyroptosis shares certain characteristics with apoptosis, such as DNA fragmentation, nuclear condensation, and caspase dependence. However, cells undergoing pyroptosis differ in that they retain intact nuclei and exhibit pore formation in the plasma membrane[80]. An increasing understanding of pyroptosis has revealed that this type of cell death can be divided into classical and non-classical pathways.

The classical pyroptotic pathway is mediated by inflammasome assembly, which consists of pattern recognition receptors (PRRs), apoptosis-related speck-like protein (ASC), and pro-caspase-1[81]. PRRs, functioning as inflammasome sensors, recognize pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs)[82]. Subsequently, they recruit the bridging protein ASC, which contains a pyrin domain (PYD) and a caspase activation and recruitment domain (CARD), through specific PYD-PYD interactions. After being recruited, ASC can interact with and activate pro-caspase-1 via CARD-CARD interactions[83]. Some PRRs containing CARD can also directly bind to pro-caspase-1, forming inflammasomes without the participation of ASC[84]. Activated caspase-1 facilitates the maturation of inflammatory cytokines interleukin (IL)-1β and IL-18, as well as cleaves the pore-forming protein gasdermin D (GSDMD) to produce its N-terminal fragment (N-GSDMD)[85]. N-GSDMDs then translocate to the plasma membrane and form pores, promoting the release of mature IL-1β and IL-18. As the number of N-GSDMD pores increases, cells swell and rupture, resulting in pyroptotic cell death[86].

In non-classical pathways, caspase-3/8 can also trigger pyroptosis by activating GSDMD or GSDME[87,88]. Additionally, caspase-8 can induce GSDMC-dependent pyroptosis as well[89]. The cleavage of GSDMD can also be mediated by caspase-4/5/11, which recognizes intracellular LPS to activate the non-canonical inflammasome[90]. Notably, GSDMB can not only be cleaved by caspase-1 to directly initiate pyroptosis, but also enhance caspase-4 activity to promote this cell death mechanism[91]. Recently, the cleavage of GSDMA has been found to be catalyzed by streptococcal pyrogenic exotoxin B (SpeB), a cysteine protease secreted by group A Streptococcus. This finding demonstrates that GSDMA can also play a role in pyroptosis by releasing the cleaved N-terminal fragments, which can bind to and disrupt specific acidic lipid-containing membranes[92] [Figure 3].

Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistance

Figure 3. In the classical pathway, PRRs recognize PAMPs and DAMPs, initiating the recruitment of ASC and pro-caspase-1. The activation of caspase-1 results in cleavage of GSDMD, forming GSDMD pores that ultimately trigger pyroptosis. Furthermore, caspase-1 also results in the maturation of IL-1β and IL-18, which are eventually released from the GSDMD pores. Alternatively, in the non-classical pathway, pyroptosis can be initiated by other members of the GSDM family. CARD: Caspase activation and recruitment domain; DAMPs: damage-associated molecular patterns; GSDMD: gasdermin D; IL: interleukin; PAMPs: pathogen-associated molecular patterns; PRR: pattern recognition receptor; PYD: pyrin domain.

Induction of pyroptosis for anti-drug resistance in cancer

In cancer treatment, the induction of pyroptosis is increasingly recognized as a promising strategy for overcoming drug resistance. NOD-like receptor family pyrin domain-containing 3 (NLRP3), a crucial inflammasome sensor in the NLR family, is recognized as a downstream target of multiple microRNAs (miRNAs) associated with cancer drug resistance. For example, by downregulating the expression of miR-556-5p in non-small cell lung cancer, NLRP3 inflammasome-mediated pyroptosis can be triggered, thereby enhancing DDP sensitivity[93].

The pore-forming proteins GSDMD and GSDME, extensively studied in the GSDM family, are potential targets for combating drug resistance and contributing to the treatment and prognosis of various cancers. Some chemotherapeutic agents have been demonstrated to exert antitumor activity when used alone or in combination to induce GSDMD-dependent pyroptosis, enhancing the efficacy of chemotherapy. For instance, the co-administration of paclitaxel and ruthenium complexes can induce cell death in paclitaxel-resistant cervical cancer cells by mediating Caspase-1/GSDMD-dependent pyroptosis[94]. GSDME-mediated pyroptosis has been observed to improve the sensitivity of various drugs across different cancer types, including increasing DDP sensitivity in esophageal squamous cell carcinoma cells and oxaliplatin sensitivity in colon cancer cells[95]. Additionally, it can alleviate the side effects of DDP in patients with oral squamous cell carcinoma.

In recent years, programmed death-ligand 1 (PD-L1), known as an immune checkpoint, has emerged as a research hotspot in tumor immunotherapy. However, PD-L1 has an additional role as a non-immune checkpoint by regulating the non-classical pyroptosis pathway mediated by GSDMC/caspase-8[96]. In this way, a variety of antibiotics can induce pyroptosis in cancer cells, indicating that it could be a novel strategy to combat antibiotic resistance in chemotherapy.

Natural compounds inducing pyroptosis for cancer treatment

Nowadays, the caspase-1/GSDMD and caspase-3/GSDME pathways have attracted significant attention in pyroptosis induction, and many natural compounds have been found to activate these pathways in cancers [Table 3]. Wedelolactone, an ingredient of Eclipta prostrata (L.) L., can simultaneously activate these two pathways by strongly increasing the activation of caspase-1, caspase-3, GSDME and GSDMD in retinoblastoma cells[97]. Ophiopogonin B, derived from Dioscorea bulbifera L., can induce caspase-1/GSDMD-dependent pyroptosis in lung cancer cells, especially exhibiting a more significant suppression of growth in DDP-resistant cancer cells[98]. As a pentacyclic triterpene compound of lupine, betulinic acid can induce caspase-1-dependent pyroptosis, thereby enhancing chemosensitivity to DDP in esophageal cancer cells[99].

Table 3

Natural compounds for anticancer drug resistance by inducing pyroptosis

CompoundsOriginStructureCancerAnti-drug resistant effectsRefs
Betulinic acidBetula platyphylla Suk.Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceEsophageal cancerEnhancing the antitumor effects of DDP by inducing pyroptosis via increasing the levels of ASC and caspase-1 and decreasing the levels of Ki67, PCNA, SOX2, and OCT4.[99]
Diosbulbin BDioscorea bulbifera L.Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceGastric cancerIncreasing DDP-sensitivity in gastric cancer by inducing pyroptosis via regulating the PD-L1/NLRP3 pathway.[106]
Ophiopogonin BOphiopogon japonicus (L. f.) Ker-Gawl.Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceLung cancerIncreasing DDP-sensitivity in gastric cancer by inducing pyroptosis via regulating the caspase-1/GSDMD pathway.[98]

By activating the caspase-3-dependent pathway, natural compounds such as curcumin, dihydroartemisinin, and germacrone can induce pyroptosis in different cancers[100-102]. Some studies have reported that the activation of caspase-9 is also involved in caspase-3-mediated pyroptosis. For example, alantolactone, a terpenoid of Inula helenium L., can promote the cleavage of caspase-9 and caspase-3 to induce GSDME-mediated pyroptosis in anaplastic thyroid cancer[103]. Some chemotherapeutic agents, such as DDP, 5-FU, and carboplatin, have been demonstrated to combat cancer drug resistance by inducing GSDME-dependent pyroptosis, while whether natural compounds can also suppress drug resistance through this pathway needs further investigation[95,104,105].

In addition, by downregulating PD-L1 to activate NLRP3-mediated pyroptosis, Diosbulbin B extracted from Dioscorea bulbifera L. can sensitize DDP-resistant gastric cancer cells to DDP[106]. At present, there are few reports on whether natural compounds induce pyroptosis via the regulation of other GSDM family proteins. This is partly because of their unclear functions in initiating pyroptotic cell death. Further elucidation of how the other members of the GSDM family contribute to pyroptosis induction in cancers may provide new insights for the search for natural compounds with anticancer and anti-drug resistance activities.

PARAPTOSIS

Overview of paraptosis

Paraptosis was first introduced as a form of programmed cell death by Sperandio et al. in 2000[107]. It derives from “para”, meaning “next to” or “related to”, and “apoptosis”, suggesting that it is distinct from apoptosis. The main morphological features of paraptosis include cytoplasmic vacuolization, swelling of the endoplasmic reticulum and/or mitochondria, and the absence of nuclear fragmentation or apoptotic body formation. In this paradigm, caspases are not activated, and thus, cells undergoing paraptosis are resistant to caspase inhibitors[108].

The MAPK pathways play a critical role in paraptosis, and research focusing on the ERK, JNK, and p38 pathways is particularly extensive. Notably, the protein AIP1/Alix was described as the first specific inhibitor of paraptosis, capable of restraining the insulin-like growth factor-I receptor (IGFIR) induced paraptotic process mediated by the MAPK/ERK and JNK pathways[109]. Furthermore, TrxR1 inhibition and GSH depletion have been observed to potentially activate the MAPK pathways by triggering the accumulation of cellular ROS[110]. Paraptosis induction is also related to the homeostasis of intracellular Ca2+, which is mainly regulated by the endoplasmic reticulum and mitochondria. Intracellular Ca2+ can be released from the endoplasmic reticulum into mitochondria when paraptosis is initiated, resulting in the endoplasmic reticulum and mitochondrial dilation and ultimately leading to cell death. The voltage- and Ca2+-activated K+ (BKCa) channels are widely expressed in body and have the ability to link changes in intracellular calcium to outward hyperpolarizing potassium currents. The activation of these channels will disrupt the osmotic balance, initiating cell swelling and vacuolization[111]. Additionally, proteasome inhibition may also promote paraptosis by inducing endoplasmic reticulum (ER) stress[112] [Figure 4].

Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistance

Figure 4. The mechanism of Paraptosis. Paraptosis was initially discovered to be induced by IGFIR and mediated through the MAPK pathways. Furthermore, it is intricately linked to various factors, including TrxR1 inhibition, GSH depletion, intracellular Ca2+ homeostasis, activation of BKCa channels, and proteasome inhibition. BKCa: Ca2+-activated K+; ER: endoplasmic reticulum; GSH: glutathione; IGFIR: insulin-like growth factor-I receptor; MAPK: MAP kinase; ROS: reactive oxygen species.

Paraptosis potential applications in cancer drug resistance

Paraptosis induction is a potential strategy for developing non-genetically modified tumor vaccines. In rat T9 glioma cells, the activation of BKCa channels promotes the overexpression of heat shock proteins and the translocation of HMGB1 from the nuclear region to the periphery, stimulating immune responses and initiating paraptosis. Rats injected with paraptotic T9 glioma cells, which are killed by prolonged BKCa channel activation, can develop specific immunity to T9 cells. This suggests the potential of using these treated cells as a functionally killed vaccine[113].

Due to its unique molecular mechanism, paraptosis induction also contributes to enhancing the activity of proteasome inhibitors in cancer cells. Although many proteasome inhibitors have shown antitumor activities, their clinical efficacy is unsatisfactory as their effectiveness can be compromised by both primary and secondary resistance mechanisms. Therefore, combination therapy can be seen as an effective strategy to address proteasome inhibitor resistance. For instance, by triggering paraptotic cell death, bortezomib (Btz), a 20S core particle inhibitor of the proteasome, can be combined with loperamide, an antidiarrheal agent, to enhance Btz sensitivity and reduce its side effects, effectively combating the colon cancer[114].

Natural compounds inducing paraptosis for cancer treatment

Currently, a number of natural compounds have shown potential in cancer treatment by inducing paraptosis [Table 4], with many of them modulating the MAPK signaling pathways to trigger ER stress. One such example is Paris Saponin II, derived from Paris polyphylla Smith, which effectively induces paraptosis by activating the JNK pathway and augmenting ER stress[115]. Moreover, the activation of this paraptosis-associated pathway enhances the sensitivity of DDP in lung cancer. Jolkinolide B, an active abietane ent-diterpenoid, is also a noteworthy compound that induces paraptosis in both sensitive and DDP-resistant bladder cancer cells by activating the ERK pathway and enhancing ER stress[110]. Additionally, Chalcomoracin, isolated from Morus alba L., has been discovered to enhance the sensitivity of non-small cell lung cancer to radiation by augmenting ER stress[116]. Elaiophylin is a natural antibiotic derived from Streptomyces melanosporus that can also induce paraptosis through the hyperactivation of the MAPK pathway. This compound demonstrates notable efficacy in eliminating ovarian cancer cells that are resistant to multiple drugs, including platinum, taxane, and poly (ADP-ribose) polymerase inhibitor (PARPi)[117].

Table 4

Natural compounds for anticancer drug resistance by inducing paraptosis

CompoundsOriginStructureCancerAnti-drug resistant effectsRefs
ChalcomoracinMorus alba L.Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceNon-small cell lung cancerIncreasing the radiosensitivity of non-small cell lung cancer by inducing ER stress-mediated paraptosis via activation of the MAPK pathway.[116]
ElaiophylinStreptomyces hygroscopicusRecent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceOvarian cancerInducing paraptosis to overcome platinum, taxane, and PARPi resistance in ovarian cancer by regulating the SHP2/SOS1/MAPK pathway.[117]
Jolkinolide BEuphorbia fischeriana SteudRecent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceBladder cancerInducing ROS-mediated paraptosis to suppress the growth of DDP-resistant bladder cancer by targeting thioredoxin and glutathione systems.[110,131]
Paris saponin IIParis polyphylla SmithRecent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistanceNon-small cell lung cancerEnhancing the antitumor effects of DDP by regulating the JNK pathway.[115]

Some natural compounds such as curcumin, morusin, and ophiobolin A can also induce paraptosis by affecting ion homeostasis. Curcumin can induce paraptosis in epithelial ovarian cancer mainly through mitochondrial Ca2+ overload, which subsequently contributes to mitochondrial swelling and dysfunction[118]. Similarly, morusin, a prenylflavonoid, can also induce paraptosis in breast cancer by triggering mitochondrial Ca2+ overload[119]. In glioblastoma cells, paraptosis induced by ophiobolin A, a sesterterpenoid phytotoxin from the genus Bipolaris, is linked to K+ homeostasis imbalance, primarily caused by the blockage of BKCa channels[120].

Furthermore, natural compounds can induce paraptosis by disrupting sulfhydryl homeostasis and suppressing proteasome functions, both of which can be significantly inhibited by thiol antioxidants. In this way, plumbagin, extracted from Plumbago zeylanica L., can induce paraptotic cell death in different cancer types[121]. Paraptosis can also be triggered in a p53-dependent manner. For instance, Ginsenoside Rh2, which is a bioactive product in Panax ginseng C. A. Meyer, can induce paraptosis in colorectal cancer via activating the p53 pathway as well as the NF-κB survival pathway[122].

CONCLUSION

Numerous natural compounds possess the ability to elicit anticancer and anti-chemoresistance effects by triggering non-apoptotic cell death mechanisms such as ferroptosis, necroptosis, pyroptosis, and paraptosis. It is noteworthy that while these forms of cell death have unique regulators, some common pathways can also govern them. This implies that multiple cell death pathways may occur concurrently and be subject to simultaneous regulation. In 2019, Malireddi et al. introduced PANoptosis, an innovative form of cell death that amalgamates essential features of pyroptosis, apoptosis, and necroptosis yet defies a straightforward classification under any one of these categories[123]. This discovery underscores the co-regulation and interplay among these cell death pathways, suggesting that drugs may possess multifaceted regulatory effects by modulating master factors within these pathways. Beyond the four aforementioned types of cell death, some other novel cell death mechanisms have surfaced in recent years, including parthanatos, disulfidptosis, and cuproptosis[124-126]. These revelations open up new avenues for identifying targets in cancer treatment and provide additional strategies for combating cancer drug resistance. As such, ongoing exploration is essential to ascertain whether natural compounds can elicit anticancer effects through these emerging forms of cell death.

DECLARATIONS

Authors’ contributions

Conceptualization: Chen GQ, Chen S, Chen JW

Data curation and original draft preparation: Chen JW, Chen GQ, Chen S

Figures and Tables: Chen JW, Chen GQ, Chen S

Review and editing: Chen GQ, Chen S, Chen JW

All authors contributed to the article and approved the submitted version.

Availability of data and materials

Not applicable.

Financial support and sponsorship

This research was funded by Shenzhen Science and Technology Innovation Commission (JCYJ20220531090802006), Innovation and Technology Fund-Mainland-Hong Kong Joint Funding Scheme (MHP/010/20), Research Centre for Chinese Medicine Innovation of The Hong Kong Polytechnic University (E-ABCT-BBBB-1) and The Hong Kong Polytechnic University Start-up Fund (P0038596).

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2023.

REFERENCES

1. Ferlay J, Colombet M, Soerjomataram I, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer 2019;144:1941-53.

2. Strasser A, Vaux DL. Cell death in the origin and treatment of cancer. Mol Cell 2020;78:1045-54.

3. Labi V, Erlacher M. How cell death shapes cancer. Cell Death Dis 2015;6:e1675.

4. Montopoli M, Ragazzi E, Froldi G, Caparrotta L. Cell-cycle inhibition and apoptosis induced by curcumin and cisplatin or oxaliplatin in human ovarian carcinoma cells. Cell Prolif 2009;42:195-206.

5. Liu SY, Song SX, Lin L, Liu X. Molecular mechanism of cell apoptosis by paclitaxel and pirarubicin in a human osteosarcoma cell line. Chemotherapy 2010;56:101-7.

6. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57-70.

7. Rajabi M, Mousa SA. The role of angiogenesis in cancer treatment. Biomedicines 2017;5:34.

8. Tossetta G. Metformin improves ovarian cancer sensitivity to paclitaxel and platinum-based drugs: a review of in vitro findings. Int J Mol Sci 2022;23:12893.

9. Tossetta G, Fantone S, Montanari E, Marzioni D, Goteri G. Role of NRF2 in ovarian cancer. Antioxidants 2022;11:663.

10. Ghareghomi S, Habibi-Rezaei M, Arese M, Saso L, Moosavi-Movahedi AA. Nrf2 modulation in breast cancer. Biomedicines 2022;10:2668.

11. Katz L, Baltz RH. Natural product discovery: past, present, and future. J Ind Microbiol Biotechnol 2016;43:155-76.

12. Bauman KD, Butler KS, Moore BS, Chekan JR. Genome mining methods to discover bioactive natural products. Nat Prod Rep 2021;38:2100-29.

13. Wu S, Pang Y, He Y, et al. A comprehensive review of natural products against atopic dermatitis: flavonoids, alkaloids, terpenes, glycosides and other compounds. Biomed Pharmacother 2021;140:111741.

14. Oberlies NH, Kroll DJ. Camptothecin and taxol: historic achievements in natural products research. J Nat Prod 2004;67:129-35.

15. Škubník J, Pavlíčková VS, Ruml T, Rimpelová S. Vincristine in combination therapy of cancer: emerging trends in clinics. Biology 2021;10:849.

16. Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 2012;149:1060-72.

17. Liang C, Zhang X, Yang M, Dong X. Recent progress in ferroptosis inducers for cancer therapy. Adv Mater 2019;31:e1904197.

18. Yang WS, Stockwell BR. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem Biol 2008;15:234-45.

19. Wang L, Liu Y, Du T, et al. ATF3 promotes erastin-induced ferroptosis by suppressing system Xc. Cell Death Differ 2020;27:662-75.

20. Sui X, Zhang R, Liu S, et al. RSL3 drives ferroptosis through GPX4 inactivation and ROS production in colorectal cancer. Front Pharmacol 2018;9:1371.

21. Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 2014;156:317-31.

22. Lin Z, Liu J, Kang R, Yang M, Tang D. Lipid metabolism in ferroptosis. Adv Biol 2021;5:e2100396.

23. Ke K, Li L, Lu C, et al. The crosstalk effect between ferrous and other ions metabolism in ferroptosis for therapy of cancer. Front Oncol 2022;12:916082.

24. Mao C, Liu X, Zhang Y, et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 2021;593:586-90.

25. Koppula P, Lei G, Zhang Y, et al. A targetable CoQ-FSP1 axis drives ferroptosis- and radiation-resistance in KEAP1 inactive lung cancers. Nat Commun 2022;13:2206.

26. Yi J, Zhu J, Wu J, Thompson CB, Jiang X. Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc Natl Acad Sci U S A 2020;117:31189-97.

27. Xiang J, Jiang M, Du X. The role of Hippo pathway in ferroptosis. Front Oncol 2022;12:1107505.

28. Ye F, Chai W, Xie M, et al. HMGB1 regulates erastin-induced ferroptosis via Ras-JNK/P38 signaling in HL-60/NrasQ61L cells. Am J Cancer Res 2019;9:730-9.

29. Mirzaei S, Mohammadi AT, Gholami MH, et al. Nrf2 signaling pathway in cisplatin chemotherapy: potential involvement in organ protection and chemoresistance. Pharmacol Res 2021;167:105575.

30. Li Y, Yan H, Xu X, Liu H, Wu C, Zhao L. Erastin/sorafenib induces cisplatin-resistant non-small cell lung cancer cell ferroptosis through inhibition of the Nrf2/xCT pathway. Oncol Lett 2020;19:323-33.

31. Liu Y, Gu W. p53 in ferroptosis regulation: the new weapon for the old guardian. Cell Death Differ 2022;29:895-910.

32. Zhou X, Zou L, Chen W, et al. Flubendazole, FDA-approved anthelmintic, elicits valid antitumor effects by targeting P53 and promoting ferroptosis in castration-resistant prostate cancer. Pharmacol Res 2021;164:105305.

33. Green YS, Ferreira Dos Santos MC, Fuja DG, et al. ISCA2 inhibition decreases HIF and induces ferroptosis in clear cell renal carcinoma. Oncogene 2022;41:4709-23.

34. Su X, Xie Y, Zhang J, et al. HIF-α activation by the prolyl hydroxylase inhibitor roxadustat suppresses chemoresistant glioblastoma growth by inducing ferroptosis. Cell Death Dis 2022;13:861.

35. Zhu YJ, Zheng B, Wang HY, Chen L. New knowledge of the mechanisms of sorafenib resistance in liver cancer. Acta Pharmacol Sin 2017;38:614-22.

36. Huang W, Chen K, Lu Y, et al. ABCC5 facilitates the acquired resistance of sorafenib through the inhibition of SLC7A11-induced ferroptosis in hepatocellular carcinoma. Neoplasia 2021;23:1227-39.

37. Wang Q, Bin C, Xue Q, et al. GSTZ1 sensitizes hepatocellular carcinoma cells to sorafenib-induced ferroptosis via inhibition of NRF2/GPX4 axis. Cell Death Dis 2021;12:426.

38. Cui Z, Wang H, Li S, et al. Dihydroartemisinin enhances the inhibitory effect of sorafenib on HepG2 cells by inducing ferroptosis and inhibiting energy metabolism. J Pharmacol Sci 2022;148:73-85.

39. Li H, Yu Y, Liu Y, et al. Ursolic acid enhances the antitumor effects of sorafenib associated with Mcl-1-related apoptosis and SLC7A11-dependent ferroptosis in human cancer. Pharmacol Res 2022;182:106306.

40. Song M, Cui M, Liu K. Therapeutic strategies to overcome cisplatin resistance in ovarian cancer. Eur J Med Chem 2022;232:114205.

41. Li M, Zhang YY, Shang J, Xu YD. LncRNA SNHG5 promotes cisplatin resistance in gastric cancer via inhibiting cell apoptosis. Eur Rev Med Pharmacol Sci 2019;23:4185-91.

42. Ni M, Zhou J, Zhu Z, et al. Shikonin and cisplatin synergistically overcome cisplatin resistance of ovarian cancer by inducing ferroptosis via upregulation of HMOX1 to promote Fe2+ accumulation. Phytomedicine 2023;112:154701.

43. Yang Y, Sun S, Xu W, et al. Piperlongumine inhibits thioredoxin reductase 1 by targeting selenocysteine residues and sensitizes cancer cells to erastin. Antioxidants 2022;11:710.

44. Zhang Y, Tan Y, Liu S, et al. Implications of Withaferin A for the metastatic potential and drug resistance in hepatocellular carcinoma cells via Nrf2-mediated EMT and ferroptosis. Toxicol Mech Methods 2023;33:47-55.

45. Yang C, Lu T, Liu M, et al. Tiliroside targets TBK1 to induce ferroptosis and sensitize hepatocellular carcinoma to sorafenib. Phytomedicine 2023;111:154668.

46. Lou JS, Zhao LP, Huang ZH, et al. Ginkgetin derived from Ginkgo biloba leaves enhances the therapeutic effect of cisplatin via ferroptosis-mediated disruption of the Nrf2/HO-1 axis in EGFR wild-type non-small-cell lung cancer. Phytomedicine 2021;80:153370.

47. Santana-Codina N, Mancias JD. The role of NCOA4-mediated ferritinophagy in health and disease. Pharmaceuticals 2018;11:114.

48. Zhou L, Yang C, Zhong W, et al. Chrysin induces autophagy-dependent ferroptosis to increase chemosensitivity to gemcitabine by targeting CBR1 in pancreatic cancer cells. Biochem Pharmacol 2021;193:114813.

49. He C, Wang C, Liu H, Shan B. Kayadiol exerted anticancer effects through p53-mediated ferroptosis in NKTCL cells. BMC Cancer 2022;22:724.

50. Markowitsch SD, Schupp P, Lauckner J, et al. Artesunate inhibits growth of sunitinib-resistant renal cell carcinoma cells through cell cycle arrest and induction of ferroptosis. Cancers 2020;12:3150.

51. Degterev A, Huang Z, Boyce M, et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol 2005;1:112-9.

52. Liu C, Zhang K, Shen H, Yao X, Sun Q, Chen G. Necroptosis: a novel manner of cell death, associated with stroke (Review). Int J Mol Med 2018;41:624-30.

53. Gong Y, Fan Z, Luo G, et al. The role of necroptosis in cancer biology and therapy. Mol Cancer 2019;18:100.

54. Liu S, Pan Y, Li T, et al. The role of regulated programmed cell death in osteoarthritis: from pathogenesis to therapy. Int J Mol Sci 2023;24:5364.

55. Chen J, Kos R, Garssen J, Redegeld F. Molecular insights into the mechanism of necroptosis: the necrosome as a potential therapeutic target. Cells 2019;8:1486.

56. de Almagro MC, Vucic D. Necroptosis: pathway diversity and characteristics. Semin Cell Dev Biol 2015;39:56-62.

57. Su Z, Yang Z, Xu Y, Chen Y, Yu Q. Apoptosis, autophagy, necroptosis, and cancer metastasis. Mol Cancer 2015;14:48.

58. Najafov A, Zervantonakis IK, Mookhtiar AK, et al. BRAF and AXL oncogenes drive RIPK3 expression loss in cancer. PLoS Biol 2018;16:e2005756.

59. Ermine KA, Chen D, Wang P, Yu J, Zhang L. Abstract A015: targeting necroptosis and RIP3 in colorectal cancer to overcome chemotherapeutic resistance. Cancer Res 2022;82:A015.

60. Wang Q, Wang P, Zhang L, et al. Epigenetic regulation of RIP3 suppresses necroptosis and increases resistance to chemotherapy in nonsmall cell lung cancer. Transl Oncol 2020;13:372-82.

61. Sun W, Yu W, Shen L, Huang T. MLKL is a potential prognostic marker in gastric cancer. Oncol Lett 2019;18:3830-6.

62. Liu X, Zhang Y, Gao H, et al. Induction of an MLKL mediated non-canonical necroptosis through reactive oxygen species by tanshinol A in lung cancer cells. Biochem Pharmacol 2020;171:113684.

63. Huang CY, Kuo WT, Huang YC, Lee TC, Yu LC. Resistance to hypoxia-induced necroptosis is conferred by glycolytic pyruvate scavenging of mitochondrial superoxide in colorectal cancer cells. Cell Death Dis 2013;4:e622.

64. Cui J, Yan M, Liu X, et al. Inorganic selenium induces nonapoptotic programmed cell death in PC-3 prostate cancer cells associated with inhibition of glycolysis. J Agric Food Chem 2019;67:10637-45.

65. Liu XD, Song CY, Kong CC, Tian X. Bufalin induces programmed necroptosis in triple-negative breast cancer drug-resistant cell lines through RIP1/ROS-mediated pathway. Chin J Integr Med 2022;28:900-8.

66. Sethy C, Kundu CN. 5-Fluorouracil (5-FU) resistance and the new strategy to enhance the sensitivity against cancer: implication of DNA repair inhibition. Biomed Pharmacother 2021;137:111285.

67. Su J, Cheng H, Zhang D, et al. Synergistic effects of 5-fluorouracil and gambogenic acid on A549 cells: activation of cell death caused by apoptotic and necroptotic mechanisms via the ROS-mitochondria pathway. Biol Pharm Bull 2014;37:1259-68.

68. Pan X, Chen G, Hu W. Piperlongumine increases the sensitivity of bladder cancer to cisplatin by mitochondrial ROS. J Clin Lab Anal 2022;36:e24452.

69. Liu L, Fan J, Ai G, et al. Berberine in combination with cisplatin induces necroptosis and apoptosis in ovarian cancer cells. Biol Res 2019;52:37.

70. Zheng W, Zhou CY, Zhu XQ, et al. Oridonin enhances the cytotoxicity of 5-FU in renal carcinoma cells by inducting necroptotic death. Biomed Pharmacother 2018;106:175-82.

71. Shao CS, Feng N, Zhou S, et al. Ganoderic acid T improves the radiosensitivity of HeLa cells via converting apoptosis to necroptosis. Toxicol Res 2021;10:531-41.

72. Ganju A, Yallapu MM, Khan S, Behrman SW, Chauhan SC, Jaggi M. Nanoways to overcome docetaxel resistance in prostate cancer. Drug Resist Updat 2014;17:13-23.

73. Guo F, Zhang C, Wang F, et al. Deubiquitinating enzyme USP33 restrains docetaxel-induced apoptosis via stabilising the phosphatase DUSP1 in prostate cancer. Cell Death Differ 2020;27:1938-51.

74. Gan L, Wang J, Xu H, Yang X. Resistance to docetaxel-induced apoptosis in prostate cancer cells by p38/p53/p21 signaling. Prostate 2011;71:1158-66.

75. Song L, Xiong H, Li J, et al. Sphingosine kinase-1 enhances resistance to apoptosis through activation of PI3K/Akt/NF-κB pathway in human non-small cell lung cancer. Clin Cancer Res 2011;17:1839-49.

76. Markowitsch SD, Juetter KM, Schupp P, et al. Shikonin reduces growth of docetaxel-resistant prostate cancer cells mainly through necroptosis. Cancers 2021;13:882.

77. Wang Y, Hao F, Nan Y, et al. PKM2 inhibitor shikonin overcomes the cisplatin resistance in bladder cancer by inducing necroptosis. Int J Biol Sci 2018;14:1883-91.

78. Lu B, Wang Z, Ding Y, et al. RIP1 and RIP3 contribute to shikonin-induced glycolysis suppression in glioma cells via increase of intracellular hydrogen peroxide. Cancer Lett 2018;425:31-42.

79. Cookson BT, Brennan MA. Pro-inflammatory programmed cell death. Trends Microbiol 2001;9:113-4.

80. Jorgensen I, Miao EA. Pyroptotic cell death defends against intracellular pathogens. Immunol Rev 2015;265:130-42.

81. Lin J, Cheng A, Cheng K, et al. New insights into the mechanisms of pyroptosis and implications for diabetic kidney disease. Int J Mol Sci 2020;21:7057.

82. Gong W, Shi Y, Ren J. Research progresses of molecular mechanism of pyroptosis and its related diseases. Immunobiology 2020;225:151884.

83. Isazadeh M, Amandadi M, Haghdoust F, Lotfollazadeh S, Orzáez M, Hosseinkhani S. Split-luciferase complementary assay of NLRP3 PYD-PYD interaction indicates inflammasome formation during inflammation. Anal Biochem 2022;638:114510.

84. Zhang KJ, Wu Q, Jiang SM, et al. Pyroptosis: a new frontier in kidney diseases. Oxid Med Cell Longev 2021;2021:6686617.

85. Yu P, Zhang X, Liu N, Tang L, Peng C, Chen X. Pyroptosis: mechanisms and diseases. Signal Transduct Target Ther 2021;6:128.

86. Ding J, Wang K, Liu W, et al. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature 2016;535:111-6.

87. Sarhan J, Liu BC, Muendlein HI, et al. Caspase-8 induces cleavage of gasdermin D to elicit pyroptosis during Yersinia infection. Proc Natl Acad Sci U S A 2018;115:E10888-97.

88. Kovacs SB, Miao EA. Gasdermins: effectors of pyroptosis. Trends Cell Biol 2017;27:673-84.

89. Zhang JY, Zhou B, Sun RY, et al. The metabolite α-KG induces GSDMC-dependent pyroptosis through death receptor 6-activated caspase-8. Cell Res 2021;31:980-97.

90. Matikainen S, Nyman TA, Cypryk W. Function and regulation of noncanonical caspase-4/5/11 inflammasome. J Immunol 2020;204:3063-9.

91. Li L, Li Y, Bai Y. Role of GSDMB in pyroptosis and cancer. Cancer Manag Res 2020;12:3033-43.

92. Deng W, Bai Y, Deng F, et al. Streptococcal pyrogenic exotoxin B cleaves GSDMA and triggers pyroptosis. Nature 2022;602:496-502.

93. Shi F, Zhang L, Liu X, Wang Y. Knock-down of microRNA miR-556-5p increases cisplatin-sensitivity in non-small cell lung cancer (NSCLC) via activating NLR family pyrin domain containing 3 (NLRP3)-mediated pyroptotic cell death. Bioengineered 2021;12:6332-42.

94. Chen D, Guo S, Tang X, et al. Combination of ruthenium (II) polypyridyl complex Δ-Ru1 and Taxol enhances the anti-cancer effect on Taxol-resistant cancer cells through Caspase-1/GSDMD-mediated pyroptosis. J Inorg Biochem 2022;230:111749.

95. Li RY, Zheng ZY, Li ZM, et al. Cisplatin-induced pyroptosis is mediated via the CAPN1/CAPN2-BAK/BAX-caspase-9-caspase-3-GSDME axis in esophageal cancer. Chem Biol Interact 2022;361:109967.

96. Hou J, Zhao R, Xia W, et al. PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis. Nat Cell Biol 2020;22:1264-75.

97. Jiang H, Niu C, Guo Y, Liu Z, Jiang Y. Wedelolactone induces apoptosis and pyroptosis in retinoblastoma through promoting ROS generation. Int Immunopharmacol 2022;111:108855.

98. Cheng Z, Li Z, Gu L, et al. Ophiopogonin B alleviates cisplatin resistance of lung cancer cells by inducing Caspase-1/GSDMD dependent pyroptosis. J Cancer 2022;13:715-27.

99. Chen J, Peng R, Niu Z, Zhou H, Kang C. Betulinic acid enhanced the chemical sensitivity of esophageal cancer cells to cisplatin by inducing cell pyroptosis and reducing cell stemness. Ann Palliat Med 2020;9:1912-20.

100. Liang WF, Gong YX, Li HF, et al. Curcumin activates ROS signaling to promote pyroptosis in hepatocellular carcinoma HepG2 cells. In Vivo 2021;35:249-57.

101. Li Y, Wang W, Li A, et al. Dihydroartemisinin induces pyroptosis by promoting the AIM2/caspase-3/DFNA5 axis in breast cancer cells. Chem Biol Interact 2021;340:109434.

102. Sun X, Zhong X, Ma W, et al. Germacrone induces caspase-3/GSDME activation and enhances ROS production, causing HepG2 pyroptosis. Exp Ther Med 2022;24:456.

103. Hu Y, Wen Q, Cai Y, et al. Alantolactone induces concurrent apoptosis and GSDME-dependent pyroptosis of anaplastic thyroid cancer through ROS mitochondria-dependent caspase pathway. Phytomedicine 2023;108:154528.

104. Li F, Xia Q, Ren L, et al. GSDME increases chemotherapeutic drug sensitivity by inducing pyroptosis in retinoblastoma cells. Oxid Med Cell Longev 2022;2022:2371807.

105. Wang Y, Yin B, Li D, Wang G, Han X, Sun X. GSDME mediates caspase-3-dependent pyroptosis in gastric cancer. Biochem Biophys Res Commun 2018;495:1418-25.

106. Li C, Qiu J, Xue Y. Low-dose Diosbulbin-B (DB) activates tumor-intrinsic PD-L1/NLRP3 signaling pathway mediated pyroptotic cell death to increase cisplatin-sensitivity in gastric cancer (GC). Cell Biosci 2021;11:38.

107. Sperandio S, de Belle I, Bredesen DE. An alternative, nonapoptotic form of programmed cell death. Proc Natl Acad Sci U S A 2000;97:14376-81.

108. Wang Y, Wen X, Zhang N, et al. Small-molecule compounds target paraptosis to improve cancer therapy. Biomed Pharmacother 2019;118:109203.

109. Wang WB, Feng LX, Yue QX, et al. Paraptosis accompanied by autophagy and apoptosis was induced by celastrol, a natural compound with influence on proteasome, ER stress and Hsp90. J Cell Physiol 2012;227:2196-206.

110. Sang J, Li W, Diao HJ, et al. Jolkinolide B targets thioredoxin and glutathione systems to induce ROS-mediated paraptosis and apoptosis in bladder cancer cells. Cancer Lett 2021;509:13-25.

111. Yoon MJ, Lee AR, Jeong SA, et al. Release of Ca2+ from the endoplasmic reticulum and its subsequent influx into mitochondria trigger celastrol-induced paraptosis in cancer cells. Oncotarget 2014;5:6816-31.

112. Lee HJ, Lee DM, Seo MJ, Kang HC, Kwon SK, Choi KS. PSMD14 targeting triggers paraptosis in breast cancer cells by inducing proteasome inhibition and Ca2+ imbalance. Int J Mol Sci 2022;23:2648.

113. Hoa N, Myers MP, Douglass TG, et al. Molecular mechanisms of paraptosis induction: implications for a non-genetically modified tumor vaccine. PLoS One 2009;4:e4631.

114. Kim IY, Shim MJ, Lee DM, et al. Loperamide overcomes the resistance of colon cancer cells to bortezomib by inducing CHOP-mediated paraptosis-like cell death. Biochem Pharmacol 2019;162:41-54.

115. Man S, Lv P, Cui J, et al. Paris saponin II-induced paraptosis-associated cell death increased the sensitivity of cisplatin. Toxicol Appl Pharmacol 2020;406:115206.

116. Zhang SR, Zhang XC, Liang JF, et al. Chalcomoracin inhibits cell proliferation and increases sensitivity to radiotherapy in human non-small cell lung cancer cells via inducing endoplasmic reticulum stress-mediated paraptosis. Acta Pharmacol Sin 2020;41:825-34.

117. Li GN, Zhao XJ, Wang Z, et al. Elaiophylin triggers paraptosis and preferentially kills ovarian cancer drug-resistant cells by inducing MAPK hyperactivation. Signal Transduct Target Ther 2022;7:317.

118. Yoon MJ, Kim EH, Kwon TK, Park SA, Choi KS. Simultaneous mitochondrial Ca2+ overload and proteasomal inhibition are responsible for the induction of paraptosis in malignant breast cancer cells. Cancer Lett 2012;324:197-209.

119. Xue J, Li R, Zhao X, et al. Morusin induces paraptosis-like cell death through mitochondrial calcium overload and dysfunction in epithelial ovarian cancer. Chem Biol Interact 2018;283:59-74.

120. Bury M, Girault A, Mégalizzi V, et al. Ophiobolin A induces paraptosis-like cell death in human glioblastoma cells by decreasing BKCa channel activity. Cell Death Dis 2013;4:e561.

121. Binoy A, Nedungadi D, Katiyar N, et al. Plumbagin induces paraptosis in cancer cells by disrupting the sulfhydryl homeostasis and proteasomal function. Chem Biol Interact 2019;310:108733.

122. Li B, Zhao J, Wang CZ, et al. Ginsenoside Rh2 induces apoptosis and paraptosis-like cell death in colorectal cancer cells through activation of p53. Cancer Lett 2011;301:185-92.

123. Malireddi RKS, Kesavardhana S, Kanneganti TD. ZBP1 and TAK1: master regulators of NLRP3 inflammasome/pyroptosis, apoptosis, and necroptosis (PAN-optosis). Front Cell Infect Microbiol 2019;9:406.

124. Wang Y, Kim NS, Haince JF, et al. Poly(ADP-ribose) (PAR) binding to apoptosis-inducing factor is critical for PAR polymerase-1-dependent cell death (parthanatos). Sci Signal 2011;4:ra20.

125. Zheng P, Zhou C, Ding Y, Duan S. Disulfidptosis: a new target for metabolic cancer therapy. J Exp Clin Cancer Res 2023;42:103.

126. Tang D, Chen X, Kroemer G. Cuproptosis: a copper-triggered modality of mitochondrial cell death. Cell Res 2022;32:417-8.

127. Han N, Yang ZY, Xie ZX, et al. Dihydroartemisinin elicits immunogenic death through ferroptosis-triggered ER stress and DNA damage for lung cancer immunotherapy. Phytomedicine 2023;112:154682.

128. Yuan B, Liao F, Shi ZZ, et al. Dihydroartemisinin inhibits the proliferation, colony formation and induces ferroptosis of lung cancer cells by inhibiting PRIM2/SLC7A11 axis. Onco Targets Ther 2020;13:10829-40.

129. Chen Y, Mi Y, Zhang X, et al. Dihydroartemisinin-induced unfolded protein response feedback attenuates ferroptosis via PERK/ATF4/HSPA5 pathway in glioma cells. J Exp Clin Cancer Res 2019;38:402.

130. Yamaguchi Y, Kasukabe T, Kumakura S. Piperlongumine rapidly induces the death of human pancreatic cancer cells mainly through the induction of ferroptosis. Int J Oncol 2018;52:1011-22.

131. Sang J, Gan L, Zou MF, et al. Jolkinolide B sensitizes bladder cancer to mTOR inhibitors via dual inhibition of Akt signaling and autophagy. Cancer Lett 2022;526:352-62.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Chen JW, Chen S, Chen GQ. Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistance. Cancer Drug Resist 2023;6:729-47. http://dx.doi.org/10.20517/cdr.2023.78

AMA Style

Chen JW, Chen S, Chen GQ. Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistance. Cancer Drug Resistance. 2023; 6(4): 729-47. http://dx.doi.org/10.20517/cdr.2023.78

Chicago/Turabian Style

Chen, Jia-Wen, Sibao Chen, Guo-Qing Chen. 2023. "Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistance" Cancer Drug Resistance. 6, no.4: 729-47. http://dx.doi.org/10.20517/cdr.2023.78

ACS Style

Chen, J.W.; Chen S.; Chen G.Q. Recent advances in natural compounds inducing non-apoptotic cell death for anticancer drug resistance. Cancer Drug Resist. 2023, 6, 729-47. http://dx.doi.org/10.20517/cdr.2023.78

About This Article

© The Author(s) 2023. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
822
Downloads
161
Citations
4
Comments
0
26

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 6 clicks
Like This Article 26 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Cancer Drug Resistance
ISSN 2578-532X (Online)

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/