Next Article in Journal
Regucalcin Is a Potential Regulator in Human Cancer: Aiming to Expand into Cancer Therapy
Next Article in Special Issue
Diallyl Trisulfide Induces ROS-Mediated Mitotic Arrest and Apoptosis and Inhibits HNSCC Tumor Growth and Cancer Stemness
Previous Article in Journal
Cardiotoxicity Associated with Immune Checkpoint Inhibitors
Previous Article in Special Issue
Differential Effect of Non-Steroidal Anti-Inflammatory Drugs Aspirin and Naproxen against TMPRSS2-ERG (Fusion)-Driven and Non-Fusion-Driven Prostate Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Promise of Piperine in Cancer Chemoprevention

by
Salma Benayad
1,
Hicham Wahnou
2,
Riad El Kebbaj
1,
Bertrand Liagre
3,
Vincent Sol
3,
Mounia Oudghiri
2,
El Madani Saad
1,
Raphaël Emmanuel Duval
4,* and
Youness Limami
1,2,*
1
Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco
2
Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20100, Morocco
3
Le Laboratoire des Agroressources, Biomolécules et Chimie pour l’Innovation en Santé (LABCiS), University Limoges, UR 22722, F-87000 Limoges, France
4
The Franch Center for Scientific Research (CNRS), Université de Lorraine, L2CM, F-54000 Nancy, France
*
Authors to whom correspondence should be addressed.
Cancers 2023, 15(22), 5488; https://doi.org/10.3390/cancers15225488
Submission received: 13 October 2023 / Revised: 14 November 2023 / Accepted: 16 November 2023 / Published: 20 November 2023
(This article belongs to the Special Issue Chemoprevention Advances in Cancer)

Abstract

:

Simple Summary

With 19.3 million new cancer cases worldwide and nearly 10.0 million cancer-related deaths cancer is a global health challenge. Cancer is a disease caused by a variety of factors that lead to uncontrolled cell growth. Cancer chemoprevention using natural or synthetic compounds is a critical strategy to mitigate the impact of cancer on healthcare systems. One of the naturally occurring compounds, piperine, has shown potential in preventing cancer. Piperine is a nitrogen-containing alkaloid molecule with pleiotropic therapeutic effects, including antioxidant, anti-inflammatory, and immunomodulatory properties. With its various health benefits, piperine shows promise in inhibiting cancer-related molecular processes and enhancing existing treatments. This review explores piperine’s potential in cancer prevention and treatment, suggesting it could be a valuable tool in managing cancer.

Abstract

Cancer, characterized by the unregulated growth and dissemination of malignantly transformed cells, presents a significant global health challenge. The multistage process of cancer development involves intricate biochemical and genetic alterations within target cells. Cancer chemoprevention has emerged as a vital strategy to address this complex issue to mitigate cancer’s impact on healthcare systems. This approach leverages pharmacologically active agents to block, suppress, prevent, or reverse invasive cancer development. Among these agents, piperine, an active alkaloid with a wide range of therapeutic properties, including antioxidant, anti-inflammatory, and immunomodulatory effects, has garnered attention for its potential in cancer prevention and treatment. This comprehensive review explores piperine’s multifaceted role in inhibiting the molecular events and signaling pathways associated with various stages of cancer development, shedding light on its promising prospects as a versatile tool in cancer chemoprevention. Furthermore, the review will also delve into how piperine enhances the effectiveness of conventional treatments such as UV-phototherapy and TRAIL-based therapy, potentially synergizing with existing therapeutic modalities to provide more robust cancer management strategies. Finally, a crucial perspective of the long-term safety and potential side effects of piperine-based therapies and the need for clinical trials is also discussed.

1. Introduction

Cancer is a global crisis responsible for a staggering number of deaths worldwide. The statistics have painted a somber picture in recent years, with millions succumbing to this relentless disease. In 2020, there were approximately 19.3 million new cancer cases worldwide, excluding nonmelanoma skin cancer, and nearly 10.0 million cancer-related deaths, also excluding nonmelanoma skin cancer. Female breast cancer emerged as the most diagnosed cancer, with approximately 2.3 million new cases, followed by lung (11.4%), colorectal (10.0%), prostate (7.3%), and stomach (5.6%) cancers. However, lung cancer remained the leading cause of cancer-related deaths, with an estimated 1.8 million fatalities (18%), followed by colorectal (9.4%), liver (8.3%), stomach (7.7%), and female breast (6.9%) cancers. Notably, there was a significant difference in cancer incidence between transitioned and transitioning countries, with incidence rates being two to three times higher in transitioned countries for both men and women. Moreover, mortality rates varied less, particularly among women [1,2].
Researchers have focused on chemopreventive natural or synthetic compounds in pursuing improved cancer therapies, uncovering promising candidates [3,4,5,6,7,8,9].
Among these natural compounds, piperine has attracted significant interest due to its diverse mechanisms of action. Piperine, chemically identified as 1-[5-[1,3-benzodioxol-5-yl]-1-oxo-2,4-pentadienyl] piperidine, represents a nitrogen-containing alkaloid molecule with a rich historical background. Its initial discovery traces back to 1820 when the Danish chemist Hans Christian Orstedt first isolated it from the dried fruit extract of pepper, presenting itself as a yellow crystalline solid (with a molecular weight of 285.33 g/mol and a melting point of 128–130 °C) [10]. Piperine’s chemical structure comprises conjugated aliphatic chains that serve as a bridge between the piperidine and 5-(3,4-methylenedioxyphenyl) moiety [11] (Figure 1).
Its pleotropic effects, from mitigating inflammation and triggering cell apoptosis to suppressing cancer stem cells and disrupting the cell cycle, underscores its potential significance in modulating cancer development and progression [12,13,14,15,16,17] (Figure 2). Piperine’s unique property of selectively targeting malignant cells while sparing healthy ones positions it as a compelling contender for cancer therapy, addressing the challenge of distinguishing cancerous cells from healthy ones often faced in conventional treatments [18].
Furthermore, piperine can complement conventional cancer treatments by enhancing their efficacy, reducing adverse effects, and improving drug bioavailability [19]. Its capacity to sensitize cancer cells to radiation therapy, surmount multidrug resistance, and synergize with treatment approaches such as TRAIL-based therapy highlights its versatility in the battle against cancer [20,21,22].
In this comprehensive review, we navigate the intricate landscape of piperine’s contributions to cancer prevention and therapy targeting various cancers including breast, cervical, prostate, lung, skin, stomach, liver, colorectal, and bone cancers, in both in vitro and in vivo models (Table 1). By unraveling its diverse mechanisms and therapeutic potential, we aim to illuminate the promising role of piperine in the ongoing battle against cancer.
To ensure the precision and comprehensiveness of our review, our team undertook a meticulous data collection and search procedure encompassing a diverse array of reputable databases, which included Google Scholar, Pubmed, Springer, Elsevier ScienceDirect, and Web of Science. Our focus was primarily on scrutinizing studies published between 2002 and 2023, with the inclusion of two earlier studies for contextual purposes.
To maintain uniformity and guarantee a thorough analysis, we exclusively selected articles containing English text. We conducted keyword and heading searches for the following terms: “piperine”, “cancer chemoprevention”, “cell death”, “antitumor”, “in vivo studies”, “in vitro studies”, and “combination cancer therapies”. Subsequently, we conducted a rigorous selection process. Prior to proceeding with a detailed examination of full-text documents, we eliminated duplicate papers and irrelevant works. Our inclusion criteria were stringent and encompassed original articles and review papers that met specific criteria, thereby ensuring the precision and quality of the information presented in this paper. In total, 96 references were selected for inclusion in our study.

2. Piperine’s Dual Mechanisms of Prevention and Destruction of Cancer

Cancer chemoprevention emboldens the use of natural and synthetic biologically active substances to prevent, inhibit, or reverse cancer progression. Chemopreventive agents have been classified into blocking agents and suppressing agents. Blocking agents impede the initiation of tumors [23,24]. Suppressing agents, on the other hand, act subsequently by suppressing the transformation of initiated cells into preneoplastic and/or neoplastic cells and malignancy [24] (Figure 2). Piperine exhibits a unique duality in its abilities, functioning as both a blocking and a suppressing agent in cancer prevention and therapy. This dual role allows piperine to target multiple pathways and aspects of cancer development and progression, ultimately enhancing the effectiveness of chemoprevention strategies (Table 1).
In this section, we will discuss the detailed mechanistic role of piperine in cancer chemoprevention and destruction.

2.1. Piperine Reduces Inflammation

Inflammation has become a target for cancer prevention and therapy. Many studies have evaluated the anti-inflammatory activity of piperine. In fact, in vitro studies using a variety of isolated cells have been used to screen the anti-inflammatory activity of piperine [12,25]. In a model of LPS-induced inflammation of nucleus pulposus cells, piperine significantly inhibited multiple inflammatory factors and oxidative stress-associated genes [26]. In human peripheral blood mononuclear cells (PBMCs), piperine inhibits IL-2 and interferon-gamma (IFN-γ) production [27]. Additionally, in a model of oxidative stress induced by UV-B in HaCaT keratinocyte cells, piperine also inhibits ROS/RNS production, leading to a decrease in inflammatory mediators such as p38, JNK, AP-1, iNOS (nitric oxide synthase), and COX-2 (cyclooxygenase-2) protein expression [12]. Piperine also inhibits LPS-induced tumor necrosis factor (TNF)-α, IL-6, IL-1β, and prostaglandin E2 (PGE2) production in BV2 microglial cells [28]. Furthermore, piperine down-regulates pathways associated with IL-1β and nuclear factor-κB (NF-κB) [29], activates the Nrf2/keap1 pathway, and inhibits TNF-α-induced expressions of cell adhesion molecules such as ICAM-1, VCAM1, and E-selectin. This leads to the blocking of neutrophil adhesion to the endothelium in a time- and concentration-dependent manner [13,14]. Similarly, in vivo studies using a wide range of acute and chronic experimental models revealed the anti-inflammatory activity of piperine in a dose-dependent way. In experiments involving carrageenan-induced paw edema, formalin-induced arthritis, croton oil-induced granuloma pouch, and cotton pellet-induced granuloma, piperine demonstrated a 56%, 40%, 40%, and 10% inhibition of inflammation, respectively [30,31]. In a research study conducted on fibroblast-like synoviocytes stimulated with IL-1β, derived from patients having rheumatoid arthritis and in an animal model of arthritis, it was found that piperine inhibits IL-6, MMP-13, and AP-1. It also reduces PGE2 levels in a dose-dependent manner. Furthermore, piperine-treated rats showed a significant reduction in nociceptive and arthritic symptoms [32].
In summary, piperine consistently proves its potent anti-inflammatory properties across diverse models. Its efficacy is evident in inhibiting inflammatory factors, modulating cytokine production, and down-regulating key pathways. The dose-dependent inhibition of inflammation in various experimental models underscores piperine’s robust potential for addressing inflammatory-related conditions in future research.

2.2. Piperine Induces Various Cell Death Types

In this section, we discuss how piperine induces various cell death types, including apoptosis, autophagy, ferroptosis, and anoikis, which play significant roles in cancer prevention and therapy.

2.2.1. Apoptosis

Apoptosis can be caused by two main pathways: the intrinsic and extrinsic pathways [33]. Apoptosis can be triggered by different factors and the way the signal is transmitted also varies, but ultimately, they converge onto the same execution pathway. This results in several changes, such as DNA fragmentation, cytoskeletal and nuclear protein degradation, protein cross-linking, the formation of apoptotic bodies, the expression of phagocytic cell receptor ligands, and finally, uptake by phagocytic cells [34].
Apoptosis is known to be induced by many chemopreventive agents and piperine can activate both the intrinsic and extrinsic pathways of apoptosis. A study performed by Tawani et al. reported the cytotoxic effects of piperine against four different human cancer cell lines (i.e., breast carcinoma (MCF-7), liver carcinoma (HepG2), cervical carcinoma (HeLa), and prostate cancer cells (PC3)), showing apoptotic characteristics such as cytoplasmic and nuclear condensation, where the externalization of membrane phospholipid phosphatidylserine and DNA cleavage were also observed [32]. Furthermore, piperine has been shown to target human G-quadruplex DNA sequences, a structure playing a vital role in regulating cellular processes that might contribute to cancer development [32]. These results were confirmed by Jafri et al., showing that piperine induces apoptosis in a dose-dependent manner by increasing reactive oxygen species (ROS) generation, nuclear condensation, the disruption of mitochondrial membrane potential, DNA fragmentation, and finally, the activation of caspase-3 [35] (Figure 3). Furthermore, in vivo studies showed that piperine reduced tumors in an osteosarcoma xenograft mouse model by up-regulating both Bax and p53 expression, as well as reducing Bcl-2 expression [36].

2.2.2. Autophagy

Autophagy is also an interesting way by which some chemopreventive agents could act. It is a fundamental cellular process that eliminates molecules and subcellular elements via lysosome-mediated degradation to promote homeostasis, differentiation, development, and survival [37]. In the context of cancer, autophagy prevents tissue damage and cell death, which can cause cancer initiation and progression [38]. Furthermore, it is known that the central pathway governing autophagy is led by PI3K/Akt/mTOR signaling [32,39]. Strikingly, piperine has been shown to induce autophagy in cancer cells by inhibiting mTORC kinase activity, allowing the formation of autophagosomes [40,41]. In this context, piperine inhibited thioredoxin reductase (TrxR) activity, increased ROS levels, reduced mitochondrial membrane potential, and induced autophagy in Bel-7402/5-FU cells via the regulation of autophagy-related proteins LC3, p62, and beclin-1 [15] (Figure 3). In vivo experiments demonstrated that piperine induces autophagy by inhibiting PI3K signaling, leading to a decrease in oral cancer tumor growth [42].

2.2.3. Ferroptosis

Ferroptosis is also an interesting target of chemopreventive agents. Ferroptosis is characterized by iron and lipidic ROS/peroxides accumulation due to the Fenton reaction and via the loss of balance in ROS production and cell glutathione (GSH)-dependent antioxidants, which protect cells from lipid peroxidation [43]. Piperine could increase the intracellular Ca2+ level and ROS in cancer cells also activate the Fenton reaction at high concentrations [24,43,44,45] (Figure 3).

2.2.4. Anoikis

Anoikis, a particular programmed cell death, could be induced by a piperine structural analogous named piperlongumine in melanoma cells in vitro by inhibiting STAT3 [46] (Figure 3).

2.3. Piperine Inhibits Cancer Stem Cells

In a tumor, cancer stem cells (CSCs) are cells that can undergo continuous self-renewal and generate heterogeneous lines of cancer cells. CSCs also contribute to tumor initiation and the relapse of cancers [47,48,49]. CSCs are characterized by the deregulation of cellular energetics, promoting an inflammatory state in the tumor, evading apoptosis, avoiding immune destruction, and resisting anticancer drugs [49]. At a molecular level, Wnt/β-catenin, Hedgehog, Notch, JAK-STAT (Janus kinase/signal transducers and activators of transcription), NF-κB, PI3K/Akt/mTOR, and TGF/SMAD are fundamental signaling pathways regulating self-renewal and differentiation in CSCs [50,51]. Therefore, finding chemopreventive agents capable of targeting those signaling pathways could help prevent tumor formation. Piperine influences all these pathways directly or indirectly.
On colorectal and breast cancer cell lines, piperine has been shown to inhibit the Wnt/β-catenin signaling pathway [51], and PI3K/Akt/mTOR signaling pathways could also be influenced by piperine [32,39]. Furthermore, human cervical cancer treated with piperine and mitomycin-C resulted in inactivating STAT3/NF-κB, leading to suppression of the Bcl-2 signaling pathway [52] (Figure 4).

2.4. Piperine Induces Cell Cycle Arrest

The dysregulation of cell cycle control, a fundamental cellular process responsible for maintaining proper cell proliferation and safeguarding cellular integrity, is commonly associated with cancer development [53,54]. Two groups of proteins, called cyclins and cyclin-dependent kinases (CDKs), are responsible for the progression of the cell through various checkpoints.
Piperine has shown an inhibition capacity of different protein regulators and checkpoints. Fofaria et al. showed that piperine treatment inhibits the growth of melanoma cells SKMEL 28 and B16F0 in a dose- and time-dependent manner by arresting the cell cycle of both cell lines in the G1 phase [55]. Their results correlated with the down-regulation of cyclin D1 and the induction of p21 [55]. Furthermore, the phosphorylation of H2AX at Ser139 supposed that intracellular ROS formation induced DNA damage, leading to apoptosis [55]. The antiproliferative effect of piperine was also achieved in HT-29 colon carcinoma cells by causing G1 phase cell cycle arrest via the inhibition of cyclins D1 and D3 and their activating partner, cyclin-dependent kinases 4 and 6. A reduction in retinoblastoma protein phosphorylation and an up-regulation of p21/WAF1 and p27/KIP1 was also promoted by piperine [55]. Piperine can also inhibit the cell cycle at the G2/M phase in cancer cells via the down-regulation of G2-associated (cyclin B, CDK1, Cdc25C) proteins, the induction of p21, and the enhancement of the phosphorylation of both CDK1 and checkpoint kinase 2 (Chk2) [18,56,57] (Figure 4).

2.5. Piperine Selectively Inhibits the Growth of Cancer Cells

One of the main limitations of anticancer drugs is their frequent and severe toxic side effects, which are caused by their inability to selectively target cancer cells [58]. In an attempt to address this limitation, piperine has emerged as a promising candidate due to its selective action on cancer cells while sparing non-transformed cells in vitro. In a study by Si et al., normal ovarian cells exposed to 20 µM piperine did not show significant effects on viability. However, exposure to 8 µM piperine caused a significant decrease in ovarian cancer cell viability after 48 h [16]. Similarly, when evaluating prostate cells, piperine displayed no cytotoxicity up to 80 µM on prostate epithelial cells, in contrast to two out of the three prostate cancer cell lines tested [41]. In addition, a 200 µM dose of piperine had a weaker growth inhibitory effect on human hFOB osteoblasts after 72 h of incubation than on human osteosarcoma cells [18] (Figure 4). Nevertheless, in vivo studies are needed to confirm and better understand the selectivity of piperine for cancer cells, as these studies will provide insights into whether piperine’s preferential action against cancer cells observed in vitro can be replicated within living organisms.

2.6. Piperine Inhibits Cancer Invasion and Metastasis Process

Piperine inhibits cancer invasion and metastasis process by having a dual therapeutic potential in cancer by manifesting anti-angiogenic effects, inhibiting the formation of new blood vessels, and displaying anti-metastatic activity, impeding cancer cells from invading other tissues. This is explored in two distinct sections below.

2.6.1. Anti-Angiogenic Effects of Piperine

Tumor progression is a step characterized by the activation of pathways that promote cell migration and the creation of further blood vessels (angiogenesis) that will allow them to self-sustain via the angiogenic factor release, such as the vascular endothelial growth factor (VEGF) [59]. In this regard, an in vivo and in vitro experimental study demonstrated that piperine inhibited tubule formation, which is a crucial step in angiogenesis [60], and inhibited angiogenic activity induced by collagen, caused by breast cancer cells. Piperine also blocked the phosphorylation of Ser 473 and Thr 308 residues of Akt involved in regulating endothelial cells and, therefore, angiogenesis [17]. To assess the effect of piperine on angiogenesis, another study evaluated VEGF expression levels in the vicinity of piperine. This study found that piperine treatment reduced VEGF expression in a dose-dependent manner, suggesting that the alkaloid negatively regulates this key growth factor in cancer cell migration [61]. The effect of piperine on crucial angiogenic factors involved in angiogenesis and tumor progression was elucidated in a study conducted in a human breast cancer cell line [61]. Piperine down-regulated MMP-9 and VEGF mRNA expressions in a dose-dependent manner. In addition, piperine treatment increased the expression levels of E-cadherin, a cell adhesion molecule required to maintain extracellular matrix integrity and cell-to-cell contact, thereby supporting the anti-metastatic potential of this alkaloid [62] (Figure 4).

2.6.2. Anti-Metastatic Activity of Piperine

Metastasis is a process by which cancer cells escape and invade other tissues. Although not all cancer cells reach this stage, metastasis is the leading cause of death from cancer [63]. An experimental test on a murine lung metastasis model also demonstrated piperine’s anti-metastatic activity. Indeed, piperine effectively reduced the size of tumor nodules and the levels of uronic acid and hexosamine, both involved in the metastasis pathway. Hydroxyproline, a collagen metabolite and a potential marker of tumor cell infiltration into the bones, indicating the presence of metastasis, was also quantitatively reduced via the treatment with piperine [64]. Another research study investigating the effect of piperine on a human gastric cancer cell line showed that it inhibits the expression of IL-6, which has a prominent role in cancer cell invasion and metastasis via the c-Src/RhoA/ROCK signaling pathway [56,65]. Further experiments found that piperine inhibited tumor migration and progression by lowering the expression of MMP-13 and -9 and that high doses of piperine significantly reduced lung metastasis [63,64]. Accordingly, experimental studies have elucidated the anti-metastatic activity of piperine by regulating the pathways that involve matrix metalloproteinases [18].
Table 1. Piperine mechanisms and targets in in vitro and in vivo models for cancer therapy.
Table 1. Piperine mechanisms and targets in in vitro and in vivo models for cancer therapy.
TargetsModelMechanismsReferences
In vitro
Inflammation
  • LPS-induced inflammation
  • PBMCs
  • UV-B induce oxidative stress
  • Inhibition of IL-2 and IFN-γ production;
  • Inhibition of ROS/RNS production;
  • Inhibition of the expression of proinflammatory mediators (e.g., p38, iNOS);
  • Down-regulation of IL-1β, NF-κB, and oxidative stress-associated genes.
[26,27,28,29]
Cell death
  • MCF-7, HepG2, HeLa, and PC3, Bel-7402/5-FU cell lines
  • Induces nuclear condensation, externalization of membrane phospholipid phosphatidylserine, DNA cleavage, increases intracellular ROS, disrupts mitochondrial membrane potential, and activates caspase-3.
  • Inhibits mTORC kinase and TrxR activity, regulates LC3, p62, and beclin-1 and induces formation of autophagosomes.
  • Increase in intracellular Ca2+, ROS, and induction of Fenton reaction.
  • Inhibition of STAT3.
[15,32,35,40,41,43,44,45,46]
Cancer stem cells
  • HCT116, SW480, DLD-1, and A549 cell lines
  • Inhibition of Wnt/β-catenin signaling pathway;
  • Inhibition of PI3K/Akt/mTOR signaling pathway;
  • Inactivation of STAT3/NF-κB;
  • Inhibition of Bcl-2 signaling pathway.
[32,39,51,52]
Cell cycle
  • SKMEL 28, B16F0, and HT-29
  • G1 and G2 phases cell cycle arrest;
  • Down-regulation of cyclin D1 and D3;
  • Inhibition of CDK4 and 6;
  • Up-regulation of p21/WAF1 and p27/KIP1;
  • Phosphorylation of H2AX;
  • Down-regulation of cyclin B, CDK1, and Cdc25C proteins.
[18,55,56,57]
Cancer cells growth
  • Hep G2, HOS, U2OS, and hFOB
  • Significantly decreased cancer cells viability;
  • Did not affect normal cells;
[16,18,41]
Invasion and metastasis
  • MDA-MB-231, U2OS, AGS, and 143B cell lines
  • Inhibition of tubule formation;
  • Down-regulation of MMP-9 and VEGF mRNA expression;
  • Inhibition IL-6 expression;
  • Down-regulation of expression of MMP-13 and MMP-9.
[56,60,61,62,63,64,65]
In vivo
Inflammation
  • Carrageenan-induced paw edema;
  • Formalin-induced arthritis;
  • Croton oil-induced gran-uloma pouch;
  • Cotton pellet-induced granuloma.
  • Significant inhibition of inflammation and arthritic symptoms (e.g., pain, edema);
  • Inhibition of IL-6, MMP-13, and AP-1.
[30,31,32]
Cell death (apoptosis, autophagy)
  • Xenograft mouse model;
  • Syngeneic Balb/c mice model.
  • Up-regulation of Bax and p53 expression;
  • Reduction in Bcl-2 expression;
  • Modulation of Wnt/β-catenin pathway;
  • Synergistically inhibits cancer cell proliferation with Celecoxib.
[36,42,66]
Invasion and metastasis
  • Murine lung metastasis;
  • Mice syngeneic to 4T1 cells.
  • Reduced uronic acid, hexosamine, and hydroxyproline levels.
[63,64]

3. Piperine Enlightens the Dark Side of Cancer Therapy

Today, chemotherapy and radiotherapy remain the most effective treatments for many types of cancer. Unfortunately, a proportion of cancer patients suffer from mild to serious side effects [67,68].
Researchers are making extraordinary efforts to improve cancer therapy efficacy, one of which is using natural products (supplements). In addition to enhancing cancer patients’ quality of life, many phytochemicals such as piperine can protect healthy cells, reduce multidrug resistance, increase drug penetration and its concentration inside cancer cells, and reduce the incidence of weight loss, malnutrition, and the severity of comorbidities [67].

3.1. Radiosensitization and UV-Phototherapy

Radiation therapy deposits high physical radiation energy on the cancer cells, leading to their destruction [69]. However, dose-limiting normal tissue toxicity, as well as the radioresistance of some tumors, limit its clinical use in cancer therapeutics [70]. At a molecular level, radiation may directly or indirectly damage DNA by producing free radicals generated when water is ionized or excited by radiation [70]. Thus, agents are needed to increase radiation-killing effectiveness and to prevent radiation-induced damage to normal cells and tissues without causing systemic toxicity. Fortunately, many chemopreventive phytochemicals could rightly fit in this approach thanks to both their cancer-preventive and therapeutic activities [71,72]. A recent study has demonstrated that piperine acts as an antioxidant in normal cells while behaving as a prooxidant in cancer cells [73]. This differential behavior enhances the radiosensitivity of cancer cells without affecting the sensitivity of normal cells. Furthermore, piperine can also increase ROS formation, leading to cell death via the dissipation of mitochondrial membrane potential [35]. Therefore, piperine can enhance ionizing radiation-induced apoptosis in cancer cells. Furthermore, in B16F10 mouse melanoma cells, the piperine combination with UVB has been reported to enhance cell death by elevating intracellular ROS formation and intracellular Ca2+ homeostasis impairment [24] (Figure 5).
On the other hand, UV-phototherapy kills cancer cells in the skin and its effectiveness is due to its capacity to expose only suitable skin areas. Nevertheless, UV-phototherapy can cause skin burns and increase skin cancer risk, like in sunlight-UV exposure. Many botanical agents, such as piperine, can prevent the risk of skin cancer [74]. Piperine attenuates the (UV)-R-induced DNA damage in human HaCaT keratinocytes, according to Jaisin et al. and Verma et al. [12,75]. Furthermore, piperine also inhibits UV-R-induced cell cycle arrest in HaCaT cells. UV-R alone induced p21 up-regulation, whereas, in combination with piperine, no significant up-regulation was observed [75]. Transcriptional and translational analysis revealed that piperine pretreatment suppresses the activation of NF-κB and attenuates the Bax/Bcl-2 expression in UV-R exposed keratinocyte cells, leading to the survival pathway switch [75,76,77] (Figure 5).

3.2. TRAIL-Based Therapy

The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), also known as Apo2L, is a type 2 membrane protein that belongs to the TNF superfamily. With the ability to kill cancer cells selectively by activating a signaling pathway involved in the innate immune system, it has become one of the few tumor-selective agents [78]. However, TRAIL therapy is limited by many cancer cells developing immunity to TRAIL and escaping destruction by the immune system [24,79]. Therefore, a study performed by Abdelhamed et al. aimed to assess the synergistic effects of 55 compounds with TRAIL on TNBC (Triple-Negative Breast Cancer) cells, including both TRAIL-sensitive and TRAIL-resistant cell lines (MDA-MB-231 and MDA-MB-468, respectively). Piperine was identified as a compound with synergistic effects with TRAIL in both cell lines, accompanied by a reduction in p65 phosphorylation and survivin expression [21].
Piperine exhibited differential cell selectivity regarding growth suppression when compared to TRAIL and its synergistic interaction with TRAIL. While the suppression of MDA-MB-468 cell growth by piperine was likely due to a cytostatic effect via G2/M cell cycle arrest, the synergistic effect with TRAIL appeared to involve direct cytotoxicity via apoptosis induction [21]. The study also indicated that the higher sensitivity of MDA-MB-468 cells to piperine was less likely to be solely due to G2/M arrest induction, as other compounds with similar cell cycle effects did not yield the same results [21]. The synergistic effect of piperine with TRAIL appeared to be linked to the suppression of p65 and survivin, which are related to TRAIL responsiveness [80,81]. Cell cycle-arrested cells are known to exhibit increased sensitivity to TRAIL, suggesting that piperine-induced G2/M arrest may contribute to both cytostatic and synergistic effects in MDA-MB-468 cells [21]. Furthermore, combination therapy involving piperine and an anti-DR5 monoclonal antibody significantly inhibited the growth of orthotopically-implanted tumors and appeared to reduce lung metastasis, showing promise in prolonging the survival rates of mice. In conclusion, the study’s findings support the potential of piperine as an adjuvant to enhance the efficacy of TRAIL-based therapy for TNBC patients [21].

3.3. Bioavailability of Drugs

Several factors determine a cancer drug’s bioavailability, including how the drug is released from its pharmaceutical dosage form, whether it is stable in the gastrointestinal tract, whether it dissolves easily, how quickly it passes through the gut wall, and whether it undergoes pre-systemic metabolism [82,83]. In the world of medicine, piperine is considered to be the first bioavailability enhancer that has been scientifically validated [19]. The bioavailability enhancement induced by piperine can be due to several mechanisms. For example, piperine can act in transporter proteins like ATP binding cassette (ABC) transporters, a transmembrane protein responsible for transporting a variety of substrates across extra- and intracellular membranes. ABC transporters are abundantly expressed in the epithelial layer of the gut wall and tumors and are responsible for anticancer drug efflux. Nevertheless, piperine can overcome this limitation. Studies have shown that piperine inhibits P-gp and cytochrome P450 3A4 (CYP3A4) [19]. Furthermore, piperine enhanced the bioavailability of various well-known chemopreventive natural agents (e.g., resveratrol, curcumin), by inhibiting glucuronidation, significantly increasing the plasma concentration of resveratrol when given with resveratrol [84] (Figure 6). Nevertheless, it is worth noting that human studies rarely reported effects that were considered adverse. Their suitability for detailed risk assessment is limited due to several factors. Firstly, there was an insufficient focus on safety parameters beyond drug interactions. Secondly, there was a lack of investigation into potentially adverse effects observed in animal studies. Moreover, the combined administration of piperine with other substances was not extensively explored in these human studies [85]. This leaves a gap in our understanding of the full spectrum of potential effects and safety considerations related to piperine and its interactions with other compounds.

3.4. Multidrug Resistance

Similar to antibiotic resistance, multidrug resistance (MDR) in cancer treatment refers to the ability of cancer cells to survive treatment with various anticancer drugs [86]. Growing evidence suggests that MDR is mediated by the increased efflux of chemotherapeutic drugs, which reduces the drug’s absorption by cancer cells [87]. Furthermore, one of the major mechanisms of MDR is the efflux of anticancer drugs by ABC transporters [88]. Oncogene mutations, tumor microenvironment (TME) changes, tumor heterogeneity, mutations at the target site, or epigenetic changes can also enhance the MDR’s development [89,90].
Phytochemicals, particularly alkaloids such as piperine, have been extensively researched for their potential to reverse MDR in cancer cells. They achieve this by affecting the activity or expression of ABC transporters, as well as by targeting additional molecular factors that can synergize with anticancer drugs [91,92]. Wojtowicz et al. discovered that piperine targets diverse drug resistance mechanisms in human ovarian cancer cell line W1 and its paclitaxel and topotecan-resistant sublines [93]. Immunofluorescence and Western blot analyses demonstrated that piperine augmented the expression of protein tyrosine phosphatase receptor type K (PTPRK). This protein was found to be down-regulated in 15 drug-resistant ovarian cancer cell lines and aldehyde dehydrogenase 1 family member A1 (ALDH1A1) positive CSCs’ population [20]. Piperine can also down-regulate P-gp and BCRP expression, limiting their ability to reject anticancer drugs [20]. Furthermore, piperine treatment down-regulates MDR1, MRP1, and BCRP genes [94,95]. Piperine can also influence the tumor microenvironment by decreasing the extracellular matrix (ECM)’s protein levels (COL3A1, TGFBI), leading to higher drug concentrations in the cell [20] (Figure 6). As a result of these combined factors, cancer cells become more susceptible to cytotoxic drugs.

4. Conclusions

This comprehensive review provides an in-depth exploration of the potential anticancer properties of piperine, shedding light on their mechanisms of action and therapeutic potential. The review emphasizes the significance of targeting key cellular processes involved in cancer development and progression. Piperine has exhibited remarkable preventive effects by inhibiting cell proliferation, inducing various cell deaths, and modulating signaling pathways implicated in cancer initiation. Moreover, piperine has emerged as a promising adjuvant therapy when combined with conventional anticancer drugs, as it enhances their efficacy by improving drug bioavailability, inhibiting drug efflux transporters, and modulating drug resistance pathways. The chemopreventive properties of piperine are further highlighted via its various mechanisms.
In addition to the current findings, several future perspectives warrant exploration to advance the understanding and application of piperine in cancer therapy. Further studies should aim to elucidate the specific molecular targets and pathways influenced by piperine. Thus, researchers can gain a deeper understanding of its precise mechanisms of action, allowing for the development of more precise and effective therapies. Identifying these specific targets and pathways can also pave the way for personalized treatment strategies, optimizing therapeutic benefits while minimizing adverse effects.
Furthermore, the investigation of piperine’s potential synergistic effects with other compounds is of paramount importance. Uncovering novel combination therapies with enhanced efficacy could lead to more potent and individualized treatment options. These combinations have the potential to increase response rates and significantly impact patient outcomes, making them a crucial area of study.
Optimizing the delivery systems for piperine administration represents a promising avenue of research. Enhancing targeting and bioavailability will ensure that piperine reaches its intended site of action within the body, maximizing its therapeutic benefits [96]. This optimization could lead to improved treatment outcomes and a higher quality of life for patients.
Conducting well-designed clinical trials is critical to assess the safety, tolerability, and effectiveness of piperine-based therapies in diverse patient populations. These trials provide valuable data on the real-world applicability of piperine in cancer therapy. If proven effective, piperine-based treatments may become a standard part of cancer care, offering patients a broader range of treatment options and potentially increasing their chances of survival. Moreover, exploring the chemopreventive effects of piperine in high-risk populations holds significant promise. This research can provide valuable insights into piperine’s potential as a preventive agent against the development of cancer. By identifying individuals at a higher risk of cancer and providing them with preventive strategies involving piperine, we can significantly reduce cancer incidence. This approach benefits individuals and society alike by reducing the societal and economic burden of cancer. These ongoing research efforts and future directions will contribute to the advancement of piperine-based therapies, paving the way for their integration into the clinical management of cancer. Ultimately, the comprehensive understanding and optimized utilization of piperine hold tremendous potential for improving patient outcomes and alleviating the burden of this devastating disease on individuals and society.

Author Contributions

Conceptualization, R.E.D. and Y.L.; data curation, S.B., H.W., R.E.K. and M.O.; figures’ conception, H.W.; writing—original draft preparation, S.B. and H.W.; writing—review and editing, R.E.K., B.L., V.S., M.O., E.M.S., R.E.D. and Y.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  3. Chien, H.J.; Liu, C.J.; Ying, T.H.; Wu, P.J.; Wang, J.W.; Ting, Y.H.; Hsieh, Y.H.; Wang, S.C. Timosaponin AIII Inhibits Migration and Invasion Abilities in Human Cervical Cancer Cells through Inactivation of p38 MAPK-Mediated uPA Expression In Vitro and In Vivo. Cancers 2022, 15, 37. [Google Scholar] [CrossRef] [PubMed]
  4. Liu, Z.; Yokoyama, N.N.; Song, L.; Xie, J.; Sadeghi, Z.; Wu, Y.X.; Yee, S.; Wu, X.R.; Wang, B.; Uchio, E.; et al. The Potent Anti-Tumor Effects of Rhodiola Drinking Are Associated with the Inhibition of the mTOR Pathway and Modification of Tumor Metabolism in the UPII-Mutant Ha-Ras Model. Cancers 2023, 15, 3086. [Google Scholar] [CrossRef]
  5. Madka, V.; Patlolla, J.M.R.; Venkatachalam, K.; Zhang, Y.; Pathuri, G.; Stratton, N.; Lightfoot, S.; Janakiram, N.B.; Mohammed, A.; Rao, C.V. Chemoprevention of Colon Cancer by DFMO, Sulindac, and NO-Sulindac Administered Individually or in Combinations in F344 Rats. Cancers 2023, 15, 4001. [Google Scholar] [CrossRef]
  6. Ismail, B.; Ghezali, L.; Gueye, R.; Limami, Y.; Pouget, C.; Leger, D.Y.; Martin, F.; Beneytout, J.L.; Duroux, J.L.; Diab-Assaf, M.; et al. Novel methylsulfonyl chalcones as potential antiproliferative drugs for human prostate cancer: Involvement of the intrinsic pathway of apoptosis. Int. J. Oncol. 2013, 43, 1160–1168. [Google Scholar] [CrossRef]
  7. Soltana, H.; Pinon, A.; Limami, Y.; Zaid, Y.; Khalki, L.; Zaid, N.; Salah, D.; Sabitaliyevich, U.Y.; Simon, A.; Liagre, B.; et al. Antitumoral activity of Ficus carica L. on colorectal cancer cell lines. Cell. Mol. Biol. 2019, 65, 6–11. [Google Scholar] [CrossRef]
  8. Hassan, L.; Pinon, A.; Limami, Y.; Seeman, J.; Fidanzi-Dugas, C.; Martin, F.; Badran, B.; Simon, A.; Liagre, B. Resistance to ursolic acid-induced apoptosis through involvement of melanogenesis and COX-2/PGE2 pathways in human M4Beu melanoma cancer cells. Exp. Cell Res. 2016, 345, 60–69. [Google Scholar] [CrossRef]
  9. da Silva Fernandes, A.; de Oliveira, C.G.; Evangelista, H.; Ds, M.S.; Araujo-Lima, C.F.; Felzenszwalb, I. In vitro chemopreventive and cytotoxic effects of Amazon mosses Leucobryum martianum (Hornsch.) and Leucobryum laevifolium (Broth) extracts. Mutagenesis 2023, gead028. [Google Scholar] [CrossRef]
  10. Gorgani, L.; Mohammadi, M.; Najafpour, G.D.; Nikzad, M. Piperine—The Bioactive Compound of Black Pepper: From Isolation to Medicinal Formulations. Compr. Rev. Food Sci. Food Saf. 2017, 16, 124–140. [Google Scholar] [CrossRef]
  11. Aziz, N.S.; Sofian-Seng, N.-S.; Mohd Razali, N.S.; Lim, S.J.; Mustapha, W.A.W. A review on conventional and biotechnological approaches in white pepper production. J. Sci. Food Agric. 2019, 99, 2665–2676. [Google Scholar] [CrossRef] [PubMed]
  12. Jaisin, Y.; Ratanachamnong, P.; Wongsawatkul, O.; Watthammawut, A.; Malaniyom, K.; Natewong, S. Antioxidant and anti-inflammatory effects of piperine on UV-B-irradiated human HaCaT keratinocyte cells. Life Sci. 2020, 263, 118607. [Google Scholar] [CrossRef]
  13. Wang, L.; Yin, Q.; Liu, C.; Tang, Y.; Sun, C.; Zhuang, J. Nanoformulations of Ursolic Acid: A Modern Natural Anticancer Molecule. Front. Pharmacol. 2021, 12, 706121. [Google Scholar] [CrossRef] [PubMed]
  14. Kumar, S.; Malhotra, S.; Prasad, A.K.; Van der Eycken, E.V.; Bracke, M.E.; Stetler-Stevenson, W.G.; Parmar, V.S.; Ghosh, B. Anti-inflammatory and antioxidant properties of Piper species: A perspective from screening to molecular mechanisms. Curr. Top. Med. Chem. 2015, 15, 886–893. [Google Scholar] [CrossRef] [PubMed]
  15. Zhu, P.; Qian, J.; Xu, Z.; Meng, C.; Liu, J.; Shan, W.; Zhu, W.; Wang, Y.; Yang, Y.; Zhang, W.; et al. Piperlonguminine and Piperine Analogues as TrxR Inhibitors that Promote ROS and Autophagy and Regulate p38 and Akt/mTOR Signaling. J. Nat. Prod. 2020, 83, 3041–3049. [Google Scholar] [CrossRef]
  16. Gunasekaran, V.; Elangovan, K.; Niranjali Devaraj, S. Targeting hepatocellular carcinoma with piperine by radical-mediated mitochondrial pathway of apoptosis: An in vitro and in vivo study. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2017, 105, 106–118. [Google Scholar] [CrossRef]
  17. Doucette, C.D.; Hilchie, A.L.; Liwski, R.; Hoskin, D.W. Piperine, a dietary phytochemical, inhibits angiogenesis. J. Nutr. Biochem. 2013, 24, 231–239. [Google Scholar] [CrossRef]
  18. Zhang, J.; Zhu, X.; Li, H.; Li, B.; Sun, L.; Xie, T.; Zhu, T.; Zhou, H.; Ye, Z. Piperine inhibits proliferation of human osteosarcoma cells via G2/M phase arrest and metastasis by suppressing MMP-2/-9 expression. Int. Immunopharmacol. 2015, 24, 50–58. [Google Scholar] [CrossRef]
  19. Atal, N.; Bedi, K.L. Bioenhancers: Revolutionary concept to market. J. Ayurveda Integr. Med. 2010, 1, 96–99. [Google Scholar] [CrossRef]
  20. Świerczewska, M.; Sterzyńska, K.; Wojtowicz, K.; Kaźmierczak, D.; Iżycki, D.; Nowicki, M.; Zabel, M.; Januchowski, R. PTPRK Expression Is Downregulated in Drug Resistant Ovarian Cancer Cell Lines, and Especially in ALDH1A1 Positive CSCs-Like Populations. Int. J. Mol. Sci. 2019, 20, 2053. [Google Scholar] [CrossRef]
  21. Abdelhamed, S.; Yokoyama, S.; Refaat, A.; Ogura, K.; Yagita, H.; Awale, S.; Saiki, I. Piperine enhances the efficacy of TRAIL-based therapy for triple-negative breast cancer cells. Anticancer Res. 2014, 34, 1893–1899. [Google Scholar] [PubMed]
  22. Limami, Y.; Pinon, A.; Riaz, A.; Simon, A. TRAIL and targeting cancer cells: Between promises and obstacles. Cell. Mol. Biol. 2015, 61, 33–38. [Google Scholar] [PubMed]
  23. Wattenberg, L.W. Chemoprevention of cancer. Cancer Res. 1985, 45, 1–8. [Google Scholar] [CrossRef] [PubMed]
  24. Rather, R.A.; Bhagat, M. Cancer Chemoprevention and Piperine: Molecular Mechanisms and Therapeutic Opportunities. Front. Cell Dev. Biol. 2018, 6, 10. [Google Scholar] [CrossRef]
  25. Woo, H.M.; Kang, J.H.; Kawada, T.; Yoo, H.; Sung, M.K.; Yu, R. Active spice-derived components can inhibit inflammatory responses of adipose tissue in obesity by suppressing inflammatory actions of macrophages and release of monocyte chemoattractant protein-1 from adipocytes. Life Sci. 2007, 80, 926–931. [Google Scholar] [CrossRef]
  26. Li, Y.; Li, K.; Hu, Y.; Xu, B.; Zhao, J. Piperine mediates LPS induced inflammatory and catabolic effects in rat intervertebral disc. Int. J. Clin. Exp. Pathol. 2015, 8, 6203–6213. [Google Scholar]
  27. Chuchawankul, S.; Khorana, N.; Poovorawan, Y. Piperine inhibits cytokine production by human peripheral blood mononuclear cells. Genet. Mol. Res. GMR 2012, 11, 617–627. [Google Scholar] [CrossRef]
  28. Wang-Sheng, C.; Jie, A.; Jian-Jun, L.; Lan, H.; Zeng-Bao, X.; Chang-Qing, L. Piperine attenuates lipopolysaccharide (LPS)-induced inflammatory responses in BV2 microglia. Int. Immunopharmacol. 2017, 42, 44–48. [Google Scholar] [CrossRef]
  29. Zazeri, G.; Povinelli, A.P.R.; Le Duff, C.S.; Tang, B.; Cornelio, M.L.; Jones, A.M. Synthesis and Spectroscopic Analysis of Piperine- and Piperlongumine-Inspired Natural Product Scaffolds and Their Molecular Docking with IL-1β and NF-κB Proteins. Molecules 2020, 25, 2841. [Google Scholar] [CrossRef]
  30. Sireeratawong, S.; Itharat, A.; Lerdvuthisopon, N.; Piyabhan, P.; Khonsung, P.; Boonraeng, S.; Jaijoy, K. Anti-Inflammatory, Analgesic, and Antipyretic Activities of the Ethanol Extract of Piper interruptum Opiz. and Piper chaba Linn. ISRN Pharmacol. 2012, 2012, 480265. [Google Scholar] [CrossRef]
  31. Bang, J.S.; Oh, D.H.; Choi, H.M.; Sur, B.-J.; Lim, S.-J.; Kim, J.Y.; Yang, H.-I.; Yoo, M.C.; Hahm, D.-H.; Kim, K.S. Anti-inflammatory and antiarthritic effects of piperine in human interleukin 1β-stimulated fibroblast-like synoviocytes and in rat arthritis models. Arthritis Res. Ther. 2009, 11, R49. [Google Scholar] [CrossRef] [PubMed]
  32. Tawani, A.; Amanullah, A.; Mishra, A.; Kumar, A. Evidences for Piperine inhibiting cancer by targeting human G-quadruplex DNA sequences. Sci. Rep. 2016, 6, 39239. [Google Scholar] [CrossRef] [PubMed]
  33. Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef] [PubMed]
  34. Fitzgerald, M.-C.; O’Halloran, P.J.; Connolly, N.M.C.; Murphy, B.M. Targeting the apoptosis pathway to treat tumours of the paediatric nervous system. Cell Death Dis. 2022, 13, 460. [Google Scholar] [CrossRef] [PubMed]
  35. Jafri, A.; Siddiqui, S.; Rais, J.; Ahmad, M.S.; Kumar, S.; Jafar, T.; Afzal, M.; Arshad, M. Induction of apoptosis by piperine in human cervical adenocarcinoma via ROS mediated mitochondrial pathway and caspase-3 activation. EXCLI J. 2019, 18, 154–164. [Google Scholar] [CrossRef]
  36. Qi, Y.; Yao, L.; Liu, J.; Wang, W. Piperine improves the sensitivity of osteosarcoma cells to doxorubicin by inducing apoptosis and inhibiting the PI3K/AKT/GSK-3β pathway. J. Orthop. Surg. Res. 2023, 18, 180. [Google Scholar] [CrossRef]
  37. Aman, Y.; Schmauck-Medina, T.; Hansen, M.; Morimoto, R.I.; Simon, A.K.; Bjedov, I.; Palikaras, K.; Simonsen, A.; Johansen, T.; Tavernarakis, N.; et al. Autophagy in healthy aging and disease. Nat. Aging 2021, 1, 634–650. [Google Scholar] [CrossRef]
  38. Chen, H.Y.; White, E. Role of autophagy in cancer prevention. Cancer Prev. Res. 2011, 4, 973–983. [Google Scholar] [CrossRef]
  39. Alvarez-Meythaler, J.G.; Garcia-Mayea, Y.; Mir, C.; Kondoh, H.; ME, L.L. Autophagy Takes Center Stage as a Possible Cancer Hallmark. Front. Oncol. 2020, 10, 586069. [Google Scholar] [CrossRef]
  40. Kaur, H.; He, B.; Zhang, C.; Rodriguez, E.; Hage, D.S.; Moreau, R. Piperine potentiates curcumin-mediated repression of mTORC1 signaling in human intestinal epithelial cells: Implications for the inhibition of protein synthesis and TNFα signaling. J. Nutr. Biochem. 2018, 57, 276–286. [Google Scholar] [CrossRef]
  41. Ouyang, D.Y.; Zeng, L.H.; Pan, H.; Xu, L.H.; Wang, Y.; Liu, K.P.; He, X.H. Piperine inhibits the proliferation of human prostate cancer cells via induction of cell cycle arrest and autophagy. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2013, 60, 424–430. [Google Scholar] [CrossRef] [PubMed]
  42. Han, E.J.; Choi, E.Y.; Jeon, S.J.; Lee, S.W.; Moon, J.M.; Jung, S.H.; Jung, J.Y. Piperine Induces Apoptosis and Autophagy in HSC-3 Human Oral Cancer Cells by Regulating PI3K Signaling Pathway. Int. J. Mol. Sci. 2023, 24, 13949. [Google Scholar] [CrossRef] [PubMed]
  43. Tang, D.; Kang, R.; Berghe, T.V.; Vandenabeele, P.; Kroemer, G. The molecular machinery of regulated cell death. Cell Res. 2019, 29, 347–364. [Google Scholar] [CrossRef] [PubMed]
  44. Mittal, R.; Gupta, R.L. In vitro antioxidant activity of piperine. Methods Find. Exp. Clin. Pharmacol. 2000, 22, 271–274. [Google Scholar] [CrossRef]
  45. 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, 1011–1022. [Google Scholar] [CrossRef] [PubMed]
  46. Fofaria, N.M.; Srivastava, S.K. STAT3 induces anoikis resistance, promotes cell invasion and metastatic potential in pancreatic cancer cells. Carcinogenesis 2015, 36, 142–150. [Google Scholar] [CrossRef]
  47. Economopoulou, P.; Kaklamani, V.G.; Siziopikou, K. The role of cancer stem cells in breast cancer initiation and progression: Potential cancer stem cell-directed therapies. Oncologist 2012, 17, 1394–1401. [Google Scholar] [CrossRef] [PubMed]
  48. Scarpa, E.S.; Ninfali, P. Phytochemicals as Innovative Therapeutic Tools against Cancer Stem Cells. Int. J. Mol. Sci. 2015, 16, 15727–15742. [Google Scholar] [CrossRef]
  49. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  50. Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Zhang, G.; Wang, X.; Dong, Z.; Chen, F.; et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct. Target. Ther. 2020, 5, 8. [Google Scholar] [CrossRef]
  51. de Almeida, G.C.; Oliveira, L.F.S.; Predes, D.; Fokoue, H.H.; Kuster, R.M.; Oliveira, F.L.; Mendes, F.A.; Abreu, J.G. Piperine suppresses the Wnt/β-catenin pathway and has anti-cancer effects on colorectal cancer cells. Sci. Rep. 2020, 10, 11681. [Google Scholar] [CrossRef] [PubMed]
  52. Stojanović-Radić, Z.; Pejčić, M.; Dimitrijević, M.; Aleksić, A.; Anil Kumar, N.V.; Salehi, B.; Cho, W.C.; Sharifi-Rad, J. Piperine-A Major Principle of Black Pepper: A Review of Its Bioactivity and Studies. Appl. Sci. 2019, 9, 4270. [Google Scholar] [CrossRef]
  53. Feitelson, M.A.; Arzumanyan, A.; Kulathinal, R.J.; Blain, S.W.; Holcombe, R.F.; Mahajna, J.; Marino, M.; Martinez-Chantar, M.L.; Nawroth, R.; Sanchez-Garcia, I.; et al. Sustained proliferation in cancer: Mechanisms and novel therapeutic targets. Semin. Cancer Biol. 2015, 35, S25–S54. [Google Scholar] [CrossRef] [PubMed]
  54. Samiry, I.; Pinon, A.; Limami, Y.; Rais, S.; Zaid, Y.; Oudghiri, M.; Liagre, B.; Mtairag, E.M. Antitumoral activity of Caralluma europaea on colorectal and prostate cancer cell lines. J. Toxicol. Environ. Health Part A 2023, 86, 230–240. [Google Scholar] [CrossRef] [PubMed]
  55. Fofaria, N.M.; Kim, S.H.; Srivastava, S.K. Piperine causes G1 phase cell cycle arrest and apoptosis in melanoma cells through checkpoint kinase-1 activation. PLoS ONE 2014, 9, e94298. [Google Scholar] [CrossRef]
  56. Greenshields, A.L.; Doucette, C.D.; Sutton, K.M.; Madera, L.; Annan, H.; Yaffe, P.B.; Knickle, A.F.; Dong, Z.; Hoskin, D.W. Piperine inhibits the growth and motility of triple-negative breast cancer cells. Cancer Lett. 2015, 357, 129–140. [Google Scholar] [CrossRef]
  57. Pressete, C.G.; Viegas, F.P.; Campos, T.G.; Caixeta, E.S.; Hanemann, J.A.; Ferreira-Silva, G.Á.; Zavan, B.; Aissa, A.F.; Miyazawa, M.; Viegas, C.; et al. Piperine–Chlorogenic Acid Hybrid Inhibits the Proliferation of the SK-MEL-147 Melanoma Cells by Modulating Mitotic Kinases. Pharmaceuticals 2023, 16, 145. [Google Scholar] [CrossRef]
  58. Suter, T.M.; Ewer, M.S. Cancer drugs and the heart: Importance and management. Eur. Heart J. 2013, 34, 1102–1111. [Google Scholar] [CrossRef]
  59. Yehya, A.H.S.; Asif, M.; Petersen, S.H.; Subramaniam, A.V.; Kono, K.; Majid, A.; Oon, C.E. Angiogenesis: Managing the Culprits behind Tumorigenesis and Metastasis. Medicina 2018, 54, 8. [Google Scholar] [CrossRef]
  60. Tobelem, G. Tumor angiogenesis. Nouv. Rev. Fr. D’hematol. 1990, 32, 405–406. [Google Scholar]
  61. Qi, Y.B.; Yang, W.; Si, M.; Nie, L. Wnt/β-catenin signaling modulates piperine-mediated antitumor effects on human osteosarcoma cells. Mol. Med. Rep. 2020, 21, 2202–2208. [Google Scholar] [CrossRef] [PubMed]
  62. Zahra, Z.; Tina Nayerpour, D.; Armaghan, L.; Zakieh Sadat, S.; Mohammad, P.; Fahimeh, H.; Vajiheh, N.; Omid, A.; Mojtaba, A.; Parisa, K. The Effect of Piperine on MMP-9, VEGF, and E-cadherin Expression in Breast Cancer MCF-7 Cell Line. Basic Clin. Cancer Res. 2020, 12, 112–119. [Google Scholar] [CrossRef]
  63. Lai, L.H.; Fu, Q.H.; Liu, Y.; Jiang, K.; Guo, Q.M.; Chen, Q.Y.; Yan, B.; Wang, Q.Q.; Shen, J.G. Piperine suppresses tumor growth and metastasis in vitro and in vivo in a 4T1 murine breast cancer model. Acta Pharmacol. Sin. 2012, 33, 523–530. [Google Scholar] [CrossRef] [PubMed]
  64. Gasser, A.B.; Depierre, D.; Courvoisier, B. Total urinary and free serum hydroxyproline in metastatic bone disease. Br. J. Cancer 1979, 39, 280–283. [Google Scholar] [CrossRef]
  65. Lin, M.T.; Lin, B.R.; Chang, C.C.; Chu, C.Y.; Su, H.J.; Chen, S.T.; Jeng, Y.M.; Kuo, M.L. IL-6 induces AGS gastric cancer cell invasion via activation of the c-Src/RhoA/ROCK signaling pathway. Int. J. Cancer 2007, 120, 2600–2608. [Google Scholar] [CrossRef]
  66. Srivastava, S.; Dewangan, J.; Mishra, S.; Divakar, A.; Chaturvedi, S.; Wahajuddin, M.; Kumar, S.; Rath, S.K. Piperine and Celecoxib synergistically inhibit colon cancer cell proliferation via modulating Wnt/β-catenin signaling pathway. Phytomed. Int. J. Phytother. Phytopharm. 2021, 84, 153484. [Google Scholar] [CrossRef]
  67. Bassam, H. Plants and Cancer Treatment. In Medicinal Plants; Bassam Abdul Rasool, H., Ed.; IntechOpen: Rijeka, Croatia, 2019; Chapter 9. [Google Scholar] [CrossRef]
  68. Wahnou, H.; Youlyouz-Marfak, I.; Liagre, B.; Sol, V.; Oudghiri, M.; Duval, R.E.; Limami, Y. Shining a Light on Prostate Cancer: Photodynamic Therapy and Combination Approaches. Pharmaceutics 2023, 15, 1767. [Google Scholar] [CrossRef]
  69. Connell, P.P.; Hellman, S. Advances in radiotherapy and implications for the next century: A historical perspective. Cancer Res. 2009, 69, 383–392. [Google Scholar] [CrossRef]
  70. Kim, B.M.; Hong, Y.; Lee, S.; Liu, P.; Lim, J.H.; Lee, Y.H.; Lee, T.H.; Chang, K.T.; Hong, Y. Therapeutic Implications for Overcoming Radiation Resistance in Cancer Therapy. Int. J. Mol. Sci. 2015, 16, 26880–26913. [Google Scholar] [CrossRef]
  71. Nambiar, D.; Rajamani, P.; Singh, R.P. Effects of phytochemicals on ionization radiation-mediated carcinogenesis and cancer therapy. Mutat. Res. 2011, 728, 139–157. [Google Scholar] [CrossRef]
  72. Kotecha, R.; Takami, A.; Espinoza, J.L. Dietary phytochemicals and cancer chemoprevention: A review of the clinical evidence. Oncotarget 2016, 7, 52517–52529. [Google Scholar] [CrossRef] [PubMed]
  73. Tak, J.K.; Lee, J.H.; Park, J.W. Resveratrol and piperine enhance radiosensitivity of tumor cells. BMB Rep. 2012, 45, 242–246. [Google Scholar] [CrossRef] [PubMed]
  74. Eli, R.; Fasciano, J.A. An adjunctive preventive treatment for cancer: Ultraviolet light and ginkgo biloba, together with other antioxidants, are a safe and powerful, but largely ignored, treatment option for the prevention of cancer. Med. Hypotheses 2006, 66, 1152–1156. [Google Scholar] [CrossRef] [PubMed]
  75. Verma, A.; Kushwaha, H.N.; Srivastava, A.K.; Srivastava, S.; Jamal, N.; Srivastava, K.; Ray, R.S. Piperine attenuates UV-R induced cell damage in human keratinocytes via NF-kB, Bax/Bcl-2 pathway: An application for photoprotection. J. Photochem. Photobiol. B Biol. 2017, 172, 139–148. [Google Scholar] [CrossRef] [PubMed]
  76. Cory, S.; Adams, J.M. The Bcl2 family: Regulators of the cellular life-or-death switch. Nat. Rev. Cancer 2002, 2, 647–656. [Google Scholar] [CrossRef] [PubMed]
  77. Shimizu, H.; Bolati, D.; Adijiang, A.; Muteliefu, G.; Enomoto, A.; Nishijima, F.; Dateki, M.; Niwa, T. NF-κB plays an important role in indoxyl sulfate-induced cellular senescence, fibrotic gene expression, and inhibition of proliferation in proximal tubular cells. Am. J. Physiol. Cell Physiol. 2011, 301, C1201–C1212. [Google Scholar] [CrossRef] [PubMed]
  78. Mérino, D.; Lalaoui, N.; Morizot, A.; Solary, E.; Micheau, O. TRAIL in cancer therapy: Present and future challenges. Expert Opin. Ther. Targets 2007, 11, 1299–1314. [Google Scholar] [CrossRef]
  79. Ismail, B.; Fagnere, C.; Limami, Y.; Ghezali, L.; Pouget, C.; Fidanzi, C.; Ouk, C.; Gueye, R.; Beneytout, J.L.; Duroux, J.L.; et al. 2′-Hydroxy-4-methylsulfonylchalcone enhances TRAIL-induced apoptosis in prostate cancer cells. Anti-Cancer Drugs 2015, 26, 74–84. [Google Scholar] [CrossRef]
  80. Chawla-Sarkar, M.; Bae, S.I.; Reu, F.J.; Jacobs, B.S.; Lindner, D.J.; Borden, E.C. Downregulation of Bcl-2, FLIP or IAPs (XIAP and survivin) by siRNAs sensitizes resistant melanoma cells to Apo2L/TRAIL-induced apoptosis. Cell Death Differ. 2004, 11, 915–923. [Google Scholar] [CrossRef]
  81. Premkumar, D.R.; Jane, E.P.; Foster, K.A.; Pollack, I.F. Survivin Inhibitor YM-155 Sensitizes Tumor Necrosis Factor–Related Apoptosis-Inducing Ligand-Resistant Glioma Cells to Apoptosis through Mcl-1 Downregulation and by Engaging the Mitochondrial Death Pathway. J. Pharmacol. Exp. Ther. 2013, 346, 201–210. [Google Scholar] [CrossRef]
  82. Pond, S.M.; Tozer, T.N. First-pass elimination. Basic concepts and clinical consequences. Clin. Pharmacokinet. 1984, 9, 1–25. [Google Scholar] [CrossRef] [PubMed]
  83. Burton, P.S.; Goodwin, J.T.; Vidmar, T.J.; Amore, B.M. Predicting drug absorption: How nature made it a difficult problem. J. Pharmacol. Exp. Ther. 2002, 303, 889–895. [Google Scholar] [CrossRef] [PubMed]
  84. Johnson, J.J.; Nihal, M.; Siddiqui, I.A.; Scarlett, C.O.; Bailey, H.H.; Mukhtar, H.; Ahmad, N. Enhancing the bioavailability of resveratrol by combining it with piperine. Mol. Nutr. Food Res. 2011, 55, 1169–1176. [Google Scholar] [CrossRef]
  85. Ziegenhagen, R.; Heimberg, K.; Lampen, A.; Hirsch-Ernst, K.I. Safety Aspects of the Use of Isolated Piperine Ingested as a Bolus. Foods 2021, 10, 2121. [Google Scholar] [CrossRef]
  86. Zahreddine, H.; Borden, K.L. Mechanisms and insights into drug resistance in cancer. Front. Pharmacol. 2013, 4, 28. [Google Scholar] [CrossRef]
  87. Sampath, D.; Cortes, J.; Estrov, Z.; Du, M.; Shi, Z.; Andreeff, M.; Gandhi, V.; Plunkett, W. Pharmacodynamics of cytarabine alone and in combination with 7-hydroxystaurosporine (UCN-01) in AML blasts in vitro and during a clinical trial. Blood 2006, 107, 2517–2524. [Google Scholar] [CrossRef] [PubMed]
  88. Choi, C.H. ABC transporters as multidrug resistance mechanisms and the development of chemosensitizers for their reversal. Cancer Cell Int. 2005, 5, 30. [Google Scholar] [CrossRef]
  89. Xue, M.; Cheng, J.; Zhao, J.; Zhang, S.; Jian, J.; Qiao, Y.; Liu, B. Outcomes of 219 chronic myeloid leukaemia patients with additional chromosomal abnormalities and/or tyrosine kinase domain mutations. Int. J. Lab. Hematol. 2019, 41, 94–101. [Google Scholar] [CrossRef]
  90. Yeldag, G.; Rice, A.; Del Río Hernández, A. Chemoresistance and the Self-Maintaining Tumor Microenvironment. Cancers 2018, 10, 471. [Google Scholar] [CrossRef]
  91. Wink, M. Molecular modes of action of cytotoxic alkaloids: From DNA intercalation, spindle poisoning, topoisomerase inhibition to apoptosis and multiple drug resistance. Alkaloids. Chem. Biol. 2007, 64, 1–47. [Google Scholar] [CrossRef]
  92. Wink, M.; Ashour, M.L.; El-Readi, M.Z. Secondary Metabolites from Plants Inhibiting ABC Transporters and Reversing Resistance of Cancer Cells and Microbes to Cytotoxic and Antimicrobial Agents. Front. Microbiol. 2012, 3, 130. [Google Scholar] [CrossRef] [PubMed]
  93. Wojtowicz, K.; Sterzyńska, K.; Świerczewska, M.; Nowicki, M.; Zabel, M.; Januchowski, R. Piperine Targets Different Drug Resistance Mechanisms in Human Ovarian Cancer Cell Lines Leading to Increased Sensitivity to Cytotoxic Drugs. Int. J. Mol. Sci. 2021, 22, 4243. [Google Scholar] [CrossRef] [PubMed]
  94. Li, S.; Lei, Y.; Jia, Y.; Li, N.; Wink, M.; Ma, Y. Piperine, a piperidine alkaloid from Piper nigrum re-sensitizes P-gp, MRP1 and BCRP dependent multidrug resistant cancer cells. Phytomed. Int. J. Phytother. Phytopharm. 2011, 19, 83–87. [Google Scholar] [CrossRef] [PubMed]
  95. Pal, M.K.; Jaiswar, S.P.; Srivastav, A.K.; Goyal, S.; Dwivedi, A.; Verma, A.; Singh, J.; Pathak, A.K.; Sankhwar, P.L.; Ray, R.S. Synergistic effect of piperine and paclitaxel on cell fate via cyt-c, Bax/Bcl-2-caspase-3 pathway in ovarian adenocarcinomas SKOV-3 cells. Eur. J. Pharmacol. 2016, 791, 751–762. [Google Scholar] [CrossRef]
  96. Wahnou, H.; Liagre, B.; Sol, V.; El Attar, H.; Attar, R.; Oudghiri, M.; Duval, R.E.; Limami, Y. Polyphenol-Based Nanoparticles: A Promising Frontier for Enhanced Colorectal Cancer Treatment. Cancers 2023, 15, 3826. [Google Scholar] [CrossRef]
Figure 1. Piperine’s chemical structure and some plant sources.
Figure 1. Piperine’s chemical structure and some plant sources.
Cancers 15 05488 g001
Figure 2. Chemopreventive blocking and suppressing agents interfere with the metabolic conversion of procarcinogens to their ultimate intermediates, where the key steps in carcinogenesis were 1. Initiation, referring to the transformation of a normal cell into an initiated cell, 2. Promotion, leading to the formation of preneoplastic cells, 3. Progression, advancing to neoplastic cells, and finally, 4. Angiogenesis and Metastasis, where VEGFR promotes the formation of new blood vessels and the migration of cancer cells to other organs. At the same time, MMPs facilitate these processes by remodeling tissues and breaking down barriers. The pleiotropic effects of piperine “P” are also briefly illustrated.
Figure 2. Chemopreventive blocking and suppressing agents interfere with the metabolic conversion of procarcinogens to their ultimate intermediates, where the key steps in carcinogenesis were 1. Initiation, referring to the transformation of a normal cell into an initiated cell, 2. Promotion, leading to the formation of preneoplastic cells, 3. Progression, advancing to neoplastic cells, and finally, 4. Angiogenesis and Metastasis, where VEGFR promotes the formation of new blood vessels and the migration of cancer cells to other organs. At the same time, MMPs facilitate these processes by remodeling tissues and breaking down barriers. The pleiotropic effects of piperine “P” are also briefly illustrated.
Cancers 15 05488 g002
Figure 3. Piperine induces various types of cell death: apoptosis via caspase 3 activation, ferroptosis via Fenton reaction, autophagy by inhibiting mTORC signaling, and anoikis via STAT3 inhibition.
Figure 3. Piperine induces various types of cell death: apoptosis via caspase 3 activation, ferroptosis via Fenton reaction, autophagy by inhibiting mTORC signaling, and anoikis via STAT3 inhibition.
Cancers 15 05488 g003
Figure 4. Piperine inhibits angiogenesis and metastasis by reducing the expression of vascular endothelial growth factor (VEGF) and down-regulating matrix metalloproteinase (MMP). Additionally, it hinders the self-renewal capacity of cancer stem cells (CSCs) via both direct and indirect mechanisms, targeting critical pathways such as Wnt/β-catenin, Hedgehog, and Notch. Moreover, piperine induces cell cycle arrest at various phases, including G1, G1/S, or G2/M.
Figure 4. Piperine inhibits angiogenesis and metastasis by reducing the expression of vascular endothelial growth factor (VEGF) and down-regulating matrix metalloproteinase (MMP). Additionally, it hinders the self-renewal capacity of cancer stem cells (CSCs) via both direct and indirect mechanisms, targeting critical pathways such as Wnt/β-catenin, Hedgehog, and Notch. Moreover, piperine induces cell cycle arrest at various phases, including G1, G1/S, or G2/M.
Cancers 15 05488 g004
Figure 5. Enhancing radiation therapy while minimizing side effects: piperine’s impact on oxidative stress, DNA damage, NF-κB activation, and cell cycle regulation in cancer and normal cells.
Figure 5. Enhancing radiation therapy while minimizing side effects: piperine’s impact on oxidative stress, DNA damage, NF-κB activation, and cell cycle regulation in cancer and normal cells.
Cancers 15 05488 g005
Figure 6. Optimizing anticancer drug bioavailability with piperine: inhibition of the up-regulation of genes (e.g., MDR1, MRP1, BCRP) and cellular proteins (e.g., P-gp and cytochrome P450) and activation of ABC Transporters.
Figure 6. Optimizing anticancer drug bioavailability with piperine: inhibition of the up-regulation of genes (e.g., MDR1, MRP1, BCRP) and cellular proteins (e.g., P-gp and cytochrome P450) and activation of ABC Transporters.
Cancers 15 05488 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Benayad, S.; Wahnou, H.; El Kebbaj, R.; Liagre, B.; Sol, V.; Oudghiri, M.; Saad, E.M.; Duval, R.E.; Limami, Y. The Promise of Piperine in Cancer Chemoprevention. Cancers 2023, 15, 5488. https://doi.org/10.3390/cancers15225488

AMA Style

Benayad S, Wahnou H, El Kebbaj R, Liagre B, Sol V, Oudghiri M, Saad EM, Duval RE, Limami Y. The Promise of Piperine in Cancer Chemoprevention. Cancers. 2023; 15(22):5488. https://doi.org/10.3390/cancers15225488

Chicago/Turabian Style

Benayad, Salma, Hicham Wahnou, Riad El Kebbaj, Bertrand Liagre, Vincent Sol, Mounia Oudghiri, El Madani Saad, Raphaël Emmanuel Duval, and Youness Limami. 2023. "The Promise of Piperine in Cancer Chemoprevention" Cancers 15, no. 22: 5488. https://doi.org/10.3390/cancers15225488

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop