Review
The potential use of natural products to negate hepatic, renal and neuronal toxicity induced by cancer therapeutics

https://doi.org/10.1016/j.bcp.2019.06.007Get rights and content

Abstract

Different types of chemotherapeutics are used for cancer treatment. These drugs act on several signal pathways, lead to programmed cell death, and damage cancer cells. Although many specific mechanisms of action have been suggested for chemotherapeutics, there are still gaps in understanding their effects. They may affect different components of the cell, particularly proteins with specific functions, such as enzymes. Recently, targeted and immuno therapies were introduced for treatment of different cancers. However, many cancer patients still depend on traditional and well-known drugs. Doxorubicin and platinum-based drugs are among the most frequently used chemotherapeutics. They are highly cytotoxic for cancer cells, but they also act on healthy cells. Hence, it is crucial to understand the mechanisms involved in order to decrease their side effects. Natural products, many of which are also available over-the-counter, may be considered to decrease various cancer drug-induced side effects. This review focuses on the use of these compounds to overcome side effects of chemotherapeutics, primarily doxorubicin and cisplatin, in the liver, kidney, and neuronal systems.

Introduction

Cancer therapies that are routinely used for the treatment of different types of cancer vary in their potency. Challenges to successful treatment include not only limited efficacy, but also acquired drug resistance and organ toxicity. Organ toxicity is not limited to chemotherapy, it also occurs in targeted- and immuno-therapy. Since toxicity associated with chemotherapeutic agents is more prevalent, in this review we will focus on this critical barrier against effective treatment and deal with side effects of doxorubicin and cisplatin in the liver, kidney, and neuronal systems.

Doxorubicin (DOX) and platinum-based drugs are among the most abundantly used chemotherapeutics. However, although they are highly cytotoxic for cancer cells, their organ cytotoxicity limits their utilization. Therefore, it is critical to understand their mechanisms of action in order to decrease their side effects. Particularly the use of natural compounds to overcome the side effects of DOX and platinum-based drugs seems to be important. DOX is an anthracycline antibiotic and a broadspectrum antineoplastic agent. It is commonly used in tumor therapy of uterine, ovarian, breast, and lung cancers, solid tumors of childhood, Hodgkin and non-Hodgkin lymphomas, and soft tissue sarcomas as well as other cancer types [1], [2]. Following its discovery in Streptomyces peucetius in the early 1960s, DOX presented a considerable leap forward in the fight against cancer. DOX antitumor effects include mechanisms of DNA alteration and the production of free radicals [3], [4]. DOX interacts with DNA by intercalation and inhibition of macromolecular biosynthesis, which inhibits the progression of the enzyme topoisomerase II (Top2) [5], [6]. Top2 is the target of a wide array of anticancer agents: the anthracyclines (e.g., DOX, epirubicin, daunorubicin), the anthracenediones (e.g., mitoxantrone), and the podophyllotoxins (e.g., etoposide, teniposide) [7]. These agents act by blocking the process of replication, since the transient double strand breaks introduced by Top2 are crucial for replication, transcription, and chromosome segregation [8].

The clinical usefulness of DOX in long-term treatment is restricted, however, due to serious side effects. These can be acute and chronic, and include dose-dependent myocardial injury, which can lead to congestive heart failure [9]. In up to 26% of patients [10], DOX administration leads to arrhythmia or cardiomyopathy induced by the formation of reactive oxygen species and cytochrome c release from mitochondria [11]. It was also confirmed that therapeutic doses of DOX enhance lipid peroxidation in microsomes and mitochondria in the liver, especially in the presence of Fe3+ ions [12]. Even applications of a single dose of DOX have been proven to cause irreversible liver damage and an increase of the apoptotic processes in hepatic tissue [13], [14]. Other tissues like the kidneys, brain, and the skeletal muscle are also affected negatively by DOX [15].

Another group of chemotherapeutic agents for the treatment of cancer patients are platinum-based drugs. The anti-cancer activity of cis-diamminedichloroplatinum (II) (cis-[PtCl2(NH3)2], CDDP, Cisplatin) was first reported in 1979, more than 40 years ago. Cisplatin and other platinum-based drugs have been routinely employed in the treatment of testicular, ovarian, cervical, bladder, and head/neck tumors [16]. The mechanism of action of cisplatin is based on the intrastrand cross-linking of the cis-Pt (NH3)2 unit to cellular DNA at two neighboring guanine bases [17] and the consequent induction of cellular apoptosis. While effective, the use of these platinum-based drugs (cisplatin, carboplatin, and oxaliplatin) is also limited by their severe, dose-limiting side effects. The dose-limiting side effect for cisplatin is nephrotoxicity, for carboplatin it is myelosuppression, and for oxaliplatin it is neurotoxicity. Patients are commonly co-prescribed nonchemotherapy-based drugs to treat these side effects, but with limited success [18].

Section snippets

Hepatotoxicity of cancer therapeutics

Therapeutic doses of DOX (1 mg/kg) cause massive hepatotoxicity in rats, manifesting on a structural level in the form of dissolution of hepatic cords, focal inflammation, and appearance of necrotic tissues [19]. Lower doses (0.2 mg/kg) also cause abnormal changes in the form of periportal fibrosis, degeneration of hepatic cords, and increased apoptosis. DOX causes pathological changes in hepatocytes’ ultrastructure, including an increase in mitochondrial vacuolization, atrophied mitochondria

Nephrotoxicity of cancer therapeutics

Nephrotoxicity refers to side effects that damage kidney function associated with filtration, reabsorption, and excretion. Nephrotoxicity of chemotherapeutic agents remains a significant impediment to the successful treatment of malignant diseases [32], not least because it is cumulative and only partially reversible. It is estimated that approximately 50–60% of patients undergoing cancer chemotherapy acquire acute kidney injury associated with increased morbidity and mortality rates [33], [34]

Neurotoxicity of cancer therapeutics

Neurotoxicity developing during cancer treatment is a crucial problem that can cause discontinuation and dose reduction of the therapy. It may be associated with both the central and peripheral nervous system. Peripheral neuropathy is the common side effect of many chemotherapeutic agents, hence the term chemotherapy induced peripheral neuropathy (CiPN). CiPN begins with the degeneration of peripheral nerves [47]. Primary symptoms include weakness, tingling, paraesthesias, dysaesthesias, and

Compounds that alleviate side effects of chemotherapeutics

We have searched and compiled studies with natural products reducing chemotherapy-induced hepatotoxicity, nephrotoxicity and neurotoxicity in humans and in different experimental models, as well as clinical trials with natural products to overcome chemotherapy-induced peripheral neuropathy. The results are summarized in Table 1 (hepatotoxicity), Table 2 (nephrotoxicity) and Table 3 (neurotoxicity).

Conclusion

Organ toxicity is associated with most cancer therapeutics. Eliminating or reducing such toxicity is crucial for treatment success. Several strategies were adopted to minimize cancer therapy-induced cytotoxicity with limited success. Phytochemicals were shown to exhibit pharmacological activity in different systems suggesting their potential for cancer therapy. In this review, we discussed the potential of several natural compounds and phytochemicals to ameliorate chemotherapy-induced

Declaration of Competing Interest

No conflict of interest.

Acknowledgements

This article/publication is based upon work from COST Action CA16112 NutRedOx supported by COST (European Cooperation in Science and Technology). For further information see www.cost.eu.

References (146)

  • M. Yagmurca et al.

    Protective effects of erdosteine on doxorubicin-induced hepatotoxicity in rats

    Arch. Med. Res.

    (2007)
  • K.D. Jhaveri et al.

    Renal effects of novel anticancer targeted therapies: a review of the Food and Drug Administration Adverse Event Reporting System

    Kidney Int.

    (2016)
  • V. Launay-Vacher et al.

    Renal effects of molecular targeted therapies in oncology: a review by the Cancer and the Kidney International Network (C-KIN)

    Ann. Oncol.

    (2015)
  • T. Karasawa et al.

    An integrated view of cisplatin-induced nephrotoxicity and ototoxicity

    Toxicol. Lett.

    (2015)
  • P. Mukhopadhyay et al.

    Cannabinoid-2 receptor limits inflammation, oxidative/nitrosative stress, and cell death in nephropathy

    Free Radic. Biol. Med.

    (2010)
  • G. Cavaletti et al.

    Current status and future prospects for the treatment of chemotherapy-induced peripheral neurotoxicity

    Eur. J. Cancer

    (2002)
  • A. Areti et al.

    Oxidative stress and nerve damage: role in chemotherapy induced peripheral neuropathy

    Redox Biol.

    (2014)
  • A.A. Argyriou et al.

    Chemotherapy-induced peripheral neurotoxicity (CIPN): an update

    Crit. Rev. Oncol./Hematol.

    (2012)
  • G. Cavaletti et al.

    Experimental peripheral neuropathy induced in adult rats by repeated intraperitoneal administration of taxol

    Exp. Neurol.

    (1995)
  • J. Cliff et al.

    The molecular genetics of chemotherapy–induced peripheral neuropathy: a systematic review and meta-analysis

    Crit. Rev. Oncol./Hematol.

    (2017)
  • L. Sung et al.

    Serial controlled N-of-1 trials of topical vitamin E as prophylaxis for chemotherapy-induced oral mucositis in paediatric patients

    Eur. J. Cancer

    (2007)
  • L. Tan et al.

    In vitro study on the individual and synergistic cytotoxicity of adriamycin and selenium nanoparticles against Bel7402 cells with a quartz crystal microbalance

    Biosens. Bioelectron.

    (2009)
  • B. Wang et al.

    NAD(+) administration decreases doxorubicin-induced liver damage of mice by enhancing antioxidation capacity and decreasing DNA damage

    Chem. Biol. Interact.

    (2014)
  • M.A. Feitelson et al.

    Sustained proliferation in cancer: mechanisms and novel therapeutic targets

    Semin. Cancer Biol.

    (2015)
  • R. Rezaee et al.

    Curcumin: a potentially powerful tool to reverse cisplatin-induced toxicity

    Pharmacol. Res.

    (2017)
  • C. Zhu et al.

    Hesperetin protects against H2O2-triggered oxidative damage via upregulation of the Keap1-Nrf2/HO-1 signal pathway in ARPE-19 cells

    Biomed. Pharmacother.

    (2017)
  • M. Kumar et al.

    The renoprotective activity of hesperetin in cisplatin induced nephrotoxicity in rats: molecular and biochemical evidence

    Biomed. Pharmacother.

    (2017)
  • Y. Li et al.

    Activation of sirtuin 3 by silybin attenuates mitochondrial dysfunction in cisplatin-induced acute kidney injury

    Front. Pharmacol.

    (2017)
  • B.D. Sahu et al.

    Naringin ameliorates gentamicin-induced nephrotoxicity and associated mitochondrial dysfunction, apoptosis and inflammation in rats: possible mechanism of nephroprotection

    Toxicol. Appl. Pharmacol.

    (2014)
  • V. Karuppagounder et al.

    Naringenin ameliorates daunorubicin induced nephrotoxicity by mitigating AT1R, ERK1/2-NFkappaB p65 mediated inflammation

    Int. Immunopharmacol.

    (2015)
  • Y. Li et al.

    Quercetin, inflammation and immunity

    Nutrients

    (2016)
  • M.H. Park

    Quercetin-induced downregulation of phospholipase D1 inhibits proliferation and invasion in U87 glioma cells

    Biochem. Biophys. Res. Commun.

    (2011)
  • C. Spagnuolo et al.

    Genistein and cancer: current status, challenges, and future directions

    Adv. Nutr.

    (2015)
  • M.J. Sung et al.

    Genistein protects the kidney from cisplatin-induced injury

    Kidney Int.

    (2008)
  • B.-J. Cho et al.

    Berberine inhibits the production of lysophosphatidylcholine-induced reactive oxygen species and the ERK1/2 pathway in vascular smooth muscle cells

    Mol. Cells

    (2005)
  • J. Li et al.

    Hepatoprotective effects of berberine on liver fibrosis via activation of AMP-activated protein kinase

    Life Sci.

    (2014)
  • Y.Y. Hsu et al.

    Berberine, a natural antidiabetes drug, attenuates glucose neurotoxicity and promotes Nrf2-related neurite outgrowth

    Toxicol. Appl. Pharmacol.

    (2013)
  • R. Domitrovic et al.

    Berberine exerts nephroprotective effect against cisplatin-induced kidney damage through inhibition of oxidative/nitrosative stress, inflammation, autophagy and apoptosis

    Food Chem. Toxicol.

    (2013)
  • C.Y. Zhang et al.

    Genipin inhibits UCP2-mediated proton leak and acutely reverses obesity- and high glucose-induced beta cell dysfunction in isolated pancreatic islets

    Cell Metab.

    (2006)
  • G. Minotti et al.

    Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity

    Pharmacol. Rev.

    (2004)
  • G.N. Hortobagyi

    Anthracyclines in the treatment of cancer. An overview

    Drugs

    (1997)
  • J.L. Quiles et al.

    Olive Oil and Mitochondrial Oxidative Stress: Studies on Adriamycin Toxicity, Physical Exercise and Ageing, Olive Oil and Health

    (2006)
  • F.A. Fornari et al.

    Interference by doxorubicin with DNA unwinding in MCF-7 breast tumor cells

    Mol. Pharmacol.

    (1994)
  • R.L. Momparler et al.

    Effect of adriamycin on DNA, RNA, and protein synthesis in cell-free systems and intact cells

    Cancer Res.

    (1976)
  • S.W. Langer

    Dexrazoxane for the treatment of chemotherapy-related side effects

    Cancer Manage. Res.

    (2014)
  • J.L. Nitiss

    DNA topoisomerase II and its growing repertoire of biological functions

    Nat. Rev. Cancer

    (2009)
  • P.K. Singal et al.

    Doxorubicin-induced cardiomyopathy

    N. Engl. J. Med.

    (1998)
  • O. Tacar et al.

    Doxorubicin: an update on anticancer molecular action, toxicity and novel drug delivery systems

    J. Pharm. Pharmacol.

    (2013)
  • A. Pedrycz et al.

    Increased apoptosis in the adult rat liver after a single dose of adriamycin administration

    Ann. Univ. Mariae Curie-Sklodowska. Sectio D: Med.

    (2004)
  • A. Pedrycz et al.

    Ultrastructural and immunohistochemical evaluation of apoptosis in foetal rat liver after adriamycin administration

    Bull. Vet. Inst. Pulawy

    (2005)
  • Cited by (26)

    • Renoprotective activity of Ribes diacanthum pall (RDP) against inflammation in cisplatin-stimulated mice model and human renal tubular epithelial cells

      2022, Journal of Ethnopharmacology
      Citation Excerpt :

      In view of the irreplaceable clinical status of cisplatin, researchers have made numerous explorations and kept seeking for effective drugs to alleviate its nephrotoxicity. In recent years, traditional herbal medicines have attracted more and more attention from scholars at home and abroad to investigate potential therapeutic agents against CDDP-induced nephrotoxicity (Ridzuan et al., 2019; Prša et al., 2020). Of note, those natural products rich in flavonoids, which have antioxidant and anti-inflammatory effects, have been demonstrated to exert renoprotective actions in diseases such as glomerulonephritis, diabetic nephropathy, and chemically-induced kidney insufficiency (Vargas et al., 2018).

    • Targeting cancer using phytoconstituents-based drug delivery

      2021, Advanced Drug Delivery Systems in the Management of Cancer
    View all citing articles on Scopus
    1

    These two authors contributed equally to the manuscript.

    View full text