Fenretinide in Cancer and Neurological Disease: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Cancer and neurodegeneration share leadership as causes of morbidity and death worldwide. They can be thought as disease mechanisms at opposite ends: while in neurodegeneration, induction of inflammatory genes and suppression of cell-cycle genes are the prominent signals; the opposite happens in cancer. Fenretinide (all-trans-N-(4-hydroxyphenyl) retinamide, 4-HPR) is a synthetic derivative of all-trans-retinoic acid initially proposed in anticancer therapy for its antitumor effects combined with limited toxicity. Currently, it is also studied in many other diseases for its ability to influence several biological pathways and provide a broad spectrum of pharmacological effects.

  • fenretinide
  • anticancer drug
  • nanomicellar formulations
  • repositioning
  • neuroprotection
  • hormesis

1. Fenretinide in Cancer

Typical retinoids induce antitumor effects mainly by decreasing the expression of anti-apoptotic bcl-2 family genes binding retinoic acid receptors (RAR) and retinoid X receptors (RXR): RXR-alpha, RXR-beta and RXR-gamma, which are ligand-activated transcription factors regulating several cellular processes including growth, differentiation and apoptosis [1].
The antitumor effect of fenretinide is more complex and encompasses at least four different mechanisms of action (Figure 1).
Figure 1. Schematic diagram of fenretinide activity in cancer. Pointed arrows represent pathway activation, and blunt arrows represent pathway inhibition. Fenretinide activates PKCδ in the mitochondrial signalosome with consequent activation of PDK2, PDHC, TCA and increased ROS production. High increase of ROS activates the p38 apoptotic pathway with CytC release, caspase-9 activation and induction of apoptosis. Moderate increase of ROS activates the JNK autophagy pathway.Fenretinide induces translocation of RARβ-Nur77 dimer from the nucleus to the cytoplasm where Nur77 binds Bcl-2 allowing their conformational change towards pro-apoptotic structures. Fenretinide inhibition of DES1 increases DhCer/Cer ratio and triggers endoplasmic reticulum stress with blocking of the PI3K/AKT/mTOR and NF-KB signaling pathways, as well as with blocking of eIF2 phosphorylation due to PERK. Moreover, fenretinide inhibits mTORC2 and mTORC1 by structural affinity with mTOR-ATP binding site.
Firstly, fenretinide selectively binds the RARβ receptors, increasing their expression and inducing translocation of the nuclear receptor Nur77, as a dimer with RARβ, from the nucleus to the cytoplasm. In the cytoplasm, Nur77 binds Bcl-2, allowing their conformational change towards pro-apoptotic structures that expose the BH3 domain [2][3][4][5].
In human neuroblastoma cells, fenretinide induced apoptosis by a caspase-dependent mechanism. Cell treatment with RARβ/γ antagonists inhibited apoptosis, while inhibitors of RARα had no effect [6].
The second mechanism is DES1 inhibition, which produces an increased DhCer/Cer ratio in cell membranes. This causes endoplasmic reticulum stress and a consequent block of the phosphatidylinositol-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) and NF-kB signaling pathways. In addition, the endoplasmic reticulum (ER) stress induces phosphorylation of the a-subunit of eukaryotic initiation factor 2 (eIF2) due to the protein kinase RNA-like endoplasmic reticulum kinase (PERK), which therefore can no longer carry out its activity as an initiator of protein translation [7].
In cancer cells, fenretinide increased DhCer levels in a dose-dependent manner, whereas no significant changes were seen in Cers content [8]. In prostate cancer cells exposed to fenretinide, DhCers promoted autophagy by inducing the formation of autophagosomes [9]; although autophagy can act as a pro-survival mechanism, it can also promote cell death and excessive autophagic activity can lead to digestion of the entire cell [10].
The third mechanism is the inhibition of mTOR. The molecular structure of fenretinide matches the binding site of mTOR with ATP, suppressing the activities of both the mTORC1 and the mTORC2 complexes and the related PI3K/AKT pathway; as a consequence, a proliferation decrease is obtained by inhibition of the PI3K/AKT pathway and the translational regulation activity of mTORC2. In vitro studies confirmed that fenretinide inhibits non-small-cell lung cancer cells growth attenuating mTOR downstream signaling. Moreover, knockdown of mTOR in cancer cells decreased their sensitivity to fenretinide [11].
The fourth mechanism is based on the ability of fenretinide to replace retinol in the mitochondrial signalosome, composed of a signal adaptor protein (p66Shc), protein kinase Cδ (PKCδ) and cytochrome c (CytC). The retinol/p66Shc-PKCδ-CytC signalosome behaves as a catalyst for the passage of electrons from PKCδ to CytC and vice versa. The electron donated by PKCδ to cytochrome oxidizes the zinc-finger domain of the kinase, resulting in a structural modification which exposes the binding site to the substrate and the binding site of ATP. This activation induces, in turn, the activation of pyruvate dehydrogenase kinase 2 (PDK2) through phosphorylation, which allows pyruvate dehydrogenase phosphatase 1 (PDP1) and 2 (PDP2) to activate the pyruvate dehydrogenase complex (PDHC) that feeds the tricarboxylic acid cycle (TCA). This makes NADH available for the oxidative phosphorylation (OXPHOS) where the electron transport chain generates ROS and ATP.
The electron flow generated by this mechanism is reversible only in the presence of physiological concentrations of retinol that allow the signalosome to switch from activation to inactivation depending on the energy demand of the cell; on the contrary, when retinol is replaced by fenretinide or retinoic acid, the mechanism is not reversible anymore and PDHC is locked in an activation state leading to ROS overproduction (Figure 1).
Cellular ROS increase is one of the triggering signals for the intrinsic pathway of apoptosis, leading to the increase in mitochondrial membrane permeability and release of pro-apoptotic proteins in the cytoplasm (such as CytC and the direct inhibitor of apoptosis-binding protein with low pI DIABLO) leading to cell death via caspase activation [12].
In tumor cells, apoptosis is activated only in the presence of high ROS levels, while low levels activate autophagy. This depends on DJ-1, a redox sensor protein that detects ROS concentration and determines the cellular response. Under moderate ROS increase, the mild oxidized form of DJ-1 links and inactivates the apoptosis signal-regulating kinase 1 (ASK1) in the p-38 apoptotic pathway, thus blocking apoptosis, but it does not interfere with ASK1 in the c-Jun N-terminal kinase (JNK) autophagy pathway; in this way, apoptosis is suppressed and cell viability is maintained by autophagy. On the contrary, in the presence of high ROS levels, perceived as lethal by the cell, an excessive oxidation of DJ-1 induces its dissociation from ASK1 with consequent activation of the p38 apoptotic pathway [13] involving CytC release, caspase-9 activation and apoptosis. The fine regulation of cell response to ROS increase is responsible for the different effect of fenretinide in tumors vs normal cells and in different tumor types.
Indeed, a cell response such as autophagy (causing cell survival) or apoptosis (causing cell death) is strictly dependent on the threshold of cell tolerance towards ROS increase that changes in different cell types and conditions. However, as a rule, high fenretinide concentrations are expected to promote apoptosis by induction of high ROS levels, that activate the p-38 apoptotic pathway and, by inhibition of PI3K/AKT/mTORC, DES1 and RARβ-Nur77 pathways that block proliferation and further stimulate apoptosis.
Conversely, low fenretinide concentrations are expected to favor cell survival over cell death due to the moderate increase of ROS production, which activates the JNK autophagy pathway. In addition, low fenretinide concentrations are not expected to inhibit the PI3K/AKT/mTORC, DES1 and RARβ-Nur77 proliferation and apoptosis-controlling pathways. This relationship between concentration and type of response has been observed in glioma cell lines in which fenretinide induced apoptosis at concentrations of 10 μM and autophagy at 5 μM; at concentrations higher than 10 μM, the glioma cells undergoing apoptosis also showed characteristic features of autophagy [14].

1.1 Clinical and Preclinical Evaluation of Fenretinide in Cancer

The activity of fenretinide has been demonstrated in vitro in a wide range of cancer cells. A minimum concentration of 10 µM has been required to provide 50–90% cell growth inhibition and apoptosis after 72 h of exposure [15].
Clinical trials of fenretinide to date have shown high variability in results, indicating that the minimum active concentration is not always achieved in the tumor mass.
As is well-known, the achievement of an active drug concentration in solid tumors depends mainly on the permeability of the tumor to drug penetration and the drug plasma concentration that controls blood-tumor drug distribution.
Low aqueous solubility has often been a major obstacle to the development and clinical use of antitumor drugs, as their administration by any route has frequently failed to provide adequate drug plasma concentrations for activity.
Fenretinide is a molecule characterized by very low aqueous solubility and high lipophilicity, which severely limits its bioavailability and provides low plasma concentrations that are often insufficient to allow drug penetration into the solid tumor at levels required for activity.
For drugs with low aqueous solubility, such as fenretinide, the formulation may become a means of increasing drug solubilization in plasma and body fluids and/or controlling the drug distribution among different body compartments, thus improving overall activity.
Clinical trials conducted so far on fenretinide have mainly used oral soft gelatin capsules consisting of a fenretinide dispersion in corn oil and polysorbate 80, which have been the most widely used formulations in clinical trials so far.
In a phase I trial, the capsules were administered to children with neuroblastoma, in a range of doses from 100 to 4000 mg/m2 (single daily dose) for 28 days followed by a seven-day drug holiday. After the first cycle (day 28), fenretinide peak plasma concentrations ranged from 1.3 to 12.9 µM in a dose-dependent manner; no complete or partial responses were observed, but 77% of the patients experienced stable disease for a median of 23 months [16]. A pharmacokinetic study with the fenretinide capsules, at the same doses and schedule, reported average plasma drug concentrations of 9.9 µM at the highest dose [17].
Doses from 350 to 3300 mg/m2 (divided twice or thrice daily administrations) for days 1 to 7 every three weeks provided 1 complete response and 13 stable disease responses out of 30 evaluable neuroblastoma patients who completed at least eight cycles. The maximum tolerated dose (2475 mg/m2) provided a fenretinide plasma concentration of 9.9 µM at the end of the first cycle [18].
In a phase II study, patients with renal cell carcinoma were treated with 1800 mg/m2 (divided twice daily) of fenretinide capsules and minimal activity was observed in 18 evaluable patients. The intratumor concentrations of fenretinide, in tumor biopsies from four patients, were within 3.6–7.9 µM, thus under the 10 µM threshold for activity [19].
The same dosage and schedule with fenretinide capsules failed to produce objective responses in patients with small-cell lung cancer (for 7 days every three weeks, with a mean plasma concentration of fenretinide of 7.4 µM on day 7 of cycle 1) [20], whereas patients showed limited activity against prostate cancer administered at 900 mg/m2 (twice daily) [21][22].
The capsule formulation failed to show beneficial effects in patients with recurrent malignant glioma [23] and ascitic ovarian cancer [24]; thus, trials were stopped due to lack of efficacy. In an ovarian cancer phase II study, 42% of patients treated with fenretinide capsules (1800 mg/m2 twice daily) showed stable disease for a median duration of 7.2 months with fenretinide plasma concentrations ranging from 3.1 to 12.5 µM and no objective responses. However, progression-free survival and overall survival improved in patients with the highest fenretinide concentration levels [25].
Again, in a phase II study, 31 patients with breast cancer or melanoma, treated daily (from 10 to 300 days) with fenretinide capsules, showed partial responses [26].
The above-mentioned studies showed that the highest average plasma concentration of 9.9 µM was obtained by repeated administration of the fenretinide capsules at the drug dose of 2475 mg/m2 for 7 days. This value approached 10 µM, the minimum concentration providing activity in in vitro studies.
Other fenretinide formulations were able to raise the drug plasma concentrations over the values achieved with the gelatin capsules, generating superior clinical responses.
A formulation based on fenretinide mixed with a lipid matrix called LYM-X-SORB™ and dispersed in a liquid nutritional drink was evaluated in a phase I study in patients with neuroblastoma at doses ranging from 352 to 2210 mg/m2 (divided twice daily) for seven days every three weeks. Out of 29 evaluable patients, 4 had complete responses and 6 had stable disease; fenretinide mean peak plasma levels reached 21 µM at 1700 mg/m2/day on day 6 of cycle 1 [27].
An intravenous lipid emulsion containing fenretinide (infused in continuo at 905–1414 mg/m2/day for 5 days in 21-day cycles) was administered in 23 patients with advanced solid tumors enrolled in a phase I study; fenretinide plasma steady-state concentrations were in the range 17.87 µM–38 µM and although no patients had objective responses, five patients showed stable disease as best response [28].
More recently, novel nanoformulations were prepared to improve fenretinide bioavailability. They were based on phospholipid–liquid triglyceride mixtures with or without cyclodextrins. In an aqueous environment, they were able to self-assemble with formation of nanomicelle-like structures that entrapped fenretinide, thus improving the drug’s aqueous solubilization. Administered by oral or intravenous routes, these nanoformulations strongly increased fenretinide plasma concentrations and provided significant antitumor activity in preclinical studies.
Among them, a nanoformulation, called bionanofenretinide (Bio-nFeR), provided drug plasma concentrations and tumor concentrations tenfold higher than those obtained with the gelatin capsules at the same administration dose. It was obtained by drug encapsulation in an ion-pair stabilized lipid matrix. Administered by oral route in animal models of human lung cancer, colon cancer and melanoma at doses 10–200 mg/kg, Bio-nFeR significantly decreased tumor growth. A single treatment at 100 mg/kg provided 9.2 µM drug plasma concentration and 9.6 µM drug concentration in tumors. The gelatin capsules at the same administered dose provided 1.0 µM plasma concentration and 1.5 µM tumor concentration [29]. Another nanoformulation, obtained by phospholipid–liquid triglyceride and cyclodextrins, was administered, in combination with lenalidomide, by intravenous injection in animal models of human neuroblastoma at 30 mg/kg fenretinide three times/week for 3 weeks. The treatment provided 100% survival at the end of the experiment with 10.06 µM plasma concentrations and 54.47 µM tumor concentrations [30][31].
The same nanoformulation also showed significant therapeutic efficacy in brain tumor models (DIPG-07 and RARE-08), indicating its ability to cross the blood–brain barrier (BBB) [32][33].

2. Fenretinide in Neurological Diseases

Due to its high metabolic rate and relatively low levels of antioxidant defenses, the brain is highly sensitive to oxidative stress and, as a consequence, neurological diseases are often characterized by increased oxidative stress and reduced ability of the antioxidant system to counter free radicals [34][35]. Other specific features of neurological diseases are increased lipid peroxidation, DNA and protein oxidation [36][37].
In the central nervous system (CNS), ROS increase generates dysregulation of the inflammatory response through the activation of microglia and astrocytes and subsequent release of cytokines and inflammatory mediators that can disrupt the blood–brain barrier’s integrity; this allows leukocytes, such as T cells and macrophages, to infiltrate the CNS and further increase neuroinflammation [38][39].
The ability of fenretinide to modulate two significant features of neurodegeneration (oxidative stress and inflammatory response), combined with its high lipophilic character that allows, in suitable formulations, to cross the blood–brain barrier [23][40], suggested to several authors its potential use in treating pathological conditions of the nervous system.
The anti-inflammatory effect of fenretinide has been attributed to its ability to downregulate the production of arachidonic acid (AA), a proinflammatory omega-6 polyunsaturated fatty acid, and to increase the levels of the anti-inflammatory omega-3 polyunsaturated docosahexaenoic acid (DHA) in macrophages [41][42][43]. In fact, DHA was shown to act as an inhibitor of various toll-like receptors (TLRs) and TLR-mediated signaling pathways, including ERK1/2 [44]. Fenretinide inhibition of the ERK1/2 pathway decreased inflammatory mediators’ genes expression, including TNF, IL-6, CCL2 and CCL5, in infected or in lipopolysaccharide endotoxin (LPS)-stimulated macrophages without generation of a widespread shutdown of signaling mechanisms [45][46].
Furthermore, fenretinide was shown to decrease proinflammatory cytokine secretion by enhancing the expression of the peroxisome proliferator-activated receptor gamma (PPARγ) in macrophages with consequent inhibition of inflammatory programs of gene expression (Figure 2). Fenretinide not only increased the expression of PPARγ but also acted as a ligand for PPARγ, thus increasing its activity [47].
Figure 2. Schematic diagram of fenretinide activity in neurological diseases. Pointed arrows represent pathway activation, and blunt arrows represent pathway inhibition. Fenretinide activates PKCδ in the mitochondrial signalosome with consequent activation of PDK2, PDHC, TCA and increased ROS production. Low doses of fenretinide induce a moderate increase of intracellular ROS with activation of JNK autophagy pathway. Fenretinide decreases oxidative stress by improving the expression of the transcription factor Nrf2 that promotes the transcription of the antioxidant responsive element (ARE). Fenretinide downregulates the production of proinflammatory cytokines in macrophages by decreasing the AA/DHA ratio and enhancing the expression of PPARγ. Fenretinide binds RBP4 in place of retinol thereby decreasing circulating levels of both retinol and RBP4.
The ability of fenretinide to decrease oxidative stress relies on improved expression of the nuclear factor erythroid 2-related factor 2 (Nrf2) and its nuclear translocation that promote the transcription of Nrf2-antioxidant responsive element (ARE), as demonstrated in mouse brain endothelial cell line [48].
Moreover, the activation of autophagy, which can take place in both tumor and normal cells at subtoxic fenretinide doses (Figure 2), can represent a positive effect in neurodegeneration as autophagy, at basal levels, is a physiologic mechanism to sustain cell survival and oppose cell death.
Finally, due to its ability to decrease RBP4 serum levels and, consequentially, to modulate insulin resistance and glucose intolerance, fenretinide could have a positive impact on neurological diseases in which glucose metabolic dysfunctions represent a risk factor [49].

2.1 Preclinical evaluation of fenretinide in neurological disease

Multiple Sclerosis

In the experimental allergic encephalomyelitis (EAE) mice, low doses (3 mg/kg/day) of oral fenretinide, very far from those required to produce cytotoxic effects in tumor cells [50], were able to reduce severity of EAE symptoms [51]. Experimental autoimmune encephalomyelitis (EAE), in which susceptible mice strains are immunized with myelin basic protein (MBP), is one of the common models for relapsing/remitting multiple sclerosis (MS), an (auto)immune driven neurological disease specifically affecting the central nervous system.
The disease course induced by MBP, characterized by an acute paralytic episode and a significant amount of Wallerian degeneration in the spinal cord, was counteracted by dietary administration of fenretinide. More importantly, fenretinide reduced inflammation and demyelination and lessened the severity of neurological deficits in EAE mice even when administered after disease onset.

Spinal Cord Injury

The omega-3 (DHA) and omega-6 (AA) polyunsaturated fatty acids (PUFA), major constituents of cell membranes, are the most susceptible to lipid peroxidation of all fatty acids. AA and the DHA act as potent signalling molecules that promote or dampen, respectively, molecular pathways involved in the inflammatory process [52]

Fenretinide, as already mentioned, down-regulates the activation of the ERK1/2 mitogen-activated protein (MAP) kinase pathway and the expression of inflammatory mediators via normalization of the AA/DHA balance [45][46]. Thus fenretinide can support the health of the central nervous system due to its ability to modulate fatty acid metabolism and lipid mediators.

This mechanism has been called into question in reducing expression of pro-inflammatory genes and decreasing oxidative stress in a mice model of spinal cord injury, in which fenretinide beneficially acted on motor deficit. Oral administration of low daily doses of fenretinide (5 mg/kg/die) modulated PUFA levels in plasma and in injured CNS tissue of mice, after spinal cord contusion. A significant improvement in locomotor recovery was also recorded [53].

Amyotrophic Lateral Sclerosis

The unpublished doctoral thesis of Skinner T [78], an author of the paper of Lopez-Vales et al. [53], describes a beneficial role of the same doses of fenretinide in a faster progressive mice model of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease characterized by selective death of motor neurons causing progressive muscle atrophy and spasticity. 

In spinal cord of ALS mice chronically treated (from presyptomatic stage of the disease) with fenretinide at low oral doses (5 mg/kg) Skinner observed increased DHA/AA ratio, decrease lipid peroxidation and gliosis (the hallmarks of neuroinflammation in ALS) and a significant reduction in the expression of TNF-α and iNOS proinflammatory mediators. These histopathologic findings well correlated with an improvement in motor functions and increased survival time in affected female mice.

Very recently we demonstrated that low doses (10 mg/kg) of a new nanomicellar fenretinide formulation, administered by intraperitoneal route, were able to extended survival of female, but non male, ALS mice even when administered after the onset of motor symptoms [54]

Alzheimer Disease

Retinoids can act upon multiple Alzheimer disease-associated targets, a neurodegenerative disease currently ranked as the most common cause of dementia among older adults [55].

Retinoid deficiency is greatest in late onset Alzheimer disease (LOAD) [56] which accounts for 90% of AD cases [57]. According, in a mouse model of AD it was shown that administration of all-trans retinoic acid recovered adult neurogenesis in the hippocampus through inhibition of microglial activation [58].

In a neuron-specific human BACE1 knock-in mice, an animal model of sporadic AD, fenretinide (0.04% w/w in diet, approximately corresponding to 40–50 mg/kg/die) has been shown to reduce adiposity gain and serum leptin, a key regulator hormone in the control of body weight, metabolism, and glucose homeostasis [59].

In the same animal model Plucińska and collaborators [60] demonstrated the ability of fenretinide (chronically administered with the diet) to decrease ER stress and neuroinflammation while boosting the expression of the sAPPα, the neuro-protective fragment of the amyloid precursor protein. In these mice fenretinide significantly prevented early signs of spatial memory deficits, restored the cerebral expression of full length APP and lowered the accumulation of toxic Aβ oligomers. However, fenretinide effects on AD phenotype appeared independent on its ability to modulate the systemic glucose homeostasis in BACE1 mice, suggesting that fenretinide may affect β-site cleavage of APP via an indirect mechanisms.

Depression

Lipopolysaccharide (LPS) administration is widely used to induce depressive-like behavior in rodents as LPS-treated mice mimick human depressive symptoms with an earlier “sickness behavior” phase which includes fever, anorexia, reduction of locomotion and a decrease in social interaction, followed by a several depressive-like behaviours [61].

LPS administration elevates the immobility time that reflects a measure of behavioral despair when placing a rodent in an uncomfortable situation from which escape is impossible; very interesting, intraperitoneal injections of fenretinide (20 and 40 mg/kg) reverted this depressive behaviour. Fenretinide also reduced BBB dysfunction in LPS-induced brain injury where promoted Nrf2-dependent antioxidative signalling [48].

Nrf2 pathway is naturally activated as an endogenous compensatory mechanism [62] and treatment with compounds able to cross the BBB and activate the Nrf2 pathway have been indicated as emerging therapeutic strategies against the oxidative stress damage in central nervous system, supporting the neuroptotective effect of fenretinide which exhibits these abilities  [48]. Fenretinide can indirectly modulate antioxidant Nrf2 activity increasing β-carotene accumulation [96] that was shown to prevent neuronal loss in ROS-associated brain diseases also by Nrf2 increase [63][64]. In a phase 3 clinical trial (ClinicalTrials.gov Identifier: NCT00534898) fenretinide (5 mg/kg/die) was proposed as add-on to stable ongoing antipsychotic treatment in patients suffered from schizophrenia. 

Retinopathies

Despite its peripheral location the retina, the neural portion of the eye, represents an extension of the central nervous system, with specialized immune responses similar to those of the brain and spinal cord.

Based on its ability to reduce circulating levels of retinol, fenretinide treatment has been proposed in geographic atrophy, an advanced form of age-related macular degeneration affecting the retina in which pathogenesis excessive accumulation of retinol-based toxins has been implicated. Unlike other organs, the uptake of retinol by the eye is largely dependent on delivery by RBP complex which can be efficiently modulated by fenretinide through a rapid elimination of the complex via urine.

In a 2-year placebo-controlled double-masked trial, 100 and 300 mg of fenretinide were orally administered every days in 246 geographic atrophy patients [65]. As expected, fenretinide administration induced a dose-dependent reversible reductions in serum RBP-retinol with visual disturbance and night blindness as the most common adverse effect in patients treated with the higher doses. More importantly, limiting retinol uptake from the serum into retinal pigment epithelium has been associated with a decrease in lesion growth rates, supporting the potential of fenretinide in the management of retinopathy.

3 Conclusion

As already reported, high doses of fenretinide can activate acute responses leading to ROS increase and cell death [13], while, at subtoxic concentrations, fenretinide can stimulate adaptive stress responses [12]. Specifically, at lethal levels of oxidative stress, induced by high fenretinide concentrations, excessive oxidized DJ-1 (an oxidative stress response protein) induces p38 activation enabling the cells to commit to apoptosis. On the contrary, in the presence of mild oxidative stress, as provided by low fenretinide concentrations, mildly oxidized DJ-1 is recruited to inhibit the activity of ASK1 thus inhibiting apoptosis, maintaining cell viability by autophagy activation [13] and providing an overall antioxidant action [66].

Thus, fenretinide behaviour appears consistent with a hormetic-biphasic dose response. The hormetic behaviour of fenretinide, already reported for other carotenoids [67][68], is demonstrated by the different drug doses necessary to provide efficacy in tumours or neurological disease, respectively. 

Therefore, the activity of fenretinide as pro-apoptotic/antitumor or pro-autophagic/adjuvant strictly depends on its concentration in the pathological site (tumor or CNS tissues). This concentration being related to the dose, the route of administration and the type of formulation.

The type of formulation has proved particularly important with fenretinide as this drug is characterized by extremely low aqueous solubility and hydrophobicity that strongly limit its availability in body fluids thus preventing its clinical use.

Among several formulations proposed, drug encapsulation and nanomicelle-like formulation (based on mixtures of phospholipids-liquid triglycerides with or without cyclodextrins, able to self-assemble in an aqueous environment) have demonstrated ability to increase fenretinide bioavailability at levels providing suitable plasma-tissues biodistribution and enhanced efficacy.

Recently, chemotherapeutic drugs such as kinase inhibitors, antimetabolites, alkylating agents and antibodies have been repurposed for neuroprotection, since it was demonstrated that some signalling pathways in neurodegeneration and cancer show peculiar sharing of genes and proteins that can become mutual therapeutic targets [69]. However, the major problems with the anticancer drugs use in neurodegenerative diseases are the presence of physical barriers in the brain, such as the blood brain barrier, drug resistance and neuronal toxicity.

The independence of fenretinide from Pgp and MRP [70]  and the ability of fenretinide to cross the blood-brain barrier, due to its highly lipophilic nature, makes it a good candidate to treat different neurological diseases

The neurotoxicity of many anticancer drugs [71], that strongly restricts their repositioning in neurological disease, was not observed with fenretinide in long term administration studies as chemopreventive agent [72] and in many other studies as a chemotherapeutic drug [16][73]. Noteworthy, the ability of fenretinide to decrease retinol plasma levels by binding RBP4 can reduce the incidence of ATRA-induced neurotoxicity observed in paediatric oncological patients  [74], thus representing a safer alternative to ATRA in cancer.

Therefore, considering the several preclinical evidences, the low toxicity profile, the independence from drug efflux transporters, the ability to penetrate the brain, the new available formulations and its hormetic behaviour, fenretinide can be regarded as potential drug candidate for selected neurological diseases, thus expanding its value beyond anticancer therapy

This entry is adapted from the peer-reviewed paper 10.3390/ijms23137426

References

  1. Simeone, A.-M.; Tari, A.M. How retinoids regulate breast cancer cell proliferation and apoptosis. Experientia 2004, 61, 1475–1484.
  2. Sani, B.P.; Shealy, Y.F.; Hill, D.L. N-(4-Hydroxyphenyl)retinamide: Interactions with retinoid-binding proteins/receptors. Carcinogenesis 1995, 16, 2531–2534.
  3. Yang, H.; Bushue, N.; Bu, P.; Wan, Y.-J.Y. Induction and intracellular localization of Nur77 dictate fenretinide-induced apoptosis of human liver cancer cells. Biochem. Pharmacol. 2010, 79, 948–954.
  4. Lin, B.; Kolluri, S.K.; Lin, F.; Liu, W.; Han, Y.-H.; Cao, X.; I Dawson, M.; Reed, J.C.; Zhang, X.-K. Conversion of Bcl-2 from Protector to Killer by Interaction with Nuclear Orphan Receptor Nur77/TR3. Cell 2004, 116, 527–540.
  5. Wu, L.; Chen, L. Characteristics of Nur77 and its ligands as potential anticancer compounds (Review). Mol. Med. Rep. 2018, 18, 4793–4801.
  6. Lovat, P.E.; Ranallic, M.; Petruzzellic, M.A.; Bernassolac, F.; Piacentinide, M.; Malcolm, A.J.; Pearson, A.D.; Melinoc, G.; Redfern, C.P. Effector Mechanisms of Fenretinide-Induced Apoptosis in Neuroblastoma. Exp. Cell Res. 2000, 260, 50–60.
  7. Lachkar, F.; Ferré, P.; Foufelle, F.; Papaioannou, A. Dihydroceramides: Their emerging physiological roles and functions in cancer and metabolic diseases. Am. J. Physiol. Metab. 2021, 320, E122–E130.
  8. Kraveka, J.M.; Li, L.; Szulc, Z.M.; Bielawski, J.; Ogretmen, B.; Hannun, Y.A.; Obeid, L.; Bielawska, A. Involvement of Dihydroceramide Desaturase in Cell Cycle Progression in Human Neuroblastoma Cells. J. Biol. Chem. 2007, 282, 16718–16728.
  9. Zheng, W.; Kollmeyer, J.; Symolon, H.; Momin, A.; Munter, E.; Wang, E.; Kelly, S.; Allegood, J.C.; Liu, Y.; Peng, Q.; et al. Ceramides and other bioactive sphingolipid backbones in health and disease: Lipidomic analysis, metabolism and roles in membrane structure, dynamics, signaling and autophagy. Biochim. Biophys. Acta Biomembr. 2006, 1758, 1864–1884.
  10. Lai, W.-L.; Wong, N.-S. The PERK/eIF2α signaling pathway of Unfolded Protein Response is essential for N-(4-hydroxyphenyl)retinamide (4HPR)-induced cytotoxicity in cancer cells. Exp. Cell Res. 2008, 314, 1667–1682.
  11. Xie, H.; Zhu, F.; Huang, Z.; Lee, M.-H.; Kim, D.J.; Li, X.; Lim, D.Y.; Jung, S.K.; Kang, S.; Li, H.; et al. Identification of mammalian target of rapamycin as a direct target of fenretinide both in vitro and in vivo. Carcinogenesis 2012, 33, 1814–1821.
  12. Kim, Y.-K.; Hammerling, U. The mitochondrial PKCδ/retinol signal complex exerts real-time control on energy homeostasis. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2020, 1865, 158614.
  13. Cao, J.; Ying, M.; Xie, N.; Lin, G.; Dong, R.; Zhang, J.; Yan, H.; Yang, X.; He, Q.; Yang, B. The Oxidation States of DJ-1 Dictate the Cell Fate in Response to Oxidative Stress Triggered by 4-HPR: Autophagy or Apoptosis? Antioxid. Redox Signal. 2014, 21, 1443–1459.
  14. Tiwari, M.; Bajpai, V.K.; Sahasrabuddhe, A.A.; Kumar, A.; Sinha, R.A.; Behari, S.; Godbole, M.M. Inhibition of N-(4-hydroxyphenyl)retinamide-induced autophagy at a lower dose enhances cell death in malignant glioma cells. Carcinogenesis 2007, 29, 600–609.
  15. Cooper, J.P.; Reynolds, C.P.; Cho, H.; Kang, M.H. Clinical development of fenretinide as an antineoplastic drug: Pharmacology perspectives. Exp. Biol. Med. 2017, 242, 1178–1184.
  16. Garaventa, A.; Luksch, R.; Piccolo, M.S.L.; Cavadini, E.; Montaldo, P.G.; Pizzitola, M.R.; Boni, L.; Ponzoni, M.; DeCensi, A.; De Bernardi, B.; et al. Phase I trial and pharmacokinetics of fenretinide in children with neuroblastoma. Clin. Cancer Res. 2003, 9, 2032–2039.
  17. Formelli, F.; Cavadini, E.; Luksch, R.; Garaventa, A.; Villani, M.G.; Appierto, V.; Persiani, S. Pharmacokinetics of oral fenretinide in neuroblastoma patients: Indications for optimal dose and dosing schedule also with respect to the active metabolite 4-oxo-fenretinide. Cancer Chemother. Pharmacol. 2007, 62, 655–665.
  18. Villablanca, J.G.; Krailo, M.D.; Ames, M.M.; Reid, J.M.; Reaman, G.H.; Reynolds, C.P. Phase I trial of oral fenretinide in children with high-risk solid tumors: A report from the children’s oncology group (CCG 09709). J. Clin. Oncol. 2006, 24, 3423–3430.
  19. Vaishampayan, U.; Heilbrun, L.K.; Parchment, R.E.; Jain, V.; Zwiebel, J.; Boinpally, R.R.; LoRusso, P.; Hussain, M. Phase II trial of fenretinide in advanced renal carcinoma. Inves. New Drugs 2005, 23, 179–185.
  20. Schneider, B.J.; Worden, F.; Gadgeel, S.; Hodges, C.; Parchment, R.; Zwiebel, J.; Kraut, M.; Kalemkerian, G. Phase II study of fenretinide in patients with small cell lung cancer (SCLC) with progression after first- or second-line chemotherapy. J. Clin. Oncol. 2004, 22, 7299.
  21. Cheung, E.; Pinski, J.; Dorff, T.; Groshen, S.; Quinn, D.; Reynolds, C.P.; Maurer, B.J.; Lara, P.N.; Tsao-Wei, D.D.; Twardowski, P.; et al. Oral Fenretinide in Biochemically Recurrent Prostate Cancer: A California Cancer Consortium Phase II Trial. Clin. Genitourin. Cancer 2009, 7, 43–50.
  22. Moore, M.M.; Stockler, M.; Lim, R.; Mok, T.S.; Millward, M.; Boyer, M.J. A phase II study of fenretinide in patients with hormone refractory prostate cancer: A trial of the Cancer Therapeutics Research Group. Cancer Chemother. Pharmacol. 2010, 66, 845–850.
  23. Puduvalli, V.K.; Yung, W.A.; Hess, K.R.; Kuhn, J.G.; Groves, M.D.; Levin, V.A.; Zwiebel, J.; Chang, S.M.; Cloughesy, T.F.; Junck, L.; et al. Phase II Study of Fenretinide (NSC 374551) in Adults with Recurrent Malignant Gliomas: A North American Brain Tumor Consortium Study. J. Clin. Oncol. 2004, 22, 4282–4289.
  24. Colombo, N.; Formelli, F.; Cantù, M.G.; Parma, G.; Gasco, M.; Argusti, A.; Santinelli, A.; Montironi, R.; Cavadini, E.; Baglietto, L.; et al. A Phase I-II Preoperative Biomarker Trial of Fenretinide in Ascitic Ovarian Cancer. Cancer Epidemiol. Biomark. Prev. 2006, 15, 1914–1919.
  25. Reynolds, C.P.; Frgala, T.; Tsao-Wei, D.D.; Groshen, S.; Morgan, R.; McNamara, M.; Scudder, S.; Zwiebel, J.A.; Lenz, H.J.; Garcia, A.A. High plasma levels of fenretinide (4-HPR) were associated with improved outcome in a phase II study of recurrent ovarian cancer: A study by the California Cancer Consortium. J. Clin. Oncol. 2007, 25, 5555.
  26. Modiano, M.R.; Dalton, W.S.; Lippman, S.M.; Joffe, L.; Booth, A.R.; Meyskens, F.L., Jr. Phase II study of Fenretinide (N-retinamide) in advanced breast cancer and melanoma. Inves. New. Drugs 1990, 8, 317–319.
  27. Maurer, B.J.; Kang, M.H.; Villablanca, J.G.; Janeba, J.; Groshen, S.; Matthay, K.K.; Sondel, P.M.; Maris, J.M.; Jackson, H.A.; Goodarzian, F.; et al. Phase I trial of fenretinide delivered orally in a novel organized lipid complex in patients with relapsed/refractory neuroblastoma: A report from the new approaches to neuroblastoma therapy (NANT) consortium. Pediatr. Blood Cancer 2013, 60, 1801–1808.
  28. Thomas, J.S.; El-Khoueiry, A.B.; Maurer, B.J.; Groshen, S.; Pinski, J.K.; Cobos, E.; Gandara, D.R.; Lenz, H.J.; Kang, M.H.; Reynolds, C.P.; et al. A phase I study of intravenous fenretinide (4-HPR) for patients with malignant solid tumors. Cancer Chemother. Pharmacol. 2021, 87, 525–532.
  29. Orienti, I.; Salvati, V.; Sette, G.; Zucchetti, M.; Bongiorno-Borbone, L.; Peschiaroli, A.; Zolla, L.; Francescangeli, F.; Ferrari, M.; Matteo, C.; et al. A novel oral micellar fenretinide formulation with enhanced bioavailability and antitumour activity against multiple tumours from cancer stem cells. J. Exp. Clin. Cancer Res. 2019, 38, 373.
  30. Orienti, I.; Nguyen, F.; Guan, P.; Kolla, V.; Calonghi, N.; Farruggia, G.; Chorny, M.; Brodeur, G.M. A Novel Nanomicellar Combination of Fenretinide and Lenalidomide Shows Marked Antitumor Activity in a Neuroblastoma Xenograft Model. Drug Des. Dev. Ther. 2019, 13, 4305–4319.
  31. Orienti, I.; Farruggia, G.; Nguyen, F.; Guan, P.; Calonghi, N.; Kolla, V.; Chorny, M.; Brodeur, G.M. Nanomicellar Lenalidomide–Fenretinide Combination Suppresses Tumor Growth in an MYCN Amplified Neuroblastoma Tumor. Int. J. Nanomed. 2020, 15, 6873–6886.
  32. Upton, D.; Valvi, S.; Liu, J.; Yeung, N.; George, S.; Ung, C.; Khan, A.; Franshaw, L.; Ehteda, A.; Shen, H.; et al. DIPG-07. High throughput drug screening identifies potential new therapies for diffuse intrinsic pontine gliomas (Dipgs). Neuro Oncol. 2020, 22, iii288.
  33. Upton, D.; Valvi, S.; Liu, J.; Yeung, N.; George, S.; Ung, C.; Khan, A.; Franshaw, L.; Ehteda, A.; Shen, H.; et al. Rare-08. Potential new therapies for diffuse intrinsic pontine gliomas identified through high throughput drug screening. Neuro Oncol. 2021, 23, i42.
  34. Uttara, B.; Singh, A.V.; Zamboni, P.; Mahajan, R.T. Oxidative Stress and Neurodegenerative Diseases: A Review of Upstream and Downstream Antioxidant Therapeutic Options. Curr. Neuropharmacol. 2009, 7, 65–74.
  35. Halliwell, B. Role of Free Radicals in the Neurodegenerative Diseases. Drugs Aging 2001, 18, 685–716.
  36. Sultana, R.; Perluigi, M.; Butterfield, D.A. Protein Oxidation and Lipid Peroxidation in Brain of Subjects with Alzheimer’s Disease: Insights into Mechanism of Neurodegeneration from Redox Proteomics. Antioxid. Redox Signal. 2006, 8, 2021–2037.
  37. Sayre, L.M.; Perry, G.; Smith, M.A. Oxidative Stress and Neurotoxicity. Chem. Res. Toxicol. 2007, 21, 172–188.
  38. Owens, T.; Babcock, A.A.; Millward, J.M.; Toft-Hansen, H. Cytokine and chemokine inter-regulation in the inflamed or injured CNS. Brain Res. Rev. 2005, 48, 178–184.
  39. Solleiro-Villavicencio, H.; Rivas-Arancibia, S. Effect of Chronic Oxidative Stress on Neuroinflammatory Response Mediated by CD4+T Cells in Neurodegenerative Diseases. Front. Cell. Neurosci. 2018, 12, 114.
  40. Bassani, B.; Bartolini, D.; Pagani, A.; Principi, E.; Zollo, M.; Noonan, D.M.; Albini, A.; Bruno, A. Fenretinide (4-HPR) Targets Caspase-9, ERK 1/2 and the Wnt3a/β-Catenin Pathway in Medulloblastoma Cells and Medulloblastoma Cell Spheroids. PLoS ONE 2016, 11, e0154111.
  41. Guilbault, C.; Wojewodka, G.; Saeed, Z.; Hajduch, M.; Matouk, E.; De Sanctis, J.B.; Radzioch, D. Cystic Fibrosis Fatty Acid Imbalance Is Linked to Ceramide Deficiency and Corrected by Fenretinide. Am. J. Respir. Cell Mol. Biol. 2009, 41, 100–106.
  42. Saeed, S.; Guilbault, C.; De Sanctis, J.; Henri, J.; Marion, D.; St-Arnaud, R.; Radzioch, D. Fenretinide prevents the development of osteoporosis in Cftr-KO mice. J. Cyst. Fibros. 2008, 7, 222–230.
  43. Garić, D.; Dumut, D.C.; Shah, J.; De Sanctis, J.; Radzioch, D. The role of essential fatty acids in cystic fibrosis and normalizing effect of fenretinide. Cell Mol. Life Sci. 2020, 77, 4255–4267.
  44. Huang, S.; Rutkowsky, J.M.; Snodgrass, R.G.; Ono-Moore, K.D.; Schneider, D.A.; Newman, J.W.; Adams, S.H.; Hwang, D.H. Saturated fatty acids activate TLR-mediated proinflammatory signaling pathways. J. Lipid Res. 2012, 53, 2002–2013.
  45. Lachance, C.; Wojewodka, G.; Skinner, T.A.A.; Guilbault, C.; De Sanctis, J.B.; Radzioch, D. Fenretinide Corrects the Imbalance between Omega-6 to Omega-3 Polyunsaturated Fatty Acids and Inhibits Macrophage Inflammatory Mediators via the ERK Pathway. PLoS ONE 2013, 8, e74875.
  46. Lachance, C.; Segura, M.; Dominguez-Punaro, M.C.; Wojewodka, G.; De Sanctis, J.B.; Radzioch, D.; Gottschalk, M. Deregulated Balance of Omega-6 and Omega-3 Polyunsaturated Fatty Acids following Infection by the Zoonotic Pathogen Streptococcus suis. Infect. Immun. 2014, 82, 1778–1785.
  47. Lin, C.-H.; Lee, S.-Y.; Zhang, C.-C.; Du, Y.-F.; Hung, H.-C.; Wu, H.-T.; Ou, H.-Y. Fenretinide inhibits macrophage inflammatory mediators and controls hypertension in spontaneously hypertensive rats via the peroxisome proliferator-activated receptor gamma pathway. Drug Des. Dev. Ther. 2016, 10, 3591–3597.
  48. Li, T.; Zheng, L.-N.; Han, X.-H. Fenretinide attenuates lipopolysaccharide (LPS)-induced blood-brain barrier (BBB) and depressive-like behavior in mice by targeting Nrf-2 signaling. Biomed. Pharmacother. 2020, 125, 109680.
  49. Barone, E.; Di Domenico, F.; Perluigi, M.; Butterfield, D.A. The interplay among oxidative stress, brain insulin resistance and AMPK dysfunction contribute to neurodegeneration in type 2 diabetes and Alzheimer disease. Free Radic. Biol. Med. 2021, 176, 16–33.
  50. Asumendi, A.; Morales, M.C.; Alvarez, A.; Aréchaga, J.; Perezyarza, G. Implication of mitochondria-derived ROS and cardiolipin peroxidation in N-(4-hydroxyphenyl)retinamide-induced apoptosis. Br. J. Cancer 2002, 86, 1951–1956.
  51. Racke, M.K.; Burnett, D.; Pak, S.H.; Albert, P.S.; Cannella, B.; Raine, C.S.; McFarlin, D.E.; Scott, D.E. Retinoid treatment of experimental allergic encephalomyelitis IL-4 production correlates with improved disease course. J. Immunol. 1995, 154, 450–458.
  52. Nadjar, A.; Leyrolle, Q.; Joffre, C.; Laye, S. Bioactive lipids as new class of microglial modulators: When nutrition meets neuroimunology. Prog. Neuropsychopharmacol. Biol. Psychiatry 2016, 79, 19–26.
  53. Lopez-Vales, R.; Redensek, A.; Skinner, T.A.A.; Rathore, K.I.; Ghasemlou, N.; Wojewodka, G.; DeSanctis, J.; Radzioch, D.; David, S. Fenretinide Promotes Functional Recovery and Tissue Protection after Spinal Cord Contusion Injury in Mice. J. Neurosci. 2010, 30, 3220–3226.
  54. Orienti, I.; Armida, M.; Dobrowolny, G.; Pepponi, R.; Sollazzini, G.; Pezzola, A.; Casola, I.; Musarò, A.; Popoli, P.; Potenza, R.L. Fenretinide Beneficial Effects on Amyotrophic Lateral Sclerosis-associated SOD1G93A Mutant Protein Toxicity: In Vitro and In Vivo Evidences. Neuroscience 2021, 473, 1–12.
  55. Beheshti, S. Chapter 35—All-Trans Retinoic Acid in Alzheimer’s Disease. In Diagnosis and Management in Dementia; Martin, C.R., Preedy, V.R., Eds.; Academic Press: Cambridge, MA, USA, 2020; pp. 559–572. ISBN 9780128158548.
  56. Das, B.C.; Dasgupta, S.; Ray, S.K. Potential therapeutic roles of retinoids for prevention of neuroinflammation and neurodegeneration in Alzheimer’s disease. Neural. Regen. Res. 2019, 14, 1880–1892.
  57. Bihaqi, S.W.; Schumacher, A.; Maloney, B.; Lahiri, D.K.; Zawia, N.H. Do epigenetic pathways initiate late onset Alzheimer disease (LOAD): Towards a new paradigm. Curr. Alzheimer Res. 2012, 9, 574–588.
  58. Takamura, R.; Watamura, N.; Nikkuni, M.; Ohshima, T. All-trans retinoic acid improved impaired proliferation of neural stem cells and suppressed microglial activation in the hippocampus in an Alzheimer’s mouse model. J. Neurosci. Res. 2016, 95, 897–906.
  59. Dekeryte, R.; Hull, C.; Plucinska, K.; Khan, S.; Kamli-Salino, S.; Mody, N.; Morrice, N.; McLaughlin, C.; Gault, V.; Platt, B.; et al. Effects of Liraglutide and Fenretinide treatments on the diabetic phenotype of neuronal human BACE1 knock-in mice. Biochem. Pharmacol. 2019, 166, 222–230.
  60. Plucińska, K.; Mody, N.; Dekeryte, R.; Shearer, K.; Mcilroy, G.D.; Delibegovic, M.; Platt, B. High-fat diet exacerbates cognitive and metabolic abnormalities in neuronal BACE1 knock-in mice—partial prevention by Fenretinide. Nutr. Neurosci. 2020, 25, 719–736.
  61. O’Connor, J.C.; A Lawson, M.; André, C.; Moreau, M.; Lestage, J.; Castanon, N.; Kelley, K.W.; Dantzer, R. Lipopolysaccharide-induced depressive-like behavior is mediated by indoleamine 2,3-dioxygenase activation in mice. Mol. Psychiatry 2008, 14, 511–522.
  62. Sun, Y.; Yang, T.; Leak, R.K.; Chen, J.; Zhang, F. Preventive and Protective Roles of Dietary Nrf2 Activators Against Central Nervous System Diseases. CNS Neurol. Disord. Drug Targets 2017, 16, 326–338.
  63. Chen, P.; Li, L.; Gao, Y.; Xie, Z.; Zhang, Y.; Pan, Z.; Tu, Y.; Wang, H.; Han, Q.; Hu, X.; et al. β-carotene provides neuro protection after experimental traumatic brain injury via the Nrf2–ARE pathway. J. Integr. Neurosci. 2019, 18, 153–161.
  64. Park, H.-A.; Hayden, M.M.; Bannerman, S.; Jansen, J.; Crowe-White, K.M. Anti-Apoptotic Effects of Carotenoids in Neurodegeneration. Molecules 2020, 25, 3453.
  65. Mata, N.L.; Lichter, J.B.; Vogel, R.; Han, Y.; Bui, T.V.; Singerman, L.J. Investigation of oral fenretinide for treatment of geographic atrophy in age-related macular degeneration. Retina 2013, 33, 498–507.
  66. Takahashi, N. Antioxidant Properties of N-(4-Hydroxyphenyl)retinamide (Fenretinide). Biol. Pharm. Bull. 2000, 23, 222–225.
  67. Cho, S.; Chae, J.S.; Shin, H.; Shin, Y.; Song, H.; Kim, Y.; Yoo, B.C.; Roh, K.; Cho, S.; Kil, E.-J.; et al. Hormetic dose response to L-ascorbic acid as an anti-cancer drug in colorectal cancer cell lines according to SVCT-2 expression. Sci. Rep. 2018, 8, 11372.
  68. Shin, J.; Saini, R.K.; Oh, J.-W. Low Dose Astaxanthin Treatments Trigger the Hormesis of Human Astroglioma Cells by Up-Regulating the Cyclin-Dependent Kinase and Down-Regulated the Tumor Suppressor Protein P53. Biomedicines 2020, 8, 434.
  69. Advani, D.; Gupta, R.; Tripathi, R.; Sharma, S.; Ambasta, R.K.; Kumar, P. Protective role of anticancer drugs in neurodegenerative disorders: A drug repurposing approach. Neurochem. Int. 2020, 140, 104841.
  70. A Roundhill, E.; A Burchill, S. Detection and characterisation of multi-drug resistance protein 1 (MRP-1) in human mitochondria. Br. J. Cancer 2012, 106, 1224–1233.
  71. Pellacani, C.; Eleftheriou, G. Neurotoxicity of antineoplastic drugs: Mechanisms, susceptibility, and neuroprotective strategies. Adv. Med Sci. 2020, 65, 265–285.
  72. Rotmensz, N.; De Palo, G.; Formelli, F.; Costa, A.; Marubini, E.; Campa, T.; Crippa, A.; Danesini, G.; Grottaglie, M.D.; Di Mauro, M.; et al. Long-term tolerability of fenretinide (4-HPR) in breast cancer patients. Eur. J. Cancer Clin. Oncol. 1991, 27, 1127–1131.
  73. Sabichi, A.L.; Modiano, M.R.; Lee, J.J.; Peng, Y.M.; Xu, M.J.; Villar, H.; Dalton, W.S.; Lippman, S.M. Breast tissue accumulation of retinamides in a randomized short-term study of fenretinide. Clin. Cancer Res. 2003, 9, 2400–2405.
  74. Lanvers, C.; Reinhardt, D.; Dübbers, A.; Wagner-Bohn, A.; Creutzig, U.; Ritter, J.; Boos, J. Pharmacology of all-trans-retinoic acid in children with acute promyelocytic leukemia. Med. Pediatr. Oncol. 2003, 40, 293–301.
More
This entry is offline, you can click here to edit this entry!
Video Production Service