1. Introduction
Neuroblastoma is a highly malignant, embryonal tumor originating in the neural crest, which develops into the adrenal medulla and sympathetic nerves. The disease has a prevalence of approximately one per 7,000 live births, and 800 cases are newly diagnosed annually in the United States [
1]. The age-standardized incidence rate in children globally ranges from 4.1 to 15.8 per million population in individuals aged 0 to 14 years [
2].
Neuroblastoma is also aggressive and capable of metastasizing to the lymph nodes, bone, and bone marrow. On the other hand, some less aggressive, early onset cases sometimes regress spontaneously or undergo tumor differentiation [
3]. Among the numerous, prognostic variables are disease stage, age at diagnosis, tumor pathology, tumor cell ploidy, and
MYCN amplification status, which can help distinguish the disease’s characteristics and predict the clinical outcome [
4].
Although neuroblastoma is highly sensitive to cytotoxic chemotherapy, relapses frequently occur in high-risk patients. However, a recent treatment strategy consisting of multiagent chemotherapy, tumor excision, local radiation therapy, and mega-dose chemotherapy plus autologous stem cell transplantation (SCT) followed by anti-disialoganglioside (GD2) immunotherapy combined with tumor differentiation therapy using retinoids has achieved a five-year event-free survival rate of 56.6 % [
5,
6]. Several, basic as well as clinical studies [
7,
8,
9,
10,
11,
12,
13,
14,
15,
16] have culminated in the development of a tumor differentiation therapy using isotretinoin, a retinoid agent and an essential element of the multimodal therapy described above. The present review article discusses both the basic and clinical research into retinoids as neuroblastoma treatment, regulatory hurdles to the approval of isotretinoin as an anti-neuroblastoma in Japan, and the prospects of tumor differentiation therapy using retinoids.
3. Preclinical evaluation of retinoids for neuroblastoma treatment
As described above, neuroblastoma occasionally undergoes spontaneous regression and/or tumor cell differentiation to become a ganglioneuroma, which is a benign tumor. This discovery motivated research into the potential of cell differentiation as a treatment for neuroblastoma. The
in vitro biological effects of isotretinoin and ATRA on neuroblastoma cell lines, including cell differentiation, sustained arrest of proliferation, and apoptosis, have been reported since the early 1980’s [
7,
8,
9,
10,
11,
12]. The following is a summary of the preclinical, pharmacologic findings of isotretinoin and/or ATRA prior to the subsequent clinical studies.
Sidell, et al. first demonstrated that ATRA induced concentration-dependent morphologic differentiation and growth inhibition in a LA-N-1 neuroblastoma cell line [
7]. An ATRA dosage ranging from 10
-9 to 10
-5 M inhibited cellular proliferation in a concentration-dependent manner [
7]. The growth curve of LA-N-1 cells showed complete inhibition from 72 to 96 hours after exposure to ATRA, which continued for five days. Even after switching the medium to one without ATRA, this effect persisted at least for eight days, and the residual cell clusters displayed normal growth characteristics [
7]. RA-induced, morphological differentiation, such as the formation of neurites, was significant at ATRA 10
-6 to 10
-5 M over 48 hours and attained maximum differentiation after approximately four days. The cellular differentiation continued for seven to ten days even after removal of ATRA from the medium [
7].
A later study using seven neuroblastoma cell lines, including LA-N-2, LA-N-5, CHP134, SK-N-SH, KA, CHP100, and IMR32, confirmed these findings [
8]. ATRA successfully inhibited cell growth and induced morphological differentiation in all but one (CHP100) cell line and induced neurite formation in four cell lines (LA-N-2, LA-N-5, CHP134, KA). On the other hand, IMR32 demonstrated cellular enlargement and vacuolization while SK-N-SH transformed into large, flat, epithelial-like cells [
8]. The same study quantified and compared cellular retinoic acid-binding protein (CRABP) in LA-N-5 and CHP100. LA-N-5, the most RA-responsive cell line, contained approximately twice as much CRABP as CHP100, the least RA-responsive cell line [
8]. Haussler et al. examined five cell lines (LA-N-1, IMR32, LA-N-5, SK-N-SH, CHP100), all of which contain CRABP, and found an association between CRABP and ATRA-induced inhibition of colony formation in soft agar [
9].
Thiele et al. described a relationship between
MYCN expression and ATRA-inducing neuronal differentiation in SMS-KCNR, another neuroblastoma cell line [
10].
MYCN expression decreased within six hours of ATRA treatment and was followed by a decrease in the growth fraction (S+G2+M) of cells at 48 hours, then by morphological differentiation [
10]. A recent study by Otsuka et al. supported a previous finding of accelerated MYCN protein degradation and neuronal differentiation in
MYCN-amplified neuroblastoma cell IMR-32 following a combination treatment of peptide TNIIIA2 and ATRA [
47].
Reynolds et al. compared the growth inhibiting ability of ATRA and isotretinoin in 16 neuroblastoma cell-lines [
12] and found the efficacy of ATRA 10 μM and isotretinoin 5 μM to be almost identical, with both achieving a growth inhibition as high as 1.7 logs [
12]. Using 12 cell lines which were sensitized to RAs, another study found that a clinically achievable level of isotretinoin (5 μM) was significantly more effective than a clinically achievable level of ATRA (0.5 μM) in inhibiting cell growth [
12]. An
in vitro study assessing two courses of isotretinoin (5 μM) exposure for 14 days alternating with 14 days of rest found complete growth arrest in a
MYCN-nonamplified SMS-LHN cell line for 120 days and a
MYCN-amplified SMS-SAN cell line for up to 60 days [
12].
4. Clinical development of isotretinoin for neuroblastoma
Although both ATRA and isotretinoin were able to induce neuronal differentiation and growth arrest in neuroblastoma cell lines, isotretinoin was chosen for further clinical research in the US in part because it was better tolerated than ATRA by children [
12,
29,
38]. The results of anecdotal clinical studies of isotretinoin for neuroblastoma, including a complete remission of bone marrow metastases and a two-year remission in one patient, provided corroboration for the choice [
11].
The first, systematic, clinical evaluation of isotretinoin for neuroblastoma treatment was conducted by the Children Cancer Group (CCG), the results of which were reported by Finklestein et al. in 1992 [
13]. Twenty-nine children aged less than 21 years with recurrent or progressive neuroblastoma following conventional therapy were enrolled. Oral isotretinoin was administered in a single, daily dose of 100 mg/m
2. Two (9%) of 22 patients whose clinical response was evaluable demonstrated a positive response to isotretinoin therapy. The median overall survival was only 46 days. Safety evaluation was difficult due to the severity of the underlying neuroblastoma, which necessitated multiple, supportive treatments, including blood transfusions. Approximately 50% of the patients were hospitalized for disease-related events for an average length of five days during the first course. The remaining patients experienced no toxicity. Adverse events, which were observed in two or more patients, included nine cases of cheilitis, five cases of fissured lip, three cases of xerosis, two cases of nausea / vomiting, three cases of abdominal pain, two cases of neutropenia < 500 /μl, four cases of thrombocytopenia < 25,000 /μl, and three cases of hemoglobin < 8 g/dl. Most of these adverse events occurred during the first 28 days of therapy.
Owing to the disappointing results of the CCG study [
13], isotretinoin was thought to be most effective
in vivo against residual neuroblastoma remaining from maximal reduction of the tumor burden by mega-dose chemotherapy and SCT. At the same time, the intermittent administration schedule, which was based with an
in vitro model published by Reynolds et al. [
12], was considered to be ideal for higher dosages. Thus, a phase I clinical trial was conducted to determine the maximal tolerated dose (MTD), toxicity, and pharmacokinetics of isotretinoin in children with neuroblastoma following SCT [
14]. Fifty-one eligible patients aged 2 to 12 years received oral isotretinoin in two, equal doses daily over 14 days followed by a 14-day rest period for up to 12 courses. In total, 407 treatment courses with dose-escalation ranging from 100 to 200 mg/m
2 were evaluated for toxicity. Dose-limiting toxicities, including hepatic dysfunction, hypercalcemia, skin rash, anemia, thrombocytopenia, and vomiting, occurred in six of nine patients at 200 mg/m
2/day [
14]. The MTD and recommended dosage were determined to be 160 mg/m
2/day. All the toxicities resolved after the discontinuation of isotretinoin, and a complete response was observed in three cases of bone marrow metastasis [
14].
These promising results led to a phase 3 clinical trial, CCG-3891, with a quasi-factorial design consisting of two, sequential randomizations. The first randomization aimed to assess the superiority of high-dose chemotherapy with autologous SCT to three, continuous courses of conventional chemotherapy in terms of event-free survival (EFS); the second part aimed to determine whether maintenance treatment with isotretinoin following cytotoxic chemotherapy could further improves EFS [
15]. The isotretinoin maintenance therapy was administered in six courses at the same dosage as in the previous phase 1 trial, namely, oral isotretinoin 160 mg/m
2/day in two, equal doses daily for 14 days followed by a 14-day rest period. A comparison of the isotretinoin maintenance arm (n=130) with a no-maintenance arm (n=128) found a three-year EFS rate of 46 % and 29 %, respectively (
p=0.027). Three-year overall survival (OS) did not differ significantly between the groups at 56 % vs 50 %, respectively (
p=0.45).
The superiority in the survival rate of the isotretinoin maintenance arm was maintained in a follow-up study of the same patient cohort with an eight-year median follow-up. A trend in the improvement of five-year EFS (42 vs 31 %;
p=0.1219) and five-year OS (50 vs 39 %;
p=0.1946) was observed in patients randomly assigned to the isotretinoin maintenance arm although the difference was statistically non-significant [
16]. A subgroup analysis in the same study found that the five-year EFS rate was 50 % in patients randomly assigned to the autologous SCT and isotretinoin (n=50) group but only 20 % in patients assigned to the no-SCT and no-isotretinoin group (n=53;
p=0.0038) [
16]. Although five-year OS was not significantly higher in the patients with isotretinoin than in the patients without isotretinoin (50 % vs 39%;
p=0.1946), OS in the isotretinoin group was significantly higher on log(log(.)) transformation of the survival estimate at five years (
p=0.0006) [
16].
Following the publication of the positive results of the CCG-3891 trial, the Children’s Oncology Group (COG) in the US declared isotretinoin maintenance therapy as the standard treatment for high-risk neuroblastoma, incorporating it into the standard therapeutic regimens in subsequent American clinical trials for high-risk neuroblastoma [
5,
49,
50].
In contrast to the positive results of the CCG-3891 trial, another randomized trial of isotretinoin enrolling 175 patients from ten European countries failed to demonstrate the superiority of isotretinoin therapy. In this double-blind randomized trial, the participants received either isotretinoin 0.75 mg/kg/day or identical, placebo capsules for up to four years or until disease recurrence [
51]. Three-year EFS in the isotretinoin arm (n=88) and placebo arm (n=87) was 37 % and 42 %, respectively (
p=0.62) [
51]. Differences in the treatment effect among the various isotretinoin dosages and schedules suggested that achieving a pharmacologically effective drug level allowing a certain duration of exposure of neuroblastoma cells to the drug is necessary for isotretinoin to work as an anticancer agent. The following section will discuss the pharmacokinetic issues.
5. Pharmacokinetic issues and countermeasures
For isotretinoin to exert its pharmacological effects
in vivo, maintaining an adequate plasma concentration is necessary. As seen in the preclinical studies mentioned above, a plasma concentration ranging from 5 to 10 μM is required to maintain growth arrest and differentiation in neuroblastoma cell lines [
11,
12]. The pharmacokinetic details of a phase 1 study [
14] demonstrated a linear increase in the mean peak serum level and area under the time-concentration curve (AUC) for a dosage range of 100 to 200 mg/m
2 [
52]. The peak serum concentrations were 4.9 ± 3.6 μM for 100 mg/m
2 (n=5), 7.2 ± 5.3 μM for 160 mg/m
2 (n=16), and 8.9 ± 10.0 μM for 200 mg/m
2 [
52]. A peak serum concentration exceeding 10 μM correlated with a high incidence (44%) of grade 3 to 4 toxicity [
52]. Effectiveness of maintenance therapy in the CCG-3891 study, which was discussed in the previous section [
15,
16], probably resulted from the same type of active dosing whereas the conservative dosing schedule in the European study [
51] and the first CCG study [
13] failed to demonstrate the therapeutic efficacy of isotretinoin.
Several factors influence the absorption, distribution, metabolism, and elimination (ADME) of isotretinoin. Isotretinoin has good permeability and poor solubility in the aqueous environment of the intestine, and its absorption is greatly enhanced by fatty foods. In contrast, the drug’s absorption and permeability are usually low during fasting. Veal et al. intensively investigated these factors in their pharmacokinetic studies [
53,
54] and found that swallowing intact capsules without extracting their contents was the biggest factor in achieving a high
Cmax (4.0 ± 2.2 vs 2.6 ± 1.8 μM) [
54]. In this regard, the development of a drug formulation suitable for pediatric patients, such as a liquid or powder, may be desirable. However, the relatively small target population of neuroblastoma patients has stymied efforts to develop a new formulation, given the extremely high costs involved.
On the other hand, strategies for improving the bioavailability of isotretinoin, which are equally important for the treatment of neuroblastoma and acne vulgaris, have been implemented. Such strategies available include lidose technology [
55] and more recently micronization technology [
56], which utilizes a novel capsule formulation with a specific lipid vehicle to deliver the drug. Micronization technology in particular can substantially increase the surface area per unit mass of the drug and thereby increase its rate of dissolution and bioavailability [
56]. Technologies such as these are likely to be crucial for achieving a higher and more stable
Cmax by improving the drug’s bioavailability.
Two, open-label, crossover studies compared micronized isotretinoin (Sun Pharmaceutical Industries, Inc., Cranbury, NJ, USA) 32 mg and its prototype, lidose isotretinoin (Absolica
TM: Sun Pharmaceutical Industries, Inc., Cranbury, NJ, USA) 40 mg [
56]. One of these, a fed bioequivalence / food-effect study enrolling 71, healthy, adult participants received a single dose of micronized isotretinoin 32 mg, lidose isotretinoin 40 mg, and micronized isotretinoin 32 mg in a fed state, fed state, and fasted state, respectively. Bioavailability was assessed using the formula, isotretinoin log-AUC
0–t, log-AUC
0–∞ and log-
Cmax in blood samples taken pre-dosing and more than 96 h post-dosing. The 90% confidence interval for the baseline-adjusted geometric least squares mean ratios for log-AUC
0–t, log-AUC
0–∞ and log-
Cmax fell within the 80–125% range for bioequivalence for micronized-isotretinoin 32 mg vs. lidose-isotretinoin 40 mg, both of which were administered in the fed state [
56]. In the fasted state, 18, healthy, adult participants received a single dose of micronized-isotretinoin 32 mg and lidose-isotretinoin 40 mg. Results showed that micronized-isotretinoin 32 mg had approximately twice the bioavailability of lidose-isotretinoin 40 mg. Food had no effect on bioavailability and only a marginal effect on the extent of absorption of micronized isotretinoin 32 mg [
56].
Another factor which might influence the pharmacokinetics of isotretinoin is the presence of biologically active metabolites. Of the major metabolites of isotretinoin, 4-oxo-isotretinoin is the most abundant [
52,
53,
54,
57]. In the phase 1 trial discussed above, the 4-oxo-isotretinoin level increased from day 1 to day 14 of isotretinoin administration in 64% of the patients [
52]. A pharmacokinetic study conducted by Veal et al. found extensive accumulation of 4-oxo-13-cisRA during each course of treatment, with the plasma concentration of the metabolite being (4.677±3.17 mM) higher than that of 13-cisRA (2.837±1.44 mM) in 16 of 23 patients on day 14 of course 2 [
53]. In another study by Veal et al., a statistically significant difference in the 4-oxo-isotretinoin
Cmax value was observed on day 14 for CYP2C8*4 and CYP3A7*1C polymorphisms although no clear impact of pharmacogenetics in determining the peak plasma concentration of isotretinoin itself was shown [
54].
Sonawane et al. performed a series of experiments aimed at determining if 4-oxo-isotretinoin is an active metabolite of isotretinoin. First, they compared the inhibitory effect of 4-oxo-isotretinoin with that of isotretinoin on six, neuroblastoma cell lines (three with, and three without, MYCN amplification). Both 4-oxo-isotretinoin and isotretinoin demonstrated a similar inhibitory effect (p > 0.2 in all six cell lines) by inhibiting more than 90% of cell growth at the highest concentration in three cell lines (SMS-KANR, CHLA-20, and SMS-LHN). Second, they tested the inhibitory effect of 4-oxo-isotretinoin against both MYCN mRNA and protein in MYCN-amplified cell lines. The extent of the decrease relative to the control appeared to be identical for both 4-oxo-isotretinoin and isotretinoin. Third, they tested the ability of the substances to induce RARβ expression, which is reportedly associated with favorable clinical outcomes in neuroblastoma, as described in the previous section. Real-time RT-PCR demonstrated that both 4-oxo-isotretinoin and isotretinoin induced significantly higher (p < 0.05) RARβ mRNA levels than in vehicle control cells on day 10 in four, MYCN gene-amplified and three, MYCN non-amplified NB cell lines. No difference in the ability of the two substances to induce RARβ (p=0.632) was observed. Finally, they confirmed neurite outgrowth and cell differentiation, which were promoted by both 4-oxo-isotretinoin and isotretinoin in SMS-KCNR and SMS-LHN cell lines.
Further investigation of the pharmacokinetics of isotretinoin and its metabolites in tandem with the development of a drug formulation suitable for childhood neuroblastoma is challenging but warranted as a step towards optimized tumor differentiation therapy using isotretinoin.
6. Pediatric off-label use and countermeasures
Although isotretinoin is an essential element of multimodal therapy for high-risk neuroblastoma, its use as a treatment for neuroblastoma is still off-label in most countries, including the US, EU, and Japan. This means that isotretinoin is not yet a part of the standard, medical treatment from the regulatory point-of-view despite the scientific evidence for its efficacy having been established more than two decades ago. Our research group recently began a registration-directed, investigator-initiated clinical trial to test the safety and efficacy of micronized-isotretinoin as a step towards the approval of its use in Japan.
The Evaluation Committee on Unapproved or Off-labeled Drugs with High Medical Needs [
58], established by the Ministry of Health, Labor and Welfare (MHLW) in 2009, recently concluded that isotretinoin qualifies as an unapproved drug with high medical need and therefore justifies urgent clinical development. Afterwards, discussions with PMDA, the regulatory agency in Japan, led to a tentative consensus that the accumulated data on the efficacy of isotretinoin for high-risk neuroblastoma in the US and EU may be extrapolatable to Japanese patients. This decision enabled planning for a relatively small clinical trial aimed at assessing the safety profile and pharmacokinetics of micronized isotretinoin (SPJ-101CA) for use as a treatment for high-risk neuroblastoma (Clinical Trial Registration: jRCT2031220687).
The inclusion criteria are (1) age 1 to 18 years; (2) a histopathological diagnosis of neuroblastoma or ganglioneuroblastoma; (3) high risk as defined by the International Neuroblastoma Risk Grouping system; (4) absence of progression after primary treatment, including chemotherapy, high-dose chemotherapy with hematopoietic stem cell transplantation, and radiotherapy; (5) 100 days or less after the most recent anticancer therapy; (6) absence of severe organ damage capable of interfering with the protocol treatment; (7) absence of an active, infectious disease; and (8) written informed consent from the patient and/or legal guardian. All the patients will receive oral SPJ-101-CA 128 mg/m2/day divided into two doses for 14 days followed by a 14-day break for six courses over a 28-day cycle. Concomitant treatment with chemotherapy will not be allowed. The primary endpoint is the severe adverse event rate with causality. The secondary endpoints include 1-year EFS, 1-year OS, the adverse event rate, and pharmacokinetics. Sixteen patients will be enrolled from March 2023 through August 2024. Follow-up observation will end in August 2025.
The clinical role of academic clinical trials in pediatric cancer drug approval has been actively discussed for decades [
59]. The impact of regulatory approval of isotretinoin for neuroblastoma treatment will likely be significant in Japan, which is a member of the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH). It is hoped that the planned study will result in advances enabling the development of effective treatment options for children with cancer, not least by providing a model of fair and efficacious collaboration among academia, industries, and regulatory agencies.
7. Prospects for retinoid therapy
As discussed above, isotretinoin is an established maintenance therapy for high-risk neuroblastoma. Current treatment regimens for high-risk neuroblastoma involve scheduling the administration of isotretinoin during the rest periods in anti-GD2 immunotherapy with the combination maintenance therapy lasting six months [
5,
6]. However, there are various, other opportunities for using retinoids as neuroblastoma treatment besides maintenance therapy; it may also be applied in consolidation therapy or salvage induction therapy if the agent or regimen is sufficiently effective. When developing a new tumor differentiation therapy, the use of retinoids should be treated as a single, independent treatment phase rather than as a part of maintenance therapy with anti-GD2 immunotherapy.
One, potential direction for further exploration involves the development of new retinoid agents with greater efficacy as a neuroblastoma treatment. Fenretinide, a synthetic retinoid which exerts a cytotoxic rather than maturational effect on neuroblastoma cell lines, has a long history of preclinical and clinical development [
38]. Two phase 1 trials using a traditional capsule formulation of fenretinide have been completed [
31,
32]. Although the safety profiles were acceptable, oral administration of a very high-dose formulation involving a large number of capsules was needed to achieve an effective plasma concentration. A later phase 2 trial testing the recommended dosage of 2,475 mg/m
2 divided over three daily administrations in patients aged <18 years) or 1,800 mg/m
2 divided into two, daily administrations in patients aged 18 years or older every 21 days for a maximum of 30 courses achieved one partial response and 13 instances of prolonged stable disease in 59 evaluable patients. To increase bioavailability, an oral, pulverized lipid complex (LXS) [
34] and an intravenous formulation [
60] are currently being developed. Another synthetic retinoid, tamibarotene, functions as a RARα and RARβ agonist and was able to induce differentiation in SH-SY5Y and NH-12 cells to a greater extent than ATRA [
61,
62]. Recently, Nitani et al. reported the results of a phase 1 study of tamibarotene monotherapy targeting recurrent / refractory pediatric solid tumors in 22 patients (median age: 8 years) [
35]. The subjects tolerated tamibarotene well, and no case of dose-limiting toxicity (DLT) occurred at any of the six dosages tested. However, none of the patients achieved a complete or partial response. The recommended dosage was determined to be 12 mg/m
2/day for 21 days in a 28-day cycle [
35]. Despite the huge effort involved in developing new retinoid therapies, no retinoid agent thus far has surpassed isotretinoin in terms of overall efficacy.
Combination therapy using RAs with other agents presents another potential area of research. As described in section 2.3, RARs function as transcription modulators by recruiting coregulator complexes having HDAC activity. RAs works as ligands to activate or block gene transcription mediated by the RARs. This finding led the idea of exploiting the synergy of combining an HDAC inhibitor with a natural RA in the treatment of neuroblastoma. Both
in vitro and
in vivo xenograft models have confirmed the synergy of retinoids and HDAC inhibitors. The combination of m-carboxycinnamic acid bis-hydroxamide (CBHA), an HDAC inhibitor, with ATRA led to synergistic cytotoxicity against neuroblastoma cell lines in
in vitro and
in vivo xenograft models [
63,
64]. Later studies using a combination of ATRA with the HDAC inhibitors, TSA, sodium butyrate or vorinostat also demonstrated a synergistic effect inhibiting the growth of neuroblastoma cell lines
in vitro [
65]. Based on these preclinical findings, Pinto et al. conducted a phase 1 trial of vorinostat combined with isotretinoin for refractory/recurrent neuroblastoma [
66]. The maximum intended dosage of vorinostat (430 mg/m
2/day on days 1–4 and 8–11) combined with isotretinoin (160 mg/m
2/day on days 1-14) was tolerable and led to more histone acetylation in surrogate tissues than at lower doses of vorinostat (
p = 0.009). Although objective responses occurred, 17% of the evaluable patients achieved prolonged stable disease [
66].
There are several, other, possible combinations, such as isotretinoin plus vandetanib [
67], fenretinide plus vorinostat [
68], or lenalidomide [
69], and tamibarotene plus 5-aza-2’-deoxycytidine [
70], all of which have demonstrated a synergic, anti-neuroblastoma effect
in vitro and
in vivo. Considering that most of the treatment phases for high-risk neuroblastoma are quite toxic for children, reducing the need for toxic tumor differentiation therapy can expand the application of such regimens not only as maintenance therapy following mega-dose chemotherapy and SCT but also as salvage treatment for heavily treated relapsed or refractory patients with neuroblastoma.