Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Apr 1.
Published in final edited form as: Invest New Drugs. 2020 Nov 16;39(2):426–441. doi: 10.1007/s10637-020-01030-0

A decision process for drug discovery in retinoblastoma

María Belen Cancela 1,2, Santiago Zugbi 1,2, Ursula Winter 3, Ana Laura Martinez 1, Claudia Sampor 4, Mariana Sgroi 5, Jasmine H Francis 6, Ralph Garippa 7, David H Abramson 6, Guillermo Chantada 1,2, Paula Schaiquevich 1,2
PMCID: PMC8488950  NIHMSID: NIHMS1733825  PMID: 33200242

Abstract

Intraocular retinoblastoma treatment has changed radically over the last decade, leading to a notable improvement in ocular survival. However, eyes that relapse remain difficult to treat, as few alternative active drugs are available. More challenging is the scenario of central nervous system (CNS) metastasis, in which almost no advancements have been made. Both clinical scenarios represent an urgent need for new drugs. Using an integrated multidisciplinary approach, we developed a decision process for prioritizing drug selection for local (intravitreal [IVi], intrathecal/intraventricular [IT/IVt]), systemic, or intra-arterial chemotherapy (IAC) treatment by means of high-throughput pharmacological screening of primary cells from two patients with intraocular tumor and CNS metastasis and a thorough database search to identify clinical and biopharmaceutical data. This process identified 169 compounds to be cytotoxic; only 8 are FDA-approved, lack serious toxicities and available for IVi administration. Four of these agents could also be delivered by IT/IVt. Twelve FDA-approved drugs were identified for systemic delivery as they are able to cross the blood-brain barrier and lack serious adverse events; four drugs are of oral usage and six compounds that lack vesicant or neurotoxicity could be delivered by IAC. We also identified promising compounds in preliminary phases of drug development including inhibitors of survivin, antiapoptotic Bcl-2 family proteins, methyltransferase, and kinesin proteins. This systematic approach may be applied more broadly to prioritize drugs to be repurposed or to identify novel hits for use in retinoblastoma treatment.

Keywords: retinoblastoma, intraocular, metastasis, decision process, high-throughput drug screening

Introduction

Retinoblastoma is the most common primary neoplasm of the eye in childhood[1]. The current use of novel routes to more selectively deliver chemotherapy to the eye, such as intraarterial chemotherapy (IAC) or intravitreal administration (IVi), has resulted in major improvements in eye preservation while reducing systemic drug exposure[14]. However, for eyes that relapse after conventional therapy or have refractory tumors, treatment options are few, and many eyes must be enucleated. Even less progress has been witnessed in the development of treatments for metastatic retinoblastoma, a stage of the disease that accounts for up to 50% of cases in low- and middle-income countries[1, 57]. Up to 60% these patients can be cured if treated with high-dose chemotherapy and autologous hematopoietic stem-cell rescue but still face a poor prognosis with an invariably fatal outcome in cases of central nervous system (CNS) involvement[8, 9]. Thus, there is a clear need to develop novel treatment strategies using more effective and less toxic chemotherapy.

The most commonly used drugs for retinoblastoma are carboplatin, etoposide, and vincristine, delivered via intravenous infusion1; melphalan, topotecan, and carboplatin intraarterially[3, 4, 10] (IAC), and melphalan or topotecan intravitreally (IVi)[1, 2]. The first three have been clinically implemented based on results in other pediatric malignancies and their pharmacological profile was only studied after their clinical use. Topotecan and melphalan activity was assessed in preclinical models before their clinical implementation but a complete pharmacokinetic profile for local administration was available only after clinical use [1113]. Comprehensive pharmacological characterization involves the use of patient-derived cell lines to mimic the biological characteristics of intraocular and metastatic disease[14]. Such approaches are often used to identify new drugs for retinoblastoma, especially in the context of high-throughput screening (HTS) for active compounds among libraries of thousands[1517]. Nonetheless, antitumor activity should not be the only factor to prioritize a candidate drug for further development; factors such as penetration across the ocular and brain barriers, toxicity profile, and the availability of adequate formulations are important when considering clinical utility.

We hereby propose a model for prioritizing potential drug candidates that most deserve further characterization, adding a selection process to assign the most suitable compounds according to intent of treatment, biopharmaceutical properties, and previous knowledge of their pharmacology.

Materials and Methods

Patient-derived tumor cells

Two patient-derived primary cell cultures, one established from an intraocular tumor of an upfront enucleated eye (HPG-RBT-12L) and the other from cerebrospinal fluid (CSF) tumor relapse after initial chemotherapy treatment (HPG-CSF-1), were used in the present study. Cell line origin, authentication, and culture conditions were previously described[1820]. The commercial cell line Y79 (ATCC HTB-18, Manassas, VA) was used as a control for HTS and cultured as well described elsewhere[14, 21].

Patient tumor samples were obtained after protocol and informed consent approval by the Institutional Review Board at Hospital de Pediatria JP Garrahan (protocol #838) and written informed consent was obtained from parents or guardians. All procedures involving human subjects were performed in accordance with the Declaration of Helsinki.

High-throughput screening

Optimal HTS assay conditions were established based on previous work published elsewhere[15, 2224] and adapted to our system conditions described in Online Resource 1. Cells were exposed to each of the 2,700 medicinally active and structurally diverse compounds contained in the Selleck Bioactive and Selleck FDA-approved libraries (SelleckChem, Houston, TX). Cells were automatically counted, seeded in 384-well plates, and cultured for 24 h. Next, compounds were diluted in 0.5% DMSO (v/v) and transferred into the well plates using an Agilent Bravo Liquid Handler, resulting in a final concentration of 4.8 μM. All plates also included positive (melphalan) and negative (vehicle) controls; melphalan was used based on its potent and reproducible in vitro cytotoxicity[21, 25]. After 72 h of drug exposure, cell viability was determined[15, 24]. Compounds that showed > 70% cytotoxic activity at 4.8 μM were considered pharmacologically active against retinoblastoma.

Decision process

A classification process was built for FDA-approved drugs as potential agents to be repurposed or new hits to be prioritized for various treatment scenarios of retinoblastoma. The decision process was based on a series of questions with binary answers (yes = 1, no = 0) identified by consensus from a multidisciplinary collaborative team of clinical experts.

The starting point was defined as pharmacologically active compounds against HPG-RBT-12L and HPG-CSF-1 cells. From the group of active drugs, we filtered for those that were FDA-approved for clinical use or were tested in clinical trials in adults and have been or are being evaluated in Phase I/II pediatric trials. The next question was intended to describe the clinical scenario for their potential use and thus, compounds were divided according to administration routeusing the following criteria:

Local treatment

  • Intravitreal (for the treatment of intraocular tumors): injectable solution, devoid of vesicant or irritant effects, and devoid of neurologic or ocular toxicity.

  • Intrathecal or intraventricular (for the treatment of CSF dissemination): same as for IVi injection, with evidence of clinical use of IT/IVt delivery[2630].

Intraarterial or intravenous administration

  • Intravenous (for chemoreduction of intraocular tumors, systemic treatment of extraocular tumors): injectable solution, able to penetrate the blood-brain barrier (BBB) or blood-ocular barrier (BOB), and not associated with serious adverse events or serious neurologic or ocular toxicity.

  • Oral: same purpose and characteristics as for i.v. administration and pharmaceutically formulated for oral administration.

  • Intraarterial chemotherapy (local treatment delivered into blood vessels): same criteria as for i.v. and does not require in vivo activation or prodrugs.

In all cases, injectable solution referred to the availability of an aqueous and sterile solution as the drug must be dissolved in a biocompatible solvent for IVi, IAC, or IT/ IVt administration. For i.v. or IAC treatment, candidate drugs must penetrate the BOB to target the tumor localized in the retina and/or vitreous humor. Thus, we searched for reports that the drugs could penetrate the BOB, and if not available, data on penetration across the BBB was considered a surrogate based on the barriers’ anatomical similarities[29, 3133]. We also searched for published reports on pharmacokinetics in preclinical models that favored drug distribution in the brain or clinical endpoints supporting CNS activity and therefore, justifying drug penetration across the BBB. Serious adverse events were defined as those that may result in death, a life-threatening adverse event, or hospitalization or prolongation of hospitalization except for hematological toxicities or other clinically manageable toxicities (grade III/IV adverse events by the Common Terminology Criteria for Adverse Events (CTCAE) v5)[34].

For each compound, we searched for answers to these detailed questions in several databases as described in Online Resource 1[3537]. To avoid missing compounds of interest, we built a parallel decision process for drug discovery to identify new hits that have been excluded because of lack of clinical use or adult or pediatric clinical studies but that still show cytotoxic activity against retinoblastoma cells and have an adequate safety profile according to clinical or preclinical studies.

Statistical analysis for sensitivity and precision assessment could not be performed because selected features consisted of questions relevant to but also mandatory for the clinical implementation of a novel treatment. Thus, the resulting features intrinsically provided the best separation of the dataset or largest information gain.

Results

In total, 169 compounds were highly active against both HPG-RBT-12L and HPG-CSF-1 cells as indicated by > 70% cytotoxic activity (Table 1). Among these, we selected 113 drugs evaluated in clinical trials in adults, of which 59 have been investigated in Phase I/II studies in pediatric patients or clinically used in children. For rapid translation into the clinic, we selected 30 FDA-approved drugs as potential candidates to be repurposed, which were then classified into two categories according to likely administration route, one for local conservative treatment (IVi) or CSF dissemination (IT/IVt) and the other including IAC, oral, or i.v. chemotherapy for conservative therapy or adjuvant or neoadjuvant therapy for extraocular disease.

Table 1. Compounds active in patient-derived retinoblastoma cells.

Compounds found to be active (> 70% cell growth inhibition) in HPG-RBT-12L (derived from intraocular tumor) and HPG-CSF1 cells (cells disseminated into the cerebrospinal fluid of a relapsed heavily pretreated patient). Name, supplier catalog number, and mechanism of action are detailed.

Compound (n = 169) Selleck code Mechanism of action
(−)-Parthenolide S2341 NF-κB /HDAC dual inhibitor
(+)-JQ1 S7110 BET inhibitor
Hydroxycamptothecin S2423 Topoisomerase I inhibitor
5-Iodotubercidin S8314 Adenosine kinase inhibitor
Abexinostat S1090 HDAC inhibitor
ABT-737 S1002 BH3 mimetic inhibitor
Alvespimycin S1142 HSP90 inhibitor
AR-42 S2244 HDAC inhibitor
ARQ 621 S7355 KSP inhibitor
AT13387 S1163 HSP90 inhibitor
AT9283 S1134 Aurora A,B / JAK2/3 dual inhibitor
AUY922 S1069 HSP90 inhibitor
AZ5104 S7298 EGFR inhibitor
AZ6102 S7767 TNKS inhibitor
AZD1480 S2162 JAK1/2 inhibitor
AZD8055 S1555 mTOR inhibitor
AZD9291 S7297 EGFR inhibitor
Bardoxolone methyl S8078 Keap1/ Nrf2 activator
BAY 11–7082 S2913 NF-κBα (IκBα) inhibitor
Bay 11–7085 S7352 NF-κB α (IκBα) inhibitor
Belinostat S1085 HDAC inhibitor
Benzethonium S4162 Antiseptic/ tensioactive
BGT226 S2749 PI3K/ mTOR dual inhibitor
BI 2536 S1109 PLK1/ BRD4 inhibitor
BIIB021 S1175 HSP90 inhibitor
BIX 01294 S8006 Histone methyltransferase G9a inhibitor
BMS-754807 S1124 IGF-1R/ IR dual inhibitor
Bortezomib S1013 Proteasome inhibitor
Brefeldin A S7046 Antimicrobial (ATPase inhibitor)
Buparlisib S2247 PI3K inhibitor
BV-6 S7597 cIAP / XIAP dual inhibitor
Caffeic acid phenethyl ester S7414 NF-κB inhibitor
Camptothecin S1288 Topoisomerase I inhibitor
Carfilzomib S2853 Proteasome inhibitor
CB-5083 S8101 p97 inhibitor
Celastrol S1290 Proteasome inhibitor
Cepharanthine S4238 Anti-inflammatory
Ceritinib S7083 ALK inhibitor
Cetrimonium S4242 Antiseptic (quaternary ammonium)
Cetylpyridinium S4172 Antiseptic (quaternary ammonium)
CH5138303 S7340 HSP90 inhibitor
Ciclopirox S2528 Antifungal/ iron chelator
Cladribine S1199 Adenosine deaminase inhibitor
CPI-203 S7304 BET inhibitor
Crystal Violet S1917 Antiseptic (antimitotic)
Cucurbitacin B S8165 Phytochemical (mitosis inhibitor)
CUDC-101 S1194 HDAC/ EGFR / HER2 triple inhibitor
CUDC-907 S2759 PI3K/ HDAC dual inhibitor
CX-6258 S7041 Pim kinase inhibitor
Dacinostat S1095 HDAC inhibitor
Daunorubicin S3035 Anthracycline-topoisomerase II inhibitor
Delanzomib S1157 Proteasome inhibitor
Deltarasin S7224 PDEδ/Kras dual inhibitor
Digoxin S4290 Cardiac glycoside (Na/K ATPase inhibitor)
Domiphen S4186 Antiseptic (quaternary ammonium)
Dovitinib Dilactic Acid S2769 RTK inhibitor
Dp44Mt S7909 Iron chelator
Entinostat S1053 HDAC inhibitor
Epoxomicin S7038 Proteasome inhibitor
Ethacridine lactate S4196 Antiseptic
Etoposide S1225 Epipodophyllotoxin (topoisomerase II inhibitor)
Fedratinib S2736 JAK-2 inhibitor
Gambogic Acid S2448 Apoptosis inducer
Ganetespib S1159 HSP90 inhibitor
GDC-0980 S2696 PI3K/ mTOR dual inhibitor
Geldanamycin S2713 HSP90 inhibitor
Gemcitabine S1714 Antimetabolite
Givinostat S2170 HDAC inhibitor
GMX1778 S8117 NAMPT inhibitor
GSK J4 S7070 JMJD3/ UTX dual inhibitor
GSK1070916 S2740 Aurora B/C inhibitor
GSK1324726A S7620 BET inhibitor
GSK2126458 S2658 PI3K inhibitor
Hesperadin S1529 Aurora B kinase inhibitor
HSP990 S7097 HSP90 inhibitor
ICG-001 S2662 Wnt pathway inhibitor
Idarubicin S1228 Anthracycline (topoisomerase II inhibitor)
IKK-16 S2882 IKK inhibitor
Irinotecan S2217 Topoisomerase I inhibitor
Ispinesib S1452 KSP inhibitor
Ixazomib S2180 Proteasome inhibitor
JTC-801 S2722 NOP antagonist
KPT-185 S7125 CRM1 inhibitor
KPT-276 S7251 CRM1 inhibitor
KW-2478 S2685 HSP90 inhibitor
Linsitinib S1091 IGF-1R/ IR dual inhibitor
LMK-235 S7569 HDAC inhibitor
LMTX™ S7762 Tau protein aggregation inhibitor
LY3023414 S8322 PI3K/ mTOR / DNA-PK triple inhibitor
M344 S2779 HDAC inhibitor
MCB-613 S7913 SRC stimulator
Melphalan S8266 Nitrogen mustard (DNA alkylator)
Methylene Blue S4535 Oxidation-reduction agent
MG-101 S7386 Cysteine protease inhibitor
MG-132 S2619 Proteasome inhibitor
MI-773 S7649 MDM2 inhibitor
Milciclib S2751 CDK/ TRK dual inhibitor
Mitomycin C S8146 Antibiotic
Mitoxantrone S1889 Topoisomerase II inhibitor
MK-1775 S1525 Wee1 inhibitor
MLN9708 S2181 Proteasome inhibitor
Mocetinostat S1122 HDAC inhibitor
Navitoclax S1001 BCL-2 inhibitor
NH125 S7436 eEF-2 inhibitor
NMS-E973 S7282 HSP90 inhibitor
NSC 319726 S7149 p53 reactivator
NVP-ADW742 S1088 IGF-1R inhibitor
NVP-AEW541 S1034 IGF-1R inhibitor
Obatoclax S1057 BCL-2 inhibitor
Oligomycin A S1478 Antibiotic (ATPase inhibitor)
Omaveloxolone S7672 NRF2 activator
ONX-0914 S7172 Proteasome inhibitor
Oprozomib S7049 Proteasome inhibitor
Oridonin S2335 PI3K inhibitor
OTX015 S7360 BET inhibitor
Ouabain S4016 Cardiac glycoside (Na/K ATPase inhibitor)
Pacritinib S8057 JAK2/ FLT3 dual inhibitor
Panobinostat S1030 HDAC inhibitor
PD0166285 S8148 Wee1 /Myt1 dual inhibitor
PF-04929113 S2656 HSP90 inhibitor
PF-477736 S2904 Chk1 inhibitor
Piperlongumine S7551 ROS inducer
Pirarubicin S1393 Antibiotic (anthracycline)
Pracinostat S1515 HDAC inhibitor
PRI-724 S8262 Wnt signaling pathway inhibitor in cancer stem cell
PU-H71 S8039 HSP90 inhibitor
Puromycin S7417 Antimicrobial/ antibiotic
Quisinostat S1096 HDAC inhibitor
RAF265 S2161 RAF/ VEGFR2 dual inhibitor
Resminostat S2693 HDAC inhibitor
Ricolinostat S8001 HDAC inhibitor
Rigosertib S1362 PLK1 inhibitor
Romidepsin S3020 HDAC inhibitor
SB743921 S2182 KSP inhibitor
SBI-0640756 S8181 EIF4G1 inhibitor
Scriptaid S8043 HDAC inhibitor
Selinexor S7252 CRM1 inhibitor
Sepantronium S1130 Survivin inhibitor
SGI-1776 S2198 Pim kinase inhibitor
SGI-7079 S7847 Axl inhibitor
SN-38 S4908 Topoisomerase I inhibitor
SNX-2112 S2639 HSP90 inhibitor
SRT1720 S1129 SIRT1 agonist
Stattic S7024 STAT3 inhibitor
Staurosporine S1421 Antifungal/ protein kinase C inhibitor
STF-118804 S7316 NAMPT inhibitor
TAE226 S2820 FAK/ IGF-1R dual inhibitor
Tanespimycin S1141 HSP90 inhibitor
Teniposide S1787 Epipodophyllotoxin (topoisomerase II inhibitor)
Tenovin-6 S4900 HDAC inhibitor
Terfenadine S4353 Antihistamine
TG101209 S2692 JAK-2 inhibitor
TIC10 S7963 TRAIL inducer
Topotecan S1231 Topoisomerase I inhibitor
Trichostatin A (TSA) S1045 HDAC inhibitor
Tubercidin S8095 Antibiotic (polymerase inhibitor)
UM171 S7608 HSC self-renewal agonist
UNC0631 S7610 Histone methyltransferase G9a inhibitor
UNC-2025 S7576 Mer/ FLT3 inhibitor
VER-49009 S7458 HSP90 inhibitor
VER-50589 S7459 HSP90 inhibitor
Verdinexor S7707 CRM1 inhibitor
Vinblastine S4505 Vinca alkaloid-mitotic inhibitor
Vincristine S1241 Vinca alkaloid-mitotic inhibitor
Vinflunine S2209 Vinca alkaloid-mitotic inhibitor
Vinorelbine S4269 Vinca alkaloid-mitotic inhibitor
Vorinostat S1047 HDAC inhibitor
VS-5584 S7016 PI3K/ mTOR dual inhibitor
XL888 S7122 HSP90 inhibitor

ALK, anaplastic lymphoma kinase inhibitor; BCL-2, B-cell lymphoma 2; BET, bromodomain and extraterminal domain; BRD4, bromodomain-containing protein 4; CDK, cyclin-dependent kinase; Chk1, checkpoint kinase 1; cIAP, cellular inhibitor of apoptosis protein-1; CRM1, chromosome region maintenance 1; DNA-PK, DNA-dependent protein kinase; Eef-2, eukaryotic elongation factor 2; EGFR, epidermal growth factor receptor; EIF4G1, eukaryotic translation initiation factor 4 gamma 1; FAK, focal adhesion kinase; FLT3, Fms like tyrosine kinase 3; HDAC, histone deacetylase inhibitors; HER2, human epidermal growth factor receptor type 2; HSC, hematopoietic stem cell; HSP90, heat shock protein 90; IGF-1R, insulin-like growth factor 1 receptor; IKK, inhibitor of nuclear factor kappa-B kinase; IR, insulin receptor; JK2/3, Janus kinase 2,3; JMJD3, Jumanji domain-containing protein D3; Keap1, Kelch-like ECH-associated protein 1; Kras, Kirsten rat sarcoma viral oncogene homolog; KSP, kinesin spindle protein; MDM2, mouse double minute 2 homolog; mTOR, mammalian target of rapamycin; NAMPT, nicotinamide phosphoribosyltransferase; NFR2, nuclear factor erythroid 2-related factor; NF-Κb, nuclear factor kappa-light-chain-enhancer of activated B cells; NOP, nociceptin Receptor; Nrf2, nuclear factor (erythroid-derived 2)-like 2; PDEδ, photoreceptor cGMP phosphodiesterase δ subunit; PI3K, phosphatidylinositol 3-kinase; PLK1, Polo-like kinase 1; RKT, receptor tyrosine kinases; SIRT1, Sirtuin 1; SRC, steroid receptor coactivator; STAT3, signal transducer and activator of transcription 3; TNKS, Tankyrase; TRAIL, TNF-related apoptosis-inducing ligand; TRK, tropomyosin receptor kinase; UTX, ubiquitously transcribed X-chromosome tetratricopeptide repeat protein; VEGFR2, vascular endothelial growth factor receptor 2; XIAP, X-linked inhibitor of apoptosis protein.

Only compounds that do not require metabolic activation and for which an injectable solution is available may be administered IVi or IT/IVt and thus were considered for local delivery. Subsequently, 13 compounds were excluded due to reports of vesicant, neurological, or ocular toxicities. Thus, 8 drugs, accounting for 5% of the total number of active compounds (n = 169), met all selection criteria for local IVi administration (Figure 1). This group of agents consisted of a histone deacetylase inhibitor (HDACi, romidepsin), one dye that also acts as an oxidation-reduction agent (methylene blue), one cardiac glycoside (digoxin), one antimetabolite (gemcitabine), and four agents already used in retinoblastoma including a topoisomerase I inhibitor (topotecan), a nitrogen mustard (melphalan), and two epidophyllotoxins (etoposide and teniposide).

Figure 1. Decision sequence to prioritize novel drugs for local and systemic administration in retinoblastoma.

Figure 1.

Serious adverse events were those classified as grade III/IV adverse events by the Common Terminology Criteria for Adverse Events (CTCAE) v5. Hematological toxicities or other clinically manageable toxicities (named as serious adverse events) were not included.

a Compounds in Phase I/II pediatric trials or that are used in routine clinical management.

b Compounds in Phase I/II pediatric trials or that are used in routine clinical management by the indicated route of administration.

Abbreviations: IT, intrathecal; IVt, intraventricular; IVi, intravitreal injection; IAC, intraarterial chemotherapy; i.v, intravenous infusion, BBB, blood-brain barrier; BOB, blood-ocular barrier.

IT or IVt administration may provide an alternative route of local CNS drug delivery for drugs that do not cross the BBB but display cytotoxicity against retinoblastoma cells. Of the 8 drugs considered candidates for local treatment (topotecan, etoposide, teniposide, and romidepsin), 4 were only eligible for IT/IVt administration after ruling out 4 active compounds with reported neurologic toxicity or serious adverse events after IT or IVt administration (i.e. digoxin, gemcitabine) [26, 38, 39].

Twelve of the 30 drugs that fulfilled the criteria for drug repurposing do cross the BBB or BOB and are not associated with serious adverse events and thus could be applied for systemic (i.v., oral, or IAC) treatment. These compounds included five agents that are already clinically used to treat metastatic retinoblastoma: two topoisomerase I inhibitors (topotecan, irinotecan), two topoisomerase II inhibitors (etoposide, teniposide), and an anthracycline (idarubicin). The other agents identified included two antimetabolites (gemcitabine, cladribine), and additional repurposing drugs consisting of one dye (methylene blue), two HDACi (vorinostat, panobinostat), an ALK inhibitor (ceritinib), and an exportin 1 (XPO1) inhibitor (selinexor) (Figure 1). Eight of these drugs are available in an aqueous formulation and could be considered for i.v. treatment. Of this set, we excluded compounds with vesicant effects (idarubicin[40]) or that require metabolic activation (irinotecan and cladribine), and thus, 5 compounds remained prioritized for IAC administration. Melphalan was retained despite its myelosuppressive effects because of its high cytotoxicity; dose reductions have been proven effective for intraocular treatment by IAC administration and eliminate such risks[3, 11, 41].

The candidates that emerged as new hits in the discovery branch included 61 agents that inhibited primary cell growth by ≥ 90% and have not been clinically tested in adults or children. Eighteen agents were eligible for systemic administration, as they are able to cross the BOB or BBB, of which four lack severe toxicity, including JTC-801, brefeldin A, parthenolide, and omaveloxolone (Figure 1, right flowchart; mechanism of action summarized in Table 2).

Table 2.

Selected agents prioritized for drug discovery.

Systemic administrationa

Compound Mechanism of action Known and mechanism-related effects
Brefeldin-A Antimicrobial; ATPase inhibitor • Macrocyclic lactone
• Exerts transcytosis inhibition resulting in impaired transcription
• Induces caspase-mediated cell death independent of p53 status in several human tumor cell lines; also inhibits VEGF secretion[88, 89]
• Safe in cultured rat photoreceptors[90]
JTC-801 Nociceptin receptor (NOP) antagonist • Cytotoxic against NCI-60 human cancer cell panel and panel of human xenograft including ortothopic and metastatic tumors[91]
• Effects mediated by pH-dependent cell death[91]
• Induces apoptosis mediated by PI3K-AKT[92]
• Inhibits invasion and migration[92]
Omaveloxolone Nuclear factor erythroid 2-related factor (Nrf2) activator • Antioxidant inflammation modulator (AIM); reduces oxidative stress and inflammation
• Antitumor activity may also involve inhibition of NF-κB, decreased cyclin D1 levels, increased p21, and JNK phosphorylation[93]
• Inhibits growth of several tumor cell lines
• Well tolerated in adult patients with advanced solid tumors after oral administration[93]
• Protects against oxidative stress-induced damage in retinal pigmented epithelium (RPE) cells[94]
(−)Parthenolide NF-κB/ histone deacetylase dual inhibitor • Sesquiterpene
• Causes downstream PI3K/AKT pathway inhibition, reducing cell proliferation and inducing apoptosis in glioblastoma as well as several adult tumor cells[95]
• Apoptosis induction may be mediated by generation of reactive oxygen species (ROS)[95]
• Combination with the plant-derived compound okadaic acid increased oxidative stress that led to apoptosis in Y79 retinoblastoma cells mediated by PTEN[96]

Local administration b

YM155 Survivin inhibitor • Inhibits transcription and expression of survivin, a member of the inhibitor of apoptosis family that also regulates mitosis, DNA damage repair and autophagy[97, 98]
• Survivin overexpression is associated with poor prognosis and chemoresistance in other cancers, in which its inhibition slows tumor growth in preclinical models[97, 98]
• Survivin is overexpressed in retinoblastoma tumors and retinoblastoma cell cultures[83, 99]
• Suppressing survivin expression reduces Y79 cell growth and migration potential, and slows tumor growth in a xenograft model[100]
• Low toxicity, no ocular toxicity[101, 102]
• Poorly penetrates the BBB[103]
•Antitumor effects are time dependent, requiring careful attention to treatment schedule [102]
ABT-737 BH3 mimetic inhibitor • Inhibits anti-apoptotic B-cell lymphoma-2 (Bcl-2) and Bcl-xL proteins, resulting in apoptosis[104, 105] Promotes apoptosis in commercial WERIRB1 cells; relatively nontoxic to normal human photoreceptor cells[84]
• Ineffective against Y79 cells, which do not express Bax and thus, not likely to be proapoptotic inall retinoblastomas
•Navitoclax, a related compound, induces apoptosis in Y79 cells; the triple combination of this agent with topotecan and RAD51 inhibitor showed synergistic effects in vitro and in vivo[106]
• Related Bcl-2 inhibitor obatoclax caused neurologic symptoms in patients in clinical trials, calling for evaluation of these adverse events after ocular administration of this agent[107, 108]
• Local administration recommended because of severe thrombocytopenia reported in humans with navitoclax, probably resulting from on-target BclxL inhibition[109]
BIX01294 and UNC0631 G9a/ G9a-like protein (GLP) • G9a and GLP are histone methyltransferases and specifically methylate the non-histone target p53 at lysine 373, leading to its inactivation[110, 111]
inhibitor • G9a is aberrantly regulated in multiple tumors[110]
• No evidence of methyltransferase dysregulation in retinoblastoma to date
Ispinesib, ARQ621, and SB743921 Kinesin spindle protein (KSP) inhibitor • Part of a promising family of antimitotic compounds that may circumvent current limitations of the antimitotic taxanes such as neurotoxicity and resistance
• Overexpression of kinesin spindle (KIF) proteins, in particular KIF14, has been associated with tumor growth and poor prognosis in several cancers, but its role in tumor development and progression is still unclear[112]
• These agents also inhibit Eg5, have shown cytotoxic activity against preclinical models, and have been evaluated human Phase I/II clinical studies in other cancers[113, 114]
• KIF14 overexpression is among the most frequent aberrations in retinoblastoma, observed in almost 50% of cases[115].
• Overexpression of KIF14 in a transgenic model of retinoblastoma promoted tumor formation and enhanced tumor burden[116]
NH125 Eukaryotic elongation factor 2 (eEF-2) inhibitor •Eukaryotic elongation factor-2 kinase (eEF2K) inactivates eEF2, limiting protein synthesis under stress[117]
• Activation of eEF2K has been detected in cells under stress such as nutrient deficiency and promotes cell survival by inhibiting apoptosis[117]
• eEF2K overexpression in some adult tumors has been associated with tumor growth and poor patient survival[117, 118]
• This agent has cytotoxic activity and causes apoptosis in several adult tumor cell lines[119]
NSC 319726 p53 reactivator •Cytotoxic in cells and mouse tumor models carrying the TP53R175H mutation, one of the most frequent TP53 mutations that causes gain-of-function by impairing Zn binding dependence for DNA binding [120]
• Functions as a zinc ionophore, delivering the metal ion to the mutant p53 and restoring wildtype conformation[120]
• Increases intracellular reactive oxygen species by this transcriptional activation of p53-R175H[121]
• Other mechanism for cytotoxicity in wild-type p53 cells involve binding to copper and boost of intracellular reactive oxygen species resulting in cell growth arrest[122].
BAY 11–7085 NF-κB (nuclear factor κB) α (IκBα) inhibitor • NF-kB transcription factors are associated with tumor growth and survival in addition to key function in innate immunity[123]
• In retinoblastoma, NF-kB is overexpressed compared with normal retin[124]
• Inhibition of NF-kB induces in vitro retinoblastoma cell apoptosis and sensitizes cells to doxorubicin [125]
SBI-0640756 Eukaryotic translation initiation factor 4 gamma 1 (EIF4G1) inhibitor • EIF4G1 is a key component of the eIF4F protein complex that mediates initiation of mRNA translation
• Several tumor cell lines overexpress EIF4G1 mRNA[126]
• Exerts anti-proliferative effects in human melanoma cell lines[127]
Stattic Signal transducer and activator of transcription 3 (STAT3) inhibitor • STAT3 is a transcription factor that regulates cell cycle progression, survival, and inflammation[128]. STAT3 is constitutively activated in several human cancers and hyperphosphorylation has been associated with aggressiveness and immune evasion[129]
•STAT3 is upregulated in retinoblastoma; STAT3 knockdown inhibits Y79 cell proliferation and suppresses tumor growth in vivo[130]
• Non-toxic in RPE cells[131]
a

Able to cross the blood-brain or blood-ocular barriers and not associated with serious adverse events

b

Cannot penetrate the blood-brain or blood-ocular barriers, or evidence that they can is not available; not associated with serious adverse events.

The remaining 43 agents were considered for local administration (IVi), from which we selected 12 compounds without vesicant or neurotoxic effects (reported for the compounds themselves or the pharmacological family to which they belong), and for which antitumor activity has been reported in models of adult or pediatric cancer. The final selection included inhibitors of survivin, antiapoptotic Bcl-2 family proteins, methyltransferase, and kinesin proteins, among others (Figure 1, right flowchart; full list with mechanisms of action in Table 2). Most of these hits’ mechanisms of action have not yet been determined in retinoblastoma, but the variety of cellular processes implicated suggests that at least some may improve tumor control.

Discussion

We report a drug selection process based on a rational, unbiased design to identify and prioritize potentially active drugs adapted to the common clinical scenarios for the treatment of retinoblastoma. This process, based on a strategy used to prioritize novel treatment combination for brain tumors [17, 29, 42, 43] combines pharmacological screening results with available data on clinical use and phase I/II trials, safety, and biopharmaceutical characteristics. Identified candidate drugs are then categorized according to their feasibility for use in various clinical scenarios (i.e., administration routes). Our screen and literature review identified 31 drugs worthy of further investigation in retinoblastoma, of which 15 have been clinically characterized and 16 that deserve further analysis.

Of note, three agents used as standard-of-care for retinoblastoma treatment were excluded during the decision process, implying that our classification prioritized drugs that demonstrated greater potency even compared with clinically used agents. Specifically, carboplatin and doxorubicin did not reach the cytotoxicity threshold of inhibiting cell growth by > 70% at the evaluated concentration, in line with previous reports[18, 44]. Vincristine was omitted due to its poor penetrance across the BBB and evidence of retinal toxicity after intravitreal injection in primates[4548].

Eight compounds fulfilled the decision criteria for consideration as IVi-administered drugs. Three of these drugs are already used as standard therapy for retinoblastoma. Two others, teniposide and gemcitabine are used to treat pediatric tumors by the i.v. route[26, 49]. A newer drug with potential for IVi administration to treat retinoblastoma was the HDACi romidepsin. HDACis are a promising class of anticancer agents because their cytotoxic effects are mediated by epigenetic regulation, leading to selective activity against cells with deregulated E2F1 activity, as occurs in retinoblastoma due to loss of pRb[50, 51]. In fact, the cytotoxic effects of HDACis were previously reported in patient-derived and murine retinoblastoma cell lines[51]. The remaining two IVi candidates were digoxin and methylene blue. The preclinical profile of digoxin was previously reported by our group, showing a favorable local disposition after intravitreal injection in a rabbit model, but local retinal toxicity warrants further study prior to clinical translation[22, 52]. For methylene blue, a dye used for sentinel lymph node mapping for breast cancer and management of drug-induced methemoglobinemia, there is ample preclinical evidence of antitumor effects that may be potentiated by photodynamic therapy[53, 54]. Its affinity for pigments such as melanin and potentially for the retinal pigmented epithelium (RPE) may add activity against the tumor but also unwanted retinal toxicity[55].

The other local administration route considered in our prioritization system is IT/IVt administration, a means of bypassing the blood-brain and blood-cerebrospinal fluid barriers [26, 27, 29, 30, 5658] to target leptomeningeal dissemination, a common and usually fatal dissemination site for retinoblastoma, which is challenging given the limited ability of chemotherapeutics that cross the BBB[1, 5, 8, 9]. Our selection process for IT/IVt administration, among the candidates listed above for the IVi route, excluded four drugs for which toxicity after IT/IVt administration has been reported[26, 38, 59]. Of the remaining four candidates, etoposide and topotecan have been given via the IT/IVt route to treat pediatric brain tumors. IVt etoposide has been used in children with metastatic brain tumors or neoplastic meningitis with good tolerability and favorable pharmacokinetics in CSF despite poor penetration into the parenchyma in some patients[26, 60, 61]. IT topotecan is currently used in retinoblastoma patients with histopathological features associated with high risk for CNS relapse[62, 63]. The remaining candidates, teniposide and romidepsin, have not been given IT/IVt to humans, though the IT safety of the HDACi panobinostat, in the same family as romidepsin, is supported by a study in nonhuman primates[64].

Most of the 12 drugs identified for systemic treatment of metastatic retinoblastoma (i.e. that can penetrate the BBB/BOB) have long been used for pediatric solid tumors (e.g. gemcitabine, irinotecan) or even for retinoblastoma (e.g. etoposide, topotecan), except for the two HDACi (vorinostat and panobinostat)[51, 6568]. Romidepsin was excluded as a candidate for systemic treatment based on its cardiac effects, association with electrolyte imbalance, and poor CSF penetration [6971].

Two new hits identified for the systemic route are noteworthy for the novelty of their mechanisms. Selinexor is an XPO1 inhibitor that alters nuclear-cytoplasmic export of cargo proteins, such as Rb, E2F1, and p53, leading to mislocalization and promoting cell survival and tumor progression[72, 73]. Preclinical evaluations of this drug by the Pediatric Preclinical Testing Program against a panel of cell lines and in vivo xenografts derived from solid and hematological malignancies showed promising responses[73]. This hit remained in the selection process despite reports of neurotoxicity in treated children at the maximum studied dose as this did not occur at a lower dose of 50 mg/m2 and even lower doses could be used to avoid other grade 3 toxicities[74]. The other hit, ceritinib, is an FDA-approved ALK inhibitor able to cross the BBB, making it a promising candidate for targeting brain tumors[75, 76]. Ceritinib also inhibits several other targets, including the type 1 insulin-like growth factor receptor (IGF1R) and insulin receptor (IR)[77]. The IGF pathway has long been implicated in tumor cell proliferation and survival and its activation has been reported in pediatric brain tumors, likely relating to the high expression of IGF1R and its ligands during embryonic development[76, 78]. Y79 retinoblastoma cells produce IGF and display autocrine IGF1R-mediated growth, supporting the likely efficacy of ceritinib in this cancer[79]. Nonetheless, aberrations in ALK in retinoblastoma should be further explored.

Among the candidates for systemic administration, 6 have an aqueous formulation, lack vesicant effects, and do not require metabolic activation and were thus prioritized for IAC. Most of these compounds are already used in the clinic to treat intraocular retinoblastoma except for methylene blue.

Novel agents in the early stages of drug development may be a source of alternative compounds. We identified 16 drugs that showed activity against our primary cell cultures and are not associated with severe toxicity. Of these, four are able to cross the BBB/BOB and thus could be further studied for their applicability in the systemic treatment of retinoblastoma[8082]. These agents display a variety of mechanisms of action including transcytosis, NF-kB/HDAC inhibition, and NRF2 activation (Table 2). The 12 hits that poorly penetrate the BOB/BBB and could be delivered by IVi or IT/IVt have similarly varied mechanisms, such as inhibition of survivin, histone methyltransferases, and kinesin spindle and antiapoptotic proteins, and p53 reactivation (Table 2). Among these compounds, the survivin inhibitor YM155 and the antiapoptotic protein inhibitor ABT737 have reported activity in retinoblastoma preclinical models[83, 84]. Other compounds have not been studied in retinoblastoma but show antitumor activity in pediatric and adult solid tumor models.

The present results may not be definitive, as a larger number of primary cell lines should be tested to characterize potential diversity in pharmacological response based on inter-individual variability in genomic features and drug sensitivity, but unfortunately retinoblastoma cells are difficult to grow in culture. Also, non-tumor cell lines should be screened to evaluate potential non-specific side effects of the prioritized compounds. Another limitation is that we based cytotoxicity on cell growth inhibition at a single concentration; further studies should characterize the complete dose-response profile for the intended candidates. Finally, we may have excluded some potentially active candidates because of toxicity at the reported concentrations; some of these may be effective at potentially safer and lower doses. One such drug was carfilzomib, a potent proteasome inhibitor that presents a lower incidence of peripheral neuropathy than bortezomib [85, 86]. Other compounds were ruled out due to scarce available information, especially regarding safety for IT/IVt use. Belinostat, an FDA-approved HDACi, was excluded because of potential cardiotoxicity that prevents i.v. administration and the incidence of severe neurologic toxicity that prohibits local administration[36, 87]. Nonetheless, our stringent criteria enhance our confidence that we selected compounds with the most promising features for development for use in retinoblastoma.

In conclusion, we developed a system for drug prioritization for retinoblastoma based on decision trees incorporating information from large-scale phenotypic screening of in vitro activity in patient-derived tumor cell lines, published clinical and preclinical evidence, and biopharmaceutical characteristics. This system identified and prioritized 36 compounds worthy of further study to determine safety and efficacy in this rare pediatric cancer.

Supplementary Material

1733825_Sup_material

Acknowledgments

The authors would like to thank Maximiliano Distefano, Hospital de Pediatria JP Garrahan (Argentina) for technical support.

Funding: This work was supported by Fund for Ophthalmic Knowledge, NY, USA; Fundación Natalie Dafne Flexer de Ayuda al Niño con Cancer, Argentina; Fundación Bunge y Born, Argentina; Fondation Nelia et Amadeo Barletta; National Agency for Science and Technology Promotion (PIDC 2014–0043; PICT 2016–1505), Argentina; and by NIH/NCI Cancer Center Support Grant P30 CA008748 to Memorial Sloan Kettering Cancer Center.

Footnotes

Declarations

Conflicts of interest/Competing interests: All authors declare that they have no competing interests.

Availability of data and materials: all data generated or analyzed during this study are included in this article.

Code availability: Not applicable

Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of a an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.

References

  • 1.Munier FL, Beck-Popovic M, Chantada GL, et al. (2019) Conservative management of retinoblastoma: Challenging orthodoxy without compromising the state of metastatic grace. “Alive, with good vision and no comorbidity.” Prog Retin Eye Res 73:100764. 10.1016/j.preteyeres.2019.05.005 [DOI] [PubMed] [Google Scholar]
  • 2.Abramson DH, Ji X, Francis JH, et al. (2019) Intravitreal chemotherapy in retinoblastoma: Expanded use beyond intravitreal seeds. Br J Ophthalmol 103:488–493. 10.1136/bjophthalmol-2018-312037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Francis JH, Levin AM, Zabor EC, et al. (2018) Ten-year experience with ophthalmic artery chemosurgery: Ocular and recurrence-free survival. PLoS One 13:e0197081. 10.1371/journal.pone.0197081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Abramson DH, Fabius AWM, Issa R, et al. (2015) Advanced unilateral retinoblastoma: The impact of ophthalmic artery chemosurgery on Enucleation rate and patient survival at MSKCC. PLoS One 10:e0145436. 10.1371/journal.pone.0145436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Fabian ID, Abdallah E, Abdullahi SU, et al. (2020) Global Retinoblastoma Presentation and Analysis by National Income Level. JAMA Oncol 6:685–695. 10.1001/jamaoncol.2019.6716 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Canturk S, Qaddoumi I, Khetan V, et al. (2010) Survival of retinoblastoma in less-developed countries impact of socioeconomic and health-related indicators. Br J Ophthalmol 94:1432–1436. 10.1136/bjo.2009.168062 [DOI] [PubMed] [Google Scholar]
  • 7.Chawla B, Hasan F, Azad R, et al. (2016) Clinical presentation and survival of retinoblastoma in Indian children. Br. J. Ophthalmol. 100:172–178 [DOI] [PubMed] [Google Scholar]
  • 8.Dunkel IJ, Krailo MD, Chantada GL, et al. (2017) Intensive multi-modality therapy for extra-ocular retinoblastoma (RB): A Children’s Oncology Group (COG) trial (ARET0321). J Clin Oncol 35:10506–10506. 10.1200/jco.2017.35.15_suppl.10506 [DOI] [Google Scholar]
  • 9.Dunkel IJ, Chan HSL, Jubran R, et al. (2010) High-dose chemotherapy with autologous hematopoietic stem cell rescue for stage 4b retinoblastoma. Pediatr Blood Cancer 55:149–152. 10.1002/pbc.22491 [DOI] [PubMed] [Google Scholar]
  • 10.Abramson DH, Francis JH, Gobin YP (2019) What’s new in intra-arterial chemotherapy for retinoblastoma? Int Ophthalmol Clin 59:87–94. 10.1097/IIO.0000000000000266 [DOI] [PubMed] [Google Scholar]
  • 11.Taich P, Ceciliano A, Buitrago E, et al. (2014) Clinical pharmacokinetics of intra-arterial melphalan and topotecan combination in patients with retinoblastoma. Ophthalmology 121:889–897. 10.1016/j.ophtha.2013.10.045 [DOI] [PubMed] [Google Scholar]
  • 12.Schaiquevich P, Buitrago E, Taich P, et al. (2012) Pharmacokinetic analysis of melphalan after superselective ophthalmic artery infusion in preclinical models and retinoblastoma patients. Investig Ophthalmol Vis Sci 53:4205–4212. 10.1167/iovs.12-9501 [DOI] [PubMed] [Google Scholar]
  • 13.Schaiquevich P, Buitrago E, Ceciliano A, et al. (2012) Pharmacokinetic analysis of topotecan after superselective ophthalmic artery infusion and periocular administration in a porcine model. Retina 32:387–395. 10.1097/IAE.0b013e31821e9f8a [DOI] [PubMed] [Google Scholar]
  • 14.Pascual-Pasto G, Olaciregui NG, Vila-Ubach M, et al. (2016) Preclinical platform of retinoblastoma xenografts recapitulating human disease and molecular markers of dissemination. Cancer Lett 380:10–19. 10.1016/j.canlet.2016.06.012 [DOI] [PubMed] [Google Scholar]
  • 15.Ben-David U, Gan QF, Golan-Lev T, et al. (2013) Selective elimination of human pluripotent stem cells by an oleate synthesis inhibitor discovered in a high-throughput screen. Cell Stem Cell 12:167–179. 10.1016/j.stem.2012.11.015 [DOI] [PubMed] [Google Scholar]
  • 16.Aherne W, Garrett M, McDonald T, Workman P (2002) Mechanism-based high-throughput screening for novel anticancer drug discovery. In: Anticancer Drug Development. Elsevier, pp 249–267 [Google Scholar]
  • 17.Nimmervoll B V, Boulos N, Bianski B, et al. (2018) Establishing a preclinical multidisciplinary board for brain tumors. Clin Cancer Res 24:1654–1666. 10.1158/1078-0432.CCR-17-2168 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zugbi S, Winter U, Castañon A, et al. (2019) Comparison of the pharmacological activity of idarubicin and doxorubicin for retinoblastoma. Pediatr Blood Cancer 66:e27441. 10.1002/pbc.27441 [DOI] [PubMed] [Google Scholar]
  • 19.Winter U, Aschero R, Fuentes F, et al. (2019) Tridimensional retinoblastoma cultures as vitreous seeds models for live-cell imaging of chemotherapy penetration. Int J Mol Sci 20:1077. 10.3390/ijms20051077 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Winter U, Ganiewich D, Ottaviani D, et al. (2020) Genomic and transcriptomic tumor heterogeneity in bilateral retinoblastoma. JAMA Ophthalmol 138:569–574. 10.1001/jamaophthalmol.2020.0427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Winter U, Mena HA, Negrotto S, et al. (2016) Schedule-dependent antiangiogenic and cytotoxic effects of chemotherapy on vascular endothelial and retinoblastoma cells. PLoS One 11:e0160094. 10.1371/journal.pone.0160094 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Antczak C, Kloepping C, Radu C, et al. (2009) Revisiting old drugs as novel agents for retinoblastoma: In vitro and in vivo antitumor activity of cardenolides. Investig Ophthalmol Vis Sci 50:3065–3073. 10.1167/iovs.08-3158 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zhang J-H (1999) A Simple Statistical Parameter for Use in Evaluation and Validation of High Throughput Screening Assays. J Biomol Screen 4:67–73. 10.1177/108705719900400206 [DOI] [PubMed] [Google Scholar]
  • 24.Zugbi S, Ganiewich D, Bhattacharyya A, et al. (2020) Clinical, Genomic, and Pharmacological Study of MYCN-Amplified RB1 Wild-Type Metastatic Retinoblastoma. Cancers (Basel) 12:2714. 10.3390/cancers12092714 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Inomata M, Kaneko A (1987) Chemosensitivity Profiles of Primary and Cultured Human Retinoblastoma Cells in a Human Tumor Clonogenic Assay. Jpn J Cancer Res 78:858–868 [PubMed] [Google Scholar]
  • 26.Conroy S, Garnett M, Vloeberghs M, et al. (2010) Medulloblastoma in childhood: Revisiting Intrathecal therapy in infants and children. Cancer Chemother Pharmacol 65:1173–1189. 10.1007/s00280-009-1127-1 [DOI] [PubMed] [Google Scholar]
  • 27.Stapleton S, Blaney S (2006) New Agents for Intrathecal Administration. Cancer Invest 24:528–534. 10.1080/07357900600815166 [DOI] [PubMed] [Google Scholar]
  • 28.Cook AM, Mieure KD, Owen RD, et al. (2009) Intracerebroventricular Administration of Drugs. Pharmacotherapy 29:832–845. 10.1592/phco.29.7.832 [DOI] [PubMed] [Google Scholar]
  • 29.Motl S, Zhuang Y, Waters CM, Stewart CF (2006) Pharmacokinetic considerations in the treatment of CNS tumours. Clin Pharmacokinet 45:871–903. 10.2165/00003088-200645090-00002 [DOI] [PubMed] [Google Scholar]
  • 30.Atkinson AJ (2017) Intracerebroventricular drug administration. Transl Clin Pharmacol 25:117–124. 10.12793/tcp.2017.25.3.117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Cunha-Vaz JG (2004) The blood-retinal barriers system. Basic concepts and clinical evaluation. Exp Eye Res 78:715–721. 10.1016/S0014-4835(03)00213-6 [DOI] [PubMed] [Google Scholar]
  • 32.Vellonen KS, Soini EM, Del Amo EM, Urtti A (2016) Prediction of ocular drug distribution from systemic blood circulation. Mol Pharm 13:2906–2911. 10.1021/acs.molpharmaceut.5b00729 [DOI] [PubMed] [Google Scholar]
  • 33.Abdul Razzak R, Florence GJ, Gunn-Moore FJ (2019) Approaches to CNS Drug Delivery with a Focus on Transporter-Mediated Transcytosis. Int J Mol Sci 20:3108. 10.3390/ijms20123108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Common Terminology Criteria for Adverse Events (CTCAE) version 5.0. https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_8.5×11.pdf.Accessed 13 Jul 2020
  • 35.FDA/CEDR resources page. Food and Drug Administration Web site. https://www.accessdata.fda.gov/scripts/cder/daf/.Accessed 13 Jul 2020
  • 36.(2015) Micromedex Solution. In: Truven Heal. Anal. Inc. http://www.micromedexsolutions.com [Google Scholar]
  • 37.Porter R, Kaplan J Merck Manual. In: Kenilworth, NJ Merck Manuals. https://www.merckmanuals.com/home.Accessed 13 Jul 2020 [Google Scholar]
  • 38.Garcia E, Calello D, Ruck B, Nelson L (2017) Abstracts from the 2017 American College of Medical Toxicology (ACMT) Annual Scientific Meeting. J Med Toxicol 13:3–46. 10.1007/s13181-017-0599-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bernardi RJ, Bomgaars L, Fox E, et al. (2008) Phase I clinical trial of intrathecal gemcitabine in patients with neoplastic meningitis. Cancer Chemother Pharmacol 62:355–361. 10.1007/s00280-007-0601-x [DOI] [PubMed] [Google Scholar]
  • 40.Kreidieh FY (2016) Overview, prevention and management of chemotherapy extravasation. World J Clin Oncol 7:87. 10.5306/wjco.v7.i1.87 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Dunkel IJ, Shi W, Salvaggio K, et al. (2014) Risk factors for severe neutropenia following intra-arterial chemotherapy for intra-ocular retinoblastoma. PLoS One 9:e108692. 10.1371/journal.pone.0108692 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Fletcher JI, Ziegler DS, Trahair TN, et al. (2018) Too many targets, not enough patients: rethinking neuroblastoma clinical trials. Nat Rev Cancer 18:389–400. 10.1038/s41568-018-0003-x [DOI] [PubMed] [Google Scholar]
  • 43.Schellens JHM, McLeod HL, Newell DR (2005) Cancer Clinical Pharmacology, 1st ed. Oxford University Press [Google Scholar]
  • 44.Laurie NA, Gray JK, Zhang J, et al. (2005) Topotecan combination chemotherapy in two new rodent models of retinoblastoma. Clin Cancer Res 11:7569–7578. 10.1158/1078-0432.CCR-05-0849 [DOI] [PubMed] [Google Scholar]
  • 45.Dareer SM El White VM, Chen FP, et al. (1977) Distribution and Metabolism of Vincristine in Mice, Rats, Dogs, and Monkeys. Cancer Treat Rep 61:1269–77 [PubMed] [Google Scholar]
  • 46.Phoenix TN, Patmore DM, Boop S, et al. (2016) Medulloblastoma Genotype Dictates Blood Brain Barrier Phenotype. Cancer Cell 29:508–522. 10.1016/j.ccell.2016.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jr DVJ, Sethi VS, Spurr CL, McWhorter JM (1981) Pharmacokinetics of Vincristine in the Cerebrospinal Fluid of Humans. Cancer Res 41:1466–8 [PubMed] [Google Scholar]
  • 48.Green WR (1975) Retinal and optic nerve atrophy induced by intravitreous vincristine in the primate. Trans Am Ophthalmol Soc 73:389–416 [PMC free article] [PubMed] [Google Scholar]
  • 49.Blatt J, Farag S, Corey SJ, et al. (2014) Expanding the scope of drug repurposing in pediatrics: The Children’s Pharmacy Collaborative™. Drug Discov Today 19:1696–1698. 10.1016/j.drudis.2014.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Zhao Y, Tan J, Zhuang L, et al. (2005) Inhibitors of histone deacetylases target the RbE2F1 pathway for apoptosis induction through activation of proapoptotic protein Bim. Proc Natl Acad Sci U S A 102:16090–16095. 10.1073/pnas.0505585102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Dalgard CL, Van Quill KR, O’Brien JM (2008) Evaluation of the in vitro and in vivo antitumor activity of histone deacetylase inhibitors for the therapy of retinoblastoma. Clin Cancer Res 14:3113–3123. 10.1158/1078-0432.CCR-07-4836 [DOI] [PubMed] [Google Scholar]
  • 52.Winter U, Buitrago E, Mena HA, et al. (2015) Pharmacokinetics, safety, and efficacy of intravitreal digoxin in preclinical models for retinoblastoma. Investig Ophthalmol Vis Sci 56:4382–4393. 10.1167/iovs.14-16239 [DOI] [PubMed] [Google Scholar]
  • 53.Kofler B, Romani A, Pritz C, et al. (2018) Photodynamic effect of methylene blue and low level laser radiation in head and neck squamous cell carcinoma cell lines. Int J Mol Sci 19:1107. 10.3390/ijms19041107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Lim EJ, Oak CH, Heo J, Kim YH (2013) Methylene blue-mediated photodynamic therapy enhances apoptosis in lung cancer cells. Oncol Rep 30:856–862. 10.3892/or.2013.2494 [DOI] [PubMed] [Google Scholar]
  • 55.Tardivo JP, Del Giglio A, De Oliveira CS, et al. (2005) Methylene blue in photodynamic therapy: From basic mechanisms to clinical applications. Photodiagnosis Photodyn Ther 2:175–191. 10.1016/S1572-1000(05)00097-9 [DOI] [PubMed] [Google Scholar]
  • 56.Triarico S, Maurizi P, Mastrangelo S, et al. (2019) Improving the Brain Delivery of Chemotherapeutic Drugs in Childhood Brain Tumors. Cancers (Basel) 11:824. 10.3390/cancers11060824 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Dimaras H, Hon E, Budning A, et al. (2009) Retinoblastoma CSF metastasis cured by multimodality chemotherapy without radiation. Ophthalmic Genet 30:121–126. 10.1080/13816810902988780 [DOI] [PubMed] [Google Scholar]
  • 58.Blaney SM, Tagen M, Onar-Thomas A, et al. (2013) A phase-1 pharmacokinetic optimal dosing study of intraventricular topotecan for children with neoplastic meningitis: A pediatric brain tumor consortium study. Pediatr Blood Cancer 60:627–632. 10.1002/pbc.24309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Berg S, Balis FM, McCully CL, Poplack DC (1994) Re: Toxicity of Intrathecal Melphalan. JNCI 86:463–463. 10.1093/jnci/86.6.463 [DOI] [PubMed] [Google Scholar]
  • 60.Fleischhack G, Reif S, Hasan C, et al. (2001) Feasibility of intraventricular administration of etoposide in patients with metastatic brain tumours. Br J Cancer 84:1453–1459. 10.1054/bjoc.2001.1841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Aguado Virginia, Lovett David, Meijer Lisethe Dr, et al. (2017) Intrathecal administration of etoposide: a review of current literature and expert opinion | Document summary | Evidence search | NICE. https://www.evidence.nhs.uk/document?id=1620070&returnUrl=Search%3Fps%3D50%26q%3DCerebrospinal%2Bfluid&q=Cerebrospinal+fluid. Accessed 10 Jul 2020 [Google Scholar]
  • 62.Sampor C, Correa Llanos MG, Oller A, et al. (2019) Intrathecal Topotecan as Part of Retinoblastoma Treatment. In: Pediatric Blood & Cancer. NLM (Medline), p e27989 [Google Scholar]
  • 63.Laurent VE, Torbidoni AV, Sampor C, et al. (2016) Minimal disseminated disease in nonmetastatic retinoblastoma with high-risk pathologic features and association with disease-free survival. JAMA Ophthalmol 134:1374–1379. 10.1001/jamaophthalmol.2016.4158 [DOI] [PubMed] [Google Scholar]
  • 64.Sandberg DI, Kharas N, Yu B, et al. (2020) High-dose MTX110 (soluble panobinostat) safely administered into the fourth ventricle in a nonhuman primate model. J Neurosurg Pediatr 1:1–9. 10.3171/2020.2.peds19786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Fouladi M, Park JR, Stewart CF, et al. (2010) Pediatric phase I trial and pharmacokinetic study of vorinostat: A children’s oncology group phase I consortium report. J Clin Oncol 28:3623–3629. 10.1200/JCO.2009.25.9119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Pinto N, DuBois SG, Marachelian A, et al. (2018) Phase I study of vorinostat in combination with isotretinoin in patients with refractory/recurrent neuroblastoma: A new approaches to Neuroblastoma Therapy (NANT) trial. Pediatr Blood Cancer 65:e27023. 10.1002/pbc.27023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Muscal JA, Thompson PA, Horton TM, et al. (2013) A phase I trial of vorinostat and bortezomib in children with refractory or recurrent solid tumors: A Children’s Oncology Group phase I consortium study (ADVL0916). Pediatr Blood Cancer 60:390–395. 10.1002/pbc.24271 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Meel MH, de Gooijer MC, Metselaar DS, et al. (2020) Combined Therapy of AXL and HDAC Inhibition Reverses Mesenchymal Transition in Diffuse Intrinsic Pontine Glioma. Clin Cancer Res 26:3319–3332. 10.1158/1078-0432.ccr-19-3538 [DOI] [PubMed] [Google Scholar]
  • 69.Noonan AM, Eisch RA, Liewehr DJ, et al. (2013) Electrocardiographic studies of romidepsin demonstrate its safety and identify a potential role for KATP channel. Clin Cancer Res 19:3095–3104. 10.1158/1078-0432.CCR-13-0109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Berg SL, Stone J, Xiao JJ, et al. (2004) Plasma and cerebrospinal fluid pharmacokinetics of depsipeptide (FR901228) in nonhuman primates. Cancer Chemother Pharmacol 54:85–88. 10.1007/s00280-004-0766-5 [DOI] [PubMed] [Google Scholar]
  • 71.Fouladi M, Furman WL, Chin T, et al. (2006) Phase I study of depsipeptide in pediatric patients with refractory solid tumors: A children’s oncology group report. J Clin Oncol 24:3678–3685. 10.1200/JCO.2006.06.4964 [DOI] [PubMed] [Google Scholar]
  • 72.Kent LN, Leone G (2019) The broken cycle: E2F dysfunction in cancer. Nat Rev Cancer 19:326–338. 10.1038/s41568-019-0143-7 [DOI] [PubMed] [Google Scholar]
  • 73.Attiyeh EF, Maris JM, Lock R, et al. (2016) Pharmacodynamic and genomic markers associated with response to the XPO1/CRM1 inhibitor selinexor (KPT-330): A report from the pediatric preclinical testing program. Pediatr Blood Cancer 63:276–286. 10.1002/pbc.25727 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Alexander TB, Lacayo NJ, Choi JK, et al. (2016) Phase I study of selinexor, a selective inhibitor of nuclear export, in combination with fludarabine and cytarabine, in pediatric relapsed or refractory acute leukemia. J Clin Oncol 34:4094–4101. 10.1200/JCO.2016.67.5066 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tsoli M, Wadham C, Pinese M, et al. (2018) Integration of genomics, high throughput drug screening, and personalized xenograft models as a novel precision medicine paradigm for high risk pediatric cancer. Cancer Biol Ther 19:1078–1087. 10.1080/15384047.2018.1491498 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Russo A, Paret C, Alt F, et al. (2019) Ceritinib-induced regression of an insulin-like growth factor-driven neuroepithelial brain tumor. Int J Mol Sci 20:4267. 10.3390/ijms20174267 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.De Pas T, Pala L, Catania C, Conforti F (2017) Molecular and clinical features of second-generation anaplastic lymphoma kinase inhibitors: Ceritinib. Futur Oncol 13:2629–2644. 10.2217/fon-2017-0262 [DOI] [PubMed] [Google Scholar]
  • 78.Lodhia KA, Tienchaiananda P, Haluska P (2015) Understanding the Key to Targeting the IGF Axis in Cancer: A Biomarker Assessment. Front Oncol 5:142. 10.3389/fonc.2015.00142 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Giuliano M, Vento R, Lauricella M, et al. (1996) Role of Insulin-Like Growth Factors in Autocrine Growth of Human Retinoblastoma Y79 Cells. Eur J Biochem 236:523–532. 10.1111/j.1432-1033.1996.00523.x [DOI] [PubMed] [Google Scholar]
  • 80.Zhang Y, Simpson-Durand CD, Standifer KM (2015) Nociceptin/orphanin FQ peptide receptor antagonist JTC-801 reverses pain and anxiety symptoms in a rat model of post-traumatic stress disorder. Br J Pharmacol 172:571–582. 10.1111/bph.12701 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Reisman SA, Gahir SS, Lee CYI, et al. (2019) Pharmacokinetics and pharmacodynamics of the novel NrF2 activator omaveloxolone in primates. Drug Des Devel Ther 13:1259–1270. 10.2147/DDDT.S193889 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Dong L, Qiao H, Zhang X, et al. (2013) Parthenolide Is Neuroprotective in Rat Experimental Stroke Model: Downregulating NF-ΚB, Phospho-P38MAPK, and Caspase1 and Ameliorating BBB Permeability. Mediat Inflamm 2013:370804. 10.1155/2013/370804 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Ferrario A, Luna M, Rucker N, et al. (2016) Targeting Survivin Enhances Chemosensitivity in Retinoblastoma Cells and Orthotopic Tumors. PLoS One 11:e0153011. 10.1371/journal.pone.0153011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Allaman-Pillet N, Oberson A, Munier F, Schorderet DF (2013) The Bcl-2/Bcl-XL inhibitor ABT-737 promotes death of retinoblastoma cancer cells. Ophthalmic Genet 34:1–13. 10.3109/13816810.2011.615077 [DOI] [PubMed] [Google Scholar]
  • 85.Franken B, van de Donk NWCJ, Cloos JC, et al. (2016) A clinical update on the role of carfilzomib in the treatment of relapsed or refractory multiple myeloma. Ther Adv Hematol 7:330–344. 10.1177/2040620716667275 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ailawadhi S, Sexton R, Lentzsch S, et al. (2020) Low-dose versus High-dose Carfilzomib with Dexamethasone (S1304) in Patients with Relapsed-Refractory Multiple Myeloma. Clin Cancer Res 26:3969–3978. 10.1158/1078-0432.CCR-19-1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Allen PB, Lechowicz MJ (2018) Hematologic toxicity is rare in relapsed patients treated with belinostat: A systematic review of belinostat toxicity and safety in peripheral T-cell lymphomas. Cancer Manag Res 10:6731–6742. 10.2147/CMAR.S149241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Pommepuy I, Terro F, Petit B, et al. (2003) Brefeldin A Induces Apoptosis and Cell Cycle Blockade in Glioblastoma Cell Lines. Oncology 64:459–467. 10.1159/000070307 [DOI] [PubMed] [Google Scholar]
  • 89.Larsson DE, Wickström M, Hassan S, et al. (2010) The Cytotoxic Agents NSC-95397, Brefeldin A, Bortezomib and Sanguinarine Induce Apoptosis in Neuroendocrine Tumors In Vitro. Anticancer Res 30:149–156 [PubMed] [Google Scholar]
  • 90.Miranda GE, Abrahan CE, Politi LE, Rotstein NP (2009) Sphingosine-1-phosphate is a key regulator of proliferation and differentiation in retina photoreceptors. Investig Ophthalmol Vis Sci 50:4416–4428. 10.1167/iovs.09-3388 [DOI] [PubMed] [Google Scholar]
  • 91.Song X, Zhu S, Xie Y, et al. (2018) JTC801 Induces pH-dependent Death Specifically in Cancer Cells and Slows Growth of Tumors in Mice. Gastroenterology 154:1480–1493. 10.1053/j.gastro.2017.12.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Li JX, Bi YP, Wang J, et al. (2018) JTC-801 inhibits the proliferation and metastasis of ovarian cancer cell SKOV3 through inhibition of the PI3K - AKT signaling pathway. Pharmazie 73:283–287. 10.1691/ph.2018.7326 [DOI] [PubMed] [Google Scholar]
  • 93.Creelan BC, Gabrilovich DI, Gray JE, et al. (2017) Safety, pharmacokinetics, and pharmacodynamics of oral omaveloxolone (RTA408), a synthetic triterpenoid, in a first-in-human trial of patients with advanced solid tumors. Onco Targets Ther 10:4239–4250. 10.2147/OTT.S136992 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Liu X, Ward K, Xavier C, et al. (2016) The novel triterpenoid RTA 408 protects human retinal pigment epithelial cells against H2O2-induced cell injury via NF-E2-related factor 2 (Nrf2) activation. Redox Biol 8:98–109. 10.1016/j.redox.2015.12.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Shields M (2017) Chemotherapeutics. In: Pharmacognosy: Fundamentals, Applications and Strategy, 1 st. Elsevier Inc., pp 295–313 [Google Scholar]
  • 96.Di Fiore R, Drago-Ferrante R, D’Anneo A, et al. (2013) In human retinoblastoma Y79 cells okadaic acid-parthenolide co-treatment induces synergistic apoptotic effects, with PTEN as a key player. Cancer Biol Ther 14:922–931. 10.4161/cbt.25944 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Jane EP, Premkumar DR, Sutera PA, et al. (2017) Survivin inhibitor YM155 induces mitochondrial dysfunction, autophagy, DNA damage and apoptosis in Bcl-xL silenced glioma cell lines. Mol Carcinog 56:1251–1265. 10.1002/mc.22587 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Khan Z, Khan AA, Yadav H, et al. (2017) Survivin, a molecular target for therapeutic interventions in squamous cell carcinoma. Cell Mol Biol Lett 22:8. 10.1186/s11658-017-0038-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Sudhakar J, Venkatesan N, Lakshmanan S, et al. (2013) Hypoxic tumor microenvironment in advanced retinoblastoma. Pediatr Blood Cancer 60:1598–1601. 10.1002/pbc.24599 [DOI] [PubMed] [Google Scholar]
  • 100.Liu K, Liu Y, Zhao G (2017) Targeting survivin suppresses proliferation and invasion of retinoblastoma cells in vitro and in vivo. Int J Clin Exp Pathol 10:9352–9361 [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 101.Satoh T, Okamoto I, Miyazaki M, et al. (2009) Phase I study of YM155, a novel survivin suppressant, in patients with advanced solid tumors. Clin Cancer Res 15:3872–3880. 10.1158/1078-0432.CCR-08-1946 [DOI] [PubMed] [Google Scholar]
  • 102.Tolcher AW, Mita A, Lewis LD, et al. (2008) Phase I and pharmacokinetic study of YM155, a small-molecule inhibitor of survivin. J Clin Oncol 26:5198–5203. 10.1200/JCO.2008.17.2064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Minematsu T, Sonoda T, Hashimoto T, et al. (2012) Pharmacokinetics, distribution and excretion of YM155 monobromide, a novel small-molecule survivin suppressant, in male and pregnant or lactating female rats. Biopharm Drug Dispos 33:160–169. 10.1002/bdd.1781 [DOI] [PubMed] [Google Scholar]
  • 104.Campbell KJ, Tait SWG (2018) Targeting BCL-2 regulated apoptosis in cancer. Open Biol 8:180002. 10.1098/rsob.180002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Oltersdorf T, Elmore SW, Shoemaker AR, et al. (2005) An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature 435:677–681. 10.1038/nature03579 [DOI] [PubMed] [Google Scholar]
  • 106.Aubry A, Pearson JD, Huang K, et al. (2020) Functional genomics identifies new synergistic therapies for retinoblastoma. Oncogene 39:5338–5357. 10.1038/s41388-020-1372-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Paik PK, Rudin CM, Brown A, et al. (2010) A phase I study of obatoclax mesylate, a Bcl-2 antagonist, plus topotecan in solid tumor malignancies. Cancer Chemother Pharmacol 66:1079–1085. 10.1007/s00280-010-1265-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Schimmer AD, O’Brien S, Kantarjian H, et al. (2008) A phase I study of the pan bcl-2 Family Inhibitor obatoclax mesylate in patients with advanced hematologic malignancies. Clin Cancer Res 14:8295–8301. 10.1158/1078-0432.CCR-08-0999 [DOI] [PubMed] [Google Scholar]
  • 109.Wilson WH, O’Connor OA, Czuczman MS, et al. (2010) Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: A phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity. Lancet Oncol 11:1149–1159. 10.1016/S1470-2045(10)70261-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Kaniskan HÜ, Konze KD, Jin J (2015) Selective inhibitors of protein methyltransferases. J Med Chem 58:1596–1629. 10.1021/jm501234a [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Casciello F, Windloch K, Gannon F, Lee JS (2015) Functional role of G9a histone methyltransferase in cancer. Front Immunol 6:487. 10.3389/fimmu.2015.00487 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Liu X, Gong H, Huang K (2013) Oncogenic role of kinesin proteins and targeting kinesin therapy. Cancer Sci 104:651–656. 10.1111/cas.12138 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Holen KD, Belani CP, Wilding G, et al. (2011) A first in human study of SB-743921, a kinesin spindle protein inhibitor, to determine pharmacokinetics, biologic effects and establish a recommended phase II dose. Cancer Chemother Pharmacol 67:447–454. 10.1007/s00280-010-1346-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Chen LC, Rosen LS, Iyengar T, et al. (2011) First-in-human study with ARQ 621, a novel inhibitor of Eg5: Final results from the solid tumors cohort. J Clin Oncol 29:3076–3076. 10.1200/jco.2011.29.15_suppl.3076 [DOI] [Google Scholar]
  • 115.Bowles E, Corson TW, Bayani J, et al. (2007) Profiling genomic copy number changes in retinoblastoma beyond loss of RB1. Genes Chromosom Cancer 46:118–129. 10.1002/gcc.20383 [DOI] [PubMed] [Google Scholar]
  • 116.O’Hare M, Shadmand M, Sulaiman RS, et al. (2016) Kif14 overexpression accelerates murine retinoblastoma development. Int J Cancer 139:1752–1758. 10.1002/ijc.30221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Karakas D, Ozpolat B (2020) Eukaryotic elongation factor-2 kinase (eEF2K) signaling in tumor and microenvironment as a novel molecular target. J Mol Med 98:775–787. 10.1007/s00109-020-01917-8 [DOI] [PubMed] [Google Scholar]
  • 118.Wang X, Xie J, Proud C (2017) Eukaryotic Elongation Factor 2 Kinase (eEF2K) in Cancer. Cancers (Basel) 9:162. 10.3390/cancers9120162 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Arora S, Yang J-M, Kinzy TG, et al. (2003) Identification and Characterization of an Inhibitor of Eukaryotic Elongation Factor 2 Kinase against Human Cancer Cell Lines. Cancer Res 63:6894–6899 [PubMed] [Google Scholar]
  • 120.Parrales A, Iwakuma T (2015) Targeting Oncogenic Mutant p53 for Cancer Therapy. Front Oncol 5:288. 10.3389/fonc.2015.00288 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Yu X, Blanden AR, Narayanan S, et al. (2014) Small molecule restoration of wildtype structure and function of mutant p53 using a novel zinc-metallochaperone based mechanism. Oncotarget 5:8879–8892. 10.18632/oncotarget.2432 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Shimada K, Reznik E, Stokes ME, et al. (2018) Copper-Binding Small Molecule Induces Oxidative Stress and Cell-Cycle Arrest in Glioblastoma-Patient-Derived Cells. Cell Chem Biol 25:585–594.e7. 10.1016/j.chembiol.2018.02.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Xia Y, Shen S, Verma IM (2014) NF- B, an Active Player in Human Cancers. Cancer Immunol Res 2:823–830. 10.1158/2326-6066.CIR-14-0112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Qu Y, Zhou F, Dai X, et al. (2011) Clinicopathologic significances of nuclear expression of nuclear factor-κB transcription factors in retinoblastoma. J Clin Pathol 64:695–700. 10.1136/jclinpath-2011-200017 [DOI] [PubMed] [Google Scholar]
  • 125.Poulaki V, Mitsiades CS, Joussen AM, et al. (2002) Constitutive nuclear factor-κB activity is crucial for human retinoblastoma cell viability. Am J Pathol 161:2229–2240. 10.1016/S0002-9440(10)64499-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Jaiswal PK, Koul S, Palanisamy N, Koul HK (2019) Eukaryotic Translation Initiation Factor 4 Gamma 1 (EIF4G1): A target for cancer therapeutic intervention? Cancer Cell Int 19:224. 10.1186/s12935-019-0947-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Feng Y, Pinkerton AB, Hulea L, et al. (2015) SBI-0640756 attenuates the growth of clinically unresponsive melanomas by disrupting the eIF4F translation initiation complex. Cancer Res 75:5211–5218. 10.1158/0008-5472.CAN-15-0885 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Yu H, Lee H, Herrmann A, et al. (2014) Revisiting STAT3 signalling in cancer: new and unexpected biological functions. Nat Rev Cancer 14:736–746. 10.1038/nrc3818 [DOI] [PubMed] [Google Scholar]
  • 129.Bharadwaj U, Kasembeli MM, Tweardy DJ (2016) STAT3 Inhibitors in Cancer: A Comprehensive Update. Humana Press, Cham, pp 95–161 [Google Scholar]
  • 130.Jo DH, Kim JH, Cho CS, et al. (2014) STAT3 inhibition suppresses proliferation of retinoblastoma through down-regulation of positive feedback loop of STAT3/miR-17–92 clusters. Oncotarget 5:11513–11525. 10.18632/oncotarget.2546 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Klettner A, Westhues D, Lassen J, et al. (2013) Regulation of constitutive vascular endothelial growth factor secretion in retinal pigment epithelium/choroid organ cultures: P38, nuclear factor kappaB, and the vascular endothelial growth factor receptor2/phosphatidylinositol 3 kinase pathway. Mol Vis 19:281–291 [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1733825_Sup_material

RESOURCES