Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2020 May 18;15(5):e0231588. doi: 10.1371/journal.pone.0231588

Patient-derived oral mucosa organoids as an in vitro model for methotrexate induced toxicity in pediatric acute lymphoblastic leukemia

E Driehuis 1,#, N Oosterom 2,#, S G Heil 3, I B Muller 4, M Lin 4, S Kolders 1, G Jansen 5, R de Jonge 4, R Pieters 2, H Clevers 1,2, M M van den Heuvel-Eibrink 2,*
Editor: Obul Reddy Bandapalli6
PMCID: PMC7233536  PMID: 32421698

Abstract

We have recently established a protocol to grow wildtype human oral mucosa organoids. These three-dimensional structures can be maintained in culture long-term, do not require immortalization, and recapitulate the multilayered composition of the epithelial lining of the oral mucosa. Here, we validate the use of this model to study the effect of Leucovorin (LV) on Methotrexate (MTX)-induced toxicity. MTX is a chemotherapeutic agent used in the treatment of pediatric acute lymphoblastic leukemia. Although effective, the use of MTX often results in severe side-effects, including oral mucositis, which is characterized by epithelial cell death. Here, we show that organoids are sensitive to MTX, and that the addition of LV reduces MTX toxicity, in both a concentration- and timing-dependent manner. Additionally, we show that a 24 hour ‘pretreatment’ with LV reduces MTX-induced cell death, suggesting that such a pretreatment could decrease mucositis in patients. Taken together, we provide the first in vitro model to study the effect of MTX on wildtype oral mucosa cells. Our findings underscore the relevance of the clinically applied LV regimen and highlight the potential of this model to further optimize modifications in dosing and timing of Leucovorin on oral mucosa cells.

Introduction

High-dose methotrexate (HD-MTX) is an important antifolate chemotherapeutic agent used in pediatric acute lymphoblastic leukemia (ALL) therapy. Currently, five-year survival rates of pediatric ALL have reached 90% in developed countries [14]. However, patients often suffer from MTX toxicities such as hepatotoxicity, nephrotoxicity, hematological malignancies and intestinal and oral mucositis [5]. Despite administration of folinic acid (Leucovorin—LV) after HD-MTX infusion, 20% of patients develop severe HD-MTX-induced oral mucositis leading to chemotherapy delays and an impaired quality of life [57]. The development of oral mucositis is a complex process, of which therapy-induced epithelial cell death is one of the main features [8, 9].

After entering the cell (via reduced folate carrier 1 (RFC1), proton-coupled folate transporter (PCFT) or membrane folate receptors (MFR)), MTX is polyglutamated (PG) by folylpolyglutamate synthetase (FPGS) [10, 11]. This polyglutamation is essential, as it increases intracellular MTX retention and augments its pharmacological activity [12]. MTX-PG inhibits DNA and RNA synthesis via inhibition of dihydrofolate reductase (DHFR), thymidylate synthase (TS) and aminoimidazole carboxamide ribonucleotide transformylase (AICARTFase), resulting in depletion of intracellular reduced folate levels (see S1 Fig for an overview of MTX transport, metabolism and action) [13]. Ultimately, this results in apoptosis in leukemia cells. However, also healthy cells with a high cell turnover, including the bone marrow and the epithelial lining of the gastrointestinal tract and oral mucosa, are affected by MTX therapy.

LV is a reduced folate that is able to restore purine/pyrimidine biosynthesis after HD-MTX therapy [14, 15]. Clinical guidelines advise to administer LV after HD-MTX to reduce toxicity [1622]. While it is nowadays generally accepted that LV rescue therapy decreases MTX-induced toxicities such as oral mucositis after MTX administration, the optimal LV dosing- and timing- regimen to reduce oral mucositis rates remains unknown and varies throughout the world.

Although LV rescue therapy was already introduced in the 1960s, preclinical studies have only retrospectively provided a biochemical rationale for the efficacy of this therapy. In mice, a decrease in MTX-induced damage to the jejunal- and oral mucosa was observed after LV administration [2326]. Importantly, when administered with a time-interval of 12 to 24 hours after MTX, LV did not compromise the anti-leukemic activity of MTX [2326]. In line with this, a selective mechanism of action for MTX and LV in tumor cells versus normal healthy cells has been proposed. A higher level of MTX-PG was observed in leukemia- and solid tumor cell lines when compared to normal intestinal and bone marrow precursor cells, both in vitro and in vivo [1622]. In contrast, several pediatric ALL studies [2730] have suggested that Leucovorin rescue therapy decreases toxicity rates, but might be accompanied by an increased risk of relapse in ALL. This phenomenon has been referred to as the folate ‘over-rescue’ principle, where not only healthy cells, but also tumor cells are rescued. There are no studies that investigate the effect of MTX and LV in healthy human oral mucosa cells and, although valuable, it has been acknowledged that 2D cell lines or mouse models do not always reliably predict the clinical utility of tested therapies [31, 32]. Taken together, there is a need for models that more closely recapitulate the in vivo situation and would allow clinicians and researchers to investigate the effects of altering LV administration regimens on MTX-induced toxicity in healthy mucosal cells.

Organoids are 3D structures grown from stem cells, that recapitulate histological and functional characteristics of their tissue of origin [33]. Since the discovery that organoids could be established from adult stem cells of the mouse gut, organoid technology has quickly evolved [34]. Nowadays, organoids can be grown from many different epithelia [3446]. These ‘mini-organs’ can be established from both tumor and normal primary patient material with high (60–70%) efficiency. Data supporting the translational potential of this technology is accumulating. For example, in vitro therapy response of tumor organoids was shown to predict the responses of corresponding patients [4752]. When derived from Cystic Fibrosis (CF) patients, organoids were also found to predict patient response in vitro [53] and could be used to find effective therapies for CF patients [54].

Recently, we have described an organoid model derived from healthy oral mucosa [38]. The resulting patient-derived structures consist of a functional stratified squamous epithelium that can be maintained and expanded in culture for over six months. Upon passaging, organoids grown from primary oral mucosa tissue can be broken into smaller fragments, which will proliferate and result in the formation of new organoids. As such, organoid technology allows us to multiply human wildtype epithelial cells for a wide variety of applications, including drug screening. Taken that others have shown that organoids are a proper model for body physiology, we set out to test the potential of different dosing- and timing- regimens of LV in search for the most optimal regimen to ‘rescue’ mucosal toxicity during treatment with HD-MTX in patient-derived oral mucosal organoids.

Methods

Establishment and culture of human organoid lines

Tissue for the generation of organoids from adult normal human oral keratinocytes was obtained from tissue biopsies in the oral cavity during ear/nose/throat surgery. Oral mucosa organoids were generated as previously described [38]. Patient material was collected from pathology material in Advanced DMEM/F12 (Life Technologies, cat.no. 12634–034), supplemented with 1x GlutaMAX (adDMEM/F12; Life Technologies, cat.no. 12634–034), Penicillin-streptomycin (Life Technologies, cat.no. 15140–122) and 10 mM HEPES (Life Technologies, cat.no. 15630–056). This medium was called +/+/+ medium. In addition, 100 μg/mL Primocin (Invivogen, cat.no. ant-pm1) was added to the +/+/+ medium for tissue collection. The tissue was cut into small fragments. When macroscopically visible, muscle or fat tissue was removed to enrich for the oral epithelium before digestion. Fragments were incubated at 37°C in 0.125% Trypsin (Sigma, cat.no. T1426) in +/+/+ medium until digested (+/- 30 minutes, never longer than 60 minutes). After trypsinization and centrifugation, the resulting pellet was resuspended in ice-cold 70% 10 mg·mL-1 cold Cultrex growth factor reduced BME type 2 (Trevigen, cat.no. 3533-010-02) in organoid medium. Organoid medium contained 1x B27 supplement (Life Technologies, cat. no. 17504–044), 1.25 mM N-acetyl-L-cysteine (Sigma-Aldrich, cat.no. A9165), 10 mM Nicotinamide (Sigma-Aldrich, cat.no. N0636), 50 ng/mL human EGF (PeproTech, cat.no. AF-100-15), 500 nM A83-01 (PeproTech, cat. no. 100–26), 10 ng/mL human FGF10 (PeproTech, cat.no. 100–26), 5 ng/mL human FGF2 (PeproTech, cat.no. 100-18B), 1 μM Prostaglandin E2 (Tocris Bioscience, cat.no. 2296), 0.3 μM CHIR 99021 (Sigma-Aldrich, cat.no. SML1046), 1 μM Forskolin (Bio-Techne R&D Systems, cat.no. 1099), 4% RSPO and 4% Noggin (produced via r-PEX protein expression platform at U-Protein Express BV). Droplets of approximately 10 μl were plated on the bottom of pre-heated suspension culture plates (Greiner, cat.no. M9312). After plating, plates were inverted and put at 37°C for 30 minutes to let the BME solidify and to prevent the cells from attaching to the bottom of the plate. Subsequently, prewarmed organoid medium was added to the plate. For the first passage of the newly established organoid line, 10 μM Rho-associated kinase (ROCK) inhibitor Y-27632 (Abmole Bioscience, cat.no. M1817) was added to the medium to aid outgrowth of organoids for the primary tissue. Organoids were split between 7 to 14 days after initial plating. For passaging, organoids were collected from the plate by disrupting the BME droplets with a P1000 and washed in 10 mL +/+/+. The pellet was resuspended in 1 mL of TrypLE Express (Life Technologies, cat.no. 12605–010) and incubated at 37°C. Digestion was closely monitored and the suspension was pipetted up and down every 5 minutes to aid disruption of the organoids into single cells. Cells were subsequently resuspended in ice-cold 70% BME in organoid medium and plated at suitable ratios (1:5 to 1:20) to allow efficient outgrowth of new organoids. After splitting, 10 μM Y-27632 (Abmole Bioscience, cat. no. M1817) was always added to aid outgrowth of organoids from single cells. In the experiments, we used organoids from five different donors. Furthermore, we performed drug screens in leukemia cell lines. T cell ALL (MOLT16, HSB2, Jurkat) and B cell ALL (Nalm6, REH) cell lines were obtained from DSMZ-German Collection of Microorganisms and Cell Cultures (DSMZ, Braunschweig, Germany).

Modification of culture conditions for MTX drug screens

For the purpose of this study, organoids were transferred to medium containing a more physiological concentration of folate rather than media with supra-physiological concentrations of folic acid usually present in regular media. We used RPMI 1640 without folic acid (Thermofisher, cat.no. 27016021) supplemented with the same supplements as in organoid medium supplemented with 5 nM folinic acid (Sigma-Aldrich, cat.no. 47612-250MG; racemic mixture of d- and l-stereoisomer of folinic acid) as sole folate source. This medium was referred to as low folate medium. Medium was changed every 2–3 days and organoids were split once every 1–2 weeks. Organoids were cultured for at least two weeks in this folate-deprived state before starting experiments. All drug screens were performed in this modified, low folate medium.

Growth curves

Organoids were disrupted into single cells using TrypLE digestion. Cells were counted and 20.000 single cells were plated per well in 24-well plates. Per well, 30 μl BME was plated and subsequently cultured as previously described. To assess growth speed of the culture on different media, each organoid line was cultured in parallel in normal and low folate medium. For each timepoint, organoid material was collected in triplicate (three wells). Material was collected by disrupting the BME drop with a P1000 pipet in a 15 mL falcon tube and 3 mL cold +/+/+ medium was added for washing. After centrifugation, supernatant was removed and pellets were stored at -20°C until readout. Material was collected at day 0, 3, 5, 7, 10, 12 and 14. For readout, pellets were thawed on ice, and 1 mL of PBS/CellTiter-Glo 3D Reagent (Promega, cat.no. G9681) (1v:1v) was added to the pellet. After a 30 minute incubation on a shaker, 100 μl of the lysate was transferred to a black 96 well plate and luminescence readout was performed to assess cell viability.

RNA collection

Cells were cultured for a week after splitting before RNA was collected. Two days after splitting, cells were cultured in the presence of 0.5 μM MTX. For collection, the QIAGEN RNA easy kit (Qiagen, cat. no. 74104) was used according to protocol. In short, pellets were collected in 350 μl RLT buffer. Subsequently, 350 μl 70% ethanol was added and mixed by pipetting up and down before transfer to the RNA binding columns provided in the kit. After washing twice with 500 μl of RPE buffer, and once with RW1, columns were centrifuged at maximal speed for 1 minute to assure that they were dry. Elution was performed by the addition of 30 μl RNAase free water. RNA was stored at -80°C until further use.

cDNA synthesis

For cDNA synthesis, 10.5 μl RNA was mixed with 1 μl 50 μg/ml 110 diluted Oligo(dT) 15 Primer (Promega, cat.no. C1101) and incubated for 5 minutes at 70°C. After that, 8.5 GoScript Reverse Transcriptase mastermix (Promega, cat.no. A5003) was added, consisting of RT buffer, MgCl2, dNTPs, RT and RNase inhibitor according to protocol. Samples were incubated for 5 minutes at 25°C, 60 minutes at 42°C and 15 minutes at 72°C. Samples were stored at -20°C until use.

Immunohistochemistry

Tissues or organoids were fixed in 4% paraformaldehyde overnight, dehydrated, and embedded in paraffin. Sections were subjected to H&E as well as IHC staining, using the antibodies shown in Table 1. Stainings were performed at the pathology department of the UMCU (Utrecht, the Netherlands).

Table 1.

Protein supplier Order nr. Host species Clone Lot nr. Dilution Antigen retrieval method
TP63 Abcam AB735 Mouse 4AB AB735 1:800 Citrate
KI67 Monosan MONX10293 Mouse MM1 10293 1:2000 Citrate autoclave
KRT13 Progen 10523 Mouse 1C7 10523 1:100 Citrate

Quantitative PCR for expression of MTX metabolism genes

For quantitative PCR, IQ SYBR green (Bio-Rad, cat.no. 1708880) was used in a 384-well format. Per well, 7.5 μl SYBR Green was used, mixed with 1 μl 10 μM FW primer and 1 μl 10 μM RV primer, 3 μl cDNA mix and 2.5 μl water. For each reaction, it was estimated that 25 ng of cDNA was loaded. For qPCR, samples were incubated for 2 minutes at 95°C and for 40 cycles at: 15 seconds at 98°C, 15 seconds at 58°C and 15 seconds at 72°C. Results were calculated by using the ΔΔCt method. Expression was calculated relative to expression in tongue tissue (total RNA, human normal tongue tissue, AmsBio, cat.no. R1234267). Melt peak analysis was performed to assure that primer had no aspecific binding. Primers used are described in S4 Table.

DNA isolation

DNA was isolated using Reliaprep gDNA tissue miniprep system (Promega, cat. no. A2052) according to protocol. DNA concentrations were measured using Nanodrop.

Whole exome sequencing

Whole exome sequencing of oral mucosa organoids was previously performed [38]. In this study, these data were reported to show the difference in number of detected mutations in both the normal human organoids used in this study compared to the tumor tissue derived from the same patient to establish that we used normal human organoids here.

LV pretreatment

For LV pretreatment, 24 hours prior to the start of the drugscreen, organoids were exposed to 0.1 μM LV, by replacing the culture media with media containing this concentration of LV. Plates were placed back in the incubator before, 24 hours later, organoids were collected for drug screening as described below.

MTX drug screens (384-well format)

Two days prior to start of the drug screen, organoids were passaged and disrupted into single cells using TrypLE. Single cells were plated in 70% BME in organoid medium. Two days later, when single cells already reformed into small organoids, the organoids were collected from the BME by addition of 1 mg/mL dispase II (Sigma-Aldrich, cat.no. D4693) to the medium of the organoids. Organoids were incubated for 30 minutes at 37°C to digest the BME. Subsequently, organoids were washed, filtered using a 70 μm nylon cell strainer (Falcon), counted and resuspended in 5% BME/growth medium (12.500 organoids/mL) prior to plating in 40 μl volume (Multi-drop Combi Reagent Dispenser, Thermo Scientific, cat.no. 5840300) in 384-well plates (Corning, cat.no. 4588). As such, 1000 organoids were plated per well. Drugs were added 1 hour after plating the organoids using the Tecan D300e Digital Dispenser (Tecan).

Methotrexate (Sigma-Aldrich, cat.no. M1000000) was dissolved in DMSO and was used in a concentration range between 5 uM–0.05 uM. Folinic acid (Sigma-Aldrich, cat.no. 47612-250MG) was dissolved in PBS containing 0.3% Tween-20, which was required to dispense the drug using the HP printer, and was used at set concentrations of 0.0125 uM–0.025 uM–0.05 uM–0.1 uM. We based our concentration range of MTX based on median MTX plasma level measured in pediatric ALL patients at T48h (0.38 μM, range 0.1–22 μM). The ratio LV:MTX in clinics is around 1:100 (~50 mg/m2: 5000 mg/m2), which was the rationale to focus on a lower range of LV concentrations (for instance 5 μM MTX versus 0.05 μM LV. All wells were normalized for solvent used. DMSO and percentage PBS/Tween-20 never exceeded 1%. Drug exposure was performed in technical triplicate and biological replicates of at least three for each concentration shown. For a lay-out of the drug screen and morphology of the organoid lines during a drugscreen, see S4 Fig.

For folinic acid (Leucovorin–LV) rescue studies, LV was added at different time points after start of MTX incubation. LV was dispensed using the Tecan Dispenser on top of plates previously started on MTX incubation. No medium change was performed (as organoids are in 5% BME, medium removal is impossible) and LV was dispensed into the medium that contained different concentrations of MTX. LV rescue was performed at 0, 12, 24, 48, 72 and 96 hours.

120 hours after adding the drugs, ATP levels were measured using the CellTiter-Glo 3D Reagent (Promega, cat.no. G9681) according to the manufacturer’s instructions and luminescence was measured using a Spark multimode microplate reader (Tecan). Results were normalized to vehicle (no drugs—100% cell viability) and baseline control (Staurosporin 1 μM—0% cell viability). IC50 values (= half maximal inhibitory concentration; the concentration at which 50% of cells are dead) were calculated for separate experiments. As a quality check for the performed assays, Z-values were calculated for each individual screen. Screens with Z-values below 0.3 were excluded from analysis (S2 Table).

MTX-polyglutamate analysis by UHPLC-MS/MS

Organoids were plated at a density of 100.000 per 4 mL in a 6-well non-repellent plate (Greiner) in low folate medium. Leukemia cell lines were cultured at a density of 10*106 cells per 20 mL low folate medium supplemented with 10% fetal calf serum (FCS). Organoids or leukemia cell lines were cultured without MTX or with MTX 0.5 μM. After a 24h incubation, cells were collected and washed twice with 15 mL medium (organoids) or PBS (cell lines). After centrifugation, cells were resuspended in 1 mL, counted and then snap frozen. Before counting organoids, they were incubated at 37°C in 0.125% Trypsin (Sigma, cat.no. T1426) until digested to be able to count single cells. Frozen cell or organoid pellets of 2–3 x 106 cells were thawed and resuspended in 50 μl ice-cold PBS (pH 7.4) (B. Braun, Melsungen, Germany) by vortexing. Next, 50 μl 13C515N-labeled custom-made stable isotopes of MTX-PG1-7 as internal standard (25 nmol/L) and 100 μl perchloric acid (10% v/v, Sigma-Aldrich cat no. 244252) were added and vortexed immediately. [55] Mixtures were incubated on ice for 30 min, followed by centrifugation at 20,160 x g for 15 min at 4°C. Supernatants were quantitatively transferred to an Eppendorf tube and 34 μl 1M phosphate buffer (pH 11.5) was added during vortexing. 100 μl of the mixture was transferred to 0.22 μm spin columns (Merck Millipore Ltd, cat no UFC30GVNB) attached to an LC vial and centrifuged at 3,200 x g for 10 min at 4°C. The resulting eluates were subjected to immediate UHPLC-MS/MS analysis of methotrexate monoglutamate to pentaglutamate (MTX-PG1-5) by injecting 20 μl samples in an Acquity Ultra Performance LC system (Waters Corporation, Milford, MA, USA) with a Kinetex 1.7 μm EVO C18 (100 Å, 100 x 2.1 mm) LC column maintained at 40°C. A linear gradient was applied, consisting of solvent A (5% acetonitrile, pH 3.77) and solvent B (55% acetonitrile, pH 3.77). Measurements were performed over total run times of 10 min. The LC system was connected to an AB Sciex 4000 Q Trap tandem quadrupole mass spectrometer (Applied Biosystems, Foster City, CA, USA), which operated in the positive ionization mode with an ion spray voltage of 5.5 kV, collision energy of 40 V, declustering potential of 100 V and collision exit potential of 7 V for all mass transitions. Mass transitions used for the MTX-PG1-5 analytes were as follows; MTX-PG1 (m/z = 455.200>308.200), MTX-PG2 (m/z = 584.2>308.2), MTX-PG3 (m/z = 713.3>308.2), MTX-PG4 (m/z = 842.3>308.2) and MTX-PG5 (m/z = 971.4>308.2), respectively. For the 13C515N-labeled MTX-PG1-5 internal standard, mass transitions used were: m/z = 461.2>308.2, m/z = 590.2>308.2, m/z = 719.3>308.2, m/z = 848.3>308.2 and m/z = 977.4>308.2, respectively. Qualification and integration of the resulting peaks were analyzed with the Analyst software version 1.6.3 (Sciex, Framingham, MA, USA), resulting in the peak area under the curve (AUC). Quantification of MTX-PG1-5 concentrations/cell number was performed with the labeled internal standards as described before. [55]

FPGS activity analysis

FPGS catalytic activity analysis in organoids and leukemic cell lines was performed essentially as described by Muller et al [56]. In short, FPGS protein was isolated from organoids and cell lines by sonication (Sonoplus Mini 20; Bandelin, Berlin, Germany) on ice for 2 · 10 seconds (10 second intervals with 90% amplitude and 30-second intervals between samples) in FPGS extraction buffer (50 mM Tris, 20 mM KCl, 10 mM MgCl2, and 5 mM DTT, pH 7.4, 4oC, in MilliQ (MilliQ Advantage A10; Merck Millipore, Burlington, MA, USA), followed by centrifugation in an Eppendorf centrifuge (12.000 • g, 15 minutes, 4°C). FPGS-mediated conversion of MTX-PG1 to MTX-PG2 is determined in cell extracts (10–200 μg protein) in a total volume of 250 μL containing final concentrations of 100 mM Tris, 20 mM KCl, 20 mM MgCl2, 10 mM DTT, 10 mM ATP, 250 mM MTX-PG1, and 4 mM 15N-labeled L glutamatic acid (Sigma-Aldrich, cat no. 332143-100MG) at a pH of 8.85 (set ith HCl) under atmospheric pressure. After 2 hour incubations at 37oC, quantities of MTX-(15N)PG2 formed were measured by LC-MS/MS as described above [56]. FPGS activity is expressed as pmol MTX-PG2 formed per microgram protein per hour (pmol•μg-1•hr-1)

Data analysis/statistical analysis

Raw luminescence values obtained after readout of MTX drug screens, were normalized to the average of untreated controls (n = 3, 100%) and staurosporin-treated controls (n = 3, 0%), using the formula: (value- 0% control)/(100% control– 0% control)*100%. Resulting percentages of viability were transferred to GraphPad v8 to generate kill curve. Curves were fit using the option ‘log inhibitor vs. normalized response–variable slope. IC50s/AUC values were obtained from GraphPad analysis. The change in IC50 values over time was assessed using a Pearson correlation, performed in Graphpad prism, using XY analysis, correlation option. AUC’s were compared as previously described. [57, 58]

Ethics approval and consent to participate

Collection of human tissues was compliant with the guidelines of the European Network of Research Ethics Committees (EUREC) and European and national laws, and written informed consent was obtained from all donors. All donors were > age 18 years. The Biobank Research Ethics Committee of the UMC Utrecht approved the biobanking protocol (12–093 HUB-Cancer).

Results

Human wildtype oral mucosa organoids can be used to model MTX-induced cell death in vitro

Organoid lines used in this study were derived from tumor-adjacent normal epithelium of patients with head and neck squamous cell carcinoma (S1 Table). Oral mucosa organoids grew as dense structures consisting of epithelial cells that recapitulate the histological organization of the oral mucosal epithelium in vivo (Fig 1A and 1B). Keeping in mind their origin and the potential risk of cancer cell contamination, wildtype status of the organoids was confirmed by whole exome sequencing. Using normal epithelial tissue as a reference, a low number of mutations was detected in these cultures (two in N1, none in N2), with no mutations found in common cancer driver genes (S2A Fig, S3 Table). To assess if oral mucosa organoids could serve as a model for MTX toxicity, expression of genes involved in MTX transport, metabolism and toxicity was assessed using quantitative PCR (Fig 1C). In addition, catalytic activity of FPGS (important for MTX retention) was compared with that of a reference human T-cell leukemia cell line CCRF-CEM. FPGS activity was >5 times lower in in oral mucosa organoids than in CCRF-CEM cells (501 versus 2715 pmol MTX-PG2/h/mg protein) (S2B Fig).

Fig 1. Human normal oral mucosa organoids can be used to model MTX-induced toxicity in vitro.

Fig 1

A. Morhpology of oral mucosa organoids, brightfield microscopy. The cells form round structures with keratinized centers. Scalebar, 500 μm. B. Immunohistochemical stainings performed on paraffin-embedded oral mucosa organoids indicate that organoids resemble the histological characteristics of in vivo oral mucosa epithelium, with a layer of proliferating basal cells (P63/KI67+) on the outside in contact with the BME (an in vitro basal lamina mimetic) and more differentiated (KRT13+) cells on the inside of the organoids. Hematoxylin and eosin, TP63 staining, KI67 staining and KRT13 staining are shown from left to right. Scalebar, 100 μm. C. Expression of genes involved in MTX transport, metabolism and toxicity was quantified in low folate medium using quantitative qPCR. Allgenes tested were expressed at detectable levels in oral mucosa organoids. D. Short chain MTX-PG1-2 (dashed bars)and long-chain MTX-PG3-5 (filled bars) accumulation in two oral mucosa organoid cultures (N1 blue, N2 green) exposed to increasing doses of MTX (0.125, 0.25 and 0.5 μM respectively), detected by UHPLC-MS/MS. E. MTX exposure induces toxicity (cell death) in oral mucosa organoids grown in low folate medium, but not in normal oral mucosa medium that was previously described in Driehuis et al [38]. Organoids were exposed for five days to MTX and viability was quantified relative to untreated organoids. Reproducible killing at physiological doses of MTX was observed in low folate medium only (dashed lines, circle data points), not in normal organoid medium (continuous lines, square data points).

Oral mucosa organoids were subsequently exposed to MTX. During MTX treatment, MTX polyglutamates (MTX-PG1-5) accumulated intracellularly in oral mucosa organoids in a dose-dependent way (Fig 1D), suggestive of functional MTX metabolism in oral mucosa organoids. Total levels of intracellular MTX-PG differed between organoids derived from different donors, which is in line with the large variation in MTX-PG levels that are detected in patients. MTX-induced cell death was only observed when organoids were cultures in a modified organoid culture medium that contained lower physiological folate levels (Fig 1E), but not the normal organoid culture media (advanced DMEM/F12 containing 6 μM folic acid, 15 μM hypoxanthine and 1.5 μM thymidine). Most likely, when exposed to MTX in this media, these high concentrations prevent MTX toxicity in vitro as previously described [59]. [60, 61]. In this low-folate medium, organoids grew at similar speed and showed similar morphology when compared to the previously defined culture medium (S3A and S3B Fig). Therefore, all subsequent drug screens were performed in low folate medium.

MTX-induced cell death in oral mucosa organoids can be rescued by LV addition, in a timing- and concentration-dependent manner

To assess the role of LV on MTX toxicity, oral mucosa organoids were exposed to a clinically relevant (at levels detected in patient plasma) concentration range of MTX in in vitro drug screens (Fig 2A). The drug screen assays showed high technical quality as measured by Z-scores (median 0.72; range 0.31–0.98, S2 Table). To model LV rescue therapy in vitro, organoids were exposed to different concentrations of LV at different timepoints after the start of MTX treatment.

Fig 2. MTX-induced toxicity in oral mucosa organoids can be rescued by the addition of LV, in both a timing- and dosing-dependent manner.

Fig 2

A. Schematic outline of the experimental set-up applied to assess the effect of LV rescue initiated post MTX exposure toxicity in vitro. Organoids were split on day 0, left to recover for two days, subsequently filtered, counted and plated in 384 well format to be exposed to MTX for five days (with or without LV added on variable timepoints after the start of MTX exposure). On day 7, viability readout was performed. As such, organoids were exposed to MTX for a total of five days. B. MTX-induced toxicity can be decreased by LV rescue and extent of LV rescue is dose dependent. MTX IC50 values were determined either without LV rescue, and with different LV rescue dosages (0.1 μM, 0.05 μM, 0.025 μM and 0.0125 μM), where rescue was initiated 24 hours post the start of MTX exposure. Each IC50 value is obtained for a 9-point dose titration of MTX, where the effect of each dose is tested in technical triplicate. Symbols indicate the different replicate experiments in which the IC50 were defined for different timepoints of LV rescue. As such, IC50 values indicated with the same symbol at different timepoints, were determined in the same experiment. Blue symbols indicate N1 organoids, turquoise symbols indicate N2 organoids. C. Timing of LV rescue influences its effect of MTX-induced toxicity in N1 organoids. Here, MTX kill curves are shown when 0.1 μM LV rescue is initiated either 0, 12, 24, 48, 72 or 96 hours after the start of MTX exposure. D. Timing of LV rescue influences its effect of MTX-induced toxicity in N2 organoids. Here, MTX kill curves are shown when 0.1 μM LV rescue is initiated either 0, 12, 24, 48, 72 or 96 hours after the start of MTX exposure. E. Effect of PT in N1 (blue symbols) and N2 (turquoise symbols) organoids when combined with LV rescue, initiated at different time-points post start of MTX exposure. MTX IC50 values are shown on the y-axis. For each timepoint, three IC50 values are show, obtained in three independent experiments. Each IC50 value is obtained for a 9-point dose titration of MTX, where the effect of each dose is tested in technical triplicate. Symbols indicate the different replicate experiments in which the IC50 were defined for different timepoints of LV rescue. As such, IC50 values indicated with the same symbol at different timepoints, were determined in the same experiment.

Administration of LV resulted in a decrease of MTX-induced cell death in a concentration-dependent manner, that was statistically significant (Fig 2B). Secondly, the extent of LV rescue was dependent on the timing of LV addition; the earlier LV was administrated, the higher the overall cell viability (Fig 2C and 2D). LV administration decreased MTX toxicity up to 72 hours after the start of MTX treatment. A correlation between timing of LV administration and MTX IC50 was observed in both N1 and N2 organoids (Fig 2E). Considering the timing of LV administration varies that can vary per treatment protocol, but is usually initiated at timepoints ranging from 24 to 48 hours after MTX infusion, these results are relevant. As in patients, MTX plasma levels have dropped by 54 hours post infusion, LV administration is rarely continued after this timepoint. Here we observe that, in vitro, LV administration still decreases MTX-induced toxicity beyond this timepoint.

A one day pre-treatment of oral mucosa cells with LV prior to MTX exposure, results in potentiation of the LV rescue effect

In patients, MTX-induced oral mucositis most frequently occurs after the first cycle of HD-MTX [62]. This has resulted in the hypothesis that intracellular LV from previous courses prevents toxicity during subsequent MTX courses. To model this in vitro, organoids were exposed to LV one day prior to the start of MTX treatment. At the start of MTX treatment, LV was removed and toxicity was assessed as previously described (Fig 3A). In organoid line N1, LV pre-treatment did not significantly alter the response to MTX (AUC no pre-treatment: 0.8044, AUC pre-treatment 0.8983, t = 1.9169, p = 0.102), although MTX IC50 values marginally increased (Fig 3B and 3C). However, in organoid line N2, a clear rescue effect of the pre-incubation with LV was observed (Fig 3D and 3E), which was also found to be statisctically significant (AUC no pre-treatment: 0.7166, AUC pre-treatment 1.034, t = 10.43, p = 0.0005),. LV pre-treatment increased the viability of N2 organoids when exposed to MTX, for all LV rescue timepoints tested here. When pre-treated, the rescue effect of LV rescue administered at 72 hours—later than currently applied in the clinic—resulted in a cell survival similar to a LV rescue that would have been given at 0 hours without this pre-treatment. This suggests that pre-treatment might increase the timeframe in which LV rescue rescues MTX toxicity in oral mucosa cells.

Fig 3. A one day LV pre-treatment of oral mucosa cells before MTX exposure results in potentiation of the LV rescue effect.

Fig 3

A. Schematic outline of the experimental set-up applied to assess the effect of LV pre-treatment (PT) on MTX toxicity in vitro. Deviations from the original experimental set-up used to determine MTX toxicity (see Fig 2A) are shown in red. In short, one day prior to the start of MTX exposure, cells were treated with 0.1 μM LV for 24 hours. Before exposure to MTX, LV was removed. From there one, experimental set-up was identical to that previously described for MTX toxicity. Organoids were exposed to MTX for 5 days, either in the presence or absence of LV rescue, that was commenced at variable times after the start of MTX exposure. B. Effect of a 24 hour 0.1 μM LV PT on MTX toxicity in N1. Cells were exposed to MTX, where MTX exposure was either preceded by a one day 0.01 μM LV PT (black squares) or not (dark blue circles). PT effect was investigated either with (dashed lines) or without (continuous lines) a 0.05 μM LV rescue, applied 24 post treatment. Toxicity was determined using a 9-point dose titration of MTX, where the effect of each dose is tested in technical triplicates. Pearson correlation coefficient was determined in GraphPad to evaluate the correlation between dose of LV rescue and MTX IC50. C. Effect of PT in N1 organoids when combined with LV rescue, initiated at different time-points post start of MTX exposure. MTX IC50 values are shown on the y-axis. For each timepoint, three IC50 values are show, obtained in three independent experiments. Each IC50 value is obtained for a 9-point dose titration of MTX, where the effect of each dose is tested in technical triplicate. Symbols indicate the different replicate experiments in which the IC50 were defined for different timepoints of LV rescue. As such, IC50 values indicated with the same symbol at different timepoints, were determined in the same experiment. D. Effect of a 24 hour 0.1 μM LV PT on MTX toxicity in N2. Cells were exposed to MTX, where MTX exposure was either preceded by a one day 0.1 μM LV PT (black squares) or not (turquoise circles). PT effect was investigated either with (dashed lines) or without (continuous lines) a 0.05 μM LV rescue, applied 24 post treatment. Toxicity was determined using a 9-point dose titration of MTX, where the effect of each dose is tested in technical triplicates. E. Effect of PT in N2 organoids when combined with LV rescue, initiated at different time-points post start of MTX exposure. MTX IC50 values are shown on the y-axis. For each timepoint, three IC50 values are show, obtained in three independent experiments. Each IC50 value is obtained for a 9-point dose titration of MTX, where the effect of each dose is tested in technical triplicate. Symbols indicate the different replicate experiments in which the IC50 were defined for different timepoints of LV rescue. As such, IC50 values indicated with the same symbol at different timepoints, were determined in the same experiment. Pearson correlation coefficient was determined in GraphPad to evaluate the correlation between timing of LV rescue and MTX IC50. F. Effect of LV pre-treatment (PT) on viability of N1, N2, N3, N4 and N5 organoids, respectively, all derived from different donors. Squares indicate PT conditions, circles indicate cells that did not receive PT. In 4/5 organoid cultures (all organoid cultures apart from N1), an increase in MTX IC50 value can be observed in response to pre-treatment, indicating that a 24 hour 0.1 μM LV PT reduces MTX toxicity in oral mucosa cells.. Here, IC50 values are shown when no LV rescue is performed. For N1 and N2, experimetns were not only performed in technical triplicate, but also biological triplicate. Here, t-tests were performed to confirm that in N2, but not N1, a statistical significant effect of LV preptreatment could be observed on MTX IC50 values.

As variable responses to LV pre-treatment were observed in N1 and N2, the effect of pre-treatment was tested in organoids established from three additional donors. In all three cultures, pre-treatment increased oral mucosa cell survival upon exposure to MTX (Fig 3F). Taken together, we conclude that a one day pre-treatment with LV decreases MTX-induced mucosal toxicity in 4/5 tested donors. This implies that LV pre-treatment may reduce the risk of oral mucositis. Regardless, the effect of such a pre-treatment on leukemia cells much be investigated before any claims can be made on clinical testing of such an intervention.

Effect of MTX and LV therapy on leukemia cell lines

To assess the effect of LV pre-treatment on leukemia cells, both T cell ALL (Jurkat, MOLT16, HSB2) and B cell ALL (Nalm6, REH) cell lines were exposed to MTX, either in the presence or absence of LV pre-treatment (Fig 4A to 4E). Although LV pre-treatment increased MTX IC50 values in the leukemia cell lines tested here, the effect was less pronounced than in oral mucosa cells (Fig 4F).

Fig 4. LV pre-treatment influences MTX toxicity in leukemia cell lines.

Fig 4

A to E. Effect of LV pre-treatment (PT) on viability of T-ALL and B-ALL leukemia cell lines. For each cell line, cells were exposed to MTX, where MTX exposure was either preceded by a one day 0.01 μM LV PT (black squares) or not (colored circles, color indicating the cell line tested). PT effect was investigated either with (dashed lines) or without (continuous lines) a 0.05 μM LV rescue, applied 24 post treatment. A. Jurkat, dark red. B. MOLT16, fuchsia. C. HSB-2, purple. D. REH, magenta. E. Nalm-6, violet. F. MTX IC50 values detected for leukemia cell lines when exposed to MTX, either preceded by a0.01 μ M LVPT (squares),or not (circles). For each cell line, three IC50 values are show, obtained in three independent experiments. Each IC50 value is obtained for a 9-point dose titration of MTX, where the effect of each dose is tested in technical triplicate. In all cases, an increase in MTX IC50 value can be observed in response to pre-treatment. Here, IC50 values are shown when no LV rescue is performed. Different colors indicate the different cell lines, colors are similar to those used in A-E. G. Viability of cells exposed for 5 days to 0.1 μM MTX, either preceded by 0.01 μM LV PT (dashed bars) or not. Viability was tested in technical triplicate, where the average viability is shown for three independent experiments. Different colors indicate the different cell lines, colors are similar to those used in A-E.

To estimate the effect of LV pre-treatment when administered systemically, we compared the effect of this treatment on both oral mucosa cells and leukemia cells at a concentration of 0.1 μM MTX (a level reached during plasma measurements of all patients) [62]. At this MTX dose, LV pre-treatment decreases the toxicity of MTX on oral mucosa cells, but does not influence the effect of MTX on 4/5 leukemia cells (Fig 4G). T-ALL cell line Jurkat did show decreased MTX toxicity when exposed to LV pre-treatment. Indeed, it has been previously reported that T-cell ALL is less sensitive than B-cell ALL [63, 64]. Indeed, MTX-PG levels in two B-ALL leukemia cell lines (REH; Nalm6) were around 3-fold higher (S2C Fig) when compared to MTX-PG levels in oral mucosa organoids (S2D Fig). MTX-PG levels in two T-ALL leukemia cell lines (Jurkat; HSB2) differed with high MTX-PG levels in Jurkat cells (~3-fold higher than organoids) and low MTX-PG levels in HSB-2 cells (same range as organoids).

Taken together, we conclude that LV pre-treatment decreases MTX-induced cell death of leukemia cells in vitro. However, as leukemia cells are much more sensitive to MTX than oral mucosa cells, the effect of this pre-treatment might be mitigated at the MTX levels reached during HD-MTX infusions. Regardless, the effect of LV pre-treatment should be explored with caution, but might prove to be an effective approach to decrease the severity and frequency of MTX-indicued mucositis. A local LV application is a feasible alternative, since it would likely not interfere with the systemic MTX effect on leukemia cells and may therefore be a safer approach to reduce the risk of mucositis.

Discussion

Using a 3D in vitro model of primary human oral mucosa organoids, we show donor-dependent MTX-induced cell death in wildtype human oral mucosa cells. To our knowledge, this is the first in vitro model (based on the model described by Driehuis et al [38] with modification in media composition) to assess the effect of MTX on proliferating, wildtype oral mucosa epithelial cells. Using this system, we show that administration of LV at dosages detected in patient plasma, reduces MTX-induced cell death in a concentration- and time-dependent manner and that MTX-induced toxicity shows variability between cells derived from different individuals.

In patients, LV infusion after HD-MTX is usually initiated 36 or 42 hours after HD-MTX, although in some protocols, LV is already applied at 24 hours post MTX infusion. LV rescue therapy is often not administered beyond the timepoint of 54 hours after start of MTX. [65] Based on our findings, we hypothesize that both an earlier start and longer continuation of LV rescue might reduce the incidence or extent of MTX-induced mucositis. However, since the model presented here only studies the effect of such interventions on either mucosal cells or leukemia cell lines, these results need to be validated (for example in mouse models) before they can be clinically tested. Alternatively, a local application of LV one day prior to the start of MTX administration might be an alternative approach to reduce mucositis in pediatric leukemia patients. A local application of LV is currently already applied in some clinics when patients present with oral mucositis after HD-MTX.

To assess the effect of LV pre-treatment on leukemia cells, leukemia-derived cell lines were exposed to the same pre-treatment to study the effect of this intervention on MTX toxicity. Although present, the effect of pre-treatment and LV rescue was less pronounced in leukemia cells than in oral mucosa cells. Differences in response to LV between leukemia cells and healthy cells have been observed before. Several pre-clinical studies showed this selective mechanism of action for MTX and LV might be due to the fact that high MTX-PG levels accumulate in leukemia cell lines compared to normal intestinal and bone marrow precursor cells [1422]. Also in oral mucosa organoids we found that MTX-PG levels and FPGS activity are lower than in the leukemia cell lines tested here. Taken together, these observations support the fact that primary patient-derived ALL cells (especially B-ALL when compared to T-ALL) are sensitive to MTX due to a high proliferation rate and high FPGS activity and thus might be less affected by LV than oral mucosa cells [22]. However, it is important to test clinical safety of the described interventions in order not to decrease anti-leukemic activity of MTX.

In clinics, only a subset of patients presents with mucositis, suggesting some patients are more sensitive to MTX treatment than others, or might respond better to LV rescue treatment. This is in line with our findings, where organoid cultures derived from different patients, show variable responses to both MTX and LV exposure in vitro. Due to this variable response, more personalized approaches to decrease the risk of MTX-induced mucositis are urgently needed and, in some clinics, already explored. Although beyond the scope of this work, oral mucosa organoids create the opportunity to study the molecular differences between sensitive and resistant cultures. If these inter-patient differences are a consequence of local rather than systemic differences in responses to MTX/LV, such comparisons could help to identify patients at risk, or contribute to the development of alternative treatment strategies to decrease the incidence of oral mucositis.

Here we only study MTX-induced cell death as a model for mucositis. Importantly, oral mucositis is a complex process of which cell death is only one of the hallmarks [8, 66]. In 2004, Sonis et al. proposed a more complicated model of oral mucositis, in which the generation of Reactive Oxygen Species (ROS) and or pro-inflammatory cytokines were also hallmarks of the clinical phenotype in addition to therapy-induced cell death [8]. Furthermore, it has been suggested that the bacterial microbiome might play a role in developing oral mucositis. It would be of interest to study the effect of these factors in future models [8]. Co-cultures of organoids and immune cells or organoids and bacteria have been described [67, 68]. Therefore, this model holds the potential to be extended to investigate the contribution of these factors in oral mucositis.

By making minor adaptations to the culture media that was previously described to culture oral mucosa organoids [38], we have shown that organoids can be used to study MTX-toxicity in vitro. These modifications can be applied to other organoid cultures such as kidney or liver organoids [69, 70]. As such, these models can perhaps be used to study MTX-toxicity that is observed in other organs than the oral mucosa.

Conclusion

Although applied in clinic for many years, the effect of LV rescue therapy to reduce oral mucositis after MTX treatment has not been shown in representative models before. Here, we report the use of normal human oral mucosa organoids that recapitulate functional and histological characteristics of this epithelium to study the potential of LV to reduce MTX-induced toxicity. Oral mucosa organoids show sensitivity to clinically relevant doses of MTX in vitro. MTX-induced toxicity could be reduced by the addition of LV after the start of MTX treatment. The extent of LV-rescue is concentration- and time- dependent and differs between organoids derived from different donors, fitting with what is observed in the clinic. Using this system, we find a pre-exposure (‘pre-treatment’) with LV before MTX treatment significantly potentiates the effect of LV rescue. The effect of this pre-treatment is present, but less pronounced in leukemia cells. Taken together, our findings support the LV rescue protocols currently applied in the clinic and, moreover, highlight the potential of this model to study the effect of modifications of the currently applied clinical regimens, such as a LV pre-treatment, on proliferation wildtype oral mucosa cells in vitro.

Supporting information

S1 Fig. Mechanism of action of MTX treatment.

Leucovorin (5-formylTHF) is represented in bold / italic. MTX enters the cell mainly through the Reduced Folate Carrier 1 (RFC1), Proton Coupled Folate Transporter (PCFT), Membrane Folate Transporters (MFR) or by passive diffusion through the cell membrane. While circulating, MTX contains one polyglutamate group (MTX-PG1). Once inside the cell, MTX is polyglutamated by Folylpolyglutamate Synthetase (FPGS) with up to seven polyglutamate groups. Long-chain MTX-PG’s (MTX-PG4-7) cannot be transported out of the cell before de-polyglutamation by Gamma-Glutamyl Hydrolase (GGH). Short-chain MTX-PG’s (MTX-PG1-3) will be actively transported out of the cell by ABCC1-4, ABCB1 and ABCG2 transporters. MTX is cytotoxic as it impairs purine- and pyrimidine synthesis by inhibiting the enzymes Dihydrofolate Reductase (DHFR) and Thymidylate Synthase (TYMS). Abbreviations: ABCB1—ATP Binding Cassette Subfamily B Member 1; ABCC1-4—ATP Binding Cassette Subfamily C Member 1–4; ABCG2—ATP Binding Cassette Subfamily G Member 2; DHFR–Dihydrofolate Reductase; FPGS–Folylpolyglutamate Synthetase; GGH–Gamma-Glutamyl Hydrolase; MFR–Membrane Folate Transporter; MTHFR—Methylene tetrahydrofolate reductase; MTHFD1—Methylenetetrahydrofolate Dehydrogenase, Cyclohydrolase And Formyltetrahydrofolate Synthetase 1; PCFT–Proton-Coupled Folate Transporter; RFC1 –Reduced Folate Carrier; SHMT—Serine hydroxymethyltransferase; TS–Thymidylate Synthase.

(PDF)

S2 Fig. Characterization of oral mucosa organoids and the effect of pretreatment on intracellular MTX-PG levels.

A. Oral mucosa organoids are derived of human normal cells, and not cancer cells. Number of mutations detected by whole exome sequencing in the healthy oral mucosa organoids used in this study, and their corresponding tumor organoids. Mutational load is low (2 for N1, 0 for T1), especially when compared to the tumor organoids. B. FPGS activity (in pmol MTX-PG2/h/mg) in organoid line versus CCRF-CEM reference leukemia cell line. C. Effect of PT on MTX-PG levels in oral mucosa organoid lines derived from two different donors. D. Effect of PT on MTX-PG levels in two B-ALL and two T-ALL cell lines.

(PDF)

S3 Fig. Organoid cultures retain their morphology and growth speed when grown in folate deprived medium.

A. Brightfield microscopy images of organoid line N1 and N2, when grown in either complete medium, or folate deprived medium. Scalebar, 500 μm. B. Growth speed of organoid cultures in both media tested. Growth was assessed by collection of cell pellets at day 0, 3, 5, 7, 10 and 14. Cell number was assessed by cell titer glow and values were made relative to day 0. C. Quantitative PCR assessing expression of genes relevant for methotrexate metabolism. Experiment was performed in triplicate, results of all three experiments are shown here.

(PDF)

S4 Fig. Technical details of drugscreen performed in this study.

A. Schematic layout of a drug screen plate as used in this study. The gradient of MTX is depicted using a color gradient (red indicates high concentration, green indicates low concentration). Here, the MTX concentrations used for organoids are depicted. Each concentration is tested in technical triplicate. Different blocks receive LV rescue at different timepoints after the start of MTX treatment, as indicated. Staurosporine treated wells are used as positive controls and are set to 0% viability, wells only receiving drug solvent are used is negative controls, and are set to 100% viability. B. Brightfield microscopy images showing the morphology of N1 organoids in drug screening plates on the day of readout. C. Brightfield microscopy images showing the morphology of N2 organoids in drug screening plates on the day of readout.

(PDF)

S1 Table. Clinical information of patients. Relevant clinical information is given on the patient that participated in this study, and form whose tissue organoids were derived.

(PDF)

S2 Table. Z-scores of drug screens performed in this study.

(PDF)

S3 Table. Comparison of mutations detected by WES in matching normal and tumor organoid lines.

All mutation detected in organoid line N1, T1, N2 and T2 are shown. Here, normal tissue was used as a reference.

(XLSX)

S4 Table. Sequences of primers used for quantitative PCR.

5’ to 3’ sequences of primers used to assess gene expression by quantitative PCR in this study.

(PDF)

Acknowledgments

We would like to acknowledge Onno Kranenburg, Anneta Brousali, Petra van der Groep, Alexander Constantinides and Anne Snelting of the Utrecht Platform for Organoid Technology (U-PORT; UMC Utrecht) for patient inclusion and tissue acquisition. We thank HUB Organoids for help with regulatory affairs regarding informed consent. We would like to thank Sacha Spelier for her help with organoid experiments. We acknowledge Eduard Struys for technical support in FPGS activity and MTX-PG level measurements. We thank the groups of Monique den Boer and Jules Meijerink at the Princess Maxima Center for providing us with leukemia cell lines.

Data Availability

All data are described within the current manuscript as it is.

Funding Statement

Funded by the Oncode Institute (partly financed by the Dutch Cancer Society), by the gravitation program CancerGenomiCs.nl from the Netherlands Organization for Scientific Research (NWO) and by a Stand Up to Cancer International Translational Cancer Research Grant, a program of the Entertainment Industry Foundation administered by the AACR (SU2C-AACR-DT1213) and a ZonMw grant (116.006.103).

References

  • 1.Hunger SP, Lu X, Devidas M, Camitta BM, Gaynon PS, Winick NJ, et al. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the children's oncology group. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2012;30(14):1663–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Pui CH, Carroll WL, Meshinchi S, Arceci RJ. Biology, risk stratification, and therapy of pediatric acute leukemias: an update. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2011;29(5):551–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kamps WA, van der Pal-de Bruin KM, Veerman AJ, Fiocco M, Bierings M, Pieters R. Long-term results of Dutch Childhood Oncology Group studies for children with acute lymphoblastic leukemia from 1984 to 2004. Leukemia. 2010;24(2):309–19. 10.1038/leu.2009.258 [DOI] [PubMed] [Google Scholar]
  • 4.Pieters R, de Groot-Kruseman H, Van der Velden V, Fiocco M, van den Berg H, de Bont E, et al. Successful Therapy Reduction and Intensification for Childhood Acute Lymphoblastic Leukemia Based on Minimal Residual Disease Monitoring: Study ALL10 From the Dutch Childhood Oncology Group. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2016;34(22):2591–601. [DOI] [PubMed] [Google Scholar]
  • 5.Howard SC, McCormick J, Pui CH, Buddington RK, Harvey RD. Preventing and Managing Toxicities of High-Dose Methotrexate. The oncologist. 2016;21(12):1471–82. 10.1634/theoncologist.2015-0164 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ackland SP, Schilsky RL. High-dose methotrexate: a critical reappraisal. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 1987;5(12):2017–31. [DOI] [PubMed] [Google Scholar]
  • 7.Bertino JR. Karnofsky memorial lecture. Ode to methotrexate. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 1993;11(1):5–14. [DOI] [PubMed] [Google Scholar]
  • 8.Sonis ST. The pathobiology of mucositis. Nature reviews Cancer. 2004;4(4):277–84. 10.1038/nrc1318 [DOI] [PubMed] [Google Scholar]
  • 9.Villa A, Sonis ST. Mucositis: pathobiology and management. Current opinion in oncology. 2015;27(3):159–64. 10.1097/CCO.0000000000000180 [DOI] [PubMed] [Google Scholar]
  • 10.Zhao R, Diop-Bove N, Visentin M, Goldman ID. Mechanisms of membrane transport of folates into cells and across epithelia. Annual review of nutrition. 2011;31:177–201. 10.1146/annurev-nutr-072610-145133 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Gonen N, Assaraf YG. Antifolates in cancer therapy: structure, activity and mechanisms of drug resistance. Drug resistance updates: reviews and commentaries in antimicrobial and anticancer chemotherapy. 2012;15(4):183–210. [DOI] [PubMed] [Google Scholar]
  • 12.Raz S, Stark M, Assaraf YG. Folylpoly-gamma-glutamate synthetase: A key determinant of folate homeostasis and antifolate resistance in cancer. Drug resistance updates: reviews and commentaries in antimicrobial and anticancer chemotherapy. 2016;28:43–64. [DOI] [PubMed] [Google Scholar]
  • 13.Zhao R, Goldman ID. Resistance to antifolates. Oncogene. 2003;22(47):7431–57. 10.1038/sj.onc.1206946 [DOI] [PubMed] [Google Scholar]
  • 14.Goldman ID, Matherly LH. Biochemical factors in the selectivity of leucovorin rescue: selective inhibition of leucovorin reactivation of dihydrofolate reductase and leucovorin utilization in purine and pyrimidine biosynthesis by methotrexate and dihydrofolate polyglutamates. NCI monographs: a publication of the National Cancer Institute. 1987(5):17–26. [PubMed] [Google Scholar]
  • 15.Matherly LH, Barlowe CK, Phillips VM, Goldman ID. The effects on 4-aminoantifolates on 5-formyltetrahydrofolate metabolism in L1210 cells. A biochemical basis of the selectivity of leucovorin rescue. J Biol Chem. 1987;262(2):710–7. [PubMed] [Google Scholar]
  • 16.Fry DW, Anderson LA, Borst M, Goldman ID. Analysis of the role of membrane transport and polyglutamation of methotrexate in gut and the Ehrlich tumor in vivo as factors in drug sensitivity and selectivity. Cancer research. 1983;43(3):1087–92. [PubMed] [Google Scholar]
  • 17.Fabre I, Fabre G, Goldman ID. Polyglutamylation, an important element in methotrexate cytotoxicity and selectivity in tumor versus murine granulocytic progenitor cells in vitro. Cancer research. 1984;44(8):3190–5. [PubMed] [Google Scholar]
  • 18.Poser RG, Sirotnak FM, Chello PL. Differential synthesis of methotrexate polyglutamates in normal proliferative and neoplastic mouse tissues in vivo. Cancer research. 1981;41(11 Pt 1):4441–6. [PubMed] [Google Scholar]
  • 19.Koizumi S, Curt GA, Fine RL, Griffin JD, Chabner BA. Formation of methotrexate polyglutamates in purified myeloid precursor cells from normal human bone marrow. The Journal of clinical investigation. 1985;75(3):1008–14. 10.1172/JCI111761 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Matherly LH, Barlowe CK, Goldman ID. Antifolate polyglutamylation and competitive drug displacement at dihydrofolate reductase as important elements in leucovorin rescue in L1210 cells. Cancer research. 1986;46(2):588–93. [PubMed] [Google Scholar]
  • 21.Rothenberg SP, Iqbal MP, da Costa M. Effect of folate compounds on the accumulation of methotrexate and the activity of dihydrofolate reductase in liver, kidney and small intestine of the mouse. The Journal of pharmacology and experimental therapeutics. 1982;223(3):631–4. [PubMed] [Google Scholar]
  • 22.Rots MG, Pieters R, Peters GJ, Noordhuis P, van Zantwijk CH, Kaspers GJ, et al. Role of folylpolyglutamate synthetase and folylpolyglutamate hydrolase in methotrexate accumulation and polyglutamylation in childhood leukemia. Blood. 1999;93(5):1677–83. [PubMed] [Google Scholar]
  • 23.Goldin A, Mantel N, Greenhouse SW, Venditti JM, Humphreys SR. Estimation of the antileukemic potency of the antimetabolite aminopterin, administered alone and in combination with citrovorum factor or folic acid. Cancer research. 1953;13(12):843–50. [PubMed] [Google Scholar]
  • 24.Goldin A, Venditti JM, Kline I, Mantel N. Eradication of leukaemic cells (L1210) by methotrexate and methotrexate plus citrovorum factor. Nature. 1966;212(5070):1548–50. 10.1038/2121548a0 [DOI] [PubMed] [Google Scholar]
  • 25.Berry CL, Germain J, Looker T. The effect of methotrexate on DNA synthesis and its reversal by folinic acid. Journal of embryology and experimental morphology. 1972;28(3):601–5. [PubMed] [Google Scholar]
  • 26.Sandberg JS, Goldin A. The use of leucovorin orally in normal and leukemic L1210 mice to prevent the toxicity and gastrointestinal lesions caused by high doses of methotrexate. Cancer research. 1970;30(5):1276–80. [PubMed] [Google Scholar]
  • 27.Borsi JD, Wesenberg F, Stokland T, Moe PJ. How much is too much? Folinic acid rescue dose in children with acute lymphoblastic leukaemia. European journal of cancer (Oxford, England: 1990). 1991;27(8):1006–9. [DOI] [PubMed] [Google Scholar]
  • 28.Wolfrom C, Hepp R, Hartmann R, Breithaupt H, Henze G. Pharmacokinetic study of methotrexate, folinic acid and their serum metabolites in children treated with high-dose methotrexate and leucovorin rescue. European journal of clinical pharmacology. 1990;39(4):377–83. 10.1007/BF00315414 [DOI] [PubMed] [Google Scholar]
  • 29.Skarby TV, Anderson H, Heldrup J, Kanerva JA, Seidel H, Schmiegelow K. High leucovorin doses during high-dose methotrexate treatment may reduce the cure rate in childhood acute lymphoblastic leukemia. Leukemia. 2006;20(11):1955–62. 10.1038/sj.leu.2404404 [DOI] [PubMed] [Google Scholar]
  • 30.Sterba J, Dusek L, Demlova R, Valik D. Pretreatment plasma folate modulates the pharmacodynamic effect of high-dose methotrexate in children with acute lymphoblastic leukemia and non-Hodgkin lymphoma: "folate overrescue" concept revisited. Clinical chemistry. 2006;52(4):692–700. 10.1373/clinchem.2005.061150 [DOI] [PubMed] [Google Scholar]
  • 31.Voskoglou-Nomikos T, Pater JL, Seymour L. Clinical predictive value of the in vitro cell line, human xenograft, and mouse allograft preclinical cancer models. Clinical cancer research: an official journal of the American Association for Cancer Research. 2003;9(11):4227–39. [PubMed] [Google Scholar]
  • 32.Johnson JI, Decker S, Zaharevitz D, Rubinstein LV, Venditti JM, Schepartz S, et al. Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials. British journal of cancer. 2001;84(10):1424–31. 10.1054/bjoc.2001.1796 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kretzschmar K, Clevers H. Organoids: Modeling Development and the Stem Cell Niche in a Dish. Developmental cell. 2016;38(6):590–600. 10.1016/j.devcel.2016.08.014 [DOI] [PubMed] [Google Scholar]
  • 34.Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459(7244):262–5. 10.1038/nature07935 [DOI] [PubMed] [Google Scholar]
  • 35.Barker N, Huch M, Kujala P, van de Wetering M, Snippert HJ, van Es JH, et al. Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell. 2010;6(1):25–36. 10.1016/j.stem.2009.11.013 [DOI] [PubMed] [Google Scholar]
  • 36.Tadokoro T, Wang Y, Barak LS, Bai Y, Randell SH, Hogan BL. IL-6/STAT3 promotes regeneration of airway ciliated cells from basal stem cells. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(35):E3641–9. 10.1073/pnas.1409781111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lee SH, Hu W, Matulay JT, Silva MV, Owczarek TB, Kim K, et al. Tumor Evolution and Drug Response in Patient-Derived Organoid Models of Bladder Cancer. Cell. 2018;173(2):515–28 e17. 10.1016/j.cell.2018.03.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Driehuis E KS, Spelier S, Lohmussaar K, Willems SM, Devriese LA, de Bree R, et al. Oral mucosal organoids as a potential platform for personalized cancer therapy. Cancer Discov. 2019. [DOI] [PubMed] [Google Scholar]
  • 39.Kopper O, de Witte CJ, Lohmussaar K, Valle-Inclan JE, Hami N, Kester L, et al. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat Med. 2019;25(5):838–49. 10.1038/s41591-019-0422-6 [DOI] [PubMed] [Google Scholar]
  • 40.DeWard AD, Cramer J, Lagasse E. Cellular heterogeneity in the mouse esophagus implicates the presence of a nonquiescent epithelial stem cell population. Cell Rep. 2014;9(2):701–11. 10.1016/j.celrep.2014.09.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Huch M, Dorrell C, Boj SF, van Es JH, Li VS, van de Wetering M, et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature. 2013;494(7436):247–50. 10.1038/nature11826 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Karthaus WR, Iaquinta PJ, Drost J, Gracanin A, van Boxtel R, Wongvipat J, et al. Identification of multipotent luminal progenitor cells in human prostate organoid cultures. Cell. 2014;159(1):163–75. 10.1016/j.cell.2014.08.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kessler M, Hoffmann K, Brinkmann V, Thieck O, Jackisch S, Toelle B, et al. The Notch and Wnt pathways regulate stemness and differentiation in human fallopian tube organoids. Nature communications. 2015;6:8989 10.1038/ncomms9989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Linnemann JR, Miura H, Meixner LK, Irmler M, Kloos UJ, Hirschi B, et al. Quantification of regenerative potential in primary human mammary epithelial cells. Development. 2015;142(18):3239–51. 10.1242/dev.123554 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Nanduri LS, Baanstra M, Faber H, Rocchi C, Zwart E, de Haan G, et al. Purification and ex vivo expansion of fully functional salivary gland stem cells. Stem Cell Reports. 2014;3(6):957–64. 10.1016/j.stemcr.2014.09.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ren W, Lewandowski BC, Watson J, Aihara E, Iwatsuki K, Bachmanov AA, et al. Single Lgr5- or Lgr6-expressing taste stem/progenitor cells generate taste bud cells ex vivo. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(46):16401–6. 10.1073/pnas.1409064111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernandez-Mateos J, Khan K, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science (New York, NY). 2018;359(6378):920–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, et al. A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell. 2018;172(1–2):373–86 e10. 10.1016/j.cell.2017.11.010 [DOI] [PubMed] [Google Scholar]
  • 49.Ooft SN, Weeber F, Dijkstra KK, McLean CM, Kaing S, van Werkhoven E, et al. Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients. Sci Transl Med. 2019;11(513). [DOI] [PubMed] [Google Scholar]
  • 50.Driehuis E, van Hoeck A, Moore K, Kolders S, Francies HE, Gulersonmez MC, et al. Pancreatic cancer organoids recapitulate disease and allow personalized drug screening. Proceedings of the National Academy of Sciences of the United States of America. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Yao Y, Xu X, Yang L, Zhu J, Wan J, Shen L, et al. Patient-Derived Organoids Predict Chemoradiation Responses of Locally Advanced Rectal Cancer. Cell Stem Cell. 2020;26(1):17–26 e6. 10.1016/j.stem.2019.10.010 [DOI] [PubMed] [Google Scholar]
  • 52.Tiriac H, Belleau P, Engle DD, Plenker D, Deschenes A, Somerville TDD, et al. Organoid Profiling Identifies Common Responders to Chemotherapy in Pancreatic Cancer. Cancer Discov. 2018;8(9):1112–29. 10.1158/2159-8290.CD-18-0349 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Berkers G, van Mourik P, Vonk AM, Kruisselbrink E, Dekkers JF, de Winter-de Groot KM, et al. Rectal Organoids Enable Personalized Treatment of Cystic Fibrosis. Cell Rep. 2019;26(7):1701–8 e3. 10.1016/j.celrep.2019.01.068 [DOI] [PubMed] [Google Scholar]
  • 54.Dekkers JF WC, de Jonge HR, Bronsveld I, Janssens HM, de Winter-de Groot KM, Brandsma AM, et al. A functional CFTR assay using primary cystic fibrosis intestinal organoids. Nat Med. 2013;19(7):939–45. 10.1038/nm.3201 [DOI] [PubMed] [Google Scholar]
  • 55.den Boer E, Meesters RJ, van Zelst BD, Luider TM, Hazes JM, Heil SG, et al. Measuring methotrexate polyglutamates in red blood cells: a new LC-MS/MS-based method. Analytical and bioanalytical chemistry. 2013;405(5):1673–81. 10.1007/s00216-012-6581-7 [DOI] [PubMed] [Google Scholar]
  • 56.Muller IB, Lin M, Struys EA, Heydari P, Hebing RCF, Nurmohamed MT, et al. Development and validation of a sensitive UHPLC-MS/MS-based method for the analysis of folylpolyglutamate synthetase enzymatic activity in peripheral blood mononuclear cells: application in rheumatoid arthritis and leukemia patients. Ther Drug Monit. 2019. [DOI] [PubMed] [Google Scholar]
  • 57.Hanley JA, McNeil BJ. A method of comparing the areas under receiver operating characteristic curves derived from the same cases. Radiology. 1983;148(3):839–43. 10.1148/radiology.148.3.6878708 [DOI] [PubMed] [Google Scholar]
  • 58.Hanley JA, McNeil BJ. The meaning and use of the area under a receiver operating characteristic (ROC) curve. Radiology. 1982;143(1):29–36. 10.1148/radiology.143.1.7063747 [DOI] [PubMed] [Google Scholar]
  • 59.Rots MG, Pieters R, Kaspers GJ, van Zantwijk CH, Noordhuis P, Mauritz R, et al. Differential methotrexate resistance in childhood T- versus common/preB-acute lymphoblastic leukemia can be measured by an in situ thymidylate synthase inhibition assay, but not by the MTT assay. Blood. 1999;93(3):1067–74. [PubMed] [Google Scholar]
  • 60.van der Laan BF JG, Kathmann GA, Westerhof GR, Schornagel JH, Hordijk GJ. In vitro activity of novel antifolates against human squamous carcinoma cell lines of the head and neck with inherent resistance to methotrexate. Int J Cancer. 1992;51(6):909–14. 10.1002/ijc.2910510613 [DOI] [PubMed] [Google Scholar]
  • 61.Jansen G, Mauritz R, Drori S, Sprecher H, Kathmann I, Bunni M, et al. A structurally altered human reduced folate carrier with increased folic acid transport mediates a novel mechanism of antifolate resistance. J Biol Chem. 1998;273(46):30189–98. 10.1074/jbc.273.46.30189 [DOI] [PubMed] [Google Scholar]
  • 62.den Hoed MA, Lopez-Lopez E, te Winkel ML, Tissing W, de Rooij JD, Gutierrez-Camino A, et al. Genetic and metabolic determinants of methotrexate-induced mucositis in pediatric acute lymphoblastic leukemia. The pharmacogenomics journal. 2015;15(3):248–54. 10.1038/tpj.2014.63 [DOI] [PubMed] [Google Scholar]
  • 63.Galpin AJ, Schuetz JD, Masson E, Yanishevski Y, Synold TW, Barredo JC, et al. Differences in folylpolyglutamate synthetase and dihydrofolate reductase expression in human B-lineage versus T-lineage leukemic lymphoblasts: mechanisms for lineage differences in methotrexate polyglutamylation and cytotoxicity. Molecular pharmacology. 1997;52(1):155–63. 10.1124/mol.52.1.155 [DOI] [PubMed] [Google Scholar]
  • 64.Pauley JL, Panetta JC, Crews KR, Pei D, Cheng C, McCormick J, et al. Between-course targeting of methotrexate exposure using pharmacokinetically guided dosage adjustments. Cancer chemotherapy and pharmacology. 2013;72(2):369–78. 10.1007/s00280-013-2206-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Van der Beek JN, Oosterom N, Pieters R, de Jonge R, van den Heuvel-Eibrink MM, Heil SG. The effect of leucovorin rescue therapy on methotrexate-induced oral mucositis in the treatment of paediatric ALL: A systematic review. Critical reviews in oncology/hematology. 2019;142:1–8. 10.1016/j.critrevonc.2019.07.003 [DOI] [PubMed] [Google Scholar]
  • 66.Sonis ST. Regimen-related gastrointestinal toxicities in cancer patients. Current opinion in supportive and palliative care. 2010;4(1):26–30. 10.1097/SPC.0b013e328335fb76 [DOI] [PubMed] [Google Scholar]
  • 67.Blutt SE, Crawford SE, Ramani S, Zou WY, Estes MK. Engineered Human Gastrointestinal Cultures to Study the Microbiome and Infectious Diseases. Cellular and molecular gastroenterology and hepatology. 2018;5(3):241–51. 10.1016/j.jcmgh.2017.12.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Noel G, Baetz NW, Staab JF, Donowitz M, Kovbasnjuk O, Pasetti MF, et al. A primary human macrophage-enteroid co-culture model to investigate mucosal gut physiology and host-pathogen interactions. Scientific reports. 2017;7:45270 10.1038/srep45270 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Schutgens F, Rookmaaker MB, Margaritis T, Rios A, Ammerlaan C, Jansen J, et al. Tubuloids derived from human adult kidney and urine for personalized disease modeling. Nat Biotechnol. 2019;37(3):303–13. 10.1038/s41587-019-0048-8 [DOI] [PubMed] [Google Scholar]
  • 70.Huch M, Gehart H, van Boxtel R, Hamer K, Blokzijl F, Verstegen MM, et al. Long-term culture of genome-stable bipotent stem cells from adult human liver. Cell. 2015;160(1–2):299–312. 10.1016/j.cell.2014.11.050 [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Obul Reddy Bandapalli

15 Jan 2020

PONE-D-19-32380

Patient-derived oral mucosa organoids as an in vitro model for methotrexate induced toxicity in pediatric acute lymphoblastic leukemia

PLOS ONE

Dear Ms Oosterom,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

Please address all the points raised by the reviewers carefully point by point.

We would appreciate receiving your revised manuscript by Feb 29 2020 11:59PM. When you are ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter.

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). This letter should be uploaded as separate file and labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. This file should be uploaded as separate file and labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. This file should be uploaded as separate file and labeled 'Manuscript'.

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

We look forward to receiving your revised manuscript.

Kind regards,

Obul Reddy Bandapalli, MSc, PhD

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements:

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at http://www.plosone.org/attachments/PLOSOne_formatting_sample_main_body.pdf and http://www.plosone.org/attachments/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. Please provide additional details regarding participant consent. In the ethics statement in the Methods and online submission information, please ensure that you have specified what type of consent you obtained (for instance, written or verbal). If your study included minors under age 18, state whether you obtained consent from parents or guardians. If the need for consent was waived by the ethics committee, please include this information.

3. We noticed you have some minor occurrence(s) of overlapping text with the following previous publication(s), which needs to be addressed:

https://doi.org/10.1158/2159-8290.CD-18-1522

https://doi.org/10.1097/FTD.0000000000000638

In your revision ensure you cite all your sources (including your own works), and quote or rephrase any duplicated text outside the Methods section. Further consideration is dependent on these concerns being addressed.

4. We note that this submission reports a functional enzymological study with kinetic and thermodynamic data. The reporting of these data should include the temperature, pH and pressure, as well as the identity of the catalyst and its origins, the method of preparation, criteria for purity and assay conditions. We recommend that you refer to the Standards for Reporting Enzymology Data (STRENDA) of the Beilstein Institut for details regarding the adequate description of experimental conditions and reporting of enzyme activity data: https://www.beilstein-strenda-db.org/strenda/public/guidelines.xhtml. Please note that the Beilstein Institut’s STRENDA database automatically checks manuscript data for guideline compliance, as well as making them publicly available after publication and assigning them a specific DOI number for reference and tracking purposes. If you obtain a STRENDA Registry number (SRN) and PDF containing all your functional enzymology data, please include these as Supplementary files.

5. Please report in your Methods a paragraph on immunohistochemistry staining.

6. In your Methods section, please give the sources of any cell lines used in your study.

7. We note that you have a patent relating to material pertinent to this article. Please provide an amended statement of Competing Interests to declare this patent (with details including name and number), along with any other relevant declarations relating to employment, consultancy, patents, products in development or modified products etc. Please confirm that this does not alter your adherence to all PLOS ONE policies on sharing data and materials, as detailed online in our guide for authors http://journals.plos.org/plosone/s/competing-interests by including the following statement: "This does not alter our adherence to  PLOS ONE policies on sharing data and materials.” If there are restrictions on sharing of data and/or materials, please state these. Please note that we cannot proceed with consideration of your article until this information has been declared.

This information should be included in your cover letter; we will change the online submission form on your behalf.

8. Please upload a copy of Supporting Information Table S3 which you refer to in your text on page 13.

 

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: No

Reviewer #3: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: No

Reviewer #3: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: In the original research article entitled, “Patient-derived oral mucosa organoids as an in vitro model for methotrexate induced toxicity in pediatric acute lymphoblastic leukemia“, Else Driehuis and colleagues describe a novel assay to assess methotrexate-induced toxicity in human squamous cells. They have established a protocol to grow human wildtype oral mucosa organoids. These three-dimensional structures can be maintained in culture, do not require immortalization, and recapitulate the multilayered composition of the epithelial lining of the oral mucosa. They have utilized their co-culture model to assess the effects of leucovorin rescue on methotrexate-induced squamous cell damage as functions of timing and concentration. The paper is interesting and well-presented, and the experiments provide compelling new information about the use of leucovorin as a rescue agent for methotrexate. Some limitations exist, which will be detailed in the sections that follow.

Major Considerations:

1. In general, methotrexate toxicity is not constrained to oral mucositis alone. Hepatic and hematological toxicities are also dose-limiting factors. The paper does not address these issues, which are common limitations in dose-intensified therapies. Moreover, leucovoroin is a highly soluble drug, and pretreatment may have unintended complications in over-rescuing methotrexate (over-rescue literature should be cited). The paper should be revised to include these limitations, and statement about the use of leucovorin pre-treatment should be modified to reflect the potential down-side of changing rescue strategies.

2. In clinical practice, methotrexate-induced oral mucositis can be somewhat mitigated by the use of oral bicarbonate rinses. Was this condition used as a control variable in the experiments that were performed? If so, what was the relative impact compared to conditions that used leucovorin pre-treatment?

3. The Discussion section could be shorter, since some of the same points are re-stated in the Conclusion paragraph.

Lesser Points:

1. At line 300, “As, clinically….” appears to be missing a word.

2. The authors should define IC50 at its first use.

3. The legend for Figure 4 could be more clearly written to help the reader. understand what effects LV rescue have on MTX toxicity in leukemia cell lines.

Reviewer #2: The authors report the use of oral mucosal organoids from adjacent normal tissue of patients who underwent surgery for head and neck squamous cell carcinoma. The development of these organoids is interesting, but while this manuscript gives the impression that these are new organoids, these are in fact previously published (ref #34 cited by the authors; this should have been made clear). The authors use these organoids to study oral toxicity of MTX and its rescue using leucovorin. There are a number of problems with this manuscript, at least two of which rise to the level of fatal flaws: Statistical analyses to support the differences claimed are lacking entirely, and critique #4 below is viewed as a completely unacceptable attempt to mislead the reviewers/readers (this may be a typo, but then this raises questions about rigor elsewhere in this work).

Critiques:

1. The drug unit labels are unnecessarily confusing on the viability plots (i.e., Fig. 1E, 2C, etc). It is difficult for most people to figure out what -0.5 log10 uM converts to, and this is the approximate effect size (based in change in IC50) induced by leucovorin treatment. It is fine to show the data on log scale, but it would be much easier for the doses to be shown as 1 uM, 0.1 uM, 0.01 uM, etc.

2. Why are the number of replicates from different timepoints/conditions so different in many experiments? For example Fig 1E, 2B, 2E, and several others. This gives the strong impression that the data shown are an amalgamation of different experiments performed at different times. If the experiments were performed simultaneously, this should be clearly stated in the methods. If the data are a mix of experiments at same vs different time points, all of the experiments should be repeated at one time point with the same number of replicates per condition. It is a good idea to repeat experiments to ensure reproducibility, but results of only one representative experiment should be shown.

3. Are the changes in IC50s induced by leucovorin in Fig 1E, 3C, 3E and 3F statistically significant? Significance of all relevant comparisons should be assessed, with appropriate correction for multiple hypothesis testing. The lack of statistical analyses to support the conclusions is a major flaw throughout this paper.

4. LV pre-treatment also protects leukemia cells. The authors claim that this is less than in normal oral mucosa, but again there is no statistical evidence to support that this is a significant difference. FURTHERMORE, IT IS GLARING THAT THE PRE-TREATMENT OF LEUKEMIA CELLS IS DONE WITH A DOSE OF LEUKOCORIN 10-FOLD LOWER THAN THAT OF THE ORGANOIDS!!! As indicated on the plots of Fig 3B, PT = 0.1 uM in organoids, and Fig 4, 0.01 uM leukemia cells. Is it any surprise that there is less rescue with a lower dose of leucovorin?? This is viewed as a completely unacceptable attempt to mislead the reviewers and readers, and in the opinion of this reviewer this alone is grounds for rejection of the paper. Unless perhaps this is a typo, but then this raises concerns about rigor elsewhere in this manuscript.

Reviewer #3: This is an original article that describes a series of experiments designed to explore the impact of methotrexate and leucovorin on the viability of cells in human oral mucosa organoids, as a model for MTX induced mucositis, an important side effect that arises in some patients in the course of treatment for childhood ALL. The authors report that, as expected, mucosal cells are killed by methotrexate in a time and concentration dependent manner. Additionally, mucosal cells also recapitulated the expected behavior of becoming more resistant to MTX-induced death upon co-incubation with leucovorin, an effect dependent upon the timing of addition as well as to the concentration of the two inhibitors.

The experiments are very well described and in sufficient detail, which should enable others to reproduce and extend these findings. Although the results are completely in accord with our understanding of the mechanisms of MTX and leucovorin and the clinical experiences with them, the manuscript provides clarity and a solid basis to support the investigators’ claim that this is a reliable model for identifying factors that modulate mucositis development in ALL patients. As pointed out by the authors, mucositis in vivo may involve additional mechanisms such as the microbiome and/or the immune system, factors absent from their model. Nonetheless, the ability to test cells from different individuals with more or less sensitivity to MTX could significantly advance our understanding of genetic factors that pre-dispose to this toxicity, above and beyond what is already known about MTX metabolism.

Mucositis occurs not infrequently during ALL treatment, and so this is an important clinical aspect of ALL therapy. The authors comments on the details of MTX and leucovorin treatment in childhood ALL are appropriate, and I agree with their note that earlier treatment with leucovorin (including pre-treatment) may impair the disease responses to MTX, so should be considered with care. Nonetheless, the idea of topical application of leucovorin to the oral mucosa is a good one, and could improve toxicity with minimal negative impact on overall cure rates.

The article is well written and the narrative flows well. The manuscript would benefit from a few additions. First of all, the authors should clarify the state of the mucosal cells during the experiment. Are they isolated as single cells, which seems to be the case since the authors report they trypsinize them just prior to adding drugs? Or do they have time to assemble into the 3d organoids, as mentioned in the title? Secondly, there is little or no discussion of why this system is superior to testing of individual isolated cell cultures, and so it would be good to comment on that. Do the authors think that penetration of drugs into the interior of 3d organoids is an important factor?

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Reviewer #3: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files to be viewed.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2020 May 18;15(5):e0231588. doi: 10.1371/journal.pone.0231588.r002

Author response to Decision Letter 0


3 Mar 2020

To: Obul Reddy Bandapalli (MSc, PhD), Academic Editor PLOS ONE

Subject: rebuttal Ms. Ref. No. PONE-D-19-32380

Dear Dr. Bandapalli,

We are pleased to hereby resubmit our revised manuscript ‘Patient-derived oral mucosa organoids as an in vitro model for methotrexate induced toxicity in pediatric acute lymphoblastic leukemia’.

We thank you and the reviewers for the time and effort put into reviewing our work, and appreciate the constructive criticism. We have addressed each the concerns raised by the reviewers, and give a point-by-point rebuttal below.

Major changes in the revised manuscript include:

- The introduction of a Log-axis in all the figures, which makes them easier to interpret for the reader.

- Statistical analysis to further support the biological differences observed between different experimental groups in our experiments.

- Rewriting of the discussion section to have less overlap with the conclusion.

- Rewriting of the Figure legends.

With these modifications, we feel that our manuscript has improved, and we hope you would consider our study for publication in Plos One.

Best regards,

Prof.Dr. Marry M. van den Heuvel-Eibrink

Editor

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at http://www.plosone.org/attachments/PLOSOne_formatting_sample_main_body.pdf and http://www.plosone.org/attachments/PLOSOne_formatting_sample_title_authors_affiliations.pdf

We have checked the style templates and the manuscript now fits the requirements.

2. Please provide additional details regarding participant consent. In the ethics statement in the Methods and online submission information, please ensure that you have specified what type of consent you obtained (for instance, written or verbal). If your study included minors under age 18, state whether you obtained consent from parents or guardians. If the need for consent was waived by the ethics committee, please include this information.

We have made the ethics statement more complete as following:

“Collection of human tissues was compliant with the guidelines of the European Network of Research Ethics Committees (EUREC) and European and national laws, and written informed consent was obtained from all donors. All donors were > age 18 years. The Biobank Research Ethics Committee of the UMC Utrecht approved the biobanking protocol (12-093 HUB-Cancer).”

3. We noticed you have some minor occurrence(s) of overlapping text with the following previous publication(s), which needs to be addressed:

https://doi.org/10.1158/2159-8290.CD-18-1522

https://doi.org/10.1097/FTD.0000000000000638

In your revision ensure you cite all your sources (including your own works), and quote or rephrase any duplicated text outside the Methods section. Further consideration is dependent on these concerns being addressed.

We have referred to the publications mentioned above.

4. We note that this submission reports a functional enzymological study with kinetic and thermodynamic data. The reporting of these data should include the temperature, pH and pressure, as well as the identity of the catalyst and its origins, the method of preparation, criteria for purity and assay conditions. We recommend that you refer to the Standards for Reporting Enzymology Data (STRENDA) of the Beilstein Institut for details regarding the adequate description of experimental conditions and reporting of enzyme activity data: https://www.beilstein-strenda-db.org/strenda/public/guidelines.xhtml. Please note that the Beilstein Institut’s STRENDA database automatically checks manuscript data for guideline compliance, as well as making them publicly available after publication and assigning them a specific DOI number for reference and tracking purposes. If you obtain a STRENDA Registry number (SRN) and PDF containing all your functional enzymology data, please include these as Supplementary files.

We have made changes accordingly:

“FPGS catalytic activity analysis in organoids and leukemic cell lines was performed essentially as described by Muller et al (2019, PMID 31008996). In short, FPGS protein was isolated from organoids and cell lines by sonication (Sonoplus Mini 20; Bandelin, Berlin, Germany) on ice for 2 · 10 seconds (10 second intervals with 90% amplitude and 30-second intervals between samples) in FPGS extraction buffer (50 mM Tris, 20 mM KCl, 10 mM MgCl2, and 5 mM DTT, pH 7.4, 4oC, in MilliQ (MilliQ Advantage A10; Merck Millipore, Burlington, MA, USA), followed by centrifugation in an Eppendorf centrifuge (12.000 • g, 15 minutes, 4°C). FPGS-mediated conversion of MTX-PG1 to MTX-PG2 is determined in cell extracts (10-200 µg protein) in a total volume of 250 µL containing final concentrations of 100 mM Tris, 20 mM KCl, 20 mM MgCl2, 10 mM DTT, 10 mM ATP, 250 mM MTX-PG1, and 4 mM 15N-labeled L glutamatic acid (Sigma-Aldrich, cat no. 332143-100MG) at a pH of 8.85 (set ith HCl) under atmospheric pressure. After 2 hour incubations at 37oC, quantities of MTX-(15N)PG2 formed were measured by LC-MS/MS as described above (Muller et al (2019, PMID 31008996). FPGS activity is expressed as pmol MTX-PG2 formed per microgram protein per hour (pmol•µg-1•hr-1).”

5. Please report in your Methods a paragraph on immunohistochemistry staining.

The following section was added in the revised manuscript:

Immunohistochemistry

Tissues or organoids were fixed in 4% paraformaldehyde overnight, dehydrated, and

embedded in paraffin. Sections were subjected to H&E as well as IHC staining, using the

antibodies shown in the table below. Stainings were performed at the pathology department

of the UMCU (Utrecht, the Netherlands).

Protein supplier Order nr. Host species Clone Lot nr. Dilution Antigen retrieval method

TP63 Abcam AB735 Mouse 4AB AB735 1:800 Citrate

KI67 Monosan MONX10293 Mouse MM1 10293 1:2000 Citrate autoclave

KRT13 Progen 10523 Mouse 1C7 10523 1:100 Citrate

6. In your Methods section, please give the sources of any cell lines used in your study.

We have added the sources of the cell lines into the methods section.

In the experiments, we used organoids from five different donors. Furthermore, we performed drug screens in leukemia cell lines. T cell ALL (MOLT16, HSB2, Jurkat) and B cell ALL (Nalm6, REH) cell lines were obtained from DSMZ-German Collection of Microorganisms and Cell Cultures (DSMZ, Braunschweig, Germany).

7. We note that you have a patent relating to material pertinent to this article. Please provide an amended statement of

Competing Interests to declare this patent (with details including name and number), along with any other relevant declarations relating to employment, consultancy, patents, products in development or modified products etc. Please confirm that this does not alter your adherence to all PLOS ONE policies on sharing data and materials, as detailed online in our guide for authors http://journals.plos.org/plosone/s/competing-interests by including the following statement: "This does not alter our adherence to PLOS ONE policies on sharing data and materials.” If there are restrictions on sharing of data and/or materials, please state these. Please note that we cannot proceed with consideration of your article until this information has been declared.

This information should be included in your cover letter; we will change the online submission

form on your behalf.

We have included the statements in our competing interest section of the revised manuscript. We have added the number of the relevant patent: WO2009/022907, WO2010/090513, WO2012/168930.

8. Please upload a copy of Supporting Information Table S3 which you refer to in your text on page 13.

This was a typo and is Table S2 in the document. We have changed this accordingly.

Reviewer #1

Major points:

1. In general, methotrexate toxicity is not constrained to oral mucositis alone. Hepatic and hematological toxicities are also dose-limiting factors. The paper does not address these issues, which are common limitations in dose-intensified therapies. Moreover, leucovorin is a highly soluble drug, and pretreatment may have unintended complications in over-rescuing methotrexate (over-rescue literature should be cited). The paper should be revised to include these limitations, and statement about the use of leucovorin pre-treatment should be modified to reflect the potential down-side of changing rescue strategies.

We agree with the reviewer that hepatotoxicity, nephrotoxicity and hematological toxicities are other dose-limiting factors in high-dose methotrexate treatment. We chose to study oral mucositis as this is one of the most frequently associated toxicities with HD-MTX courses - occurring in ~20% of patients. Since this paper addresses the effect of methotrexate on the oral mucosa (using an oral mucosa epithelial cell model), we did not explore these other toxicities in this current study. Regardless, we did not mean to mitigate the effect of these toxicities, neither claim that the suggested LV pre-treatment would not result in systemic effect that might be negative. To discuss these potential systemic effects, we have added a sentence in both the introduction and the discussion of the revised document to clarify that these other toxicities could be explored further in a similar manner. Organoid cultures of liver- and kidney- cells to perform similar studies have been described in literature.

Introduction (line 27 – 28): ‘However, patients often suffer from MTX toxicities such as hepatotoxicity, nephrotoxicity, hematological malignancies and mucositis.’

Discussion: ‘By making minor adaptations to the culture media that was previously described to culture oral mucosa organoids, we have shown that organoids can be used to study MTX-toxicity in vitro. These modifications can be applied to organoid cultures from other organs, such as kidney or liver organoids. As such, these models can perhaps be used to study relevant MTX-toxicity that is observed in other organs.’

Moreover, in the abstract, we have changed the sentence ‘Although effective, the use of MTX often results in oral mucositis, which is characterized by epithelial cell death’ to: ‘Although effective, the use of MTX often results in severe side-effects, including oral mucositis, characterized by epithelial cell death’ (line 12 – line 13) in the revised manuscript. In studying these toxicities, it would be worthwhile to explore the effect of MTX, but also of 6-Mercaptopurine and PEG-Asparaginase, as these chemotherapeutic regimens are often administered within the same treatment course and the different toxicity profiles should be distinguished.

Secondly, we fully agree with the reviewer that we should be cautious of the possible ‘rescuing’ effects of a leucovorin pretreatment on leukemia cells. We have added more ‘overrescue’ literature into the introduction and have discussed the possible downsides of a leucovorin pretreatment more clearly in the discussion section. We would not like to state that this intervention should be directly applied into clinics, but it is a promising in vitro strategy of which the clinical safety should be assessed first.

Introduction (line 66 - line 69): In contrast, several pediatric ALL studies (38-41) have suggested that Leucovorin rescue therapy decreases toxicity rates, but might be accompanied by an increased risk of relapse in ALL. This phenomenon has been referred to as the folate ‘over-rescue’ principle, where not only healthy cells, but also tumor cells are rescued.

Discussion (line 473 – 474): However, it is important to test clinical safety of the described interventions in order not to decrease anti-leukemic activity of MTX.

Currently, a Leucovorin formula prepared in a concentration of 0.1 mg/mL to apply locally to the oral mucosa is available in our clinics. Before introducing such an application to the clinics, we would need to assess whether locally applied Leucovorin to the oral mucosa could lead to systemic levels interfering with leukemia treatment. Hypothetically, when we would locally administer 5 mL of a formula with 0.1 mg/mL to the oral mucosa of a patient of 10 kg with a distribution volume of 3.2 L/kg, in a scenario where 100% would enter the systemic circulation, this would lead to a concentration of 0.5 mg*3.2*10 kg = 16 ng/mL. Previous studies showed that intravenous administration of 50 mg Leucovorin leads to much higher peak plasma levels around 1 ug/mL with a short plasma half-life of around 30 minutes. A pharmacokinetic study, which assesses the plasma levels in health individuals after application of such an intervention to the oral mucosa could aid in determining clinical safety.

2. In clinical practice, methotrexate-induced oral mucositis can be somewhat mitigated by the use of oral bicarbonate rinses. Was this condition used as a control variable in the experiments that were performed? If so, what was the relative impact compared to conditions that used leucovorin pre-treatment?

We thank the reviewer for the suggestion of studying the effect of oral bicarbonate rinses on mitigating methotrexate-induced oral mucositis. This intervention was not studied here as previous studies, including several meta-analyses, showed conflicting evidence concerning the effect of oral bicarbonate rinses, even though we know it is often applied in clinics. We agree with the reviewer that this would be a very clean model to study the effect of MTX and oral bicarbonate rinses on the oral mucosa cells. However, for now this was outside the scope of this study.

3. The Discussion section could be shorter, since some of the same points are re-stated in the Conclusion paragraph.

We agree with the Reviewer that the discussion section of the original manuscript summarized our findings, rather than discussing implication etc. Therefore, we have extensively rewritten the discussion section of our revised manuscript to address this point. We expect this rewriting will resolve the issue raised by the Reviewer.

Minor points:

1. At line 300, “As, clinically….” appears to be missing a word.

We thank the reviewer for his/her critical reading and have modified this sentence as suggested by the reviewer: “As MTX plasma levels have dropped by 54 hours post infusion in clinics…” to correct this typo.

2. The authors should define IC50 at its first use.

We have added a definition of IC50 at line 212-213: “IC50 values (=half maximal inhibitory concentration; the concentration at which 50% of cells are dead)…” to correct this issue.

3. The legend for Figure 4 could be more clearly written to help the reader understand what effects LV rescue have on MTX toxicity in leukemia cell lines.

We agree with the Reviewer that the legends of the Figures could be improved to aid the reader understand the data shown in the Figure. Therefore, we have revised the Legend of this figure in the revised manuscript. In addition, we have also improved the legends for Figure 1 – 4 of the revised manuscript. We hope these modifications will resolve the Reviewer’s concerns.

See for changes: line 618 – 789.

Reviewer #2

1. The drug unit labels are unnecessarily confusing on the viability plots (i.e., Fig. 1E, 2C, etc). It is difficult for most people to figure out what -0.5 log10 uM converts to, and this is the approximate effect size (based in change in IC50) induced by leucovorin treatment. It is fine to show the data on log scale, but it would be much easier for the doses to be shown as 1 uM, 0.1 uM, 0.01 uM, etc.

We agree with this comment of the Reviewer. In the revised manuscript, we have amended all graphs in all the figures of the original manuscript that contain a log-axis according to the Reviewers suggestion. We agree with the Reviewer that this modification makes the graphs easier to interpret for the reader and therefore thank the Reviewer for his/her suggestion.

2. Why are the number of replicates from different timepoints/conditions so different in many experiments? For example, Fig 1E, 2B, 2E, and several others. This gives the strong impression that the data shown are an amalgamation of different experiments performed at different times. If the experiments were performed simultaneously, this should be clearly stated in the methods. If the data are a mix of experiments at same vs different time points, all of the experiments should be repeated at one time point with the same number of replicates per condition. It is a good idea to repeat experiments to ensure reproducibility, but results of only one representative experiment should be shown.

In the revised manuscript, we have amended the figures to only show matching replicates. With matching, we mean they were obtained in the same experiment (for example IC50 value MTX + 0.05 LV rescue 12 h post MTX matching to the IC50 values MTX +0.05 uM LV rescue 24 hours post MTX since these were values obtained from plates where the MTX exposure was initiated at the same time (in the same 384 well plate) and the LV was added at different timepoints. To further facilitate the reader to understand which IC50 matches to which other (since we show three replicate experiments, we have indicated this with different symbols. Hence, all the IC50 values obtained in one experiment have round symbols, whereas those obtained in a different experiment (performed in a different week for example) are shown with square symbols.

Furthermore, we have removed any control (MTX only) values that could not be matched to other IC50 values (for example obtained after LV rescue) from these graphs. We initially placed these values also in the Figures of the original manuscript, in light of transparency. However, we agree with the Reviewer that these values, since obtained separately from the other datapoints, do not add to the figures and only generate confusion for the reader.

Taken together, we hope these modifications will satisfy the Reviewers comments.

3. Are the changes in IC50s induced by leucovorin in Fig 1E, 3C, 3E and 3F statistically significant? Significance of all relevant comparisons should be assessed, with appropriate correction for multiple hypothesis testing. The lack of statistical analyses to support the conclusions is a major flaw throughout this paper.

We agree with the reviewer that statistical analyses of the data is of importance and have analyzed the data as described in our new methods section:

Methods (line 286 - line 293): Raw luminescence values obtained after readout of MTX drug screens, were normalized to the average of untreated controls (n=3, 100%) and staurosporin-treated controls (n=3, 0%), using the formula: (value- 0% control)/(100% control – 0% control)*100%. Resulting percentages of viability were transferred to GraphPad v8 to generate kill curve. Curves were fit using the option ‘log inhibitor vs. normalized response – variable slope. IC50s/AUC values were obtained from GraphPad analysis. The change in IC50 values over time was assessed using a Pearson correlation, performed in Graphpad prism, using XY analysis, correlation option. AUC’s were compared as previously described.

We have added the correlations and p-values in our results and figures. The addition of these statistics does not alter our conclusions.

4. LV pre-treatment also protects leukemia cells. The authors claim that this is less than in normal oral mucosa, but again there is no statistical evidence to support that this is a significant difference. FURTHERMORE, IT IS GLARING THAT THE PRE-TREATMENT OF LEUKEMIA CELLS IS DONE WITH A DOSE OF LEUKOCORIN 10-FOLD LOWER THAN THAT OF THE ORGANOIDS!!! As indicated on the plots of Fig 3B, PT = 0.1 uM in organoids, and Fig 4, 0.01 uM leukemia cells. Is it any surprise that there is less rescue with a lower dose of leucovorin?? This is viewed as a completely unacceptable attempt to mislead the reviewers and readers, and in the opinion of this reviewer this alone is grounds for rejection of the paper. Unless perhaps this is a typo, but then this raises concerns about rigor elsewhere in this manuscript.

We understand the concern of the reviewer, and would like to sincerely apologize for this typo, which we have amended in the revised manuscript Of course, we would not have made these statements if the used concentration of LV was indeed 10 times lower, since we have written this manuscript to share our findings with the research community, and thus would not at all attempt to mislead the reader in any way. Although we understand the reviewers concern, we have gone over the remainder of the manuscript to assure no other typos are present, and can assure him/her that we would not purposely introduce such differences between the controls of our experiments.

Reviewer #3

1. First of all, the authors should clarify the state of the mucosal cells during the experiment. Are they isolated as single cells, which seems to be the case since the authors report they trypsinize them just prior to adding drugs? Or do they have time to assemble into the 3d organoids, as mentioned in the title?

This study was performed with organoids derived from human oral mucosa resections. These cultures are established from tissue digest. Once established, cultures can be passaged regularly, by digestion (using TrypLE) of the formed 3D organoids into smaller fragments (single cells/clumps of cells). Once replated, these single cells/clumps of cells, will proliferate and form new organoids, which contain both proliferating basal cells, and differentiated keratinocytes (see Figure 1A). In this study, MTX toxicity was determined on established organoid cultures (>3 passages), not fresh tissue isolates.

In the method section of the paper, we state that organoids were split to single cells 2 days prior to the start of drug screening. This means that by the time of screening, organoids have already reformed from these single cells, to form 3D structures. In the revised manuscript, we now state this more clearly: ‘Two days later, when single cells already reformed into small organoids, the organoids were collected from the BME by addition of 1 mg/mL dispase II (Sigma-Aldrich, cat.no. D4693) to the medium of the organoids.’

Regardless, since this was apparently not made clear by us in the original manuscript, we have modified the introduction section on organoids in our revised manuscript. It now contains the following section:

Introduction (line 88 - line 91): ‘Recently, we described an organoid model derived from healthy oral mucosa (34). The resulting patient-derived structures consist of a functional stratified squamous epithelium that can be maintained and expanded in culture for over six months. *NEW SECTION STARTING* Upon passaging, organoids grown from primary oral mucosa tissue, can be broken into smaller fragments, which will proliferate and result in the formation of new organoids. As such, organoid technology allows us to multiply human wildtype epithelial cells for a wide variety of applications, including drug screening.’

Taken together, we hope these modifications will satisfy the Reviewers concern.

2. Secondly, there is little or no discussion of why this system is superior to testing of individual isolated cell cultures, and so it would be good to comment on that. Do the authors think that penetration of drugs into the interior of 3D organoids is an important factor?

In the introduction and in the answers above, we have more clearly described the advantages of a 3D model as opposed to single cell cultures.

The penetration of Methotrexate into intracellular compartments is mediated through different transporters, of which the RFC1, the PCFT and the FR receptors are most important. As methotrexate is administered intravenously in pediatric ALL patients, the main mode of transportation is through the RFC1 transporter, which is both located apically as basolaterally, whereas for instance the PCFT transporter is mainly located apically in the gastro-intestinal tract. As transporters are located at all sides of the organoid, we expect that penetration of the drugs will not hamper results in a 3D-organoid model.

We hope that we have addressed the concerns of the reviewers sufficiently and are looking forward to hear your decision concerning the revised manuscript.

Kind regards,

Prof.Dr. Marry M. van den Heuvel-Eibrink

Attachment

Submitted filename: 20200228 Rebuttal PLOS ONE_FINAL.docx

Decision Letter 1

Obul Reddy Bandapalli

27 Mar 2020

Patient-derived oral mucosa organoids as an in vitro model for methotrexate induced toxicity in pediatric acute lymphoblastic leukemia

PONE-D-19-32380R1

Dear Dr. Oosterom,

We are pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it complies with all outstanding technical requirements.

Within one week, you will receive an e-mail containing information on the amendments required prior to publication. When all required modifications have been addressed, you will receive a formal acceptance letter and your manuscript will proceed to our production department and be scheduled for publication.

Shortly after the formal acceptance letter is sent, an invoice for payment will follow. To ensure an efficient production and billing process, please log into Editorial Manager at https://www.editorialmanager.com/pone/, click the "Update My Information" link at the top of the page, and update your user information. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to enable them to help maximize its impact. If they will be preparing press materials for this manuscript, you must inform our press team as soon as possible and no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

With kind regards,

Obul Reddy Bandapalli, MSc, PhD

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: All comments have been addressed

Reviewer #2: All comments have been addressed

Reviewer #3: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: (No Response)

Reviewer #2: The authors have addressed all of my concerns. My major critique turned out to be a typo that has been corrected.

Reviewer #3: none

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Reviewer #3: No

Acceptance letter

Obul Reddy Bandapalli

4 May 2020

PONE-D-19-32380R1

Patient-derived oral mucosa organoids as an in vitro model for methotrexate induced toxicity in pediatric acute lymphoblastic leukemia

Dear Dr. Oosterom:

I am pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please notify them about your upcoming paper at this point, to enable them to help maximize its impact. If they will be preparing press materials for this manuscript, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

For any other questions or concerns, please email plosone@plos.org.

Thank you for submitting your work to PLOS ONE.

With kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Obul Reddy Bandapalli

Academic Editor

PLOS ONE

Associated Data

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

    Supplementary Materials

    S1 Fig. Mechanism of action of MTX treatment.

    Leucovorin (5-formylTHF) is represented in bold / italic. MTX enters the cell mainly through the Reduced Folate Carrier 1 (RFC1), Proton Coupled Folate Transporter (PCFT), Membrane Folate Transporters (MFR) or by passive diffusion through the cell membrane. While circulating, MTX contains one polyglutamate group (MTX-PG1). Once inside the cell, MTX is polyglutamated by Folylpolyglutamate Synthetase (FPGS) with up to seven polyglutamate groups. Long-chain MTX-PG’s (MTX-PG4-7) cannot be transported out of the cell before de-polyglutamation by Gamma-Glutamyl Hydrolase (GGH). Short-chain MTX-PG’s (MTX-PG1-3) will be actively transported out of the cell by ABCC1-4, ABCB1 and ABCG2 transporters. MTX is cytotoxic as it impairs purine- and pyrimidine synthesis by inhibiting the enzymes Dihydrofolate Reductase (DHFR) and Thymidylate Synthase (TYMS). Abbreviations: ABCB1—ATP Binding Cassette Subfamily B Member 1; ABCC1-4—ATP Binding Cassette Subfamily C Member 1–4; ABCG2—ATP Binding Cassette Subfamily G Member 2; DHFR–Dihydrofolate Reductase; FPGS–Folylpolyglutamate Synthetase; GGH–Gamma-Glutamyl Hydrolase; MFR–Membrane Folate Transporter; MTHFR—Methylene tetrahydrofolate reductase; MTHFD1—Methylenetetrahydrofolate Dehydrogenase, Cyclohydrolase And Formyltetrahydrofolate Synthetase 1; PCFT–Proton-Coupled Folate Transporter; RFC1 –Reduced Folate Carrier; SHMT—Serine hydroxymethyltransferase; TS–Thymidylate Synthase.

    (PDF)

    S2 Fig. Characterization of oral mucosa organoids and the effect of pretreatment on intracellular MTX-PG levels.

    A. Oral mucosa organoids are derived of human normal cells, and not cancer cells. Number of mutations detected by whole exome sequencing in the healthy oral mucosa organoids used in this study, and their corresponding tumor organoids. Mutational load is low (2 for N1, 0 for T1), especially when compared to the tumor organoids. B. FPGS activity (in pmol MTX-PG2/h/mg) in organoid line versus CCRF-CEM reference leukemia cell line. C. Effect of PT on MTX-PG levels in oral mucosa organoid lines derived from two different donors. D. Effect of PT on MTX-PG levels in two B-ALL and two T-ALL cell lines.

    (PDF)

    S3 Fig. Organoid cultures retain their morphology and growth speed when grown in folate deprived medium.

    A. Brightfield microscopy images of organoid line N1 and N2, when grown in either complete medium, or folate deprived medium. Scalebar, 500 μm. B. Growth speed of organoid cultures in both media tested. Growth was assessed by collection of cell pellets at day 0, 3, 5, 7, 10 and 14. Cell number was assessed by cell titer glow and values were made relative to day 0. C. Quantitative PCR assessing expression of genes relevant for methotrexate metabolism. Experiment was performed in triplicate, results of all three experiments are shown here.

    (PDF)

    S4 Fig. Technical details of drugscreen performed in this study.

    A. Schematic layout of a drug screen plate as used in this study. The gradient of MTX is depicted using a color gradient (red indicates high concentration, green indicates low concentration). Here, the MTX concentrations used for organoids are depicted. Each concentration is tested in technical triplicate. Different blocks receive LV rescue at different timepoints after the start of MTX treatment, as indicated. Staurosporine treated wells are used as positive controls and are set to 0% viability, wells only receiving drug solvent are used is negative controls, and are set to 100% viability. B. Brightfield microscopy images showing the morphology of N1 organoids in drug screening plates on the day of readout. C. Brightfield microscopy images showing the morphology of N2 organoids in drug screening plates on the day of readout.

    (PDF)

    S1 Table. Clinical information of patients. Relevant clinical information is given on the patient that participated in this study, and form whose tissue organoids were derived.

    (PDF)

    S2 Table. Z-scores of drug screens performed in this study.

    (PDF)

    S3 Table. Comparison of mutations detected by WES in matching normal and tumor organoid lines.

    All mutation detected in organoid line N1, T1, N2 and T2 are shown. Here, normal tissue was used as a reference.

    (XLSX)

    S4 Table. Sequences of primers used for quantitative PCR.

    5’ to 3’ sequences of primers used to assess gene expression by quantitative PCR in this study.

    (PDF)

    Attachment

    Submitted filename: 20200228 Rebuttal PLOS ONE_FINAL.docx

    Data Availability Statement

    All data are described within the current manuscript as it is.


    Articles from PLoS ONE are provided here courtesy of PLOS

    RESOURCES