Abstract
Brain tumor is the leading cause of cancer related death in children. Clinically relevant animals are critical for new therapy development. To address the potential impact of animal gender on tumorigenicity rate, xenograft growth and in vivo drug responses, we retrospectively analyzed 99 of our established patient derived orthotopic xenograft mouse models (orthotopic PDX or PDOX). From 27 patient tumors, including 5 glioblastomas (GBMs), 11 medulloblastomas (MBs), 4 ependymomas (EPNs), 4 atypical teratoid/rhabdoid tumors (ATRTs) and 3 diffuse intrinsic pontine gliomas (DIPGs), that were directly implanted into matching locations in the brains of approximately equal numbers of male and female animals (n = 310) in age-matched (within 2-week age-difference) SCID mice, the tumor formation rate was 50.6 ± 21.5% in male and 52.7 ± 23.5% in female mice with animal survival times of 192.6 ± 31.7 days in male and 173.9 ± 34.5 days in female mice (P = 0.46) regardless of pathological diagnosis. Once established, PDOX tumors were serially subtransplanted for up to VII passage. Analysis of 1,595 mice from 59 PDOX models (18 GBMs, 18 MBs, 5 ATRTs, 6 EPNs, 7 DIPGs and 5 PENTs) during passage II and VII revealed similar tumor take rates of the 6 different tumor types between male (85.4 ± 15.5%) and female mice (84.7 ± 15.2%) (P = 0.74), and animal survival times were 96.7 ± 23.3 days in male mice and 99.7 ± 20 days in female (P = 0.25). A total of 284 mice from 7 GBM, 2 MB, 1 ATRT, 1 EPN, 2 DIPG and 1 PNET were treated with a series of standard and investigational drugs/compounds. The overall survival times were 106.9 ± 25.7 days in male mice, and 110.9 ± 31.8 days in female mice (P = 0.41), similar results were observed when different types/models were analyzed separately. In conclusion, our data demonstrated that the gender of SCID mice did not have a major impact on animal model development nor drug responses in vivo, and SCID mice of both genders are appropriate for use.
Keywords: Gender, Brain tumor, Tumorigenicity, Survival time, Drug treatment
1. Introduction
Brain tumor is the most common childhood cancer other than leukemia and remain the leading cause of cancer-related death in children [1-7]. Despite recent advances of imaging diagnosis, surgical techniques and continually improved chemo-, radiation- and many other adjuvant therapies, the overall survival in many types of malignant pediatric brain tumors has not improved significantly. The five-year survival rate in children with high grade glioma is still <25%, and nearly all patients with diffuse intrinsic pontine glioma (DIPG) succumb to this disease within 9–12 months [8]. Even among brain tumor survivors, many patients are left with long-term cognitive and/or neuroendocrine sequalae [9-13]. Clinically relevant and molecularly accurate animal models are therefore needed to understand tumor biology and to develop novel and less-toxic therapies.
Direct implantation of patient tumor cells into the matching locations in the brains of SCID mice, i.e. cerebral tumor into mouse cerebra, cerebellar tumors to mouse cerebella and brain stem tumors into mouse brain stem, has led to the successful establishment of multiple pediatric brain tumor models [14-23]. Detailed characterization has also shown faithful replication of key histopathology, invasion and metastasis, as well as genetic features of the original tumors in these patient derived orthotopic xenograft (orthotopic PDX or PDOX) mouse models [14-17,20,24-26]. Many of them have been successfully utilized for understanding tumor biology and testing various new therapies [27-32].
While it is well established that host gender is important for cancers predominantly or exclusively occur in female (such as breast cancer) or male (such as prostate cancer) patients, recent studies have further shown that the gender of patient can have significant impact on tumor biology and drug responses in cancers that affect both female and male patients as well. In GBMs, for example, a recent study showed that standard therapy is more effective in female patients compared with male patients and cell cycle and integrin signaling were shown to be critical determinants of survival for male and female patients, respectively [33]. An analysis of GBM patient data also showed that proliferating monocytic myeloid-derived suppressor cells (mMDSC) that block antitumor immunity MDSCs were predominant in male tumors and that a high granulocytic myeloid-derived suppressor cell (gMDSC)/IL1β gene signature correlated with poor prognosis in female patients [34]. A large-scale glioma genome-wide association studies (GWAS) even identified sex-specific risk locus [35].
Despite widespread use of patient-derived xenograft animal models, the impact of host gender (e.g. male v. s female mice) on xenograft tumor growth and therapy responsiveness has not been well established. While male animals were frequently used for acute genotoxicity [36], female mice are primarily favored for preclinical drug testing [37-40] because male mice from separate litters usually do not get along in the cage and some mice are lost to injuries during the experiments. Recent studies, however, showed that tumor biology and drug responses in female mice were not always identical to tumors in male animals [34,41]. Since the genes and signaling pathways targeted by different treatment as well as drug metabolism and toxicity can potentially be influenced by the sex hormone and/or gender-related host differences, it is important to systematically analyze the similarities and differences between female and male immunodeficient animals on the xenograft tumor formation rate and in vivo responsiveness toward standard therapies and various investigational anti-cancer agents using a large panel of PDOX models. Indeed, the issue of gender-drug effect in vivo has gained prominence in recent years and National Institute of Health (NIH) announced the requirement for the use of both male and female animals in preclinical testing of drug safety and efficacy [42].
To evaluate the impact of animal sex difference in PDOX models of pediatric brain tumors, we retrospectively analyzed our panel of 99 PDOX models in which childhood brain tumors were implanted with approximately equal numbers of male and female SCID mice, on their tumorigenicity rate and xenograft growth changes with primary patient tumors and during serial in vivo subtransplantations as well as their responses toward standard and investigational anti-cancer agents. Our aim is to provide a large series of experimental data to inform future selection of animal genders during model development and preclinical drug testing.
2. Materials and Methods
2.1. Tumor tissues
Fresh surgical tumor tissues were collected in patients undergoing surgery at Texas Children’s Hospital, including patients with glioblastoma (GBM), medulloblastoma (MB), ependymoma (EPN), atypical teratoid rhabdoid tumor (ATRT) and primitive neuroectodermal tumor (PENT). Samples from consented autopsied diffuse intrinsic pontine glioma (DIPG) were also collected. Signed informed consent was obtained from the patient or legal guardian prior to sample acquisition in accordance with Institutional Review Board (IRB) policy as we described previously [14,15,17,20].
2.2. Patient-derived orthotopic xenograft mouse models
Tumor tissues were washed and minced with fine scissors into small fragments. Single cells and small clumps (3–5 cells per clump) of tumor cells were collected with a 40 μm cell strainer, then resuspended in Dulbecco’s modified Eagle’s medium (DMEM) to achieve a final concentration of 0.5 × 108 live cells per ml, as assessed by trypan blue staining, and transferred to animal facility on ice. The SCID mice were bred and housed in a specific pathogen free animal facility at Texas Children’s Hospital. All the experiments were conducted using an Institutional Animal Care and Use Committee approved protocol. Surgical transplantation of tumor cells into mouse cerebellum, cerebrum and brain stem, usually completed within 60 min of tumor removal, was performed as described previously [14]. Both male and female mice, aged 4–6 weeks, were anesthetized with sodium pentobarbital (50 mg/kg, i. p. Injections) or isoflurane . Tumor cells (1 × 105) were suspended in 2 μL of culture medium and injected into the right cerebellum (1 mm to the right of the midline, 1 mm posterior to the lamboidal suture, and 3 mm deep), or right cerebral hemisphere (1 mm to the right of the midline, 1.5 mm anterior to the lamboidal suture and 3 mm deep) or brain stem (1 mm to the right of the midline, 1 mm posterior to the lamboidal suture, and 5.2 mm deep) via a 10 μL 26-gauge Hamilton Gastight 1701 syringe needle (Hamilton Company, Reno, NV, http://www.hamiltoncompany.com). The animals were then monitored daily for development of neurological deficits, at which time they were euthanized, and their brains removed for histopathologic examination. Those mice without any neurological deficit after 12 months were euthanized and examined for tumor development.
2.3. Serial subtransplantation of xenografts in vivo in mouse brains
Whole brains of donor mice were aseptically removed, coronal cut into halves, and transferred back to the tissue culture laboratory. Xenograft tumors were then dissected, mechanically dissociated into cell suspensions, and injected into the brains of recipient SCID mice as described above [14,15,17,20].
2.4. In vivo treatment for PDOX model
Two weeks after tumor cell implantation, mice were administered standard and investigational drugs/compounds, including radiation, oncolytic virus (SVV-001) [43], ABT888, MLN8237, TMZ, Flavopiridol, VPA, SAHA, Cisplatin and Panobinostat [25] as showed in Table 2. To determine any survival benefits from drug treatment, the mice were monitored daily until they developed signs of neurologic deficit or became moribund, at which time they were euthanized, and their brains were removed for analysis.
Table 2.
Tumorigenicity of established PDOX models during serial subransplantation between male and female SCID mice.
Tumor type | Model number | Total mouse number | Passages |
---|---|---|---|
GBM | 18 (34a) | 467 | II-VII |
MB | 18 (33) | 523 | II-V |
ATRT | 5 (10) | 120 | II-V |
EPN | 6 (8) | 91 | II-V |
DIPG | 7 (20) | 310 | II-VII |
PNET | 5 (7) | 84 | II-V |
Total | 59 (112) | 1595 | II-VII |
Model number including different passages in vivo.
2.5. Statistical analysis
Comparison of the numbers of PDOX models between male and female mice were performed with the Student t-test, Animal survival times were evaluated by log-rank analysis followed by pairwise multiple comparison procedures using the Holm-Sidak method with SigmaPlot 13 (Systat Software). P values < 0.05 are considered significant.
3. Results
3.1. Development of PDOX mouse models of pediatric brain tumors
As detailed in the Materials and Methods section, patient tumor tissues obtained from surgery or autopsy were mechanically dissociated and injected into the matching locations in the brains of SCID mice at 1 × 105 cells for all intra-cerebral (IC) and intra-cerebellar (ICb) tumors and 5 × 104 for intra-brain stem tumors (Fig. 1A and B) [44]. All the animals were carefully monitored, and the mice that developed signs of neurological deficit or became moribund were euthanized following our Institutional Animal Care and Use Committee (IACUC) approved protocol. Gross examination usually revealed the enlargement of the mouse brains with frequent visualization of tumor growth, particularly intra-cerebral or intra-cerebellar tumors (Fig. 1C). Serial section of paraffin embedded whole mouse brains was also performed to confirm tumor formation (Fig. 1C).
Fig. 1.
Xenograft tumor of PDOX in the mouse brain. (A, B) Images illustrating intra-cerebral (IC) and intra-cerebellar (ICb) and intra-brain stem (IBs) injections of pediatric brain tumor cells in the brains of SCID mice. 1 × 105 brain tumor cells was injected into SCID mouse within 60 min of tumor resection into cerebellum, cerebrum or brain stem to establish the medulloblastoma (MB), glioblastoma (GBM), ependymoma (EPN), diffuse intrinsic pontine glioma (DIPG), Atypical Teratoid Rhabdoid Tumor (ATRT) and primitive neuroectodermal tumor (PNET) models. (C) Gross appearance of H&E-stained mouse brain with huge xenograft tumors.
3.2. The impact of animal gender on tumorigenicity and xenograft growth at passage I
To determine if animal gender affected xenograft forming capabilities of patient tumors, we retrospectively analyzed gender distribution in 99 PDOX models in which at least 1 mouse developed xenograft tumor and identified 27 models of which the human tumor cells were implanted into both male and female mice that were age-matched (within 2 weeks age-difference) (Table 1). The remaining 72 models were excluded because the tumors were injected to SCID mice of single gender as the gender differences were not considered an issue when human brain tumors were implanted several years earlier in our laboratory and the selection of mice were primarily based on the availability of young (4–6 week) animals (to mimic the developing brain of childhood brain tumors). Among 27 PDOX models, there were 5 GBMs, 11 MBs, 4 EPNs, 4 ATRTs and 3 DIPGs (Supplemental Table 1) with a comparable average number of male ranging from 3.8 ± 1 (in EPNs) to 10.3 ± 4.7 (in DIPGs), and of female mice from 4.2 ± 0.8 (in GBMs) to 8.7 ± 3.2 (in DIPGs). Since the overall number differences between male and female mice in all the five models were not significant (P > 0.05) (Fig. 2A and Table S1), we proceed to compare the tumorigenicity in these models.
Table 1.
Tumorigenicity of pediatric brain tumors in male and female SCID mice.
Model name |
Formed tumor in both gender |
Formed tumor in single gender |
||
---|---|---|---|---|
Model number |
Mouse number |
Model number |
Mouse number |
|
GBM | 5 | 48 | 24 | 296 |
MB | 11 | 124 | 39 | 411 |
ATRT | 4 | 47 | 7 | 93 |
EPN | 4 | 34 | 18 | 169 |
DIPG | 3 | 57 | 11 | 217 |
Subtotal | 27 | 310 | 99 | 1186 |
GBM: Glioblastoma Multiforme.
MB: Medulloblastoma.
ATRT: Atypical Teratoid Rhabdoid Tumor.
EPN: Anaplastic Ependymoma.
DIPG: Brain Stem Glioma.
Fig. 2.
The impact of animal gender on tumorigenicity and survival time of PDOX models of primary passage in vivo. From total 1186 mice of passage I (P–I), the animal number (A), tumor formation rate (B, C) and survival times (D, E) of male mice bearing tumors were compared with that of female mice from 5 GBMs, 11 MBs, 4 ATRTs, 4 EPNs and 3 DIPGs models when the mice reached the endpoint. P > 0.05 means no difference between male and female mice.
Tumor forming rate in the 5 GBM models were 78.2 ± 33.4% in male and 84 ± 35.8% in female mice. The difference was not significant (P = 0.18). Similar results were found in MBs (78.4 ± 27.9% in male vs. 75 ± 33.2% in female) (P = 0.28); ATRTs (61.7 ± 32.8% in male vs. 70.6 ± 25.5% in female) (P = 0.26); EPNs (47.5 ± 14.5% in male: 53.8 ± 17% in female) (P = 0.39) and DIPGs (37.9 ± 20.3% in male: 22.7 ± 15.5% in female) (P = 0.24) (Fig. 2B). When all the male and female mice were grouped together regardless of the tumor type, the tumor take rate was 50.6 ± 21.5% in male and 52.7 ± 23.5% in female, not statistically significant (P = 0.92) (Fig. 2C).
We next examined if there were any differences of tumor growth between male and female mice by comparing animal survival times of tumor-bearing mice. In the 5 GBM models, the animal survival times was 126 ± 21.8 days in male and 112.6 ± 20.5 days in female mice (P = 0.31) and in the 11 MB models, 183.5 ± 40.3 days in male and 191.1 ± 43.2 days in female mice (P = 0.6). Similarly findings were revealed in the 4 ATRT models (141.4 ± 20.8 days in male and 129.6 ± 15.7 days in female mice) (P = 0.43), 4 EPN models (225.5 ± 75.7 days in male and 255.4 ± 90.3 days in female mice) (P = 0.58), and 3 DIPG models (286.8 ± 31.7 days in male and 180.8 ± 37.5 days in female mice) (P = 0.52) (Fig. 2D). When combined, the survival times of male mice of the 5 types of pediatric brain tumors was 192.6 ± 31.7 days, similar to that in female mice (173.9 ± 34.5 days) (P = 0.46) (Fig. 2E). Altogether, our data did not reveal significant differences in tumor take rate and tumor growth between male and female mice during PDOX model establishment from patient tumor tissues of 5 types of pediatric brain tumors.
3.3. The impact of animal gender on tumor take and growth during serial subtransplantations
Similar to human brain tumors, animals bearing malignant tumor xenografts in their brain eventually all died of the disease. Serial subtransplantation, in which PDOX tumors harvested from donor mice were implanted serially in recipient mice, provided a critical means for sustained supply of PDOX models. It is therefore very important to examine if animal gender would affect tumor take and tumor growth during serial in vivo subtransplantations. From our database, we identified 59 PDOX models (with a total of 1595 mice), including 18 GBMs, 18 MBs, 5 ATRTs, 6 EPNs, 7 DIPGs and 5 PNETs (note: PNET was discontinued in the 2016 WHO classification), that were implanted with approximately equal numbers of male and female animals during serial subtransplantation from passage II up to passage VII. Among these PDOX models, the average number of male animals ranged from 5.1 ± 2 (in EPNs) to 8.2 ± 4.8 (in MBs), and of female mice from 6.3 ± 3 (in EPNs) to 7.4 ± 3.1 (in DIPGs). Since the overall number differences between male and female mice in all the five models were not significant (P > 0.05) (Table 2 and Fig. 3A), we proceeded to compare the tumorigenicity in these models.
Fig. 3.
The impact of animal gender on tumorigenicity and survival time of subtransplanted PDOX models in vivo. From total 1595 mice of passage II (P-II) to passage VII (P-VII), the animal number (A), tumor formation rate (B, C) and survival times (D, E) of male mice bearing tumors were compared with that of female mice from 18 GBMs, 18 MBs, 5 ATRTs, 6 EPNs, 7 DIPGs and 5 PNETs models when the mice reached the endpoint; P > 0.05 means no difference between male and female mice.
Tumor forming rate in the 18 GBM models were 93.9 ± 12.8% in male and 96.5 ± 10.1% in female mice. The difference was not significant (P = 0.33). Similar results were found in MBs (93.4 ± 11.7% in male vs. 94.9 ± 9.4% in female) (P = 0.18); ATRTs (94.2 ± 13.5% in male vs. 94.4 ± 9.8% in female) (P = 0.23); EPNs (83.1 ± 27.6% in male and 72.5 ± 31.5% in female) (P = 0.45); DIPGs (48.1 ± 27.6% in male and 52.8 ± 24.9% in female) (P = 0.9) and PNETs (100 ± 0% in male vs. 96.8 ± 5.5% in female) (P = 0.16) (Fig. 3B). When all the male and female mice were grouped together regardless of the tumor type, the tumor take rate was 85.4 ± 15.5% in male and 85 ± 15.2% in female, not statistically significant (P = 0.74) (Fig. 3C).
With the similar tumor take rate and animal number per group, we next compared the animal survival times of tumor bearing mice. In GBM models, it was 84.4 ± 18.3 days in male mice and 77.5 ± 15.7 days in female mice (P = 0.92); whereas in MB models, 119 ± 34.4 days in male and 124.2 ± 32.4 days in female mice (P = 0.89). Similar results were found in ATRT models (94.5 ± 21.6 days in male mice vs 96.9 ± 15.2 days in female, P = 0.06); in EPN models (129.7 ± 23.9 days vs 138.9 ± 24.9 days, P = 0.62); in DIPG models (74.9 ± 28.4 days vs and 82 ± 21.9 days, P = 0.81); in PNET models (77.8 ± 13.4 days vs 79.3 ± 9.8 days, P = 0.76) (Fig. 3D). When combined, the survival times of tumor-bearing mice was 97 ± 23.3% days in male and 100 ± 20% days in female (P = 0.25) (Fig. 3E), indicating that the SCID mice of both genders are appropriate for tumorigenicity and xenograft growth in PDOX model development.
3.4. The impact of animal gender on in vivo drug responses in preclinical drug testing
A total of 284 mice from 7 GBM, 2 MB, 1 ATRT, 1 EPN, 2 DIPG and 1 PNET were treated with a series of standard and investigational drugs/compounds, including radiation, oncolytic virus (SVV-001), ABT888, MLN8237, temozolomide (TMZ), Flavopiridol, valproic acid (VPA), SAHA, Echinomycin, MCB613, Cisplatin, Panobinostat (Table 3A). The scheme of administration was described as in Table 3B. The overall survival times were 106.93 ± 25.7 days in male mice, and 110.87 ± 31.76 in female mice (P = 0.41) in models with relative even numbers of male and female animals (7.9 ± 8.7 mice in male and 7.4 ± 6.3) (P = 0.46) (Table 4). The overall differences of drug response between male and female mice were not significant (P > 0.05) (Fig. 4). suggesting that both genders of SCID mice are appropriate for preclinical drug testing in PDOX models of pediatric brain tumors.
Table 3a.
Compounds for treatment of 6 types of PDOX models.
Tumor type |
Model number |
Treatment/compounds |
---|---|---|
GBM | 7 | XRT; ABT888; MLN8237; TMZ; Flavopiridol; VPA; SAHA |
MB | 2 | SVV-001; TMZ; ABT888 |
ATRT | 1 | Cisplatin |
EPN | 1 | XRT; ABT888 |
DIPG | 2 | Flavopiridol; Panobinostat |
PNET | 1 | SVV-001 |
Total | 14 | 10 |
Table 3b.
In Vivo treatment scheme of PDOX models.
Compounds | Name in the study | Target | Solution for compounds | Treatment scheme in PDOX model |
Route of administration |
---|---|---|---|---|---|
Alvocidib | Flavopiridol | CDK9 kinase inhibitor | 1 mg/mL in 0.2 mL DMSO add 14.8 mL PBS | 5 mg/kg/day ×12 days | i.p. |
Alisertib | MLN8237 | Aurora A Kinase inhibitor | 5 mg/mL in 10% HPBCD/1% sodium bicarbonate | 30 mg/kg/day x 12 days | gavage |
NA | Radiation | DNA | NA | 2 Gy/day x 5 days | In located brain |
Temozolomide | TMZ | Alkylate/methylate DNA | In water | 50 mg/kg/day ×5 day | gavage |
Panobinostat | Panobinostat | HDACi | 2 mg/mL in DMSO (1.3%)/DPBS | 10 mg/kg (5 d on-2 d off) x 2 cycle | i.p. |
Valproic acid | VPA | HDACi | In 0.9% sodium chloride solution | 600 mg/mL x 7 days (1 μl/hr), | Pump |
Suberoylanilide Hydroxamic Acid | SAHA | HDACi | In PBS/DMSO (1:1) | 100 mg/kg, Qd, 2 weeks | i.p. |
Veliparib | ABT888 | PARPi inhibitor | In 2% DMSO | 10μl/g ×5 days, Qd | i.p. |
Cisplatin | Cisplatin | DNA | Sterile water | 5 mL/kg on days 1 and 4 | i.p. |
Seneca Valley Virus | SVV-001 | Oncolytic virus | In PBS | 20μ1*10^8vp/ul for one time | Tail vein |
Table 4.
Survival time in male/female mice after treatment for 6 types of PDOX models.
Gender | Number of mouse |
Mouse number per model |
Survival time (days) |
---|---|---|---|
Male | 144 | 7.6 ± 8.7 | 106.9 ± 25.7 |
Female | 140 | 7.4 ± 6.3 (P = 0.46) | 110.9 ± 31.8 (P = 0.41) |
Fig. 4.
The impact of animal gender on drug responses in PDOX models with the preclinical drug testing. A total of 284 mice from 7 GBMs, 2 MBs, 1 ATRT, 1 EPNs, 2 DIPGs and 1 PNET models were treated 10 of standard and investigational drugs/compounds, including radiation, oncolytic virus (SVV-001), ABT888, MLN8237, TMZ, Flavopiridol, VPA, SAHA, Cisplatin and Panobinostat. The survival times of male mice bearing tumors was compared with that of female mice when the mice reached the endpoint. P > 0.05 means no difference between male and female mice.
We also analyzed a total of 316 mice from 5 GBMs, 3 MBs and 1 ATRT treated with a series of 11 standard and investigational drugs/compounds from the NIH/NCI Pediatric Preclincial Testing Consortium (PPTC) (Table S2 and Fig. S1). The overall survival times were 65.4 ± 9.3 days in male mice, and 64.5 ± 10.7 in female mice (P = 0.86) with approximately numbers of male and female animals (5.4 ± 1.1 mice in male and 5.4 ± 1.1) (P = 1) (Table S2). All the mice had no significantly difference in drug response between male and female (Fig. S1). Our data showed gender of SCID mice did not have major impact on PPTC drug response in PDOX model development, that both genders of SCID mice are appropriate for preclinical drug testing in PDOX models of pediatric brain tumors.
4. Discussion
In this study, we examined if animal gender has a significant impact on the development and the drug responsiveness of pediatric brain tumor PDOX models. Our retrospective analysis of a large cohorts of PDOX models demonstrated that the gender of SCID mice did not significantly affect tumor formation, in vivo growth nor in vivo drug responses. Our data suggested that SCID mice of both genders are appropriate for brain tumor PDOX model development and preclinical drug testing.
A fundamental assumption using human tumor xenografts for cancer research is that the xenograft tumors preserve key features of the originating patient tumors. We and many other groups have been actively engaged in the establishment and characterization of PDOX mouse models to support the understanding of tumor biology and to facilitate preclinical drug testing [14-16,21,24,45-49]. Since patient tumor tissues are very difficult to obtain, every effort should be made to increase the success of model development and to enhance the predictability of preclinical drug testing in future clinical applications. For pediatric brain tumor PDOX models, we have tried to recapitulate the microenvironment of the developing brain, which is known to critically affect the biological behavior of xenograft tumors [50,51], by using young (4–6 week old) mice and by implanting brain tumor cells into the matching locations in the mouse brains [14-16,21,22,24,45,46]. While many studies favored the use of single gender (male or female) SCID mice due to various reasons, our systematic analysis support the use of both male and female mice, particularly when they were young (4–6 week), for pediatric brain tumor studies. In addition to the fact that pediatric brain tumor is not a sex-specific or gender-predominant cancer (e.g., breast cancer and prostate cancer), young age of animals and the universally low levels of sex hormones in these mice in our study may have minimized the microenvironmental differences between male and female SCID mice and contributed to their relatively homogeneous accommodation of newly implanted human tumor cells and the subsequent responses of the xenografts toward anti-cancer treatments.
One added benefit of using both genders of SCID mice, particularly in those laboratories that breed their own SCID mice, is the increased availability of animals (as all the litters can be used). In this way, large cohorts of models can be established to support biological and preclinical drug testing with significantly reduced cost of SCID mice (50% reduction as compared with the use of single gender mouse).
As preclinical drug development is one of the primary goals of animal model development, it is also important to evaluate if there is any difference of animal gender on drug responses in vivo. For pediatric brain tumors, however, no previous studies have been published about the impact of animal gender on drug response. Recognizing the vast differences of therapeutic targets and mechanisms of action, we analyzed a large collection of anti-cancer agents to minimize or prevent our risk of inaccurate or biased conclusion caused by single drugs. Analysis of 284 mice from 14 models (7 GBM, 2 MB, 1 ATRT, 1 EPN, 2 DIPG and 1 PNET) treated a series of standard and investigational drugs/compounds, including fractionated radiation, Temozolamide (TMZ), oncolytic virus (SVV-001), ABT888, MLN8237, Flavopiridol, valproic acid, SAHA, Panobinostat and Cisplatin, did not find major differences between male and female SCID mice in our PDOX models. These data combined with the results obtained from a panel of target therapies completed with the Pediatric Preclinical Testing Consortium provided the much-needed experimental data to justify and support the use of SCIC mice of both genders in preclinical drug testing for pediatric brain tumor models.
In conclusion, our retrospective analysis of a large panel of PDOX models derived from different types of pediatric brain tumors and a series of anti-cancer drugs in vivo did not identify significant differences between male and female SCID mice on tumorigenicity, growth, sub-transplantability, and in vivo drug responses. Our data, accumulated over 15 years, support the use of both genders of SCID mice for PDOX model development and preclinical drug testing.
This study demonstrated that the gender of SCID mice dids not have major impact on animal development nor in drug responses, and SCID mice of both genders are appropriate for brain tumor PDOX model development as well as preclinical drug testing.
Supplementary Material
Acknowledgments
The authors wish to thank all the veterinarians and veterinary technicians of the Center of Comparative Medicine in Baylor College of Medicineand staff members of the Feigin Center animal facility at Texas Children’s Hospital for their excellent support of our animal experiments;
Funding
This work was funded by NIH RO1 CA185402 (Li XN), NIH/NCI Pediatric Preclinical Testing Consortium UO1 (1U01CA199288) (Li XN), Cancer Prevention & Research Institute of Texas (CPRIT) RP150032 and RP-170169 (Li XN), Cure Starts Now Foundation (Li XN), Golfers against Cancer (Li XN), CDMRP DOD PRCRP CA100735 (Li XN), Childhood brain tumor foundation (Li XN), National Brain Tumor Foundation (LI XN) and St. Baldrick’s Foundation (Grant 2532341503, Su JM).
Abbreviations
- ABT888
Veliparib
- ATRTs
Atypical teratoid/rhabdoid tumors
- DIPGs
Diffuse intrinsic pontine gliomas
- EPNs
Ependymomas
- GBMs
Glioblastomas
- MBs
Medulloblastomas
- PDOX
Patient derived orthotopic xenograft mouse models
- PENTs
Primitive neuroectodermal tumor
- SAHA
Suberoylanilide Hydroxamic Acid
- SVV-001
Seneca Valley Virus
- TMZ
Temozolomide
- VPA
Valproic acid
- XRT
Radiation
Footnotes
Appendix A. Supplementary data
Supplementary data to this article can be found online at https://doi.org/10.1016/j.canlet.2020.08.035.
Declaration of competing interest
The authors declare no conflict of interest.
References
- [1].Ostrom QT, Gittleman H, de Blank PM, Finlay JL, Gurney JG, McKean-Cowdin R, Stearns DS, Wolff JE, Liu M, Wolinsky Y, Kruchko C, Barnholtz-Sloan JS, American brain tumor association adolescent and young adult primary brain and central nervous system tumors diagnosed in the United States in 2008-2012, Neuro Oncol. 18 (Suppl 1) (2016) il–i50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Hwang EI, Kool M, Burger PC, Capper D, Chavez L, Brabetz S, Williams-Hughes C, Billups C, Heier L, Jaju A, Michalski J, Li Y, Leary S, Zhou T, von Deimling A, Jones DTW, Fouladi M, Pollack IF, Gajjar A, Packer RJ, Pfister SM, Olson JM, Extensive molecular and clinical heterogeneity in patients with histologically diagnosed CNS-PNET treated as a single entity: a report from the Children’s oncology group randomized ACNS0332 trial, J. Clin. Oncol (2018), JCO2017764720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Wolff JE, Driever PH, Erdlenbruch B, Kortmann RD, Rutkowski S, Pietsch T, Parker C, Metz MW, Gnekow A, Kramm CM, Intensive chemotherapy improves survival in pediatric high-grade glioma after gross total resection: results of the HIT-GBM-C protocol, Cancer 116 (2010) 705–712. [DOI] [PubMed] [Google Scholar]
- [4].Wu G, Diaz AK, Paugh BS, Rankin SL, Ju B, Li Y, Zhu X, Qu C, Chen X, Zhang J, Easton J, Edmonson M, Ma X, Lu C, Nagahawatte P, Hedlund E, Rusch M, Pounds S, Lin T, Onar-Thomas A, Huether R, Kriwacki R, Parker M, Gupta P, Becksfort J, Wei L, Mulder HL, Boggs K, Vadodaria B, Yergeau D, Russell JC, Ochoa K, Fulton RS, Fulton LL, Jones C, Boop FA, Broniscer A, Wetmore C, Gajjar A, Ding L, Mardis ER, Wilson RK, Taylor MR, Downing JR, Ellison DW, Zhang J, Baker SJ, The genomic landscape of diffuse intrinsic pontine glioma and pediatric non-brainstem high-grade glioma, Nat. Genet 46 (2014) 444–450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Thompson EM, Hielscher T, Bouffet E, Remke M, Luu B, Gururangan S, McLendon RE, Bigner DD, Lipp ES, Perreault S, Cho YJ, Grant G, Kim SK, Lee JY, Rao AAN, Giannini C, Li KKW, Ng HK, Yao Y, Kumabe T, Tominaga T, Grajkowska WA, Perek-Polnik M, Low DCY, Seow WT, Chang KTE, Mora J, Pollack IF, Hamilton RL, Leary S, Moore AS, Ingram WJ, Hallahan AR, Jouvet A, Fevre-Montange M, Vasiljevic A, Faure-Conter C, Shofuda T, Kagawa N, Hashimoto N, Jabado N, Weil AG, Gayden T, Wataya T, Shalaby T, Grotzer M, Zitterbart K, Sterba J, Kren L, Hortobagyi T, Klekner A, Laszlo B, Pocza T, Hauser P, Schuller U, Jung S, Jang WY, French PJ, Kros JM, van Veelen MC, Massimi L, Leonard JR, Rubin JB, Vibhakar R, Chambless LB, Cooper MK, Thompson RC, Faria CC, Carvalho A, Nunes S, Pimentel J, Fan X, Muraszko KM, Lopez-Aguilar E, Lyden D, Garzia L, Shih DJH, Kijima N, Schneider C, Adamski J, Northcott PA, Kool M, Jones DTW, Chan JA, Nikolic A, Garre ML, Van Meir EG, Osuka S, Olson JJ, Jahangiri A, Castro BA, Gupta N, Weiss WA, Moxon-Emre I, Mabbott DJ, Lassaletta A, Hawkins CE, Tabori U, Drake J, Kulkarni A, Dirks P, Rutka JT, Korshunov A, Pfister SM, Packer RJ, Ramaswamy V, Taylor MD, Prognostic value of medulloblastoma extent of resection after accounting for molecular subgroup: a retrospective integrated clinical and molecular analysis, Lancet Oncol. 17 (2016) 484–495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Mack SC, Pajtler KW, Chavez L, Okonechnikov K, Bertrand KC, Wang X, Erkek S, Federation A, Song A, Lee C, Wang X, McDonald L, Morrow JJ, Saiakhova A, Sin-Chan P, Wu Q, Michaelraj KA, Miller TE, Hubert CG, Ryzhova M, Garzia L, Donovan L, Dombrowski S, Factor DC, Luu B, Valentim CLL, Gimple RC, Morton A, Kim L, Prager BC, Lee JJY, Wu X, Zuccaro J, Thompson Y, Holgado BL, Reimand J, Ke SQ, Tropper A, Lai S, Vijayarajah S, Doan S, Mahadev V, Minan AF, Grobner SN, Lienhard M, Zapatka M, Huang Z, Aldape KD, Carcaboso AM, Houghton PJ, Keir ST, Milde T, Witt H, Li Y, Li CJ, Bian XW, Jones DTW, Scott I, Singh SK, Huang A, Dirks PB, Bouffet E, Bradner JE, Ramaswamy V, Jabado N, Rutka JT, Northcott PA, Lupien M, Lichter P, Korshunov A, Scacheri PC, Pfister SM, Kool M, Taylor MD, Rich JN, Therapeutic targeting of ependymoma as informed by oncogenic enhancer profiling, Nature 553 (2018) 101–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Mackay A, Burford A, Molinari V, Jones DTW, Izquierdo E, Brouwer-Visser J, Giangaspero F, Haberler C, Pietsch T, Jacques TS, Figarella-Branger D, Rodriguez D, Morgan PS, Raman P, Waanders AJ, Resnick AC, Massimino M, Garrè ML, Smith H, Capper D, Pfister SM, Würdinger T, Tam R, Garcia J, Thakur MD, Vassal G, Grill J, Jaspan T, Varlet P, Jones C, Molecular, pathological, radiological, and immune profiling of non-brainstem pediatric high-grade glioma from the HERBY phase II randomized trial, Cane. Cell 33 (2018) 829–842, e825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Johung TB, Monje M, Diffuse intrinsic pontine glioma: new pathophysiological insights and emerging therapeutic targets, Curr. Neuropharmacol 15 (2017) 88–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Sun LR, Cooper S, Neurological complications of the treatment of pediatric neoplastic disorders, Pediatr. Neurol 85 (2018) 33–42. [DOI] [PubMed] [Google Scholar]
- [10].Li MD, Forkert ND, Kundu P, Ambler C, Lober RM, Burns TC, Barnes PD, Gibbs IC, Grant GA, Fisher PG, Cheshier SH, Campen CJ, Monje M, Yeom KW, Brain perfusion and diffusion abnormalities in children treated for posterior fossa brain tumors, J. Pediatr 185 (2017) 173–180 e173. [DOI] [PubMed] [Google Scholar]
- [11].Krishnatry R, Zhukova N, Guerreiro Stucklin AS, Pole JD, Mistry M, Fried I, Ramaswamy V, Bartels U, Huang A, Laperriere N, Dirks P, Nathan PC, Greenberg M, Malkin D, Hawkins C, Bandopadhayay P, Kieran MW, Manley PE, Bouffet E, Tabori U, Clinical and treatment factors determining long-term outcomes for adult survivors of childhood low-grade glioma: a population-based study, Cancer 122 (2016) 1261–1269. [DOI] [PubMed] [Google Scholar]
- [12].Hoffman LM, Veldhuijzen van Zanten SEM, Colditz N, Baugh J, Chaney B, Hoffmann M, Lane A, Fuller C, Miles L, Hawkins C, Bartels U, Bouffet E, Goldman S, Leary S, Foreman NK, Packer R, Warren KE, Broniscer A, Kieran MW, Minturn J, Comito M, Broxson E, Shih CS, Khatua S, Chintagumpala M, Carret AS, Escorza NY, Hassall T, Ziegler DS, Gottardo N, Dholaria H, Doughman R, Benesch M, Drissi R, Nazarian J, Jabado N, Boddaert N, Varlet P, Giraud G, Castel D, Puget S, Jones C, Hulleman E, Modena P, Giagnacovo M, Antonelli M, Pietsch T, Gielen GH, Jones DTW, Sturm D, Pfister SM, Gerber NU, Grotzer MA, Pfaff E, von Bueren AO, Hargrave D, Solanki GA, Jadrijevic Cvrlje F, Kaspers GJL, Vandertop WP, Grill J, Bailey S, Biassoni V, Massimino M, Calmon R, Sanchez E, Bison B, Warmuth-Metz M, Leach J, Jones B, van Vuurden DG, Kramm CM, Fouladi M, Clinical, radiologic, pathologic, and molecular characteristics of long-term survivors of diffuse intrinsic pontine glioma (DIPG): a collaborative report from the international and European society for pediatric oncology DIPG registries, J. Clin. Oncol 36 (2018) 1963–1972, official journal of the American Society of Clinical Oncology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Ullrich NJ, Pomeroy SL, Kapur K, Manley PE, Goumnerova LC, Loddenkemper T, Incidence, risk factors, and longitudinal outcome of seizures in long-term survivors of pediatric brain tumors, Epilepsia 56 (2015) 1599–1604. [DOI] [PubMed] [Google Scholar]
- [14].Shu Q, Wong KK, Su JM, Adesina AM, Yu LT, Tsang YT, Antalffy BC, Baxter P, Perlaky L, Yang J, Dauser RC, Chintagumpala M, Blaney SM, Lau CC, Li XN, Direct orthotopic transplantation of fresh surgical specimen preserves CD133+ tumor cells in clinically relevant mouse models of medulloblastoma and glioma, Stem Cell. 26 (2008) 1414–1424. [DOI] [PubMed] [Google Scholar]
- [15].Yu L, Baxter PA, Voicu H, Gurusiddappa S, Zhao Y, Adesina A, Man TK, Shu Q, Zhang YJ, Zhao XM, Su JM, Perlaky L, Dauser R, Chintagumpala M, Lau CC, Blaney SM, Rao PH, Leung HC, Li XN, A clinically relevant orthotopic xenograft model of ependymoma that maintains the genomic signature of the primary tumor and preserves cancer stem cells in vivo, Neuro Oncol. 12 (2010) 580–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Zhao X, Liu Z, Yu L, Zhang Y, Baxter P, Voicu H, Gurusiddappa S, Luan J, Su JM, Leung HC, Li XN, Global gene expression profiling confirms the molecular fidelity of primary tumor-based orthotopic xenograft mouse models of medulloblastoma, Neuro Oncol. 14 (2012) 574–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Liu Z, Zhao X, Wang Y, Mao H, Huang Y, Kogiso M, Qi L, Baxter PA, Man TK, Adesina A, Su JM, Picard D, Ching Ho K, Huang A, Perlaky L, Lau CC, Chintagumpala M, Li XN, A patient tumor-derived orthotopic xenograft mouse model replicating the group 3 supratentorial primitive neuroectodermal tumor in children, Neuro Oncol. 16 (2014) 787–799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Baxter PA, Lin Q, Mao H, Kogiso M, Zhao X, Liu Z, Huang Y, Voicu H, Gurusiddappa S, Su JM, Adesina AM, Perlaky L, Dauser RC, Leung HC, Muraszko KM, Heth JA, Fan X, Lau CC, Man TK, Chintagumpala M, Li XN, Silencing BMI1 eliminates tumor formation of pediatric glioma CD133+ cells not by affecting known targets but by down-regulating a novel set of core genes, Acta Neuropathol. Commun 2 (2014) 160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Grasso CS, Tang Y, Truffaux N, Berlow NE, Liu L, Debily MA, Quist MJ, Davis LE, Huang EC, Woo PJ, Ponnuswami A, Chen S, Johung TB, Sun W, Kogiso M, Du Y, Qi L, Huang Y, Hutt-Cabezas M, Warren KE, Le DL, Meltzer PS, Mao H, Quezado M, van Vuurden DG, Abraham J, Fouladi M, Svalina MN, Wang N, Hawkins C, Nazarian J, Alonso MM, Raabe EH, Hulleman E, Spellman PT, Li XN, Keller C, Pal R, Grill J, Monje M, Functionally defined therapeutic targets in diffuse intrinsic pontine glioma, Nat. Med 21 (2015) 555–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Lindsay H, Huang Y, Du Y, Braun FK, Teo WY, Kogiso M, Qi L, Zhang H, Zhao S, Mao H, Lin F, Baxter P, Su JM, Terashima K, Perlaky L, Chintagumpala M, Adesina A, Lau CC, Williams Parsons D, Li XN, Preservation of KIT genotype in a novel pair of patient-derived orthotopic xenograft mouse models of metastatic pediatric CNS germinoma, J. Neuro Oncol 128 (2016) 47–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Kogiso M, Qi L, Lindsay H, Huang Y, Zhao X, Liu Z, Braun FK, Du Y, Zhang H, Bae G, Zhao S, Injac SG, Sobieski M, Brunell D, Mehta V, Tran D, Murray J, Baxter PA, Yuan XJ, Su JM, Adesina A, Perlaky L, Chintagumpala M, Parsons DW, Lau CC, Stephan CC, Lu X, Li XN, Xenotransplantation of pediatric low grade gliomas confirms the enrichment of BRAF V600E mutation and preservation of CDKN2A deletion in a novel orthotopic xenograft mouse model of progressive pleomorphic xanthoastrocytoma, Oncotarget 8 (2017) 87455–87471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Kogiso M, Qi L, Braun FK, Injac SG, Zhang L, Du Y, Zhang H, Lin FY, Zhao S, Lindsay H, Su JM, Baxter PA, Adesina AM, Liao D, Qian MG, Berg S, Muscal JA, Li XN, Concurrent inhibition of neurosphere and monolayer cells of pediatric glioblastoma by aurora A inhibitor MLN8237 predicted survival extension in PDOX models, Clin. Cane. Res 24 (2018) 2159–2170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Brabetz S, Leary SES, Grobner SN, Nakamoto MW, Seker-Cin H, Girard EJ, Cole B, Strand AD, Bloom KL, Hovestadt V, Mack NL, Pakiam F, Schwalm B, Korshunov A, Balasubramanian GP, Northcott PA, Pedro KD, Dey J, Hansen S, Ditzler S, Lichter P, Chavez L, Jones DTW, Koster J, Pfister SM, Kool M, Olson JM, A biobank of patient-derived pediatric brain tumor models, Nat. Med 24 (2018) 1752–1761. [DOI] [PubMed] [Google Scholar]
- [24].Zhao X, Zhao YJ, Lin Q, Yu L, Liu Z, Lindsay H, Kogiso M, Rao P, Li XN, Lu X, Cytogenetic landscape of paired neurospheres and traditional monolayer cultures in pediatric malignant brain tumors, Neuro Oncol. 17 (2015) 965–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Grasso CS, Tang Y, Truffaux N, Berlow NE, Liu L, Debily MA, Quist MJ, Davis LE, Huang EC, Woo PJ, Ponnuswami A, Chen S, Johung TB, Sun W, Kogiso M, Du Y, Qi L, Huang Y, Hutt-Cabezas M, Warren KE, Le Dret L, Meltzer PS, Mao H, Quezado M, van Vuurden DG, Abraham J, Fouladi M, Svalina MN, Wang N, Hawkins C, Nazarian J, Alonso MM, Raabe EH, Hulleman E, Spellman PT, Li XN, Keller C, Pal R, Grill J, Monje M, Functionally defined therapeutic targets in diffuse intrinsic pontine glioma, Nat. Med 21 (2015) 555–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Rokita JL, Rathi KS, Cardenas MF, Upton KA, Jayaseelan J, Cross KL, Pfeil J, Egolf LE, Way GP, Farrel A, Kendsersky NM, Patel K, Gaonkar KS, Modi A, Berko ER, Lopez G, Vaksman Z, Mayoh C, Nance J, McCoy K, Haber M, Evans K, McCalmont H, Bendak K, Bohm JW, Marshall GM, Tyrrell V, Kalletla K, Braun FK, Qi L, Du Y, Zhang H, Lindsay HB, Zhao S, Shu J, Baxter P, Morton C, Kurmashev D, Zheng S, Chen Y, Bowen J, Bryan AC, Leraas KM, Coppens SE, Doddapaneni H, Momin Z, Zhang W, Sacks GI, Hart LS, Krytska K, Mosse YP, Gatto GJ, Sanchez Y, Greene CS, Diskin SJ, Vaske OM, Haussler D, Gastier-Foster JM, Kolb EA, Gorlick R, Li XN, Reynolds CP, Kurmasheva RT, Houghton PJ, Smith MA, Lock RB, Raman P, Wheeler DA, Maris JM, Genomic profiling of childhood tumor patient-derived xenograft models to enable rational clinical trial design, Cell Rep. 29 (2019) 1675–1689, e1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Sin-Chan P, Mumal I, Suwal T, Ho B, Fan X, Singh I, Du Y, Lu M, Patel N, Torchia J, Popovski D, Fouladi M, Guilhamon P, Hansford JR, Leary S, Hoffman LM, Mulcahy Levy JM, Lassaletta A, Solano-Paez P, Rivas E, Reddy A, Gillespie GY, Gupta N, Van Meter TE, Nakamura H, Wong TT, Ra YS, Kim SK, Massimi L, Grundy RG, Fangusaro J, Johnston D, Chan J, Lafay-Cousin L, Hwang EI, Wang Y, Catchpoole D, Michaud J, Ellezam B, Ramanujachar R, Lindsay H, Taylor MD, Hawkins CE, Bouffet E, Jabado N, Singh SK, Kleinman CL, Barsyte-Lovejoy D, Li XN, Dirks PB, Lin CY, Mack SC, Rich JN, Huang A, A C19MC-LIN28A-MYCN oncogenic circuit driven by hijacked super-enhancers is a distinct therapeutic vulnerability in ETMRs: a lethal brain tumor, Canc. Cell 36 (2019) 51–67 e57. [DOI] [PubMed] [Google Scholar]
- [28].Yao M, Ventura PB, Jiang Y, Rodriguez FJ, Wang L, Perry JSA, Yang Y, Wahl K, Crittenden RB, Bennett ML, Qi L, Gong CC, Li XN, Barres BA, Bender TP, Ravichandran KS, Janes KA, Eberhart CG, Zong H, Astrocytic trans-differentiation completes a multicellular paracrine feedback loop required for medulloblastoma tumor growth, Cell 180 (2020) 502–520 e519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Baxter PA, Su JM, Onar-Thomas A, Billups CA, Li XN, Poussaint TY, Smith ER, Thompson PA, Adekunsle A, Ansell P, Giranda VL, Paulino AC, Kilburn L, Quaddoumi I, Broniscer A, Blaney SM, Dunkel IJ, Fouladi M, A phase I/II study of veliparib (ABT-888) with radiation and temozolomide in newly diagnosed diffuse pontine glioma: a pediatric brain tumor consortium study, Neuro Oncol. 22 (6) (2020) 875–885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Hovestadt V, Smith KS, Bihannic L, Filbin MG, Shaw ML, Baumgartner A, DeWitt JC, Groves A, Mayr L, Weisman HR, Richman AR, Shore ME, Goumnerova L, Rosencrance C, Carter RA, Phoenix TN, Hadley JL, Tong Y, Houston J, Ashmun RA, DeCuypere M, Sharma T, Flasch D, Silkov A, Ligon KL, Pomeroy SL, Rivera MN, Rozenblatt-Rosen O, Rusert JM, Wechsler-Reya RJ, Li XN, Peyrl A, Gojo J, Kirchhofer D, Lotsch D, Czech T, Dorfer C, Haberler C, Geyeregger R, Halfmann A, Gawad C, Easton J, Pfister SM, Regev A, Gajjar A, Orr BA, Slavc I, Robinson GW, Bernstein BE, Suva ML, Northcott PA, Resolving medulloblastoma cellular architecture by single-cell genomics, Nature 572 (2019) 74–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Dobson THW, Tao RH, Swaminathan J, Maegawa S, Shaik S, Bravo-Alegria J, Sharma A, Kennis B, Yang Y, Callegari K, Haltom AR, Taylor P, Kogiso M, Qi L, Khatua S, Goldman S, Lulla RR, Fangusaro J, MacDonald TJ, Li XN, Hawkins C, Rajaram V, Gopalakrishnan V, Transcriptional repressor REST drives lineage stage-specific chromatin compaction at Ptch 1 and increases AKT activation in a mouse model of medulloblastoma, Sci. Signal 12 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Lambo S, Grobner SN, Rausch T, Waszak SM, Schmidt C, Gorthi A, Romero JC, Mauermann M, Brabetz S, Krausert S, Buchhalter I, Roster J, Zwijnenburg DA, Sill M, Hubner JM, Mack N, Schwalm B, Ryzhova M, Hovestadt V, Papillon-Cavanagh S, Chan JA, Landgraf P, Ho B, Milde T, Witt O, Ecker J, Sahm F, Sumerauer D, Ellison DW, Orr BA, Darabi A, Haberler C, Figarella-Branger D, Wesseling P, Schittenhelm J, Remke M, Taylor MD, Gil-da-Costa MJ, Lastowska M, Grajkowska W, Hasselblatt M, Hauser P, Pietsch T, Uro-Coste E, Bourdeaut F, Masliah-Planchon J, Rigau V, Alexandrescu S, Wolf S, Li XN, Schuller U, Snuderl M, Karajannis MA, Giangaspero F, Jabado N, von Deimling A, Jones DTW, Korbel JO, von Hoff K, Lichter P, Huang A, Bishop AJR, Pfister SM, Korshunov A, Kool M, The molecular landscape of ETMR at diagnosis and relapse, Nature 576 (2019) 274–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Yang W, Warrington NM, Taylor SJ, Whitmire P, Carrasco E, Singleton KW, Wu N, Lathia JD, Berens ME, Kim AH, Barnholtz-Sloan JS, Swanson KR, Luo J, Rubin JB, Sex differences in GBM revealed by analysis of patient imaging, transcriptome, and survival data, Sci. Transl. Med 11 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Bayik D, Zhou Y, Park C, Hong C, Vail D, Silver DJ, Lauko A, Roversi G, Watson DC, Lo A, Alban TJ, McGraw M, Sorensen M, Grabowski MM, Otvos B, Vogelbaum MA, Horbinski C, Kristensen BW, Khalil AM, Hwang TH, Ahluwalia MS, Cheng F, Lathia JD, Myeloid-derived suppressor cell subsets drive glioblastoma growth in a sex-specific manner, Canc. Discov 10 (2020) 1210–1225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Ostrom QT, Kinnersley B, Wrensch MR, Eckel-Passow JE, Armstrong G, Rice T, Chen Y, Wiencke JK, McCoy LS, Hansen HM, Amos CI, Bernstein JL, Claus EB, Il’yasova D, Johansen C, Lachance DH, Lai RK, Merrell RT, Olson SH, Sadetzki S, Schildkraut JM, Shete S, Rubin JB, Lathia JD, Berens ME, Andersson U, Rajaraman P, Chanock SJ, Linet MS, Wang Z, Yeager M, Houlston RS, Jenkins RB, Melin B, Bondy ML, Barnholtz-Sloan JS, Sex-specific glioma genome-wide association study identifies new risk locus at 3p21.31 in females, and finds sex-differences in risk at 8q24.21, Sci. Rep 8 (2018) 7352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Ding W, Bishop ME, Pearce MG, Davis KJ, White GA, Lyn-Cook LE, Manjanatha MG, Sex-specific dose-response analysis of genotoxicity in cyproterone acetate-treated F344 rats, Mutat. Res. Genet. Toxicol. Environ. Mutagen 774 (2014) 1–7. [DOI] [PubMed] [Google Scholar]
- [37].Houghton PJ, Lock R, Carol H, Morton CL, Gorlick R, Anders Kolb E, Keir ST, Reynolds CP, Kang MH, Maris JM, Billups CA, Zhang MX, Madden SL, Teicher BA, Smith MA, Testing of the topoisomerase 1 inhibitor Genz-644282 by the pediatric preclinical testing program, Pediatr. Blood Canc 58 (2012) 200–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Kolb EA, Houghton PJ, Kurmasheva RT, Mosse YP, Maris JM, Erickson SW, Guo Y, Teicher BA, Smith MA, Gorlick R, Preclinical evaluation of the combination of AZD1775 and irinotecan against selected pediatric solid tumors: a Pediatric Preclinical Testing Consortium report, Pediatr. Blood Canc 67 (2020), e28098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Kurmasheva RT, Bandyopadhyay A, Favours E, Pozo VD, Ghilu S, Phelps DA, McGeehan GM, Erickson SW, Smith MA, Houghton PJ, Evaluation of VTP-50469, a menin-MLL1 inhibitor, against Ewing sarcoma xenograft models by the pediatric preclinical testing consortium, Pediatr. Blood Canc 67 (2020), e28284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Robles AJ, Kurmasheva RT, Bandyopadhyay A, Phelps DA, Erickson SW, Lai Z, Kurmashev D, Chen Y, Smith MA, Houghton PJ, Evaluation of eribulin combined with irinotecan for treatment of pediatric cancer xenografts, Clin. Canc. Res 26 (2020) 3012–3023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Turaga SM, Silver DJ, Bayik D, Paouri E, Peng S, Lauko A, Alban TJ, Borjini N, Stanko S, Naik U, Keri RA, Connor JR, Barnholtz-Sloan JS, Rubin JB, Berens M, Davalos D, Lathia JD, JAM-A functions as a female microglial tumor suppressor in glioblastoma, Neuro Oncol. (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Clayton JA, Collins FS, Policy: NIH to balance sex in cell and animal studies, Nature 509 (2014) 282–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Liu Z, Zhao X, Mao H, Baxter PA, Huang Y, Yu L, Wadhwa L, Su JM, Adesina A, Perlaky L, Hurwitz M, Idamakanti N, Police SR, Hallenbeck PL, Hurwitz RL, Lau CC, Chintagumpala M, Blaney SM, Li XN, Intravenous injection of oncolytic picornavirus SVV-001 prolongs animal survival in a panel of primary tumor-based orthotopic xenograft mouse models of pediatric glioma, Neuro Oncol. 15 (2013) 1173–1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Shu Q, Wong KK, Su JM, Adesina AM, Yu LT, Tsang YT, Antalffy BC, Baxter P, Perlaky L, Yang J, Dauser RC, Chintagumpala M, Blaney SM, Lau CC, Li XN, Direct orthotopic transplantation of fresh surgical specimen preserves CD133+ tumor cells in clinically relevant mouse models of medulloblastoma and glioma, Stem Cell. 26 (2008) 1414–1424. [DOI] [PubMed] [Google Scholar]
- [45].Huang L, Garrett Injac S, Cui K, Braun F, Lin Q, Du Y, Zhang H, Kogiso M, Lindsay H, Zhao S, Baxter P, Adekunle A, Man TK, Zhao H, Li XN, Lau CC, Wong STC, Systems biology-based drug repositioning identifies digoxin as a potential therapy for groups 3 and 4 medulloblastoma, Sci. Transl. Med 10 (2018), eaat0150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Yu L, Baxter PA, Zhao X, Liu Z, Wadhwa L, Zhang Y, Su JM, Tan X, Yang J, Adesina A, Perlaky L, Hurwitz M, Idamakanti N, Police SR, Hallenbeck PL, Blaney SM, Chintagumpala M, Hurwitz RL, Li XN, A single intravenous injection of oncolytic picornavirus SVV-001 eliminates medulloblastomas in primary tumor-based orthotopic xenograft mouse models, Neuro Oncol. 13 (2010) 14–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Dong H, Liu S, Zhang X, Chen S, Kang L, Chen Y, Ma S, Fu X, Liu Y, Zhang H, Zou B, EED inhibition suppresses cancers by modulating the immune response, Canc. Res 79 (21) (2019) 5587–5596. [DOI] [PubMed] [Google Scholar]
- [48].Chalishazar MD, Wait SJ, Huang F, Ireland AS, Mukhopadhyay A, Lee Y, Schuman SS, Guthrie MR, Berrett KC, Vahrenkamp JM, Hu Z, Kudla M, Modzelewska K, Wang G, Ingolia NT, Gertz J, Lum DH, Cosulich SC, Bomalaski JS, DeBerardinis RJ, Oliver TG, MYC-driven small-cell lung cancer is metabolically distinct and vulnerable to arginine depletion, Clin. Canc. Res 25 (2019) 5107–5121, an official journal of the American Association for Cancer Research. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Kita K, Fukuda K, Takahashi H, Tanimoto A, Nishiyama A, Arai S, Takeuchi S, Yamashita K, Ohtsubo K, Otani S, Yanagimura N, Suzuki C, Ikeda H, Tamura M, Matsumoto I, Yano S, Patient-derived xenograft models of non-small cell lung cancer for evaluating targeted drug sensitivity and resistance, Canc. Sci 110 (2019) 3215–3224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Truong D, Fiorelli R, Barrientos ES, Melendez EL, Sanai N, Mehta S, Nikkhah M, A three-dimensional (3D) organotypic microfluidic model for glioma stem cells - vascular interactions, Biomaterials 198 (2019) 63–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Burns MB, Montassier E, Abrahante J, Priya S, Niccum DE, Khoruts A, Starr TK, Knights D, Blekhman R, Colorectal cancer mutational profiles correlate with defined microbial communities in the tumor microenvironment, PLoS Genet. 14 (2018), e1007376. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.