Skip to main content

Failure of a patient-derived xenograft for brain tumor model prepared by implantation of tissue fragments

Abstract

Background

With the continuing development of new anti-cancer drugs comes a need for preclinical experimental models capable of predicting the clinical activity of these novel agents in cancer patients. However existing models have a limited ability to recapitulate the clinical characteristics and associated drug sensitivity of tumors. Among the more promising approaches for improving preclinical models is direct implantation of patient-derived tumor tissue into immunocompromised mice, such as athymic nude or non-obese diabetic/severe combined immunodeficient (NOD/SCID) mice. In the current study, we attempted to develop patient-derived xenograft (PDX) models using tissue fragments from surgical samples of brain tumors.

Methods

In this approach, tiny tissue fragments of tumors were biopsied from eight brain tumor patients—seven glioblastoma patients and one primitive neuroectodermal tumor patient. Two administration methods—a cut-down syringe and a pipette—were used to implant tissue fragments from each patient into the brains of athymic nude mice.

Results

In contrast to previous reports, and contrary to our expectations, we found that none of these fragments from brain tumor biopsies resulted in the successful establishment of xenograft tumors.

Conclusions

These results suggest that fragments of surgical specimens from brain tumor patients are unsuitable for implementation of brain tumor PDX models, and instead recommend other in vivo testing platforms for brain tumors, such as cell-based brain tumor models.

Background

The multiple genetic lesions and complex signaling cascades associated with the etiology and progression of cancer contribute to difficulties in understanding the biology of this disease [1]. Of the various kinds of cancers, brain tumors, especially glioblastoma (GBM), are among the most lethal [2]. Thus not surprisingly, considerable research effort has been devoted to increasing the efficacy of new treatments for GBM [36]. Although these efforts have been ongoing for decades, preclinical tumor models suitable for evaluating the effects of anti-cancer therapies are lacking [79].

A number of animal models of primary brain tumors have been developed to understand brain tumors, including chemically induced models, genetically engineered mouse models, and tumor xenograft models [8]. Among these animal models, tumor xenografts derived from human GBM cell lines have traditionally been used as a preclinical tool to understand cancer biology and therapeutic efficacy. However, GBM cell line xenografts have a number of shortcomings for preclinical use, including (i) the failure of tumor xenografts to mirror patient responses [10, 11] because they do not accurately reflect the clinical characteristics of the patient tumor, (ii) differences in pharmacokinetics between animal models and as humans [12], and (iii) the loss of biological properties of cancer cell lines during the process of establishing them [13, 14].

To overcome the shortcomings of previous tumor models and to preserve the oncological heterogeneity observed in patients, researchers are continuing to focus on new animal testing platforms for cancer. Several recent studies have reported innovative preclinical animal models generated by transplanting sectioned patient-derived tumor tissue fragments, rather than cell lines, into immunocompromised mice, such as athymic nude or non-obese diabetic/severe combined immunodeficient (NOD/SCID) mice [7, 1518], a concept generally referred to in the literature as patient-derived xenograft (PDX) [15, 18]. Maintaining the histological characteristics and primary architecture of tumors is one of the major advantages of the PDX model [18, 19]. In addition, the propagation of tumors in successive generations of mouse hosts enables PDX cells to avoid the stressful conditions that possibly occur during cell culture [20]. These advantages make PDXs biologically stable and enable them to maintain the molecular characteristics of the primary tumors from which they were derived [20, 21]. Because of these strengths, PDXs are useful not only for the preclinical testing of drugs, but also to verify molecular changes and signaling pathways in oncology [1].

A recent series of studies in the brain tumor field has made meaningful progress in animal testing platforms using patient-derived tumorspheres [10, 2224] and enzymatically dissociated cells [25]. However, because these previous studies used tumor cells, the resulting animal models may not satisfy the classic definition of a PDX [1, 15, 16, 18]. Making PDXs more similar to the originating GBM patient tumors may instead require injection of patient-derived tumor tissue fragments. In this context, one report concluded that PDXs formed by implanting GBM tissue fragments were similar to the original tumors of GBM patients [26]. In the present study, we attempted to develop PDX models using tissue fragments from surgical samples of brain tumors using two different direct injection methods: a cut-down syringe and a pipette.

Methods

Patient population

Eight patients with primary brain tumors, including seven glioblastoma (GBM) patients and one primitive neuroectodermal tumor (PNET) patient, treated at our institution between June 2013 and October 2013, were included in this study (Table 1). All patients were histologically diagnosed and graded by neuro-pathologists according to 2007 World Health Organization (WHO) classification criteria [27]. O-6-methylguanine-DNA methyltransferase (MGMT) promotor methylation and isocitrate dehydrogenase (IDH)-1 mutations were assessed by polymerase chain reaction (PCR) and immunohistochemistry (IHC). Epidermal growth factor receptor (EGFR) amplification and loss of heterozygosity (LOH) at chromosomes 1p and 19q were evaluated by fluorescent in situ hybridization (FISH). P53 and ki-67 were examined by IHC. All patients provided written informed consent, and the study was approved by the Institutional Review Boards of our institution.

Table 1 Patient characteristics

From fresh tumor specimen to small tissue fragments

Specimens from GBM patients were freshly obtained from the operating room, placed in sterile centrifuge tubes (SPL Life Sciences Co., Ltd, Seoul, Korea) in ice, and then chopped with a sterile stainless steel surgical blade (Paragon, Sheffield, UK) and suspended in phosphate-buffered saline (PBS; Mediatech, Manassas, VA, USA) within 30 min of being sent from the surgical room. Fragmented specimens, approximately 3 mm3 in size (Aliquots of 3 μl of suspended tumor fragments were prepared for injection), were prepared for each sample [15, 26].

Animals

Male athymic nude mice (Central Lab Animal Inc., Seoul, Korea), aged 4–8 weeks, were used in this study. Mice were housed in micro-isolator cages under sterile conditions and were observed for at least 1 week before study initiation to ensure proper health. Lighting, temperature, and humidity were centrally controlled. All experimental procedures were approved by the Institutional Animal Care and Use Committee of Yonsei University College of Medicine.

Orthotopic xenograft of patient-derived brain tumor tissue fragments

Brain tumor tissue fragments from each patient (n = 8) were directly injected into mice (n = 3/group) using two methods: a 21 gauge, cut-down syringe (Korea Vaccine Co., Ltd., Gyeonggi-do, Korea), and a pipette (Gilson, Middleton, WI, USA) (Fig. 1). Mice were first anesthetized with a solution of Zoletil (30 mg/kg; Virbac Korea, Seoul, Korea) and xylazine (10 mg/kg; Bayer Korea, Seoul, Korea), delivered intraperitoneally. Then, a hole was drilled in the right side of the skull at the implantation position (2.5 mm lateral from sagittal suture, 1 mm anterior from coronal suture) using a small, hand-controlled twist drill (Plastics One, Roanoke, VA, USA). Thereafter, patient-derived brain tumor tissue fragments (~3 mm3) were implanted to a depth of 4.5 mm in the right frontal lobe of nude mice via an inserted cut-down syringe or pipette.

Fig. 1
figure 1

Methods for developing PDX mouse models. Athymic mice received patient-derived surgical brain tumor tissue fragments (~3 mm3) via two different injection methods: 21 gauge cut-down syringe and pipette

Results

Patient-derived xenograft (PDX) models were prepared for eight brain tumor patients (Table 1) by implanting tiny tissue fragments biopsied from each patient into the brains of athymic mice (n = 3/group) using the two injection methods—cut-down syringe and pipette—described in “Methods” section. A dataset was obtained for each PDX that included a pathological assessment and magnetic resonance imaging (MRI; T1 axial enhancement) of the corresponding donor, the injection method, the lifetime of the resulting PDX, and images of hematoxylin and eosin (H&E)-stained histological sections of PDX brain tissue (Fig. 2).

Fig. 2
figure 2

Example of a PDX data set. Each data set included the pathology of the donor patient, an MRI (T1 axial enhancement) image of the donor patient, PDX number, injection method, lifetime of the PDX, and an H&E-stained histological section of PDX brain tissue. This data set corresponds to that for tumor tissue fragments from a glioblastoma patient with a T1 contrast-enhanced lesion in the right temporoparietal lobe implanted into PDX mouse #22 through a pipette. No xenograft patient-derived tumor tissue fragments were detected in H&E-stained histological sections of brain tissue from this mouse, as confirmed by Neuro-pathologists

The neuro-pathologists examined each PDX brain. Tissues were fixed in 4 % phosphate-buffered paraformaldehyde for 24 h, embedded in paraffin, sectioned, and stained with H&E to examine whether the xenografted tumor tissue fragments had been successfully established. A total of 17 PDXs were prepared by injecting tissue fragments via a cut-down syringe and 8 PDXs were prepared by injecting with a pipette. Among these PDXs, four in the cut-down syringe group and one in the pipette group died because of surgical complications. Apart from these dead PDXs, no PDX-bearing patient-derived brain tumor was detected, yielding an overall establishment rate of 0 %. The neuro-pathologists verified that the PDX brain tissue sections showed inflammation and was thus unlikely to be necrotic (Table 2).

Table 2 Tissue fragment implantation results

Discussion

Previous studies have reported testing platforms that attempt to recapitulate the properties of patients’ tumors using tumorspheres [10] or dissociated cells [25]. One additional previous study also suggested that a xenograft formed from glioblastoma (GBM) fragments inserted into the brain was similar to that of the originating GBM tumor [26]. On the basis of these results, we hypothesized that brain tumor patient-derived xenograft (PDX) models could be successfully established through direct implantation of tumor tissue fragments. To create these models, we inserted tiny tumor tissue fragments (~3 mm3), biopsied from brain tumor patients, into the brains of athymic mice using two injection methods: a cut-down syringe and a pipette. Among these PDXs, ~3 % died due to surgical complication. Inflammation was the only fatal surgical complication observed, and mice subject to it died an average of 5 days post-op. The post-op survival of all other mice was longer than 6 months. An analysis of each PDX brain tissue section to determine whether the xenografted tumor tissue fragments successfully became established unexpectedly revealed a tumor-establishment rate for xenografted brain tumor tissue fragments of 0 %. Thus, contrary to our hypothesis, we could not develop brain tumor PDXs from tumor tissue fragments, considered an ideal method for immortalization of a patient’s cancer.

Considerable effort has been devoted to identifying potential animal hosts for xenotransplantation of human cells for use in the development of animal models for human diseases. As a consequence, a number of immunocompromised mouse models, including athymic nude mice and severe combined immunodeficient (SCID) mice, have been developed. Motivated by previous research [26], we used T cell deficient athymic nude mice as xenografts animal subjects. However, these mice still have functional B cells, which could cause failure of engraftment. Accordingly, a change in animal model to SCID mice, which are defective in functional T and B cell, could facilitate engraftment. One report has also suggested NOD/SCID/γ nullc mice, a modified SCID mouse with multiple immunological dysfunctions, as an animal recipient for improving the engraftment success of xenografts [28].

From the standpoint of transplanting tumors most like the original, our study is similar to a previous report [26]. Fei et al. injected 2 mm3 of tumor tissue fragments per mouse in a total maximum volume of 20 μl injected cell suspension [26, 29, 30]. Here, we implanted 3 mm3 of suspended tumor tissue fragments per mouse, in an effort to increase the amount of tissue fragments implanted. In both studies, the tumor tissue fragments were injected into mice within 5 min (from anesthesia to closure of skull hole). In our study, it took about 15 min to transfer the samples from the operating room to the laboratory, and 15 min to chop the samples, but no specific sample movement time was reported in the previous study. We used a cut-down syringe and pipette to insert tumor tissue fragments, whereas the previous study used a trocar for injection. This single known difference in methodology may have caused differences in the total volume of injected tissue fragments, resulting in the implantation of a relatively small number tumor cells in our study. It might also have produced differences in injection pressure or speed that ultimately proved detrimental.

In this study, no PDX-bearing patient-derived brain tumor was detected. It is possible that this might reflect a loss of cell viability, perhaps due to our brief placement of fresh patient-derived brain tumor tissue fragments in ice, or the suspension of brain tumor tissue fragments in PBS (which was required because otherwise the chopped tissue fragments were too sticky). We do not believe, however, that there was any issue with loss of cell viability. We used PBS instead of medium to exclude any effects from factors found in the medium. Specimens were chopped with a surgical blade and suspended in PBS within 30 min being sent from the surgical room, and were then injected within 30 min of the chopping step. Moreover, we performed cell culture using specimens placed in ice and suspended with PBS, as described in our protocol, and successfully obtained cultured tumor cells (Additional file 1: Figure S1). Additional method for cell isolation was described in Additional file 2. Finally, previous reports have described the successful isolation of viable cells using the same method [23, 24]. Thus, we believe that our patient-derived brain tumor tissue fragments maintained their viability during the processing steps.

The 0 % tumor-establishment rate for xenografted brain tumor tissue fragments may be attributable to these differences. However, despite the differences between our method and the trocar injection system, the basic concept of both methods is similar. Thus, notwithstanding a previous report of GBM PDX models prepared using athymic mice, our results suggest that tissue fragments of surgical brain tumor specimens from patients may be unsuitable for implementation of brain tumor PDX models. Previous reports described the successful development of brain tumor xenograft models using the same methods applied in the present work [23, 24, 31]. Though the methods in this study were proved through previous reports, alteration of some method conditions such as alternative tissue preparation, injection techniques, and selection of immunodeficient animals can prove more successful in establishing brain tumor PDX models.

Abbreviations

PDX:

patient-derived xenograft

GBM:

glioblastoma

NOD-SCID:

non-obese diabetic/severe combined immunodeficient

PNET:

primitive neuroectodermal tumor

References

  1. Malaney P, Nicosia SV, Dave V. One mouse, one patient paradigm: new avatars of personalized cancer therapy. Cancer Lett. 2014;344:1–12.

    Article  CAS  PubMed  Google Scholar 

  2. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–96.

    Article  CAS  PubMed  Google Scholar 

  3. Zhang S, Han L, Wei J, Shi Z, Pu P, Zhang J, et al. Combination treatment with doxorubicin and microRNA-21 inhibitor synergistically augments anticancer activity through upregulation of tumor suppressing genes. Int J Oncol. 2015. doi:10.3892/ijo.2015.2841.

    Google Scholar 

  4. Jia PF, Gu WT, Zhang WF, Li F. Treatment of recurrent malignant gliomas with 13-cis-retinoic acid naphthalene triazole. Neurol Sci. 2015. doi:10.1007/s10072-014-2025-9.

    Google Scholar 

  5. Marrero L, Wyczechowska D, Musto AE, Wilk A, Vashistha H, Zapata A, et al. Therapeutic efficacy of aldoxorubicin in an intracranial xenograft mouse model of human glioblastoma. Neoplasia. 2014;16:874–82.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Wohlfart S, Khalansky AS, Gelperina S, Maksimenko O, Bernreuther C, Glatzel M, et al. Efficient chemotherapy of rat glioblastoma using doxorubicin-loaded PLGA nanoparticles with different stabilizers. PLoS One. 2011;6:e19121.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wong NC, Bhadri VA, Maksimovic J, Parkinson-Bates M, Ng J, Craig JM, et al. Stability of gene expression and epigenetic profiles highlights the utility of patient-derived paediatric acute lymphoblastic leukaemia xenografts for investigating molecular mechanisms of drug resistance. BMC Genom. 2014;15:416.

    Article  Google Scholar 

  8. Huszthy PC, Daphu I, Niclou SP, Stieber D, Nigro JM, Sakariassen PO, et al. In vivo models of primary brain tumors: pitfalls and perspectives. Neuro Oncol. 2012;14:979–93.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Qazi M, Mann A, van Ommeren R, Venugopal C, McFarlane N, Vora P, et al. Generation of murine xenograft models of brain tumors from primary human tissue for in vivo analysis of the brain tumor-initiating cell. Methods Mol Biol. 2014;1210:37–49.

    Article  CAS  PubMed  Google Scholar 

  10. Wang J, Miletic H, Sakariassen PO, Huszthy PC, Jacobsen H, Brekka N, et al. A reproducible brain tumour model established from human glioblastoma biopsies. BMC Cancer. 2009;9:465.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Bjerkvig R, Tonnesen A, Laerum OD, Backlund EO. Multicellular tumor spheroids from human gliomas maintained in organ culture. J Neurosurg. 1990;72:463–75.

    Article  CAS  PubMed  Google Scholar 

  12. Peterson JK, Houghton PJ. Integrating pharmacology and in vivo cancer models in preclinical and clinical drug development. Eur J Cancer. 2004;40:837–44.

    Article  CAS  PubMed  Google Scholar 

  13. Sausville EA, Burger AM. Contributions of human tumor xenografts to anticancer drug development. Cancer Res. 2006;66:3351–4 (discussion 4).

    Article  CAS  PubMed  Google Scholar 

  14. Martens T, Laabs Y, Gunther HS, Kemming D, Zhu Z, Witte L, et al. Inhibition of glioblastoma growth in a highly invasive nude mouse model can be achieved by targeting epidermal growth factor receptor but not vascular endothelial growth factor receptor-2. Clin Cancer Res. 2008;14:5447–58.

    Article  CAS  PubMed  Google Scholar 

  15. Rubio-Viqueira B, Jimeno A, Cusatis G, Zhang X, Iacobuzio-Donahue C, Karikari C, et al. An in vivo platform for translational drug development in pancreatic cancer. Clin Cancer Res. 2006;12:4652–61.

    Article  CAS  PubMed  Google Scholar 

  16. Jin K, Teng L, Shen Y, He K, Xu Z, Li G. Patient-derived human tumour tissue xenografts in immunodeficient mice: a systematic review. Clin Transl Oncol. 2010;12:473–80.

    Article  PubMed  Google Scholar 

  17. Reyal F, Guyader C, Decraene C, Lucchesi C, Auger N, Assayag F, et al. Molecular profiling of patient-derived breast cancer xenografts. Breast Cancer Res. 2012;14:R11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Hidalgo M, Amant F, Biankin AV, Budinska E, Byrne AT, Caldas C, et al. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov. 2014;4:998–1013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sanz L, Cuesta AM, Salas C, Corbacho C, Bellas C, Alvarez-Vallina L. Differential transplantability of human endothelial cells in colorectal cancer and renal cell carcinoma primary xenografts. Lab Invest. 2009;89:91–7.

    Article  CAS  PubMed  Google Scholar 

  20. Daniel VC, Marchionni L, Hierman JS, Rhodes JT, Devereux WL, Rudin CM, et al. A primary xenograft model of small-cell lung cancer reveals irreversible changes in gene expression imposed by culture in vitro. Cancer Res. 2009;69:3364–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fichtner I, Rolff J, Soong R, Hoffmann J, Hammer S, Sommer A, et al. Establishment of patient-derived non-small cell lung cancer xenografts as models for the identification of predictive biomarkers. Clin Cancer Res. 2008;14:6456–68.

    Article  CAS  PubMed  Google Scholar 

  22. Kang SG, Cheong JH, Huh YM, Kim EH, Kim SH, Chang JH. Potential use of glioblastoma tumorsphere: clinical credentialing. Arch Pharm Res. 2015;38:402–7.

    Article  CAS  PubMed  Google Scholar 

  23. Kong BH, Park NR, Shim JK, Kim BK, Shin HJ, Lee JH, et al. Isolation of glioma cancer stem cells in relation to histological grades in glioma specimens. Childs Nerv Syst. 2013;29:217–29.

    Article  PubMed  Google Scholar 

  24. Kwak J, Shin HJ, Kim SH, Shim JK, Lee JH, Huh YM, et al. Isolation of tumor spheres and mesenchymal stem-like cells from a single primitive neuroectodermal tumor specimen. Childs Nerv Syst. 2013;29:2229–39.

    Article  PubMed  Google Scholar 

  25. Joo KM, Kim J, Jin J, Kim M, Seol HJ, Muradov J, et al. Patient-specific orthotopic glioblastoma xenograft models recapitulate the histopathology and biology of human glioblastomas in situ. Cell Rep. 2013;3:260–73.

    Article  CAS  PubMed  Google Scholar 

  26. Fei XF, Zhang QB, Dong J, Diao Y, Wang ZM, Li RJ, et al. Development of clinically relevant orthotopic xenograft mouse model of metastatic lung cancer and glioblastoma through surgical tumor tissues injection with trocar. J Exp Clin Cancer Res. 2010;29:84.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114:97–109.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Ito M, Hiramatsu H, Kobayashi K, Suzue K, Kawahata M, Hioki K, et al. NOD/SCID/gamma(c)(null) mouse: an excellent recipient mouse model for engraftment of human cells. Blood. 2002;100:3175–82.

    Article  CAS  PubMed  Google Scholar 

  29. Engebraaten O, Hjortland GO, Hirschberg H, Fodstad O. Growth of precultured human glioma specimens in nude rat brain. J Neurosurg. 1999;90:125–32.

    Article  CAS  PubMed  Google Scholar 

  30. Yamada S, Khankaldyyan V, Bu X, Suzuki A, Gonzalez-Gomez I, Takahashi K, et al. A method to accurately inject tumor cells into the caudate/putamen nuclei of the mouse brain. Tokai J Exp Clin Med. 2004;29:167–73.

    PubMed  Google Scholar 

  31. Shin GY, Shim JK, Lee JH, Shin HJ, Lee SJ, Huh YM, et al. Changes in the biological characteristics of glioma cancer stem cells after serial in vivo subtransplantation. Childs Nerv Syst. 2013;29:55–64.

    Article  PubMed  Google Scholar 

Download references

Authors’ contributions

SGK provided the concept design for the studies. KMK, JKS, and JHC interpreted the data and wrote this paper. JHL and JKS performed animal experiments. SHK and JC provided expert pathological analysis, clinical insight and pathological study in animal experiments. JP, EHK, SHK and YMH provided expert clinical insight. SJL, JHC and SGK conceived and supervised the project. All authors read and approved the final manuscript.

Acknowledgements

This study was supported by Grants from the Korean Health Technology R&D Project, Ministry of Health & Welfare, Republic of Korea (HI14C0042), the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (NRF-2013R1A1A2006427) and a faculty research grant of Yonsei University College of Medicine for 2013 (6-2013-0035).

Competing interests

The authors declare that they have no competing interests.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Seok-Gu Kang.

Additional information

Kyung-Min Kim, Jin-Kyoung Shim and Jong Hee Chang contributed equally to this manuscript

Additional files

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, KM., Shim, JK., Chang, J.H. et al. Failure of a patient-derived xenograft for brain tumor model prepared by implantation of tissue fragments. Cancer Cell Int 16, 43 (2016). https://doi.org/10.1186/s12935-016-0319-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-016-0319-0

Keywords