Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


For many tumours there is a lack of randomised data from which we can guide systemic treatments. Although gene expression profiling along with proteomics has led to advances in diagnosis, classification and prognosis, our ability to target many cancers has been further limited due to a lack of therapeutic options. The use of patient-derived xenograft (PDX) models in the setting of a rare malignancy is discussed here by Kamili et al, with the successful establishment of new model systems.

Free full text 


Logo of brjcancerBJC HomepageBJC Advance online publicationBJC Current IssueSubmitting an article to BJCWeb feeds
Br J Cancer. 2020 Mar 3; 122(5): 601–602.
Published online 2020 Jan 10. https://doi.org/10.1038/s41416-019-0678-0
PMCID: PMC7054515
PMID: 31919403

Patient-derived xenograft models—the future of personalised cancer treatment

Associated Data

Data Availability Statement

Abstract

For many tumours there is a lack of randomised data from which we can guide systemic treatments. Although gene expression profiling along with proteomics has led to advances in diagnosis, classification and prognosis, our ability to target many cancers has been further limited due to a lack of therapeutic options. The use of patient-derived xenograft (PDX) models in the setting of a rare malignancy is discussed here by Kamili et al, with the successful establishment of new model systems.

Main

There is almost always a discrepancy between preclinical efficacy in trials and actual clinical outcomes. This generates a demand for improving preclinical modelling. The rapidly evolving field of targeted personalised therapy is the future of oncological practice and sometimes this cannot be evaluated through traditional research methodology, such as randomised control trials (RCTs).1 Biomarker-driven therapy has become integral to treatment of cancer patients, leading to the introduction of novel trial designs with populations of biomarker-identified patient groups.2 A review of predictive and prognostic tumour biomarkers advocated that reliance on clinical judgement and expertise is vital in developing personalised cancer medicine, rather than utilisation of published clinical data.3

Despite the variability and heterogeneity of cancer types, most treatments remain ‘generic’ and usually involve chemotherapy as the mainstay.4 Chemotherapy has often been shown to be only minimally beneficial to overall survival, and is often ineffective with intolerable side effects—though one could argue that this is the case with all treatments. The hidden costs of managing chemotherapy toxicities, with repeated admissions and discussions about side effects, are likely to be substantial. Targeted monoclonal antibodies, immune checkpoint inhibitors and CAR T-cell therapies have shown promising advances in individualised cancer treatment, however, only a few are available for standard clinical practice.4 In Phase 1 clinical trials, both response rate and progression-free survival were greatly improved with personalised oncology therapy by using biomarker selection strategies compared with those undergoing generic treatment.5 Currently, developments in the detection of cancer drug targets, comprehensive molecular profiling and personalised combined treatment regimens are all contributing to increasing availability of personalised oncology to a wider range of patients.6

Patient-derived xenograft (PDX) models have been increasingly used in translational research since their development.7 Currently, cell-line xenografts are the standard for preclinical research, able to create a tumour microenvironment.8 More often than not however, they do not accurately reflect the true behaviour of the host tumour and are able to adapt to in vitro growth, losing the original properties of the host tumour.7 Other models for tumour graft strategies have previously had limited success.8

Trials examining PDX models have shown that they can produce samples that are authentic to the host tumour.9 They are able to accurately replicate tumour growth, diversity of tumour cells and tumour progression, including metastatic potential.7,9,10 PDX models have been shown to be used for prognostication: studies have shown that successful engraftment is associated with a poorer prognosis that can be correlated clinically.10 Another study, by exploring heterogeneous sarcoma patients with a wide range of prognoses and tumour subtypes, demonstrated that PDX models aided therapeutic decision-making in the case of a collection of disparate tumours where each one is a rare subtype.11

While neuroblastomas are the most common extracranial solid tumour in children, they are generally rare, and have a wide variety of outcomes depending on the specific, albeit variable, biology of the tumour.1214 Children with high-risk neuroblastoma have a less than 50% chance of cure.10 This has led to recognition of an increasing need for personalised treatment for patients with high-risk neuroblastomas.15 This is especially relevant as current treatment regimens have a range of acute toxicities and long-term side effects.15,16,17 RCTs effectively test new interventions, remove allocation bias, are ethically conducted and ensure that no subject receives less-than-baseline care; however, they are expensive and can take many years to complete. RCTs are also ultimately not appropriate for the requirement for rapid developments in any field, let alone a rare cancer18—where they are less valid, almost impossible to recruit adequate numbers for and are thus performed less frequently, leading to a dearth of evidence.19 In children there is a scarcity of trials conducted, with issues relating to feasibility as well as ethics. This leads to an absence of evidence, and alternatives need to be sought.20 Existing in vivo and in vitro data will always remain the preclinical vanguards of drug development, but the clinical use of patient-derived xenografts can enhance the robustness of preclinical studies.14

In this edition of the BJC, Kamili et al. investigated the reliability of establishing PDX models for high-risk neuroblastomas.16 They examine different techniques including different engraftment sites and different biological sample types, such as metastatic and primary tumour samples. Previous papers have reported that PDX models are more informative than cell-line xenografts; however, they have demonstrated limited engraftment success rates, prolonged establishment of grafts and high costs.12,13,21 This paper’s key finding is that of successful engraftment via orthotopic implantation, a method leading to more rapid model development. This is in keeping with previous research in advanced sarcoma patients, which highlighted the need for time-efficient engraftment in order to see a benefit within a clinically appropriate timescale.16

Kamili et al. are able to address the lengthy establishment time that has been a limiting factor in previous studies.16 All of the orthotopically inoculated tumours were successfully engrafted and resulted in the quickest time to engraftment compared with subcutaneously and intramuscularly inoculated tumours. Indeed, PDX models were established for 4 of 9 of patients at diagnosis and all patients tested (5/5) at relapse.16 These findings show that PDX can be established quickly, which is key in high-risk and rapidly developing tumours. Although the orthotopic model was found to be the most rapid engraftment approach, there were also subcutaneous and intramuscular xenografts that were found to be equally as representative of the donor tumour. This suggests that in future, the xenograft type can be selected depending on its clinical utility. It has also shown that PDX models can reliably be established from a diverse range of samples, depending on which is most accessible clinically.16 This is going to be especially central to rare cancers, paediatric tumours or those with a range of subtypes, though one can argue that oncology is heading in that direction already. It is also recognised that tissue samples from tumours have often been inaccessible, and this has been a limiting factor.21 This is significant for development of personalised models that can be used in future clinical trials and clinical practice. Another important aspect of this paper is the expansion of patient material for ex vivo and in vivo drug testing. Personalised PDX models would allow for prioritisation of therapeutic options and provide an evidence-based platform for decisions regarding personalised therapy.16

This research explores the challenges of developing PDX models, including xenogeneic graft versus host disease and proliferation of EBV-infected cells. It does, however, propose a strategy to overcome and limit using this T-lymphocyte depletion.16 It emphasises the possibility that a personalised approach to cancer treatment and research can be developed with these models. With the development of reliable engraftment, this could lead to informative preclinical models for individual patients.16,22 It also proposes the use of xenografts to expand the current limited basis for drug testing in cancer patients, minimising the need for expensive and prolonged randomised controlled trials.7,9 Changes in research practice are needed to adapt to the current medical climate. Innovations, such as PDX models, have the ability to change what is considered standard practice and improve access to personalised treatment.

Acknowledgements

None

Author contributions

All authors contributed to the writing of the paper and approved the final submitted version.

Ethics approval and consent to participate

Not applicable

Consent to publish

Not applicable.

Data availability

Not applicable.

Competing interests

In 2018—present Professor Stebbing, the Editor-in-Chief of Oncogene sat on SABs for Celltrion, Singapore Biotech, Vor Biopharma, TLC Biopharmaceuticals and Benevolent AI, has consulted with Lansdowne partners, Vitruvian and Social Impact Capital and he chairs the Board of Directors for BB Biotech Healthcare Trust and Xerion Healthcare. Jenna Bhimani and Katie Ball declare no competing interests.

Funding information

This work is supported by the Imperial BRC and ECMC.

Footnotes

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

1. Jackson SE, Chester JD. Personalised cancer medicine. Int. J. Cancer. 2015;137:262–266. 10.1002/ijc.28940. [Abstract] [CrossRef] [Google Scholar]
2. Janiaud P, Serghiou S, Ioannidis J. New Clinical trial designs in the era of precision medicine: an overview of definitions, strengths, weaknesses and current view in oncology. Cancer Treat. Rev. 2019;73:20–30. 10.1016/j.ctrv.2018.12.003. [Abstract] [CrossRef] [Google Scholar]
3. Kelley RK, Van Bebber SL, Phillips KA, Venook AP. Personalized medicine and oncology practice guidelines: a case study of contemporary biomarkers in colorectal cancer. J. Natl Compr. Canc. Netw. 2011;9:13–25. 10.6004/jnccn.2011.0004. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
4. Krzyszczyk P, Acevedo A, Davidoff EJ, Timmins LM, Marrero-Berrios I, Patel M, et al. The growing role of precision and personalized medicine for cancer treatment. Technology Singap. World Sci. 2018;6:79–100. [Europe PMC free article] [Abstract] [Google Scholar]
5. Schwaederle M, Zhao M, Lee J, Lazar V, Leyland-Jones V, Schilsky RL, et al. Association of biomarker-based treatment strategies with response rates and progression-free survival in refractory malignant neoplasms: a meta-analysis. JAMA Oncol. 2016;2:1452–1459. 10.1001/jamaoncol.2016.2129. [Abstract] [CrossRef] [Google Scholar]
6. Hannah C, Wise, David B. Solit, precision oncology: three small steps forward. Cancer Cell. 2019;35:825–826. 10.1016/j.ccell.2019.05.009. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
7. Hildago 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. 10.1158/2159-8290.CD-14-0001. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
8. Yada, E., Wada, S., Yoshida, S. & Sasada T. Use of patient-derived xenograft mouse models in cancer research and treatment. Future Sci. OA. 4, FSO271 (2017) [Europe PMC free article] [Abstract]
9. Tentler JJ, Tan AC, Weekes CD, Jimeno A, Leong S, Pitts TM, et al. Patient derived tumour xenografts as models for oncology drug development. Nat. Rev. Clin. Oncol. 2012;9:338–350. 10.1038/nrclinonc.2012.61. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
10. DeRose YS, Wang G, Lin YC, Bernard PS, Buys SS, Ebbert MT, et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat Med. 2011;17:1514–1520. 10.1038/nm.2454. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
11. Stebbing J, Paz K, Schwartz GK, Wexler LH, Maki R, Pollock RE, et al. Patient-derived xenografts for individualized care in advanced sarcoma. Cancer. 2014;120:3588. 10.1002/cncr.28696. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
12. Van Arendonk KJ, Chung DH. Neuroblastoma: Tumor Biology and Its Implications for Staging and Treatment. Children. 2019;6:12. 10.3390/children6010012. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
13. Kaatsch P. Epidemiology of childhood cancer. Cancer Treat. Rev. 2010;36:277–285. 10.1016/j.ctrv.2010.02.003. [Abstract] [CrossRef] [Google Scholar]
14. Matthay KK, Maris JM, Schleiermacher G, Nakagawara A, Mackall CL, Diller L, et al. Neuroblastoma. Nat. Rev. Dis. Primers. 2016;2:16078. 10.1038/nrdp.2016.78. [Abstract] [CrossRef] [Google Scholar]
15. Pinto NR, Applebaum MA, Volchenboum SL, Matthay KK, London WB, Ambros BF, et al. Advances in risk classification and treatment strategies for Neuroblastoma. J. Clin. Oncol. 2015;33:3008–3017. 10.1200/JCO.2014.59.4648. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
16. Kamili A., Gifford A., Li N., Mayoh C., Chow S. O., Failes T. W. et al. Accelerating development of high-risk neuroblastoma patient-derived xenograft models for pre-clinical testing and personalised therapy. Br. J. Cancer (2019); 10.1038/s41416-019-0682-4. [Europe PMC free article] [Abstract]
17. Cohen LE, Gordon JH, Popvsky EY, Gunawardene S, Duffey-Lind E, Lehmann LE, et al. Late effects in children treated with multimodel therapy for high risk neuroblastoma: high incidence of endocrine and growth problems. Bone Marrow Transplant. 2014;49:502–508. 10.1038/bmt.2013.218. [Abstract] [CrossRef] [Google Scholar]
18. Perez-Gomez, A., Mejia-Trujillo, J. & Mejia, A. How useful are randomized controlled trials in a rapidly changing world? Glob. Ment. Health (Camb). 3, e6 2016) [Europe PMC free article] [Abstract]
19. Behera M, Kumar A, Soares H, Sokol L, Djulbegovic B. Evidence Based medicine for rare diseases: implications for data interpretation and clinical trial design. Cancer Control. 2017;14:160–166. 10.1177/107327480701400209. [Abstract] [CrossRef] [Google Scholar]
20. Joseph PD, Craig JC, Caldwell P. Clinical trials in children. Bri. J.Clin. Pharmacol. 2015;79:357–369. 10.1111/bcp.12305. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
21. Applebaum MA. Second malignancies in patients with neuroblastoma: the effects of risk based therapy. Paediatr. Blood Cancer. 2015;62:128–133. 10.1002/pbc.25249. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
22. Zarzosa P, Navarro N, Giralt I, Molist C, Almazán-Moga A, Vidal I, et al. Patient derived xenografts for childhood solid tumours; a valuable tool to test new drugs and personalize treatments. Clin. Transl. Oncol. 2017;19:44–50. 10.1007/s12094-016-1557-2. [Abstract] [CrossRef] [Google Scholar]

Articles from British Journal of Cancer are provided here courtesy of Cancer Research UK

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/74144592
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/74144592

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1038/s41416-019-0678-0

Supporting
Mentioning
Contrasting
1
44
0

Article citations


Go to all (38) article citations

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.


Funding 


Funders who supported this work.

Engineering and Physical Sciences Research Council (1)

National Institute for Health Research (NIHR) (1)