“Of mice and men”: values and liabilities of the athymic nude mouse model in anticancer drug development

https://doi.org/10.1016/j.ejca.2003.11.028Get rights and content

Abstract

Human tumour xenografts implanted subcutaneously (s.c.) into immunosuppressed mice have played a significant role in preclinical anticancer drug development for the past 25 years. Their use as a predictive indicator of probable clinical activity has been validated for cytotoxics. A retrospective analysis for 39 compounds where both extensive xenograft testing and Phase II clinical data were available, performed by the National Cancer Institute (NCI), has shown that 15/33 agents (45%) with activity in more than one-third of xenografts showed clinical activity (P=0.04). However, with the exception of non-small cell lung cancer, activity within a particular histological type of the xenograft generally did not predict for clinical activity in the same tumour. Today, the question (largely unanswered) is how useful is the xenograft model (particularly the traditional s.c. model) in contemporary cancer drug discovery? There are many variables when conducting xenograft experiments which impact on outcome; viz, site of implantation, growth properties of the xenograft and size when treatment is initiated, agent formulation, scheduling, route of administration and dose and the selected endpoint for assessing activity. The xenograft model remains of value in current preclinical cancer drug development, especially when such studies give due consideration to the above variables and are based on sound mechanistic (e.g. status of the selected target in the chosen model) and pharmacological (e.g. use of formulated agent) principles. Dependent upon the drug target, a slowing of xenograft tumour growth (cytostatic effect) rather than tumour shrinkage might be the major observed effect. Human tumour xenografts are also particularly useful in determining pharmacodynamic markers of response for subsequent clinical application. Nevertheless, it needs to be kept in mind that the use of xenografts is relatively time-consuming and expensive, raises animal ethical issues and there are instances where the model is inappropriate as a likely predictor of clinical outcome (e.g. inhibitors of the metastatic process and anti-angiogenic strategies as the vasculature is of murine origin).

Introduction

Contemporary anticancer drug development is a multi-million dollar and time-consuming business. Typically, from concept to the completion of Phase III clinical trials and gaining regulatory approval requires in excess of 10 years and as much as 500 million dollars. For the first quarter of a century of modern cancer drug development (circa 1945–1969), thousands of generally randomly produced molecules were tested in mice bearing rapidly growing murine leukaemias (e.g. P388 and L1210) [1]. In 1969, came the first report of the growth of a human tumour in an immunodeficient “nude” (athymic) mouse [2]. Since then, human tumour xenografts grown in nude [3] or in mice with severe combined immunodeficiency (SCID) [4] have covered all of the major tumour types and represented the mainstay of preclinical anticancer drug development testing in vivo.

The modern paradigm for anticancer drug discovery, as widely used by drug companies and within some academic groups, comprises a series of carefully constructed steps that are designed to rapidly and efficiently allow “proof of principle”, pharmaceutically-tractable, molecules to be tested in Phase I and II clinical trials. Such a cascade (Fig. 1) may be envisaged as a large number of molecules feeding into a series of iterative stop/go tests of increasing biological complexity. The concept of “therapeutic index”, that is the demonstration of antitumour efficacy at doses well below those causing severe toxicities, is also a long-established paradigm in preclinical drug discovery [5]. A key part of the preclinical stage of the process, and often representing a significant bottleneck, is the demonstration of antitumour efficacy in a “relevant” tumour model in vivo.

However, what is the place for the nude mouse xenograft model in cancer drug development today in this post-genomic era? What are its values? What are its limitations? Is testing in vivo even required—could predictive answers be obtained solely using human tumour cells in culture or even using in silico methodology, thus avoiding the increasing ethical issues raised by animal experimentation? As opposed to the previous era of “cytotoxic” cancer drugs, contemporary cancer drug development encompasses a wide variety of approaches generally based on attacking specific molecular targets where often, cytostatic rather than cytotoxic effects may be predicted. Thus, a slowing of tumour growth rather than shrinkage may occur. This may require a re-evaluation of the in vivo models developed and validated using cytotoxic drugs when testing such agents. Even within mouse models, is it the case that, in some instances, murine syngeneic models (often dismissed since the advent of human tumour models), transgenic models or orthotopic models may be more appropriate to use than human subcutaneously (s.c.) implanted xenografts in immune-suppressed animals? In some cases, maybe there is no appropriate preclinical model? In an industry where time is paramount, should we dispense with relatively slow and laborious xenograft efficacy determinations in favour of some more rapid, higher-throughput alternative (e.g. the hollow fibre assay [6])?

Hence, this article aims to provide a critical review of the role of human tumour xenografts transplanted in athymic (or SCID) mice in contemporary cancer drug development. Both values and liabilities will be discussed specifically in the context of experience with firstly cytotoxic platinum-based molecules and secondly with small molecules targeted against contemporary cancer targets (e.g. farnesyltransferase, heat shock protein 90, telomerase and tumour vasculature).

Section snippets

Human tumour xenografts: a validated model in the development of cytotoxic drugs

Within a few years of the original description of the nude mouse model, the Developmental Therapeutics Program at the National Cancer Institute (NCI) adopted, in 1976, the use of 3 human tumour s.c. xenografts (one representing each of colon, CX-1, breast, MX-1, and lung, LX-1, cancers) into its in vivo cancer drug screening programme [7]. Since then, although far greater emphasis has been placed on initial screening using panels of disease-oriented human tumour cell lines [8], the xenograft

Use in cytotoxic cancer drug development

In addition, during the 1980s, many groups established disease-specific panels of xenografts from patient biopsies for the purposes of either studying tumour biology, responses to existing therapy (radiotherapy or chemotherapy) or for the discovery of specific classes of new drugs. A particular emphasis at the Institute of Cancer Research, London, was to establish parallel panels of in vitro cell lines (as a renewable source for biochemical and molecular studies) and corresponding in vivo

A standard operating procedure (SOP) for conducting xenograft experiments? 99 ways to determine efficacy in a xenograft model: values and limitations

Before considering whether xenografts should still be used in contemporary cancer drug discovery (see below), it is critical to appreciate and understand each of the many variables that exist in the use of xenografts in drug testing. The cancer drug discovery literature is overwhelmed with preclinical studies invariably describing “active” new molecules where it is then proposed that these should be tested in the clinic. Commonly, such studies have involved human tumour xenografts, but used in

The value (and limitations) of the xenograft model in contemporary cancer drug discovery

Today there is far less focus on the development of further cytotoxics. In addition, there has been a considerable move away from the “black-box” approach to Phase I clinical trials where many agents of unknown mechanisms of action and poorly defined preclinical pharmacokinetics were introduced into the clinic. Does this necessarily mean that the xenograft model is of no further value in contemporary mechanism-directed cancer drug development? I believe that this should be considered on a

Summary

During the era of cytotoxic cancer drug discovery, human tumour xenografts s.c. transplanted in athymic mice played a pivotal role in late preclinical agent optimisation and guiding the selection of candidates for Phase I clinical trials. A retrospective review conducted by the NCI for 39 agents for which Phase II activity data were available showed that, where compounds were active in at least one-third of xenograft models tested, there was a statistically significant correlation with activity

References (38)

  • Plowman J, Dykes DJ, Hollingshead M, Simpson-Herren L, Alley M.C. Human tumor xenograft models in NCI drug development....
  • M.C. Alley et al.

    Feasibility of drug screening with panels of human tumor cell lines using a microculture tetrazolium assay

    Cancer Research

    (1988)
  • G.G. Steel et al.

    The response to chemotherapy of a variety of human tumour xenografts

    British Journal of Cancer

    (1983)
  • V.D. Courtenay et al.

    An in vitro colony assay for human tumours grown in immune suppressed mice and treated in vivo with cytotoxic agents

    British Journal of Cancer

    (1978)
  • H.-H. Fiebig et al.

    Human tumor xenografts and explants

  • L.R. Kelland et al.

    Characterization of four new cell lines derived from human squamous carcinomas of the uterine cervix

    Cancer Research

    (1987)
  • L.R Kelland et al.

    Establishment and response to chemotherapy of human cervical carcinoma xenografts

  • L.R. Kelland et al.

    Human ovarian carcinoma cell lines and companion xenograftsa disease oriented approach to new platinum anticancer drug development

    Cancer Chemother. Pharmacol.

    (1992)
  • M. Jones et al.

    Acquisition of platinum drug resistance and platinum cross resistance patterns in a panel of human ovarian carcinoma xenografts

    British Journal of Cancer

    (1993)
  • Cited by (313)

    • Experimental models for cancer brain metastasis

      2024, Cancer Pathogenesis and Therapy
    • Preclinical tumor mouse models for studying esophageal cancer

      2023, Critical Reviews in Oncology/Hematology
    • Integrated pharmacokinetic/pharmacodynamic/efficacy analysis in oncology: importance of pharmacodynamic/efficacy relationships

      2023, Overcoming Obstacles in Drug Discovery and Development: Surmounting the Insurmountable-Case Studies for Critical Thinking
    View all citing articles on Scopus
    View full text