You are on page 1of 3

AVIAN INFLUENZA Shift expertise

COMMENT EARTH SYSTEMS Past climates HISTORY OF SCIENCE Descartes’ OBITUARY Wylie Vale
to track mutations where give valuable clues to future lost letter tracked using and an elusive stress
they emerge p.534 warming p.537 Google p.540 hormone p.542
S. GSCHMEISSNER/SPL

Many landmark findings in preclinical oncology research are not reproducible, in part because of inadequate cell lines and animal models.

Raise standards for


preclinical cancer research
C. Glenn Begley and Lee M. Ellis propose how methods, publications and
incentives must change if patients are to benefit.

E
fforts over the past decade to trials in oncology have the highest failure investigators must reassess their approach to
characterize the genetic alterations rate compared with other therapeutic areas. translating discovery research into greater
in human cancers have led to a better Given the high unmet need in oncology, it clinical success and impact.
understanding of molecular drivers of this is understandable that barriers to clinical Many factors are responsible for the high
complex set of diseases. Although we in the development may be lower than for other failure rate, notwithstanding the inher-
cancer field hoped that this would lead to disease areas, and a larger number of drugs ently difficult nature of this disease. Cer-
more effective drugs, historically, our ability with suboptimal preclinical validation will tainly, the limitations of preclinical tools
to translate cancer research to clinical suc- enter oncology trials. However, this low suc- such as inadequate cancer-cell-line and
cess has been remarkably low1. Sadly, clinical cess rate is not sustainable or acceptable, and mouse models2 make it difficult for even

2 9 M A RC H 2 0 1 2 | VO L 4 8 3 | NAT U R E | 5 3 1
© 2012 Macmillan Publishers Limited. All rights reserved
COMMENT

the best scientists working in optimal to drive a drug-development programme subjected themselves to a trial of a regimen
conditions to make a discovery that will ulti- it is essential that findings are sufficiently or agent that probably wouldn’t work.
mately have an impact in the clinic. Issues robust and applicable beyond the one nar- These results, although disturbing, do not
related to clinical-trial design — such as row experimental model that may have mean that the entire system is flawed. There
uncontrolled phase II studies, a reliance been enough for publication. To address are many examples of outstanding research
on standard criteria for evaluating tumour these concerns, when findings could not be that has been rapidly and reliably translated
response and the challenges of selecting reproduced, an attempt was made to contact into clinical benefit. In 2011, several new
patients prospectively — also play a signifi- the original authors, cancer drugs were approved, built on robust
cant part in the dismal success rate3. “The scientific discuss the discrep- preclinical data. However, the inability of
Unquestionably, a significant contribu- process ant findings, exchange industry and clinical trials to validate results
tor to failure in oncology trials is the qual- reagents and repeat from the majority of publications on poten-
demands
ity of published preclinical data. Drug experiments under tial therapeutic targets suggests a general,
development relies heavily on the literature,
the highest the authors’ direction, systemic problem. On speaking with many
especially with regards to new targets and standards of occasionally even in investigators in academia and industry, we
biology. Moreover, clinical endpoints in can- quality, ethics the laboratory of the found widespread recognition of this issue.
cer are defined mainly in terms of patient and rigour.” original investigator.
survival, rather than by the intermediate These investigators IMPROVING THE PRECLINICAL ENVIRONMENT
endpoints seen in other disciplines (for were all competent, well-meaning scientists How can the robustness of published pre-
example, cholesterol levels for statins). Thus, who truly wanted to make advances in can- clinical cancer research be increased? Clearly
it takes many years before the clinical appli- cer research. there are fundamental problems in both aca-
cability of initial preclinical observations In studies for which findings could be demia and industry in the way such research
is known. The results of preclinical studies reproduced, authors had paid close attention is conducted and reported. Addressing these
must therefore be very robust to withstand to controls, reagents, investigator bias and systemic issues will require tremendous
the rigours and challenges of clinical trials, describing the complete data set. For results commitment and a desire to change the
stemming from the heterogeneity of both that could not be reproduced, however, data prevalent culture. Perhaps the most crucial
tumours and patients. were not routinely analysed by investigators element for change is to acknowledge that
blinded to the experimental versus control the bar for reproducibility in performing and
CONFIRMING RESEARCH FINDINGS groups. Investigators frequently presented presenting preclinical studies must be raised.
The scientific community assumes that the the results of one experiment, such as a sin- An enduring challenge in cancer-drug
claims in a preclinical study can be taken at gle Western-blot analysis. They sometimes development lies in the erroneous use and
face value — that although there might be said they presented specific experiments that misinterpretation of preclinical data from
some errors in detail, the main message of the supported their underlying hypothesis, but cell lines and animal models. The limita-
paper can be relied on and the data will, for that were not reflective of the entire data set. tions of preclinical cancer models have been
the most part, stand the test of time. Unfor- There are no guidelines that require all data widely reviewed and are largely acknowl-
tunately, this is not always the case. Although sets to be reported in a paper; often, original edged by the field. They include the use
the issue of irreproducible data has been data are removed during the peer review and of small numbers of poorly characterized
discussed between scientists for decades, it publication process. tumour cell lines that inadequately recapitu-
has recently received greater attention (see Unfortunately, Amgen’s findings are con- late human disease, an inability to capture
go.nature.com/q7i2up) as the costs of drug sistent with those of others in industry. A the human tumour environment, a poor
development have increased along with the team at Bayer HealthCare in Germany last appreciation of pharmacokinetics and phar-
number of late-stage clinical-trial failures and year reported4 that only about 25% of pub- macodynamics, and the use of problematic
the demand for more effective therapies. lished preclinical studies could be validated endpoints and testing strategies. In addition,
Over the past decade, before pursu- to the point at which projects could con- preclinical testing rarely includes predictive
ing a particular line of research, scientists tinue. Notably, published cancer research biomarkers that, when advanced to clinical
(including C.G.B.) in the haematology and represented 70% of the studies analysed in trials, will help to distinguish those patients
oncology department at the biotechnology that report, some of which might overlap who are likely to benefit from a drug.
firm Amgen in Thousand Oaks, Califor- with the 53 papers examined at Amgen. Wide recognition of the limitations in
nia, tried to confirm published findings Some non-reproducible preclinical papers preclinical cancer studies means that busi-
related to that work. Fifty-three papers were had spawned an entire field, with hundreds ness as usual is no longer an option. Can-
deemed ‘landmark’ studies (see ‘Repro- of secondary publications that expanded on cer researchers must be more rigorous in
ducibility of research findings’). It was elements of the original observation, but their approach to preclinical studies. Given
acknowledged from the outset that some of did not actually seek to confirm or falsify its the inherent difficulties of mimicking the
the data might not hold up, because papers fundamental basis. More troubling, some of human micro-environment in preclini-
were deliberately selected that described the research has triggered a series of clinical cal research, reviewers and editors should
something completely new, such as fresh studies — suggesting that many patients had demand greater thoroughness.
approaches to targeting cancers or alterna-
tive clinical uses for existing therapeutics.
Nevertheless, scientific findings were con- REPRODUCIBILITY OF RESEARCH FINDINGS
Preclinical research generates many secondary publications, even when results cannot be reproduced.
firmed in only 6 (11%) cases. Even knowing
the limitations of preclinical research, this Journal Number of Mean number of citations of Mean number of citations of
impact factor articles non-reproduced articles* reproduced articles
was a shocking result.
Of course, the validation attempts may >20 21 248 (range 3–800) 231 (range 82–519)
have failed because of technical differences 5–19 32 169 (range 6–1,909) 13 (range 3–24)
or difficulties, despite efforts to ensure that Results from ten-year retrospective analysis of experiments performed prospectively. The term ‘non-reproduced’ was
this was not the case. Additional models assigned on the basis of findings not being sufficiently robust to drive a drug-development programme.
*Source of citations: Google Scholar, May 2011.
were also used in the validation, because

5 3 2 | NAT U R E | VO L 4 8 3 | 2 9 M A RC H 2 0 1 2
© 2012 Macmillan Publishers Limited. All rights reserved
COMMENT

lines may allow elucidation of markers of


R EC O MME N DAT I O N S sensitivity or resistance. Journals and grant
reviewers must allow for the presentation of
Improving the reliability of preclinical cancer studies imperfect stories, and recognize and reward
reproducible results, so that scientists feel
We recommend the following steps to ●●Greater dialogue should be encouraged less pressure to tell an impossibly perfect
change the culture of oncology research between physicians, scientists, patient story to advance their careers.
and improve the relevance of translational advocates and patients. Scientists benefit Although reviewers, editors and grant-
studies: from learning about clinical reality. committee members share some responsi-
●●There must be more opportunities to Physicians need better knowledge of the bility for flaws in the system, investigators
present negative data. It should be the challenges and limitations of preclinical must be accountable for the data they gener-
expectation that negative preclinical data studies. Both groups benefit from improved ate, analyse and submit. We in the field must
will be presented at conferences and in understanding of patients’ concerns. remain focused on the purpose of cancer
publications. Preclinical investigators ●●Institutions and committees should give research: to improve the lives of patients.
should be required to report all findings, more credit for teaching and mentoring: Success in our own careers should be a con-
regardless of the outcome. To facilitate this, relying solely on publications in top-tier sequence of outstanding research that has an
funding agencies, reviewers and journal journals as the benchmark for promotion impact on patients.
editors must agree that negative data can or grant funding can be misleading, The lack of rigour that currently exists
be just as informative as positive data. and does not recognize the valuable around generation and analysis of preclinical
●●Journal editors must play an active part contributions of great mentors, educators data is reminiscent of the situation in clini-
in initiating a cultural change. There must and administrators. cal research about 50 years ago. The changes
be mechanisms to report negative data that ●●Funding organizations must recognize that have taken place in clinical-trials pro-
are accessible through PubMed or other and embrace the need for new cancer- cesses over that time indicate that changes
search engines. There should be links to research tools and assist in their in prevailing attitudes and philosophies can
journal articles in which investigators have development, and in providing greater occur (see ‘Improving the reliability of pre-
reported alternative findings to those in an community access to those tools. Examples clinical cancer studies’).
initial (sometimes considered landmark) include support for establishing large Improving preclinical cancer research
publication. One suggestion is to include cancer cell-line collections with easy to the point at which it is reproducible and
‘tags’ that report whether the key findings investigator access (a simple, universal translatable to clinical-trial success will
of a seminal paper were confirmed. material-transfer agreement); capabilities be an extraordinarily difficult challenge.
●●There should be transparent for genetic characterization of newly However, it is important to remember that
opportunities for trainees, technicians and derived tumour cell lines and xenografts; patients are at the centre of all these efforts.
colleagues to discuss and report troubling identification of patient selection If we in the field forget this, it is easy to
or unethical behaviours without fearing biomarkers; and generation of more robust, lose our sense of focus, transparency and
adverse consequences. predictive tumour models. C.G.B. and L.M.E. urgency. Cancer researchers are funded
by community taxes and by the hard work
and philanthropic donations of advocates.
As with clinical studies, preclinical inves- for design, analysis and presentation of More importantly, patients rely on us to
tigators should be blinded to the control data rests with investigators, the laboratory embrace innovation, make advances and
and treatment arms, and use only rigor- and the host institution. All are account- deliver new therapies that will improve their
ously validated reagents. All experiments able for poor experimental design, a lack lives. Although hundreds of thousands of
should include and show appropriate posi- of robust supportive data or selective research papers are published annually, too
tive and negative controls. Critical experi- data presentation. The scientific process few clinical successes have been produced
ments should be repeated, preferably by demands the highest standards of quality, given the public investment of significant
different investigators in the same lab, and ethics and rigour. financial resources. We need a system that
the entire data set must be represented in will facilitate a transparent discovery pro-
the final publication. For example, showing BUILDING A STRONGER SYSTEM cess that frequently and consistently leads to
data from tumour models in which a drug What reasons underlie the publication of significant patient benefit. ■ SEE EDITORIAL P.509
is inactive, and may not completely fit an erroneous, selective or irreproducible data?
original hypothesis, is just as important as The academic system and peer-review pro- C. Glenn Begley is a consultant and
showing models in which the hypothesis was cess tolerates and perhaps even inadvertently former vice-president and global head of
confirmed. encourages such conduct5. To obtain fund- Hematology and Oncology Research at
Studies should not be published using a ing, a job, promotion or tenure, research- Amgen, Thousand Oaks, California 91359,
single cell line or model, but should include ers need a strong publication record, often USA. Lee M. Ellis is at the University of
a number of well-characterized cancer cell including a first-authored high-impact Texas M. D. Anderson Cancer Center,
lines that are representative of the intended publication. Journal editors, reviewers and Houston, Texas 77030, USA.
patient population. Cancer researchers grant-review committees often look for a e-mail: lellis@mdanderson.org
must commit to making the difficult, time- scientific finding that is simple, clear and
consuming and costly transition towards complete — a ‘perfect’ story. It is therefore 1. Hutchinson, L. & Kirk, R. Nature Rev. Clin. Oncol.
new research tools, as well as adopting tempting for investigators to submit selected 8, 189–190 (2011).
more robust, predictive tumour models and data sets for publication, or even to massage 2. Francia, G. & Kerbel, R. S. Nature Biotechnol. 28,
561–562 (2010).
improved validation strategies. Similarly, data to fit the underlying hypothesis. 3. Rubin, E. H. & Gilliland, D. G. Nature Rev.
efforts to identify patient-selection bio- But there are no perfect stories in biology. Clin. Oncol. http://dx.doi.org/10.1038/
markers should be mandatory at the outset In fact, gaps in stories can provide opportu- nrclinonc.2012.22 (2012).
4. Prinz, F., Schlange, T. & Asadullah, K. Nature Rev.
of drug development. nities for further research — for example, a Drug Discov. 10, 712 (2011).
Ultimately, however, the responsibility treatment that may work in only some cell 5. Fanelli, D. PLoS ONE 5, e10271 (2010).

2 9 M A RC H 2 0 1 2 | VO L 4 8 3 | NAT U R E | 5 3 3
© 2012 Macmillan Publishers Limited. All rights reserved

You might also like