Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

The future of clinical trials in urological oncology

Abstract

Well-designed clinical trials in urological oncology help to guide treatment decisions and aid in counselling patients, ultimately serving to improve outcomes. Since the term evidence-based medicine was first used by Gordon Guyatt in 1991, a renewed emphasis on methodology, transparent trial design and study reporting has helped to improve clinical research and in turn, the landscape of medical literature. Novel clinical trial designs (including multi-arm, multistage trials, basket and umbrella studies and research from big data sources, such as electronic health records, administrative claims databases and quality monitoring registries) are well suited to advance innovation in urological oncology. Existing urological clinical trials are often limited by small numbers, are statistically underpowered and many face difficulties with accrual. Thus, efforts to improve trial design are of considerable importance. The development and use of standard outcome sets and adherence to reporting guidelines offer researchers the opportunity to guide value-oriented care, minimize research waste and efficiently identify solutions to the unanswered questions in urology cancer care.

Key points

  • Innovations in trial design, such as the multi-arm, multistage (MAMS) study type as well as basket and umbrella trial designs, are being used to investigate therapies for urological cancers in a cost-efficient and rapid manner.

  • Urologists should become familiar with the strengths and limitations of big data sources, such as information gathered from systems including electronic health records, administrative claims databases, prescription monitoring registries, whole-genome sequencing and social media.

  • Standard (or core) outcome sets are agreed sets of standardized outcomes that should be measured and reported as a minimum in all clinical trials in a specific area of health or health care; these are developed in conjunction with a panel of treatment experts and patient advocates. Their development and adoption can improve the utility of clinical trial data.

  • Familiarity and adherence to reporting guidelines such as CONSORT and STARD by trial designers, researchers, publishers and users of the medical literature can help to standardize and improve the transparency of trial reporting.

  • Future trials in urological oncology should focus on opportunities to fill gaps within areas of existing guideline deficiency, including areas where current clinical guidance is driven primarily by retrospective observational data or expert opinion.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: MAMS trial versus traditional RCTs.
Fig. 2: Basket and umbrella trials.

Similar content being viewed by others

References

  1. Hopewell, S., Dutton, S., Yu, L. M., Chan, A. W. & Altman, D. G. The quality of reports of randomised trials in 2000 and 2006: comparative study of articles indexed in PubMed. BMJ 340, c723 (2010).

    Article  Google Scholar 

  2. Carthon, B. C. & Antonarakis, E. S. The STAMPEDE trial: paradigm-changing data through innovative trial design. Transl Cancer Res. 5, S485–S490 (2016).

    PubMed  Google Scholar 

  3. Parmar, M. K. B. et al. Speeding up the evaluation of new agents in cancer. J. Natl Cancer Inst. 100, 1204–1214 (2008). Provides an overview of the MAMS trial design with a comprehensive overview of its methodology, strengths and limitations.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Pariser, A. R., Robb, M. & Sherman, R. E. Expedited programs for drug development and approval. Expert Opin. Orphan Drugs 1, 507–510 (2013).

    Google Scholar 

  5. US Food and Drug Administration. Food and Drug Administration Safety and Innovation Act (FDASIA) 112–144 (FDA, 2012).

  6. US Food and Drug Administration. Section 506(a) of the Food Drug and Cosmetic Act (FDCA), as added by section 902 of the Food and Drug Administration and Safety and Innovation Act (FDASIA) (FDA, 2018).

  7. Kesselheim, A. & Darrow, J. FDA designations for therapeutics and their impact on drug development and regulatory review outcomes. Clin. Pharmacol. Ther. 97, 29–36 (2015).

    CAS  PubMed  Google Scholar 

  8. Hwang, T. J. et al. Efficacy, safety, and regulatory approval of food and drug administration-designated breakthrough and nonbreakthrough cancer medicines. J. Clin. Oncol. 36, 1805–1812 (2018). An important study that highlighted the fact that despite the earlier regulatory approval of breakthrough-therapy-designated drugs, no evidence exists that these drugs provide improvements in safety or novelty.

    CAS  PubMed  Google Scholar 

  9. Shea, M. et al. Impact of breakthrough therapy designation on cancer drug development. Nat. Rev. Drug Discov. 15, 152–152 (2016).

    CAS  PubMed  Google Scholar 

  10. Krishnamurti, T., Woloshin, S., Schwartz, L. M. & Fischhoff, B. A randomized trial testing US Food and Drug Administration “breakthrough” language. JAMA Intern. Med. 175, 1856 (2015).

    PubMed  Google Scholar 

  11. European Medicines Agency. Human regulatory. Accelerated assessment (EMA, 2019).

  12. European Medicines Agency. Human regulatory. Conditional marketing authorisation (EMA, 2019).

  13. European Medicines Agency. Human regulatory. PRIME: priority medicines (EMA, 2019).

  14. Ellis, L. M. et al. American Society of Clinical Oncology perspective: raising the bar for clinical trials by defining clinically meaningful outcomes. J. Clin. Oncol. 32, 1277–1280 (2014). A highly cited and well-written summary of the American Society of Clinical Oncology Cancer Research Committee working group deliberations establishing end points that are clinically meaningful to patients. Although focused on pancreas, breast, lung and colon cancers, this reference provides a blueprint for similar efforts to be undertaken by trialists in urological oncology.

    PubMed  Google Scholar 

  15. US Food and Drug Administration. Adaptive designs for clinical trials of drugs and biologics, guidance for industry (draft guidance) (FDA, 2018).

  16. Proschan, M. A. Sample size re-estimation in clinical trials. Biom. J. 51, 348–357 (2009).

    PubMed  Google Scholar 

  17. Mehta, C. R. & Patel, N. R. Adaptive, group sequential and decision theoretic approaches to sample size determination. Stat. Med. 25, 3250–3269 (2006).

    PubMed  Google Scholar 

  18. Simon, N. & Simon, R. Adaptive enrichment designs for clinical trials. Biostatistics 14, 613–625 (2013).

    PubMed  PubMed Central  Google Scholar 

  19. Bretz, F., Schmidli, H., König, F., Racine, A. & Maurer, W. Confirmatory seamless phase II/III clinical trials with hypotheses selection at interim: general concepts. Biom. J. 48, 623–634 (2006).

    PubMed  Google Scholar 

  20. Sydes, M. R. et al. Flexible trial design in practice — stopping arms for lack-of-benefit and adding research arms mid-trial in STAMPEDE: a multi-arm multi-stage randomized controlled trial. Trials 13, 168 (2012). A review of both practical and statistical issues encountered by the STAMPEDE authors in implementing MAMS methodology, with an overview of how these issues were addressed.

    PubMed  PubMed Central  Google Scholar 

  21. Schaid, D. J., Wieand, S. & Therneau, T. M. Optimal two-stage screening designs for survival comparisons. Biometrika 77, 507–513 (1990).

    Google Scholar 

  22. Royston, P., Parmar, M. K. B. & Qian, W. Novel designs for multi-arm clinical trials with survival outcomes with an application in ovarian cancer. Stat. Med. 22, 2239–2256 (2003).

    PubMed  Google Scholar 

  23. Sydes, M. R. et al. Issues in applying multi-arm multi-stage methodology to a clinical trial in prostate cancer: the MRC STAMPEDE trial. Trials 10, 39 (2009).

    PubMed  PubMed Central  Google Scholar 

  24. Medical Research Council. STAMPEDE. Systemic Therapy in Advancing or Metastatic Prostate Cancer: Evaluation of Drug Efficacy (MRC, 2018).

  25. James, N. D. et al. Addition of docetaxel, zoledronic acid, or both to first-line long-term hormone therapy in prostate cancer (STAMPEDE): survival results from an adaptive, multiarm, multistage, platform randomised controlled trial. Lancet 387, 1163–1177 (2016). Established docetaxel chemotherapy as standard of care along with androgen-deprivation therapy for men with metastatic prostate cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. James, N. D. et al. Abiraterone for prostate cancer not previously treated with hormone therapy. N. Engl. J. Med. 377, 338–351 (2017). Established abiraterone and prednisolone as a standard of care with androgen-deprivation therapy for men with locally advanced or metastatic prostate cancer.

    CAS  PubMed Central  PubMed  Google Scholar 

  27. Parker, C. C. et al. Radiotherapy to the primary tumour for newly diagnosed, metastatic prostate cancer (STAMPEDE): a randomised controlled phase 3 trial. Lancet. 392, 2353–2366 (2018).

    PubMed Central  PubMed  Google Scholar 

  28. Gillessen, S. et al. Repurposing metformin as therapy for prostate cancer within the STAMPEDE trial platform. Eur. Urol. 70, 906–908 (2016).

    PubMed  Google Scholar 

  29. James, N. D. et al. Celecoxib plus hormone therapy versus hormone therapy alone for hormone-sensitive prostate cancer: first results from the STAMPEDE multiarm, multistage, randomised controlled trial. Lancet Oncol. 13, 549–558 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Lachin, J. M. Introduction to sample size determination and power analysis for clinical trials. Control. Clin. Trials. 2, 93–113 (1981).

    CAS  PubMed  Google Scholar 

  31. Freidlin, B., Korn, E. L., Gray, R. & Martin, A. Multi-arm clinical trials of new agents: some design considerations. Clin. Cancer Res. 14, 4368–4371 (2008).

    CAS  PubMed  Google Scholar 

  32. Wason, J. M. S. & Jaki, T. Optimal design of multi-arm multi-stage trials. Stat. Med. 31, 4269–4279 (2012).

    PubMed  Google Scholar 

  33. Bratton, D. J., Parmar, M. K. B., Phillips, P. P. J. & Choodari-Oskooei, B. Type I error rates of multi-arm multi-stage clinical trials: strong control and impact of intermediate outcomes. Trials 17, 309 (2016).

    PubMed  PubMed Central  Google Scholar 

  34. Wason, J. M. S., Stecher, L. & Mander, A. P. Correcting for multiple-testing in multi-arm trials: is it necessary and is it done? Trials 15, 364 (2014).

    PubMed  PubMed Central  Google Scholar 

  35. Bland, J. M. & Altman, D. G. Multiple significance tests: the Bonferroni method. BMJ 310, 170–170 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Moher, D. et al. CONSORT 2010 Explanation and elaboration: updated guidelines for reporting parallel group randomised trials. BMJ 340, c869–c869 (2010). A key reference summarizing the Consolidated Standards of Reporting Trials (CONSORT) 2010 statement, with explanation of CONSORT principles to guide investigators when preparing trial manuscripts.

    PubMed  PubMed Central  Google Scholar 

  37. Sweeney, C. J. et al. Chemohormonal therapy in metastatic hormone-sensitive prostate cancer. N. Engl. J. Med. 373, 737–746 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Bratton D. Design Issues and Extensions of Multi-arm Multi-stage Clinical Trials. Thesis, UCL (2015).

  39. Jitlal, M., Khan, I., Lee, S. M. & Hackshaw, A. Stopping clinical trials early for futility: retrospective analysis of several randomised clinical studies. Br. J. Cancer 107, 910–917 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Renfro, L. A. & Sargent, D. J. Statistical controversies in clinical research: basket trials, umbrella trials, and other master protocols: a review and examples. Ann. Oncol. J. Eur. Soc. Med. Oncol. 28, 34–43 (2017). Detailed overview of basket and umbrella trial designs, including definitions, examples, and a review of their strengths and limitations in practice.

    CAS  Google Scholar 

  41. Pili, R. et al. Phase II randomized, double-blind, placebo-controlled study of tasquinimod in men with minimally symptomatic metastatic castrate-resistant prostate cancer. J. Clin. Oncol. 29, 4022–4028 (2011).

    CAS  PubMed  Google Scholar 

  42. Kelly, W. K. et al. Randomized, double-blind, placebo-controlled phase III trial comparing docetaxel and prednisone with or without bevacizumab in men with metastatic castration-resistant prostate cancer: CALGB 90401. J. Clin. Oncol. 30, 1534–1540 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Seiler, R. et al. Impact of molecular subtypes in muscle-invasive bladder cancer on predicting response and survival after neoadjuvant chemotherapy. Eur. Urol. 72(4), 544–554 (2017).

    CAS  PubMed  Google Scholar 

  44. Plimack, E. R. et al. Safety and activity of pembrolizumab in patients with locally advanced or metastatic urothelial cancer (KEYNOTE-012): a non-randomised, open-label, phase 1b study. Lancet Oncol. 18, 212–220 (2017).

    CAS  PubMed  Google Scholar 

  45. US National Library of Medicine. ClinicalTrials.gov http://www.clinicaltrials.gov/ct2/show/NCT02152254 (2019).

  46. Hyman, D. M. et al. Precision medicine at Memorial Sloan Kettering Cancer Center: clinical next-generation sequencing enabling next-generation targeted therapy trials. Drug Discov. Today 20, 1422–1428 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Lopez-Chavez, A. et al. Molecular profiling and targeted therapy for advanced thoracic malignancies: a biomarker-derived, multiarm, multihistology phase II basket trial. J. Clin. Oncol. 33, 1000–1007 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Hazim, A. & Prasad, V. A pooled analysis of published, basket trials in cancer medicine. Eur. J. Cancer 101, 244–250 (2018).

    PubMed  Google Scholar 

  49. Alden, R. S., Mandrekar, S. J. & Oxnard, G. R. Designing a definitive trial for adjuvant targeted therapy in genotype defined lung cancer: the ALCHEMIST trials. Chin. Clin. Oncol. 4, 37 (2015).

    PubMed  Google Scholar 

  50. US National Library of Medicine. ClinicalTrials.gov http://www.clinicaltrials.gov/ct2/show/NCT03385655 (2019).

  51. Simon, R. Critical review of umbrella, basket, and platform designs for oncology clinical trials. Clin. Pharmacol. Ther. 102, 934–941 (2017).

    PubMed  Google Scholar 

  52. Cunanan, K. M., Iasonos, A., Shen, R., Begg, C. B. & Gönen, M. An efficient basket trial design. Stat. Med. 36, 1568–1579 (2017).

    PubMed  PubMed Central  Google Scholar 

  53. Murdoch, T. B. & Detsky, A. S. The inevitable application of big data to health care. JAMA 309, 1351 (2013).

    CAS  PubMed  Google Scholar 

  54. Ghani, K. R., Zheng, K., Wei, J. T. & Friedman, C. P. Harnessing big data for health care and research: are urologists ready? Eur. Urol. 66, 975–977 (2014).

    PubMed  Google Scholar 

  55. Auffray, C. et al. Making sense of big data in health research: towards an EU action plan. Genome Med. 8, 71 (2016).

    PubMed  PubMed Central  Google Scholar 

  56. ELIXIR. A distributed infrastructure for life-science information. elixir https://elixir-europe.org/ (2019).

  57. Yu, J. B., Gross, C. P., Wilson, L. D. & Smith, B. D. NCI SEER public-use data: applications and limitations in oncology research. Oncology 23, 288–295 (2009).

    PubMed  Google Scholar 

  58. Giordano, S. H. et al. Limits of observational data in determining outcomes from cancer therapy. Cancer 112, 2456–2466 (2008). This paper highlights the limitations of observational data, particularly those derived from claims registries such as SEER.

    PubMed  Google Scholar 

  59. Soni, P. D. et al. Comparison of population-based observational studies with randomized trials in oncology. J. Clin. Oncol. 37, 1209–1216 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Schlomer, B. J. & Copp, H. L. Secondary data analysis of large data sets in urology: successes and errors to avoid. J. Urol. 191, 587–596 (2014).

    PubMed  Google Scholar 

  61. Howerton, M. W. et al. Provider roles in the recruitment of underrepresented populations to cancer clinical trials. Cancer 109, 465–476 (2007). A systematic review that highlights the role that providers and caregivers have in adversely influencing the recruitment of under-represented groups to clinical trials.

    PubMed  Google Scholar 

  62. TSSAHA Panel. Final report of the Tuskegee Syphilis Study Ad Hoc Advisory Panel. https://biotech.law.lsu.edu/cphl/history/reports/tuskegee/report1.pdf (1973).

  63. Green, B. L., Maisiak, R., Wang, M. Q., Britt, M. F. & Ebeling, N. Participation in health education, health promotion, and health research by African Americans: effects of the Tuskegee Syphilis Experiment. J. Health Educ. 28, 196–201 (1997).

    Google Scholar 

  64. Simon, M. S. et al. Factors associated with breast cancer clinical trials participation and enrollment at a large academic medical center. J. Clin. Oncol. 22, 2046–2052 (2004).

    PubMed  Google Scholar 

  65. Ni, Y. et al. Increasing the efficiency of trial-patient matching: automated clinical trial eligibility pre-screening for pediatric oncology patients. BMC Med. Inf. Decis. Mak. 15, 28 (2015).

    Google Scholar 

  66. Murff, H. J. et al. Automated identification of postoperative complications within an electronic medical record using natural language processing. JAMA 306, 848–855 (2011).

    CAS  PubMed  Google Scholar 

  67. Choi, W. et al. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell 25, 152–165 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Jeong, I. G. et al. Association of robotic-assisted vs laparoscopic radical nephrectomy with perioperative outcomes and health care costs, 2003 to 2015. JAMA 318, 1561 (2017).

    PubMed  PubMed Central  Google Scholar 

  69. Cooperberg, M. R., Fang, R., Schlossberg, S., Wolf, J. S. & Clemens, J. Q. The AUA quality registry: engaging stakeholders to improve the quality of care for patients with prostate cancer. Urol. Pract. 4, 30–35 (2017).

    Google Scholar 

  70. Schwartz, D. & Lellouch, J. Explanatory and pragmatic attitudes in therapeutical trials. J. Chronic. Dis. 20, 637–648 (1967).

    CAS  PubMed  Google Scholar 

  71. Thorpe, K. E. et al. A pragmatic-explanatory continuum indicator summary (PRECIS): a tool to help trial designers. CMAJ 180, E47–E57 (2009).

    PubMed  PubMed Central  Google Scholar 

  72. Barocas, D. A. et al. Using a population-based observational cohort study to address difficult comparative effectiveness research questions: the CEASAR study. J. Comp. Eff. Res. 2, 445–460 (2013).

    PubMed  Google Scholar 

  73. Chalmers, I. & Glasziou, P. Avoidable waste in the production and reporting of research evidence. Lancet 374, 86–89 (2009). An important editorial regarding the concept of 'research waste', and an exploration of the various stages of research design and development that can contribute to waste production.

    PubMed  Google Scholar 

  74. Stuckler, D., King, L., Robinson, H. & McKee, M. WHO’s budgetary allocations and burden of disease: a comparative analysis. Lancet 372, 1563–1569 (2008).

    PubMed  PubMed Central  Google Scholar 

  75. Tallon, D., Chard, J. & Dieppe, P. Relation between agendas of the research community and the research consumer. Lancet 355, 2037–2040 (2000).

    CAS  PubMed  Google Scholar 

  76. Williamson, P. R. et al. Developing core outcome sets for clinical trials: issues to consider. Trials 13, 132 (2012).

    PubMed  PubMed Central  Google Scholar 

  77. Martin, N. E. et al. Defining a standard set of patient-centered outcomes for men with localized prostate cancer. Eur. Urol. 67, 460–467 (2015). An example of a standard (core) outcome set for prostate cancer, as defined by the ICHOM working group.

    PubMed  Google Scholar 

  78. Foust-Wright, C. et al. Development of a core set of outcome measures for OAB treatment. Int. Urogynecol. J. 28, 1785–1793 (2017).

    PubMed  PubMed Central  Google Scholar 

  79. Ackerman, I. N., Cavka, B., Lippa, J. & Bucknill, A. The feasibility of implementing the ICHOM standard set for hip and knee osteoarthritis: a mixed-methods evaluation in public and private hospital settings. J. Patient Rep. Outcomes 2, 32 (2017).

    PubMed  Google Scholar 

  80. Curran, G. M., Bauer, M., Mittman, B., Pyne, J. M. & Stetler, C. Effectiveness-implementation hybrid designs: combining elements of clinical effectiveness and implementation research to enhance public health impact. Med. Care 50, 217–226 (2012).

    PubMed  PubMed Central  Google Scholar 

  81. Bossuyt, P. M. et al. The STARD statement for reporting studies of diagnostic accuracy: explanation and elaboration. Ann. Intern. Med. 138, W1–W12 (2003). Similar to CONSORT, the STARD statement provides a checklist for ensuring transparent reporting of diagnostic accuracy test studies.

    PubMed  Google Scholar 

  82. Narayan, V. M., Cone, E. B., Smith, D., Scales, C. D. & Dahm, P. Improved reporting of randomized controlled trials in the urologic literature. Eur. Urol. 70, 1044–1049 (2016).

    PubMed  Google Scholar 

  83. Turner, L. et al. Consolidated standards of reporting trials (CONSORT) and the completeness of reporting of randomised controlled trials (RCTs) published in medical journals. Cochrane Database Syst. Rev. 11, MR000030 (2012).

    PubMed  Google Scholar 

  84. Smith, D. W., Gandhi, S. & Dahm, P. The reporting quality of studies of diagnostic accuracy in the urologic literature. World J. Urol. 37, 969–974 (2019).

    PubMed  Google Scholar 

  85. Narayan V. M. A critical appraisal of biomarkers in prostate cancer. World J. Urol. https://doi.org/10.1007/s00345-019-02759-x (2019).

Download references

Author information

Authors and Affiliations

Authors

Contributions

Both authors made substantial contribution to the discussion of content, wrote the article and reviewed and edited the article before submission. V.M.N. researched data for the article.

Corresponding author

Correspondence to Philipp Dahm.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Urology thanks K. Cunanan, T. Skolarus and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Glossary

Type I error

The incorrect rejection of a true null hypothesis, resulting in a false-positive conclusion.

Family-wise error

The occurrence of making a type I (false-positive) error, and doing so within the context of multiple testing (for example, when more than one hypothesis is being tested).

Natural language processing software

Software that is able to impute discrete data from free-text information entered using human syntax; for example, natural language processing software could be used to scan a patient’s progress note and identify the patient’s age, demographic information, current symptoms and treatment plan.

Meaningful use reporting initiatives

US government standards for electronic medical record use, specifically focused on promoting the adoption of electronic medical records, exchange of patient information electronically and standardizing methods for electronic prescribing, care coordination, case reporting and billing.

Cohort inception tool

A method of identifying study groups (cohorts), usually used within the contexts of claims or quality registries.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Narayan, V.M., Dahm, P. The future of clinical trials in urological oncology. Nat Rev Urol 16, 722–733 (2019). https://doi.org/10.1038/s41585-019-0243-x

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41585-019-0243-x

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing