The Application of Evidence-Based Medicine in Individualized Medicine
Abstract
:1. Introduction
- Strengthening of the standard of care anticancer treatments with immunogenic cell death (ICD) immunotherapy using oncolytic viruses (e.g., Newcastle Disease Virus) in combination with local modulated electrohyperthermia to induce in situ ICD of tumor cells, aimed at immune modulation in the tumor micro-environment plus conditioning of the patient’s immune system before vaccination.
- Active specific immunotherapy after completion of chemotherapy, aimed to induce a strong specific T cell immune response against tumor-associated antigens, which is realized through the production of an individualized IO-VAC® DC vaccine (DE_NW_04_MIA_2015_0033 and DE_NW_04_MIA_2020_0017 since 27 May 2015; IOZK GmbH, Cologne, Germany). This vaccine is an approved advanced therapy medicinal product (ATMP) that consists of autologous mature DCs loaded with autologous tumor antigens and matured with danger signals (in this case, a Newcastle Disease Virus with cytokines cocktail). The vaccine is unique for each patient due to the use of autologous DCs and tumor antigens. During this phase of treatment, individualized modulatory immunotherapies are implemented.
- Maintenance of anticancer tumor control and expansion of immune-covered antigenic spectrum through repetitive 5-day ICD immunotherapy cycles, which target and kill potential newly developed tumor cells; and through vaccination with more universal long-peptide vaccines like WT1 and/or survivin.
- Immunization through a booster IO-VAC® vaccine, at least 6 months after the second IO-VAC® (DE_NW_04_MIA_2015_0033 and DE_NW_04_MIA_2020_0017 since 27 May 2015; IOZK GmbH, Cologne, Germany), to increase immune response and induce a memory response.
- This treatment is an experimental application;
- Concrete studies on the combination of the chosen methods are missing;
- Efficacy can only be demonstrated through controlled clinical trials with a large number of patients. Therefore, treatment with new DC vaccines should be carried out exclusively in trials until efficacy is proven. The basic rule here is that these studies should take place in centers, and participation must be voluntary and free of charge as this is an experimental therapy in which the benefits and risks are still unknown.
2. Evidence-Based Medicine
- Disregarding patient values leads to hasty or routine-protocol medicine;
- Not considering best research evidence leads to application of empirical treatments; and
- Ignoring own clinical expertise leads to patient exposure to non-suitable treatments.
2.1. Best Research Evidence
- Traditional RCTs focus on hypothesis testing by comparing an experimental arm (e.g., therapeutic intervention) to a control arm (no intervention). For the concept to work as intended, the administration of the experimental treatment should be the sole difference between the experimental and the control group. By nature, these are study designs, not treatment designs. The primary end of the trial is not to treat patients, but rather to generate generalizable medical knowledge. Aside from the financial and logistical complications (such trials take years to design and run, time these patients simply do not have), it implies that half of the patients that might benefit from a novel intervention are denied such a benefit, as they do not meet scientific design criteria. Of the patients who are accepted, half will be placed in the control group, again not receiving treatment. This conflicts with the right of the patient to consent to individualized treatment decisions based on to their condition. It is argued that patients explicitly consent to this when they endorse trial participation, but, in almost all countries, patients often have no other options left outside palliative treatment or euthanasia. This is a pertinent ethical concern, given that the Helsinki Declaration requires that “the well-being of the individual research subject must take precedence over all other interests” [16,17].
- Traditional clinical trials produce “average” results for a given outcome variable and cannot answer questions related to why therapies work in certain situations but not in others. Even the most rigorous and clearly reported RCT cannot predict if a given intervention will be effective in any specific individual. Ironically, these questions are of most value to patients, and thus of most interest to clinicians. This is especially valid in (immune-)oncological research, where the importance of evaluating patient-unique genetic and molecular hallmarks to design and administer situationally the least harmful and most effective treatment (tailored treatment), gains momentum out of apparent necessity [18,19].
- Traditional clinical trials require multiple clear and strict eligibility criteria to ensure that the study population is similar in all baseline factors that may affect the potential benefits and risks from the intervention studied. This not only requires large study populations, but also implies that patients considered at greater risk of adverse events from the trial or not expected to benefit will be excluded, as well as patients with comorbid conditions or receiving concurrent therapies. In addition, patients who are not expected to live through a clinical study, arguably those in greatest need, cannot be included. While these criteria make sense to eliminate bias and balance for unknown covariates in such experimental setup, it introduces its own “optimized patient”-bias. Furthermore, it is argued that overly strict eligibility criteria result in lower patient accrual, which is already a challenge for rare or orphan diseases where the sample sizes are small and overall survival is low. This problem is clearly presented in recent work by Liau et al. [20], who had to modify their trial design testing an autologous tumor lysate-loaded dendritic cell vaccine for treatment of glioblastoma, for feasibility and/or ethical reasons. Patients who need it most cannot benefit from an experimental treatment, even though these are the patients who will receive the treatment in clinical reality. West [21] argues that for patients who cannot await results from RCTs, which often take years to become available, retrospective clinical data may provide assurance instead. These pertinent issues in immune-oncology research result in study populations that are unrepresentative of the actual clinical population of patients with cancer, disregarding concerns and complications which occur in real-life treatment, while simultaneously limiting patient access to new treatments [6,22,23,24,25].
- Traditional clinical trials do not consider rapid advances in tumor biology, which slices and dices cancer into ever smaller subsets. Indeed, it is assumed that all randomized individuals are, and will remain, homogeneous, and that no change within the set of investigated subjects occurs during the study period except the changes due to treatment. This is not true for cancers, which are known to evolve through continuous and rapid accumulation of genomic mutations. This calls for smaller, shorter, focused approaches, as targeted therapies will, by nature, become more individualized [14,26,27]. This complexity of personalized medicine means that a model subset cannot accurately present patient heterogeneity.
- Rapid expansion of novel immune-oncology agents may result in a need to compare new therapies and new combination treatments against each other and/or against an expanding list of standard care treatments. Conducting multiple RCTs to facilitate these comparisons gets overly complicated, expensive and resource-intensive. Due to the complicated and time-expensive setup of RCTs, they simply cannot keep up with current technological advancements, which partially explains why only 1 in 5000 to 100,000 new therapeutic inventions make it to market application [14,28].
- As RCTs are study designs in support of, but not creating, EBM, it is important that “all” evidence should be publicly available, both published and unpublished, to avoid evidence-biased medicine. Transparency in reporting is essential; if methods and data are not shared in an unbiased and open format, it contributes to the so-called reproducibility crisis [29]. It is commonly understood that about 50% of research is not published, with a strong bias towards positive results in published data (publication and reporting bias) [30,31]. Furthermore, a sample review suggested that 45% of the industry-funded trials were not required to report any results, as opposed to 6% of trials funded by the National Institutes of Health and 9% of the trials that were funded by other government or academic institutions [32]. This is of concern, given that sponsorship of drug and device studies by manufacturing companies leads to more favorable efficacy results than sponsorship by other sources (industry/sponsorship bias) [33,34].
2.2. Clinical Evidence
2.3. Patient Expectations
3. Moving Forward: Managing Evidence-Based Data in Personalized and Individualized Medicine
4. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Van Gool, S.W.; Van de Vliet, P.; Kampers, L.; Makalowski, J.; Sprenger, T.; Reich, E.; Schirrmacher, V.; Stuecker, W. Methods in anticancer cellular immunotherapy. In Methods in Cell Biology; Garg, A., Ed.; Elsevier: Amsterdam, The Netherlands, 2023. [Google Scholar]
- Schirrmacher, V.; Sprenger, T.; Stuecker, W.; Van Gool, S.W. Evidence-Based Medicine in Oncology: Commercial Versus Patient Benefit. Biomedicines 2020, 8, 237. [Google Scholar] [CrossRef]
- Huber, F. Individueller Heilversuch und Klinisches Experiment. Ph.D. Thesis, Universität Augsburg, Augsburg, Germany, 2014. [Google Scholar]
- Van Gool, S.W.; Makalowski, J.; Van de Vliet, P.; Van Gool, S.; Sprenger, T.; Schirrmacher, V.; Stuecker, W. Individualized multimodal immunotherapy for adults with IDH1 wild-type GBM: A single institute experience. Cancers 2023, 15, 1194. [Google Scholar] [CrossRef] [PubMed]
- Schirrmacher, V. Cancer Vaccines and Oncolytic Viruses Exert Profoundly Lower Side Effects in Cancer Patients than Other Systemic Therapies: A Comparative Analysis. Biomedicines 2020, 8, 61. [Google Scholar] [CrossRef] [Green Version]
- Deaton, A.; Cartwright, N. Understanding and misunderstanding randomized controlled trials. Soc. Sci. Med. 2018, 210, 2–21. [Google Scholar] [CrossRef]
- Roussos Torres, E.T.; Epstein, A.L. Adopting an alternative structure for clinical trials in immunotherapy. Expert. Rev. Anticancer Ther. 2021, 21, 373–375. [Google Scholar] [CrossRef]
- Sackett, D.L. Evidence-based medicine. Semin. Perinatol. 1997, 21, 3–5. [Google Scholar] [CrossRef] [PubMed]
- Masic, I.; Miokovic, M.; Muhamedagic, B. Evidence based medicine—New approaches and challenges. Acta Inform. Med. 2008, 16, 219–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Donnell, J.C.; Le, T.K.; Dobrin, R.; Higashi, M.; Pereira, A.; Wagner, S.; Yang, A.; Hukkelhoven, M. Evolving use of real-world evidence in the regulatory process: A focus on immuno-oncology treatment and outcomes. Future Oncol. 2021, 17, 333–347. [Google Scholar] [CrossRef]
- The Centre for Evidence-Based Medicine. The Oxford Levels of Evidence 2. Available online: www.cebm.net/index.aspx?o=5653 (accessed on 25 April 2023).
- Jones, D.S.; Podolsky, S.H. The history and fate of the gold standard. Lancet 2015, 385, 1502–1503. [Google Scholar] [CrossRef] [Green Version]
- Marshall, J. The standard of care in oncology is unacceptable. Oncology 2006, 20, 806. [Google Scholar]
- Subbiah, V. The next generation of evidence-based medicine. Nat. Med. 2023, 29, 49–58. [Google Scholar] [CrossRef] [PubMed]
- Jureidini, J.; McHenry, L.B. The illusion of evidence based medicine. BMJ 2022, 376, o702. [Google Scholar] [CrossRef] [PubMed]
- Nardini, C. The ethics of clinical trials. Ecancermedicalscience 2014, 8, 387. [Google Scholar] [CrossRef] [PubMed]
- Kyr, M.; Svobodnik, A.; Stepanova, R.; Hejnova, R. N-of-1 Trials in Pediatric Oncology: From a Population-Based Approach to Personalized Medicine-A Review. Cancers 2021, 13, 5428. [Google Scholar] [CrossRef] [PubMed]
- Sackett, D.L.; Rosenberg, W.M.; Gray, J.A.; Haynes, R.B.; Richardson, W.S. Evidence based medicine: What it is and what it isn’t. BMJ 1996, 312, 71–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ellis, L.M.; Bernstein, D.S.; Voest, E.E.; Berlin, J.D.; Sargent, D.; Cortazar, P.; Garrett-Mayer, E.; Herbst, R.S.; Lilenbaum, R.C.; Sima, C.; et al. American Society of Clinical Oncology perspective: Raising the bar for clinical trials by defining clinically meaningful outcomes. J. Clin. Oncol. 2014, 32, 1277–1280. [Google Scholar] [CrossRef] [Green Version]
- Liau, L.M.; Ashkan, K.; Brem, S.; Campian, J.L.; Trusheim, J.E.; Iwamoto, F.M.; Tran, D.D.; Ansstas, G.; Cobbs, C.S.; Heth, J.A.; et al. Association of Autologous Tumor Lysate-Loaded Dendritic Cell Vaccination with Extension of Survival among Patients with Newly Diagnosed and Recurrent Glioblastoma: A Phase 3 Prospective Externally Controlled Cohort Trial. JAMA Oncol. 2023, 9, 112–121. [Google Scholar] [CrossRef]
- West, H.J. Clinical Decision Making in the Real World-The Perfect as the Enemy of the Good. JAMA Oncol. 2023, in press. [Google Scholar] [CrossRef]
- Jin, S.; Pazdur, R.; Sridhara, R. Re-Evaluating Eligibility Criteria for Oncology Clinical Trials: Analysis of Investigational New Drug Applications in 2015. J. Clin. Oncol. 2017, 35, 3745–3752. [Google Scholar] [CrossRef]
- Kyr, M.; Klement, G.L.; Zdrazilova-Dubska, L.; Demlova, R.; Valik, D.; Slaby, O.; Slavc, I.; Sterba, J. Editorial: Precision/Personalized Pediatric Oncology and Immune Therapies: Rather Customize Than Randomize. Front. Oncol. 2020, 10, 377. [Google Scholar] [CrossRef] [Green Version]
- Skaga, E.; Skretteberg, M.A.; Johannesen, T.B.; Brandal, P.; Vik-Mo, E.O.; Helseth, E.; Langmoen, I.A. Real-world validity of randomized controlled phase III trials in newly diagnosed glioblastoma: To whom do the results of the trials apply? Neurooncol. Adv. 2021, 3, vdab008. [Google Scholar] [CrossRef]
- Fortin, M.; Dionne, J.; Pinho, G.; Gignac, J.; Almirall, J.; Lapointe, L. Randomized controlled trials: Do they have external validity for patients with multiple comorbidities? Ann. Fam. Med. 2006, 4, 104–108. [Google Scholar] [CrossRef]
- Rodon, J.; Soria, J.C.; Berger, R.; Batist, G.; Tsimberidou, A.; Bresson, C.; Lee, J.J.; Rubin, E.; Onn, A.; Schilsky, R.L.; et al. Challenges in initiating and conducting personalized cancer therapy trials: Perspectives from WINTHER, a Worldwide Innovative Network (WIN) Consortium trial. Ann. Oncol. 2015, 26, 1791–1798. [Google Scholar] [CrossRef]
- Catani, J.P.P.; Riechelmann, R.P.; Adjemian, S.; Strauss, B.E. Near future of tumor immunology: Anticipating resistance mechanisms to immunotherapies, a big challenge for clinical trials. Hum. Vaccin. Immunother. 2017, 13, 1109–1111. [Google Scholar] [CrossRef] [Green Version]
- Stremersch, S.; Van Dyck, W. Marketing of the life sciences: A new framework and research agenda for a nascent field. J. Mark. 2009, 73, 4–30. [Google Scholar] [CrossRef]
- Munafo, M.R.; Chambers, C.D.; Collins, A.M.; Fortunato, L.; Macleod, M.R. Research Culture and Reproducibility. Trends Cogn. Sci. 2020, 24, 91–93. [Google Scholar] [CrossRef]
- Fosse, V.; Oldoni, E.; Bietrix, F.; Budillon, A.; Daskalopoulos, E.P.; Fratelli, M.; Gerlach, B.; Groenen, P.M.A.; Holter, S.M.; Menon, J.M.L.; et al. Recommendations for robust and reproducible preclinical research in personalised medicine. BMC Med. 2023, 21, 14. [Google Scholar] [CrossRef] [PubMed]
- Team, E.J.A. Levels of Evidence—Applicability of Evidence for the Context of a Relative Effectiveness Assessment. Guidance Document. Available online: https://www.eunethta.eu/ (accessed on 25 April 2023).
- Anderson, M.L.; Chiswell, K.; Peterson, E.D.; Tasneem, A.; Topping, J.; Califf, R.M. Compliance with results reporting at ClinicalTrials.gov. N. Engl. J. Med. 2015, 372, 1031–1039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lundh, A.; Lexchin, J.; Mintzes, B.; Schroll, J.B.; Bero, L. Industry sponsorship and research outcome. Cochrane Database Syst. Rev. 2017, 2, MR000033. [Google Scholar] [CrossRef] [Green Version]
- Moynihan, R.; Bero, L.; Hill, S.; Johansson, M.; Lexchin, J.; Macdonald, H.; Mintzes, B.; Pearson, C.; Rodwin, M.A.; Stavdal, A.; et al. Pathways to independence: Towards producing and using trustworthy evidence. BMJ 2019, 367, l6576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haynes, B. Can it work? Does it work? Is it worth it? The testing of healthcareinterventions is evolving. BMJ 1999, 319, 652–653. [Google Scholar] [CrossRef]
- Ioannidis, J.P. Why Most Clinical Research Is Not Useful. PLoS Med. 2016, 13, e1002049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hoffmann, T.C.; Del Mar, C. Clinicians’ Expectations of the Benefits and Harms of Treatments, Screening, and Tests: A Systematic Review. JAMA Intern. Med. 2017, 177, 407–419. [Google Scholar] [CrossRef] [PubMed]
- Nejstgaard, C.H.; Bero, L.; Hrobjartsson, A.; Jorgensen, A.W.; Jorgensen, K.J.; Le, M.; Lundh, A. Association between conflicts of interest and favourable recommendations in clinical guidelines, advisory committee reports, opinion pieces, and narrative reviews: Systematic review. BMJ 2020, 371, m4234. [Google Scholar] [CrossRef]
- Bendow, J.H.; Rivera, D.R.; Lund, J.L.; Feldman, J.E.; Kim, E.S. Increasing inclusiveness of patient-centric clinical evidence generation in oncology: Real-World Data and Clinical Trials. In American Society of Clinical Oncology Educational Book 42; ASCO: Alexandria, VA, USA, 2022; pp. 116–126. [Google Scholar]
- Ioannidis, J.P.A.; Stuart, M.E.; Brownlee, S.; Strite, S.A. How to survive the medical misinformation mess. Eur. J. Clin. Investig. 2017, 47, 795–802. [Google Scholar] [CrossRef] [Green Version]
- FDA. Considerations for the Use of Real-World Data and Real-World Evidence to Support Regulatory Decision-Making for Drug and Biological Products. Guidance or Industry. Available online: https://www.fda.gov/media/154714/download (accessed on 25 April 2023).
- Makady, A.; de Boer, A.; Hillege, H.; Klungel, O.; Goettsch, W. What Is Real-World Data? A Review of Definitions Based on Literature and Stakeholder Interviews. Value Health 2017, 20, 858–865. [Google Scholar] [CrossRef] [Green Version]
- Liu, F.; Panagiotakos, D. Real-world data: A brief review of the methods, applications, challenges and opportunities. BMC Med. Res. Methodol 2022, 22, 287. [Google Scholar] [CrossRef]
- Ihrig, A.; Richter, J.; Grullich, C.; Apostolidis, L.; Horak, P.; Villalobos, M.; Grapp, M.; Friederich, H.C.; Maatouk, I. Patient expectations are better for immunotherapy than traditional chemotherapy for cancer. J. Cancer Res. Clin. Oncol. 2020, 146, 3189–3198. [Google Scholar] [CrossRef]
- EUnetHTA. Guideline: Endpoints Used for Relative Effectiveness Assessment of Pharmaceuticals, Health-Related Quality of Life and Utility Measures. Available online: https://www.eunethta.eu/wp-content/uploads/2018/01/Endpoints-used-for-Relative-Effectiveness-Assessment-Health-related-quality-of-life-and-utility-measures_Amended-JA1-Guideline_Final-Nov-2015 (accessed on 25 April 2023).
- Bottomley, A. The cancer patient and quality of life. Oncologist 2002, 7, 120–125. [Google Scholar] [CrossRef]
- Cella, D.; Chang, C.H.; Lai, J.S.; Webster, K. Advances in quality of life measurements in oncology patients. Semin. Oncol. 2002, 29, 60–68. [Google Scholar] [CrossRef] [PubMed]
- Wiklund, I. Assessment of patient-reported outcomes in clinical trials: The example of health-related quality of life. Fundam. Clin. Pharmacol. 2004, 18, 351–363. [Google Scholar] [CrossRef] [PubMed]
- Sibeoni, J.; Picard, C.; Orri, M.; Labey, M.; Bousquet, G.; Verneuil, L.; Revah-Levy, A. Patients’ quality of life during active cancer treatment: A qualitative study. BMC Cancer 2018, 18, 951. [Google Scholar] [CrossRef] [Green Version]
- Elwyn, G.; Frosch, D.; Thomson, R.; Joseph-Williams, N.; Lloyd, A.; Kinnersley, P.; Cording, E.; Tomson, D.; Dodd, C.; Rollnick, S.; et al. Shared decision making: A model for clinical practice. J. Gen. Intern. Med. 2012, 27, 1361–1367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stewart, D.J.; Kurzrock, R. Fool’s gold, lost treasures, and the randomized clinical trial. BMC Cancer 2013, 13, 193. [Google Scholar] [CrossRef] [Green Version]
- Ferrara, R.; Pilotto, S.; Caccese, M.; Grizzi, G.; Sperduti, I.; Giannarelli, D.; Milella, M.; Besse, B.; Tortora, G.; Bria, E. Do immune checkpoint inhibitors need new studies methodology? J. Thorac. Dis. 2018, 10, S1564–S1580. [Google Scholar] [CrossRef] [PubMed]
- Jayasundara, K.; Hollis, A.; Krahn, M.; Mamdani, M.; Hoch, J.S.; Grootendorst, P. Estimating the clinical cost of drug development for orphan versus non-orphan drugs. Orphanet. J. Rare Dis. 2019, 14, 12. [Google Scholar] [CrossRef] [Green Version]
- Gagne, J.J.; Thompson, L.; O’Keefe, K.; Kesselheim, A.S. Innovative research methods for studying treatments for rare diseases: Methodological review. BMJ 2014, 349, g6802. [Google Scholar] [CrossRef] [Green Version]
- Subbiah, V.; Kurzrock, R. Challenging Standard-of-Care Paradigms in the Precision Oncology Era. Trends Cancer 2018, 4, 101–109. [Google Scholar] [CrossRef]
- FDA. Framework for FDA’s Real-World Evidence Program. 2018. Available online: https://www.fda.gov/media/120060/download (accessed on 2 May 2023).
- Couespel, N.; Price, R. Strengthening Europe in the Fight Against Cancer—Going Further, Faster. Available online: http://www.europarl.europa.eu/supporting-analyses (accessed on 26 May 2023).
- Verweij, J.; Hendriks, H.R.; Zwierzina, H.; Cancer Drug Development Forum. Innovation in oncology clinical trial design. Cancer Treat Rev. 2019, 74, 15–20. [Google Scholar] [CrossRef]
- Fountzilas, E.; Tsimberidou, A.M.; Vo, H.H.; Kurzrock, R. Clinical trial design in the era of precision medicine. Genome Med. 2022, 14, 101. [Google Scholar] [CrossRef]
- Alharbi, E.; Skeva, R.; Juty, N.; Jay, C.; Goble, C. A FAIR-Decide framework for pharmaceutical R&D: FAIR data cost-benefit assessment. Drug Discov. Today 2023, 28, 103510. [Google Scholar] [CrossRef] [PubMed]
- Parmar, A.; Dai, W.F.; Dionne, F.; Geirnaert, M.; Denburg, A.; Ahuja, T.; Beca, J.; Bouchard, S.; Chambers, C.; Hunt, M.J.; et al. Development of a Multi-Criteria Decision Analysis Rating Tool to Prioritize Real-World Evidence Questions for the Canadian Real-World Evidence for Value of Cancer Drugs (CanREValue) Collaboration. Curr. Oncol. 2023, 30, 3776–3786. [Google Scholar] [CrossRef]
- Nikles, J.; Mitchell, G.K.; Schluter, P.; Good, P.; Hardy, J.; Rowett, D.; Shelby-James, T.; Vohra, S.; Currow, D. Aggregating single patient (n-of-1) trials in populations where recruitment and retention was difficult: The case of palliative care. J. Clin. Epidemiol. 2011, 64, 471–480. [Google Scholar] [CrossRef] [PubMed]
- Vohra, S.; Shamseer, L.; Sampson, M.; Bukutu, C.; Schmid, C.H.; Tate, R.; Nikles, J.; Zucker, D.R.; Kravitz, R.; Guyatt, G.; et al. CONSORT extension for reporting N-of-1 trials (CENT) 2015 Statement. BMJ 2015, 350, h1738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- European Medicines Agency. Guideline on Clinical Trials in Small Populations. EMA CHMP EWP 83561/2005. Available online: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-clinical-trials-small-populations_en.pdf (accessed on 2 May 2023).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Van de Vliet, P.; Sprenger, T.; Kampers, L.F.C.; Makalowski, J.; Schirrmacher, V.; Stücker, W.; Van Gool, S.W. The Application of Evidence-Based Medicine in Individualized Medicine. Biomedicines 2023, 11, 1793. https://doi.org/10.3390/biomedicines11071793
Van de Vliet P, Sprenger T, Kampers LFC, Makalowski J, Schirrmacher V, Stücker W, Van Gool SW. The Application of Evidence-Based Medicine in Individualized Medicine. Biomedicines. 2023; 11(7):1793. https://doi.org/10.3390/biomedicines11071793
Chicago/Turabian StyleVan de Vliet, Peter, Tobias Sprenger, Linde F. C. Kampers, Jennifer Makalowski, Volker Schirrmacher, Wilfried Stücker, and Stefaan W. Van Gool. 2023. "The Application of Evidence-Based Medicine in Individualized Medicine" Biomedicines 11, no. 7: 1793. https://doi.org/10.3390/biomedicines11071793
APA StyleVan de Vliet, P., Sprenger, T., Kampers, L. F. C., Makalowski, J., Schirrmacher, V., Stücker, W., & Van Gool, S. W. (2023). The Application of Evidence-Based Medicine in Individualized Medicine. Biomedicines, 11(7), 1793. https://doi.org/10.3390/biomedicines11071793