Skip to main content

REVIEW article

Front. Oncol., 16 March 2023
Sec. Gastrointestinal Cancers: Gastric and Esophageal Cancers
This article is part of the Research Topic Clinical Trials, Practice and Design in Gastrointestinal Cancers View all 23 articles

Is it feasible and ethical to randomize patients between surgery and non-surgical treatments for gastrointestinal cancers?

  • Department of Visceral, Vascular and Endocrine Surgery, University Hospital Halle (Saale), Martin-Luther-University Halle-Wittenberg, Halle, Germany

Background: In several settings in the treatment of gastrointestinal cancers, it is unclear if the addition of surgery to a multimodal treatment strategy, or in some circumstances its omission, lead to a better outcome for patients. In such situations of clinical equipoise, high-quality evidence from randomised-controlled trials is needed to decide which treatment approach is preferable.

Objective: In this article, we outline the importance of randomised trials comparing surgery with non-surgical therapies for specific scenarios in the treatment of gastrointestinal cancers. We explain the difficulties and solutions of designing these trials and recruiting patients in this context.

Methods: We performed a selective review based on a not systematic literature search in core databases, supplemented by browsing health information journals and citation searching. Only articles in English were selected. Based on this search, we discuss the results and methodological characteristics of several trials which randomised patients with gastrointestinal cancers between surgery and non-surgical treatments, highlighting their differences, advantages, and limitations.

Results and conclusions: Innovative and effective cancer treatment requires randomised trials, also comparing surgery and non-surgical treatments for defined scenarios in the treatment of gastrointestinal malignancies. Nevertheless, potential obstacles to designing and carrying out these trials must be recognised ahead of time to avoid problems before or during the trial.

Introduction

In several settings in the treatment of gastrointestinal cancers, the available data cannot answer the question whether surgery or non-surgical treatments lead to a better outcome for patients. In such situations of clinical equipoise, to provide a valid answer to this question, as for any treatment recommendation in medicine, high-quality evidence is needed. Despite improvements in the quality of clinical research in surgical oncology, several aspects regarding the design of studies comparing surgery to no surgery are still a problem. Only some surgical treatments have been assessed in randomised controlled trials (RCTs), and a relevant proportion of surgical treatments is based on scarce and conflicting evidence (1). Practical and personal experience drives the apparent progress in surgery to a much higher extent than in drug treatments. Surgical RCTs represent only 15% of the published RCTs, and only about 24% of surgical therapies are supported by evidence from RCTs (24). A large proportion of published studies in surgical oncology have a retrospective observational design with several limitations and inherent risk of bias. Despite of recent efforts in designing surgical RCTs, in a systematic review of 388 randomised clinical trials, the sizes of surgical trials were small (5). Also, discrepancies with the published protocol and reporting bias were frequent (611). Randomising patients between additional surgery and no surgery involves confronting several problems: commercial interests in the light of high reimbursements for many surgeries, lack of cooperation between surgical and non-surgical departments, hesitancy and ethical concerns of patients and investigators to randomise between surgery and non-surgical treatments with the knowledge that surgery is a viable option, and blinding of patients and surgeons.

In this article, we outline the importance of conceiving randomised trials comparing surgery with non-surgical therapies for specific scenarios in the treatment of gastrointestinal cancers, highlighting the difficulties and solutions of designing these trials and recruiting patients in this context.

Why do we need randomization between surgery and non-surgical treatments?

An RCT has several advantages. The prospective nature of the study implies a planned assessment, documentation, and follow-up (1). The blinded RCT provides the highest level of evidence in evidence-based medicine and minimizes bias. Randomization is the best design to establish causal relationship between exposure and outcome. Non-randomized comparative cohort studies provide important data, but only with statistical adjustments (a. e. propensity score analysis) from covariates, an association between intervention and outcome may be shown, and a considerable risk of bias persists.

Regarding RCTs comparing surgery and non-surgical treatments, different types of comparison groups are possible: no active intervention, medical management, deferred surgery, active monitoring (“watch and wait”), physical or manual therapy and placebo (sham surgery).

In a systematic review comparing quality domains in trials of surgical interventions to a previously reported control sample of trials of medical interventions, although reporting of quality domains was suboptimal, surgical trials compared favorably to medical trials (12). “They were 24% more likely to have an adequate method of random sequence generation, and 71% more likely to have an adequate method of allocation concealment. However, blinding was 40% less likely to be adequate in surgical trials, and sources of funding were 33% less likely to be reported” (12). Although it is not a specific limitation of RCTs, publication bias is also a problem that has to be faced when designing these trials. Selective outcome reporting is a known problem of RCTs (10). For example, in neurosurgery, it was shown that RCTs comparing surgical to non-operative treatment fairly frequently changed their outcome measures, which may distort the available results of a given trial und undermines the trials’ credibility (13).

Randomized controlled trials comparing surgery with non-surgical treatments are rare, but with the development of new multimodal therapy regimens in gastrointestinal cancer surgery, randomized comparisons of different medical and surgical approaches are needed (14). For example, conversion surgery is defined as an operation aiming to clear all tumor sites after tumors that had initially been considered technically unresectable or where a resection was deemed to be of no oncological benefit, responded to chemotherapy and become resectable (15). Another example is the possible omission of surgery after very good response to chemotherapy or chemoradiotherapy, as it is now discussed regarding complete response after total neoadjuvant therapy for rectal cancer (16).

Advantages and disadvantages of non-randomized trials

Usually, observational studies have some advantages when compared to RCTs: lower cost, greater timeliness, and a broader range of patients eligible for study inclusion. Despite its limitations on comparing treatments, they are used to identify risk factors and prognostic factors (17). Furthermore, in some clinical scenarios, non-randomized prospective cohort studies categorizing and comparing observational data may represent better alternatives than RCTs (1821). These types of studies potentially lead to a higher participation of the patients in the interventional group, mostly according to the preferences of the clinician or the patient. Despite of the risk of selection bias, these studies give insights on the outcomes of the effects of surgical treatments and provide, in some cases, quality evidence comparable to RCTs. The level of evidence gained from a poor quality RCT is not necessarily better than that from a well-conducted cohort study. A priori registration of protocols is still not required in observational studies but would be a major strength to avoid explorative data analyses. Conducting and reporting observational studies according to the Strengthening the Reporting of Observational studies in Epidemiology (STROBE) Statement is a requirement for publication in some journals (22). Nevertheless, prospective observational studies usually represent complementary evidence or are the basis for designing RCTs (23, 24). Chalmers et al. reported that 56 percent of non-randomized trials reported on favorable treatment effects, as compared with 30 percent of blinded, randomized-controlled trials. This potential selection bias was also reported in other studies (2528).

As demonstrated, the potential for bias in RCTs is normally lower when compared to non-randomized studies. Bias is defined as the systematic difference between the study results and an RCT, addressing the same question and conducted on the same participant group that had no flaws in its conduct. Assessing bias of a non-randomized study involves comparing it to a hypothetical pragmatic RCT that compares the health effects of the same interventions and is conducted in the same participants without features putting it at risk of bias. The assessment of risk of bias in non-randomized studies involves pre-intervention, at-intervention, and post-intervention features of the study (29, 30). The bias related to non-random allocation results in over- or underestimations of treatment effects, being large enough to lead studies to false conclusions. Even when applying case-mix adjustment methods (i. e. logistic regression, propensity score) bias stays significant (31). The absence of reliable methods to prevent the biasing consequences of selection bias in observational research leaves non-randomized studies for situations when RCTs are unfeasible or unethical. Unfeasibility of RCTs usually is present when the disease or indication is very rare, and ethical problems often arise when very large treatment effects can already be seen in non-randomized studies, so that equipoise can no longer be assumed (32).

Disadvantages of randomized trials

Surgical trials are difficult to conceive, and only half of the initiated trials reach their recruitment target (3335). When performing these studies, surgical clinician scientists face several obstacles such as the surgical learning curve and the lack of financial support. Furthermore, blinding problems, poor generalizability of the trial population and difficulties with randomization in emergency situations represent important adversities that researchers must overcome. These and other problems result in 21% of RCTs in surgery being discontinued and 34% being unpublished (36, 37)

Surgical trials face patient and surgeon related challenges: a radical choice between treatments, patients’ discomfort with randomization between an operation and no operation, patients’ or clinicians’ a priori preferences for one or the other treatment, and an imbalanced presentation of the treatment options to patients (38). Regarding trials comparing surgical and non-surgical interventions, slow recruitment is mentioned to be the most common problem that researchers have to confront, with the consequence that no evidence-based treatment recommendations can be made (3942).

Furthermore, historical and cultural limitations are relevant when designing RCTs comparing surgery with non-surgical treatments. Most surgical treatments were developed to treat conditions that were untreatable with other means and were potentially life-threatening. Once a surgical treatment is established, it is difficult and sometimes appears ethically questionable to compare it to a medical treatment or surveillance. Structural, political, and commercial aspects also play an important role. Regarding ethical aspects, the possible adverse effects of surgery and non-surgical treatments usually differ substantially, and surgery is mostly irreversible with organs or parts thereof being removed. Due to these limitations, an indirect selection bias may be present in these RCTs, as only a small subgroup of patients may agree to participate on them.

Placebo controlled trials represent another option in this context. In the context of surgery, placebo means sham surgery, i.e. general anesthesia without an actual operation, or a surgical procedure intended to mimic the actual operation. However, the conception of a placebo control in a surgical RCT may be challenging and ethically difficult because the surgical unlike the medical “placebo” bears a relevant degree of invasiveness. If there is no expected benefit (beside the placebo effect), patients are usually resistant to undergo the low-risk anesthesia required for a sham surgery intervention. Blinding is also very difficult in this kind of trials. Nevertheless, surgical RCTs with a placebo arm are feasible, with the recruitment of patients remaining the leading challenge (43).

Advantages of randomized trials

Notwithstanding the challenges outlined above, randomized-controlled trials remain the gold standard for generating evidence on what is the best treatment for a given condition or in a specific setting. This holds equally true with regard to both medical treatments as well as surgical procedures and is of particular importance for patients with gastrointestinal cancers, where the choice of treatment has direct implications on survival, treatment-related morbidity and mortality, and quality of life, among other outcomes. Therefore, all reasonable efforts should be made to design and carry out randomized-controlled trials also for comparing surgical treatments with no surgery in patients with gastrointestinal cancers. Motivating patients for enrolling into such trials requires open, patient-centered, and evidence-based communication. Only by thoroughly explaining all expected risks and benefits, both in terms of procedural and long-term oncological outcomes, in an impartial way, patients can be empowered to make an informed decision on trial participation, which will ultimately enhance the probability of enrollment (44, 45). In a situation of assumed clinical equipoise, which is the foundation of all RCTs, surgery should neither be regarded only as a chance for cure or prolongation of life without appreciating its associated risks nor as a mere invasive procedure with morbidity and mortality risks without considering possible beneficial effects on oncological outcomes like survival. Quality of life, which can possibly be affected in both a positive and negative direction by a surgical procedure, is of high importance for many patients when deciding for or against surgery and must be specifically addressed in such conversations (46). Pre-existing preferences of patients towards one or the other treatment need to be considered, addressed openly and discussed using available evidence (47). The general advantages of participating in a controlled clinical trial, such as close monitoring, possibly more frequent follow-up visits and access to novel treatments, need to be well explained to patients, but should not be overstated in a promotional manner (48). While discontinuation of the trial by single patients should obviously not be encouraged, the freedom of choice to quit trial participation at any time, and eventually even to seek the alternative treatment, i.e. surgery for patients who had been randomized into the no surgery arm or no surgery for patients who had been randomized into the surgery arm (as long as the operation has not been carried out) should be addressed, too. “Placebo”-controlled trials are almost impossible to realize in surgical oncology. Sham surgery, which would potentially delay further non-surgical treatments such as chemotherapy, seems ethically not acceptable for cancer patients. Sham anesthesia could be a theoretical less invasive option, but a lack of scars would still render long-term blinding of patients not feasible. Therefore, RCTs in surgical oncology including those enrolling patients with gastrointestinal cancer are usually open-label studies.

Given that the likelihood of selective participation in RCTs randomizing between surgery and no surgery based on patients’ characteristics is considerable, efforts should be made to collect baseline but also outcome data from patients who are screened and offered trial participation, but who ultimately choose not to enroll. Observational cohorts comprising patients who refused trial participation or did not meet all inclusion criteria but were treated with identical interventions as if they had participated in the respective trial, can support evidence generated by RCTs. In a specific example of an RCT comparing preoperative radiotherapy plus surgery with surgery alone in patients with retroperitoneal sarcoma, results from such an observational cohort closely resembled the results from the actual RCT (49).

Another possible solution is the use of adaptive randomized trial designs. This allows modifications to the trial design during the collection of patient outcome data and despite its challenges, may present several advantages when compared to standard trial designs (50).

Examples of successful randomization between surgery and non-surgical treatments

Several examples show that RCTs comparing surgery and no surgery in specific treatment settings of gastrointestinal cancers can be successfully conducted.

In a potentially curative setting, the FFCD 9102 trial randomized patients with thoracic esophageal squamous cell carcinoma or adenocarcinoma who had shown clinical response to neoadjuvant chemoradiotherapy to either resection or continuation of chemoradiotherapy (51). Only 14 of 273 patients (5.1%) fulfilling all eligibility criteria refused randomization. Compliance with the allocated treatment was high with only 10 of 129 patients (7.8%) randomized to surgery deciding against the operation and only 1 of 130 patients (0.8%) randomized to continuation of chemoradiotherapy demanding surgery. A trial with a similar design randomized 37 of 38 eligible patients (97.4%) with squamous cell carcinoma of the esophagus, who showed complete clinical and metabolic response to chemoradiotherapy, to esophagectomy or observation (52). While all 18 patients allocated to observation were compliant with that treatment with some patients being operated on later because of secondary progression, 6 of 19 patients (31.6%) allocated to surgery chose not to have the operation. Overall enrolment into the trial was much slower than expected which together with the low compliance with treatment in the surgery arm led to premature trial closure. The trialists assumed that compliance of patients allocated to surgery was low due to the timing of randomization after complete response had been confirmed and with a general change of local treatment patterns towards observation instead of surgery. The ongoing RENAISSANCE trial randomizes patients with oligometastatic gastroesophageal adenocarcinoma and no disease progression following chemotherapy between additional chemotherapy or resection of the primary tumor and the metastatic lesions followed by chemotherapy (53). In a similar population, i.e. patients with gastric adenocarcinoma and one metastatic site, the REGATTA trial randomized between gastrectomy followed by chemotherapy and chemotherapy alone (54). All enrolled 175 patients were successfully randomized. While 7 of 86 patients (8.1%) allocated to chemotherapy alone withdrew consent, 1 of 89 (1.1%) patients allocated to gastrectomy plus chemotherapy decided not to undergo the operation.

In rectal adenocarcinoma, which often shows very good or even complete response to neoadjuvant chemoradiotherapy, several trials randomizing between rectal resection and organ preservation, either through a watch-and-wait strategy or local excision, have been or are being conducted. The GRECCAR-2 trial randomized 145 out of 146 eligible patients (99.3%) who demonstrated good response to chemoradiotherapy (55). Only 1 of 74 patients (1.4%) allocated to local excision underwent rectal resection while 8 of 73 patients (11.0%) allocated to rectal resection underwent local excision and 3 of 73 patients (4.1%) no surgery at all. In the TREC trial, 55 of 152 identified eligible patients (36.2%) consented to randomization between organ preservation by transanal microsurgery and radical rectal resection (56). Of the 27 patients allocated to organ preservation, 3 patients (11.1%) crossed over to the rectal resection arm, and one patient had to end protocol treatment because of metastatic disease. Of the 28 patients allocated to rectal resection, 3 patients (10.7%) refused surgery and crossed over to the organ preservation arm.

The SYNCHRONOUS trial randomized patients with colon cancer and unresectable synchronous metastases to resection of the primary before starting chemotherapy (187 patients) and chemotherapy without prior resection (206 patients). Results have so far only been published in abstract form, and no information on the proportion of eligible screened patients who were randomized and on compliance with the allocated treatments are available (57).

A Chinese trial randomized patients with metastatic gastrointestinal stromal tumor responding to imatinib treatment either to surgery of residual disease followed by continuation of imatinib treatment or to continuation of imatinib treatment without surgery (58). Although only 5 of 46 screened eligible patients (10.9%) refused entering the trial, the trial had to be closed prematurely due to slow accrual. However, all patients received the treatment they were allocated to with no crossing over or refusal of therapy.

Conclusions

As in all other fields of medicine, guidelines, and recommendations for when and if surgery for gastrointestinal cancers should be performed need to be based on evidence of the highest possible level. Such evidence can only be provided by well-designed RCTs with other study designs bearing a non-negligible risk of bias, which compromises the validity of their results. A randomization between an operation and no operation with either a watch-and-wait approach or an alternative non-surgical treatment is ethically fully acceptable if there is clinical equipoise between the two treatments. However, it is often more difficult for patients and physicians to accept than a randomization between two drugs or even between a presumably active drug and a placebo. Frequently, there is an a priori preference towards either the surgical treatment or against surgery, even if such preferences are not supported by available data. A dedicated explanation of all expected risks and benefits associated with trial participation, and the open discussion of patients’ pre-existing preferences are key factors for achieving fast and unselected recruitment into these RCTs. Several trials conducted in esophageal cancer and colorectal cancer show that randomization between surgery and no surgery or microsurgery can be successfully done both in a setting with curative intent and in metastatic disease. These examples should be encouraging for researchers to conceive of, design, and carry out more of these RCTs to provide high-level evidence for unanswered treatment questions for gastrointestinal cancers.

Author contributions

AR, JKlo, JKle and UR performed research, selected manuscripts wrote and edited the paper. All authors contributed to the article and approved the submitted version.

Funding

This work was partially supported by the Advanced Clinician Scientist Program of the Medical Faculty of the Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany. The content is solely the responsibility of the authors and does not represent the official views of the funding agencies.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Solheim O. Randomised controlled trials in surgery and the glass ceiling effect. Acta Neurochirurgica (2019) 161(4):623–25. doi: 10.1007/s00701-019-03850-3

CrossRef Full Text | Google Scholar

2. Gelijns AC, Ascheim DD, Parides MK, Kent KC, Moskowitz AJ. Randomised trials in surgery. Surgery (2009) 145:581–7. doi: 10.1016/j.surg.2009.04.003

CrossRef Full Text | Google Scholar

3. Wente MN, Seiler CM, Uhl W, Büchler MW. Perspectives of evidence-based surgery. Dig Surg (2003) 20:263–9. doi: 10.1159/000071183

CrossRef Full Text | Google Scholar

4. Howes N, Chagla L, Thorpe M, McCulloch P. Surgical practice is evidence based. BrJ Surg (1997) 84:1220–3.

Google Scholar

5. Robinson NB, Fremes S, Hameed I, Rahouma M, Weidenmann V, Demetres M, et al. Characteristics of randomised clinical trials in surgery from 2008 to 2020: A systematic review. JAMA (2021) 2008:1–13. doi: 10.1001/jamanetworkopen.2021.14494

CrossRef Full Text | Google Scholar

6. McCulloch P, Altman DG, Campbell WB, Flum DR, Glasziou P, Marshall JC, et al. No surgical innovation without evaluation: The IDEAL recommendations. Lancet (2009) 374(9695):1105–12. doi: 10.1016/S0140-6736(09)61116-8

CrossRef Full Text | Google Scholar

7. Riaz A, Cooper D, Muir G. The reporting quality of randomised controlled trials in surgery: A systematic review. Int J Surg (2007) 5(6):413–22. doi: 10.1016/j.ijsu.2007.06.002

CrossRef Full Text | Google Scholar

8. Hajibandeh S, Hajibandeh S, Antoniou GA, Green PA, Maden M, Torella F. Reporting and methodological quality of randomised controlled trials in vascular and endovascular surgery. Eur J Vasc Endovascular Surg (2015) 50(5):664–70. doi: 10.1016/j.ejvs.2015.06.114

CrossRef Full Text | Google Scholar

9. Jones CW, Keil LG, Holland WC, Caughey MC, Platts-Mills TF. Comparison of registered and published outcomes in randomised controlled trials: A systematic review. BMC Med (2015) 13(1). doi: 10.1186/s12916-015-0520-3

CrossRef Full Text | Google Scholar

10. Mathieu S, Boutron I, Moher D, Altman DG, Ravaud P. Comparison of registered and published primary outcomes in randomised controlled trials. Jama (2009) 302(9):977–84. doi: 10.1001/jama.2009.1242

CrossRef Full Text | Google Scholar

11. Stubenrouch FE, Cohen ES, Bossuyt PMM, Koelemay MJW, van der Vet PCR, Ubbink DT. Systematic review of reporting benefits and harms of surgical interventions in randomised clinical trials. BJS Open (2020) 4(2):171–81. doi: 10.1002/bjs5.50240

CrossRef Full Text | Google Scholar

12. Adie S, Harris IA, Naylor JM, Mittal R. The quality of surgical versus non-surgical randomised controlled trials. Contemp Clin Trials Commun (2017) 5:63–6. doi: 10.1016/j.conctc.2016.12.001

CrossRef Full Text | Google Scholar

13. Martin E, Muskens IS, Senders JT, DiRisio AC, Karhade AV, Zaidi HA, et al. Randomised controlled trials comparing surgery to non-operative management in neurosurgery: A systematic review. Acta Neurochirurgica (2019) 161(4):627–34. doi: 10.1007/s00701-019-03849-w

CrossRef Full Text | Google Scholar

14. Grayling MJ, Dimairo M, Mander AP, Jaki TF. A review of perspectives on the use of randomization in phase II oncology trials. JNCI: J Natl Cancer Institute (2019) 111(12):1255–62. doi: 10.1093/jnci/djz126

CrossRef Full Text | Google Scholar

15. Zhang F, Huang X, Song Y, Gao P, Zhou C, Guo Z, et al. Conversion surgery for stage IV gastric cancer. Front Oncol (2019) 9:1158. doi: 10.3389/fonc.2019.01158

CrossRef Full Text | Google Scholar

16. Smith CA, Kachnic LA. Evolving treatment paradigm in the treatment of locally advanced rectal cancer. J Natl Compr Canc Netw (2018) 16(7):909–15. doi: 10.6004/jnccn.2018.7032

CrossRef Full Text | Google Scholar

17. Benson K, Hartz AJ. A comparison of observational studies and randomised, controlled trials. N Engl J Med (2000) 342(25):1878–86. doi: 10.1056/NEJM200006223422506

CrossRef Full Text | Google Scholar

18. Slim K. Limits of evidence-based surgery. World J Surg (2005) 29(5):606–9. doi: 10.1007/s00268-005-7922-x

CrossRef Full Text | Google Scholar

19. Young JM, Solomon MJ. Improving the evidence-base in surgery: Evaluating surgical effectiveness. ANZ J Surg (2003) 73(7):507–10. doi: 10.1046/j.1445-1433.2003.02662.x

CrossRef Full Text | Google Scholar

20. Lilford R, Braunholtz D, Harris J, Gill T. Trials in surgery. Br J Surg (2004) 91(1):6–16. doi: 10.1002/bjs.4418

CrossRef Full Text | Google Scholar

21. McCulloch P, Taylor I, Sasako M, Lovett B, Griffin D. Randomised trials in surgery: problems and possible solutions. BMJ (2002) 324(7351):1448–51. doi: 10.1136/bmj.324.7351.1448

CrossRef Full Text | Google Scholar

22. von Elm E, Altman DG, Egger M, Pocock SJ, Gøtzsche PC, Vandenbroucke JP, et al. The strengthening the reporting of observational studies in epidemiology (STROBE) statement: Guidelines for reporting observational studies. J Clin Epidemiol. (2008) 61(4):344–9. doi: 10.1016/j.jclinepi.2007.11.008

CrossRef Full Text | Google Scholar

23. Abraham NS, Byrne CJ, Young JM, Solomon MJ. Meta-analysis of well-designed nonrandomised comparative studies of surgical procedures is as good as randomised controlled trials. J Clin Epidemiol (2010) 63(3):238–45. doi: 10.1016/j.jclinepi.2009.04.005

CrossRef Full Text | Google Scholar

24. Lassen K, Hoye A, Myrmel T. Randomised trials in surgery: The burden of evidence. Rev Recent Clin Trials (2012) 7(3):244–48. doi: 10.2174/157488712802281402

CrossRef Full Text | Google Scholar

25. Sacks H, Chalmers TC, Smith H Jr. Randomised versus historical con- trols for clinical trials. Am J Med (1982) 72:233–40. doi: 10.1016/0002-9343(82)90815-4

CrossRef Full Text | Google Scholar

26. Colditz GA, Miller JN, Mosteller F. How study design affects outcomes in comparisons of therapy. I. Medical Stat Med (1989) 8:441–54. doi: 10.1002/sim.4780080408

CrossRef Full Text | Google Scholar

27. Miller JN, Colditz GA, Mosteller F. How study design affects outcomes in comparisons of therapy. II. Surgical Stat Med (1989) 8:455–66. doi: 10.1002/sim.4780080409

CrossRef Full Text | Google Scholar

28. Chalmers TC, Celano P, Sacks HS, Smith H Jr. Bias in treatment as- signment in controlled clinical trials. N Engl J Med (1983) 309:1358–61. doi: 10.1056/NEJM198312013092204

CrossRef Full Text | Google Scholar

29. Lonjon G, Porcher R, Ergina P, Fouet M, Boutron I. Potential pitfalls of reporting and bias in observational studies with propensity score analysis assessing a surgical procedure: A methodological systematic review. Ann Surg (2017) 265(5):901–9. doi: 10.1097/SLA.0000000000001797

CrossRef Full Text | Google Scholar

30. Sterne JA, Hernán MA, Reeves BC, Savović J, Berkman ND, Viswanathan M, et al. ROBINS-I: A tool for assessing risk of bias in non-randomised studies of interventions. BMJ (2016) 355:i4919. doi: 10.1136/bmj.i4919

CrossRef Full Text | Google Scholar

31. Stürmer T, Joshi M, Glynn RJ, Avorn J, Rothman KJ, Schneeweiss S. A review of the application of propensity score methods yielded increasing use, advantages in specific settings, but not substantially different estimates compared with conventional multivariable methods. J Clin Epidemiol (2006) 59(5):437–47. doi: 10.1016/j.jclinepi.2005.07.004

CrossRef Full Text | Google Scholar

32. Deeks J, Dinnes J, D’Amico R, Sowden A, Sakarovitch C, Song F, et al. Evaluating non-randomised intervention studies. Health Technol Assess (2003) 7(27):iii–x, 1–173. doi: 10.3310/hta7270

CrossRef Full Text | Google Scholar

33. McDonald AM, Knight RC, Campbell MK, Entwistle VA, Grant AM, Cook JA. What influences recruitment to randomised controlled trials? A review of trials funded by two UK funding agencies. Trials (2006) 7(1). doi: 10.1186/1745-6215-7-9

CrossRef Full Text | Google Scholar

34. Walters SJ, dos Anjos Henriques-Cadby IB, Bortolami O, Flight L, Hind D, Jacques RM, et al. Recruitment and retention of participants in randomised controlled trials: A review of trials funded and published by the united kingdom health technology assessment programme. BMJ Open (2017) 7(3):e015276. doi: 10.1136/bmjopen-2016-015276

CrossRef Full Text | Google Scholar

35. Sully BG, Julious SA, Nicholl J. A reinvestigation of recruitment to randomised, controlled, multicenter trials: A review of trials funded by two UK funding agencies. Trials (2013) 14(1):166. doi: 10.1186/1745-6215-14-166

CrossRef Full Text | Google Scholar

36. Chapman SJ, Shelton B, Mahmood H, Fitzgerald JE, Harrison EM, Bhangu A, et al. Discontinuation and non-publication of surgical randomised controlled trials: observational study. BMJ (2014) 349:g6870. doi: 10.1136/bmj.g6870

CrossRef Full Text | Google Scholar

37. Augustinus S, van Goor IWJM, Berkhof J, Daamen LA, Koerkamp BG, Mackay TM, et al. Alternative randomised trial designs in surgery: Systematic review. Ann Surgery Ann Surg (2022) 276(5):753–60. doi: 10.1097/SLA.0000000000005620

CrossRef Full Text | Google Scholar

38. Davies L, Beard D, Cook JA, Price A, Osbeck I, Toye F. The challenge of equipoise in trials with a surgical and non-surgical comparison: A qualitative synthesis using meta-ethnography. Trials (2021) 22(1):1–24. doi: 10.1186/s13063-021-05403-5

CrossRef Full Text | Google Scholar

39. Watson JM, Torgerson DJ. Increasing recruitment to randomised trials: A review of randomised controlled trials. BMC Med Res Methodol (2006) 6(1). doi: 10.1186/1471-2288-6-34

CrossRef Full Text | Google Scholar

40. Cook JA, Ramsay CR, Norrie J. Recruitment to publicly funded trials–are surgical trials really different? Contemp Clin Trials (2008) 29(5):631–4. doi: 10.1016/j.cct.2008.02.005

CrossRef Full Text | Google Scholar

41. Lim CT, Roberts HJ, Collins JE, Losina E, Katz JN. Factors influencing the enrollment in randomised controlled trials in orthopedics. Contemp Clin Trials Commun (2017) 8:203–8. doi: 10.1016/j.conctc.2017.10.005

CrossRef Full Text | Google Scholar

42. Nguyen TK, Nguyen EK, Warner A, Louie AV, Palma DA. Failed randomised clinical trials in radiation oncology: What can we learn? Int J Radiat Oncol Biol Phys (2018) 101(5):1018–24. doi: 10.1016/j.ijrobp.2018.04.030

CrossRef Full Text | Google Scholar

43. Wartolowska K, Collins GS, Hopewell S, Judge A, Dean BJF, Rombach I, et al. Feasibility of surgical randomised controlled trials with a placebo arm: A systematic review. BMJ Open (2016) 6(3):1–9. doi: 10.1136/bmjopen-2015-010194

CrossRef Full Text | Google Scholar

44. Harrison JD, Solomon MJ, Young JM, Meagher A, Hruby G, Salkeld G, et al. Surgical and oncology trials for rectal cancer: who will participate? Surgery (2007) 142(1):94–101. doi: 10.1016/j.surg.2007.01.013

CrossRef Full Text | Google Scholar

45. Wright JR, Whelan TJ, Schiff S, Dubois S, Crooks D, Haines PT, et al. Why cancer patients enter randomised clinical trials: exploring the factors that influence their decision. J Clin Oncol (2004) 22(21):4312–8. doi: 10.1200/JCO.2004.01.187

CrossRef Full Text | Google Scholar

46. Holmes E, Crome P, Arora A. Patients' preferences and existential perspective: what to consider and how should patient's expectations be guided? Aging Clin Exp Res (2018) 30(3):271–5. doi: 10.1007/s40520-017-0868-7

CrossRef Full Text | Google Scholar

47. Ubel PA, Merz JF, Shea J, Asch DA. How preliminary data affect people's stated willingness to enter a hypothetical randomised controlled trial. J Investig Med (1997) 45(9):561–6.

Google Scholar

48. So YJ, Jameson M, Newton V, O'Donnell A, Jeffery M, Jackson C, et al. Investigating strategies to improve clinical trial opportunities for patients with cancer in new Zealand-INSIGHT. N Z Med J (2019) 132(1498):10–31.

Google Scholar

49. Callegaro D, Raut CP, Ajayi T, Strauss D, Bonvalot S, Ng D, et al. Preoperative radiotherapy in patients with primary retroperitoneal sarcoma: EORTC-62092 trial (STRASS) versus off-trial (STREXIT) results. Ann Surg (2022). doi: 10.1097/SLA.0000000000005492

CrossRef Full Text | Google Scholar

50. Mukherjee A, Grayling MJ, Wason JMS. Adaptive designs: Benefits and cautions for neurosurgery trials. World Neurosurg (2022) 161:316–22. doi: 10.1016/j.wneu.2021.07.061

CrossRef Full Text | Google Scholar

51. Bedenne L, Michel P, Bouché O, Milan C, Mariette C, Conroy T, et al. Chemoradiation followed by surgery compared with chemoradiation alone in squamous cancer of the esophagus: FFCD 9102. J Clin Oncol (2007) 25(10):1160–8. doi: 10.1200/JCO.2005.04.7118

CrossRef Full Text | Google Scholar

52. Park SR, Yoon DH, Kim JH, Kim YH, Kim HR, Lee HJ, et al. A randomised phase III trial on the role of esophagectomy in complete responders to preoperative chemoradiotherapy for esophageal squamous cell carcinoma (ESOPRESSO). Anticancer Res (2019) 39(9):5123–33. doi: 10.21873/anticanres.13707

CrossRef Full Text | Google Scholar

53. Al-Batran SE, Goetze TO, Mueller DW, Vogel A, Winkler M, Lorenzen S, et al. The RENAISSANCE (AIO-FLOT5) trial: effect of chemotherapy alone vs. chemotherapy followed by surgical resection on survival and quality of life in patients with limited-metastatic adenocarcinoma of the stomach or esophagogastric junction - a phase III trial of the German AIO/CAO-V/CAOGI. BMC Cancer (2017) 17(1):893. doi: 10.1186/s12885-017-3918-9

CrossRef Full Text | Google Scholar

54. Fujitani K, Yang HK, Mizusawa J, Kim YW, Terashima M, Han SU, et al. Gastrectomy plus chemotherapy versus chemotherapy alone for advanced gastric cancer with a single non-curable factor (REGATTA): A phase 3, randomised controlled trial. Lancet Oncol (2016) 17(3):309–18. doi: 10.1016/S1470-2045(15)00553-7

CrossRef Full Text | Google Scholar

55. Rullier E, Rouanet P, Tuech JJ, Valverde A, Lelong B, Rivoire M, et al. Organ preservation for rectal cancer (GRECCAR 2): A prospective, randomised, open-label, multicentre, phase 3 trial. Lancet (2017) 390(10093):469–79. doi: 10.1016/S0140-6736(17)31056-5

CrossRef Full Text | Google Scholar

56. Bach SP, Gilbert A, Brock K, Korsgen S, Geh I, Hill J, et al. Radical surgery versus organ preservation via short-course radiotherapy followed by transanal endoscopic microsurgery for early-stage rectal cancer (TREC): A randomised, open-label feasibility study. TREC collaborators.Lancet Gastroenterol Hepatol (2021) 6(2):92–105. doi: 10.1016/S2468-1253(20)30333-2

CrossRef Full Text | Google Scholar

57. Rahbari NN, Biondo S, Feißt M, Bruckner T, Rossion I, Luntz S, et al. Randomized clinical trial on resection of the primary tumor versus no resection prior to systemic therapy in patients with colon cancer and synchronous unresectable metastases. J Clin Oncol (2022) 40(Suppl 17):LBA3507–LBA3507. doi: 10.1200/JCO.2022.40.17_suppl.LBA3507

CrossRef Full Text | Google Scholar

58. Du CY, Zhou Y, Song C, Wang YP, Jie ZG, He YL, et al. Is there a role of surgery in patients with recurrent or metastatic gastrointestinal stromal tumours responding to imatinib: A prospective randomised trial in China. Eur J Cancer (2014) 50(10):1772–8. doi: 10.1016/j.ejca.2014.03.280

CrossRef Full Text | Google Scholar

Keywords: cancer, randomized control trial (RCT), ethical, gastrointestinal, oncology

Citation: Rebelo A, Klose J, Kleeff J and Ronellenfitsch U (2023) Is it feasible and ethical to randomize patients between surgery and non-surgical treatments for gastrointestinal cancers? Front. Oncol. 13:1119436. doi: 10.3389/fonc.2023.1119436

Received: 08 December 2022; Accepted: 08 March 2023;
Published: 16 March 2023.

Edited by:

Zhibo Yan, Qilu Hospital, Shandong University, China

Reviewed by:

Stefan Sauerland, Institut für Qualität und Wirtschaftlichkeit im Gesundheitswesen (IQWiG), Germany
Michael Grayling, Janssen Pharmaceuticals, United Kingdom

Copyright © 2023 Rebelo, Klose, Kleeff and Ronellenfitsch. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Artur Rebelo, artur.rebelo@uk-halle.de

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.