Abstract
Cancer treatment has entered the era of personalised or precision medicine. Biomarker-driven therapies provide improved treatment efficacy and manageable toxicity profiles compared to systemic standard-of-care therapies. They also drive the development of combining non-surgical treatments, extending indications to early-stage tumours and further refining treatment lines with more precise options. The current treatment landscape, however, has introduced a complexity of approaches to cancer treatment, including the optimal timing of when to initiate and discontinue these treatments. Of note, treatment timing usually lacks evaluation in clinical trials and can be variable in real-world settings due to the impacts of medical, healthcare, and social factors. Given that more patients can benefit from multi-modality strategies, a better understanding of the prognostic impact of treatment-to-treatment intervals (TTIs) – the intervals between combined treatments and between treatment lines – is needed. Studies for this purpose can rely on existing trial and real-world data and be context-specific for treatment options, therapeutic settings, cancer types and biomarkers, healthcare settings or systems. This perspective article calls for emerging evidence of the optimal timing of cancer treatments. We anticipate that new studies on the optimal timing will bring new insights into how to better use cancer treatments, further improving treatment efficacy.
Similar content being viewed by others
Early detection and timely treatment are essential to ensure optimal survival and avoid long-term morbidities associated with complex therapies for advanced cancers. However, delayed diagnosis and long wait times for treatment remain significant healthcare problems globally, risking the progression of disease to more advanced stages and potentially shifting the treatment intent from curative to palliative [1].
Cancer treatment has entered the era of personalised or precision medicine. Cancer cells and the tumour microenvironment can be targeted by advanced therapies like targeted therapies and immunotherapies based on the expression of biomarkers, providing superior treatment efficacy and manageable toxicity profiles to systemic standard-of-care therapies [2, 3]. Such features drive progressive development of existing treatment options: combining non-surgical treatment options, extending indications to earlier-stage, resectable cancers in the adjuvant or neoadjuvant settings, and further refining treatment lines with more precise options [1,2,3]. Chemotherapy and radiotherapy continue to evolve, improving the above strategies for better outcomes [2, 3].
Optimal timing of cancer treatments
With the increasing complexity of approaches to cancer treatment, especially biomarker-driven therapies, the impact of treatment timing on prognosis becomes a pressing question to answer. Despite randomised clinical trials (RCTs) being the gold standard of evidence-based medicine, few oncological RCTs have been designed to investigate the optimal timing of cancer treatments before trial implementation. During implementation, trialists administer treatments to eligible patients; especially for adjuvant treatments, eligible patients can be selected given that they have successfully tolerated the previous treatment. Therefore, they commonly miss pre-specified time intervals between the treatments and lack an evaluation of their associations with patient outcomes. Practice guidelines and expert consensuses do not often suggest the optimal timing of treatment uses [4]. Some do [5, 6]; for example, no more than six weeks for postoperative radiation therapy (PORT) for surgically managed head and neck squamous cell carcinoma (HNSCC) [5] and surgery within 3–6 weeks of neoadjuvant treatment for early-stage non-small-cell lung cancer (NSCLC) [6]. However, at the same time, they claim no or low level of evidence to support the specific time windows [6]. Even with these practice guidelines, real-world studies have found that many patients do not receive treatment within the optimal timeframes (e.g., >50% of people with HNSCC do not receive PORT within the optimal timeframe) [7,8,9]. Selecting the time points for treatment initiation and discontinuation may have been arbitrary.
Treatment-to-treatment interval
One key focus is the treatment-to-treatment interval (TTI) between multiple modalities within one single treatment strategy (e.g., the time between neoadjuvant immunotherapy [e.g., ipilimumab, nivolumab or both] and surgery for clinical stage III melanoma [3]), and between treatment lines (e.g., the time between first-line first-generation epidermal growth factor receptor [EGFR] tyrosine kinase inhibitor [TKI] and second-line third-generation EGFR TKI for advanced NSCLC [2]). Studies should be conducted to determine optimal TTIs so that patients will not risk poorer survival or quality of life.
Associations between time intervals and outcomes such as survival are often non-linear (U- or J-shaped) [10]. We assume similar findings apply to TTI. Particularly, the right slope of the U- or J-shape could demonstrate an increasing risk of poorer survival when patients encounter a longer TTI than the day at the nadir (the lowest point on the curve). The left slope could demonstrate the increased mortality risk when the subsequent treatment is initiated immediately; then, the risk would decrease in patients with a longer TTI (but less than the day at the nadir).
Regarding the impact of TTI on patient outcomes, several studies (including one RCT) in the settings of adjuvant chemotherapy or radiotherapy have been explicitly conducted, but most did not consider non-linear associations given the study design and analytic approach, treating the exposure TTI as a continuous or categorical variable in a traditional Cox-regression analysis [8, 9, 11,12,13,14,15,16,17,18,19,20,21]. However, a few studies had considered then demonstrated the association to be non-linear via a more advanced analytic method such as the restricted cubic spline model [17,18,19,20,21].
Below is an example of a study that demonstrated a U-shaped association. In brief, the study evaluated the impact of TTI between surgery (primary radical hysterectomy and pelvic lymphadenectomy) and adjuvant radiotherapy (whole pelvic irradiation) on all-cause mortality and disease-free survival (DFS) in 1541 women with clinical stage IB-IIB cervical cancer [21]. Based on a median of 5.6 years follow-up after surgery, the authors applied a Cox regression analysis with the restricted cubic spline model and found that patients with a TTI larger than six weeks had a higher risk of all-cause mortality (Fig. 1). Specifically, the adjusted hazard ratio was 1.45 (95%CI 1.21, 1.74) for week 8 and 2.91 (1.71, 4.95) for week 12. A similar pattern can be found in DFS. Given the pattern on the right slopes, the authors recommended adjuvant radiotherapy be administered within six weeks of surgery [21]. From the U-shaped association of all-cause mortality with the nadir at weeks 4–6, we can also see the pattern on the left slope: the risk of worse outcomes was increased right at the beginning when patients immediately received adjuvant radiotherapy after surgery; the risk started to decrease until reaching a minimum among patients with a larger TTI but less than 4–6 weeks (Fig. 1) [21].
Example of treatment-to-treatment interval (between surgery and adjuvant radiotherapy) associated with all-cause mortality (a) and disease-free survival (b) [21]. WPRT Whole pelvic radiotherapy, HR Hazard ratio, CI Confidence interval.
Our assumptions on the U- or J-shaped associations were inspired by studies on traditional diagnostic and treatment intervals (e.g., the time between diagnosis and first treatment initiation). Many of these studies have debunked the typical assumption that prognosis worsens with a longer time in a linear association [10, 22,23,24,25]. Some have found non-linear associations, even U- or J-shape associations, relying on an analytic approach like the restricted cubic spline model to consider the non-linear pattern [10, 25,26,27].
Relying on the U- or J-shaped associations, an important indication for clinical and policy practice is that the pattern helps suggest the optimal timing between treatment modalities. Specifically, using the days at the nadir as the maximum timeframe [24], theoretically, patients would have a minimal mortality risk when receiving the subsequent treatment before the timeframe. At the same time, patients would potentially avoid an increased risk of mortality if they did not receive the subsequent treatment within a very short period of time after the former treatment.
In addition to the prognostic impact of TTI, studies can investigate the reasons or factors explaining the patterns in the U- or J- shape so that future interventions can address the barriers to treatment initiation within the optimal timing window. Particularly, studies can answer why patients have treatment delays with a longer TTI than the days at the nadir and why they have to immediately receive the subsequent treatment after the former treatment is discontinued.
To explain the U- or J- shaped associations, we assume medical, healthcare, and social rather than biological reasons and factors would play a significant role. To explain the pattern on the right side of the graph, studies explained that patients who delay receiving the subsequent treatment have a risk of experiencing a challenging course after the former treatment (e.g., complications, treatment toxicity, increased length of stay at the hospital, unplanned 30-day readmission) [7, 8, 10, 19, 28], and that these patients are more likely to have prognosis-related sociodemographic characteristics such female, Black, Asian and Hispanic races, older age, being uninsured, lower education and income levels, and being unmarried [7, 11, 14, 19, 28]. Of note, delay in subsequent treatment is common in real-world practice. For example, a study found over 90% of patients with stage II NSCLC received adjuvant chemotherapy outside of the 6-week timeframe usually used in RCTs [9]. In that study, reasons for delay included medical conditions like poor postoperative recovery (23%) and complications (16%), healthcare conditions like referral delay to medical oncology for treatment (16%), logistical delay due to treatment in remote areas (10%) and full re-staging before adjuvant chemotherapy as requested by the treating oncologists (8%), as well as social reasons like patient decision (18%) [9]. According to previous studies, reasons or factors for delays also include: 1) medical factors such as advanced stage [7, 11, 28], poorer performance status [11], comorbidities [7, 15, 20, 28], no completion of planned former treatment [15], type or regiment(s) of former treatment [2]; 2) healthcare factors such as long wait between the date of biomarker testing and the date of available results [29], delay in follow-up after the former treatment [8], academic or integrated network treatment facilities [7, 19] and treatments at more than one facility [7]; and 3) social factors such as long distance from treatment facility [7, 28] and states/regions for living [7, 28].
To explain the pattern on the left side of the graph, it is understandable that these patients with a higher mortality risk after immediately receiving the subsequent treatment may be yet to fully recover from the former treatment [12, 20]. This condition might partially explain a certain number of patients who failed to complete the subsequent treatment, as the main reasons include inability to tolerate the treatment (due to adverse events) and patient choice [8]. Of note, in the adjuvant setting, especially for advanced treatments such as antiangiogenic agents, an essential condition requiring patients to wait a certain time for the subsequent treatment is that surgical wound should be healed entirely; otherwise, patients who receive these treatments in the short term after surgery can have increased risks of surgical wound dehiscence, bleeding or infection, leading to treatment failure and potentially risking survival [20, 30, 31]. Among the patients who immediately receive subsequent treatment, some might be directly provided with the subsequent treatment like chemotherapy rather than taking time for a new pathological assessment so that a biomarker-driven treatment can possibly be eligible. This situation has been found in a certain proportion (22–24%) of patients with NSCLC [29].
Regarding biological reasons or factors accounting for the U- or J-shaped associations, we have not found strong evidence except for a few indications from previous studies [8, 9, 11,12,13,14,15,16,17,18,19,20,21]. For example, baseline results in one study show that patients who had estrogen receptor (ER) positive, progesterone receptor (PgR) positive, or HER2 negative early breast cancer were more likely to delay adjuvant chemotherapy [11]. Another study discussed immunosuppression induced by neoadjuvant treatments, which might play a role in mortality risk if patients have yet to recover and then shortly afterwards receive further treatments [20]. The study also discussed the downside of adjuvant radiotherapy as it could impair host immunologic effects, induce vascular damage, remodulate the tumour microenvironment, upregulate key molecules, and ultimately stimulate tumour invasion and metastasis [20, 32,33,34]. Another study reported that clinical practice has been shifting towards a longer TTI for oesophageal cancer surgery after neoadjuvant chemoradiotherapy, because of an increased probability of a complete histological response from neoadjuvant treatment [12, 35]. Confirmation and more discoveries are needed by future research.
Other research focuses
In addition to TTI, we hold similar assumptions of the U- or J-shaped association of prognosis with the cycle-to-cycle interval of a treatment. Current evidence includes a lower survival probability in people with breast cancer who experienced inter-cycle delays of more than 7 days during the period of receiving neoadjuvant or adjuvant chemotherapy [36].
An area of focus could include the effect of treatment duration or discontinuation. Decisions on discontinuing treatments can be made for indications such as unacceptable or intolerable adverse events, disease progression or metastasis, recurrence, or patient choice (e.g., due to financial toxicity) [37, 38]. However, given that patients have significantly prolonged survival with advanced therapies, treatment discontinuation has become ambiguous, particularly when extending these treatments to the adjuvant setting for early-stage surgically treated cancers [2, 5, 6] and long-term use of immune checkpoint inhibitors [39,40,41]. Of note, even though the maximal duration of immunotherapies was two years (35 cycles) in clinical trials [39, 40], in the real world, patients can be on treatment continuously over the period [39,40,41], mainly given the impressive duration of treatment response (e.g., median of 23.2 months in nivolumab plus ipilimumab for advanced NSCLC [42]) so that medicine authorities like the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) allow the continuation after two years. However, we have yet to see strong evidence of efficacious benefits from longer usage [39, 40], and there are concerns about health, financial, and societal toxicities [41]. These issues in treatment discontinuation reflect another under-researched area of focus – treatment deintensification or de-escalation [43, 44]. Studies should investigate the optimal duration of treatment that balances prognosis, quality of life, and financial costs to patients and the healthcare system.
Research on treatment timing may also include the order of advanced therapies before (neoadjuvant strategy), during (perioperative strategy), or after (adjuvant strategy) surgery or radiotherapy. Although several studies have started evaluating the prognostic impact of the treatment order, a clear, evidence-based consensus might yet be achieved [5]. For example, neoadjuvant versus adjuvant immunotherapy for NSCLC [5, 45], neoadjuvant versus adjuvant chemotherapy for early-stage breast cancer [46], and even the “sandwich approach” using treatment in both the neoadjuvant and adjuvant settings [47,48,49].
Of note, enriched treatment options and strategies require more considerate assessments in radiology and pathology and more frequent use of multidisciplinary meetings. Therefore, research is also needed to keep track of the length of traditional treatment intervals (including the period before the first treatment initiation), understand the current length of TTI between treatment lines, and evaluate the impact on patient experience and healthcare resourcing. We assume, given the enriched radiological and pathological assessments, the lengths of these time intervals might have yet to be improved, even longer than before. For example, a study reported a median of 21 days for biomarker testing results to be available after cancer diagnosis in patients who can receive first-line targeted therapies for advanced NSCLC [29]; the time period has been beyond the timeframe (14 days) of treatment interval between cancer diagnosis and treatment as recommended in international guidelines [50, 51]. We are concerned that many patients could still experience treatment delays [23]; at the same time, healthcare utilisation and expenditure may never decrease. These issues have been even of concern due to the COVID-19 pandemic period [52]. In addition to suggesting optimal timing and providing reasons or factors to explain the association patterns between timing and patient outcomes, research on the above focuses also brings implications of improving healthcare delivery through medical, healthcare and policy interventions.
Opportunities for clinical trials and real-world studies
The above research questions and assumptions provide new opportunities for fostering evidence of the optimal timing of cancer treatments using the existing clinical trial and real-world data. Specific to the timing between treatments, we rarely see studies in advanced treatments like neoadjuvant/adjuvant targeted therapies or immunotherapies [8, 9, 11,12,13,14,15,16,17,18,19,20,21, 35].
As previously mentioned, current RCTs normally lack evaluation on the optimal timing of treatments; this might result from the restricted study designs influenced by the research questions from medicine authorities (e.g., FDA, EMA). Relying on the completed clinical trials, however, a post hoc analysis can be conducted if participants have varying lengths of treatment intervals or treatment duration. As these data had been obtained for drug approval, reusing them for post hoc analysis should be feasible and could provide direct insights into the optimal timing of treatments. Such an analysis might allow another opportunity for re-assessing treatment efficacy, especially for superiority trials that yield non-positive results based on the pre-specified design. At the same time, RCTs can bring better records of medical reasons for treatment delays or discontinuation (e.g., details of adverse effects and comorbidities) [12, 13, 16, 31, 38], which can explain the associations between treatment timing and patient outcomes.
Limitations of the study may include poor generalisability and potentially insufficient statistical power, given the nature of RCTs. Typically, due to the inclusion and exclusion criteria for recruiting patients, the RCT sample may not represent the whole patient population in the real world [53]. At the same time, the actual number of enroled patients in RCTs is subject to the pre-specified estimate for the minimum sample size for efficacy comparison [54], as well as difficulties in enrolment as reported with no more than half (49%) of enrolment success rate [55]. These conditions may render the post hoc analysis underpowered to estimate the optimal timing, especially in understanding the non-linear associations between TTI and patient outcomes and in analyses by various patient subgroups.
Using real-world data (RWD) from electronic health records or population-based registries can address the above limitations and may provide more relevant results. Real-world studies often provide a large and diverse study sample that typically represents the target real-world population, enabling investigation of the optimal timing with large statistical power and even allowing investigation of the potential reasons and factors accounting for treatment delays or discontinuation. Real-world studies could also provide insight into the impact on healthcare utilisation. In the real world, particularly, time intervals and treatment duration should be more variable as they are more impacted by multifaceted reasons or factors at the medical, healthcare, and social levels [56]; using RWD may be potentially more relevant to investigate optimal timing, typically, demonstrating and explaining the assumed U- or J-shaped associations between treatment timing and patient outcomes. However, study limitations include the lack of accurate recording of clinical information, difficulties in constructing study cohort and key variables if lacking multiple data sources and data linkage capacity, and challenges in addressing potential biases, given the nature of administrative or health electronic data that are not designed for research [57]. These limitations might have less impact if using hospital-level data with more detailed health records instead of population-level data.
Given the feasibility of using the current clinical trial data and RWD, we call for emerging evidence of the optimal timing of cancer treatments. We envision that various studies can be conducted, but the results might be specific, since the studies for this purpose can be context-specific based on study characteristics like particular treatment options, therapeutic settings, cancer types and biomarkers, healthcare settings, or systems. Given the large variations in study characteristics, we suggest careful study methodology and detailed reporting, allowing more cautious interpretation and comparative assessment in the future [1]. Particularly, we emphasise appropriate analytic approaches given the potential of non-linear associations between treatment intervals and patient outcomes, as assumed and carefully described above. Methodological frameworks and recommendations like the Aarhus statement are valuable to guide the research design and conduct [1, 24, 56, 58, 59], especially for traditional treatment intervals on which these recommendations mainly focus. In addition to using clinical trial data and RWD independently, further studies may utilise complementary features of both data sources to develop accurate and robust timing evaluations for a target patient population routinely seen in real-world clinical practice [60]. We anticipate that studies on the optimal timing will bring new insights into using cancer treatments better, even further improving treatment efficacy.
References
Zhang J, IJzerman MJ, Emery JD. Timely cancer diagnosis and treatment: towards a generalisable research framework studying timeliness to appropriate care. Ann Cancer Epidemiol. 2023;7:3 https://doi.org/10.21037/ace-23-2.
Wang M, Herbst RS, Boshoff C. Toward personalized treatment approaches for non-small-cell lung cancer. Nat Med. 2021;27:1345–56. https://doi.org/10.1038/s41591-021-01450-2.
Garbe C, Amaral T, Peris K, Hauschild A, Arenberger P, Basset-Seguin N, et al. European consensus-based interdisciplinary guideline for melanoma. Part 2: Treatment - Update 2024. Eur J Cancer. 2025;215:115153. https://doi.org/10.1016/j.ejca.2024.115153.
Baxter NN, Kennedy EB, Bergsland E, Berlin J, George TJ, Gill S, et al. Adjuvant Therapy for Stage II Colon Cancer: ASCO Guideline Update. J Clin Oncol. 2022;40:892–910. https://doi.org/10.1200/JCO.21.02538.
Graboyes EM, Divi V, Moore BA. Head and Neck Oncology Is on the National Quality Sidelines No Longer-Put Me in, Coach. JAMA Otolaryngol Head Neck Surg. 2022;148:715–6. https://doi.org/10.1001/jamaoto.2022.1389.
Spicer JD, Cascone T, Wynes MW, Ahn MJ, Dacic S, Felip E, et al. Neoadjuvant and Adjuvant Treatments for Early Stage Resectable NSCLC: Consensus Recommendations From the International Association for the Study of Lung Cancer. J Thorac Oncol. 2024;19:1373–414. https://doi.org/10.1016/j.jtho.2024.06.010.
Graboyes EM, Garrett‐Mayer E, Sharma AK, Lentsch EJ, Day TA. Adherence to National Comprehensive Cancer Network guidelines for time to initiation of postoperative radiation therapy for patients with head and neck cancer. Cancer. 2017;123:2651–60. https://doi.org/10.1002/cncr.30651.
Alnajjar S, Shoucair S, Almanzar A, Zheng K, Lisle D, Gupta V. Predictors of Timely Initiation and Completion of Adjuvant Chemotherapy in Stage II/III Colorectal Adenocarcinoma. Am Surg. 2024;90:2724–32. https://doi.org/10.1177/00031348241248689.
Ramsden K, Laskin J, Ho C. Adjuvant Chemotherapy in Resected Stage II Non-small Cell Lung Cancer: Evaluating the Impact of Dose Intensity and Time to Treatment. Clin Oncol R Coll Radio. 2015;27:394–400. https://doi.org/10.1016/j.clon.2015.03.001.
Tørring ML, Frydenberg M, Hamilton W, Hansen RP, Lautrup MD, Vedsted P. Diagnostic interval and mortality in colorectal cancer: U-shaped association demonstrated for three different datasets. J Clin Epidemiol. 2012;65:669–78. https://doi.org/10.1016/j.jclinepi.2011.12.006.
Okines AF, Kipps E, Irfan T, Coakley M, Angelis V, Asare B, et al. Impact of timing of adjuvant chemothapy for early breast cancer: the Royal Marsden Hospital experience. Br J Cancer. 2021;125:299–304. https://doi.org/10.1038/s41416-021-01428-4.
Nilsson K, Klevebro F, Sunde B, Rouvelas I, Lindblad M, Szabo E, et al. Oncological outcomes of standard versus prolonged time to surgery after neoadjuvant chemoradiotherapy for oesophageal cancer in the multicentre, randomised, controlled NeoRes II trial. Ann Oncol. 2023;34:1015–24. https://doi.org/10.1016/j.annonc.2023.08.010.
Nilsson K, Klevebro F, Rouvelas I, Lindblad M, Szabo E, Halldestam I, et al. Surgical Morbidity and Mortality From the Multicenter Randomized Controlled NeoRes II Trial: Standard Versus Prolonged Time to Surgery After Neoadjuvant Chemoradiotherapy for Esophageal Cancer. Ann Surg. 2022;272:684–9. https://doi.org/10.1097/SLA.0000000000004340.
Kang KM, Hong KS, Noh GT, Oh BY, Chung SS, Lee RA, et al. Optimal time of initiating adjuvant chemotherapy after curative surgery in colorectal cancer patients. Ann Coloproctol. 2013;29:150–4. https://doi.org/10.3393/ac.2013.29.4.150.
Maeng CH, Kim H, Kim M. Time interval between surgery and adjuvant chemotherapy in patients with gastric cancer after gastrectomy: a population-based cohort study using a nationwide claim database. Ther Adv Med Oncol. 2024;16:17588359241241972. https://doi.org/10.1177/17588359241241972.
Gögenur M, Rosen AW, Iveson T, Kerr RS, Saunders MP, Cassidy J, et al. Time From Colorectal Cancer Surgery to Adjuvant Chemotherapy: Post Hoc Analysis of the SCOT Randomized Clinical Trial. JAMA Surg. 2024;159:865–71. https://doi.org/10.1001/jamasurg.2024.1555.
Dumont F, Kepenekian V, De Franco V, Eveno C, Rat P, Sabbagh C, et al. Delaying Surgery After Neoadjuvant Chemotherapy Affects Survival in Patients with Colorectal Peritoneal Metastases: A BIG-RENAPE Network Multicentric Study. Ann Surg Oncol. 2023;30:3549–59. https://doi.org/10.1038/10.1245/s10434-023-13224-w.
Ho AS, Kim S, Tighiouart M, Mita A, Scher KS, Epstein JB, et al. Quantitative survival impact of composite treatment delays in head and neck cancer. Cancer. 2018;124:3154–62. https://doi.org/10.1002/cncr.31533.
Salazar MC, Rosen JE, Wang Z, Arnold BN, Thomas DC, Herbst RS, et al. Association of Delayed Adjuvant Chemotherapy With Survival After Lung Cancer Surgery. JAMA Oncol. 2017;3:610–9. https://doi.org/10.1001/jamaoncol.2016.5829.
Cao L, Xu C, Wang MD, Qi WX, Cai G, Cai R, et al. Influence of Adjuvant Radiotherapy Timing on Survival Outcomes in High-Risk Patients Receiving Neoadjuvant Treatments. Front Oncol. 2022;12:905223. https://doi.org/10.3389/fonc.2022.905223.
Matsuo K, Shimada M, Matsuzaki S, Enomoto T, Mikami M. Wait-time for adjuvant radiotherapy and oncologic outcome in early-stage cervical cancer: A treatment implication during the coronavirus pandemic. Eur J Cancer. 2021;148:117–20. https://doi.org/10.1016/j.ejca.2021.02.013.
Neal RD, Tharmanathan P, France B, Din NU, Cotton S, Fallon-Ferguson J, et al. Is increased time to diagnosis and treatment in symptomatic cancer associated with poorer outcomes? Systematic review. Br J Cancer. 2015;112:S92–S107. https://doi.org/10.1038/bjc.2015.48.
Zhang J, IJzerman MJ, Oberoi J, Karnchanachari N, Bergin RJ, Franchini F, et al. Time to diagnosis and treatment of lung cancer: A systematic overview of risk factors, interventions and impact on patient outcomes. Lung Cancer. 2022;166:27–39. https://doi.org/10.1016/j.lungcan.2022.01.015.
Drosdowsky A, Lamb KE, Bergin RJ, Boyd L, Milley K, IJzerman MJ, et al. A systematic review of methodological considerations in time to diagnosis and treatment in colorectal cancer research. Cancer Epidemiol. 2023;83:102323. https://doi.org/10.1016/j.canep.2023.102323.
Drosdowsky A, Lamb KE, Karahalios A, Bergin RJ, Milley K, Boyd L, et al. The effect of time before diagnosis and treatment on colorectal cancer outcomes: systematic review and dose-response meta-analysis. Br J Cancer. 2023;129:993–1006. https://doi.org/10.1038/s41416-023-02377-w.
Castelo M, Paszat L, Hansen BE, Scheer AS, Faught N, Nguyen L, et al. Analysis of time to treatment and survival among adults younger than 50 years of age with colorectal cancer in Canada. JAMA Netw Open. 2023;6:e2327109. https://doi.org/10.1001/jamanetworkopen.2023.27109.
Lee S, Fujita K, Negoro E, Morishita T, Yamauchi H, Oiwa K, et al. The impact of diagnostic wait time on the survival of patients with diffuse large B‐cell lymphoma: effect modification by the International Prognostic Index. Br J Haematol. 2019;187:195–205. https://doi.org/10.1111/bjh.16078.
Duckett KA, Kassir MF, Nguyen SA, Brennan EA, Chera BS, Sterba KR, et al. Factors Associated with Head and Neck Cancer Postoperative Radiotherapy Delays: A Systematic Review and Meta-analysis. Otolaryngol Head Neck Surg. 2024;171:1265–82. https://doi.org/10.1002/ohn.835.
Stricker T, Jain N, Ma E, Yu E, Wang R, Schuldt R, et al. Clinical Value of Timely Targeted Therapy for Patients With Advanced Non-Small Cell Lung Cancer With Actionable Driver Oncogenes. Oncologist. 2024;29:534–42. https://doi.org/10.1093/oncolo/oyae022.
Bailey CE, Parikh AA. Assessment of the risk of antiangiogenic agents before and after surgery. Cancer Treat Rev. 2018;68:38–46. https://doi.org/10.1016/j.ctrv.2018.05.002.
Wang C, Hu X, Yang L, Xu Y, Zheng B, Yang J, et al. Anlotinib versus placebo as adjuvant therapy for localized high-grade soft tissue sarcomas: a phase 2, double-blinded, randomized controlled trial. Clin Cancer Res. 2025;31:1194–203. https://doi.org/10.1158/1078-0432.CCR-24-2531.
Madani I, De Neve W, Mareel M. Does ionizing radiation stimulate cancer invasion and metastasis? Bull Cancer. 2008;95:292–300. https://doi.org/10.1684/bdc.2008.0598.
Kuonen F, Secondini C, Rüegg C. Molecular pathways: emerging pathways mediating growth, invasion, and metastasis of tumors progressing in an irradiated microenvironment. Clin Cancer Res. 2012;18:5196–202. https://doi.org/10.1158/1078-0432.CCR-11-1758.
Verginadis II, Citrin DE, Ky B, Feigenberg SJ, Georgakilas AG, Hill-Kayser CE, et al. Radiotherapy toxicities: mechanisms, management, and future directions. Lancet. 2025;405:338–52. https://doi.org/10.1016/S0140-6736(24)02319-5.
Shapiro J, van Hagen P, Lingsma HF, Wijnhoven BP, Biermann K, ten Kate FJ, et al. Prolonged time to surgery after neoadjuvant chemoradiotherapy increases histopathological response without affecting survival in patients with esophageal or junctional cancer. Ann Surg. 2014;260:807–13. https://doi.org/10.1097/SLA.0000000000000966.
Steventon L, Kipps E, Man KK, Roylance R, Forster MD, Wong IC, et al. The impact of inter-cycle treatment delays on 5-year all-cause mortality in early-stage breast cancer: A retrospective cohort study. Eur J Cancer. 2024;210:114301. https://doi.org/10.1016/j.ejca.2024.114301.
Virtanen S, Pihlman H, Silvoniemi M, Vihinen P, Jaakkola P, Mattila KE. Reasons for Treatment Discontinuation and Their Effect on Outcomes of Immunotherapy in Southwest Finland: A Retrospective, Real-World Cohort Study. Cancers. 2024;16:709. https://doi.org/10.3390/cancers16040709.
Yang LZ, He Q, Zhang J, Ganti AK, Stinchcombe TE, Pang H, et al. Characteristics of toxicity occurrence patterns in concurrent chemoradiotherapy after induction chemotherapy for patients with locally advanced non-small cell lung cancer: a pooled analysis based on individual patient data of CALGB/Alliance trials. Transl Cancer Res. 2022;11:3506–21. https://doi.org/10.21037/tcr-22-2006.
Sun L, Bleiberg B, Hwang WT, Marmarelis ME, Langer CJ, Singh A, et al. Association Between Duration of Immunotherapy and Overall Survival in Advanced Non-Small Cell Lung Cancer. JAMA Oncol. 2023;9:1075–82. https://doi.org/10.1001/jamaoncol.2023.1891.
Rousseau A, Michiels S, Simon-Tillaux N, Lolivier A, Bonastre J, Planchard D, et al. Impact of pembrolizumab treatment duration on overall survival and prognostic factors in advanced non-small cell lung cancer: a nationwide retrospective cohort study. Lancet Reg Health Eur. 2024;43:100970. https://doi.org/10.1016/j.lanepe.2024.100970.
Marron TU, Ryan AE, Reddy SM, Kaczanowska S, Younis RH, Thakkar D, et al. Considerations for treatment duration in responders to immune checkpoint inhibitors. J Immunother Cancer. 2021;9:e001901. https://doi.org/10.1136/jitc-2020-001901.
Hellmann MD, Paz-Ares L, Bernabe Caro R, Zurawski B, Kim SW, Carcereny Costa E, et al. Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N Engl J Med. 2019;381:2020–31. https://doi.org/10.1056/NEJMoa1910231.
Soon JA, Franchini F, IJzerman MJ, McArthur GA. Leveraging the potential for deintensification in cancer care. Nat Cancer. 2024. https://doi.org/10.1038/s43018-024-00827-9.
Remon, J, Bortolot, M, Bironzo, P, Cortiula, F, Menis, J, Brandao, M, et al. De-Escalation Strategies With Immune Checkpoint Blockers in Non-Small Cell Lung Cancer: Do We Already Have Enough Evidence? J Clin Oncol. 2025;JCO2402347. https://doi.org/10.1200/JCO-24-02347.
John AO, Ramnath N. Neoadjuvant Versus Adjuvant Systemic Therapy for Early-Stage Non-Small Cell Lung Cancer: The Changing Landscape Due to Immunotherapy. Oncologist. 2023;28:752–64. https://doi.org/10.1093/oncolo/oyad125.
Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of individual patient data from ten randomised trials. Lancet Oncol. 2018;19:27–39. https://doi.org/10.1016/S1470-2045(17)30777-5.
Kang YK, Yook JH, Park YK, Lee JS, Kim YW, Kim JY, et al. PRODIGY: A Phase III Study of Neoadjuvant Docetaxel, Oxaliplatin, and S-1 Plus Surgery and Adjuvant S-1 Versus Surgery and Adjuvant S-1 for Resectable Advanced Gastric Cancer. J Clin Oncol. 2021;39:2903–13. https://doi.org/10.1200/JCO.20.02914.
Spicer JD, Garassino MC, Wakelee H, Liberman M, Kato T, Tsuboi M, et al. Neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab compared with neoadjuvant chemotherapy alone in patients with early-stage non-small-cell lung cancer (KEYNOTE-671): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2024;404:1240–52. https://doi.org/10.1016/S0140-6736(24)01756-2.
Patel SP, Othus M, Chen Y, Wright GP Jr, Yost KJ, Hyngstrom JR, et al. Neoadjuvant-Adjuvant or Adjuvant-Only Pembrolizumab in Advanced Melanoma. N Engl J Med. 2023;388:813–23. https://doi.org/10.1056/NEJMoa2211437.
Cancer Council Victoria and Department of Health Victoria. Optimal care pathway for people with lung cancer (2nd edition). Cancer Council Victoria. 2021. https://www.cancer.org.au/assets/pdf/lung-cancer-optimal-cancer-care-pathway. Accessed on 26 January 2025.
England National Health Service (NHS). Implementing a timed lung cancer diagnostic pathway: A handbook for local health and care systems. NHS Rapid cancer diagnostic and assessment pathways. 2023. https://www.england.nhs.uk/long-read/implementing-a-timed-lung-cancer-diagnostic-pathway. Accessed on 26 January 2025.
Shah R, Hanna NM, Loo CE, David M, Mafra A, Fink H, et al. The global impact of the COVID-19 pandemic on delays and disruptions in cancer care services: a systematic review and meta-analysis. Nat Cancer. 2025;6:194–204. https://doi.org/10.1038/s43018-024-00880-4.
Tan YY, Papez V, Chang WH, Mueller SH, Denaxas S, Lai AG. Comparing clinical trial population representativeness to real-world populations: an external validity analysis encompassing 43 895 trials and 5 685 738 individuals across 989 unique drugs and 286 conditions in England. Lancet Healthy Longev. 2022;3:e674–e689. https://doi.org/10.1016/S2666-7568(22)00186-6.
Wang X, Ji X. Sample Size Estimation in Clinical Research: From Randomized Controlled Trials to Observational Studies. Chest. 2020;158:S12–S20. https://doi.org/10.1016/j.chest.2020.03.010.
Zhang S, Zhang J, Liu S, Pang H, Stinchcombe TE, Wang X. Enrollment Success, Factors, and Prediction Models in Cancer Trials (2008–19). JCO Oncol Pr. 2023;19:1058–68. https://doi.org/10.1200/OP.23.00147.
Walter F, Webster A, Scott S, Emery J. The Andersen Model of Total Patient Delay: a systematic review of its application in cancer diagnosis. J Health Serv Res Policy. 2012;17:110–8. https://doi.org/10.1258/jhsrp.2011.010113.
Zhang K, Wang D, Zhang J. How to optimize real-world study: concept, opportunities, and evidence quality. Transl Breast Cancer Res. 2020;1:12. https://doi.org/10.21037/tbcr-20-30.
Weller D, Vedsted P, Rubin G, Walter FM, Emery J, Scott S, et al. The Aarhus statement: improving design and reporting of studies on early cancer diagnosis. Br J Cancer. 2012;106:1262–7. https://doi.org/10.1038/bjc.2012.68.
Olesen F, Hansen RP, Vedsted P. Delay in diagnosis: the experience in Denmark. Br J Cancer. 2009;101:S5–S8. https://doi.org/10.1038/sj.bjc.6605383.
Lee D, Yang S, Berry M, Stinchcombe T, Cohen HJ, Wang X. genRCT: a statistical analysis framework for generalizing RCT findings to real-world population. J Biopharm Stat. 2024;1–20. https://doi.org/10.1080/10543406.2024.2333136.
Funding
JZ is a recipient of the Melbourne Research Scholarship, funded by the University of Melbourne. Open Access funding enabled and organized by CAUL and its Member Institutions.
Author information
Authors and Affiliations
Contributions
JZ wrote the first draft, which was critically revised by RV, XW and QH. All authors approved the final paper.
Corresponding author
Ethics declarations
Competing interests
JZ declares that the conceptualisation and raised assumptions are inspired by his PhD research and involvement in the ongoing work updating the Arhus statement. However, both works mainly focus on the time to cancer diagnosis and treatment from primary care instead of the timing within or between treatments on which this paper primarily focuses. The rest of the authors declare no conflict of interests.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Zhang, J., Venchiarutti, R., Wang, X. et al. Optimal timing of cancer treatments: a call for emerging evidence from clinical trials and real-world studies. Br J Cancer 132, 1085–1090 (2025). https://doi.org/10.1038/s41416-025-03030-4
Received:
Revised:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41416-025-03030-4