Intermittent afatinib treatment suppresses the growth of resistant T790M-H1975 cells in non-small cell lung cancer (NSCLC) co-culture
-
Amir Imran Faisal Hamdi
, Wen Tsin Poh , Jonathan Chee Woei Lim , Ummi Nadira Daut , Soon Hin How , Yong Kek Pang and Johnson Stanslas
Abstract
Objectives
Most non-small-cell lung cancers (NSCLC) that harbour an epidermal growth factor receptor (EGFR) mutation are treated with afatinib. Resistance emerges in most patients, making treatment ineffective. Reports suggest intermittent treatment (IT) delays the emergence of resistance compared to standard-of-care continuous treatment (CT). Thus, this study aimed to investigate the efficacy of afatinib through both treatments in NSCLC in vitro.
Methods
The growth inhibition of afatinib was evaluated in mixed co-culture lines of HCC827 (sensitising with exon 19 deletion) and H1975 (resistant with L858R/T790M), with resistant percentages of 0.1% and 0.5 % treated with afatinib at 100 and 500 pM (pM). CT was treated for 96 h, while IT was treated for 24 h and was treatment-free for 72 h. They were assessed at 192 h with cell counting and qPCR with cell line-specific primers targeting exon 19 deletion and L858R, followed by images of fluorescent resistance.
Results
At 96 and 192 h, IT had a higher cell count than CT in both 0.1 % and 0.5 % resistance at 100 and 500 pM (p<0.05). The qPCR analysis showed the gene expression for resistant cells in IT was lower than in CT (p<0.05). However, nanomolar concentrations in patients’ pharmacokinetics resulted in resistance dominance and progression.
Conclusions
Overall, in both 0.1% and 0.5 % resistance in co-cultures, intermittent treatment allows a significant portion of viable sensitive cells to suppress the growth of resistant cells at concentrations of both 100 and 500 pM. Collectively, IT delays the emergence of de novo resistance with low resistance at specific concentrations. These findings offer the potential for utilising alternative treatment strategies as opposed to continuous treatment to improve therapeutic outcomes in NSCLC.
Introduction
The discovery of tyrosine kinase inhibitors (TKIs) that selectively target activating mutations in the protein kinase epidermal growth factor receptor (EGFR) led to a breakthrough in the treatment of non-small cell lung cancer (NSCLC) [1], [2], [3], [4]. EGFR-TKIs target common activating mutations, such as the exon 19 deletion (E746_A750del) and L858R, which show clinical benefit in the majority of patients with EGFR-mutated NSCLC when administered continuously [5], 6]. However, resistance invariably develops, with the emergence of de novo (pre-existing) T790M resistance reported in approximately half of the treated patients [7]. Previously, in NSCLC, the variant allelic frequency (VAF) of de novo T790M was almost absent in diagnosed patients with predominant common activating mutations [8], [9], [10]. This notion coincides with older detection technologies such as polymerase chain reaction (PCR) clamp, scorpion amplified refractory mutation system methods, and direct sequencing [11], 12]. However, as detection technology advances over time, the prevalence of de novo T790M mutation increases with the aid of better sensitivity in detection methods, which include matrix-assisted laser desorption ionisation-time of flight mass spectrometry (MALDI-TOF MS) [7], 13], TaqMan assay [14], 15], colony hybridisation [16], and peptide nucleic acid (PNA)-clamping PCR [17], 18]. Digital [19], droplet digital PCR, and next-generation sequencing [20], 21] were able to have an analytical sensitivity of approximately 0.001 %, which detected 66 % of the overall incidence of ultra-low de novo T790M [22], 23].
An emerging body of evidence has suggested that intermittent dosing schedules with elongated treatment-free periods might have advantages in delaying the emergence of resistance over continuous treatment [24], [25], [26]. Preclinical studies with H3255 lung cancer xenografts showed that intermittent weekly gefitinib dosing showed better therapeutic outcomes than continuous dosing [27]. Multiple case reports have detailed how allowing an elongated treatment-free period in a dosing regimen prolongs progression-free survival with reduced side effects in erlotinib [28] and afatinib [29], 30].
The success of intermittent treatment is supported by a cell-cell competition treatment regimen called “adaptive therapy.” This concept emphasises a significant proportion of viable treatment-sensitive cells to allow cellular competition with de novo resistant cells for nutrients and space during the treatment-free period [31]. The application of this treatment regimen is aimed at being patient-specific, as tumour growth and heterogeneity vary between individuals. Thus, it is one of the biggest hurdles to having it in the clinical setting. Multiple reports of the successes of adaptive therapy come from mathematical modelling and simulations to predict the treatment outcome [32], 33]. Under adaptive therapy, the same tumour could be managed with an average of just 35 % of the maximum-tolerated dose and a total dose that was 70 % of that given with continuous treatment [34]. In addition, the adaptive therapy cycling demonstrated that control may be durably maintained for up to 20 cycles, which is significantly longer than continuous treatment [35]. Although it is understood that intermittent treatment is shown to be therapeutically inferior to continuous treatment, it is unknown whether optimising intermittent treatment with adaptive therapy principles can improve the treatment regimen.
In the present study, we used an in vitro strategy to examine the effect of afatinib with a treatment-free period in intermittent treatment to spare a significant proportion of sensitive cells and delay de novo resistant cell lines.
Materials and methods
Chemicals and reagents
Fetal bovine serum (FBS), ethylenediaminetetraacetic acid (EDTA), and cell culture-grade dimethyl sulfoxide (DMSO) were purchased from Sigma-Aldrich (MO, USA). Roswell Park Memorial Institute (RPMI) 1640 Medium (with l-glutamine and phenol red), 2.5 % trypsin, and 10,000 U/mL penicillin–10 mg/mL streptomycin (pen-strep) were purchased from Gibco (NY, USA); phosphate-buffered saline (PBS) tablets were purchased from Invitrogen (CA, USA); 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) was obtained from Molecular Probes (OR, USA); whereas the analytical-grade DMSO was purchased Fisher Scientific (NH, USA). Ultrapure water was obtained using Merck Millipore’s Milli-Q water purification system from Merck KGaA, Darmstadt, Germany.
Test compounds
Afatinib was purchased from Cayman Chemical (purity ≥95 %, Item No.: 11492; MI, USA) in a lyophilised form. A stock solution of 100 mM for each test compound was made up of cell culture-grade DMSO, aliquoted, and stored at −20 °C until further use. All test compounds were diluted using the corresponding media to different concentrations according to the experimental design.
Cell lines and cell culture
HCC827 (CRL-2868) and NCI-H1975 (CRL-5908) human NSCLC cell lines were purchased from American Type Culture Collection (VA, USA). The HCC827 cell line has the presence of the sensitising mutation, exon 19 deletion (E746-A750 deletion), and the absence of the resistance mutation EGFR T790M. Conversely, H1975 has the sensitising mutation L858R and the resistance mutation EGFR T790M. Thus, HCC827 will be denoted as sensitive cells and H1975 as resistant cells. All cell culture procedures were carried out in sterile conditions, and aseptic techniques were applied. All cell lines were grown in complete growth media (CGM) described as follows: both cell lines were grown in RPMI 1640 and supplemented with 10 % heat-inactivated cc and 1 % penicillin. The cells were maintained at 37 °C in a humidified atmosphere containing 5 % CO2 and 95 % air. Cell morphology was observed under an inverted phase-contrast microscope (Carl Zeiss Microscopy GmbH, Jena, Germany). These cell lines are authenticated for the absence of mycoplasma by Universal Mycoplasma Detection Kit - 30–1012K, ATCC.
In vitro growth inhibitory assay
Briefly, cells were seeded in 96-well flat-bottomed cell culture plates, with 2000 cells per well. Following incubation overnight at 37 °C for cell attachment, the cells were treated with each test compound at final concentrations ranging from 0.1 to 100.0 µM, with 20 µL of each concentration added into appropriate wells in replicates of four. The control cells were treated with 0.01 % DMSO, equivalent to the highest amount of DMSO used as a vehicle in the compound-treated wells after 96 h of incubation at 37 °C. Growth inhibition was calculated as a percentage relative to vehicle-treated wells using the MTT (3-[4,5-Dimethylthiazol-2-yl]-2,5-Diphenyltetrazolium Bromide) assay by Invitrogen (CA, USA). Absorbance was measured at 550 nm using a VersaMax microplate reader with Molecular Devices (CA, USA) with SoftMax® Pro software (version 5.4). Semi-log dose-response growth curves were constructed by plotting the percent of cell growth against the test concentrations, from which the concentration that inhibits cell growth by 50 % (IC50) was determined.
Response of the co-culture of sensitive and resistant cells to afatinib
The experimental design in co-culture containing 0.1 and 0.5 % resistance was carried out with continuous therapy of 100 and 500 pM afatinib up to 96 and 192 h to represent 2 treatment cycles. Meanwhile, in intermittent therapy, co-cultures were exposed to concentrations of 100 and 500 pM afatinib for 24 h and replaced with treatment-free media for 72 h. The process was repeated after 96 h of design, as shown in Figure 1. Following that, the pharmacokinetic calculation from the area under the curve (AUC) to experimental concentrations was done with Equation (1). The AUC has been extrapolated to 96 h via PharmaCalc (Zurich, Switzerland).

Illustration representation of the experimental setup for continuous and intermittent treatments.
Viable cell counting technique
Viable cell counting was performed using the Trypan Blue exclusion method with a hemocytometer at 96 and 192 h. Cells were harvested, centrifuged, and resuspended in PBS or culture medium to form a uniform suspension. An equal volume of 0.4 % Trypan Blue was mixed with the cell suspension and incubated briefly to distinguish viable (unstained) from non-viable (blue-stained) cells. A small volume of the mixture was loaded into a hemocytometer, and cells were counted under a ZEISS Primo Vert inverted phase-contrast microscope with monitor (Carl Zeiss Microscopy GmbH, Jena, Germany) in four quadrants. The viable cell concentration was calculated using a standard formula, factoring in dilution and chamber volume, and cell viability was expressed as a percentage of total cells. Counts were repeated across multiple chambers to ensure accuracy and reproducibility.
Quantitative polymerase chain reaction
RNA extraction was performed using the Monarch® Total RNA Miniprep Kit (MA, USA), and the RNA was measured with an ND-1000 spectrophotometer from Thermo Fisher Scientific (MA, USA). The concentration and purity of the extracted RNA were determined according to the instructions of the LunaScript® RT SuperMix Kit for quantitative PCR (qPCR). The extracted RNA was reverse transcribed into cDNA, and Luna® Universal qPCR Master Mix reagent was used for PCR on the Mastercycler® ep realplex Real-time PCR System from Eppendorf (CT, USA). The following steps were performed under the following conditions: first, pre-denaturation at 95 °C for 30 s, then 95 °C for 10 s, 60 °C for 30 s, and 72 °C for 45 s, for a total of 40 cycles. The primer sets were shown in Supplementary Table S1.
Fluorescent-labelled resistant cells
Resistant cells were fluorescently labelled in 0.1 % resistance co-cultures and were grown in a 6-well plate with a separate microscope glass cover pre-added to it. Resistant cells were fluorescently labelled with CellTracker™ orange CMRA dye from Thermo Fisher Scientific (MA, USA) as per the manufacturer’s protocol, followed by mixing with sensitive cells to make a 0.1 % resistance co-culture for seeding. Upon imaging, a microscope glass cover was taken out of each well and fixed by cross-linking with a 4 % paraformaldehyde solution (diluted in PBS). Incubation was done for 10–20 min at room temperature. The microscope glass cover was placed on top of a microscopic cover glass and viewed under selected filters using Olympus bx51 fluorescent microscope (Tokyo, Japan).
Statistical analysis
All experiments were performed in triplicate. Each data point was presented as an average value with the corresponding standard deviation. Statistical analysis was performed using SPSS version 28 software for Windows (IBM Corp., Armonk, NY, USA). All assays used one-way ANOVA with Tukey’s post hoc test to determine significant differences between the control and treatment groups. Student’s t-test was used to determine significant differences between treatment groups (continuous and intermittent treatments). A p-value of less than 0.05 was considered statistically significant.
Results
In the absence of afatinib, both cell lines were evaluated for their proliferation rate (Figure 2), followed by the calculated doubling time listed as Equation (2). In Table 1, the mean doubling time of the HCC827 was longer than that of the H1975, indicating that resistant cells (H1975) proliferate faster than sensitive cells (HCC827) by approximately 10 h.

Growth kinetics of EGFR-mutant NSCLC cell lines over 96 h. Cell proliferation of HCC827 (afatinib-sensitive) and NCI-H1975 (afatinib-resistant) cell lines was monitored over 96 h. Graphs represent the average of triplicate wells ±SD.
Calculated doubling time for both HCC827 and H1975 cells.
| Cell line | Doubling time, hoursa | p-Value |
|---|---|---|
| H1975 | 28.83 ± 1.20 | <0.05 |
| HCC827 | 39.44 ± 2.40 |
-
aThe values are presented in mean ± SD (n=3).
The sensitivity of sensitive, resistant, and mixed co-cultures with varying resistance percentages to afatinib was identified in Figure 3 via MTT assay as described in the Section “In vitro growth inhibitory assay”. At 50 % growth inhibition, the fold resistance between the two was calculated to be more than 1000-fold (Table 2). This is evident from the presence of the EGFR T790M mutation in the H1975 cell line. The percentage of resistant cells in the co-culture is necessary to mimic the heterogeneous tumour sub-populations in patients. This ensures the efficacy of continuous and intermittent treatment cycles. Resistant cells were mixed with sensitive cells at a known percentage, and the cell mixture’s drug sensitivity was measured by growth inhibition assays (Figure 3). Co-cultures with resistance percentages of 0.1 and 1.0 displayed the closest sensitivity to pure sensitive cells (0 %), whereas sensitivity was gradually reduced when resistant cells made up >10 % of the population. Therefore, monitoring the progression of resistant cells with subsequent experiments was done with resistance percentages of 0.1% and 0.5 %. Additionally, this low resistance percentage has been documented to be present in metastatic prostate cancer patients [6], 9]. The in vitro resistance percentages of 0.1% and 0.5 % were selected to mimic the clinical real-world tumour composition diagnosed in tumour-sensitive patients harbouring minute drug-resistant cells.

Dose-response curve with varying resistance percentages to afatinib. Mixed populations of cells were treated with increasing concentrations of afatinib for 96 h, at which point growth inhibition was measured. The total cell number was determined after 96 h and graphed as the percent growth compared to the DMSO control ± SD.
IC50 of mixed co-culture with afatinib.
| Resistance percentage, % | IC50, pM |
|---|---|
| 0 | 300 |
| 0.1 | 500 |
| 1 | 1,000 |
| 10 | 10,500 |
| 25 | 14,000 |
| 50 | 1,00,000 |
| 100 | 1,25,000 |
The concentrations used were kept below 500 pM to observe the role of sensitive cells in competing with resistant cells. Thus, according to Figure 3, specific final concentrations of 100 and 500 pM were used alongside the resistance as mentioned above percentages in continuous and intermittent treatments. It is important to note that in Figure 4A and B, the cell count for continuous treatment at 100 pM is almost similar to that for intermittent treatment at 500 pM. This suggests that under these conditions, equal drug efficacy can be achieved, as reported in case reports, particularly in high-dose intermittent treatment regimens in clinics [36], 37]. In both resistance percentages, it was observed that the treatment exposure of 96 h in intermittent treatment significantly (p<0.05) sustained a higher viable cell count than continuous treatment in the concentration of 100 pM. A similar trend was observed for 500 pM but with fewer viable cells. In 192 h, the growth trend for continuous and intermittent treatments at 100 and 500 pM was similarly replicated in 96 h, with more cell growth. This implied that at these concentrations, afatinib only slows the growth of the cells rather than killing them.

Cell growth of co-culture cells treated with DMSO control (0.01 %), continuous and intermittent treatments for 100 and 500 pM in 96 and 192 h. (A) 0.1 %, and (B) 0.5 % resistance. A similar growth pattern was shown for both 0.1% and 0.5 % resistance, with intermittent treatment having a higher cell count than continuous treatment across 96 hours and 192 h. Data are shown as means ± SD of triplicate cultures of three independent experiments. Significant differences between control and treatment groups were evaluated using one-way ANOVA, and significant differences between treatment groups (continuous and intermittent) were evaluated using an independent sample t-test (*p<0.05).
To quantify the respective sensitive and resistant levels in the treated cell counts in Figure 4A and B, qPCR was used with cell line-specific primers that target the gene of interest, which is indistinguishable between both cell types, namely, exon 19 deletion in sensitive cells and L858R in T790 M-resistant cells. Although the primer sets were designed from other publications (Supplementary Table S1) [38], optimisation was done with evident similarity in both cell numbers increasing linearly with total RNA extracted, followed by a standard curve with specific primers targeting cell line-specific cDNA sequences showing R-values of more than 0.99 for both cell lines (Supplementary Figure S1). Both Figure 5A and B showed similar expression trends, with genes of interest for sensitive cells all below normalised control, in contrast to resistant cells, which were all above normalised control. In Figure 5A and B, upon normalisation at 96 h of DMSO control (0.01 %), the gene expression for sensitive cells reduces gradually with increasing concentration. This correlates with a gradual increase in resistant cells. This trend was similarly replicated between the two gene expressions at 192 h, which is statistically significant compared to the DMSO control. In intermittent treatment, at 96 h, gene expression for sensitive cells is higher, while, interestingly, the expression for resistant cells in intermittent treatment is lower than in continuous treatment at both concentrations. This trend is replicated at 192 h, with the gene expression of resistant cells in intermittent treatment being lower than in continuous treatment, which is statistically significant (p-value < 0.05).

Gene expression analysis of co-culture cells treated with DMSO and both continuous and intermittent treatments at 100 and 500 pM in 96 and 192 h. (A) 0.1 %, and (B) 0.5 % resistance. The PCR product for sensitive cells is the exon 19 deletion, and the PCR product for resistant cells is L858R. Significant differences between control and treatment groups were evaluated using one-way ANOVA, and significant differences between treatment groups (continuous and intermittent) were evaluated using an independent sample t-test (ns, no significance; *p<0.05).
While it has been shown that gene expression for resistant cells in intermittent treatment is lower than in continuous treatment, resistant cells were fluorescently labelled in a 0.1 % resistance co-culture to observe the cellular growth pattern and to support the qPCR data. The treatment period lasted only 96 h because the fluorescent signals would become even lower beyond this period. Images in Figure 6A show the proliferation of the resistant cells directly in response to the treatment regimen used in this study. In continuous treatment, fluorescently labelled resistant cells proliferate with less sensitive cells surrounding them, while a growing cluster of resistant cells was “caged” in intermittent treatment, showing a slight difference in morphology for resistant cells in both treatments. Both fluorescent cell count (Figure 6B) and fluorescent total co-culture read (Figure 6C) showed that fluorescent resistant cells were higher in continuous treatment as compared to intermittent treatment, and the efficacy of growth inhibition posed by intermittent treatment was calculated to inhibit 50.74 % growth resistance as compared to continuous treatment (Figure 6D).

Fluorescently labelled resistant cells analysis (labelled with CellTracker™ Orange CMRA) in 0.1 % resistance co-culture. (A) Fluorescent images were captured at 100 × magnification at 96 h. Brightfield images were visualised by an inverted phase-contrast microscope. (B) Total cell count of resistant cells at 96 h (labelled with CellTracker™ Orange CMRA). The fluorescent cell count was done from the top left, top right, middle, bottom left, and bottom right to obtain a representative count of the total co-culture from the microscope glass cover per well. (C) Total fluorescence read at 96 h. The absorbance of the cell lysate was measured using a fluorescent microplate reader. (D) The calculated resistance inhibition efficacy is achieved by intermittent suppression of the growth of resistant cells. Significant differences between control and treatment groups were evaluated using one-way ANOVA, and significant differences between treatment groups (continuous and intermittent) were evaluated using an independent sample t-test (*p<0.05).
Although qPCR analysis and fluorescent analysis showed that intermittent treatment has been shown to delay the emergence of resistance over 192 h, the concentrations of 100 and 500 pM are not what is being experienced in patients. To emulate the plasma drug concentration in patients, the pharmacokinetic profile of afatinib in 24 h was reported as the area under the plasma concentration-time curve (AUC) [39]. The AUC has been extrapolated to 96 h to make the treatment regimen change from once daily (continuous treatment) to once every 4 days (intermittent treatment) (Figure 6). The extrapolated AUC was converted to experimental concentrations in nanomolar according to Equation (1). The experimental plan of both continuous and intermittent treatments was remodelled with high concentrations to analyse their efficacy with pharmacokinetic parameters (Figure 7). While the treatment-free period from the previous experimental setup in Figure 2 was replaced with drug concentrations, this supports the residual concentration in the plasma drug during the clearance period in the pharmacokinetics of afatinib.

Extrapolated plasma concentration-time curve of afatinib in 96 h using PharmaCalc (Zurich, Switzerland). (Refer to Equation (2)).
Cell count analysis revealed a significant reduction in the number of viable cells between 0 and 96 h. Interestingly, the viable cell count increased from 96 to 192 h for both 0.1% and 0.5 % resistance percentages in continuous and intermittent treatments based on Figure 8. From 0 to 192 h, intermittent treatment sustained a higher cell count than continuous treatment (Figure 9A). In the qPCR analysis, intermittent treatment showed higher gene expression for resistant cells as compared to continuous treatment at 96 h, and it is significantly replicated at 192 h for both resistance percentages, 0.1% and 0.5 % (Figure 9B). A statistically significant reduction of approximately 85 % of viable cells was observed for both 0.1 and 0.5 % resistance at 96 h, followed by an increase in viable cells at 192 h, particularly for 0.5 % resistance (Figure 9A). The gene expression of sensitive cells reduces over time, reaching approximately a 50 % reduction at 192 h (Figure 9B). Inversely, the expression increased by more than 1.5-fold for resistant cells for both 0.1% and 0.5 % resistance percentages, while being statistically significant at 96 and 192 h.

Illustration representation of the experimental setup for ‘once daily’ and ‘once every 4 days’ treatment regimen.

Progression of cell growth and gene expression analysis of co-culture cells with 0.1 and 0.5 % resistance treated in 96 and 192 h. (A) The cell count for continuous and intermittent for 0.1% and 0.5 % resistance at 96 hours and 192 h. (B) The gene expression of sensitive and resistant cells, respectively, at 0.1% and 0.5 % resistance. Significant differences between control and treatment groups were evaluated using one-way ANOVA, and significant differences between treatment groups (continuous and intermittent) were evaluated using an independent sample t-test (ns, no significance; *p<0.05).
Discussion
Although adding an elongated treatment-free period to intermittent treatment delayed the emergence of resistance cells, this study showed it required specific conditions, such as pM concentrations and less than 0.5 % de novo resistance. Not only can resistance be delayed, but it is also paired with a significantly higher cell count than continuous treatment. This means that patients who undergo intermittent treatment have a larger tumour burden than those who undergo continuous treatment [40], [41], [42], [43]. Maintaining a potentially large tumour burden while focusing on shrinking tumours as quickly as possible can make both clinicians and patients uneasy. However, the results contradicted each other upon applying a similar concept with nM concentrations converted from pharmacokinetic parameters. Interestingly, resistant cells grow more with lower drug pressure intermittently as opposed to growing slowly with higher concentrations. This might explain the ineffectiveness of intermittent therapy in clinics, as allowing such an extensive period between treatments can lower the plasma drug concentration [44]. One of the attempts to employ intermittent treatment involves high-dose intermittent strategies as opposed to using a similar dosage for continuous treatment [45], [46], [47]. Reports suggested this strategy was mainly used to reduce the toxicity effect of the drug while trying to keep the drug concentration within the therapeutic window [48], [49], [50].
One of the notable reasons that delays the emergence of resistant cells is the presence of sensitive cells, which imposes a survival competition during the treatment-free period for nutrients and space. This effect has been proven at pM concentrations. Although the concentration used was similar for both continuous and intermittent treatments, with a fixed experimental timepoint as illustrated in Figure 1, the presence of a 72-h treatment-free interval appeared to play a role in delaying the growth of resistant cells (Figure 6). According to adaptive therapy, to achieve control in tumour burden that is able to suppress the growth of resistance, it is necessary to have sensitive cells that grow faster than resistant cells because the latter is likely to acquire a resistant mutation. Although Figure 2 has shown that the expected doubling time is the complete opposite, resistance can still be managed below 0.5 % resistance, and this notion has been supported by multiple reports [51], 52]. In Figure 6A, it can be seen in intermittent treatment that a cluster of growing resistant cells is “covered” by surrounding sensitive cells, which stops the growth of resistance via contact inhibition. This observation is contradictory to continuous treatment, as vacant spaces left unattended by dying sensitive cells only allow resistant cells to occupy them and continuously grow without hindrance. This phenomenon has been coined “competitive release” [53], 54]. The importance of keeping the majority of sensitive cells was further emphasised in Figure 9A and B, where higher concentrations that drastically kill sensitive cells allow for the resistance to become the dominant sub-population and drive the tumour growth without any suppression from sensitive cells. Therefore, we are led to believe that continuous treatment at high concentrations promotes the competitive release of de novo resistance cells [55].
Even though intermittent treatment with adaptive therapy principles can delay the emergence of resistance, the question becomes: how long can it delay the inevitable? In this study, the experimental design of intermittent treatment was developed to represent two treatment cycles against continuous treatment (Figure 1). This is to prevent the co-culture from reaching more than 80 % confluence, which would have prevented contact inhibition. It is unknown how many cycles it took for the sensitive cells to no longer be able to suppress the resistant cells. Adaptive therapy is primarily studied based on mathematical modelling to simulate the treatment cycles until tumour progression using the cell-competitive Lotka–Volterra [51], 56] and agent-based models [34]. It has been reported that even with such models, de novo resistance tends to increase slightly with each treatment cycle, eventually leading to therapeutic failure. Thus, making an important point about changing treatment strategies for cancer can only delay it, but not eradicate it, as summarised in Figure 10. However, mathematical models demonstrate the effectiveness of an elongated treatment-free period, and low-dose treatment strategies may be durably maintained for up to 20 cycles – significantly longer than continuous therapy in metastatic castrate-resistant prostate cancer [27]. In melanoma treated with BRAF/MEK inhibitors, it was predicted in a patient-calibrated Lotka–Volterra model that it would have delayed time to progression by 6–25 months with dose rates of 6–74 % as compared to continuous therapy [56].

Illustrated summarisation of experimental results.
Some notable limitations of the study needed to be mentioned, such as the absence of a tumour microenvironment in the co-culture experimental setup. The presence of immune cells and fibroblasts is said to affect the outcome of adaptive therapy [57], 58]. In addition, the setup was done entirely with a 2-dimensional setup instead of a 3-dimensional one. The dimension of the co-culture does not represent the actual cellular interactions that might be present in vivo or patient tumours [59]. Although we were able to quantify the gene expression level of sensitive and resistant cells via specific primers in qPCR, we were not able to quantify the level of housekeeping genes that are specific for each cell line in the co-culture. This method poses difficulty in establishing a relative fold of gene expression normalised with housekeeping genes via the delta-delta Ct method; instead, the relative readings are based on normalising based on Ct values. Multiple better detection technologies, such as genetically encoded fluorescent tags and fluorescent in situ hybridisation (FISH), are suggested. Besides that, the treatment setup in this study is based on a fixed concentration for 24 h instead of a constantly changing plasma drug concentration-time curve. Even though the AUC of the curve has been converted directly to experimental concentrations, the constant-changing concentrations, which are affected by maximum drug concentration in plasma (Cmax), clearance of the drug from plasma (CL/F), terminal elimination half-life (t1/2), time to reach Cmax (tmax), and volume of distribution (V) over 24 h, were not replicated.
Conclusions
Collectively, intermittent treatment with 72 h of treatment-free period suppresses the growth of de novo resistance with 0.1% and 0.5 % resistance percentages at specific concentrations as compared to continuous treatment. These findings proved to be effective in vitro in NSCLC cell lines.
Funding source: Putra Grant
Award Identifier / Grant number: GP/2022/ 9734600
Acknowledgements
The authors gratefully acknowledge the financial support from the Putra Grant (Grant number: GP/2022/9734600).
-
Research ethics: Not applicable.
-
Informed consent: Not applicable.
-
Author contributions: Amir Imran Faisal Hamdi (Conducting experiments, data collection and data analysis, wrote the manuscript); Wen Tsin Poh (Writing – review and editing); Jonathan Chee Woei Lim (Supervision); Ummi Nadira Daut (Supervision); Soon Hin How (Conceptualisation and supervision); Yong Kek Pang (Supervision); Johnson Stanslas* (Study conceptualisation and design, review, editing and supervision). All authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Use of Large Language Models, AI and Machine Learning Tools: None declared.
-
Conflict of interest: The authors state no conflict of interest.
-
Research funding: This work was supported by Putra Grant (Grant number: GP/2022/9734600).
-
Data availability: The raw data can be obtained on request from the corresponding author.
References
1. Mok, TS, Wu, YL, Thongprasert, S, Yang, CH, Chu, DT, Saijo, N, et al.. Gefitinib or carboplatin–paclitaxel in pulmonary adenocarcinoma. New Engl J Med 2009;361:947–57. https://doi.org/10.1056/nejmoa0810699.Search in Google Scholar PubMed
2. Kuiper, JL, Smit, EF. High-dose, pulsatile erlotinib in two NSCLC patients with leptomeningeal metastases–One with a remarkable thoracic response as well. Lung Cancer 2013;80:102–5. https://doi.org/10.1016/j.lungcan.2012.12.024.Search in Google Scholar PubMed
3. Yang, JCH, Sequist, LV, Zhou, C, Schuler, M, Geater, SL, Mok, T, et al.. Effect of dose adjustment on the safety and efficacy of afatinib for EGFR mutation-positive lung adenocarcinoma: post hoc analyses of the randomized LUX-lung 3 and 6 trials. Ann Oncol 2016;27:2103–10. https://doi.org/10.1093/annonc/mdw322.Search in Google Scholar PubMed
4. Noguchi, S, Kawachi, H, Fukao, A, Terashita, S, Tajiri, T, Ikeue, T, et al.. Osimertinib Administration as the Primary Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Therapy for Brain Metastasis of De Novo T790M-positive Lung Cancer. Intern Med 2019;58:3029–31. https://doi.org/10.2169/internalmedicine.3169-19.Search in Google Scholar PubMed PubMed Central
5. Kumari, R, James, E, Pavithran, K. Complete response to afatinib in lung adenocarcinoma of epidermal growth factor receptor exon-19 deletion mutation and disease recurrence on drug discontinuation. J Pharmacol Pharmacother 2018;9:191–4. https://doi.org/10.4103/jpp.jpp-111-18.Search in Google Scholar
6. Saxena, A, Nagasaka, M, Li, Z, Becker, DJ, Levy, BP. Double trouble: a case of concurrent de novo T790M and L858R EGFR mutations in treatment-naive advanced non-small-cell lung cancer. Oncology (Williston Park, N.Y.) 2014;28, 526, 528, 530, 534.Search in Google Scholar
7. Lettig, L, Sahnane, N, Pepe, F, Cerutti, R, Albeni, C, Franzi, F, et al.. EGFR T790M detection rate in lung adenocarcinomas at baseline using droplet digital PCR and validation by ultra-deep next generation sequencing. Transl Lung Cancer Res 2019;8:584–92. https://doi.org/10.21037/tlcr.2019.09.18.Search in Google Scholar PubMed PubMed Central
8. Fujita, Y, Suda, K, Kimura, H, Matsumoto, K, Arao, T, Nagai, T, et al.. Highly sensitive detection of EGFR T790M mutation using colony hybridization predicts favorable prognosis of patients with lung cancer harboring activating EGFR mutation. J Thorac Oncol: Official Publication Int Assoc Study Lung Cancer 2012;7:1640–4. https://doi.org/10.1097/jto.0b013e3182653d7f.Search in Google Scholar
9. Zhu, G, Ye, X, Dong, Z, Lu, YC, Sun, Y, Liu, Y, et al.. Highly sensitive droplet digital PCR method for detection of EGFR-activating mutations in plasma cell-free DNA from patients with advanced non-small cell lung cancer. J Mod Dynam 2015;17:265–72. https://doi.org/10.1016/j.jmoldx.2015.01.004.Search in Google Scholar PubMed
10. Wang, X, Li, X, Guo, H, Zhu, L, Peng, Z, Wang, J, et al.. Highly Sensitive Droplet Digital PCR Method for Detection of de novo EGFR T790M Mutation in Patients with Non-Small Cell Lung Cancer. OncoTargets Ther 2020;13:10621–30. https://doi.org/10.2147/ott.s267677.Search in Google Scholar
11. Inukai, M, Toyooka, S, Ito, S, Asano, H, Ichihara, S, Soh, J, et al.. Presence of epidermal growth factor receptor gene T790M mutation as a minor clone in non-small cell lung cancer. Can Res 2006;66:7854–8. https://doi.org/10.1158/0008-5472.can-06-1951.Search in Google Scholar PubMed
12. Wu, JY, Yu, CJ, Chang, YC, Yang, CH, Shih, JY, Yang, PC. Effectiveness of tyrosine kinase inhibitors on “uncommon” epidermal growth factor receptor mutations of unknown clinical significance in non-small cell lung cancer. Clin Cancer Res: Official J Am Assoc Can Res 2011;17:3812–21. https://doi.org/10.1158/1078-0432.ccr-10-3408.Search in Google Scholar
13. Su, KY, Tseng, JS, Liao, KM, Yang, TY, Chen, KC, Hsu, KH, et al.. Mutational monitoring of EGFR T790M in cfDNA for clinical outcome prediction in EGFR-mutant lung adenocarcinoma. PLoS ONE 2018;13:e0207001. https://doi.org/10.1371/journal.pone.0207001.Search in Google Scholar PubMed PubMed Central
14. Yang, Y, Meng, Y, Zhang, H, Shen, X, Li, R, Yu, L, et al.. Detection of EGFR and BRAF mutations by competitive allele-specific TaqMan polymerase chain reaction in lung adenocarcinoma. Oncol Lett 2018;15:3295–304. https://doi.org/10.3892/ol.2017.7652.Search in Google Scholar PubMed PubMed Central
15. Xue, Z, You, M, Peng, P, Tong, H, He, W, Li, A, et al.. Taqman-MGB nanoPCR for highly specific detection of single-base mutations. Int J Nanomed 2021;16:3695–705. https://doi.org/10.2147/ijn.s310254.Search in Google Scholar PubMed PubMed Central
16. Lee, SH, Kim, EY, Kim, A, Chang, YS. Clinical implication and usefulness of de novo EGFR T790M mutation in lung adenocarcinoma with EGFR-tyrosine kinase inhibitor sensitizing mutation. Can Biol Ther 2020;21:741–8. https://doi.org/10.1080/15384047.2020.1776579.Search in Google Scholar PubMed PubMed Central
17. Billaud, A, Verriele, V, Dauvé, J, Chevalier, LM, Morel, A. Non-Small-Cell lung cancer-sensitive detection of the p.Thr790Met EGFR alteration by preamplification before PNA-Mediated PCR clamping and pyrosequencing. Diagnostics 2020;10:E527. https://doi.org/10.3390/diagnostics10080527.Search in Google Scholar PubMed PubMed Central
18. Siggillino, A, Ulivi, P, Pasini, L, Reda, MS, Chiadini, E, Tofanetti, FR, et al.. Detection of EGFR mutations in plasma cell-free tumor DNA of TKI-Treated Advanced-NSCLC patients by three methodologies: scorpion-ARMS, PNAClamp, and digital PCR. Diagnostics 2020;10:E1062. https://doi.org/10.3390/diagnostics10121062.Search in Google Scholar PubMed PubMed Central
19. Zhou, R, Cai, Y, Li, Z, Shen, S, Sha, M, Head, SR, et al.. A digital PCR assay development to detect EGFR T790M mutation in NSCLC patients. Front Lab Med 2018;2:89–96. https://doi.org/10.1016/j.flm.2018.08.002.Search in Google Scholar
20. Bria, E, Pilotto, S, Amato, E, Fassan, M, Novello, S, Peretti, U, et al.. Molecular heterogeneity assessment by next-generation sequencing and response to gefitinib of EGFR mutant advanced lung adenocarcinoma. Oncotarget 2015;6:12783–95. https://doi.org/10.18632/oncotarget.3727.Search in Google Scholar PubMed PubMed Central
21. Guo, G, Li, G, Liu, Y, Li, H, Guo, Q, Liu, J, et al.. Next-generation sequencing reveals high uncommon EGFR mutations and tumour mutation burden in a subgroup of lung cancer patients. Front Oncol 2021;11:621422. https://doi.org/10.3389/fonc.2021.621422.Search in Google Scholar PubMed PubMed Central
22. Vendrell, JA, Mazieres, J, Senal, R, Rouquette, I, Quantin, X, Pujol, JL, et al.. Ultra-sensitive EGFR T790M detection as an independent prognostic marker for lung cancer patients harboring EGFR del19 mutations and treated with first-generation TKIs. Clin Cancer Res: Official J Am Assoc Can Res 2019;25:4280–9. https://doi.org/10.1158/1078-0432.ccr-18-2683.Search in Google Scholar
23. de Kock, R, Knoops, C, Baselmans, M, van den Borne, B, Brunsveld, L, Scharnhorst, V, et al.. Sensitive cell-free tumor DNA analysis in supernatant pleural effusions supports therapy selection and disease monitoring of lung cancer patients. Can Treat Res Commun 2021;29:100449. https://doi.org/10.1016/j.ctarc.2021.100449.Search in Google Scholar PubMed
24. Dooley, AJ, Gupta, A, Bhattacharyya, M, Middleton, MR. Intermittent dosing with vemurafenib in BRAF V600E-mutant melanoma: review of a case series. Ther Adv Med Oncol 2014;6:262–6. https://doi.org/10.1177/1758834014548187.Search in Google Scholar PubMed PubMed Central
25. Holderfield, M, Deuker, MM, McCormick, F, McMahon, M. Targeting RAF kinases for cancer therapy: BRAF-Mutated melanoma and beyond. Nat Rev Cancer 2014;14:455–67. https://doi.org/10.1038/nrc3760.Search in Google Scholar PubMed PubMed Central
26. Tétu, P, Vercellino, L, Reger de Moura, C, Baroudjian, B, Dumaz, N, Mourah, S, et al.. Mitogen-activated protein kinase blockade in melanoma: intermittent versus continuous therapy, from preclinical to clinical data. Curr Opin Oncol 2021;33:127–32. https://doi.org/10.1097/cco.0000000000000706.Search in Google Scholar PubMed
27. Zhang, J, Cunningham, JJ, Brown, JS, Gatenby, RA. Integrating evolutionary dynamics into treatment of metastatic castrate-resistant prostate cancer. Nat Commun 2017;8:1816. https://doi.org/10.1038/s41467-017-01968-5.Search in Google Scholar PubMed PubMed Central
28. Matsuzaki, T, Iwami, E, Sasahara, K, Kuroda, A, Nakajima, T, Terashima, T. A case report of metastatic lung adenocarcinoma with long-term survival for over 11 years. Medicine 2019;98:e14100. https://doi.org/10.1097/md.0000000000014100.Search in Google Scholar
29. Tu, HY, Li, YS, Yang, JJ, Chen, HJ, Jiang, BY, Zhong, WZ, et al.. Long-term survival of over 6 years with afatinib sequential treatment in a patient with EGFR mutation-positive non-small cell lung cancer: a case report. Clin Drug Invest 2021;41:483–8. https://doi.org/10.1007/s40261-021-01025-6.Search in Google Scholar PubMed PubMed Central
30. Yamanaka, Y, Seki, Y, Ishikawa, T, Kuwano, K. Long-lasting response to afatinib that persisted after treatment discontinuation in a case of EGFR-mutated lung adenocarcinoma. BMJ Case Rep 2019;12:e227383. https://doi.org/10.1136/bcr-2018-227383.Search in Google Scholar PubMed PubMed Central
31. Gatenby, RA, Silva, AS, Gillies, RJ, Frieden, BR. Adaptive therapy. Can Res 2009;69:4894–903. https://doi.org/10.1158/0008-5472.can-08-3658.Search in Google Scholar PubMed PubMed Central
32. Gallaher, J, Strobl, M, West, J, Gatenby, R, Zhang, J, Robertson-Tessi, M, et al.. Inter- and intra-metastatic heterogeneity shapes adaptive therapy cycling dynamics. Can Res 2023;83:2775–89. CAN-22-2558. https://doi.org/10.1158/0008-5472.CAN-22-2558.Search in Google Scholar PubMed PubMed Central
33. Masud, MA, Kim, JY, Kim, E. Effective dose window for containing tumor burden under tolerable level. Npj Syst Biol Appl 2023;9:17. Article 1. https://doi.org/10.1038/s41540-023-00279-4.Search in Google Scholar PubMed PubMed Central
34. Gallaher, JA, Enriquez-Navas, PM, Luddy, KA, Gatenby, RA, Anderson, ARA. Spatial heterogeneity and evolutionary dynamics modulate time to recurrence in continuous and adaptive cancer therapies. Can Res 2018;78:2127–39. https://doi.org/10.1158/0008-5472.can-17-2649.Search in Google Scholar
35. Zhang, Q, Li, R, Chen, X, Lee, SB, Pan, J, Xiong, D, et al.. Effect of weekly or daily dosing regimen of Gefitinib in mouse models of lung cancer. Oncotarget 2017;8:72447–56. https://doi.org/10.18632/oncotarget.19785.Search in Google Scholar PubMed PubMed Central
36. Camidge, DR, Sequist, LV, Jänne, PA, Weickhardt, AJ, Dowling, ES, Alicea, J, et al.. Phase Ib study of high-dose intermittent afatinib in patients with advanced solid tumors. Clin Lung Cancer 2018;19:e655–e665. https://doi.org/10.1016/j.cllc.2018.04.015.Search in Google Scholar PubMed
37. Gerritse, SL, Labots, M, Ter Heine, R, Dekker, H, Poel, D, Tauriello, DVF, et al.. High-dose intermittent treatment with the multikinase inhibitor sunitinib leads to high intra-tumor drug exposure in patients with advanced solid tumors. Cancers 2022;14:6061. https://doi.org/10.3390/cancers14246061.Search in Google Scholar PubMed PubMed Central
38. Takata, M, Chikumi, H, Matsunami, K, Kodani, M, Sakamoto, T, Hashimoto, K, et al.. A new rapid method for detecting epidermal growth factor receptor mutations in non-small cell lung cancer. Oncol Rep 2015;33:1040–8. https://doi.org/10.3892/or.2015.3716.Search in Google Scholar PubMed PubMed Central
39. Wind, S, Schnell, D, Ebner, T, Freiwald, M, Stopfer, P. Clinical pharmacokinetics and pharmacodynamics of afatinib. Clin Pharmacokinet 2017;56:235–50. https://doi.org/10.1007/s40262-016-0440-1.Search in Google Scholar PubMed PubMed Central
40. Hata, A, Fujita, S, Kaji, R, Nanjo, S, Katakami, N. Dose reduction or intermittent administration of Erlotinib: which is better for patients suffering from intolerable toxicities? Intern Med 2013;5.10.2169/internalmedicine.52.8543Search in Google Scholar PubMed
41. Brady-Nicholls, R, Nagy, JD, Gerke, TA, Zhang, T, Wang, AZ, Zhang, J, et al.. Prostate-specific antigen dynamics predict individual responses to intermittent androgen deprivation. Nat Commun 2020;11:1750. https://doi.org/10.1038/s41467-020-15424-4.Search in Google Scholar PubMed PubMed Central
42. Cunningham, J, Thuijsman, F, Peeters, R, Viossat, Y, Brown, J, Gatenby, R, et al.. Optimal control to reach eco-evolutionary stability in metastatic castrate-resistant prostate cancer. PLoS ONE 2020;15:e0243386. https://doi.org/10.1371/journal.pone.0243386.Search in Google Scholar PubMed PubMed Central
43. Brady-Nicholls, R, Zhang, J, Zhang, T, Wang, AZ, Butler, R, Gatenby, RA, et al.. Predicting patient-specific response to adaptive therapy in metastatic castration-resistant prostate cancer using prostate-specific antigen dynamics. Neoplasia 2021;23:851–8. https://doi.org/10.1016/j.neo.2021.06.013.Search in Google Scholar PubMed PubMed Central
44. Junaid, T, Wu, X, Thanukrishnan, H, Venkataramanan, R. Chapter 30–Therapeutic drug monitoring. In: Thomas, D, editor. Clinical pharmacy education, practice and research. Amsterdam: Elsevier; 2019:425–36 pp.10.1016/B978-0-12-814276-9.00030-1Search in Google Scholar
45. Rovithi, M, de Haas, RR, Honeywell, RJ, Poel, D, Peters, GJ, Griffioen, AW, et al.. Alternative scheduling of pulsatile, high dose sunitinib efficiently suppresses tumor growth. J Exp Clin Can Res 2016;35:138. https://doi.org/10.1186/s13046-016-0411-2.Search in Google Scholar PubMed PubMed Central
46. Zhu, Y, Du, Y, Liu, H, Ma, T, Shen, Y, Pan, Y. Study of efficacy and safety of pulsatile administration of high-dose gefitinib or erlotinib for advanced non-small cell lung cancer patients with secondary drug resistance: a single center, single arm, phase II clinical trial. Thorac Cancer 2016;7:663–9. https://doi.org/10.1111/1759-7714.12384.Search in Google Scholar PubMed PubMed Central
47. Wang, X, Zhang, L, Goldberg, SN, Bhasin, M, Brown, V, Alsop, DC, et al.. High dose intermittent sorafenib shows improved efficacy over conventional continuous dose in renal cell carcinoma. J Transl Med 2011;9:220. https://doi.org/10.1186/1479-5876-9-220.Search in Google Scholar PubMed PubMed Central
48. Faber, E, Nausová, J, Jarosová, M, Egorin, MJ, Holzerová, M, Rozmanová, S, et al.. Intermittent dosage of imatinib mesylate in CML patients with a history of significant hematologic toxicity after standard dosing. Leuk Lymphoma 2006;47:1082–90. https://doi.org/10.1080/10428190600565057.Search in Google Scholar PubMed
49. Russo, D, Malagola, M, Skert, C, Cancelli, V, Turri, D, Pregno, P, et al.. Managing chronic myeloid leukaemia in the elderly with intermittent imatinib treatment. Blood Can J 2015;5:e347. https://doi.org/10.1038/bcj.2015.75. Article 9.Search in Google Scholar PubMed PubMed Central
50. Giona, F, Malaspina, F, Putti, MC, Ladogana, S, Mura, R, Burnelli, R, et al.. Results and outcome of intermittent imatinib (ON/OFF schedule) in children and adolescents with chronic myeloid leukaemia. Br J Haematol 2020;188:e101–5. https://doi.org/10.1111/bjh.16388.Search in Google Scholar PubMed
51. Strobl, MAR, Gallaher, J, West, J, Robertson-Tessi, M, Maini, PK, Anderson, ARA. Spatial structure impacts adaptive therapy by shaping intra-tumoral competition. Commun Med 2022;2:46. https://doi.org/10.1038/s43856-022-00110-x.Search in Google Scholar PubMed PubMed Central
52. Viossat, Y, Noble, R. A theoretical analysis of tumour containment. Nat Ecol Evol 2021;5:826–35. https://doi.org/10.1038/s41559-021-01428-w.Search in Google Scholar PubMed PubMed Central
53. West, J, Ma, Y, Newton, PK. Capitalizing on competition: an evolutionary model of competitive release in metastatic castration resistant prostate cancer treatment. J Theor Biol 2018;455:249–60. https://doi.org/10.1016/j.jtbi.2018.07.028.Search in Google Scholar PubMed PubMed Central
54. Newton, PK, Ma, Y. Nonlinear adaptive control of competitive release and chemotherapeutic resistance. Phys Rev E 2019;9:022404. https://doi.org/10.1103/physreve.99.022404.Search in Google Scholar
55. West, J, Adler, F, Gallaher, J, Strobl, M, Brady-Nicholls, R, Brown, J, et al.. A survey of open questions in adaptive therapy: bridging mathematics and clinical translation. Elife 2023;12:e84263. https://doi.org/10.7554/elife.84263.Search in Google Scholar PubMed PubMed Central
56. Kim, E, Brown, JS, Eroglu, Z, Anderson, ARA. Adaptive therapy for metastatic melanoma: predictions from patient calibrated mathematical models. Cancers 2021;13:823. https://doi.org/10.3390/cancers13040823.Search in Google Scholar PubMed PubMed Central
57. A, MM, Kim, JY, Pan, CH, Kim, E. The impact of the spatial heterogeneity of resistant cells and fibroblasts on treatment response. PLoS Comput Biol 2022;18:e1009919. https://doi.org/10.1371/journal.pcbi.1009919.Search in Google Scholar PubMed PubMed Central
58. Ma, S, Lei, J, Lai, X. Modeling tumour heterogeneity of PD-L1 expression in tumour progression and adaptive therapy. J Math Biol 2023;86:38. https://doi.org/10.1007/s00285-023-01872-1.Search in Google Scholar PubMed
59. Jensen, C, Teng, Y. Is it time to start transitioning from 2D to 3D cell culture? Front Mol Biosci 2020;7:33. https://doi.org/10.3389/fmolb.2020.00033.Search in Google Scholar PubMed PubMed Central
Supplementary Material
This article contains supplementary material (https://doi.org/10.1515/oncologie-2025-0178).
© 2025 the author(s), published by De Gruyter on behalf of Tech Science Press (TSP)
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Review Articles
- Liquid biopsy – a promising and effective method for surveying non-small cell lung cancer minimal residual diseases and anti-cancer drug response after treatment
- Current application status of proton beam therapy for gastrointestinal tumors
- Research progress on the regulation of cuproptosis-related genes by non-coding RNAs in tumors
- Deep learning in hepatic oncology imaging: a narrative review of computed tomography applications
- Synergistic approaches: a narrative mini-review of radiotherapy and immunotherapy in the treatment of lung cancer
- Research Articles
- Intravesical prostatic protrusion as a predictor of acute urinary retention following stereotactic body radiation therapy for localised prostate cancer: a retrospective study
- The differential effect of glutamine supplementation on the orthotopic and subcutaneous growth of two syngeneic murine models of glioma
- Intermittent afatinib treatment suppresses the growth of resistant T790M-H1975 cells in non-small cell lung cancer (NSCLC) co-culture
- Prognostic stratification of colorectal cancer by immune profiling reveals SPP1 as a key indicator for tumor immune status
- The activity of base excision repair is positively correlated with the infiltration of CD4+ T cells in melanoma
- Integrated analysis of immunity and ferroptosis related tumor microenvironment in a novel risk score model for lung adenocarcinoma prognosis
- Retrospective analysis of risk factors for early recurrence after hepatocellular carcinoma resection
- The ENST00000539930 transcript predicts sensitivity to PARP inhibitors and clinical prognosis in cancers
- VTA1 and breast cancer: a potential indicator for diagnostic and prognostic evaluation
- USP24 stabilizes VDAC2 via deubiquitination to promote apoptosis and ferroptosis in clear cell renal cell carcinoma (ccRCC)
- Clinicopathological characteristics, prognosis, and therapeutic implications in breast cancer with pathologically confirmed bone marrow metastases: an observational retrospective study
- Short Commentaries
- Cancer cell mitochondria: the missing puzzle in predicting response to PD-1/PD-L1 inhibitors
- From mitochondrial cristae pathobiology to metabolic reprogramming in cancer: the α and ω of Malignancies?
- Article Commentary
- Stopping SOAT1 sparks an immune attack on liver cancer: a metabolic-immune axis in hepatocellular carcinoma
Articles in the same Issue
- Frontmatter
- Review Articles
- Liquid biopsy – a promising and effective method for surveying non-small cell lung cancer minimal residual diseases and anti-cancer drug response after treatment
- Current application status of proton beam therapy for gastrointestinal tumors
- Research progress on the regulation of cuproptosis-related genes by non-coding RNAs in tumors
- Deep learning in hepatic oncology imaging: a narrative review of computed tomography applications
- Synergistic approaches: a narrative mini-review of radiotherapy and immunotherapy in the treatment of lung cancer
- Research Articles
- Intravesical prostatic protrusion as a predictor of acute urinary retention following stereotactic body radiation therapy for localised prostate cancer: a retrospective study
- The differential effect of glutamine supplementation on the orthotopic and subcutaneous growth of two syngeneic murine models of glioma
- Intermittent afatinib treatment suppresses the growth of resistant T790M-H1975 cells in non-small cell lung cancer (NSCLC) co-culture
- Prognostic stratification of colorectal cancer by immune profiling reveals SPP1 as a key indicator for tumor immune status
- The activity of base excision repair is positively correlated with the infiltration of CD4+ T cells in melanoma
- Integrated analysis of immunity and ferroptosis related tumor microenvironment in a novel risk score model for lung adenocarcinoma prognosis
- Retrospective analysis of risk factors for early recurrence after hepatocellular carcinoma resection
- The ENST00000539930 transcript predicts sensitivity to PARP inhibitors and clinical prognosis in cancers
- VTA1 and breast cancer: a potential indicator for diagnostic and prognostic evaluation
- USP24 stabilizes VDAC2 via deubiquitination to promote apoptosis and ferroptosis in clear cell renal cell carcinoma (ccRCC)
- Clinicopathological characteristics, prognosis, and therapeutic implications in breast cancer with pathologically confirmed bone marrow metastases: an observational retrospective study
- Short Commentaries
- Cancer cell mitochondria: the missing puzzle in predicting response to PD-1/PD-L1 inhibitors
- From mitochondrial cristae pathobiology to metabolic reprogramming in cancer: the α and ω of Malignancies?
- Article Commentary
- Stopping SOAT1 sparks an immune attack on liver cancer: a metabolic-immune axis in hepatocellular carcinoma