Skip to main content
PLOS Biology logoLink to PLOS Biology
. 2020 May 15;18(5):e3000713. doi: 10.1371/journal.pbio.3000713

Antibiotics can be used to contain drug-resistant bacteria by maintaining sufficiently large sensitive populations

Elsa Hansen 1,#, Jason Karslake 2,#, Robert J Woods 3, Andrew F Read 4, Kevin B Wood 2,5,*
Editor: David S Schneider6
PMCID: PMC7266357  PMID: 32413038

Abstract

Standard infectious disease practice calls for aggressive drug treatment that rapidly eliminates the pathogen population before resistance can emerge. When resistance is absent, this elimination strategy can lead to complete cure. However, when resistance is already present, removing drug-sensitive cells as quickly as possible removes competitive barriers that may slow the growth of resistant cells. In contrast to the elimination strategy, a containment strategy aims to maintain the maximum tolerable number of pathogens, exploiting competitive suppression to achieve chronic control. Here, we combine in vitro experiments in computer-controlled bioreactors with mathematical modeling to investigate whether containment strategies can delay failure of antibiotic treatment regimens. To do so, we measured the “escape time” required for drug-resistant Escherichia coli populations to eclipse a threshold density maintained by adaptive antibiotic dosing. Populations containing only resistant cells rapidly escape the threshold density, but we found that matched resistant populations that also contain the maximum possible number of sensitive cells could be contained for significantly longer. The increase in escape time occurs only when the threshold density—the acceptable bacterial burden—is sufficiently high, an effect that mathematical models attribute to increased competition. The findings provide decisive experimental confirmation that maintaining the maximum number of sensitive cells can be used to contain resistance when the size of the population is sufficiently large.


Adding antibiotic-susceptible cells to otherwise resistant bacterial populations can be used to slow microbial growth in the presence of antibiotics; this provides support for a containment strategy that aims to maintain the maximum tolerable number of pathogens, exploiting competitive suppression to achieve chronic control.

Introduction

The ability to successfully treat infectious disease is often undermined by drug resistance [16]. When resistance poses a major threat to the quality and duration of a patient's life, the goal of treatment is to restore patient health while delaying treatment failure for as long as possible. To do so, standard practice calls for aggressive drug treatment to rapidly remove the drug-sensitive pathogen population and prevent resistance-conferring mutations [717]. Aggressive treatment can involve either single-drug or combination therapies, which have been shown to modulate the emergence of resistance [1825]. Here, we are interested in situations in which such aggressive regimens do not completely prevent the emergence of resistance—for example, scenarios in which resistance is already present at the onset of treatment.

If aggressive treatment cannot prevent the emergence of resistance, an alternative approach is to use competition between drug-sensitive and drug-resistant cells to slow the expansion of the drug-resistant population. There is ample evidence that competition between sensitive and resistant cells can be intense [2629] and may be over limited resources like glucose or target cells [3033]. Competition can also be immune mediated or occur via direct interference (e.g., bacteriocins) [26, 3437]. There are numerous theoretical studies [35, 3849] suggesting that sensitive cells can competitively suppress resistant cells, and this suppression has even been observed experimentally in parasites and cancer [42, 5055]. Ideally, resistance never emerges, but if it does, delaying the time to treatment failure can potentially prolong life (chronic infections [56]) or give immunity time to prevent resistance emergence (e.g., acute infections, or when immunosuppression is medically induced and temporary). Because sensitive cells can both generate de novo resistance and also competitively suppress existing resistant mutants, making good treatment decisions requires understanding the relative importance of these opposing effects (Fig 1).

Fig 1. Containment strategies may leverage competition to extend time below treatment failure threshold.

Fig 1

(A) Aggressive treatment uses high drug concentrations (lightning flashes), which eliminates sensitive cells (blue) but may fail when resistant cells (red) emerge and the population exceeds the failure threshold (“acceptable burden”, light-blue circle). (B) Containment strategies attempt to maintain the population just below the failure threshold, leveraging competition between sensitive (blue) and emergent resistant (red) cells to potentially prolong time to failure. (C) Schematic of potential feedback between growth processes in mixed populations. Drug (lightning flash) inhibits sensitive cells (blue), which in turn inhibit resistant cells (red) through competition but may also contribute to the resistant population via mutation.

Recent theoretical work compares two extreme treatment strategies: a strategy that removes all drug-sensitive cells (what we call elimination) and a strategy that maximizes the sensitive population (what we call containment) [45]. The elimination strategy removes sensitive cells as fast as possible, minimizing the risk of mutation but also removing competitive barriers that may slow the growth of existing resistant cells [35] (Fig 1A). Containment, on the other hand, maintains as many sensitive pathogens as is clinically acceptable (i.e., a pathogen density that is deemed to be safe and below which treatment is not necessary), using drug treatment only to alleviate symptoms. Containment maintains the total pathogen density at this acceptable burden (Fig 1B). This maximizes competitive suppression but leaves sensitive pathogens that can generate resistance. Cases in which containment better slows the expansion of the resistant population represent situations in which standard practice can potentially be improved.

Theory predicts that neither strategy is always best; there are situations in which containment will better control resistance and others in which it will make a bad prognosis worse [45]. The latter case arises when the benefit of maintaining sensitive pathogens (competitive suppression of resistance) is outweighed by the rate at which sensitive pathogens become resistant by mutation (or horizontal gene transfer). Mathematical modeling suggests that across a wide variety of diseases and settings, the same fundamental principles determine when containment is better than elimination. First, there has to be sufficient competition (the clinically acceptable burden high enough), and second, the growth of the resistant population has to be driven primarily by the replication of resistant cells, not by mutational inputs (resistant population large enough).

Here, we experimentally test the hypothesis that containment strategies that maintain a subpopulation of sensitive cells can slow the expansion of resistant bacteria. We combine simple mathematical models with in vitro experiments in computer-controlled bioreactors, in which “treatment failure” is defined by bacterial populations eclipsing a threshold density (the acceptable burden). The experimental design directly tests the effect of maximizing the size of the sensitive subpopulation on escape time. We find that containment strategies can increase escape times when the acceptable burden is sufficiently high but are ineffective at low densities, in which competition is small. Although there is empirical evidence for competitive suppression in parasites and cancer [5055], our work provides an explicit demonstration of competitive suppression of resistance to antibiotic treatment and a direct test of a competition-maximizing containment strategy in a bacterial pathogen. The findings are particularly striking because they occur in well-mixed populations with a continual renewal of resources and using an acceptable burden well below the natural carrying capacity—all conditions not typically associated with strong competition.

Results

The aim of this study is to investigate whether a containment strategy that maintains subpopulations of sensitive cells can improve our ability to keep drug-resistant populations below a predefined threshold density. To do so, we develop an experimental assay based on adaptive drug dosing that allows us to directly compare escape times for resistant-only populations with those of matched resistant populations supplemented with sensitive populations of different sizes.

Model system

We grew bacterial populations in well-mixed bioreactors in which environmental conditions, including drug concentration and nutrient levels, can be modulated using a series of computer-controlled peristaltic pumps (see, for example, [57, 58]). Population density is measured using light scattering (optical density [OD]), and drug concentration can be adjusted in real time in response to population dynamics or predetermined protocols (S1 Fig).

To obtain bacterial populations with different drug sensitivities, we began with E. coli strains REL606 and REL607, which are well-characterized ancestral strains used in the long-term evolution experiment (LTEE) in E. coli [59]. These strains differ by a single point mutation in araA, which serves as a neutral marker for competition experiments; REL606 (REL607) appears red (pink) when grown on tetrazolium arabinose (TA) plates. To generate a “drug-resistant” strain, we used laboratory evolution to isolate a mutant of the REL606 strain that was resistant to doxycycline, a frequently used protein synthesis inhibitor (Methods). Whole-genome sequencing confirmed that the ancestor REL606 strain was identical to the published sequence (accession number NC_012967), whereas the REL606-derived resistant strain had five mutations, including mutations in OmpF and AcrR, genes that have been previously linked with increased resistance to multiple antibiotics, including tetracyclines [60, 61]. We quantified the phenotypic responses of the drug-sensitive (REL607) and drug-resistant (REL606-derived mutant) cells to doxycycline by measuring real-time per capita growth rate for isogenic populations of each strain exposed to different concentrations of drug (Fig 2). Briefly, growth rate was estimated using influx rate of media required to maintain populations at a constant density (Methods). The resistant isolate exhibits both increased resistance to doxycycline (increased half-maximal inhibitory concentration) as well as decreased growth in the absence of drug (Fig 2). We note that in the experiments that follow, drug concentrations are sufficiently high such that resistant cells generally have a selective advantage over sensitive cells despite this fitness cost.

Fig 2. Resistant cells exhibit increased resistance to doxycycline and a small fitness cost.

Fig 2

Left panels: per capita growth rate in bioreactors for ancestral (sensitive, blue) and resistant (red) populations exposed to increasing concentrations of doxycycline (top to bottom in each panel). Real-time per capita growth rate (light blue or red curves) is estimated from flow rates required to maintain constant cell density at each drug concentration (Methods). Mean growth rate (thick solid lines) is estimated between 200 and 300 minutes post–drug addition (shaded regions), when the system has reached steady state. Doxycycline concentrations are 10, 30, 50, and 80 ng/mL (top panel, top to bottom) and 50, 150, 300, and 500 ng/mL (bottom panel, top to bottom). Right panel: dose-response curve for sensitive (blue) and resistant (red) populations. Circles are time-averaged growth rates between 200 and 300 minutes post–drug addition (shaded regions in [A]), with error bars ± one standard deviation over the measured interval; filled circles correspond to the specific examples shown in left panels. Drug-free growth rates are 0.017±0.001 minutes−1 (sensitive) and 0.015±0.002 minutes−1 (resistant), indicating that resistant cells exhibit a fitness cost. Solid lines, fit to Hill-like dose-response function r = r0(1+(D/h)k)−1, with r the growth rate, D the drug concentration, r0 the growth in the absence of drug, h the IC50, and k the Hill coefficient. IC50 values are estimated to be h = 49 ng/mL (sensitive cells) and h = 210 ng/mL (resistant cells). Data are deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943 [62]. IC50, half-maximal inhibitory concentration.

Experimental design to measure effect of sensitive cells on escape time

Our experimental design aims to measure the effect of sensitive cells on a resistant population. First, we seed two vials with the same low density of resistant cells. Then, drug-sensitive cells are added to one of these vials (the “mixed” vial) to achieve a total cell density equal to a predetermined threshold density—the acceptable burden (Pmax). We use an additive design because we are interested in comparing the dynamics of the resistant population in the presence/absence of the sensitive population. Because high concentrations of drug are expected to completely inhibit growth of sensitive cells and therefore eliminate any potential competition, we designed an adaptive drug dosing protocol intended to maintain the mixed population at a fixed density (Pmax) using minimal drug. The dosing protocol uses simple feedback control to adjust the drug concentration in real time in response to changes in population density (Fig 3A and Methods). Finally, as a control, a third vial is seeded with the same density of sensitive bacteria as that added to the mixed vial (Fig 3A, sensitive-only). This control allows for an indirect measure of the effect of sensitive cells in the mixed vial because, in the absence of competition or other intercellular interactions, the dynamics of the mixed population should be a simple sum of the dynamics in the two single-strain populations.

Fig 3. Experimental design and dynamics for comparison to measure effect of “competition maximization”.

Fig 3

(A) Schematic of experiment. Three different populations (sensitive-only, resistant-only, and mixed) were exposed to identical antibiotic treatments in separate bioreactors. The medium in each bioreactor was also refreshed at a constant rate of FN = 0.067 mL/min. The drug treatment was determined in real time by measuring the density (OD) of the mixed population and adjusting drug influx to maintain a constant density (Pmax) while minimizing drug used (Methods). Although the dynamics of the mixed population fully determine the temporal profile of the drug dosing, all three populations then receive identical treatments. In the high-density experiment, mixed populations started at an OD of Pmax = 0.2, with a 90–10 ratio of sensitive to resistant cells. The initial OD of resistant cells is therefore 0.02. Resistant-only populations started from an initial density of 0.02 and contained no sensitive cells, whereas sensitive-only populations started from an initial density of 0.18 and contained no resistant cells. In the low-density experiment, mixed populations started at an OD of Pmax = 0.1, and the initial OD of resistant cells was unchanged (OD = 0.02). Therefore, the starting conditions of the high- and low-density experiments differ only in the number of sensitive cells. (B and C) Experiments (left) and model (right) in high-density (Pmax = 0.2 [B]) and low-density (Pmax = 0.1 [C]) regimes. Red curves are resistant only, blue are sensitive only, and black are the mixed populations. Lightly shaded curves correspond to individual experiments, and dark curves show the median across experiments. Horizontal dashed lines show the treatment failure threshold Pmax = 0.025, in which the 0.025 term allows for small experimental fluctuations without triggering a threshold crossing event. Data are deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943 [62]. OD, optical density; Pmax, acceptable burden.

The temporal dynamics of the mixed population—but not the other populations—completely determine the drug dosing protocol, but all three populations receive identical drug dosing and therefore experience identical drug concentrations over time. This strategy ensures that any increased resistance suppression in the mixed vial can be attributed to competitive suppression by sensitive cells and not drug inhibition. It is essential that all vials have the same drug concentration—otherwise, differences in resistance suppression could be attributed to the differences in drug concentration. Because drug concentration in the vials is restricted to a finite range (0–125 ng/mL), populations containing resistant cells cannot be contained indefinitely and will eventually eclipse the threshold density (Pmax). The time required for this crossover is defined as the escape time, and the goal of the experiment is to compare escape times—which correspond, intuitively, to times of treatment failure—in the absence of sensitive cells and the presence of the largest acceptable sensitive population.

There are three possibilities. If mutation dominates, then the absence of sensitive cells is expected to be best, and the escape time of the resistant-only population should exceed that of the mixed population. On the other hand, if competition dominates, then the mixed population should take the longest to escape. Finally, if the effects of sensitive cells (both mutational input and competitive suppression) are negligible, then the escape times of the resistant-only and mixed populations should be similar. To quantitatively guide our experiments and refine this intuition, we developed and parameterized a simple mathematical model for population growth in the bioreactors (see Methods and S1 Text for a detailed description of the model). A simple analysis suggests that, for our experimental design, the effect of competition will always dominate over the effect of mutational input (see S2 Text). As a result, we neglect mutation and focus on the role of competition. This allows us to identify two values of acceptable burden (Pmax), which are predicted to produce different results. For high Pmax (OD = 0.2), competition dominates, and the mixed vial should have the longest escape time. For low Pmax (OD = 0.1), competition is minimal, and the escape times of the resistant-only and mixed vials should be similar.

Benefit of competition maximization depends on acceptable burden

To test these predictions, we first performed the experiment at the threshold density that the model predicts will lead to competitive suppression (Pmax = 0.2 [Fig 3B]). Note that this density falls in the range of exponential growth and falls below the stationary phase limit in unperturbed populations (S2 Fig). To account for batch effects and day-to-day experimental fluctuations, we repeated the experiment multiple times across different days, using different media and drug preparations. Unsurprisingly, the experiments confirm that sensitive-only populations are significantly inhibited under this treatment protocol and never reach the containment threshold; in fact, the overall density decreases slowly over time because of a combination of strong drug inhibition and effluent flow (Fig 3B, blue curves). By contrast, the resistant-only population grows steadily and eclipses the threshold in 6–9 hours (Fig 3B, red curves). Remarkably, however, the mixed population (black curves) is contained below threshold—in almost all cases—for the entire length of the experiment, which spans more than 18 hours. At the end of the experiment, we plated representative examples of resistant-only and mixed populations (S4 Fig), which confirmed that the mixed vial was predominantly resistant at the end of the experiment. In addition, these sensitive and resistant isolates exhibited similar dose-response curves to the original sensitive and resistant strains (S5 Fig). Matched drug-free controls indicate that containment in the mixed vial is not due to artifacts from media inflow or outflow (S1 Text and S3 Fig). The experiments also show remarkable agreement with the model (with no adjustable parameters; compare left and right panels in Fig 3B).

If competition were driving the increased escape time, one would expect the effect to be reduced as the threshold density (Pmax) is decreased. To test this hypothesis, we repeated the experiments at Pmax = 0.1 (Fig 3C). As before, the sensitive-only population is strongly inhibited by the drug and decreases in size over time (blue). Also as before, the resistant-only population (red) escapes the containment threshold, typically between 5 and 8 hours (faster than in the high Pmax experiment because of the lower threshold). In contrast to the previous experiment, however, the mixed population also escapes the containment threshold, and furthermore, it does so on similar timescales as the resistant-only population. This density-dependent discrepancy reflects the fact that sensitive and resistant populations interact when the density is sufficiently high. Again, the agreement between model and experiment is quite good, though the model does predict a slightly longer escape time in the mixed population. The small discrepancy between the model and the experiment suggests that at low densities the growth in the resistant-only vial is slower than the model predicts—suggesting that competition at low densities may be greater than the model assumes.

To quantify these results, we calculated the time to escape for each experiment. We defined time to escape for a particular experiment as the first time at which the growth curve (OD) exceeded the threshold density Pmax by at least 0.025 OD units (note that the 0.025 was chosen to allow for noise fluctuations in the OD time series without triggering a threshold crossing event). For low values of acceptable burden (Pmax), the escape times for resistant-only and mixed populations are nearly identical (Fig 4, left). By contrast, at higher values of Pmax, the escape time is dramatically increased (more than doubled) in the mixed population relative to the resistant-only population (Fig 4, right), even though both receive identical drug treatment and start with identically sized resistant populations. For our specific experimental setup, this corresponds to extending the escape time by more than 10 hours. Importantly, our experiments suggest that sensitive cells are beneficial at high values of Pmax and have little effect at low Pmax, consistent with the assumption that mutation-driven costs of sensitive cells in our system are negligible and that the acceptable burden must be high enough to derive benefit from competitive suppression.

Fig 4. Increased escape time under “competition maximization” requires a high threshold density.

Fig 4

Time to escape for populations maintained at low (Pmax = 0.1, left) and high (Pmax = 0.2, right) threshold densities (“acceptable burdens”). Small circles: escape times for individual experiments in mixed (black) or R-only (red) populations. Large circles: mean escape time across experiments, with error bars corresponding to ±1 standard deviation. Time to escape is defined as the time at which the population exceeds the threshold OD of Pmax+0.025, in which the 0.025 is padding provided to account for noise fluctuations. Time to escape is normalized by the total length of the experiment (mean length 22.5 hours). Note that in the high Pmax case (right), the mixed population (black) reached the threshold density during the course of the experiment in only one case, so the escape times are set to 1 in all other cases. Data are deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943 [62]. OD, optical density; Pmax, acceptable burden; R, resistant.

To further quantify the relationship between Pmax and escape time, we performed similar experiments for a range of Pmax values (Fig 5). As predicted by the model, we found that escape time increases rapidly as Pmax increases for the mixed, but not for the resistant-only, population. Furthermore, the quantitative trends we observe are generally well captured by the model, though the theory consistently underestimates the escape time for the resistant-only vial (colored ×’s are above dashed curve) and the escape time at low Pmax values (blue ×’s and squares lie above both the dashed and dotted curve). This is consistent with the trend observed in Fig 3C and may suggest that the model underestimates the strength of competition at low densities. Finally, we performed one series of experiments over an extended time period (more than 35 hours, Fig 5C). To probe the extreme limits of containment, we chose Pmax to be greater than the predicted steady-state population size. In this scenario, both populations (resistant-only and mixed) eventually approach a population size below Pmax. Interestingly, however, the resistant-only population transiently crosses Pmax, whereas the mixed population can be held below the threshold for the entirety of the experiment. This example illustrates that there are situations in which populations containing resistant and sensitive cells can be held below threshold for extended periods, avoiding the transient escape dynamics of resistant-only populations.

Fig 5. Escape time depends sensitively on Pmax for mixed, but not for R-only, population.

Fig 5

(A) Growth curves for R-only (left) and mixed (right) populations for different values of Pmax, ranging from 0.2 (blue) to 0.5 (red). Curves transition from opaque to transparent as they eclipse Pmax, and the crossover point is marked with an “×” (R-only populations) or square (mixed populations). (B) Failure time (i.e., time to cross threshold) for experiments in panel A (“×” for R-only, squares for mixed populations; note that one yellow square falls below the axis limits). Dotted (dashed) curves are predictions from theoretical model for mixed (R-only) populations. (C) Example long-term (35+hours) experiment in the regime in which Pmax is larger than the steady-state population density. As in panel A, growth curves become transparent after crossing threshold. In all curves, density is measured relative to carrying capacity in a unperturbed bioreactors (see S2 Fig). Data are deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943 [62]. Pmax, acceptable burden; R, resistant; S, sensitive.

Discussion

In this work, we provide direct experimental evidence that the presence of drug-sensitive cells can lead to improved antibiotic-driven control of bacterial populations in vitro. Specifically, we show that a “competition-maximizing” strategy can contain mixed populations of sensitive and resistant cells below a threshold density for significantly longer than matched populations containing only resistant cells. The increase in escape time occurs only when the threshold density is sufficiently high that competition is significant. The findings are particularly remarkable given that experiments are performed in well-mixed bioreactors with continuous resource renewal, and even the highest density thresholds occur in the exponential growth regime for unperturbed populations. The surprisingly strong effect of competition under these conditions suggests that similar approaches may yield even more dramatic results in natural environments, in which spatial heterogeneity and limited diffusion may enhance competition [6367].

Notably, our experiments did not uncover scenarios in which sensitive cells may actually be detrimental and accelerate the expansion of resistance. Theory suggests that these scenarios do indeed exist [45], but because of the typical mutation rates observed in bacteria, they cannot be reliably produced with our experimental system (see S2 Text for extended discussion). Indeed, the dose-response curves of isolates from the beginning of the experiment were similar to isolates taken a day later at the end of the experiment (S5 Fig), further supporting the idea that mutation plays a negligible role in this model. Although reservoir sizes and contamination risks limited the length of our experiments, it is possible that accumulated mutations over longer periods of time could change the average characteristics of the two populations. We note that we did observe similar dynamics (i.e., containment consistent with model predictions) in an isolated experiment run for more than 35 hours (Fig 5C).

Theory indicates that whether or not containment is better than elimination depends on a number of factors, including the frequency of resistance and how resistance impacts the basic growth (intrinsic fitness costs) and competitive ability (competitive fitness costs) of the resistant population (S6 Fig and [45]). These factors are important because they affect the relative amounts of competition and mutational input. Here, we have fixed all of these factors and manipulated only the acceptable burden Pmax, which we find must be sufficiently large to get significant benefit. In general, the threshold that is “high enough” will depend on the details of these other factors in potentially complex ways, and confirming these relationships experimentally is an exciting avenue for future work. Additionally, although in our experiment the mixed populations eventually became dominated by drug-resistant bacteria, in practice this may not always be the case. For certain combinations of fitness costs and competitive abilities, containment could drive the resistant population to very low levels. Importantly—even if this is not the case—our experimental results suggest that there are situations in which containment can extend the time before the resistant population dominates and treatment failure occurs.

It is important to keep in mind several technical limitations of our study. First, we measured population density using light scattering (OD), which is a widely used experimental surrogate for microbial population size but is sensitive to changes in cell shape [68]. Because we use protein synthesis inhibitors primarily at sub–minimum inhibitory concentrations, we do not anticipate significant artifacts from this limitation, though it may pose challenges when trying to extend these results to drugs such as fluoroquinolones, which are known to induce filamentation [69, 70]. In addition, in the absence of cell lysis, OD cannot distinguish between dead and living cells. However, our experiments include a slow background flow that adds fresh media and removes waste, leading to a clear distinction between nongrowing and growing populations. Under these conditions, fully inhibited (or dead) populations would experience a decrease in OD over time, whereas populations maintained at a constant density are required to divide at an effective rate equal to this background refresh rate.

Most importantly, our results are based entirely on in vitro experiments, which allow for precise environmental control and quantitative measurements but clearly lack important complexities of realistic in vivo and clinical scenarios. Developing drug protocols for clinical use is an extremely challenging problem. Our goal was not to design clinically realistic containment strategies but, instead, was to provide proof-of-principle experimental evidence that protocols aimed at maximally maintaining sensitive cells can contain resistance. The drug dosing protocol applied here attempts to supply the minimum possible amount of drug to control the population. However, the results show that it sometimes reduces the size of the sensitive population more than necessary, suggesting that our protocol is suboptimal, and therefore, our experimental results are likely underestimating the potential benefit of maximizing the sensitive population. The fact that we are still able to detect a benefit to maintaining a (slightly nonmaximal) sensitive population indicates that there is room for implementing these types of strategies in the nonidealized setting of real life. In fact, adaptive strategies designed to leverage (but not maximize) competition between drug-sensitive and drug-resistant cancer cells have been successful in both mouse models [42] and clinical cancer trials [46]. Our results suggest that these successes may be further improved by modifying treatment strategies to further increase competition—working toward competition-maximizing strategies.

We do not want to make a strong case that containment itself will soon become a widespread treatment strategy. The idea of a clinically acceptable pathogen burden will make many clinicians uneasy and, in many cases (e.g., bacterial meningitis), there is no acceptable burden. But in some settings, containment is not completely far-fetched. In acute infections, containment would be a temporary strategy, enough to relieve symptoms until immunity controls the infection [43]. Moreover, there is ample justification for the idea of an acceptable burden in nonsterile sites (asymptomatic bacteriuria, gastrointestinal bacteria) and increasing evidence that a low burden of pathogen can be tolerated even in the lung or blood [71, 72]. Tolerance-promoting and antivirulence drugs are increasingly being sought [7375], and these work by alleviating symptoms rather than pathogens. Additionally, there are settings in which adding drug-sensitive microbes is being considered (microbiome or bacteriotherapy [76]) or, in the case of fecal transplants, being enacted [77]. Moreover, in chronic infections with uncontrolled source populations, in which the evolution of resistance is the main threat to patient well-being [56], it is very possible that containment could be a key part of more complex resistance-controlling regimens. In cancer, in which resistance is responsible for many deaths, Gatenby and colleagues have argued that containment may better prolong life [31, 78, 79].

A basic science understanding of containment has the potential for real-world impact, as containment strategies are being tested on people now. If our hypothesis is confirmed, there are definable situations in which existing strategies could be improved by maximizing competition as opposed to simply using “less aggressive” adaptive approaches. We hope these experiments will motivate continued experimental, theoretical, and perhaps even clinical investigations, particularly in situations in which the primary threat to the well-being of the patient is resistance-induced treatment failure (e.g., [46, 56]).

Methods

Bacterial strains, media, and growth conditions

Experiments were performed with Escherichia coli strains REL606 and REL607 [59]. A resistant strain (“resistant mutant”) was isolated from lab-evolved populations of REL606 undergoing daily dilutions (200×) into fresh media with increasing doxycycline (Research Products International) concentrations for 3 days. A single resistant isolate was used for all experiments. Stock solutions were frozen at −80°C in 30% glycerol and streaked onto fresh agar plates (Davis Minimal Media [Sigma] with 2,000 g/ml glucose) as needed. Overnight cultures of resistant and sensitive cells for each experiment were grown from single colonies and then incubated in sterile Davis Minimal Media with 1,000 g/ml glucose liquid media overnight at 30°C while rotating at 240 rpm. All bioreactor experiments were performed in a temperature-controlled warm room at 30°C.

Continuous culture bioreactors

Experiments were performed in custom-built, computer-controlled bioreactors as described in [58], which are based, in part, on similar designs from [57, 80]. Briefly, constant-volume bacterial cultures (17 mL) are grown in glass vials with customized Teflon tops that allow inflow and outflow of fluid via silicone tubing. Flow is managed by a series of computer-controlled peristaltic pumps—up to 6 per vial—which are connected to media and drug reservoirs and allow for precise control of various environmental conditions. Cell density is monitored by light scattering using infrared LED/detector pairs on the side of each vial holder. Voltage readings are converted to OD using a calibration curve based on separate readings with a tabletop OD reader. Up to nine cultures can be grown simultaneously using a series of multiposition magnetic stirrers. The entire system is controlled by custom Matlab software.

Experimental mixtures and setup

Before the experiments begin, vials are seeded with sensitive or resistant strains of E. coli and allowed to grow to the desired density in the bioreactor vials. Cells were then mixed (to create the desired population compositions) and diluted as appropriate to achieve the desired starting densities. Each vial is connected to (1) a drug reservoir containing media and doxycycline (500 μg/ml), (2) a drug-free media reservoir that provides constant renewal of media, and (3) an effluent waste reservoir. Flow from reservoir 1 (drug reservoir) is determined in real time according to a simple feedback algorithm intended to maintain cells at a constant target density with minimal drug. Flow to/from reservoirs 2 and 3 provides a slow renewal of media and nutrients while maintaining a constant culture volume in each vial.

Drug dosing protocols

To determine the appropriate antibiotic dosing strategy, the computer records the OD in each vial every 3 seconds. Every 3 minutes, the computer computes (1) the average OD, ODavg, in the mixed vial over the last 30 seconds and (2) the current drug concentration in the vial. If ODavg is greater than Pmax, the desired containment density, and the current drug concentration is less than dmax = 125 ng/mL (dmax = 100 ng/mL for the experiments used to generate Fig 5), then drug and media will be added to the vial for 21 seconds at a flow rate of 1 mL per minute. For the experiments corresponding to Fig 5, the drug threshold was lowered to 100 ng/mL to increase the long-term steady state of the system—allowing escape times for containment to be measured for a wider range of Pmax values. In a typical experiment, this control algorithm is applied to one of the mixed populations to determine, in real time, the drug dosing protocol (i.e., influx of drug solution over time). The exact same drug dosing protocol is then simultaneously applied to all experimental populations (mixed, resistant-only, sensitive-only, Fig 3). In parallel, an identical dosing protocol is applied to a series of control populations, but in these populations, the drug solution is replaced by drug-free media (S3 Text and S3 Fig). Finally, each experiment includes an unperturbed (no flow, no drug) control vial containing only resistant cells. To minimize effects of day-to-day fluctuations (in temperature, media batch, etc.), we measure population density in Fig 5 in units of carrying capacity estimated from these control populations each day.

Whole-genome sequencing

Genomic DNA was isolated from single colony isolates using a Quick-DNA Fungal/Bacterial Kit (Zymo Reserach) according to the manufacturer’s instructions. Libraries were prepared using the Swift 2S turbo flexible library prep kit and the swift normalase kit to normalize and pool libraries for sequencing. Sequencing was performed on the NovaSeq-6000, with 150-bp paired-end reads. Mutations were identified by mapping the Illumina sequencing reads to the reference strain REL606 (accession number NC_012967) using breseq [81].

Sequencing confirmed that the sensitive clone is identical to the published REL606 sequences (accession number NC_012967). The resistant strain has five mutations: OmpF P138H (CCT→CAT) porin mutation, a 1-bp deletion in fimbrial biogenesis outer membrane usher protein, an intergenic (+275/−54) C→T mutation between methylenetetrahydrofolate reductase/catalase/peroxidase HPI, an IS1 insertion into multidrug efflux transporter transcriptional repressor AcrR, and an IS150 mediated deletion of part of the ribose operon that has been previously described in the LTEE and confers a slight fitness advantage under those conditions [82]. Mutations in OmpF and AcrR genes have been previously linked with increased resistance to multiple antibiotics, including tetracyclines [60, 61].

Mathematical model

The mathematical model used in the simulations is

S˙=rS1+(D(tτS)hS)kS(1S+RC)SFDχD+FNVS,
R˙=rR1+(D(tτR)hR)kR(1S+RC)RFDχD+FNVR,
R˙only=rR1+(D(tτR)hR)kR(1RonlyC)RonlyFDχD+FNVRonly,
S˙only=rS1+(D(tτS)hS)kS(1SonlyC)SonlyFDχD+FNVSonly,
D˙=FDχDVDinFDχD+FNVD, (1)

where S and R are the drug-sensitive and drug-resistant densities in the mixed vial, Ronly is the bacterial density in the vial that contains only drug-resistant bacteria, Sonly is the bacterial density in the vial that contains only drug-sensitive bacteria, and D is the drug concentration in the vials. The model parameters and initial conditions for the simulations (and experiments) are given in Tables 12. The effect of drug on growth rate is modeled as a Hill function with parameters rS, kS, and hS for the sensitive strain and parameters rR, kR, and hR for the resistant strain. There is also a time delay associated with the effect of drug (denoted by τS for the sensitive strain and τR for the resistant strain). Competition in the model is captured by using a logistic growth term with carrying capacity C. It is assumed that the sensitive and resistant strains have similar carrying capacities. Finally, the bioreactor has a continual efflux to maintain constant volume. The rate of this outflow is the sum of the constant background nutrient flow FN and any additional outflow required to compensate for the inflow of drug, which enters at a rate FDχD. FD is a constant rate, and χD is an indicator function, which is 1 when drug is being added to the vials and 0 when it is not. In the simulations, the decision of when to add drug is based on the same control algorithm that was used in the actual experiment (see Methods: Drug dosing protocols). Since the model describes the rate of change of bacterial density, the total efflux (FN+FDχD) is divided by the volume of the vials V. The drug concentration in the vials is determined by the rate of drug flow into the vials (FDχDDin, where Din is the concentration of drug in the reservoir) and the rate of efflux out of the vials (FN+FDχD). The values of Din, V, FD, and FN were chosen to match the associated values in the experimental system; all other parameters in the model were fit using independent experimental data (see S3 Text for details) and are given in Table 1.

Table 1. Model parameter description.

Parameter Definition Value
V Volume of vial 17 mL
FD Flow rate of drug reservoir 1 mLmin
χD Function indicating when drug is being added 1 when drug is being added
FN Constant background flow rate of nutrients 0.067 mLmin
Din Drug concentration in drug reservoir 500 ngmL
C Carrying capacity Range: (2.4 × 108)–(3.2 × 108) cellsmL
(OD: 0.3–0.4)*
rS Intrinsic per capita growth rate of drug-sensitive strain 0.0169 1min
rR Intrinsic per capita growth rate of drug-resistant strain 0.0152 1min
hS IC50 for sensitive strain 49.0639 ngmL
hR IC50 for resistant strain 209.9995 ngmL
kS Hill function coefficient 2.2023
kR Hill function coefficient 2.4849
τS Time delay for sensitive strain 79.04 minutes
τR Time delay for resistant strain 96.72 minutes

*Each experiment includes a drug-free control vial with no inflow or outflow.

Carrying capacity is estimated daily from this growth curve.

Abbreviation: IC50, half-maximal inhibitory concentration; OD, optical density

Table 2. Initial optical densities for simulations and experiments.

Pmax D(0) S(0) R(0) Sonly(0) Ronly(0)
High 0.2 0 0.175 0.02 0.175 0.02
Low 0.1 0 0.075 0.02 0.075 0.02

Multiply optical densities by 8 × 108 to obtain cells per mL.

Abbreviation: Pmax, acceptable burden

Supporting information

S1 Text. Details on parameter estimation.

(PDF)

S2 Text. Details on mutation.

(PDF)

S3 Text. Drug-free controls.

(PDF)

S1 Fig. Computer-controlled bioreactors.

Constant-volume bacterial cultures (17 mL) are grown in glass vials with customized Teflon tops that allow inflow and outflow of fluid via silicone tubing. Flow is managed by a series of computer-controlled peristaltic pumps that are connected to media and drug reservoirs. Cell density is monitored by light scattering using infrared LED/detector pairs on the side of each vial holder. Voltage readings are converted to OD using a calibration curve based on separate readings with a tabletop OD reader. Up to nine cultures can be grown simultaneously using a series of multiposition magnetic stirrers. The entire system is controlled by custom Matlab software. Flow chart (above) depicts adaptive drug therapy (lower branches) intended to maintain constant OD by adding drug in response to changes in cell density. LED, light-emitting diode; OD, optical density.

(PNG)

S2 Fig. Growth of resistant cells in unperturbed bioreactors.

Cell density (OD) over time for REL607-derived resistant strains in bioreactors without influx or outflow of media. Transparent black lines correspond to growth curves performed in parallel with each bioreactor experiment. Thick black curve is the median over replicates. Dashed lines indicate threshold densities used in experiments (Pmax = 0.2 and Pmax = 0.1). Data are deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943 [62]. OD, optical density; Pmax, acceptable burden.

(PDF)

S3 Fig. Matched drug-free control populations are not contained by adaptive dosing protocol.

Conditions are identical to those in Fig 3B and 3C except that all populations receive drug-free media rather than drug solution media as part of the adaptive dosing protocol. Data are deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943 [62].

(PDF)

S4 Fig. Mixed populations contain primarily resistant cells at final time point of escape time experiment.

The REL606-derived resistant strain appears red, and the sensitive REL607 strain appears pink when grown on TA plates. Upper panels: samples from two mixed vials taken at the end of a high-density escape time experiment (as in Fig 3B). Arrows indicate sensitive colonies. Bottom row: samples from the end of a high-density escape time experiment for a vial seeded with only resistant bacteria (left) and a vial seeded with only sensitive bacteria (right). TA, tetrazolium arabinose.

(PDF)

S5 Fig. Sensitive and resistant isolates show similar dose-response curves before and after experiment.

Dose-response curves measured in 96-well microplates for sensitive (REL607; blue circles) and REL606-derived resistant strains (red circles). Dark line, mean across replicates. Thin (transparent) curves correspond to colonies isolated from population mixture at the end of an escape time experiment. Red curve, resistant isolate (appears red on plate); blue curves, sensitive isolates (appear pink on plate). Data are deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943 [62].

(PDF)

S6 Fig. Amount of benefit from containment depends on multiple factors.

Simulation showing fold increase in escape time gained by using containment instead of elimination. Each shaded region corresponds to a different Pmax (green, red, and blue are 20%, 30%, and 40% of the carrying capacity, respectively). Upper bounds of each shaded region correspond to an intrinsic fitness cost for resistance of 25% (rR = rS(0.75)), and lower bounds assume no fitness cost (rR = rS). Simulation uses mathematical model from main text and parameter values given in Table 1 (except for rR, which is modified as described above). Trends show that increasing the intrinsic fitness cost and decreasing the frequency of resistance will increase the benefit of containment. Importantly, these simulations assume that there is no mutation. The role of fitness costs and frequency of resistance are more complicated when there is appreciable mutational input. Data are deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943 [62]. Pmax, acceptable burden.

(JPG)

Abbreviations

LTEE

long-term evolution experiment

OD

optical density

Pmax

acceptable burden

TA

tetrazolium arabinose

Data Availability

Data deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943.

Funding Statement

This work is supported by the National Science Foundation (NSF No. 1553028 to KBW), the National Institutes of Health (NIH No. 1R35GM124875-01 to KBW; NIH No. R01 GM089932 to AFR; NIH K08 AI119182 to RJW), the Hartwell Foundation for Biomedical Research (to KBW), and the Eberly Family (to AFR). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Neu HC. The Crisis in Antibiotic Resistance. Science. 1992;257(5073):1064–1073. 10.1126/science.257.5073.1064 [DOI] [PubMed] [Google Scholar]
  • 2.Spellberg B, Blaser M, Guidos RJ, Boucher HW, Bradley JS, et al. ; Infectious Diseases Society of America (IDSA). Combating antimicrobial resistance: policy recommendations to save lives. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America. 2011;52 Suppl 5:S397–428. 10.1093/cid/cir153 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Dryden M, Johnson AP, Ashiru-Oredope D, Sharland M. Using antibiotics responsibly: right drug, right time, right dose, right duration. The Journal of Antimicrobial Chemotherapy. 2011;66(11):2441–2443. 10.1093/jac/dkr370 [DOI] [PubMed] [Google Scholar]
  • 4.Brown ED, Wright GD. Antibacterial drug discovery in the resistance era. Nature. 2016;529(7586):336–343. 10.1038/nature17042 [DOI] [PubMed] [Google Scholar]
  • 5.Laxminarayan R, Matsoso P, Pant S, Brower C, Røttingen JA, Klugman K, et al. Access to effective antimicrobials: a worldwide challenge. The Lancet. 2016;387(10014):168–175. 10.1016/S0140-6736(15)00474-2 [DOI] [PubMed] [Google Scholar]
  • 6.Marston HD, Dixon DM, Knisely JM, Palmore TN, Fauci AS. Antimicrobial Resistance. JAMA. 2016;316(11):1193–1204. 10.1001/jama.2016.11764 [DOI] [PubMed] [Google Scholar]
  • 7.Madaras-Kelly KJ, Demasters TA. In vitro characterization of fluoroquinolone concentration/MIC antimicrobial activity and resistance while simulating clinical pharmacokinetics of levofloxacin, ofloxacin, or ciprofloxacin against Streptococcus pneumoniae. Diagnostic Microbiology and Infectious Disease. 2000;37(4):253–260. 10.1016/s0732-8893(00)00147-4 [DOI] [PubMed] [Google Scholar]
  • 8.Craig WA. Re-evaluating current antibiotic therapy. Respiratory Medicine. 2001;95:S12–S19. 10.1016/S0954-6111(01)90023-X [DOI] [PubMed] [Google Scholar]
  • 9.Mehrotra R, De Gaudio R, Palazzo M. Antibiotic pharmacokinetic and pharmacodynamic considerations in critical illness. Intensive Care Medicine. 2004;30(12):2145–2156. 10.1007/s00134-004-2428-9 [DOI] [PubMed] [Google Scholar]
  • 10.Drlica K, Zhao X. Mutant selection window hypothesis updated. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America. 2007;44(5):681–688. 10.1086/511642 [DOI] [PubMed] [Google Scholar]
  • 11.Roberts JA, Kruger P, Paterson DL, Lipman J. Antibiotic resistance–what's dosing got to do with it? Critical Care Medicine. 2008;36(8):2433–2440. 10.1097/CCM.0b013e318180fe62 [DOI] [PubMed] [Google Scholar]
  • 12.Owens RC, Shorr AF. Rational dosing of antimicrobial agents: pharmacokinetic and pharmacodynamic strategies. American journal of health-system pharmacy: AJHP: official journal of the American Society of Health-System Pharmacists. 2009;66(12 Suppl 4):S23–30. 10.2146/090087d [DOI] [PubMed] [Google Scholar]
  • 13.Cantón R, Morosini MI. Emergence and spread of antibiotic resistance following exposure to antibiotics. FEMS microbiology reviews. 2011;35(5):977–991. 10.1111/j.1574-6976.2011.00295.x [DOI] [PubMed] [Google Scholar]
  • 14.Yoshida K, Niki Y, Okimoto N, Kishimoto M, Fukano H, Hara H, et al. Efficacy and safety of moxifloxacin for community-acquired bacterial pneumonia based on pharmacokinetic analysis. Journal of Infection and Chemotherapy. 2011;17(5):678–685. 10.1007/s10156-011-0282-6 [DOI] [PubMed] [Google Scholar]
  • 15.Downes KJ, Hahn A, Wiles J, Courter JD, Vinks AA. Dose optimisation of antibiotics in children: application of pharmacokinetics/pharmacodynamics in paediatrics. International journal of antimicrobial agents. 2014;43(3):223–230. 10.1016/j.ijantimicag.2013.11.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Marcusson LL, Komp Lindgren P, Olofsson SK, Hughes D, Cars O. Mutant prevention concentrations of pradofloxacin for susceptible and mutant strains of Escherichia coli with reduced fluoroquinolone susceptibility. International Journal of Antimicrobial Agents. 2014;44(4):354–357. 10.1016/j.ijantimicag.2014.06.010 [DOI] [PubMed] [Google Scholar]
  • 17.Abdul-Aziz MH, Lipman J, Mouton JW, Hope WW, Roberts JA. Applying pharmacokinetic/pharmacodynamic principles in critically ill patients: optimizing efficacy and reducing resistance development. Seminars in Respiratory and Critical Care Medicine. 2015;36(1):136–153. 10.1055/s-0034-1398490 [DOI] [PubMed] [Google Scholar]
  • 18.Balzarini J, Pelemans H, Karlsson A, De ClercQ E, Kleim JP. Concomitant combination therapy for HIV infection preferable over sequential therapy with 3TC and non-nucleoside reverse transcriptase inhibitors. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(23):13152–13157. 10.1073/pnas.93.23.13152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Chait R, Craney A, Kishony R. Antibiotic interactions that select against resistance. Nature. 2007;446(7136):668–671. 10.1038/nature05685 [DOI] [PubMed] [Google Scholar]
  • 20.Abbate E, Vescovo M, Natiello M, Cufré M, García A, Gonzalez Montaner P, et al. Successful alternative treatment of extensively drug-resistant tuberculosis in Argentina with a combination of linezolid, moxifloxacin and thioridazine. The Journal of Antimicrobial Chemotherapy. 2012;67(2):473–477. 10.1093/jac/dkr500 [DOI] [PubMed] [Google Scholar]
  • 21.Lienhardt C, Raviglione M, Spigelman M, Hafner R, Jaramillo E, Hoelscher M, et al. New drugs for the treatment of tuberculosis: needs, challenges, promise, and prospects for the future. The Journal of Infectious Diseases. 2012;205 Suppl 2:S241–249. 10.1093/infdis/jis034 [DOI] [PubMed] [Google Scholar]
  • 22.Horwitz JA, Halper-Stromberg A, Mouquet H, Gitlin AD, Tretiakova A, Eisenreich TR, et al. HIV-1 suppression and durable control by combining single broadly neutralizing antibodies and antiretroviral drugs in humanized mice. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(41):16538–16543. 10.1073/pnas.1315295110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Baym M, Stone LK, Kishony R. Multidrug evolutionary strategies to reverse antibiotic resistance. Science (New York, NY). 2016;351(6268):aad3292 10.1126/science.aad3292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Dean Z, Maltas J, Wood KB. Antibiotic interactions shape short-term evolution of resistance in E. faecalis. PLoS Pathog 2020; 16(3): e1008278 10.1371/journal.ppat.1008278 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.McAdams D, Wollein Waldetoft K, Tedijanto C, Lipsitch M, Brown SP. Resistance diagnostics as a public health tool to combat antibiotic resistance: A model-based evaluation. PLoS Biol. 2019;17(5). 10.1371/journal.pbio.3000250 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Reaz AF, Taylor LH. The ecology of genetically diverse infections. Science (New York, NY). 2001;292(5519):1099–1102. [DOI] [PubMed] [Google Scholar]
  • 27.Roche-Lestienne C, Preudhomme C. Mutations in the ABL kinase domain pre-exist the onset of imatinib treatment. Seminars in Hematology. 2003;40(2 Suppl 2):80–82. 10.1053/shem.2003.50046 [DOI] [PubMed] [Google Scholar]
  • 28.Greaves M, Maley CC. Clonal evolution in cancer. Nature. 2012;481(7381):306–313. 10.1038/nature10762 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kommineni S, Bretl DJ, Lam V, Chakraborty R, Hayward M, Simpson P, et al. Bacteriocin production augments niche competition by enterococci in the mammalian gastrointestinal tract. Nature. 2015;526(7575):719–722. 10.1038/nature15524 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hsu PP, Sabatini DM. Cancer cell metabolism: Warburg and beyond. Cell. 2008;134(5):703–707. 10.1016/j.cell.2008.08.021 [DOI] [PubMed] [Google Scholar]
  • 31.Gillies RJ, Verduzco D, Gatenby RA. Evolutionary dynamics of carcinogenesis and why targeted therapy does not work. Nature Reviews Cancer. 2012;12(7):487–493. 10.1038/nrc3298 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ross-Gillespie A, Weigert M, Brown SP, Kümmerli R. Gallium-mediated siderophore quenching as an evolutionarily robust antibacterial treatment. Evolution, Medicine, and Public Health. 2014;2014(1):18–29. 10.1093/emph/eou003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Garber K. Targeting copper to treat breast cancer. Science. 2015;349(6244):128–129. 10.1126/science.349.6244.128 [DOI] [PubMed] [Google Scholar]
  • 34.Balmer O, Tanner M. Prevalence and implications of multiple-strain infections. The Lancet Infectious Diseases. 2011;11(11):868–878. 10.1016/S1473-3099(11)70241-9 [DOI] [PubMed] [Google Scholar]
  • 35.Day T, Huijben S, Read AF. Is selection relevant in the evolutionary emergence of drug resistance? Trends in Microbiology. 2015;23(3):126–133. 10.1016/j.tim.2015.01.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Mideo N. Parasite adaptations to within-host competition. Trends in Parasitology. 2009;25(6):261–268. 10.1016/j.pt.2009.03.001 [DOI] [PubMed] [Google Scholar]
  • 37.Wale N, Sim DG, Read AF. A nutrient mediates intraspecific competition between rodent malaria parasites in vivo. Proceedings of the Royal Society B: Biological Sciences. 2017;284(1859). 10.1098/rspb.2017.1067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Geli P, Laxminarayan R, Dunne M, Smith DL. “One-Size-Fits-All”? Optimizing Treatment Duration for Bacterial Infections. PLoS ONE. 2012;7(1). 10.1371/journal.pone.0029838 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Colijn C, Cohen T. How competition governs whether moderate or aggressive treatment minimizes antibiotic resistance. eLife. 2015;4:e10559 10.7554/eLife.10559 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Fuentes-Hernandez A, Plucain J, Gori F, Pena-Miller R, Reding C, Jansen G, et al. Using a Sequential Regimen to Eliminate Bacteria at Sublethal Antibiotic Dosages. PLoS Biol. 2015;13(4):e1002104 10.1371/journal.pbio.1002104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Day T, Read AF. Does High-Dose Antimicrobial Chemotherapy Prevent the Evolution of Resistance? PLoS Comput Biol. 2016;12(1):e1004689 10.1371/journal.pcbi.1004689 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Enriquez-Navas PM, Kam Y, Das T, Hassan S, Silva A, Foroutan P, et al. Exploiting evolutionary principles to prolong tumor control in preclinical models of breast cancer. Science Translational Medicine. 2016;8(327):327ra24 10.1126/scitranslmed.aad7842 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Gjini E, Brito PH. Integrating Antimicrobial Therapy with Host Immunity to Fight Drug-Resistant Infections: Classical vs. Adaptive Treatment. PLoS Comput Biol. 2016;12(4):e1004857 10.1371/journal.pcbi.1004857 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bauer M, Graf IR, Ngampruetikorn V, Stephens GJ, Frey E. Exploiting ecology in drug pulse sequences in favour of population reduction. PLoS Comput Biol. 2017;13(9):e1005747 10.1371/journal.pcbi.1005747 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Hansen E, Woods RJ, Read AF. How to use a chemotherapeutic agent when resistance to it threatens the patient. PLoS Biol. 2017;15(2):e2001110 10.1371/journal.pbio.2001110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Zhang J, Cunningham JJ, Brown JS, Gatenby RA. Integrating evolutionary dynamics into treatment of metastatic castrate-resistant prostate cancer. Nature Communications. 2017;8(1):1816 10.1038/s41467-017-01968-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.West J, Ma Y, Newton PK. Capitalizing on competition: An evolutionary model of competitive release in metastatic castration resistant prostate cancer treatment. Journal of Theoretical Biology. 2018;455:249–260. 10.1016/j.jtbi.2018.07.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.West J, Newton PK. Cellular interactions constrain tumor growth. Proceedings of the National Academy of Sciences of the United States of America. 2019;116(6):1918–1923. 10.1073/pnas.1804150116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Newton PK, Ma Y. Nonlinear adaptive control of competitive release and chemotherapeutic resistance. Physical Review E. 2019;99(2):022404 10.1103/PhysRevE.99.022404 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wargo AR, Huijben S, de Roode JC, Shepherd J, Read AF. Competitive release and facilitation of drug-resistant parasites after therapeutic chemotherapy in a rodent malaria model. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(50):19914–19919. 10.1073/pnas.0707766104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Huijben S, Nelson WA, Wargo AR, Sim DG, Drew DR, Read AF. Chemotherapy, within-host ecology and the fitness of drug-resistant malaria parasites. Evolution; International Journal of Organic Evolution. 2010;64(10):2952–2968. 10.1111/j.1558-5646.2010.01068.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Read AF, Day T, Huijben S. The evolution of drug resistance and the curious orthodoxy of aggressive chemotherapy. Proceedings of the National Academy of Sciences of the United States of America. 2011;108 Suppl 2:10871–10877. 10.1073/pnas.1100299108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Huijben S, Bell AS, Sim DG, Tomasello D, Mideo N, Day T, et al. Aggressive Chemotherapy and the Selection of Drug Resistant Pathogens. PLoS Pathog. 2013;9(9):e1003578 10.1371/journal.ppat.1003578 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Kouyos RD, Metcalf CJE, Birger R, Klein EY, Abel zur Wiesch P, Ankomah P, et al. The path of least resistance: aggressive or moderate treatment? Proceedings Biological Sciences. 2014;281(1794):20140566 10.1098/rspb.2014.0566 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Pollitt LC, Huijben S, Sim DG, Salathé RM, Jones MJ, Read AF. Rapid Response to Selection, Competitive Release and Increased Transmission Potential of Artesunate-Selected Plasmodium chabaudi Malaria Parasites. PLoS Pathog. 2014;10(4):e1004019 10.1371/journal.ppat.1004019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Woods RJ, Read AF. Clinical management of resistance evolution in a bacterial infection: A case study. Evolution, Medicine, and Public Health. 2015;2015(1):281–288. 10.1093/emph/eov025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Toprak E, Veres A, Michel JB, Chait R, Hartl DL, Kishony R. Evolutionary paths to antibiotic resistance under dynamically sustained drug selection. Nature Genetics. 2012;44(1):101–105. 10.1038/ng.1034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Karslake J, Maltas J, Brumm P, Wood KB. Population Density Modulates Drug Inhibition and Gives Rise to Potential Bistability of Treatment Outcomes for Bacterial Infections. PLoS Comput Biol. 2016;12(10):e1005098 10.1371/journal.pcbi.1005098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Lenski RE, Rose MR, Simpson SC, Tadler SC. Long-Term Experimental Evolution in Escherichia coli. I. Adaptation and Divergence During 2,000 Generations. The American Naturalist. 1991;138(6):1315–1341. [Google Scholar]
  • 60.Alekshun MN, Levy SB. Regulation of chromosomally mediated multiple antibiotic resistance: the mar regulon. Antimicrobial agents and chemotherapy. 1997;41(10):2067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cohen SP, McMurry L, Hooper D, Wolfson J, Levy S. Cross-resistance to fluoroquinolones in multiple-antibiotic-resistant (Mar) Escherichia coli selected by tetracycline or chloramphenicol: decreased drug accumulation associated with membrane changes in addition to OmpF reduction. Antimicrobial agents and chemotherapy. 1989;33(8):1318–1325. 10.1128/aac.33.8.1318 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hansen E, Karslake J, Woods RJ, Read AF, Wood KB. Data from: Antibiotics can be used to contain drug-resistant bacteria by maintaining sufficiently large sensitive populations. 2020. Dryad Digital Repository. Available from: 10.5061/dryad.s4mw6m943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.González-García I, Solé RV, Costa J. Metapopulation dynamics and spatial heterogeneity in cancer. Proceedings of the National Academy of Sciences. 2002;99(20):13085–13089. 10.1073/pnas.202139299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Liu W, Røder HL, Madsen JS, Bjarnsholt T, Sørensen SJ, Burmølle M. Interspecific Bacterial Interactions are Reflected in Multispecies Biofilm Spatial Organization. Frontiers in Microbiology. 2016;7 10.3389/fmicb.2016.01366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Steenackers HP, Parijs I, Foster KR, Vanderleyden J. Experimental evolution in biofilm populations. FEMS Microbiology Reviews. 2016;40(3):373–397. 10.1093/femsre/fuw002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Fusco D, Gralka M, Kayser J, Anderson A, Hallatschek O. Excess of mutational jackpot events in expanding populations revealed by spatial Luria-Delbrück experiments. Nature Communications. 2016;7(1):1–9. 10.1038/ncomms12760 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Bacevic K, Noble R, Soffar A, Wael Ammar O, Boszonyik B, Prieto S, et al. Spatial competition constrains resistance to targeted cancer therapy. Nature Communications. 2017;8(1):1995 10.1038/s41467-017-01516-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Stevenson K, McVey AF, Clark IBN, Swain PS, Pilizota T. General calibration of microbial growth in microplate readers. Scientific Reports. 2016;6:38828 10.1038/srep38828 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Diver JM, Wise R. Morphological and biochemical changes in Escherichia coli after exposure to ciprofloxacin. Journal of Antimicrobial Chemotherapy. 1986;18(Supplement_D):31–41. 10.1093/jac/18.Supplement-D.31 [DOI] [PubMed] [Google Scholar]
  • 70.Wickens HJ, Pinney RJ, Mason DJ, Gant VA. Flow Cytometric Investigation of Filamentation, Membrane Patency, and Membrane Potential in Escherichia coli following Ciprofloxacin Exposure. Antimicrobial Agents and Chemotherapy. 2000;44(3):682–687. 10.1128/aac.44.3.682-687.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Nicolle LE, Bradley S, Colgan R, Rice JC, Schaeffer A, Hooton TM. Infectious Diseases Society of America Guidelines for the Diagnosis and Treatment of Asymptomatic Bacteriuria in Adults. Clinical Infectious Diseases. 2005;40(5):643–654. 10.1086/427507 [DOI] [PubMed] [Google Scholar]
  • 72.Morris A, Beck JM, Schloss PD, Campbell TB, Crothers K, Curtis JL, et al. Comparison of the respiratory microbiome in healthy nonsmokers and smokers. American Journal of Respiratory and Critical Care Medicine. 2013;187(10):1067–1075. 10.1164/rccm.201210-1913OC [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Rasko DA, Sperandio V. Anti-virulence strategies to combat bacteria-mediated disease. Nature Reviews Drug Discovery. 2010;9(2):117–128. 10.1038/nrd3013 [DOI] [PubMed] [Google Scholar]
  • 74.Bender KO, Garland M, Ferreyra JA, Hryckowian AJ, Child MA, Puri AW, et al. A small-molecule antivirulence agent for treating Clostridium difficile infection. Science Translational Medicine. 2015;7(306):306ra148 10.1126/scitranslmed.aac9103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Garland M, Loscher S, Bogyo M. Chemical Strategies To Target Bacterial Virulence. Chemical Reviews. 2017;117(5):4422–4461. 10.1021/acs.chemrev.6b00676 [DOI] [PubMed] [Google Scholar]
  • 76.Ouwehand AC, Forssten S, Hibberd AA, Lyra A, Stahl B. Probiotic approach to prevent antibiotic resistance. Annals of Medicine. 2016;48(4):246–255. 10.3109/07853890.2016.1161232 [DOI] [PubMed] [Google Scholar]
  • 77.Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A, et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature. 2015;517(7533):205–208. 10.1038/nature13828 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Gatenby RA. A change of strategy in the war on cancer. Nature. 2009;459:508–509. 10.1038/459508a [DOI] [PubMed] [Google Scholar]
  • 79.Gatenby RA, Silva AS, Gillies RJ, Frieden BR. Adaptive therapy. Cancer Research. 2009;69(11):4894–4903. 10.1158/0008-5472.CAN-08-3658 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Toprak E, Veres A, Yildiz S, Pedraza JM, Chait R, Paulsson J, et al. Building a morbidostat: an automated continuous-culture device for studying bacterial drug resistance under dynamically sustained drug inhibition. Nature Protocols. 2013;8(3):555–567. 10.1038/nprot.nprot.2013.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Deatherage DE, Barrick JE. Identification of mutations in laboratory-evolved microbes from next-generation sequencing data using breseq In: Engineering and analyzing multicellular systems. Springer; 2014. p. 165–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Cooper VS, Schneider D, Blot M, Lenski RE. Mechanisms causing rapid and parallel losses of ribose catabolism in evolving populations of Escherichia coli B. Journal of Bacteriology. 2001;183(9):2834–2841. 10.1128/JB.183.9.2834-2841.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Roland G Roberts

14 Jun 2019

Dear Dr Wood,

Thank you for submitting your manuscript entitled "Antibiotics can be used to contain drug-resistant bacteria by maintaining sufficiently large sensitive populations" for consideration as a Research Article by PLOS Biology.

Your manuscript has now been evaluated by the PLOS Biology editorial staff, as well as by an academic editor with relevant expertise, and I'm writing to let you know that we would like to send your submission out for external peer review.

However, before we can send your manuscript to reviewers, we need you to complete your submission by providing the metadata that is required for full assessment. To this end, please login to Editorial Manager where you will find the paper in the 'Submissions Needing Revisions' folder on your homepage. Please click 'Revise Submission' from the Action Links and complete all additional questions in the submission questionnaire.

**Important**: Please also see below for further information regarding completing the MDAR reporting checklist. The checklist can be accessed here: https://plos.io/MDARChecklist

Please re-submit your manuscript and the checklist, within two working days, i.e. by Jun 18 2019 11:59PM.

Login to Editorial Manager here: https://www.editorialmanager.com/pbiology

During resubmission, you will be invited to opt-in to posting your pre-review manuscript as a bioRxiv preprint. Visit http://journals.plos.org/plosbiology/s/preprints for full details. If you consent to posting your current manuscript as a preprint, please upload a single Preprint PDF when you re-submit.

Once your full submission is complete, your paper will undergo a series of checks in preparation for peer review. Once your manuscript has passed all checks it will be sent out for review.

Feel free to email us at plosbiology@plos.org if you have any queries relating to your submission.

Kind regards,

Roli Roberts

Roland G Roberts, PhD,

Senior Editor

PLOS Biology

==================

INFORMATION REGARDING THE REPORTING CHECKLIST:

PLOS Biology is pleased to support the "minimum reporting standards in the life sciences" initiative (https://osf.io/preprints/metaarxiv/9sm4x/). This effort brings together a number of leading journals and reproducibility experts to develop minimum expectations for reporting information about Materials (including data and code), Design, Analysis and Reporting (MDAR) in published papers. We believe broad alignment on these standards will be to the benefit of authors, reviewers, journals and the wider research community and will help drive better practise in publishing reproducible research.

We are therefore participating in a community pilot involving a small number of life science journals to test the MDAR checklist. The checklist is intended to help authors, reviewers and editors adopt and implement the minimum reporting framework.

IMPORTANT: We have chosen your manuscript to participate in this trial. The relevant documents can be located here:

MDAR reporting checklist (to be filled in by you): https://plos.io/MDARChecklist

**We strongly encourage you to complete the MDAR reporting checklist and return it to us with your full submission, as described above. We would also be very grateful if you could complete this author survey:

https://forms.gle/seEgCrDtM6GLKFGQA

Additional background information:

Interpreting the MDAR Framework: https://plos.io/MDARFramework

Please note that your completed checklist and survey will be shared with the minimum reporting standards working group. However, the working group will not be provided with access to the manuscript or any other confidential information including author identities, manuscript titles or abstracts. Feedback from this process will be used to consider next steps, which might include revisions to the content of the checklist. Data and materials from this initial trial will be publicly shared in September 2019. Data will only be provided in aggregate form and will not be parsed by individual article or by journal, so as to respect the confidentiality of responses.

Please treat the checklist and elaboration as confidential as public release is planned for September 2019.

We would be grateful for any feedback you may have.

Decision Letter 1

Roland G Roberts

28 Jul 2019

Dear Dr Wood,

Thank you very much for submitting your manuscript "Antibiotics can be used to contain drug-resistant bacteria by maintaining sufficiently large sensitive populations" for consideration as a Research Article at PLOS Biology. Your manuscript has been evaluated by the PLOS Biology editors, an Academic Editor with relevant expertise, and by three independent reviewers.

The reviews of your manuscript are appended below. You will see that the reviewers find the work potentially interesting. However, based on their specific comments and following discussion with the academic editor, I regret that we cannot accept the current version of the manuscript for publication. We remain interested in your study and we would be willing to consider resubmission of a comprehensively revised version that thoroughly addresses all the reviewers' comments. We cannot make any decision about publication until we have seen the revised manuscript and your response to the reviewers' comments. Your revised manuscript would be sent for further evaluation by the reviewers.

We appreciate that these requests represent a great deal of extra work, and we are willing to relax our standard revision time to allow you six months to revise your manuscript. Please email us (plosbiology@plos.org) to discuss this if you have any questions or concerns, or think that you would need longer than this. At this stage, your manuscript remains formally under active consideration at our journal; please notify us by email if you do not wish to submit a revision and instead wish to pursue publication elsewhere, so that we may end consideration of the manuscript at PLOS Biology.

Your revisions should address the specific points made by each reviewer. Please submit a file detailing your responses to the editorial requests and a point-by-point response to all of the reviewers' comments that indicates the changes you have made to the manuscript. In addition to a clean copy of the manuscript, please upload a 'track-changes' version of your manuscript that specifies the edits made. This should be uploaded as a "Related" file type. You should also cite any additional relevant literature that has been published since the original submission and mention any additional citations in your response.

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

Before you revise your manuscript, please review the following PLOS policy and formatting requirements checklist PDF: http://journals.plos.org/plosbiology/s/file?id=9411/plos-biology-formatting-checklist.pdf. It is helpful if you format your revision according to our requirements - should your paper subsequently be accepted, this will save time at the acceptance stage.

Please note that as a condition of publication PLOS' data policy (http://journals.plos.org/plosbiology/s/data-availability) requires that you make available all data used to draw the conclusions arrived at in your manuscript. If you have not already done so, you must include any data used in your manuscript either in appropriate repositories, within the body of the manuscript, or as supporting information (N.B. this includes any numerical values that were used to generate graphs, histograms etc.). For an example see here: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5.

For manuscripts submitted on or after 1st July 2019, we require the original, uncropped and minimally adjusted images supporting all blot and gel results reported in an article's figures or Supporting Information files. We will require these files before a manuscript can be accepted so please prepare them now, if you have not already uploaded them. Please carefully read our guidelines for how to prepare and upload this data: https://journals.plos.org/plosbiology/s/figures#loc-blot-and-gel-reporting-requirements.

Upon resubmission, the editors will assess your revision and if the editors and Academic Editor feel that the revised manuscript remains appropriate for the journal, we will send the manuscript for re-review. We aim to consult the same Academic Editor and reviewers for revised manuscripts but may consult others if needed.

If you still intend to submit a revised version of your manuscript, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' where you will find your submission record.

Thank you again for your submission to our journal. We hope that our editorial process has been constructive thus far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Roli Roberts

Roland G Roberts, PhD,

Senior Editor

PLOS Biology

*****************************************************

REVIEWERS' COMMENTS:

Reviewer #1:

The authors develop a computer controled bioreactor experimental model to assess whether a strategy of ‘containment’ (drug treatment only to limit growth below a threshold) can increase time to escape from drug control. They provide exciting experimental support for the containment strategy, illustrating a significant delay to population growth under a containment regime (compared to a limit case of total S annihilation) – even when the containment target is substantially below the bacterial carrying capacity.

These results present an important avenue for future development. My two main issues with the current MS are related and fixable primarily in the discussion – first, how could a containment strategy be implemented in the clinic? Second, how does a containment strategy relate to other conditional strategies and to an unconditional low dose strategy?

1) implementation.

The experimental feedback control mechanism doesn’t appear to be a plausible clinical strategy (and I’m fine with the proof of concept goals) – but this leaves the key question of translation. What do the authors see as a path to capturing elements of this feedback control in the clinic? It might be interesting at this point to borrow from analogous approaches in the cancer adaptive therapy field, which I believe is more developed. Does the approach work, given uncertainty over the R/S composition of the infection? How can clinicians track infection growth? Imaging? Symptoms??

2) relationship to other conditional/unconditional strategies

A full clinical implementation of this experimental model would require information on the initial resistance distributions and also the temporal dynamics of an infection – this is a tough goal (see point 1). However just having resistance profiles alone could open simple and effective treatments – use a drug that is effective against both co-infecting strains. (https://journals.plos.org/plosbiology/article?id=10.1371/journal.pbio.3000250). In light of this simpler strategy (treat conditional on resistances), why invest in containment? One argument is pan-resistant strains – conditioning on resistance is no longer effective, and conditioning on pathogen growth might buy time for … immune control? It would also be useful to discuss relationships with the most simple and low tech strategy of reducing drug dose – which will tend to reduce competitive release. This would be an interesting avenue for a control experiment / simulation (same total drug, constant administration)

other concerns:

Fig 4: please use time in hours on y axis. The timescale here is important in my view to discuss. You’re potentially adding hours to the control window – not a substantial time period. This ties into another key translational theme – what is the clinical advantage of adding hours to the window of time until loss of infection control? In an acute infection context this might be of practical significance for development of adaptive immune control – not sure here, would be interested in authors thoughts.

Related to timescale issue - what were the obstacles to running the experiment for longer? It would have been useful to compare the ultimate escape outcomes in all treatments and replicates. More data on longer timecourses would be helpful- the authors should at least comment on what the road block was to longer implementation of this device.

Minor:

How resistant is the ‘R’ E. coli strain? Sounds like a low ½ concentration. Not a big concern as this is plainly a ‘proof of concept’ implementation.

Reviewer #2:

In the light of the worldwide spread of multi-drug resistant bacterial pathogens, it is important to develop new approaches that limit the spread of superbugs and make current antibiotic therapies more sustainable. The paper by Hansen et al. contributes to this aim and asks whether less aggressive antibiotic treatments, allowing competition between susceptible and resistant strains could repress overall population growth, thereby keeping pathogen burden below an acceptable level and extend treatment efficacy.

I concur with the authors that inter-strain competition could be a powerful way to limit the spread of antibiotic resistant strains. But unfortunately the experimental design and the data presented are quite weak, and I am not convinced that this study will extend our knowledge on the role of competition in making antibiotic therapies more sustainable.

The authors performed bioreactor (chemostat) experiments with Escherichia coli bacteria. They have three treatments: (1) a monoculture of a susceptible strain, (2) a monoculture of an antibiotic resistant strain, and (3) a mixture of the two. Through the automated adjustment of the flow in the bioreactor, population growth rate can be kept constant under a range of antibiotic concentrations. The authors chose two arbitrary “acceptable” pathogen levels (OD = 0.225 and OD = 0.125), and measured the time for the treatments needed to surpass this threshold. The time needed to do so is called “escape time” and is interpreted as time until treatment failure. The authors found that (i) the escape time was very short for the antibiotic resistant strain, (ii) sensitive populations declined under treatment and never surpassed the threshold, and (iii) mixed populations had either an extended escape time or did not surpass the threshold. The authors conclude that competition helps to keep bacterial load at a manageable level. While I have no problem with these results, I question the overall relevance of the finding.

(1) The authors consider treatment 2 (resistant bacteria only) as a “mutation minimization” treatment, matching aggressive antibiotic treatment. However, aggressive treatment is thought to minimize the risk of resistance to arise in susceptible populations and not in already resistant populations as implemented by the authors. In this context, it is not surprising that resistant monocultures grow best. Anything else would be surprising, and this treatment does not match a “mutation minimization” treatment as one would deploy in clinical settings.

(2) The authors consider treatment 3 as a “competition maximization” treatment, and conclude that competition is only efficient in extending escape time at higher population densities. I have multiple problems with this conclusion. (a) Higher bacterial burden is likely bad for the patient and this is not a practicable way to take. (b) the chosen thresholds are arbitrary and the growth increase observed for the mixed treatment under low density (Fig. 3C) is minimal. It surpasses the threshold, yes, but compared to the resistant only treatment, mixed cultures are still much more stable and show a dramatically lower growth increase. Thus, competition also has an effect at low cell density. (c) the observed effects are transient. The authors plated the mixed cultures at the end of their experiment and found that populations are dominated by resistant clones. This means that in the long-run competition cannot constrain the spread of resistance.

(3) The advantage of in vitro proof-of-concept studies is that there is full control over the experimental set up and a large number of conditions and treatment combinations can be tested. Unfortunately, the authors did not make use of this advantage and the data presented is thin both in terms of quality and quantity. (a) The authors use an undefined laboratory evolved antibiotic resistant mutant. We don’t know the resistant mechanism involved and the potential mechanisms of competition that could drive interaction patterns in mixed cultures. (b) Only one antibiotic was used for hypothesis testing. So we don’t learn whether the observed patterns are of general relevance or specific to this antibiotic. (c) Only two populations densities were examined and we thus don’t learn what the overall relationship between escape time and population density is. This is particularly problematic because OD = 0.2 is called high density although in absolute terms OD = 0.2 is still very low density. From a proof-of-concept study, which is far away from anything that is clinically relevant, I would expect a much larger parameter space to be explored, especially with the mathematical model at hand.

More minor comments:

- It is unclear what should be extracted from the left panels of Figure 2. Is this just to demonstrate that real-time growth rates can be measured in the chemostats? This is quite trivial because this is what chemostats are made for.

- Why does the resistant strain treatment start with a much lower OD than the other two treatments? This induces a massive bias towards longer escape times for this treatment.

- page 2, line 3: “aggressive treatment may actually promote the emergence of resistance” sounds like the treatment itself will actively give raise to resistance mutation. Consider rephrasing to “promote selection for resistance” or “promote the spread of resistance”

- end of page 2 and beginning of page 3: I don’t think that statements like “this is the first explicit demonstration” and “we are also unaware of any direct tests …” really help to increase the impact/relevance of the paper. Please remove.

- page 3: “the celebrated long term evolution experiment” reads odd. Please be more objective.

Reviewer #3:

The aim of this study is to provide a proof-of-concept experimental illustration that under given conditions, containment (i.e. more moderate) drug regimens can better manage antibiotic resistance than elimination (i.e. aggressive) strategies. For this the authors design three scenarios in an in-vitro experimental setup with automated drug and nutrient delivery to study the effect of treatment strategy on resistance emergence: i) resistant-only (no competition), ii) resistant+sensitive cells (competition scenario) and iii) sensitive-only dynamics. They set the drug concentration adaptively in the mixed scenario, to maintain the population at a certain density, and apply the same drug concentration to the other homogeneous populations. The readout for time of escape is the time it takes each population to cross a given target threshold density. The study finds that in the mixed case, the time of escape is higher than in the resistant only case, thus indicating the important role of competition in delaying resistance selection in real settings. This finding is also corroborated by a mathematical model. The authors go on to test this effect at another population size, and report that at lower population sizes the benefit of competition in reducing resistance is lower. Overall, I consider the study interesting and novel in the combination of ecological theory, experimental in vitro-approach with computer-controlled bioreactors and mathematical modeling. Indeed, the question of optimal drug therapies is a very urgent one in the face of antibiotic resistance. Previous mathematical models using population dynamic arguments have shown that competition between drug-sensitive and drug-resistant pathogens can slow down the selection of resistance. This particular study goes one step further by providing experimental validation of this expectation. However, there are some parts of the study and analysis that I find confusing and that I would need the authors to clarify.

1. I am a bit confused by the authors' focus and repeated reference to "emergence of resistance" and "escape time", since they do not talk explicitly about the mechanisms underlying this escape. Is it because of selection of pre-existing sub-populations, or because of further or de-novo mutations? In particular, are the mechanisms of "escape" the same in the Resistant-only, mixed case, and Sensitive-only scenarios? In general I guess not, and I would invite the authors to comment a bit more on this biological aspect. If I understand it correctly, in the Resistant-only scenario, the process governing escape is basically growth of resistant cells under sub-inhibitory antibiotics and the time it takes to exceed a certain population size threshold. In the mixed case, escape is basically due to the selection of the resistant sub-population in the context of a heterogeneous population, where the majority of sensitive cells by competitive suppression, delay any growth advantage of the resistant minority in the presence of the drug. In the sensitive-only case, escape can only happen if there is an allegedly very rare beneficial mutation conferring drug resistance in the time frame of the experiment followed by subsequent growth. Thus, there should be no emergence per se in this study, only selection. This is also confirmed by the dose-response curve for R-only population at the end of the experiment, which is the same as the original strain.

2. Another issue the authors seem to overlook is the fact that mutational processes can happen both in the sensitive cells and already resistant cells. There are many studies that show that drug-resistant cells may experience even higher mutation rates that effectively accelerate further resistance evolution. In the present study, this process is completely neglected. Why?

3. Regarding, the definition and approach to competition adopted in this study, I would also invite the authors to elaborate more and be more explicit on:

i) the confounding between population size and heterogeneity in their 3 scenarios: Besides the fact that we are dealing with a homogeneous vs. heterogeneous population, there is also a big difference in initial population size in the R-only (much lower than Pmax) and mixed case (equal to Pmax). This by default creates an advantage for the population of cells growing in the R-only regime. And this advantage is higher, the more space there is for growth from P(0) to Pmax. Thus, in one case we are dealing with a growing ('epidemic scenario') population, in the other case, we are dealing with an 'endemic' scenario, of constant population size and simple replacement. It is known also from epidemiological models and studies that patterns of evolution can be different.

ii) What is the rationale behind equalizing the drug concentrations across the scenarios? By equalizing drug concentrations (set by the mixed regime) the authors are comparing dynamics using minimal inhibitory treatment vs. sub-inhibitory treatment relatively in the "mixed/sensitive-only" and R-only scenarios. Thus, unlike the schematic shown in Figure 1A, the resistant-only scenario, is experiencing much lower concentration of drug than its corresponding aggressive regime, amplifying the total growth advantage relative to the mixed case. Thus, not only they are starting with dR/dt>0, not only resistant cells are not competing with sensitive cells, they are also under less drug-inhibition overall. It seems that the authors are simulating in the R-only regime a treatment of the type "assumed prior aggressive treatment that removed the S population + moderate treatment when there is only the resistant population left".

iii) Competition maximization to slow down resistance is one thing, infection clearance is another. What are the novel prospects for infection clearance generated by this study? In light of this work, could addition of drug-sensitive cells to a drug-resistant infection be used to increase competition and leverage its benefits?

4. It is necessary for this study to address more clearly what the effect of containment strategies is along the 2-d continuum: pop. size vs. initial frequency of R. The authors explore indeed two population sizes (achieved with different drug dosing), but these correspond at the same time to two different initial frequencies of resistance in the bacterial population, namely 10% in the high population size and 20% in the small population size. Thus, we cannot really disentangle whether the sensitivity of escape time is a population size effect or frequency-dependent effect, or of both. The higher the initial frequency of resistance in the mixed case (i.e. here the lower the Pmax), the closer we are to the R-only extreme, and that is why no difference is found in "escape time." I recommend the authors to comment on this issue when interpreting their results.

In my view, a contour plot of the model-derived expected time to escape as a function of both population size and initial frequency would be an important and insightful addition to the paper. I would even suggest to provide two versions of such contour plot, for example by changing the growth differential between the sensitive and resistant strains or efficacy of antibiotic. In this way, it will become clear that the quantitative benefit of containment does not depend just on population size (as suggested in the paper and abstract), but is indeed much more complex, and may vary also with all these other parameters and initial conditions.

5. The worst-case scenario for containment is when the total bacterial population is resistant (frequency of resistance=1) and when the population size is large. This scenario cannot be tested with the current framework because the maximum dose feasible is well below the half-maximum inhibitory concentration for the resistant population (129<209). Can the authors comment on this scenario? What would be the role of competition in that case?

Since this study assumes that sensitive and resistant cells compete equally, can we expect a positive effect of competition also in the R-only adaptive regime? Would a moderate regime delay further resistance evolution compared to an aggressive regime?

6. Some more links with existing models from the literature are warranted, e.g. Colijn and Cohen (eLife 2015, https://doi.org/10.7554/eLife.10559.001) where competition between a drug-sensitive strain and drug-resistant strain is modeled in more detail. Similarly, Gjini & Brito (Plos Comp. Biol. 2016 https://doi.org/10.1371/journal.pcbi.1004857) also consider in detail adaptive treatment, and along the lines of this study, conclude that at higher symptom thresholds, where indirect competition via immune stimulation is maximized, resistance selection risk is also lower, as opposed to lower symptom thresholds. This may be relevant for in vivo settings where antigen-dependent immunity acts as another layer of competition on heterogeneous bacterial populations.

Similarly, some discussion on the possibility of asymmetric competition is also needed. Here it is assumed that the carrying capacities are the same for both S and R. Indeed, if the competition were asymmetric and S could inhibit S more or less than inhibiting R, and so on (there are in principle 4 pairwise competitive interactions), additional indirect effects could manifest on R dynamics due to S removal by the drug. This phenomenon has been discussed widely in pneumococcus epidemiology and serotype replacement due to vaccination, where asymmetric competition can accelerate or slow down non-vaccine type 'selection' (similar to R selection in the mixed experimental setting of this study).

Minor comments:

- Why is the main result in Figure 4 not divided in Probability of Escape and Time of Escape | Escape?

- In Table 2, the R-only and S-only initial conditions have to be corrected. Please also put the initial conditions in terms of bacterial concentrations as required by the differential equation model.

- More details on the Model fitting and parameter estimation, especially the delays associated with drug effect on resistant strain and sensitive strain are needed. How important are the assumed differences in these delays?

-Figure 3 should be accompanied in the Supplements with the corresponding underlying dynamics of replacement of S by R in the mixed regime during drug administration. The R-dynamics in that case could be compared with the R-only dynamics by overlaying the two graphs.

- In Figure 3, please clarify what generates the variability in model predictions. What is the stochastic component of the simulations?

Decision Letter 2

Roland G Roberts

20 Mar 2020

Dear Dr Wood,

Thank you for submitting your revised Research Article entitled "Antibiotics can be used to contain drug-resistant bacteria by maintaining sufficiently large sensitive populations" for publication in PLOS Biology. I have now obtained advice from two of the original reviewers and have discussed their comments with the Academic Editor.

Based on the reviews, we will probably accept this manuscript for publication, assuming that you will modify the manuscript to address the remaining points raised by the reviewers. Please also make sure to address the data and other policy-related requests noted at the end of this email.

Specifically, please address the remaining requests from reviewer #3 regarding the Supplementary Figs, and attend to my Data Policy request below.

We expect to receive your revised manuscript within two weeks. Your revisions should address the specific points made by each reviewer. In addition to the remaining revisions and before we will be able to formally accept your manuscript and consider it "in press", we also need to ensure that your article conforms to our guidelines. A member of our team will be in touch shortly with a set of requests. As we can't proceed until these requirements are met, your swift response will help prevent delays to publication.

*Copyediting*

Upon acceptance of your article, your final files will be copyedited and typeset into the final PDF. While you will have an opportunity to review these files as proofs, PLOS will only permit corrections to spelling or significant scientific errors. Therefore, please take this final revision time to assess and make any remaining major changes to your manuscript.

NOTE: If Supporting Information files are included with your article, note that these are not copyedited and will be published as they are submitted. Please ensure that these files are legible and of high quality (at least 300 dpi) in an easily accessible file format. For this reason, please be aware that any references listed in an SI file will not be indexed. For more information, see our Supporting Information guidelines:

https://journals.plos.org/plosbiology/s/supporting-information

*Published Peer Review History*

Please note that you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out. Please see here for more details:

https://blogs.plos.org/plos/2019/05/plos-journals-now-open-for-published-peer-review/

*Early Version*

Please note that an uncorrected proof of your manuscript will be published online ahead of the final version, unless you opted out when submitting your manuscript. If, for any reason, you do not want an earlier version of your manuscript published online, uncheck the box. Should you, your institution's press office or the journal office choose to press release your paper, you will automatically be opted out of early publication. We ask that you notify us as soon as possible if you or your institution is planning to press release the article.

*Protocols deposition*

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosbiology/s/submission-guidelines#loc-materials-and-methods

*Submitting Your Revision*

To submit your revision, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' to find your submission record. Your revised submission must include a cover letter, a Response to Reviewers file that provides a detailed response to the reviewers' comments (if applicable), and a track-changes file indicating any changes that you have made to the manuscript.

Please do not hesitate to contact me should you have any questions.

Sincerely,

Roli Roberts

Roland G Roberts, PhD,

Senior Editor

PLOS Biology

------------------------------------------------------------------------

DATA POLICY:

You may be aware of the PLOS Data Policy, which requires that all data be made available without restriction: http://journals.plos.org/plosbiology/s/data-availability. For more information, please also see this editorial: http://dx.doi.org/10.1371/journal.pbio.1001797

Note that we do not require all raw data. Rather, we ask that all individual quantitative observations that underlie the data summarized in the figures and results of your paper be made available in one of the following forms:

1) Supplementary files (e.g., excel). Please ensure that all data files are uploaded as 'Supporting Information' and are invariably referred to (in the manuscript, figure legends, and the Description field when uploading your files) using the following format verbatim: S1 Data, S2 Data, etc. Multiple panels of a single or even several figures can be included as multiple sheets in one excel file that is saved using exactly the following convention: S1_Data.xlsx (using an underscore).

2) Deposition in a publicly available repository. Please also provide the accession code or a reviewer link so that we may view your data before publication.

Regardless of the method selected (we note that you have expressed an intention to deposit your data in Dryad), please ensure that you provide the individual numerical values that underlie the summary data displayed in the following figure panels as they are essential for readers to assess your analysis and to reproduce it: Figs 2, 3, 4, 5, S2, S3, S4, S6, NOTE: the numerical data provided should include all replicates AND the way in which the plotted mean and errors were derived (it should not present only the mean/average values).

Please also ensure that figure legends in your manuscript include information on where the underlying data can be found, and ensure your supplemental data file/s has a legend.

Please ensure that your Data Statement in the submission system accurately describes where your data can be found.

------------------------------------------------------------------------

REVIEWERS' COMMENTS:

Reviewer #1:

The authors have made valuable edits in response to my earlier comments. I have no further concerns.

minor: fix typo in title of figure 5 (which is a very nice addition)

Reviewer #3:

I appreciate the effort gone into this revision and the clarifications made by the authors to address most of the points raised. I do believe this has improved substantially the manuscript. While still some caveats remain, the authors have mentioned them more transparently in this version, opening the space for future work to fill the gaps. Overall, this is a valuable contribution to the empirical understanding of competition and intervention processes on the dynamics of bacterial populations, which may help address therapeutic challenges for resistant infections in clinical settings.

Some final comments below:

- As a further clarification on Figure 3, the provided Figure S4 is not what I had in mind when I asked for a comparison between Resistant dynamics visualization in the R-only vs mixed case. In its current version, I don't find figure S4 informative. I understand the explicit R-dynamics in the mixed case may be inaccessible in retrospect, thus making my request unfeasible with the current data, where only total population size is tracked over time. I would recommend to remove figure S4.

- I would appreciate seeing the figure about Frequency of Resistance and escape time in the response file (response to my previous comment 4) as an added Figure to the Supplements, with appropriate referencing in the main text.

- I suggest removing unnecessary brackets from the model equations (e.g. from the numerator in some fractions), this would make the reading of the model less cumbersome.

Decision Letter 3

Roland G Roberts

23 Apr 2020

Dear Dr Wood,

On behalf of my colleagues and the Academic Editor, David S. Schneider, I am pleased to inform you that we will be delighted to publish your Research Article in PLOS Biology.

The files will now enter our production system. You will receive a copyedited version of the manuscript, along with your figures for a final review. You will be given two business days to review and approve the copyedit. Then, within a week, you will receive a PDF proof of your typeset article. You will have two days to review the PDF and make any final corrections. If there is a chance that you'll be unavailable during the copy editing/proof review period, please provide us with contact details of one of the other authors whom you nominate to handle these stages on your behalf. This will ensure that any requested corrections reach the production department in time for publication.

Early Version

The version of your manuscript submitted at the copyedit stage will be posted online ahead of the final proof version, unless you have already opted out of the process. The date of the early version will be your article's publication date. The final article will be published to the same URL, and all versions of the paper will be accessible to readers.

PRESS

We frequently collaborate with press offices. If your institution or institutions have a press office, please notify them about your upcoming paper at this point, to enable them to help maximise its impact. If the press office is planning to promote your findings, we would be grateful if they could coordinate with biologypress@plos.org. If you have not yet opted out of the early version process, we ask that you notify us immediately of any press plans so that we may do so on your behalf.

We also ask that you take this opportunity to read our Embargo Policy regarding the discussion, promotion and media coverage of work that is yet to be published by PLOS. As your manuscript is not yet published, it is bound by the conditions of our Embargo Policy. Please be aware that this policy is in place both to ensure that any press coverage of your article is fully substantiated and to provide a direct link between such coverage and the published work. For full details of our Embargo Policy, please visit http://www.plos.org/about/media-inquiries/embargo-policy/.

Thank you again for submitting your manuscript to PLOS Biology and for your support of Open Access publishing. Please do not hesitate to contact me if I can provide any assistance during the production process.

Kind regards,

Vita Usova

Publication Assistant,

PLOS Biology

on behalf of

Roland Roberts,

Senior Editor

PLOS Biology

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Text. Details on parameter estimation.

    (PDF)

    S2 Text. Details on mutation.

    (PDF)

    S3 Text. Drug-free controls.

    (PDF)

    S1 Fig. Computer-controlled bioreactors.

    Constant-volume bacterial cultures (17 mL) are grown in glass vials with customized Teflon tops that allow inflow and outflow of fluid via silicone tubing. Flow is managed by a series of computer-controlled peristaltic pumps that are connected to media and drug reservoirs. Cell density is monitored by light scattering using infrared LED/detector pairs on the side of each vial holder. Voltage readings are converted to OD using a calibration curve based on separate readings with a tabletop OD reader. Up to nine cultures can be grown simultaneously using a series of multiposition magnetic stirrers. The entire system is controlled by custom Matlab software. Flow chart (above) depicts adaptive drug therapy (lower branches) intended to maintain constant OD by adding drug in response to changes in cell density. LED, light-emitting diode; OD, optical density.

    (PNG)

    S2 Fig. Growth of resistant cells in unperturbed bioreactors.

    Cell density (OD) over time for REL607-derived resistant strains in bioreactors without influx or outflow of media. Transparent black lines correspond to growth curves performed in parallel with each bioreactor experiment. Thick black curve is the median over replicates. Dashed lines indicate threshold densities used in experiments (Pmax = 0.2 and Pmax = 0.1). Data are deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943 [62]. OD, optical density; Pmax, acceptable burden.

    (PDF)

    S3 Fig. Matched drug-free control populations are not contained by adaptive dosing protocol.

    Conditions are identical to those in Fig 3B and 3C except that all populations receive drug-free media rather than drug solution media as part of the adaptive dosing protocol. Data are deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943 [62].

    (PDF)

    S4 Fig. Mixed populations contain primarily resistant cells at final time point of escape time experiment.

    The REL606-derived resistant strain appears red, and the sensitive REL607 strain appears pink when grown on TA plates. Upper panels: samples from two mixed vials taken at the end of a high-density escape time experiment (as in Fig 3B). Arrows indicate sensitive colonies. Bottom row: samples from the end of a high-density escape time experiment for a vial seeded with only resistant bacteria (left) and a vial seeded with only sensitive bacteria (right). TA, tetrazolium arabinose.

    (PDF)

    S5 Fig. Sensitive and resistant isolates show similar dose-response curves before and after experiment.

    Dose-response curves measured in 96-well microplates for sensitive (REL607; blue circles) and REL606-derived resistant strains (red circles). Dark line, mean across replicates. Thin (transparent) curves correspond to colonies isolated from population mixture at the end of an escape time experiment. Red curve, resistant isolate (appears red on plate); blue curves, sensitive isolates (appear pink on plate). Data are deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943 [62].

    (PDF)

    S6 Fig. Amount of benefit from containment depends on multiple factors.

    Simulation showing fold increase in escape time gained by using containment instead of elimination. Each shaded region corresponds to a different Pmax (green, red, and blue are 20%, 30%, and 40% of the carrying capacity, respectively). Upper bounds of each shaded region correspond to an intrinsic fitness cost for resistance of 25% (rR = rS(0.75)), and lower bounds assume no fitness cost (rR = rS). Simulation uses mathematical model from main text and parameter values given in Table 1 (except for rR, which is modified as described above). Trends show that increasing the intrinsic fitness cost and decreasing the frequency of resistance will increase the benefit of containment. Importantly, these simulations assume that there is no mutation. The role of fitness costs and frequency of resistance are more complicated when there is appreciable mutational input. Data are deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943 [62]. Pmax, acceptable burden.

    (JPG)

    Attachment

    Submitted filename: ReviewerResponseFinal.pdf

    Attachment

    Submitted filename: ReviewerResponseRd2.pdf

    Data Availability Statement

    Data deposited in the Dryad repository: https://doi.org/10.5061/dryad.s4mw6m943.


    Articles from PLoS Biology are provided here courtesy of PLOS

    RESOURCES