Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
1.
Nat Biotechnol ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965430

ABSTRACT

Most targeted anticancer therapies fail due to drug resistance evolution. Here we show that tumor evolution can be reproducibly redirected to engineer therapeutic opportunity, regardless of the exact ensemble of pre-existing genetic heterogeneity. We develop a selection gene drive system that is stably introduced into cancer cells and is composed of two genes, or switches, that couple an inducible fitness advantage with a shared fitness cost. Using stochastic models of evolutionary dynamics, we identify the design criteria for selection gene drives. We then build prototypes that harness the selective pressure of multiple approved tyrosine kinase inhibitors and employ therapeutic mechanisms as diverse as prodrug catalysis and immune activity induction. We show that selection gene drives can eradicate diverse forms of genetic resistance in vitro. Finally, we demonstrate that model-informed switch engagement effectively targets pre-existing resistance in mouse models of solid tumors. These results establish selection gene drives as a powerful framework for evolution-guided anticancer therapy.

2.
Virus Evol ; 10(1): vead084, 2024.
Article in English | MEDLINE | ID: mdl-38516655

ABSTRACT

In secondary lymphoid tissues, human immunodeficiency virus (HIV) can replicate in both the follicular and extrafollicular compartments. Yet, virus is concentrated in the follicular compartment in the absence of antiretroviral therapy, in part due to the lack of cytotoxic T lymphocyte (CTL)-mediated activity there. CTLs home to the extrafollicular compartment, where they can suppress virus load to relatively low levels. We use mathematical models to show that this compartmentalization can explain seemingly counter-intuitive observations. First, it can explain the observed constancy of the viral decline slope during antiviral therapy in the peripheral blood, irrespective of the presence of CTL in Simian Immunodeficiency Virus (SIV)-infected macaques, under the assumption that CTL-mediated lysis significantly contributes to virus suppression. Second, it can account for the relatively long times it takes for CTL escape mutants to emerge during chronic infection even if CTL-mediated lysis is responsible for virus suppression. The reason is the heterogeneity in CTL activity and the consequent heterogeneity in selection pressure between the follicular and extrafollicular compartments. Hence, to understand HIV dynamics more thoroughly, this analysis highlights the importance of measuring virus populations separately in the extrafollicular and follicular compartments rather than using virus load in peripheral blood as an observable; this hides the heterogeneity between compartments that might be responsible for the particular patterns seen in the dynamics and evolution of the HIV in vivo.

3.
Nat Commun ; 14(1): 6642, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37863909

ABSTRACT

The literature about mutant invasion and fixation typically assumes populations to exist in isolation from their ecosystem. Yet, populations are part of ecological communities, and enemy-victim (e.g. predator-prey or pathogen-host) interactions are particularly common. We use spatially explicit, computational pathogen-host models (with wild-type and mutant hosts) to re-visit the established theory about mutant fixation, where the pathogen equally attacks both wild-type and mutant individuals. Mutant fitness is assumed to be unrelated to infection. We find that pathogen presence substantially weakens selection, increasing the fixation probability of disadvantageous mutants and decreasing it for advantageous mutants. The magnitude of the effect rises with the infection rate. This occurs because infection induces spatial structures, where mutant and wild-type individuals are mostly spatially separated. Thus, instead of mutant and wild-type individuals competing with each other, it is mutant and wild-type "patches" that compete, resulting in smaller fitness differences and weakened selection. This implies that the deleterious mutant burden in natural populations might be higher than expected from traditional theory.


Subject(s)
Ecosystem , Models, Biological , Humans , Probability , Population Dynamics
4.
J Evol Biol ; 36(2): 444-460, 2023 02.
Article in English | MEDLINE | ID: mdl-36514852

ABSTRACT

Mutant dynamics in fragmented populations have been studied extensively in evolutionary biology. Yet, open questions remain, both experimentally and theoretically. Some of the fundamental properties predicted by models still need to be addressed experimentally. We contribute to this by using a combination of experiments and theory to investigate the role of migration in mutant distribution. In the case of neutral mutants, while the mean frequency of mutants is not influenced by migration, the probability distribution is. To address this empirically, we performed in vitro experiments, where mixtures of GFP-labelled ("mutant") and non-labelled ("wid-type") murine cells were grown in wells (demes), and migration was mimicked via cell transfer from well to well. In the presence of migration, we observed a change in the skewedness of the distribution of the mutant frequencies in the wells, consistent with previous and our own model predictions. In the presence of de novo mutant production, we used modelling to investigate the level at which disadvantageous mutants are predicted to exist, which has implications for the adaptive potential of the population in case of an environmental change. In panmictic populations, disadvantageous mutants can persist around a steady state, determined by the rate of mutant production and the selective disadvantage (selection-mutation balance). In a fragmented system that consists of demes connected by migration, a steady-state persistence of disadvantageous mutants is also observed, which, however, is fundamentally different from the mutation-selection balance and characterized by higher mutant levels. The increase in mutant frequencies above the selection-mutation balance can be maintained in small ( N < N c ) demes as long as the migration rate is sufficiently small. The migration rate above which the mutants approach the selection-mutation balance decays exponentially with N / N c . The observed increase in the mutant numbers is not explained by the change in the effective population size. Implications for evolutionary processes in diseases are discussed, where the pre-existence of disadvantageous drug-resistant mutant cells or pathogens drives the response of the disease to treatments.


Subject(s)
Models, Genetic , Selection, Genetic , Animals , Mice , Mutation , Population Dynamics , Biological Evolution
5.
Bull Math Biol ; 84(12): 144, 2022 11 05.
Article in English | MEDLINE | ID: mdl-36334172

ABSTRACT

It is well known in the literature that human behavior can change as a reaction to disease observed in others, and that such behavioral changes can be an important factor in the spread of an epidemic. It has been noted that human behavioral traits in disease avoidance are under selection in the presence of infectious diseases. Here, we explore a complementary trend: the pathogen itself might experience a force of selection to become less "visible," or less "symptomatic," in the presence of such human behavioral trends. Using a stochastic SIR agent-based model, we investigated the co-evolution of two viral strains with cross-immunity, where the resident strain is symptomatic while the mutant strain is asymptomatic. We assumed that individuals exercised self-regulated social distancing (SD) behavior if one of their neighbors was infected with a symptomatic strain. We observed that the proportion of asymptomatic carriers increased over time with a stronger effect corresponding to higher levels of self-regulated SD. Adding mandated SD made the effect more significant, while the existence of a time-delay between the onset of infection and the change of behavior reduced the advantage of the asymptomatic strain. These results were consistent under random geometric networks, scale-free networks, and a synthetic network that represented the social behavior of the residents of New Orleans.


Subject(s)
Epidemics , Models, Biological , Humans , Mathematical Concepts
6.
PLoS Comput Biol ; 18(5): e1010039, 2022 05.
Article in English | MEDLINE | ID: mdl-35522694

ABSTRACT

Feedback mechanisms within cell lineages are thought to be important for maintaining tissue homeostasis. Mathematical models that assume well-mixed cell populations, together with experimental data, have suggested that negative feedback from differentiated cells on the stem cell self-renewal probability can maintain a stable equilibrium and hence homeostasis. Cell lineage dynamics, however, are characterized by spatial structure, which can lead to different properties. Here, we investigate these dynamics using spatially explicit computational models, including cell division, differentiation, death, and migration / diffusion processes. According to these models, the negative feedback loop on stem cell self-renewal fails to maintain homeostasis, both under the assumption of strong spatial restrictions and fast migration / diffusion. Although homeostasis cannot be maintained, this feedback can regulate cell density and promote the formation of spatial structures in the model. Tissue homeostasis, however, can be achieved if spatially restricted negative feedback on self-renewal is combined with an experimentally documented spatial feedforward loop, in which stem cells regulate the fate of transit amplifying cells. This indicates that the dynamics of feedback regulation in tissue cell lineages are more complex than previously thought, and that combinations of spatially explicit control mechanisms are likely instrumental.


Subject(s)
Models, Biological , Stem Cells , Cell Differentiation/physiology , Cell Lineage , Feedback
7.
Dev Cell ; 57(10): 1226-1240.e8, 2022 05 23.
Article in English | MEDLINE | ID: mdl-35483358

ABSTRACT

Rhabdomyosarcoma (RMS) is a pediatric cancer with features of skeletal muscle; patients with unresectable or metastatic RMS fare poorly due to high rates of disease recurrence. Here, we use single-cell and single-nucleus RNA sequencing to show that RMS tumors recapitulate the spectrum of embryonal myogenesis. Using matched patient samples from a clinical trial and orthotopic patient-derived xenografts (O-PDXs), we show that chemotherapy eliminates the most proliferative component with features of myoblasts within embryonal RMS; after treatment, the immature population with features of paraxial mesoderm expands to reconstitute the developmental hierarchy of the original tumor. We discovered that this paraxial mesoderm population is dependent on EGFR signaling and is sensitive to EGFR inhibitors. Taken together, these data serve as a proof of concept that targeting each developmental state in embryonal RMS is an effective strategy for improving outcomes by preventing disease recurrence.


Subject(s)
Rhabdomyosarcoma, Embryonal , Rhabdomyosarcoma , Child , Drug Resistance , ErbB Receptors , Humans , Muscle Development/genetics , Neoplasm Recurrence, Local , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma, Embryonal/drug therapy , Rhabdomyosarcoma, Embryonal/genetics , Rhabdomyosarcoma, Embryonal/pathology
8.
Elife ; 112022 04 13.
Article in English | MEDLINE | ID: mdl-35416770

ABSTRACT

Aspirin intake has been shown to lead to significant protection against colorectal cancer, for example with an up to twofold reduction in colorectal adenoma incidence rates at higher doses. The mechanisms contributing to protection are not yet fully understood. While aspirin is an anti-inflammatory drug and can thus influence the tumor microenvironment, in vitro and in vivo experiments have recently shown that aspirin can also have a direct effect on cellular kinetics and fitness. It reduces the rate of tumor cell division and increases the rate of cell death. The question arises whether such changes in cellular fitness are sufficient to significantly contribute to the epidemiologically observed protection. To investigate this, we constructed a class of mathematical models of in vivo evolution of advanced adenomas, parameterized it with available estimates, and calculated population level incidence. Fitting the predictions to age incidence data revealed that only a model that included colonic crypt competition can account for the observed age-incidence curve. This model was then used to predict modified incidence patterns if cellular kinetics were altered as a result of aspirin treatment. We found that changes in cellular fitness that were within the experimentally observed ranges could reduce advanced adenoma incidence by a sufficient amount to account for age incidence data in aspirin-treated patient cohorts. While the mechanisms that contribute to the protective effect of aspirin are likely complex and multi-factorial, our study demonstrates that direct aspirin-induced changes of tumor cell fitness can significantly contribute to epidemiologically observed reduced incidence patterns.


Subject(s)
Adenoma , Colorectal Neoplasms , Adenoma/epidemiology , Adenoma/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aspirin/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/prevention & control , Humans , Incidence , Kinetics , Tumor Microenvironment
9.
Nat Commun ; 13(1): 121, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013215

ABSTRACT

HIV is difficult to eradicate due to the persistence of a long-lived reservoir of latently infected cells. Previous studies have shown that natural killer cells are important to inhibiting HIV infection, but it is unclear whether the administration of natural killer cells can reduce rebound viremia when anti-retroviral therapy is discontinued. Here we show the administration of allogeneic human peripheral blood natural killer cells delays viral rebound following interruption of anti-retroviral therapy in humanized mice infected with HIV-1. Utilizing genetically barcoded virus technology, we show these natural killer cells efficiently reduced viral clones rebounding from latency. Moreover, a kick and kill strategy comprised of the protein kinase C modulator and latency reversing agent SUW133 and allogeneic human peripheral blood natural killer cells during anti-retroviral therapy eliminated the viral reservoir in a subset of mice. Therefore, combinations utilizing latency reversal agents with targeted cellular killing agents may be an effective approach to eradicating the viral reservoir.


Subject(s)
Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/therapy , HIV-1/drug effects , Killer Cells, Natural/immunology , Protein Kinase Inhibitors/pharmacology , Viremia/therapy , Animals , Bone Marrow/drug effects , Bone Marrow/immunology , Bone Marrow/virology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Coculture Techniques , Female , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Killer Cells, Natural/transplantation , Male , Mice , Mice, Transgenic , Protein Kinase C/genetics , Protein Kinase C/immunology , Spleen/drug effects , Spleen/immunology , Spleen/virology , Viral Load/drug effects , Viremia/genetics , Viremia/immunology , Viremia/virology , Virus Latency/drug effects , Virus Replication/drug effects
10.
PLoS Comput Biol ; 17(12): e1009713, 2021 12.
Article in English | MEDLINE | ID: mdl-34936647

ABSTRACT

To study viral evolutionary processes within patients, mathematical models have been instrumental. Yet, the need for stochastic simulations of minority mutant dynamics can pose computational challenges, especially in heterogeneous systems where very large and very small sub-populations coexist. Here, we describe a hybrid stochastic-deterministic algorithm to simulate mutant evolution in large viral populations, such as acute HIV-1 infection, and further include the multiple infection of cells. We demonstrate that the hybrid method can approximate the fully stochastic dynamics with sufficient accuracy at a fraction of the computational time, and quantify evolutionary end points that cannot be expressed by deterministic models, such as the mutant distribution or the probability of mutant existence at a given infected cell population size. We apply this method to study the role of multiple infection and intracellular interactions among different virus strains (such as complementation and interference) for mutant evolution. Multiple infection is predicted to increase the number of mutants at a given infected cell population size, due to a larger number of infection events. We further find that viral complementation can significantly enhance the spread of disadvantageous mutants, but only in select circumstances: it requires the occurrence of direct cell-to-cell transmission through virological synapses, as well as a substantial fitness disadvantage of the mutant, most likely corresponding to defective virus particles. This, however, likely has strong biological consequences because defective viruses can carry genetic diversity that can be incorporated into functional virus genomes via recombination. Through this mechanism, synaptic transmission in HIV might promote virus evolvability.


Subject(s)
HIV Infections , HIV-1 , Host-Pathogen Interactions/genetics , Algorithms , Cells/virology , Computational Biology , Evolution, Molecular , HIV Infections/genetics , HIV Infections/transmission , HIV Infections/virology , HIV-1/genetics , HIV-1/pathogenicity , Humans , Mutation/genetics , Stochastic Processes , Virus Replication/genetics
11.
Virus Evol ; 7(1): veab026, 2021 Jan.
Article in English | MEDLINE | ID: mdl-34012557

ABSTRACT

Recombination has been shown to contribute to human immunodeficiency virus-1 (HIV-1) evolution in vivo, but the underlying dynamics are extremely complex, depending on the nature of the fitness landscapes and of epistatic interactions. A less well-studied determinant of recombinant evolution is the mode of virus transmission in the cell population. HIV-1 can spread by free virus transmission, resulting largely in singly infected cells, and also by direct cell-to-cell transmission, resulting in the simultaneous infection of cells with multiple viruses. We investigate the contribution of these two transmission pathways to recombinant evolution, by applying mathematical models to in vitro experimental data on the growth of fluorescent reporter viruses under static conditions (where both transmission pathways operate), and under gentle shaking conditions, where cell-to-cell transmission is largely inhibited. The parameterized mathematical models are then used to extrapolate the viral evolutionary dynamics beyond the experimental settings. Assuming a fixed basic reproductive ratio of the virus (independent of transmission pathway), we find that recombinant evolution is fastest if virus spread is driven only by cell-to-cell transmission and slows down if both transmission pathways operate. Recombinant evolution is slowest if all virus spread occurs through free virus transmission. This is due to cell-to-cell transmission 1, increasing infection multiplicity; 2, promoting the co-transmission of different virus strains from cell to cell; and 3, increasing the rate at which point mutations are generated as a result of more reverse transcription events. This study further resulted in the estimation of various parameters that characterize these evolutionary processes. For example, we estimate that during cell-to-cell transmission, an average of three viruses successfully integrated into the target cell, which can significantly raise the infection multiplicity compared to free virus transmission. In general, our study points towards the importance of infection multiplicity and cell-to-cell transmission for HIV evolution.

12.
Epidemics ; 35: 100463, 2021 06.
Article in English | MEDLINE | ID: mdl-34000693

ABSTRACT

Non-pharmaceutical intervention measures, such as social distancing, have so far been the only means to slow the spread of SARS-CoV-2. In the United States, strict social distancing during the first wave of virus spread has resulted in different types of infection dynamics. In some states, such as New York, extensive infection spread was followed by a pronounced decline of infection levels. In other states, such as California, less infection spread occurred before strict social distancing, and a different pattern was observed. Instead of a pronounced infection decline, a long-lasting plateau is evident, characterized by similar daily new infection levels. Here we show that network models, in which individuals and their social contacts are explicitly tracked, can reproduce the plateau if network connections are cut due to social distancing measures. The reason is that in networks characterized by a 2D spatial structure, infection tends to spread quadratically with time, but as edges are randomly removed, the infection spreads along nearly one-dimensional infection "corridors", resulting in plateau dynamics. Further, we show that plateau dynamics are observed only if interventions start sufficiently early; late intervention leads to a "peak and decay" pattern. Interestingly, the plateau dynamics are predicted to eventually transition into an infection decline phase without any further increase in social distancing measures. Additionally, the models suggest that a second wave becomes significantly less pronounced if social distancing is only relaxed once the dynamics have transitioned to the decline phase. The network models analyzed here allow us to interpret and reconcile different infection dynamics during social distancing observed in various US states.


Subject(s)
COVID-19/epidemiology , COVID-19/prevention & control , COVID-19/transmission , Contact Tracing , Humans , Models, Theoretical , Physical Distancing , SARS-CoV-2 , United States/epidemiology
13.
Cancer Res ; 81(4): 811-812, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33822746

ABSTRACT

The concept of adaptive cancer therapy proposes that the use of drugs at less than maximum tolerated dose can provide clinical benefits by allowing persisting drug-sensitive cells to competitively suppress drug-resistant cells; this can delay the outgrowth of these cell clones. The adaptive therapy concept has been developed with mathematical models and has subsequently been explored in clinical trials with promising results. In studies performed so far, a fitness cost of drug-resistant cells has been invoked for this treatment approach to be beneficial. In new work, it is shown that a clinical benefit can be achieved even in the absence of a fitness cost for resistant cells, which broadens the applicability of adaptive therapy.See related article by Strobl et al., p. 1135.


Subject(s)
Pharmaceutical Preparations , Maximum Tolerated Dose
14.
J R Soc Interface ; 18(176): 20200916, 2021 03.
Article in English | MEDLINE | ID: mdl-33784886

ABSTRACT

Epidemiological data about SARS-CoV-2 spread indicate that the virus is not transmitted uniformly in the population. The transmission tends to be more effective in select settings that involve exposure to relatively high viral dose, such as in crowded indoor settings, assisted living facilities, prisons or food processing plants. To explore the effect on infection dynamics, we describe a new mathematical model where transmission can occur (i) in the community at large, characterized by low-dose exposure and mostly mild disease, and (ii) in so-called transmission hot zones, characterized by high-dose exposure that can be associated with more severe disease. The model yields different types of epidemiological dynamics, depending on the relative importance of hot zone and community transmission. Interesting dynamics occur if the rate of virus release/deposition from severely infected people is larger than that of mildly infected individuals. Under this assumption, we find that successful infection spread can hinge upon high-dose hot zone transmission, yet the majority of infections are predicted to occur in the community at large with mild disease. In this regime, residual hot zone transmission can account for continued virus spread during community lockdowns, and the suppression of hot zones after community interventions are relaxed can cause a prolonged lack of infection resurgence following the reopening of society. This gives rise to the notion that targeted interventions specifically reducing virus transmission in the hot zones have the potential to suppress overall infection spread, including in the community at large. Epidemiological trends in the USA and Europe are interpreted in light of this model.


Subject(s)
COVID-19/epidemiology , COVID-19/transmission , Models, Biological , Pandemics , SARS-CoV-2 , Basic Reproduction Number/statistics & numerical data , COVID-19/virology , Computer Simulation , Humans , Mathematical Concepts , Pandemics/prevention & control , Pandemics/statistics & numerical data , Quarantine , Viral Load/statistics & numerical data
15.
J Theor Biol ; 509: 110499, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33130064

ABSTRACT

While resistance mutations are often implicated in the failure of cancer therapy, lack of response also occurs without such mutants. In bladder cancer mouse xenografts, repeated chemotherapy cycles have resulted in cancer stem cell (CSC) enrichment, and consequent loss of therapy response due to the reduced susceptibility of CSCs to drugs. A particular feedback loop present in the xenografts has been shown to promote CSC enrichment in this system. Yet, many other regulatory loops might also be operational and might promote CSC enrichment. Their identification is central to improving therapy response. Here, we perform a comprehensive mathematical analysis to define what types of regulatory feedback loops can and cannot contribute to CSC enrichment, providing guidance to the experimental identification of feedback molecules. We derive a formula that reveals whether or not the cell population experiences CSC enrichment over time, based on the properties of the feedback. We find that negative feedback on the CSC division rate or positive feedback on differentiated cell death rate can lead to CSC enrichment. Further, the feedback mediators that achieve CSC enrichment can be secreted by either CSCs or by more differentiated cells. The extent of enrichment is determined by the CSC death rate, the CSC self-renewal probability, and by feedback strength. Defining these general characteristics of feedback loops can guide the experimental screening for and identification of feedback mediators that can promote CSC enrichment in bladder cancer and potentially other tumors. This can help understand and overcome the phenomenon of CSC-based therapy resistance.


Subject(s)
Drug Resistance, Neoplasm , Neoplasms , Animals , Cell Differentiation , Cell Line, Tumor , Feedback , Mice , Neoplastic Stem Cells
16.
medRxiv ; 2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33052356

ABSTRACT

Epidemiological data on the spread of SARS-CoV-2 in the absence and presence of various non-pharmaceutical interventions indicate that the virus is not transmitted uniformly in the population. Transmission tends to be more effective in select settings that involve exposure to relatively high viral dose, such as in crowded indoor settings, assisted living facilities, prisons, or food processing plants. To explore the effect on infection dynamics, we describe a new mathematical model where transmission can occur (i) in the community at large, characterized by low dose exposure and mostly mild disease, and (ii) in so called transmission hot zones, characterized by high dose exposure that can be associated with more severe disease. Interestingly, we find that successful infection spread can hinge upon high-dose hot zone transmission, yet the majority of infections are predicted to occur in the community at large with mild disease. This gives rise to the prediction that targeted interventions that specifically reduce virus transmission in the hot zones (but not in the community at large) have the potential to suppress overall infection spread, including in the community at large. The model can further reconcile seemingly contradicting epidemiological observations. While in some locations like California, strict stay-home orders failed to significantly reduce infection prevalence, in other locations, such as New York and several European countries, stay-home orders lead to a pronounced fall in infection levels, which remained suppressed for some months after re-opening of society. Differences in hot zone transmission levels during and after social distancing interventions can account for these diverging infection patterns. These modeling results warrant further epidemiological investigations into the role of high dose hot zone transmission for the maintenance of SARS-CoV-2 spread.

17.
J R Soc Interface ; 17(170): 20200518, 2020 09.
Article in English | MEDLINE | ID: mdl-32993434

ABSTRACT

We have analysed the COVID-19 epidemic data of more than 174 countries (excluding China) in the period between 22 January and 28 March 2020. We found that some countries (such as the USA, the UK and Canada) follow an exponential epidemic growth, while others (like Italy and several other European countries) show a power law like growth. Regardless of the best fitting law, many countries can be shown to follow a common trajectory that is similar to Italy (the epicentre at the time of analysis), but with varying degrees of delay. We found that countries with 'younger' epidemics, i.e. countries where the epidemic started more recently, tend to exhibit more exponential like behaviour, while countries that were closer behind Italy tend to follow a power law growth. We hypothesize that there is a universal growth pattern of this infection that starts off as exponential and subsequently becomes more power law like. Although it cannot be excluded that this growth pattern is a consequence of social distancing measures, an alternative explanation is that it is an intrinsic epidemic growth law, dictated by a spatially distributed community structure, where the growth in individual highly mixed communities is exponential but the longer term, local geographical spread (in the absence of global mixing) results in a power law. This is supported by computer simulations of a metapopulation model that gives rise to predictions about the growth dynamics that are consistent with correlations found in the epidemiological data. Therefore, seeing a deviation from straight exponential growth may be a natural progression of the epidemic in each country. On the practical side, this indicates that (i) even in the absence of strict social distancing interventions, exponential growth is not an accurate predictor of longer term infection spread, and (ii) a deviation from exponential spread and a reduction of estimated doubling times do not necessarily indicate successful interventions, which are instead indicated by a transition to a reduced power or by a deviation from power law behaviour.


Subject(s)
Computer Simulation , Coronavirus Infections/epidemiology , Pneumonia, Viral/epidemiology , Public Health Informatics , Betacoronavirus , COVID-19 , Communicable Disease Control , Data Collection , Geography , Global Health , Humans , Kinetics , Pandemics , SARS-CoV-2
18.
Genetics ; 216(1): 191-203, 2020 09.
Article in English | MEDLINE | ID: mdl-32661138

ABSTRACT

Mutant evolution in spatially structured systems is important for a range of biological systems, but aspects of it still require further elucidation. Adding to previous work, we provide a simple derivation of growth laws that characterize the number of mutants of different relative fitness in expanding populations in spatial models of different dimensionalities. These laws are universal and independent of "microscopic" modeling details. We further study the accumulation of mutants and find that, with advantageous and neutral mutants, more of them are present in spatially structured, compared to well-mixed colonies of the same size. The behavior of disadvantageous mutants is subtle: if they are disadvantageous through a reduction in division rates, the result is the same, and it is the opposite if the disadvantage is due to a death rate increase. Finally, we show that in all cases, the same results are observed in fragmented, nonspatial patch models. This suggests that the patterns observed are the consequence of population fragmentation, and not spatial restrictions per se We provide an intuitive explanation for the complex dependence of disadvantageous mutant evolution on spatial restriction, which relies on desynchronized dynamics in different locations/patches, and plays out differently depending on whether the disadvantage is due to a lower division rate or a higher death rate. Implications for specific biological systems, such as the evolution of drug-resistant cell mutants in cancer or bacterial biofilms, are discussed.


Subject(s)
Evolution, Molecular , Models, Genetic , Bacteria/genetics , Genetic Fitness , Mutation
19.
J Theor Biol ; 503: 110384, 2020 10 21.
Article in English | MEDLINE | ID: mdl-32603669

ABSTRACT

The cancer stem cell hypothesis claims that tumor growth and progression are driven by a (typically) small niche of the total cancer cell population called cancer stem cells (CSCs). These CSCs can go through symmetric or asymmetric divisions to differentiate into specialised, progenitor cells or reproduce new CSCs. While it was once held that this differentiation pathway was unidirectional, recent research has demonstrated that differentiated cells are more plastic than initially considered. In particular, differentiated cells can de-differentiate and recover their stem-like capacity. Two recent papers have considered how this rate of plasticity affects the evolutionary dynamic of an invasive, malignant population of stem cells and differentiated cells into existing tissue (Mahdipour-Shirayeh et al., 2017; Wodarz, 2018). These papers arrive at seemingly opposing conclusions, one claiming that increased plasticity results in increased invasive potential, and the other that increased plasticity decreases invasive potential. Here, we show that what is most important, when determining the effect on invasive potential, is how one distributes this increased plasticity between the compartments of resident and mutant-type cells. We also demonstrate how these results vary, producing non-monotone fixation probability curves, as inter-compartmental plasticity changes when differentiated cell compartments are allowed to continue proliferating, highlighting a fundamental difference between the two models. We conclude by demonstrating the stability of these qualitative results over various parameter ranges. Keywords: cancer stem cells, plasticity, de-differentiation, fixation probability.


Subject(s)
Neoplasms , Neoplastic Stem Cells , Adaptation, Physiological , Cell Differentiation , Humans , Neoplasms/genetics , Probability
20.
Phys Rev E ; 101(3-1): 032404, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32289892

ABSTRACT

The process of range expansion (colonization) is one of the basic types of biological dynamics, whereby a species grows and spreads outwards, occupying new territories. Spatial modeling of this process is naturally implemented as a stochastic cellular automaton, with individuals occupying nodes on a rectangular grid, births and deaths occurring probabilistically, and individuals only reproducing onto unoccupied neighboring spots. In this paper we derive several approximations that allow prediction of the expected range expansion dynamics, based on the reproduction and death rates. We derive several approximations, where the cellular automaton is described by a system of ordinary differential equations that preserves correlations among neighboring spots (up to a distance). This methodology allows us to develop accurate approximations of the population size and the expected spatial shape, at a fraction of the computational time required to simulate the original stochastic system. In addition, we provide simple formulas for the steady-state population densities for von Neumann and Moore neighborhoods. Finally, we derive concise approximations for the speed of range expansion in terms of the reproduction and death rates, for both types of neighborhoods. The methodology is generalizable to more complex scenarios, such as different interaction ranges and multiple-species systems.


Subject(s)
Models, Statistical , Population Dynamics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...