Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
PLoS Comput Biol ; 20(8): e1012256, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39093897

RESUMO

Patient-derived tumor organoids (PDTOs) are novel cellular models that maintain the genetic, phenotypic and structural features of patient tumor tissue and are useful for studying tumorigenesis and drug response. When integrated with advanced 3D imaging and analysis techniques, PDTOs can be used to establish physiologically relevant high-throughput and high-content drug screening platforms that support the development of patient-specific treatment strategies. However, in order to effectively leverage high-throughput PDTO observations for clinical predictions, it is critical to establish a quantitative understanding of the basic properties and variability of organoid growth dynamics. In this work, we introduced an innovative workflow for analyzing and understanding PDTO growth dynamics, by integrating a high-throughput imaging deep learning platform with mathematical modeling, incorporating flexible growth laws and variable dormancy times. We applied the workflow to colon cancer organoids and demonstrated that organoid growth is well-described by the Gompertz model of growth. Our analysis showed significant intrapatient heterogeneity in PDTO growth dynamics, with the initial exponential growth rate of an organoid following a lognormal distribution within each dataset. The level of intrapatient heterogeneity varied between patients, as did organoid growth rates and dormancy times of single seeded cells. Our work contributes to an emerging understanding of the basic growth characteristics of PDTOs, and it highlights the heterogeneity in organoid growth both within and between patients. These results pave the way for further modeling efforts aimed at predicting treatment response dynamics and drug resistance timing.


Assuntos
Organoides , Humanos , Organoides/crescimento & desenvolvimento , Organoides/efeitos dos fármacos , Organoides/patologia , Modelos Biológicos , Neoplasias do Colo/diagnóstico por imagem , Neoplasias do Colo/patologia , Neoplasias do Colo/tratamento farmacológico , Biologia Computacional , Aprendizado Profundo , Modelos Teóricos , Imageamento Tridimensional/métodos
2.
PLoS Comput Biol ; 20(3): e1011888, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38446830

RESUMO

Tumor heterogeneity is a complex and widely recognized trait that poses significant challenges in developing effective cancer therapies. In particular, many tumors harbor a variety of subpopulations with distinct therapeutic response characteristics. Characterizing this heterogeneity by determining the subpopulation structure within a tumor enables more precise and successful treatment strategies. In our prior work, we developed PhenoPop, a computational framework for unravelling the drug-response subpopulation structure within a tumor from bulk high-throughput drug screening data. However, the deterministic nature of the underlying models driving PhenoPop restricts the model fit and the information it can extract from the data. As an advancement, we propose a stochastic model based on the linear birth-death process to address this limitation. Our model can formulate a dynamic variance along the horizon of the experiment so that the model uses more information from the data to provide a more robust estimation. In addition, the newly proposed model can be readily adapted to situations where the experimental data exhibits a positive time correlation. We test our model on simulated data (in silico) and experimental data (in vitro), which supports our argument about its advantages.


Assuntos
Fenômenos Genéticos , Neoplasias , Humanos , Avaliação Pré-Clínica de Medicamentos , Neoplasias/tratamento farmacológico , Neoplasias/patologia
3.
J Theor Biol ; 568: 111497, 2023 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-37087049

RESUMO

Recent evidence suggests that nongenetic (epigenetic) mechanisms play an important role at all stages of cancer evolution. In many cancers, these mechanisms have been observed to induce dynamic switching between two or more cell states, which commonly show differential responses to drug treatments. To understand how these cancers evolve over time, and how they respond to treatment, we need to understand the state-dependent rates of cell proliferation and phenotypic switching. In this work, we propose a rigorous statistical framework for estimating these parameters, using data from commonly performed cell line experiments, where phenotypes are sorted and expanded in culture. The framework explicitly models the stochastic dynamics of cell division, cell death and phenotypic switching, and it provides likelihood-based confidence intervals for the model parameters. The input data can be either the fraction of cells or the number of cells in each state at one or more time points. Through a combination of theoretical analysis and numerical simulations, we show that when cell fraction data is used, the rates of switching may be the only parameters that can be estimated accurately. On the other hand, using cell number data enables accurate estimation of the net division rate for each phenotype, and it can even enable estimation of the state-dependent rates of cell division and cell death. We conclude by applying our framework to a publicly available dataset.


Assuntos
Neoplasias , Humanos , Funções Verossimilhança , Divisão Celular , Fenótipo
4.
J Math Biol ; 87(4): 59, 2023 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-37707631

RESUMO

Tumor recurrence, driven by the evolution of drug resistance is a major barrier to therapeutic success in cancer. Tumor drug resistance is often caused by genetic alterations such as point mutation, which refers to the modification of a single genomic base pair, or gene amplification, which refers to the duplication of a region of DNA that contains a gene. These mechanisms typically confer varying degrees of resistance, and they tend to occur at vastly different frequencies. Here we investigate the dependence of tumor recurrence dynamics on these mechanisms of resistance, using stochastic multi-type branching process models. We derive tumor extinction probabilities and deterministic estimates for the tumor recurrence time, defined as the time when an initially drug sensitive tumor surpasses its original size after developing resistance. For models of amplification-driven and mutation-driven resistance, we prove law of large numbers results regarding the convergence of the stochastic recurrence times to their mean. Additionally, we prove sufficient and necessary conditions for a tumor to escape extinction under the gene amplification model, discuss behavior under biologically relevant parameters, and compare the recurrence time and tumor composition in the mutation and amplification models both analytically and using simulations. In comparing these mechanisms, we find that the ratio between recurrence times driven by amplification versus mutation depends linearly on the number of amplification events required to acquire the same degree of resistance as a mutation event, and we find that the relative frequency of amplification and mutation events plays a key role in determining the mechanism under which recurrence is more rapid for any specific system. In the amplification-driven resistance model, we also observe that increasing drug concentration leads to a stronger initial reduction in tumor burden, but that the eventual recurrent tumor population is less heterogeneous, more aggressive and harbors higher levels of drug-resistance.


Assuntos
Agressão , Recidiva Local de Neoplasia , Humanos , Recidiva Local de Neoplasia/genética , Mutação , Genômica , Probabilidade
5.
Phys Biol ; 19(3)2022 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-35078159

RESUMO

The role of plasticity and epigenetics in shaping cancer evolution and response to therapy has taken center stage with recent technological advances including single cell sequencing. This roadmap article is focused on state-of-the-art mathematical and experimental approaches to interrogate plasticity in cancer, and addresses the following themes and questions: is there a formal overarching framework that encompasses both non-genetic plasticity and mutation-driven somatic evolution? How do we measure and model the role of the microenvironment in influencing/controlling non-genetic plasticity? How can we experimentally study non-genetic plasticity? Which mathematical techniques are required or best suited? What are the clinical and practical applications and implications of these concepts?


Assuntos
Epigênese Genética , Neoplasias , Epigenômica , Humanos , Mutação , Neoplasias/tratamento farmacológico , Neoplasias/genética , Microambiente Tumoral
6.
Theor Popul Biol ; 142: 67-90, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34560155

RESUMO

The site frequency spectrum (SFS) is a popular summary statistic of genomic data. While the SFS of a constant-sized population undergoing neutral mutations has been extensively studied in population genetics, the rapidly growing amount of cancer genomic data has attracted interest in the spectrum of an exponentially growing population. Recent theoretical results have generally dealt with special or limiting cases, such as considering only cells with an infinite line of descent, assuming deterministic tumor growth, or taking large-time or large-population limits. In this work, we derive exact expressions for the expected SFS of a cell population that evolves according to a stochastic branching process, first for cells with an infinite line of descent and then for the total population, evaluated either at a fixed time (fixed-time spectrum) or at the stochastic time at which the population reaches a certain size (fixed-size spectrum). We find that while the rate of mutation scales the SFS of the total population linearly, the rates of cell birth and cell death change the shape of the spectrum at the small-frequency end, inducing a transition between a 1/j2 power-law spectrum and a 1/j spectrum as cell viability decreases. We show that this insight can in principle be used to estimate the ratio between the rate of cell death and cell birth, as well as the mutation rate, using the site frequency spectrum alone. Although the discussion is framed in terms of tumor dynamics, our results apply to any exponentially growing population of individuals undergoing neutral mutations.


Assuntos
Modelos Genéticos , Neoplasias , Sobrevivência Celular/genética , Genética Populacional , Humanos , Mutação , Neoplasias/genética , Processos Estocásticos
7.
Am Nat ; 195(2): 300-314, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32017618

RESUMO

The history of a trait within a lineage may influence its future evolutionary trajectory, but macroevolutionary theory of this process is not well developed. For example, consider the simplified binary trait of living in cave versus surface habitat. The longer a species has been cave dwelling, the more accumulated loss of vision, pigmentation, and defense may restrict future adaptation if the species encounters the surface environment. However, the Markov model of discrete trait evolution that is widely adopted in phylogenetics does not allow the rate of cave-to-surface transition to decrease with longer duration as a cave dweller. Here we describe three models of evolution that remove this memoryless constraint, using a renewal process to generalize beyond the typical Poisson process of discrete trait macroevolution. We then show how the two-state renewal process can be used for inference, and we investigate the potential of phylogenetic comparative data to reveal different influences of trait duration, or memory in trait evolution. We hope that such approaches may open new avenues for modeling trait evolution and for broad comparative tests of hypotheses that some traits become entrenched.


Assuntos
Evolução Biológica , Modelos Teóricos , Filogenia , Cadeias de Markov , Fenótipo , Fatores de Tempo
8.
J Theor Biol ; 490: 110162, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-31953135

RESUMO

The emergence of acquired drug resistance in cancer represents a major barrier to treatment success. While research has traditionally focused on genetic sources of resistance, recent findings suggest that cancer cells can acquire transient resistant phenotypes via epigenetic modifications and other non-genetic mechanisms. Although these resistant phenotypes are eventually relinquished by individual cells, they can temporarily 'save' the tumor from extinction and enable the emergence of more permanent resistance mechanisms. These observations have generated interest in the potential of epigenetic therapies for long-term tumor control or eradication. In this work, we develop a mathematical model to study how phenotypic switching at the single-cell level affects resistance evolution in cancer. We highlight unique features of non-genetic resistance, probe the evolutionary consequences of epigenetic drugs and explore potential therapeutic strategies. We find that even short-term epigenetic modifications and stochastic fluctuations in gene expression can drive long-term drug resistance in the absence of any bona fide resistance mechanisms. We also find that an epigenetic drug that slightly perturbs the average retention of the resistant phenotype can turn guaranteed treatment failure into guaranteed success. Lastly, we find that combining an epigenetic drug with an anti-cancer agent can significantly outperform monotherapy, and that treatment outcome is heavily affected by drug sequencing.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Neoplasias , Resistencia a Medicamentos Antineoplásicos/genética , Epigênese Genética , Humanos , Modelos Teóricos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Fenótipo
9.
PLoS Comput Biol ; 13(7): e1005482, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28683103

RESUMO

Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) is characterized by a very poor prognosis and a high likelihood of acquired chemo-resistance. Although tyrosine kinase inhibitor (TKI) therapy has improved clinical outcome, most ALL patients relapse following treatment with TKI due to the development of resistance. We developed an in vitro model of Nilotinib-resistant Ph+ leukemia cells to investigate whether low dose radiation (LDR) in combination with TKI therapy overcome chemo-resistance. Additionally, we developed a mathematical model, parameterized by cell viability experiments under Nilotinib treatment and LDR, to explain the cellular response to combination therapy. The addition of LDR significantly reduced drug resistance both in vitro and in computational model. Decreased expression level of phosphorylated AKT suggests that the combination treatment plays an important role in overcoming resistance through the AKT pathway. Model-predicted cellular responses to the combined therapy provide good agreement with experimental results. Augmentation of LDR and Nilotinib therapy seems to be beneficial to control Ph+ leukemia resistance and the quantitative model can determine optimal dosing schedule to enhance the effectiveness of the combination therapy.


Assuntos
Quimiorradioterapia/métodos , Modelos Biológicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/fisiopatologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirimidinas/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Simulação por Computador , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Camundongos , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Resultado do Tratamento
10.
PLoS Comput Biol ; 12(10): e1005129, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27764087

RESUMO

Over the past decade, several targeted therapies (e.g. imatinib, dasatinib, nilotinib) have been developed to treat Chronic Myeloid Leukemia (CML). Despite an initial response to therapy, drug resistance remains a problem for some CML patients. Recent studies have shown that resistance mutations that preexist treatment can be detected in a substantial number of patients, and that this may be associated with eventual treatment failure. One proposed method to extend treatment efficacy is to use a combination of multiple targeted therapies. However, the design of such combination therapies (timing, sequence, etc.) remains an open challenge. In this work we mathematically model the dynamics of CML response to combination therapy and analyze the impact of combination treatment schedules on treatment efficacy in patients with preexisting resistance. We then propose an optimization problem to find the best schedule of multiple therapies based on the evolution of CML according to our ordinary differential equation model. This resulting optimization problem is nontrivial due to the presence of ordinary different equation constraints and integer variables. Our model also incorporates drug toxicity constraints by tracking the dynamics of patient neutrophil counts in response to therapy. We determine optimal combination strategies that maximize time until treatment failure on hypothetical patients, using parameters estimated from clinical data in the literature.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Sistemas de Apoio a Decisões Clínicas , Monitoramento de Medicamentos/métodos , Quimioterapia Assistida por Computador/métodos , Leucemia Mielogênica Crônica BCR-ABL Positiva/diagnóstico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Antineoplásicos/administração & dosagem , Esquema de Medicação , Humanos , Resultado do Tratamento
11.
PLoS Comput Biol ; 12(8): e1005077, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27560187

RESUMO

Experimental studies have shown that one key factor in driving the emergence of drug resistance in solid tumors is tumor hypoxia, which leads to the formation of localized environmental niches where drug-resistant cell populations can evolve and survive. Hypoxia-activated prodrugs (HAPs) are compounds designed to penetrate to hypoxic regions of a tumor and release cytotoxic or cytostatic agents; several of these HAPs are currently in clinical trial. However, preliminary results have not shown a survival benefit in several of these trials. We hypothesize that the efficacy of treatments involving these prodrugs depends heavily on identifying the correct treatment schedule, and that mathematical modeling can be used to help design potential therapeutic strategies combining HAPs with standard therapies to achieve long-term tumor control or eradication. We develop this framework in the specific context of EGFR-driven non-small cell lung cancer, which is commonly treated with the tyrosine kinase inhibitor erlotinib. We develop a stochastic mathematical model, parametrized using clinical and experimental data, to explore a spectrum of treatment regimens combining a HAP, evofosfamide, with erlotinib. We design combination toxicity constraint models and optimize treatment strategies over the space of tolerated schedules to identify specific combination schedules that lead to optimal tumor control. We find that (i) combining these therapies delays resistance longer than any monotherapy schedule with either evofosfamide or erlotinib alone, (ii) sequentially alternating single doses of each drug leads to minimal tumor burden and maximal reduction in probability of developing resistance, and (iii) strategies minimizing the length of time after an evofosfamide dose and before erlotinib confer further benefits in reduction of tumor burden. These results provide insights into how hypoxia-activated prodrugs may be used to enhance therapeutic effectiveness in the clinic.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Hipóxia/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Pró-Fármacos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Biologia Computacional , Cloridrato de Erlotinib/farmacologia , Cloridrato de Erlotinib/uso terapêutico , Humanos , Pró-Fármacos/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos
12.
PLoS Comput Biol ; 11(9): e1004350, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26379039

RESUMO

The traditional view of cancer as a genetic disease that can successfully be treated with drugs targeting mutant onco-proteins has motivated whole-genome sequencing efforts in many human cancer types. However, only a subset of mutations found within the genomic landscape of cancer is likely to provide a fitness advantage to the cell. Distinguishing such "driver" mutations from innocuous "passenger" events is critical for prioritizing the validation of candidate mutations in disease-relevant models. We design a novel statistical index, called the Hitchhiking Index, which reflects the probability that any observed candidate gene is a passenger alteration, given the frequency of alterations in a cross-sectional cancer sample set, and apply it to a mutational data set in colorectal cancer. Our methodology is based upon a population dynamics model of mutation accumulation and selection in colorectal tissue prior to cancer initiation as well as during tumorigenesis. This methodology can be used to aid in the prioritization of candidate mutations for functional validation and contributes to the process of drug discovery.


Assuntos
Neoplasias Colorretais/genética , Biologia Computacional/métodos , Modelos Genéticos , Mutação/genética , Estudos Transversais , Evolução Molecular , Humanos , Modelos Estatísticos , Dinâmica Populacional
13.
J Theor Biol ; 355: 10-20, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-24681298

RESUMO

Acquired drug resistance is a major limitation for the successful treatment of cancer. Resistance can emerge due to a variety of reasons including host environmental factors as well as genetic or epigenetic alterations in the cancer cells. Evolutionary theory has contributed to the understanding of the dynamics of resistance mutations in a cancer cell population, the risk of resistance pre-existing before the initiation of therapy, the composition of drug cocktails necessary to prevent the emergence of resistance, and optimum drug administration schedules for patient populations at risk of evolving acquired resistance. Here we review recent advances towards elucidating the evolutionary dynamics of acquired drug resistance and outline how evolutionary thinking can contribute to outstanding questions in the field.


Assuntos
Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Evolução Molecular , Modelos Biológicos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Humanos , Neoplasias/patologia
14.
J Theor Biol ; 355: 170-84, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-24735903

RESUMO

Primary tumors often emerge within genetically altered fields of premalignant cells that appear histologically normal but have a high chance of progression to malignancy. Clinical observations have suggested that these premalignant fields pose high risks for emergence of recurrent tumors if left behind after surgical removal of the primary tumor. In this work, we develop a spatio-temporal stochastic model of epithelial carcinogenesis, combining cellular dynamics with a general framework for multi-stage genetic progression to cancer. Using the model, we investigate how various properties of the premalignant fields depend on microscopic cellular properties of the tissue. In particular, we provide analytic results for the size-distribution of the histologically undetectable premalignant fields at the time of diagnosis, and investigate how the extent and the geometry of these fields depend upon key groups of parameters associated with the tissue and genetic pathways. We also derive analytical results for the relative risks of local vs. distant secondary tumors for different parameter regimes, a critical aspect for the optimal choice of post-operative therapy in carcinoma patients. This study contributes to a growing literature seeking to obtain a quantitative understanding of the spatial dynamics in cancer initiation.


Assuntos
Transformação Celular Neoplásica/metabolismo , Modelos Biológicos , Neoplasias/metabolismo , Transformação Celular Neoplásica/patologia , Humanos , Neoplasias/patologia
15.
bioRxiv ; 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-39005329

RESUMO

Circulating tumor DNA assays are promising tools for the prediction of cancer treatment response. Here, we build a framework for the design of ctDNA biomarkers of therapy response that incorporate variations in ctDNA dynamics driven by specific treatment mechanisms. We develop mathematical models of ctDNA kinetics driven by tumor response to several therapy classes, and utilize them to simulate randomized virtual patient cohorts to test candidate biomarkers. Using this approach, we propose specific biomarkers, based on ctDNA longitudinal features, for targeted therapy, chemotherapy and radiation therapy. We evaluate and demonstrate the efficacy of these biomarkers in predicting treatment response within a randomized virtual patient cohort dataset. These biomarkers are based on novel proposals for ctDNA sampling protocols, consisting of frequent sampling within a compact time window surrounding therapy initiation - which we hypothesize to hold valuable prognostic information on longer-term treatment response. This study highlights a need for tailoring ctDNA sampling protocols and interpretation methodology to specific biological mechanisms of therapy response, and it provides a novel modeling and simulation framework for doing so. In addition, it highlights the potential of ctDNA assays for making early, rapid predictions of treatment response within the first days or weeks of treatment, and generates hypotheses for further clinical testing.

16.
ArXiv ; 2023 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-37396604

RESUMO

Tumor recurrence, driven by the evolution of drug resistance is a major barrier to therapeutic success in cancer. Resistance is often caused by genetic alterations such as point mutation, which refers to the modification of a single genomic base pair, or gene amplification, which refers to the duplication of a region of DNA that contains a gene. Here we investigate the dependence of tumor recurrence dynamics on these mechanisms of resistance, using stochastic multi-type branching process models. We derive tumor extinction probabilities and deterministic estimates for the tumor recurrence time, defined as the time when an initially drug sensitive tumor surpasses its original size after developing resistance. For models of amplification-driven and mutation-driven resistance, we prove law of large numbers results regarding the convergence of the stochastic recurrence times to their mean. Additionally, we prove sufficient and necessary conditions for a tumor to escape extinction under the gene amplification model, discuss behavior under biologically relevant parameters, and compare the recurrence time and tumor composition in the mutation and amplification models both analytically and using simulations. In comparing these mechanisms, we find that the ratio between recurrence times driven by amplification vs. mutation depends linearly on the number of amplification events required to acquire the same degree of resistance as a mutation event, and we find that the relative frequency of amplification and mutation events plays a key role in determining the mechanism under which recurrence is more rapid. In the amplification-driven resistance model, we also observe that increasing drug concentration leads to a stronger initial reduction in tumor burden, but that the eventual recurrent tumor population is less heterogeneous, more aggressive, and harbors higher levels of drug-resistance.

17.
ArXiv ; 2023 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-37396613

RESUMO

Recent evidence suggests that nongenetic (epigenetic) mechanisms play an important role at all stages of cancer evolution. In many cancers, these mechanisms have been observed to induce dynamic switching between two or more cell states, which commonly show differential responses to drug treatments. To understand how these cancers evolve over time, and how they respond to treatment, we need to understand the state-dependent rates of cell proliferation and phenotypic switching. In this work, we propose a rigorous statistical framework for estimating these parameters, using data from commonly performed cell line experiments, where phenotypes are sorted and expanded in culture. The framework explicitly models the stochastic dynamics of cell division, cell death and phenotypic switching, and it provides likelihood-based confidence intervals for the model parameters. The input data can be either the fraction of cells or the number of cells in each state at one or more time points. Through a combination of theoretical analysis and numerical simulations, we show that when cell fraction data is used, the rates of switching may be the only parameters that can be estimated accurately. On the other hand, using cell number data enables accurate estimation of the net division rate for each phenotype, and it can even enable estimation of the state-dependent rates of cell division and cell death. We conclude by applying our framework to a publicly available dataset.

18.
Cell Rep Methods ; 3(3): 100417, 2023 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-37056380

RESUMO

Tumor heterogeneity is an important driver of treatment failure in cancer since therapies often select for drug-tolerant or drug-resistant cellular subpopulations that drive tumor growth and recurrence. Profiling the drug-response heterogeneity of tumor samples using traditional genomic deconvolution methods has yielded limited results, due in part to the imperfect mapping between genomic variation and functional characteristics. Here, we leverage mechanistic population modeling to develop a statistical framework for profiling phenotypic heterogeneity from standard drug-screen data on bulk tumor samples. This method, called PhenoPop, reliably identifies tumor subpopulations exhibiting differential drug responses and estimates their drug sensitivities and frequencies within the bulk population. We apply PhenoPop to synthetically generated cell populations, mixed cell-line experiments, and multiple myeloma patient samples and demonstrate how it can provide individualized predictions of tumor growth under candidate therapies. This methodology can also be applied to deconvolution problems in a variety of biological settings beyond cancer drug response.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Detecção Precoce de Câncer , Neoplasias/tratamento farmacológico , Antineoplásicos/farmacologia , Linhagem Celular , Genômica
19.
Haematologica ; 97(10): 1553-61, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22419579

RESUMO

BACKGROUND: Chronic myeloid leukemia is successfully managed by imatinib therapy, but the question remains whether treatment must be administered indefinitely. Imatinib discontinuation trials have led to two distinct outcomes: about 60% of patients experienced disease relapse within 6 months of treatment cessation, while the remaining 40% remained disease-free throughout the duration of follow-up. We aimed to investigate the mechanisms underlying these disparate clinical outcomes. DESIGN AND METHODS: We utilized molecular data from the "Stop Imatinib" trial together with a mathematical framework of chronic myeloid leukemia, based on a four-compartment model that can explain the kinetics of the molecular response to imatinib. This approach was complemented by statistical analyses to estimate system parameters and investigate whether chronic myeloid leukemia can be cured by imatinib therapy alone. RESULTS: We found that there are insufficient follow-up data from the "Stop Imatinib" trial in order to conclude whether the absence of a relapse signifies cure of the disease. We determined that selection of less aggressive leukemic phenotypes by imatinib therapy recapitulates the trial outcomes. This postulated mechanism agrees with the observation that most patients who have a complete molecular response after discontinuation of imatinib continue to harbor minimal residual disease, and might work in concert with other factors suppressing leukemic cell expansion when the tumor burden remains low. CONCLUSIONS: Our analysis provides evidence for a mechanistic model of chronic myeloid leukemia selection by imatinib treatment and suggests that it may not be safe to discontinue therapy outside a clinical trial.


Assuntos
Antineoplásicos/uso terapêutico , Benzamidas/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Humanos , Mesilato de Imatinib , Leucemia Mielogênica Crônica BCR-ABL Positiva/mortalidade , Modelos Teóricos , Recidiva , Resultado do Tratamento
20.
RNA Biol ; 9(10): 1275-87, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22995834

RESUMO

MicroRNAs (miRNAs) are important players of post-transcriptional gene regulation. Individual miRNAs can target multiple mRNAs and a single mRNA can be targeted by many miRNAs. We hypothesized that miRNAs select and regulate their targets based on their own expression levels, those of their target mRNAs and triggered feedback loops. We studied the effects of varying concentrations of let-7a-7f and the miR-17-92 cluster plasmids on the reporter genes carrying either DICER- or cMYC -3'UTR in Huh-7 cells. We showed that let-7 significantly downregulated expression of DICER 3'UTR reporter at lower concentrations, but selectively downregulated expression of a cMYC 3'UTR reporter at higher dose. This miRNA dose-dependent target selection was also confirmed in other target genes, including CCND1, CDKN1 and E2F1. After overexpressing let-7a-7f or the miR-17-92 clusters at wide-ranging doses, the target genes displayed a nonlinear correlation to the transfected miRNA. Further, by comparing the expression levels of let-7a and miR-17-5p, along with their selected target genes in 3 different cell lines, we found that the knockdown dose of each miRNA was directly related to their baseline expression level, that of the target gene and feedback loops. These findings were supported by gene modulation studies using endogenous levels of miR-29, -1 and -206 and a luciferase reporter system in multiple cell lines. Finally, we determined that the miR-17-92 cluster affected cell viability in a dose-dependent manner. In conclusion, we have shown that miRNAs potentially select their targets in a dose-dependent and nonlinear fashion that affects biological function; and this represents a novel mechanism by which miRNAs orchestrate the finely tuned balance of cell function.


Assuntos
Regiões 3' não Traduzidas , Retroalimentação Fisiológica , Regulação da Expressão Gênica , MicroRNAs/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Ciclina D1/genética , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Genes Reporter , Humanos , Luciferases , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Longo não Codificante , Ribonuclease III/genética , Ribonuclease III/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA