Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Bull Math Biol ; 86(8): 89, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38884815

ABSTRACT

Cancer metastasis accounts for a majority of cancer-related deaths worldwide. Metastasis occurs when the primary tumor sheds cells into the blood and lymphatic circulation, thereby becoming circulating tumor cells (CTCs) that transverse through the circulatory system, extravasate the circulation and establish a secondary distant tumor. Accumulating evidence suggests that circulating effector CD 8 + T cells are able to recognize and attack arrested or extravasating CTCs, but this important antitumoral effect remains largely undefined. Recent studies highlighted the supporting role of activated platelets in CTCs's extravasation from the bloodstream, contributing to metastatic progression. In this work, a simple mathematical model describes how the primary tumor, CTCs, activated platelets and effector CD 8 + T cells participate in metastasis. The stability analysis reveals that for early dissemination of CTCs, effector CD 8 + T cells can present or keep secondary metastatic tumor burden at low equilibrium state. In contrast, for late dissemination of CTCs, effector CD 8 + T cells are unlikely to inhibit secondary tumor growth. Moreover, global sensitivity analysis demonstrates that the rate of the primary tumor growth, intravascular CTC proliferation, as well as the CD 8 + T cell proliferation, strongly affects the number of the secondary tumor cells. Additionally, model simulations indicate that an increase in CTC proliferation greatly contributes to tumor metastasis. Our simulations further illustrate that the higher the number of activated platelets on CTCs, the higher the probability of secondary tumor establishment. Intriguingly, from a mathematical immunology perspective, our simulations indicate that if the rate of effector CD 8 + T cell proliferation is high, then the secondary tumor formation can be considerably delayed, providing a window for adjuvant tumor control strategies. Collectively, our results suggest that the earlier the effector CD 8 + T cell response is enhanced the higher is the probability of preventing or delaying secondary tumor metastases.


Subject(s)
Blood Platelets , CD8-Positive T-Lymphocytes , Models, Immunological , Neoplasm Metastasis , Neoplastic Cells, Circulating , Humans , Blood Platelets/immunology , Blood Platelets/pathology , CD8-Positive T-Lymphocytes/immunology , Computer Simulation , Mathematical Concepts , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/immunology , Platelet Activation/immunology
2.
Math Biosci Eng ; 19(5): 4429-4457, 2022 03 02.
Article in English | MEDLINE | ID: mdl-35430822

ABSTRACT

Combining chimeric antigen receptor T (CAR-T) cells with oncolytic viruses (OVs) has recently emerged as a promising treatment approach in preclinical studies that aim to alleviate some of the barriers faced by CAR-T cell therapy. In this study, we address by means of mathematical modeling the main question of whether a single dose or multiple sequential doses of CAR-T cells during the OVs therapy can have a synergetic effect on tumor reduction. To that end, we propose an ordinary differential equations-based model with virus-induced synergism to investigate potential effects of different regimes that could result in efficacious combination therapy against tumor cell populations. Model simulations show that, while the treatment with a single dose of CAR-T cells is inadequate to eliminate all tumor cells, combining the same dose with a single dose of OVs can successfully eliminate the tumor in the absence of virus-induced synergism. However, in the presence of virus-induced synergism, the same combination therapy fails to eliminate the tumor. Furthermore, it is shown that if the intensity of virus-induced synergy and/or virus oncolytic potency is high, then the induced CAR-T cell response can inhibit virus oncolysis. Additionally, the simulations show a more robust synergistic effect on tumor cell reduction when OVs and CAR-T cells are administered simultaneously compared to the combination treatment where CAR-T cells are administered first or after OV injection. Our findings suggest that the combination therapy of CAR-T cells and OVs seems unlikely to be effective if the virus-induced synergistic effects are included when genetically engineering oncolytic viral vectors.


Subject(s)
Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Receptors, Chimeric Antigen , Humans , Models, Theoretical , Neoplasms/therapy , Oncolytic Viruses/physiology , T-Lymphocytes
3.
J Math Biol ; 84(4): 28, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35239041

ABSTRACT

Estrogen is known to stimulate the growth of breast cancer, but is also effective in treating the disease. This is referred to as the"estrogen paradox". Furthermore, short-term treatment with estrogen can successfully eliminate breast cancer, whereas long-term treatment can cause cancer recurrence. Studies highlighted clinical correlations between estrogen and the protein p53 which plays a pivotal role in breast cancer suppression. We sought to investigate how the interplay between estrogen and p53 impacts the dynamics of breast cancer, and further explore if this could be a plausible explanation for the estrogen paradox and the paradoxical tumor recurrence that results from prolonged treatment with estrogen. For this, we propose a novel ODE based mathematical model that accounts for dormant and active cancer cells, along with the estrogen hormone and the p53 protein. We analyze the model's global stability behavior using the Poincaré-Bendixson theorem and results from differential inequalities. We also perform a bifurcation analysis and carry out numerical simulations that elucidate the roles of estrogen and p53 in the estrogen paradox and its long term estrogen paradoxical effect. The mathematical and numerical analyses suggest that the apparent paradoxical role of estrogen could be the result of an interplay between estrogen and p53, and provide explicit conditions under which the paradoxical effect of long-term treatment may be prevented.


Subject(s)
Breast Neoplasms , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Estrogens/metabolism , Estrogens/therapeutic use , Female , Humans , Models, Theoretical , Neoplasm Recurrence, Local
4.
Bull Math Biol ; 83(7): 75, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34008149

ABSTRACT

In this paper, we investigate how natural killer (NK) cell recruitment to the tumor microenvironment (TME) affects oncolytic virotherapy. NK cells play a major role against viral infections. They are, however, known to induce early viral clearance of oncolytic viruses, which hinders the overall efficacy of oncolytic virotherapy. Here, we formulate and analyze a simple mathematical model of the dynamics of the tumor, OV and NK cells using currently available preclinical information. The aim of this study is to characterize conditions under which the synergistic balance between OV-induced NK responses and required viral cytopathicity may or may not result in a successful treatment. In this study, we found that NK cell recruitment to the TME must take place neither too early nor too late in the course of OV infection so that treatment will be successful. NK cell responses are most influential at either early (partly because of rapid response of NK cells to viral infections or antigens) or later (partly because of antitumoral ability of NK cells) stages of oncolytic virotherapy. The model also predicts that: (a) an NK cell response augments oncolytic virotherapy only if viral cytopathicity is weak; (b) the recruitment of NK cells modulates tumor growth; and (c) the depletion of activated NK cells within the TME enhances the probability of tumor escape in oncolytic virotherapy. Taken together, our model results demonstrate that OV infection is crucial, not just to cytoreduce tumor burden, but also to induce the stronger NK cell response necessary to achieve complete or at least partial tumor remission. Furthermore, our modeling framework supports combination therapies involving NK cells and OV which are currently used in oncolytic immunovirotherapy to treat several cancer types.


Subject(s)
Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Humans , Killer Cells, Natural , Mathematical Concepts , Models, Theoretical , Neoplasms/therapy , Tumor Microenvironment
5.
Sci Rep ; 10(1): 425, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949228

ABSTRACT

Mesenchymal stem cells (MSCs) loaded with oncolytic viruses are presently being investigated as a new modality of advanced/metastatic tumors treatment and enhancement of virotherapy. MSCs can, however, either promote or suppress tumor growth. To address the critical question of how MSCs loaded with oncolytic viruses affect virotherapy outcomes and tumor growth patterns in a tumor microenvironment, we developed and analyzed an integrated mathematical-experimental model. We used the model to describe both the growth dynamics in our experiments of firefly luciferase-expressing Hep3B tumor xenografts and the effects of the immune response during the MSCs-based virotherapy. We further employed it to explore the conceptual clinical feasibility, particularly, in evaluating the relative significance of potential immune promotive/suppressive mechanisms induced by MSCs loaded with oncolytic viruses. We were able to delineate conditions which may significantly contribute to the success or failure of MSC-based virotherapy as well as generate new hypotheses. In fact, one of the most impactful outcomes shown by this investigation, not inferred from the experiments alone, was the initially counter-intuitive fact that using tumor-promoting MSCs as carriers is not only helpful but necessary in achieving tumor control. Considering the fact that it is still currently a controversial debate whether MSCs exert a pro- or anti-tumor action, mathematical models such as this one help to quantitatively predict the consequences of using MSCs for delivering virotherapeutic agents in vivo. Taken together, our results show that MSC-mediated systemic delivery of oncolytic viruses is a promising strategy for achieving synergistic anti-tumor efficacy with improved safety profiles.


Subject(s)
Adenoviridae/physiology , Mesenchymal Stem Cells/metabolism , Models, Biological , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Adenoviridae/metabolism , Cell Proliferation , Oncolytic Viruses/metabolism
6.
Math Biosci ; 320: 108304, 2020 02.
Article in English | MEDLINE | ID: mdl-31883985

ABSTRACT

South Sudan accounts for a large proportion of all annual malaria cases in Africa. In recent years, the country has witnessed an unprecedented number of people on the move, refugees, internally displaced people, people who have returned to their counties or areas of origin, stateless people and other populations of concern, posing challenges to malaria control. Thus, one can claim that human mobility is one of the contributing factors to the resurgence of malaria. The aim of this paper is to assess the impact of human mobility on the burden of malaria disease in South Sudan. For this, we formulate an SIR-type model that describes the transmission dynamics of malaria disease between multiple patches. The proposed model is a system of stochastic differential equations consisting of ordinary differential equations perturbed by a stochastic Wiener process. For the deterministic part of the model, we calculate the basic reproduction number. Concerning the whole stochastic model, we use the maximum likelihood approach to fit the model to weekly malaria data of 2011 from Central Equatoria State, Western Bahr El Ghazal State and Warrap State. Using the parameters estimated on the fitted model, we simulate the future observation of the disease pattern. The disease was found to persist in the low transmission patches when there is human inflow in these patches and although the intervention coverage reaches 75%.


Subject(s)
Malaria/transmission , Models, Theoretical , Population Dynamics/statistics & numerical data , Humans , Malaria/prevention & control , South Sudan/epidemiology
7.
Math Biosci ; 310: 13-23, 2019 04.
Article in English | MEDLINE | ID: mdl-30711479

ABSTRACT

Malaria is endemic in South Sudan and it is one of the most severe diseases in the war-torn nation. There has been much concern about whether the severity of its transmission might depend upon climatic conditions that are related to the reproduction of the single-cell parasite attaching to female mosquitoes, especially in high altitude areas. The country experiences two different climatic conditions; namely one tropical and the other hot and semi-arid. In this study, we aim to assess the potential impact of climatic conditions on malaria prevalence in these two climatically distinct regions of South Sudan. We develop and analyze a host-mosquito disease-based model that includes temperature and rainfall. The model has also been parameterized in a Bayesian framework using Bayesian Markov Chain Monte Carlo (MCMC). The mathematical analysis for this study has included equilibria, stability and a sensitivity index on the basic reproduction number R0. The threshold R0 is also used to provide a numerical basis for further refinement and prediction of the impact of climate variability on malaria transmission intensity over the study region. The study highlights the impact of various temperature values on the population dynamics of the mosquito.


Subject(s)
Basic Reproduction Number , Malaria/transmission , Models, Biological , Models, Statistical , Seasons , Bayes Theorem , Humans , Markov Chains , Monte Carlo Method , South Sudan/epidemiology
8.
J Theor Biol ; 481: 202-222, 2019 11 21.
Article in English | MEDLINE | ID: mdl-30610868

ABSTRACT

A modelling framework that describes the dynamics of populations of the female Anopheles sp mosquitoes is used to develop and analyse a deterministic ordinary differential equation model for dynamics and transmission of malaria amongst humans and varying mosquito populations. The framework includes a characterization of the gonotrophic cycle of the female mosquito. The epidemiological model also captures a novel feature whereby treated human's blood can become mosquitocidal to the questing mosquitoes upon the successful ingestion of the treated human's blood. Analysis of the disease free system, that is the model in the absence of infection in the human and mosquito populations, reveals the presence of a basic offspring number, N, whose size determines the existence and stability of a thriving mosquito population in the sense that when N≤1 we have only the mosquito extinction steady state which is globally asymptotically stable, while for N > 1 we have the persistent mosquito population steady state which is also globally asymptotically stable for these range of values of N. In the presence of disease, N still strongly affects the properties of the epidemiological model in the sense that for N≤1 the only steady state for the system is the mosquito extinction steady state, which is globally and asymptotically stable. As N increases beyond unity in the epidemiological model, we obtained the epidemiological basic reproduction number, R0. For R0 < 1, the disease free equilibrium, with both healthy thriving susceptible human and mosquito populations, is globally asymptotically stable. Both N and R0 are studied for control purposes and our study highlights that multiple control schemes would have a stronger impact on reducing both N and R0 to values small enough for a possible disease vector control and disease eradication. Our model further illustrates that newly emerged mosquitoes that are infected with the malaria parasite during their first blood meal play an important and strong role in the malaria disease dynamics. Additionally, mosquitoes at later gonotrophic cycle stages also impact the dynamics but their contributions to the total mosquito population size decreases with increasing number of gonotrophic cycles. The size of the contribution into the young mosquito population is also dependent on the length of the gonotrophic cycles, an important bionomic parameter, as well as on how the mosquitoes at the final gonotrophic cycles are incorporated into the modelling scheme.


Subject(s)
Anopheles , Malaria , Models, Biological , Mosquito Vectors , Animals , Anopheles/parasitology , Anopheles/physiology , Humans , Larva/parasitology , Larva/physiology , Malaria/epidemiology , Malaria/transmission , Mosquito Vectors/parasitology , Mosquito Vectors/physiology , Population Dynamics
9.
J Theor Biol ; 460: 37-55, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30296448

ABSTRACT

The cancer stem cell hypothesis has gained currency in recent times but concerns remain about its scientific foundations because of significant gaps that exist between research findings and comprehensive knowledge about cancer stem cells (CSCs). In this light, a mathematical model that considers hematopoietic dynamics in the diseased state of the bone marrow and peripheral blood is proposed and used to address findings about CSCs. The ensuing model, resulting from a modification and refinement of a recent model, develops out of the position that mathematical models of CSC development, that are few at this time, are needed to provide insightful underpinnings for biomedical findings about CSCs as the CSC idea gains traction. Accordingly, the mathematical challenges brought on by the model that mirror general challenges in dealing with nonlinear phenomena are discussed and placed in context. The proposed model describes the logical occurrence of discrete time delays, that by themselves present mathematical challenges, in the evolving cell populations under consideration. Under the challenging circumstances, the steady state properties of the model system of delay differential equations are obtained, analyzed, and the resulting mathematical predictions arising therefrom are interpreted and placed within the framework of findings regarding CSCs. Simulations of the model are carried out by considering various parameter scenarios that reflect different experimental situations involving disease evolution in human hosts. Model analyses and simulations suggest that the emergence of the cancer stem cell population alongside other malignant cells engenders higher dimensions of complexity in the evolution of malignancy in the bone marrow and peripheral blood at the expense of healthy hematopoietic development. The model predicts the evolution of an aberrant environment in which the malignant population particularly in the bone marrow shows tendencies of reaching an uncontrollable equilibrium state. Essentially, the model shows that a structural relationship exists between CSCs and non-stem malignant cells that confers on CSCs the role of temporally enhancing and stimulating the expansion of non-stem malignant cells while also benefitting from increases in their own population and these CSCs may be the main protagonists that drive the ultimate evolution of the uncontrollable equilibrium state of such malignant cells and these may have implications for treatment.


Subject(s)
Bone Marrow/pathology , Models, Theoretical , Neoplastic Stem Cells/pathology , Cell Proliferation , Computer Simulation , Hematopoiesis , Humans , Models, Biological
10.
Math Biosci Eng ; 15(6): 1435-1463, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30418793

ABSTRACT

Oncolytic virotherapy has been emerging as a promising novel cancer treatment which may be further combined with the existing therapeutic modalities to enhance their effects. To investigate how virotherapy could enhance chemotherapy, we propose an ODE based mathematical model describing the interactions between tumour cells, the immune response, and a treatment combination with chemotherapy and oncolytic viruses. Stability analysis of the model with constant chemotherapy treatment rates shows that without any form of treatment, a tumour would grow to its maximum size. It also demonstrates that chemotherapy alone is capable of clearing tumour cells provided that the drug efficacy is greater than the intrinsic tumour growth rate. Furthermore, virotherapy alone may not be able to clear tumour cells from body tissue but would rather enhance chemotherapy if viruses with high viral potency are used. To assess the combined effect of virotherapy and chemotherapy we use the forward sensitivity index to perform a sensitivity analysis, with respect to chemotherapy key parameters, of the virus basic reproductive number and the tumour endemic equilibrium. The results from this sensitivity analysis indicate the existence of a critical dose of chemotherapy above which no further significant reduction in the tumour population can be observed. Numerical simulations show that a successful combinational therapy of the chemotherapeutic drugs and viruses depends mostly on the virus burst size, infection rate, and the amount of drugs supplied. Optimal control analysis was performed, by means of the Pontryagin's maximum principle, to further refine predictions of the model with constant treatment rates by accounting for the treatment costs and sides effects. Results from this analysis suggest that the optimal drug and virus combination correspond to half their maximum tolerated doses. This is in agreement with the results from stability and sensitivity analyses.


Subject(s)
Models, Biological , Neoplasms/therapy , Oncolytic Virotherapy , Animals , Combined Modality Therapy/methods , Combined Modality Therapy/statistics & numerical data , Computer Simulation , Humans , Mathematical Concepts , Neoplasms/drug therapy , Neoplasms/pathology , Oncolytic Virotherapy/statistics & numerical data
11.
PLoS One ; 13(6): e0198280, 2018.
Article in English | MEDLINE | ID: mdl-29879166

ABSTRACT

A campaign for malaria control, using Long Lasting Insecticide Nets (LLINs) was launched in South Sudan in 2009. The success of such a campaign often depends upon adequate available resources and reliable surveillance data which help officials understand existing infections. An optimal allocation of resources for malaria control at a sub-national scale is therefore paramount to the success of efforts to reduce malaria prevalence. In this paper, we extend an existing SIR mathematical model to capture the effect of LLINs on malaria transmission. Available data on malaria is utilized to determine realistic parameter values of this model using a Bayesian approach via Markov Chain Monte Carlo (MCMC) methods. Then, we explore the parasite prevalence on a continued rollout of LLINs in three different settings in order to create a sub-national projection of malaria. Further, we calculate the model's basic reproductive number and study its sensitivity to LLINs' coverage and its efficacy. From the numerical simulation results, we notice a basic reproduction number, [Formula: see text], confirming a substantial increase of incidence cases if no form of intervention takes place in the community. This work indicates that an effective use of LLINs may reduce [Formula: see text] and hence malaria transmission. We hope that this study will provide a basis for recommending a scaling-up of the entry point of LLINs' distribution that targets households in areas at risk of malaria.


Subject(s)
Malaria/epidemiology , Malaria/transmission , Mosquito Control/instrumentation , Humans , Insecticide-Treated Bednets , Malaria/prevention & control , Markov Chains , Models, Theoretical , Monte Carlo Method , Population Surveillance , Prevalence , South Sudan/epidemiology
12.
PLoS One ; 12(9): e0184347, 2017.
Article in English | MEDLINE | ID: mdl-28934210

ABSTRACT

In the present paper, we address by means of mathematical modeling the following main question: How can oncolytic virus infection of some normal cells in the vicinity of tumor cells enhance oncolytic virotherapy? We formulate a mathematical model describing the interactions between the oncolytic virus, the tumor cells, the normal cells, and the antitumoral and antiviral immune responses. The model consists of a system of delay differential equations with one (discrete) delay. We derive the model's basic reproductive number within tumor and normal cell populations and use their ratio as a metric for virus tumor-specificity. Numerical simulations are performed for different values of the basic reproduction numbers and their ratios to investigate potential trade-offs between tumor reduction and normal cells losses. A fundamental feature unravelled by the model simulations is its great sensitivity to parameters that account for most variation in the early or late stages of oncolytic virotherapy. From a clinical point of view, our findings indicate that designing an oncolytic virus that is not 100% tumor-specific can increase virus particles, which in turn, can further infect tumor cells. Moreover, our findings indicate that when infected tissues can be regenerated, oncolytic viral infection of normal cells could improve cancer treatment.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Algorithms , Computer Simulation , Humans , Models, Biological , Neoplasms/immunology , Neoplasms/virology
13.
J Theor Biol ; 404: 312-330, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27317864

ABSTRACT

We present a novel mathematical model involving various immune cell populations and tumor cell populations. The model describes how tumor cells evolve and survive the brief encounter with the immune system mediated by natural killer (NK) cells and the activated CD8(+) cytotoxic T lymphocytes (CTLs). The model is composed of ordinary differential equations describing the interactions between these important immune lymphocytes and various tumor cell populations. Based on up-to-date knowledge of immune evasion and rational considerations, the model is designed to illustrate how tumors evade both arms of host immunity (i.e. innate and adaptive immunity). The model predicts that (a) an influx of an external source of NK cells might play a crucial role in enhancing NK-cell immune surveillance; (b) the host immune system alone is not fully effective against progression of tumor cells; (c) the development of immunoresistance by tumor cells is inevitable in tumor immune surveillance. Our model also supports the importance of infiltrating NK cells in tumor immune surveillance, which can be enhanced by NK cell-based immunotherapeutic approaches.


Subject(s)
Models, Immunological , Neoplasms/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Communication , Computer Simulation , Cytotoxicity, Immunologic , Humans , Immune Evasion , Immunotherapy , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Neoplasms/pathology
14.
Math Biosci ; 274: 83-93, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26877072

ABSTRACT

Stemming from current emerging paradigms related to the cancer stem cell hypothesis, an existing mathematical model is expanded and used to study cell interaction dynamics in the bone marrow and peripheral blood. The proposed mathematical model is described by a system of nonlinear differential equations with delay, to quantify the dynamics in abnormal hematopoiesis. The steady states of the model are analytically and numerically obtained. Some conditions for the local asymptotic stability of such states are investigated. Model analyses suggest that malignancy may be irreversible once it evolves from a nonmalignant state into a malignant one and no intervention takes place. This leads to the proposition that a great deal of emphasis be placed on cancer prevention. Nevertheless, should malignancy arise, treatment programs for its containment or curtailment may have to include a maximum and extensive level of effort to protect normal cells from eventual destruction. Further model analyses and simulations predict that in the untreated disease state, there is an evolution towards a situation in which malignant cells dominate the entire bone marrow - peripheral blood system. Arguments are then advanced regarding requirements for quantitatively understanding cancer stem cell behavior. Among the suggested requirements are, mathematical frameworks for describing the dynamics of cancer initiation and progression, the response to treatment, the evolution of resistance, and malignancy prevention dynamics within the bone marrow - peripheral blood architecture.


Subject(s)
Blood Cells/pathology , Bone Marrow Cells/pathology , Models, Biological , Neoplastic Stem Cells/pathology , Computer Simulation , Hematologic Neoplasms/etiology , Hematologic Neoplasms/pathology , Hematopoiesis , Humans , Mathematical Concepts
15.
AIDS ; 28 Suppl 3: S269-75, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24991900

ABSTRACT

We outline the benefits, challenges and possible approaches to developing mathematical models that could be used to estimate the magnitude of negative consequences of adult HIV infection for children. Adult HIV infection can lead to numerous negative consequences for dependent children, including depression, anxiety, withdrawal from school and early sexual debut, among others. For advocacy and planning purposes, it is important to highlight and consider as many of these as possible. A focus solely on orphan numbers, which is the typical summary measure for children affected by HIV and AIDS, can be misleading. The complexity of child development that is characterized by the interaction of a multitude of proximal and distal factors, coupled with a significant lack of data on child development in the context of adult HIV infection make the development of models a challenging task. Although it may not be possible in the first attempt to develop a population-based model capable of examining family dynamics, the negative consequences together with the impact of interventions, steps in that direction can be taken. We propose approaches and assumptions that we believe will allow the development of a useful first set of models. We conclude with a brief discussion of the type of data that, if collected, would facilitate refinement and development of these models.


Subject(s)
Family Health , HIV Infections/psychology , Mental Disorders/psychology , Models, Theoretical , Parent-Child Relations , Adult , Child , Child, Preschool , Female , Humans
16.
Bull Math Biol ; 76(3): 673-96, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24584715

ABSTRACT

We present a mathematical model for the transmission of Trypanosoma brucei rhodesiense by tsetse vectors to a multi-host population. To control tsetse and T. b. rhodesiense, a proportion, ψ, of cattle (one of the hosts considered in the model) is taken to be kept on treatment with insecticides. Analytical expressions are obtained for the basic reproduction number, R0n in the absence, and R(0n)(T) in the presence of insecticide-treated cattle (ITC). Stability analysis of the disease-free equilibrium was carried out for the case when there is one vertebrate host untreated with insecticide. By considering three vertebrate hosts (cattle, humans and wildlife) the sensitivity analysis was carried out on the basic reproduction number (R(0n)(T)) in the absence and presence of ITC. The results show that R(03)(T) is more sensitive to changes in the tsetse mortality. The model is then used to study the control of tsetse and T. b. rhodesiense in humans through application insecticides to cattle either over the whole-body or to restricted areas of the body known to be favoured tsetse feeding sites. Numerical results show that while both ITC strategies result in decreases in tsetse density and in the incidence of T. b. rhodesiense in humans, the restricted application technique results in improved cost-effectiveness, providing a cheap, safe, environmentally friendly and farmer based strategy for the control of vectors and T. b. rhodesiense in humans.


Subject(s)
Cattle Diseases/prevention & control , Models, Biological , Trypanosoma brucei rhodesiense , Trypanosomiasis, African/veterinary , Tsetse Flies/parasitology , Africa South of the Sahara , Animals , Basic Reproduction Number , Cattle , Cattle Diseases/transmission , Humans , Insect Control/methods , Insect Control/statistics & numerical data , Insect Vectors/parasitology , Insecticides/administration & dosage , Mathematical Concepts , Trypanosomiasis, African/prevention & control , Trypanosomiasis, African/transmission
17.
PLoS One ; 7(9): e44377, 2012.
Article in English | MEDLINE | ID: mdl-22984497

ABSTRACT

We derive a new method to estimate the age specific incidence of an infection with a differential mortality, using individual level infection status data from successive surveys. The method consists of a) an SI-type model to express the incidence rate in terms of the prevalence and its derivatives as well as the difference in mortality rate, and b) a maximum likelihood approach to estimate the prevalence and its derivatives. Estimates can in principle be obtained for any chosen age and time, and no particular assumptions are made about the epidemiological or demographic context. This is in contrast with earlier methods for estimating incidence from prevalence data, which work with aggregated data, and the aggregated effect of demographic and epidemiological rates over the time interval between prevalence surveys. Numerical simulation of HIV epidemics, under the presumption of known excess mortality due to infection, shows improved control of bias and variance, compared to previous methods. Our analysis motivates for a) effort to be applied to obtain accurate estimates of excess mortality rates as a function of age and time among HIV infected individuals and b) use of individual level rather than aggregated data in order to estimate HIV incidence rates at times between two prevalence surveys.


Subject(s)
Epidemics , HIV Infections/epidemiology , HIV/metabolism , Adolescent , Adult , Age Distribution , Age Factors , Computer Simulation , HIV Infections/mortality , HIV Seropositivity , HIV Seroprevalence , Humans , Incidence , Likelihood Functions , Middle Aged , Models, Statistical , Prevalence , Probability , Time Factors
18.
PLoS Negl Trop Dis ; 6(5): e1615, 2012.
Article in English | MEDLINE | ID: mdl-22616017

ABSTRACT

BACKGROUND: In Uganda, Rhodesian sleeping sickness, caused by Trypanosoma brucei rhodesiense, and animal trypanosomiasis caused by T. vivax and T. congolense, are being controlled by treating cattle with trypanocides and/or insecticides. We used a mathematical model to identify treatment coverages required to break transmission when host populations consisted of various proportions of wild and domestic mammals, and reptiles. METHODOLOGY/PRINCIPAL FINDINGS: An Ro model for trypanosomiasis was generalized to allow tsetse to feed off multiple host species. Assuming populations of cattle and humans only, pre-intervention Ro values for T. vivax, T. congolense, and T. brucei were 388, 64 and 3, respectively. Treating cattle with trypanocides reduced R(0) for T. brucei to <1 if >65% of cattle were treated, vs 100% coverage necessary for T. vivax and T. congolense. The presence of wild mammalian hosts increased the coverage required and made control of T. vivax and T. congolense impossible. When tsetse fed only on cattle or humans, R(0) for T. brucei was <1 if 20% of cattle were treated with insecticide, compared to 55% for T. congolense. If wild mammalian hosts were also present, control of the two species was impossible if proportions of non-human bloodmeals from cattle were <40% or <70%, respectively. R(0) was <1 for T. vivax only when insecticide treatment led to reductions in the tsetse population. Under such circumstances R(0)<1 for T. brucei and T. congolense if cattle make up 30% and 55%, respectively of the non-human tsetse bloodmeals, as long as all cattle are treated with insecticide. CONCLUSIONS/SIGNIFICANCE: In settled areas of Uganda with few wild hosts, control of Rhodesian sleeping sickness is likely to be much more effectively controlled by treating cattle with insecticide than with trypanocides.


Subject(s)
Biota , Insecticides/administration & dosage , Trypanocidal Agents/administration & dosage , Trypanosomiasis/drug therapy , Trypanosomiasis/veterinary , Tsetse Flies/growth & development , Animals , Cattle , Humans , Livestock , Mammals , Models, Theoretical , Reptiles , Treatment Outcome , Trypanosomiasis/prevention & control , Trypanosomiasis/transmission , Uganda
19.
Biosystems ; 106(2-3): 136-45, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21843591

ABSTRACT

We derive and analyse a deterministic model for the transmission of malaria disease with mass action form of infection. Firstly, we calculate the basic reproduction number, R(0), and investigate the existence and stability of equilibria. The system is found to exhibit backward bifurcation. The implication of this occurrence is that the classical epidemiological requirement for effective eradication of malaria, R(0)<1, is no longer sufficient, even though necessary. Secondly, by using optimal control theory we derive the conditions under which it is optimal to eradicate the disease and examine the impact of a possible combined vaccination and treatment strategy on the disease transmission. When eradication is impossible, we derive the necessary conditions for optimal control of the disease using Pontryagin's Maximum Principle. The results obtained from the numerical simulations of the model show that a possible vaccination combined with effective treatment regime would reduce the spread of the disease appreciably.


Subject(s)
Disease Eradication/methods , Insect Vectors , Malaria/drug therapy , Malaria/prevention & control , Malaria/transmission , Models, Biological , Animals , Computer Simulation , Humans , Vaccination/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...