1
|
Frisbie L, Pressimone C, Dyer E, Baruwal R, Garcia G, St Croix C, Watkins S, Calderone M, Gorecki G, Javed Z, Atiya HI, Hempel N, Pearson A, Coffman LG. Carcinoma-associated mesenchymal stem cells promote ovarian cancer heterogeneity and metastasis through mitochondrial transfer. Cell Rep 2024; 43:114551. [PMID: 39067022 PMCID: PMC11420855 DOI: 10.1016/j.celrep.2024.114551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/03/2024] [Accepted: 07/12/2024] [Indexed: 07/30/2024] Open
Abstract
Ovarian cancer is characterized by early metastatic spread. This study demonstrates that carcinoma-associated mesenchymal stromal cells (CA-MSCs) enhance metastasis by increasing tumor cell heterogeneity through mitochondrial donation. CA-MSC mitochondrial donation preferentially occurs in ovarian cancer cells with low levels of mitochondria ("mito poor"). CA-MSC mitochondrial donation rescues the phenotype of mito poor cells, restoring their proliferative capacity, resistance to chemotherapy, and cellular respiration. Receipt of CA-MSC-derived mitochondria induces tumor cell transcriptional changes leading to the secretion of ANGPTL3, which enhances the proliferation of tumor cells without CA-MSC mitochondria, thus amplifying the impact of mitochondrial transfer. Donated CA-MSC mitochondrial DNA persisted in recipient tumor cells for at least 14 days. CA-MSC mitochondrial donation occurs in vivo, enhancing tumor cell heterogeneity and decreasing mouse survival. Collectively, this work identifies CA-MSC mitochondrial transfer as a critical mediator of ovarian cancer cell survival, heterogeneity, and metastasis and presents a unique therapeutic target in ovarian cancer.
Collapse
Affiliation(s)
- Leonard Frisbie
- Department of Integrative Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Emma Dyer
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Roja Baruwal
- Molecular Pharmacology Graduate Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Geyon Garcia
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Claudette St Croix
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Simon Watkins
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Calderone
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Grace Gorecki
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Zaineb Javed
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Huda I Atiya
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Nadine Hempel
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Alexander Pearson
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA; Comprehensive Cancer Center, The University of Chicago, Chicago, IL, USA
| | - Lan G Coffman
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee Women's Research Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Mahasa KJ, Ouifki R, de Pillis L, Eladdadi A. A Role of Effector CD 8 + T Cells Against Circulating Tumor Cells Cloaked with Platelets: Insights from a Mathematical Model. Bull Math Biol 2024; 86:89. [PMID: 38884815 DOI: 10.1007/s11538-024-01323-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
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.
Collapse
Affiliation(s)
- Khaphetsi Joseph Mahasa
- Department of Mathematics and Computer Science, National University of Lesotho, Roma, Maseru, Lesotho.
| | - Rachid Ouifki
- Department of Mathematics and Applied Mathematics, Mafikeng Campus, North-West University, Private Bag X2046, Mmabatho, 2735, South Africa
| | | | - Amina Eladdadi
- Division of Mathematical Sciences, The National Science Foundation, Alexandria, VA, USA
| |
Collapse
|
3
|
Thompson JF, Williams GJ. When does a melanoma metastasize? Implications for management. Oncotarget 2024; 15:374-378. [PMID: 38870033 PMCID: PMC11174830 DOI: 10.18632/oncotarget.28591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024] Open
Abstract
Selecting which patients with clinically-localized melanoma require treatment other than wide excision of the primary tumor is based on the risk or presence of metastatic disease. This in turn is linked to survival. Knowing if and when a melanoma is likely to metastasize is therefore of great importance. Several studies employing a range of different methodologies have suggested that many melanomas metastasize long before the primary lesion is diagnosed. Therefore, waiting for dissemination of metastatic disease to become evident before making systemic therapy available to these patients may be less effective than giving them post-operative adjuvant therapy initially if the metastatic risk is high. The identification of these high-risk patients will assist in selecting those to whom adjuvant systemic therapy can most appropriately be offered. Further studies are required to better identify high-risk patients whose primary melanoma is likely to have already metastasized.
Collapse
Affiliation(s)
- John F. Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Faculty of Health and Medical Sciences, University of Western Australia, Perth, WA, Australia
| | - Gabrielle J. Williams
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
4
|
Etehadtavakol M, Etehadtavakol M, Ng EYK. Enhanced thyroid nodule segmentation through U-Net and VGG16 fusion with feature engineering: A comprehensive study. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 251:108209. [PMID: 38723436 DOI: 10.1016/j.cmpb.2024.108209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/28/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND AND OBJECTIVE The thyroid gland, a key component of the endocrine system, is pivotal in regulating bodily functions. Thermography, a non-invasive imaging technique utilizing infrared cameras, has emerged as a diagnostic tool for thyroid-related conditions, offering advantages such as early detection and risk stratification. Artificial intelligence (AI) has demonstrated success in medical diagnostics, and its integration into thermal imaging analysis holds promise for improving diagnostic capabilities. This study aims to explore the potential of AI, specifically convolutional neural networks (CNNs), in enhancing the analysis of thyroid thermograms for the detection of nodules and abnormalities. METHODS Artificial intelligence (AI) and machine learning techniques are integrated to enhance thyroid thermal image analysis. Specifically, a fusion of U-Net and VGG16, combined with feature engineering (FE), is proposed for accurate thyroid nodule segmentation. The novelty of this research lies in leveraging feature engineering in transfer learning for the segmentation of thyroid nodules, even in the presence of a limited dataset. RESULTS The study presents results from four conducted studies, demonstrating the efficacy of this approach even with a limited dataset. It's observed that in study 4, using FE has led to a significant improvement in the value of the dice coefficient. Even for the small size of the masked region, incorporating radiomics with FE resulted in significant improvements in the segmentation dice coefficient. It's promising that one can achieve higher dice coefficients by employing different models and refining them. CONCLUSION The findings here underscore the potential of AI for precise and efficient segmentation of thyroid nodules, paving the way for improved thyroid health assessment.
Collapse
Affiliation(s)
- Mehdi Etehadtavakol
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran; Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahnaz Etehadtavakol
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81745-33871, Iran
| | - Eddie Y K Ng
- School of Mechanical and Aerospace Engineering, College of Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore.
| |
Collapse
|
5
|
Zhdanov VP. Kinetics of cancer metastasis. Biosystems 2024; 235:105098. [PMID: 38056592 DOI: 10.1016/j.biosystems.2023.105098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/14/2023] [Accepted: 12/02/2023] [Indexed: 12/08/2023]
Abstract
The formation of metastases during cancer is now considered to be induced by migrating metastatic stem cells (MetSCs) in preexisting niches or niches induced by MetSCs or tumor-derived exosomes (TDEs). I propose and compare two simplest generic models describing these two scenarios. The number of tumors is predicted (i) to increase exponentially in the case of preexisting niches and (ii) to diverge during a finite time interval in the case of induced niches. The latter prediction is novel and of interest because rapid collapse in the end of a finite time interval is a well-known feature of the cancer metastasis. Two advanced models describing the two scenarios of cancer metastasis have been scrutinized as well. These models clarify the likely role of various specific factors in the metastasis. In particular, the equations derived in the framework of the advanced model with preexisting niches have been solved analytically allowing (i) to clarify the factors determining the duration of the period from the initiation of the primary tumor to the phase when the metastases start to dominate, (ii) to estimate the number of metastases in the end of this period, and (iii) to explains why the use of chemotherapy typically results in the improvement of the patient state only for a relatively short period. The equations derived in the framework of the advanced model with induced niches have no analytical solution, and their analysis merits additional attention.
Collapse
Affiliation(s)
- Vladimir P Zhdanov
- Boreskov Institute of Catalysis, Russian Academy of Sciences, Novosibirsk, Russia.
| |
Collapse
|
6
|
Placha W, Suder P, Panek A, Bronowicka-Adamska P, Zarzycka M, Szczygieł M, Zagajewski J, Piwowar MW. The Blocking of Drug Resistance Channels by Selected Hydrophobic Statins in Chemoresistance Human Melanoma. Biomolecules 2023; 13:1682. [PMID: 38136555 PMCID: PMC10741734 DOI: 10.3390/biom13121682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
Despite the development of modern drugs, drug resistance in oncology remains the main factor limiting the curability of patients. This paper shows the use of a group of hydrophobic statins to inhibit drug resistance (Pgp protein). In a chemoresistance melanoma cell model, viability, necroptosis with DNA damage, the absorption of the applied pharmaceuticals, and the functional activity of the ABCB1 drug transporter after administration of docetaxel or docetaxel with a selected hydrophobic statin were studied. Taxol-resistant human melanoma cells from three stages of development were used as a model: both A375P and WM239A metastatic lines and radial growth phase WM35 cells. An animal model (Mus musculus SCID) was developed for the A375P cell line. The results show that hydrophobic statins administered with docetaxel increase the accumulation of the drug in the tumor cell a.o. by blocking the ABCB1 channel. They reduce taxol-induced drug resistance. The tumor size reduction was observed after the drug combination was administrated. It was shown that the structural similarity of statins is of secondary importance, e.g., pravastatin and simvastatin. Using cytostatics in the presence of hydrophobic statins increases their effectiveness while reducing their overall toxicity.
Collapse
Affiliation(s)
- Wojciech Placha
- Department of Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7b St., 31-034 Krakow, Poland; (P.B.-A.); (M.Z.); (J.Z.)
| | - Piotr Suder
- Department of Analytical Chemistry and Biochemistry, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, 31-007 Krakow, Poland;
| | - Agnieszka Panek
- Institute of Nuclear Physics Polish Academy of Sciences, 31-342 Krakow, Poland;
| | - Patrycja Bronowicka-Adamska
- Department of Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7b St., 31-034 Krakow, Poland; (P.B.-A.); (M.Z.); (J.Z.)
| | - Marta Zarzycka
- Department of Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7b St., 31-034 Krakow, Poland; (P.B.-A.); (M.Z.); (J.Z.)
| | - Małgorzata Szczygieł
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, 31-007 Krakow, Poland;
| | - Jacek Zagajewski
- Department of Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7b St., 31-034 Krakow, Poland; (P.B.-A.); (M.Z.); (J.Z.)
| | - Monika Weronika Piwowar
- Department of Bioinformatics and Telemedicine, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7e St., 31-034 Krakow, Poland;
| |
Collapse
|
7
|
Gallaher J, Strobl M, West J, Gatenby R, Zhang J, Robertson-Tessi M, Anderson AR. Intermetastatic and Intrametastatic Heterogeneity Shapes Adaptive Therapy Cycling Dynamics. Cancer Res 2023; 83:2775-2789. [PMID: 37205789 PMCID: PMC10425736 DOI: 10.1158/0008-5472.can-22-2558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/11/2023] [Accepted: 05/17/2023] [Indexed: 05/21/2023]
Abstract
Adaptive therapies that alternate between drug applications and drug-free vacations can exploit competition between sensitive and resistant cells to maximize the time to progression. However, optimal dosing schedules depend on the properties of metastases, which are often not directly measurable in clinical practice. Here, we proposed a framework for estimating features of metastases through tumor response dynamics during the first adaptive therapy treatment cycle. Longitudinal prostate-specific antigen (PSA) levels in 16 patients with metastatic castration-resistant prostate cancer undergoing adaptive androgen deprivation treatment were analyzed to investigate relationships between cycle dynamics and clinical variables such as Gleason score, the change in the number of metastases over a cycle, and the total number of cycles over the course of treatment. The first cycle of adaptive therapy, which consists of a response period (applying therapy until 50% PSA reduction), and a regrowth period (removing treatment until reaching initial PSA levels), delineated several features of the computational metastatic system: larger metastases had longer cycles; a higher proportion of drug-resistant cells slowed the cycles; and a faster cell turnover rate sped up drug response time and slowed regrowth time. The number of metastases did not affect cycle times, as response dynamics were dominated by the largest tumors rather than the aggregate. In addition, systems with higher intermetastasis heterogeneity responded better to continuous therapy and correlated with dynamics from patients with high or low Gleason scores. Conversely, systems with higher intrametastasis heterogeneity responded better to adaptive therapy and correlated with dynamics from patients with intermediate Gleason scores. SIGNIFICANCE Multiscale mathematical modeling combined with biomarker dynamics during adaptive therapy helps identify underlying features of metastatic cancer to inform treatment decisions.
Collapse
Affiliation(s)
- Jill Gallaher
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Maximilian Strobl
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Jeffrey West
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Robert Gatenby
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida
- Department of Radiology, Moffitt Cancer Center, Tampa, Florida
| | - Jingsong Zhang
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Mark Robertson-Tessi
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida
| | | |
Collapse
|
8
|
Silva-Aravena F, Núñez Delafuente H, Gutiérrez-Bahamondes JH, Morales J. A Hybrid Algorithm of ML and XAI to Prevent Breast Cancer: A Strategy to Support Decision Making. Cancers (Basel) 2023; 15:cancers15092443. [PMID: 37173910 PMCID: PMC10177162 DOI: 10.3390/cancers15092443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Worldwide, the coronavirus has intensified the management problems of health services, significantly harming patients. Some of the most affected processes have been cancer patients' prevention, diagnosis, and treatment. Breast cancer is the most affected, with more than 20 million cases and at least 10 million deaths by 2020. Various studies have been carried out to support the management of this disease globally. This paper presents a decision support strategy for health teams based on machine learning (ML) tools and explainability algorithms (XAI). The main methodological contributions are: first, the evaluation of different ML algorithms that allow classifying patients with and without cancer from the available dataset; and second, an ML methodology mixed with an XAI algorithm, which makes it possible to predict the disease and interpret the variables and how they affect the health of patients. The results show that first, the XGBoost Algorithm has a better predictive capacity, with an accuracy of 0.813 for the train data and 0.81 for the test data; and second, with the SHAP algorithm, it is possible to know the relevant variables and their level of significance in the prediction, and to quantify the impact on the clinical condition of the patients, which will allow health teams to offer early and personalized alerts for each patient.
Collapse
Affiliation(s)
- Fabián Silva-Aravena
- Facultad de Ciencias Sociales y Económicas, Universidad Católica del Maule, Avenida San Miguel 3605, Talca 3460000, Chile
| | - Hugo Núñez Delafuente
- Doctorado en Sistemas de Ingeniería, Facultad de Ingeniería, Universidad de Talca, Camino Los Niches Km 1, Curicó 3340000, Chile
| | - Jimmy H Gutiérrez-Bahamondes
- Doctorado en Sistemas de Ingeniería, Facultad de Ingeniería, Universidad de Talca, Camino Los Niches Km 1, Curicó 3340000, Chile
| | - Jenny Morales
- Facultad de Ciencias Sociales y Económicas, Universidad Católica del Maule, Avenida San Miguel 3605, Talca 3460000, Chile
| |
Collapse
|
9
|
Metastasis prevention: How to catch metastatic seeds. Biochim Biophys Acta Rev Cancer 2023; 1878:188867. [PMID: 36842768 DOI: 10.1016/j.bbcan.2023.188867] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/09/2023] [Accepted: 02/18/2023] [Indexed: 02/26/2023]
Abstract
Despite considerable advances in the evolution of anticancer therapies, metastasis still remains the main cause of cancer mortality. Therefore, current strategies for cancer cure should be redirected towards prevention of metastasis. Targeting metastatic pathways represents a promising therapeutic opportunity aimed at obstructing tumor cell dissemination and metastatic colonization. In this review, we focus on preclinical studies and clinical trials over the last five years that showed high efficacy in suppressing metastasis through targeting lymph node dissemination, tumor cell extravasation, reactive oxygen species, pre-metastatic niche, exosome machinery, and dormancy.
Collapse
|
10
|
Li X, Li M. The application of zebrafish patient-derived xenograft tumor models in the development of antitumor agents. Med Res Rev 2023; 43:212-236. [PMID: 36029178 DOI: 10.1002/med.21924] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/09/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023]
Abstract
The cost of antitumor drug development is enormous, yet the clinical outcomes are less than satisfactory. Therefore, it is of great importance to develop effective drug screening methods that enable accurate, rapid, and high-throughput discovery of lead compounds in the process of preclinical antitumor drug research. An effective solution is to use the patient-derived xenograft (PDX) tumor animal models, which are applicable for the elucidation of tumor pathogenesis and the preclinical testing of novel antitumor compounds. As a promising screening model organism, zebrafish has been widely applied in the construction of the PDX tumor model and the discovery of antineoplastic agents. Herein, we systematically survey the recent cutting-edge advances in zebrafish PDX models (zPDX) for studies of pathogenesis mechanisms and drug screening. In addition, the techniques used in the construction of zPDX are summarized. The advantages and limitations of the zPDX are also discussed in detail. Finally, the prospects of zPDX in drug discovery, translational medicine, and clinical precision medicine treatment are well presented.
Collapse
Affiliation(s)
- Xiang Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
11
|
Gasparini A, Humphreys K. Estimating latent, dynamic processes of breast cancer tumour growth and distant metastatic spread from mammography screening data. Stat Methods Med Res 2022; 31:862-881. [PMID: 35103530 PMCID: PMC9099158 DOI: 10.1177/09622802211072496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We propose a framework for jointly modelling tumour size at diagnosis and time to
distant metastatic spread, from diagnosis, based on latent dynamic sub-models of
growth of the primary tumour and of distant metastatic detection. The framework
also includes a sub-model for screening sensitivity as a function of latent
tumour size. Our approach connects post-diagnosis events to the natural history
of cancer and, once refined, may prove useful for evaluating new interventions,
such as personalised screening regimes. We evaluate our model-fitting procedure
using Monte Carlo simulation, showing that the estimation algorithm can retrieve
the correct model parameters, that key patterns in the data can be captured by
the model even with misspecification of some structural assumptions, and that,
still, with enough data it should be possible to detect strong
misspecifications. Furthermore, we fit our model to observational data from an
extension of a case-control study of post-menopausal breast cancer in Sweden,
providing model-based estimates of the probability of being free from detected
distant metastasis as a function of tumour size, mode of detection (of the
primary tumour), and screening history. For women with screen-detected cancer
and two previous negative screens, the probabilities of being free from detected
distant metastases 5 years after detection and removal of the primary tumour are
0.97, 0.89 and 0.59 for tumours of diameter 5, 15 and 35 mm, respectively. We
also study the probability of having latent/dormant metastases at detection of
the primary tumour, estimating that 33% of patients in our study had such
metastases.
Collapse
Affiliation(s)
- Alessandro Gasparini
- Alessandro Gasparini, Department of Medical
Epidemiology and Biostatistics, Karolinska Institutet, PO Box 281, SE-17177,
Stockholm, Sweden.
| | | |
Collapse
|
12
|
Zhang S, Gong C, Ruiz-Martinez A, Wang H, Davis-Marcisak E, Deshpande A, Popel AS, Fertig EJ. Integrating single cell sequencing with a spatial quantitative systems pharmacology model spQSP for personalized prediction of triple-negative breast cancer immunotherapy response. ACTA ACUST UNITED AC 2021; 1-2. [PMID: 34708216 PMCID: PMC8547770 DOI: 10.1016/j.immuno.2021.100002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Response to cancer immunotherapies depends on the complex and dynamic interactions between T cell recognition and killing of cancer cells that are counteracted through immunosuppressive pathways in the tumor microenvironment. Therefore, while measurements such as tumor mutational burden provide biomarkers to select patients for immunotherapy, they neither universally predict patient response nor implicate the mechanisms that underlie immunotherapy resistance. Recent advances in single-cell RNA sequencing technology measure cellular heterogeneity within cells of an individual tumor but have yet to realize the promise of predictive oncology. In addition to data, mechanistic multiscale computational models are developed to predict treatment response. Incorporating single-cell data from tumors to parameterize these computational models provides deeper insights into prediction of clinical outcome in individual patients. Here, we integrate whole-exome sequencing and scRNA-seq data from Triple-Negative Breast Cancer patients to model neoantigen burden in tumor cells as input to a spatial Quantitative System Pharmacology model. The model comprises a four-compartmental Quantitative System Pharmacology sub-model to represent a whole patient and a spatial agent-based sub-model to represent tumor volumes at the cellular scale. We use the high-throughput single-cell data to model the role of antigen burden and heterogeneity relative to the tumor microenvironment composition on predicted immunotherapy response. We demonstrate how this integrated modeling and single-cell analysis framework can be used to relate neoantigen heterogeneity to immunotherapy treatment outcomes. Our results demonstrate feasibility of merging single-cell data to initialize cell states in multiscale computational models such as the spQSP for personalized prediction of clinical outcomes to immunotherapy.
Collapse
Affiliation(s)
- Shuming Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Chang Gong
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alvaro Ruiz-Martinez
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Emily Davis-Marcisak
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Atul Deshpande
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Elana J Fertig
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
13
|
Neinavaie F, Ibrahim-Hashim A, Kramer AM, Brown JS, Richards CL. The Genomic Processes of Biological Invasions: From Invasive Species to Cancer Metastases and Back Again. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.681100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The concept of invasion is useful across a broad range of contexts, spanning from the fine scale landscape of cancer tumors up to the broader landscape of ecosystems. Invasion biology provides extraordinary opportunities for studying the mechanistic basis of contemporary evolution at the molecular level. Although the field of invasion genetics was established in ecology and evolution more than 50 years ago, there is still a limited understanding of how genomic level processes translate into invasive phenotypes across different taxa in response to complex environmental conditions. This is largely because the study of most invasive species is limited by information about complex genome level processes. We lack good reference genomes for most species. Rigorous studies to examine genomic processes are generally too costly. On the contrary, cancer studies are fortified with extensive resources for studying genome level dynamics and the interactions among genetic and non-genetic mechanisms. Extensive analysis of primary tumors and metastatic samples have revealed the importance of several genomic mechanisms including higher mutation rates, specific types of mutations, aneuploidy or whole genome doubling and non-genetic effects. Metastatic sites can be directly compared to primary tumor cell counterparts. At the same time, clonal dynamics shape the genomics and evolution of metastatic cancers. Clonal diversity varies by cancer type, and the tumors’ donor and recipient tissues. Still, the cancer research community has been unable to identify any common events that provide a universal predictor of “metastatic potential” which parallels findings in evolutionary ecology. Instead, invasion in cancer studies depends strongly on context, including order of events and clonal composition. The detailed studies of the behavior of a variety of human cancers promises to inform our understanding of genome level dynamics in the diversity of invasive species and provide novel insights for management.
Collapse
|
14
|
González JR, Damião C, Moran M, Pantaleão CA, Cruz RA, Balarini GA, Conci A. A Computational Study on the Role of Parameters for Identification of Thyroid Nodules by Infrared Images (and Comparison with Real Data). SENSORS (BASEL, SWITZERLAND) 2021; 21:4459. [PMID: 34209986 PMCID: PMC8272175 DOI: 10.3390/s21134459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 01/03/2023]
Abstract
According to experts and medical literature, healthy thyroids and thyroids containing benign nodules tend to be less inflamed and less active than those with malignant nodules. It seems to be a consensus that malignant nodules have more blood veins and more blood circulation. This may be related to the maintenance of the nodule's heat at a higher level compared with neighboring tissues. If the internal heat modifies the skin radiation, then it could be detected by infrared sensors. The goal of this work is the investigation of the factors that allow this detection, and the possible relation with any pattern referent to nodule malignancy. We aim to consider a wide range of factors, so a great number of numerical simulations of the heat transfer in the region under analysis, based on the Finite Element method, are performed to study the influence of each nodule and patient characteristics on the infrared sensor acquisition. To do so, the protocol for infrared thyroid examination used in our university's hospital is simulated in the numerical study. This protocol presents two phases. In the first one, the body under observation is in steady state. In the second one, it is submitted to thermal stress (transient state). Both are simulated in order to verify if it is possible (by infrared sensors) to identify different behavior referent to malignant nodules. Moreover, when the simulation indicates possible important aspects, patients with and without similar characteristics are examined to confirm such influences. The results show that the tissues between skin and thyroid, as well as the nodule size, have an influence on superficial temperatures. Other thermal parameters of thyroid nodules show little influence on surface infrared emissions, for instance, those related to the vascularization of the nodule. All details of the physical parameters used in the simulations, characteristics of the real nodules and thermal examinations are publicly available, allowing these simulations to be compared with other types of heat transfer solutions and infrared examination protocols. Among the main contributions of this work, we highlight the simulation of the possible range of parameters, and definition of the simulation approach for mapping the used infrared protocol, promoting the investigation of a possible relation between the heat transfer process and the data obtained by infrared acquisitions.
Collapse
Affiliation(s)
- José R. González
- Institute of Computing, Fluminense Federal University, Niterói, Rio de Janeiro 24220-900, Brazil; (M.M.); (A.C.)
| | - Charbel Damião
- Department of Internal Medicine, Fluminense Federal University, Niterói, Rio de Janeiro 24033-900, Brazil; (C.D.); (C.A.P.); (R.A.C.); (G.A.B.)
| | - Maira Moran
- Institute of Computing, Fluminense Federal University, Niterói, Rio de Janeiro 24220-900, Brazil; (M.M.); (A.C.)
| | - Cristina A. Pantaleão
- Department of Internal Medicine, Fluminense Federal University, Niterói, Rio de Janeiro 24033-900, Brazil; (C.D.); (C.A.P.); (R.A.C.); (G.A.B.)
| | - Rubens A. Cruz
- Department of Internal Medicine, Fluminense Federal University, Niterói, Rio de Janeiro 24033-900, Brazil; (C.D.); (C.A.P.); (R.A.C.); (G.A.B.)
| | - Giovanna A. Balarini
- Department of Internal Medicine, Fluminense Federal University, Niterói, Rio de Janeiro 24033-900, Brazil; (C.D.); (C.A.P.); (R.A.C.); (G.A.B.)
| | - Aura Conci
- Institute of Computing, Fluminense Federal University, Niterói, Rio de Janeiro 24220-900, Brazil; (M.M.); (A.C.)
| |
Collapse
|
15
|
Sun R, Nikolakopoulos AN. Elements and evolutionary determinants of genomic divergence between paired primary and metastatic tumors. PLoS Comput Biol 2021; 17:e1008838. [PMID: 33730105 PMCID: PMC8007046 DOI: 10.1371/journal.pcbi.1008838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/29/2021] [Accepted: 02/26/2021] [Indexed: 12/24/2022] Open
Abstract
Can metastatic-primary (M-P) genomic divergence measured from next generation sequencing reveal the natural history of metastatic dissemination? This remains an open question of utmost importance in facilitating a deeper understanding of metastatic progression, and thereby, improving its prevention. Here, we utilize mathematical and computational modeling to tackle this question as well as to provide a framework that illuminates the fundamental elements and evolutionary determinants of M-P divergence. Our framework facilitates the integration of sequencing detectability of somatic variants, and hence, paves the way towards bridging the measurable between-tumor heterogeneity with analytical modeling and interpretability. We show that the number of somatic variants of the metastatic seeding cell that are experimentally undetectable in the primary tumor, can be characterized as the path of the phylogenetic tree from the last appearing variant of the seeding cell back to the most recent detectable variant. We find that the expected length of this path is principally determined by the decay in detectability of the variants along the seeding cell's lineage; and thus, exhibits a significant dependence on the underlying tumor growth dynamics. A striking implication of this fact, is that dissemination from an advanced detectable subclone of the primary tumor can lead to an abrupt drop in the expected measurable M-P divergence, thereby breaking the previously assumed monotonic relation between seeding time and M-P divergence. This is emphatically verified by our single cell-based spatial tumor growth simulation, where we find that M-P divergence exhibits a non-monotonic relationship with seeding time when the primary tumor grows under branched and linear evolution. On the other hand, a monotonic relationship holds when we condition on the dynamics of progressive diversification, or by restricting the seeding cells to always originate from undetectable subclones. Our results highlight the fact that a precise understanding of tumor growth dynamics is the sine qua non for exploiting M-P divergence to reconstruct the chronology of metastatic dissemination. The quantitative models presented here enable further careful evaluation of M-P divergence in association with crucial evolutionary and sequencing parameters.
Collapse
Affiliation(s)
- Ruping Sun
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Athanasios N. Nikolakopoulos
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|