1
|
O'Connor CM, Zawacki KP, Durrant JR, Barrie K, Walton RG, King D, Onishi GH, Noto FK, Hinderman M, Begemann D, Branyan T, Panwell K, DiFeo A, Schlosser MJ, Narla G. The SRG RAT® supports human cell xenotransplantation through enhanced tumor microenvironment interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645250. [PMID: 40236141 PMCID: PMC11996424 DOI: 10.1101/2025.03.27.645250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The use of immunodeficient mice for human tumor engraftment is an essential model of human cancer, with uses ranging from basic science to translational research. However, low engraftment rates, slow growth, and smaller tumor volumes can be limitations. Previously, we reported a highly immunodeficient rat strain with the functional deletion of both the Rag2 and Il2rg genes on the Sprague-Dawley background ( SRG RAT ® ), which lacks B, T, and NK cells. Here, we subcutaneously engrafted two cell-derived xenograft (CDX) and seven patient-derived xenograft (PDX) models, including prostate, lung, ovarian, and uterine cancer models, into the SRG rat or NSG mouse models and tracked tumor growth. In all cases, the engraftment and tumor growth rates were better supported in the SRG rat compared to the NSG mouse. Interestingly, the SRG rat is not more immunocompromised than the NSG mouse, suggesting alternative mechanisms leading to the supportive growth in the SRG rat. Therefore, we explored potential differences in the tumor microenvironment (TME) between models grown in the two host animals. Lung PDX models grown in SRG rats showed enhanced formation of vasculature and stroma and were morphologically more consistent with the originating patient tumors. IHC analysis of the NCI-H660 CDX model showed differences in the tumors' stroma, vasculature, and macrophages when grown in the two host species. Single-cell spatial imaging of engrafted tumors showed upregulation of the human CXCL2 and TCIM in NCI-H660 tumors grown in the SRG rat versus the NSG mouse, both of which have been linked to poor prognosis in cancer. Combined, our data demonstrate that the SRG rat supports the growth of multiple human cancer types and displays enhanced tumor microenvironment interactions compared to NSG mice.
Collapse
|
2
|
Lanari C, Novaro V, Rossi M, C Kordon E. Buenos Aires Breast Cancer Symposium (BA-BCS 2024) A Second Successful "Trial" for Bringing Together both World Hemispheres To Debate the Future of Translational Breast Cancer Research. J Mammary Gland Biol Neoplasia 2025; 30:5. [PMID: 40088327 DOI: 10.1007/s10911-025-09577-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
The second edition of the Buenos Aires Breast Cancer Symposium (BA-BCS), the first held in person, took place from September 3rd to 6th in Buenos Aires, Argentina. This report provides an overview of the talks delivered as individual lectures or as part of the mini-symposia illustrating the diversity and complexity of the topics discussed throughout the meeting. The event brought together leading scientists and clinical experts dedicated to advancing breast cancer research and improving therapeutic strategies and patient care. The contributions from both speakers and attendees fostered an extraordinary atmosphere at the gathering in Buenos Aires. We hope these interactions will lead to more translational approaches and promote future collaborations.
Collapse
Affiliation(s)
- Claudia Lanari
- Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Virginia Novaro
- Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Mario Rossi
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Pilar, Argentina
| | - Edith C Kordon
- Instituto de Fisiología, Biología Molecular y Neurociencias, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Tecnológicas (IFIBYNE-UBA-CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
3
|
Blanchard Z, Brown EA, Ghazaryan A, Welm AL. PDX models for functional precision oncology and discovery science. Nat Rev Cancer 2025; 25:153-166. [PMID: 39681638 PMCID: PMC12124142 DOI: 10.1038/s41568-024-00779-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 12/18/2024]
Abstract
Precision oncology relies on detailed molecular analysis of how diverse tumours respond to various therapies, with the aim to optimize treatment outcomes for individual patients. Patient-derived xenograft (PDX) models have been key to preclinical validation of precision oncology approaches, enabling the analysis of each tumour's unique genomic landscape and testing therapies that are predicted to be effective based on specific mutations, gene expression patterns or signalling abnormalities. To extend these standard precision oncology approaches, the field has strived to complement the otherwise static and often descriptive measurements with functional assays, termed functional precision oncology (FPO). By utilizing diverse PDX and PDX-derived models, FPO has gained traction as an effective preclinical and clinical tool to more precisely recapitulate patient biology using in vivo and ex vivo functional assays. Here, we explore advances and limitations of PDX and PDX-derived models for precision oncology and FPO. We also examine the future of PDX models for precision oncology in the age of artificial intelligence. Integrating these two disciplines could be the key to fast, accurate and cost-effective treatment prediction, revolutionizing oncology and providing patients with cancer with the most effective, personalized treatments.
Collapse
Affiliation(s)
- Zannel Blanchard
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Elisabeth A Brown
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Arevik Ghazaryan
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Alana L Welm
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA.
| |
Collapse
|
4
|
Pataccini G, Elia A, Sequeira G, Ambrosio L, Coianis M, Lamb CA, Rojas PA, Martínez Vázquez P, Burruchaga J, Spengler E, Vanzulli SI, Abba M, Lanari C. Steroid hormone receptors, exome sequencing and treatment responsiveness of breast cancer patient-derived xenografts originated in a South American country. Sci Rep 2025; 15:2415. [PMID: 39827242 PMCID: PMC11742900 DOI: 10.1038/s41598-025-86389-x] [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: 09/10/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Breast cancer (BC) patient-derived xenografts (PDX) are relevant models for precision medicine. However, there are no collections derived from South American BC patients. Since ethnicity significantly impacts clinical outcomes, it is necessary to develop PDX models from different lineages. Our goals were to a) develop BC PDX from our population; b) characterize the expression of estrogen (ER), progesterone (PR), androgen (AR) and glucocorticoid (GR) receptors, basal and luminal cytokeratins, EGFR and HER2; c) identify PDX mutations; d) evaluate the response to treatments selected based on their biological and genetic features, and e) perform BC tissue cultures (BCTC) from PDX tissues and compare in vivo and ex vivo results. Surgical fragments were maintained in a culture medium and inoculated subcutaneously into untreated NSG female mice, or treated with estradiol pellets. Other fragments were fixed in formalin for diagnosis and immunohistochemistry, and a third piece was frozen at -80°C for molecular studies or whole exome-sequencing. Tumors were serially transplanted into NSG mice. Once the PDX was established, in vivo and ex vivo drug responses were evaluated. Eight PDX were established: two ER + [BC-AR685 (PR +) and BC-AR707 (PR-)], one from a triple-negative (TN) recurrence whose primary tumor was ER + (BC-AR485), one HER2 + (BC-AR474) and four TN primary tumors (BC-AR553, BC-AR546, BC-AR631 and BC-AR687). BC-AR685 had higher levels of PR isoform A than isoform B and was sensitive to mifepristone, tamoxifen, and palbociclib. BC-AR707 was inhibited by tamoxifen and testosterone. BC-AR474 was inhibited by trastuzumab and trastuzumab emtansine. BC-AR485 was sensitive to doxorubicin and resistant to paclitaxel in vivo and ex vivo. BC-AR687 carried a PIK3CA (C420R) mutation and was sensitive to alpelisib and mTOR inhibitors. All PDX expressed AR with varying intensities. GR and AR were co-expressed in the ER + tumors and in 3 TN PDX. We report the first PDX originated from South American countries that were genetically and biologically characterized and may be used in precision medicine studies. PDX expressing AR and/or GR are powerful tools to evaluate different endocrine treatment combinations even in TN tumors.
Collapse
Affiliation(s)
- Gabriela Pataccini
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Andrés Elia
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Gonzalo Sequeira
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Luisa Ambrosio
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Marcela Coianis
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Caroline A Lamb
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Paola A Rojas
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | | | - Javier Burruchaga
- Hospital de Agudos Magdalena V. de Martínez, General Pacheco, Buenos Aires, Argentina
| | - Eunice Spengler
- Hospital de Agudos Magdalena V. de Martínez, General Pacheco, Buenos Aires, Argentina
| | | | - Martin Abba
- Universidad Nacional de La Plata, La Plata, Argentina
| | - Claudia Lanari
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina.
| |
Collapse
|
5
|
Khairani AF, Harmonia S, Chou Y, Alfarafisa NM, Ramadhanti J. Optimizing Xenograft Models for Breast Cancer: A Comparative Analysis of Cell-Derived and Patient-Derived Implantation Techniques in Pre-Clinical Research. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:1-10. [PMID: 39811602 PMCID: PMC11727321 DOI: 10.2147/bctt.s490532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025]
Abstract
Purpose The high mortality rate of breast cancer motivates researchers to search for effective treatments. Due to their ability to simulate human conditions, xenograft models such as CDX (Cell line-Derived Xenografts) and PDX (Patient-Derived Xenografts) have gained popularity in pre-clinical research. The choice of xenograft technique is influenced by the type of tumor employed, particularly in more aggressive tumor models like TNBC with metastases. Subcutaneous or orthotopic implantation may influence tumor engraftment rates and the applicability of the models for drug testing. To optimize xenograft models and support the development of breast cancer drugs, selecting a suitable transplantation technique is essential to attaining the best results. Methods This scoping review used PRISMA-Scr methodology to summarize findings from eleven articles published between 2012 and 2024 on pre-clinical trials related to xenograft models for breast cancer considering PDX began traction after 2010. Using specific criteria, the review included studies from electronic platforms. The inclusion criteria ensured relevant English sources were available in full text, while the exclusion criteria eliminated certain types of articles and inadequately comprehensive studies. Results Subcutaneous and orthotopic implantation are critical methods for xenograft models in cancer research. Subcutaneous implantation is less invasive and more manageable but does not fully mimic the tumor's natural environment. Orthotopic implantation accurately mimic the migration, invasion, and molecular characteristics of the original tumor, although the procedure is more complex and requires specialized techniques. The specific research objectives determine their choice, the need for accurate tumor replication, and the testing convenience. Conclusion Orthotopic implantation is the preferable method for developing PDX and CDX models of breast cancer because it closely mimics the tumor microenvironment and metastatic behavior, yielding clinically relevant results for drug testing. Subcutaneous implantation may result in higher engraftment rates, but it cannot accurately represent the complexity of tumors.
Collapse
Affiliation(s)
- Astrid Feinisa Khairani
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Graduate School of Master Program in Anti Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Shella Harmonia
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Yoan Chou
- Graduate School of Master Program in Anti Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Nayla Majeda Alfarafisa
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Julia Ramadhanti
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
6
|
Silva J, Oliveira PA, Duarte JA, Faustino-Rocha AI. Mammary Cancer Models: An Overview from the Past to the Future. In Vivo 2025; 39:1-16. [PMID: 39740866 PMCID: PMC11705154 DOI: 10.21873/invivo.13800] [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: 09/12/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 01/02/2025]
Abstract
Breast cancer research heavily relies on diverse model systems to comprehend disease progression, develop novel diagnostics, and evaluate new therapeutic strategies. This review offers a comprehensive overview of mammary cancer models, covering both ex vivo and in vivo approaches. We delve into established techniques, such as cell culture and explore cutting-edge advancements, like tumor-on-a-chip and bioprinting. The in vivo section encompasses spontaneous, induced, and transplanted models, genetically engineered models, chick chorioallantoic membrane assays, and the burgeoning field of in silico models. Additionally, this article briefly highlights the key discoveries made using these models, significantly enhancing our understanding of breast cancer. In essence, this article serves as a comprehensive compass, charting the trajectory of mammary cancer modeling from its early beginnings to the promising vistas of tomorrow.
Collapse
Affiliation(s)
- Jessica Silva
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal;
- Institute for Innovation, Capacity Building and Sustainability of Agri-food Production (Inov4Agro), Vila Real, Portugal
| | - Paula A Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-food Production (Inov4Agro), Vila Real, Portugal
- Department of Veterinary Sciences, UTAD, Vila Real, Portugal
| | - José Alberto Duarte
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, Gandra, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, Portugal
| | - Ana I Faustino-Rocha
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-food Production (Inov4Agro), Vila Real, Portugal
- Department of Zootechnics, School of Sciences and Technology, University of Évora, Évora, Portugal
- Comprehensive Health Research Center (CHRC), University of Évora, Évora, Portugal
| |
Collapse
|
7
|
Uijttewaal ECH, Lee J, Sell AC, Botay N, Vainorius G, Novatchkova M, Baar J, Yang J, Potzler T, van der Leij S, Lowden C, Sinner J, Elewaut A, Gavrilovic M, Obenauf A, Schramek D, Elling U. CRISPR-StAR enables high-resolution genetic screening in complex in vivo models. Nat Biotechnol 2024:10.1038/s41587-024-02512-9. [PMID: 39681701 DOI: 10.1038/s41587-024-02512-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024]
Abstract
Pooled genetic screening with CRISPR-Cas9 has enabled genome-wide, high-resolution mapping of genes to phenotypes, but assessing the effect of a given genetic perturbation requires evaluation of each single guide RNA (sgRNA) in hundreds of cells to counter stochastic genetic drift and obtain robust results. However, resolution is limited in complex, heterogeneous models, such as organoids or tumors transplanted into mice, because achieving sufficient representation requires impractical scaling. This is due to bottleneck effects and biological heterogeneity of cell populations. Here we introduce CRISPR-StAR, a screening method that uses internal controls generated by activating sgRNAs in only half the progeny of each cell subsequent to re-expansion of the cell clone. Our method overcomes both intrinsic and extrinsic heterogeneity as well as genetic drift in bottlenecks by generating clonal, single-cell-derived intrinsic controls. We use CRISPR-StAR to identify in-vivo-specific genetic dependencies in a genome-wide screen in mouse melanoma. Benchmarking against conventional screening demonstrates the improved data quality provided by this technology.
Collapse
Affiliation(s)
- Esther C H Uijttewaal
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria
- Vienna BioCenter PhD Program, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Joonsun Lee
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria
- Vienna BioCenter PhD Program, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Annika Charlotte Sell
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria
| | - Naomi Botay
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria
| | - Gintautas Vainorius
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria
- Vienna BioCenter PhD Program, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Maria Novatchkova
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria
- Research Institute of Molecular Pathology, Campus-Vienna-Biocenter 1, Vienna BioCenter (VBC), Vienna, Austria
| | - Juliane Baar
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria
| | - Jiaye Yang
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria
| | - Tobias Potzler
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria
| | - Sophie van der Leij
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria
| | - Christopher Lowden
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Julia Sinner
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria
| | - Anais Elewaut
- Vienna BioCenter PhD Program, University of Vienna and Medical University of Vienna, Vienna, Austria
- Research Institute of Molecular Pathology, Campus-Vienna-Biocenter 1, Vienna BioCenter (VBC), Vienna, Austria
| | - Milanka Gavrilovic
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria
| | - Anna Obenauf
- Research Institute of Molecular Pathology, Campus-Vienna-Biocenter 1, Vienna BioCenter (VBC), Vienna, Austria
| | - Daniel Schramek
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Ulrich Elling
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), Vienna, Austria.
- Viverita Discovery, Vienna, Austria.
| |
Collapse
|
8
|
Li M, Guo X, Verma A, Rudkouskaya A, McKenna AM, Intes X, Wang G, Barroso M. Contrast-enhanced photon-counting micro-CT of tumor xenograft models. Phys Med Biol 2024; 69:155011. [PMID: 38670143 PMCID: PMC11258216 DOI: 10.1088/1361-6560/ad4447] [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: 01/12/2024] [Revised: 04/11/2024] [Accepted: 04/26/2024] [Indexed: 04/28/2024]
Abstract
Objective. Photon-counting micro-computed tomography (micro-CT) is a major advance in small animal preclinical imaging. Small molecule- and nanoparticle-based contrast agents have been widely used to enable the differentiation of liver tumors from surrounding tissues using photon-counting micro-CT. However, there is a notable gap in the application of these market-available agents to the imaging of breast and ovarian tumors using photon-counting micro-CT. Herein, we have used photon-counting micro-CT to determine the effectiveness of these contrast agents in differentiating ovarian and breast tumor xenografts in live, intact mice.Approach. Nude mice carrying different types of breast and ovarian tumor xenografts (AU565, MDA-MB-231 and SKOV-3 human cancer cells) were injected with ISOVUE-370 (a small molecule-based agent) or Exitron Nano 12000 (a nanoparticle-based agent) and subjected to photon-counting micro-CT. To improve tumor visualization using photon-counting micro-CT, we developed a novel color visualization method, which changes color tones to highlight contrast media distribution, offering a robust alternative to traditional material decomposition methods with less computational demand.Main results. Ourin vivoexperiments confirm the effectiveness of this color visualization approach, showing distinct enhancement characteristics for each contrast agent. Qualitative and quantitative analyses suggest that Exitron Nano 12000 provides superior vasculature enhancement and better quantitative consistency across scans, while ISOVUE-370 delivers a more comprehensive tumor enhancement but with significant variance between scans due to its short blood half-time. Further, a paired t-test on mean and standard deviation values within tumor volumes showed significant differences between the AU565 and SKOV-3 tumor models with the nanoparticle-based contrast agent (p-values < 0.02), attributable to their distinct vascularity, as confirmed by immunohistochemical analysis.Significance. These findings underscore the utility of photon-counting micro-CT in non-invasively assessing the morphology and anatomy of different tumor xenografts, which is crucial for tumor characterization and longitudinal monitoring of tumor progression and response to treatments.
Collapse
Affiliation(s)
- Mengzhou Li
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Xiaodong Guo
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Amit Verma
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, United States of America
| | - Alena Rudkouskaya
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, United States of America
| | - Antigone M McKenna
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Xavier Intes
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Ge Wang
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, United States of America
| |
Collapse
|
9
|
Naser IH, Zaid M, Ali E, Jabar HI, Mustafa AN, Alubiady MHS, Ramadan MF, Muzammil K, Khalaf RM, Jalal SS, Alawadi AH, Alsalamy A. Unveiling innovative therapeutic strategies and future trajectories on stimuli-responsive drug delivery systems for targeted treatment of breast carcinoma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3747-3770. [PMID: 38095649 DOI: 10.1007/s00210-023-02885-9] [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: 09/10/2023] [Accepted: 12/02/2023] [Indexed: 05/23/2024]
Abstract
This comprehensive review delineates the latest advancements in stimuli-responsive drug delivery systems engineered for the targeted treatment of breast carcinoma. The manuscript commences by introducing mammary carcinoma and the current therapeutic methodologies, underscoring the urgency for innovative therapeutic strategies. Subsequently, it elucidates the logic behind the employment of stimuli-responsive drug delivery systems, which promise targeted drug administration and the minimization of adverse reactions. The review proffers an in-depth analysis of diverse types of stimuli-responsive systems, including thermoresponsive, pH-responsive, and enzyme-responsive nanocarriers. The paramount importance of material choice, biocompatibility, and drug loading strategies in the design of these systems is accentuated. The review explores characterization methodologies for stimuli-responsive nanocarriers and probes preclinical evaluations of their efficacy, toxicity, pharmacokinetics, and biodistribution in mammary carcinoma models. Clinical applications of stimuli-responsive systems, ongoing clinical trials, the potential of combination therapies, and the utility of multifunctional nanocarriers for the co-delivery of assorted drugs and therapies are also discussed. The manuscript addresses the persistent challenge of drug resistance in mammary carcinoma and the potential of stimuli-responsive systems in surmounting it. Regulatory and safety considerations, including FDA guidelines and biocompatibility assessments, are outlined. The review concludes by spotlighting future trajectories and emergent technologies in stimuli-responsive drug delivery, focusing on pioneering approaches, advancements in nanotechnology, and personalized medicine considerations. This review aims to serve as a valuable compendium for researchers and clinicians interested in the development of efficacious and safe stimuli-responsive drug delivery systems for the treatment of breast carcinoma.
Collapse
Affiliation(s)
- Israa Habeeb Naser
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Muhaned Zaid
- Department of Pharmacy, Al-Manara College for Medical Sciences, Maysan, Amarah, Iraq
| | - Eyhab Ali
- Al-Zahraa University for Women, Karbala, Iraq
| | - Hayder Imad Jabar
- Department of Pharmaceutics, College of Pharmacy, University of Al-Ameed, Karbala, Iraq
| | | | | | | | - Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University, Abha, Saudi Arabia
| | | | - Sarah Salah Jalal
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Ahmed Hussien Alawadi
- College of Technical Engineering, the Islamic University, Najaf, Iraq
- College of Technical Engineering, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, the Islamic University of Babylon, Babylon, Iraq
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq.
| |
Collapse
|
10
|
Brugnoli F, Dell’Aira M, Tedeschi P, Grassilli S, Pierantoni M, Foschi R, Bertagnolo V. Effects of Garlic on Breast Tumor Cells with a Triple Negative Phenotype: Peculiar Subtype-Dependent Down-Modulation of Akt Signaling. Cells 2024; 13:822. [PMID: 38786044 PMCID: PMC11119207 DOI: 10.3390/cells13100822] [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: 03/29/2024] [Revised: 04/30/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Breast cancer includes tumor subgroups with morphological, molecular, and clinical differences. Intrinsic heterogeneity especially characterizes breast tumors with a triple negative phenotype, often leading to the failure of even the most advanced therapeutic strategies. To improve breast cancer treatment, the use of natural agents to integrate conventional therapies is the subject of ever-increasing attention. In this context, garlic (Allium sativum) shows anti-cancerous potential, interfering with the proliferation, motility, and malignant progression of both non-invasive and invasive breast tumor cells. As heterogeneity could be at the basis of variable effects, the main objective of our study was to evaluate the anti-tumoral activity of a garlic extract in breast cancer cells with a triple negative phenotype. Established triple negative breast cancer (TNBC) cell lines from patient-derived xenografts (PDXs) were used, revealing subtype-dependent effects on morphology, cell cycle, and invasive potential, correlated with the peculiar down-modulation of Akt signaling, a crucial regulator in solid tumors. Our results first demonstrate that the effects of garlic on TNBC breast cancer are not unique and suggest that only more precise knowledge of the mechanisms activated by this natural compound in each tumor will allow for the inclusion of garlic in personalized therapeutic approaches to breast cancer.
Collapse
Affiliation(s)
- Federica Brugnoli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.B.); (M.D.); (M.P.); (R.F.)
| | - Marcello Dell’Aira
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.B.); (M.D.); (M.P.); (R.F.)
| | - Paola Tedeschi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Silvia Grassilli
- Department of Environmental Sciences and Prevention and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Marina Pierantoni
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.B.); (M.D.); (M.P.); (R.F.)
| | - Rebecca Foschi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.B.); (M.D.); (M.P.); (R.F.)
| | - Valeria Bertagnolo
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.B.); (M.D.); (M.P.); (R.F.)
| |
Collapse
|
11
|
Li M, Guo X, Verma A, Rudkouskaya A, McKenna AM, Intes X, Wang G, Barroso M. Contrast-enhanced photon-counting micro-CT of tumor xenograft models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.574097. [PMID: 38260707 PMCID: PMC10802390 DOI: 10.1101/2024.01.03.574097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Photon-counting micro computed tomography (micro-CT) offers new potential in preclinical imaging, particularly in distinguishing materials. It becomes especially helpful when combined with contrast agents, enabling the differentiation of tumors from surrounding tissues. There are mainly two types of contrast agents in the market for micro-CT: small molecule-based and nanoparticle-based. However, despite their widespread use in liver tumor studies, there is a notable gap in research on the application of these commercially available agents for photon-counting micro-CT in breast and ovarian tumors. Herein, we explored the effectiveness of these agents in differentiating tumor xenografts from various origins (AU565, MDA-MB-231, and SKOV-3) in nude mice, using photon-counting micro-CT. Specifically, ISOVUE-370 (a small molecule-based agent) and Exitrone Nano 12000 (a nanoparticle-based agent) were investigated in this context. To improve tumor visualization, we proposed a novel color visualization method for photon-counting micro-CT, which changes color tones to highlight contrast media distribution, offering a robust alternative to traditional material decomposition methods with less computational demand. Our in vivo experiments confirm its effectiveness, showing distinct enhancement characteristics for each contrast agent. Qualitative and quantitative analyses suggested that Exitrone Nano 12000 provides superior vasculature enhancement and better quantitative consistency across scans, while ISOVUE-370 gives more comprehensive tumor enhancement but with a significant variance between scans due to its short blood half-time. This variability leads to high sensitivity to timing and individual differences among mice. Further, a paired t-test on mean and standard deviation values within tumor volumes showed significant differences between the AU565 and SKOV-3 tumor models with the nanoparticle-based (p-values < 0.02), attributable to their distinct vascularity, as confirmed by immunohistochemistry. These findings underscore the utility of photon-counting micro-CT in non-invasively assessing the morphology and anatomy of different tumor xenografts, which is crucial for tumor characterization and longitudinal monitoring of tumor development and response to treatments.
Collapse
Affiliation(s)
- Mengzhou Li
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Xiaodong Guo
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Amit Verma
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Alena Rudkouskaya
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Antigone M. McKenna
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Xavier Intes
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Ge Wang
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
12
|
Zhang C, Sui Y, Liu S, Yang M. In vitro and in vivo experimental models for cancer immunotherapy study. CURRENT RESEARCH IN BIOTECHNOLOGY 2024; 7:100210. [DOI: 10.1016/j.crbiot.2024.100210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025] Open
|
13
|
Jain M, Goel A. Current Insights in Murine Models for Breast Cancer: Present, Past and Future. Curr Pharm Des 2024; 30:2267-2275. [PMID: 38910416 DOI: 10.2174/0113816128306053240604074142] [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: 01/23/2024] [Revised: 04/25/2024] [Accepted: 05/09/2024] [Indexed: 06/25/2024]
Abstract
Breast cancer is an intricate disease that is increasing at a fast pace, and numerous heterogeneities within it further make it difficult to investigate. We have always used animal models to understand cancer pathology and create an in vivo microenvironment that closely resembles human cancer. They are considered an indispensable part of any clinical investigation regarding cancer. Animal models have a high potency in identifying the relevant biomarkers and genetic pathways involved in the course of disease prognosis. Researchers have previously explored a variety of organisms, including Drosophila melanogaster, zebrafish, and guinea pigs, to analyse breast cancer, but murine models have proven the most comprehensive due to their homologous nature with human chromosomes, easy availability, simple gene editing, and high adaptability. The available models have their pros and cons, and it depends on the researcher to select the one most relevant to their research question. Chemically induced models are cost-effective and simple to create. Transplantation models such as allografts and xenografts can mimic the human breast cancer environment reliably. Genetically engineered mouse models (GEMMs) help to underpin the genetic alterations involved and test novel immunotherapies. Virus-mediated models and gene knockout models have also provided new findings regarding breast cancer progression and metastasis. These mouse models have also enabled the visualization of breast cancer metastases. It is also imperative to consider the cost-effectiveness of these models. Despite loopholes, mouse models have evolved and are required for disease analysis.
Collapse
Affiliation(s)
- Mansi Jain
- Department of Biotechnology, GLA University, Mathura, India
| | - Anjana Goel
- Department of Biotechnology, GLA University, Mathura, India
| |
Collapse
|
14
|
Witt BL, Tollefsbol TO. Molecular, Cellular, and Technical Aspects of Breast Cancer Cell Lines as a Foundational Tool in Cancer Research. Life (Basel) 2023; 13:2311. [PMID: 38137912 PMCID: PMC10744609 DOI: 10.3390/life13122311] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Breast cancer comprises about 30% of all new female cancers each year and is the most common malignant cancer in women in the United States. Breast cancer cell lines have been harnessed for many years as a foundation for in vitro analytic studies to understand the use of cancer prevention and therapy. There has yet to be a compilation of works to analyze the pitfalls, novel discoveries, and essential techniques for breast cancer cell line studies in a scientific context. In this article, we review the history of breast cancer cell lines and their origins, as well as analyze the molecular pathways that pharmaceutical drugs apply to breast cancer cell lines in vitro and in vivo. Controversies regarding the origins of certain breast cancer cell lines, the benefits of utilizing Patient-Derived Xenograft (PDX) versus Cell-Derived Xenograft (CDX), and 2D versus 3D cell culturing techniques will be analyzed. Novel outcomes from epigenetic discovery with dietary compound usage are also discussed. This review is intended to create a foundational tool that will aid investigators when choosing a breast cancer cell line to use in multiple expanding areas such as epigenetic discovery, xenograft experimentation, and cancer prevention, among other areas.
Collapse
Affiliation(s)
- Brittany L. Witt
- Department of Biology, University of Alabama at Birmingham, 902 14th Street, Birmingham, AL 35228, USA;
| | - Trygve O. Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 902 14th Street, Birmingham, AL 35228, USA;
- Integrative Center for Aging Research, University of Alabama at Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, 1802 6th Avenue South, Birmingham, AL 35294, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, 1675 University Boulevard, Birmingham, AL 35294, USA
- Comprehensive Diabetes Center, University of Alabama at Birmingham, 1825 University Boulevard, Birmingham, AL 35294, USA
- University Wide Microbiome Center, University of Alabama at Birmingham, 845 19th Street South, Birmingham, AL 35294, USA
| |
Collapse
|
15
|
Cao C, Lu X, Guo X, Zhao H, Gao Y. Patient-derived models: Promising tools for accelerating the clinical translation of breast cancer research findings. Exp Cell Res 2023; 425:113538. [PMID: 36871856 DOI: 10.1016/j.yexcr.2023.113538] [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: 12/12/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/06/2023]
Abstract
Breast cancer has become the highest incidence of cancer in women. It was extensively and deeply studied by biologists and medical workers worldwide. However, the meaningful results in lab researches cannot be realized in clinical, and a part of new drugs in clinical experiments do not obtain as good results as the preclinical researches. It is urgently that promote a kind of breast cancer research models that can get study results closer to the physiological condition of the human body. Patient-derived models (PDMs) originating from clinical tumor, contain primary elements of tumor and maintain key clinical features of tumor. So they are promising research models to facilitate laboratory researches translate to clinical application, and predict the treatment outcome of patients. In this review, we summarize the establishment of PDMs of breast cancer, reviewed the application of PDMs in clinical translational researches and personalized precision medicine with breast cancer as an example, to improve the understanding of PDMs among researchers and clinician, facilitate them to use PDMs on a large scale of breast cancer researches and promote the clinical translation of laboratory research and new drug development.
Collapse
Affiliation(s)
- Changqing Cao
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, China; State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, China
| | - Xiyan Lu
- Department of Outpatient, The Second Affiliated Hospital of Air Force Medical University, China
| | - Xinyan Guo
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, China
| | - Huadong Zhao
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, China.
| | - Yuan Gao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, China.
| |
Collapse
|