New Experimental Models in Prevalent Cancers

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Methods and Technologies Development".

Deadline for manuscript submissions: 31 January 2025 | Viewed by 1842

Special Issue Editors


E-Mail Website
Guest Editor
Department of Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, Caserta, 81100, Italy
Interests: cancer therapy; drug resistance; colorectal cancer; cancer stem cells; apoptosis
Special Issues, Collections and Topics in MDPI journals

E-Mail
Guest Editor
Department of Experimental Medicine and Surgery, Università degli Studi di Roma "Tor Vergata", 00133 Rome, Italy
Interests: signaling pathway; resistance; leukemia; cancer stem cells; apoptosis
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
Interests: drug resistance; apoptosis; colorectal cancer; cancer stem cells; leukemia

E-Mail
Guest Editor
School of Biomedical Convergence Engineering, Pusan National University, Yangsan, Republic of Korea
Interests: cancer stem cells; drug resistance; drug delivery; regenerative medicine; 3D bioprinting; organ-on-a-chip (OOC); iPSC; CRISPR-Cas9

Special Issue Information

Dear Colleagues,

Today, cancer is responsible for approximately 8 million deaths annually worldwide. The Global Burden Of Cancer (GLOBOCAN) 2020 reported 19.3 million new cases of cancer every year, which is projected to increase to 28.4 million by 2040. In the future, female breast cancer will be the most common cancer (11.7%), followed by lung (11.4%), colorectal (10.0%), prostate (7.3%), and stomach (5.6%). As far as cancer-related mortality is concerned, lung cancer is the leading cause, responsible for 1.8 million deaths (18%), followed by colorectal (9.4%), liver (8.3%), stomach (7.7%), and female breast (6.9%). In light of this, it is becoming increasingly important to have a deep understanding of the biology and progression of cancer. New models for research can play a crucial role in better understanding the invasion and metastasis of cancer. Cancer is not a characterizable disease,; instead, it represents a heterogeneous and variable system, so it is crucial to create models that accurately reflect the tumor system. In vitro cancer models often fail to represent the heterogeneity of human cancer cells, their microenvironment, and the stromal compartment, which are important for understanding tumor pathogenesis, therapeutic responses, and adverse reactions. There are various experimental systems available to study human cancer, including cancer cell lines, 3D model organoids, and organisms like Drosophila melanogaster, zebrafish, genetically engineered mouse models, pigs, patient-derived xenografts (PDXs), and computational cancer models. These models allow one to investigate biochemical or genetic pathways and cancer pathologies, such as the best form of therapy and the resistance to drugs. Furthermore, to analyze clinical feedback in patients based on the model utilized, it is important to obtain a 50% hindrance in tumor development to better understand the “response” to treatment and to utilize clinically applicable dosages of curative agents to observe survival agents. It is crucial to choose the right therapy against tumor progression and understand when growth resumes after stopping or delaying treatment. Advanced preclinical models can be vital in elucidating the mechanisms of action of single drugs and their combinations in different types of cancer. Therefore, addressing investigations with more advanced preclinical models is critical to better show the mechanisms of action of single drugs and their combinations in different kinds of cancer. In this regard, it is possible to use different preclinical models such as cell lines, 3D cultures systems, and animal studies. Furthermore, models of patient-derived xenografts (PDXs) and patient-derived cells are grown and then injected into mice to test the effects of several treatments. Another method that can play a crucial role in cancer research is represented by a combination between patient-derived tumor organoids and a microfluidic organ-on-chip system to investigate the cancer’s invasion in the tumor microenvironment (TME). This can enable us to create patient-specific tumor models to study cancer biology and the effect of different drugs. Specifically, this model is very suitable for evaluating therapies’ efficacy and effects, which are very close to the response observed in patients.

This Special Issue will focus on studies regarding the following topics:

  1. The importance of modeling in cancer research and therapeutic development.
  2. Acknowledgment of suitable models in different types of cancer (such as lung, colorectal, breast, prostate, ovarian, and melanoma cancer).
  3. Systems biology approaches for understanding cancer heterogeneity and treatment.
  4. Mathematical modeling of cancer progression and treatment response resistance.
  5. Computational modeling of cancer metastasis and therapeutics. 

Dr. Gabriella Marfe
Dr. Carla Di Stefano
Dr. Giuseppe Mirone
Dr. Arvind Kumar Shukla
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • cell lines
  • 3D cultures systems and animal studies
  • PDXs
  • Drosophila melanogaster
  • zebrafish

Benefits of Publishing in a Special Issue

  • Ease of navigation: Grouping papers by topic helps scholars navigate broad scope journals more efficiently.
  • Greater discoverability: Special Issues support the reach and impact of scientific research. Articles in Special Issues are more discoverable and cited more frequently.
  • Expansion of research network: Special Issues facilitate connections among authors, fostering scientific collaborations.
  • External promotion: Articles in Special Issues are often promoted through the journal's social media, increasing their visibility.
  • e-Book format: Special Issues with more than 10 articles can be published as dedicated e-books, ensuring wide and rapid dissemination.

Further information on MDPI's Special Issue polices can be found here.

Published Papers (2 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

12 pages, 3268 KiB  
Article
Development of New Diffuse Large B Cell Lymphoma Mouse Models
by Syed Hassan Mehdi, Ying-Zhi Xu, Leonard D. Shultz, Eunkyung Kim, Yong Gu Lee, Samantha Kendrick and Donghoon Yoon
Cancers 2024, 16(17), 3006; https://doi.org/10.3390/cancers16173006 - 29 Aug 2024
Viewed by 603
Abstract
Diffuse large B cell lymphoma (DLBCL) is the most diagnosed, aggressive non-Hodgkin lymphoma, with ~40% of patients experiencing refractory or relapsed disease. Given the low response rates to current therapy, alternative treatment strategies are necessary to improve patient outcomes. Here, we sought to [...] Read more.
Diffuse large B cell lymphoma (DLBCL) is the most diagnosed, aggressive non-Hodgkin lymphoma, with ~40% of patients experiencing refractory or relapsed disease. Given the low response rates to current therapy, alternative treatment strategies are necessary to improve patient outcomes. Here, we sought to develop an easily accessible new xenograft mouse model that better recapitulates the human disease for preclinical studies. We generated two Luciferase (Luc)-EGFP-expressing human DLBCL cell lines representing the different DLBCL cell-of-origin subtypes. After intravenous injection of these cells into humanized NSG mice, we monitored the tumor growth and evaluated the organ-specific engraftment/progression period. Our results showed that human IL6-expressing NSG (NSG-IL6) mice were highly permissive for DLBCL cell growth. In NSG-IL6 mice, systemic engraftments of both U2932 activated B cell-like- and VAL germinal B cell-like-DLBCL (engraftment rate; 75% and 82%, respectively) were detected within 2nd-week post-injection. In the organ-specific ex vivo evaluation, both U2932-Luc and VAL-Luc cells were initially engrafted and expanded in the spleen, liver, and lung and subsequently in the skeleton, ovary, and brain. Consistent with the dual BCL2/MYC translocation association with poor patient outcomes, VAL cells showed heightened proliferation in human IL6-conditioned media and caused rapid tumor expansion and early death in the engrafted mice. We concluded that the U2932 and VAL cell-derived human IL6-expressing mouse models reproduced the clinical features of an aggressive DLBCL with a highly consistent pattern of tumor development. Based on these findings, NSG mice expressing human IL6 have the potential to serve as a new tool to develop DLBCL xenograft models to overcome the limitations of standard subcutaneous DLBCL xenografts. Full article
(This article belongs to the Special Issue New Experimental Models in Prevalent Cancers)
Show Figures

Figure 1

14 pages, 4305 KiB  
Article
Development and Characterization of Syngeneic Orthotopic Transplant Models of Obesity-Responsive Triple-Negative Breast Cancer in C57BL/6J Mice
by Meredith S. Carson, Patrick D. Rädler, Jody Albright, Melissa VerHague, Erika T. Rezeli, Daniel Roth, John E. French, Charles M. Perou, Stephen D. Hursting and Michael F. Coleman
Cancers 2024, 16(16), 2803; https://doi.org/10.3390/cancers16162803 - 9 Aug 2024
Viewed by 830
Abstract
Obesity is an established risk and progression factor for triple-negative breast cancer (TNBC), but preclinical studies to delineate the mechanisms underlying the obesity-TNBC link as well as strategies to break that link are constrained by the lack of tumor models syngeneic to obesity-prone [...] Read more.
Obesity is an established risk and progression factor for triple-negative breast cancer (TNBC), but preclinical studies to delineate the mechanisms underlying the obesity-TNBC link as well as strategies to break that link are constrained by the lack of tumor models syngeneic to obesity-prone mouse strains. C3(1)/SV40 T-antigen (C3-TAg) transgenic mice on an FVB genetic background develop tumors with molecular and pathologic features that closely resemble human TNBC, but FVB mice are resistant to diet-induced obesity (DIO). Herein, we sought to develop transplantable C3-TAg cell lines syngeneic to C57BL/6 mice, an inbred mouse strain that is sensitive to DIO. We backcrossed FVB-Tg(C3-1-TAg)cJeg/JegJ to C57BL/6 mice for ten generations, and spontaneous tumors from those mice were excised and used to generate four clonal cell lines (B6TAg1.02, B6TAg2.03, B6TAg2.10, and B6TAg2.51). We characterized the growth of the four cell lines in both lean and DIO C57BL/6J female mice and performed transcriptomic profiling. Each cell line was readily tumorigenic and had transcriptional profiles that clustered as claudin-low, yet markedly differed from each other in their rate of tumor progression and transcriptomic signatures for key metabolic, immune, and oncogenic signaling pathways. DIO accelerated tumor growth of orthotopically transplanted B6TAg1.02, B6TAg2.03, and B6TAg2.51 cells. Thus, the B6TAg cell lines described herein offer promising and diverse new models to augment the study of DIO-associated TNBC. Full article
(This article belongs to the Special Issue New Experimental Models in Prevalent Cancers)
Show Figures

Figure 1

Back to TopTop