Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (142)

Search Parameters:
Keywords = ex vivo expansion

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 3664 KiB  
Article
Feasibility of Manufacturing and Antitumor Activity of TIL for Advanced Endometrial Cancers
by Yongliang Zhang, Kathleen N. Moore, Amir A. Jazaeri, Judy Fang, Ilabahen Patel, Andrew Yuhas, Patrick Innamarato, Nathan Gilbert, Joseph W. Dean, Behzad Damirchi, Joe Yglesias, Rongsu Qi, Michelle R. Simpson-Abelson, Erwin Cammaart, Sean R. R. Hall and Hequn Yin
Int. J. Mol. Sci. 2025, 26(15), 7151; https://doi.org/10.3390/ijms26157151 - 24 Jul 2025
Viewed by 582
Abstract
Lifileucel, a tumor-infiltrating lymphocyte (TIL) cell therapy approved for advanced melanoma, demonstrates promise for treating other solid tumors, including endometrial cancer (EC). The current study evaluates the feasibility of manufacturing TILs from EC tumors using Iovance’s proprietary 22-day Gen2 manufacturing process. Key parameters, [...] Read more.
Lifileucel, a tumor-infiltrating lymphocyte (TIL) cell therapy approved for advanced melanoma, demonstrates promise for treating other solid tumors, including endometrial cancer (EC). The current study evaluates the feasibility of manufacturing TILs from EC tumors using Iovance’s proprietary 22-day Gen2 manufacturing process. Key parameters, including TIL yield, viability, immune phenotype, T-cell receptor clonality, and cytotoxic activity, were assessed. Of the 11 EC tumor samples processed at research scale, 10 (91%) successfully generated >1 × 109 viable TIL cells, with a median yield of 1.1 × 1010 cells and a median viability of 82.8%. Of the four EC tumor samples processed at full scale, all achieved the pre-specified TVC and viability targets. Putative tumor-reactive T-cell clones were maintained throughout the manufacturing process. Functional reactivity was evidenced by the upregulation of 4-1BB in CD8+ T cells, OX40 in CD4+ T cells, and increased production of IFN-γ and TNF-α upon autologous tumor stimulation. Furthermore, antitumor activity was confirmed using an in vitro autologous tumor organoid killing assay. These findings demonstrate the feasibility of ex vivo TIL expansion from EC tumors. This study provides a rationale for the initiation of the phase II clinical trial IOV-END-201 (NCT06481592) to evaluate lifileucel in patients with advanced EC. Full article
(This article belongs to the Special Issue Endometrial Cancer: From Basic Science to Novel Therapeutics)
Show Figures

Figure 1

16 pages, 5794 KiB  
Article
A More Rapid Method for Culturing LUHMES-Derived Neurons Provides Greater Cell Numbers and Facilitates Studies of Multiple Viruses
by Adam W. Whisnant, Stephanie E. Clark, José Alberto Aguilar-Briseño, Lorellin A. Durnell, Arnhild Grothey, Ann M. Miller, Steven M. Varga, Jeffery L. Meier, Charles Grose, Patrick L. Sinn, Jessica M. Tucker, Caroline C. Friedel, Wendy J. Maury, David H. Price and Lars Dölken
Viruses 2025, 17(7), 1001; https://doi.org/10.3390/v17071001 - 16 Jul 2025
Viewed by 385
Abstract
The ability to study mature neuronal cells ex vivo is complicated by their non-dividing nature and difficulty in obtaining large numbers of primary cells from organisms. Thus, numerous transformed progenitor models have been developed that can be routinely cultured, then scaled, and differentiated [...] Read more.
The ability to study mature neuronal cells ex vivo is complicated by their non-dividing nature and difficulty in obtaining large numbers of primary cells from organisms. Thus, numerous transformed progenitor models have been developed that can be routinely cultured, then scaled, and differentiated to mature neurons. In this paper, we present a new method for differentiating one such model, the Lund human mesencephalic (LUHMES) dopaminergic neurons. This method is two days faster than some established protocols, results in nearly five times greater numbers of mature neurons, and involves fewer handling steps that could introduce technical variability. Moreover, it overcomes the problem of cell aggregate formation that commonly impedes high-resolution imaging, cell dissociation, and downstream analysis. While recently established for herpes simplex virus type 1, we demonstrate that LUHMES neurons can facilitate studies of other herpesviruses, as well as RNA viruses associated with childhood encephalitis and hemorrhagic fever. This protocol provides an improvement in the generation of large-scale neuronal cultures, which may be readily applicable to other neuronal 2D cell culture models and provides a system for studying neurotrophic viruses. We named this method the Streamlined Protocol for Enhanced Expansion and Differentiation Yield, or SPEEDY, method. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Figure 1

15 pages, 1192 KiB  
Review
Natural Killer Cell and Extracellular Vesicle-Based Immunotherapy in Thyroid Cancer: Advances, Challenges, and Future Perspectives
by Kruthika Prakash, Ramya Lakshmi Rajendran, Sanjana Dhayalan, Prakash Gangadaran, Byeong-Cheol Ahn and Kandasamy Nagarajan Aruljothi
Cells 2025, 14(14), 1087; https://doi.org/10.3390/cells14141087 - 16 Jul 2025
Viewed by 613
Abstract
Thyroid cancer, the most frequently occurring endocrine neoplasm, comprises a heterogeneous group of histological subtypes, spanning from the indolent papillary thyroid carcinoma (PTC) to the rapidly progressive and lethal anaplastic thyroid carcinoma (ATC). Although conventional therapies, such as surgery and radioactive iodine (RAI), [...] Read more.
Thyroid cancer, the most frequently occurring endocrine neoplasm, comprises a heterogeneous group of histological subtypes, spanning from the indolent papillary thyroid carcinoma (PTC) to the rapidly progressive and lethal anaplastic thyroid carcinoma (ATC). Although conventional therapies, such as surgery and radioactive iodine (RAI), are effective for differentiated thyroid cancers, treatment resistance and poor prognosis remain major challenges in advanced and undifferentiated forms. In current times, growing attention has been directed toward the potential of Natural Killer (NK) cells as a promising immunotherapeutic avenue. These innate immune cells are capable of direct cytotoxicity against tumor cells, but their efficiency is frequently compromised by the immunosuppressive tumor microenvironment (TME), which inhibits NK cell activation, infiltration, and persistence. This review explores the dynamic interaction between NK cells and the TME in thyroid cancer, detailing key mechanisms of immune evasion, including the impact of suppressive cytokines, altered chemokine landscapes, and inhibitory ligand expression. We further discuss latest advancements in NK cell-based immunotherapies, including strategies for ex vivo expansion, genetic modification, and combinatorial approaches with checkpoint inhibitors or cytokines. Additionally, emerging modalities, such as NK cell-derived extracellular vesicles, are addressed. By combining mechanistic insights with advancing therapeutic techniques, this review provides a comprehensive perspective on NK cell-based interventions and their future potential in improving outcomes for patients with thyroid cancer. Full article
Show Figures

Figure 1

21 pages, 9022 KiB  
Article
Ex Vivo and Simulation Comparison of Leakage in End-to-End Versus End-to-Side Anastomosed Porcine Large Intestine
by Youssef Fahmy, Mohamed Trabia, Brian Ward, Lucas Gallup and Whitney Elks
Bioengineering 2025, 12(7), 676; https://doi.org/10.3390/bioengineering12070676 - 20 Jun 2025
Viewed by 482
Abstract
Anastomotic leaks after colorectal resection are serious surgical complications. We have compared the integrity of two common colorectal anastomosis techniques, end-to-side (ES) and end-to-end (EE), to control specimens using a novel experimental setup that mimics anastomotic air leak tests, which are typically performed [...] Read more.
Anastomotic leaks after colorectal resection are serious surgical complications. We have compared the integrity of two common colorectal anastomosis techniques, end-to-side (ES) and end-to-end (EE), to control specimens using a novel experimental setup that mimics anastomotic air leak tests, which are typically performed during surgeries. Freshly harvested porcine colonic sections from 23 F1 cross-species pigs were used. Pressure measurements and video imaging were used to monitor the ex vivo experiments on EE, ES, and Control specimens. Using EE (n = 16), ES (n = 12), and Control (n = 22) specimens, leak pressure was 282.6 ± 3.0 mm Hg for EE, 282.8 ± 2.6 mm Hg for ES, and 294.4 ± 12.1 for the Control. Time to leakage was 106.3 ± 28.1 s for EE, 263.9 ± 2127.0 s for ES, and 194.5 ± 90.2 s for the Control. We found that, while EE and ES have nearly identical leak pressures, ES was superior in terms of time to leakage and tissue expansion, which may explain why ES anastomoses have a lower clinical anastomotic leak rate. Two dependent variables representing stress and strain of colonic tissues were introduced. These variables showed ES was comparable to the Control. The experiments were simulated successfully using the finite element method (FEM). This research provides a reproducible ex vivo system with a corresponding FEM system to study the differences between anastomosis techniques and may help design anastomoses with lower leak rates and improve patient outcomes in colorectal surgeries. Full article
(This article belongs to the Special Issue Advanced Assessment of Medical Devices)
Show Figures

Figure 1

25 pages, 1678 KiB  
Review
Progress in Pseudotyping Lentiviral Vectors Towards Cell-Specific Gene Delivery In Vivo
by Ariana Arduini, Harshita Katiyar and Chen Liang
Viruses 2025, 17(6), 802; https://doi.org/10.3390/v17060802 - 31 May 2025
Viewed by 1515
Abstract
Lentiviral vectors (LVs) have become a fundamental tool in gene therapy due to their unique ability to transduce both dividing and non-dividing cells, transfer large genes of up to 10 kb, and facilitate stable, long-term expression of therapeutic genes into target cells. A [...] Read more.
Lentiviral vectors (LVs) have become a fundamental tool in gene therapy due to their unique ability to transduce both dividing and non-dividing cells, transfer large genes of up to 10 kb, and facilitate stable, long-term expression of therapeutic genes into target cells. A key application of LVs is the ex vivo genetic modification of patient-derived cells, such as the production of CAR-T cells by transducing isolated T cells with LVs to express the CAR gene, enabling them to target and destroy cancer cells once infused back into the patient. However, these ex vivo gene therapy drugs are often dismally unaffordable due to the complex procedures involved, including cell isolation, genetic modification, and expansion, along with the significant risks associated with immune conditioning to ensure successful engraftment. To overcome these barriers, direct in vivo transgene delivery to physiologically relevant cells has been explored, bypassing the need for ex vivo manipulations and reducing costs. Yet, a major challenge in this approach is engineering LV cell tropism to ensure the precise targeting of specific cells while avoiding off-target effects. Recent advances in modifying LV surface proteins have shown promise, including the successful in vivo generation of CAR T cells and ensuing clinical trials. This review is aimed at providing an up-to-date account of the progress in engineering LV tropism, covering the utility of different heterologous viral envelopes and their engineering to achieve cell-type-specific delivery and host immune evasion, and highlighting the potential of in vivo gene therapy to improve the affordability and accessibility of life-saving treatments. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Figure 1

19 pages, 3450 KiB  
Article
BET Protein Inhibition Relieves MDSC-Mediated Immune Suppression in Chronic Lymphocytic Leukemia
by Erin M. Drengler, Audrey L. Smith, Sydney A. Skupa, Elizabeth Schmitz, Eslam Mohamed and Dalia El-Gamal
Hemato 2025, 6(2), 14; https://doi.org/10.3390/hemato6020014 - 24 May 2025
Viewed by 983
Abstract
Background: Myeloid-derived suppressor cells (MDSCs) contribute to immune suppression observed in chronic lymphocytic leukemia (CLL). MDSCs are immature myeloid cells that are hijacked during development and further reprogrammed by the tumor microenvironment (TME) to harbor immune-suppressive properties and inhibit T-cell functions. Bromodomain [...] Read more.
Background: Myeloid-derived suppressor cells (MDSCs) contribute to immune suppression observed in chronic lymphocytic leukemia (CLL). MDSCs are immature myeloid cells that are hijacked during development and further reprogrammed by the tumor microenvironment (TME) to harbor immune-suppressive properties and inhibit T-cell functions. Bromodomain and extraterminal domain (BET) proteins, including BRD4, are epigenetic modulators that regulate genes implicated in CLL pathogenesis and TME interactions. Previously, we investigated how the novel BET inhibitor OPN-51107 (OPN5) prevents CLL disease expansion, modulates T-cell immune function, and alters gene expression related to MDSCs. In turn, we hypothesize that BET proteins such as BRD4 regulate MDSC functions, and subsequent pharmacological inhibition of BRD4 will alleviate MDSC-mediated immune suppression in CLL. Methods: Utilizing the Eµ-TCL1 mouse model of CLL, we evaluated BRD4 protein expression in MDSCs derived from the bone marrow of transgenic and age-matched wild-type (WT) mice. We then investigated the ex vivo functionality of OPN5-treated MDSCs, expanded from Eµ-TCL1 and WT bone marrow in MDSC-supportive medium. Finally, we conducted an in vivo study utilizing the Eµ-TCL1 adoptive transfer mouse model to determine the in vivo effects of OPN5 on MDSCs and other immune populations. Results: Through the course of this study, we found that MDSCs isolated from Eμ-TCL1 mice upregulate BRD4 expression and are more immune-suppressive than their WT counterparts. Furthermore, we demonstrated ex vivo OPN5 treatment reverses the immune-suppressive capacity of MDSCs isolated from leukemic mice, evident via enhanced T-cell proliferation and IFNγ production. Finally, we showed in vivo OPN5 treatment slows CLL disease progression and modulates immune cell populations, including MDSCs. Conclusions: Altogether, these data support BET inhibition as a useful therapeutic approach to reverse MDSC-mediated immune suppression in CLL. Full article
Show Figures

Figure 1

23 pages, 347 KiB  
Review
Advancements in Melanoma Treatment: A Review of PD-1 Inhibitors, T-VEC, mRNA Vaccines, and Tumor-Infiltrating Lymphocyte Therapy in an Evolving Landscape of Immunotherapy
by Apoorva Mehta, Mateen Motavaf, Ikenna Nebo, Sophia Luyten, Kofi D. Osei-Opare and Alejandro A. Gru
J. Clin. Med. 2025, 14(4), 1200; https://doi.org/10.3390/jcm14041200 - 12 Feb 2025
Cited by 4 | Viewed by 4081
Abstract
Melanoma, an aggressive skin cancer, presents significant therapeutic challenges. Consequently, innovative treatment strategies beyond conventional chemotherapy, radiation, and surgery are actively explored. This review discusses the evolution of immunotherapy in advanced melanoma, highlighting PD-1/PD-L1 inhibitors, mRNA vaccines, Talimogene Laherparepvec (T-VEC), and tumor-infiltrating lymphocyte [...] Read more.
Melanoma, an aggressive skin cancer, presents significant therapeutic challenges. Consequently, innovative treatment strategies beyond conventional chemotherapy, radiation, and surgery are actively explored. This review discusses the evolution of immunotherapy in advanced melanoma, highlighting PD-1/PD-L1 inhibitors, mRNA vaccines, Talimogene Laherparepvec (T-VEC), and tumor-infiltrating lymphocyte (TIL) therapies. PD-1/PD-L1 inhibitors such as pembrolizumab and nivolumab block immune checkpoints, promoting T-cell cytotoxic activity and improving overall survival in patients with advanced melanoma. T-VEC, a modified oncolytic herpes virus, promotes a systemic anti-tumor response while simultaneously lysing malignant cells. mRNA vaccines, such as Moderna’s mRNA-4157/V940, take advantage of malignant-cell-specific neoantigens to amplify the adaptive immune response while protecting healthy tissue. TIL therapy is a form of therapy involving ex vivo expansion and reinfusion of the patient’s tumor-specific lymphocytes and has been shown to provide durable tumor control. While these therapies have demonstrated promising clinical outcomes, challenges such as tumor resistance, high financial burden, and limited accessibility pose challenges to their widespread use. This review explores combination therapies such as PD-L1 inhibitors with mRNA vaccines, or TIL therapy, which aim to enhance treatment through synergistic approaches. Further research is required to optimize these combinations, address barriers preventing their use, and control adverse events. Full article
(This article belongs to the Section Dermatology)
19 pages, 4778 KiB  
Article
Development of a Competitive Nutrient-Based T-Cell Immunotherapy Designed to Block the Adaptive Warburg Effect in Acute Myeloid Leukemia
by Huynh Cao, Jeffrey Xiao, David J. Baylink, Vinh Nguyen, Nathan Shim, Jae Lee, Dave J. R. Mallari, Samiksha Wasnik, Saied Mirshahidi, Chien-Shing Chen, Hisham Abdel-Azim, Mark E. Reeves and Yi Xu
Biomedicines 2024, 12(10), 2250; https://doi.org/10.3390/biomedicines12102250 - 3 Oct 2024
Cited by 1 | Viewed by 2502
Abstract
Background: T-cell-based adoptive cell therapies have emerged at the forefront of cancer immunotherapies; however, failed long-term survival and inevitable exhaustion of transplanted T lymphocytes in vivo limits clinical efficacy. Leukemia blasts possess enhanced glycolysis (Warburg effect), exploiting their microenvironment to deprive nutrients (e.g., [...] Read more.
Background: T-cell-based adoptive cell therapies have emerged at the forefront of cancer immunotherapies; however, failed long-term survival and inevitable exhaustion of transplanted T lymphocytes in vivo limits clinical efficacy. Leukemia blasts possess enhanced glycolysis (Warburg effect), exploiting their microenvironment to deprive nutrients (e.g., glucose) from T cells, leading to T-cell dysfunction and leukemia progression. Methods: Thus, we explored whether genetic reprogramming of T-cell metabolism could improve their survival and empower T cells with a competitive glucose-uptake advantage against blasts and inhibit their uncontrolled proliferation. Results: Here, we discovered that high-glucose concentration reduced the T-cell expression of glucose transporter GLUT1 (SLC2A1) and TFAM (mitochondrion transcription factor A), an essential transcriptional regulator of mitochondrial biogenesis, leading to their impaired expansion ex vivo. To overcome the glucose-induced genetic deficiency in metabolism, we engineered T cells with lentiviral overexpression of SLC2A1 and/or TFAM transgene. Multi-omics analyses revealed that metabolic reprogramming promoted T-cell proliferation by increasing IL-2 release and reducing exhaustion. Moreover, the engineered T cells competitively deprived glucose from allogenic blasts and lessened leukemia burden in vitro. Conclusions: Our findings propose a novel T-cell immunotherapy that utilizes a dual strategy of starving blasts and cytotoxicity for preventing uncontrolled leukemia proliferation. Full article
(This article belongs to the Section Immunology and Immunotherapy)
Show Figures

Figure 1

24 pages, 1404 KiB  
Review
Pushing the Survival Bar Higher: Two Decades of Innovation in Lung Transplantation
by Khalil Aburahma, Nunzio Davide de Manna, Christian Kuehn, Jawad Salman, Mark Greer and Fabio Ius
J. Clin. Med. 2024, 13(18), 5516; https://doi.org/10.3390/jcm13185516 - 18 Sep 2024
Cited by 4 | Viewed by 3234
Abstract
Survival after lung transplantation has significantly improved during the last two decades. The refinement of the already existing extracorporeal life support (ECLS) systems, such as extracorporeal membrane oxygenation (ECMO), and the introduction of new techniques for donor lung optimization, such as ex vivo [...] Read more.
Survival after lung transplantation has significantly improved during the last two decades. The refinement of the already existing extracorporeal life support (ECLS) systems, such as extracorporeal membrane oxygenation (ECMO), and the introduction of new techniques for donor lung optimization, such as ex vivo lung perfusion (EVLP), have allowed the extension of transplant indication to patients with end-stage lung failure after acute respiratory distress syndrome (ARDS) and the expansion of the donor organ pool, due to the better evaluation and optimization of extended-criteria donor (ECD) lungs and of donors after circulatory death (DCD). The close monitoring of anti-HLA donor-specific antibodies (DSAs) has allowed the early recognition of pulmonary antibody-mediated rejection (AMR), which requires a completely different treatment and has a worse prognosis than acute cellular rejection (ACR). As such, the standardization of patient selection and post-transplant management has significantly contributed to this positive trend, especially at high-volume centers. This review focuses on lung transplantation after ARDS, on the role of EVLP in lung donor expansion, on ECMO as a principal cardiopulmonary support system in lung transplantation, and on the diagnosis and therapy of pulmonary AMR. Full article
(This article belongs to the Special Issue Clinical Outcomes of Cardiac Surgery)
Show Figures

Figure 1

17 pages, 4521 KiB  
Article
A Novel Recombinant Vitronectin Variant Supports the Expansion and Differentiation of Pluripotent Stem Cells in Defined Animal-Free Workflows
by Xi Lu, Eli Perr, Tahmina Naqvi, David Galitz, Marnelle Andersen, David Grabowski, Anthony Person, Alex Kalyuzhny and Kevin C. Flynn
Cells 2024, 13(18), 1566; https://doi.org/10.3390/cells13181566 - 17 Sep 2024
Cited by 1 | Viewed by 1965
Abstract
An essential aspect of harnessing the potential of pluripotent stem cells (PSCs) and their derivatives for regenerative medicine is the development of animal-free and chemically defined conditions for ex vivo cultivation. PSCs, including embryonic and induced PSCs (iPSCs), are in the early stages [...] Read more.
An essential aspect of harnessing the potential of pluripotent stem cells (PSCs) and their derivatives for regenerative medicine is the development of animal-free and chemically defined conditions for ex vivo cultivation. PSCs, including embryonic and induced PSCs (iPSCs), are in the early stages of clinical trials for various indications, including degenerative diseases and traumatic injury. A key step in the workflows generating these cells for more widespread clinical use is their safe and robust ex vivo cultivation. This entails optimization of cell culture media and substrates that are safe and consistent while maintaining robust functionality. Here, we describe the design of a human vitronectin (hVTN) variant with improved manufacturability in a bacterial expression system along with improved function in comparison to wild-type VTN and other previously characterized polypeptide fragments. In conjunction with an animal component-free media formulation, our hVTN fragment provides animal-free conditions for the enhanced expansion of iPSCs. This hVTN variant also supports the reprogramming of PBMCs into iPSCs. Furthermore, we show that these iPSCs can be efficiently differentiated into the three major germ layers and cortical neurons, thereby closing the loop on a completely defined animal-free workflow for cell types relevant for regenerative medicine. Full article
(This article belongs to the Special Issue Advances and Breakthroughs in Stem Cell Research)
Show Figures

Figure 1

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
Cited by 1 | Viewed by 2232
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

22 pages, 1700 KiB  
Review
Strategies for Improving CAR T Cell Persistence in Solid Tumors
by Megen C. Wittling, Anna C. Cole, Brianna Brammer, Kailey G. Diatikar, Nicole C. Schmitt and Chrystal M. Paulos
Cancers 2024, 16(16), 2858; https://doi.org/10.3390/cancers16162858 - 16 Aug 2024
Cited by 8 | Viewed by 5662
Abstract
CAR T cells require optimization to be effective in patients with solid tumors. There are many barriers affecting their ability to succeed. One barrier is persistence, as to achieve an optimal antitumor response, infused CAR T cells must engraft and persist. This singular [...] Read more.
CAR T cells require optimization to be effective in patients with solid tumors. There are many barriers affecting their ability to succeed. One barrier is persistence, as to achieve an optimal antitumor response, infused CAR T cells must engraft and persist. This singular variable is impacted by a multitude of factors—the CAR T cell design, lymphodepletion regimen used, expansion method to generate the T cell product, and more. Additionally, external agents can be utilized to augment CAR T cells, such as the addition of novel cytokines, pharmaceutical drugs that bolster memory formation, or other agents during either the ex vivo expansion process or after CAR T cell infusion to support them in the oppressive tumor microenvironment. This review highlights many strategies being used to optimize T cell persistence as well as future directions for improving the persistence of infused cells. Full article
(This article belongs to the Special Issue CAR T Therapy of Solid Cancers: Problems and Progress)
Show Figures

Figure 1

37 pages, 4373 KiB  
Review
The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair?
by Fereshteh Sadat Younesi and Boris Hinz
Int. J. Mol. Sci. 2024, 25(16), 8712; https://doi.org/10.3390/ijms25168712 - 9 Aug 2024
Cited by 2 | Viewed by 3522
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is [...] Read more.
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring—called fibrosis—that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies. Full article
Show Figures

Figure 1

12 pages, 2212 KiB  
Review
Cell Culture Models for Translational Research on Thymomas and Thymic Carcinomas: Current Status and Future Perspectives
by Denise Müller, Jürgen Loskutov, Stefan Küffer, Alexander Marx, Christian R. A. Regenbrecht, Philipp Ströbel and Manuela J. Regenbrecht
Cancers 2024, 16(15), 2762; https://doi.org/10.3390/cancers16152762 - 4 Aug 2024
Cited by 1 | Viewed by 2089
Abstract
Cell culture model systems are fundamental tools for studying cancer biology and identifying therapeutic vulnerabilities in a controlled environment. TET cells are notoriously difficult to culture, with only a few permanent cell lines available. The optimal conditions and requirements for the ex vivo [...] Read more.
Cell culture model systems are fundamental tools for studying cancer biology and identifying therapeutic vulnerabilities in a controlled environment. TET cells are notoriously difficult to culture, with only a few permanent cell lines available. The optimal conditions and requirements for the ex vivo establishment and permanent expansion of TET cells have not been systematically studied, and it is currently unknown whether different TET subtypes require different culture conditions or specific supplements. The few permanent cell lines available represent only type AB thymomas and thymic carcinomas, while attempts to propagate tumor cells derived from type B thymomas so far have been frustrated. It is conceivable that epithelial cells in type B thymomas are critically dependent on their interaction with immature T cells or their three-dimensional scaffold. Extensive studies leading to validated cell culture protocols would be highly desirable and a major advance in the field. Alternative methods such as tumor cell organoid models, patient-derived xenografts, or tissue slices have been sporadically used in TETs, but their specific contributions and advantages remain to be shown. Full article
Show Figures

Figure 1

10 pages, 6344 KiB  
Article
Distraction Enterogenesis in Rats: A Novel Approach for the Treatment of Short Bowel Syndrome
by Collyn O’Quin, Sean D. Clayton, Lexus Trosclair, Hannah Meyer, Nhi H. Dao, Andrew Minagar, Luke White, Valerie Welch, Giovanni Solitro, Jonathan Steven Alexander and Donald Sorrells
Pathophysiology 2024, 31(3), 388-397; https://doi.org/10.3390/pathophysiology31030029 - 30 Jul 2024
Cited by 2 | Viewed by 1402
Abstract
Background: Surgeons often encounter patients with intestinal failure due to inadequate intestinal length (“short bowel syndrome”/SBS). Treatment in these patients remains challenging and the process of physiologic adaptation may take years to complete, which frequently requires parenteral nutrition. We propose a proof-of-concept mechanical [...] Read more.
Background: Surgeons often encounter patients with intestinal failure due to inadequate intestinal length (“short bowel syndrome”/SBS). Treatment in these patients remains challenging and the process of physiologic adaptation may take years to complete, which frequently requires parenteral nutrition. We propose a proof-of-concept mechanical bowel elongation approach using a self-expanding prototype of an intestinal expansion sleeve (IES) for use in SBS to accelerate the adaptation process. Methods: IESs were deployed in the small intestines of Sprague Dawley rats. Mechanical characterization of these prototypes was performed. IES length–tension relationships and post-implant bowel expansion were measured ex vivo. Bowel histology before and after implantation was evaluated. Results: IES mechanical studies demonstrated decreasing expansive force with elongation. The deployment of IES devices produced an immediate 21 ± 8% increase in bowel length (p < 0.001, n = 11). Mechanical load testing data showed that the IESs expressed maximum expansive forces at 50% compression of the initial pre-contracted length. The small-intestine failure load in the rats was 1.88 ± 21 N. Intestinal histology post deployment of the IES showed significant expansive changes compared to unstretched bowel tissue. Conclusions: IES devices were scalable to the rat intestinal model in our study. The failure load of the rat small intestine was many times higher than the force exerted by the contraction of the IES. Histology demonstrated preservation of intestinal structure with some mucosal erosion. Future in vivo rat studies on distraction enterogenesis with this IES should help to define this organogenesis phenomenon. Full article
Show Figures

Figure 1

Back to TopTop