1
|
Crouigneau R, Li YF, Auxillos J, Goncalves-Alves E, Marie R, Sandelin A, Pedersen SF. Mimicking and analyzing the tumor microenvironment. CELL REPORTS METHODS 2024; 4:100866. [PMID: 39353424 DOI: 10.1016/j.crmeth.2024.100866] [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: 01/28/2024] [Revised: 07/22/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024]
Abstract
The tumor microenvironment (TME) is increasingly appreciated to play a decisive role in cancer development and response to therapy in all solid tumors. Hypoxia, acidosis, high interstitial pressure, nutrient-poor conditions, and high cellular heterogeneity of the TME arise from interactions between cancer cells and their environment. These properties, in turn, play key roles in the aggressiveness and therapy resistance of the disease, through complex reciprocal interactions between the cancer cell genotype and phenotype, and the physicochemical and cellular environment. Understanding this complexity requires the combination of sophisticated cancer models and high-resolution analysis tools. Models must allow both control and analysis of cellular and acellular TME properties, and analyses must be able to capture the complexity at high depth and spatial resolution. Here, we review the advantages and limitations of key models and methods in order to guide further TME research and outline future challenges.
Collapse
Affiliation(s)
- Roxane Crouigneau
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Yan-Fang Li
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Jamie Auxillos
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Eliana Goncalves-Alves
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Rodolphe Marie
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Albin Sandelin
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| | - Stine Falsig Pedersen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Taylor S, Mueller E, Jones LR, Makela AV, Ashammakhi N. Translational Aspects of 3D and 4D Printing and Bioprinting. Adv Healthc Mater 2024; 13:e2400463. [PMID: 38979857 DOI: 10.1002/adhm.202400463] [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: 02/06/2024] [Revised: 05/22/2024] [Indexed: 07/10/2024]
Abstract
Three-dimensional (3D) printed medical devices include orthopedic and craniofacial implants, surgical tools, and external prosthetics that have been directly used in patients. While the advances of additive manufacturing techniques in the production of medical devices have been on the rise, clinical translation of living cellular constructs face significant limitations in terms of regulatory affairs, process technology, and materials development. In this perspective, the current status-quo of 3D and four-dimensional (4D) (bio)printing is summarized, current advancements are discussed and the challenges that need to be addressed for improved industrial translation and clinical applications of bioprinting are highlighted. It is focused on a multidisciplinary approach in discussing the key translational considerations, from the perspective of industry, regulatory bodies, funding strategies, and future directions.
Collapse
Affiliation(s)
| | - Eva Mueller
- Ricoh 3D for Healthcare, Ricoh USA, Winston-Salem, NC 27101, USA
| | - Lamont R Jones
- Department of Otolaryngology, Henry Ford Heath, Detroit, MI 48322, USA
| | - Ashley V Makela
- Institute for Quantitative Health Science & Engineering and Department of Engineering, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science & Engineering and Department of Engineering, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
3
|
Cordts SC, Yuki K, Henao Echeverri MF, Narasimhan B, Kuo CJ, Tang SKY. Microdissection tools to generate organoids for modeling the tumor immune microenvironment. MICROSYSTEMS & NANOENGINEERING 2024; 10:126. [PMID: 39251611 PMCID: PMC11385579 DOI: 10.1038/s41378-024-00756-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 09/11/2024]
Abstract
Patient-derived tumor organoids have emerged as promising models for predicting personalized drug responses in cancer therapy, but they typically lack immune components. Preserving the in vivo association between tumor cells and endogenous immune cells is critical for accurate testing of cancer immunotherapies. Mechanical dissection of tumor specimens into tumor fragments, as opposed to enzymatic digestion into single cells, is essential for maintaining these native tumor-immune cell spatial relationships. However, conventional mechanical dissection relying on manual mincing is time-consuming and irreproducible. This study describes two microdissection devices, the µDicer and µGrater, to facilitate the generation of intact tumor fragments from mouse B16 melanoma, a common model of human melanoma. The µDicer- and µGrater-cut tumor fragments were used to generate air‒liquid interface (ALI) organoids that copreserve tumor cells with infiltrating immune subsets without artificial reconstitution. The µDicer, consisting of a hexagonal array of silicon microblades, was employed to investigate the effect of organoid size. The viability of ALI organoid immune cells appeared insensitive to organoid sizes exceeding ~400 µm but diminished in organoids ~200 µm in size. The µGrater, consisting of an array of submillimeter holes in stainless steel, was employed to accelerate dissection. For the samples studied, the µGrater was 4.5 times faster than manual mincing. Compared with those generated by manual mincing, ALI organoids generated by the µGrater demonstrated similar viability, immune cell composition, and responses to anti-PD-1 immunotherapy. With further optimization, the µGrater holds potential for integration into clinical workflows to support the advancement of personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Seth C Cordts
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Kanako Yuki
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
| | | | | | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
| | - Sindy K Y Tang
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
4
|
Liu Z, Fan Y, Cui M, Wang X, Zhao P. Investigation of tumour environments through advancements in microtechnology and nanotechnology. Biomed Pharmacother 2024; 178:117230. [PMID: 39116787 DOI: 10.1016/j.biopha.2024.117230] [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: 06/05/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer has a significant negative social and economic impact on both developed and developing countries. As a result, understanding the onset and progression of cancer is critical for developing therapies that can improve the well-being and health of individuals with cancer. With time, study has revealed, the tumor microenvironment has great influence on this process. Micro and nanoscale engineering techniques can be used to study the tumor microenvironment. Nanoscale and Microscale engineering use Novel technologies and designs with small dimensions to recreate the TME. Knowing how cancer cells interact with one another can help researchers develop therapeutic approaches that anticipate and counteract cancer cells' techniques for evading detection and fighting anti-cancer treatments, such as microfabrication techniques, microfluidic devices, nanosensors, and nanodevices used to study or recreate the tumor microenvironment. Nevertheless, a complicated action just like the growth and in cancer advancement, and their intensive association along the environment around it that has to be studied in more detail.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Radiology, Shengjing Hospital of China Medical University, China
| | - Yan Fan
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mengyao Cui
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xu Wang
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Pengfei Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
5
|
Kazempour H, Teymouri F, Khatami M, Hosseini SN. Computational modelling of the therapeutic outputs of photodynamic therapy on spheroid-on-chip models. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 258:112960. [PMID: 38991293 DOI: 10.1016/j.jphotobiol.2024.112960] [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: 03/02/2024] [Revised: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024]
Abstract
Photodynamic therapy (PDT) is a medical radio chemotherapeutic method that uses light, photosensitizing agents, and oxygen to produce cytotoxic compounds, which eliminate malignant cells. Recently, Microfluidic systems have been used to analyse photosensitizers (PSs) due to their potential to replicate in vivo environments. While prior studies have established a strong correlation between reacted singlet oxygen concentration and PDT-induced cellular death, the effects that the ambient fluid flow might have on the concentration of oxygen and PS have been disregarded in many, which limits the reliability of the results. Herein, we coupled the transport of oxygen and PS throughout the ambient medium and within the spheroidal multicellular aggregate to initially study the profiles of oxygen and PS concentration alongside PDT-induced cellular death throughout the spheroid before and after radiation. The attained results indicate that the PDT-induced cellular death initiates on the surface of the spheroids and subsequently spreads to the neighbouring regions, which is in great accordance with experimental results. Afterward, the effects that drug-light interval (DLI), fluence rate, PS composition, microchannel height, and inlet flow rate have on the therapeutic outcomes are studied. The findings show that adequate DLI is critical to ensure uniform distribution of PS throughout the medium, and a value of 5 h was found to be sufficient. The composition of PS is critical, as ALA-PpIX induces earlier cell death but accelerates oxygen consumption, especially in the outer layers, depriving the inner layers of oxygen necessary for PDT, which in turn disrupts and prolongs the exposure time compared to mTHPC and Photofrin. Despite the fluence rate directly influencing the singlet oxygen generation rate, increasing the fluence rate by 189 mW/cm2 would not significantly benefit us. Microwell height and inlet flow rate involve competing phenomena-increasing height or decreasing flow reduces oxygen supply and increases PS "washout" and its concentration.
Collapse
Affiliation(s)
- Hossein Kazempour
- Chemical and Petroleum Engineering Department, Sharif University of Technology, Tehran, Iran
| | - Fatemeh Teymouri
- Chemical and Petroleum Engineering Department, Sharif University of Technology, Tehran, Iran
| | - Maryam Khatami
- Research and Production Complex, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
6
|
Brasino DSK, Speese SD, Schilling K, Schutt CE, Barton MC. A Linkable, Polycarbonate Gut Microbiome-Distal Tumor Chip Platform for Interrogating Cancer Promoting Mechanisms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309220. [PMID: 39023197 PMCID: PMC11425222 DOI: 10.1002/advs.202309220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/31/2024] [Indexed: 07/20/2024]
Abstract
Gut microbiome composition is tied to diseases ranging from arthritis to cancer to depression. However, mechanisms of action are poorly understood, limiting development of relevant therapeutics. Organ-on-chip platforms, which model minimal functional units of tissues and can tightly control communication between them, are ideal platforms to study these relationships. Many gut microbiome models are published to date but devices are typically fabricated using oxygen permeable polydimethylsiloxane, requiring interventions to support anaerobic bacteria. To address this challenge, a platform is developed where the chips are fabricated entirely from gas-impermeable polycarbonate without tapes or gaskets. These chips replicate polarized villus-like structures of the native tissue. Further, they enable co-cultures of commensal anaerobic bacteria Blautia coccoides on the surface of gut epithelia for two days within a standard incubator. Another complication of commonly used materials in organ-on-chip devices is high ad-/absorption, limiting applications in high-resolution microscopy and biomolecule interaction studies. For future communication studies between gut microbiota and distal tumors, an additional polycarbonate chip design is developed to support hydrogel-embedded tissue culture. These chips enable high-resolution microscopy with all relevant processing done on-chip. Designed for facile linking, this platform will make a variety of mechanistic studies possible.
Collapse
Affiliation(s)
- Danielle S K Brasino
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Sean D Speese
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Kevin Schilling
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Carolyn E Schutt
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Michelle C Barton
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| |
Collapse
|
7
|
Horowitz LF, Rodriguez-Mias R, Chan M, Zhu S, Gottshall NR, Stepanov I, Stiles C, Yeung M, Nguyen TN, Lockhart EJ, Yeung RS, Villen J, Gujral TS, Folch A. Microdissected tumor cuboids: a microscale cancer model for large-scale testing that retains a complex tumor microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586189. [PMID: 38585935 PMCID: PMC10996559 DOI: 10.1101/2024.03.22.586189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Current cancer disease models fail to faithfully recapitulate key features of the human tumor microenvironment (TME), such as immune and vascular cells, while simultaneously enabling high-throughput drug tests. We have recently developed a precision slicing method that optimizes the yield of large numbers of cuboidal microtissues ("cuboids", ∼(400 µm) 3 ) from a single tumor biopsy. Here we demonstrate that cuboids from syngeneic mouse tumor models and human tumors retain a complex TME, making them amenable for drug and immunotherapy evaluation. We characterize relevant TME parameters, such as cellular architecture, cytokine secretion, proteomics profiles, and response to drug panels in multi-well arrays. Despite the cutting procedure and the time spent in culture (up to 7 days), the cuboids display strong cytokine and drug responses, including to immunotherapy. Overall, our results suggest that cuboids could provide invaluable therapeutic information for personalized oncology applications, and could help the development of TME-dependent therapeutics and cancer disease models, including for clinical trials.
Collapse
|
8
|
Stepanov I, Gottshall NR, Ahmadianyazdi A, Sinha D, Lockhart EJ, Nguyen TNH, Hassan S, Horowitz LF, Yeung RS, Gujral TS, Folch A. Low-Cost Robotic Manipulation of Live Microtissues for Cancer Drug Testing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586169. [PMID: 38586030 PMCID: PMC10996467 DOI: 10.1101/2024.03.21.586169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The scarcity of human biopsies available for drug testing is a paramount challenge for developing new therapeutics, disease models, and personalized treatments. Microtechnologies that combine the microscale manipulation of tissues and fluids offer the exciting possibility of miniaturizing both disease models and drug testing workflows on scarce human biopsies. Unfortunately, these technologies presently require microfluidic devices or robotic dispensers that are not widely accessible. We have rapidly-prototyped an inexpensive platform based on an off-the-shelf robot that can microfluidically manipulate live microtissues into/out of culture plates without using complicated accessories such as microscopes or pneumatic controllers. The robot integrates complex functions with a simple, cost-effective and compact construction, allowing placement inside a tissue culture hood for sterile workflows. We demonstrated a proof-of-concept cancer drug evaluation workflow of potential clinical utility using patient tumor biopsies with multiple drugs on 384-well plates. Our user-friendly, low-cost platform promises to make drug testing of microtissues broadly accessible to pharmaceutical, clinical, and biological laboratories. Teaser A low-cost robot for handling microtissues and catalyzing their use in cancer drug evaluation and personalized oncology.
Collapse
|
9
|
Mes-Masson AM. The journey from bench to bedside-it takes a science village. Biochem Cell Biol 2024; 102:299-304. [PMID: 38640502 DOI: 10.1139/bcb-2024-0075] [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] [Indexed: 04/21/2024] Open
Abstract
I was fortunate enough to start my career at what was the dawn of modern-day molecular biology and to apply it to an important health problem. While my early work focused on fundamental science, the desire to understand human disease better and to find practical applications for research discoveries resulted, over the following decades, in creating a stream of translational research directed specifically toward epithelial cancers. This could only have been possible through multiple collaborations. This type of team science would eventually become a hallmark of my career. With the development of higher throughput molecular techniques, the pace of research and discovery has quickened, and the concept of personalized medicine based on genomics is now coming to fruition. I hope my legacy will not just reflect my published works, but will also include the impact I have had on the development of the next generation of scientists and clinician scientists who inspired me with their dedication, knowledge, and enthusiasm.
Collapse
Affiliation(s)
- Anne-Marie Mes-Masson
- Institut du cancer de Montréal and Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
10
|
Lopez-Vince E, Wilhelm C, Simon-Yarza T. Vascularized tumor models for the evaluation of drug delivery systems: a paradigm shift. Drug Deliv Transl Res 2024; 14:2216-2241. [PMID: 38619704 PMCID: PMC11208221 DOI: 10.1007/s13346-024-01580-3] [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] [Accepted: 03/13/2024] [Indexed: 04/16/2024]
Abstract
As the conversion rate of preclinical studies for cancer treatment is low, user-friendly models that mimic the pathological microenvironment and drug intake with high throughput are scarce. Animal models are key, but an alternative to reduce their use would be valuable. Vascularized tumor-on-chip models combine great versatility with scalable throughput and are easy to use. Several strategies to integrate both tumor and vascular compartments have been developed, but few have been used to assess drug delivery. Permeability, intra/extravasation, and free drug circulation are often evaluated, but imperfectly recapitulate the processes at stake. Indeed, tumor targeting and chemoresistance bypass must be investigated to design promising cancer therapeutics. In vitro models that would help the development of drug delivery systems (DDS) are thus needed. They would allow selecting good candidates before animal studies based on rational criteria such as drug accumulation, diffusion in the tumor, and potency, as well as absence of side damage. In this review, we focus on vascularized tumor models. First, we detail their fabrication, and especially the materials, cell types, and coculture used. Then, the different strategies of vascularization are described along with their classical applications in intra/extravasation or free drug assessment. Finally, current trends in DDS for cancer are discussed with an overview of the current efforts in the domain.
Collapse
Affiliation(s)
- Elliot Lopez-Vince
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005, Paris, France
- Université Paris Cité, Université Sorbonne Paris Nord, LVTS Inserm U1148, 75018, Paris, France
| | - Claire Wilhelm
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005, Paris, France
| | - Teresa Simon-Yarza
- Université Paris Cité, Université Sorbonne Paris Nord, LVTS Inserm U1148, 75018, Paris, France.
| |
Collapse
|
11
|
Mehta V, Vilikkathala Sudhakaran S, Nellore V, Madduri S, Rath SN. 3D stem-like spheroids-on-a-chip for personalized combinatorial drug testing in oral cancer. J Nanobiotechnology 2024; 22:344. [PMID: 38890730 PMCID: PMC11186147 DOI: 10.1186/s12951-024-02625-y] [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: 04/15/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Functional drug testing (FDT) with patient-derived tumor cells in microfluidic devices is gaining popularity. However, the majority of previously reported microfluidic devices for FDT were limited by at least one of these factors: lengthy fabrication procedures, absence of tumor progenitor cells, lack of clinical correlation, and mono-drug therapy testing. Furthermore, personalized microfluidic models based on spheroids derived from oral cancer patients remain to be thoroughly validated. Overcoming the limitations, we develop 3D printed mold-based, dynamic, and personalized oral stem-like spheroids-on-a-chip, featuring unique serpentine loops and flat-bottom microwells arrangement. RESULTS This unique arrangement enables the screening of seven combinations of three drugs on chemoresistive cancer stem-like cells. Oral cancer patients-derived stem-like spheroids (CD 44+) remains highly viable (> 90%) for 5 days. Treatment with a well-known oral cancer chemotherapy regimen (paclitaxel, 5 fluorouracil, and cisplatin) at clinically relevant dosages results in heterogeneous drug responses in spheroids. These spheroids are derived from three oral cancer patients, each diagnosed with either well-differentiated or moderately-differentiated squamous cell carcinoma. Oral spheroids exhibit dissimilar morphology, size, and oral tumor-relevant oxygen levels (< 5% O2). These features correlate with the drug responses and clinical diagnosis from each patient's histopathological report. CONCLUSIONS Overall, we demonstrate the influence of tumor differentiation status on treatment responses, which has been rarely carried out in the previous reports. To the best of our knowledge, this is the first report demonstrating extensive work on development of microfluidic based oral cancer spheroid model for personalized combinatorial drug screening. Furthermore, the obtained clinical correlation of drug screening data represents a significant advancement over previously reported personalized spheroid-based microfluidic devices. Finally, the maintenance of patient-derived spheroids with high viability under oral cancer relevant oxygen levels of less than 5% O2 is a more realistic representation of solid tumor microenvironment in our developed device.
Collapse
Affiliation(s)
- Viraj Mehta
- Regenerative Medicine and Stem Cell Laboratory (RMS), Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Kandi, 502285, Telangana, India
| | - Sukanya Vilikkathala Sudhakaran
- Regenerative Medicine and Stem Cell Laboratory (RMS), Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Kandi, 502285, Telangana, India
| | - Vijaykumar Nellore
- Regenerative Medicine and Stem Cell Laboratory (RMS), Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Kandi, 502285, Telangana, India
| | - Srinivas Madduri
- Department of Surgery, University of Geneva, 1205, Geneva, Switzerland
| | - Subha Narayan Rath
- Regenerative Medicine and Stem Cell Laboratory (RMS), Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Kandi, 502285, Telangana, India.
| |
Collapse
|
12
|
Lockhart EJ, Horowitz LF, Rodríguez A, Zhu S, Nguyen T, Mehrabi M, Gujral TS, Folch A. Drug testing of monodisperse arrays of live microdissected tumors using a valved multiwell microfluidic platform. LAB ON A CHIP 2024; 24:2683-2699. [PMID: 38651213 DOI: 10.1039/d4lc00016a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Cancer drug testing in animals is an extremely poor predictor of the drug's safety and efficacy observed in humans. Hence there is a pressing need for functional testing platforms that better predict traditional and immunotherapy responses in human, live tumor tissue or tissue constructs, and at the same time are compatible with the use of mouse tumor tissue to facilitate building more accurate disease models. Since many cancer drug actions rely on mechanisms that depend on the tumor microenvironment (TME), such platforms should also retain as much of the native TME as possible. Additionally, platforms based on miniaturization technologies are desirable to reduce animal use and sensitivity to human tissue scarcity. Present high-throughput testing platforms that have some of these features, e.g. based on patient-derived tumor organoids, require a growth step that alters the TME. On the other hand, microdissected tumors (μDTs) or "spheroids" that retain an intact TME have shown promising responses to immunomodulators acting on native immune cells. However, difficult tissue handling after microdissection has reduced the throughput of drug testing on μDTs, thereby constraining the inherent advantages of producing numerous TME-preserving units of tissue for drug testing. Here we demonstrate a microfluidic 96-well platform designed for drug treatment of hundreds of similarly-sized, cuboidal μDTs ("cuboids") produced from a single tumor sample. The platform organizes a monodisperse array of four cuboids per well in 384 hydrodynamic traps. The microfluidic device, entirely fabricated in thermoplastics, features 96 microvalves that fluidically isolate each well after the cuboid loading step for straightforward multi-drug testing. Since our platform makes the most of scarce tumor tissue, it can potentially be applied to human biopsies that preserve the human TME while minimizing animal testing.
Collapse
Affiliation(s)
- Ethan J Lockhart
- Department of Bioengineering, University of Washington, Seattle, USA.
| | - Lisa F Horowitz
- Department of Bioengineering, University of Washington, Seattle, USA.
| | - Adán Rodríguez
- Department of Bioengineering, University of Washington, Seattle, USA.
| | - Songli Zhu
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Tran Nguyen
- Department of Bioengineering, University of Washington, Seattle, USA.
| | | | - Taranjit S Gujral
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Albert Folch
- Department of Bioengineering, University of Washington, Seattle, USA.
| |
Collapse
|
13
|
Brennen WN, Le Magnen C, Karkampouna S, Anselmino N, Bock N, Choo N, Clark AK, Coleman IM, Dolgos R, Ferguson AM, Goode DL, Krutihof-de Julio M, Navone NM, Nelson PS, O'Neill E, Porter LH, Ranasinghe W, Sunada T, Williams ED, Butler LM, Corey E, van Weerden WM, Taylor RA, Risbridger GP, Lawrence MG. Defining the challenges and opportunities for using patient-derived models in prostate cancer research. Prostate 2024; 84:623-635. [PMID: 38450798 PMCID: PMC11014775 DOI: 10.1002/pros.24682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/29/2024] [Accepted: 02/15/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND There are relatively few widely used models of prostate cancer compared to other common malignancies. This impedes translational prostate cancer research because the range of models does not reflect the diversity of disease seen in clinical practice. In response to this challenge, research laboratories around the world have been developing new patient-derived models of prostate cancer, including xenografts, organoids, and tumor explants. METHODS In May 2023, we held a workshop at the Monash University Prato Campus for researchers with expertise in establishing and using a variety of patient-derived models of prostate cancer. This review summarizes our collective ideas on how patient-derived models are currently being used, the common challenges, and future opportunities for maximizing their usefulness in prostate cancer research. RESULTS An increasing number of patient-derived models for prostate cancer are being developed. Despite their individual limitations and varying success rates, these models are valuable resources for exploring new concepts in prostate cancer biology and for preclinical testing of potential treatments. Here we focus on the need for larger collections of models that represent the changing treatment landscape of prostate cancer, robust readouts for preclinical testing, improved in vitro culture conditions, and integration of the tumor microenvironment. Additional priorities include ensuring model reproducibility, standardization, and replication, and streamlining the exchange of models and data sets among research groups. CONCLUSIONS There are several opportunities to maximize the impact of patient-derived models on prostate cancer research. We must develop large, diverse and accessible cohorts of models and more sophisticated methods for emulating the intricacy of patient tumors. In this way, we can use the samples that are generously donated by patients to advance the outcomes of patients in the future.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Pharmacology & Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Clémentine Le Magnen
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Urology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sofia Karkampouna
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Nicolas Anselmino
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nathalie Bock
- School of Biomedical Sciences at Translational Research Institute, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
| | - Nicholas Choo
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
| | - Ashlee K Clark
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
| | - Ilsa M Coleman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Robin Dolgos
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Urology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alison M Ferguson
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Katharina Gaus Light Microscopy Facility, Mark Wainwright Analytical Centre, Division of Research and Enterprise, University of New South Wales, Sydney, NSW, Australia
| | - David L Goode
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Marianna Krutihof-de Julio
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, Translational Organoid Resource, University of Bern, Bern, Switzerland
| | - Nora M Navone
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Edward O'Neill
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Laura H Porter
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
| | - Weranja Ranasinghe
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
- Department of Surgery, Monash University, Melbourne, VIC, Australia
- Department of Urology, Monash Health, Melbourne, VIC, Australia
- Department of Urology, Austin Health, Melbourne, VIC, Australia
| | - Takuro Sunada
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Elizabeth D Williams
- School of Biomedical Sciences at Translational Research Institute, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Australian Prostate Cancer Research Centre-Queensland, Brisbane, QLD, Australia
- Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Lisa M Butler
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, SA, Australia
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | | | - Renea A Taylor
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Physiology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
- Cabrini Institute, Cabrini Health, Malvern, VIC, Australia
- Melbourne Urological Research Alliance, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Gail P Risbridger
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Cabrini Institute, Cabrini Health, Malvern, VIC, Australia
- Melbourne Urological Research Alliance, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Mitchell G Lawrence
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Cabrini Institute, Cabrini Health, Malvern, VIC, Australia
- Melbourne Urological Research Alliance, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Ahmad Zawawi SS, Salleh EA, Musa M. Spheroids and organoids derived from colorectal cancer as tools for in vitro drug screening. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:409-431. [PMID: 38745769 PMCID: PMC11090692 DOI: 10.37349/etat.2024.00226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/02/2024] [Indexed: 05/16/2024] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease. Conventional two-dimensional (2D) culture employing cell lines was developed to study the molecular properties of CRC in vitro. Although these cell lines which are isolated from the tumor niche in which cancer develop, the translation to human model such as studying drug response is often hindered by the inability of cell lines to recapture original tumor features and the lack of heterogeneous clinical tumors represented by this 2D model, differed from in vivo condition. These limitations which may be overcome by utilizing three-dimensional (3D) culture consisting of spheroids and organoids. Over the past decade, great advancements have been made in optimizing culture method to establish spheroids and organoids of solid tumors including of CRC for multiple purposes including drug screening and establishing personalized medicine. These structures have been proven to be versatile and robust models to study CRC progression and deciphering its heterogeneity. This review will describe on advances in 3D culture technology and the application as well as the challenges of CRC-derived spheroids and organoids as a mode to screen for anticancer drugs.
Collapse
Affiliation(s)
| | - Elyn Amiela Salleh
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Marahaini Musa
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| |
Collapse
|
15
|
Chermat R, Refet-Mollof E, Kamio Y, Carrier JF, Wong P, Gervais T. Brachytherapy on-a-chip: a clinically-relevant approach for radiotherapy testing in 3d biology. LAB ON A CHIP 2024; 24:2335-2346. [PMID: 38568477 DOI: 10.1039/d4lc00032c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
We describe the first microfluidic device for in vitro testing of brachytherapy (BT), with applications in translational cancer research. Our PDMS-made BT-on-chip system allows highly precise manual insertion of clinical BT seeds, reliable dose calculation using standard clinically-used TG-43 formalism and easy culture of naturally hypoxic spheroids in less than 3 days, thereby increasing the translational potential of the device. As the BT-on-chip platform is designed to be versatile, we showcase three different gold-standard post-irradiation bioassays and recapitulate, for the first time on-chip, key clinical observations such as dose rate effect and hypoxia-induced radioresistance. Our results suggest that BT-on-chip can be used to safely and efficiently integrate BT and radiotherapy to translational research and drug development pipelines, without expensive equipment or complex workflows.
Collapse
Affiliation(s)
- Rodin Chermat
- μFO Lab, Institute of Biomedical Engineering, Polytechnique Montréal, Montréal, Canada.
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Institut du Cancer de Montréal (ICM), Montréal, Canada
| | - Elena Refet-Mollof
- μFO Lab, Institute of Biomedical Engineering, Polytechnique Montréal, Montréal, Canada.
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Institut du Cancer de Montréal (ICM), Montréal, Canada
| | - Yuji Kamio
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Département de radio-oncologie, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Canada
| | - Jean-François Carrier
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Institut du Cancer de Montréal (ICM), Montréal, Canada
- Département de radio-oncologie, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Canada
- Département de Physique, Université de Montréal, Canada
| | - Philip Wong
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Thomas Gervais
- μFO Lab, Institute of Biomedical Engineering, Polytechnique Montréal, Montréal, Canada.
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Canada
- Institut du Cancer de Montréal (ICM), Montréal, Canada
| |
Collapse
|
16
|
Bitounis D, Jacquinet E, Rogers MA, Amiji MM. Strategies to reduce the risks of mRNA drug and vaccine toxicity. Nat Rev Drug Discov 2024; 23:281-300. [PMID: 38263456 DOI: 10.1038/s41573-023-00859-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/25/2024]
Abstract
mRNA formulated with lipid nanoparticles is a transformative technology that has enabled the rapid development and administration of billions of coronavirus disease 2019 (COVID-19) vaccine doses worldwide. However, avoiding unacceptable toxicity with mRNA drugs and vaccines presents challenges. Lipid nanoparticle structural components, production methods, route of administration and proteins produced from complexed mRNAs all present toxicity concerns. Here, we discuss these concerns, specifically how cell tropism and tissue distribution of mRNA and lipid nanoparticles can lead to toxicity, and their possible reactogenicity. We focus on adverse events from mRNA applications for protein replacement and gene editing therapies as well as vaccines, tracing common biochemical and cellular pathways. The potential and limitations of existing models and tools used to screen for on-target efficacy and de-risk off-target toxicity, including in vivo and next-generation in vitro models, are also discussed.
Collapse
Affiliation(s)
- Dimitrios Bitounis
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
- Moderna, Inc., Cambridge, MA, USA
| | | | | | - Mansoor M Amiji
- Departments of Pharmaceutical Sciences and Chemical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
17
|
Chuaychob S, Lyu R, Tanaka M, Haginiwa A, Kitada A, Nakamura T, Yokokawa R. Mimicking angiogenic microenvironment of alveolar soft-part sarcoma in a microfluidic coculture vasculature chip. Proc Natl Acad Sci U S A 2024; 121:e2312472121. [PMID: 38502703 PMCID: PMC10990104 DOI: 10.1073/pnas.2312472121] [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: 07/27/2023] [Accepted: 01/24/2024] [Indexed: 03/21/2024] Open
Abstract
Alveolar soft-part sarcoma (ASPS) is a slow-growing soft tissue sarcoma with high mortality rates that affects adolescents and young adults. ASPS resists conventional chemotherapy; thus, decades of research have elucidated pathogenic mechanisms driving the disease, particularly its angiogenic capacities. Integrated blood vessels that are rich in pericytes (PCs) and metastatic potential are distinctive of ASPS. To mimic ASPS angiogenic microenvironment, a microfluidic coculture vasculature chip has been developed as a three-dimensional (3D) spheroid composed of mouse ASPS, a layer of PCs, and endothelial cells (ECs). This ASPS-on-a-chip provided functional and morphological similarity as the in vivo mouse model to elucidate the cellular crosstalk within the tumor vasculature before metastasis. We successfully reproduce ASPS spheroid and leaky vessels representing the unique tumor vasculature to assess effective drug delivery into the core of a solid tumor. Furthermore, this ASPS angiogenesis model enabled us to investigate the role of proteins in the intracellular trafficking of bioactive signals from ASPS to PCs and ECs during angiogenesis, including Rab27a and Sytl2. The results can help to develop drugs targeting the crosstalk between ASPS and the adjacent cells in the tumoral microenvironment.
Collapse
Affiliation(s)
- Surachada Chuaychob
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto615-8540, Japan
| | - Ruyin Lyu
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto615-8540, Japan
| | - Miwa Tanaka
- Project for Cancer Epigenomics, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo135-8550, Japan
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo160-8402, Japan
| | - Ayumi Haginiwa
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto615-8540, Japan
| | - Atsuya Kitada
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto615-8540, Japan
| | - Takuro Nakamura
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo160-8402, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto615-8540, Japan
| |
Collapse
|
18
|
Wu Z, Huang D, Wang J, Zhao Y, Sun W, Shen X. Engineering Heterogeneous Tumor Models for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304160. [PMID: 37946674 PMCID: PMC10767453 DOI: 10.1002/advs.202304160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/16/2023] [Indexed: 11/12/2023]
Abstract
Tumor tissue engineering holds great promise for replicating the physiological and behavioral characteristics of tumors in vitro. Advances in this field have led to new opportunities for studying the tumor microenvironment and exploring potential anti-cancer therapeutics. However, the main obstacle to the widespread adoption of tumor models is the poor understanding and insufficient reconstruction of tumor heterogeneity. In this review, the current progress of engineering heterogeneous tumor models is discussed. First, the major components of tumor heterogeneity are summarized, which encompasses various signaling pathways, cell proliferations, and spatial configurations. Then, contemporary approaches are elucidated in tumor engineering that are guided by fundamental principles of tumor biology, and the potential of a bottom-up approach in tumor engineering is highlighted. Additionally, the characterization approaches and biomedical applications of tumor models are discussed, emphasizing the significant role of engineered tumor models in scientific research and clinical trials. Lastly, the challenges of heterogeneous tumor models in promoting oncology research and tumor therapy are described and key directions for future research are provided.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Danqing Huang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Jinglin Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| | - Weijian Sun
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Xian Shen
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| |
Collapse
|
19
|
Bouquerel C, Dubrova A, Hofer I, Phan DTT, Bernheim M, Ladaigue S, Cavaniol C, Maddalo D, Cabel L, Mechta-Grigoriou F, Wilhelm C, Zalcman G, Parrini MC, Descroix S. Bridging the gap between tumor-on-chip and clinics: a systematic review of 15 years of studies. LAB ON A CHIP 2023; 23:3906-3935. [PMID: 37592893 DOI: 10.1039/d3lc00531c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Over the past 15 years, the field of oncology research has witnessed significant progress in the development of new cell culture models, such as tumor-on-chip (ToC) systems. In this comprehensive overview, we present a multidisciplinary perspective by bringing together physicists, biologists, clinicians, and experts from pharmaceutical companies to highlight the current state of ToC research, its unique features, and the challenges it faces. To offer readers a clear and quantitative understanding of the ToC field, we conducted an extensive systematic analysis of more than 300 publications related to ToC from 2005 to 2022. ToC offer key advantages over other in vitro models by enabling precise control over various parameters. These parameters include the properties of the extracellular matrix, mechanical forces exerted on cells, the physico-chemical environment, cell composition, and the architecture of the tumor microenvironment. Such fine control allows ToC to closely replicate the complex microenvironment and interactions within tumors, facilitating the study of cancer progression and therapeutic responses in a highly representative manner. Importantly, by incorporating patient-derived cells or tumor xenografts, ToC models have demonstrated promising results in terms of clinical validation. We also examined the potential of ToC for pharmaceutical industries in which ToC adoption is expected to occur gradually. Looking ahead, given the high failure rate of clinical trials and the increasing emphasis on the 3Rs principles (replacement, reduction, refinement of animal experimentation), ToC models hold immense potential for cancer research. In the next decade, data generated from ToC models could potentially be employed for discovering new therapeutic targets, contributing to regulatory purposes, refining preclinical drug testing and reducing reliance on animal models.
Collapse
Affiliation(s)
- Charlotte Bouquerel
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
- Fluigent, 67 avenue de Fontainebleau, 94270, Le Kremlin-Bicêtre, France
| | - Anastasiia Dubrova
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Isabella Hofer
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Duc T T Phan
- Biomedicine Design, Pfizer Inc., San Diego, CA, USA
| | - Moencopi Bernheim
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Ségolène Ladaigue
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Charles Cavaniol
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Danilo Maddalo
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Luc Cabel
- Institut Curie, Department of Medical Oncology, 26 rue d'Ulm, 75005, Paris, France
| | - Fatima Mechta-Grigoriou
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Claire Wilhelm
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Gérard Zalcman
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
- Université Paris Cité, Thoracic Oncology Department, INSERM CIC1425, Bichat Hospital, Cancer Institute AP-HP. Nord, Paris, France.
| | - Maria Carla Parrini
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Stéphanie Descroix
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| |
Collapse
|
20
|
Boudina M, Elfring GJ. Capillary Imbibition in a Diverging Flexible Channel. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:12174-12181. [PMID: 37594738 DOI: 10.1021/acs.langmuir.3c01488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
We study the imbibition of a wetting liquid between flexible sheets that are fixed at both ends. Assuming a narrow gap between the sheets, we solve the lubrication equation coupled with a slender body deformation. When the sheets are parallel, we find that the deformation initially speeds up the flow, as shown in previous studies, but only up to the middle of the channel. As the channel contracts, the hydrodynamic resistance increases and ultimately slows down the filling process. Below a threshold stiffness, the channel collapses and imbibition stops. We propose a scaling of the filling duration near this threshold. Next, we show that if the sheets are initially tilted with a minimal angle, the channel avoids collapse. The liquid front pulls the diverging sheets and spreads in a nearly parallel portion, which maintains the capillary propulsion and enhances the wicking. Therefore, while it is established that diverging rigid plates imbibe liquids slower than parallel ones do, we show that elasticity reverses this principle: diverging flexible sheets imbibe liquids faster than parallel ones. We find an optimal tilt angle that gives the shortest filling time.
Collapse
Affiliation(s)
- Mouad Boudina
- Department of Mechanical Engineering, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
| | - Gwynn J Elfring
- Department of Mechanical Engineering, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
| |
Collapse
|
21
|
Pillai S, Kwan JC, Yaziji F, Yu H, Tran SD. Mapping the Potential of Microfluidics in Early Diagnosis and Personalized Treatment of Head and Neck Cancers. Cancers (Basel) 2023; 15:3894. [PMID: 37568710 PMCID: PMC10417175 DOI: 10.3390/cancers15153894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Head and neck cancers (HNCs) account for ~4% of all cancers in North America and encompass cancers affecting the oral cavity, pharynx, larynx, sinuses, nasal cavity, and salivary glands. The anatomical complexity of the head and neck region, characterized by highly perfused and innervated structures, presents challenges in the early diagnosis and treatment of these cancers. The utilization of sub-microliter volumes and the unique phenomenon associated with microscale fluid dynamics have facilitated the development of microfluidic platforms for studying complex biological systems. The advent of on-chip microfluidics has significantly impacted the diagnosis and treatment strategies of HNC. Sensor-based microfluidics and point-of-care devices have improved the detection and monitoring of cancer biomarkers using biological specimens like saliva, urine, blood, and serum. Additionally, tumor-on-a-chip platforms have allowed the creation of patient-specific cancer models on a chip, enabling the development of personalized treatments through high-throughput screening of drugs. In this review, we first focus on how microfluidics enable the development of an enhanced, functional drug screening process for targeted treatment in HNCs. We then discuss current advances in microfluidic platforms for biomarker sensing and early detection, followed by on-chip modeling of HNC to evaluate treatment response. Finally, we address the practical challenges that hinder the clinical translation of these microfluidic advances.
Collapse
Affiliation(s)
| | | | | | | | - Simon D. Tran
- McGill Craniofacial Tissue Engineering and Stem Cell Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada; (S.P.); (J.C.K.); (F.Y.); (H.Y.)
| |
Collapse
|
22
|
He Y, Zhan L, Shi J, Xiao M, Zuo R, Wang C, Liu Z, Gong W, Chen L, Luo Y, Zhang S, Wang Y, Chen L, Guo H. The Combination of R848 with Sorafenib Enhances Antitumor Effects by Reprogramming the Tumor Immune Microenvironment and Facilitating Vascular Normalization in Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207650. [PMID: 37083239 PMCID: PMC10288281 DOI: 10.1002/advs.202207650] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/20/2023] [Indexed: 05/03/2023]
Abstract
Novel promising strategies for combination with sorafenib are urgently needed to enhance its clinical benefit and overcome toxicity in hepatocellular carcinoma (HCC). the molecular and immunomodulatory antitumor effects of sorafenib alone and in combination with the new immunotherapeutic agent R848 are presented. Syngeneic HCC mouse model is presented to explore the antitumor effect and safety of three sorafenib doses alone, R848 alone, or their combination in vivo. R848 significantly enhances the sorafenib antitumor activity at a low subclinical dose with no obvious toxic side effects. Furthermore, the combination therapy reprograms the tumor immune microenvironment by increasing antitumor macrophages and neutrophils and preventing immunosuppressive signaling. Combination treatment promotes classical M1 macrophage-to-FTH1high M1 macrophage transition. The close interaction between neutrophils/classical M1 macrophages and dendritic cells promotes tumor antigen presentation to T cells, inducing cytotoxic CD8+ T cell-mediated antitumor immunity. Additionally, low-dose sorafenib, alone or combined with R848, normalizes the tumor vasculature, generating a positive feedback loop to support the antitumor immune environment. Therefore, the combination therapy reprograms the HCC immune microenvironment and normalizes the vasculature, improving the therapeutic benefit of low-dose sorafenib and minimizing toxicity, suggesting a promising novel immunotherapy (R848) and targeted therapy (tyrosine kinase inhibitors) combination strategy for HCC treatment.
Collapse
Affiliation(s)
- Yuchao He
- Department of Tumor Cell BiologyTianjin Medical University Cancer Institute and HospitalTianjin300060China
- National Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerTianjin300060China
| | - Linlin Zhan
- Department of Tumor Cell BiologyTianjin Medical University Cancer Institute and HospitalTianjin300060China
- National Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerTianjin300060China
| | - Jian Shi
- Institute of Precision MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhou510080China
| | - Manyu Xiao
- School of PharmacyMinzu University of ChinaBeijing10081China
| | - Ran Zuo
- Department of Tumor Cell BiologyTianjin Medical University Cancer Institute and HospitalTianjin300060China
- National Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerTianjin300060China
| | - Chengmeng Wang
- Department of Tumor Cell BiologyTianjin Medical University Cancer Institute and HospitalTianjin300060China
- National Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerTianjin300060China
| | - Zhiyong Liu
- Department of Tumor Cell BiologyTianjin Medical University Cancer Institute and HospitalTianjin300060China
- National Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerTianjin300060China
| | - Wenchen Gong
- National Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerTianjin300060China
- Department of PathologyTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Liwei Chen
- Department of Tumor Cell BiologyTianjin Medical University Cancer Institute and HospitalTianjin300060China
- National Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerTianjin300060China
| | - Yi Luo
- Department of Tumor Cell BiologyTianjin Medical University Cancer Institute and HospitalTianjin300060China
- National Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerTianjin300060China
| | - Shaojun Zhang
- Medical Research InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Youwei Wang
- Institute of Medical Engineering & Translational MedicineTianjin UniversityTianjin300072China
| | - Lu Chen
- National Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerTianjin300060China
- Department of Hepatobiliary CancerLiver Cancer Research CenterTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Hua Guo
- Department of Tumor Cell BiologyTianjin Medical University Cancer Institute and HospitalTianjin300060China
- National Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerTianjin300060China
| |
Collapse
|
23
|
Wu KZ, Adine C, Mitriashkin A, Aw BJJ, Iyer NG, Fong ELS. Making In Vitro Tumor Models Whole Again. Adv Healthc Mater 2023; 12:e2202279. [PMID: 36718949 PMCID: PMC11469124 DOI: 10.1002/adhm.202202279] [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: 09/06/2022] [Revised: 01/04/2023] [Indexed: 02/01/2023]
Abstract
As a reductionist approach, patient-derived in vitro tumor models are inherently still too simplistic for personalized drug testing as they do not capture many characteristics of the tumor microenvironment (TME), such as tumor architecture and stromal heterogeneity. This is especially problematic for assessing stromal-targeting drugs such as immunotherapies in which the density and distribution of immune and other stromal cells determine drug efficacy. On the other end, in vivo models are typically costly, low-throughput, and time-consuming to establish. Ex vivo patient-derived tumor explant (PDE) cultures involve the culture of resected tumor fragments that potentially retain the intact TME of the original tumor. Although developed decades ago, PDE cultures have not been widely adopted likely because of their low-throughput and poor long-term viability. However, with growing recognition of the importance of patient-specific TME in mediating drug response, especially in the field of immune-oncology, there is an urgent need to resurrect these holistic cultures. In this Review, the key limitations of patient-derived tumor explant cultures are outlined and technologies that have been developed or could be employed to address these limitations are discussed. Engineered holistic tumor explant cultures may truly realize the concept of personalized medicine for cancer patients.
Collapse
Affiliation(s)
- Kenny Zhuoran Wu
- Department of Biomedical EngineeringCollege of Design and EngineeringNational University of SingaporeSingapore119276Singapore
| | - Christabella Adine
- Department of Biomedical EngineeringCollege of Design and EngineeringNational University of SingaporeSingapore119276Singapore
| | - Aleksandr Mitriashkin
- Department of Biomedical EngineeringCollege of Design and EngineeringNational University of SingaporeSingapore119276Singapore
| | - Benjamin Jun Jie Aw
- Department of Biomedical EngineeringCollege of Design and EngineeringNational University of SingaporeSingapore119276Singapore
| | - N. Gopalakrishna Iyer
- Department of Head and Neck Surgery, Division of Surgery and Surgical OncologyDuke‐NUS Medical SchoolSingapore169857Singapore
- Department of Head and Neck SurgeryNational Cancer Centre SingaporeSingapore169610Singapore
| | - Eliza Li Shan Fong
- Department of Biomedical EngineeringCollege of Design and EngineeringNational University of SingaporeSingapore119276Singapore
- The N.1 Institute for HealthNational University of SingaporeSingapore117456Singapore
- Cancer Science Institute (CSI)National University of SingaporeSingapore117599Singapore
| |
Collapse
|
24
|
Barry A, Samuel SF, Hosni I, Moursi A, Feugere L, Sennett CJ, Deepak S, Achawal S, Rajaraman C, Iles A, Wollenberg Valero KC, Scott IS, Green V, Stead LF, Greenman J, Wade MA, Beltran-Alvarez P. Investigating the effects of arginine methylation inhibitors on microdissected brain tumour biopsies maintained in a miniaturised perfusion system. LAB ON A CHIP 2023; 23:2664-2682. [PMID: 37191188 DOI: 10.1039/d3lc00204g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Arginine methylation is a post-translational modification that consists of the transfer of one or two methyl (CH3) groups to arginine residues in proteins. Several types of arginine methylation occur, namely monomethylation, symmetric dimethylation and asymmetric dimethylation, which are catalysed by different protein arginine methyltransferases (PRMTs). Inhibitors of PRMTs have recently entered clinical trials to target several types of cancer, including gliomas (NCT04089449). People with glioblastoma (GBM), the most aggressive form of brain tumour, are among those with the poorest quality of life and likelihood of survival of anyone diagnosed with cancer. There is currently a lack of (pre)clinical research on the possible application of PRMT inhibitors to target brain tumours. Here, we set out to investigate the effects of clinically-relevant PRMT inhibitors on GBM biopsies. We present a new, low-cost, easy to fabricate perfusion device that can maintain GBM tissue in a viable condition for at least eight days post-surgical resection. The miniaturised perfusion device enables the treatment of GBM tissue with PRMT inhibitors ex vivo, and we observed a two-fold increase in apoptosis in treated samples compared to parallel control experiments. Mechanistically, we show thousands of differentially expressed genes after treatment, and changes in the type of arginine methylation of the RNA binding protein FUS that are consistent with hundreds of differential gene splicing events. This is the first time that cross-talk between different types of arginine methylation has been observed in clinical samples after treatment with PRMT inhibitors.
Collapse
Affiliation(s)
- Antonia Barry
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull, UK.
| | - Sabrina F Samuel
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull, UK.
| | - Ines Hosni
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull, UK.
| | - Amr Moursi
- Department of Neurosurgery, Hull University Teaching Hospitals NHS Trust, Hull Royal Infirmary, Hull, UK
| | - Lauric Feugere
- Department of Biological and Marine Sciences, University of Hull, Hull, UK
| | | | - Srihari Deepak
- Department of Neurosurgery, Hull University Teaching Hospitals NHS Trust, Hull Royal Infirmary, Hull, UK
| | - Shailendra Achawal
- Department of Neurosurgery, Hull University Teaching Hospitals NHS Trust, Hull Royal Infirmary, Hull, UK
| | - Chittoor Rajaraman
- Department of Neurosurgery, Hull University Teaching Hospitals NHS Trust, Hull Royal Infirmary, Hull, UK
| | | | | | - Ian S Scott
- Neuroscience Laboratories, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - Vicky Green
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull, UK.
| | - Lucy F Stead
- Leeds Institute of Medical Research at St James's, St James's University Hospital, Leeds, UK
| | - John Greenman
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull, UK.
| | - Mark A Wade
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull, UK.
| | | |
Collapse
|
25
|
Ozulumba T, Montalbine AN, Ortiz-Cárdenas JE, Pompano RR. New tools for immunologists: models of lymph node function from cells to tissues. Front Immunol 2023; 14:1183286. [PMID: 37234163 PMCID: PMC10206051 DOI: 10.3389/fimmu.2023.1183286] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The lymph node is a highly structured organ that mediates the body's adaptive immune response to antigens and other foreign particles. Central to its function is the distinct spatial assortment of lymphocytes and stromal cells, as well as chemokines that drive the signaling cascades which underpin immune responses. Investigations of lymph node biology were historically explored in vivo in animal models, using technologies that were breakthroughs in their time such as immunofluorescence with monoclonal antibodies, genetic reporters, in vivo two-photon imaging, and, more recently spatial biology techniques. However, new approaches are needed to enable tests of cell behavior and spatiotemporal dynamics under well controlled experimental perturbation, particularly for human immunity. This review presents a suite of technologies, comprising in vitro, ex vivo and in silico models, developed to study the lymph node or its components. We discuss the use of these tools to model cell behaviors in increasing order of complexity, from cell motility, to cell-cell interactions, to organ-level functions such as vaccination. Next, we identify current challenges regarding cell sourcing and culture, real time measurements of lymph node behavior in vivo and tool development for analysis and control of engineered cultures. Finally, we propose new research directions and offer our perspective on the future of this rapidly growing field. We anticipate that this review will be especially beneficial to immunologists looking to expand their toolkit for probing lymph node structure and function.
Collapse
Affiliation(s)
- Tochukwu Ozulumba
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Alyssa N. Montalbine
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Jennifer E. Ortiz-Cárdenas
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia (UVA) Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
26
|
Fröhlich E. The Variety of 3D Breast Cancer Models for the Study of Tumor Physiology and Drug Screening. Int J Mol Sci 2023; 24:ijms24087116. [PMID: 37108283 PMCID: PMC10139112 DOI: 10.3390/ijms24087116] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/01/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Breast cancer is the most common cancer in women and responsible for multiple deaths worldwide. 3D cancer models enable a better representation of tumor physiology than the conventional 2D cultures. This review summarizes the important components of physiologically relevant 3D models and describes the spectrum of 3D breast cancer models, e.g., spheroids, organoids, breast cancer on a chip and bioprinted tissues. The generation of spheroids is relatively standardized and easy to perform. Microfluidic systems allow control over the environment and the inclusion of sensors and can be combined with spheroids or bioprinted models. The strength of bioprinting relies on the spatial control of the cells and the modulation of the extracellular matrix. Except for the predominant use of breast cancer cell lines, the models differ in stromal cell composition, matrices and fluid flow. Organoids are most appropriate for personalized treatment, but all technologies can mimic most aspects of breast cancer physiology. Fetal bovine serum as a culture supplement and Matrigel as a scaffold limit the reproducibility and standardization of the listed 3D models. The integration of adipocytes is needed because they possess an important role in breast cancer.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, 8010 Graz, Austria
- Research Center Pharmaceutical Engineering GmbH, 8010 Graz, Austria
| |
Collapse
|
27
|
Zhou L, Liu L, Chang MA, Ma C, Chen W, Chen P. Spatiotemporal dissection of tumor microenvironment via in situ sensing and monitoring in tumor-on-a-chip. Biosens Bioelectron 2023; 225:115064. [PMID: 36680970 PMCID: PMC9918721 DOI: 10.1016/j.bios.2023.115064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Real-time monitoring in the tumor microenvironment provides critical insights of cancer progression and mechanistic understanding of responses to cancer treatments. However, clinical challenges and significant questions remain regarding assessment of limited clinical tissue samples, establishment of validated, controllable pre-clinical cancer models, monitoring of static versus dynamic markers, and the translation of insights gained from in vitro tumor microenvironments to systematic investigation and understanding in clinical practice. State-of-art tumor-on-a-chip strategies will be reviewed herein, and emerging real-time sensing and monitoring platforms for on-chip analysis of tumor microenvironment will also be examined. The integration of the sensors with tumor-on-a-chip platforms to provide spatiotemporal information of the tumor microenvironment and the associated challenges will be further evaluated. Though optimal integrated systems for in situ monitoring are still in evolution, great promises lie ahead that will open new paradigm for rapid, comprehensive analysis of cancer development and assist clinicians with powerful tools to guide the diagnosis, prognosis and treatment course in cancer.
Collapse
Affiliation(s)
- Lang Zhou
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Lunan Liu
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Muammar Ali Chang
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Pengyu Chen
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
28
|
Sailer V, von Amsberg G, Duensing S, Kirfel J, Lieb V, Metzger E, Offermann A, Pantel K, Schuele R, Taubert H, Wach S, Perner S, Werner S, Aigner A. Experimental in vitro, ex vivo and in vivo models in prostate cancer research. Nat Rev Urol 2023; 20:158-178. [PMID: 36451039 DOI: 10.1038/s41585-022-00677-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2022] [Indexed: 12/02/2022]
Abstract
Androgen deprivation therapy has a central role in the treatment of advanced prostate cancer, often causing initial tumour remission before increasing independence from signal transduction mechanisms of the androgen receptor and then eventual disease progression. Novel treatment approaches are urgently needed, but only a fraction of promising drug candidates from the laboratory will eventually reach clinical approval, highlighting the demand for critical assessment of current preclinical models. Such models include standard, genetically modified and patient-derived cell lines, spheroid and organoid culture models, scaffold and hydrogel cultures, tissue slices, tumour xenograft models, patient-derived xenograft and circulating tumour cell eXplant models as well as transgenic and knockout mouse models. These models need to account for inter-patient and intra-patient heterogeneity, the acquisition of primary or secondary resistance, the interaction of tumour cells with their microenvironment, which make crucial contributions to tumour progression and resistance, as well as the effects of the 3D tissue network on drug penetration, bioavailability and efficacy.
Collapse
Affiliation(s)
- Verena Sailer
- Institute for Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Gunhild von Amsberg
- Department of Oncology and Hematology, University Cancer Center Hamburg Eppendorf and Martini-Klinik, Prostate Cancer Center, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Stefan Duensing
- Section of Molecular Urooncology, Department of Urology, University Hospital Heidelberg and National Center for Tumour Diseases, Heidelberg, Germany
| | - Jutta Kirfel
- Institute for Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Verena Lieb
- Research Division Molecular Urology, Department of Urology and Paediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Eric Metzger
- Department of Urology, Center for Clinical Research, University of Freiburg Medical Center, Freiburg, Germany
| | - Anne Offermann
- Institute for Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Klaus Pantel
- Institute for Tumour Biology, Center for Experimental Medicine, University Clinics Hamburg-Eppendorf, Hamburg, Germany
- Mildred-Scheel-Nachwuchszentrum HaTRiCs4, University Cancer Center Hamburg, Hamburg, Germany
| | - Roland Schuele
- Department of Urology, Center for Clinical Research, University of Freiburg Medical Center, Freiburg, Germany
| | - Helge Taubert
- Research Division Molecular Urology, Department of Urology and Paediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Sven Wach
- Research Division Molecular Urology, Department of Urology and Paediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Sven Perner
- University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Stefan Werner
- Institute for Tumour Biology, Center for Experimental Medicine, University Clinics Hamburg-Eppendorf, Hamburg, Germany
- Mildred-Scheel-Nachwuchszentrum HaTRiCs4, University Cancer Center Hamburg, Hamburg, Germany
| | - Achim Aigner
- Clinical Pharmacology, Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of Leipzig, Medical Faculty, Leipzig, Germany.
| |
Collapse
|
29
|
Komar ZM, van Gent DC, Chakrabarty S. Establishing a Microfluidic Tumor Slice Culture Platform to Study Drug Response. Curr Protoc 2023; 3:e693. [PMID: 36912175 DOI: 10.1002/cpz1.693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Accurate models for tumor biology and prediction of drug responses of individual tumors require novel technology to grow tumor tissue ex vivo to maintain tumor growth characteristics in situ. Models containing only tumor cells, without the stromal components of the tumor, are suboptimal for many purposes and are generally problematic because the cells are passed through extensive culture and selection. Therefore, direct culture of (human) tumors is of considerable interest for basic tumor biology and diagnostic purposes. Microfluidic technologies have been proposed to accurately mimic physiological conditions for tissue growth. Most published systems build tissues from individual cell types in so-called Organ-on-Chip (OoC) cultures. We here describe a novel OoC device for growing tumor specimens. Thin tumor slices are grown in a microfluidic 'chip' that allows precisely controlled in vitro culture conditions. The performance of the OoC device was extensively validated for predicting therapeutic responses in human breast cancer patient-derived xenograft (PDX) tumor material. The system is amenable to primary tumor material from surgery or biopsies. In addition to using the model to predict and evaluate therapeutic responses, the model can also be used for mechanistic studies of human cancers, such as clonal evolution or immune responses, or to validate new or repurposed (cancer) drugs. The Bi/ond Cancer-on-Chip (CoC) device is designed to culture tumor slices and investigate aspects of tumor growth and drug responses. Here, we describe the step-by-step process of setting up tumor slice cultures using a Bi/ond CoC device and performing in vitro drug response evaluation. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Establishment of breast cancer tumor slice culture using a microfluidic cancer-on-chip platform for chemotherapy testing ex vivo Basic Protocol 2: Histology and immunohistochemistry-based analysis of tumor tissue architecture, cell proliferation, and cell death.
Collapse
Affiliation(s)
- Zofia M Komar
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dik C van Gent
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands.,Oncode Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sanjiban Chakrabarty
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
30
|
Pixelated Microfluidics for Drug Screening on Tumour Spheroids and Ex Vivo Microdissected Tumour Explants. Cancers (Basel) 2023; 15:cancers15041060. [PMID: 36831403 PMCID: PMC9954565 DOI: 10.3390/cancers15041060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Anticancer drugs have the lowest success rate of approval in drug development programs. Thus, preclinical assays that closely predict the clinical responses to drugs are of utmost importance in both clinical oncology and pharmaceutical research. 3D tumour models preserve the tumoral architecture and are cost- and time-efficient. However, the short-term longevity, limited throughput, and limitations of live imaging of these models have so far driven researchers towards less realistic tumour models such as monolayer cell cultures. Here, we present an open-space microfluidic drug screening platform that enables the formation, culture, and multiplexed delivery of several reagents to various 3D tumour models, namely cancer cell line spheroids and ex vivo primary tumour fragments. Our platform utilizes a microfluidic pixelated chemical display that creates isolated adjacent flow sub-units of reagents, which we refer to as fluidic 'pixels', over tumour models in a contact-free fashion. Up to nine different treatment conditions can be tested over 144 samples in a single experiment. We provide a proof-of-concept application by staining fixed and live tumour models with multiple cellular dyes. Furthermore, we demonstrate that the response of the tumour models to biological stimuli can be assessed using the platform. Upscaling the microfluidic platform to larger areas can lead to higher throughputs, and thus will have a significant impact on developing treatments for cancer.
Collapse
|
31
|
Vega JMDH, Hong HJ, Loutherback K, Stybayeva G, Revzin A. A Microfluidic Device for Long-Term Maintenance of Organotypic Liver Cultures. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201121. [PMID: 36818276 PMCID: PMC9937715 DOI: 10.1002/admt.202201121] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Indexed: 06/03/2023]
Abstract
Liver cultures may be used for disease modeling, testing therapies and predicting drug-induced injury. The complexity of the liver cultures has evolved from hepatocyte monocultures to co-cultures with non-parenchymal cells and finally to precision-cut liver slices. The latter culture format retains liver's native biomolecular and cellular complexity and therefore holds considerable promise for in vitro testing. However, liver slices remain functional for ~72 h in vitro and display limited utility for some disease modeling and therapy testing applications that require longer culture times. This paper describes a microfluidic device for longer-term maintenance of functional organotypic liver cultures. Our microfluidic culture system was designed to enable direct injection of liver tissue into a culture chamber through a valve-enabled side port. Liver tissue was embedded in collagen and remained functional for up to 31 days, highlighted by continued production of albumin and urea. These organotypic cultures also expressed several enzymes involved in xenobiotic metabolism. Conversely, matched liver tissue embedded in collagen in a 96-well plate lost its phenotype and function within 3-5 days. The microfluidic organotypic liver cultures described here represent a significant advance in liver cultivation and may be used for future modeling of liver diseases or for individualized liver-directed therapies.
Collapse
Affiliation(s)
- José M. de Hoyos Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Hye Jin Hong
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kevin Loutherback
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
32
|
Victorious A. Current Applications of Organ-on-a-Chip: A Step Closer to Personalized Medicine. BIO INTEGRATION 2022. [DOI: 10.15212/bioi-2022-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Abstract In the pharmaceutical industry, a critical need exists for effective drug development approaches that better account for factors imposed by the physiological microenvironment. Organ-on-a-chip (OOAC)—a revolutionary technology that simulates human organs’
physiological milieu and performance on a chip—has applications in curing illnesses and drug screening, and enormous potential to transform the drug discovery workflow. However, the effective integration of this unique engineering system into ordinary pharmacological and medical contexts
remains in development. This Editorial summarizes current research on OOAC systems, and offers insight into future development prospects and the need for a next-generation OOAC framework.
Collapse
|
33
|
Manzano-Muñoz A, Yeste J, Ortega MA, Martín F, López A, Rosell J, Castro S, Serrano C, Samitier J, Ramón-Azcón J, Montero J. Microfluidic-based dynamic BH3 profiling predicts anticancer treatment efficacy. NPJ Precis Oncol 2022; 6:90. [PMID: 36456699 PMCID: PMC9715649 DOI: 10.1038/s41698-022-00333-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 11/08/2022] [Indexed: 12/04/2022] Open
Abstract
Precision medicine is starting to incorporate functional assays to evaluate anticancer agents on patient-isolated tissues or cells to select for the most effective. Among these new technologies, dynamic BH3 profiling (DBP) has emerged and extensively been used to predict treatment efficacy in different types of cancer. DBP uses synthetic BH3 peptides to measure early apoptotic events ('priming') and anticipate therapy-induced cell death leading to tumor elimination. This predictive functional assay presents multiple advantages but a critical limitation: the cell number requirement, that limits drug screening on patient samples, especially in solid tumors. To solve this problem, we developed an innovative microfluidic-based DBP (µDBP) device that overcomes tissue limitations on primary samples. We used microfluidic chips to generate a gradient of BIM BH3 peptide, compared it with the standard flow cytometry based DBP, and tested different anticancer treatments. We first examined this new technology's predictive capacity using gastrointestinal stromal tumor (GIST) cell lines, by comparing imatinib sensitive and resistant cells, and we could detect differences in apoptotic priming and anticipate cytotoxicity. We then validated µDBP on a refractory GIST patient sample and identified that the combination of dactolisib and venetoclax increased apoptotic priming. In summary, this new technology could represent an important advance for precision medicine by providing a fast, easy-to-use and scalable microfluidic device to perform DBP in situ as a routine assay to identify the best treatment for cancer patients.
Collapse
Grants
- Ramon y Cajal Programme, Ministerio de Economia y Competitividad grant RYC-2015-18357. (JM) Ministerio de Ciencia, Innovación y Universidades grant RTI2018-094533-A-I00 (JM) CELLEX foundation (JM, AM). Beca Trienal Fundación Mari Paz Jiménez Casado (JM)
- Fundación Cellex (Cellex Foundation)
- Networking Biomedical Research Center (CIBER). CIBER is an initiative funded by the VI National R & D &i Plan 2008–2011, Iniciativa Ingenio 2010, Consolider Program, CIBER Actions, and the Instituto de Salud Carlos III (RD16/0006/0012), with the support of the European Regional Development Fund (JS). Generalitat de Catalunya. CERCA Programme 2017-SGR-1079 (JR-A, JS)
- European Research Council, grant ERC-StG-DAMOC 714317 (JR-A) European Research Council, H2020 EU framework FET-open BLOC 863037 (JR-A) Spanish Ministry of Economy and Competitiveness, "Severo Ochoa" Program for Centers of Excellence in R&D SEV-2020-2023 (JR-A) Generalitat de Catalunya. CERCA Programme 2017-SGR-1079 (JR-A, JS) Fundación Bancaria "la Caixa"- Obra Social "la Caixa" (project IBEC-La Caixa Health Ageing) (JR-A)
Collapse
Affiliation(s)
- Albert Manzano-Muñoz
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - José Yeste
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - María A Ortega
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Vitala Technologies, Barcelona, Spain
| | - Fernando Martín
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Anna López
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Jordi Rosell
- Sarcoma Translational Research Program, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitario Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Sandra Castro
- Surgical Oncology Division, Vall d'Hebron University Hospital, Barcelona, Spain
| | - César Serrano
- Sarcoma Translational Research Program, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitario Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
- Department of Electronics and Biomedical Engineering, Faculty of Physics, University of Barcelona, Barcelona, Spain
| | - Javier Ramón-Azcón
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Institució Catalana de Reserca i Estudis Avançats (ICREA), Passeig de Lluís Companys, 23, E08010, Barcelona, Spain
| | - Joan Montero
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Casanova 143, Barcelona, 08036, Spain.
| |
Collapse
|
34
|
Fang X, Wang Y, Wang S, Liu B. Nanomaterials assisted exosomes isolation and analysis towards liquid biopsy. Mater Today Bio 2022; 16:100371. [PMID: 35937576 PMCID: PMC9352971 DOI: 10.1016/j.mtbio.2022.100371] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/13/2022] [Accepted: 07/17/2022] [Indexed: 11/18/2022] Open
Abstract
Exosomes has attracted tremendous research interests as they are emerging as a new paradigm of liquid biopsy. Although the concentration of exosomes in blood is relatively abundant, there still exists various vesicle-like nanoparticles, such as microvesicles, apoptotic bodies. It's an urgent need to isolate and enrich exosomes from the complex contaminants in biofluid samples. Moreover, the expressing level of exosomal biomarkers varies a lot, which make the sensitive molecular detection of exosomes in high demand. Unfortunately, the efficient isolation and sensitive molecular quantification of exosomes is still a major obstacle hindering the further development and clinical application of exosome-based liquid biopsy. Nanomaterials, with unique physiochemical properties, have been widely used in biosensing and analysis aspects, thus they are thought as powerful tools for effective purification and molecular analysis of exosomes. In this review, we summarized the most recent progresses in nanomaterials assisted exosome isolation and analysis towards liquid biopsy. On the one hand, nanomaterials can be used as capture substrates to afford large binding area and specific affinity to exosomes. Meanwhile, nanomaterials can also be served as promising signal transducers and amplifiers for molecular detection of exosomes. Furthermore, we also pointed out several potential and promising research directions in nanomaterials assisted exosome analysis. It's envisioned that this review will give the audience a complete outline of nanomaterials in exosome study, and further promote the intersection of nanotechnology and bio-analysis.
Collapse
Affiliation(s)
- Xiaoni Fang
- School of Pharmacy, Shanghai Stomatological Hospital, Department of Chemistry, Fudan University, Shanghai, 200438, China
| | - Yuqing Wang
- School of Pharmacy, Shanghai Stomatological Hospital, Department of Chemistry, Fudan University, Shanghai, 200438, China
| | - Shurong Wang
- School of Pharmacy, Shanghai Stomatological Hospital, Department of Chemistry, Fudan University, Shanghai, 200438, China
| | - Baohong Liu
- School of Pharmacy, Shanghai Stomatological Hospital, Department of Chemistry, Fudan University, Shanghai, 200438, China
| |
Collapse
|
35
|
Utharala R, Grab A, Vafaizadeh V, Peschke N, Ballinger M, Turei D, Tuechler N, Ma W, Ivanova O, Ortiz AG, Saez-Rodriguez J, Merten CA. A microfluidic Braille valve platform for on-demand production, combinatorial screening and sorting of chemically distinct droplets. Nat Protoc 2022; 17:2920-2965. [PMID: 36261631 DOI: 10.1038/s41596-022-00740-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 06/16/2022] [Indexed: 11/09/2022]
Abstract
Droplet microfluidics is a powerful tool for a variety of biological applications including single-cell genetics, antibody discovery and directed evolution. All these applications make use of genetic libraries, illustrating the difficulty of generating chemically distinct droplets for screening applications. This protocol describes our Braille Display valving platform for on-demand generation of droplets with different chemical contents (16 different reagents and combinations thereof), as well as sorting droplets with different chemical properties, on the basis of fluorescence signals. The Braille Display platform is compact, versatile and cost efficient (only ~US$1,000 on top of a standard droplet microfluidics setup). The procedure includes manufacturing of microfluidic chips, assembly of custom hardware, co-encapsulation of cells and drugs into droplets, fluorescence detection of readout signals and data analysis using shared, freely available LabVIEW and Python packages. As a first application, we demonstrate the complete workflow for screening cancer cell drug sensitivities toward 74 conditions. Furthermore, we describe here an assay enabling the normalization of the observed drug sensitivity to the number of cancer cells per droplet, which additionally increases the robustness of the system. As a second application, we also demonstrate the sorting of droplets according to enzymatic activity. The drug screening application can be completed within 2 d; droplet sorting takes ~1 d; and all preparatory steps for manufacturing molds, chips and setting up the Braille controller can be accomplished within 1 week.
Collapse
Affiliation(s)
- Ramesh Utharala
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Anna Grab
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Clinical Cooperation Unit Molecular Hematology/Oncology, DKFZ Heidelberg and Translational Myeloma Research Group, Department of Internal Medicine V, University Hospital, Heidelberg, Germany
| | - Vida Vafaizadeh
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nicolas Peschke
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Martine Ballinger
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Denes Turei
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Faculty of Medicine and Heidelberg University Hospital, Institute of Computational Biomedicine, Heidelberg University, Heidelberg, Germany
| | - Nadine Tuechler
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Wenwei Ma
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Olga Ivanova
- Faculty of Medicine and Heidelberg University Hospital, Institute of Computational Biomedicine, Heidelberg University, Heidelberg, Germany
| | | | - Julio Saez-Rodriguez
- Faculty of Medicine and Heidelberg University Hospital, Institute of Computational Biomedicine, Heidelberg University, Heidelberg, Germany
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen University, Aachen, Germany
| | - Christoph A Merten
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
36
|
Budhwani KI, Patel ZH, Guenter RE, Charania AA. A hitchhiker's guide to cancer models. Trends Biotechnol 2022; 40:1361-1373. [PMID: 35534320 PMCID: PMC9588514 DOI: 10.1016/j.tibtech.2022.04.003] [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: 01/25/2022] [Revised: 03/31/2022] [Accepted: 04/08/2022] [Indexed: 01/21/2023]
Abstract
Cancer is a complex and uniquely personal disease. More than 1.7 million people in the United States are diagnosed with cancer every year. As the burden of cancer grows, so does the need for new, more effective therapeutics and for predictive tools to identify optimal, personalized treatment options for every patient. Cancer models that recapitulate various aspects of the disease are fundamental to making advances along the continuum of cancer treatment from benchside discoveries to bedside delivery. In this review, we use a thought experiment as a vehicle to arrive at four broad categories of cancer models and explore the strengths, weaknesses, opportunities, and threats for each category in advancing our understanding of the disease and improving treatment strategies.
Collapse
Affiliation(s)
- Karim I Budhwani
- CerFlux, Inc., Birmingham, AL, USA; Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA; Department of Physics, Coe College, Cedar Rapids, IA, USA.
| | | | | | | |
Collapse
|
37
|
Foglizzo V, Cocco E, Marchiò S. Advanced Cellular Models for Preclinical Drug Testing: From 2D Cultures to Organ-On-A-Chip Technology. Cancers (Basel) 2022; 14:cancers14153692. [PMID: 35954355 PMCID: PMC9367322 DOI: 10.3390/cancers14153692] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/18/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Novel strategies that aim at personalizing cancer therapy are in rapid evolution. In the past decade, new methods to test for the efficacy of either standard-of-care medicines or novel targeted compounds have been implemented. In this review, we introduce the reader to experimental studies that employ patient-derived material to produce spheroids, organoids, or organs-on-a-chip as platforms that allow a more accurate representation of cancer complexity compared to bidimensional cell cultures. We discuss on the versatility and reliability of these model systems, provide evidence of their usage in drug screenings, and describe potential downfalls. The open question is whether or not tumor mimicry in vitro will be, in the near future, advanced enough to prospectively inform about treatment outcome on a certain patient. Abstract Cancer is a complex disease arising from a homeostatic imbalance of cell-intrinsic and microenvironment-related mechanisms. A multimodal approach to treat cancer that includes surgery, chemotherapy, and radiation therapy often fails in achieving tumor remission and produces unbearable side effects including secondary malignancies. Novel strategies have been implemented in the past decades in order to replace conventional chemotherapeutics with targeted, less toxic drugs. Up to now, scientists have relied on results achieved in animal research before proceeding to clinical trials. However, the high failure rate of targeted drugs in early phase trials leaves no doubt about the inadequacy of those models. In compliance with the need of reducing, and possibly replacing, animal research, studies have been conducted in vitro with advanced cellular models that more and more mimic the tumor in vivo. We will here review those methods that allow for the 3D reconstitution of the tumor and its microenvironment and the implementation of the organ-on-a-chip technology to study minimal organ units in disease progression. We will make specific reference to the usability of these systems as predictive cancer models and report on recent applications in high-throughput screenings of innovative and targeted drug compounds.
Collapse
Affiliation(s)
- Valentina Foglizzo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (V.F.); (E.C.)
| | - Emiliano Cocco
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (V.F.); (E.C.)
| | - Serena Marchiò
- Department of Oncology, University of Torino, 10060 Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
- Correspondence:
| |
Collapse
|
38
|
|
39
|
Chermat R, Ziaee M, Mak DY, Refet-Mollof E, Rodier F, Wong P, Carrier JF, Kamio Y, Gervais T. Radiotherapy on-chip: microfluidics for translational radiation oncology. LAB ON A CHIP 2022; 22:2065-2079. [PMID: 35477748 DOI: 10.1039/d2lc00177b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The clinical importance of radiotherapy in the treatment of cancer patients justifies the development and use of research tools at the fundamental, pre-clinical, and ultimately clinical levels, to investigate their toxicities and synergies with systemic agents on relevant biological samples. Although microfluidics has prompted a paradigm shift in drug discovery in the past two decades, it appears to have yet to translate to radiotherapy research. However, the materials, dimensions, design versatility and multiplexing capabilities of microfluidic devices make them well-suited to a variety of studies involving radiation physics, radiobiology and radiotherapy. This review will present the state-of-the-art applications of microfluidics in these fields and specifically highlight the perspectives offered by radiotherapy on-a-chip in the field of translational radiobiology and precision medicine. This body of knowledge can serve both the microfluidics and radiotherapy communities by identifying potential collaboration avenues to improve patient care.
Collapse
Affiliation(s)
- Rodin Chermat
- μFO Lab, Polytechnique Montréal, Montréal, QC, Canada.
- Institut du Cancer de Montréal, (ICM), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Maryam Ziaee
- μFO Lab, Polytechnique Montréal, Montréal, QC, Canada.
- Institut du Cancer de Montréal, (ICM), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - David Y Mak
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Elena Refet-Mollof
- μFO Lab, Polytechnique Montréal, Montréal, QC, Canada.
- Institut du Cancer de Montréal, (ICM), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Francis Rodier
- Institut du Cancer de Montréal, (ICM), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Département de radiologie, radio-oncologie et médecine nucléaire, Université de Montréal, Montreal, QC, Canada
| | - Philip Wong
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Institut du Cancer de Montréal, (ICM), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Jean-François Carrier
- Département de radiologie, radio-oncologie et médecine nucléaire, Université de Montréal, Montreal, QC, Canada
- Département de Physique, Université de Montréal, Montréal, QC, Canada
- Département de Radio-oncologie, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Yuji Kamio
- Département de Radio-oncologie, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Thomas Gervais
- μFO Lab, Polytechnique Montréal, Montréal, QC, Canada.
- Institut du Cancer de Montréal, (ICM), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| |
Collapse
|
40
|
Parsian M, Mutlu P, Yildirim E, Ildiz C, Ozen C, Gunduz U. Development of a microfluidic platform to maintain viability of micro-dissected tumor slices in culture. BIOMICROFLUIDICS 2022; 16:034103. [PMID: 35547184 PMCID: PMC9076128 DOI: 10.1063/5.0087532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/06/2022] [Indexed: 05/07/2023]
Abstract
One of the issues limiting the development of personalized medicine is the absence of realistic models that reflect the nature and complexity of tumor tissues. We described a new tissue culture approach that combines a microfluidic chip with the microdissected breast cancer tumor. "Tumor-on-a-chip" devices are suitable for precision medicine since the viability of tissue samples is maintained during the culture period by continuously feeding fresh media and eliminating metabolic wastes from the tissue. However, the mass transport of oxygen, which arguably is the most critical nutrient, is rarely assessed. According to our results, transportation of oxygen provides satisfactory in vivo oxygenation within the system. A high level of dissolved oxygen, around 98%-100% for every 24 h, was measurable in the outlet medium. The microfluidic chip system developed within the scope of this study allows living and testing tumor tissues under laboratory conditions. In this study, tumors were generated in CD-1 mice using MDA-MB-231 and SKBR-3 cell lines. Microdissected tumor tissues were cultured both in the newly developed microfluidic chip system and in conventional 24-well culture plates. Two systems were compared for two different types of tumors. The confocal microscopy analyses, lactate dehydrogenase release, and glucose consumption values showed that the tissues in the microfluidic system remained more viable with respect to the conventional well plate culturing method, up to 96 h. The new culturing technique described here may be superior to conventional culturing techniques for developing new treatment strategies, such as testing chemotherapeutics on tumor samples from individual patients.
Collapse
Affiliation(s)
- Maryam Parsian
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey
| | - Pelin Mutlu
- Department of Biotechnology, Ankara University, Ankara, Turkey
- Author to whom correspondence should be addressed:
| | - Ender Yildirim
- Department of Mechanical Engineering, Middle East Technical University, Ankara, Turkey
| | - Can Ildiz
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Can Ozen
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey
| | | |
Collapse
|
41
|
Azizipour N, Avazpour R, Sawan M, Ajji A, H Rosenzweig D. Surface Optimization and Design Adaptation toward Spheroid Formation On-Chip. SENSORS (BASEL, SWITZERLAND) 2022; 22:s22093191. [PMID: 35590879 DOI: 10.1039/d2sd00004k] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/07/2022] [Accepted: 04/19/2022] [Indexed: 05/27/2023]
Abstract
Spheroids have become an essential tool in preclinical cancer research. The uniformity of spheroids is a critical parameter in drug test results. Spheroids form by self-assembly of cells. Hence, the control of homogeneity of spheroids in terms of size, shape, and density is challenging. We developed surface-optimized polydimethylsiloxane (PDMS) biochip platforms for uniform spheroid formation on-chip. These biochips were surface modified with 10% bovine serum albumin (BSA) to effectively suppress cell adhesion on the PDMS surface. These surface-optimized platforms facilitate cell self-aggregations to produce homogenous non-scaffold-based spheroids. We produced uniform spheroids on these biochips using six different established human cell lines and a co-culture model. Here, we observe that the concentration of the BSA is important in blocking cell adhesion to the PDMS surfaces. Biochips treated with 3% BSA demonstrated cell repellent properties similar to the bare PDMS surfaces. This work highlights the importance of surface modification on spheroid production on PDMS-based microfluidic devices.
Collapse
Affiliation(s)
- Neda Azizipour
- Institut de Génie Biomédical, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada
| | - Rahi Avazpour
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada
| | - Mohamad Sawan
- Institut de Génie Biomédical, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada
- Polystim Neurotech Laboratory, Electrical Engineering Department, Polytechnique Montréal, Montréal, QC H3T 1J4, Canada
- CenBRAIN Laboratory, Westlake Institute for Advanced Study, School of Engineering, Westlake University, Hangzhou 310024, China
| | - Abdellah Ajji
- Institut de Génie Biomédical, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada
- The Research Center for High Performance Polymer and Composite Systems, Chemical Engineering Department, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada
| | - Derek H Rosenzweig
- Department of Surgery, McGill University, Montréal, QC H3G 1A4, Canada
- Injury, Repair and Recovery Program, Research Institute of McGill University Health Centre, Montréal, QC H3H 2R9, Canada
| |
Collapse
|
42
|
Azizipour N, Avazpour R, Sawan M, Ajji A, H. Rosenzweig D. Surface Optimization and Design Adaptation toward Spheroid Formation On-Chip. SENSORS (BASEL, SWITZERLAND) 2022; 22:3191. [PMID: 35590879 PMCID: PMC9104470 DOI: 10.3390/s22093191] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/07/2022] [Accepted: 04/19/2022] [Indexed: 12/17/2022]
Abstract
Spheroids have become an essential tool in preclinical cancer research. The uniformity of spheroids is a critical parameter in drug test results. Spheroids form by self-assembly of cells. Hence, the control of homogeneity of spheroids in terms of size, shape, and density is challenging. We developed surface-optimized polydimethylsiloxane (PDMS) biochip platforms for uniform spheroid formation on-chip. These biochips were surface modified with 10% bovine serum albumin (BSA) to effectively suppress cell adhesion on the PDMS surface. These surface-optimized platforms facilitate cell self-aggregations to produce homogenous non-scaffold-based spheroids. We produced uniform spheroids on these biochips using six different established human cell lines and a co-culture model. Here, we observe that the concentration of the BSA is important in blocking cell adhesion to the PDMS surfaces. Biochips treated with 3% BSA demonstrated cell repellent properties similar to the bare PDMS surfaces. This work highlights the importance of surface modification on spheroid production on PDMS-based microfluidic devices.
Collapse
Affiliation(s)
- Neda Azizipour
- Institut de Génie Biomédical, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada; (N.A.); (M.S.)
| | - Rahi Avazpour
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada;
| | - Mohamad Sawan
- Institut de Génie Biomédical, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada; (N.A.); (M.S.)
- Polystim Neurotech Laboratory, Electrical Engineering Department, Polytechnique Montréal, Montréal, QC H3T 1J4, Canada
- CenBRAIN Laboratory, Westlake Institute for Advanced Study, School of Engineering, Westlake University, Hangzhou 310024, China
| | - Abdellah Ajji
- Institut de Génie Biomédical, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada; (N.A.); (M.S.)
- The Research Center for High Performance Polymer and Composite Systems, Chemical Engineering Department, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada
| | - Derek H. Rosenzweig
- Department of Surgery, McGill University, Montréal, QC H3G 1A4, Canada
- Injury, Repair and Recovery Program, Research Institute of McGill University Health Centre, Montréal, QC H3H 2R9, Canada
| |
Collapse
|
43
|
Azizipour N, Avazpour R, Weber MH, Sawan M, Ajji A, Rosenzweig DH. Uniform Tumor Spheroids on Surface-Optimized Microfluidic Biochips for Reproducible Drug Screening and Personalized Medicine. MICROMACHINES 2022; 13:587. [PMID: 35457892 PMCID: PMC9028696 DOI: 10.3390/mi13040587] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 01/27/2023]
Abstract
Spheroids are recognized for resembling the important characteristics of natural tumors in cancer research. However, the lack of controllability of the spheroid size, form, and density in conventional spheroid culture methods reduces the reproducibility and precision of bioassay results and the assessment of drug-dose responses in spheroids. Nonetheless, the accurate prediction of cellular responses to drug compounds is crucial for developing new efficient therapeutic agents and optimizing existing therapeutic strategies for personalized medicine. We developed a surface-optimized PDMS microfluidic biochip to produce uniform and homogenous multicellular spheroids in a reproducible manner. This platform is surface optimized with 10% bovine serum albumin (BSA) to provide cell-repellent properties. Therefore, weak cell-surface interactions lead to the promotion of cell self-aggregations and the production of compact and uniform spheroids. We used a lung cancer cell line (A549), a co-culture model of lung cancer cells (A549) with (primary human osteoblasts, and patient-derived spine metastases cells (BML, bone metastasis secondary to lung). We observed that the behavior of cells cultured in three-dimensional (3D) spheroids within this biochip platform more closely reflects in vivo-like cellular responses to a chemotherapeutic drug, Doxorubicin, rather than on 24-well plates (two-dimensional (2D) model). It was also observed that the co-culture and patient-derived spheroids exhibited resistance to anti-cancer drugs more than the mono-culture spheroids. The repeatability of drug test results in this optimized platform is the hallmark of the reproducibility of uniform spheroids on a chip. This surface-optimized biochip can be a reliable platform to generate homogenous and uniform spheroids to study and monitor the tumor microenvironment and for drug screening.
Collapse
Affiliation(s)
- Neda Azizipour
- Institut de Génie Biomédical, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada
| | - Rahi Avazpour
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada
| | - Michael H Weber
- Department of Surgery, Division of Orthopaedic Surgery, McGill University, Montréal, QC H3G 1A4, Canada
- Injury, Repair and Recovery Program, Research Institute of McGill University Health Centre, Montréal, QC H3H 2R9, Canada
| | - Mohamad Sawan
- Institut de Génie Biomédical, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada
- Polystim Neurotech Laboratory, Electrical Engineering Department, Polytechnique Montréal, Montréal, QC H3T 1J4, Canada
- CenBRAIN Laboratory, School of Engineering, Westlake Institute for Advanced Study, Westlake University, Hangzhou 310024, China
| | - Abdellah Ajji
- Institut de Génie Biomédical, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada
- NSERC-Industry Chair, CREPEC, Chemical Engineering Department, Polytechnique Montréal, Montréal, QC H3C 3A7, Canada
| | - Derek H Rosenzweig
- Department of Surgery, Division of Orthopaedic Surgery, McGill University, Montréal, QC H3G 1A4, Canada
- Injury, Repair and Recovery Program, Research Institute of McGill University Health Centre, Montréal, QC H3H 2R9, Canada
| |
Collapse
|
44
|
Quintard C, Tubbs E, Achard JL, Navarro F, Gidrol X, Fouillet Y. Microfluidic device integrating a network of hyper-elastic valves for automated glucose stimulation and insulin secretion collection from a single pancreatic islet. Biosens Bioelectron 2022; 202:113967. [DOI: 10.1016/j.bios.2022.113967] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/22/2021] [Accepted: 01/03/2022] [Indexed: 01/15/2023]
|
45
|
Cahuzac M, Langlois P, Péant B, Fleury H, Mes-Masson AM, Saad F. Pre-activation of autophagy impacts response to olaparib in prostate cancer cells. Commun Biol 2022; 5:251. [PMID: 35318456 PMCID: PMC8940895 DOI: 10.1038/s42003-022-03210-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 03/01/2022] [Indexed: 01/01/2023] Open
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) plays an essential role in DNA repair and is targeted by anticancer therapies using PARP inhibitors (PARPi) such as olaparib. PARPi treatment in prostate cancer (PC) is currently used as a monotherapy or in combination with standard therapies (hormonotherapy) in clinical trials for patients with DNA damage response mutation. Unfortunately, 20% of these patients did not respond to this new treatment. This resistance mechanism in PC is still not well understood. Here, we report that autophagy affects differently the response of PC cell lines to olaparib depending on its activation status. Pre-activation of autophagy before olaparib resulted in an increase of DNA repair activity by homologous recombination (HR) to repair double-strand breaks induced by olaparib and enhanced cell proliferation. When autophagy was activated after olaparib treatment, or completely inhibited, PC cells demonstrated an increased sensitivity to this PARPi. This autophagy-mediated resistance is, in part, regulated by the nuclear localization of sequestrosome 1 (SQSTM1/p62). Decrease of SQSTM1/p62 nuclear localization due to autophagy pre-activation leads to an increase of filamin A (FLNA) protein expression and BRCA1/Rad51 recruitment involved in the HR pathway. Our results reveal that autophagy basal levels may in part determine amenability to PARPi treatment. Pre-activation of autophagy mediates resistance to olaparib by decreasing nuclear SQSTM1/p62, which increases homologous recombination-mediated repair through filamin A expression and BRCA1/Rad51 recruitment.
Collapse
Affiliation(s)
- Maxime Cahuzac
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal, Montreal, QC, Canada
| | - Patricia Langlois
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal, Montreal, QC, Canada
| | - Benjamin Péant
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal, Montreal, QC, Canada
| | - Hubert Fleury
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal, Montreal, QC, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada. .,Institut du cancer de Montréal, Montreal, QC, Canada. .,Department of Surgery, Université de Montréal, Montreal, QC, Canada.
| | - Fred Saad
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal, Montreal, QC, Canada.,Department of Surgery, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
46
|
Dsouza VL, Kuthethur R, Kabekkodu SP, Chakrabarty S. Organ-on-Chip platforms to study tumor evolution and chemosensitivity. Biochim Biophys Acta Rev Cancer 2022; 1877:188717. [PMID: 35304293 DOI: 10.1016/j.bbcan.2022.188717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
Abstract
Despite tremendous advancements in oncology research and therapeutics, cancer remains a primary cause of death worldwide. One of the significant factors in this critical challenge is a precise diagnosis and limited knowledge on how the tumor microenvironment (TME) behaves to the treatment and its role in chemo-resistance. Therefore, it is critical to understand the contribution of a heterogeneous TME in cancer drug response in individual patients for effective therapy management. Micro-physiological systems along with tissue engineering have facilitated the development of more physiologically relevant platforms, known as Organ-on-Chips (OoC). OoC platforms recapitulate the critical hallmarks of the TME in vitro and subsequently abet in sensitivity and efficacy testing of anti-cancer drugs before clinical trials. The OoC platforms incorporating conventional in vitro models enable researchers to control the cellular, molecular, chemical, and biophysical parameters of the TME in precise combinations while analyzing how they contribute to tumor progression and therapy response. This review discusses the application of OoC platforms integrated with conventional 2D cell lines, 3D organoids and spheroid models, and the organotypic tissue slices, including patient-derived and xenograft tumor slice cultures in cancer treatment responses. We summarize the relevance and drawbacks of conventional in vitro models in assessing cancer treatment response, challenges and limitations associated with OoC models, and future opportunities enabled by the OoC technologies towards developing personalized cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Venzil Lavie Dsouza
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Raviprasad Kuthethur
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
47
|
Chakrabarty S, Quiros-Solano WF, Kuijten MM, Haspels B, Mallya S, Lo CSY, Othman A, Silvestri C, van de Stolpe A, Gaio N, Odijk H, van de Ven M, de Ridder CM, van Weerden WM, Jonkers J, Dekker R, Taneja N, Kanaar R, van Gent DC. A Microfluidic Cancer-on-Chip Platform Predicts Drug Response Using Organotypic Tumor Slice Culture. Cancer Res 2022; 82:510-520. [PMID: 34872965 PMCID: PMC9397621 DOI: 10.1158/0008-5472.can-21-0799] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/31/2021] [Accepted: 11/30/2021] [Indexed: 01/07/2023]
Abstract
Optimal treatment of cancer requires diagnostic methods to facilitate therapy choice and prevent ineffective treatments. Direct assessment of therapy response in viable tumor specimens could fill this diagnostic gap. Therefore, we designed a microfluidic platform for assessment of patient treatment response using tumor tissue slices under precisely controlled growth conditions. The optimized Cancer-on-Chip (CoC) platform maintained viability and sustained proliferation of breast and prostate tumor slices for 7 days. No major changes in tissue morphology or gene expression patterns were observed within this time frame, suggesting that the CoC system provides a reliable and effective way to probe intrinsic chemotherapeutic sensitivity of tumors. The customized CoC platform accurately predicted cisplatin and apalutamide treatment response in breast and prostate tumor xenograft models, respectively. The culture period for breast cancer could be extended up to 14 days without major changes in tissue morphology and viability. These culture characteristics enable assessment of treatment outcomes and open possibilities for detailed mechanistic studies. SIGNIFICANCE: The Cancer-on-Chip platform with a 6-well plate design incorporating silicon-based microfluidics can enable optimal patient-specific treatment strategies through parallel culture of multiple tumor slices and diagnostic assays using primary tumor material.
Collapse
Affiliation(s)
- Sanjiban Chakrabarty
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - William F. Quiros-Solano
- Department of Microelectronics, Electronic Components, Technology and Materials, Delft University of Technology, Delft, the Netherlands.,BIOND Solutions B.V., Delft, the Netherlands
| | - Maayke M.P. Kuijten
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands.,Oncode Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ben Haspels
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sandeep Mallya
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Calvin Shun Yu Lo
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Amr Othman
- BIOND Solutions B.V., Delft, the Netherlands
| | | | | | | | - Hanny Odijk
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marieke van de Ven
- Preclinical Intervention Unit, Mouse Clinic for Cancer and Ageing, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Corrina M.A. de Ridder
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Wytske M. van Weerden
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jos Jonkers
- Preclinical Intervention Unit, Mouse Clinic for Cancer and Ageing, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ronald Dekker
- Department of Microelectronics, Electronic Components, Technology and Materials, Delft University of Technology, Delft, the Netherlands.,Philips Research, Eindhoven, the Netherlands
| | - Nitika Taneja
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Roland Kanaar
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands.,Oncode Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Dik C. van Gent
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands.,Oncode Institute, Erasmus University Medical Center, Rotterdam, the Netherlands.,Corresponding Author: Dik C. van Gent, Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, Rotterdam 3015GD, the Netherlands. Phone: 31-10-7043932; E-mail:
| |
Collapse
|
48
|
Micro/nanofluidic devices for drug delivery. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 187:9-39. [PMID: 35094782 DOI: 10.1016/bs.pmbts.2021.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Micro/nanofluidic drug delivery systems have attracted significant attention as they offer unique advantages in targeted and controlled drug delivery. Based on the desired application, these systems can be categorized into three different groups: in vitro, in situ and in vivo microfluidic drug delivery platforms. In vitro microfluidic drug delivery platforms are closely linked with the emerging concept of lab-on-a-chip for cell culture studies. These systems can be used to administer drugs or therapeutic agents, mostly at the cellular or tissue level, to find the therapeutic index and can potentially be used for personalized medicine. In situ and in vivo microfluidic drug delivery platforms are still at the developmental stage and can be used for drug delivery at tissue or organ levels. A famous example of these systems are microneedles that can be used for painless and controllable delivery of drugs or vaccines through human skin. This chapter presents the cutting edge advances in the design and fabrication of in vitro microfluidic drug delivery systems that can be used for both cellular and tissue drug delivery. It also briefly discusses the in situ drug delivery platforms using microneedles.
Collapse
|
49
|
Lafontaine J, Refet-Mollof E, Najyb O, Gervais T, Wong P. Cell Death Analysis in Cancer Spheroids from a Microfluidic Device. Methods Mol Biol 2022; 2543:13-25. [PMID: 36087255 DOI: 10.1007/978-1-0716-2553-8_2] [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] [Indexed: 06/15/2023]
Abstract
Microfluidic technology facilitates the generation of 3D spheroids from cancer cells, a more suitable model for preclinical therapeutic studies. This system opens the possibility to test many drugs combination at a low cost. Here we describe the use of microfluidic devices for cytotoxicity evaluation on cancer spheroids for the discovery of drugs that could be used in combination with radiotherapy. Device fabrication, preparation, and seeding are also covered. Cell death arising following treatment is detected and characterized according to spheroid size, colony formation assays, and flow cytometry analysis of apoptotic marker annexin V.
Collapse
Affiliation(s)
- Julie Lafontaine
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Elena Refet-Mollof
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Institute of Biomedical Engineering, École Polytechnique de MontrÉal, Montréal, QC, Canada
| | - Ouafa Najyb
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Thomas Gervais
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Institute of Biomedical Engineering, École Polytechnique de MontrÉal, Montréal, QC, Canada
- Department of Engineering Physics, École Polytechnique de MontrÉal, Montréal, Canada
| | - Philip Wong
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.
- Département de Radio-Oncologie, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON, Canada.
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
50
|
Francés-Herrero E, Lopez R, Hellström M, de Miguel-Gómez L, Herraiz S, Brännström M, Pellicer A, Cervelló I. OUP accepted manuscript. Hum Reprod Update 2022; 28:798-837. [PMID: 35652272 PMCID: PMC9629485 DOI: 10.1093/humupd/dmac025] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 04/13/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND To provide the optimal milieu for implantation and fetal development, the female reproductive system must orchestrate uterine dynamics with the appropriate hormones produced by the ovaries. Mature oocytes may be fertilized in the fallopian tubes, and the resulting zygote is transported toward the uterus, where it can implant and continue developing. The cervix acts as a physical barrier to protect the fetus throughout pregnancy, and the vagina acts as a birth canal (involving uterine and cervix mechanisms) and facilitates copulation. Fertility can be compromised by pathologies that affect any of these organs or processes, and therefore, being able to accurately model them or restore their function is of paramount importance in applied and translational research. However, innate differences in human and animal model reproductive tracts, and the static nature of 2D cell/tissue culture techniques, necessitate continued research and development of dynamic and more complex in vitro platforms, ex vivo approaches and in vivo therapies to study and support reproductive biology. To meet this need, bioengineering is propelling the research on female reproduction into a new dimension through a wide range of potential applications and preclinical models, and the burgeoning number and variety of studies makes for a rapidly changing state of the field. OBJECTIVE AND RATIONALE This review aims to summarize the mounting evidence on bioengineering strategies, platforms and therapies currently available and under development in the context of female reproductive medicine, in order to further understand female reproductive biology and provide new options for fertility restoration. Specifically, techniques used in, or for, the uterus (endometrium and myometrium), ovary, fallopian tubes, cervix and vagina will be discussed. SEARCH METHODS A systematic search of full-text articles available in PubMed and Embase databases was conducted to identify relevant studies published between January 2000 and September 2021. The search terms included: bioengineering, reproduction, artificial, biomaterial, microfluidic, bioprinting, organoid, hydrogel, scaffold, uterus, endometrium, ovary, fallopian tubes, oviduct, cervix, vagina, endometriosis, adenomyosis, uterine fibroids, chlamydia, Asherman’s syndrome, intrauterine adhesions, uterine polyps, polycystic ovary syndrome and primary ovarian insufficiency. Additional studies were identified by manually searching the references of the selected articles and of complementary reviews. Eligibility criteria included original, rigorous and accessible peer-reviewed work, published in English, on female reproductive bioengineering techniques in preclinical (in vitro/in vivo/ex vivo) and/or clinical testing phases. OUTCOMES Out of the 10 390 records identified, 312 studies were included for systematic review. Owing to inconsistencies in the study measurements and designs, the findings were assessed qualitatively rather than by meta-analysis. Hydrogels and scaffolds were commonly applied in various bioengineering-related studies of the female reproductive tract. Emerging technologies, such as organoids and bioprinting, offered personalized diagnoses and alternative treatment options, respectively. Promising microfluidic systems combining various bioengineering approaches have also shown translational value. WIDER IMPLICATIONS The complexity of the molecular, endocrine and tissue-level interactions regulating female reproduction present challenges for bioengineering approaches to replace female reproductive organs. However, interdisciplinary work is providing valuable insight into the physicochemical properties necessary for reproductive biological processes to occur. Defining the landscape of reproductive bioengineering technologies currently available and under development for women can provide alternative models for toxicology/drug testing, ex vivo fertility options, clinical therapies and a basis for future organ regeneration studies.
Collapse
Affiliation(s)
| | | | - Mats Hellström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lucía de Miguel-Gómez
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain
- Fundación IVI, IVI-RMA Global, Valencia, Spain
| | - Sonia Herraiz
- Fundación IVI, IVI-RMA Global, Valencia, Spain
- Reproductive Medicine Research Group, IIS La Fe, Valencia, Spain
| | - Mats Brännström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Stockholm IVF-EUGIN, Stockholm, Sweden
| | - Antonio Pellicer
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain
- IVI Roma Parioli, IVI-RMA Global, Rome, Italy
| | | |
Collapse
|