1
|
Peng Z, Lv X, Sun H, Zhao L, Huang S. 3D tumor cultures for drug resistance and screening development in clinical applications. Mol Cancer 2025; 24:93. [PMID: 40119343 PMCID: PMC11927140 DOI: 10.1186/s12943-025-02281-2] [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: 01/25/2025] [Accepted: 02/24/2025] [Indexed: 03/24/2025] Open
Abstract
Tumor drug resistance presents a growing challenge in medical practice, particularly during anti-cancer therapies, where the emergence of drug-resistant cancer cells significantly complicates clinical treatment. In recent years, three-dimensional (3D) tumor culture technology, which more effectively simulates the in vivo physiological environment, has gained increasing attention in tumor drug resistance research and clinical applications. By mimicking the in vivo cellular microenvironment, 3D tumor culture technology not only recapitulates cell-cell interactions but also more faithfully reproduces the biological effects of therapeutic agents. Consequently, 3D tumor culture technology is emerging as a crucial tool in biomedical and clinical research. We summarize the benefits of 3D culture models and organoid technology, explore their application in the realm of drug resistance, drug screening, and personalized therapy, and discuss their potential application prospects and challenges in clinical transformation, with the aim of providing insights for optimizing cancer treatment strategies and advancing precision therapy.
Collapse
Affiliation(s)
- Zheng Peng
- Department of Clinical Laboratory, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, Guangxi, China
| | - Xiaolan Lv
- Department of Clinical Laboratory, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Hao Sun
- Faculty of Science, Autonomous University of Madrid, Spainish National Research Council -Consejo Superior de Investigaciones Científicas,(UAM-CSIC), Madrid, 28049, Spain
| | - Lina Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Shigao Huang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
2
|
Tripathi S, Sharma Y, Kumar D. Unveiling the link between chronic inflammation and cancer. Metabol Open 2025; 25:100347. [PMID: 39876904 PMCID: PMC11772974 DOI: 10.1016/j.metop.2025.100347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
The highly nuanced transition from an inflammatory process to tumorigenesis is of great scientific interest. While it is well known that environmental stimuli can cause inflammation, less is known about the oncogenic modifications that chronic inflammation in the tissue microenvironment can bring about, as well as how these modifications can set off pro-tumorigenic processes. It is clear that no matter where the environmental factors come from, maintaining an inflammatory microenvironment encourages carcinogenesis. In addition to encouraging angiogenesis and metastatic processes, sustaining the survival and proliferation of malignant transformed cells, and possibly altering the efficacy of therapeutic agents, inflammation can negatively regulate the antitumoral adaptive and innate immune responses. Because chronic inflammation has multiple pathways involved in tumorigenesis and metastasis, it has gained recognition as a marker of cancer and a desirable target for cancer therapy. Recent advances in our knowledge of the molecular mechanisms that drive cancer's progression demonstrate that inflammation promotes tumorigenesis and metastasis while suppressing anti-tumor immunity. In many solid tumor types, including breast, lung, and liver cancer, inflammation stimulates the activation of oncogenes and impairs the body's defenses against the tumor. Additionally, it alters the microenvironment of the tumor. As a tactical approach to cancer treatment, these findings have underscored the importance of targeting inflammatory pathways. This review highlights the role of inflammation in cancer development and metastasis, focusing on its impact on tumor progression, immune suppression, and therapy resistance. It examines current anti-inflammatory strategies, including NSAIDs, cytokine modulators, and STAT3 inhibitors, while addressing their potential and limitations. The review emphasizes the need for further research to unravel the complex mechanisms linking inflammation to cancer progression and identify molecular targets for specific cancer subtypes.
Collapse
Affiliation(s)
- Siddhant Tripathi
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Yashika Sharma
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Dileep Kumar
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
3
|
Högström JM, Muranen T. An Optimized Protocol for Simultaneous Propagation of Patient-derived Organoids and Matching CAFs. Bio Protoc 2025; 15:e5160. [PMID: 39872717 PMCID: PMC11769750 DOI: 10.21769/bioprotoc.5160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/03/2024] [Accepted: 11/05/2024] [Indexed: 01/30/2025] Open
Abstract
Recurrent hormone receptor-positive (HR+) breast cancer is a leading cause of cancer mortality in women. Recurrence and resistance to targeted therapies have been difficult to study due to the long clinical course of the disease, the complex nature of resistance, and the lack of clinically relevant model systems. Existing models are limited to a few HR+ cell lines, organoid models, and patient-derived xenograft models, all lacking components of the human tumor microenvironment. Furthermore, the low take rate and loss of estrogen receptor (ER) expression in patient-derived organoids (PDOs) has been challenging. Our protocol allows simultaneous isolation of PDOs and matching cancer-associated fibroblasts (CAFs) from primary and metastatic HR+ breast cancers. Importantly, our protocol has a higher take rate and enables long-term culturing of PDOs that retain ER expression. Our matching PDOs and CAFs will provide researchers with a new resource to study the influence of the tumor microenvironment on various aspects of cancer biology such as cell growth and drug resistance in HR+ breast cancer. Key features • Propagation of patient-derived organoids and matching cancer-associated fibroblasts from primary and metastatic hormone receptor (HR+) positive breast cancer. • Optimized media for long-term culturing of HR+ organoids from primary tumors and bone metastasis. • Co-culture model to assess the influence of the tumor stroma on breast cancer progression.
Collapse
Affiliation(s)
- Jenny M. Högström
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Taru Muranen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Vitacolonna M, Bruch R, Schneider R, Jabs J, Hafner M, Reischl M, Rudolf R. A spheroid whole mount drug testing pipeline with machine-learning based image analysis identifies cell-type specific differences in drug efficacy on a single-cell level. BMC Cancer 2024; 24:1542. [PMID: 39696122 DOI: 10.1186/s12885-024-13329-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The growth and drug response of tumors are influenced by their stromal composition, both in vivo and 3D-cell culture models. Cell-type inherent features as well as mutual relationships between the different cell types in a tumor might affect drug susceptibility of the tumor as a whole and/or of its cell populations. However, a lack of single-cell procedures with sufficient detail has hampered the automated observation of cell-type-specific effects in three-dimensional stroma-tumor cell co-cultures. METHODS Here, we developed a high-content pipeline ranging from the setup of novel tumor-fibroblast spheroid co-cultures over optical tissue clearing, whole mount staining, and 3D confocal microscopy to optimized 3D-image segmentation and a 3D-deep-learning model to automate the analysis of a range of cell-type-specific processes, such as cell proliferation, apoptosis, necrosis, drug susceptibility, nuclear morphology, and cell density. RESULTS This demonstrated that co-cultures of KP-4 tumor cells with CCD-1137Sk fibroblasts exhibited a growth advantage compared to tumor cell mono-cultures, resulting in higher cell counts following cytostatic treatments with paclitaxel and doxorubicin. However, cell-type-specific single-cell analysis revealed that this apparent benefit of co-cultures was due to a higher resilience of fibroblasts against the drugs and did not indicate a higher drug resistance of the KP-4 cancer cells during co-culture. Conversely, cancer cells were partially even more susceptible in the presence of fibroblasts than in mono-cultures. CONCLUSION In summary, this underlines that a novel cell-type-specific single-cell analysis method can reveal critical insights regarding the mechanism of action of drug substances in three-dimensional cell culture models.
Collapse
Affiliation(s)
- Mario Vitacolonna
- CeMOS, Mannheim University of Applied Sciences, 68163, Mannheim, Germany.
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany.
| | - Roman Bruch
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, 76344, Eggen-stein-Leopoldshafen, Germany
| | | | - Julia Jabs
- Merck Healthcare KGaA, 64293, Darmstadt, Germany
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
- Institute of Medical Technology, Medical Faculty Mannheim of Heidelberg University, Mannheim University of Applied Sciences, 68167, Mannheim, Germany
| | - Markus Reischl
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, 76344, Eggen-stein-Leopoldshafen, Germany
| | - Rüdiger Rudolf
- CeMOS, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
| |
Collapse
|
5
|
Domenici G, Lopes NF, Trindade G, Ramella Gal I, Miret Minard J, Rebelo SP, Freitas C, Duarte N, Brito C. Assessing Novel Antibody-Based Therapies in Reconstructive 3D Cell Models of the Tumor Microenvironment. Adv Biol (Weinh) 2024; 8:e2400431. [PMID: 39601467 DOI: 10.1002/adbi.202400431] [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: 07/23/2024] [Revised: 10/21/2024] [Indexed: 11/29/2024]
Abstract
Targeted, combinatorial, and immunomodulatory therapies, such as antibody-drug conjugates (ADCs) and immunomodulatory antibodies (Abs), are powerful weapons against tumor cells and immune cells within the tumor microenvironment (TME). Therefore, the evaluation of such therapies should be conducted in pre-clinical models able to recapitulate the complex cellular and molecular crosstalk of the TME. To build-in critical hallmarks of the TME, a breast cancer heterotypic 3D cell model (3D-3) is devised using a microencapsulation strategy with an inert biomaterial (alginate) and agitation-based cultures. Both stromal and immune components are added to multicellular tumor spheroids, therefore fostering cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) immunomodulatory interactions. The potential of the methodology to assess Ab-based therapies is then addressed by employing a series of anti-HER2-based ADCs. ADCs induced tumor-cell specific cytotoxicity toward HER2+ breast cancer spheroids while sparing HER2-negative CAFs. In addition, an immunomodulatory blocking Ab against colony-stimulating factor 1 receptor (CSF1R) decreases the expression of immunosuppressive and anti-inflammatory markers in TAMs, like what is previously observed upon in vivo α-CSF1R administration. Collectively, the human TME-based 3D-3 cell model is a suitable tool to evaluate the anti-tumor and immunomodulatory potential of novel antibody-based therapies directed against TME targets, such as cancer cells and macrophages.
Collapse
Affiliation(s)
- Giacomo Domenici
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| | - Nuno F Lopes
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| | - Gonçalo Trindade
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| | - Isabella Ramella Gal
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| | - Joan Miret Minard
- Department of Chemical, Biological and Environmental Engineering, Universitat Autònoma de Barcelona, Plaça Cívica, Bellaterra, 08193, Spain
| | - Sofia P Rebelo
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
| | - Catarina Freitas
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| | - Nádia Duarte
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| |
Collapse
|
6
|
Jiang RY, Zhu JY, Zhang HP, Yu Y, Dong ZX, Zhou HH, Wang X. STAT3: Key targets of growth-promoting receptor positive breast cancer. Cancer Cell Int 2024; 24:356. [PMID: 39468521 PMCID: PMC11520424 DOI: 10.1186/s12935-024-03541-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
Breast cancer has become the malignant tumor with the first incidence and the second mortality among female cancers. Most female breast cancers belong to luminal-type breast cancer and HER2-positive breast cancer. These breast cancer cells all have different driving genes, which constantly promote the proliferation and metastasis of breast cancer cells. Signal transducer and activator of transcription 3 (STAT3) is an important breast cancer-related gene, which can promote the progress of breast cancer. It has been proved in clinical and basic research that over-expressed and constitutively activated STAT3 is involved in the progress, proliferation, metastasis and chemotherapy resistance of breast cancer. STAT3 is an important key target in luminal-type breast cancer and HER2-positive cancer, which has an important impact on the curative effect of related treatments. In breast cancer, the activation of STAT3 will change the spatial position of STAT3 protein and cause different phenotypic changes of breast cancer cells. In the current basic research and clinical research, small molecule inhibitors activated by targeting STAT3 can effectively treat breast cancer, and enhance the efficacy level of related treatment methods for luminal-type and HER2-positive breast cancers.
Collapse
Affiliation(s)
- Rui-Yuan Jiang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Jia-Yu Zhu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Huan-Ping Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Department of Graduate Student, Wenzhou Medical University, No.270, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China
| | - Yuan Yu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Zhi-Xin Dong
- Department of Oncology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No.89-9, Dongge Road, Qingxiu District, Nanning, 530000, Guangxi, China
| | - Huan-Huan Zhou
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China.
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Xiaojia Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
7
|
Poskus MD, McDonald J, Laird M, Li R, Norcoss K, Zervantonakis IK. Rational Design of HER2-Targeted Combination Therapies to Reverse Drug Resistance in Fibroblast-Protected HER2+ Breast Cancer Cells. Cell Mol Bioeng 2024; 17:491-506. [PMID: 39513002 PMCID: PMC11538110 DOI: 10.1007/s12195-024-00823-0] [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: 03/30/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction Fibroblasts, an abundant cell type in the breast tumor microenvironment, interact with cancer cells and orchestrate tumor progression and drug resistance. However, the mechanisms by which fibroblast-derived factors impact drug sensitivity remain poorly understood. Here, we develop rational combination therapies that are informed by proteomic profiling to overcome fibroblast-mediated therapeutic resistance in HER2+ breast cancer cells. Methods Drug sensitivity to the HER2 kinase inhibitor lapatinib was characterized under conditions of monoculture and exposure to breast fibroblast-conditioned medium. Protein expression was measured using reverse phase protein arrays. Candidate targets for combination therapy were identified using differential expression and multivariate regression modeling. Follow-up experiments were performed to evaluate the effects of HER2 kinase combination therapies in fibroblast-protected cancer cell lines and fibroblasts. Results Compared to monoculture, fibroblast-conditioned medium increased the expression of plasminogen activator inhibitor-1 (PAI1) and cell cycle regulator polo like kinase 1 (PLK1) in lapatinib-treated breast cancer cells. Combination therapy of lapatinib with inhibitors targeting either PAI1 or PLK1, eliminated fibroblast-protected cancer cells, under both conditions of direct coculture with fibroblasts and protection by fibroblast-conditioned medium. Analysis of publicly available, clinical transcriptomic datasets revealed that HER2-targeted therapy fails to suppress PLK1 expression in stroma-rich HER2+ breast tumors and that high PAI1 gene expression associates with high stroma density. Furthermore, we showed that an epigenetics-directed approach using a bromodomain and extraterminal inhibitor to globally target fibroblast-induced proteomic adaptions in cancer cells, also restored lapatinib sensitivity. Conclusions Our data-driven framework of proteomic profiling in breast cancer cells identified the proteolytic degradation regulator PAI1 and the cell cycle regulator PLK1 as predictors of fibroblast-mediated treatment resistance. Combination therapies targeting HER2 kinase and these fibroblast-induced signaling adaptations eliminates fibroblast-protected HER2+ breast cancer cells. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00823-0.
Collapse
Affiliation(s)
- Matthew D. Poskus
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA USA
| | - Jacob McDonald
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA USA
| | - Matthew Laird
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA USA
| | - Ruxuan Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA USA
| | - Kyle Norcoss
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA USA
| | - Ioannis K. Zervantonakis
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA USA
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA USA
| |
Collapse
|
8
|
Alekseenko I, Zhukova L, Kondratyeva L, Buzdin A, Chernov I, Sverdlov E. Tumor Cell Communications as Promising Supramolecular Targets for Cancer Chemotherapy: A Possible Strategy. Int J Mol Sci 2024; 25:10454. [PMID: 39408784 PMCID: PMC11476449 DOI: 10.3390/ijms251910454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Fifty-two years have passed since President Nixon launched the "War on Cancer". Despite unparalleled efforts and funds allocated worldwide, the outlined goals were not achieved because cancer treatment approaches such as chemotherapy, radiation therapy, hormonal and targeted therapies have not fully met the expectations. Based on the recent literature, a new direction in cancer therapy can be proposed which targets connections between cancer cells and their microenvironment by chemical means. Cancer-stromal synapses such as immunological synapses between cancer and immune cells provide an attractive target for this approach. Such synapses form ligand-receptor clusters on the interface of the interacting cells. They share a common property of involving intercellular clusters of spatially proximate and cooperatively acting proteins. Synapses provide the space for the focused intercellular signaling molecules exchange. Thus, the disassembly of cancer-stromal synapses may potentially cause the collapse of various tumors. Additionally, the clustered arrangement of synapse components offers opportunities to enhance treatment safety and precision by using targeted crosslinking chemical agents which may inactivate cancer synapses even in reduced concentrations. Furthermore, attaching a cleavable cell-permeable toxic agent(s) to a crosslinker may further enhance the anti-cancer effect of such therapeutics. The highlighted approach promises to be universal, relatively simple and cost-efficient. We also hope that, unlike chemotherapeutic and immune drugs that interact with a single target, by using supramolecular large clusters that include many different components as a target, the emergence of a resistance characteristic of chemo- and immunotherapy is extremely unlikely.
Collapse
Affiliation(s)
- Irina Alekseenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | - Lyudmila Zhukova
- Department of Oncology, SBIH “Moscow Clinical Scientific and Practical Center Named After A.S. Loginov” DHM, 111123 Moscow, Russia;
| | - Liya Kondratyeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119992 Moscow, Russia
- Oncobox LLC, 121205 Moscow, Russia
| | - Igor Chernov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
| | - Eugene Sverdlov
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| |
Collapse
|
9
|
Miti T, Desai B, Miroshnychenko D, Basanta D, Marusyk A. Dissecting the Spatially Restricted Effects of Microenvironment-Mediated Resistance on Targeted Therapy Responses. Cancers (Basel) 2024; 16:2405. [PMID: 39001467 PMCID: PMC11240540 DOI: 10.3390/cancers16132405] [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: 05/09/2024] [Revised: 06/16/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
The response of tumors to anti-cancer therapies is defined not only by cell-intrinsic therapy sensitivities but also by local interactions with the tumor microenvironment. Fibroblasts that make tumor stroma have been shown to produce paracrine factors that can strongly reduce the sensitivity of tumor cells to many types of targeted therapies. Moreover, a high stroma/tumor ratio is generally associated with poor survival and reduced therapy responses. However, in contrast to advanced knowledge of the molecular mechanisms responsible for stroma-mediated resistance, its effect on the ability of tumors to escape therapeutic eradication remains poorly understood. To a large extent, this gap of knowledge reflects the challenge of accounting for the spatial aspects of microenvironmental resistance, especially over longer time frames. To address this problem, we integrated spatial inferences of proliferation-death dynamics from an experimental animal model of targeted therapy responses with spatial mathematical modeling. With this approach, we dissected the impact of tumor/stroma distribution, magnitude and distance of stromal effects. While all of the tested parameters affected the ability of tumor cells to resist elimination, spatial patterns of stroma distribution within tumor tissue had a particularly strong impact.
Collapse
Affiliation(s)
- Tatiana Miti
- Department of Integrative Mathematical Oncology, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA;
| | - Bina Desai
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA (D.M.)
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33620, USA
| | - Daria Miroshnychenko
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA (D.M.)
| | - David Basanta
- Department of Integrative Mathematical Oncology, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA;
| | - Andriy Marusyk
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA (D.M.)
| |
Collapse
|
10
|
Poskus MD, McDonald J, Laird M, Li R, Norcoss K, Zervantonakis IK. Rational design of HER2-targeted combination therapies to reverse drug resistance in fibroblast-protected HER2+ breast cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.18.594826. [PMID: 38798591 PMCID: PMC11118562 DOI: 10.1101/2024.05.18.594826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Introduction Fibroblasts, an abundant cell type in the breast tumor microenvironment, interact with cancer cells and orchestrate tumor progression and drug resistance. However, the mechanisms by which fibroblast-derived factors impact drug sensitivity remain poorly understood. Here, we develop rational combination therapies that are informed by proteomic profiling to overcome fibroblast-mediated therapeutic resistance in HER2+ breast cancer cells. Methods Drug sensitivity to the HER2 kinase inhibitor lapatinib was characterized under conditions of monoculture and exposure to breast fibroblast-conditioned medium. Protein expression was measured using reverse phase protein arrays. Candidate targets for combination therapy were identified using differential expression and multivariate regression modeling. Follow-up experiments were performed to evaluate the effects of HER2 kinase combination therapies in fibroblast-protected cancer cell lines and fibroblasts. Results Compared to monoculture, fibroblast-conditioned medium increased the expression of plasminogen activator inhibitor-1 (PAI1) and cell cycle regulator polo like kinase 1 (PLK1) in lapatinib-treated breast cancer cells. Combination therapy of lapatinib with inhibitors targeting either PAI1 or PLK1, eliminated fibroblast-protected cancer cells, under both conditions of direct coculture with fibroblasts and protection by fibroblast-conditioned medium. Analysis of publicly available, clinical transcriptomic datasets revealed that HER2-targeted therapy fails to suppress PLK1 expression in stroma-rich HER2+ breast tumors and that high PAI1 gene expression associates with high stroma density. Furthermore, we showed that an epigenetics-directed approach using a bromodomain and extraterminal inhibitor to globally target fibroblast-induced proteomic adaptions in cancer cells, also restored lapatinib sensitivity. Conclusions Our data-driven framework of proteomic profiling in breast cancer cells identified the proteolytic degradation regulator PAI1 and the cell cycle regulator PLK1 as predictors of fibroblast-mediated treatment resistance. Combination therapies targeting HER2 kinase and these fibroblast-induced signaling adaptations eliminates fibroblast-protected HER2+ breast cancer cells.
Collapse
|
11
|
Goyette MA, Stevens LE, DePinho CR, Seehawer M, Nishida J, Li Z, Wilde CM, Li R, Qiu X, Pyke AL, Zhao S, Lim K, Tender GS, Northey JJ, Riley NM, Long HW, Bertozzi CR, Weaver VM, Polyak K. Cancer-stromal cell interactions in breast cancer brain metastases induce glycocalyx-mediated resistance to HER2-targeting therapies. Proc Natl Acad Sci U S A 2024; 121:e2322688121. [PMID: 38709925 PMCID: PMC11098130 DOI: 10.1073/pnas.2322688121] [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: 12/22/2023] [Accepted: 03/27/2024] [Indexed: 05/08/2024] Open
Abstract
Brain metastatic breast cancer is particularly lethal largely due to therapeutic resistance. Almost half of the patients with metastatic HER2-positive breast cancer develop brain metastases, representing a major clinical challenge. We previously described that cancer-associated fibroblasts are an important source of resistance in primary tumors. Here, we report that breast cancer brain metastasis stromal cell interactions in 3D cocultures induce therapeutic resistance to HER2-targeting agents, particularly to the small molecule inhibitor of HER2/EGFR neratinib. We investigated the underlying mechanisms using a synthetic Notch reporter system enabling the sorting of cancer cells that directly interact with stromal cells. We identified mucins and bulky glycoprotein synthesis as top-up-regulated genes and pathways by comparing the gene expression and chromatin profiles of stroma-contact and no-contact cancer cells before and after neratinib treatment. Glycoprotein gene signatures were also enriched in human brain metastases compared to primary tumors. We confirmed increased glycocalyx surrounding cocultures by immunofluorescence and showed that mucinase treatment increased sensitivity to neratinib by enabling a more efficient inhibition of EGFR/HER2 signaling in cancer cells. Overexpression of truncated MUC1 lacking the intracellular domain as a model of increased glycocalyx-induced resistance to neratinib both in cell culture and in experimental brain metastases in immunodeficient mice. Our results highlight the importance of glycoproteins as a resistance mechanism to HER2-targeting therapies in breast cancer brain metastases.
Collapse
Affiliation(s)
- Marie-Anne Goyette
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medicine, Harvard Medical School, Boston, MA02115
- Department of Medicine, Brigham and Women's Hospital, Boston, MA02115
| | - Laura E. Stevens
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medicine, Harvard Medical School, Boston, MA02115
- Department of Medicine, Brigham and Women's Hospital, Boston, MA02115
| | - Carolyn R. DePinho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Marco Seehawer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medicine, Harvard Medical School, Boston, MA02115
- Department of Medicine, Brigham and Women's Hospital, Boston, MA02115
| | - Jun Nishida
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medicine, Harvard Medical School, Boston, MA02115
- Department of Medicine, Brigham and Women's Hospital, Boston, MA02115
| | - Zheqi Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medicine, Harvard Medical School, Boston, MA02115
- Department of Medicine, Brigham and Women's Hospital, Boston, MA02115
| | - Callahan M. Wilde
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Rong Li
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA02215
| | - Xintao Qiu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA02215
| | - Alanna L. Pyke
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Stephanie Zhao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Klothilda Lim
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA02215
| | | | - Jason J. Northey
- Center for Bioengineering and Tissue Regeneration, University of California San Francisco, San Francisco, CA94143
| | | | - Henry W. Long
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA02215
| | - Carolyn R. Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, University of California San Francisco, San Francisco, CA94143
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA94143
- Department of Surgery, University of California San Francisco, San Francisco, CA94143
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA94143
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA94143
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA94143
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medicine, Harvard Medical School, Boston, MA02115
- Department of Medicine, Brigham and Women's Hospital, Boston, MA02115
| |
Collapse
|
12
|
Desai B, Miti T, Prabhakaran S, Miroshnychenko D, Henry M, Marusyk V, Gatenbee C, Bui M, Scott J, Altrock PM, Haura E, Anderson ARA, Basanta D, Marusyk A. Peristromal niches protect lung cancers from targeted therapies through a combined effect of multiple molecular mediators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590626. [PMID: 38712093 PMCID: PMC11071426 DOI: 10.1101/2024.04.24.590626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Targeted therapies directed against oncogenic signaling addictions, such as inhibitors of ALK in ALK+ NSCLC often induce strong and durable clinical responses. However, they are not curative in metastatic cancers, as some tumor cells persist through therapy, eventually developing resistance. Therapy sensitivity can reflect not only cell-intrinsic mechanisms but also inputs from stromal microenvironment. Yet, the contribution of tumor stroma to therapeutic responses in vivo remains poorly defined. To address this gap of knowledge, we assessed the contribution of stroma-mediated resistance to therapeutic responses to the frontline ALK inhibitor alectinib in xenograft models of ALK+ NSCLC. We found that stroma-proximal tumor cells are partially protected against cytostatic effects of alectinib. This effect is observed not only in remission, but also during relapse, indicating the strong contribution of stroma-mediated resistance to both persistence and resistance. This therapy-protective effect of the stromal niche reflects a combined action of multiple mechanisms, including growth factors and extracellular matrix components. Consequently, despite improving alectinib responses, suppression of any individual resistance mechanism was insufficient to fully overcome the protective effect of stroma. Focusing on shared collateral sensitivity of persisters offered a superior therapeutic benefit, especially when using an antibody-drug conjugate with bystander effect to limit therapeutic escape. These findings indicate that stroma-mediated resistance might be the major contributor to both residual and progressing disease and highlight the limitation of focusing on suppressing a single resistance mechanism at a time.
Collapse
|
13
|
Dou T, Li J, Zhang Y, Pei W, Zhang B, Wang B, Wang Y, Jia H. The cellular composition of the tumor microenvironment is an important marker for predicting therapeutic efficacy in breast cancer. Front Immunol 2024; 15:1368687. [PMID: 38487526 PMCID: PMC10937353 DOI: 10.3389/fimmu.2024.1368687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
At present, the incidence rate of breast cancer ranks first among new-onset malignant tumors in women. The tumor microenvironment is a hot topic in tumor research. There are abundant cells in the tumor microenvironment that play a protumor or antitumor role in breast cancer. During the treatment of breast cancer, different cells have different influences on the therapeutic response. And after treatment, the cellular composition in the tumor microenvironment will change too. In this review, we summarize the interactions between different cell compositions (such as immune cells, fibroblasts, endothelial cells, and adipocytes) in the tumor microenvironment and the treatment mechanism of breast cancer. We believe that detecting the cellular composition of the tumor microenvironment is able to predict the therapeutic efficacy of treatments for breast cancer and benefit to combination administration of breast cancer.
Collapse
Affiliation(s)
- Tingyao Dou
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaochen Zhang
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Wanru Pei
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Binyue Zhang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Bin Wang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi, China
| | - Hongyan Jia
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi, China
| |
Collapse
|
14
|
Adhikari E, Liu Q, Johnson J, Stewart P, Marusyk V, Fang B, Izumi V, Bowers K, Guzman KM, Koomen JM, Marusyk A, Lau EK. Brain metastasis-associated fibroblasts secrete fucosylated PVR/CD155 that induces breast cancer invasion. Cell Rep 2023; 42:113463. [PMID: 37995180 DOI: 10.1016/j.celrep.2023.113463] [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: 05/22/2023] [Revised: 09/19/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023] Open
Abstract
Brain metastasis cancer-associated fibroblasts (bmCAFs) are emerging as crucial players in the development of breast cancer brain metastasis (BCBM), but our understanding of the underlying molecular mechanisms is limited. In this study, we aim to elucidate the pathological contributions of fucosylation (the post-translational modification of proteins by the dietary sugar L-fucose) to tumor-stromal interactions that drive the development of BCBM. Here, we report that patient-derived bmCAFs secrete high levels of polio virus receptor (PVR), which enhance the invasive capacity of BC cells. Mechanistically, we find that HIF1α transcriptionally upregulates fucosyltransferase 11, which fucosylates PVR, triggering its secretion from bmCAFs. Global phosphoproteomic analysis of BC cells followed by functional verification identifies cell-cell junction and actin cytoskeletal signaling as modulated by bmCAF-secreted, -fucosylated PVR. Our findings delineate a hypoxia- and fucosylation-regulated mechanism by which bmCAFs contribute to the invasiveness of BCBM in the brain.
Collapse
Affiliation(s)
- Emma Adhikari
- Department of Tumor Microenvironment & Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33612, USA; Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Qian Liu
- Department of Tumor Microenvironment & Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33612, USA; Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Joseph Johnson
- Department of Analytic Microscopy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Paul Stewart
- Biostatistics and Bioinformatics Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Viktoriya Marusyk
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Bin Fang
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Victoria Izumi
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kiah Bowers
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kelly M Guzman
- Department of Analytic Microscopy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - John M Koomen
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Andriy Marusyk
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Eric K Lau
- Department of Tumor Microenvironment & Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
15
|
Choi J. Spatial simulation of autologous cell defection for cancer treatment. Evol Med Public Health 2023; 11:461-471. [PMID: 38111808 PMCID: PMC10727474 DOI: 10.1093/emph/eoad042] [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: 08/09/2023] [Revised: 11/02/2023] [Indexed: 12/20/2023] Open
Abstract
Cancer cells are highly cooperative in a nepotistic way and evolutionarily dynamic. Present cancer treatments often overlook these aspects, inducing the selection of resistant cancer cells and the corresponding relapse. As an alternative method of cancer elimination, autologous cell defection (ACD) was suggested by which modified cancer cells parasitically reliant on other cancer cells are implemented to the cancer cluster. Specifically, modified cancer cells should not produce costly growth factors that promote the growth of other cancer cells while receiving the benefit of exposure to such growth factors. Analytical models and rudimentary experiments up to date provide the medical feasibility of this method. In this study, I built comprehensive spatial simulation models by embracing the effects of the multiple growth factors, the Warburg effect, mutations and immunity. The simulation results based on planar spatial structures indicate that implementation of the defective modified tumours may replace the existing cancer cluster and defective cells would later collapse by themselves. Furthermore, I built a mathematical model that compares the fitness of the cells adjacent to the hypertumour-cancer interface. I also calculated whether anticancer drugs that reduce the effects of the growth factors promote or demote the utility of ACD under diverse fitness functions. The computational examination implies that anticancer drugs may impede the therapeutic effect of ACD when there is a strong concavity in the fitness function. The analysis results could work as a general guidance for effective ACD that may expand the paradigm of cancer treatment.
Collapse
Affiliation(s)
- Jibeom Choi
- Department of Applied Mathematics, College of Applied Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| |
Collapse
|
16
|
Miroshnychenko D, Miti T, Kumar P, Miller A, Laurie M, Giraldo N, Bui MM, Altrock PM, Basanta D, Marusyk A. Stroma-Mediated Breast Cancer Cell Proliferation Indirectly Drives Chemoresistance by Accelerating Tumor Recovery between Chemotherapy Cycles. Cancer Res 2023; 83:3681-3692. [PMID: 37791818 PMCID: PMC10646478 DOI: 10.1158/0008-5472.can-23-0398] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/28/2023] [Accepted: 09/29/2023] [Indexed: 10/05/2023]
Abstract
The ability of tumors to survive therapy reflects both cell-intrinsic and microenvironmental mechanisms. Across many cancers, including triple-negative breast cancer (TNBC), a high stroma/tumor ratio correlates with poor survival. In many contexts, this correlation can be explained by the direct reduction of therapy sensitivity induced by stroma-produced paracrine factors. We sought to explore whether this direct effect contributes to the link between stroma and poor responses to chemotherapies. In vitro studies with panels of TNBC cell line models and stromal isolates failed to detect a direct modulation of chemoresistance. At the same time, consistent with prior studies, fibroblast-produced secreted factors stimulated treatment-independent enhancement of tumor cell proliferation. Spatial analyses indicated that proximity to stroma is often associated with enhanced tumor cell proliferation in vivo. These observations suggested an indirect link between stroma and chemoresistance, where stroma-augmented proliferation potentiates the recovery of residual tumors between chemotherapy cycles. To evaluate this hypothesis, a spatial agent-based model of stroma impact on proliferation/death dynamics was developed that was quantitatively parameterized using inferences from histologic analyses and experimental studies. The model demonstrated that the observed enhancement of tumor cell proliferation within stroma-proximal niches could enable tumors to avoid elimination over multiple chemotherapy cycles. Therefore, this study supports the existence of an indirect mechanism of environment-mediated chemoresistance that might contribute to the negative correlation between stromal content and poor therapy outcomes. SIGNIFICANCE Integration of experimental research with mathematical modeling reveals an indirect microenvironmental chemoresistance mechanism by which stromal cells stimulate breast cancer cell proliferation and highlights the importance of consideration of proliferation/death dynamics. See related commentary by Wall and Echeverria, p. 3667.
Collapse
Affiliation(s)
- Daria Miroshnychenko
- Department of Metabolism and Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Tatiana Miti
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Pragya Kumar
- Department of Metabolism and Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Cancer Biology PhD Program, University of South Florida, Tampa, Florida
| | - Anna Miller
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mark Laurie
- Department of Metabolism and Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Nathalia Giraldo
- Department of Metabolism and Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Molecular Medicine, University of South Florida, Tampa, Florida
| | - Marilyn M. Bui
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Philipp M. Altrock
- Department of Theoretical Biology, Max Planck Institute for Evolutionary Biology, Ploen, Schleswig-Holstein, Germany
| | - David Basanta
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Andriy Marusyk
- Department of Metabolism and Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Molecular Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
17
|
Wall SW, Echeverria GV. Avoiding Extinction: Cancer-Associated Fibroblasts Help Triple-Negative Breast Cancer Outrun Chemotherapy. Cancer Res 2023; 83:3667-3669. [PMID: 37964615 DOI: 10.1158/0008-5472.can-23-2770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2023]
Abstract
Neoadjuvant chemotherapy is a staple of triple-negative breast cancer (TNBC) treatment. Predicated on the principle of fractional cell killing, chemotherapy regimens are typically cycles of short drug exposure followed by a period of recovery from the toxic side effects. However, many patients experience chemotherapy resistance for a variety of reasons, resulting in tumors that are not sufficiently reduced with treatment. Response to chemotherapy prior to surgical resection is a strong predictor of patient prognosis; therefore, finding ways to improve efficacy is a critical need. Tremendous effort has gone into understanding the relationship between the tumor microenvironment and treatment sensitivity in many tumor types. In this issue of Cancer Research, Miroshnychenko and colleagues investigate how the well-characterized phenomenon of cancer-associated fibroblast (CAF)-induced proliferation of tumor cells allows TNBC to evade extinction after multiple cycles of cytotoxic chemotherapies. Their findings imply CAF-promoted tumor cell proliferation allows tumor cells to push through stressful conditions caused by treatment and to avoid tumor elimination. This mechanism of 'indirect' chemoresistance contrasts with the dogma that tumor cell proliferation enhances chemosensitivity. This study opens the door for the discovery of mechanisms and therapeutic targets to limit the ability of CAFs to rescue tumor cells from the brink of extinction. See related article by Miroshnychenko et al., p. 3681.
Collapse
Affiliation(s)
- Steven W Wall
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Gloria V Echeverria
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
18
|
Cogliati B, Yashaswini CN, Wang S, Sia D, Friedman SL. Friend or foe? The elusive role of hepatic stellate cells in liver cancer. Nat Rev Gastroenterol Hepatol 2023; 20:647-661. [PMID: 37550577 PMCID: PMC10671228 DOI: 10.1038/s41575-023-00821-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2023] [Indexed: 08/09/2023]
Abstract
Liver fibrosis is a substantial risk factor for the development and progression of liver cancer, which includes hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA). Studies utilizing cell fate mapping and single-cell transcriptomics techniques have identified quiescent perisinusoidal hepatic stellate cells (HSCs) as the primary source of activated collagen-producing HSCs and liver cancer-associated fibroblasts (CAFs) in HCC and liver metastasis, complemented in iCCA by contributions from portal fibroblasts. At the same time, integrative computational analysis of single-cell, single-nucleus and spatial RNA sequencing data have revealed marked heterogeneity among HSCs and CAFs, with distinct subpopulations displaying unique gene expression signatures and functions. Some of these subpopulations have divergent roles in promoting or inhibiting liver fibrogenesis and carcinogenesis. In this Review, we discuss the dual roles of HSC subpopulations in liver fibrogenesis and their contribution to liver cancer promotion, progression and metastasis. We review the transcriptomic and functional similarities between HSC and CAF subpopulations, highlighting the pathways that either promote or prevent fibrosis and cancer, and the immunological landscape from which these pathways emerge. Insights from ongoing studies will yield novel strategies for developing biomarkers, assessing prognosis and generating new therapies for both HCC and iCCA prevention and treatment.
Collapse
Affiliation(s)
- Bruno Cogliati
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | | | - Shuang Wang
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniela Sia
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
19
|
Januškevičienė I, Petrikaitė V. Interaction of phenotypic sublines isolated from triple-negative breast cancer cell line MDA-MB-231 modulates their sensitivity to paclitaxel and doxorubicin in 2D and 3D assays. Am J Cancer Res 2023; 13:3368-3383. [PMID: 37693129 PMCID: PMC10492099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/23/2023] [Indexed: 09/12/2023] Open
Abstract
Breast cancer is a rapidly evolving, multifactorial disease that accumulates numerous genetic and epigenetic alterations. These result in molecular and phenotypic heterogeneity within the tumor, the complexity of which is further amplified through specific interactions between cancer cells. We aimed to analyze cell phenotypic sublines and the influence of their interaction on drug resistance, spheroid formation, and migration. Seven sublines were derived from the MDA-MB-231 breast cancer cell line using a multiple-cell suspension dilution. The growth rate, CD133 receptor expression, migration ability, and chemosensitivity of these sublines to anticancer drugs doxorubicin (DOX) and paclitaxel (PTX) were determined. Three sublines (F5, D8, H2) have been chosen to study their interaction in 2D and 3D assays. In the 2D model, the resistance of all sublines composition to DOX decreased, but in the 3D model, the resistance of all sublines except H2, increased to both PTX and DOX. In the 3D model, the combined sublines F5 and D8 had higher resistance to DOX and statistically significantly lower resistance for PTX compared to the control. The interaction between cancer stem-like cells (F5) and increased migration cells (D8) increased resistance to PTX in cell monolayer and increased resistance against both DOX and PTX in the spheroids. The interaction of DOX-resistant (H2) cells with other cell subpopulations (D8, F5, HF) decreased the resistance to DOX in cell monolayer and both DOX and PTX in spheroids.
Collapse
Affiliation(s)
- Indrė Januškevičienė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Sukilėlių pr., LT-50162, Kaunas, Lithuania
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Sukilėlių pr., LT-50162, Kaunas, Lithuania
| |
Collapse
|
20
|
Hogstrom JM, Cruz KA, Selfors LM, Ward MN, Mehta TS, Kanarek N, Philips J, Dialani V, Wulf G, Collins LC, Patel JM, Muranen T. Simultaneous isolation of hormone receptor-positive breast cancer organoids and fibroblasts reveals stroma-mediated resistance mechanisms. J Biol Chem 2023; 299:105021. [PMID: 37423299 PMCID: PMC10415704 DOI: 10.1016/j.jbc.2023.105021] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/11/2023] Open
Abstract
Recurrent hormone receptor-positive (HR+) breast cancer kills more than 600,000 women annually. Although HR+ breast cancers typically respond well to therapies, approximately 30% of patients relapse. At this stage, the tumors are usually metastatic and incurable. Resistance to therapy, particularly endocrine therapy is typically thought to be tumor intrinsic (e.g., estrogen receptor mutations). However, tumor-extrinsic factors also contribute to resistance. For example, stromal cells, such as cancer-associated fibroblasts (CAFs), residing in the tumor microenvironment, are known to stimulate resistance and disease recurrence. Recurrence in HR+ disease has been difficult to study due to the prolonged clinical course, complex nature of resistance, and lack of appropriate model systems. Existing HR+ models are limited to HR+ cell lines, a few HR+ organoid models, and xenograft models that all lack components of the human stroma. Therefore, there is an urgent need for more clinically relevant models to study the complex nature of recurrent HR+ breast cancer, and the factors contributing to treatment relapse. Here, we present an optimized protocol that allows a high take-rate, and simultaneous propagation of patient-derived organoids (PDOs) and matching CAFs, from primary and metastatic HR+ breast cancers. Our protocol allows for long-term culturing of HR+ PDOs that retain estrogen receptor expression and show responsiveness to hormone therapy. We further show the functional utility of this system by identifying CAF-secreted cytokines, such as growth-regulated oncogene α , as stroma-derived resistance drivers to endocrine therapy in HR+ PDOs.
Collapse
Affiliation(s)
- Jenny M Hogstrom
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Kayla A Cruz
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Laura M Selfors
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Madelyn N Ward
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Tejas S Mehta
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Naama Kanarek
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jordana Philips
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Vandana Dialani
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Gerburg Wulf
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Laura C Collins
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jaymin M Patel
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Taru Muranen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
21
|
Null JL, Kim DJ, McCann JV, Pramoonjago P, Fox JW, Zeng J, Kumar P, Edatt L, Pecot CV, Dudley AC. Periostin+ Stromal Cells Guide Lymphovascular Invasion by Cancer Cells. Cancer Res 2023; 83:2105-2122. [PMID: 37205636 PMCID: PMC10330490 DOI: 10.1158/0008-5472.can-22-2412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/16/2023] [Accepted: 05/17/2023] [Indexed: 05/21/2023]
Abstract
Cancer cell dissemination to sentinel lymph nodes is associated with poor patient outcomes, particularly in breast cancer. The process by which cancer cells egress from the primary tumor upon interfacing with the lymphatic vasculature is complex and driven by dynamic interactions between cancer cells and stromal cells, including cancer-associated fibroblasts (CAF). The matricellular protein periostin can distinguish CAF subtypes in breast cancer and is associated with increased desmoplasia and disease recurrence in patients. However, as periostin is secreted, periostin-expressing CAFs are difficult to characterize in situ, limiting our understanding of their specific contribution to cancer progression. Here, we used in vivo genetic labeling and ablation to lineage trace periostin+ cells and characterize their functions during tumor growth and metastasis. Periostin-expressing CAFs were spatially found at periductal and perivascular margins, were enriched at lymphatic vessel peripheries, and were differentially activated by highly metastatic cancer cells versus poorly metastatic counterparts. Surprisingly, genetically depleting periostin+ CAFs slightly accelerated primary tumor growth but impaired intratumoral collagen organization and inhibited lymphatic, but not lung, metastases. Periostin ablation in CAFs impaired their ability to deposit aligned collagen matrices and inhibited cancer cell invasion through collagen and across lymphatic endothelial cell monolayers. Thus, highly metastatic cancer cells mobilize periostin-expressing CAFs in the primary tumor site that promote collagen remodeling and collective cell invasion within lymphatic vessels and ultimately to sentinel lymph nodes. SIGNIFICANCE Highly metastatic breast cancer cells activate a population of periostin-expressing CAFs that remodel the extracellular matrix to promote escape of cancer cells into lymphatic vessels and drive colonization of proximal lymph nodes.
Collapse
Affiliation(s)
- Jamie L. Null
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA 22908, USA
| | - Dae Joong Kim
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA 22908, USA
| | - James V. McCann
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Patcharin Pramoonjago
- Department of Pathology, The University of Virginia, Charlottesville, VA 22908, USA
- UVA Biorepository and Tissue Research Facility
| | - Jay W. Fox
- Emily Couric Comprehensive Cancer Center, The University of Virginia
| | - Jianhao Zeng
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA 22908, USA
| | - Pankaj Kumar
- UVA Bioinformatics Core
- Department of Biochemistry and Molecular Genetics, The University of Virginia, Charlottesville, VA 22908, USA
| | | | - Chad V. Pecot
- Lineberger Comprehensive Cancer Center
- Division of Hematology/Oncology, Chapel Hill, North Carolina
- UNC RNA Discovery Center
- Department of Medicine, Chapel Hill, North Carolina, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Andrew C. Dudley
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA 22908, USA
- Emily Couric Comprehensive Cancer Center, The University of Virginia
| |
Collapse
|
22
|
Shimpi AA, Tan ML, Vilkhovoy M, Dai D, Roberts LM, Kuo J, Huang L, Varner JD, Paszek M, Fischbach C. Convergent Approaches to Delineate the Metabolic Regulation of Tumor Invasion by Hyaluronic Acid Biosynthesis. Adv Healthc Mater 2023; 12:e2202224. [PMID: 36479976 PMCID: PMC10238572 DOI: 10.1002/adhm.202202224] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/21/2022] [Indexed: 12/13/2022]
Abstract
Metastasis is the leading cause of breast cancer-related deaths and is often driven by invasion and cancer-stem like cells (CSCs). Both the CSC phenotype and invasion are associated with increased hyaluronic acid (HA) production. How these independent observations are connected, and which role metabolism plays in this process, remains unclear due to the lack of convergent approaches integrating engineered model systems, computational tools, and cancer biology. Using microfluidic invasion models, metabolomics, computational flux balance analysis, and bioinformatic analysis of patient data, the functional links between the stem-like, invasive, and metabolic phenotype of breast cancer cells as a function of HA biosynthesis are investigated. These results suggest that CSCs are more invasive than non-CSCs and that broad metabolic changes caused by overproduction of HA play a role in this process. Accordingly, overexpression of hyaluronic acid synthases (HAS) 2 or 3 induces a metabolic phenotype that promotes cancer cell stemness and invasion in vitro and upregulates a transcriptomic signature predictive of increased invasion and worse patient survival. This study suggests that HA overproduction leads to metabolic adaptations to satisfy the energy demands for 3D invasion of breast CSCs highlighting the importance of engineered model systems and multidisciplinary approaches in cancer research.
Collapse
Affiliation(s)
- Adrian A. Shimpi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Matthew L. Tan
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Michael Vilkhovoy
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - David Dai
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - L. Monet Roberts
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 14853, USA
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Joe Kuo
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Lingting Huang
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Jeffrey D. Varner
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Matthew Paszek
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York, 14853, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York, 14853, USA
| |
Collapse
|
23
|
Masud MA, Kim JY, Kim E. Effective dose window for containing tumor burden under tolerable level. NPJ Syst Biol Appl 2023; 9:17. [PMID: 37221258 DOI: 10.1038/s41540-023-00279-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/05/2023] [Indexed: 05/25/2023] Open
Abstract
A maximum-tolerated dose (MTD) reduces the drug-sensitive cell population, though it may result in the competitive release of drug resistance. Alternative treatment strategies such as adaptive therapy (AT) or dose modulation aim to impose competitive stress on drug-resistant cell populations by maintaining a sufficient number of drug-sensitive cells. However, given the heterogeneous treatment response and tolerable tumor burden level of individual patients, determining an effective dose that can fine-tune competitive stress remains challenging. This study presents a mathematical model-driven approach that determines the plausible existence of an effective dose window (EDW) as a range of doses that conserve sufficient sensitive cells while maintaining the tumor volume below a threshold tolerable tumor volume (TTV). We use a mathematical model that explains intratumor cell competition. Analyzing the model, we derive an EDW determined by TTV and the competitive strength. By applying a fixed endpoint optimal control model, we determine the minimal dose to contain cancer at a TTV. As a proof of concept, we study the existence of EDW for a small cohort of melanoma patients by fitting the model to longitudinal tumor response data. We performed identifiability analysis, and for the patients with uniquely identifiable parameters, we deduced patient-specific EDW and minimal dose. The tumor volume for a patient could be theoretically contained at the TTV either using continuous dose or AT strategy with doses belonging to EDW. Further, we conclude that the lower bound of the EDW approximates the minimum effective dose (MED) for containing tumor volume at the TTV.
Collapse
Affiliation(s)
- M A Masud
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
| | - Jae-Young Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Eunjung Kim
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea.
| |
Collapse
|
24
|
Salemme V, Centonze G, Avalle L, Natalini D, Piccolantonio A, Arina P, Morellato A, Ala U, Taverna D, Turco E, Defilippi P. The role of tumor microenvironment in drug resistance: emerging technologies to unravel breast cancer heterogeneity. Front Oncol 2023; 13:1170264. [PMID: 37265795 PMCID: PMC10229846 DOI: 10.3389/fonc.2023.1170264] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Breast cancer is a highly heterogeneous disease, at both inter- and intra-tumor levels, and this heterogeneity is a crucial determinant of malignant progression and response to treatments. In addition to genetic diversity and plasticity of cancer cells, the tumor microenvironment contributes to tumor heterogeneity shaping the physical and biological surroundings of the tumor. The activity of certain types of immune, endothelial or mesenchymal cells in the microenvironment can change the effectiveness of cancer therapies via a plethora of different mechanisms. Therefore, deciphering the interactions between the distinct cell types, their spatial organization and their specific contribution to tumor growth and drug sensitivity is still a major challenge. Dissecting intra-tumor heterogeneity is currently an urgent need to better define breast cancer biology and to develop therapeutic strategies targeting the microenvironment as helpful tools for combined and personalized treatment. In this review, we analyze the mechanisms by which the tumor microenvironment affects the characteristics of tumor heterogeneity that ultimately result in drug resistance, and we outline state of the art preclinical models and emerging technologies that will be instrumental in unraveling the impact of the tumor microenvironment on resistance to therapies.
Collapse
Affiliation(s)
- Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Alessio Piccolantonio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Pietro Arina
- UCL, Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Ugo Ala
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| |
Collapse
|
25
|
Amini P, Hajihosseini M, Pyne S, Dinu I. Geographically weighted linear combination test for gene-set analysis of a continuous spatial phenotype as applied to intratumor heterogeneity. Front Cell Dev Biol 2023; 11:1065586. [PMID: 36998245 PMCID: PMC10044624 DOI: 10.3389/fcell.2023.1065586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Background: The impact of gene-sets on a spatial phenotype is not necessarily uniform across different locations of cancer tissue. This study introduces a computational platform, GWLCT, for combining gene set analysis with spatial data modeling to provide a new statistical test for location-specific association of phenotypes and molecular pathways in spatial single-cell RNA-seq data collected from an input tumor sample.Methods: The main advantage of GWLCT consists of an analysis beyond global significance, allowing the association between the gene-set and the phenotype to vary across the tumor space. At each location, the most significant linear combination is found using a geographically weighted shrunken covariance matrix and kernel function. Whether a fixed or adaptive bandwidth is determined based on a cross-validation cross procedure. Our proposed method is compared to the global version of linear combination test (LCT), bulk and random-forest based gene-set enrichment analyses using data created by the Visium Spatial Gene Expression technique on an invasive breast cancer tissue sample, as well as 144 different simulation scenarios.Results: In an illustrative example, the new geographically weighted linear combination test, GWLCT, identifies the cancer hallmark gene-sets that are significantly associated at each location with the five spatially continuous phenotypic contexts in the tumors defined by different well-known markers of cancer-associated fibroblasts. Scan statistics revealed clustering in the number of significant gene-sets. A spatial heatmap of combined significance over all selected gene-sets is also produced. Extensive simulation studies demonstrate that our proposed approach outperforms other methods in the considered scenarios, especially when the spatial association increases.Conclusion: Our proposed approach considers the spatial covariance of gene expression to detect the most significant gene-sets affecting a continuous phenotype. It reveals spatially detailed information in tissue space and can thus play a key role in understanding the contextual heterogeneity of cancer cells.
Collapse
Affiliation(s)
- Payam Amini
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
- School of Medicine, Keele University, Keele, Staffordshire, United Kingdom
| | - Morteza Hajihosseini
- School of Public Health, University of Alberta, Edmonton, AB, Canada
- Stanford Department of Urology, Center for Academic Medicine, Palo Alto, CA, United States
| | - Saumyadipta Pyne
- Health Analytics Network, Pittsburgh, PA, United States
- University of California, Santa Barbara, Santa Barbara, CA, United States
- *Correspondence: Saumyadipta Pyne, ; Irina Dinu,
| | - Irina Dinu
- School of Public Health, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Saumyadipta Pyne, ; Irina Dinu,
| |
Collapse
|
26
|
Miroshnychenko D, Miti T, Miller A, Kumar P, Laurie M, Bui MM, Altrock PM, Basanta D, Marusyk A. Paracrine enhancement of tumor cell proliferation provides indirect stroma-mediated chemoresistance via acceleration of tumor recovery between chemotherapy cycles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527543. [PMID: 36798328 PMCID: PMC9934626 DOI: 10.1101/2023.02.07.527543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The ability of tumors to survive therapy is mediated not only by cell-intrinsic but also cell-extrinsic, microenvironmental mechanisms. Across many cancers, including triple-negative breast cancer (TNBC), a high stroma/tumor ratio correlates with poor survival. In many contexts, this correlation can be explained by the direct reduction of therapy sensitivity by stroma-produced paracrine factors through activating pro-survival signaling and stemness. We sought to explore whether this direct effect contributes to the link between stroma and poor responses to chemotherapies in TNBC. Our in vitro studies with panels of TNBC cell line models and stromal isolates failed to detect a direct modulation of chemoresistance. However, we found that fibroblasts often enhance baseline tumor cell proliferation. Consistent with this in vitro observation, we found evidence of stroma-enhanced TNBC cell proliferation in vivo , in xenograft models and patient samples. Based on these observations, we hypothesized an indirect link between stroma and chemoresistance, where stroma-augmented proliferation potentiates the recovery of residual tumors between chemotherapy cycles. To test this hypothesis, we developed a spatial agent-based model of tumor response to repeated dosing of chemotherapy. The model was quantitatively parameterized from histological analyses and experimental studies. We found that even a slight enhancement of tumor cell proliferation within stroma-proximal niches can strongly enhance the ability of tumors to survive multiple cycles of chemotherapy under biologically and clinically feasible parameters. In summary, our study uncovered a novel, indirect mechanism of chemoresistance. Further, our study highlights the limitations of short-term cytotoxicity assays in understanding chemotherapy responses and supports the integration of experimental and in silico modeling.
Collapse
|
27
|
Brumskill S, Barrera LN, Calcraft P, Phillips C, Costello E. Inclusion of cancer-associated fibroblasts in drug screening assays to evaluate pancreatic cancer resistance to therapeutic drugs. J Physiol Biochem 2023; 79:223-234. [PMID: 34865180 PMCID: PMC9905179 DOI: 10.1007/s13105-021-00857-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/28/2021] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterised by a pro-inflammatory stroma and multi-faceted microenvironment that promotes and maintains tumorigenesis. However, the models used to test new and emerging therapies for PDAC have not increased in complexity to keep pace with our understanding of the human disease. Promising therapies that pass pre-clinical testing often fail in pancreatic cancer clinical trials. The objective of this study was to investigate whether changes in the drug-dosing regimen or the addition of cancer-associated fibroblasts (CAFs) to current existing models can impact the efficacy of chemotherapy drugs used in the clinic. Here, we reveal that gemcitabine and paclitaxel markedly reduce the viability of pancreatic cell lines, but not CAFs, when cultured in 2D. Following the use of an in vitro drug pulsing experiment, PDAC cell lines showed sensitivity to gemcitabine and paclitaxel. However, CAFs were less sensitive to pulsing with gemcitabine compared to their response to paclitaxel. We also identify that a 3D co-culture model of MIA PaCa-2 or PANC-1 with CAFs showed an increased chemoresistance to gemcitabine when compared to standard 2D mono-cultures a difference to paclitaxel which showed no measurable difference between the 2D and 3D models, suggesting a complex interaction between the drug in study and the cell type used. Changes to standard 2D mono-culture-based assays and implementation of 3D co-culture assays lend complexity to established models and could provide tools for identifying therapies that will match clinically the success observed with in vitro models, thereby aiding in the discovery of novel therapies.
Collapse
Affiliation(s)
- Sarah Brumskill
- Institute of Translational Medicine, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, 2nd Floor Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
- Redx Oncology, Alderley Park, Macclesfield, Cheshire, UK
| | - Lawrence N Barrera
- Institute of Translational Medicine, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, 2nd Floor Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Peter Calcraft
- Redx Oncology, Alderley Park, Macclesfield, Cheshire, UK
| | | | - Eithne Costello
- Institute of Translational Medicine, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, 2nd Floor Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK.
| |
Collapse
|
28
|
Liu X, Lu Y, Huang J, Xing Y, Dai H, Zhu L, Li S, Feng J, Zhou B, Li J, Xia Q, Li J, Huang M, Gu Y, Su S. CD16 + fibroblasts foster a trastuzumab-refractory microenvironment that is reversed by VAV2 inhibition. Cancer Cell 2022; 40:1341-1357.e13. [PMID: 36379207 DOI: 10.1016/j.ccell.2022.10.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/16/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022]
Abstract
The leukocyte Fcγ receptor (FcγR)-mediated response is important for the efficacy of therapeutic antibodies; however, little is known about the role of FcγRs in other cell types. Here we identify a subset of fibroblasts in human breast cancer that express CD16 (FcγRIII). An abundance of these cells in HER2+ breast cancer patients is associated with poor prognosis and response to trastuzumab. Functionally, upon trastuzumab stimulation, CD16+ fibroblasts reduce drug delivery by enhancing extracellular matrix stiffness. Interaction between trastuzumab and CD16 activates the intracellular SYK-VAV2-RhoA-ROCK-MLC2-MRTF-A pathway, leading to elevated contractile force and matrix production. Targeting of a Rho family guanine nucleotide exchange factor, VAV2, which is indispensable for the function of CD16 in fibroblasts rather than leukocytes, reverses desmoplasia provoked by CD16+ fibroblasts. Collectively, our study reveals a role for the fibroblast FcγR in drug resistance, and suggests that VAV2 is an attractive target to augment the effects of antibody treatments.
Collapse
Affiliation(s)
- Xinwei Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Breast Surgery, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Yiwen Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jingying Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yue Xing
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Huiqi Dai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Liling Zhu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Shunrong Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jingwei Feng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Boxuan Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jiaqian Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Qidong Xia
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jiang Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yuanting Gu
- Department of Breast Surgery, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Shicheng Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China; Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Biotherapy Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
29
|
Ma Y, Zhan L, Yang J, Zhang J. SLC11A1 associated with tumor microenvironment is a potential biomarker of prognosis and immunotherapy efficacy for colorectal cancer. Front Pharmacol 2022; 13:984555. [PMID: 36438826 PMCID: PMC9681808 DOI: 10.3389/fphar.2022.984555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/20/2022] [Indexed: 11/04/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most lethal cancers of the digestive system. The tumor microenvironment (TME) plays a central role in the initiation and development of CRC. However, little is known about the modulation mechanism of the TME in CRC. In our study, we attempted to identify a biomarker related to the TME modulation that could serve as a potential prognostic biomarker for CRC. We identified differentially expressed genes between the ImmuneScore high/low and StromalScore high/low groups. Using univariate COX regression analysis and hub gene analysis (cytoHubba), SLC11A1 was identified as the only candidate gene for subsequent analysis. CIBERSORT, EPIC, MCPcounter, and immunogenic cell death were performed to evaluate the effect of SLC11A1 on the TME. We also collected samples and performed Real-time quantitative PCR to verify the expression levels of SLC11A1 in CRC and adjacent normal tissues. The IMvigor210 cohort, TIDE score, and immunophenoscore (IPS) were used to analyze the association between SLC11A1 and immunotherapy efficacy. SLC11A1 was highly expressed in CRC tissues compared with its expression in normal colorectal tissues and was associated with poor prognosis and advanced clinicopathological stages. Gene set enrichment analysis showed that TGF-β pathways, JAK-STAT pathways, and angiogenesis were significantly enriched in the high-SLC11A1 group. Single-cell analysis validated the correlation between SLC11A1 and the TME. Using CIBERSORT, EPIC, and MCPcounter algorithms, we found that there was more macrophage and fibroblast infiltration in the SLC11A1 high-expression group. Meanwhile, high-SLC11A1 patients had lower IPS scores, higher TIDE scores, and fewer immunotherapy benefits than those of low-SLC11A1 patients. In conclusion, SLC11A1 plays a crucial role in the TME and could serve as a potential biomarker for poor prognosis and immunotherapy efficacy in CRC.
Collapse
Affiliation(s)
- Yiming Ma
- Medical Oncology Department of Gastrointestinal Tumors, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Lei Zhan
- Medical Oncology Department of Gastrointestinal Tumors, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Jun Yang
- Medical Oncology Department of Breast Tumors, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Jingdong Zhang
- Medical Oncology Department of Gastrointestinal Tumors, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
30
|
Rizzolio S, Giordano S, Corso S. The importance of being CAFs (in cancer resistance to targeted therapies). J Exp Clin Cancer Res 2022; 41:319. [PMID: 36324182 PMCID: PMC9632140 DOI: 10.1186/s13046-022-02524-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/23/2022] [Indexed: 05/09/2023] Open
Abstract
In the last two decades, clinical oncology has been revolutionized by the advent of targeted drugs. However, the efficacy of these therapies is significantly limited by primary and acquired resistance, that relies not only on cell-autonomous mechanisms but also on tumor microenvironment cues. Cancer-associated fibroblasts (CAFs) are extremely plastic cells of the tumor microenvironment. They not only produce extracellular matrix components that build up the structure of tumor stroma, but they also release growth factors, chemokines, exosomes, and metabolites that affect all tumor properties, including response to drug treatment. The contribution of CAFs to tumor progression has been deeply investigated and reviewed in several works. However, their role in resistance to anticancer therapies, and in particular to molecular therapies, has been largely overlooked. This review specifically dissects the role of CAFs in driving resistance to targeted therapies and discusses novel CAF targeted therapeutic strategies to improve patient survival.
Collapse
Affiliation(s)
| | - Silvia Giordano
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
- Department of Oncology, University of Torino, Torino, Italy
| | - Simona Corso
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
- Department of Oncology, University of Torino, Torino, Italy.
| |
Collapse
|
31
|
Li J, Li X, Guo Q. Drug Resistance in Cancers: A Free Pass for Bullying. Cells 2022; 11:3383. [PMID: 36359776 PMCID: PMC9654341 DOI: 10.3390/cells11213383] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 08/13/2023] Open
Abstract
The cancer burden continues to grow globally, and drug resistance remains a substantial challenge in cancer therapy. It is well established that cancerous cells with clonal dysplasia generate the same carcinogenic lesions. Tumor cells pass on genetic templates to subsequent generations in evolutionary terms and exhibit drug resistance simply by accumulating genetic alterations. However, recent evidence has implied that tumor cells accumulate genetic alterations by progressively adapting. As a result, intratumor heterogeneity (ITH) is generated due to genetically distinct subclonal populations of cells coexisting. The genetic adaptive mechanisms of action of ITH include activating "cellular plasticity", through which tumor cells create a tumor-supportive microenvironment in which they can proliferate and cause increased damage. These highly plastic cells are located in the tumor microenvironment (TME) and undergo extreme changes to resist therapeutic drugs. Accordingly, the underlying mechanisms involved in drug resistance have been re-evaluated. Herein, we will reveal new themes emerging from initial studies of drug resistance and outline the findings regarding drug resistance from the perspective of the TME; the themes include exosomes, metabolic reprogramming, protein glycosylation and autophagy, and the relates studies aim to provide new targets and strategies for reversing drug resistance in cancers.
Collapse
Affiliation(s)
| | | | - Qie Guo
- The Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
32
|
Leighow SM, Landry B, Lee MJ, Peyton SR, Pritchard JR. Agent-Based Models Help Interpret Patterns of Clinical Drug Resistance by Contextualizing Competition Between Distinct Drug Failure Modes. Cell Mol Bioeng 2022; 15:521-533. [PMID: 36444351 PMCID: PMC9700548 DOI: 10.1007/s12195-022-00748-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction Modern targeted cancer therapies are carefully crafted small molecules. These exquisite technologies exhibit an astonishing diversity of observed failure modes (drug resistance mechanisms) in the clinic. This diversity is surprising because back of the envelope calculations and classic modeling results in evolutionary dynamics suggest that the diversity in the modes of clinical drug resistance should be considerably smaller than what is observed. These same calculations suggest that the outgrowth of strong pre-existing genetic resistance mutations within a tumor should be ubiquitous. Yet, clinically relevant drug resistance occurs in the absence of obvious resistance conferring genetic alterations. Quantitatively, understanding the underlying biological mechanisms of failure mode diversity may improve the next generation of targeted anticancer therapies. It also provides insights into how intratumoral heterogeneity might shape interpatient diversity during clinical relapse. Materials and Methods We employed spatial agent-based models to explore regimes where spatial constraints enable wild type cells (that encounter beneficial microenvironments) to compete against genetically resistant subclones in the presence of therapy. In order to parameterize a model of microenvironmental resistance, BT20 cells were cultured in the presence and absence of fibroblasts from 16 different tissues. The degree of resistance conferred by cancer associated fibroblasts in the tumor microenvironment was quantified by treating mono- and co-cultures with letrozole and then measuring the death rates. Results and Discussion Our simulations indicate that, even when a mutation is more drug resistant, its outgrowth can be delayed by abundant, low magnitude microenvironmental resistance across large regions of a tumor that lack genetic resistance. These observations hold for different modes of microenvironmental resistance, including juxtacrine signaling, soluble secreted factors, and remodeled ECM. This result helps to explain the remarkable diversity of resistance mechanisms observed in solid tumors, which subverts the presumption that the failure mode that causes the quantitatively fastest growth in the presence of drug should occur most often in the clinic. Conclusion Our model results demonstrate that spatial effects can interact with low magnitude of resistance microenvironmental effects to successfully compete against genetic resistance that is orders of magnitude larger. Clinical outcomes of solid tumors are intrinsically connected to their spatial structure, and the tractability of spatial agent-based models like the ones presented here enable us to understand this relationship more completely.
Collapse
Affiliation(s)
- Scott M. Leighow
- Department of Biomedical Engineering, 211 Wartik Laboratory, Pennsylvania State University, University Park, State College, PA 16802 USA
| | - Ben Landry
- Department of Systems Biology, University of Massachusetts Medical School, Worcester, MA USA
| | - Michael J. Lee
- Department of Systems Biology, University of Massachusetts Medical School, Worcester, MA USA
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA USA
| | - Justin R. Pritchard
- Department of Biomedical Engineering, 211 Wartik Laboratory, Pennsylvania State University, University Park, State College, PA 16802 USA
| |
Collapse
|
33
|
Stupnikov A, Sizykh A, Budkina A, Favorov A, Afsari B, Wheelan S, Marchionni L, Medvedeva Y. Hobotnica: exploring molecular signature quality. F1000Res 2022; 10:1260. [PMID: 36204675 PMCID: PMC9513410 DOI: 10.12688/f1000research.74846.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 11/24/2022] Open
Abstract
A Molecular Features Set (MFS), is a result of a vast diversity of bioinformatics pipelines. The lack of a “gold standard” for most experimental data modalities makes it difficult to provide valid estimation for a particular MFS's quality. Yet, this goal can partially be achieved by analyzing inner-sample Distance Matrices (DM) and their power to distinguish between phenotypes. The quality of a DM can be assessed by summarizing its power to quantify the differences of inner-phenotype and outer-phenotype distances. This estimation of the DM quality can be construed as a measure of the MFS's quality. Here we propose Hobotnica, an approach to estimate MFSs quality by their ability to stratify data, and assign them significance scores, that allow for collating various signatures and comparing their quality for contrasting groups.
Collapse
Affiliation(s)
- Alexey Stupnikov
- Moscow Institute of Physics and Technology, Moscow, Russian Federation
- National Medical Research Center for Endocrinology, Moscow, Russian Federation
| | - Alexey Sizykh
- Moscow Institute of Physics and Technology, Moscow, Russian Federation
| | - Anna Budkina
- Moscow Institute of Physics and Technology, Moscow, Russian Federation
| | - Alexander Favorov
- Johns Hopkins University, Baltimore, USA
- Vavilov Institute for General Genetics RAS, Moscow, Russian Federation
| | | | | | | | - Yulia Medvedeva
- Moscow Institute of Physics and Technology, Moscow, Russian Federation
- National Medical Research Center for Endocrinology, Moscow, Russian Federation
- Center of Biotechnology RAS, Moscow, Russian Federation
| |
Collapse
|
34
|
Labrie M, Brugge JS, Mills GB, Zervantonakis IK. Therapy resistance: opportunities created by adaptive responses to targeted therapies in cancer. Nat Rev Cancer 2022; 22:323-339. [PMID: 35264777 PMCID: PMC9149051 DOI: 10.1038/s41568-022-00454-5] [Citation(s) in RCA: 142] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/09/2022] [Indexed: 02/08/2023]
Abstract
Normal cells explore multiple states to survive stresses encountered during development and self-renewal as well as environmental stresses such as starvation, DNA damage, toxins or infection. Cancer cells co-opt normal stress mitigation pathways to survive stresses that accompany tumour initiation, progression, metastasis and immune evasion. Cancer therapies accentuate cancer cell stresses and invoke rapid non-genomic stress mitigation processes that maintain cell viability and thus represent key targetable resistance mechanisms. In this Review, we describe mechanisms by which tumour ecosystems, including cancer cells, immune cells and stroma, adapt to therapeutic stresses and describe three different approaches to exploit stress mitigation processes: (1) interdict stress mitigation to induce cell death; (2) increase stress to induce cellular catastrophe; and (3) exploit emergent vulnerabilities in cancer cells and cells of the tumour microenvironment. We review challenges associated with tumour heterogeneity, prioritizing actionable adaptive responses for optimal therapeutic outcomes, and development of an integrative framework to identify and target vulnerabilities that arise from adaptive responses and engagement of stress mitigation pathways. Finally, we discuss the need to monitor adaptive responses across multiple scales and translation of combination therapies designed to take advantage of adaptive responses and stress mitigation pathways to the clinic.
Collapse
Affiliation(s)
- Marilyne Labrie
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
- Department of Obstetrics and Gynecology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Ludwig Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Gordon B Mills
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Ioannis K Zervantonakis
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
35
|
Pritchard JR, Lee MJ, Peyton SR. Materials-driven approaches to understand extrinsic drug resistance in cancer. SOFT MATTER 2022; 18:3465-3472. [PMID: 35445686 PMCID: PMC9380814 DOI: 10.1039/d2sm00071g] [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] [Indexed: 05/14/2023]
Abstract
Metastatic cancer has a poor prognosis, because it is broadly disseminated and associated with both intrinsic and acquired drug resistance. Critical unmet needs in effectively killing drug resistant cancer cells include overcoming the drug desensitization characteristics of some metastatic cancers/lesions, and tailoring therapeutic regimens to both the tumor microenvironment and the genetic profiles of the resident cancer cells. Bioengineers and materials scientists are developing technologies to determine how metastatic sites exclude therapies, and how extracellular factors (including cells, proteins, metabolites, extracellular matrix, and abiotic factors) at metastatic sites significantly affect drug pharmacodynamics. Two looming challenges are determining which feature, or combination of features, from the tumor microenvironment drive drug resistance, and what the relative impact is of extracellular signals vs. intrinsic cell genetics in determining drug response. Sophisticated systems biology tools that can de-convolve a crowded network of signals and responses, as well as controllable microenvironments capable of providing discrete and tunable extracellular cues can help us begin to interrogate the high dimensional interactions governing drug resistance in patients.
Collapse
Affiliation(s)
- Justin R Pritchard
- Department of Biomedical Engineering, Pennsylvania State University, State College PA, USA
| | - Michael J Lee
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shelly R Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, 240 Thatcher Way, Life Sciences Laboratory N531, Amherst, MA 01003, USA.
| |
Collapse
|
36
|
Hu D, Li Z, Zheng B, Lin X, Pan Y, Gong P, Zhuo W, Hu Y, Chen C, Chen L, Zhou J, Wang L. Cancer-associated fibroblasts in breast cancer: Challenges and opportunities. Cancer Commun (Lond) 2022; 42:401-434. [PMID: 35481621 PMCID: PMC9118050 DOI: 10.1002/cac2.12291] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/06/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment is proposed to contribute substantially to the progression of cancers, including breast cancer. Cancer-associated fibroblasts (CAFs) are the most abundant components of the tumor microenvironment. Studies have revealed that CAFs in breast cancer originate from several types of cells and promote breast cancer malignancy by secreting factors, generating exosomes, releasing nutrients, reshaping the extracellular matrix, and suppressing the function of immune cells. CAFs are also becoming therapeutic targets for breast cancer due to their specific distribution in tumors and their unique biomarkers. Agents interrupting the effect of CAFs on surrounding cells have been developed and applied in clinical trials. Here, we reviewed studies examining the heterogeneity of CAFs in breast cancer and expression patterns of CAF markers in different subtypes of breast cancer. We hope that summarizing CAF-related studies from a historical perspective will help to accelerate the development of CAF-targeted therapeutic strategies for breast cancer.
Collapse
Affiliation(s)
- Dengdi Hu
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, P. R. China
| | - Zhaoqing Li
- Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| | - Bin Zheng
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, P. R. China
| | - Xixi Lin
- Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| | - Yuehong Pan
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, P. R. China
| | - Peirong Gong
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, P. R. China
| | - Wenying Zhuo
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, P. R. China.,Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| | - Yujie Hu
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, P. R. China
| | - Cong Chen
- Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| | - Lini Chen
- Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| | - Jichun Zhou
- Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| | - Linbo Wang
- Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Hangzhou, Zhejiang, 310016, P. R. China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, P. R. China
| |
Collapse
|
37
|
Foo J, Basanta D, Rockne RC, Strelez C, Shah C, Ghaffarian K, Mumenthaler SM, Mitchell K, Lathia JD, Frankhouser D, Branciamore S, Kuo YH, Marcucci G, Vander Velde R, Marusyk A, Huang S, Hari K, Jolly MK, Hatzikirou H, Poels KE, Spilker ME, Shtylla B, Robertson-Tessi M, Anderson ARA. Roadmap on plasticity and epigenetics in cancer. Phys Biol 2022; 19:10.1088/1478-3975/ac4ee2. [PMID: 35078159 PMCID: PMC9190291 DOI: 10.1088/1478-3975/ac4ee2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 01/25/2022] [Indexed: 11/22/2022]
Abstract
The role of plasticity and epigenetics in shaping cancer evolution and response to therapy has taken center stage with recent technological advances including single cell sequencing. This roadmap article is focused on state-of-the-art mathematical and experimental approaches to interrogate plasticity in cancer, and addresses the following themes and questions: is there a formal overarching framework that encompasses both non-genetic plasticity and mutation-driven somatic evolution? How do we measure and model the role of the microenvironment in influencing/controlling non-genetic plasticity? How can we experimentally study non-genetic plasticity? Which mathematical techniques are required or best suited? What are the clinical and practical applications and implications of these concepts?
Collapse
Affiliation(s)
- Jasmine Foo
- School of Mathematics, University of Minnesota, Twin Cities, MN 55455, United States of America
| | - David Basanta
- Integrated Mathematical Oncology Department, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States of America
| | - Russell C Rockne
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, City of Hope National Medical Center, Beckman Research Institute, Duarte, CA 91010, United States of America
| | - Carly Strelez
- Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, CA 90064, United States of America
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States of America
| | - Curran Shah
- Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, CA 90064, United States of America
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America
| | - Kimya Ghaffarian
- Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, CA 90064, United States of America
| | - Shannon M Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, CA 90064, United States of America
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States of America
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America
| | - Kelly Mitchell
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Justin D Lathia
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, United States of America
- Case Comprehensive Cancer Center, Cleveland, OH 44106, United States of America
- Rose Ella Burkhardt Brain Tumor & Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - David Frankhouser
- Department of Population Sciences, City of Hope National Medical Center, Beckman Research Institute, Duarte, CA 91010, United States of America
| | - Sergio Branciamore
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, City of Hope National Medical Center, Beckman Research Institute, Duarte, CA 91010, United States of America
| | - Ya-Huei Kuo
- Department of Hematologic Malignancies Translational Science, City of Hope National Medical Center, Beckman Research Institute, Duarte, CA 91010, United States of America
| | - Guido Marcucci
- Department of Hematologic Malignancies Translational Science, City of Hope National Medical Center, Beckman Research Institute, Duarte, CA 91010, United States of America
| | - Robert Vander Velde
- Department of Cancer Physiology, Moffitt Cancer Center, Tampa, FL 33612, United States of America
- Department of Molecular Biology, University of South Florida Health, Tampa, FL 33612, United States of America
| | - Andriy Marusyk
- Department of Cancer Physiology, Moffitt Cancer Center, Tampa, FL 33612, United States of America
| | - Sui Huang
- Institute for Systems Biology, Seattle, WA 98109, United States of America
| | - Kishore Hari
- Centre for BioSystems Science and Engineering, Indian Institute of Science, 560012 Bangalore, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, 560012 Bangalore, India
| | - Haralampos Hatzikirou
- Mathematics Department, Khalifa University, PO Box 127788, Abu Dhabi, United Arab Emirates
- Centre for Information Services and High Performance Computing, TU Dresden, 01062, Dresden, Germany
| | - Kamrine E Poels
- Early Clinical Development, Pfizer Worldwide Research and Development and Medical, United States of America
| | - Mary E Spilker
- Medicine Design, Pfizer Worldwide Research and Development and Medical, United States of America
| | - Blerta Shtylla
- Early Clinical Development, Pfizer Worldwide Research and Development and Medical, United States of America
| | - Mark Robertson-Tessi
- Integrated Mathematical Oncology Department, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States of America
| | - Alexander R A Anderson
- Integrated Mathematical Oncology Department, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States of America
| |
Collapse
|
38
|
Kokoretsis D, Maniaki EK, Kyriakopoulou K, Koutsakis C, Piperigkou Z, Karamanos NK. Hyaluronan as "Agent Smith" in cancer extracellular matrix pathobiology: Regulatory roles in immune response, cancer progression and targeting. IUBMB Life 2022; 74:943-954. [PMID: 35261139 DOI: 10.1002/iub.2608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 12/16/2022]
Abstract
Extracellular matrix (ECM) critically regulates cancer cell behavior by governing cell signaling and properties. Hyaluronan (HA) acts as a structural and functional ECM component that mediates critical properties of cancer cells in a molecular size-dependent manner. HA fragments secreted by cancer-associated fibroblasts (CAFs) reveal the correlation of HA to CAF-mediated matrix remodeling, a key step for the initiation of metastasis. The main goal of this article is to highlight the vital functions of HA in cancer cell initiation and progression as well as HA-mediated paracrine interactions among cancer and stromal cells. Furthermore, the HA implication in mediating immune responses to cancer progression is also discussed. Novel data on the role of HA in the formation of pre-metastatic niche may contribute towards the improvement of current theranostic approaches that benefit cancer management.
Collapse
Affiliation(s)
- Dimitris Kokoretsis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Evangelia-Konstantina Maniaki
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Konstantina Kyriakopoulou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Christos Koutsakis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
39
|
M A M, Kim JY, Pan CH, Kim E. The impact of the spatial heterogeneity of resistant cells and fibroblasts on treatment response. PLoS Comput Biol 2022; 18:e1009919. [PMID: 35263336 PMCID: PMC8906648 DOI: 10.1371/journal.pcbi.1009919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/11/2022] [Indexed: 01/03/2023] Open
Abstract
A long-standing practice in the treatment of cancer is that of hitting hard with the maximum tolerated dose to eradicate tumors. This continuous therapy, however, selects for resistant cells, leading to the failure of the treatment. A different type of treatment strategy, adaptive therapy, has recently been shown to have a degree of success in both preclinical xenograft experiments and clinical trials. Adaptive therapy is used to maintain a tumor's volume by exploiting the competition between drug-sensitive and drug-resistant cells with minimum effective drug doses or timed drug holidays. To further understand the role of competition in the outcomes of adaptive therapy, we developed a 2D on-lattice agent-based model. Our simulations show that the superiority of the adaptive strategy over continuous therapy depends on the local competition shaped by the spatial distribution of resistant cells. Intratumor competition can also be affected by fibroblasts, which produce microenvironmental factors that promote cancer cell growth. To this end, we simulated the impact of different fibroblast distributions on treatment outcomes. As a proof of principle, we focused on five types of distribution of fibroblasts characterized by different locations, shapes, and orientations of the fibroblast region with respect to the resistant cells. Our simulation shows that the spatial architecture of fibroblasts modulates tumor progression in both continuous and adaptive therapy. Finally, as a proof of concept, we simulated the outcomes of adaptive therapy of a virtual patient with four metastatic sites composed of different spatial distributions of fibroblasts and drug-resistant cell populations. Our simulation highlights the importance of undetected metastatic lesions on adaptive therapy outcomes.
Collapse
Affiliation(s)
- Masud M A
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Republic of Korea
| | - Jae-Young Kim
- Graduate School of Science and Technology, Chungnam National University, Daejeon, Republic of Korea
| | - Cheol-Ho Pan
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Republic of Korea
| | - Eunjung Kim
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Republic of Korea
| |
Collapse
|
40
|
Bouchard G, Garcia Marques FJ, Karacosta LG, Zhang W, Bermudez A, Riley NM, Varma S, Mehl LC, Benson JA, Shrager JB, Bertozzi CR, Pitteri S, Giaccia AJ, Plevritis SK. Multiomics Analysis of Spatially Distinct Stromal Cells Reveals Tumor-Induced O-Glycosylation of the CDK4-pRB Axis in Fibroblasts at the Invasive Tumor Edge. Cancer Res 2022; 82:648-664. [PMID: 34853070 PMCID: PMC9075699 DOI: 10.1158/0008-5472.can-21-1705] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/02/2021] [Accepted: 11/24/2021] [Indexed: 11/16/2022]
Abstract
The invasive leading edge represents a potential gateway for tumor metastasis. The role of fibroblasts from the tumor edge in promoting cancer invasion and metastasis has not been comprehensively elucidated. We hypothesize that cross-talk between tumor and stromal cells within the tumor microenvironment results in activation of key biological pathways depending on their position in the tumor (edge vs. core). Here we highlight phenotypic differences between tumor-adjacent-fibroblasts (TAF) from the invasive edge and tumor core fibroblasts from the tumor core, established from human lung adenocarcinomas. A multiomics approach that includes genomics, proteomics, and O-glycoproteomics was used to characterize cross-talk between TAFs and cancer cells. These analyses showed that O-glycosylation, an essential posttranslational modification resulting from sugar metabolism, alters key biological pathways including the cyclin-dependent kinase 4 (CDK4) and phosphorylated retinoblastoma protein axis in the stroma and indirectly modulates proinvasive features of cancer cells. In summary, the O-glycoproteome represents a new consideration for important biological processes involved in tumor-stroma cross-talk and a potential avenue to improve the anticancer efficacy of CDK4 inhibitors. SIGNIFICANCE A multiomics analysis of spatially distinct fibroblasts establishes the importance of the stromal O-glycoproteome in tumor-stroma interactions at the leading edge and provides potential strategies to improve cancer treatment. See related commentary by De Wever, p. 537.
Collapse
Affiliation(s)
- Gina Bouchard
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
- Department of Radiology, Canary Center for Cancer Early Detection, Palo Alto CA, 94304, USA
- Department of Radiation Oncology, Stanford, CA 94305, USA
| | | | | | - Weiruo Zhang
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Abel Bermudez
- Department of Radiology, Canary Center for Cancer Early Detection, Palo Alto CA, 94304, USA
| | | | - Sushama Varma
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | | | - Jalen Anthony Benson
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Joseph B Shrager
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
| | | | - Sharon Pitteri
- Department of Radiology, Canary Center for Cancer Early Detection, Palo Alto CA, 94304, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford, CA 94305, USA
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Sylvia Katina Plevritis
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
- Department of Radiology, Canary Center for Cancer Early Detection, Palo Alto CA, 94304, USA
| |
Collapse
|
41
|
Kawiak A, Kostecka A. Regulation of Bcl-2 Family Proteins in Estrogen Receptor-Positive Breast Cancer and Their Implications in Endocrine Therapy. Cancers (Basel) 2022; 14:279. [PMID: 35053443 PMCID: PMC8773933 DOI: 10.3390/cancers14020279] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022] Open
Abstract
Estrogen receptor (ER)-positive breast cancer accounts for around two-thirds of breast cancer occurrences, with endocrine therapy serving as first-line therapy in most cases. Targeting estrogen signaling pathways, which play a central role in regulating ER+ breast cell proliferation and survival, has proven to improve patient outcomes. However, despite the undeniable advantages of endocrine therapy, a subset of breast cancer patients develop acquired or intrinsic resistance to ER-targeting agents, limiting their efficacy. The activation of downstream ER signaling pathways upregulates pro-survival mechanisms that have been shown to influence the response of cells to endocrine therapy. The Bcl-2 family proteins play a central role in cell death regulation and have been shown to contribute to endocrine therapy resistance, supporting the survival of breast cancer cells and enhancing cell death evasion. Due to the overexpression of anti-apoptotic Bcl-2 proteins in ER-positive breast cancer, the role of these proteins as potential targets in hormone-responsive breast cancer is growing in interest. In particular, recent advances in the development of BH3 mimetics have enabled their evaluation in preclinical studies with ER+ breast cancer models, and BH3 mimetics have entered early ER+ breast cancer clinical trials. This review summarizes the molecular mechanisms underlying the regulation of Bcl-2 family proteins in ER+ breast cancer. Furthermore, an overview of recent advances in research regarding the efficacy of BH3 mimetics in ER+ breast cancer has been provided.
Collapse
Affiliation(s)
- Anna Kawiak
- Intercollegiate Faculty of Biotechnology, University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland
| | - Anna Kostecka
- Faculty of Pharmacy, Medical University of Gdansk, Hallera 107, 80-416 Gdansk, Poland;
| |
Collapse
|
42
|
Thomas SC, Madaan T, Kamble NS, Siddiqui NA, Pauletti GM, Kotagiri N. Engineered Bacteria Enhance Immunotherapy and Targeted Therapy through Stromal Remodeling of Tumors. Adv Healthc Mater 2022; 11:e2101487. [PMID: 34738725 PMCID: PMC8770579 DOI: 10.1002/adhm.202101487] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/28/2021] [Indexed: 01/03/2023]
Abstract
Desmoplastic solid tumors are characterized by the rapid build-up of extracellular matrix (ECM) macromolecules, such as hyaluronic acid (HA). The resulting physiological barrier prevents the infiltration of immune cells and also impedes the delivery of anticancer agents. The development of a hypervesiculating Escherichia coli Nissle (ΔECHy) based tumor targeting bacterial system capable of distributing a fusion peptide, cytolysin A (ClyA)-hyaluronidase (Hy) via outer membrane vesicles (OMVs) is reported. The capability of targeting hypoxic tumors, manufacturing recombinant proteins in situ and the added advantage of an on-site OMV based distribution system makes the engineered bacterial vector a unique candidate for peptide delivery. The HA degrading potential of Hy for stromal modulation is combined with the cytolytic activity of ClyA followed by testing it within syngeneic cancer models. ΔECHy is combined with immune checkpoint antibodies and tyrosine kinase inhibitors (TKIs) to demonstrate that remodeling the tumor stroma results in the improvement of immunotherapy outcomes and enhancing the efficacy of biological signaling inhibitors. The biocompatibility of ΔECHy is also investigated to show that the engineered bacteria are effectively cleared, elicit minimal inflammatory and immune responses, and therefore could be a reliable candidate as a live biotherapeutic.
Collapse
Affiliation(s)
- Shindu C. Thomas
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Tushar Madaan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Nitin S. Kamble
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Nabil A. Siddiqui
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Giovanni M. Pauletti
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, 1 Pharmacy Place, St. Louis, MO 63110, USA
| | - Nalinikanth Kotagiri
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| |
Collapse
|
43
|
Patient-derived functional organoids as a personalized approach for drug screening against hepatobiliary cancers. Adv Cancer Res 2022; 156:319-341. [DOI: 10.1016/bs.acr.2022.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
44
|
Chen M, Sheu MT, Cheng TL, Roffler SR, Lin SY, Chen YJ, Cheng YA, Cheng JJ, Chang HY, Wu TY, Kao AP, Ho YS, Chuang KH. A novel anti-tumor/anti-tumor-associated fibroblast/anti-mPEG tri-specific antibody to maximize the efficacy of mPEGylated nanomedicines against fibroblast-rich solid tumor. Biomater Sci 2021; 10:202-215. [PMID: 34826322 DOI: 10.1039/d1bm01218e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The therapeutic efficacy of methoxypolyethylene glycol (mPEG)-coated nanomedicines in solid tumor treatment is hindered by tumor-associated fibroblasts (TAFs), which promote tumor progression and form physical barriers. We developed an anti-HER2/anti-FAP/anti-mPEG tri-specific antibody (TsAb) for one-step conversion of mPEG-coated liposomal doxorubicin (Lipo-Dox) to immunoliposomes, which simultaneously target HER2+ breast cancer cells and FAP+ TAFs. The non-covalent modification did not adversely alter the physical characteristics and stability of Lipo-Dox. The TsAb-Lipo-Dox exhibited specific targeting and enhanced cytotoxicity against mono- and co-cultured HER2+ breast cancer cells and FAP+ TAFs, compared to bi-specific antibody (BsAb) modified or unmodified Lipo-Dox. An in vivo model of human breast tumor containing TAFs also revealed the improved tumor accumulation and therapeutic efficacy of TsAb-modified mPEGylated liposomes without signs of toxicity. Our data indicate that arming clinical mPEGylated nanomedicines with the TsAb is a feasible and applicable approach for overcoming the difficulties caused by TAFs in solid tumor treatment.
Collapse
Affiliation(s)
- Michael Chen
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Ming-Thau Sheu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Tian-Lu Cheng
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Shyr-Yi Lin
- Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei, Taiwan.,Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jou Chen
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Yi-An Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Jing-Jy Cheng
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan
| | - Hsin-Yu Chang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Tung-Yun Wu
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan
| | - An-Pei Kao
- Stemforce Biotechnology Co., Ltd, Chiayi City, Taiwan
| | - Yuan-Soon Ho
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.,School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan. .,Cancer Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan.,Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei, Taiwan. .,Ph.D Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan.,Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
45
|
Stupnikov A, Sizykh A, Budkina A, Favorov A, Afsari B, Wheelan S, Marchionni L, Medvedeva Y. Hobotnica: exploring molecular signature quality. F1000Res 2021; 10:1260. [PMID: 36204675 PMCID: PMC9513410 DOI: 10.12688/f1000research.74846.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 12/26/2023] Open
Abstract
A Molecular Features Set (MFS), is a result of a vast diversity of bioinformatics pipelines. The lack of a "gold standard" for most experimental data modalities makes it difficult to provide valid estimation for a particular MFS's quality. Yet, this goal can partially be achieved by analyzing inner-sample Distance Matrices (DM) and their power to distinguish between phenotypes. The quality of a DM can be assessed by summarizing its power to quantify the differences of inner-phenotype and outer-phenotype distances. This estimation of the DM quality can be construed as a measure of the MFS's quality. Here we propose Hobotnica, an approach to estimate MFSs quality by their ability to stratify data, and assign them significance scores, that allow for collating various signatures and comparing their quality for contrasting groups.
Collapse
Affiliation(s)
- Alexey Stupnikov
- Moscow Institute of Physics and Technology, Moscow, Russian Federation
- National Medical Research Center for Endocrinology, Moscow, Russian Federation
| | - Alexey Sizykh
- Moscow Institute of Physics and Technology, Moscow, Russian Federation
| | - Anna Budkina
- Moscow Institute of Physics and Technology, Moscow, Russian Federation
| | - Alexander Favorov
- Johns Hopkins University, Baltimore, USA
- Vavilov Institute for General Genetics RAS, Moscow, Russian Federation
| | | | | | | | - Yulia Medvedeva
- Moscow Institute of Physics and Technology, Moscow, Russian Federation
- National Medical Research Center for Endocrinology, Moscow, Russian Federation
- Center of Biotechnology RAS, Moscow, Russian Federation
| |
Collapse
|
46
|
Becker L, Janssen N, Layland SL, Mürdter TE, Nies AT, Schenke-Layland K, Marzi J. Raman Imaging and Fluorescence Lifetime Imaging Microscopy for Diagnosis of Cancer State and Metabolic Monitoring. Cancers (Basel) 2021; 13:cancers13225682. [PMID: 34830837 PMCID: PMC8616063 DOI: 10.3390/cancers13225682] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Hurdles for effective tumor therapy are delayed detection and limited effectiveness of systemic drug therapies by patient-specific multidrug resistance. Non-invasive bioimaging tools such as fluorescence lifetime imaging microscopy (FLIM) and Raman-microspectroscopy have evolved over the last decade, providing the potential to be translated into clinics for early-stage disease detection, in vitro drug screening, and drug efficacy studies in personalized medicine. Accessing tissue- and cell-specific spectral signatures, Raman microspectroscopy has emerged as a diagnostic tool to identify precancerous lesions, cancer stages, or cell malignancy. In vivo Raman measurements have been enabled by recent technological advances in Raman endoscopy and signal-enhancing setups such as coherent anti-stokes Raman spectroscopy or surface-enhanced Raman spectroscopy. FLIM enables in situ investigations of metabolic processes such as glycolysis, oxidative stress, or mitochondrial activity by using the autofluorescence of co-enzymes NADH and FAD, which are associated with intrinsic proteins as a direct measure of tumor metabolism, cell death stages and drug efficacy. The combination of non-invasive and molecular-sensitive in situ techniques and advanced 3D tumor models such as patient-derived organoids or microtumors allows the recapitulation of tumor physiology and metabolism in vitro and facilitates the screening for patient-individualized drug treatment options.
Collapse
Affiliation(s)
- Lucas Becker
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
| | - Nicole Janssen
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Shannon L Layland
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, University of Tübingen, 72076 Tübingen, Germany
| | - Thomas E Mürdter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Anne T Nies
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Katja Schenke-Layland
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
- Cardiovascular Research Laboratories, Department of Medicine/Cardiology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90073, USA
| | - Julia Marzi
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
| |
Collapse
|
47
|
Jones JO, Moody WM, Shields JD. Microenvironmental modulation of the developing tumour: an immune-stromal dialogue. Mol Oncol 2021; 15:2600-2633. [PMID: 32741067 PMCID: PMC8486574 DOI: 10.1002/1878-0261.12773] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/03/2020] [Accepted: 07/27/2020] [Indexed: 12/17/2022] Open
Abstract
Successful establishment of a tumour relies on a cascade of interactions between cancer cells and stromal cells within an evolving microenvironment. Both immune and nonimmune cellular components are key factors in this process, and the individual players may change their role from tumour elimination to tumour promotion as the microenvironment develops. While the tumour-stroma crosstalk present in an established tumour is well-studied, aspects in the early tumour or premalignant microenvironment have received less attention. This is in part due to the challenges in studying this process in the clinic or in mouse models. Here, we review the key anti- and pro-tumour factors in the early microenvironment and discuss how understanding this process may be exploited in the clinic.
Collapse
Affiliation(s)
- James O. Jones
- MRC Cancer UnitHutchison/MRC Research CentreUniversity of CambridgeCambridgeUK
- Department of OncologyCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - William M. Moody
- MRC Cancer UnitHutchison/MRC Research CentreUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
48
|
Hamalian S, Güth R, Runa F, Sanchez F, Vickers E, Agajanian M, Molnar J, Nguyen T, Gamez J, Humphries JD, Nayak A, Humphries MJ, Tchou J, Zervantonakis IK, Kelber JA. A SNAI2-PEAK1-INHBA stromal axis drives progression and lapatinib resistance in HER2-positive breast cancer by supporting subpopulations of tumor cells positive for antiapoptotic and stress signaling markers. Oncogene 2021; 40:5224-5235. [PMID: 34239043 PMCID: PMC8376636 DOI: 10.1038/s41388-021-01906-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 05/31/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023]
Abstract
Intercellular mechanisms by which the stromal microenvironment contributes to solid tumor progression and targeted therapy resistance remain poorly understood, presenting significant clinical hurdles. PEAK1 (Pseudopodium-Enriched Atypical Kinase One) is an actin cytoskeleton- and focal adhesion-associated pseudokinase that promotes cell state plasticity and cancer metastasis by mediating growth factor-integrin signaling crosstalk. Here, we determined that stromal PEAK1 expression predicts poor outcomes in HER2-positive breast cancers high in SNAI2 expression and enriched for MSC content. Specifically, we identified that the fibroblastic stroma in HER2-positive breast cancer patient tissue stains positive for both nuclear SNAI2 and cytoplasmic PEAK1. Furthermore, mesenchymal stem cells (MSCs) and cancer-associated fibroblasts (CAFs) express high PEAK1 protein levels and potentiate tumorigenesis, lapatinib resistance and metastasis of HER2-positive breast cancer cells in a PEAK1-dependent manner. Analysis of PEAK1-dependent secreted factors from MSCs revealed INHBA/activin-A as a necessary factor in the conditioned media of PEAK1-expressing MSCs that promotes lapatinib resistance. Single-cell CycIF analysis of MSC-breast cancer cell co-cultures identified enrichment of p-Akthigh/p-gH2AXlow, MCL1high/p-gH2AXlow and GRP78high/VIMhigh breast cancer cell subpopulations by the presence of PEAK1-expressing MSCs and lapatinib treatment. Bioinformatic analyses on a PEAK1-centric stroma-tumor cell gene set and follow-up immunostaining of co-cultures predict targeting antiapoptotic and stress pathways as a means to improve targeted therapy responses and patient outcomes in HER2-positive breast cancer and other stroma-rich malignancies. These data provide the first evidence that PEAK1 promotes tumorigenic phenotypes through a previously unrecognized SNAI2-PEAK1-INHBA stromal cell axis.
Collapse
Affiliation(s)
- Sarkis Hamalian
- Department of Biology, California State University Northridge, Northridge, CA, USA
| | - Robert Güth
- Department of Biology, California State University Northridge, Northridge, CA, USA
| | - Farhana Runa
- Department of Biology, California State University Northridge, Northridge, CA, USA
| | - Francesca Sanchez
- Department of Biology, California State University Northridge, Northridge, CA, USA
| | - Eric Vickers
- Department of Biology, California State University Northridge, Northridge, CA, USA
| | - Megan Agajanian
- Department of Biology, California State University Northridge, Northridge, CA, USA
| | - Justin Molnar
- Department of Biology, California State University Northridge, Northridge, CA, USA
| | - Tuan Nguyen
- Department of Biology, California State University Northridge, Northridge, CA, USA
| | - Joshua Gamez
- Department of Biology, California State University Northridge, Northridge, CA, USA
| | - Jonathan D Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Anupma Nayak
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania and Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| | - Martin J Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Julia Tchou
- Division of Endocrine and Oncologic Surgery, Department of Surgery, Rena Rowan Breast Center, Abramson Cancer Center, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis K Zervantonakis
- Department of Bioengineering, University of Pittsburgh, Center for Bioengineering, Pittsburgh, PA, USA
| | - Jonathan A Kelber
- Department of Biology, California State University Northridge, Northridge, CA, USA.
| |
Collapse
|
49
|
Tang LJW, Zaseela A, Toh CCM, Adine C, Aydar AO, Iyer NG, Fong ELS. Engineering stromal heterogeneity in cancer. Adv Drug Deliv Rev 2021; 175:113817. [PMID: 34087326 DOI: 10.1016/j.addr.2021.05.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/19/2021] [Accepted: 05/29/2021] [Indexed: 02/09/2023]
Abstract
Based on our exponentially increasing knowledge of stromal heterogeneity from advances in single-cell technologies, the notion that stromal cell types exist as a spectrum of unique subpopulations that have specific functions and spatial distributions in the tumor microenvironment has significant impact on tumor modeling for drug development and personalized drug testing. In this Review, we discuss the importance of incorporating stromal heterogeneity and tumor architecture, and propose an overall approach to guide the reconstruction of stromal heterogeneity in vitro for tumor modeling. These next-generation tumor models may support the development of more precise drugs targeting specific stromal cell subpopulations, as well as enable improved recapitulation of patient tumors in vitro for personalized drug testing.
Collapse
Affiliation(s)
- Leon Jia Wei Tang
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Ayshath Zaseela
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | | | - Christabella Adine
- Department of Biomedical Engineering, National University of Singapore, Singapore; The N.1 Institute for Health, National University of Singapore, Singapore
| | - Abdullah Omer Aydar
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - N Gopalakrishna Iyer
- National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, Singapore.
| | - Eliza Li Shan Fong
- Department of Biomedical Engineering, National University of Singapore, Singapore; The N.1 Institute for Health, National University of Singapore, Singapore.
| |
Collapse
|
50
|
Fu T, Dai LJ, Wu SY, Xiao Y, Ma D, Jiang YZ, Shao ZM. Spatial architecture of the immune microenvironment orchestrates tumor immunity and therapeutic response. J Hematol Oncol 2021; 14:98. [PMID: 34172088 PMCID: PMC8234625 DOI: 10.1186/s13045-021-01103-4] [Citation(s) in RCA: 262] [Impact Index Per Article: 65.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 06/03/2021] [Indexed: 02/08/2023] Open
Abstract
Tumors are not only aggregates of malignant cells but also well-organized complex ecosystems. The immunological components within tumors, termed the tumor immune microenvironment (TIME), have long been shown to be strongly related to tumor development, recurrence and metastasis. However, conventional studies that underestimate the potential value of the spatial architecture of the TIME are unable to completely elucidate its complexity. As innovative high-flux and high-dimensional technologies emerge, researchers can more feasibly and accurately detect and depict the spatial architecture of the TIME. These findings have improved our understanding of the complexity and role of the TIME in tumor biology. In this review, we first epitomized some representative emerging technologies in the study of the spatial architecture of the TIME and categorized the description methods used to characterize these structures. Then, we determined the functions of the spatial architecture of the TIME in tumor biology and the effects of the gradient of extracellular nonspecific chemicals (ENSCs) on the TIME. We also discussed the potential clinical value of our understanding of the spatial architectures of the TIME, as well as current limitations and future prospects in this novel field. This review will bring spatial architectures of the TIME, an emerging dimension of tumor ecosystem research, to the attention of more researchers and promote its application in tumor research and clinical practice.
Collapse
Affiliation(s)
- Tong Fu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lei-Jie Dai
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Song-Yang Wu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi Xiao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ding Ma
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|