1
|
Zhou S, Zhao Z, Wang Z, Xu H, Li Y, Xu K, Li W, Yang J. Cancer-associated fibroblasts in carcinogenesis. J Transl Med 2025; 23:50. [PMID: 39806363 PMCID: PMC11727299 DOI: 10.1186/s12967-025-06071-8] [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: 11/12/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
In contemporary times, cancer poses the most significant threat to human life and safety. Scientists have relentlessly pursued the intricacies of carcinogenesis and explored ways to prevent and treat cancer. Carcinogenesis is a complex, multi-faceted, and multi-stage process, with numerous underlying causes, including inflammation and fibrosis. Cancer-associated fibroblasts (CAFs), however, occupy a pivotal and substantial role within the tumor microenvironment, facilitating carcinogenesis through diverse mechanisms such as creating inflammation, fostering a fibrotic tumor microenvironment, and immunosuppression. In this paper, we introduce the concept of carcinogenesis, explain its causes, describe the characteristics of CAFs and their sources, and highlight the roles and mechanisms of CAFs in promoting carcinogenesis. Ultimately, our aim is to contribute to the development of novel therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Shufen Zhou
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zekun Zhao
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zhaojun Wang
- Department of Thyroid and Breast Surgery, The DingLi Clinical, The Wenzhou Central Hospital, College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hanzheng Xu
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yijie Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China.
| | - Wei Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Jiahua Yang
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| |
Collapse
|
2
|
Butler G, Amend SR, Venditti C, Pienta KJ. Punctuational evolution is pervasive in distal site metastatic colonization. Proc Biol Sci 2025; 292:20242850. [PMID: 39837515 PMCID: PMC11750355 DOI: 10.1098/rspb.2024.2850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/23/2025] Open
Abstract
The evolution of metastasis, the spread of cancer to distal sites within the body, represents a lethal stage of cancer progression. Yet, the evolutionary dynamics that shape the emergence of metastatic disease remain unresolved. Here, using single-cell lineage tracing data in combination with phylogenetic statistical methods, we show that the evolutionary trajectory of metastatic disease is littered with bursts of rapid molecular change as new cellular subpopulations appear, a pattern known as punctuational evolution. Next, by measuring punctuational evolution across the metastatic cascade, we show that punctuational effects are concentrated within the formation of secondary tumours at distal metastatic sites, suggesting that qualitatively different modes of evolution may drive primary and metastatic tumour progression. Taken as a whole, our findings provide empirical evidence for distinct patterns of molecular evolution at early and late stages of metastatic disease and our approach provides a framework to study the evolution of metastasis at a more nuanced level than has been previously possible.
Collapse
Affiliation(s)
- George Butler
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD21287, USA
| | - Sarah R. Amend
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD21287, USA
| | - Chris Venditti
- School of Biological Sciences, University of Reading, ReadingRG6 6AS, UK
| | - Kenneth J. Pienta
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD21287, USA
| |
Collapse
|
3
|
Bhattacharya R, Brown JS, Gatenby RA, Ibrahim-Hashim A. A gene for all seasons: The evolutionary consequences of HIF-1 in carcinogenesis, tumor growth and metastasis. Semin Cancer Biol 2024; 102-103:17-24. [PMID: 38969311 DOI: 10.1016/j.semcancer.2024.06.003] [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: 02/06/2024] [Revised: 04/23/2024] [Accepted: 06/06/2024] [Indexed: 07/07/2024]
Abstract
Oxygen played a pivotal role in the evolution of multicellularity during the Cambrian Explosion. Not surprisingly, responses to fluctuating oxygen concentrations are integral to the evolution of cancer-a disease characterized by the breakdown of multicellularity. Poorly organized tumor vasculature results in chaotic patterns of blood flow characterized by large spatial and temporal variations in intra-tumoral oxygen concentrations. Hypoxia-inducible growth factor (HIF-1) plays a pivotal role in enabling cells to adapt, metabolize, and proliferate in low oxygen conditions. HIF-1 is often constitutively activated in cancers, underscoring its importance in cancer progression. Here, we argue that the phenotypic changes mediated by HIF-1, in addition to adapting the cancer cells to their local environment, also "pre-adapt" them for proliferation at distant, metastatic sites. HIF-1-mediated adaptations include a metabolic shift towards anaerobic respiration or glycolysis, activation of cell survival mechanisms like phenotypic plasticity and epigenetic reprogramming, and formation of tumor vasculature through angiogenesis. Hypoxia induced epigenetic reprogramming can trigger epithelial to mesenchymal transition in cancer cells-the first step in the metastatic cascade. Highly glycolytic cells facilitate local invasion by acidifying the tumor microenvironment. New blood vessels, formed due to angiogenesis, provide cancer cells a conduit to the circulatory system. Moreover, survival mechanisms acquired by cancer cells in the primary site allow them to remodel tissue at the metastatic site generating tumor promoting microenvironment. Thus, hypoxia in the primary tumor promoted adaptations conducive to all stages of the metastatic cascade from the initial escape entry into a blood vessel, intravascular survival, extravasation into distant tissues, and establishment of secondary tumors.
Collapse
Affiliation(s)
- Ranjini Bhattacharya
- Department of Cancer Biology, University of South Florida, United States; Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, United States
| | - Joel S Brown
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, United States; Department of Evolutionary Biology, University of Illinois, at Chicago, United States
| | - Robert A Gatenby
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, United States; Department of Radiology, H. Lee Moffitt Cancer Center, United States.
| | - Arig Ibrahim-Hashim
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, United States.
| |
Collapse
|
4
|
Zhou X, Wu C, Wang X, Pan N, Sun X, Chen B, Zheng S, Wei Y, Chen J, Wu Y, Zhu F, Chen J, Chen H, Wang LX. Tumor cell-released autophagosomes (TRAPs) induce PD-L1-decorated NETs that suppress T-cell function to promote breast cancer pulmonary metastasis. J Immunother Cancer 2024; 12:e009082. [PMID: 38926151 PMCID: PMC11216055 DOI: 10.1136/jitc-2024-009082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Lung metastasis is the primary cause of breast cancer-related mortality. Neutrophil extracellular traps (NETs) are involved in the progression of breast cancer. However, the mechanism of NET formation is not fully understood. This study posits that tumor cell-released autophagosomes (TRAPs) play a crucial role in this process. METHODS TRAPs were isolated from breast cancer cell lines to analyze their impact on NET formation in both human and mouse neutrophils. The study used both in vitro and in vivo models, including Toll-like receptor 4 (TLR4-/-) mice and engineered breast cancer cell lines. Immunofluorescence, ELISA, Western blotting, RNA sequencing, and flow cytometry were employed to dissect the signaling pathways leading to NET production and to explore their immunosuppressive effects, particularly focusing on the impact of NETs on T-cell function. The therapeutic potential of targeting TRAP-induced NETs and their immunosuppressive functions was evaluated using DNase I and αPD-L1 antibodies. Clinical relevance was assessed by correlating circulating levels of TRAPs and NETs with lung metastasis in patients with breast cancer. RESULTS This study showed that TRAPs induced the formation of NETs in both human and mouse neutrophils by using the high mobility group box 1 and activating the TLR4-Myd88-ERK/p38 signaling axis. More importantly, PD-L1 carried by TRAP-induced NETs inhibited T-cell function in vitro and in vivo, thereby contributing to the formation of lung premetastatic niche (PMN) immunosuppression. In contrast, Becn1 KD-4T1 breast tumors with decreased circulating TRAPs in vivo reduced the formation of NETs, which in turn attenuated the immunosuppressive effects in PMN and resulted in a reduction of breast cancer pulmonary metastasis in murine models. Moreover, treatment with αPD-L1 in combination with DNase I that degraded NETs restored T-cell function and significantly reduced tumor metastasis. TRAP levels in the peripheral blood positively correlated with NET levels and lung metastasis in patients with breast cancer. CONCLUSIONS Our results demonstrate a novel role of TRAPs in the formation of PD-L1-decorated NETs, which may provide a new strategy for early detection and treatment of pulmonary metastasis in patients with breast cancer.
Collapse
Affiliation(s)
- Xiaohe Zhou
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Microbiology and Immunology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Chengdong Wu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Microbiology and Immunology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xuru Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Microbiology and Immunology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Ning Pan
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Microbiology and Immunology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xiaotong Sun
- Department of Laboratory Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Bohao Chen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Microbiology and Immunology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Shiya Zheng
- Department of Oncology, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
| | - Yiting Wei
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Microbiology and Immunology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Jing Chen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Microbiology and Immunology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Yuyang Wu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Microbiology and Immunology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Fengjiao Zhu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Microbiology and Immunology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Jinpeng Chen
- Department of general surgery, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
| | - Huabiao Chen
- Institute of Biomedical Engineering and Technology, School of Medicine, Ningbo University, Ningbo, China
| | - Li-xin Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Microbiology and Immunology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Mallin MM, Rolle LTA, Pienta KJ, Amend SR. Multiparameter flow cytometric detection and analysis of rare cells in in vivo models of cancer metastasis. Biol Methods Protoc 2024; 9:bpae026. [PMID: 38737789 PMCID: PMC11088742 DOI: 10.1093/biomethods/bpae026] [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: 04/03/2024] [Revised: 04/12/2024] [Accepted: 04/25/2024] [Indexed: 05/14/2024] Open
Abstract
Rapid and reliable circulating tumor cell (CTC) and disseminated tumor cell (DTC) detection are critical for rigorous evaluation of in vivo metastasis models. Clinical data show that each step of the metastatic cascade presents increasing barriers to success, limiting the number of successful metastatic cells to fewer than 1 in 1,500,000,000. As such, it is critical for scientists to employ approaches that allow for the evaluation of metastatic competency at each step of the cascade. Here, we present a flow cytometry-based method that enables swift and simultaneous comparison of both CTCs and DTCs from single animals, enabling evaluation of multiple metastatic steps within a single model system. We present the necessary gating strategy and optimized sample preparation conditions necessary to capture CTCs and DTCs using this approach. We also provide proof-of-concept experiments emphasizing the appropriate limits of detection of these conditions. Most importantly, we successfully recover CTCs and DTCs from murine blood and bone marrow. In Supplemental materials, we expand the applicability of our method to lung tissue and exemplify a potential multi-plexing strategy to further characterize recovered CTCs and DTCs. This approach to multiparameter flow cytometric detection and analysis of rare cells in in vivo models of metastasis is reproducible, high throughput, broadly applicable, and highly adaptable to a wide range of scientific inquiries. Most notably, it simplifies the recovery and analysis of CTCs and DTCs from the same animal, allowing for a rapid first look at the comparative metastatic competency of various model systems throughout multiple steps of the metastatic cascade.
Collapse
Affiliation(s)
- Mikaela M Mallin
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, United States
- Cellular and Molecular Medicine Graduate Training Program, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Louis T A Rolle
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, United States
| | - Kenneth J Pienta
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, United States
| | - Sarah R Amend
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, United States
| |
Collapse
|
6
|
Butler G, Amend SR, Axelrod R, Venditti C, Pienta KJ. Punctuational evolution is pervasive in distal site metastatic colonization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588529. [PMID: 38645078 PMCID: PMC11030309 DOI: 10.1101/2024.04.08.588529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The evolution of metastasis represents a lethal stage of cancer progression. Yet, the evolutionary kinetics of metastatic disease remain unresolved. Here, using single cell CRISPR-Cas9 lineage tracing data, we show that in metastatic disease, gradual molecular evolution is punctuated by episodes of rapid evolutionary change associated with lineage divergence. By measuring punctuational effects across the metastatic cascade, we show that punctuational effects contribute more to the molecular diversity at distal site metastases compared to the paired primary tumor, suggesting qualitatively different modes of evolution may drive primary and metastatic tumor progression. This is the first empirical evidence for distinct patterns of molecular evolution at early and late stages of metastasis and demonstrates the complex interplay of cell intrinsic and extrinsic factors that shape lethal cancer.
Collapse
Affiliation(s)
- George Butler
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sarah R. Amend
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Robert Axelrod
- Geral R. Ford School of Public Policy, University of Michigan, Ann Arbor, MI, USA
| | - Chris Venditti
- School of Biological Sciences, University of Reading, Reading, UK
| | - Kenneth J. Pienta
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
7
|
Cao Y, Meng F, Cai T, Gao L, Lee J, Solomevich SO, Aharodnikau UE, Guo T, Lan M, Liu F, Li Q, Viktor T, Li D, Cai Y. Nanoparticle drug delivery systems responsive to tumor microenvironment: Promising alternatives in the treatment of triple-negative breast cancer. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1950. [PMID: 38528388 DOI: 10.1002/wnan.1950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/04/2024] [Accepted: 02/11/2024] [Indexed: 03/27/2024]
Abstract
The conventional therapeutic treatment of triple-negative breast cancer (TNBC) is negatively influenced by the development of tumor cell drug resistant, and systemic toxicity of therapeutic agents due to off-target activity. In accordance with research findings, nanoparticles (NPs) responsive to the tumor microenvironment (TME) have been discovered for providing opportunities to selectively target tumor cells via active targeting or Enhanced Permeability and Retention (EPR) effect. The combination of the TME control and therapeutic NPs offers promising solutions for improving the prognosis of the TNBC because the TME actively participates in tumor growth, metastasis, and drug resistance. The NP-based systems leverage stimulus-responsive mechanisms, such as low pH value, hypoxic, excessive secretion enzyme, concentration of glutathione (GSH)/reactive oxygen species (ROS), and high concentration of Adenosine triphosphate (ATP) to combat TNBC progression. Concurrently, NP-based stimulus-responsive introduces a novel approach for drug dosage design, administration, and modification of the pharmacokinetics of conventional chemotherapy and immunotherapy drugs. This review provides a comprehensive examination of the strengths, limitations, applications, perspectives, and future expectations of both novel and traditional stimulus-responsive NP-based drug delivery systems for improving outcomes in the medical practice of TNBC. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Ye Cao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/Guangdong Key Lab of Traditional Chinese Medicine Informatization/International Science and Technology Cooperation Base of Guangdong Province/School of Pharmacy, Jinan University, Guangzhou, China
| | - Fansu Meng
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, China
| | - Tiange Cai
- College of Life Sciences, Liaoning University, Shenyang, China
| | - Lanwen Gao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/Guangdong Key Lab of Traditional Chinese Medicine Informatization/International Science and Technology Cooperation Base of Guangdong Province/School of Pharmacy, Jinan University, Guangzhou, China
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sergey O Solomevich
- Research Institute for Physical Chemical Problems of the Belarusian State University, Minsk, Belarus
| | - Uladzislau E Aharodnikau
- Research Institute for Physical Chemical Problems of the Belarusian State University, Minsk, Belarus
| | - Tingting Guo
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/Guangdong Key Lab of Traditional Chinese Medicine Informatization/International Science and Technology Cooperation Base of Guangdong Province/School of Pharmacy, Jinan University, Guangzhou, China
| | - Meng Lan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/Guangdong Key Lab of Traditional Chinese Medicine Informatization/International Science and Technology Cooperation Base of Guangdong Province/School of Pharmacy, Jinan University, Guangzhou, China
| | - Fengjie Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/Guangdong Key Lab of Traditional Chinese Medicine Informatization/International Science and Technology Cooperation Base of Guangdong Province/School of Pharmacy, Jinan University, Guangzhou, China
| | - Qianwen Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/Guangdong Key Lab of Traditional Chinese Medicine Informatization/International Science and Technology Cooperation Base of Guangdong Province/School of Pharmacy, Jinan University, Guangzhou, China
| | - Timoshenko Viktor
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia
| | - Detang Li
- The First Clinical Medical School of Guangzhou University of Chinese Medicine/Department of Pharmacy, The First Affiliated Hospital of Guangzhou University of Chinese Medicine/Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Yu Cai
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/Guangdong Key Lab of Traditional Chinese Medicine Informatization/International Science and Technology Cooperation Base of Guangdong Province/School of Pharmacy, Jinan University, Guangzhou, China
| |
Collapse
|
8
|
Zieren RC, Zondervan PJ, Pienta KJ, Bex A, de Reijke TM, Bins AD. Diagnostic liquid biopsy biomarkers in renal cell cancer. Nat Rev Urol 2024; 21:133-157. [PMID: 37758847 DOI: 10.1038/s41585-023-00818-y] [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] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
The clinical presentation of renal cell cancer (RCC) is shifting towards incidental and early detection, creating new challenges in RCC diagnosis. Overtreatment might be reduced with the development of new diagnostic biomarkers to distinguish benign from malignant small renal masses (SRMs). Differently from tissue biopsies, liquid biopsies are obtained from a patient's blood or urine and, therefore, are minimally invasive and suitable for longitudinal monitoring. The most promising types of liquid biopsy biomarkers for RCC diagnosis are circulating tumour cells, extracellular vesicles (EVs) and cell-free DNA. Circulating tumour cell assays have the highest specificity, with low processing time and costs. However, the biological characteristics and low sensitivity limit the use of these markers in SRM diagnostics. Cell-free DNA might complement the diagnosis of high-volume RCC, but the potential for clinical application in SRMs is limited. EVs have the highest biological abundance and the highest sensitivity in identifying low-volume disease; moreover, the molecular characteristics of these markers make EVs suitable for multiple analytical applications. Thus, currently, EV assays have the greatest potential for diagnostic application in RCC (including identification of SRMs). All these liquid biomarkers have potential in clinical practice, pending validation studies. Biomarker implementation will be needed to also improve characterization of RCC subtypes. Last, diagnostic biomarkers might be extended to prognostic or predictive applications.
Collapse
Affiliation(s)
- Richard C Zieren
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Patricia J Zondervan
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Kenneth J Pienta
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Axel Bex
- Specialist Centre for Kidney Cancer, Royal Free Hospital, London, United Kingdom
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
- The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Theo M de Reijke
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Adriaan D Bins
- Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Jiao Y, Yu Y, Zheng M, Yan M, Wang J, Zhang Y, Zhang S. Dormant cancer cells and polyploid giant cancer cells: The roots of cancer recurrence and metastasis. Clin Transl Med 2024; 14:e1567. [PMID: 38362620 PMCID: PMC10870057 DOI: 10.1002/ctm2.1567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Tumour cell dormancy is critical for metastasis and resistance to chemoradiotherapy. Polyploid giant cancer cells (PGCCs) with giant or multiple nuclei and high DNA content have the properties of cancer stem cell and single PGCCs can individually generate tumours in immunodeficient mice. PGCCs represent a dormant form of cancer cells that survive harsh tumour conditions and contribute to tumour recurrence. Hypoxic mimics, chemotherapeutics, radiation and cytotoxic traditional Chinese medicines can induce PGCCs formation through endoreduplication and/or cell fusion. After incubation, dormant PGCCs can recover from the treatment and produce daughter cells with strong proliferative, migratory and invasive abilities via asymmetric cell division. Additionally, PGCCs can resist hypoxia or chemical stress and have a distinct protein signature that involves chromatin remodelling and cell cycle regulation. Dormant PGCCs form the cellular basis for therapeutic resistance, metastatic cascade and disease recurrence. This review summarises regulatory mechanisms governing dormant cancer cells entry and exit of dormancy, which may be used by PGCCs, and potential therapeutic strategies for targeting PGCCs.
Collapse
Affiliation(s)
- Yuqi Jiao
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Yongjun Yu
- Department of PathologyTianjin Union Medical CenterTianjinChina
| | - Minying Zheng
- Department of PathologyTianjin Union Medical CenterNankai UniversityTianjinChina
| | - Man Yan
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Jiangping Wang
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Yue Zhang
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Shiwu Zhang
- Department of PathologyTianjin Union Medical CenterTianjinChina
| |
Collapse
|
10
|
Castillo SP, Rebolledo RA, Arim M, Hochberg ME, Marquet PA. Metastatic cells exploit their stoichiometric niche in the network of cancer ecosystems. SCIENCE ADVANCES 2023; 9:eadi7902. [PMID: 38091399 PMCID: PMC10848726 DOI: 10.1126/sciadv.adi7902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023]
Abstract
Metastasis is a nonrandom process with varying degrees of organotropism-specific source-acceptor seeding. Understanding how patterns between source and acceptor tumors emerge remains a challenge in oncology. We hypothesize that organotropism results from the macronutrient niche of cells in source and acceptor organs. To test this, we constructed and analyzed a metastatic network based on 9303 records across 28 tissue types. We found that the topology of the network is nested and modular with scale-free degree distributions, reflecting organotropism along a specificity/generality continuum. The variation in topology is significantly explained by the matching of metastatic cells to their stoichiometric niche. Specifically, successful metastases are associated with higher phosphorus content in the acceptor compared to the source organ, due to metabolic constraints in proliferation crucial to the invasion of new tissues. We conclude that metastases are codetermined by processes at source and acceptor organs, where phosphorus content is a limiting factor orchestrating tumor ecology.
Collapse
Affiliation(s)
- Simon P. Castillo
- Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, C.P. 8331150, Santiago, Chile
| | - Rolando A. Rebolledo
- Instituto de Ingeniería Biológica y Médica (IIBM), Pontificia Universidad Católica de Chile, Santiago, Chile
- Hepato-Pancreato-Biliary Surgery Unit, Surgery Service, Complejo Asistencial Dr. Sótero Del Río, Santiago, Chile
| | - Matías Arim
- Departamento de Ecologia y Gestion Ambiental, Centro Universitario Regional Este (CURE), Universidad de la República, Maldonado, Uruguay
| | - Michael E. Hochberg
- ISEM, University of Montpellier, Montpellier, France
- Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Pablo A. Marquet
- Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, C.P. 8331150, Santiago, Chile
- Santa Fe Institute, Santa Fe, NM 87501, USA
- Centro de Modelamiento Matemático, Universidad de Chile, International Research Laboratory 2807, CNRS, C.P. 8370456, Santiago, Chile
- Instituto de Sistemas Complejos de Valparaíso (ISCV), Valparaíso, Chile
| |
Collapse
|
11
|
Mallin MM, Kim N, Choudhury MI, Lee SJ, An SS, Sun SX, Konstantopoulos K, Pienta KJ, Amend SR. Cells in the polyaneuploid cancer cell (PACC) state have increased metastatic potential. Clin Exp Metastasis 2023:10.1007/s10585-023-10216-8. [PMID: 37326720 DOI: 10.1007/s10585-023-10216-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 06/02/2023] [Indexed: 06/17/2023]
Abstract
Although metastasis is the leading cause of cancer deaths, it is quite rare at the cellular level. Only a rare subset of cancer cells (~ 1 in 1.5 billion) can complete the entire metastatic cascade: invasion, intravasation, survival in the circulation, extravasation, and colonization (i.e. are metastasis competent). We propose that cells engaging a Polyaneuploid Cancer Cell (PACC) phenotype are metastasis competent. Cells in the PACC state are enlarged, endocycling (i.e. non-dividing) cells with increased genomic content that form in response to stress. Single-cell tracking using time lapse microscopy reveals that PACC state cells have increased motility. Additionally, cells in the PACC state exhibit increased capacity for environment-sensing and directional migration in chemotactic environments, predicting successful invasion. Magnetic Twisting Cytometry and Atomic Force Microscopy reveal that cells in the PACC state display hyper-elastic properties like increased peripheral deformability and maintained peri-nuclear cortical integrity that predict successful intravasation and extravasation. Furthermore, four orthogonal methods reveal that cells in the PACC state have increased expression of vimentin, a hyper-elastic biomolecule known to modulate biomechanical properties and induce mesenchymal-like motility. Taken together, these data indicate that cells in the PACC state have increased metastatic potential and are worthy of further in vivo analysis.
Collapse
Affiliation(s)
- Mikaela M Mallin
- Cellular and Molecular Medicine Graduate Training Program, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, USA.
| | - Nicholas Kim
- Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ, USA
| | | | - Se Jong Lee
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Steven S An
- Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ, USA
| | - Sean X Sun
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | | - Kenneth J Pienta
- Cellular and Molecular Medicine Graduate Training Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Sarah R Amend
- Cellular and Molecular Medicine Graduate Training Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Cancer Ecology Center, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD, USA
| |
Collapse
|
12
|
Chhabra Y, Weeraratna AT. Fibroblasts in cancer: Unity in heterogeneity. Cell 2023; 186:1580-1609. [PMID: 37059066 PMCID: PMC11422789 DOI: 10.1016/j.cell.2023.03.016] [Citation(s) in RCA: 129] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 04/16/2023]
Abstract
Tumor cells do not exist in isolation in vivo, and carcinogenesis depends on the surrounding tumor microenvironment (TME), composed of a myriad of cell types and biophysical and biochemical components. Fibroblasts are integral in maintaining tissue homeostasis. However, even before a tumor develops, pro-tumorigenic fibroblasts in close proximity can provide the fertile 'soil' to the cancer 'seed' and are known as cancer-associated fibroblasts (CAFs). In response to intrinsic and extrinsic stressors, CAFs reorganize the TME enabling metastasis, therapeutic resistance, dormancy and reactivation by secreting cellular and acellular factors. In this review, we summarize the recent discoveries on CAF-mediated cancer progression with a particular focus on fibroblast heterogeneity and plasticity.
Collapse
Affiliation(s)
- Yash Chhabra
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Department of Oncology, Sidney Kimmel Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Department of Oncology, Sidney Kimmel Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
13
|
Deng Y, Lu L, Zhang H, Fu Y, Liu T, Chen Y. The role and regulation of Maf proteins in cancer. Biomark Res 2023; 11:17. [PMID: 36750911 PMCID: PMC9903618 DOI: 10.1186/s40364-023-00457-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/22/2023] [Indexed: 02/09/2023] Open
Abstract
The Maf proteins (Mafs) belong to basic leucine zipper transcription factors and are members of the activator protein-1 (AP-1) superfamily. There are two subgroups of Mafs: large Mafs and small Mafs, which are involved in a wide range of biological processes, such as the cell cycle, proliferation, oxidative stress, and inflammation. Therefore, dysregulation of Mafs can affect cell fate and is closely associated with diverse diseases. Accumulating evidence has established both large and small Mafs as mediators of tumor development. In this review, we first briefly describe the structure and physiological functions of Mafs. Then we summarize the upstream regulatory mechanisms that control the expression and activity of Mafs. Furthermore, we discuss recent studies on the critical role of Mafs in cancer progression, including cancer proliferation, apoptosis, metastasis, tumor/stroma interaction and angiogenesis. We also review the clinical implications of Mafs, namely their potential possibilities and limitations as biomarkers and therapeutic targets in cancer.
Collapse
Affiliation(s)
- Yalan Deng
- grid.452223.00000 0004 1757 7615Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Liqing Lu
- grid.452223.00000 0004 1757 7615Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China ,grid.452223.00000 0004 1757 7615Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Huajun Zhang
- grid.452223.00000 0004 1757 7615Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China ,grid.452223.00000 0004 1757 7615Department of Ultrasonic Imaging, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Ying Fu
- grid.452223.00000 0004 1757 7615Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Ting Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
14
|
Yao M, Liang S, Cheng B. Role of exosomes in hepatocellular carcinoma and the regulation of traditional Chinese medicine. Front Pharmacol 2023; 14:1110922. [PMID: 36733504 PMCID: PMC9886889 DOI: 10.3389/fphar.2023.1110922] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/03/2023] [Indexed: 01/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) usually occurs on the basis of chronic liver inflammatory diseases and cirrhosis. The liver microenvironment plays a vital role in the tumor initiation and progression. Exosomes, which are nanometer-sized membrane vesicles are secreted by a number of cell types. Exosomes carry multiple proteins, DNAs and various forms of RNA, and are mediators of cell-cell communication and regulate the tumor microenvironment. In the recent decade, many studies have demonstrated that exosomes are involved in the communication between HCC cells and the stromal cells, including endothelial cells, macrophages, hepatic stellate cells and the immune cells, and serve as a regulator in the tumor proliferation and metastasis, immune evasion and immunotherapy. In addition, exosomes can also be used for the diagnosis and treatment HCC. They can potentially serve as specific biomarkers for early diagnosis and drug delivery vehicles of HCC. Chinese herbal medicine, which is widely used in the prevention and treatment of HCC in China, may regulate the release of exosomes and exosomes-mediated intercellular communication. In this review, we summarized the latest progresses on the role of the exosomes in the initiation, progression and treatment of HCC and the potential value of Traditional Chinese medicine in exosomes-mediated biological behaviors of HCC.
Collapse
Affiliation(s)
- Man Yao
- Oncology Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University (The Second Military Medical University), Shanghai, China
| | - Shufang Liang
- Oncology Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University (The Second Military Medical University), Shanghai, China
| | - Binbin Cheng
- Oncology Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University (The Second Military Medical University), Shanghai, China,Faculty of Traditional Chinese Medicine, Naval Medical University (The Second Military Medical University), Shanghai, China,*Correspondence: Binbin Cheng,
| |
Collapse
|
15
|
Pontis F, Roz L, Fortunato O, Bertolini G. The metastatic niche formation: focus on extracellular vesicle-mediated dialogue between lung cancer cells and the microenvironment. Front Oncol 2023; 13:1116783. [PMID: 37207158 PMCID: PMC10189117 DOI: 10.3389/fonc.2023.1116783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/21/2023] [Indexed: 05/21/2023] Open
Abstract
Lung cancer is the deadliest cancer in the world, with the majority of patients presenting with advanced or metastatic disease at first diagnosis. The lungs are also one of the most common sites of metastasis from lung cancer and other tumors. Understanding the mechanisms that regulate metastasis formation from primary lung cancer and in the lungs is therefore fundamental unmet clinical need. One of the first steps during the establishment of lung cancer metastases includes the formation of the pre-metastatic niche (PMN) at distant organs, which may occur even during the early phases of cancer development. The PMN is established through intricate cross-talk between primary tumor-secreted factors and stromal components at distant sites. Mechanisms controlling primary tumor escape and seeding of distant organs rely on specific properties of tumor cells but are also tightly regulated by interactions with stromal cells at the metastatic niche that finally dictate the success of metastasis establishment. Here, we summarize the mechanisms underlying pre-metastatic niche formation starting from how lung primary tumor cells modulate distant sites through the release of several factors, focusing on Extracellular Vesicles (EVs). In this context, we highlight the role of lung cancer-derived EVs in the modulation of tumor immune escape. Then, we illustrate the complexity of Circulating Tumor Cells (CTCs) that represent the seeds of metastasis and how interactions with stromal and immune cells can help their metastatic dissemination. Finally, we evaluate the contribution of EVs in dictating metastasis development at the PMN through stimulation of proliferation and control of disseminated tumor cell dormancy. Overall, we present an overview of different steps in the lung cancer metastatic cascade, focusing on the EV-mediated interactions between tumor cells and stromal/immune cells.
Collapse
|
16
|
Pardella E, Pranzini E, Nesi I, Parri M, Spatafora P, Torre E, Muccilli A, Castiglione F, Fambrini M, Sorbi F, Cirri P, Caselli A, Puhr M, Klocker H, Serni S, Raugei G, Magherini F, Taddei ML. Therapy-Induced Stromal Senescence Promoting Aggressiveness of Prostate and Ovarian Cancer. Cells 2022; 11:cells11244026. [PMID: 36552790 PMCID: PMC9776582 DOI: 10.3390/cells11244026] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Cancer progression is supported by the cross-talk between tumor cells and the surrounding stroma. In this context, senescent cells in the tumor microenvironment contribute to the development of a pro-inflammatory milieu and the acquisition of aggressive traits by cancer cells. Anticancer treatments induce cellular senescence (therapy-induced senescence, TIS) in both tumor and non-cancerous cells, contributing to many detrimental side effects of therapies. Thus, we focused on the effects of chemotherapy on the stromal compartment of prostate and ovarian cancer. We demonstrated that anticancer chemotherapeutics, regardless of their specific mechanism of action, promote a senescent phenotype in stromal fibroblasts, resulting in metabolic alterations and secretion of paracrine factors, sustaining the invasive and clonogenic potential of both prostate and ovarian cancer cells. The clearance of senescent stromal cells, through senolytic drug treatment, reverts the malignant phenotype of tumor cells. The clinical relevance of TIS was validated in ovarian and prostate cancer patients, highlighting increased accumulation of lipofuscin aggregates, a marker of the senescent phenotype, in the stromal compartment of tissues from chemotherapy-treated patients. These data provide new insights into the potential efficacy of combining traditional anticancer strategies with innovative senotherapy to potentiate anticancer treatments and overcome the adverse effects of chemotherapy.
Collapse
Affiliation(s)
- Elisa Pardella
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Ilaria Nesi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Matteo Parri
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Pietro Spatafora
- Department of Minimally Invasive and Robotic Urologic Surgery and Kidney Transplantation, University of Florence, 50134 Florence, Italy
| | - Eugenio Torre
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Angela Muccilli
- Department of Health Sciences, Section of Pathology, University of Florence, 50134 Florence, Italy
| | - Francesca Castiglione
- Histopathology and Molecular Diagnostics, Careggi Teaching Hospital, 50134 Florence, Italy
| | - Massimiliano Fambrini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Flavia Sorbi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Paolo Cirri
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Anna Caselli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Martin Puhr
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Helmut Klocker
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Sergio Serni
- Department of Minimally Invasive and Robotic Urologic Surgery and Kidney Transplantation, University of Florence, 50134 Florence, Italy
| | - Giovanni Raugei
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Francesca Magherini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Medicine, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
- Correspondence:
| |
Collapse
|
17
|
Wang M, Qin Z, Wan J, Yan Y, Duan X, Yao X, Jiang Z, Li W, Qin Z. Tumor-derived exosomes drive pre-metastatic niche formation in lung via modulating CCL1+ fibroblast and CCR8+ Treg cell interactions. Cancer Immunol Immunother 2022; 71:2717-2730. [DOI: 10.1007/s00262-022-03196-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/25/2022] [Indexed: 10/18/2022]
|
18
|
Yue Y, Liang J, Wu Y, Tong W, Li D, Cao X, Wang X. A Nomogram for Predicting Liver Metastasis of Lymph-Node Positive Luminal B HER2 Negative Subtype Breast Cancer by Analyzing the Clinicopathological Characteristics of Patients with Breast Cancer. Technol Cancer Res Treat 2022; 21:15330338221132669. [PMID: 36254567 PMCID: PMC9580102 DOI: 10.1177/15330338221132669] [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] [Indexed: 01/19/2023] Open
Abstract
Background: Luminal B-like human epidermal growth factor receptor 2 negative (Luminal B [HER2-]) is the most common molecular subtype of breast cancer (BC). Since the relationship between Luminal B (HER2-) BC and liver metastasis (LM) is poorly defined, this retrospective study aimed to develop an LM risk nomogram for patients with lymph node-related (N + Luminal B [HER2-]) BC. Methods: Data were obtained for patients initially diagnosed with BC from the Tianjin Medical University Cancer Institute and Hospital. There were 30,975 Chinese female patients with stage I-III BC and follow-up confirming 1217 subsequent patients with LM, and 427 patients with N + Luminal B (HER2-). The LM risk was assessed using Cox proportional hazards regression, histogram, Venn diagram, and Kaplan-Meier survival analysis, with further analysis for patients with N + Luminal B (HER2-) BC. A nomogram was established based on the N + Luminal B (HER2-) BC data, which was validated using calibration plots. Results: The median age of 427 patients with N + Luminal B (HER2-) liver metastasis of breast cancer (BCLM) was 49 years. The largest number of patients with BCLM was diagnosed between the second to the 6th year, the longest interval from initial BC diagnosis to subsequent LM was 145 months. The patients with LM as the first site of distant metastasis which is associated with better survival were analyzed by Kaplan-Meier. The nomogram was constructed for the risk of LM that included age, menstrual status, unilateral oophorectomy, pregnancy, hepatitis B antigen, region of residence, tumor size, lymph node, clavicular lymph nodes, progesterone receptor, and lymph vessel invasion. Conclusion: We described the clinicopathological characteristics of patients with stage I-III BC, and constructed a nomogram for calculating personalized LM probabilities for patients with N + Luminal B (HER2-), which could guide future prolonged or early extensive treatment decisions.
Collapse
Affiliation(s)
- Yuhan Yue
- First Department of Breast cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin, China,Department of Breast Tumor Center, Affiliated People's Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia Autonomous Region, China
| | - Junqing Liang
- Department of Breast Tumor Center, Affiliated People's Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia Autonomous Region, China,Department of cytotherapy for tumors, Affiliated People's Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia Autonomous Region, China
| | - Yuruo Wu
- Department of cytotherapy for tumors, Affiliated People's Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia Autonomous Region, China
| | - Weibing Tong
- Department of Breast Tumor Center, Affiliated People's Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia Autonomous Region, China
| | - Dan Li
- First Department of Breast cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin, China
| | - Xuchen Cao
- First Department of Breast cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin, China
| | - Xin Wang
- First Department of Breast cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin, China,Xin Wang, First Department of Breast cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 300060, China.
| |
Collapse
|
19
|
The oligometastatic spectrum in the era of improved detection and modern systemic therapy. Nat Rev Clin Oncol 2022; 19:585-599. [PMID: 35831494 DOI: 10.1038/s41571-022-00655-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 12/11/2022]
Abstract
Metastases remain the leading cause of cancer-related mortality. The oligometastasis hypothesis postulates that a spectrum of metastatic spread exists and that some patients with a limited burden of metastases can be cured with ablative therapy. Over the past decade, substantial advances in systemic therapies have resulted in considerable improvements in the outcomes of patients with metastatic cancers, warranting re-examination of the oligometastatic paradigm and the role of local ablative therapies within the context of the improved therapeutic responses, shifting patterns of disease recurrence and possible synergy with systemic treatments. Herein, we reframe the oligometastatic phenotype as a dynamic state for which locally ablative, metastasis-directed therapy improves clinical outcomes, including by prolonging survival and increasing cure rates. Important risk factors defining the metastatic spectrum are highlighted that inform both staging and therapy. Finally, we synthesize the literature on combining local therapies with modern systemic treatments, identifying general themes to optimally integrate ablative therapies in this context.
Collapse
|
20
|
Mallin MM, Pienta KJ, Amend SR. Cancer cell foraging to explain bone-specific metastatic progression. Bone 2022; 158:115788. [PMID: 33279670 DOI: 10.1016/j.bone.2020.115788] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/18/2020] [Accepted: 12/01/2020] [Indexed: 01/06/2023]
Abstract
Metastatic cancer is lethal and patients who suffer bone metastases fare especially poorly. Bone-specific metastatic progression in prostate and breast cancers is a highly observed example of organ-specific metastasis, or organotropism. Though research has delineated the sequential steps of the metastatic cascade, the determinants of bone-specific metastasis have remained elusive for decades. Applying fundamental ecological principles to cancer biology models of metastasis provides novel insights into metastatic organotropism. We use critical concepts from foraging theory and movement ecology to propose that observed bone-specific metastasis is the result of habitat selection by foraging cancer cells. Furthermore, we posit that cancer cells can only perform habitat selection if and when they employ a reversible motile foraging strategy. Only a very small percentage of cells in a primary tumor harbor this ability. Therefore, our habitat selection model emphasizes the importance of identifying the rare subset of cancer cells that might exhibit habitat selection, ergo achieve bone-specific metastatic colonization.
Collapse
Affiliation(s)
- Mikaela M Mallin
- Cellular and Molecular Medicine Graduate Training Program, Johns Hopkins School of Medicine, 1830 E. Monument St. Suite 2-103, Baltimore, MD 21205, USA.
| | - Kenneth J Pienta
- The James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, 600 North Wolfe St., Marburg 105, Baltimore, MD 21287, USA
| | - Sarah R Amend
- The James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, 600 North Wolfe St., Marburg 105, Baltimore, MD 21287, USA
| |
Collapse
|
21
|
Tagirasa R, Yoo E. Role of Serine Proteases at the Tumor-Stroma Interface. Front Immunol 2022; 13:832418. [PMID: 35222418 PMCID: PMC8873516 DOI: 10.3389/fimmu.2022.832418] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/24/2022] [Indexed: 01/19/2023] Open
Abstract
During tumor development, invasion and metastasis, the intimate interaction between tumor and stroma shapes the tumor microenvironment and dictates the fate of tumor cells. Stromal cells can also influence anti-tumor immunity and response to immunotherapy. Understanding the molecular mechanisms that govern this complex and dynamic interplay, thus is important for cancer diagnosis and therapy. Proteolytic enzymes that are expressed and secreted by both cancer and stromal cells play important roles in modulating tumor-stromal interaction. Among, several serine proteases such as fibroblast activation protein, urokinase-type plasminogen activator, kallikrein-related peptidases, and granzymes have attracted great attention owing to their elevated expression and dysregulated activity in the tumor microenvironment. This review highlights the role of serine proteases that are mainly derived from stromal cells in tumor progression and associated theranostic applications.
Collapse
|
22
|
Abstract
Integration of ecological and evolutionary features has begun to understand the interplay of tumor heterogeneity, microenvironment, and metastatic potential. Developing a theoretical framework is intrinsic to deciphering tumors' tremendous spatial and longitudinal genetic variation patterns in patients. Here, we propose that tumors can be considered evolutionary island-like ecosystems, that is, isolated systems that undergo evolutionary and spatiotemporal dynamic processes that shape tumor microenvironments and drive the migration of cancer cells. We examine attributes of insular systems and causes of insularity, such as physical distance and connectivity. These properties modulate migration rates of cancer cells through processes causing spatial and temporal isolation of the organs and tissues functioning as a supply of cancer cells for new colonizations. We discuss hypotheses, predictions, and limitations of tumors as islands analogy. We present emerging evidence of tumor insularity in different cancer types and discuss their relevance to the islands model. We suggest that the engagement of tumor insularity into conceptual and mathematical models holds promise to illuminate cancer evolution, tumor heterogeneity, and metastatic potential of cells.
Collapse
Affiliation(s)
- Antonia Chroni
- Institute for Genomics and Evolutionary Medicine, Temple University, USA
- Department of Biology, Temple University, USA
| | - Sudhir Kumar
- Institute for Genomics and Evolutionary Medicine, Temple University, USA
- Department of Biology, Temple University, USA
- Center for Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
23
|
Neinavaie F, Ibrahim-Hashim A, Kramer AM, Brown JS, Richards CL. The Genomic Processes of Biological Invasions: From Invasive Species to Cancer Metastases and Back Again. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.681100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The concept of invasion is useful across a broad range of contexts, spanning from the fine scale landscape of cancer tumors up to the broader landscape of ecosystems. Invasion biology provides extraordinary opportunities for studying the mechanistic basis of contemporary evolution at the molecular level. Although the field of invasion genetics was established in ecology and evolution more than 50 years ago, there is still a limited understanding of how genomic level processes translate into invasive phenotypes across different taxa in response to complex environmental conditions. This is largely because the study of most invasive species is limited by information about complex genome level processes. We lack good reference genomes for most species. Rigorous studies to examine genomic processes are generally too costly. On the contrary, cancer studies are fortified with extensive resources for studying genome level dynamics and the interactions among genetic and non-genetic mechanisms. Extensive analysis of primary tumors and metastatic samples have revealed the importance of several genomic mechanisms including higher mutation rates, specific types of mutations, aneuploidy or whole genome doubling and non-genetic effects. Metastatic sites can be directly compared to primary tumor cell counterparts. At the same time, clonal dynamics shape the genomics and evolution of metastatic cancers. Clonal diversity varies by cancer type, and the tumors’ donor and recipient tissues. Still, the cancer research community has been unable to identify any common events that provide a universal predictor of “metastatic potential” which parallels findings in evolutionary ecology. Instead, invasion in cancer studies depends strongly on context, including order of events and clonal composition. The detailed studies of the behavior of a variety of human cancers promises to inform our understanding of genome level dynamics in the diversity of invasive species and provide novel insights for management.
Collapse
|
24
|
Li M, Gonye AL, Truskowski K, Loftus LV, Urbanski LA, Myers KV, Mallin MM, Yang ME, Mendez SA, Kostecka LG, Udedibor CR, Kim CJ, Kuczler MD, Shin GH, Amend SR, Pienta KJ. Twelve unanswered questions in cancer inspired by the life and work of Leland Chung: "if this is true, what does it imply"? AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2021; 9:254-260. [PMID: 34541023 PMCID: PMC8446763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 06/13/2023]
Affiliation(s)
- Melvin Li
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Anna Lk Gonye
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Kevin Truskowski
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Luke V Loftus
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Lanie A Urbanski
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Kayla V Myers
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Mikaela M Mallin
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Margaret E Yang
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Sabrina A Mendez
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Laurie G Kostecka
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Chiamaka R Udedibor
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Chi-Ju Kim
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Morgan D Kuczler
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Gloria H Shin
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Sarah R Amend
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| | - Kenneth J Pienta
- The Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD 21287, USA
| |
Collapse
|
25
|
Li Y, Jin J, Bai F. Cancer biology deciphered by single-cell transcriptomic sequencing. Protein Cell 2021; 13:167-179. [PMID: 34405376 PMCID: PMC8901819 DOI: 10.1007/s13238-021-00868-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
Tumors are complex ecosystems in which heterogeneous cancer cells interact with their microenvironment composed of diverse immune, endothelial, and stromal cells. Cancer biology had been studied using bulk genomic and gene expression profiling, which however mask the cellular diversity and average the variability among individual molecular programs. Recent advances in single-cell transcriptomic sequencing have enabled a detailed dissection of tumor ecosystems and promoted our understanding of tumorigenesis at single-cell resolution. In the present review, we discuss the main topics of recent cancer studies that have implemented single-cell RNA sequencing (scRNA-seq). To study cancer cells, scRNA-seq has provided novel insights into the cancer stem-cell model, treatment resistance, and cancer metastasis. To study the tumor microenvironment, scRNA-seq has portrayed the diverse cell types and complex cellular states of both immune and non-immune cells interacting with cancer cells, with the promise to discover novel targets for future immunotherapy.
Collapse
Affiliation(s)
- Yanmeng Li
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, 100871, China
| | - Jianshi Jin
- RIKEN Center for Biosystems Dynamics Research (BDR), 6-2-3, Furuedai, Suita, Osaka, Japan
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| |
Collapse
|
26
|
Paul S, Sa G. Curcumin as an Adjuvant to Cancer Immunotherapy. Front Oncol 2021; 11:675923. [PMID: 34485117 PMCID: PMC8415504 DOI: 10.3389/fonc.2021.675923] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/19/2021] [Indexed: 01/21/2023] Open
Abstract
The components of the immune system play a very sincere and crucial role in combating tumors. However, despite their firm efforts of elimination, tumor cells cleverly escape the surveillance process by adopting several immune evasion mechanisms. The conversion of immunogenicity of tumor microenvironment into tolerogenic is considered as a prime reason for tumor immune escape. Therapeutically, different immunotherapies have been adopted to block such immune escaping routes along with better clinical outcomes. Still, the therapies are haunted by several drawbacks. Over time, curcumin has been considered as a potential anti-cancer molecule. Its potentialities have been recorded against the standard hallmarks of cancer such as continuous proliferation, escaping apoptosis, continuous angiogenesis, insensitivity to growth inhibitors, tissue invasion, and metastasis. Hence, the diversity of curcumin functioning has already been established and exploration of its application with immunotherapies might open up a new avenue for scientists and clinicians. In this review, we briefly discuss the tumor's way of immune escaping, followed by various modern immunotherapies that have been used to encounter the escaping paths and their minute flaws. Finally, the conclusion has been drawn with the application of curcumin as a potential immune-adjuvant, which fearlessly could be used with immunotherapies for best outcomes.
Collapse
Affiliation(s)
| | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
27
|
Pressley M, Salvioli M, Lewis DB, Richards CL, Brown JS, Staňková K. Evolutionary Dynamics of Treatment-Induced Resistance in Cancer Informs Understanding of Rapid Evolution in Natural Systems. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.681121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Rapid evolution is ubiquitous in nature. We briefly review some of this quite broadly, particularly in the context of response to anthropogenic disturbances. Nowhere is this more evident, replicated and accessible to study than in cancer. Curiously cancer has been late - relative to fisheries, antibiotic resistance, pest management and evolution in human dominated landscapes - in recognizing the need for evolutionarily informed management strategies. The speed of evolution matters. Here, we employ game-theoretic modeling to compare time to progression with continuous maximum tolerable dose to that of adaptive therapy where treatment is discontinued when the population of cancer cells gets below half of its initial size and re-administered when the cancer cells recover, forming cycles with and without treatment. We show that the success of adaptive therapy relative to continuous maximum tolerable dose therapy is much higher if the population of cancer cells is defined by two cell types (sensitive vs. resistant in a polymorphic population). Additionally, the relative increase in time to progression increases with the speed of evolution. These results hold with and without a cost of resistance in cancer cells. On the other hand, treatment-induced resistance can be modeled as a quantitative trait in a monomorphic population of cancer cells. In that case, when evolution is rapid, there is no advantage to adaptive therapy. Initial responses to therapy are blunted by the cancer cells evolving too quickly. Our study emphasizes how cancer provides a unique system for studying rapid evolutionary changes within tumor ecosystems in response to human interventions; and allows us to contrast and compare this system to other human managed or dominated systems in nature.
Collapse
|
28
|
Cancer recurrence and lethality are enabled by enhanced survival and reversible cell cycle arrest of polyaneuploid cells. Proc Natl Acad Sci U S A 2021; 118:2020838118. [PMID: 33504594 PMCID: PMC7896294 DOI: 10.1073/pnas.2020838118] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We present a unifying theory to explain cancer recurrence, therapeutic resistance, and lethality. The basis of this theory is the formation of simultaneously polyploid and aneuploid cancer cells, polyaneuploid cancer cells (PACCs), that avoid the toxic effects of systemic therapy by entering a state of cell cycle arrest. The theory is independent of which of the classically associated oncogenic mutations have already occurred. PACCs have been generally disregarded as senescent or dying cells. Our theory states that therapeutic resistance is driven by PACC formation that is enabled by accessing a polyploid program that allows an aneuploid cancer cell to double its genomic content, followed by entry into a nondividing cell state to protect DNA integrity and ensure cell survival. Upon removal of stress, e.g., chemotherapy, PACCs undergo depolyploidization and generate resistant progeny that make up the bulk of cancer cells within a tumor.
Collapse
|
29
|
Aghdam MA, Tohidkia MR, Ghamghami E, Ahmadikhah A, Khanmahamadi M, Baradaran B, Mokhtarzadeh A. Implementation of a Design of Experiments to Improve Periplasmic Yield of Functional ScFv Antibodies in a Phage Display Platform. Adv Pharm Bull 2021; 12:583-592. [PMID: 35935041 PMCID: PMC9348535 DOI: 10.34172/apb.2022.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/01/2021] [Accepted: 07/02/2021] [Indexed: 11/16/2022] Open
Abstract
Purpose: Production of functional recombinant antibody fragments in the periplasm of E. coli is a prerequisite step to achieve sufficient reagent for preclinical studies. Thus, the cost-effective and lab-scale production of antibody fragments demands the optimization of culture conditions.
Methods: The culture conditions such as temperature, optical density (OD600) at induction, induction time, and IPTG concentration were investigated to optimize the functional expression of a phage-derived scFv molecule using a design of experiment (DoE). Additionally, the effects of different culture media and osmolyte supplements on the expression yield of scFv were examined.
Results: The developed 2FI regression model indicated the significant linear effect of the incubation temperature, the induction time, and the induction OD600 on the expression yield of functional scFv. Besides, the statistical analysis indicated that two significant interactions of the temperature/induction time and the temperature/induction OD600 significantly interplay to increase the yield. Further optimization showed that the expression level of functional scFv was the most optimal when the cultivation was undertaken either in the TB medium or in the presence of media supplements of 0.5 M sorbitol or 100 mM glycine betaine.
Conclusion: In the present study, for the first time, we successfully implemented DoE to comprehensively optimize the culture conditions for the expression of scFv molecules in a phage antibody display setting, where scFv molecules can be isolated from a tailor-made phage antibody library known as "Human Single Fold scFv Library I."
Collapse
Affiliation(s)
- Marjan Abri Aghdam
- Department of Biological Science, Faculty of Basic Science, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Mohammad Reza Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ghamghami
- Department of Biological Science, Faculty of Basic Science, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Asadollah Ahmadikhah
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, G.C Velenjak, Tehran, Iran
| | - Morteza Khanmahamadi
- Chemical Engineering Faculty, Sahand University of Technology, Sahand New Town, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
30
|
Breast Cancer Cell Re-Dissemination from Lung Metastases-A Mechanism for Enhancing Metastatic Burden. J Clin Med 2021; 10:jcm10112340. [PMID: 34071839 PMCID: PMC8199463 DOI: 10.3390/jcm10112340] [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: 04/24/2021] [Revised: 05/17/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022] Open
Abstract
Although metastatic disease is the primary cause of mortality in cancer patients, the mechanisms leading to overwhelming metastatic burden are still incompletely understood. Metastases are the endpoint of a series of multi-step events involving cancer cell intravasation, dissemination to distant organs, and outgrowth to metastatic colonies. Here we show, for the first-time, that breast cancer cells do not solely disseminate to distant organs from primary tumors and metastatic nodules in the lymph nodes, but also do so from lung metastases. Thus, our findings indicate that metastatic dissemination could continue even after the removal of the primary tumor. Provided that the re-disseminated cancer cells initiate growth upon arrival to distant sites, cancer cell re-dissemination from metastatic foci could be one of the crucial mechanisms leading to overt metastases and patient demise. Therefore, the development of new therapeutic strategies to block cancer cell re-dissemination would be crucial to improving survival of patients with metastatic disease.
Collapse
|
31
|
Chen WK, Wu ZG, Xiao YB, Wang QQ, Yu DD, Cai J, Zhou CF. Prognostic Value of Site-Specific Metastases and Therapeutic Roles of Surgery and Chemotherapy for Patients With Metastatic Renal Pelvis Cancer: A SEER Based Study. Technol Cancer Res Treat 2021; 20:15330338211004914. [PMID: 33929915 PMCID: PMC8111551 DOI: 10.1177/15330338211004914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background and Aims: There is a lack of research on metastatic renal pelvis cell carcinoma in the current literature. In this study, we aimed to detect distant metastatic patterns in renal pelvis cell carcinoma, and illustrated the affection of different metastatic sites, surgery to primary site and chemotherapy on prognosis outcomes in patients with diverse conditions. Methods: We collected data between 2010 and 2015 from the Surveillance, Epidemiology and End Results database. Kaplan–Meier analysis with log-rank test was used for survival comparisons. Multivariate Cox regression model was employed to analyze the effect of distant metastatic sites on overall survival (OS) and cancer-specific survival (CSS). Results: A total of 424 patients were included in the analysis, the median follow-up time was 5 months (interquartile range (IQR): 2-12) and 391 deaths (92.2%) in all patients were recorded. Among them, 192 (45.3%), 153 (36.1%), 137 (32.3%) and 127 (30.0%) patients were diagnosed with lung, bone, liver and brain metastases, respectively, while only 12 (2.8%) patients had brain metastases. The bi-organ, tri-organ and tetra-organ metastatic pattern was found in 135 (31.8%), 32 (7.5%) and 11 (2.6%) patients, respectively. The multivariate Cox analyses showed that distant lymph nodes (DL) metastases was not an independent prognostic factor for both OS and CSS (OS: Hazard ratios (HR) = 1.1, 95% CI = 0.8-1.4, P = 0.622; CSS: HR = 1.0, 95% CI = 0.8-1.3, P = 0.906). Besides, there was no significant difference of survival in patients with T3-T4 stage (OS: HR = 0.8, 95% CI = 0.5–1.2, P = 0.296; CSS: HR = 0.8, 95% CI = 0.5–1.2, P = 0.224), N2-3 stage (OS: HR = 0.8, 95% CI = 0.5–1.3, P = 0.351; CSS: HR = 0.7, 95% CI = 0.4–1.2, P = 0.259) and multi-organ metastases (OS: HR = 0.8, 95% CI = 0.5–1.3, P = 0.359; CSS: HR = 0.7, 95% CI = 0.4–1.2, P = 0.179) between surgery to primary site group and no-surgery to primary site group. Conclusion: we described the metastatic patterns of mRPCC and the prognosis outcomes of DL metastases, surgery to primary site and chemotherapy. Our findings provide more information for clinical therapeutic intervention and translational study designs.
Collapse
Affiliation(s)
- Wei-Kang Chen
- Department of Andrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Zhi-Gang Wu
- Department of Andrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Yun-Bei Xiao
- Department of Andrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Qin-Quan Wang
- Department of Andrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Dong-Dong Yu
- Department of Andrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Jian Cai
- Department of Andrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Chao-Feng Zhou
- Department of Andrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| |
Collapse
|
32
|
G-protein subunit gamma-4 expression has potential for detection, prediction and therapeutic targeting in liver metastasis of gastric cancer. Br J Cancer 2021; 125:220-228. [PMID: 33854208 DOI: 10.1038/s41416-021-01366-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 02/25/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The liver is the most common site for haematogenous metastasis of gastric cancer, and liver metastasis is fatal. METHODS We conducted a transcriptomic analysis between metastatic foci in the liver, primary tumour and adjacent tissues from gastric cancer patients with metastasis limited to the liver. We determined mRNA expression levels in tumour tissues of 300 patients with gastric cancer via quantitative RT-PCR. The oncogenic phenotypes of GNG4 were determined with knockdown, knockout and forced expression experiments. We established and compared subcutaneous and liver metastatic mouse xenograft models of gastric cancer to reveal the roles of GNG4 in tumorigenesis in the liver. RESULTS GNG4 was upregulated substantially in primary gastric cancer tissues as well as liver metastatic lesions. High levels of GNG4 in primary cancer tissues were associated with short overall survival and the likelihood of liver recurrence. Functional assays revealed that GNG4 promoted cancer cell proliferation, the cell cycle and adhesiveness. Tumour formation by GNG4-knockout cells was moderately reduced in the subcutaneous mouse model and strikingly attenuated in the liver metastasis mouse model. CONCLUSIONS GNG4 expression may provide better disease monitoring for liver metastasis, and GNG4 may be a novel candidate therapeutic target for liver metastasis.
Collapse
|
33
|
Dujon AM, Aktipis A, Alix‐Panabières C, Amend SR, Boddy AM, Brown JS, Capp J, DeGregori J, Ewald P, Gatenby R, Gerlinger M, Giraudeau M, Hamede RK, Hansen E, Kareva I, Maley CC, Marusyk A, McGranahan N, Metzger MJ, Nedelcu AM, Noble R, Nunney L, Pienta KJ, Polyak K, Pujol P, Read AF, Roche B, Sebens S, Solary E, Staňková K, Swain Ewald H, Thomas F, Ujvari B. Identifying key questions in the ecology and evolution of cancer. Evol Appl 2021; 14:877-892. [PMID: 33897809 PMCID: PMC8061275 DOI: 10.1111/eva.13190] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/24/2020] [Accepted: 12/26/2020] [Indexed: 12/17/2022] Open
Abstract
The application of evolutionary and ecological principles to cancer prevention and treatment, as well as recognizing cancer as a selection force in nature, has gained impetus over the last 50 years. Following the initial theoretical approaches that combined knowledge from interdisciplinary fields, it became clear that using the eco-evolutionary framework is of key importance to understand cancer. We are now at a pivotal point where accumulating evidence starts to steer the future directions of the discipline and allows us to underpin the key challenges that remain to be addressed. Here, we aim to assess current advancements in the field and to suggest future directions for research. First, we summarize cancer research areas that, so far, have assimilated ecological and evolutionary principles into their approaches and illustrate their key importance. Then, we assembled 33 experts and identified 84 key questions, organized around nine major themes, to pave the foundations for research to come. We highlight the urgent need for broadening the portfolio of research directions to stimulate novel approaches at the interface of oncology and ecological and evolutionary sciences. We conclude that progressive and efficient cross-disciplinary collaborations that draw on the expertise of the fields of ecology, evolution and cancer are essential in order to efficiently address current and future questions about cancer.
Collapse
Affiliation(s)
- Antoine M. Dujon
- School of Life and Environmental SciencesCentre for Integrative EcologyDeakin UniversityWaurn PondsVic.Australia
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRDMontpellierFrance
| | - Athena Aktipis
- Biodesign InstituteDepartment of PsychologyArizona State UniversityTempeAZUSA
| | - Catherine Alix‐Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH)University Medical Center of MontpellierMontpellierFrance
| | - Sarah R. Amend
- Brady Urological InstituteThe Johns Hopkins School of MedicineBaltimoreMDUSA
| | - Amy M. Boddy
- Department of AnthropologyUniversity of California Santa BarbaraSanta BarbaraCAUSA
| | - Joel S. Brown
- Department of Integrated MathematicsMoffitt Cancer CenterTampaFLUSA
| | - Jean‐Pascal Capp
- Toulouse Biotechnology InstituteINSA/University of ToulouseCNRSINRAEToulouseFrance
| | - James DeGregori
- Department of Biochemistry and Molecular GeneticsIntegrated Department of ImmunologyDepartment of PaediatricsDepartment of Medicine (Section of Hematology)University of Colorado School of MedicineAuroraCOUSA
| | - Paul Ewald
- Department of BiologyUniversity of LouisvilleLouisvilleKYUSA
| | - Robert Gatenby
- Department of RadiologyH. Lee Moffitt Cancer Center & Research InstituteTampaFLUSA
| | - Marco Gerlinger
- Translational Oncogenomics LabThe Institute of Cancer ResearchLondonUK
| | - Mathieu Giraudeau
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRDMontpellierFrance
- Littoral Environnement et Sociétés (LIENSs)UMR 7266CNRS‐Université de La RochelleLa RochelleFrance
| | | | - Elsa Hansen
- Center for Infectious Disease Dynamics, Biology DepartmentPennsylvania State UniversityUniversity ParkPAUSA
| | - Irina Kareva
- Mathematical and Computational Sciences CenterSchool of Human Evolution and Social ChangeArizona State UniversityTempeAZUSA
| | - Carlo C. Maley
- Arizona Cancer Evolution CenterBiodesign Institute and School of Life SciencesArizona State UniversityTempeAZUSA
| | - Andriy Marusyk
- Department of Cancer PhysiologyH Lee Moffitt Cancer Centre and Research InstituteTampaFLUSA
| | - Nicholas McGranahan
- Translational Cancer Therapeutics LaboratoryThe Francis Crick InstituteLondonUK
- Cancer Research UK Lung Cancer Centre of ExcellenceUniversity College London Cancer InstituteLondonUK
| | | | | | - Robert Noble
- Department of Biosystems Science and EngineeringETH ZurichBaselSwitzerland
- Department of Evolutionary Biology and Environmental StudiesUniversity of ZurichZurichSwitzerland
| | - Leonard Nunney
- Department of Evolution, Ecology, and Organismal BiologyUniversity of California RiversideRiversideCAUSA
| | - Kenneth J. Pienta
- Brady Urological InstituteThe Johns Hopkins School of MedicineBaltimoreMDUSA
| | - Kornelia Polyak
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMAUSA
- Department of MedicineHarvard Medical SchoolBostonMAUSA
| | - Pascal Pujol
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRDMontpellierFrance
- Centre Hospitalier Universitaire Arnaud de VilleneuveMontpellierFrance
| | - Andrew F. Read
- Center for Infectious Disease DynamicsHuck Institutes of the Life SciencesDepartments of Biology and EntomologyPennsylvania State UniversityUniversity ParkPAUSA
| | - Benjamin Roche
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRDMontpellierFrance
- Unité Mixte Internationale de Modélisation Mathématique et Informatique des Systèmes ComplexesUMI IRD/Sorbonne UniversitéUMMISCOBondyFrance
| | - Susanne Sebens
- Institute for Experimental Cancer Research Kiel University and University Hospital Schleswig‐HolsteinKielGermany
| | - Eric Solary
- INSERM U1287Gustave RoussyVillejuifFrance
- Faculté de MédecineUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - Kateřina Staňková
- Department of Data Science and Knowledge EngineeringMaastricht UniversityMaastrichtThe Netherlands
- Delft Institute of Applied MathematicsDelft University of TechnologyDelftThe Netherlands
| | | | - Frédéric Thomas
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRDMontpellierFrance
| | - Beata Ujvari
- School of Life and Environmental SciencesCentre for Integrative EcologyDeakin UniversityWaurn PondsVic.Australia
| |
Collapse
|
34
|
Fan B, Bu Z, Zhang J, Zong X, Ji X, Fu T, Jia Z, Zhang Y, Wu X. Phase II trial of prophylactic hyperthermic intraperitoneal chemotherapy in patients with locally advanced gastric cancer after curative surgery. BMC Cancer 2021; 21:216. [PMID: 33653317 PMCID: PMC7923597 DOI: 10.1186/s12885-021-07925-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/17/2021] [Indexed: 11/10/2022] Open
Abstract
Background HIPEC is an emerging procedure to treat peritoneal metastasis of gastric cancer. Data about HIPEC in locally advanced gastric cancer is scarce. The purpose of this trial is to evaluate the safety and toxicity of prophylactic HIPEC with cisplatin for patients with locally advanced gastric cancer. Methods From March 2015 to November 2016, a prospective, randomized phase II trial was conducted. After radical gastrectomy, patients in the experimental group underwent HIPEC with cisplatin followed by adjuvant chemotherapy with SOX regime. Patients in the other group were treated with SOX regime alone. Postoperative complications and patient survival were compared. Results In total, 50 patients were eligible for analyses. No significant difference was found in the incidence of postoperative complications including anastomotic/intestinal leakage, liver dysfunction, bone marrow suppression, wound infection and ileus (P > 0.05). Mean duration of hospitalization after radical gastrectomy was 11.7 days. 12.2 days in experimental group and 10.8 days in control group respectively (P = 0.255). The percentage of patients with elevated tumor markers was 12.1% in experimental group, which was significantly lower than 41.2% in control group (P = 0.02). 3-year RFS of patients who treated with or without prophylactic HIPEC were 84.8 and 88.2% respectively (P = 0.986). In the multivariate analysis, pathological T stage was the only independent risk factor for the RFS of patients (P = 0.012, HR =15.071). Conclusion Additional intraoperative HIPEC with cisplatin did not increase postoperative complications for locally advanced gastric cancer after curative surgery. Prophylactic HIPEC with cisplatin was safe and tolerable, while it did not reduce the risk of peritoneal recurrence in this trial, supporting further studies to validate the efficacy of it. Trial registration Chinese Clinical Trial Registry, ChiCTR2000038331. Registered 18 September 2020 - Retrospectively registered, http://www.chictr.org.cn/showproj.aspx?proj=59692.
Collapse
Affiliation(s)
- Biao Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Zhaode Bu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Ji Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Xianglong Zong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Xin Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Tao Fu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Ziyu Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Yinan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Xiaojiang Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China.
| |
Collapse
|
35
|
Crean D, Murphy EP. Targeting NR4A Nuclear Receptors to Control Stromal Cell Inflammation, Metabolism, Angiogenesis, and Tumorigenesis. Front Cell Dev Biol 2021; 9:589770. [PMID: 33634114 PMCID: PMC7901948 DOI: 10.3389/fcell.2021.589770] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/06/2021] [Indexed: 12/17/2022] Open
Abstract
The NR4A1–NR4A3 (Nur77, Nurr1, and Nor-1) subfamily of nuclear receptors is a group of immediate early genes induced by a pleiotropy of stimuli including peptide hormones, growth factors, cytokines, inflammatory, and physiological stimuli, and cellular stress. NR4A receptors function as potent sensors of changes in the cellular microenvironment to control physiological and pathological processes through genomic and non-genomic actions. NR4A receptors control metabolism and cardiovascular and neurological functions and mediate immune cell homeostasis in inflammation and cancer. This receptor subfamily is increasingly recognized as an important molecular connection between chronic inflammation, altered immune cell responses, and cancer development. In this review, we examine how transcriptome analysis identified NR4A1/NR4A2 receptors as transcriptional regulators in mesenchymal stromal cell (MSC) migration, cell cycle progression, and cytokine production to control local immune responses. In chronic inflammatory conditions, such as rheumatoid arthritis, NR4A receptors have been shown to modify the activity of MSC and fibroblast-like stromal cells to regulate synovial tissue hyperplasia, pathological angiogenesis, and cartilage turnover in vivo. Additionally, as NR4A1 has been observed as a major transcriptional regulator in tumor–stromal communication controlling tumorigenesis, we discuss how advances in the pharmacological control of these receptors lead to important new mechanistic insights into understanding the role of the tumor microenvironment in health and disease.
Collapse
Affiliation(s)
- Daniel Crean
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Evelyn P Murphy
- School of Medicine, University of Limerick, Limerick, Ireland
| |
Collapse
|
36
|
Grzywa TM, Klicka K, Włodarski PK. Regulators at Every Step-How microRNAs Drive Tumor Cell Invasiveness and Metastasis. Cancers (Basel) 2020; 12:E3709. [PMID: 33321819 PMCID: PMC7763175 DOI: 10.3390/cancers12123709] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor cell invasiveness and metastasis are the main causes of mortality in cancer. Tumor progression is composed of many steps, including primary tumor growth, local invasion, intravasation, survival in the circulation, pre-metastatic niche formation, and metastasis. All these steps are strictly controlled by microRNAs (miRNAs), small non-coding RNA that regulate gene expression at the post-transcriptional level. miRNAs can act as oncomiRs that promote tumor cell invasion and metastasis or as tumor suppressor miRNAs that inhibit tumor progression. These miRNAs regulate the actin cytoskeleton, the expression of extracellular matrix (ECM) receptors including integrins and ECM-remodeling enzymes comprising matrix metalloproteinases (MMPs), and regulate epithelial-mesenchymal transition (EMT), hence modulating cell migration and invasiveness. Moreover, miRNAs regulate angiogenesis, the formation of a pre-metastatic niche, and metastasis. Thus, miRNAs are biomarkers of metastases as well as promising targets of therapy. In this review, we comprehensively describe the role of various miRNAs in tumor cell migration, invasion, and metastasis.
Collapse
Affiliation(s)
- Tomasz M. Grzywa
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Klaudia Klicka
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Paweł K. Włodarski
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
| |
Collapse
|
37
|
Cancer cells employ an evolutionarily conserved polyploidization program to resist therapy. Semin Cancer Biol 2020; 81:145-159. [PMID: 33276091 DOI: 10.1016/j.semcancer.2020.11.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/24/2022]
Abstract
Unusually large cancer cells with abnormal nuclei have been documented in the cancer literature since 1858. For more than 100 years, they have been generally disregarded as irreversibly senescent or dying cells, too morphologically misshapen and chromatin too disorganized to be functional. Cell enlargement, accompanied by whole genome doubling or more, is observed across organisms, often associated with mitigation strategies against environmental change, severe stress, or the lack of nutrients. Our comparison of the mechanisms for polyploidization in other organisms and non-transformed tissues suggest that cancer cells draw from a conserved program for their survival, utilizing whole genome doubling and pausing proliferation to survive stress. These polyaneuploid cancer cells (PACCs) are the source of therapeutic resistance, responsible for cancer recurrence and, ultimately, cancer lethality.
Collapse
|
38
|
Coste A, Karagiannis GS, Wang Y, Xue EA, Lin Y, Skobe M, Jones JG, Oktay MH, Condeelis JS, Entenberg D. Hematogenous Dissemination of Breast Cancer Cells From Lymph Nodes Is Mediated by Tumor MicroEnvironment of Metastasis Doorways. Front Oncol 2020; 10:571100. [PMID: 33194666 PMCID: PMC7649363 DOI: 10.3389/fonc.2020.571100] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/17/2020] [Indexed: 11/24/2022] Open
Abstract
In primary breast tumors, cancer cells hematogenously disseminate through doorways in the vasculature composed of three-cell complexes (known as Tumor MicroEnvironment of Metastasis) comprising a perivascular macrophage, a tumor cell overexpressing the actin-regulatory protein Mammalian Enabled (Mena), and an endothelial cell, all in direct physical contact. It has been previously shown that once tumor cells establish lymph node metastases in patients, TMEM doorways form in the metastatic tumor cell nests. However, it has not been established if such lymph node-TMEM doorways actively transit tumor cells into the peripheral circulation and on to tertiary sites. To address this question in this short report, we used a mouse model of lymph node metastasis to demonstrate that TMEM doorways: (1) exist in tumor-positive lymph nodes of mice, (2) are restricted to the blood vascular endothelium, (3) serve as a mechanism for further dissemination to peripheral sites such as to the lungs, and (4) their activity can be abrogated by a pharmaceutical intervention. Our data suggest that cancer cell dissemination via TMEM doorways is a common mechanism of breast cancer cell dissemination to distant sites and thus the pharmacological targeting of TMEM may be necessary, even after resection of the primary tumor, to suppress cancer cell dissemination.
Collapse
Affiliation(s)
- Anouchka Coste
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Department of Surgery, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States
| | - George S Karagiannis
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Integrated Imaging Program, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States
| | - Yarong Wang
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Integrated Imaging Program, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States
| | - Emily A Xue
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States
| | - Yu Lin
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States
| | - Mihaela Skobe
- Department of Oncological Sciences and Tisch Cancer Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Joan G Jones
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Integrated Imaging Program, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Department of Pathology, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Department of Epidemiology and Population Health, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States
| | - Maja H Oktay
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Integrated Imaging Program, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Department of Pathology, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Department of Surgery, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Integrated Imaging Program, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States
| | - David Entenberg
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States.,Integrated Imaging Program, Einstein College of Medicine/Montefiore Medical Center, New York, NY, United States
| |
Collapse
|
39
|
Ahrens TD, Bang-Christensen SR, Jørgensen AM, Løppke C, Spliid CB, Sand NT, Clausen TM, Salanti A, Agerbæk MØ. The Role of Proteoglycans in Cancer Metastasis and Circulating Tumor Cell Analysis. Front Cell Dev Biol 2020; 8:749. [PMID: 32984308 PMCID: PMC7479181 DOI: 10.3389/fcell.2020.00749] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
Circulating tumor cells (CTCs) are accessible by liquid biopsies via an easy blood draw. They represent not only the primary tumor site, but also potential metastatic lesions, and could thus be an attractive supplement for cancer diagnostics. However, the analysis of rare CTCs in billions of normal blood cells is still technically challenging and novel specific CTC markers are needed. The formation of metastasis is a complex process supported by numerous molecular alterations, and thus novel CTC markers might be found by focusing on this process. One example of this is specific changes in the cancer cell glycocalyx, which is a network on the cell surface composed of carbohydrate structures. Proteoglycans are important glycocalyx components and consist of a protein core and covalently attached long glycosaminoglycan chains. A few CTC assays have already utilized proteoglycans for both enrichment and analysis of CTCs. Nonetheless, the biological function of proteoglycans on clinical CTCs has not been studied in detail so far. Therefore, the present review describes proteoglycan functions during the metastatic cascade to highlight their importance to CTCs. We also outline current approaches for CTC assays based on targeting proteoglycans by their protein cores or their glycosaminoglycan chains. Lastly, we briefly discuss important technical aspects, which should be considered for studying proteoglycans.
Collapse
Affiliation(s)
- Theresa D. Ahrens
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sara R. Bang-Christensen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
- VarCT Diagnostics, Copenhagen, Denmark
| | | | - Caroline Løppke
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Charlotte B. Spliid
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Nicolai T. Sand
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thomas M. Clausen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Ali Salanti
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mette Ø. Agerbæk
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
- VarCT Diagnostics, Copenhagen, Denmark
| |
Collapse
|
40
|
O'Connor RÍ, Kiely PA, Dunne CP. The relationship between post-surgery infection and breast cancer recurrence. J Hosp Infect 2020; 106:522-535. [PMID: 32800825 DOI: 10.1016/j.jhin.2020.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023]
Abstract
Breast cancer is the second most prevalent form of cancer in women worldwide, with surgery remaining the standard treatment. The adverse impact of the surgery remains controversial. It has been suggested that systemic factors during the postoperative period may increase the risk of recurrence, specifically surgical site infection (SSI). The aim of this review was to critically appraise current published literature regarding the influence of SSIs, after primary breast cancer surgery, on breast cancer recurrence, and to delve into potential links between these. This systematic review adopted two approaches: to identify the incidence rates and risk factors related to SSI after primary breast cancer surgery; and, secondly, to examine breast cancer recurrence following SSI occurrence. Ninety-nine studies with 484,605 patients were eligible in the SSI-focused searches, and 53 studies with 17,569 patients for recurrence-focused. There was a 13.07% mean incidence of SSI. Six-hundred and thirty-eight Gram-positive and 442 Gram-negative isolates were identified, with methicillin-susceptible Staphylococcus aureus and Escherichia coli most commonly identified. There were 2077 cases of recurrence (11.8%), with 563 cases of local recurrence, 1186 cases of distant and 25 cases which recurred both locally and distantly. Five studies investigated the association between SSI and breast cancer recurrence with three concluding that an association did exist. In conclusion, there is association between SSI and adverse cancer outcomes, but the cellular link between them remains elusive. Confounding factors of retrospective study design, surgery type and SSI definition make results challenging to compare and interpret. A standardized prospective study with appropriate statistical power is justified.
Collapse
Affiliation(s)
- R Í O'Connor
- School of Medicine, University of Limerick, Ireland; Health Research Institute, University of Limerick, Ireland; Bernal Institute, University of Limerick, Ireland
| | - P A Kiely
- School of Medicine, University of Limerick, Ireland; Health Research Institute, University of Limerick, Ireland; Bernal Institute, University of Limerick, Ireland; Centre for Interventions in Infection, Inflammation & Immunity (4i), University of Limerick, Limerick, Ireland
| | - C P Dunne
- School of Medicine, University of Limerick, Ireland; Health Research Institute, University of Limerick, Ireland; Centre for Interventions in Infection, Inflammation & Immunity (4i), University of Limerick, Limerick, Ireland.
| |
Collapse
|
41
|
Borriello L, Karagiannis GS, Duran CL, Coste A, Oktay MH, Entenberg D, Condeelis JS. The role of the tumor microenvironment in tumor cell intravasation and dissemination. Eur J Cell Biol 2020; 99:151098. [PMID: 32800278 DOI: 10.1016/j.ejcb.2020.151098] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/15/2020] [Accepted: 06/29/2020] [Indexed: 01/11/2023] Open
Abstract
Metastasis, a process that requires tumor cell dissemination followed by tumor growth, is the primary cause of death in cancer patients. An essential step of tumor cell dissemination is intravasation, a process by which tumor cells cross the blood vessel endothelium and disseminate to distant sites. Studying this process is of utmost importance given that intravasation in the primary tumor, as well as the secondary and tertiary metastases, is the key step in the systemic spread of tumor cells, and that this process continues even after removal of the primary tumor. High-resolution intravital imaging of the tumor microenvironment of breast carcinoma has revealed that tumor cell intravasation exclusively occurs at doorways, termed "Tumor MicroEnvironment of Metastasis" (TMEM), composed of three different cell types: a Tie2high/VEGFhigh perivascular macrophage, a Mena overexpressing tumor cell, and an endothelial cell, all in direct contact. In this review article, we discuss the interactions between these cell types, the subsequent signaling events which lead to tumor cell intravasation, and the role of invadopodia in supporting tumor cell invasion and dissemination. We end our review by discussing how the knowledge acquired from the use of intravital imaging is now leading to new clinical trials targeting tumor cell dissemination and preventing metastatic progression.
Collapse
Affiliation(s)
- Lucia Borriello
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA.
| | - George S Karagiannis
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Camille L Duran
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Anouchka Coste
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Department of Surgery, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Maja H Oktay
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Department of Pathology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - David Entenberg
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA.
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Department of Surgery, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
42
|
Wang Z, Wang H, Ding X, Chen X, Shen K. A large-cohort retrospective study of metastatic patterns and prognostic outcomes between inflammatory and non-inflammatory breast cancer. Ther Adv Med Oncol 2020; 12:1758835920932674. [PMID: 32550867 PMCID: PMC7278308 DOI: 10.1177/1758835920932674] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 05/15/2020] [Indexed: 01/11/2023] Open
Abstract
Background and aims: Breast cancer-related death is attributable mainly to metastasis. Inflammatory breast cancer (IBC) is an infrequent subtype of breast cancer that shows a relatively high rate of metastasis. In this study, we aimed to compare the metastatic patterns and prognostic outcomes of IBC and non-inflammatory breast cancer (non-IBC). Methods: We extracted data between 2010 and 2014 from the Surveillance, Epidemiology and End Results (SEER) database. The Chi-square test and Fisher’s exact test were used to compare the categorical parameters among different groups. Logistic regression was applied for multivariate analysis. The Kaplan–Meier method and multivariate Cox regression models were performed to analyze prognosis. Results: We enrolled 233,686 breast cancer patients between 2010 and 2014 in our research, including 2806 IBC and 230,880 non-IBC patients. Compared with the non-IBC group, the IBC group tended to have a higher incidence of the human epidermal growth factor receptor 2 positive (HER2+) and triple-negative breast cancer (TNBC) subtypes, older age, a higher rate of unmarried status, a lower incidence of black race, poorer tumor differentiation, larger tumor sizes, and a higher frequency of regional lymph node invasion. IBC and non-IBC shared similar trends in molecular subtypes among different metastatic organs. The percentage of the hormone receptor positive (HR+)/human epidermal growth factor receptor 2 negative (HER2–) subtype decreased gradually in patients with lung (IBC 42.5%, non-IBC 55.7%), distant lymph node (IBC 41.5%, non-IBC 54.6%), liver (IBC 31.1%, non-IBC 46.7%), and brain (IBC 30.6%, non-IBC 47.9%) metastases compared with that in patients with bone (IBC 50.8%, non-IBC 69.0%) metastasis in both cohorts. In both the IBC and non-IBC cases, the proportion of visceral metastases increased in the TNBC subtype, especially brain metastasis (IBC 26.4%, non-IBC 21.2%), which had the largest increase. The frequencies of all sites (bone, lung, liver, brain, and distant lymph node) in IBC were much higher than those in non-IBC (bone: IBC 21.1%, non-IBC 3.0%; lung: IBC 11.4%, non-IBC 1.4%; liver: IBC 9.6%, non-IBC 1.2%; brain: IBC 2.6%, non-IBC 0.3%; distant lymph node: IBC 12.9%, non-IBC 1.0%). The most frequent bi-site metastasis was the bone and liver (IBC 2.5%, non-IBC 0.3%), and the most frequent tri-site combination was the bone, lung, and liver (IBC 1.1%, non-IBC 0.2%). Kaplan–Meier curves and multivariate Cox regression models suggested that the IBC cohort had poorer overall survival [hazard ratio (HR) 1.602, 95% confidence interval (CI) 1.496–1.716, p < 0.001] and breast cancer-specific survival (HR 1.511, 95% CI 1.402–1.628, p < 0.001) than the non-IBC cohort. Furthermore, univariate and multivariate analyses indicated that IBC was an independent prognostic factor in patients with different metastatic sites. Conclusion: IBC and non-IBC patients presented with different metastatic frequencies, clinical features and prognostic outcomes. Our findings provide more information for therapeutic decision making and clinical study designs.
Collapse
Affiliation(s)
- Zheng Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai 200025, China
| | - Hui Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyuan Ding
- Department of Pharmacy, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Xiaosong Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai 200025, China
| | - Kunwei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai 200025, China
| |
Collapse
|
43
|
Pienta KJ, Hammarlund EU, Axelrod R, Amend SR, Brown JS. Convergent Evolution, Evolving Evolvability, and the Origins of Lethal Cancer. Mol Cancer Res 2020; 18:801-810. [PMID: 32234827 PMCID: PMC7272288 DOI: 10.1158/1541-7786.mcr-19-1158] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/03/2020] [Accepted: 03/26/2020] [Indexed: 01/20/2023]
Abstract
Advances in curative treatment to remove the primary tumor have increased survival of localized cancers for most solid tumor types, yet cancers that have spread are typically incurable and account for >90% of cancer-related deaths. Metastatic disease remains incurable because, somehow, tumors evolve resistance to all known compounds, including therapies. In all of these incurable patients, de novo lethal cancer evolves capacities for both metastasis and resistance. Therefore, cancers in different patients appear to follow the same eco-evolutionary path that independently manifests in affected patients. This convergent outcome, that always includes the ability to metastasize and exhibit resistance, demands an explanation beyond the slow and steady accrual of stochastic mutations. The common denominator may be that cancer starts as a speciation event when a unicellular protist breaks away from its multicellular host and initiates a cancer clade within the patient. As the cancer cells speciate and diversify further, some evolve the capacity to evolve: evolvability. Evolvability becomes a heritable trait that influences the available variation of other phenotypes that can then be acted upon by natural selection. Evolving evolvability may be an adaptation for cancer cells. By generating and maintaining considerable heritable variation, the cancer clade can, with high certainty, serendipitously produce cells resistant to therapy and cells capable of metastasizing. Understanding that cancer cells can swiftly evolve responses to novel and varied stressors create opportunities for adaptive therapy, double-bind therapies, and extinction therapies; all involving strategic decision making that steers and anticipates the convergent coevolutionary responses of the cancers.
Collapse
Affiliation(s)
- Kenneth J Pienta
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, Maryland.
| | - Emma U Hammarlund
- Nordic Center for Earth Evolution, University of Southern Denmark, Odense, Denmark
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Robert Axelrod
- Gerald R. Ford School of Public Policy, University of Michigan, Ann Arbor, Michigan
| | - Sarah R Amend
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Joel S Brown
- Cancer Biology and Evolution Program and Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
44
|
Dong L, Zhang Z, Smith K, Kuczler MD, Reyes D, Amend SR, Cho YK, Xue W, Pienta KJ. The combination of size-based separation and selection-free technology provides higher circulating tumour cells detection sensitivity than either method alone in patients with metastatic prostate cancer. BJU Int 2020; 126:191-201. [PMID: 32115854 DOI: 10.1111/bju.15041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2020] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To investigate the circulating tumour cells (CTCs) capture abilities of two technologies that are not dependent on cell-surface marker expression: a selection-free platform [AccuCyte® -CyteFinder® system (Rarecyte)] and a size-based platform [fluid-assisted separation technology (FAST)]. In addition, the combination of the two systems to more completely assess CTCs was investigated. PATIENTS AND METHODS In all, 28 patients with metastatic prostate cancer were included. Two 6 mL peripheral blood samples were taken from each patient at the same time-point. The samples were then subjected to the two different technology platforms in parallel. An additional group of samples was acquired by applying the waste chamber material from the FAST-group tests (flow-through that goes through the FAST filter membrane) to the Rarecyte system for the detection any CTCs that were not captured by FAST. RESULTS The three groups had significantly different putative CTC-positive tests, with positive rates of 29% for Rarecyte, 57% for FAST, and 79% for the combination. We also assessed CTC phenotype: 56.6% of the CTCs were cytokeratin (CK)+/epithelial cell adhesion molecule (EpCAM)-, 3.1% were CK-/EpCAM+, and 40.3% were CK+/EPCAM+. The captured CTCs diameter ranged from 5.2 to 16.9 µm. The mean CTC size from the FAST waste chamber was significantly smaller. The diameters for each of the phenotypic groups were significantly different. CONCLUSIONS These data highlight disparities in the positive rates and enumerated CTC numbers detected by the two techniques. Notably, the combination of the two technologies resulted in the highest CTC-capture rates. Smaller CTCs were more likely to be missed by the FAST as they passed through the filter system. Sizes of CTCs varied with different cell surface marker phenotypes.
Collapse
Affiliation(s)
- Liang Dong
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhongyuan Zhang
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kimberly Smith
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Morgan D Kuczler
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Diane Reyes
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah R Amend
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yoon-Kyoung Cho
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Korea.,Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Korea
| | - Wei Xue
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kenneth J Pienta
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
45
|
Resveratrol Suppresses Prostate Cancer Epithelial Cell Scatter/Invasion by Targeting Inhibition of Hepatocyte Growth Factor (HGF) Secretion by Prostate Stromal Cells and Upregulation of E-cadherin by Prostate Cancer Epithelial Cells. Int J Mol Sci 2020; 21:ijms21051760. [PMID: 32143478 PMCID: PMC7084722 DOI: 10.3390/ijms21051760] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/23/2020] [Accepted: 03/02/2020] [Indexed: 12/17/2022] Open
Abstract
Cancer mortality is primarily attributed to metastasis and the resulting compromise of organs secondary to the initial tumor site. Metastasis is a multi-step process in which the tumor cells must first acquire a migratory phenotype and invade through the surrounding tissue for spread to distant organs in the body. The ability of malignant cells to migrate and breach surrounding tissue/matrix barriers is among the most daunting challenges to disease management for men in the United States diagnosed with prostate cancer (CaP), especially since, at diagnosis, a high proportion of patients already have occult or clinically-detectable metastasis. The interaction between hepatocyte growth factor (HGF) secreted by the stroma, with its receptor c-Met located in the epithelium, must occur for epithelial CaP cells to become migratory. We studied the effects of grape-derived phytochemical resveratrol on the transition of epithelial tumor cells from sedentary to a mobile, penetrant phenotype. A time lapse microscopy assay was used to monitor the acquisition of the migratory phenotype by resveratrol. The results show that resveratrol inhibits HGF-mediated interaction between the stroma and epithelium and suppresses epithelial CaP cell migration by attenuating the control of epithelial-to-mesenchymal transition (EMT).
Collapse
|
46
|
Salari B, Sheinbein DM, Rosman IS, Dehner LP. Metastatic pancreatic adenosquamous carcinoma to the scalp: A case report and review of the literature. J Cutan Pathol 2020; 47:263-268. [PMID: 31587324 DOI: 10.1111/cup.13582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/09/2019] [Accepted: 09/13/2019] [Indexed: 12/30/2022]
Abstract
Metastatic carcinoma to the skin occurs in only a minority of patients with a visceral or internal malignancy, with breast, lung, and colorectum accounting for the majority of cases. We present the case of a 66-year-old man with a recent violaceous nodule of the left scalp (1.2 × 1.0 × 0.2 cm) that was a metastatic pancreatic adenosquamous carcinoma, representing a seemingly rare event. Two months prior, after complaining of right hip pain, an image revealed a right femoral lesion. A biopsy of that lesion showed moderately differentiated adenocarcinoma. Subsequent imaging showed a mass in the pancreatic tail and also markedly elevated serum tumor markers, CA 19-9 and carcinoembryonic antigen (5325 and 111.5 U/mL, respectively). Before the appearance of the scalp nodule, the patient received radiotherapy and was started on chemotherapy, which was continued after diagnosis and resection of the nodule. Subsequent metastases developed in the liver, lung and additional cutaneous lesions. He died 11 months after initial presentation with right hip pain. As this case shows, cutaneous metastases confer a poor prognosis, often with less than a year survival following their appearance.
Collapse
Affiliation(s)
- Behzad Salari
- Lauren V. Ackerman Laboratory of Surgical Pathology, School of Medicine, Washington University Medical Center, St. Louis, Missouri
| | - David M Sheinbein
- Division of Dermatology, School of Medicine, Washington University Medical Center, St. Louis, Missouri
| | - Ilana S Rosman
- Lauren V. Ackerman Laboratory of Surgical Pathology, School of Medicine, Washington University Medical Center, St. Louis, Missouri
- Division of Dermatology, School of Medicine, Washington University Medical Center, St. Louis, Missouri
| | - Louis P Dehner
- Lauren V. Ackerman Laboratory of Surgical Pathology, School of Medicine, Washington University Medical Center, St. Louis, Missouri
| |
Collapse
|
47
|
Bader DA, McGuire SE. Tumour metabolism and its unique properties in prostate adenocarcinoma. Nat Rev Urol 2020; 17:214-231. [PMID: 32112053 DOI: 10.1038/s41585-020-0288-x] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2020] [Indexed: 12/14/2022]
Abstract
Anabolic metabolism mediated by aberrant growth factor signalling fuels tumour growth and progression. The first biochemical descriptions of the altered metabolic nature of solid tumours were reported by Otto Warburg almost a century ago. Now, the study of tumour metabolism is being redefined by the development of new molecular tools, tumour modelling systems and precise instrumentation together with important advances in genetics, cell biology and spectroscopy. In contrast to Warburg's original hypothesis, accumulating evidence demonstrates a critical role for mitochondrial metabolism and substantial variation in the way in which different tumours metabolize nutrients to generate biomass. Furthermore, computational and experimental approaches suggest a dominant influence of the tissue-of-origin in shaping the metabolic reprogramming that enables tumour growth. For example, the unique metabolic properties of prostate adenocarcinoma are likely to stem from the distinct metabolism of the prostatic epithelium from which it emerges. Normal prostatic epithelium employs comparatively glycolytic metabolism to sustain physiological citrate secretion, whereas prostate adenocarcinoma consumes citrate to power oxidative phosphorylation and fuel lipogenesis, enabling tumour progression through metabolic reprogramming. Current data suggest that the distinct metabolic aberrations in prostate adenocarcinoma are driven by the androgen receptor, providing opportunities for functional metabolic imaging and novel therapeutic interventions that will be complementary to existing diagnostic and treatment options.
Collapse
Affiliation(s)
- David A Bader
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA. .,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA.
| | - Sean E McGuire
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA. .,Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
48
|
Wang Z, Wang H, Sun X, Fang Y, Lu SS, Ding SN, Chen XS, Shen KW. A Risk Stratification Model for Predicting Overall Survival and Surgical Benefit in Triple-Negative Breast Cancer Patients With de novo Distant Metastasis. Front Oncol 2020; 10:14. [PMID: 32038988 PMCID: PMC6992581 DOI: 10.3389/fonc.2020.00014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/07/2020] [Indexed: 12/22/2022] Open
Abstract
Background and Aims: This research aimed to construct a novel model for predicting overall survival (OS) and surgical benefit in triple-negative breast cancer (TNBC) patients with de novo distant metastasis. Methods: We collected data from the Surveillance, Epidemiology, and End Results (SEER) database for TNBC patients with distant metastasis between 2010 and 2016. Patients were excluded if the data regarding metastatic status, follow-up time, or clinicopathological information were incomplete. Univariate and multivariate analyses were applied to identify significant prognostic parameters. By integrating these variables, a predictive nomogram and risk stratification model were constructed and assessed with C-indexes and calibration curves. Results: A total of 1,737 patients were finally identified. Patients enrolled from 2010 to 2014 were randomly assigned to two cohorts, 918 patients in the training cohort and 306 patients in the validation cohort I, and 513 patients enrolled from 2015 to 2016 were assigned to validation cohort II. Seven clinicopathological factors were included as prognostic variables in the nomogram: age, marital status, T stage, bone metastasis, brain metastasis, liver metastasis, and lung metastasis. The C-indexes were 0.72 [95% confidence interval [CI] 0.68-0.76] in the training cohort, 0.71 (95% CI 0.68-0.74) in validation cohort I and 0.71 (95% CI 0.67-0.75) in validation cohort II. Calibration plots indicated that the nomogram-based predictive outcome had good consistency with the recoded prognosis. A risk stratification model was further generated to accurately differentiate patients into three prognostic groups. In all cohorts, the median overall survival time in the low-, intermediate- and high-risk groups was 17.0 months (95% CI 15.6-18.4), 11.0 months (95% CI 10.0-12.0), and 6.0 months (95% CI 4.7-7.3), respectively. Locoregional surgery improved prognosis in both the low-risk [hazard ratio [HR] 0.49, 95% CI 0.41-0.60, P < 0.0001] and intermediate-risk groups (HR 0.55, 95% CI 0.46-0.67, P < 0.0001), but not in high-risk group (HR 0.73, 95% CI 0.52-1.03, P = 0.068). All stratified groups could prognostically benefit from chemotherapy (low-risk group: HR 0.50, 95% CI 0.35-0.69, P < 0.0001; intermediate-risk group: HR 0.34, 95% CI 0.26-0.44, P < 0.0001; and high-risk group: HR 0.16, 95% CI 0.10-0.25, P < 0.0001). Conclusion: A predictive nomogram and risk stratification model were constructed to assess prognosis in TNBC patients with de novo distant metastasis; these methods may provide additional introspection, integration and improvement for therapeutic decisions and further studies.
Collapse
Affiliation(s)
- Zheng Wang
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi Sun
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Fang
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuang-Shuang Lu
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu-Ning Ding
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Song Chen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun-Wei Shen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
49
|
Raskov H, Orhan A, Salanti A, Gögenur I. Premetastatic niches, exosomes and circulating tumor cells: Early mechanisms of tumor dissemination and the relation to surgery. Int J Cancer 2020; 146:3244-3255. [PMID: 31808150 DOI: 10.1002/ijc.32820] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/15/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022]
Abstract
The physiological stress response to surgery promotes wound healing and functional recovery and includes the activation of neural, inflammatory and proangiogenic signaling pathways. Paradoxically, the same pathways also promote metastatic spread and growth of residual cancer. Human and animal studies show that cancer surgery can increase survival, migration and proliferation of residual tumor cells. To secure the survival and growth of disseminated tumor cells, the formation of premetastatic niches in target organs involves a complex interplay between microenvironment, immune system, circulating tumor cells, as well as chemical mediators and exosomes secreted by the primary tumor. This review describes the current understanding of the early mechanisms of dissemination, as well as how surgery may facilitate disease progression.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ali Salanti
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
50
|
Du L, Sun Y. Skeletal muscle metastasis from squamous cell lung cancer was first found by ultrasound: a case report. Transl Cancer Res 2019; 8:2936-2940. [PMID: 35117052 PMCID: PMC8798471 DOI: 10.21037/tcr.2019.12.55] [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: 08/20/2019] [Accepted: 12/16/2019] [Indexed: 11/24/2022]
Abstract
Skeletal muscle metastasis (SMM) is rare and very difficult to diagnosis using routine ultrasound, computed tomography (CT), and magnetic resonance imaging (MRI), especially in patients without a definitive tumor history. In this report, we describe a 64-year-old man who came to hospital because of pain from a touchable mass at his left thigh. Ultrasonography showed a substantive hypoechoic mass within the left adductor longus muscle with an anechoic area, which was ill-defined and rich in blood supply on color Doppler flow imaging (CDFI). The mass was biopsied under ultrasound guidance. The pathological findings displayed an infiltration of moderately differentiated squamous cell lung cancer (SCLC) and nests of atypical cells. Thoracic plain CT images displayed thickening of the bronchus wall of the right inferior lobe, with obstructive pneumonia and atelectasis at the distal bronchus. The final main diagnosis for this patient was central bronchogenic carcinoma, with intramuscular metastasis of the left adductor longus muscle, and the TNM classification was T2N0M1. Although the incidence of IM originating from SCLC is very rare, patients with an occasional found mass in soft tissue should be examined to exclude SMM, and ultrasound-guided percutaneous biopsy can provide vital information regarding indeterminate soft tissue masses.
Collapse
Affiliation(s)
- Lin Du
- Department of Developmental and Behavioral Pediatrics, the First Hospital of Jilin University, Changchun 130021, China
| | - Ying Sun
- Department of Abdominal Ultrasonography, the First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|