Cellular Plasticity and the Untapped Therapeutic Potential in Cancer

A special issue of Cancers (ISSN 2072-6694).

Deadline for manuscript submissions: closed (1 April 2021) | Viewed by 39942

Special Issue Editors


E-Mail Website
Guest Editor
Curtin University, Perth, Australia
Interests: TGFB signaling; PI3K signaling; cancer cell plasticity; ubiquitin modifying enzymes

E-Mail Website
Guest Editor
The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Australia
Interests: cancer cell plasticity; metastasis; cell signaling; chemotherapy-resistance; heterogeneity

Special Issue Information

Dear Colleagues,

The administration of targeted therapies in cancer patients with well-defined tumour-driving mutations has markedly improved overall survival. Response rates to these therapies, however, remain disappointing, with quantifiable tumour regression limited by the development of acquired resistance. More recent evidence has indicated that targeted therapy can also rapidly induce diverse, genetically-independent, transcriptional programmes resulting in a “drug-tolerant” or “drug persister” cell population. Consequently, during this nongenetic evolutionary phase, cells are able to undergo an adaptive phenotype switch. This cellular or phenotype plasticity exhibited by a subpopulation by tumour cells has been demonstrated to release cells from their dependence on the tumour-driving alteration, resulting in a population of dedifferentiated, slow-cycling cells, capable of surviving continuous drug treatment. The pool of these drug-tolerant cells that survive initial therapy may eventually acquire validated genetic resistance mechanisms, limiting the efficacy of the therapy in the patient. Importantly, phenotype plasticity may be reversible with discontinued drug treatment resulting in resentisation of cells to the original therapy “drug holiday”. The identification of this subpopulation of slow-cycling drug-tolerant cells paves the way for the identification of a number of novel and incredible therapeutic opportunities to target cellular plasticity prior to the development of acquired resistance, with the potential to significantly improve overall survival in patients.

In this Special Issue, we seek to highlight the mechanisms driving cellular plasticity through alterations including chromatin remodelling, activation of reprogramming factors, and development programmes, such as the epithelial-to-mesenchymal and mesenchymal-to-epithelial transitions. In so doing, we aim to gain a greater understanding of the role of cellular plasticity in disease progression, therapy-resistance and metastasis.

Assoc. Prof. Pieter Eichhorn
Dr. Christine Chaffer
Guest Editors

Manuscript Submission Information

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

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

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

Keywords

  • Cellular plasticity
  • Phenotype switching
  • EMT
  • Drug tolerance
  • Chemotherapy
  • Targeted therapy
  • Metastasis
  • Therapy-resistance

Published Papers (8 papers)

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

Research

Jump to: Review

19 pages, 2327 KiB  
Article
Inactivating Mutations of the IK Gene Weaken Ku80/Ku70-Mediated DNA Repair and Sensitize Endometrial Cancer to Chemotherapy
by Chao Gao, Guangxu Jin, Elizabeth Forbes, Lingegowda S. Mangala, Yingmei Wang, Cristian Rodriguez-Aguayo, Paola Amero, Emine Bayraktar, Ye Yan, Gabriel Lopez-Berestein, Russell R. Broaddus, Anil K. Sood, Fengxia Xue and Wei Zhang
Cancers 2021, 13(10), 2487; https://doi.org/10.3390/cancers13102487 - 20 May 2021
Cited by 1 | Viewed by 2991
Abstract
IK is a mitotic factor that promotes cell cycle progression. Our previous investigation of 271 endometrial cancer (EC) samples from the Cancer Genome Atlas (TCGA) dataset showed IK somatic mutations were enriched in a cluster of patients with high-grade and high-stage cancers, and [...] Read more.
IK is a mitotic factor that promotes cell cycle progression. Our previous investigation of 271 endometrial cancer (EC) samples from the Cancer Genome Atlas (TCGA) dataset showed IK somatic mutations were enriched in a cluster of patients with high-grade and high-stage cancers, and this group had longer survival. This study provides insight into how IK somatic mutations contribute to EC pathophysiology. We analyzed the somatic mutational landscape of IK gene in 547 EC patients using expanded TCGA dataset. Co-immunoprecipitation and mass spectrometry were used to identify protein interactions. In vitro and in vivo experiments were used to evaluate IK’s role in EC. The patients with IK-inactivating mutations had longer survival during 10-year follow-up. Frameshift and stop-gain were common mutations and were associated with decreased IK expression. IK knockdown led to enrichment of G2/M phase cells, inactivation of DNA repair signaling mediated by heterodimerization of Ku80 and Ku70, and sensitization of EC cells to cisplatin treatment. IK/Ku80 mutations were accompanied by higher mutation rates and associated with significantly better overall survival. Inactivating mutations of IK gene and loss of IK protein expression were associated with weakened Ku80/Ku70-mediated DNA repair, increased mutation burden, and better response to chemotherapy in patients with EC. Full article
(This article belongs to the Special Issue Cellular Plasticity and the Untapped Therapeutic Potential in Cancer)
Show Figures

Figure 1

14 pages, 2457 KiB  
Article
A Theoretical Approach to Coupling the Epithelial-Mesenchymal Transition (EMT) to Extracellular Matrix (ECM) Stiffness via LOXL2
by Youyuan Deng, Priyanka Chakraborty, Mohit Kumar Jolly and Herbert Levine
Cancers 2021, 13(7), 1609; https://doi.org/10.3390/cancers13071609 - 31 Mar 2021
Cited by 26 | Viewed by 4401
Abstract
The epithelial-mesenchymal transition (EMT) plays a critical role in cancer progression, being responsible in many cases for the onset of the metastatic cascade and being integral in the ability of cells to resist drug treatment. Most studies of EMT focus on its induction [...] Read more.
The epithelial-mesenchymal transition (EMT) plays a critical role in cancer progression, being responsible in many cases for the onset of the metastatic cascade and being integral in the ability of cells to resist drug treatment. Most studies of EMT focus on its induction via chemical signals such as TGF-β or Notch ligands, but it has become increasingly clear that biomechanical features of the microenvironment such as extracellular matrix (ECM) stiffness can be equally important. Here, we introduce a coupled feedback loop connecting stiffness to the EMT transcription factor ZEB1, which acts via increasing the secretion of LOXL2 that leads to increased cross-linking of collagen fibers in the ECM. This increased cross-linking can effectively increase ECM stiffness and increase ZEB1 levels, thus setting a positive feedback loop between ZEB1 and ECM stiffness. To investigate the impact of this non-cell-autonomous effect, we introduce a computational approach capable of connecting LOXL2 concentration to increased stiffness and thereby to higher ZEB1 levels. Our results indicate that this positive feedback loop, once activated, can effectively lock the cells in a mesenchymal state. The spatial-temporal heterogeneity of the LOXL2 concentration and thus the mechanical stiffness also has direct implications for migrating cells that attempt to escape the primary tumor. Full article
(This article belongs to the Special Issue Cellular Plasticity and the Untapped Therapeutic Potential in Cancer)
Show Figures

Figure 1

18 pages, 3259 KiB  
Article
Identifying Therapies to Combat Epithelial Mesenchymal Plasticity-Associated Chemoresistance to Conventional Breast Cancer Therapies Using An shRNA Library Screen
by Sugandha Bhatia, Tony Blick, Cletus Pinto, Mark Waltham, James Monkman, Ekaterina Ivanova, Pamela M. Pollock, Shivashankar H. Nagaraj, Adrian P. Wiegmans, Izhak Haviv, Kaylene J. Simpson and Erik W. Thompson
Cancers 2020, 12(5), 1123; https://doi.org/10.3390/cancers12051123 - 30 Apr 2020
Cited by 6 | Viewed by 3303
Abstract
Background: Breast cancer (BC) is a heterogeneous disease for which the commonly used chemotherapeutic agents primarily include the anthracyclines (doxorubicin, epirubicin), microtubule inhibitors (paclitaxel, docetaxel, eribulin), and alkylating agents (cyclophosphamide). While these drugs can be highly effective, metastatic tumours are frequently refractory to [...] Read more.
Background: Breast cancer (BC) is a heterogeneous disease for which the commonly used chemotherapeutic agents primarily include the anthracyclines (doxorubicin, epirubicin), microtubule inhibitors (paclitaxel, docetaxel, eribulin), and alkylating agents (cyclophosphamide). While these drugs can be highly effective, metastatic tumours are frequently refractory to treatment or become resistant upon tumour relapse. Methods: We undertook a cell polarity/epithelial mesenchymal plasticity (EMP)-enriched short hairpin RNA (shRNA) screen in MDA-MB-468 breast cancer cells to identify factors underpinning heterogeneous responses to three chemotherapeutic agents used clinically in breast cancer: Doxorubicin, docetaxel, and eribulin. shRNA-transduced cells were treated for 6 weeks with the EC10 of each drug, and shRNA representation assessed by deep sequencing. We first identified candidate genes with depleted shRNA, implying that their silencing could promote a response. Using the Broad Institute’s Connectivity Map (CMap), we identified partner inhibitors targeting the identified gene families that may induce cell death in combination with doxorubicin, and tested them with all three drug treatments. Results: In total, 259 shRNAs were depleted with doxorubicin treatment (at p < 0.01), 66 with docetaxel, and 25 with eribulin. Twenty-four depleted hairpins overlapped between doxorubicin and docetaxel, and shRNAs for TGFB2, RUNX1, CCDC80, and HYOU1 were depleted across all the three drug treatments. Inhibitors of MDM/TP53, TGFBR, and FGFR were identified by CMap as the top pharmaceutical perturbagens and we validated the combinatorial benefits of the TGFBR inhibitor (SB525334) and MDM inhibitor (RITA) with doxorubicin treatment, and also observed synergy between the inhibitor SB525334 and eribulin in MDA-MB-468 cells. Conclusions: Taken together, a cell polarity/EMP-enriched shRNA library screen identified relevant gene products that could be targeted alongside current chemotherapeutic agents for the treatment of invasive BC. Full article
(This article belongs to the Special Issue Cellular Plasticity and the Untapped Therapeutic Potential in Cancer)
Show Figures

Graphical abstract

Review

Jump to: Research

15 pages, 1078 KiB  
Review
Harnessing Carcinoma Cell Plasticity Mediated by TGF-β Signaling
by Xuecong Wang and Jean Paul Thiery
Cancers 2021, 13(14), 3397; https://doi.org/10.3390/cancers13143397 - 7 Jul 2021
Cited by 8 | Viewed by 2696
Abstract
Epithelial cell plasticity, a hallmark of carcinoma progression, results in local and distant cancer dissemination. Carcinoma cell plasticity can be achieved through epithelial–mesenchymal transition (EMT), with cells positioned seemingly indiscriminately across the spectrum of EMT phenotypes. Different degrees of plasticity are achieved by [...] Read more.
Epithelial cell plasticity, a hallmark of carcinoma progression, results in local and distant cancer dissemination. Carcinoma cell plasticity can be achieved through epithelial–mesenchymal transition (EMT), with cells positioned seemingly indiscriminately across the spectrum of EMT phenotypes. Different degrees of plasticity are achieved by transcriptional regulation and feedback-loops, which confer carcinoma cells with unique properties of tumor propagation and therapy resistance. Decoding the molecular and cellular basis of EMT in carcinoma should enable the discovery of new therapeutic strategies against cancer. In this review, we discuss the different attributes of plasticity in carcinoma and highlight the role of the canonical TGFβ receptor signaling pathway in the acquisition of plasticity. We emphasize the potential stochasticity of stemness in carcinoma in relation to plasticity and provide data from recent clinical trials that seek to target plasticity. Full article
(This article belongs to the Special Issue Cellular Plasticity and the Untapped Therapeutic Potential in Cancer)
Show Figures

Figure 1

26 pages, 2150 KiB  
Review
Emerging Insights into Targeted Therapy-Tolerant Persister Cells in Cancer
by Heidie Frisco Cabanos and Aaron N. Hata
Cancers 2021, 13(11), 2666; https://doi.org/10.3390/cancers13112666 - 28 May 2021
Cited by 73 | Viewed by 9507
Abstract
Drug resistance is perhaps the greatest challenge in improving outcomes for cancer patients undergoing treatment with targeted therapies. It is becoming clear that “persisters,” a subpopulation of drug-tolerant cells found in cancer populations, play a critical role in the development of drug resistance. [...] Read more.
Drug resistance is perhaps the greatest challenge in improving outcomes for cancer patients undergoing treatment with targeted therapies. It is becoming clear that “persisters,” a subpopulation of drug-tolerant cells found in cancer populations, play a critical role in the development of drug resistance. Persisters are able to maintain viability under therapy but are typically slow cycling or dormant. These cells do not harbor classic drug resistance driver alterations, and their partial resistance phenotype is transient and reversible upon removal of the drug. In the clinic, the persister state most closely corresponds to minimal residual disease from which relapse can occur if treatment is discontinued or if acquired drug resistance develops in response to continuous therapy. Thus, eliminating persister cells will be crucial to improve outcomes for cancer patients. Using lung cancer targeted therapies as a primary paradigm, this review will give an overview of the characteristics of drug-tolerant persister cells, mechanisms associated with drug tolerance, and potential therapeutic opportunities to target this persister cell population in tumors. Full article
(This article belongs to the Special Issue Cellular Plasticity and the Untapped Therapeutic Potential in Cancer)
Show Figures

Figure 1

27 pages, 1465 KiB  
Review
Mechanisms of Resistance to PI3K Inhibitors in Cancer: Adaptive Responses, Drug Tolerance and Cellular Plasticity
by Sarah Christine Elisabeth Wright, Natali Vasilevski, Violeta Serra, Jordi Rodon and Pieter Johan Adam Eichhorn
Cancers 2021, 13(7), 1538; https://doi.org/10.3390/cancers13071538 - 26 Mar 2021
Cited by 35 | Viewed by 6009
Abstract
The phosphatidylinositol-3-kinase (PI3K) pathway plays a central role in the regulation of several signalling cascades which regulate biological processes such as cellular growth, survival, proliferation, motility and angiogenesis. The hyperactivation of this pathway is linked to tumour progression and is one of the [...] Read more.
The phosphatidylinositol-3-kinase (PI3K) pathway plays a central role in the regulation of several signalling cascades which regulate biological processes such as cellular growth, survival, proliferation, motility and angiogenesis. The hyperactivation of this pathway is linked to tumour progression and is one of the most common events in human cancers. Additionally, aberrant activation of the PI3K pathway has been demonstrated to limit the effectiveness of a number of anti-tumour agents paving the way for the development and implementation of PI3K inhibitors in the clinic. However, the overall effectiveness of these compounds has been greatly limited by inadequate target engagement due to reactivation of the pathway by compensatory mechanisms. Herein, we review the common adaptive responses that lead to reactivation of the PI3K pathway, therapy resistance and potential strategies to overcome these mechanisms of resistance. Furthermore, we highlight the potential role in changes in cellular plasticity and PI3K inhibitor resistance. Full article
(This article belongs to the Special Issue Cellular Plasticity and the Untapped Therapeutic Potential in Cancer)
Show Figures

Figure 1

22 pages, 957 KiB  
Review
Shifting the Gears of Metabolic Plasticity to Drive Cell State Transitions in Cancer
by Zhengwei Wu, Yi Fei Lee, Xun Hui Yeo, Ser Yue Loo and Wai Leong Tam
Cancers 2021, 13(6), 1316; https://doi.org/10.3390/cancers13061316 - 15 Mar 2021
Cited by 6 | Viewed by 2925
Abstract
Cancer metabolism is a hallmark of cancer. Metabolic plasticity defines the ability of cancer cells to reprogram a plethora of metabolic pathways to meet unique energetic needs during the various steps of disease progression. Cell state transitions are phenotypic adaptations which confer distinct [...] Read more.
Cancer metabolism is a hallmark of cancer. Metabolic plasticity defines the ability of cancer cells to reprogram a plethora of metabolic pathways to meet unique energetic needs during the various steps of disease progression. Cell state transitions are phenotypic adaptations which confer distinct advantages that help cancer cells overcome progression hurdles, that include tumor initiation, expansive growth, resistance to therapy, metastasis, colonization, and relapse. It is increasingly appreciated that cancer cells need to appropriately reprogram their cellular metabolism in a timely manner to support the changes associated with new phenotypic cell states. We discuss metabolic alterations that may be adopted by cancer cells in relation to the maintenance of cancer stemness, activation of the epithelial–mesenchymal transition program for facilitating metastasis, and the acquisition of drug resistance. While such metabolic plasticity is harnessed by cancer cells for survival, their dependence and addiction towards certain metabolic pathways also present therapeutic opportunities that may be exploited. Full article
(This article belongs to the Special Issue Cellular Plasticity and the Untapped Therapeutic Potential in Cancer)
Show Figures

Figure 1

14 pages, 1720 KiB  
Review
Fighting Drug Resistance through the Targeting of Drug-Tolerant Persister Cells
by Giulia De Conti, Matheus Henrique Dias and René Bernards
Cancers 2021, 13(5), 1118; https://doi.org/10.3390/cancers13051118 - 5 Mar 2021
Cited by 45 | Viewed by 7117
Abstract
Designing specific therapies for drug-resistant cancers is arguably the ultimate challenge in cancer therapy. While much emphasis has been put on the study of genetic alterations that give rise to drug resistance, much less is known about the non-genetic adaptation mechanisms that operate [...] Read more.
Designing specific therapies for drug-resistant cancers is arguably the ultimate challenge in cancer therapy. While much emphasis has been put on the study of genetic alterations that give rise to drug resistance, much less is known about the non-genetic adaptation mechanisms that operate during the early stages of drug resistance development. Drug-tolerant persister cells have been suggested to be key players in this process. These cells are thought to have undergone non-genetic adaptations that enable survival in the presence of a drug, from which full-blown resistant cells may emerge. Such initial adaptations often involve engagement of stress response programs to maintain cancer cell viability. In this review, we discuss the nature of drug-tolerant cancer phenotypes, as well as the non-genetic adaptations involved. We also discuss how malignant cells employ homeostatic stress response pathways to mitigate the intrinsic costs of such adaptations. Lastly, we discuss which vulnerabilities are introduced by these adaptations and how these might be exploited therapeutically. Full article
(This article belongs to the Special Issue Cellular Plasticity and the Untapped Therapeutic Potential in Cancer)
Show Figures

Figure 1

Back to TopTop