tumor cell plasticity
Recently Published Documents


TOTAL DOCUMENTS

61
(FIVE YEARS 23)

H-INDEX

17
(FIVE YEARS 3)

2021 ◽  
Vol 1 (3) ◽  
Author(s):  
Can Zhang ◽  
Changrong Shao ◽  
Xiaopei Jiao ◽  
Yue Bai ◽  
Miao Li ◽  
...  

2021 ◽  
Vol 22 (14) ◽  
pp. 7340
Author(s):  
Alvina I. Khamidullina ◽  
Ekaterina A. Varlamova ◽  
Nour Alhuda Hammoud ◽  
Margarita A. Yastrebova ◽  
Alexandra V. Bruter

Blood malignancies often arise from undifferentiated hematopoietic stem cells or partially differentiated stem-like cells. A tight balance of multipotency and differentiation, cell division, and quiescence underlying normal hematopoiesis requires a special program governed by the transcriptional machinery. Acquisition of drug resistance by tumor cells also involves reprogramming of their transcriptional landscape. Limiting tumor cell plasticity by disabling reprogramming of the gene transcription is a promising strategy for improvement of treatment outcomes. Herein, we review the molecular mechanisms of action of transcription-targeted drugs in hematological malignancies (largely in leukemia) with particular respect to the results of clinical trials.


2021 ◽  
Author(s):  
Xin Li ◽  
Dave Thirumalai

Inevitably, almost all cancer patients develop resistance to targeted therapy. Intratu- mor heterogeneity (ITH), which refers to coexistence of distinct clones within a single tumor, is a major cause of drug resistance. Mathematical models that explain exper- iments quantitatively is useful in understanding the origin of ITH, which then could be used to explore scenarios for efficacious therapy. Here, we develop a mathematical model to investigate ITH in breast cancer by exploiting the observation that HER2+ and HER2- cells could divide symmetrically (producing two identical daughter cells) or asymmetrically (HER2+ produces one HER2+ and one HER2- cell, for example). Our predictions for the evolution of cell fractions of HER2+ and HER2- cells are in quantitative agreement with single-cell experiments. Remarkably, the colony size of HER2+ cells emerging from a single HER2- cell (or vice versa), which occurs in about four cell doublings, agrees perfectly with experimental results, without tweaking any parameter in the model. The theory also quantitatively explains experimental data on the responses of breast cancer tumor under different treatment protocols. We then used the model to predict that, not only the order of two drugs, but also the treatment period for each drug and also the tumor cell plasticity could be manipulated to improve the treatment efficacy. Mathematical models, when integrated with data on patients, make possible exploration of a broad range of parameters readily, which might provide insights in devising effective therapies.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Erik Jung ◽  
Matthias Osswald ◽  
Miriam Ratliff ◽  
Helin Dogan ◽  
Ruifan Xie ◽  
...  

AbstractBoth the perivascular niche (PVN) and the integration into multicellular networks by tumor microtubes (TMs) have been associated with progression and resistance to therapies in glioblastoma, but their specific contribution remained unknown. By long-term tracking of tumor cell fate and dynamics in the live mouse brain, differential therapeutic responses in both niches are determined. Both the PVN, a preferential location of long-term quiescent glioma cells, and network integration facilitate resistance against cytotoxic effects of radiotherapy and chemotherapy—independently of each other, but with additive effects. Perivascular glioblastoma cells are particularly able to actively repair damage to tumor regions. Population of the PVN and resistance in it depend on proficient NOTCH1 expression. In turn, NOTCH1 downregulation induces resistant multicellular networks by TM extension. Our findings identify NOTCH1 as a central switch between the PVN and network niche in glioma, and demonstrate robust cross-compensation when only one niche is targeted.


2021 ◽  
Vol 10 ◽  
Author(s):  
Tamir Baram ◽  
Linor Rubinstein-Achiasaf ◽  
Hagar Ben-Yaakov ◽  
Adit Ben-Baruch

Cellular heterogeneity poses an immense therapeutic challenge in cancer due to a constant change in tumor cell characteristics, endowing cancer cells with the ability to dynamically shift between states. Intra-tumor heterogeneity is largely driven by cancer cell plasticity, demonstrated by the ability of malignant cells to acquire stemness and epithelial-to-mesenchymal transition (EMT) properties, to develop therapy resistance and to escape dormancy. These different aspects of cancer cell remodeling are driven by intrinsic as well as by extrinsic signals, the latter being dominated by factors of the tumor microenvironment. As part of the tumor milieu, chronic inflammation is generally regarded as a most influential player that supports tumor development and progression. In this review article, we put together recent findings on the roles of inflammatory elements in driving forward key processes of tumor cell plasticity. Using breast cancer as a representative research system, we demonstrate the critical roles played by inflammation-associated myeloid cells (mainly macrophages), pro-inflammatory cytokines [such as tumor necrosis factor α (TNFα) and interleukin 6 (IL-6)] and inflammatory chemokines [primarily CXCL8 (interleukin 8, IL-8) and CXCL1 (GROα)] in promoting tumor cell remodeling. These inflammatory components form a common thread that is involved in regulation of the three plasticity levels: stemness/EMT, therapy resistance, and dormancy. In view of the fact that inflammatory elements are a common denominator shared by different aspects of tumor cell plasticity, it is possible that their targeting may have a critical clinical benefit for cancer patients.


2021 ◽  
Vol 7 ◽  
Author(s):  
Satyendra Chandra Tripathi ◽  
Mohit Kumar Jolly ◽  
Sendurai A. Mani ◽  
Herbert Levine

2021 ◽  
Vol 10 (4) ◽  
pp. 66
Author(s):  
A.I. Stukan ◽  
R.A. Murashko ◽  
V.A. Porkhanov ◽  
A.G. Baryshev ◽  
I.V. Gilevich ◽  
...  

Cancers ◽  
2020 ◽  
Vol 12 (12) ◽  
pp. 3674 ◽  
Author(s):  
Ralf Hass ◽  
Juliane von der Ohe ◽  
Hendrik Ungefroren

Intratumoral heterogeneity is considered the major cause of drug unresponsiveness in cancer and accumulating evidence implicates non-mutational resistance mechanisms rather than genetic mutations in its development. These non-mutational processes are largely driven by phenotypic plasticity, which is defined as the ability of a cell to reprogram and change its identity (phenotype switching). Tumor cell plasticity is characterized by the reactivation of developmental programs that are closely correlated with the acquisition of cancer stem cell properties and an enhanced potential for retrodifferentiation or transdifferentiation. A well-studied mechanism of phenotypic plasticity is the epithelial-mesenchymal transition (EMT). Current evidence suggests a complex interplay between EMT, genetic and epigenetic alterations, and clues from the tumor microenvironment in cell reprogramming. A deeper understanding of the connections between stem cell, epithelial–mesenchymal, and tumor-associated reprogramming events is crucial to develop novel therapies that mitigate cell plasticity and minimize the evolution of tumor heterogeneity, and hence drug resistance. Alternatively, vulnerabilities exposed by tumor cells when residing in a plastic or stem-like state may be exploited therapeutically, i.e., by converting them into less aggressive or even postmitotic cells. Tumor cell plasticity thus presents a new paradigm for understanding a cancer’s resistance to therapy and deciphering its underlying mechanisms.


Sign in / Sign up

Export Citation Format

Share Document