Adaptive Resistance to Targeted Cancer Therapies and Rational Development of Combination Therapies

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Cancer Therapy".

Deadline for manuscript submissions: 30 April 2024 | Viewed by 891

Special Issue Editors


E-Mail
Guest Editor
Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
Interests: targeted therapies; signal transduction; breast cancer

E-Mail Website
Guest Editor
1. Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
2. Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
3. Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
Interests: systems biology; targeted therapies; resistance; structure-based modeling; cell state transitions

Special Issue Information

Dear Colleagues,

Although genetic events are important determinants of resistance to molecularly targeted therapies, the sensitivity of tumors to drugs is also substantially shaped by the plasticity of tumor cells, which, through epigenetic mechanisms and remodulation of gene expression and through the complex dynamics of intracellular signaling networks, underlies adaptation to drug-induced perturbations. Drug resistance can be deciphered as an adaptation of a complex system to a perturbation affecting one or a few of its elements.

Adaptive resistance has been defined as a form of nongenomic resistance that intervenes rapidly and is potentially reversible and targetable. A thorough understanding of this form of resistance would facilitate the development of more effective therapies. Phenomena such as paradoxical activation of the ERK pathway by BRAF inhibitors, the rapid onset of BRAF inhibitor resistance in melanoma patients, the compensatory activation of one pathway following inhibition of a parallel pathway (e.g., PI3K/AKT and RAS/ERK), and the variable effects of targeted therapies on apoptosis and cellular senescence, underscore the importance of a thorough understanding of cancer cell responses to drugs. This underlies the rational development of effective drug combinations specific to any single tumor, aiming to avoid or delay the development of resistance.

This Special Issue will explore the phenomena of adaptive resistance to molecularly targeted therapies, with a systems perspective, to stimulate research in the area of rational drug combinations.

Dr. Andrea Rocca
Prof. Dr. Boris Kholodenko
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

  • targeted therapies
  • cancer drug resistance
  • adaptive resistance
  • signal transduction networks
  • network adaptation
  • cell state transitions

Published Papers (1 paper)

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

Research

16 pages, 6044 KiB  
Article
Reducing State Conflicts between Network Motifs Synergistically Enhances Cancer Drug Effects and Overcomes Adaptive Resistance
by Yunseong Kim, Sea Rom Choi and Kwang-Hyun Cho
Cancers 2024, 16(7), 1337; https://doi.org/10.3390/cancers16071337 - 29 Mar 2024
Viewed by 494
Abstract
Inducing apoptosis in cancer cells is a primary goal in anti-cancer therapy, but curing cancer with a single drug is unattainable due to drug resistance. The complex molecular network in cancer cells causes heterogeneous responses to single-target drugs, thereby inducing an adaptive drug [...] Read more.
Inducing apoptosis in cancer cells is a primary goal in anti-cancer therapy, but curing cancer with a single drug is unattainable due to drug resistance. The complex molecular network in cancer cells causes heterogeneous responses to single-target drugs, thereby inducing an adaptive drug response. Here, we showed that targeted drug perturbations can trigger state conflicts between multi-stable motifs within a molecular regulatory network, resulting in heterogeneous drug responses. However, we revealed that properly regulating an interconnecting molecule between these motifs can synergistically minimize the heterogeneous responses and overcome drug resistance. We extracted the essential cellular response dynamics of the Boolean network driven by the target node perturbation and developed an algorithm to identify a synergistic combinatorial target that can reduce heterogeneous drug responses. We validated the proposed approach using exemplary network models and a gastric cancer model from a previous study by showing that the targets identified with our algorithm can better drive the networks to desired states than those with other control theories. Of note, our approach suggests a new synergistic pair of control targets that can increase cancer drug efficacy to overcome adaptive drug resistance. Full article
Show Figures

Figure 1

Planned Papers

The below list represents only planned manuscripts. Some of these manuscripts have not been received by the Editorial Office yet. Papers submitted to MDPI journals are subject to peer-review.

Title: Model-based analysis reveals different dosing strategies for beneficial combination of pan-RAF and MEK inhibitors in BRAF vs NRAS mutant melanomas
Authors: Gerosa, Luca
Affiliation: Harvard Medical School., Boston, United States

Title: Cell state transition models enable engineering breast cancer cell phenotypes
Authors: Oleksii S. Rukhlenko; Hiroaki Imoto; Ayush Tambde; Amy McGillycuddy; Philipp Junk; Anna Tuliakova; Walter Kolch; Boris N. Kholodenko
Affiliation: 1 Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland; 2 Stratford college, Dublin, Ireland; 3 Technological University Dublin, Ireland; 4 Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland; 5 Department of Pharmacology, Yale University School of Medicine, New Haven, USA.
Abstract: Understanding signalling patterns of transformation and controlling cell phenotypes is a challenge of current biology. Here we applied a cell State Transition Assessment and Regulation (cSTAR) approach to a publicly available perturbation dataset of single cell phosphoproteomic patterns of multiple breast cancer (BC) and normal breast tissue-derived cell lines. Following a separation of luminal, basal, and normal cell states, we determined signalling nodes and causal connections of core controlling network, and main drivers of oncogenic transformation and transitions between different BC subtypes. Whereas cell lines within the same BC subtype have different mutational and expression profiles, the architecture of core network was similar for all luminal BC cells, and mTOR was a main oncogenic driver. In contrast, core networks of basal BC were heterogeneous and segregated into two major subclasses that featured distinct main oncogenic and phenotype drivers. Likewise, normal breast tissue cells separated into two different subclasses. Based on the data and quantified network topologies we derived mechanistic cSTAR models that serve as digital cell twins and allow to deliberately control cells’ movements in a Waddington landscape of different cell states. These cSTAR models suggested strategies of normalizing phosphorylation networks of BC cell lines using small molecule inhibitors.

Back to TopTop