Killing Cancer: Discovery and Selection of New Target Molecules

A special issue of Cells (ISSN 2073-4409).

Deadline for manuscript submissions: closed (12 December 2019) | Viewed by 81437

Printed Edition Available!
A printed edition of this Special Issue is available here.

Special Issue Editor


E-Mail Website1 Website2
Guest Editor
1. Cancer Invasion and Resistance, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
2. Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
Interests: HER2; breast cancer; invasion; metastasis; drug screening; lysosome; transcription factor; signal transduction; autophagy
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Cancer incidence is rising steadily worldwide. Despite the fact that cancer treatments are better than ever, millions of people die of cancer every year. Currently, in developed countries, the lifetime risk of a cancer patient to die of cancer is about 20% when all cancers are considered. Over 90% of cancer deaths are due to cancer invasion and metastasis, which often occurs despite the most efficient, state-of-the art treatment. Thus, new therapies that can target invasion and metastasis and overcome the cancers primary and acquired resistance to existing treatments are needed. Novel therapies require innovative thinking and, for example, changes in old paradigms such as what indeed is “undruggable”.

This Special Issue presents some novel and promising research on new target molecules and innovative ideas to fight cancer.

Prof. Tuula Kallunki
Guest Editor

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. Cells 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 2700 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

  • treatment resistant
  • invasion
  • metastasis
  • drug discovery
  • cancer target

Published Papers (16 papers)

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

Editorial

Jump to: Research, Review, Other

5 pages, 188 KiB  
Editorial
Seeking and Exploring Efficient Ways to Target Cancer
by Tuula Kallunki
Cells 2020, 9(9), 2117; https://doi.org/10.3390/cells9092117 - 17 Sep 2020
Viewed by 1659
Abstract
Anti-cancer treatments have never been so numerous and so efficient [...] Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)

Research

Jump to: Editorial, Review, Other

22 pages, 3666 KiB  
Article
The Pyrazolo[3,4-d]pyrimidine Derivative, SCO-201, Reverses Multidrug Resistance Mediated by ABCG2/BCRP
by Sophie E. B. Ambjørner, Michael Wiese, Sebastian Christoph Köhler, Joen Svindt, Xamuel Loft Lund, Michael Gajhede, Lasse Saaby, Birger Brodin, Steffen Rump, Henning Weigt, Nils Brünner and Jan Stenvang
Cells 2020, 9(3), 613; https://doi.org/10.3390/cells9030613 - 04 Mar 2020
Cited by 12 | Viewed by 3641
Abstract
ATP-binding cassette (ABC) transporters, such as breast cancer resistance protein (BCRP), are key players in resistance to multiple anti-cancer drugs, leading to cancer treatment failure and cancer-related death. Currently, there are no clinically approved drugs for reversal of cancer drug resistance caused by [...] Read more.
ATP-binding cassette (ABC) transporters, such as breast cancer resistance protein (BCRP), are key players in resistance to multiple anti-cancer drugs, leading to cancer treatment failure and cancer-related death. Currently, there are no clinically approved drugs for reversal of cancer drug resistance caused by ABC transporters. This study investigated if a novel drug candidate, SCO-201, could inhibit BCRP and reverse BCRP-mediated drug resistance. We applied in vitro cell viability assays in SN-38 (7-Ethyl-10-hydroxycamptothecin)-resistant colon cancer cells and in non-cancer cells with ectopic expression of BCRP. SCO-201 reversed resistance to SN-38 (active metabolite of irinotecan) in both model systems. Dye efflux assays, bidirectional transport assays, and ATPase assays demonstrated that SCO-201 inhibits BCRP. In silico interaction analyses supported the ATPase assay data and suggest that SCO-201 competes with SN-38 for the BCRP drug-binding site. To analyze for inhibition of other transporters or cytochrome P450 (CYP) enzymes, we performed enzyme and transporter assays by in vitro drug metabolism and pharmacokinetics studies, which demonstrated that SCO-201 selectively inhibited BCRP and neither inhibited nor induced CYPs. We conclude that SCO-201 is a specific, potent, and potentially non-toxic drug candidate for the reversal of BCRP-mediated resistance in cancer cells. Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
Show Figures

Figure 1

18 pages, 6198 KiB  
Article
Targeting the NPL4 Adaptor of p97/VCP Segregase by Disulfiram as an Emerging Cancer Vulnerability Evokes Replication Stress and DNA Damage while Silencing the ATR Pathway
by Dusana Majera, Zdenek Skrott, Katarina Chroma, Joanna Maria Merchut-Maya, Martin Mistrik and Jiri Bartek
Cells 2020, 9(2), 469; https://doi.org/10.3390/cells9020469 - 18 Feb 2020
Cited by 28 | Viewed by 5133
Abstract
Research on repurposing the old alcohol-aversion drug disulfiram (DSF) for cancer treatment has identified inhibition of NPL4, an adaptor of the p97/VCP segregase essential for turnover of proteins involved in multiple pathways, as an unsuspected cancer cell vulnerability. While we reported that NPL4 [...] Read more.
Research on repurposing the old alcohol-aversion drug disulfiram (DSF) for cancer treatment has identified inhibition of NPL4, an adaptor of the p97/VCP segregase essential for turnover of proteins involved in multiple pathways, as an unsuspected cancer cell vulnerability. While we reported that NPL4 is targeted by the anticancer metabolite of DSF, the bis-diethyldithiocarbamate-copper complex (CuET), the exact, apparently multifaceted mechanism(s) through which the CuET-induced aggregation of NPL4 kills cancer cells remains to be fully elucidated. Given the pronounced sensitivity to CuET in tumor cell lines lacking the genome integrity caretaker proteins BRCA1 and BRCA2, here we investigated the impact of NPL4 targeting by CuET on DNA replication dynamics and DNA damage response pathways in human cancer cell models. Our results show that CuET treatment interferes with DNA replication, slows down replication fork progression and causes accumulation of single-stranded DNA (ssDNA). Such a replication stress (RS) scenario is associated with DNA damage, preferentially in the S phase, and activates the homologous recombination (HR) DNA repair pathway. At the same time, we find that cellular responses to the CuET-triggered RS are seriously impaired due to concomitant malfunction of the ATRIP-ATR-CHK1 signaling pathway that reflects an unorthodox checkpoint silencing mode through ATR (Ataxia telangiectasia and Rad3 related) kinase sequestration within the CuET-evoked NPL4 protein aggregates. Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
Show Figures

Figure 1

22 pages, 10429 KiB  
Article
The Vacuolar H+ ATPase α3 Subunit Negatively Regulates Migration and Invasion of Human Pancreatic Ductal Adenocarcinoma Cells
by Mette Flinck, Sofie Hagelund, Andrej Gorbatenko, Marc Severin, Elena Pedraz-Cuesta, Ivana Novak, Christian Stock and Stine Falsig Pedersen
Cells 2020, 9(2), 465; https://doi.org/10.3390/cells9020465 - 18 Feb 2020
Cited by 14 | Viewed by 3845
Abstract
Increased metabolic acid production and upregulation of net acid extrusion render pH homeostasis profoundly dysregulated in many cancers. Plasma membrane activity of vacuolar H+ ATPases (V-ATPases) has been implicated in acid extrusion and invasiveness of some cancers, yet often on the basis [...] Read more.
Increased metabolic acid production and upregulation of net acid extrusion render pH homeostasis profoundly dysregulated in many cancers. Plasma membrane activity of vacuolar H+ ATPases (V-ATPases) has been implicated in acid extrusion and invasiveness of some cancers, yet often on the basis of unspecific inhibitors. Serving as a membrane anchor directing V-ATPase localization, the a subunit of the V0 domain of the V-ATPase (ATP6V0a1-4) is particularly interesting in this regard. Here, we map the regulation and roles of ATP6V0a3 in migration, invasion, and growth in pancreatic ductal adenocarcinoma (PDAC) cells. a3 mRNA and protein levels were upregulated in PDAC cell lines compared to non-cancer pancreatic epithelial cells. Under control conditions, a3 localization was mainly endo-/lysosomal, and its knockdown had no detectable effect on pHi regulation after acid loading. V-ATPase inhibition, but not a3 knockdown, increased HIF-1α expression and decreased proliferation and autophagic flux under both starved and non-starved conditions, and spheroid growth of PDAC cells was also unaffected by a3 knockdown. Strikingly, a3 knockdown increased migration and transwell invasion of Panc-1 and BxPC-3 PDAC cells, and increased gelatin degradation in BxPC-3 cells yet decreased it in Panc-1 cells. We conclude that in these PDAC cells, a3 is upregulated and negatively regulates migration and invasion, likely in part via effects on extracellular matrix degradation. Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
Show Figures

Figure 1

24 pages, 9154 KiB  
Article
Dexamethasone Inhibits Spheroid Formation of Thyroid Cancer Cells Exposed to Simulated Microgravity
by Daniela Melnik, Jayashree Sahana, Thomas J. Corydon, Sascha Kopp, Mohamed Zakaria Nassef, Markus Wehland, Manfred Infanger, Daniela Grimm and Marcus Krüger
Cells 2020, 9(2), 367; https://doi.org/10.3390/cells9020367 - 05 Feb 2020
Cited by 20 | Viewed by 5391
Abstract
Detachment and the formation of spheroids under microgravity conditions can be observed with various types of intrinsically adherent human cells. In particular, for cancer cells this process mimics metastasis and may provide insights into cancer biology and progression that can be used to [...] Read more.
Detachment and the formation of spheroids under microgravity conditions can be observed with various types of intrinsically adherent human cells. In particular, for cancer cells this process mimics metastasis and may provide insights into cancer biology and progression that can be used to identify new drug/target combinations for future therapies. By using the synthetic glucocorticoid dexamethasone (DEX), we were able to suppress spheroid formation in a culture of follicular thyroid cancer (FTC)-133 cells that were exposed to altered gravity conditions on a random positioning machine. DEX inhibited the growth of three-dimensional cell aggregates in a dose-dependent manner. In the first approach, we analyzed the expression of several factors that are known to be involved in key processes of cancer progression such as autocrine signaling, proliferation, epithelial–mesenchymal transition, and anoikis. Wnt/β-catenin signaling and expression patterns of important genes in cancer cell growth and survival, which were further suggested to play a role in three-dimensional aggregation, such as NFKB2, VEGFA, CTGF, CAV1, BCL2(L1), or SNAI1, were clearly affected by DEX. Our data suggest the presence of a more complex regulation network of tumor spheroid formation involving additional signal pathways or individual key players that are also influenced by DEX. Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
Show Figures

Graphical abstract

13 pages, 2072 KiB  
Article
The Oncogene AF1Q is Associated with WNT and STAT Signaling and Offers a Novel Independent Prognostic Marker in Patients with Resectable Esophageal Cancer
by Elisabeth S. Gruber, Georg Oberhuber, Peter Birner, Michaela Schlederer, Michael Kenn, Wolfgang Schreiner, Gerd Jomrich, Sebastian F. Schoppmann, Michael Gnant, William Tse and Lukas Kenner
Cells 2019, 8(11), 1357; https://doi.org/10.3390/cells8111357 - 30 Oct 2019
Cited by 6 | Viewed by 3045
Abstract
AF1q impairs survival in hematologic and solid malignancies. AF1q expression is associated with tumor progression, migration and chemoresistance and acts as a transcriptional co-activator in WNT and STAT signaling. This study evaluates the role of AF1q in patients with resectable esophageal cancer (EC). [...] Read more.
AF1q impairs survival in hematologic and solid malignancies. AF1q expression is associated with tumor progression, migration and chemoresistance and acts as a transcriptional co-activator in WNT and STAT signaling. This study evaluates the role of AF1q in patients with resectable esophageal cancer (EC). A total of 278 patients operated on for EC were retrospectively included and the expression of AF1q, CD44 and pYSTAT3 was analyzed following immunostaining. Quantified data were processed to correlational and survival analysis. In EC tissue samples, an elevated expression of AF1q was associated with the expression of CD44 (p = 0.004) and pYSTAT3 (p = 0.0002). High AF1q expression in primary tumors showed high AF1q expression in the corresponding lymph nodes (p = 0.016). AF1q expression was higher after neoadjuvant therapy (p = 0.0002). Patients with AF1q-positive EC relapsed and died earlier compared to patients with AF1q-negative EC (disease-free survival (DFS), p = 0.0005; disease-specific survival (DSS), p = 0.003); in the multivariable Cox regression model, AF1q proved to be an independent prognostic marker (DFS, p = 0.01; DSS, p = 0.03). AF1q is associated with WNT and STAT signaling; it impairs and independently predicts DFS and DSS in patients with resectable EC. Testing AF1q could facilitate prognosis estimation and provide a possibility of identifying the patients responsive to the therapeutic blockade of its oncogenic downstream targets. Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
Show Figures

Figure 1

13 pages, 4636 KiB  
Article
Disruption of the NF-κB/IL-8 Signaling Axis by Sulconazole Inhibits Human Breast Cancer Stem Cell Formation
by Hack Sun Choi, Ji-Hyang Kim, Su-Lim Kim and Dong-Sun Lee
Cells 2019, 8(9), 1007; https://doi.org/10.3390/cells8091007 - 30 Aug 2019
Cited by 27 | Viewed by 5499
Abstract
Breast cancer stem cells (BCSCs) are tumor-initiating cells that possess the capacity for self-renewal. Cancer stem cells (CSCs) are responsible for poor outcomes caused by therapeutic resistance. In our study, we found that sulconazole—an antifungal medicine in the imidazole class—inhibited cell proliferation, tumor [...] Read more.
Breast cancer stem cells (BCSCs) are tumor-initiating cells that possess the capacity for self-renewal. Cancer stem cells (CSCs) are responsible for poor outcomes caused by therapeutic resistance. In our study, we found that sulconazole—an antifungal medicine in the imidazole class—inhibited cell proliferation, tumor growth, and CSC formation. This compound also reduced the frequency of cells expressing CSC markers (CD44high/CD24low) as well as the expression of another CSC marker, aldehyde dehydrogenase (ALDH), and other self-renewal-related genes. Sulconazole inhibited mammosphere formation, reduced the protein level of nuclear NF-κB, and reduced extracellular IL-8 levels in mammospheres. Knocking down NF-κB expression using a p65-specific siRNA reduced CSC formation and secreted IL-8 levels in mammospheres. Sulconazole reduced nuclear NF-κB protein levels and secreted IL-8 levels in mammospheres. These new findings show that sulconazole blocks the NF-κB/IL-8 signaling pathway and CSC formation. NF-κB/IL-8 signaling is important for CSC formation and may be an important therapeutic target for BCSC treatment. Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
Show Figures

Figure 1

Review

Jump to: Editorial, Research, Other

12 pages, 254 KiB  
Review
Oncofetal Chondroitin Sulfate: A Putative Therapeutic Target in Adult and Pediatric Solid Tumors
by Nastaran Khazamipour, Nader Al-Nakouzi, Htoo Zarni Oo, Maj Ørum-Madsen, Anne Steino, Poul H Sorensen and Mads Daugaard
Cells 2020, 9(4), 818; https://doi.org/10.3390/cells9040818 - 28 Mar 2020
Cited by 8 | Viewed by 3830
Abstract
Solid tumors remain a major challenge for targeted therapeutic intervention strategies such as antibody-drug conjugates and immunotherapy. At a minimum, clear and actionable solid tumor targets have to comply with the key biological requirement of being differentially over-expressed in solid tumors and metastasis, [...] Read more.
Solid tumors remain a major challenge for targeted therapeutic intervention strategies such as antibody-drug conjugates and immunotherapy. At a minimum, clear and actionable solid tumor targets have to comply with the key biological requirement of being differentially over-expressed in solid tumors and metastasis, in contrast to healthy organs. Oncofetal chondroitin sulfate is a cancer-specific secondary glycosaminoglycan modification to proteoglycans expressed in a variety of solid tumors and metastasis. Normally, this modification is found to be exclusively expressed in the placenta, where it is thought to facilitate normal placental implantation during pregnancy. Informed by this biology, oncofetal chondroitin sulfate is currently under investigation as a broad and specific target in solid tumors. Here, we discuss oncofetal chondroitin sulfate as a potential therapeutic target in childhood solid tumors in the context of current knowhow obtained over the past five years in adult cancers. Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
18 pages, 2794 KiB  
Review
Immunotherapy, Inflammation and Colorectal Cancer
by Charles Robert Lichtenstern, Rachael Katie Ngu, Shabnam Shalapour and Michael Karin
Cells 2020, 9(3), 618; https://doi.org/10.3390/cells9030618 - 04 Mar 2020
Cited by 164 | Viewed by 13509
Abstract
Colorectal cancer (CRC) is the third most common cancer type, and third highest in mortality rates among cancer-related deaths in the United States. Originating from intestinal epithelial cells in the colon and rectum, that are impacted by numerous factors including genetics, environment and [...] Read more.
Colorectal cancer (CRC) is the third most common cancer type, and third highest in mortality rates among cancer-related deaths in the United States. Originating from intestinal epithelial cells in the colon and rectum, that are impacted by numerous factors including genetics, environment and chronic, lingering inflammation, CRC can be a problematic malignancy to treat when detected at advanced stages. Chemotherapeutic agents serve as the historical first line of defense in the treatment of metastatic CRC. In recent years, however, combinational treatment with targeted therapies, such as vascular endothelial growth factor, or epidermal growth factor receptor inhibitors, has proven to be quite effective in patients with specific CRC subtypes. While scientific and clinical advances have uncovered promising new treatment options, the five-year survival rate for metastatic CRC is still low at about 14%. Current research into the efficacy of immunotherapy, particularly immune checkpoint inhibitor therapy (ICI) in mismatch repair deficient and microsatellite instability high (dMMR–MSI-H) CRC tumors have shown promising results, but its use in other CRC subtypes has been either unsuccessful, or not extensively explored. This Review will focus on the current status of immunotherapies, including ICI, vaccination and adoptive T cell therapy (ATC) in the treatment of CRC and its potential use, not only in dMMR–MSI-H CRC, but also in mismatch repair proficient and microsatellite instability low (pMMR-MSI-L). Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
Show Figures

Figure 1

18 pages, 2252 KiB  
Review
Zinc Finger Transcription Factor MZF1—A Specific Regulator of Cancer Invasion
by Ditte Marie Brix, Knut Kristoffer Bundgaard Clemmensen and Tuula Kallunki
Cells 2020, 9(1), 223; https://doi.org/10.3390/cells9010223 - 16 Jan 2020
Cited by 30 | Viewed by 4770
Abstract
Over 90% of cancer deaths are due to cancer cells metastasizing into other organs. Invasion is a prerequisite for metastasis formation. Thus, inhibition of invasion can be an efficient way to prevent disease progression in these patients. This could be achieved by targeting [...] Read more.
Over 90% of cancer deaths are due to cancer cells metastasizing into other organs. Invasion is a prerequisite for metastasis formation. Thus, inhibition of invasion can be an efficient way to prevent disease progression in these patients. This could be achieved by targeting the molecules regulating invasion. One of these is an oncogenic transcription factor, Myeloid Zinc Finger 1 (MZF1). Dysregulated transcription factors represent a unique, increasing group of drug targets that are responsible for aberrant gene expression in cancer and are important nodes driving cancer malignancy. Recent studies report of a central involvement of MZF1 in the invasion and metastasis of various solid cancers. In this review, we summarize the research on MZF1 in cancer including its function and role in lysosome-mediated invasion and in the expression of genes involved in epithelial to mesenchymal transition. We also discuss possible means to target it on the basis of the current knowledge of its function in cancer. Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
Show Figures

Figure 1

42 pages, 1085 KiB  
Review
Mutations That Confer Drug-Resistance, Oncogenicity and Intrinsic Activity on the ERK MAP Kinases—Current State of the Art
by Karina Smorodinsky-Atias, Nadine Soudah and David Engelberg
Cells 2020, 9(1), 129; https://doi.org/10.3390/cells9010129 - 06 Jan 2020
Cited by 24 | Viewed by 3816
Abstract
Unique characteristics distinguish extracellular signal-regulated kinases (Erks) from other eukaryotic protein kinases (ePKs). Unlike most ePKs, Erks do not autoactivate and they manifest no basal activity; they become catalysts only when dually phosphorylated on neighboring Thr and Tyr residues and they possess unique [...] Read more.
Unique characteristics distinguish extracellular signal-regulated kinases (Erks) from other eukaryotic protein kinases (ePKs). Unlike most ePKs, Erks do not autoactivate and they manifest no basal activity; they become catalysts only when dually phosphorylated on neighboring Thr and Tyr residues and they possess unique structural motifs. Erks function as the sole targets of the receptor tyrosine kinases (RTKs)-Ras-Raf-MEK signaling cascade, which controls numerous physiological processes and is mutated in most cancers. Erks are therefore the executers of the pathway’s biology and pathology. As oncogenic mutations have not been identified in Erks themselves, combined with the tight regulation of their activity, Erks have been considered immune against mutations that would render them intrinsically active. Nevertheless, several such mutations have been generated on the basis of structure-function analysis, understanding of ePK evolution and, mostly, via genetic screens in lower eukaryotes. One of the mutations conferred oncogenic properties on Erk1. The number of interesting mutations in Erks has dramatically increased following the development of Erk-specific pharmacological inhibitors and identification of mutations that cause resistance to these compounds. Several mutations have been recently identified in cancer patients. Here we summarize the mutations identified in Erks so far, describe their properties and discuss their possible mechanism of action. Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
Show Figures

Figure 1

51 pages, 2711 KiB  
Review
Targeting mTOR and Metabolism in Cancer: Lessons and Innovations
by Cedric Magaway, Eugene Kim and Estela Jacinto
Cells 2019, 8(12), 1584; https://doi.org/10.3390/cells8121584 - 06 Dec 2019
Cited by 140 | Viewed by 16278
Abstract
Cancer cells support their growth and proliferation by reprogramming their metabolism in order to gain access to nutrients. Despite the heterogeneity in genetic mutations that lead to tumorigenesis, a common alteration in tumors occurs in pathways that upregulate nutrient acquisition. A central signaling [...] Read more.
Cancer cells support their growth and proliferation by reprogramming their metabolism in order to gain access to nutrients. Despite the heterogeneity in genetic mutations that lead to tumorigenesis, a common alteration in tumors occurs in pathways that upregulate nutrient acquisition. A central signaling pathway that controls metabolic processes is the mTOR pathway. The elucidation of the regulation and functions of mTOR can be traced to the discovery of the natural compound, rapamycin. Studies using rapamycin have unraveled the role of mTOR in the control of cell growth and metabolism. By sensing the intracellular nutrient status, mTOR orchestrates metabolic reprogramming by controlling nutrient uptake and flux through various metabolic pathways. The central role of mTOR in metabolic rewiring makes it a promising target for cancer therapy. Numerous clinical trials are ongoing to evaluate the efficacy of mTOR inhibition for cancer treatment. Rapamycin analogs have been approved to treat specific types of cancer. Since rapamycin does not fully inhibit mTOR activity, new compounds have been engineered to inhibit the catalytic activity of mTOR to more potently block its functions. Despite highly promising pre-clinical studies, early clinical trial results of these second generation mTOR inhibitors revealed increased toxicity and modest antitumor activity. The plasticity of metabolic processes and seemingly enormous capacity of malignant cells to salvage nutrients through various mechanisms make cancer therapy extremely challenging. Therefore, identifying metabolic vulnerabilities in different types of tumors would present opportunities for rational therapeutic strategies. Understanding how the different sources of nutrients are metabolized not just by the growing tumor but also by other cells from the microenvironment, in particular, immune cells, will also facilitate the design of more sophisticated and effective therapeutic regimen. In this review, we discuss the functions of mTOR in cancer metabolism that have been illuminated from pre-clinical studies. We then review key findings from clinical trials that target mTOR and the lessons we have learned from both pre-clinical and clinical studies that could provide insights on innovative therapeutic strategies, including immunotherapy to target mTOR signaling and the metabolic network in cancer. Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
Show Figures

Figure 1

21 pages, 3514 KiB  
Review
Ferlin Overview: From Membrane to Cancer Biology
by Olivier Peulen, Gilles Rademaker, Sandy Anania, Andrei Turtoi, Akeila Bellahcène and Vincent Castronovo
Cells 2019, 8(9), 954; https://doi.org/10.3390/cells8090954 - 22 Aug 2019
Cited by 16 | Viewed by 4690
Abstract
In mammal myocytes, endothelial cells and inner ear cells, ferlins are proteins involved in membrane processes such as fusion, recycling, endo- and exocytosis. They harbour several C2 domains allowing their interaction with phospholipids. The expression of several Ferlin genes was described as altered [...] Read more.
In mammal myocytes, endothelial cells and inner ear cells, ferlins are proteins involved in membrane processes such as fusion, recycling, endo- and exocytosis. They harbour several C2 domains allowing their interaction with phospholipids. The expression of several Ferlin genes was described as altered in several tumoural tissues. Intriguingly, beyond a simple alteration, myoferlin, otoferlin and Fer1L4 expressions were negatively correlated with patient survival in some cancer types. Therefore, it can be assumed that membrane biology is of extreme importance for cell survival and signalling, making Ferlin proteins core machinery indispensable for cancer cell adaptation to hostile environments. The evidences suggest that myoferlin, when overexpressed, enhances cancer cell proliferation, migration and metabolism by affecting various aspects of membrane biology. Targeting myoferlin using pharmacological compounds, gene transfer technology, or interfering RNA is now considered as an emerging therapeutic strategy. Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
Show Figures

Graphical abstract

Other

3 pages, 178 KiB  
Erratum
Erratum: Gruber, E.S.; et al. The Oncogene AF1Q is Associated with WNT and STAT Signaling and Offers a Novel Independent Prognostic Marker in Patients with Resectable Esophageal Cancer. Cells 2019, 8, 1357
by Elisabeth S. Gruber, Georg Oberhuber, Peter Birner, Michaela Schlederer, Michael Kenn, Wolfgang Schreiner, Gerd Jomrich, Sebastian F. Schoppmann, Michael Gnant, William Tse and Lukas Kenner
Cells 2020, 9(12), 2724; https://doi.org/10.3390/cells9122724 - 21 Dec 2020
Viewed by 1446
Abstract
The authors wish to make the following change to their paper [...] Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
3 pages, 177 KiB  
Reply
Science between Bioreactors and Space Research—Response to Comments by Joseph J. Bevelacqua et al. on “Dexamethasone Inhibits Spheroid Formation of Thyroid Cancer Cells Exposed to Simulated Microgravity”
by Marcus Krüger, Sascha Kopp, Markus Wehland, Thomas J. Corydon and Daniela Grimm
Cells 2020, 9(8), 1763; https://doi.org/10.3390/cells9081763 - 23 Jul 2020
Viewed by 1592
Abstract
We would like to thank Bevelacqua et al [...] Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
3 pages, 171 KiB  
Comment
Comment on “Dexamethasone Inhibits Spheroid Formation of Thyroid Cancer Cells Exposed to Simulated Microgravity”
by Joseph J. Bevelacqua, James Welsh and S.M.J. Mortazavi
Cells 2020, 9(7), 1738; https://doi.org/10.3390/cells9071738 - 21 Jul 2020
Cited by 5 | Viewed by 2186
Abstract
This letter addresses our concerns about a paper by Melnik et al [...] Full article
(This article belongs to the Special Issue Killing Cancer: Discovery and Selection of New Target Molecules)
Back to TopTop