Kinases and Cancer

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

Deadline for manuscript submissions: 29 March 2024 | Viewed by 140052

Special Issue Editor

1. Proteomics Center, Institute of Biochemistry, Vilnius University Life Sciences Center, Sauletekio al. 7, LT-10257 Vilnius, Lithuania
2. MAP Kinase Resource, Bioinformatics, Melchiorstrasse 9, CH-3027 Bern, Switzerland
Interests: breast cancer; prostate cancer; acute myeloid leukemia (AML); kinases
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Protein phosphorylation is one of the most critical mechanisms for regulating various cellular functions. Consequently, protein kinase dysfunctions have been associated with the development of various types and subtypes of human cancers. Some examples include amplification and overexpression of the epidermal growth factor receptor (EGFR) and the ERBB2 receptor in lung and breast tumors, mutation of the BRAF oncogene in a wide range of human tumors, and the chromosomal translocation known as the Philadelphia chromosome, which results in ABL1 tyrosine kinase activation.
In the drug discovery field, despite early skepticism on the potential and selectivity of protein kinase inhibitors, enormous progress has been made. New classes of drugs, such as antibodies to receptors, and small-molecule inhibitors, were developed and tested in the clinics. The success of such drugs (e.g., imatinib and trastuzumab) encouraged development of new, second-generation drugs, which target protein kinases in cancer. Moreover, kinases have been shown to be invaluable diagnostic, prognostic, and predictive biomarkers for many types of cancers. In addition to protein kinases, lipid kinases, such as phosphatidylinositol-3-kinase (PI3K) and phosphatidylinositol-4-kinase (PI4K), have also been involved in the pathology of many tumors. In this issue of Cancers, “Kinases and Cancers”, experts are invited to contribute original research papers or review articles that will provide further insights on the various functions of kinases in cancers, and on their role as drug targets and biomarkers.

Dr. Jonas Cicenas
Collection 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. 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

  • kinase inhibitors in cancer therapy
  • monoclonal antibodies against kinases in cancer therapy
  • kinases as oncogenes
  • kinases as tumor supressors
  • kinases as biomarkers
  • protein phosphorylation and cancer

Published Papers (15 papers)

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

Editorial

Jump to: Research, Review

4 pages, 577 KiB  
Editorial
Anti-Cancer Drugs Targeting Protein Kinases Approved by FDA in 2020
by Jonas Cicenas and Asta Račienė
Cancers 2021, 13(5), 947; https://doi.org/10.3390/cancers13050947 - 24 Feb 2021
Cited by 10 | Viewed by 2745
Abstract
Cancers are a large group of diseases that mostly emerge because of the uncontrollable action of many different genes in human cells [...] Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Figure 1

7 pages, 858 KiB  
Editorial
Kinases and Cancer
by Jonas Cicenas, Egle Zalyte, Amos Bairoch and Pascale Gaudet
Cancers 2018, 10(3), 63; https://doi.org/10.3390/cancers10030063 - 01 Mar 2018
Cited by 78 | Viewed by 7551
Abstract
Protein kinases are a large family of enzymes catalyzing protein phosphorylation. The human genome contains 518 protein kinase genes, 478 of which belong to the classical protein kinase family and 40 are atypical protein kinases [...]
Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Figure 1

1068 KiB  
Editorial
JNK, p38, ERK, and SGK1 Inhibitors in Cancer
by Jonas Cicenas, Egle Zalyte, Arnas Rimkus, Dalius Dapkus, Remigijus Noreika and Sigitas Urbonavicius
Cancers 2018, 10(1), 1; https://doi.org/10.3390/cancers10010001 - 21 Dec 2017
Cited by 68 | Viewed by 13201
Abstract
Mitogen-activated protein kinases (MAP kinases) are a family of kinases that regulates a range of biological processes implicated in the response to growth factors like latelet-derived growth factor (PDGF), epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), and stress, such as ultraviolet [...] Read more.
Mitogen-activated protein kinases (MAP kinases) are a family of kinases that regulates a range of biological processes implicated in the response to growth factors like latelet-derived growth factor (PDGF), epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), and stress, such as ultraviolet irradiation, heat shock, and osmotic shock. The MAP kinase family consists of four major subfamilies of related proteins (extracellular regulated kinases 1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), p38, and extracellular regulated kinase 5 (ERK5)) and regulates numerous cellular activities, such as apoptosis, gene expression, mitosis, differentiation, and immune responses. The deregulation of these kinases is shown to be involved in human diseases, such as cancer, immune diseases, inflammation, and neurodegenerative disorders. The awareness of the therapeutic potential of the inhibition of MAP kinases led to a thorough search for small-molecule inhibitors. Here, we discuss some of the most well-known MAP kinase inhibitors and their use in cancer research. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Figure 1

Research

Jump to: Editorial, Review

17 pages, 2746 KiB  
Article
Tumour-Secreted Protein S (ProS1) Activates a Tyro3-Erk Signalling Axis and Protects Cancer Cells from Apoptosis
by Nour Al Kafri and Sassan Hafizi
Cancers 2019, 11(12), 1843; https://doi.org/10.3390/cancers11121843 - 22 Nov 2019
Cited by 18 | Viewed by 4584
Abstract
The TAM subfamily (Tyro3, Axl, MerTK) of receptor tyrosine kinases are implicated in several cancers, where they have been shown to support primary tumorigenesis as well as secondary resistance to cancer therapies. Relatively little is known about the oncogenic role of Tyro3, including [...] Read more.
The TAM subfamily (Tyro3, Axl, MerTK) of receptor tyrosine kinases are implicated in several cancers, where they have been shown to support primary tumorigenesis as well as secondary resistance to cancer therapies. Relatively little is known about the oncogenic role of Tyro3, including its ligand selectivity and signalling in cancer cells. Tyro3 showed widespread protein and mRNA expression in a variety of human cancer cell lines. In SCC-25 head and neck cancer cells expressing both Tyro3 and Axl, Western blotting showed that both natural TAM ligands ProS1 and Gas6 rapidly stimulated Tyro3 and Erk kinase phosphorylation, with ProS1 eliciting a greater effect. In contrast, Gas6 was the sole stimulator of Axl and Akt kinase phosphorylation. In MGH-U3 bladder cancer cells, which express Tyro3 alone, ProS1 was again the stronger stimulator of Tyro3 and Erk stimulation but additionally stimulated Akt phosphorylation. Conditioned medium from ProS1-secreting 786-0 kidney cancer cells replicated the kinase activation effects of recombinant ProS1 in SCC-25 cells, with specificity confirmed by ProS1 ligand traps and warfarin. In addition, ProS1 protected cancer cells from acute apoptosis induced by staurosporine, as well as additionally, long-term serum starvation-induced apoptosis in MGH-U3 cells (Tyro3 only), which reflects its additional coupling to Akt signalling in these cells. In conclusion, we have shown that ProS1 is a tumour-derived functional ligand for Tyro3 that supports cancer cell survival. Furthermore, the ProS1-Tyro3 interaction is primarily coupled to Erk signalling although it displays signalling diversity dependent upon its representative expression as a TAM receptor in tumour cells. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Figure 1

16 pages, 2779 KiB  
Article
Synergistic Anti-Tumor Effect of mTOR Inhibitors with Irinotecan on Colon Cancer Cells
by Damien Reita, Cyril Bour, Radhia Benbrika, Audrey Groh, Erwan Pencreach, Eric Guérin and Dominique Guenot
Cancers 2019, 11(10), 1581; https://doi.org/10.3390/cancers11101581 - 17 Oct 2019
Cited by 23 | Viewed by 4628
Abstract
Advanced colorectal cancer has a poor prognosis because of metastasis formation and resistance to combined therapies. Downstream of PI3K/Akt and Ras/MAPK pathways, the mTOR kinase plays a decisive role in treatment failure. We previously established that irinotecan has antiangiogenic properties and it is [...] Read more.
Advanced colorectal cancer has a poor prognosis because of metastasis formation and resistance to combined therapies. Downstream of PI3K/Akt and Ras/MAPK pathways, the mTOR kinase plays a decisive role in treatment failure. We previously established that irinotecan has antiangiogenic properties and it is known that new mammalian target of rapamycin (mTOR) catalytic AZD inhibitors, unlike rapamycin, target both mTORC1 and mTORC2. Thus, we hypothesized that the complete inhibition of the PI3K/AKT/mTOR/HIF-1α axis with mTOR catalytic inhibitors and low doses of irinotecan may have antitumor effects. We showed that the AZD8055 and AZD2014 inhibitors were much more potent than rapamycin to reduce cell viability of four colon cell lines. On the other hand, whereas AZD2014 alone inhibits migration by 40%, the drug combination led to 70% inhibition. Similarly, neither irinotecan nor AZD2014 significantly reduced cell invasion, whereas a combination of the two inhibits invasion by 70%. In vivo, irinotecan and AZD2014 combination drastically reduced ectopic patient-derived colon tumor growth and this combination was more potent than Folfox or Folfiri. Finally, the combination totally inhibited liver and lung metastases developed from orthotopic implantation of SW480 cells. Thus, the use of mTOR catalytic inhibitors, in association with other chemotherapeutic agents like irinotecan at low doses, is potentially a hope for colon cancer treatment. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Graphical abstract

24 pages, 3870 KiB  
Article
Proteomic Analysis of Breast Cancer Resistance to the Anticancer Drug RH1 Reveals the Importance of Cancer Stem Cells
by Dalius Kuciauskas, Nadezda Dreize, Marija Ger, Algirdas Kaupinis, Kristijonas Zemaitis, Vaidotas Stankevicius, Kestutis Suziedelis, Jonas Cicenas, Lee M. Graves and Mindaugas Valius
Cancers 2019, 11(7), 972; https://doi.org/10.3390/cancers11070972 - 11 Jul 2019
Cited by 4 | Viewed by 5782
Abstract
Antitumor drug resistance remains a major challenge in cancer chemotherapy. Here we investigated the mechanism of acquired resistance to a novel anticancer agent RH1 designed to be activated in cancer cells by the NQO1 enzyme. Data show that in some cancer cells RH1 [...] Read more.
Antitumor drug resistance remains a major challenge in cancer chemotherapy. Here we investigated the mechanism of acquired resistance to a novel anticancer agent RH1 designed to be activated in cancer cells by the NQO1 enzyme. Data show that in some cancer cells RH1 may act in an NQO1-independent way. Differential proteomic analysis of breast cancer cells with acquired resistance to RH1 revealed changes in cell energy, amino acid metabolism and G2/M cell cycle transition regulation. Analysis of phosphoproteomics and protein kinase activity by multiplexed kinase inhibitor beads showed an increase in the activity of protein kinases involved in the cell cycle and stemness regulation and downregulation of proapoptotic kinases such as JNK in RH1-resistant cells. Suppression of JNK leads to the increase of cancer cell resistance to RH1. Moreover, resistant cells have enhanced expression of stem cell factor (SCF) and stem cell markers. Inhibition of SCF receptor c-KIT resulted in the attenuation of cancer stem cell enrichment and decreased amounts of tumor-initiating cells. RH1-resistant cells also acquire resistance to conventional therapeutics while remaining susceptible to c-KIT-targeted therapy. Data show that RH1 can be useful to treat cancers in the NQO1-independent way, and targeting of the cancer stem cells might be an effective approach for combating resistance to RH1 therapy. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Graphical abstract

25 pages, 5063 KiB  
Article
SYK Inhibition Potentiates the Effect of Chemotherapeutic Drugs on Neuroblastoma Cells In Vitro
by Conny Tümmler, Gianina Dumitriu, Malin Wickström, Peter Coopman, Andrey Valkov, Per Kogner, John Inge Johnsen, Ugo Moens and Baldur Sveinbjörnsson
Cancers 2019, 11(2), 202; https://doi.org/10.3390/cancers11020202 - 10 Feb 2019
Cited by 6 | Viewed by 5671
Abstract
Neuroblastoma is a malignancy arising from the developing sympathetic nervous system and the most common and deadly cancer of infancy. New therapies are needed to improve the prognosis for high-risk patients and to reduce toxicity and late effects. Spleen tyrosine kinase (SYK) has [...] Read more.
Neuroblastoma is a malignancy arising from the developing sympathetic nervous system and the most common and deadly cancer of infancy. New therapies are needed to improve the prognosis for high-risk patients and to reduce toxicity and late effects. Spleen tyrosine kinase (SYK) has previously been identified as a promising drug target in various inflammatory diseases and cancers but has so far not been extensively studied as a potential therapeutic target in neuroblastoma. In this study, we observed elevated SYK gene expression in neuroblastoma compared to neural crest and benign neurofibroma. While SYK protein was detected in the majority of examined neuroblastoma tissues it was less frequently observed in neuroblastoma cell lines. Depletion of SYK by siRNA and the use of small molecule SYK inhibitors significantly reduced the cell viability of neuroblastoma cell lines expressing SYK protein. Moreover, SYK inhibition decreased ERK1/2 and Akt phosphorylation. The SYK inhibitor BAY 61-3606 enhanced the effect of different chemotherapeutic drugs. Transient expression of a constitutive active SYK variant increased the viability of neuroblastoma cells independent of endogenous SYK levels. Collectively, our findings suggest that targeting SYK in combination with conventional chemotherapy should be further evaluated as a treatment option in neuroblastoma. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Graphical abstract

23 pages, 1962 KiB  
Article
Focal Adhesion Genes Refine the Intermediate-Risk Cytogenetic Classification of Acute Myeloid Leukemia
by Victor Pallarès, Montserrat Hoyos, M. Carmen Chillón, Eva Barragán, M. Isabel Prieto Conde, Marta Llop, Aïda Falgàs, María Virtudes Céspedes, Pau Montesinos, Josep F. Nomdedeu, Salut Brunet, Miguel Ángel Sanz, Marcos González-Díaz, Jorge Sierra, Ramon Mangues and Isolda Casanova
Cancers 2018, 10(11), 436; https://doi.org/10.3390/cancers10110436 - 13 Nov 2018
Cited by 8 | Viewed by 3824
Abstract
In recent years, several attempts have been made to identify novel prognostic markers in patients with intermediate-risk acute myeloid leukemia (IR-AML), to implement risk-adapted strategies. The non-receptor tyrosine kinases are proteins involved in regulation of cell growth, adhesion, migration and apoptosis. They associate [...] Read more.
In recent years, several attempts have been made to identify novel prognostic markers in patients with intermediate-risk acute myeloid leukemia (IR-AML), to implement risk-adapted strategies. The non-receptor tyrosine kinases are proteins involved in regulation of cell growth, adhesion, migration and apoptosis. They associate with metastatic dissemination in solid tumors and poor prognosis. However, their role in haematological malignancies has been scarcely studied. We hypothesized that PTK2/FAK, PTK2B/PYK2, LYN or SRC could be new prognostic markers in IR-AML. We assessed PTK2, PTK2B, LYN and SRC gene expression in a cohort of 324 patients, adults up to the age of 70, classified in the IR-AML cytogenetic group. Univariate and multivariate analyses showed that PTK2B, LYN and PTK2 gene expression are independent prognostic factors in IR-AML patients. PTK2B and LYN identify a patient subgroup with good prognosis within the cohort with non-favorable FLT3/NPM1 combined mutations. In contrast, PTK2 identifies a patient subgroup with poor prognosis within the worst prognosis cohort who display non-favorable FLT3/NPM1 combined mutations and underexpression of PTK2B or LYN. The combined use of these markers can refine the highly heterogeneous intermediate-risk subgroup of AML patients, and allow the development of risk-adapted post-remission chemotherapy protocols to improve their response to treatment. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Figure 1

624 KiB  
Article
Comparison of Intracellular Stress Response of NCI-H526 Small Cell Lung Cancer (SCLC) Cells to Platinum(II) Cisplatin and Platinum(IV) Oxoplatin
by Gerhard Hamilton
Cancers 2014, 6(3), 1487-1499; https://doi.org/10.3390/cancers6031487 - 08 Jul 2014
Cited by 4 | Viewed by 7345
Abstract
In attempts to develop an orally applicable platinum-based drug, platinum(IV) drugs which exhibit higher in vivo stability compared to the platinum(II) drug cisplatin were formulated. The first such chemotherapeutic agent, namely satraplatin, failed to receive approval. In the present work, we checked the [...] Read more.
In attempts to develop an orally applicable platinum-based drug, platinum(IV) drugs which exhibit higher in vivo stability compared to the platinum(II) drug cisplatin were formulated. The first such chemotherapeutic agent, namely satraplatin, failed to receive approval. In the present work, we checked the initial cellular stress response of the chemosensitive NCI-H526 small cell lung cancer (SCLC) cells by determination of the relative phosphorylation of 46 specific phosphorylation sites of 38 selected proteins in a six hours response to cisplatin (platinum(II)) or oxoplatin (platinum(IV)), respectively. Oxoplatin is considered as prodrug of cisplatin, although several findings point to differences in intracellular effects. Cisplatin induced hyperphosphorylation of p38α MAPK and AMPKα1, whereas oxoplatin treatment resulted in increased phosphorylation of a large number of signaling proteins involved in stress response/drug resistance, including JNK, GSK-3α, AMPKα1, src kinases, STATs, CHK-2 and especially focal adhesion kinase (FAK). Cisplatin exerts markedly higher cytotoxicity upon four hours short-term exposure in comparison to oxoplatin and, correspondingly, the extended initial stress response to the platinum(IV) drug oxoplatin thus is expected to increase clinical drug resistance. Induction of a substantial stress response to any prodrug of a platinum-based compound may likewise limit the effectivity of its active metabolite(s), such contributing to the failure of selected derivatized platinum complexes. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Figure 1

734 KiB  
Article
Characterization of a Dual CDC7/CDK9 Inhibitor in Multiple Myeloma Cellular Models
by Alessandro Natoni, Mark R. E. Coyne, Alan Jacobsen, Michael D. Rainey, Gemma O'Brien, Sandra Healy, Alessia Montagnoli, Jürgen Moll, Michael O'Dwyer and Corrado Santocanale
Cancers 2013, 5(3), 901-918; https://doi.org/10.3390/cancers5030901 - 24 Jul 2013
Cited by 18 | Viewed by 8105
Abstract
Two key features of myeloma cells are the deregulation of the cell cycle and the dependency on the expression of the BCL2 family of anti-apoptotic proteins. The cell division cycle 7 (CDC7) is an essential S-phase kinase and emerging CDC7 inhibitors are effective [...] Read more.
Two key features of myeloma cells are the deregulation of the cell cycle and the dependency on the expression of the BCL2 family of anti-apoptotic proteins. The cell division cycle 7 (CDC7) is an essential S-phase kinase and emerging CDC7 inhibitors are effective in a variety of preclinical cancer models. These compounds also inhibit CDK9 which is relevant for MCL-1 expression. The activity and mechanism of action of the dual CDC7/CDK9 inhibitor PHA-767491 was assessed in a panel of multiple myeloma cell lines, in primary samples from patients, in the presence of stromal cells and in combination with drugs used in current chemotherapeutic regimens. We report that in all conditions myeloma cells undergo cell death upon PHA-767491 treatment and we report an overall additive effect with melphalan, bortezomib and doxorubicin, thus supporting further assessment of targeting CDC7 and CDK9 in multiple myeloma. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Figure 1

Review

Jump to: Editorial, Research

22 pages, 1045 KiB  
Review
FGF/FGFR Signaling in Hepatocellular Carcinoma: From Carcinogenesis to Recent Therapeutic Intervention
by Yijun Wang, Danfei Liu, Tongyue Zhang and Limin Xia
Cancers 2021, 13(6), 1360; https://doi.org/10.3390/cancers13061360 - 17 Mar 2021
Cited by 23 | Viewed by 4009
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, ranking third in cancer deaths worldwide. Over the last decade, several studies have emphasized the development of tyrosine kinase inhibitors (TKIs) to target the aberrant pathways in HCC. However, the outcomes [...] Read more.
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, ranking third in cancer deaths worldwide. Over the last decade, several studies have emphasized the development of tyrosine kinase inhibitors (TKIs) to target the aberrant pathways in HCC. However, the outcomes are far from satisfactory due to the increasing resistance and adverse effects. The family of fibroblast growth factor (FGF) and its receptors (FGFR) are involved in various biological processes, including embryogenesis, morphogenesis, wound repair, and cell growth. The aberrant FGF/FGFR signaling is also observed in multiple cancers, including HCC. Anti-FGF/FGFR provides delightful benefits for cancer patients, especially those with FGF signaling alteration. More and more multi-kinase inhibitors targeting FGF signaling, pan-FGFR inhibitors, and selective FGFR inhibitors are now under preclinical and clinical investigation. This review summarizes the aberrant FGF/FGFR signaling in HCC initiating, development and treatment status, and provide new insights into the treatment of HCC. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Figure 1

2383 KiB  
Review
Targeting Cyclin-Dependent Kinases in Human Cancers: From Small Molecules to Peptide Inhibitors
by Marion Peyressatre, Camille Prével, Morgan Pellerano and May C. Morris
Cancers 2015, 7(1), 179-237; https://doi.org/10.3390/cancers7010179 - 23 Jan 2015
Cited by 246 | Viewed by 22437
Abstract
Cyclin-dependent kinases (CDK/Cyclins) form a family of heterodimeric kinases that play central roles in regulation of cell cycle progression, transcription and other major biological processes including neuronal differentiation and metabolism. Constitutive or deregulated hyperactivity of these kinases due to amplification, overexpression or mutation [...] Read more.
Cyclin-dependent kinases (CDK/Cyclins) form a family of heterodimeric kinases that play central roles in regulation of cell cycle progression, transcription and other major biological processes including neuronal differentiation and metabolism. Constitutive or deregulated hyperactivity of these kinases due to amplification, overexpression or mutation of cyclins or CDK, contributes to proliferation of cancer cells, and aberrant activity of these kinases has been reported in a wide variety of human cancers. These kinases therefore constitute biomarkers of proliferation and attractive pharmacological targets for development of anticancer therapeutics. The structural features of several of these kinases have been elucidated and their molecular mechanisms of regulation characterized in depth, providing clues for development of drugs and inhibitors to disrupt their function. However, like most other kinases, they constitute a challenging class of therapeutic targets due to their highly conserved structural features and ATP-binding pocket. Notwithstanding, several classes of inhibitors have been discovered from natural sources, and small molecule derivatives have been synthesized through rational, structure-guided approaches or identified in high throughput screens. The larger part of these inhibitors target ATP pockets, but a growing number of peptides targeting protein/protein interfaces are being proposed, and a small number of compounds targeting allosteric sites have been reported. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Graphical abstract

637 KiB  
Review
Highlights of the Latest Advances in Research on CDK Inhibitors
by Jonas Cicenas, Karthik Kalyan, Aleksandras Sorokinas, Asta Jatulyte, Deividas Valiunas, Algirdas Kaupinis and Mindaugas Valius
Cancers 2014, 6(4), 2224-2242; https://doi.org/10.3390/cancers6042224 - 27 Oct 2014
Cited by 109 | Viewed by 14321
Abstract
Uncontrolled proliferation is the hallmark of cancer and other proliferative disorders and abnormal cell cycle regulation is, therefore, common in these diseases. Cyclin-dependent kinases (CDKs) play a crucial role in the control of the cell cycle and proliferation. These kinases are frequently deregulated [...] Read more.
Uncontrolled proliferation is the hallmark of cancer and other proliferative disorders and abnormal cell cycle regulation is, therefore, common in these diseases. Cyclin-dependent kinases (CDKs) play a crucial role in the control of the cell cycle and proliferation. These kinases are frequently deregulated in various cancers, viral infections, neurodegenerative diseases, ischemia and some proliferative disorders. This led to a rigorous pursuit for small-molecule CDK inhibitors for therapeutic uses. Early efforts to block CDKs with nonselective CDK inhibitors led to little specificity and efficacy but apparent toxicity, but the recent advance of selective CDK inhibitors allowed the first successful efforts to target these kinases for the therapies of several diseases. Major ongoing efforts are to develop CDK inhibitors as monotherapies and rational combinations with chemotherapy and other targeted drugs. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Figure 1

802 KiB  
Review
The Role of PI3K/Akt/mTOR Signaling in Gastric Carcinoma
by Tasuku Matsuoka and Masakazu Yashiro
Cancers 2014, 6(3), 1441-1463; https://doi.org/10.3390/cancers6031441 - 07 Jul 2014
Cited by 152 | Viewed by 21202
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is one of the key signaling pathways induced by various receptor-tyrosine kinases. Accumulating evidence shows that this pathway is an important promoter of cell growth, metabolism, survival, metastasis, and resistance to chemotherapy. Genetic alterations [...] Read more.
The phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is one of the key signaling pathways induced by various receptor-tyrosine kinases. Accumulating evidence shows that this pathway is an important promoter of cell growth, metabolism, survival, metastasis, and resistance to chemotherapy. Genetic alterations in the PI3K/Akt/mTOR pathway in gastric carcinoma have often been demonstrated. Many kinds of molecular targeting therapies are currently undergoing clinical testing in patients with solid tumors. However, with the exception of the ErbB2-targeting antibody, targeting agents, including PI3K/Akt/mTOR inhibitors, have not been approved for treatment of patients with gastric carcinoma. This review summarizes the current knowledge on PI3K/Akt/mTOR signaling in the pathogenesis of gastric carcinoma and the possible therapeutic targets for gastric carcinoma. Improved knowledge of the PI3K/Akt/mTOR pathway in gastric carcinoma will be useful in understanding the mechanisms of tumor development and for identifying ideal targets of anticancer therapy for gastric carcinoma. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Figure 1

656 KiB  
Review
The Multifunctional Protein Kinase C-ε in Cancer Development and Progression
by Kirti Jain and Alakananda Basu
Cancers 2014, 6(2), 860-878; https://doi.org/10.3390/cancers6020860 - 10 Apr 2014
Cited by 46 | Viewed by 12135
Abstract
The protein kinase C (PKC) family proteins are important signal transducers and have long been the focus of cancer research. PKCɛ, a member of this family, is overexpressed in most solid tumors and plays critical roles in different processes that lead to cancer [...] Read more.
The protein kinase C (PKC) family proteins are important signal transducers and have long been the focus of cancer research. PKCɛ, a member of this family, is overexpressed in most solid tumors and plays critical roles in different processes that lead to cancer development. Studies using cell lines and animal models demonstrated the transforming potential of PKCɛ. While earlier research established the survival functions of PKCɛ, recent studies revealed its role in cell migration, invasion and cancer metastasis. PKCɛ has also been implicated in epithelial to mesenchymal transition (EMT), which may be the underlying mechanism by which it contributes to cell motility. In addition, PKCɛ affects cell-extracellular matrix (ECM) interactions by direct regulation of the cytoskeletal elements. Recent studies have also linked PKCɛ signaling to cancer stem cell functioning. This review focuses on the role of PKCɛ in different processes that lead to cancer development and progression. We also discussed current literatures on the pursuit of PKCɛ as a target for cancer therapy. Full article
(This article belongs to the Special Issue Kinases and Cancer)
Show Figures

Graphical abstract

Back to TopTop