Protein Kinases and Cancer

A special issue of Pharmaceuticals (ISSN 1424-8247). This special issue belongs to the section "Pharmacology".

Deadline for manuscript submissions: closed (30 April 2022) | Viewed by 55539

Special Issue Editors

Minneapolis V.A. Health Care System, and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
Interests: protein kinases; protein kinase CK2; prostate cancer; androgens; cancer; head and neck cancer; signaling; protein kinases as targets for therapy; cancer therapy; nuclear matrix; chromatin; intracellular shuttling; apoptosis; mitochondria; cell death; cell calcium
Special Issues, Collections and Topics in MDPI journals
1. Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
2. Research Service, Minneapolis VA Health Care System, Minneapolis, MN 55417, USA
Interests: protein kinase CK2; CDK11 signal mechanisms; therapy targeting

Special Issue Information

Dear Colleagues,

During the latter part of the twentieth century, it has continuously been recognized that protein kinases represent a large group of enzymes involved in the phosphorylation of proteins at various sites, thereby altering their functional activities. Protein phosphorylation has emerged as a major post-translation modification encountered by proteins occurring in normal, as well as in abnormal, cell functions. Virtually every aspect of biology is affected by activity of protein kinases. Diverse protein kinases modify proteins largely at serine, threonine, and tyrosine, although other phosphorylation sites are also known. Importantly, a protein may be modified by several protein kinases at the same sites, as well as at multiple sites by different kinases, thereby resulting in complex regulation of the function of a particular protein.

Because of the vast involvement of protein kinases in cell biological functions and their role in various disease states, it stands to reason that protein kinases have attracted much attention as possible targets for therapeutic intervention. In this regard, the role of dysregulated function of protein kinases in cancer has attracted particular consideration and there is a large body of ongoing investigations in this subject area as many different protein kinases are implicated in cancer development and survival. The involvement of protein kinases in cancer is also of focal interest because they serve as existing and potential targets for cancer therapy, and this is also a highly active area of research.  

The present Special Issue of Pharmaceuticals is specially dedicated to “Protein Kinases and Cancer”, and represents a compendium of manuscripts and review articles involving different protein kinases with respect to their biological function in various cancers and their potential as therapeutic targets. Pharmaceuticals has previously published a Special Issue dealing with Protein Kinase CK2 and Cancer; you may refer to:https://www.mdpi.com/journal/pharmaceuticals/special_issues/protein_kinase_CK2. However, the present issue includes a broader range of topics covering Protein Kinases and Cancer. We sincerely hope that you will be able to contribute a research manuscript or review article dealing with your research in this area of investigation. The quality of the publications will be ensured by peer review of all the manuscripts prior to their publication.

We look forward to your participation in this endeavor.

Prof. Dr. Khalil Ahmed
Dr. Janeen Trembley
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. Pharmaceuticals is an international peer-reviewed open access monthly 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

  • Protein kinases
  • Cancer biology
  • Cancer therapy
  • Signaling
  • Protein kinases as target for cancer therapy

Related Special Issue

Published Papers (12 papers)

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

Research

Jump to: Review

19 pages, 3377 KiB  
Communication
Antiproliferative Activity of a New Quinazolin-4(3H)-One Derivative via Targeting Aurora Kinase A in Non-Small Cell Lung Cancer
by Ji Yun Lee, Huarong Yang, Donghwa Kim, Kay Zin Kyaw, Ruoci Hu, Yanhua Fan and Sang Kook Lee
Pharmaceuticals 2022, 15(6), 698; https://doi.org/10.3390/ph15060698 - 02 Jun 2022
Cited by 3 | Viewed by 2058
Abstract
Non-small cell lung cancer (NSCLC) is the most common lung cancer subtype. Although chemotherapy and targeted therapy are used for the treatment of patients with NSCLC, the survival rate remains very low. Recent findings suggested that aurora kinase A (AKA), a cell cycle [...] Read more.
Non-small cell lung cancer (NSCLC) is the most common lung cancer subtype. Although chemotherapy and targeted therapy are used for the treatment of patients with NSCLC, the survival rate remains very low. Recent findings suggested that aurora kinase A (AKA), a cell cycle regulator, is a potential target for NSCLC therapy. Previously, we reported that a chemical entity of quinazolin-4(3H)-one represents a new template for AKA inhibitors, with antiproliferative activity against cancer cells. A quinazolin-4(3H)-one derivative was further designed and synthesized in order to improve the pharmacokinetic properties and antiproliferation activity against NSCLC cell lines. The derivative, BIQO-19 (Ethyl 6-(4-oxo-3-(pyrimidin-2-ylmethyl)-3,4-dihydroquinazolin-6-yl)imidazo [1,2-a]pyridine-2-carboxylate), exhibited improved solubility and antiproliferative activity in NSCLC cells, including epidermal growth factor receptor–tyrosine kinase inhibitor (EGFR-TKI)-resistant NSCLC cells. BIQO-19 effectively inhibited the growth of the EGFR-TKI-resistant H1975 NSCLC cells, with the suppression of activated AKA (p-AKA) expression in these cells. The inhibition of AKA by BIQO-19 significantly induced G2/M phase arrest and subsequently evoked apoptosis in H1975 cells. In addition, the combination of gefitinib and BIQO-19 exhibited synergistic antiproliferative activity in NSCLC cells. These findings suggest the potential of BIQO-19 as a novel therapeutic agent for restoring the sensitivity of gefitinib in EGFR-TKI-resistant NSCLC cells. Full article
(This article belongs to the Special Issue Protein Kinases and Cancer)
Show Figures

Graphical abstract

19 pages, 11816 KiB  
Article
Artonin F Induces the Ubiquitin-Proteasomal Degradation of c-Met and Decreases Akt-mTOR Signaling
by Rapeepun Soonnarong, Ismail Dwi Putra, Nicharat Sriratanasak, Boonchoo Sritularak and Pithi Chanvorachote
Pharmaceuticals 2022, 15(5), 633; https://doi.org/10.3390/ph15050633 - 21 May 2022
Cited by 4 | Viewed by 2077
Abstract
Targeted therapies that selectively inhibit certain molecules in cancer cells have been considered promising for cancer treatment. In lung cancer, evidence has suggested that mesenchymal-epithelial transition factor (c-Met) oncoprotein drives cancer progression through its signaling transduction pathway. In this paper, we report the [...] Read more.
Targeted therapies that selectively inhibit certain molecules in cancer cells have been considered promising for cancer treatment. In lung cancer, evidence has suggested that mesenchymal-epithelial transition factor (c-Met) oncoprotein drives cancer progression through its signaling transduction pathway. In this paper, we report the downregulation of c-Met by artonin F, a flavonoid isolated from Artocarpus gomezianus. Artonin F was found to be dominantly toxic to lung cancer cells by mediating apoptosis. With regard to its mechanism of action, artonin F downregulated c-Met expression, consequently suppressed the phosphatidylinositol-3 kinase/Akt/mammalian target of rapamycin signaling, increased Bax expression, decreased Bcl-2 expression, and activated caspase-3. The depletion of c-Met was mediated by ubiquitin-proteasomal degradation following co-treatment with artonin F, with the proteasome inhibitor MG132 reversing its c-Met-targeting effect. The immunoprecipitation analysis revealed that artonin F significantly promoted the formation of the c-Met–ubiquitin complex. Given that ubiquitin-specific protease 8 (USP8) prevents c-Met degradation by deubiquitination, we performed a preliminary in silico molecular docking and observed that artonin F blocked the catalytic site of USP8. In addition, artonin F interacted with the catalytic residues of palmitoylating enzymes. By acting as a competitive inhibitor, artonin F could reduce the degree of palmitoylation of c-Met, which affected its stability and activity. In conclusion, c-Met is critical for cancer cell survival and the failure of chemotherapeutic regimens. This novel information on the c-Met downregulating effect of artonin F will be beneficial for the development of efficient anticancer strategies or targeted therapies. Full article
(This article belongs to the Special Issue Protein Kinases and Cancer)
Show Figures

Graphical abstract

14 pages, 2831 KiB  
Article
Cyclin-Dependent Kinase and Antioxidant Gene Expression in Cancers with Poor Therapeutic Response
by George S. Scaria, Betsy T. Kren and Mark A. Klein
Pharmaceuticals 2020, 13(2), 26; https://doi.org/10.3390/ph13020026 - 05 Feb 2020
Cited by 1 | Viewed by 2537
Abstract
Pancreatic cancer, hepatocellular carcinoma (HCC), and mesothelioma are treatment-refractory cancers, and patients afflicted with these cancers generally have a very poor prognosis. The genomics of these tumors were analyzed as part of The Cancer Genome Atlas (TCGA) project. However, these analyses are an [...] Read more.
Pancreatic cancer, hepatocellular carcinoma (HCC), and mesothelioma are treatment-refractory cancers, and patients afflicted with these cancers generally have a very poor prognosis. The genomics of these tumors were analyzed as part of The Cancer Genome Atlas (TCGA) project. However, these analyses are an overview and may miss pathway interactions that could be exploited for therapeutic targeting. In this study, the TCGA Pan-Cancer datasets were queried via cBioPortal for correlations among mRNA expression of key genes in the cell cycle and mitochondrial (mt) antioxidant defense pathways. Here we describe these correlations. The results support further evaluation to develop combination treatment strategies that target these two critical pathways in pancreatic cancer, hepatocellular carcinoma, and mesothelioma. Full article
(This article belongs to the Special Issue Protein Kinases and Cancer)
Show Figures

Graphical abstract

11 pages, 2533 KiB  
Article
Prostate-Derived ETS Factor (PDEF) Modulates Yes Associated Protein 1 (YAP1) in Prostate Cancer Cells: A Potential Cross-Talk between PDEF and Hippo Signaling
by Praveen Kumar Jaiswal, Suman Mohajan, Sweaty Koul, Fengtian Wang, Runhua Shi and Hari K. Koul
Pharmaceuticals 2019, 12(4), 181; https://doi.org/10.3390/ph12040181 - 10 Dec 2019
Cited by 3 | Viewed by 3675
Abstract
PDEF (prostate-derived ETS factor, also known as SAM-pointed domain containing ETS transcription factor (SPDEF)) is expressed in luminal epithelial cells of the prostate gland and associates with luminal phenotype. The Hippo pathway regulates cell growth/proliferation, cellular homeostasis, and organ development by modulating phosphorylation [...] Read more.
PDEF (prostate-derived ETS factor, also known as SAM-pointed domain containing ETS transcription factor (SPDEF)) is expressed in luminal epithelial cells of the prostate gland and associates with luminal phenotype. The Hippo pathway regulates cell growth/proliferation, cellular homeostasis, and organ development by modulating phosphorylation of its downstream effectors. In previous studies, we observed decreased levels of PDEF during prostate cancer progression. In the present study, we evaluated the effects of the expression of PDEF on total/phosphoprotein levels of YAP1 (a downstream effector of the Hippo pathway). We observed that the PC3 and DU145 cells transfected with PDEF (PDEF-PC3 and PDEF-DU145) showed an increased phospho-YAP1 (Ser127) and total YAP1 levels as compared to the respective PC3 vector control (VC-PC3) and DU145 vector control cells (VC-DU145). We also observed an increased cytoplasmic YAP1 levels in PDEF-PC3 cells as compared to VC-PC3 cells. Moreover, our gene set enrichment analysis (GSEA) of mRNA expression in PDEF-PC3 and VC-PC3 cells revealed that PDEF resulted in inhibition of YAP1 target genes, directly demonstrating that PDEF plays a critical role in modulating YAP1 activity, and by extension in the regulation of the Hippo pathway. We also observed a decrease in YAP1 mRNA levels in prostate cancer tissues as compared to normal prostate tissues. Our analysis of multiple publicly available clinical cohorts revealed a gradual decrease in YAP1 mRNA expression during prostate cancer progression and metastasis. This decrease was similar to the decrease in PDEF levels, which we had reported earlier, and we observed a direct correlation between PDEF and YAP1 expression in CRPC data set. To the best of our knowledge, these results provide the first demonstration of inhibiting YAP1 activity by PDEF in any system and suggest a cross-talk between PDEF and the Hippo signaling pathway. Full article
(This article belongs to the Special Issue Protein Kinases and Cancer)
Show Figures

Figure 1

22 pages, 3119 KiB  
Article
CK2 Pro-Survival Role in Prostate Cancer Is Mediated via Maintenance and Promotion of Androgen Receptor and NFκB p65 Expression
by Janeen H. Trembley, Betsy T. Kren, Md. J. Abedin, Daniel P. Shaughnessy, Yingming Li, Scott M. Dehm and Khalil Ahmed
Pharmaceuticals 2019, 12(2), 89; https://doi.org/10.3390/ph12020089 - 14 Jun 2019
Cited by 11 | Viewed by 4181
Abstract
The prosurvival protein kinase CK2, androgen receptor (AR), and nuclear factor kappa B (NFκB) interact in the function of prostate cells, and there is evidence of crosstalk between these signals in the pathobiology of prostate cancer (PCa). As CK2 is elevated in PCa, [...] Read more.
The prosurvival protein kinase CK2, androgen receptor (AR), and nuclear factor kappa B (NFκB) interact in the function of prostate cells, and there is evidence of crosstalk between these signals in the pathobiology of prostate cancer (PCa). As CK2 is elevated in PCa, and AR and NFκB are involved in the development and progression of prostate cancer, we investigated their interaction in benign and malignant prostate cells in the presence of altered CK2 expression. Our results show that elevation of CK2 levels caused increased levels of AR and NFκB p65 in prostate cells of different phenotypes. Analysis of TCGA PCa data indicated that AR and CK2α RNA expression are strongly correlated. Small molecule inhibition or molecular down-regulation of CK2 caused reduction in AR mRNA expression and protein levels in PCa cells and in orthotopic xenograft tumors by various pathways. Among these, regulation of AR protein stability plays a unifying role in CK2 maintenance of AR protein levels. Our results show induction of various endoplasmic reticulum stress signals after CK2 inhibition, which may play a role in the PCa cell death response. Of note, CK2 inhibition caused loss of cell viability in both parental and enzalutamide-resistant castrate-resistant PCa cells. The present work elucidates the specific link of CK2 to the pathogenesis of PCa in association with AR and NFκB expression; further, the observation that inhibition of CK2 can exert a growth inhibitory effect on therapy-resistant PCa cells emphasizes the potential utility of CK2 inhibition in patients who are on enzalutamide treatment for advanced cancer. Full article
(This article belongs to the Special Issue Protein Kinases and Cancer)
Show Figures

Figure 1

20 pages, 3420 KiB  
Article
CDK11 Loss Induces Cell Cycle Dysfunction and Death of BRAF and NRAS Melanoma Cells
by Rehana L. Ahmed, Daniel P. Shaughnessy, Todd P. Knutson, Rachel I. Vogel, Khalil Ahmed, Betsy T. Kren and Janeen H. Trembley
Pharmaceuticals 2019, 12(2), 50; https://doi.org/10.3390/ph12020050 - 02 Apr 2019
Cited by 9 | Viewed by 4370
Abstract
Cyclin dependent kinase 11 (CDK11) is a protein kinase that regulates RNA transcription, pre-mRNA splicing, mitosis, and cell death. Targeting of CDK11 expression levels is effective in the experimental treatment of breast and other cancers, but these data are lacking in melanoma. To [...] Read more.
Cyclin dependent kinase 11 (CDK11) is a protein kinase that regulates RNA transcription, pre-mRNA splicing, mitosis, and cell death. Targeting of CDK11 expression levels is effective in the experimental treatment of breast and other cancers, but these data are lacking in melanoma. To understand CDK11 function in melanoma, we evaluated protein and RNA levels of CDK11, Cyclin L1 and Cyclin L2 in benign melanocytes and BRAF- as well as NRAS-mutant melanoma cell lines. We investigated the effectiveness of reducing expression of this survival kinase using RNA interference on viability, clonal survival, and tumorsphere formation in melanoma cell lines. We examined the impact of CDK11 loss in BRAF-mutant melanoma on more than 700 genes important in cancer signaling pathways. Follow-up analysis evaluated how CDK11 loss alters cell cycle function in BRAF- and NRAS-mutant melanoma cells. We present data on CDK11, CCNL1 and CCNL2 mRNA expression in melanoma patients, including prognosis for survival. In sum, we found that CDK11 is necessary for melanoma cell survival, and a major impact of CDK11 loss in melanoma is to cause disruption of the cell cycle distribution with accumulation of G1- and loss of G2/M-phase cancer cells. Full article
(This article belongs to the Special Issue Protein Kinases and Cancer)
Show Figures

Figure 1

Review

Jump to: Research

8 pages, 567 KiB  
Review
Small Molecule Induced FLT3 Degradation
by Sun-Young Han
Pharmaceuticals 2022, 15(3), 320; https://doi.org/10.3390/ph15030320 - 08 Mar 2022
Cited by 3 | Viewed by 3016
Abstract
Target protein degrader is a new paradigm in the small molecule drug discovery field and relates to the term ‘event-driven pharmacology’. Fms-like tyrosine kinase 3 (FLT3) is a significant target for treating acute myeloid leukemia (AML). A few FLT3 kinase inhibitors are currently [...] Read more.
Target protein degrader is a new paradigm in the small molecule drug discovery field and relates to the term ‘event-driven pharmacology’. Fms-like tyrosine kinase 3 (FLT3) is a significant target for treating acute myeloid leukemia (AML). A few FLT3 kinase inhibitors are currently used in the clinic for AML patients. However, resistance to current FLT3 inhibitors has emerged, and strategies to overcome this resistance are required. Small molecules downregulating FLT3 protein level are reported, exhibiting antileukemic effects against AML cell lines. Small molecules with various mechanisms such as Hsp90 inhibition, proteasome inhibition, RET inhibition, and USP10 inhibition are explained. In addition, reports of FLT3 as a client of Hsp90, current knowledge of the ubiquitin proteasome system for FLT3 degradation, the relationship with FLT3 phosphorylation status and susceptibility of FLT3 degradation are discussed. Full article
(This article belongs to the Special Issue Protein Kinases and Cancer)
Show Figures

Figure 1

26 pages, 1128 KiB  
Review
CDK4/6 and MAPK—Crosstalk as Opportunity for Cancer Treatment
by Lisa Scheiblecker, Karoline Kollmann and Veronika Sexl
Pharmaceuticals 2020, 13(12), 418; https://doi.org/10.3390/ph13120418 - 24 Nov 2020
Cited by 28 | Viewed by 6197
Abstract
Despite the development of targeted therapies and novel inhibitors, cancer remains an undefeated disease. Resistance mechanisms arise quickly and alternative treatment options are urgently required, which may be partially met by drug combinations. Protein kinases as signaling switchboards are frequently deregulated in cancer [...] Read more.
Despite the development of targeted therapies and novel inhibitors, cancer remains an undefeated disease. Resistance mechanisms arise quickly and alternative treatment options are urgently required, which may be partially met by drug combinations. Protein kinases as signaling switchboards are frequently deregulated in cancer and signify vulnerable nodes and potential therapeutic targets. We here focus on the cell cycle kinase CDK6 and on the MAPK pathway and on their interplay. We also provide an overview on clinical studies examining the effects of combinational treatments currently explored for several cancer types. Full article
(This article belongs to the Special Issue Protein Kinases and Cancer)
Show Figures

Figure 1

11 pages, 2207 KiB  
Review
Mitogen-Activated Protein Kinase Inhibitors and T-Cell-Dependent Immunotherapy in Cancer
by Sandeep Kumar, Daniel R. Principe, Sunil Kumar Singh, Navin Viswakarma, Gautam Sondarva, Basabi Rana and Ajay Rana
Pharmaceuticals 2020, 13(1), 9; https://doi.org/10.3390/ph13010009 - 07 Jan 2020
Cited by 22 | Viewed by 5796
Abstract
Mitogen-activated protein kinase (MAPK) signaling networks serve to regulate a wide range of physiologic and cancer-associated cell processes. For instance, a variety of oncogenic mutations often lead to hyperactivation of MAPK signaling, thereby enhancing tumor cell proliferation and disease progression. As such, several [...] Read more.
Mitogen-activated protein kinase (MAPK) signaling networks serve to regulate a wide range of physiologic and cancer-associated cell processes. For instance, a variety of oncogenic mutations often lead to hyperactivation of MAPK signaling, thereby enhancing tumor cell proliferation and disease progression. As such, several components of the MAPK signaling network have been proposed as viable targets for cancer therapy. However, the contributions of MAPK signaling extend well beyond the tumor cells, and several MAPK effectors have been identified as key mediators of the tumor microenvironment (TME), particularly with respect to the local immune infiltrate. In fact, a blockade of various MAPK signals has been suggested to fundamentally alter the interaction between tumor cells and T lymphocytes and have been suggested a potential adjuvant to immune checkpoint inhibition in the clinic. Therefore, in this review article, we discuss the various mechanisms through which MAPK family members contribute to T-cell biology, as well as circumstances in which MAPK inhibition may potentiate or limit cancer immunotherapy. Full article
(This article belongs to the Special Issue Protein Kinases and Cancer)
Show Figures

Figure 1

12 pages, 1351 KiB  
Review
CDK8-Novel Therapeutic Opportunities
by Ingeborg Menzl, Agnieszka Witalisz-Siepracka and Veronika Sexl
Pharmaceuticals 2019, 12(2), 92; https://doi.org/10.3390/ph12020092 - 19 Jun 2019
Cited by 32 | Viewed by 6685
Abstract
Improvements in cancer therapy frequently stem from the development of new small-molecule inhibitors, paralleled by the identification of biomarkers that can predict the treatment response. Recent evidence supports the idea that cyclin-dependent kinase 8 (CDK8) may represent a potential drug target for breast [...] Read more.
Improvements in cancer therapy frequently stem from the development of new small-molecule inhibitors, paralleled by the identification of biomarkers that can predict the treatment response. Recent evidence supports the idea that cyclin-dependent kinase 8 (CDK8) may represent a potential drug target for breast and prostate cancer, although no CDK8 inhibitors have entered the clinics. As the available inhibitors have been recently reviewed, we focus on the biological functions of CDK8 and provide an overview of the complexity of CDK8-dependent signaling throughout evolution and CDK8-dependent effects that may open novel treatment avenues. Full article
(This article belongs to the Special Issue Protein Kinases and Cancer)
Show Figures

Figure 1

29 pages, 1261 KiB  
Review
The Multi-Functional Calcium/Calmodulin Stimulated Protein Kinase (CaMK) Family: Emerging Targets for Anti-Cancer Therapeutic Intervention
by Joshua S. Brzozowski and Kathryn A. Skelding
Pharmaceuticals 2019, 12(1), 8; https://doi.org/10.3390/ph12010008 - 07 Jan 2019
Cited by 49 | Viewed by 6997
Abstract
The importance of Ca2+ signalling in key events of cancer cell function and tumour progression, such as proliferation, migration, invasion and survival, has recently begun to be appreciated. Many cellular Ca2+-stimulated signalling cascades utilise the intermediate, calmodulin (CaM). The Ca [...] Read more.
The importance of Ca2+ signalling in key events of cancer cell function and tumour progression, such as proliferation, migration, invasion and survival, has recently begun to be appreciated. Many cellular Ca2+-stimulated signalling cascades utilise the intermediate, calmodulin (CaM). The Ca2+/CaM complex binds and activates a variety of enzymes, including members of the multifunctional Ca2+/calmodulin-stimulated protein kinase (CaMK) family. These enzymes control a broad range of cancer-related functions in a multitude of tumour types. Herein, we explore the cancer-related functions of these kinases and discuss their potential as targets for therapeutic intervention. Full article
(This article belongs to the Special Issue Protein Kinases and Cancer)
Show Figures

Figure 1

21 pages, 7040 KiB  
Review
Natural Compounds and Derivatives as Ser/Thr Protein Kinase Modulators and Inhibitors
by Barbara Guerra and Olaf-Georg Issinger
Pharmaceuticals 2019, 12(1), 4; https://doi.org/10.3390/ph12010004 - 01 Jan 2019
Cited by 23 | Viewed by 6581
Abstract
The need for new drugs is compelling, irrespective of the disease. Focusing on medical problems in the Western countries, heart disease and cancer are at the moment predominant illnesses. Owing to the fact that ~90% of all 21,000 cellular proteins in humans are [...] Read more.
The need for new drugs is compelling, irrespective of the disease. Focusing on medical problems in the Western countries, heart disease and cancer are at the moment predominant illnesses. Owing to the fact that ~90% of all 21,000 cellular proteins in humans are regulated by phosphorylation/dephosphorylation it is not surprising that the enzymes catalysing these reactions (i.e., protein kinases and phosphatases, respectively) have attracted considerable attention in the recent past. Protein kinases are major team players in cell signalling. In tumours, these enzymes are found to be mutated disturbing the proper function of signalling pathways and leading to uncontrolled cellular growth and sustained malignant behaviour. Hence, the search for small-molecule inhibitors targeting the altered protein kinase molecules in tumour cells has become a major research focus in the academia and pharmaceutical companies. Full article
(This article belongs to the Special Issue Protein Kinases and Cancer)
Show Figures

Figure 1

Back to TopTop