Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Apoptosis ; 27(5-6): 342-353, 2022 06.
Article in English | MEDLINE | ID: mdl-35267106

ABSTRACT

Actinomycin D (ActD) was the first anticancer antibiotic approved for the management of human cancers. However, the notorious toxicity profile limits its widespread application in cancers, including cancers of the aerodigestive tract. Recent studies show that combining low-dose ActD with existing chemotherapies could potentially protect normal cells from the toxicity of chemotherapy drugs through p53 activation (cyclotherapy). An understanding of ActD's effect on p53 signaling is critical for the meaningful application of ActD in cyclotherapy-based combinations. This study evaluated the anti-tumor efficacy and mechanism of action of ActD in aerodigestive tract cancers. We found that ActD strongly inhibited the growth of a panel of aerodigestive tract cancer cell lines and induced efficient apoptosis, although the sensitivity varies among cell lines. The IC50 values of ActD spanned between 0.021 and 2.96 nM. Mechanistic studies revealed that ActD increased the expression of total and phosphorylated p53 (ser15) in a time- and dose-dependent manner. Moreover, ActD-induced apoptosis is dependent on p53 in cells expressing wild-type p53 and that ActD induced context-dependent differential expression of downstream targets p21 and PUMA without significant effects on p27. In the final analysis, this study revealed that p53-p21 is the predominant pathway activated by low-dose ActD, supporting further development of ActD in cyclotherapy.


Subject(s)
Neoplasms , Tumor Suppressor Protein p53 , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Dactinomycin/metabolism , Dactinomycin/pharmacology , Dactinomycin/therapeutic use , Humans , Neoplasms/drug therapy , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
Semin Cancer Biol ; 35 Suppl: S55-S77, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25749195

ABSTRACT

The evasion of anti-growth signaling is an important characteristic of cancer cells. In order to continue to proliferate, cancer cells must somehow uncouple themselves from the many signals that exist to slow down cell growth. Here, we define the anti-growth signaling process, and review several important pathways involved in growth signaling: p53, phosphatase and tensin homolog (PTEN), retinoblastoma protein (Rb), Hippo, growth differentiation factor 15 (GDF15), AT-rich interactive domain 1A (ARID1A), Notch, insulin-like growth factor (IGF), and Krüppel-like factor 5 (KLF5) pathways. Aberrations in these processes in cancer cells involve mutations and thus the suppression of genes that prevent growth, as well as mutation and activation of genes involved in driving cell growth. Using these pathways as examples, we prioritize molecular targets that might be leveraged to promote anti-growth signaling in cancer cells. Interestingly, naturally occurring phytochemicals found in human diets (either singly or as mixtures) may promote anti-growth signaling, and do so without the potentially adverse effects associated with synthetic chemicals. We review examples of naturally occurring phytochemicals that may be applied to prevent cancer by antagonizing growth signaling, and propose one phytochemical for each pathway. These are: epigallocatechin-3-gallate (EGCG) for the Rb pathway, luteolin for p53, curcumin for PTEN, porphyrins for Hippo, genistein for GDF15, resveratrol for ARID1A, withaferin A for Notch and diguelin for the IGF1-receptor pathway. The coordination of anti-growth signaling and natural compound studies will provide insight into the future application of these compounds in the clinical setting.


Subject(s)
Carcinogenesis/genetics , Cell Proliferation/genetics , Neoplasms/genetics , Neoplasms/therapy , Signal Transduction , DNA-Binding Proteins , Growth Differentiation Factor 15/genetics , Hippo Signaling Pathway , Humans , Kruppel-Like Transcription Factors/genetics , Molecular Targeted Therapy , Nuclear Proteins/genetics , PTEN Phosphohydrolase/genetics , Protein Serine-Threonine Kinases/genetics , Retinoblastoma Protein/genetics , Somatomedins/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics
3.
Semin Cancer Biol ; 35 Suppl: S276-S304, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26590477

ABSTRACT

Targeted therapies and the consequent adoption of "personalized" oncology have achieved notable successes in some cancers; however, significant problems remain with this approach. Many targeted therapies are highly toxic, costs are extremely high, and most patients experience relapse after a few disease-free months. Relapses arise from genetic heterogeneity in tumors, which harbor therapy-resistant immortalized cells that have adopted alternate and compensatory pathways (i.e., pathways that are not reliant upon the same mechanisms as those which have been targeted). To address these limitations, an international task force of 180 scientists was assembled to explore the concept of a low-toxicity "broad-spectrum" therapeutic approach that could simultaneously target many key pathways and mechanisms. Using cancer hallmark phenotypes and the tumor microenvironment to account for the various aspects of relevant cancer biology, interdisciplinary teams reviewed each hallmark area and nominated a wide range of high-priority targets (74 in total) that could be modified to improve patient outcomes. For these targets, corresponding low-toxicity therapeutic approaches were then suggested, many of which were phytochemicals. Proposed actions on each target and all of the approaches were further reviewed for known effects on other hallmark areas and the tumor microenvironment. Potential contrary or procarcinogenic effects were found for 3.9% of the relationships between targets and hallmarks, and mixed evidence of complementary and contrary relationships was found for 7.1%. Approximately 67% of the relationships revealed potentially complementary effects, and the remainder had no known relationship. Among the approaches, 1.1% had contrary, 2.8% had mixed and 62.1% had complementary relationships. These results suggest that a broad-spectrum approach should be feasible from a safety standpoint. This novel approach has potential to be relatively inexpensive, it should help us address stages and types of cancer that lack conventional treatment, and it may reduce relapse risks. A proposed agenda for future research is offered.


Subject(s)
Genetic Heterogeneity , Molecular Targeted Therapy , Neoplasms/therapy , Precision Medicine , Antineoplastic Agents, Phytogenic/therapeutic use , Drug Resistance, Neoplasm/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/prevention & control , Signal Transduction , Tumor Microenvironment/genetics
4.
Cancers (Basel) ; 16(1)2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38201459

ABSTRACT

The aberrant transformation of normal cells into cancer cells, known as carcinogenesis, is a complex process involving numerous genetic and molecular alterations in response to innate and environmental stimuli. The Src family kinases (SFK) are key components of signaling pathways implicated in carcinogenesis, with c-Src and its oncogenic counterpart v-Src often playing a significant role. The discovery of c-Src represents a compelling narrative highlighting groundbreaking discoveries and valuable insights into the molecular mechanisms underlying carcinogenesis. Upon oncogenic activation, c-Src activates multiple downstream signaling pathways, including the PI3K-AKT pathway, the Ras-MAPK pathway, the JAK-STAT3 pathway, and the FAK/Paxillin pathway, which are important for cell proliferation, survival, migration, invasion, metastasis, and drug resistance. In this review, we delve into the discovery of c-Src and v-Src, the structure of c-Src, and the molecular mechanisms that activate c-Src. We also focus on the various signaling pathways that c-Src employs to promote oncogenesis and resistance to chemotherapy drugs as well as molecularly targeted agents.

5.
Sci Rep ; 13(1): 353, 2023 01 07.
Article in English | MEDLINE | ID: mdl-36611126

ABSTRACT

Head and neck cancer is the sixth most common malignancy, and there is an urgent need to identify physiological processes contributing to tumorigenesis. Extracellular acidification caused by aerobic glycolysis within tumor microenvironments can stimulate proton-sensing receptors. GPR68, or ovarian cancer G protein-coupled receptor 1, responds to extracellular acidity and is highly expressed in head and neck squamous cell carcinoma (HNSCC) as well as normal esophageal tissue. To study the role of GPR68 in oral dysplasia, wild-type and GPR68-/- mice were treated with 4-Nitroquinoline N-oxide (4NQO) in drinking water for 11-13 weeks, followed by normal water for 11-12 weeks. 4NQO treatment resulted in 45 percent of GPR68-/- mice developing severe dysplasia or squamous cell carcinoma compared to only 10.5 percent of GPR68+/+ mice. This correlated with increased frequencies of regulatory T cells in the spleens of male GPR68-/- mice. Dysplastic regions of the tongue had increased CD31 staining compared to normal regions in both GPR68-/- and GPR68+/+ mice, suggesting that angiogenesis was GPR68-independent. RNA knockdown studies using HNSCC cell lines demonstrated no direct effect of GPR68 on survival or growth. Overall, we demonstrate that GPR68-deficiency worsens the severity of chemical-induced oral dysplasia, suggesting a protective role for this gene in tumorigenesis.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Male , Mice , Animals , Squamous Cell Carcinoma of Head and Neck , Carcinoma, Squamous Cell/pathology , Carcinogenesis/pathology , 4-Nitroquinoline-1-oxide/toxicity , Cell Transformation, Neoplastic , Head and Neck Neoplasms/chemically induced , Head and Neck Neoplasms/genetics , Hyperplasia , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Tumor Microenvironment
6.
J Biol Chem ; 285(45): 34557-65, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20826787

ABSTRACT

Natural dietary agents have drawn a great deal of attention toward cancer prevention because of their wide safety margin. However, single agent intervention has failed to bring the expected outcome in clinical trials; therefore, combinations of chemopreventive agents are gaining increasingly popularity. In the present study, we investigated a combinatorial approach using two natural dietary polyphenols, luteolin and EGCG, and found that their combination at low doses (at which single agents induce minimal apoptosis) synergistically increased apoptosis (3-5-fold more than the additive level of apoptosis) in both head and neck and lung cancer cell lines. This combination also significantly inhibited growth of xenografted tumors in nude mice. The in vivo findings also were supported by significant inhibition of Ki-67 expression and increase in TUNEL-positive cells in xenografted tissues. Mechanistic studies revealed that the combination induced mitochondria-dependent apoptosis in some cell lines and mitochondria-independent apoptosis in others. Moreover, we found more efficient stabilization and ATM-dependent Ser(15) phosphorylation of p53 due to DNA damage by the combination, and ablation of p53 using shRNA strongly inhibited apoptosis as evidenced by decreased poly(ADP-ribose) polymerase and caspase-3 cleavage. In addition, we observed mitochondrial translocation of p53 after treatment with luteolin or the combination of EGCG and luteolin. Taken together, our results for the first time suggest that the combination of luteolin and EGCG has synergistic/additive growth inhibitory effects and provides an important rationale for future chemoprevention trials of head and neck and lung cancers.


Subject(s)
Anticarcinogenic Agents/pharmacology , Catechin/analogs & derivatives , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/prevention & control , Luteolin/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Anticarcinogenic Agents/agonists , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins , Caspase 3/metabolism , Catechin/agonists , Catechin/pharmacology , Cell Cycle Proteins/pharmacology , Cell Line, Tumor , DNA Damage/drug effects , DNA-Binding Proteins/pharmacology , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ki-67 Antigen/biosynthesis , Luteolin/agonists , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasm Transplantation , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/pharmacology , Protein Stability/drug effects , Tumor Suppressor Proteins/pharmacology , Xenograft Model Antitumor Assays
7.
Proc Natl Acad Sci U S A ; 105(17): 6314-9, 2008 Apr 29.
Article in English | MEDLINE | ID: mdl-18434539

ABSTRACT

p53-dependent G(1) and G(2) cell cycle checkpoints are activated in response DNA damage that help to maintain genomic stability. p53 also helps to protect cells from damage that occurs during S phase, for example, when the cells are starved for DNA precursors or irradiated with a low dose of UV. p53 is activated in normal cells starved for pyrimidine nucleotides by treatment with N-(phosphonacetyl)-l-aspartate (PALA). The treated cells progress through a first S phase with kinetics similar to those of untreated cells. However, the DNA of the treated cells begins to become damaged rapidly, within 12 h, as revealed by a comet assay, which detects broken DNA, and by staining for phosphorylated histone H2AX, which accumulates at sites of DNA damage. Because the cells survive, the damage must be reversible, suggesting single-strand breaks or gaps as the most likely possibility. The transiently damaged DNA stimulates activation of ATR and CHK1, which in turn catalyze the phosphorylation and accumulation of p53. Although PALA-induced DNA damage occurs only in dividing cells, the p53 that is activated is only competent to transcribe genes such as p21 and macrophage inhibitory cytokine 1 (whose products regulate G(2) and G(1) or S phase checkpoints, respectively) after the cells have exited the S phase during which damage occurs. We propose that p53 is activated by stimulation of mismatch repair in response to the misincorporation of deoxynucleotides into newly synthesized DNA, long before the lack of pyrimidine nucleoside triphosphates causes the rate of DNA synthesis to slow appreciably.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , DNA/biosynthesis , Nucleotides/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Aspartic Acid/analogs & derivatives , Aspartic Acid/pharmacology , Ataxia Telangiectasia Mutated Proteins , Cell Line , Checkpoint Kinase 1 , DNA-Binding Proteins/metabolism , Humans , Models, Biological , Phosphonoacetic Acid/analogs & derivatives , Phosphonoacetic Acid/pharmacology , Phosphorylation/drug effects , Pyrimidines/metabolism , S Phase/drug effects , Tumor Suppressor Proteins/metabolism
8.
Eur J Pharmacol ; 906: 174266, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34146588

ABSTRACT

Curcumin, a pure compound extracted from the flowering plant, turmeric (Curcuma longa. Zingiberaceae), is a common dietary ingredient found in curry powder. It has been studied extensively for its anti-inflammatory, antioxidant, antimicrobial and anti-tumour activities. Evidence is accumulating demonstrating its potential in chemoprevention and as an anti-tumour agent for the treatment of cancer. Despite demonstrated safety and tolerability, the clinical application of curcumin is frustrated by its poor solubility, metabolic instability and low oral bioavailability. Consequently researchers have tried novel techniques of formulation and delivery as well as synthesis of analogues with enhanced properties to overcome these barriers. This review presents the synthetic analogues of curcumin that have proven their anticancer potential from different studies. It also highlights studies that combined these analogues with approved chemotherapies and delivered them via novel techniques. Currently, there are no reports of clinical studies on any of the synthetic congeners of curcumin and this presents an opportunity for future research. This review presents the synthetic analogues of curcumin and makes a compelling argument for their potential application in the management of cancerous disease.


Subject(s)
Curcumin/therapeutic use , Neoplasms/drug therapy , Neoplasms/prevention & control , Animals , Cell Line, Tumor , Curcumin/analogs & derivatives , Curcumin/chemical synthesis , Humans , Molecular Structure , Structure-Activity Relationship , Xenograft Model Antitumor Assays
9.
Oncol Rep ; 45(5)2021 05.
Article in English | MEDLINE | ID: mdl-33864659

ABSTRACT

Despite widespread interest in chemoprevention and therapy due to the high margin of safety of dietary natural compounds, clinical intervention with single agents has failed to yield the expected outcomes, mostly due to poor bioavailability and low potency. Combinations of natural agents with synergistic effects are gaining increasing attention. In the present study, in vitro and in vivo antitumor effects of a combination of two natural dietary agents, green tea epigallocatechin gallate (EGCG) and resveratrol were investigated. It was revealed that their combination at low doses (at which single agents induce minimal apoptosis) synergistically increased apoptosis (combination index < 1) in head and neck cancer cell lines. Synergistic apoptosis was also supported by caspase­3 and PARP cleavage. The combination also significantly inhibited growth of xenografted head and neck tumors in nude mice as supported by significant inhibition of tumor volume, tumor weight and Ki67 expression, and increase in TUNEL­positive cells. Mechanistic studies revealed that the combination inhibited AKT­mTOR signaling both in vitro and in vivo. In addition, overexpression of constitutively active AKT protected cells from apoptosis induced by the combination of EGCG and resveratrol. Collectively, the present results for the first time suggest that the combination of EGCG and resveratrol has synergistic growth inhibitory effects and provide an important rationale for future clinical development for chemoprevention and treatment of head and neck cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Catechin/analogs & derivatives , Head and Neck Neoplasms/prevention & control , Resveratrol/pharmacology , Animals , Anticarcinogenic Agents/therapeutic use , Apoptosis/drug effects , Carcinogenesis/drug effects , Catechin/pharmacology , Catechin/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Head and Neck Neoplasms/pathology , Humans , Mice , Resveratrol/therapeutic use , Signal Transduction/drug effects , Tea/chemistry , Xenograft Model Antitumor Assays
10.
ACS Med Chem Lett ; 12(5): 758-767, 2021 May 13.
Article in English | MEDLINE | ID: mdl-34055223

ABSTRACT

TRPM8 antagonists derived from its cognate ligand, (-)-menthol, are underrepresented. We determine the absolute stereochemistry of a well-known TRPM8 antagonist, (-)-menthyl 1, using VCD and 2D NMR. We explore 1 for its antagonist effects of the human TRPM8 (hTRPM8) orthologue to uncover species-dependent inhibition versus rat channels. (-)-Menthyl 1 inhibits menthol- and icilin-evoked Ca2+ responses at hTRPM8 with IC50 values of 805 ± 200 nM and 1.8 ± 0.6 µM, respectively, while more potently inhibiting agonist responses at the rat orthologue (rTRPM8 IC50 (menthol) = 117 ± 18 nM, IC50 (icilin) = 521 ± 20 nM). Whole-cell patch-clamp recordings of hTRPM8 confirm the 1 inhibition of menthol-stimulated currents, with an IC50 of 700 ± 200 nM. We demonstrate that 1 possesses ≥400-fold selectivity for hTRPM8 versus hTRPA1/hTRPV1. (-)-menthyl 1 can be used as a novel chemical tool to study hTRPM8 pharmacology and differences in species commonly used in drug discovery.

11.
Nutr Cancer ; 62(7): 973-87, 2010.
Article in English | MEDLINE | ID: mdl-20924973

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is one of the most fatal cancers worldwide. Despite advances in the management of HNSCC, the overall survival for patients has not improved significantly due to advanced stages at diagnosis, high recurrence rate after surgical removal, and second primary tumor development, which underscore the importance of novel strategies for cancer prevention. Cancer chemoprevention, the use of natural or synthetic compounds to prevent, arrest, or reverse the process of carcinogenesis at its earliest stages, aims to reverse premalignancies and prevent second primary tumors. Genomics and proteomics information including initial mutation, cancer promotion, progression, and susceptibility has brought molecularly targeted therapies for drug development. The development of preventive approaches using specific natural or synthetic compounds, or both, requires a depth of understanding of the cross-talk between cancer signaling pathways and networks to retain or enhance chemopreventive activity while reducing known toxic effects. Many natural dietary compounds have been identified with multiple molecular targets, effective in the prevention and treatment of cancer. This review describes recent advances in the understanding of the complex signaling networks driving cancer progression and of molecularly targeted natural compounds under preclinical and clinical investigation.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinoma, Squamous Cell/prevention & control , Head and Neck Neoplasms/prevention & control , Animals , Catechin/analogs & derivatives , Catechin/pharmacology , Curcumin/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Humans , NF-kappa B/antagonists & inhibitors , Resveratrol , Stilbenes/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Suppressor Protein p53/physiology
12.
ACS Chem Neurosci ; 11(3): 268-290, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31850745

ABSTRACT

Structure-activity relationship studies of a reported menthol-based transient receptor potential cation channel subfamily M member 8 channel (TRPM8) antagonist, guided by computational simulations and structure-based design, uncovers a novel series of TRPM8 antagonists with >10-fold selectivity versus related TRP subtypes. Spiro[4.5]decan-8-yl analogue 14 inhibits icilin-evoked Ca2+ entry in HEK-293 cells stably expressing human TRPM8 (hTRPM8) with an IC50 of 2.4 ± 1.0 nM, while in whole-cell patch-clamp recordings this analogue inhibits menthol-evoked currents with a hTRPM8 IC50 of 64 ± 2 nM. Molecular dynamics (MD) simulations of compound 14 in our homology model of hTRPM8 suggest that this antagonist forms extensive hydrophobic contacts within the orthosteric site. In the wet dog shakes (WDS) assay, compound 14 dose-dependently blocks icilin-triggered shaking behaviors in mice. Upon local administration, compound 14 dose dependently inhibits cold allodynia evoked by the chemotherapy oxaliplatin in a murine model of peripheral neuropathy at microgram doses. Our findings suggest that 14 and other biphenyl amide analogues within our series can find utility as potent antagonist chemical probes derived from (-)-menthol as well as small molecule therapeutic scaffolds for chemotherapy-induced peripheral neuropathy (CIPN) and other sensory neuropathies.


Subject(s)
Biphenyl Compounds/antagonists & inhibitors , Hyperalgesia/drug therapy , Peripheral Nervous System Diseases/drug therapy , Structure-Activity Relationship , TRPM Cation Channels/metabolism , Amides , Calcium/metabolism , HEK293 Cells , Humans , Menthol/analogs & derivatives , Patch-Clamp Techniques/methods , TRPM Cation Channels/drug effects , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/metabolism
13.
Cancer Res ; 67(12): 5617-21, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575126

ABSTRACT

Virtually all human cancers encounter disruption of the "p53 network." From a therapeutic point of view, it is important to devise strategies that eliminate cancer cells, which are often defective in functional p53 and protect p53-expressing normal cells. By comparing the response of a pair of isogenic cell lines, we identify a plant-derived compound, Concanavalin A (Con A), which differentially kills p53-null cells. Further, we find that p53 family member, p73, plays a critical role that is unmasked in the absence of p53. Con A treatment leads to induction of p73 and several others that are important mediators of apoptosis and act downstream, such as p21, Bax, Foxo1a, and Bim. Inactivation of p73 reverses the expression of these proteins and apoptosis. Inhibition of Akt activation sensitizes otherwise resistant cells. These observations thus reveal a novel role for p73 in the regulation of Akt-Foxo1a-Bim signaling and apoptosis especially when p53 is absent.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Concanavalin A/pharmacology , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/drug effects , Bcl-2-Like Protein 11 , Blotting, Western , Cell Line , Fibroblasts/drug effects , Fibroblasts/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , In Situ Nick-End Labeling , Membrane Proteins/drug effects , Plant Lectins/pharmacology , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Protein p73 , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism
14.
Mol Cancer Ther ; 16(4): 729-738, 2017 04.
Article in English | MEDLINE | ID: mdl-28119490

ABSTRACT

We previously reported that the EGFR-targeted inhibitor erlotinib induces G1 arrest of squamous cell carcinoma of the head and neck (SCCHN) cell lines without inducing significant apoptosis. Large-scale genomic studies suggest that >50% of SCCHN cases have activation of PI3K pathways. This study investigated whether cotargeting of EGFR and PI3K has synergistic antitumor effects and apoptosis induction. We examined growth suppression, apoptosis, and signaling pathway modulation resulting from single and combined targeting of EGFR and PI3K with erlotinib and BKM120, respectively, in a panel of SCCHN cell lines and a xenograft model of SCCHN. In a panel of 12 cell lines, single targeting of EGFR with erlotinib or PI3K with BKM120 suppressed cellular growth without inducing significant apoptosis. Cotargeting of EGFR and PI3K synergistically inhibited SCCHN cell line and xenograft tumor growth, but induced variable apoptosis; some lines were highly sensitive, others were resistant. Mechanistic studies revealed that the combination inhibited both axes of the mTORC1 (S6 and 4EBP1) pathway in apoptosis-sensitive cell lines along with translational inhibition of Bcl-2, Bcl-xL, and Mcl-1, but failed to inhibit p-4EBP1, Bcl-2, Bcl-xL, and Mcl-1 in an apoptosis-resistant cell line. siRNA-mediated knockdown of eIF4E inhibited Bcl-2 and Mcl-1 and sensitized this cell line to apoptosis. Our results strongly suggest that cotargeting of EGFR and PI3K is synergistic and induces apoptosis of SCCHN cell lines by inhibiting both axes of the AKT-mTOR pathway and translational regulation of antiapoptotic Bcl-2 proteins. These findings may guide the development of clinical trials using this combination of agents. Mol Cancer Ther; 16(4); 729-38. ©2017 AACR.


Subject(s)
Aminopyridines/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Drug Resistance, Neoplasm/drug effects , Erlotinib Hydrochloride/administration & dosage , Head and Neck Neoplasms/drug therapy , Morpholines/administration & dosage , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/genetics , Aminopyridines/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cytoprotection/drug effects , Drug Synergism , Erlotinib Hydrochloride/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/genetics , Humans , Mice , Morpholines/pharmacology , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck , Xenograft Model Antitumor Assays
15.
Clin Cancer Res ; 23(3): 677-686, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27358485

ABSTRACT

PURPOSE: Our previous work suggested that HER3 inhibition sensitizes head and neck squamous cell carcinoma (HNSCC) to EGFR inhibition with cetuximab. This study aimed to define the role of HER3 in cetuximab resistance and the antitumor mechanisms of EGFR/HER3 dual targeting in HNSCC. EXPERIMENTAL DESIGN: We treated cetuximab-resistant HNSCC UMSCC1-C and parental UMSCC1-P cell lines with anti-EGFR antibody cetuximab, anti-HER3 antibody MM-121, and their combination. We assessed activities of HER2, HER3, and downstream signaling pathways by Western blotting and cell growth by sulforhodamine B (SRB) and colony formation assays. HER3-specific shRNA was used to confirm the role of HER3 in cetuximab response. The combined efficacy and alterations in biomarkers were evaluated in UMSCC1-C xenograft and patient-derived xenograft (PDX) models. RESULTS: Cetuximab treatment induced HER3 activation and HER2/HER3 dimerization in HNSCC cell lines. Combined treatment with cetuximab and MM-121 blocked EGFR and HER3 activities and inhibited the PI3K/AKT and ERK signaling pathways and HNSCC cell growth more effectively than each antibody alone. HER3 knockdown reduced HER2 activation and resensitized cells to cetuximab. Cetuximab-resistant xenografts and PDX models revealed greater efficacy of dual EGFR and HER3 inhibition compared with single antibodies. In PDX tissue samples, cetuximab induced HER3 expression and MM-121 reduced AKT activity. CONCLUSIONS: Clinically relevant PDX models demonstrate that dual targeting of EGFR and HER3 is superior to EGFR targeting alone in HNSCC. Our study illustrates the upregulation of HER3 by cetuximab as one mechanism underlying resistance to EGFR inhibition in HNSCC, supporting further clinical investigations using multiple targeting strategies in patients who have failed cetuximab-based therapy. Clin Cancer Res; 23(3); 677-86. ©2016 AACR.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Squamous Cell/pathology , Cetuximab/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/pathology , Neoplasm Proteins/chemistry , Receptor, ErbB-2/chemistry , Receptor, ErbB-3/chemistry , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/metabolism , Cetuximab/administration & dosage , Cetuximab/therapeutic use , Dimerization , Drug Resistance, Neoplasm/physiology , Enzyme Induction/drug effects , Head and Neck Neoplasms/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Protein Conformation/drug effects , RNA Interference , Random Allocation , Receptor, ErbB-3/biosynthesis , Receptor, ErbB-3/genetics , Receptor, ErbB-3/immunology , Signal Transduction/drug effects , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
17.
PLoS One ; 11(12): e0168450, 2016.
Article in English | MEDLINE | ID: mdl-28033401

ABSTRACT

The anti-tumor effect of a chelating phen-based ligand 2,9-di-sec-butyl-1,10-phenanthroline (dsBPT) and its combination with cisplatin were examined in both lung and head and neck cancer cell lines and xenograft animal models in this study. The effects of this agent on cell cycle and apoptosis were investigated. Protein markers relevant to these mechanisms were also assessed. We found that the inhibitory effect of dsBPT on lung and head and neck cancer cell growth (IC50 ranged between 0.1-0.2 µM) was 10 times greater than that on normal epithelial cells. dsBPT alone induced autophagy, G1 cell cycle arrest, and apoptosis. Our in vivo studies indicated that dsBPT inhibited tumor growth in a dose-dependent manner in a head and neck cancer xenograft mouse model. The combination of dsBPT with cisplatin synergistically inhibited cancer cell growth with a combination index of 0.3. Moreover, the combination significantly reduced tumor volume as compared with the untreated control (p = 0.0017) in a head and neck cancer xenograft model. No organ related toxicities were observed in treated animals. Our data suggest that dsBPT is a novel and potent antitumor drug that warrants further preclinical and clinical development either as a single agent or in combination with known chemotherapy drugs such as cisplatin.


Subject(s)
Antineoplastic Agents/pharmacology , Phenanthrolines/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Drug Synergism , Head and Neck Neoplasms/pathology , Humans , Inhibitory Concentration 50 , Lung Neoplasms/pathology , Mice , Mice, Nude , Xenograft Model Antitumor Assays
18.
Cancer Prev Res (Phila) ; 9(1): 63-73, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26511491

ABSTRACT

Despite its high promise for cancer prevention and therapy, the potential utility of curcumin in cancer is compromised by its low bioavailability and weak potency. The purpose of the current study was to assess the in vitro and in vivo efficacy and pharmacokinetic parameters of the potent curcumin analogue FLLL12 in SCCHN and identify the mechanisms of its antitumor effect. IC50 values against a panel of one premalignant and eight malignant head and neck cancer cell lines as well as apoptosis assay results suggested that FLLL12 is 10- to 24-fold more potent than natural curcumin depending on the cell line and induces mitochondria-mediated apoptosis. In vivo efficacy (xenograft) and pharmacokinetic studies also suggested that FLLL12 is significantly more potent and has more favorable pharmacokinetic properties than curcumin. FLLL12 strongly inhibited the expression of p-EGFR, EGFR, p-AKT, AKT, Bcl-2, and Bid and increased the expression of Bim. Overexpression of constitutively active AKT or Bcl-2 or ablation of Bim or Bid significantly inhibited FLLL12-induced apoptosis. Further mechanistic studies revealed that FLLL12 regulated EGFR and AKT at transcriptional levels, whereas Bcl-2 was regulated at the translational level. Finally, FLLL12 strongly inhibited the AKT downstream targets mTOR and FOXO1a and 3a. Taken together, our results strongly suggest that FLLL12 is a potent curcumin analogue with more favorable pharmacokinetic properties that induces apoptosis of head and neck cancer cell lines by inhibition of survival proteins including EGFR, AKT, and Bcl-2 and increasing of the proapoptotic protein Bim.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Curcumin/analogs & derivatives , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/prevention & control , Animals , Apoptosis , Biological Availability , Cell Line, Tumor , Curcumin/administration & dosage , Curcumin/pharmacokinetics , Drug Screening Assays, Antitumor , ErbB Receptors/metabolism , Female , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Mitochondria , Neoplasm Transplantation , Polymerase Chain Reaction , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/metabolism , Reproducibility of Results
19.
Oncogene ; 22(9): 1411-7, 2003 Mar 06.
Article in English | MEDLINE | ID: mdl-12618767

ABSTRACT

To evaluate the function of cysteine residues of the Src kinase, we constructed a series of Src mutants in which some of cysteines were replaced to alanines. With these mutants, we studied the effect of SH-alkylating agents, N-[p-(2-benzimidazolyl)phenyl] maleimide (BIPM) and N-(9-acridinyl) maleimide (NAM), on their kinase activity. Of 10 cysteine residues scattered over v-Src, either a single mutation at Cys520 or multiple mutations at the four clustered cyteines, Cys483, Cys487, Cys496 and Cys498, yielded clear resistance to the treatment with 10 microM BIPM or 1 microM NAM. In contrast, other cysteines including those in the SH2 domain and those in the catalytic cleft of the kinase domain were dispensable for the inactivation by BIPM and NAM. Similarly, deletion of SH2 and SH3 did not confer the resistance to v-Src, suggesting the inactivation by the SH-alkylating agents is SH2/SH3-independent. Although Cys520-mutated v-Src was resistant to 1 microM NAM, it was inactivated by 5 microM NAM. However, combined mutation including all of Cys483, Cys487, Cys496, Cys498 and Cys520 yielded clear resistance to 5 microM NAM. Among these mutants, those with double mutations in the four clustered cysteines yielded a temperature sensitive phenotype in the transfected cells, whereas Cys520 did not, suggesting that Cys520 has, at least in part, a discrete function. In contrast to v-Src, c-Src, which lacks cysteine at position 520, was resistant to 1 microM NAM but sensitive to 5 microM NAM. While replacement of Phe520 of c-Src to cysteine made it sensitive to 1 microM NAM, double mutation in clustered cysteines again yielded resistance to 5 microM NAM. Taken together, our results strongly suggest that the multiple cysteine residues clustered at the end of the C-terminal lobe are critical for the inhibition by the SH-alkylating agents and, thereby, have an allosteric repressor effect on the catalytic activity of Src in a SH2-phosphoTyr527 independent manner.


Subject(s)
Cysteine/chemistry , Oncogene Protein pp60(v-src)/chemistry , Protein-Tyrosine Kinases/chemistry , Alkylating Agents/pharmacology , Allosteric Regulation , Amino Acid Sequence , Animals , Avian Sarcoma Viruses/enzymology , Avian Sarcoma Viruses/genetics , COS Cells , CSK Tyrosine-Protein Kinase , Catalysis , Catalytic Domain , Chlorocebus aethiops , Codon , Drug Resistance , Enzyme Inhibitors/pharmacology , Maleimides/pharmacology , Molecular Sequence Data , Oncogene Protein pp60(v-src)/antagonists & inhibitors , Phosphotyrosine/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship , src Homology Domains , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL