Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 133(24)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37847564

ABSTRACT

A paucity of chemotherapeutic options for metastatic brain cancer limits patient survival and portends poor clinical outcomes. Using a CNS small-molecule inhibitor library of 320 agents known to be blood-brain barrier permeable and approved by the FDA, we interrogated breast cancer brain metastasis vulnerabilities to identify an effective agent. Metixene, an antiparkinsonian drug, was identified as a top therapeutic agent that was capable of decreasing cellular viability and inducing cell death across different metastatic breast cancer subtypes. This agent significantly reduced mammary tumor size in orthotopic xenograft assays and improved survival in an intracardiac model of multiorgan site metastases. Metixene further extended survival in mice bearing intracranial xenografts and in an intracarotid mouse model of multiple brain metastases. Functional analysis revealed that metixene induced incomplete autophagy through N-Myc downstream regulated 1 (NDRG1) phosphorylation, thereby leading to caspase-mediated apoptosis in both primary and brain-metastatic cells, regardless of cancer subtype or origin. CRISPR/Cas9 KO of NDRG1 led to autophagy completion and reversal of the metixene apoptotic effect. Metixene is a promising therapeutic agent against metastatic brain cancer, with minimal reported side effects in humans, which merits consideration for clinical translation.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Humans , Animals , Mice , Female , Cell Proliferation , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Autophagy , Cell Line, Tumor , Xenograft Model Antitumor Assays
2.
Nat Commun ; 14(1): 1566, 2023 03 22.
Article in English | MEDLINE | ID: mdl-36949040

ABSTRACT

Whereas the contribution of tumor microenvironment to the profound immune suppression of glioblastoma (GBM) is clear, tumor-cell intrinsic mechanisms that regulate resistance to CD8 T cell mediated killing are less understood. Kinases are potentially druggable targets that drive tumor progression and might influence immune response. Here, we perform an in vivo CRISPR screen to identify glioma intrinsic kinases that contribute to evasion of tumor cells from CD8 T cell recognition. The screen reveals checkpoint kinase 2 (Chek2) to be the most important kinase contributing to escape from CD8 T-cell recognition. Genetic depletion or pharmacological inhibition of Chek2 with blood-brain-barrier permeable drugs that are currently being evaluated in clinical trials, in combination with PD-1 or PD-L1 blockade, lead to survival benefit in multiple preclinical glioma models. Mechanistically, loss of Chek2 enhances antigen presentation, STING pathway activation and PD-L1 expression in mouse gliomas. Analysis of human GBMs demonstrates that Chek2 expression is inversely associated with antigen presentation and T-cell activation. Collectively, these results support Chek2 as a promising target for enhancement of response to immune checkpoint blockade therapy in GBM.


Subject(s)
Glioblastoma , Glioma , Humans , Animals , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , B7-H1 Antigen , Checkpoint Kinase 1 , Glioma/drug therapy , Glioma/genetics , Glioma/pathology , Glioblastoma/drug therapy , Glioblastoma/genetics , CD8-Positive T-Lymphocytes , Immunity , Tumor Microenvironment
3.
Mol Cancer Res ; 21(5): 389-396, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36652630

ABSTRACT

Immunotherapy has emerged as a powerful strategy for halting cancer progression. However, primary malignancies affecting the brain have been exempt to this success. Indeed, brain tumors continue to portend severe morbidity and remain a globally lethal disease. Extensive efforts have been directed at understanding how tumor cells survive and propagate within the unique microenvironment of the central nervous system (CNS). Cancer genetic aberrations and metabolic abnormalities provoke a state of persistent endoplasmic reticulum (ER) stress that in turn promotes tumor growth, invasion, therapeutic resistance, and the dynamic reprogramming of the infiltrating immune cells. Consequently, targeting ER stress is a potential therapeutic approach. In this work, we provide an overview of how ER stress response is advantageous to brain tumor development, discuss the significance of ER stress in governing antitumor immunity, and put forth therapeutic strategies of regulating ER stress to augment the effect of immunotherapy for primary CNS tumors.


Subject(s)
Brain Neoplasms , Brain , Humans , Brain Neoplasms/genetics , Oncogenes , Immunotherapy , Endoplasmic Reticulum Stress , Tumor Microenvironment
4.
Biochim Biophys Acta Mol Basis Dis ; 1867(11): 166228, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34311079

ABSTRACT

Breast cancer stem cells (CSCs) are distinct CD44+-subpopulations that are involved in metastasis and chemoresistance. However, the underlying molecular mechanism of CD44 in breast CSCs-mediated tumorigenesis remains elusive. We observed high CD44 expression in advanced-stage clinical breast tumor samples. CD44 activation in breast CSCs sorted from various triple negative breast cancer (TNBC) cell lines induced proliferation, migration, invasion, mammosphere formation that were reversed in presence of inhibitor, 4-methyl umbelliferone or CD44 silencing. CD44 activation in breast CSCs induced Src, Akt, and nuclear translocation of pSTAT3. PCR arrays revealed differential expression of a metabolic gene, Lipoprotein lipase (LPL), and transcription factor, SNAI3. Differential transcriptional regulation of LPL by pSTAT3 and SNAI3 was confirmed by promoter-reporter and chromatin immunoprecipitation analysis. Orthotopic xenograft murine breast tumor model revealed high tumorigenicity of CD24-/CD44+-breast CSCs as compared with CD24+-breast cancer cells. Furthermore, stable breast CSCs-CD44 shRNA and/or intratumoral administration of Tetrahydrolipstatin (LPL inhibitor) abrogated tumor progression and neoangiogenesis. Thus, LPL serves as a potential target for an efficacious therapeutics against aggressive breast cancer.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Hyaluronan Receptors/metabolism , Lipoprotein Lipase/genetics , Neoplastic Stem Cells/pathology , Animals , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinogenesis/drug effects , Cell Movement/drug effects , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Hyaluronan Receptors/genetics , Lipoprotein Lipase/antagonists & inhibitors , Mice , Orlistat/pharmacology , Orlistat/therapeutic use , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Xenograft Model Antitumor Assays
5.
Biochim Biophys Acta Mol Basis Dis ; 1867(7): 166141, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33845139

ABSTRACT

Triple-negative breast cancer (TNBC) patients often exhibit poor prognosis and breast cancer relapse due to metastasis. This results in secondary tumor generation at distant-unrelated organs that account for the majority of breast cancer-related deaths. Although breast cancer stem cells (CSCs) have been attributed to metastasis, a mechanistic understanding is essential for developing therapeutic interventions to combat breast cancer relapse. Breast CSCs are generated due to Epithelial-to-mesenchymal transition (EMT), regulated by transcription factors (EMT-TF) that are implicated in tumorigenesis and metastasis. However, the underlying mechanisms mediating these processes remain elusive. In the present study, we have reported that TWIST1, an EMT-TF, exhibits positive transcriptional regulation on PDGFRß promoter, thus identifying PDGFRß as one of the downstream targets of EMT regulation in breast CSCs. Breast cancer cells overexpressing PDGFRß exhibited a significant increase in physiological and molecular properties comparable to that of breast CSCs, while molecular silencing of PDGFRß in breast CSCs perturbed these phenomena. Mechanistically, PDGFRß overexpression induced the activation of FAK and Src leading to cell migration and invasion. Orthotopic xenograft transplantation of stable breast cancer cells and CSCs with PDGFRß overexpression in nude mice led to a significant increase in tumorigenesis, and metastasis to lung and liver as depicted by the significant increase in human gene-specific PDGFRß and CD44 expression, and colocalization along with an expression of human-specific Alu sequences which were perturbed with stable silencing of PDGFRß in breast CSCs. Thus, PDGFRß plays a crucial role in inducing breast cancer tumorigenesis and metastasis that can be a plausible therapeutic target to treat TNBC patients.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Neoplastic Stem Cells/pathology , Nuclear Proteins/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Twist-Related Protein 1/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinogenesis , Cell Cycle , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , Neoplastic Stem Cells/metabolism , Nuclear Proteins/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Transcriptional Activation , Tumor Cells, Cultured , Twist-Related Protein 1/genetics , Xenograft Model Antitumor Assays
6.
Hematol Oncol Stem Cell Ther ; 14(4): 275-289, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33744312

ABSTRACT

Breast cancer has the highest incidence rate of malignancy in women worldwide. A major clinical challenge faced by patients with breast cancer treated by conventional therapies is frequent relapse. This relapse has been attributed to the cancer stem cell (CSC) population that resides within the tumor and possess stemness properties. Breast CSCs are generated when breast cancer cells undergo epithelial-mesenchymal transition resulting in aggressive, highly metastatic, and invasive phenotypes that exhibit resistance towards chemotherapeutics. Metastasis, a phenomenon that aids in the migration of breast CSCs, occurs through any of three different routes: hematogenous, lymphatic, and transcoelomic. Hematogenous dissemination of breast CSCs leads to metastasis towards distant unrelated organs like lungs, liver, bone, and brain causing secondary tumor generation. Activation of metastasis genes or silencing of metastasis suppressor genes often leads to the advancement of metastasis. This review focuses on various genes and molecular factors that have been implicated to regulate organ-specific breast cancer metastasis by defying the available therapeutic interventions.


Subject(s)
Breast Neoplasms , Female , Humans , Neoplasm Metastasis , Recurrence
7.
Anticancer Agents Med Chem ; 21(14): 1783-1792, 2021.
Article in English | MEDLINE | ID: mdl-33355057

ABSTRACT

Epidermal Growth Factor Receptor (EGFR), a type-I transmembrane protein with intrinsic tyrosine kinase activity, is activated by peptide growth factors such as EGF, epigen, amphiregulin, etc. EGFR plays a vital role in regulating cell growth, migration, and differentiation in various tissue-specific cancers. It has been reported to be overexpressed in lung, head, and neck, colon, brain, pancreatic, and breast cancer that triggers tumor progression and drug resistance. EGFR overexpression alters the signaling pathway and induces cell division, invasion, and cell survival. Our prior studies demonstrated that EGFR inhibition modulates chemosensitivity in breast cancer stem cells, thereby serving as a potential drug target for breast cancer mitigation. Tyrosine kinase inhibitors (Lapatinib, Neratinib) and monoclonal antibodies (Trastuzumab) targeting EGFR have been developed and approved by the US FDA for clinical use against breast cancer. This review highlights the critical role of EGFR in breast cancer progression and enumerates the various approaches being undertaken to inhibit aggressive breast cancers by suppressing the downstream pathways. Furthermore, the mechanisms of action of potential molecules at various stages of drug development, as well as clinically approved drugs for breast cancer treatment, are illustrated.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Humans , Lapatinib/chemistry , Lapatinib/pharmacology , Protein Kinase Inhibitors/chemistry , Quinolines/chemistry , Quinolines/pharmacology , Trastuzumab/chemistry , Trastuzumab/pharmacology
8.
Anticancer Agents Med Chem ; 20(12): 1469-1474, 2020.
Article in English | MEDLINE | ID: mdl-32324523

ABSTRACT

BACKGROUND: Discovery of small molecules that inhibit tubulin polymerization is an attractive strategy for the development of new and improved anti-proliferative agents. OBJECTIVE: A series of novel 2-sulfonyl-1,1-diarylethenes were designed towards this end keeping in view the favorable chemical and pharmacological virtues of unsaturated sulfones. METHODS: Rapid, convenient and efficient two-step assembly of the designed molecules was achieved by the vicinal iodo-sulfonylation-Suzuki coupling sequence. RESULTS: As hypothesized, these compounds showed good anti-proliferative activity against different tissuespecific cancer cell lines: MCF-7, DU-145, A-549, HepG2, and HeLa. The most active compound, pnitrophenyl ring-bearing analog, exhibited an IC50 value of 0.90µM against A-549 cells. Flow cytometry studies on this derivative revealed that it arrests the cell cycle of A-549 cells at the G2/M phase. This compound exhibited molecular binding to tubulin as well as tubulin polymerization inhibition comparable to that of colchicine. CONCLUSION: A new class of potent, tubulin binding anticancer agents based on 1,1,-diarylvinyl sulfone scaffold has been designed and synthesized.


Subject(s)
Antineoplastic Agents/pharmacology , Sulfones/pharmacology , Tubulin Modulators/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/chemistry , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
9.
Comput Biol Chem ; 83: 107156, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31710991

ABSTRACT

BACKGROUND & OBJECTIVE: Epidermal growth factor receptor (EGFR) signaling pathway is one of the promising and well-established targets for anticancer therapy. The objective of the present study was to identify new EGFR inhibitors using ligand and structure-based drug designing methods, followed by a synthesis of selected inhibitors and evaluation of their activity. METHODS: A series of C-7-hydroxyproton substituted chrysin derivatives were virtually drawn to generate a small compound library that was screened using 3D QSAR model created from forty-two known EGFR tyrosine kinase inhibitors. Next, the obtained hits with fitness score ≥ 1.0 were subjected to molecular docking analysis. Based on the predicted activity and XP glide score, three EGFR inhibitors were synthesized and characterized using 1H-NMR, 13C-NMR and MS. Finally, comparative in vitro investigation of the biological activity of synthesized inhibitors was performed with that of the parent molecule, chrysin. RESULTS: The data depicted a 3.2-fold enhanced cytotoxicity of chrysin derivative, CHM-04 against breast cancer cells as compared with chrysin as well as its binding with EGFR protein. Furthermore, the biological activity of CHM-04 was comparable to the standard EGFR inhibitor, AG1478 in increasing apoptosis and decreasing the migratory potential of triple-negative breast cancer cells as well as significantly lowering the mammosphere forming ability of breast cancer stem cells. CONCLUSION: The present study suggests CHM-04, an EGFR inhibitor possessing drug-like properties as a plausible therapeutic candidate against breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Computer Simulation , Drug Design , Flavonoids/pharmacology , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Flavonoids/chemical synthesis , Flavonoids/chemistry , HEK293 Cells , Humans , Models, Molecular , Molecular Structure , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Quantitative Structure-Activity Relationship , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
10.
ACS Omega ; 3(1): 63-75, 2018 Jan 31.
Article in English | MEDLINE | ID: mdl-30023766

ABSTRACT

The total synthesis of highly potent and scarcely available marine natural product (-)-jahanyne was attempted resulting in a solution-phase synthesis of pruned versions with comparable activity. A simple and facile synthetic route was employed for the preparation of pruned congeners and would be scalable. The lipophilic tail of the natural product was synthesized from R-(+)-citronellol, utilizing easily available chemicals. All the synthesized compounds were screened for apoptotic activity against a panel of cell lines. These compounds depicted marked binding to B cell lymphoma 2 till 50 °C in cellular thermal shift analysis.

11.
FEBS J ; 284(12): 1830-1854, 2017 06.
Article in English | MEDLINE | ID: mdl-28398698

ABSTRACT

The recurrence of breast cancer in patients is a persistent challenge to the medical fraternity. Breast tumor contains a small population of cells with high tumor initiating and metastatic potential, known as cancer stem cells (CSCs), which are resistant to existing chemotherapeutics. CSCs contribute to the aggressiveness of triple negative breast cancers (TNBCs), thereby necessitating the identification of molecular targets on breast CSCs. TNBC cell line MDA-MB-231, in comparison with MCF-7, demonstrated a higher expression of epidermal growth factor receptor (EGFR). Thus, the naturally occurring flavanone, chrysin, with limited potential as a chemotherapeutic agent, was structurally modified by designing an analog with EGFR binding affinity using a molecular docking approach and subsequently synthesised. Chrysin analog CHM-09 and known EGFR inhibitors demonstrated a comparable anti-proliferative, anti-migratory activity along with the induction of apoptosis and cell cycle arrest in MDA-MB-231. Furthermore, sorted CD24- /CD44+ -breast CSCs and CD24+ -breast cancer cells from MDA-MB-231 demonstrated a markedly high expression of EGFR in the former than in the latter. CHM-09 and EGFR inhibitors could perturb EGF-induced EGFR signalling of breast CSC proliferation, migration, mammosphere formation and mesenchymal tri-lineage differentiation. CHM-09 or EGFR inhibitors not only led to inactivation of EGFR downstream signalling pathways such as Akt, extracellular signal regulated kinase and signal transducer and activator of transcription 3, but also induction of mesenchymal-epithelial transition as confirmed by decreased N-cadherin and increased E-cadherin expression. Finally, combinatorial treatment of EGFR inhibitors and doxorubicin led to significant increase in breast CSCs responsiveness to a chemotherapeutic drug. The results of the present study suggest that EGFR is a therapeutic target in breast CSCs and that abrogation of EGFR signalling along with chemotherapeutic drugs is an effective approach against breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/antagonists & inhibitors , Neoplastic Stem Cells/pathology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Female , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/drug therapy , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...