Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 479
Filter
1.
N Engl J Med ; 386(21): 1998-2010, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35613022

ABSTRACT

BACKGROUND: Although hypomethylating agents are currently used to treat patients with cancer, whether they can also reactivate and up-regulate oncogenes is not well elucidated. METHODS: We examined the effect of hypomethylating agents on SALL4, a known oncogene that plays an important role in myelodysplastic syndrome and other cancers. Paired bone marrow samples that were obtained from two cohorts of patients with myelodysplastic syndrome before and after treatment with a hypomethylating agent were used to explore the relationships among changes in SALL4 expression, treatment response, and clinical outcome. Leukemic cell lines with low or undetectable SALL4 expression were used to study the relationship between SALL4 methylation and expression. A locus-specific demethylation technology, CRISPR-DNMT1-interacting RNA (CRISPR-DiR), was used to identify the CpG island that is critical for SALL4 expression. RESULTS: SALL4 up-regulation after treatment with hypomethylating agents was observed in 10 of 25 patients (40%) in cohort 1 and in 13 of 43 patients (30%) in cohort 2 and was associated with a worse outcome. Using CRISPR-DiR, we discovered that demethylation of a CpG island within the 5' untranslated region was critical for SALL4 expression. In cell lines and patients, we confirmed that treatment with a hypomethylating agent led to demethylation of the same CpG region and up-regulation of SALL4 expression. CONCLUSIONS: By combining analysis of patient samples with CRISPR-DiR technology, we found that demethylation and up-regulation of an oncogene after treatment with a hypomethylating agent can indeed occur and should be further studied. (Funded by Associazione Italiana per la Ricerca sul Cancro and others.).


Subject(s)
Antineoplastic Agents , Demethylation , Myelodysplastic Syndromes , Oncogenes , Up-Regulation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Clustered Regularly Interspaced Short Palindromic Repeats , Demethylation/drug effects , Humans , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Oncogenes/drug effects , Oncogenes/physiology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation/drug effects
2.
Clin Cancer Res ; 28(13): 2762-2770, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35486638

ABSTRACT

PURPOSE: Lurbinectedin suppresses the oncogenic transcription factor EWS-FLI1 through relocalization to the nucleolus, and delays tumor growth in mice bearing Ewing sarcoma xenografts. On the basis of this rationale, lurbinectedin was evaluated in patients with relapsed Ewing sarcoma. PATIENTS AND METHODS: This open-label, single-arm, Basket phase II trial included a cohort of 28 treated adult patients with confirmed Ewing sarcoma, measurable disease as per Response Evaluation Criteria In Solid Tumors (RECIST) v.1.1, Eastern Cooperative Oncology Group performance status ≤2, adequate organ function, no central nervous system metastasis, and pretreated with ≤2 chemotherapy lines for metastatic/recurrent disease. Patients received lurbinectedin 3.2 mg/m2 as a 1-hour infusion every 3 weeks. Primary endpoint was overall response rate (ORR) as per RECIST v.1.1. Secondary endpoints included time-to-event parameters and safety profile. RESULTS: ORR was 14.3% [95% confidence interval (CI), 4.0%-32.7%], with median duration of response of 4.2 months (95% CI, 2.9-5.5 months). Median progression-free survival was 2.7 months (95% CI, 1.4-4.3 months), clinical benefit rate was 39.3%, and disease control rate was 57.1%. With 39% censoring, median overall survival was 12.0 months (95% CI, 8.5-18.5 months). Most common grade 3/4 adverse events were neutropenia (57%), anemia, thrombocytopenia, and treatment-related febrile neutropenia (14% each). No deaths or discontinuations were due to toxicity. CONCLUSIONS: Lurbinectedin was active in the treatment of relapsed Ewing sarcoma and had a manageable safety profile. Lurbinectedin could represent a valuable addition to therapies for Ewing sarcoma, and is currently being evaluated in combination with irinotecan in advanced Ewing sarcoma in a phase Ib/II trial.


Subject(s)
Bone Neoplasms , Neoplasm Recurrence, Local , Sarcoma, Ewing , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Carbolines/adverse effects , Carbolines/pharmacology , Heterocyclic Compounds, 4 or More Rings/adverse effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Oncogenes/drug effects , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/genetics
3.
Biochem Pharmacol ; 196: 114725, 2022 02.
Article in English | MEDLINE | ID: mdl-34384758

ABSTRACT

Aberrant cholesterol metabolism and homeostasis in the form of elevated cholesterol biosynthesis and dysregulated efflux and metabolism is well recognized as a major feature of metabolic reprogramming in solid tumors. Recent studies have emphasized on major drivers and regulators such as Myc, mutant p53, SREBP2, LXRs and oncogenic signaling pathways that play crucial roles in tumor cholesterol metabolic reprogramming. Therapeutics such as statins targeting the mevalonate pathway were tried at the clinic without showing consistent benefits to cancer patients. Nuclear receptors are prominent regulators of mammalian metabolism. Their de-regulation often drives tumorigenesis. RORγ and its immune cell-specific isoform RORγt play important functions in control of mammalian metabolism, circadian rhythm and immune responses. Although RORγ, together with its closely related members RORα and RORß were identified initially as orphan receptors, recent studies strongly support the conclusion that specific intermediates and metabolites of cholesterol pathways serve as endogenous ligands of RORγ. More recent studies also reveal a critical role of RORγ in tumorigenesis through major oncogenic pathways including acting a new master-like regulator of tumor cholesterol biosynthesis program. Importantly, an increasing number of RORγ orthosteric and allosteric ligands are being identified that display potent activities in blocking tumor growth and autoimmune disorders in preclinical models. This review summarizes the recent preclinical and clinical progress on RORγ with emphasis on its role in reprogramming tumor cholesterol metabolism and its regulation. It will also discuss RORγ functional mechanisms, context-specificity and its value as a therapeutic target for effective cancer treatment.


Subject(s)
Anticholesteremic Agents/administration & dosage , Autoimmune Diseases/metabolism , Cholesterol/biosynthesis , Neoplasms/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Oncogenes/physiology , Animals , Antineoplastic Agents/administration & dosage , Autoimmune Diseases/drug therapy , Drug Delivery Systems/trends , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Neoplasms/drug therapy , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Oncogenes/drug effects
4.
Anticancer Agents Med Chem ; 22(3): 499-514, 2022.
Article in English | MEDLINE | ID: mdl-34802408

ABSTRACT

Phytochemicals are being used for thousands of years to prevent dreadful malignancy. Side effects of existing allopathic treatment have also initiated intense research in the field of bioactive phytochemicals. Gallic acid, a natural polyphenolic compound, exists freely as well as in polymeric forms. The anti-cancer properties of gallic acid are indomitable by a variety of cellular pathways such as induction of programmed cell death, cell cycle apprehension, reticence of vasculature and tumor migration, and inflammation. Furthermore, gallic acid is found to show synergism with other existing chemotherapeutic drugs. Therefore, the antineoplastic role of gallic acid suggests its promising therapeutic candidature in the near future. The present review describes all these aspects of gallic acid at a single platform. In addition nanotechnology-mediated approaches are also discussed to enhance bioavailability and therapeutic efficacy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Gallic Acid/pharmacology , Neoplasms/drug therapy , Oncogenes/drug effects , Phytochemicals/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Cell Proliferation/drug effects , Gallic Acid/chemistry , Humans , Neoplasms/pathology , Phytochemicals/chemistry
5.
Org Biomol Chem ; 19(45): 9953-9965, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34747958

ABSTRACT

Aiming at discovering novel, putative anticancer drugs featuring low-to-null side effects, natural compounds isolated from Juncaceae were studied here for their ability to target G-quadruplex structures originating from cancer-related telomeric and oncogene DNA sequences. Particularly, various dihydrophenanthrene, benzocoumarin and dihydrodibenzoxepin derivatives were firstly screened by the affinity chromatography-based G4-CPG assay, and the compound with the highest affinity and selectivity for G-quadruplexes (named J10) was selected for further studies. Fluorescence spectroscopy and circular dichroism experiments corroborated its capability to selectively recognize and stabilize G-quadruplexes over duplex DNA, also showing a preference for parallel G-quadruplexes. Molecular docking proved that the selective G-quadruplex interactions over duplex interactions could be due to the ability of J10 to bind to the grooves of the telomeric and oncogene G-quadruplex structures. Finally, biological assays demonstrated that J10 induces significant antiproliferative effects on human leukemia cells, with no relevant effects on healthy human fibroblasts. Interestingly, J10 exerts its antiproliferative action on tumor cells by activating the apoptotic pathway.


Subject(s)
Antineoplastic Agents , G-Quadruplexes , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Circular Dichroism , Humans , Molecular Docking Simulation , Oncogenes/drug effects , Telomere
6.
Nat Commun ; 12(1): 5906, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34625563

ABSTRACT

Mouse models are critical in pre-clinical studies of cancer therapy, allowing dissection of mechanisms through chemical and genetic manipulations that are not feasible in the clinical setting. In studies of the tumour microenvironment (TME), multiplexed imaging methods can provide a rich source of information. However, the application of such technologies in mouse tissues is still in its infancy. Here we present a workflow for studying the TME using imaging mass cytometry with a panel of 27 antibodies on frozen mouse tissues. We optimise and validate image segmentation strategies and automate the process in a Nextflow-based pipeline (imcyto) that is scalable and portable, allowing for parallelised segmentation of large multi-image datasets. With these methods we interrogate the remodelling of the TME induced by a KRAS G12C inhibitor in an immune competent mouse orthotopic lung cancer model, highlighting the infiltration and activation of antigen presenting cells and effector cells.


Subject(s)
Image Cytometry/methods , Oncogenes , Tumor Microenvironment/immunology , Animals , Antibodies , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/immunology , Disease Models, Animal , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/drug therapy , Macrophages , Mice , Mice, Inbred C57BL , Oncogenes/drug effects , T-Lymphocytes , Tumor Microenvironment/drug effects
7.
Clin Transl Med ; 11(8): e503, 2021 08.
Article in English | MEDLINE | ID: mdl-34459131

ABSTRACT

Intrinsic resistance to CDK4/6 inhibitors hinders their clinical utility in cancer treatment. Furthermore, the predictive markers of CDK4/6 inhibitors in gastric cancer (GC) remain incompletely described. Here, we found that PAX6 expression was negatively correlated with the response to palbociclib in vitro and in vivo in GC. We observed that the PAX6 expression level was negatively correlated with the overall survival of GC patients and further showed that PAX6 can promote GC cell proliferation and the cell cycle. The cell cycle is regulated by the interaction of cyclins with their partner serine/threonine cyclin-dependent kinases (CDKs), and the G1/S-phase transition is the main target of CDK4/6 inhibitors. Therefore, we tested whether PAX6 expression was correlated with the GC response to palbociclib. We found that PAX6 hypermethylates the promoter of LATS2 and inactivates the Hippo pathway, which upregulates cyclin D1 (CCND1) expression. This results in a suppressed response to palbociclib in GC. Furthermore, we found that the induction of the Hippo signaling pathway or treatment with a DNA methylation inhibitor could overcome PAX6-induced palbociclib resistance in GC. These findings uncover a tumor promoter function of PAX6 in GC and establish overexpressed PAX6 as a mechanism of resistance to palbociclib.


Subject(s)
Cyclin-Dependent Kinase 4/drug effects , Cyclin-Dependent Kinase 6/drug effects , Hippo Signaling Pathway/drug effects , PAX6 Transcription Factor/drug effects , Piperazines/pharmacology , Protein Serine-Threonine Kinases/drug effects , Pyridines/pharmacology , Stomach Neoplasms/drug therapy , Tumor Suppressor Proteins/drug effects , Aged , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , China , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , Disease Models, Animal , Female , Hippo Signaling Pathway/genetics , Humans , Male , Mice , Mice, Nude , Middle Aged , Oncogenes/drug effects , Oncogenes/genetics , PAX6 Transcription Factor/genetics , Protein Serine-Threonine Kinases/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Proteins/genetics
8.
Theranostics ; 11(17): 8172-8184, 2021.
Article in English | MEDLINE | ID: mdl-34373735

ABSTRACT

Chromosome translocations involving mixed lineage leukemia (MLL) gene cause acute leukemia with a poor prognosis. MLL is frequently fused with transcription cofactors AF4 (~35%), AF9 (25%) or its paralog ENL (10%). The AHD domain of AF9/ENL binds to AF4, its paralog AFF4, or histone-H3 lysine-79 (H3K79) methyltransferase DOT1L. Formation of AF9/ENL/AF4/AFF4-containing super elongation complexes (SEC) and the catalytic activity of DOT1L are essential for MLL-rearranged leukemia. Protein-protein interactions (PPI) between AF9/ENL and DOT1L/AF4/AFF4 are therefore a potential drug target. Methods: Compound screening followed by medicinal chemistry was used to find inhibitors of such PPIs, which were examined for their biological activities against MLL-rearranged leukemia and other cancer cells. Results: Compound-1 was identified to be a novel small-molecule inhibitor of the AF9/ENL-DOT1L/AF4/AFF4 interaction with IC50s of 0.9-3.5 µM. Pharmacological inhibition of the PPIs significantly reduced SEC and DOT1L-mediated H3K79 methylation in the leukemia cells. Gene profiling shows compound-1 significantly suppressed the gene signatures related to onco-MLL, DOT1L, HoxA9 and Myc. It selectively inhibited proliferation of onco-MLL- or Myc-driven cancer cells and induced cell differentiation and apoptosis. Compound-1 exhibited strong antitumor activity in a mouse model of MLL-rearranged leukemia. Conclusions: The AF9/ENL-DOT1L/AF4/AFF4 interactions are validated to be an anticancer target and compound-1 is a useful in vivo probe for biological studies as well as a pharmacological lead for further drug development.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute , Oncogene Proteins, Fusion , Animals , Gene Expression/drug effects , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/drug effects , Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mice , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/drug effects , Oncogene Proteins, Fusion/genetics , Oncogenes/drug effects , Protein Interaction Domains and Motifs , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/drug effects , Transcriptional Elongation Factors/genetics
9.
Neoplasia ; 23(9): 1016-1027, 2021 09.
Article in English | MEDLINE | ID: mdl-34403880

ABSTRACT

Tyrosine kinase inhibitors (TKIs) such as imatinib, nilotinib, dasatinib, and ponatinib have significantly improved the life expectancy of Philadelphia chromosome-positive (Ph+) acute lymphocytic leukemia (ALL) patients; however, resistance to TKIs remains a major clinical challenge. Point mutations in the tyrosine kinase domain (TKD) of BCR-ABL1 have emerged as the predominant cause of acquired resistance. In approximately 30% of patients, the mechanism of resistance to TKIs remains elusive. This study aimed to investigate mechanisms of nonmutational resistance in Ph+ ALL. Here we report the development of a nonmutational resistance cell line SupB15-RT; conferring resistance to approved ABL kinase inhibitors (AKIs) and allosteric inhibitors GNF-2, ABL001, and crizotinib, except for dasatinib (IC90 50nM), a multitarget kinase inhibitor. We found that the AKT/mTOR pathway is activated in these cells and their proliferation inhibited by Torin-1 with an IC50 of 24.7 nM. These observations were confirmed using 3 different ALL patient-derived long term cultures (PDLTCs): (1) HP (BCR-ABL1 negative), (2) PH (BCR-ABL1 positive and responsive to TKIs) and (3) BV (BCR-ABL1 positive and nonmutational resistant to TKIs). Furthermore, Torin-1 and NVP-BEZ235 induced apoptosis in PH and BV cells but not in HP cells. Our experiments provide evidence of the involvement of AKT/mTOR pathway in the evolution of nonmutational resistance in Ph+ ALL which will assist in developing novel targeted therapy for Ph+ ALL patients with BCR-ABL1 independent nonmutational resistance.


Subject(s)
Drug Resistance, Neoplasm/physiology , Oncogenes/physiology , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Humans , Jurkat Cells , MTOR Inhibitors/pharmacology , MTOR Inhibitors/therapeutic use , Oncogenes/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured
10.
Cell Rep Med ; 2(5): 100279, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34095881

ABSTRACT

Aberrant NOTCH3 signaling and overexpression is oncogenic, associated with cancer stem cells and drug resistance, yet therapeutic targeting remains elusive. Here, we develop NOTCH3-targeted antibody drug conjugates (NOTCH3-ADCs) by bioconjugation of an auristatin microtubule inhibitor through a protease cleavable linker to two antibodies with differential abilities to inhibit signaling. The signaling inhibitory antibody rapidly induces ligand-independent receptor clustering and internalization through both caveolin and clathrin-mediated pathways. The non-inhibitory antibody also efficiently endocytoses via clathrin without inducing receptor clustering but with slower lysosomal co-localization kinetics. In addition, DLL4 ligand binding to the NOTCH3 receptor mediates transendocytosis of NOTCH3-ADCs into ligand-expressing cells. NOTCH3-ADCs internalize into receptor and ligand cells independent of signaling and induce cell death in both cell types representing an atypical mechanism of ADC cytotoxicity. Treatment of xenografts with NOTCH3-ADCs leads to sustained tumor regressions, outperforms standard-of-care chemotherapy, and allows targeting of tumors that overexpress NOTCH3 independent of signaling inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Immunoconjugates/pharmacology , Receptor, Notch3/metabolism , Cell Line, Tumor/drug effects , Humans , Immunoconjugates/metabolism , Oncogenes/drug effects , Receptor, Notch3/immunology , Receptors, Notch/antagonists & inhibitors , Xenograft Model Antitumor Assays
11.
Cells ; 10(6)2021 05 23.
Article in English | MEDLINE | ID: mdl-34071075

ABSTRACT

Chronic hepatitis B virus (HBV) infection is the major etiology of hepatocellular carcinoma (HCC), frequently with HBV integrating into the host genome. HBV integration, found in 85% of HBV-associated HCC (HBV-HCC) tissue samples, has been suggested to be oncogenic. Here, we investigated the potential of HBV-HCC driver identification via the characterization of recurrently targeted genes (RTGs). A total of 18,596 HBV integration sites from our in-house study and others were analyzed. RTGs were identified by applying three criteria: at least two HCC subjects, reported by at least two studies, and the number of reporting studies. A total of 396 RTGs were identified. Among the 28 most frequent RTGs, defined as affected in at least 10 HCC patients, 23 (82%) were associated with carcinogenesis and 5 (18%) had no known function. Available breakpoint positions from the three most frequent RTGs, TERT, MLL4/KMT2B, and PLEKHG4B, were analyzed. Mutual exclusivity of TERT promoter mutation and HBV integration into TERT was observed. We present an RTG consensus through comprehensive analysis to enable the potential identification and discovery of HCC drivers for drug development and disease management.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/etiology , Hepatitis B virus/drug effects , Hepatitis B, Chronic/drug therapy , Liver Neoplasms/drug therapy , Aged , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Disease Management , Female , Humans , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Male , Middle Aged , Oncogenes/drug effects , Oncogenes/physiology
12.
Oncogene ; 40(24): 4079-4093, 2021 06.
Article in English | MEDLINE | ID: mdl-34079087

ABSTRACT

Receptor tyrosine kinases (RTKs) are transmembrane receptors of great clinical interest due to their role in disease, notably cancer. Since their discovery, several mechanisms of RTK dysregulation have been identified, resulting in multiple cancer types displaying 'oncogenic addiction' to RTKs. As a result, RTKs have represented a major class for targeted therapeutics over the past two decades, with numerous small molecule-based tyrosine kinase inhibitor (TKI) therapeutics having been developed and clinically approved for several cancers. However, many of the current RTK inhibitor treatments eventually result in the rapid development of acquired resistance and subsequent tumor relapse. Recent technological advances and tools are being generated for the identification of novel RTK small molecule therapeutics. These newer technologies will be important for the identification of diverse types of RTK inhibitors, targeting both the receptors themselves as well as key cellular factors that play important roles in the RTK signaling cascade.


Subject(s)
Neoplasms/drug therapy , Neoplasms/metabolism , Oncogenes/drug effects , Protein Kinase Inhibitors/pharmacology , Tyrosine/metabolism , Animals , Humans , Molecular Targeted Therapy/methods
13.
Int J Mol Sci ; 22(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807608

ABSTRACT

Oncogenic kinases contribute to immunosuppression and modulate the tumor microenvironment in solid tumors. Increasing evidence supports the fundamental role of oncogenic kinase signaling networks in coordinating immunosuppressive tumor microenvironments. This has led to numerous studies examining the efficacy of kinase inhibitors in inducing anti-tumor immune responses by increasing tumor immunogenicity. Kinase inhibitors are the second most common FDA-approved group of drugs that are deployed for cancer treatment. With few exceptions, they inevitably lead to intrinsic and/or acquired resistance, particularly in patients with metastatic disease when used as a monotherapy. On the other hand, cancer immunotherapies, including immune checkpoint inhibitors, have revolutionized cancer treatment for malignancies such as melanoma and lung cancer. However, key hurdles remain to successfully incorporate such therapies in the treatment of other solid cancers. Here, we review the recent literature on oncogenic kinases that regulate tumor immunogenicity, immune suppression, and anti-tumor immunity. Furthermore, we discuss current efforts in clinical trials that combine kinase inhibitors and immune checkpoint inhibitors to treat breast cancer and other solid tumors.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Animals , Humans , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Oncogenes/drug effects , Tumor Microenvironment/drug effects
14.
Can J Urol ; 28(2): 10643-10647, 2021 04.
Article in English | MEDLINE | ID: mdl-33872566

ABSTRACT

Primary scrotal melanoma represents the rarest genitourinary malignancy. We describe the 25th reported case. The 79-year-old patient presented with a rapidly enlarging right cutaneous scrotal mass which after local excision demonstrated pT4b nodular malignant melanoma (BRAF V600E mutation positive). The patient underwent wide local excision of his hemiscrotum and inguinal lymph node dissection demonstrating nodes positive for melanoma (pN2b). Postoperatively, the patient developed a left sided malignant pleural effusion (M1b). Per American Joint Commission Cancer staging, BRAF mutant targeted therapy (dabrafenib) was initiated. This case documents the first instance in which metastatic scrotal melanoma will be treated with oncogene targeted therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Genital Neoplasms, Male/surgery , Imidazoles/therapeutic use , Melanoma/secondary , Oximes/therapeutic use , Scrotum , Skin Neoplasms/pathology , Aged , Antineoplastic Agents/pharmacology , Genital Neoplasms, Male/genetics , Humans , Imidazoles/pharmacology , Male , Melanoma/genetics , Oncogenes/drug effects , Oximes/pharmacology
15.
Balkan Med J ; 38(1): 43-49, 2021 01.
Article in English | MEDLINE | ID: mdl-32975519

ABSTRACT

BACKGROUND: Multiple myeloma remains a virtually incurable hematologic malignancy, which is featured with the aberrant growth of malignant plasma cells. AIMS: To elucidate the functions of miR-19a-3p in multiple myeloma. STUDY DESIGN: Cell study. METHODS: Cell counting kit-8 assay was performed to detect cell viability, and flow cytometry was conducted to detect cell apoptosis. Bioinformatics analysis predicted miR-19a-3p-associated biological function, pathway, core regulatory network, and target genes. Luciferase reporter assay verified the target sequence of miR-19a-3p regulating FBXO32. RESULTS: miR-19a-3p is upregulated in multiple myeloma cells (p<0.01) and patients with multiple myeloma (p<0.001). Overexpressed miR-19a-3p significantly increased cell viability (p<0.05) and inhibited cell apoptosis (p<0.01). FBXO32 is a target gene of miR-19a-3p (p<0.01). Moreover, FBXO32 is downregulated in MM, and it significantly decreased cell viability (p<0.05) and promoted cell apoptosis (p<0.01). FBXO32 significantly rescued the influence of miR-19a-3p-inhibiting cell apoptosis (p<0.05). CONCLUSION: miR-19a-3p promoted cell proliferation and inhibited cell apoptosis by degrading the target FBXO32 mRNA in multiple myeloma.


Subject(s)
MicroRNAs/pharmacology , Multiple Myeloma/drug therapy , Muscle Proteins/antagonists & inhibitors , Oncogenes/drug effects , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Adult , Apoptosis/drug effects , Female , Humans , Male , MicroRNAs/therapeutic use , Middle Aged , Multiple Myeloma/physiopathology , Muscle Proteins/therapeutic use , Oncogenes/physiology , SKP Cullin F-Box Protein Ligases/therapeutic use
16.
Anticancer Agents Med Chem ; 21(2): 278-286, 2021.
Article in English | MEDLINE | ID: mdl-32698745

ABSTRACT

BACKGROUND: Oxaliplatin (L-OHP)-based chemotherapy, such as FOLFOX4 (5-fluorouracil, leucovorin, and L-OHP), improves the prognosis of patients with late-stage Hepatocellular Carcinoma (HCC). However, the development of resistance to L-OHP leads to the failure of chemotherapy. The aim of this study was to investigate the role of linc01559 and miR-6783-3p in regulating resistance to L-OHP. METHODS: Quantitative reverse transcription-polymerase chain reaction was used to determine the expression profile. The Cell Counting Kit-8 test and wound healing assay were also used. Dual-luciferase reporter gene assay, RNA pull-down assay, and RNA immunoprecipitation were used to evaluate the interaction between linc01559 and miR-6783-3p. RESULT: linc01559 expression was associated with response to FOLFOX4, as well as miR-1343-3p and miR- 6783-3p expression in vivo. A nomogram, including linc01559 and miR-1343-3p, precisely and accurately predicted the overall survival of patients with HCC. Regarding the in vitro tests, linc01559 showed higher expression in L-OHP-resistant cell lines, whereas miR-6783-3p was downregulated. Knockdown of linc01559 led to decreased proliferation and migration ability, and increased expression of miR-6783-3p; however, it did not influence the expression of miR-1343-3p. We also found that linc01559 directly interacted with miR-6783-3p. Furthermore, linc01559 and miR-6783-3p regulated the viability of L-OHP-resistant cells following treatment with L-OHP. CONCLUSION: linc01559 promoted the proliferation of HCC by sponging miR-6783-3p. This suggests that linc01559/miR-6783-3p may be key factors in regulating resistance and response to L-OHP. Moreover, they may be potential therapeutic targets for improving sensitivity to L-OHP in patients with HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Drug Resistance, Neoplasm , Liver Neoplasms/genetics , MicroRNAs/genetics , Oxaliplatin/pharmacology , RNA, Long Noncoding/genetics , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/drug therapy , Oncogenes/drug effects , Oxaliplatin/therapeutic use
17.
Br J Haematol ; 193(1): 160-170, 2021 04.
Article in English | MEDLINE | ID: mdl-32945549

ABSTRACT

Panobinostat is a pan-deacetylase inhibitor that modulates the expression of oncogenic and immune-mediating genes involved in tumour cell growth and survival. We evaluated panobinostat-induced post-transplant responses and identified correlative biomarkers in patients with multiple myeloma who had failed to achieve a complete response after autologous transplantation. Patients received panobinostat 45 mg administered three-times weekly (TIW) on alternate weeks of 28-day cycles commencing 8-12 weeks post-transplant. Twelve of 25 patients (48%) improved their depth of response after a median (range) of 4·3 (1·9-9·7) months of panobinostat. In responders, T-lymphocyte histone acetylation increased after both three cycles (P < 0·05) and six cycles (P < 0·01) of panobinostat when compared to baseline, with no differences in non-responders. The reduction in the proportion of CD127+ CD8+ T cells and CD4:CD8 ratio was significantly greater, after three and six cycles of panobinostat compared to pre-transplant, in non-responders when compared to responders. Whole marrow RNA-seq revealed widespread transcriptional changes only in responders with baseline differences in genes involved in ribosome biogenesis, oxidative phosphorylation and metabolic pathways. This study confirmed the efficacy of panobinostat as a single agent in multiple myeloma and established acetylation of lymphocyte histones, modulation of immune subsets and transcriptional changes as pharmacodynamic biomarkers of clinical benefit.


Subject(s)
Histone Deacetylase Inhibitors/therapeutic use , Multiple Myeloma/metabolism , Multiple Myeloma/therapy , Panobinostat/therapeutic use , Transplantation, Autologous/adverse effects , Adult , Aged , CD4 Antigens/drug effects , CD4 Antigens/immunology , CD8 Antigens/drug effects , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Female , Follow-Up Studies , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/adverse effects , Histones/drug effects , Histones/metabolism , Humans , Interleukin-7 Receptor alpha Subunit/drug effects , Interleukin-7 Receptor alpha Subunit/immunology , Male , Middle Aged , Multiple Myeloma/immunology , Multiple Myeloma/mortality , Neoplasm Staging/methods , Oncogenes/drug effects , Panobinostat/administration & dosage , Panobinostat/adverse effects , Remission Induction , Survival Analysis , Transplantation, Autologous/statistics & numerical data , Treatment Outcome
18.
Expert Rev Gastroenterol Hepatol ; 15(1): 65-79, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32946312

ABSTRACT

INTRODUCTION: In colorectal carcinogenesis, genetic alterations in RAS and BRAF oncogenes play an important role for cancer initiation and/or progression and represent a key focus in the search for targeted therapies. Despite many years of research and a great amount of studies, until very recently this pathway was considered extremely hard to downregulate to obtain a significant clinical impact in colorectal cancer patients. But better times are coming with the advent of new promising drugs and combinations strategies. AREAS COVERED: In this review, we go over the biological characteristics of the MAPK pathway in colorectal tumors, while illustrating the clinical correlation of RAS and BRAF mutations, particularly its prognostic and predictive value. We also present newly data about recent improvements in the treatment strategy for patients harboring these types of tumors. EXPERT COMMENTARY: With great advances in the knowledge of molecular basis of RAS and BRAF mutant colorectal cancer in conjunction with biotechnology development and the constant effort for improvement, in the near future many new therapeutic options would be available for the management of this group of patient with dismal prognosis.


Subject(s)
Colorectal Neoplasms/genetics , Genes, ras/genetics , Proto-Oncogene Proteins B-raf/genetics , Biomarkers, Tumor/genetics , Colorectal Neoplasms/drug therapy , Genes, ras/drug effects , Humans , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/genetics , Mutation , Oncogenes/drug effects , Oncogenes/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/drug effects
19.
Life Sci ; 265: 118797, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33285162

ABSTRACT

AIMS: Lung cancer was the most fatal malignancy, dominated the cancer related mortality list for years, and we tried to distinguish the lung adenocarcinoma patients at higher risk from those bearing lower therapy resistance and recurrence risk. MATERIALS: Patients information from clinical Sequencing Cohorts and from the Regional Medical Center of the Middle-West China were included. The whole-exome sequencing was analyzed for risk evaluation. KEY FINDINGS: We found that Smoking stimulated the oncogenic genes mutations, and the most frequently mutated genes of EGFR, KRAS, and TP53 (E/K/P) were identified. Different N stage affected the survival prognosis of patients bearing E/K/P mutations, but the T stage and AJCC stage did not. Radiation failed to prolong survival of phase II/III patients who didn't receive surgery. In those received surgery, radiation also failed to prolong survival of phase II/III patients. Radiation did not improve the prognosis in patients bearing E/K/P mutation burdens, whose differences were identified in gender or smoking-history classified groups. SIGNIFICANCE: Smoking status and history contributed to oncogenic mutation burdens associated therapy resistance, and the aggressive treatment, especially to radiation, may lead to worse therapy response to current and past smoking behavior.


Subject(s)
Adenocarcinoma of Lung/genetics , Carcinogenesis/genetics , Lung Neoplasms/genetics , Mutation/genetics , Oncogenes/genetics , Smoking/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/therapy , Aged , Biomarkers, Tumor/genetics , Carcinogenesis/drug effects , Carcinogenesis/pathology , Female , Humans , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Middle Aged , Mutation/drug effects , Oncogenes/drug effects , Smoking/pathology , Smoking/therapy , Survival Rate/trends , Treatment Outcome
20.
Eur J Med Chem ; 213: 113082, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33309163

ABSTRACT

KRAS is the most commonly altered oncogene of the RAS family, especially the G12C mutant (KRASG12C), which has been a promising drug target for many cancers. On the basis of the bicyclic pyridopyrimidinone framework of the first-in-class clinical KRASG12C inhibitor AMG510, a scaffold hopping strategy was conducted including a F-OH cyclization approach and a pyridinyl N-atom working approach leading to new tetracyclic and bicyclic analogues. Compound 26a was identified possessing binding potency of 1.87 µM against KRASG12C and cell growth inhibition of 0.79 µM in MIA PaCa-2 pancreatic cancer cells. Treatment of 26a with NCI-H358 cells resulted in down-regulation of KRAS-GTP levels and reduction of phosphorylation of downstream ERK and AKT dose-dependently. Molecular docking suggested that the fluorophenol moiety of 26a occupies a hydrophobic pocket region thus forming hydrogen bonding to Arg68. These results will be useful to guide further structural modification.


Subject(s)
Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Pancreatic Neoplasms/drug therapy , Piperazines/chemical synthesis , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Pyridines/chemical synthesis , Pyrimidines/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclization , Drug Screening Assays, Antitumor , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Molecular Docking Simulation , Oncogenes/drug effects , Phosphorylation/drug effects , Piperazines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Structure-Activity Relationship , Pancreatic Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL
...