Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Pharm Sin B ; 14(1): 319-334, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38261830

ABSTRACT

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) functions as a key regulator in inflammation and cell death and is involved in mediating a variety of inflammatory or degenerative diseases. A number of allosteric RIPK1 inhibitors (RIPK1i) have been developed, and some of them have already advanced into clinical evaluation. Recently, selective RIPK1i that interact with both the allosteric pocket and the ATP-binding site of RIPK1 have started to emerge. Here, we report the rational development of a new series of type-II RIPK1i based on the rediscovery of a reported but mechanistically atypical RIPK3i. We also describe the structure-guided lead optimization of a potent, selective, and orally bioavailable RIPK1i, 62, which exhibits extraordinary efficacies in mouse models of acute or chronic inflammatory diseases. Collectively, 62 provides a useful tool for evaluating RIPK1 in animal disease models and a promising lead for further drug development.

2.
J Med Chem ; 66(4): 2608-2621, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36723914

ABSTRACT

There is growing interest in covalent targeted inhibitors in drug discovery against previously "undruggable" sites and targets. These molecules typically feature an electrophilic warhead that reacts with nucleophilic groups of protein residues, most notably the thiol group of cysteines. One main challenge in the field is to develop versatile utilizable warheads. Here, we characterize the unique features of novel arsenous warheads for reaction with thiol species in a reversible manner and further demonstrate that organoarsenic probes can be chemically tuned toward specific molecular targets by developing selective and potent inhibitors of pyruvate kinase M2 (PKM2). We show that compound 24 is a covalent and allosteric inhibitor of PKM2 and its orally bioavailable prodrug 25 exerts efficacious inhibition of PKM2-dependent tumor growth in vitro and in vivo. Our results introduce 25 and its derivatives as useful pharmacological tools and provide a general road map for targeting the protein cysteinome using arsenous warheads.


Subject(s)
Drug Discovery , Pyruvate Kinase , Cysteine/chemistry
3.
Int J Anthropol Ethnol ; 6(1): 18, 2022.
Article in English | MEDLINE | ID: mdl-36532330

ABSTRACT

The rise of neo-nationalism has been an important political phenomenon since the 21st century. Neo-nationalism is not a single form of nationalism. It is not only a generalization of a specific type of nationalism at present, but also a description of a series of new nationalism phenomena. From the point of what it may include, it has at least four forms: far-right nationalism, evangelical nationalism, separatist nationalism, and (the third world) religious nationalism. Compared with traditional nationalism, neo-nationalism has undergone major changes in terms of guiding ideology or values behind, epochal character, propulsion mechanism, function, propagation mode, influence, and field of occurrence. At the same time, neo-nationalism is also a kind of high-intensity identity politics with a sort of quasi-fundamentalist characteristics. It discards the core value principles of traditional nationalism and the basic etiquette of polite society. Some dangerous and even crazy essential factors of nationalism have been developed to extremes in the new era. This means that its destructiveness to specific countries and societies is far greater than that caused by the reactive identity politics of minorities. From the perspective of trends in development, while the neo-nationalism shows the general characteristics of co-advance and retreat in the general trend, its four specific forms also show certain differences in the development direction. With the further setback of the globalization process, neo-nationalism will have a significant impact on the security of relevant countries, regions and even the world.

4.
Cancer Res ; 82(19): 3588-3602, 2022 Oct 04.
Article in English | MEDLINE | ID: mdl-35857807

ABSTRACT

Therapeutic perturbation of cyclin-dependent kinase 12 (CDK12) is proposed to have pleiotropic effects in ovarian cancer, including direct cytotoxicity against tumor cells and indirect induction of immunogenicity that confer synthetic sensitivity to immune-based treatment. However, formal testing of this hypothesis has been hindered by an insufficient mechanistic understanding of CDK12 and its close homolog CDK13, as well as generally unfavorable pharmacokinetics of available CDK12/CDK13 covalent inhibitors. In this study, we used an innovative arsenous warhead modality to develop an orally bioavailable CDK12/CDK13 covalent compound. The dual CDK12/CDK13 inhibitors ZSQ836 exerted potent anticancer activity in cell culture and mouse models and induced transcriptional reprogramming, including downregulation of DNA damage response genes. CDK12 and CDK13 were both ubiquitously expressed in primary and metastatic ovarian cancer, and the two kinases performed independent and synergistic functions to promote tumorigenicity. Unexpectedly, although ZSQ836 triggered genomic instability in malignant cells, it counterintuitively impaired lymphocytic infiltration in neoplastic lesions by interfering with T-cell proliferation and activation. These findings highlight the Janus-faced effects of dual CDK12/CDK13 inhibitors by simultaneously suppressing tumor and immune cells, offering valuable insights into the future direction of drug discovery to pharmacologically target CDK12. SIGNIFICANCE: This study dissects the specific roles of CDK12 and CDK13 in ovarian cancer and develops a CDK12/CDK13 inhibitor that impairs both tumor and immune cells, which could guide future CDK12 inhibitor development.


Subject(s)
CDC2 Protein Kinase , Ovarian Neoplasms , Animals , Carcinoma, Ovarian Epithelial/genetics , Cyclin-Dependent Kinases/genetics , Female , Genes, cdc , Humans , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics
5.
Oncogene ; 41(12): 1767-1779, 2022 03.
Article in English | MEDLINE | ID: mdl-35124696

ABSTRACT

Müllerian tissue-specific oncogenes, prototyped by PAX8, underlie ovarian tumorigenesis and represent unique molecular vulnerabilities. Further delineating such lineage-dependency factors and associated therapeutic implications would provide valuable insights into ovarian cancer biology and treatment. In this study, we identified SOX17 as a new lineage-survival master transcription factor, which shared co-expression pattern with PAX8 in epithelial ovarian carcinoma. Genetic disruption of SOX17 or PAX8 analogously inhibited neoplastic cell viability and downregulated a spectrum of lineage-related transcripts. Mechanistically, we showed that SOX17 physically interacted with PAX8 in cultured cell lines and clinical tumor specimens. The two nuclear proteins bound to overlapping genomic regions and regulated a common set of downstream genes, including those involved in cell cycle and tissue morphogenesis. In addition, we revealed that small-molecule inhibitors of transcriptional cyclin-dependent kinases (CDKs) effectively reduced SOX17 and PAX8 expression. ZSQ1722, a novel orally bioavailable CDK12/13 covalent antagonist, exerted potent anti-tumor activity in xenograft models. These findings shed light on an actionable lineage-survival transcriptional complex in ovarian cancer, and facilitated drug discovery by generating a serial of candidate compounds to pharmacologically target this difficult-to-treat malignancy.


Subject(s)
Ovarian Neoplasms , PAX8 Transcription Factor , SOXF Transcription Factors , Cell Cycle , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
6.
Cancer Immunol Immunother ; 67(12): 1939-1947, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29943070

ABSTRACT

Prostate cancer is one of the leading causes of cancer deaths, with no curative treatments once it spreads. Alternative therapies, including immunotherapy, have shown limited efficacy. Dendritic cells (DC) have been widely used in the treatment of various malignancies. DC capture antigens and move to the lymphoid organs where they prime naive T cells. Interaction between DC and T cells are most active in lymph nodes and suppression of DC trafficking to lymph nodes impairs the immune response. In this work, we aimed to study trafficking of DC in vivo via various routes of delivery, to optimize the effectiveness of DC-based therapy. A DC labeling system was developed using 1,1'-dioctadecyltetramethyl indotricarbocyanine Iodine for in vivo fluorescent imaging. DC harvested from C57B/6 mice were matured, labeled, and injected intravenously, subcutaneously, or intratumorally, with or without antigen loading with whole tumor lysate, into C57B/6 mice inoculated with RM-1 murine prostate tumor cells. Signal intensity was measured in vivo and ex vivo. Signal intensity at the tumor site increased over time, suggesting trafficking of DC to the tumor with all modes of injection. Subcutaneous injection showed preferential trafficking to lymph nodes and tumor. Intravenous injection showed trafficking to lungs, intestines, and spleen. Subcutaneous injection of DC pulsed with whole tumor lysate resulted in the highest increase in signal intensity at the tumor site and lymph nodes, suggesting subcutaneous injection of primed DC leads to highest preferential trafficking of DC to the immunocompetent organs.


Subject(s)
Dendritic Cells/immunology , Neoplasms/immunology , Neoplasms/metabolism , Animals , Biomarkers , Cell Line, Tumor , Cell Movement/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Disease Models, Animal , Heterografts , Humans , Immunity , Immunomodulation , Male , Mice , Neoplasms/diagnostic imaging , Neoplasms/pathology , Optical Imaging/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Sci Rep ; 7(1): 80, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28250439

ABSTRACT

Breast cancer-related lymphoedema (BCRL) is a common and intractable complication. To evaluate the possible complications of using lymphatic transverse rectus abdominis myocutaneous/deep inferior epigastric perforator (TRAM/DIEP) flaps for breast reconstruction and BCRL treatment, 20 patients with moderate or severe BCRL were retrospectively enrolled between November 2012 and October 2014. 10 patients had undergone lymphatic TRAM/DIEP flap surgery were assigned to the surgery group. 10 patients unwilling to undergo reconstruction were assigned to the physiotherapy group treated with traditional physical therapy. Upper-limb movement and circumference were measured and patients' subjective assessment was assessed using a questionnaire. In the surgery group, all flaps were successfully transferred. BCRL in 8 patients was improved by one level. The upper-limb circumference returned to normal in 1 case, and only 1 patient did not improve. In the physiotherapy group, a slight improvement was noted in 6 patients and unchanged in four cases. From the questionnaires, patients underwent lymphatic TRAM/DIEP flap surgery reported a significantly greater improvement in the affected limb (p < 0.05). In the physiotherapy group, the limb subjective did not improve as well as in the surgery group. Lymphatic TRAM/DIEP is a safe and effective option for patients who suffer from post-mastectomy lymphoedema.


Subject(s)
Breast Cancer Lymphedema/therapy , Epigastric Arteries/surgery , Mammaplasty , Rectus Abdominis/transplantation , Upper Extremity/pathology , Breast Cancer Lymphedema/physiopathology , Breast Cancer Lymphedema/rehabilitation , Breast Cancer Lymphedema/surgery , Female , Free Tissue Flaps/transplantation , Humans , Myocutaneous Flap/transplantation , Perforator Flap/transplantation , Physical Therapy Modalities , Retrospective Studies , Surveys and Questionnaires , Treatment Outcome
8.
Prostate ; 77(4): 361-373, 2017 03.
Article in English | MEDLINE | ID: mdl-27862100

ABSTRACT

INTRODUCTION: Decreased expression of highly immunogenic cancer-testis antigens (CTA) might help tumor to achieve low immunogenicity, escape immune surveillance and grow unimpeded. Our aim was to evaluate CTA expression in tumor and normal tissues and to investigate possible means of improving the immune response in a murine prostate cancer (CaP) model by using the combination of epigenetic modifier 5-azacitidine (5-AzaC) and immunomodulator lenalidomide. No study to date has examined the effect of this combination on the prostate cancer or its impact on antigen-presenting cells (APC). MATERIALS AND METHODS: Gene microarrays were performed to compare expression of several CTA in murine prostate cancer (RM-1 cells) and normal prostate. RM-1 cells were treated with 5-AzaC and real-time PCR was performed to investigate the expression of several CTA. Western blotting was used to determine whether expression of CTA-specific mRNA induced by 5-AzaC resulted in increase in the corresponding protein. Effect of the epigenetic agents and immunomodulators was assessed on dendritic cells (DC) using flow cytometry, ELISA and T-cell proliferation assay. RESULTS: Gene arrays demonstrated decreased expression of 35 CTA in CaP tissue compared to normal prostate. 5-AzaC treatment of RM-1 prostate cancer cells upregulated the expression of all 13 CTA tested in a dose-dependent fashion. DC were treated with 5-AzaC and lenalidomide and the expression of surface markers MHC Class I, MHC Class II, CD80, CD86, CD 205, and CD40 was increased. Combination of 5-AzaC and lenalidomide enhances the ability of DC to stimulate T-cell proliferation in mixed leukocyte reaction. Secretion of IL-12 and IL-15 by DC increased significantly with addition of 5-AzaC or 5-AzaC and lenalidomide. CONCLUSIONS: Decreased expression of CTA by prostate cancer may be a means of escaping immune monitoring. Combination of epigenetic modifications and immunomodulation by 5-AzaC and lenalidomide increased tumor immunogenicity and enhanced DC function and may be used in the treatment of advanced prostate cancer. Prostate 77: 361-373, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Epigenesis, Genetic/physiology , Immunomodulation/physiology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Animals , Antigens, Neoplasm/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Epigenesis, Genetic/drug effects , Immunologic Factors/pharmacology , Immunomodulation/drug effects , Lenalidomide , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Prostatic Neoplasms/metabolism , Thalidomide/analogs & derivatives , Thalidomide/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...