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.
Molecules ; 28(19)2023 Oct 06.
Article in English | MEDLINE | ID: mdl-37836790

ABSTRACT

Src homology 2 domain-containing phosphatase 2 (SHP2) is an attractive target for cancer therapy due to its multifaceted roles in both tumor and immune cells. Herein, we designed and synthesized a novel series of proteolysis targeting chimeras (PROTACs) using a SHP2 allosteric inhibitor as warhead, with the goal of achieving SHP2 degradation both inside the cell and in vivo. Among these molecules, compound P9 induces efficient degradation of SHP2 (DC50 = 35.2 ± 1.5 nM) in a concentration- and time-dependent manner. Mechanistic investigation illustrates that the P9-mediated SHP2 degradation requires the recruitment of the E3 ligase and is ubiquitination- and proteasome-dependent. P9 shows improved anti-tumor activity in a number of cancer cell lines over its parent allosteric inhibitor. Importantly, administration of P9 leads to a nearly complete tumor regression in a xenograft mouse model, as a result of robust SHP2 depletion and suppression of phospho-ERK1/2 in the tumor. Hence, P9 represents the first SHP2 PROTAC molecule with excellent in vivo efficacy. It is anticipated that P9 could serve not only as a new chemical tool to interrogate SHP2 biology but also as a starting point for the development of novel therapeutics targeting SHP2.


Subject(s)
Neoplasms , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Humans , Animals , Mice , Neoplasms/drug therapy , Cell Line , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/metabolism , Proteolysis
2.
Front Immunol ; 14: 1203073, 2023.
Article in English | MEDLINE | ID: mdl-37671162

ABSTRACT

Cancer is one of the deadliest diseases, causing million of deaths each year globally. Conventional anti-cancer therapies are non-targeted and have systemic toxicities limiting their versatile applications in many cancers. So, there is an unmet need for more specific therapeutic options that will be effective as well as free from toxicities. Antibody-drug conjugates (ADCs) are suitable alternatives with the right potential and improved therapeutic index for cancer therapy. The ADCs are highly precise new class of biopharmaceutical products that covalently linked a monoclonal antibody (mAb) (binds explicitly to a tumor-associated surface antigen) with a customized cytotoxic drug (kills cancer cells) and tied via a chemical linker (releases the drug). Due to its precise design, it brings about the target cell killing sparing the normal counterpart and free from the toxicities of conventional chemotherapy. It has never been so easy to develop potential ADCs for successful therapeutic usage. With relentless efforts, it took almost a century for scientists to advance the formula and design ADCs for its current clinical applications. Until now, several ADCs have passed successfully through preclinical and clinical trials and because of proven efficacy, a few are approved by the FDA to treat various cancer types. Even though ADCs posed some shortcomings like adverse effects and resistance at various stages of development, with continuous efforts most of these limitations are addressed and overcome to improve their efficacy. In this review, the basics of ADCs, physical and chemical properties, the evolution of design, limitations, and future potentials are discussed.


Subject(s)
Biological Products , Drug-Related Side Effects and Adverse Reactions , Immunoconjugates , Neoplasms , Humans , Antibodies, Monoclonal
3.
J Clin Invest ; 2021 Jul 20.
Article in English | MEDLINE | ID: mdl-34283806

ABSTRACT

Both epidemiologic and cellular studies in the context of autoimmune diseases have established that protein tyrosine phosphatase non-receptor type 22 (PTPN22) is a key regulator of T cell receptor (TCR) signaling. However, its mechanism of action in tumors and its translatability as a target for cancer immunotherapy have not been established. Here we show that a germline variant of PTPN22, rs2476601, portended a lower likelihood of cancer in patients. PTPN22 expression was also associated with markers of immune regulation in multiple cancer types. In mice, lack of PTPN22 augmented antitumor activity with greater infiltration and activation of macrophages, natural killer (NK) cells, and T cells. Notably, we generated a novel small molecule inhibitor of PTPN22, named L-1, that phenocopied the antitumor effects seen in genotypic PTPN22 knockout. PTPN22 inhibition promoted activation of CD8+ T cells and macrophage subpopulations toward MHC-II expressing M1-like phenotypes, both of which were necessary for successful antitumor efficacy. Increased PD1-PDL1 axis in the setting of PTPN22 inhibition could be further leveraged with PD1 inhibition to augment antitumor effects. Similarly, cancer patients with the rs2476601 variant responded significantly better to checkpoint inhibitor immunotherapy. Our findings suggest that PTPN22 is a druggable systemic target for cancer immunotherapy.

4.
Life Sci ; 265: 118834, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33249096

ABSTRACT

Renal injury might originate from multiple factors like ischemia reperfusion (I/R), drug toxicity, cystic fibrosis, radio contrast agent etc. The four adenosine receptor subtypes have been identified and found to show diverse physiological and pathological roles in kidney diseases. The activation of A1 adenosine receptor (A1) protects against acute kidney injury by improving renal hemodynamic alterations, decreasing tubular necrosis and its inhibition might facilitate removal of toxin or drug metabolite in chronic kidney disease models. Furthermore, recent findings revealed that A2A receptor subtype activation regulates macrophage phenotype in experimental models of nephritis. Interestingly the emerging role of adenosine kinase inhibitors in kidney diseases has been discussed which act by increasing adenosine availability at target sites and thereby promote A2A receptor stimulation. In addition, the least explored adenosine receptor subtype A3 inhibition was observed to exert anti- oxidant, immunosuppressive and anti-fibrotic effects, but more studies are required to confirm its benefits in other renal injury models. The clinical studies targeting A1 receptor in patients with pre-existing kidney disease have yielded disappointing results, perhaps owing to the origin of unexpected neurological complications during the course of trial. Importantly, conducting well designed clinical trials and testing adenosine modulators with lesser brain penetrability could clear the way for clinical approval of these agents for patients with renal functional impairments.


Subject(s)
Adenosine/metabolism , Kidney Diseases/physiopathology , Receptors, Purinergic P1/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/physiopathology , Adenosine Kinase/antagonists & inhibitors , Adenosine Kinase/metabolism , Animals , Humans , Kidney Diseases/drug therapy , Reperfusion Injury/physiopathology
5.
Microorganisms ; 9(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374544

ABSTRACT

Tuberculosis (TB) is an airborne infectious disease caused by Mycobacterium tuberculosis (Mtb). According to the World Health Organization, an estimated 10 million people developed TB in 2018. The occurrence of drug-resistant TB demands therapeutic agents with novel mechanisms of action. Antivirulence is an alternative strategy that targets bacterial virulence factors instead of central growth pathways to treat disease. Mycobacterium protein tyrosine phosphatases, mPTPA and mPTPB, are secreted by Mtb into the cytoplasm of macrophages and are required for survival and growth of infection within the host. Here we present recent advances in understanding the roles of mPTPA and mPTPB in the pathogenesis of TB. We also focus on potent, selective, and well-characterized small molecule inhibitors reported in the last decade for mPTPA and mPTPB.

6.
J Med Chem ; 63(17): 9212-9227, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32787087

ABSTRACT

Tuberculosis is an infectious disease caused by the bacterium Mycobacterium tuberculosis (Mtb). Mtb protein tyrosine phosphatase B (mPTPB) is a virulence factor required for Mtb survival in host macrophages. Consequently, mPTPB represents an exciting target for tuberculosis treatment. Here, we identified N-phenyl oxamic acid as a highly potent and selective monoacid-based phosphotyrosine mimetic for mPTPB inhibition. SAR studies on the initial hit, compound 4 (IC50 = 257 nM), resulted in several highly potent inhibitors with IC50 values lower than 20 nM for mPTPB. Among them, compound 4t showed a Ki of 2.7 nM for mPTPB with over 4500-fold preference over 25 mammalian PTPs. Kinetic, molecular docking, and site-directed mutagenesis analyses confirmed these compounds as active site-directed reversible inhibitors of mPTPB. These inhibitors can reverse the altered host cell immune responses induced by the bacterial phosphatase. Furthermore, the inhibitors possess molecular weights <400 Da, log D7.4 < 2.5, topological polar surface area < 75, ligand efficiency > 0.43, and good aqueous solubility and metabolic stability, thus offering excellent starting points for further therapeutic development.


Subject(s)
Antitubercular Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Oxamic Acid/chemistry , Protein Tyrosine Phosphatases/antagonists & inhibitors , Animals , Antitubercular Agents/metabolism , Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Catalytic Domain , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Kinetics , Mice , Microsomes, Liver/metabolism , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mycobacterium tuberculosis/drug effects , Oxamic Acid/metabolism , Oxamic Acid/pharmacology , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , RAW 264.7 Cells , Structure-Activity Relationship
7.
Naunyn Schmiedebergs Arch Pharmacol ; 393(4): 615-627, 2020 04.
Article in English | MEDLINE | ID: mdl-31773183

ABSTRACT

Ischemic reperfusion (I/R) is the primary cause of acute kidney injury (AKI) in hospitalized patients. Although AKI resolution occurs in few days, it predisposes kidneys to progressive renal injury. Previously, administration of rennin-angiotensin-aldosterone system (RAAS) blocker spironolactone in acute phase was reported to attenuate various manifestations of chronic kidney disease (CKD) in rats. The present study investigates the effects of RAAS blockade during progressive kidney disease (30 days onwards) on CKD outcomes in rodent model of I/R injury. CKD was induced by clamping both renal pedicles for 45 min followed by 90 days of reperfusion in rats. Single and dual RAAS blocker therapy was initiated at 30 days post-I/R injury and continued until the end of the study period. Evaluation of proteinuria and creatinine levels was done every 30 days in various study groups. Assessment of CKD was done by analyzing renal tissue oxidative stress, inflammatory biomarker levels, and histological changes after 90 days of I/R injury. After 90 days, I/R rat kidneys displayed hypertrophy, reduced body weight, increased oxidative stress, elevated inflammatory biomarker levels, and histological abnormalities such as glomerulosclerosis, mesangial expansion, and tubulointerstitial fibrosis. Treatment with losartan or spironolactone alone significantly reduced various CKD-associated features. Remarkably, combined treatment with dual RAAS blocker in low dose or high dose exhibited highest beneficial effects on various parameters in CKD model, with low-dose combination showing fewer side effects. Therefore, we propose that combined low-dose RAAS blockade therapy might serve as a better therapeutic approach for retarding progressive kidney disease transition to CKD.


Subject(s)
Kidney Diseases/drug therapy , Losartan/therapeutic use , Renin-Angiotensin System/drug effects , Reperfusion Injury/drug therapy , Spironolactone/therapeutic use , Animals , Catalase/metabolism , Cytokines/metabolism , Glutathione/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Losartan/pharmacology , Male , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Spironolactone/pharmacology , Thiobarbituric Acid Reactive Substances/metabolism
8.
Eur J Pharmacol ; 865: 172711, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31589870

ABSTRACT

Acute kidney injury (AKI) is defined as a rapid loss of kidney function characterised by inflammation and cell death, ultimately leading to further functional and structural renal alterations. Based on experimental and epidemiological pieces of evidence, AKI may progress to chronic kidney disease (CKD) even after a recovery period due to maladaptive repair and other underlying mechanisms such as heightened Wnt signalling, overstimulation of the renin-angiotensin-aldosterone-system (RAAS) pathway, epigenetic alterations and inhibition of hypoxia-inducible factor (HIF) dependent defences. It has been reported that RAAS activation subsequent to renal insult mediates inflammatory and fibrotic mechanisms, which are a hallmark of CKD. Moreover, interesting evidence regarding the exposure-dependent dual role of Wnt signalling in both injury and repair, epigenetic changes underlying kidney disease suggest a potential therapeutic role of these pathways in AKI to CKD continuum. In addition, the hypoxia-independent renal benefits of erythropoietin such as anti-apoptosis and tubular regeneration also present an auspicious target which could be useful in clinical settings. In this review, the specific roles of these pathways in kidney disease, their pathological mechanisms and therapeutic strategies are discussed. Moreover, notable reports concerning stem cell therapy which hold promise in halting AKI-CKD continuum will be elaborated.


Subject(s)
Acute Kidney Injury/pathology , Disease Progression , Renal Insufficiency, Chronic/pathology , Acute Kidney Injury/genetics , Acute Kidney Injury/therapy , Epigenesis, Genetic , Humans , Signal Transduction , Stem Cell Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...