Synthesis and biological evaluation of orally active prodrugs (1a-d) of indomethacin are described. Prodrugs 1a-c showed a similar degree of anti-inflammatory activity, and prodrug 1d was found to be less potent than the parent drug indomethacin (1). Ulcer index (UI) data indicated that 1a (UI = 19), 1c (UI = 0), and 1d (UI = 0) were substantially less ulcerogenic and 1b (UI = 62) was more ulcerogenic than parent drug 1 (UI = 47). These prodrugs demonstrated good stability at acidic and basic pH and found to be more lipophilic than parent drug compound 1, indicated by partition coefficients measured in octanol-buffer system at pH 7.4 and 3.0. On the basis of in vivo studies, 1a and 1c compounds were selected for metabolic stability in rat liver microsome (RLM) and rat plasma (RP), and both were found to be enzymatically labile. Prodrugs 1a and 1c emerged as potent anti-inflammatory agents with a lesser potential for ulcer than the parent drug indomethacin.
Diclofenac ester pro drugs (4, 5, 6) were synthesized and evaluated in vitro and in vivo for their potential use for oral delivery, with the aim of obtaining enzymatically labile and less ulceration drugs than the parent drug diclofenac sodium (1a). Prodrugs 4, 5, 6 were found to be potent anti-inflammatory drugs with less ulcerogenic potential than the parent diclofenac sodium. Prodrugs 4, 5, 6 rapidly underwent enzymatic hydrolysis to release the parent drug diclofenac in 30-60 min in rat liver microsomes (RLM) and rat plasma (RP). Prodrugs were found to be more lipophilic when the partition coefficient was measured in 1-octanol and buffer system at pH 7.4 and 3.0. Diclofenac prodrugs 4, 5, 6 were found to be crystalline in nature (analyzed by PXRD). Prodrug 4 was found to be a superior candidate for the treatment of chronic inflammatory diseases.
Introduction: Lysine Specific Demethylase 1 (LSD1) is over-expressed in many cancers and down-regulation of LSD1 has been shown to effectively treat cancers by inducing re-expression of aberrantly silenced genes. Studies have shown that LSD1 may contribute to acute myelogenous leukaemia pathogenesis by inhibiting the normal pro-differentiative function of ATRA, paving the way for new combinatorial therapies for AML. In addition, LSD1 inhibition also leads to convertion of ‘cold' tumors to ‘hot' tumors by activating type 1 interferon response. Similarly, HDAC6 inhibition enhances immune response by over-coming immune suppression. Further, combined inhibition of LSD1 and HDAC has been shown to be more efficacious in inhibiting multiple cancers. Here, we show that JBI-802, a dual inhibitor targeting both LSD1 and HDAC6/8 shows stronger efficacy in several cancers, including AML, lymphoma and others and well tolerated at efficacious doses. Methods: Computational chemistry approaches were used to design LSD1 specific and LSD1-HDAC dual inhibitors. To assess in vitro LSD1 potency, TR-FRET assay was used. For assessing in vitro HDAC activity fluorescence based HDAC6 activity assay was performed. Western blotting and quantitative PCR were used to assess biomarkers of LSD1 and HDAC inhibition. Alamar blue cytotoxicity assay was used to assess cell proliferation. Xenograft and syngeneic models were used to assess in vivo efficacy. Results: Several compounds from this series show strong in vitro potency against LSD1 with excellent selectivity against MAOs. JBI-802 our lead dual molecule, showed an IC50 of 0.05 μM against LSD1 and isoform selective HDAC6/8 activity (IC50 of 0.011 and 0.098 µM on HDAC6 and HDAC8, respectively), with about 100 fold selectivity against other HDAC isoforms. When assessed in a panel of 25 cell lines, JBI-802 showed strong anti-proliferative activity on select AML, CLL, SCLC, sarcoma and multiple myeloma cell lines. In cell based and in vivo target engagement studies there was significant dose-dependent increase in CD11b, CD86 and GFI1b and tubulin acetylation levels. JBI-802 had a much stronger effect in inhibiting the growth of HEL92.1.7 erythroleukemia xenograft by oral administration when compared to ACY-1215, a HDAC6 selective inhibitor or ORY-1001 an LSD1 selective inhibitor. ED50 of JBI-802 in this model was ~6.25 mg/kg BID. Further, dose-dependent modulation of above mentioned biomarkers and inhibition of c-Myc could be observed in tumors. Stronger tumor growth inhibition was also observed in other haematological cancers models such as CMK-1 (leukemia) and Z-138 (lymphoma). In addition, JBI-802 showed comparable single agent activity to anti-PD-1/PD-L1 mAbs in a syngeneic murine colon cancer model CT26 and resulted in stronger tumor growth inhibition when combined with these antibodies. 14-day dose ranging finding toxicology studies clearly show that the molecule was well tolerated up to 300 mg/kg. Conclusion: The data obtained to date demonstrate that dual LSD1-HDAC6/8 inhibitors could serve as novel, effective therapeutic agents for treatment of select subset of AML and other cancers. IND-enabling studies are in progress with this inhibitor to be developed as a clinical candidate Citation Format: Dhanalakshmi Sivanandhan, Sridharan Rajagopal, Sreekala Nair, Basavaprabhu B, Reshma Dhkar, Santosh Viswakarma, Amir Siddiqui, Mohd Zainuddin, Rudresh G, Prashanthi Daram, Sunil Mohire, Krishnakumar V. JBI-802, novel dual inhibitor of LSD1-HDAC6 for treatment of cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 1756.
2597 Background: Programmed Cell Death 1 (PD-1) protein plays a key role in inhibiting immune responses and enhancing self-tolerance via modulation of T-cell activity, inducing T-cell apoptosis and inhibiting apoptosis of regulatory T cells. PD-L1 also plays an important role in various malignancies where it can attenuate the host immune response to tumor cells thereby favouring tumor progression and metastasis. High expression of PD-L1 in glioblastoma tumor tissues is associated with poor survival of patients, and PD-L1 may act as a prognostic predictor and an effective therapeutic target for glioblastoma. A number of monoclonal antibodies targeting PD-1/PD-L1 have approved for various malignancies. Still, efficacy of these antibodies in glioblastoma and brain metastasis continues to be moderate potentially owing to lack of or poor brain penetrance of these agents. Therefore, there is still a need for potent, selective small molecule PD-1/PD-L1 inhibitors with enhanced brain penetration in the treatment of such cancers. Methods: Rational design approaches were used to design novel small molecule PD-1/PD-L1 pathway inhibitors; potency of these inhibitors was assessed in an in-vitro TR-FRET assay. Checkpoints signalling reporter assays as well cell based PD-L1 dimerization assays were used to assess the mechanistic and functional effects. In vivo efficacy was assessed in orthotopic GBM as well as in syngeneic and humanized subcutaneous tumor models in mice. Results: Our lead PD-L1 inhibitor JBI-2174 showed strong in vitro IC50 of ̃1 nM in TR-FRET assay that measures interaction between hPD-1 and hPD-L1 and a picomolar IC50 against monkey PD-L1. In selectivity assays for immunooncology targets, JBI-2174 was highly selective for PD-L1. JBI-2174 also inhibited PD-L1/PD-1 mediated signalling essential for T-cell modulation. JBI-2174 induced dimerization of PD-L1 as observed by size exclusion chromatography, which was confirmed by co-crystal structure and recapitulated in cell based dimerization assay. Elucidation of the co-crystal structure, clearly demonstrated that JBI-2174 clearly interacts with multiple amino acids on PD-L1 that are critical for PD-1 binding. JBI-2174 showed excellent oral bioavailability across pre-clinical species and sustained brain exposure. In the in vivo efficacy studies, JBI-2174 showed comparable efficacy to the anti-PD-L1 antibody or Atezolizumab in syngeneic (4T1, CT-26) and in partially humanized models (MC-38/hPD-L1). Further, oral administration of JBI-2174 resulted in statistically significant increase in survival (Day 27 in control vs day 38 in treated, p < 0.05) in a mouse glioma orthotopic model. Conclusions: The oral bioavailability and brain exposure of this molecule will make it attractive for cancers with unmet medical needs such as GBM and brain metastasis. IND enabling studies are being initiated for this compound.
scite is a Brooklyn-based organization that helps researchers better discover and understand research articles through Smart Citations–citations that display the context of the citation and describe whether the article provides supporting or contrasting evidence. scite is used by students and researchers from around the world and is funded in part by the National Science Foundation and the National Institute on Drug Abuse of the National Institutes of Health.
customersupport@researchsolutions.com
10624 S. Eastern Ave., Ste. A-614
Henderson, NV 89052, USA
This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.
Copyright © 2024 scite LLC. All rights reserved.
Made with 💙 for researchers
Part of the Research Solutions Family.