Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Susanta Samajdar is active.

Publication


Featured researches published by Susanta Samajdar.


Cancer Research | 2016

Abstract 4649: ODM-207, a novel BET-bromodomain inhibitor as a therapeutic approach for the treatment of prostate and breast cancer

Mari Björkman; Elina Mattila; Reetta Riikonen; Chandra Sekhar; Mahaboobi Jaleel; Sivapriya Marappan; Tarja Ikonen; Daniel Nicorici Nicorici; Juha Rantala; Susanta Samajdar; Murali Ramachandra; Pekka Kallio; Anu-Maarit Moilanen

Introduction: BET (bromodomain and extraterminal) family proteins (BRD2, BRD3, BRD4, and BRDT) are epigenetic readers that bind to acetylated-lysine residues in histones and recruit protein complexes to promote transcription elongation. In many cancers, BET proteins have been shown to regulate expression of MYC and other oncogenic drivers that are important for cell proliferation and survival. Pharmacologic inhibition of the BET-histone interaction has been shown to result in transcriptional downregulation of a number of oncogenes and inhibition of tumor growth providing a novel strategy for treatment of cancer. Therefore, in this study, we evaluated the in vitro and in vivo antitumor activity of ODM-207, a novel, potent and highly selective BET bromodomain inhibitor using cell lines derived from prostate and breast cancer as well as patient-derived tumor cell cultures of breast cancer. Methods and Results: ODM-207 has antiproliferative effects on several hematological and solid tumor cell lines. In a panel of prostate cancer cell lines, ODM-207 attenuates cell growth of androgen receptor (AR)-positive cell lines such as VCaP and 22Rv1. RNA-sequencing and Western blot studies revealed that the exposure of sensitive prostate cancer cells to ODM-207 is associated with rapid down-regulation c-Myc expression levels while wtAR was not affected. In 22Rv1 prostate cancer xenograft, which expresses both the full-length androgen receptor and androgen receptor splice variant V7, oral administration of ODM-207 was very efficacious in suppressing tumor growth at well tolerated doses whereas enzalutamide had only a modest effect. Contrary to our findings in prostate cancer cells, BET-inhibitor treatment of estrogen-dependent MCF-7 breast cancer cell line inhibits tumor growth in cell proliferation assays but is associated with down-regulation of ERα while the c-Myc levels are very low, highlighting the context-dependent functional effects of BET inhibition. Interestingly, ODM-207 produces potent antiproliferative effects associated with cell-cycle arrest and cellular senescence in patient-derived breast cancer cells. Conclusions: In summary, ODM-207 is a new generation BET inhibitor found to possess excellent pharmacological properties and antitumor activity both in vitro and in vivo. Our data suggest the potential utilization of ODM-207 for the treatment of prostate and breast cancer. Citation Format: Mari Bjorkman, Elina Mattila, Reetta Riikonen, Chandra Sekhar, Mahaboobi Jaleel, Sivapriya Marappan, Tarja Ikonen, Daniel Nicorici Nicorici, Juha Rantala, Susanta Samajdar, Murali Ramachandra, Pekka Kallio, Anu-Maarit Moilanen. ODM-207, a novel BET-bromodomain inhibitor as a therapeutic approach for the treatment of prostate and breast cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4649.


Cancer Research | 2016

Abstract 3070: Potent and selective inhibition of CDK7 by novel covalent inhibitors

Leena K. Satyam; Ramulu Poddutoori; Subhendu Mukherjee; Sivapriya Marappan; Sreevalsam Gopinath; Raghuveer Ramachandra; Manoj Kumar Pothuganti; Shilpa Nayak; Nandish C; Chandranath Naik; Ravindra Mv; Madhu Dabbeeru; Nagaraju A; Mahankali B; Thomas Antony; Chetan Pandit; Shekar Chelur; Girish Daginakatte; Susanta Samajdar; Murali Ramachandra

Background: Phosphorylation of the RNA polymerase II (RNAPII) in C-terminal domain (CTD) by Cyclin-dependent kinase 7 (CDK7) is an important step in cellular transcription process. Hence pharmacological modulation of CDK7 kinase activity is considered as an interesting approach to treat cancers that critically dependent on transcription to maintain their oncogenic state. Experimental procedures: Multiple series of novel covalent CDK7 inhibitors were identified by SBDD approach based on the binding mode of known CDK7 inhibitors to find early hits. Iterative medicinal chemistry efforts were performed to identify several lead compounds by optimizing the initial hits to achieve good physicochemical properties, high potency, good selectivity and desirable pharmacokinetic profile. Summary: Highly potent ATP competitive covalent inhibitors of CDK7 from two distinct chemical series were identified. They show time-dependent inhibition of CDK7 enzyme activity as a proof of covalent binding and exhibit potent anti-proliferative activity in cell lines derived from various tumor types. CDK7 modulation by these compounds was also confirmed by monitoring cellular pS5RNAPII levels. Representative compounds from each series showed very good selectivity profile in broad kinase (332) panel. Lead molecules were identified based on excellent drug-like properties (solubility, permeability and good oral bioavailability). Tolerability and efficacy studies in rodent xenograft models are ongoing with selected leads to test their impact on tumor growth inhibition and to determine therapeutic window by oral administration. Conclusion: We have identified novel and selective CDK7 covalent inhibitors from two distinct chemical series with optimized drug-like properties including oral bioavailability. These compounds are being evaluated for anti-tumor activity in mouse xenograft models. Citation Format: Leena Khare Satyam, Ramulu Poddutoori, Subhendu Mukherjee, Sivapriya Marappan, Sreevalsam Gopinath, Raghuveer Ramachandra, Manoj Kumar Pothuganti, Shilpa S. Nayak, Nandish C, Chandranath Naik, Ravindra MV, Madhu B. Dabbeeru, Nagaraju A, Mahankali B, Thomas Antony, Chetan Pandit, Shekar Chelur, Girish Daginakatte, Susanta Samajdar, Murali Ramachandra. Potent and selective inhibition of CDK7 by novel covalent inhibitors. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3070.


Molecular Cancer Therapeutics | 2015

Abstract C190: Potent and selective inhibition of CDK7 by novel covalent inhibitors

Ramulu Poddutoori; Leena K. Satyam; Girish Daginakatte; Subhendu Mukherjee; Sivapriya Marappan; Sreevalsam Gopinath; Raghuveer Ramachandra; Anirudha Lakshminarasimhan; Manoj Kumar Pothuganti; Shilpa Nayak; Nandish C; Chandranath Naik; Ravindra Mv; Madhu Dabbeeru; Thomas Antony; Chetan Pandit; Murali Ramachandra; Shekar Chelur; Susanta Samajdar

Cyclin-dependent kinase 7 (CDK7) is an important constituent of the cellular transcriptional machinery, where it phosphorylates the C-terminal domain (CTD) of RNAP polymerase II (RNAPII). Because many tumor types are critically dependent on transcription for maintenance of their oncogenic state, pharmacological modulation of CDK7 kinase activity is considered as an approach to treat cancer. Multiple series of CDK7 inhibitors were identified by iterative medicinal chemistry efforts and SAR based approach. Early compounds were optimized towards attaining good physicochemical properties, high potency, good selectivity and desirable pharmacokinetic profile to achieve anti-tumor activity. We have identified compounds from two distinct chemical series that are highly potent in inhibiting CDK7 in biochemical assays. These inhibitors demonstrate time-dependent inhibition of CDK7 indicating covalent nature of binding. The compounds showed potent anti-proliferative activity in cell lines derived from various tumor types and this was accompanied by CDK7 modulation in cells as monitored by pS5RNAPII levels. They have excellent drug-like characteristics including solubility, permeability, metabolic stability and good oral bioavailability. In a broad panel of kinases (332 kinase), selected compounds from both series showed good selectivity profile. Tolerability and efficacy studies are ongoing with selected early leads to test their impact on tumor growth inhibition in xenograft models. We have identified novel and selective CDK7 covalent inhibitors from two series with desirable drug-like properties, which are being evauated for anti-tumor activity in xenograft models. Citation Format: Ramulu Poddutoori, Leena K. Satyam, Girish Daginakatte, Subhendu Mukherjee, Sivapriya Marappan, Sreevalsam Gopinath, Raghuveer Ramachandra, Anirudha Lakshminarasimhan, Manoj Pothuganti, Shilpa Nayak, Nandish C, Chandranath Naik, Ravindra MV, Madhu Dabbeeru, Thomas Antony, Chetan Pandit, Murali Ramachandra, Shekar Chelur, Susanta Samajdar. Potent and selective inhibition of CDK7 by novel covalent inhibitors. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr C190.


Cancer Research | 2017

Abstract LB-317: Identification of a novel preclinical candidate for CDK7 inhibition

Leena K. Satyam; Ramulu Poddutoori; Subhendu Mukherjee; Sivapriya Marappan; Sreevalsam Gopinath; Aravind Basavaraju; Lakshmi Narayana Kaza; Manoj Kumar Pothuganti; Shilpa Nayak; Nandish C; Amith A; Ravindra Mv; Dabbeeru Madhu Babu; Nagaraju A; Suraj Tgore; Thomas Antony; Chetan Pandit; Murali Ramachandra; Shekar Chelur; Girish Daginakatte; Susanta Samajdar

Cyclin-dependent kinase 7 (CDK7) is an important constituent of the cellular transcriptional machinery, where it phosphorylates the C-terminal domain (CTD) of RNAP polymerase II (RNAPII). Because many tumor types are critically dependent on transcription for maintenance of their oncogenic state, pharmacological modulation of CDK7 kinase activity is considered as an approach to treat cancer. Multiple series of covalent CDK7 inhibitors were identified by iterative medicinal chemistry efforts and SAR based approach. These compounds were optimized towards attaining good physicochemical properties, high potency, good selectivity and desirable pharmacokinetic profile to achieve anti-tumor activity. We have now identified a pre-clinical candidate AU-BGB-002 which is highly potent in inhibiting CDK7 in biochemical as well as cellular assays while fully efficiently engaging the target. In a panel of kinases, AU-BGB-002 shows selectivity for CDK7. A panel of cell lines derived from a diverse set of indications are sensitive to AU-BGB-002. AU-BGB-002 exhibits excellent drug-like characteristics including solubility, permeability, metabolic stability and good oral bioavailability. When tested in a xenograft model, AU-BGB-002 treatment resulted in dose dependent tumor growth inhibition in AML xenograft model with tumor stasis at a dose of 10 mg/kg. Potent inhibiton of tumor growth was accompanied by complete target engagement and suppression of pS5RNAPII RNAPolII Ser5 phosphorylation in a parallel PK-PD study. Efficacy studies in additional xenograft models, advanced DMPK and toxicity studies are ongoing for this compound. In summary, we have identified a novel and selective CDK7 covalent inhibitor candidate with desirable drug-like properties that shows excellent efficacy in an AML xenograft model. Findings presented here support further development of AU-BGB-002 for the treatment of cancer. Citation Format: Leena K. Satyam, Ramulu Poddutoori, Subhendu Mukherjee, Sivapriya Marappan, Sreevalsam Gopinath, Aravind Basavaraju, Lakshmi Narayana Kaza, Manoj Kumar Pothuganti, Shilpa Nayak, Nandish C, Amith A, Ravindra MV, Dabbeeru Madhu Babu, Nagaraju A, Suraj Tgore, Thomas Antony, Chetan Pandit, Murali Ramachandra, Shekar Chelur, Girish Daginakatte, Susanta Samajdar. Identification of a novel preclinical candidate for CDK7 inhibition [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr LB-317. doi:10.1158/1538-7445.AM2017-LB-317


Cancer Research | 2017

Abstract LB-113: Immune-mediated anti-tumor activity with a clinical stage BET bromodomain inhibitor ODM-207 in pre-clinical models

Pratima Deshpande; Ravi Krishna Babu; Prashant Yallappa Vadnal; Mahaboobi Jaleel; Murali Ramachandra; Chandrasekhar Abbineni; Susanta Samajdar; Anu-Maarit Moilanen; Pekka Kallio

Background: ODM-207 is a potent and selective BET inhibitor that is structurally unrelated to the benzodiazepine-based inhibitors including JQ1, I-BET762, and OTX015. Phase I clinical trials have now been initiated with this agent based on its potent anti-tumor activity in various in vitro and in vivo models of hematologic malignancies and solid tumors. In view of the recent publications implicating a role for BET protein BRD4 in the suppression of PD-L1 expression, an immune checkpoint ligand for PD-1, we sought to evaluate ODM-207 for its effect on immune-mediated anti-tumor efficacy in pre-clinical models. Methods and Results: Mouse splenocytes were stimulated with anti-CD3 and anti-CD28 in the presence or absence of ODM-207 for four days and changes in immune cell population were analyzed by FACS. Results revealed an increase in the level of activated cytotoxic CD8+ T cells as indicated by increased intracellular IFNγ and granzyme B with ODM-207 treatment. After confirming the lack of direct anti-proliferative activity on the mouse colon carcinoma cell line CT26, in vivo evaluation of ODM-207 was carried out in the syngeneic CT26 subcutaneous tumor model established in BALB/c mice. Daily oral administration of ODM-207 at 30 mg/kg was well tolerated in this model and resulted in a statistically significant inhibition of tumor growth. Interestingly, the tumor growth inhibition observed with ODM-207 was comparable to that with a commercially available anti-mouse PD1 antibody. Studies to characterize the immune changes in the tumor and anti-tumor activity of ODM-207 in combination with an anti-mouse PD1 antibody are currently underway and the results will be presented. Conclusions: In summary, these studies demonstrate the anti-tumor activity of BET inhibitor in a syngeneic model of colon carcinoma in the absence of a direct anti-proliferative activity on tumor cells. Observed tumor growth inhibition correlated with the in vitro activation of cytotoxic CD8+ T cells supporting the immune-mediated effect leading to tumor growth inhibition. In view of the remarkable success with the immune-based therapeutic approaches, these findings are relevant in devising appropriate strategies for the continued clinical development of ODM-207. Citation Format: Pratima Deshpande, Ravi Krishna Babu, Prashant Yallappa Vadnal, Mahaboobi Jaleel, Murali Ramachandra, Chandrasekhar Abbineni, Susanta Samajdar, Anu-Maarit Moilanen, Pekka Kallio. Immune-mediated anti-tumor activity with a clinical stage BET bromodomain inhibitor ODM-207 in pre-clinical models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr LB-113. doi:10.1158/1538-7445.AM2017-LB-113


Cancer Research | 2016

Abstract 4798: Efficacy and safety of highly selective novel IRAK4 inhibitors for treatment of ABC-DLBCL

Wesley Roy Balasubramanian; Venkateshwar Rao Gummadi; Kavitha Nellore; Subhendu Mukherjee; Sivapriya Marappan; Aravind Basavaraju; Bharathi Raja Ainan; Girish Daginakatte; Sreevalsam Gopinath; Sanjeev Giri; Thomas Antony; Shekar Chelur; Susanta Samajdar; Chetan Pandit; Murali Ramachandra

Interleukin-1 receptor associated kinases (IRAKs) are serine/threonine protein kinases belonging to the tyrosine-like kinase (TLK) family. IRAKs function as mediators of Toll-like receptor (TLR) and interleukin-1 receptor (IL-1R) signaling pathways and play an important role in innate immune signaling. TLR/IL-1R stimulation leads to recruitment of MYD88, an adaptor molecule, to the activated receptor complex, which then complexes with IRAK4 and activates IRAK1. TRAF6 is then activated by IRAK1 leading to NFkB activation. Recent studies have reported the occurrence of gain of function oncogenic mutation (L265P) in MYD88 in ∼30% of activated B cell diffuse large B-cell lymphoma(ABC DLBCL) and ∼90% of Waldenstrom9s macroglobulinemia (WM) leading to constitutive activation of IRAK4 and NFkB pathway. Among the DLBCL subtypes (GCB, ABC DLBCL and PMBL), ABC DLBCL is the most refractory. Inhibition of constitutive IRAK4 signalling can be used as a therapeutic strategy to treat ABC DLBCL Small molecule inhibitors of IRAK4 were synthesized based on hits originating from Aurigene9s compound library. Structure guided drug design approach was used to further improve the potency. Lead compounds demonstrated moderate to very high selectivity towardsIRAK4 (S35 score of 0.03) when screened against a large panel of 329 kinases. Aurigene9s lead compounds have excellent PK profile and good oral bioavailability in mice, leading to good in-vivo activity in TLR4 induced cytokine release model. Selected lead compounds were tested in a OCI-Ly3 xenograft model, which has a MYD88(L265P) mutation leading to constitutive activation of IRAK4 signaling. An advanced lead compound has demonstrated excellent efficacy in OCI-Ly3 model, with tumor stasis at low doses and tumor regression at higher doses. The compound is well tolerated and has a good therapeutic window as determined in a 14 day rodent toxicity study. In summary, a selective IRAK4 inhibitor has been identified with excellent efficacy and good safety profile. Citation Format: Wesley Roy Balasubramanian, Venkateshwar Rao Gummadi, Kavitha Nellore, Subhendu Mukherjee, Sivapriya Marappan, Aravind Basavaraju, Bharathi Raja Ainan, Girish Daginakatte, Sreevalsam Gopinath, Sanjeev Giri, Thomas Antony, Shekar Chelur, Susanta Samajdar, Chetan Pandit, Murali Ramachandra. Efficacy and safety of highly selective novel IRAK4 inhibitors for treatment of ABC-DLBCL. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4798.


Molecular Cancer Therapeutics | 2015

Abstract C191: Efficacy of novel IRAK4 inhibitors in ABC-DLBCL and AML models

Venkateshwar Rao; Wesley Roy Balasubramanian; Kavitha Nellore; Sivapriya Marappan; Aravind Basavaraju; Bharathi Raja Ainan; Girish Daginakatte; Sreevalsam Gopinath; Sanjeev Giri; Thomas Antony; Shekar Chelur; Susanta Samajdar; Chetan Pandit; Murali Ramachandra

Interleukin-1 receptor associated kinases (IRAKs) are serine/threonine protein kinases belonging to tyrosine-like kinase (TLK) family. The IRAK family consists of IRAK1, IRAK2, IRAK3 and IRAK4 out of which only IRAK1 and IRAK4 exhibit kinase activity. IRAKs function as mediators of Toll-like receptor (TLR) and interleukin-1 receptor (IL-1R) signaling pathways and play an important role in innate immune signaling. Recent studies have reported the occurrence of oncogenic mutations in MYD88 in 30% of activated B cell diffuse large B-cell lymphoma (ABC DLBCL) and 90% of Waldenstrom9s macroglobulinemia (WM) leading to constitutive activation of the IRAK4 and NFkB pathway. Recent studies have also highlighted the association of dysregulated innate immune signaling with Myelodysplastic syndrome (MDS) and Acute Myeloid leukaemia (AML). TLRs and their associated signal transducers are frequently overexpressed and/or constitutively activated in MDS. Overexpression and activation of IRAK1 is observed in AML. Thus IRAKs are attractive therapeutic targets for treatment of tumors with altered innate immune signaling such as ABC DLBCL and AML. We have designed, synthesized and tested small molecule inhibitors of IRAK4 based on hits originating from Aurigene9s compound library. We have identified a series of novel bicyclic heterocycles as potent inhibitors of IRAK-4 with moderate to very high selectivity (S35 score = 0.03) in a 329 kinase panel. Lead compounds were profiled in proliferation and mechanistic assays (p-IRAK1 and p-TAK1 inhibition) in appropriate ABC DLBCL/AML cell lines. Aurigene lead compounds demonstrate potent inhibition of cellular proliferation with a good correlation to inhibition of phosphorylation of signaling intermediates in mechanistic assays. Lead compounds exhibit excellent PK profile and good oral bioavailability in mice. Preliminary in-vitro toxicology studies indicate a clean safety profile. Selected compounds demonstrate excellent in-vivo efficacy in relevant tumor models with >90% tumor growth inhibition and good in-vivo PD modulation. In summary, a series of potent and selective IRAK4 inhibitors have been discovered and are being evaluated for treatment of cancers with dysregulated innate immune signaling. Citation Format: Venkateshwar Rao, Wesley Roy Balasubramanian, Kavitha Nellore, Sivapriya Marappan, Aravind Basavaraju, Bharathi Raja Ainan, Girish Daginakatte, Sreevalsam Gopinath, Sanjeev Giri, Thomas Antony, Shekar Chelur, Susanta Samajdar, Chetan Pandit, Murali Ramachandra. Efficacy of novel IRAK4 inhibitors in ABC-DLBCL and AML models. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr C191.


Archive | 2016

Indazole Compounds as IRAK4 Inhibitors

Venkateshwar Rao Gummadi; Susanta Samajdar; Ajay Gupta


Archive | 2015

Compounds and compositions as inhibitors of MEK

Mark G. Bock; Henrik Moebitz; Sunil Kumar Panigrahi; Ramulu Poddutoori; Susanta Samajdar


Archive | 2015

FUSED PYRIDINE AND PYRIMIDINE DERIVATIVES AS ROR GAMMA MODULATORS

Ravi Kotrabasaiah Ujjinamatada; Susanta Samajdar; Subramanya Hosahalli; Mallesham Bejugam

Collaboration


Dive into the Susanta Samajdar's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge