Network


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

Hotspot


Dive into the research topics where Kishan Patel is active.

Publication


Featured researches published by Kishan Patel.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Activin A promotes multiple myeloma-induced osteolysis and is a promising target for myeloma bone disease

Sonia Vallet; Siddhartha Mukherjee; Nileshwari Vaghela; Teru Hideshima; Mariateresa Fulciniti; Samantha Pozzi; Loredana Santo; Diana Cirstea; Kishan Patel; Aliyah R. Sohani; Alexander R. Guimaraes; Wanling Xie; Dharminder Chauhan; Jesse Schoonmaker; Eyal C. Attar; Michael Churchill; Edie Weller; Nikhil C. Munshi; Jasbir Seehra; Ralph Weissleder; Kenneth C. Anderson; David T. Scadden; Noopur Raje

Understanding the pathogenesis of cancer-related bone disease is crucial to the discovery of new therapies. Here we identify activin A, a TGF-β family member, as a therapeutically amenable target exploited by multiple myeloma (MM) to alter its microenvironmental niche favoring osteolysis. Increased bone marrow plasma activin A levels were found in MM patients with osteolytic disease. MM cell engagement of marrow stromal cells enhanced activin A secretion via adhesion-mediated JNK activation. Activin A, in turn, inhibited osteoblast differentiation via SMAD2-dependent distal-less homeobox–5 down-regulation. Targeting activin A by a soluble decoy receptor reversed osteoblast inhibition, ameliorated MM bone disease, and inhibited tumor growth in an in vivo humanized MM model, setting the stage for testing in human clinical trials.


Molecular Cancer Therapeutics | 2010

Dual inhibition of akt/mammalian target of rapamycin pathway by nanoparticle albumin-bound-rapamycin and perifosine induces antitumor activity in multiple myeloma.

Diana Cirstea; Teru Hideshima; Scott J. Rodig; Loredana Santo; Samantha Pozzi; Sonia Vallet; Hiroshi Ikeda; Giulia Perrone; Gullu Gorgun; Kishan Patel; Neil Desai; Peter Sportelli; Shweta Kapoor; Shireen Vali; Siddhartha Mukherjee; Nikhil C. Munshi; Kenneth C. Anderson; Noopur Raje

The phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway mediates multiple myeloma (MM) cell proliferation, survival, and development of drug resistance, underscoring the role of mTOR inhibitors, such as rapamycin, with potential anti-MM activity. However, recent data show a positive feedback loop from mTOR/S6K1 to Akt, whereby Akt activation confers resistance to mTOR inhibitors. We confirmed that suppression of mTOR signaling in MM cells by rapamycin was associated with upregulation of Akt phosphorylation. We hypothesized that inhibiting this positive feedback by a potent Akt inhibitor perifosine would augment rapamycin-induced cytotoxicity in MM cells. Perifosine inhibited rapamycin-induced phosphorylated Akt, resulting in enhanced cytotoxicity in MM.1S cells even in the presence of interleukin-6, insulin-like growth factor-I, or bone marrow stromal cells. Moreover, rapamycin-induced autophagy in MM.1S MM cells, as evidenced by electron microscopy and immunocytochemistry, was augmented by perifosine. Combination therapy increased apoptosis detected by Annexin V/propidium iodide analysis and caspase/poly(ADP-ribose) polymerase cleavage. Importantly, in vivo antitumor activity and prolongation of survival in a MM mouse xenograft model after treatment was enhanced with combination of nanoparticle albumin-bound–rapamycin and perifosine. Utilizing the in silico predictive analysis, we confirmed our experimental findings of this drug combination on PI3K, Akt, mTOR kinases, and the caspases. Our data suggest that mutual suppression of the PI3K/Akt/mTOR pathway by rapamycin and perifosine combination induces synergistic MM cell cytotoxicity, providing the rationale for clinical trials in patients with relapsed/refractory MM. Mol Cancer Ther; 9(4); 963–75. ©2010 AACR.


Leukemia | 2011

A Novel Role for CCL3 (MIP-1α) in Myeloma-induced Bone Disease via Osteocalcin Downregulation and Inhibition of Osteoblast Function

Sonia Vallet; Samantha Pozzi; Kishan Patel; Nileshwari Vaghela; Mariateresa Fulciniti; Petter Veiby; Teru Hideshima; Loredana Santo; Diana Cirstea; David T. Scadden; Kenneth C. Anderson; Noopur Raje

Upregulation of cytokines and chemokines is a frequent finding in multiple myeloma (MM). CCL3 (also known as MIP-1α) is a pro-inflammatory chemokine, levels of which in the MM microenvironment correlate with osteolytic lesions and tumor burden. CCL3 and its receptors, CCR1 and CCR5, contribute to the development of bone disease in MM by supporting tumor growth and regulating osteoclast (OC) differentiation. In this study, we identify inhibition of osteoblast (OB) function as an additional pathogenic mechanism in CCL3-induced bone disease. MM-derived and exogenous CCL3 represses mineralization and osteocalcin production by primary human bone marrow stromal cells and HS27A cells. Our results suggest that CCL3 effects on OBs are mediated by ERK activation and subsequent downregulation of the osteogenic transcription factor osterix. CCR1 inhibition reduced ERK phosphorylation and restored both osterix and osteocalcin expression in the presence of CCL3. Finally, treating SCID-hu mice with a small molecule CCR1 inhibitor suggests an upregulation of osteocalcin expression along with OC downregulation. Our results show that CCL3, in addition to its known catabolic activity, reduces bone formation by inhibiting OB function, and therefore contributes to OB/OC uncoupling in MM.


Oncogene | 2010

AT7519, A novel small molecule multi-cyclin-dependent kinase inhibitor, induces apoptosis in multiple myeloma via GSK-3β activation and RNA polymerase II inhibition

Loredana Santo; Sonia Vallet; Teru Hideshima; Diana Cirstea; Hiroshi Ikeda; Samantha Pozzi; Kishan Patel; Yutaka Okawa; Gullu Gorgun; Giulia Perrone; Elisabetta Calabrese; Murray Yule; Matt Squires; Marco Ladetto; Mario Boccadoro; Paul G. Richardson; Nikhil C. Munshi; Kenneth C. Anderson; Noopur Raje

Dysregulated cell cycling is a universal hallmark of cancer and is often mediated by abnormal activation of cyclin-dependent kinases (CDKs) and their cyclin partners. Overexpression of individual complexes are reported in multiple myeloma (MM), making them attractive therapeutic targets. In this study, we investigate the preclinical activity of a novel small-molecule multi-CDK inhibitor, AT7519, in MM. We show the anti-MM activity of AT7519 displaying potent cytotoxicity and apoptosis; associated with in vivo tumor growth inhibition and prolonged survival. At the molecular level, AT7519 inhibited RNA polymerase II (RNA pol II) phosphorylation, a CDK9, 7 substrate, associated with decreased RNA synthesis confirmed by [3H] Uridine incorporation. In addition, AT7519 inhibited glycogen synthase kinase 3β (GSK-3β) phosphorylation; conversely pretreatment with a selective GSK-3 inhibitor and shRNA GSK-3β knockdown restored MM survival, suggesting the involvement of GSK-3β in AT7519-induced apoptosis. GSK-3β activation was independent of RNA pol II dephosphorylation confirmed by α-amanitin, a specific RNA pol II inihibitor, showing potent inhibition of RNA pol II phosphorylation without corresponding effects on GSK-3β phosphorylation. These results offer new insights into the crucial, yet controversial role of GSK-3β in MM and show significant anti-MM activity of AT7519, providing the rationale for its clinical evaluation in MM.


Bone | 2013

In vivo and in vitro effects of a novel anti-Dkk1 neutralizing antibody in multiple myeloma

Samantha Pozzi; Mariateresa Fulciniti; Hua Yan; Sonia Vallet; Homare Eda; Kishan Patel; Loredana Santo; Diana Cirstea; Teru Hideshima; Linda Schirtzinge; Stuart Kuhstoss; Kenneth C. Anderson; Nikhil C. Munshi; David T. Scadden; Henry M. Kronenberg; Noopur Raje

Over-expression of the protein Dickkopf-1 (Dkk1) has been associated with multiple myeloma bone disease. Previous reports with the use of anti-Dkk1 neutralizing Ab directed strategies have demonstrated a pro-anabolic effect with associated anti-myeloma activity in 2 in vivo mouse models. However new insights on the role of the wnt pathway in osteoclasts (OC) are emerging and the potential effect of a neutralizing Ab to Dkk1 in osteoclastogenesis remains to be elucidated. In order to better define the effect of an anti-Dkk1 neutralizing Ab on osteoclastogenesis and myeloma, we studied a novel anti-Dkk1 monoclonal Ab in our preclinical models. In vivo data confirmed the pro-anabolic and anti-MM effect. In vitro data in part confirmed the in vivo observation, suggesting an indirect anti-MM effect secondary to inhibition of osteoclastogenesis and thus the interaction between MM and bone microenvironment. However, when studies on osteoclastogenesis were extended to samples derived from MM patients, we observed a variable response to anti-Dkk1 treatment without correlation to expression of surface receptors for Dkk1 in OCs suggesting potential heterogeneity in the efficacy of such a strategy. In conclusion, Dkk1 is a promising target for the treatment of both MM and bone disease, and ongoing clinical studies will help elucidate its efficacy.


Clinical Cancer Research | 2011

Antimyeloma Activity of a Multitargeted Kinase Inhibitor, AT9283, via Potent Aurora Kinase and STAT3 Inhibition Either Alone or in Combination with Lenalidomide

Loredana Santo; Teru Hideshima; Diana Cirstea; Madhavi Bandi; Erik A. Nelson; Gullu Gorgun; Scott J. Rodig; Sonia Vallet; Samantha Pozzi; Kishan Patel; Christine Unitt; Matthew Squires; Yiguo Hu; Dharminder Chauhan; Anuj Mahindra; Nikhil C. Munshi; Kenneth C. Anderson; Noopur Raje

Purpose: Aurora kinases, whose expression is linked to genetic instability and cellular proliferation, are being investigated as novel therapeutic targets in multiple myeloma (MM). In this study, we investigated the preclinical activity of a small-molecule multitargeted kinase inhibitor, AT9283, with potent activity against Aurora kinase A, Aurora kinase B, and Janus kinase 2/3. Experimental Design: We evaluated the in vitro antimyeloma activity of AT9283 alone and in combination with lenalidomide and the in vivo efficacy by using a xenograft mouse model of human MM. Results: Our data showed that AT9283 induced cell-growth inhibition and apoptosis in MM. Studying the apoptosis mechanism of AT9283 in MM, we observed features consistent with both Aurora kinase A and Aurora kinase B inhibition, such as increase of cells with polyploid DNA content, decrease in phospho-histone H3, and decrease in phospho-Aurora A. Importantly, AT9283 also inhibited STAT3 tyrosine phosphorylation in MM cells. Genetic depletion of STAT3, Aurora kinase A, or Aurora kinase B showed growth inhibition of MM cells, suggesting a role of AT9283-induced inhibition of these molecules in the underlying mechanism of MM cell death. In vivo studies showed decreased MM cell growth and prolonged survival in AT9283-treated mice compared with controls. Importantly, combination studies of AT9283 with lenalidomide showed significant synergistic cytotoxicity in MM cells, even in the presence of bone marrow stromal cells. Enhanced cytotoxicity was associated with increased inhibition of phosphorylated STAT3 and phosphorylated extracellular signal–regulated kinase. Conclusions: Demonstration of in vitro and in vivo anti-MM activity of AT9283 provides the rationale for the clinical evaluation of AT9283 as monotherapy and in combination therapy for treating patients with MM. Clin Cancer Res; 17(10); 3259–71. ©2011 AACR.


Leukemia | 2013

Lenalidomide in combination with an activin A-neutralizing antibody: preclinical rationale for a novel anti-myeloma strategy

Tyler A. Scullen; Loredana Santo; Sonia Vallet; Mariateresa Fulciniti; Homare Eda; Diana Cirstea; Kishan Patel; Neeharika Nemani; Andrew Yee; Anuj Mahindra; Noopur Raje

Given the prevalence of osteolytic bone disease in multiple myeloma (MM), novel therapies targeting bone microenvironment are essential. Previous studies have identified activin A to be of critical importance in MM-induced osteolysis. Lenalidomide is a known and approved treatment strategy for relapsed MM. Our findings demonstrate that lenalidomide acts directly on bone marrow stromal cells via an Akt-mediated increase in Jun N-terminal kinase-dependent signaling resulting in activin A secretion, with consequent inhibition of osteoblastogenesis. Here, we attempted to augment the antitumor benefits of lenalidomide while overcoming its effects on osteoblastogenesis by combining it with a neutralizing antibody to activin A. Increased activin A secretion induced by lenalidomide was abrogated by the addition of activin A-neutralizing antibody, which effectively restored osteoblast function and inhibited MM-induced osteolysis without negating the cytotoxic effects of lenalidomide on malignant cells. This provides the rationale for an ongoing clinical trial (NCT01562405) combining lenalidomide with an anti-activin A strategy.


Cancer Research | 2010

Abstract 2530: Antimyeloma activity of a small molecule multi-targeted kinase inhibitor, AT9283, via potent aurora kinase and STAT3 inhibition

Loredana Santo; Teru Hideshima; Diana Cirstea; Erik A. Nelson; Madhavi Bandi; Gullu Gorgun; Sonia Vallet; Samantha Pozzi; Kishan Patel; Hiroshi Ikeda; Giulia Perrone; Yiguo Hu; Dharminder Chauhan; Matthew Squires; Nikhil C. Munshi; Kenneth C. Anderson; Noopur Raje

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC Aurora Kinases are a family of mitotic regulators. Aurora Kinase A (AURKA) plays a crucial role in centrosome separation and spindle assembly and is required for mitosis and bipolar mitotic spindle formation. Aurora Kinase B (AURKB), a member of the chromosomal passenger complex, is required for chromosome segregation, spindle assembly checkpoint and cytokinesis. Both AURKA and AURKB are significantly overexpressed in MM cells, which has prompted the investigation of aurora kinase inhibitors as a therapeutic strategy in MM. Here, we investigated the preclinical activity of a small molecule multitargeted inhibitor, AT9283, with potent in vitro kinase activity against AURKA and AURKB kinases (3 nM), JAK2 and 3 (at 1.2 and 1.1 nM) and Abl T315I (at 4 nM). Growth inhibitory effects of AT9283 on MM cell lines and patient derived cells was observed with IC50 values of 0.25µM −0.5 µM at 48 hours using a [3H]thymidine incorporation assay. Cell cycle analysis following AT9283 treatment resulted in increased G2/M phase and polyploidy consistent with failed cytokinesis (associated with AURKB inhibition) confirmed by immunofluorescence assay. This was followed by induction of apoptosis assessed by Annexin V+PI+ staining peaking at 48 − 72 hours with associated −8-9 cleavage. Decreased levels of phosphorylated histone H3 at serine-10, a direct downstream substrate of AURKB, confirmed the role of AURKB inhibition by AT9283. Importantly, besides aurora kinase inhibition, we observed that AT9283 also inhibited STAT3 tyrosine phosphorylation in MM cells within 30 minutes of treatment. The effect of AT9283 on STAT3 inhibition was further investigated by using U3A cells stably expressing a luciferase reporter gene under the control of a STAT-dependent promoter. AT9283 inhibited STAT3-dependent luciferase activity with an EC50 of approximately 0.125 μM. Since MM cell lines with constitutive STAT3 tyrosine phosphorylation were more sensitive to AT9283, we investigated whether AT9283-induced effects on the JAK/STAT pathway correlated with Aurora inhibition. Genetic depletion by RNA interference showed that STAT3 knockdown in U266 cells did not affect the expression levels of AURKA and AURKB. In contrast, in cells with knocked-down AURK A and B, we observed a downregulation in the expression level of STAT3, due to either an off-target effect or the possibility that STAT3 is downstream of Aurora Kinases. Ongoing studies are aimed at understanding whether AT9283-induced effects on the JAK/STAT pathway enhance the efficacy of aurora kinase inhibition in the context of MM. Finally, in vivo data using a xenograft mouse model of human MM show that mice treated with AT9283 demonstrated slower tumor growth compared to the control group without adverse effects. In conclusion, these results show significant anti-MM activity of AT9283, and provide the rationale for its clinical evaluation in MM. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2530.


Blood | 2010

Selective Inhibition of HDAC6 with a New Prototype Inhibitor (ACY-1215) Overcomes Bortezomib Resistance In Multiple Myeloma (MM)

Loredana Santo; Teru Hideshima; Andrew L. Kung; Matthew Jarpe; Diana Cirstea; Kishan Patel; Samantha Pozzi; Jen C. Tseng; Scott J. Rodig; James E. Bradner; Kenneth C. Anderson; Simon Arnett Jones; Noopur Raje


Blood | 2010

Lenalidomide In Combination with the Activin Receptor Type II Murine Fc Protein RAP-011: Preclinical Rationale for a Novel Anti-Myeloma Strategy

Sonia Vallet; Kishan Patel; Diana Cirstea; Katie Luly; Samantha Pozzi; Loredana Santo; Homare Eda; Jasbir Seehra; Anuj Mahindra; David T. Scadden; Noopur Raje

Collaboration


Dive into the Kishan Patel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Samantha Pozzi

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge