Sangeetha Palakurthi
Harvard University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Sangeetha Palakurthi.
Nature Communications | 2016
Shohei Koyama; Esra A. Akbay; Yvonne Y. Li; Grit S. Herter-Sprie; Kevin A. Buczkowski; William G. Richards; Leena Gandhi; Amanda J. Redig; Scott J. Rodig; Hajime Asahina; Robert E. Jones; Meghana M. Kulkarni; Mari Kuraguchi; Sangeetha Palakurthi; Peter E. Fecci; Bruce E. Johnson; Pasi A. Jänne; Jeffrey A. Engelman; Sidharta P. Gangadharan; Daniel B. Costa; Gordon J. Freeman; Raphael Bueno; F. Stephen Hodi; Glenn Dranoff; Kwok-Kin Wong; Peter S. Hammerman
Despite compelling antitumour activity of antibodies targeting the programmed death 1 (PD-1): programmed death ligand 1 (PD-L1) immune checkpoint in lung cancer, resistance to these therapies has increasingly been observed. In this study, to elucidate mechanisms of adaptive resistance, we analyse the tumour immune microenvironment in the context of anti-PD-1 therapy in two fully immunocompetent mouse models of lung adenocarcinoma. In tumours progressing following response to anti-PD-1 therapy, we observe upregulation of alternative immune checkpoints, notably T-cell immunoglobulin mucin-3 (TIM-3), in PD-1 antibody bound T cells and demonstrate a survival advantage with addition of a TIM-3 blocking antibody following failure of PD-1 blockade. Two patients who developed adaptive resistance to anti-PD-1 treatment also show a similar TIM-3 upregulation in blocking antibody-bound T cells at treatment failure. These data suggest that upregulation of TIM-3 and other immune checkpoints may be targetable biomarkers associated with adaptive resistance to PD-1 blockade.
Nature | 2016
Yong Jia; Cai-Hong Yun; Eunyoung Park; Dalia Ercan; Mari Manuia; Jose Juarez; Chunxiao Xu; Kevin Rhee; Ting Chen; Haikuo Zhang; Sangeetha Palakurthi; Jaebong Jang; Gerald Lelais; Michael DiDonato; Badry Bursulaya; Pierre-Yves Michellys; Robert Epple; Thomas H. Marsilje; Matthew McNeill; Wenshuo Lu; Jennifer L. Harris; Steven Bender; Kwok-Kin Wong; Pasi A. Jänne; Michael J. Eck
EGFR tyrosine kinase inhibitors (TKIs) gefitinib, erlotinib and afatinib are approved treatments for non-small cell lung cancers harboring activating mutations in the EGFR kinase1,2, but resistance arises rapidly, most frequently due to the secondary T790M mutation within the ATP-site of the receptor.3,4 Recently developed mutant-selective irreversible inhibitors are highly active against the T790M mutant5,6, but their efficacy can be compromised by acquired mutation of C797, the cysteine residue with which they form a key covalent bond7. All current EGFR TKIs target the ATP-site of the kinase, highlighting the need for therapeutic agents with alternate mechanisms of action. Here we describe rational discovery of EAI045, an allosteric inhibitor that targets selected drug-resistant EGFR mutants but spares the wild type receptor. A crystal structure shows that the compound binds an allosteric site created by the displacement of the regulatory C-helix in an inactive conformation of the kinase. The compound inhibits L858R/T790M-mutant EGFR with low-nanomolar potency in biochemical assays, but as a single agent is not effective in blocking EGFR-driven proliferation in cells due to differential potency on the two subunits of the dimeric receptor, which interact in an asymmetric manner in the active state8. We observe dramatic synergy of EAI045 with cetuximab, an antibody therapeutic that blocks EGFR dimerization9,10, rendering the kinase uniformly susceptible to the allosteric agent. EAI045 in combination with cetuximab is effective in mouse models of lung cancer driven by L858R/T790M EGFR and by L858R/T790M/C797S EGFR, a mutant that is resistant to all currently available EGFR TKIs. More generally, our findings illustrate the utility of purposefully targeting allosteric sites to obtain mutant-selective inhibitors.
Cancer Discovery | 2017
Russell W. Jenkins; Amir R. Aref; Patrick H. Lizotte; Elena Ivanova; Susanna Stinson; Chensheng W. Zhou; Michaela Bowden; Jiehui Deng; Hongye Liu; Diana Miao; Meng Xiao He; William F. Walker; Gao Zhang; Tian Tian; Chaoran Cheng; Zhi Wei; Sangeetha Palakurthi; Mark Bittinger; Hans Vitzthum; Jong Wook Kim; Ashley A. Merlino; Max M. Quinn; Chandrasekar Venkataramani; Joshua A. Kaplan; Andrew Portell; Prafulla C. Gokhale; Bart Phillips; Alicia Smart; Asaf Rotem; Robert E. Jones
Ex vivo systems that incorporate features of the tumor microenvironment and model the dynamic response to immune checkpoint blockade (ICB) may facilitate efforts in precision immuno-oncology and the development of effective combination therapies. Here, we demonstrate the ability to interrogate ex vivo response to ICB using murine- and patient-derived organotypic tumor spheroids (MDOTS/PDOTS). MDOTS/PDOTS isolated from mouse and human tumors retain autologous lymphoid and myeloid cell populations and respond to ICB in short-term three-dimensional microfluidic culture. Response and resistance to ICB was recapitulated using MDOTS derived from established immunocompetent mouse tumor models. MDOTS profiling demonstrated that TBK1/IKKε inhibition enhanced response to PD-1 blockade, which effectively predicted tumor response in vivo Systematic profiling of secreted cytokines in PDOTS captured key features associated with response and resistance to PD-1 blockade. Thus, MDOTS/PDOTS profiling represents a novel platform to evaluate ICB using established murine models as well as clinically relevant patient specimens.Significance: Resistance to PD-1 blockade remains a challenge for many patients, and biomarkers to guide treatment are lacking. Here, we demonstrate feasibility of ex vivo profiling of PD-1 blockade to interrogate the tumor immune microenvironment, develop therapeutic combinations, and facilitate precision immuno-oncology efforts. Cancer Discov; 8(2); 196-215. ©2017 AACR.See related commentary by Balko and Sosman, p. 143See related article by Deng et al., p. 216This article is highlighted in the In This Issue feature, p. 127.
Annals of Oncology | 2016
Ursula A. Matulonis; Gerburg Wulf; William T. Barry; Michael J. Birrer; S. N. Westin; Sarah Farooq; K. M. Bell-McGuinn; Elizabeth Obermayer; Christin Whalen; Tatum Spagnoletti; Weixiu Luo; H. Liu; R. C. Hok; Carol Aghajanian; David B. Solit; Gordon B. Mills; Barry S. Taylor; Helen H. Won; Michael F. Berger; Sangeetha Palakurthi; J. Liu; Lewis C. Cantley
Background Based upon preclinical synergy in murine models, we carried out a phase I trial to determine the maximum tolerated dose (MTD), toxicities, pharmacokinetics, and biomarkers of response for the combination of BKM120, a PI3K inhibitor, and olaparib, a PARP inhibitor. Patients and methods Olaparib was administered twice daily (tablet formulation) and BKM120 daily on a 28-day cycle, both orally. A 3u2009+u20093 dose-escalation design was employed with the primary objective of defining the combination MTD, and secondary objectives were to define toxicities, activity, and pharmacokinetic profiles. Eligibility included recurrent breast (BC) or ovarian cancer (OC); dose-expansion cohorts at the MTD were enrolled for each cancer. Results In total, 69 of 70 patients enrolled received study treatment; one patient never received study treatment because of ineligibility. Twenty-four patients had BC; 46 patients had OC. Thirty-five patients had a germline BRCA mutation (gBRCAm). Two DLTs (grade 3 transaminitis and hyperglycemia) were observed at DL0 (BKM120 60u2009mg/olaparib and 100u2009mg b.i.d.). The MTD was determined to be BKM120 50u2009mg q.d. and olaparib 300u2009mg b.i.d. (DL8). Additional DLTs included grade 3 depression and transaminitis, occurring early in cycle 2 (DL7). Anticancer activity was observed in BC and OC and in gBRCAm and gBRCA wild-type (gBRCAwt) patients. Conclusions BKM120 and olaparib can be co-administered, but the combination requires attenuation of the BKM120 dose. Clinical benefit was observed in both gBRCAm and gBRCAwt pts. Randomized phase II studies will be needed to further define the efficacy of PI3K/PARP-inhibitor combinations as compared with a PARP inhibitor alone.
Cancer Discovery | 2017
Jiehui Deng; Eric S. Wang; Russell W. Jenkins; Shuai Li; Ruben Dries; Kathleen Yates; Sandeep Chhabra; Wei Huang; Hongye Liu; Amir R. Aref; Elena Ivanova; Cloud P. Paweletz; Michaela Bowden; Chensheng W. Zhou; Grit S. Herter-Sprie; Jessica A. Sorrentino; John E. Bisi; Patrick H. Lizotte; Ashley A. Merlino; Max M. Quinn; Lauren E. Bufe; Annan Yang; Yanxi Zhang; Hua Zhang; Peng Gao; Ting Chen; Megan E. Cavanaugh; Amanda J. Rode; Eric Haines; Patrick J. Roberts
Immune checkpoint blockade, exemplified by antibodies targeting the PD-1 receptor, can induce durable tumor regressions in some patients. To enhance the efficacy of existing immunotherapies, we screened for small molecules capable of increasing the activity of T cells suppressed by PD-1. Here, we show that short-term exposure to small-molecule inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6) significantly enhances T-cell activation, contributing to antitumor effects in vivo, due in part to the derepression of NFAT family proteins and their target genes, critical regulators of T-cell function. Although CDK4/6 inhibitors decrease T-cell proliferation, they increase tumor infiltration and activation of effector T cells. Moreover, CDK4/6 inhibition augments the response to PD-1 blockade in a novel ex vivo organotypic tumor spheroid culture system and in multiple in vivo murine syngeneic models, thereby providing a rationale for combining CDK4/6 inhibitors and immunotherapies.Significance: Our results define previously unrecognized immunomodulatory functions of CDK4/6 and suggest that combining CDK4/6 inhibitors with immune checkpoint blockade may increase treatment efficacy in patients. Furthermore, our study highlights the critical importance of identifying complementary strategies to improve the efficacy of immunotherapy for patients with cancer. Cancer Discov; 8(2); 216-33. ©2017 AACR.See related commentary by Balko and Sosman, p. 143See related article by Jenkins et al., p. 196This article is highlighted in the In This Issue feature, p. 127.
Clinical Cancer Research | 2017
Joyce Liu; Sangeetha Palakurthi; Qing Zeng; Shan Zhou; Elena Ivanova; Wei Huang; Ioannis K. Zervantonakis; Laura M. Selfors; Yiping Shen; Colin C. Pritchard; Mei Zheng; Vilmos Adleff; Eniko Papp; Huiying Piao; Marian Novak; Susan Fotheringham; Gerburg Wulf; Jessie M. English; Paul Kirschmeier; Victor E. Velculescu; Cloud P. Paweletz; Gordon B. Mills; David M. Livingston; Joan S. Brugge; Ursula A. Matulonis; Ronny Drapkin
Purpose: Ovarian cancer is the leading cause of death from gynecologic malignancy in the United States, with high rates of recurrence and eventual resistance to cytotoxic chemotherapy. Model systems that allow for accurate and reproducible target discovery and validation are needed to support further drug development in this disease. Experimental Design: Clinically annotated patient-derived xenograft (PDX) models were generated from tumor cells isolated from the ascites or pleural fluid of patients undergoing clinical procedures. Models were characterized by IHC and by molecular analyses. Each PDX was luciferized to allow for reproducible in vivo assessment of intraperitoneal tumor burden by bioluminescence imaging (BLI). Plasma assays for CA125 and human LINE-1 were developed as secondary tests of in vivo disease burden. Results: Fourteen clinically annotated and molecularly characterized luciferized ovarian PDX models were generated. Luciferized PDX models retain fidelity to both the nonluciferized PDX and the original patient tumor, as demonstrated by IHC, array CGH, and targeted and whole-exome sequencing analyses. Models demonstrated diversity in specific genetic alterations and activation of PI3K signaling pathway members. Response of luciferized PDX models to standard-of-care therapy could be reproducibly monitored by BLI or plasma markers. Conclusions: We describe the establishment of a collection of 14 clinically annotated and molecularly characterized luciferized ovarian PDX models in which orthotopic tumor burden in the intraperitoneal space can be followed by standard and reproducible methods. This collection is well suited as a platform for proof-of-concept efficacy and biomarker studies and for validation of novel therapeutic strategies in ovarian cancer. Clin Cancer Res; 23(5); 1263–73. ©2016 AACR.
Nature Communications | 2017
Haikuo Zhang; Christine Fillmore Brainson; Shohei Koyama; Amanda J. Redig; Ting Chen; Shuai Li; Manav Gupta; Carolina Garcia-De-Alba; Margherita Paschini; Grit S. Herter-Sprie; Gang Lu; Xin Zhang; Bryan P. Marsh; Stephanie Tuminello; Chunxiao Xu; Zhao Chen; Xiaoen Wang; Esra A. Akbay; Mei Zheng; Sangeetha Palakurthi; Lynette M. Sholl; Anil K. Rustgi; David J. Kwiatkowski; J. Alan Diehl; Adam J. Bass; Norman E. Sharpless; Glenn Dranoff; Peter S. Hammerman; Hongbin Ji; Nabeel Bardeesy
Adenosquamous lung tumours, which are extremely poor prognosis, may result from cellular plasticity. Here, we demonstrate lineage switching of KRAS+ lung adenocarcinomas (ADC) to squamous cell carcinoma (SCC) through deletion of Lkb1 (Stk11) in autochthonous and transplant models. Chromatin analysis reveals loss of H3K27me3 and gain of H3K27ac and H3K4me3 at squamous lineage genes, including Sox2, ΔNp63 and Ngfr. SCC lesions have higher levels of the H3K27 methyltransferase EZH2 than the ADC lesions, but there is a clear lack of the essential Polycomb Repressive Complex 2 (PRC2) subunit EED in the SCC lesions. The pattern of high EZH2, but low H3K27me3 mark, is also prevalent in human lung SCC and SCC regions within ADSCC tumours. Using FACS-isolated populations, we demonstrate that bronchioalveolar stem cells and club cells are the likely cells-of-origin for SCC transitioned tumours. These findings shed light on the epigenetics and cellular origins of lineage-specific lung tumours.
Science Translational Medicine | 2016
Suming Wang; Anna Blois; Tina El Rayes; Joyce Liu; Michelle S. Hirsch; Karsten Gravdal; Sangeetha Palakurthi; Diane R. Bielenberg; Lars A. Akslen; Ronny Drapkin; Vivek Mittal; Randolph S. Watnick
A cyclic prosaposin-derived peptide targets ovarian cancer cells through the fatty acid translocase CD36. Running rings around ovarian cancer Although approved drugs for ovarian cancer are available, this remains a difficult disease to overcome, and most ovarian cancer patients cannot be successfully treated, particularly in the setting of advanced disease. Wang et al. determined that prosaposin, a naturally occurring protein with antimetastatic properties, can promote regression of ovarian cancer because of its effects on thrombospondin, another antitumorigenic protein, which targets a receptor called CD36. The authors generated a cyclic peptide modeled on the active site of prosaposin and showed that the new peptide is very effective in treating mice with patient-derived xenografts of metastatic ovarian cancer, suggesting that this peptide is a candidate for future testing in human patients. The vast majority of ovarian cancer–related deaths are caused by metastatic dissemination of tumor cells, resulting in subsequent organ failure. However, despite our increased understanding of the physiological processes involved in tumor metastasis, there are no clinically approved drugs that have made a major impact in increasing the overall survival of patients with advanced, metastatic ovarian cancer. We identified prosaposin (psap) as a potent inhibitor of tumor metastasis, which acts via stimulation of p53 and the antitumorigenic protein thrombospondin-1 (TSP-1) in bone marrow–derived cells that are recruited to metastatic sites. We report that more than 97% of human serous ovarian tumors tested express CD36, the receptor that mediates the proapoptotic activity of TSP-1. Accordingly, we sought to determine whether a peptide derived from psap would be effective in treating this form of ovarian cancer. To that end, we developed a cyclic peptide with drug-like properties derived from the active sequence in psap. The cyclic psap peptide promoted tumor regression in a patient-derived tumor xenograft model of metastatic ovarian cancer. Thus, we hypothesize that a therapeutic agent based on this psap peptide would have efficacy in treating patients with metastatic ovarian cancer.
Nature Communications | 2017
Ioannis K. Zervantonakis; Claudia Iavarone; Hsing Yu Chen; Laura M. Selfors; Sangeetha Palakurthi; Joyce Liu; Ronny Drapkin; Ursula A. Matulonis; Joel D. Leverson; Deepak Sampath; Gordon B. Mills; Joan S. Brugge
The lack of effective chemotherapies for high-grade serous ovarian cancers (HGS-OvCa) has motivated a search for alternative treatment strategies. Here, we present an unbiased systems-approach to interrogate a panel of 14 well-annotated HGS-OvCa patient-derived xenografts for sensitivity to PI3K and PI3K/mTOR inhibitors and uncover cell death vulnerabilities. Proteomic analysis reveals that PI3K/mTOR inhibition in HGS-OvCa patient-derived xenografts induces both pro-apoptotic and anti-apoptotic signaling responses that limit cell killing, but also primes cells for inhibitors of anti-apoptotic proteins. In-depth quantitative analysis of BCL-2 family proteins and other apoptotic regulators, together with computational modeling and selective anti-apoptotic protein inhibitors, uncovers new mechanistic details about apoptotic regulators that are predictive of drug sensitivity (BIM, caspase-3, BCL-XL) and resistance (MCL-1, XIAP). Our systems-approach presents a strategy for systematic analysis of the mechanisms that limit effective tumor cell killing and the identification of apoptotic vulnerabilities to overcome drug resistance in ovarian and other cancers.High-grade serous ovarian cancers (HGS-OvCa) frequently develop chemotherapy resistance. Here, the authors through a systematic analysis of proteomic and drug response data of 14 HGS-OvCa PDXs demonstrate that targeting apoptosis regulators can improve response of these tumors to inhibitors of the PI3K/mTOR pathway.
Cancer immunology research | 2017
Sangeetha Palakurthi; Mari Kuraguchi; Sima Zacharek; Jeff Liu; Dennis M. Bonal; Wei Huang; Kristin Depeaux; Abha Dhaneshwar; Sam Regan; Dyane Bailey; Martha Gowaski; Mei Zheng; Roderick T. Bronson; Catherine Ferrante; Enrique Zudaire; Sylvie Laquerre; Mark Bittinger; Kirschmeier Paul; Kathryn Packman; Raluca Verona; Kwok-Kin Wong; Matthew V. Lorenzi
Targeted therapies against activated oncogenes, such as receptor tyrosine kinases, have significantly prolonged non-small cell lung cancer (NSCLC) patient survival, but the development of resistance limits the durability of clinical response. Genetic alterations which constitutively activate Fibroblast Growth Factor Receptors (FGFR) have been observed in patients with NSCLC. Erdafitinib (JNJ-42756493), an orally bioavailable pan-FGFR inhibitor discovered as part of a collaboration between Janssen and Astex Pharmaceuticals, has been shown to inhibit FGFR signaling pathways resulting in cell death and tumor growth inhibition in both in vitro and in vivo models of FGFR pathway aberration. Further, erdafitinib has been shown to have favorable pharmaceutical properties with manageable side effects in humans and several clinical trials are currently underway. One potential strategy to enhance the durability of response to targeted therapies, such as FGFR inhibitors, is to couple them with immunotherapy. In this setting, T cell responses primed and activated by increased antigen release resulting from the tumor cell targeted therapy could be enhanced and maintained by T-cell directed checkpoint blockade. To test this hypothesis, we evaluated erdafitinib in combination with an anti-programmed death-1 (PD-1) blocking antibody in an autochthonous FGFR2K660N/p53 genetically engineered mouse model (GEMM) of lung cancer, in which tumors develop within the context of an intact immune microenvironment. Cohorts of tumor bearing FGFR2K660N/p53 mutant mice treated with erdafitinib with or without anti-PD-1 showed significant tumor regressions compared to control and anti-PD-1 alone groups. Despite lack of differences in acute tumor responses between erdafitinib monotherapy and combination therapy, we observed significant survival benefit in the combination group erdafitinib alone (median survival 19.7 weeks vs 13.4 weeks, p Citation Format: Sangeetha Palakurthi, Mari Kuraguchi, Sima Zacharek, Jeff Liu, Dennis Bonal, Wei Huang, Kristin Depeaux, Abha Dhaneshwar, Sam Regan, Dyane Bailey, Martha Gowaski, Mei Zheng, Roderick Bronson, Catherine Ferrante, Enrique Zudaire, Sylvie Laquerre, Mark Bittinger, Kirschmeier Paul, Kathryn Packman, Raluca I. Verona, Kwok-Kin Wong, Matthew V. Lorenzi. Improved survival with erdafitinib (JNJ-42756493) and PD-1 blockade mediated by enhancement of anti-tumor immunity in an FGFR2-driven genetically engineered mouse model of lung cancer. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2016 Oct 20-23; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2017;5(3 Suppl):Abstract nr B27.